Next Article in Journal
Dental Caries in Children and Its Relationship with Parenting Styles: A Systematic Review
Previous Article in Journal
Evaluation of Physical and Mental Health in Adults Who Underwent Limb-Lengthening Procedures with Circular External Fixators During Childhood or Adolescence
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unveiling Tumorigenesis Mechanisms and Drug Therapy in Neuroblastoma by Mass Spectrometry Based Proteomics

1
Department of Surgical Oncology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
2
Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou 310052, China
3
State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
4
Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250118, China
5
Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou 310052, China
6
Cancer Center, Zhejiang University, Hangzhou 310058, China
*
Authors to whom correspondence should be addressed.
Children 2024, 11(11), 1323; https://doi.org/10.3390/children11111323
Submission received: 7 October 2024 / Revised: 24 October 2024 / Accepted: 25 October 2024 / Published: 30 October 2024
(This article belongs to the Section Translational Pediatrics)

Abstract

:
Neuroblastoma (NB) is the most common type of extracranial solid tumors in children. Despite the advancements in treatment strategies over the past years, the overall survival rate in patients within the high-risk NB group remains less than 50%. Therefore, new treatment options are urgently needed for this group of patients. Compared with genomic aberrations, proteomic alterations are more dynamic and complex, as well as more directly related to pathological phenotypes and external perturbations such as environmental changes and drug treatments. This review focuses on specific examples of proteomics application in various fundamental aspects of NB research, including tumorigenesis, drug treatment, drug resistance, and highlights potential protein signatures and related signaling pathways with translational values for clinical practice. Moreover, emerging cutting-edge proteomic techniques, such as single cell and spatial proteomics, as well as mass spectrometry imaging, are discussed for their potentials to probe intratumor heterogeneity of NB.

1. Introduction

Neuroblastoma (NB) poses a significant challenge as the most common extracranial pediatric solid tumor, accounting for approximately 7–8% of all childhood malignancies and contributing to approximately 15% of childhood cancer-related mortality [1,2]. It originates from the embryonal sympathoadrenal lineage of the neural crest and can develop anywhere in the sympathetic nervous system, with the adrenal gland being the most common primary site [3,4]. Its heterogeneous nature leads to variable outcomes, from complete regression to aggressive metastasis despite multiple aggressive treatments [5,6]. Patients with low- and intermediate-risk NB have an excellent five-year survival rate of more than 90%. Surgery and reduced chemotherapy are typically provided for this group of patients. However, the five-year survival rate for high-risk NB patients, which account for approximately 60% of all NB cases, remains less than 50% [5,6,7]. Current treatments for high-risk NB include chemotherapy, surgical resection, high-dose chemotherapy with autologous stem cell transplant (ASCT), radiation therapy, immunotherapy, and isotretinoin treatment. Newly introduced trial treatments, such as anti-disialoganglioside (GD2) immunotherapy and 131I-meta-iodobenzylguanidine (131I-MIBG) therapy, has improved overall survival rates, but further advances in treatment are still needed [8,9]. Currently, NB diagnosis relies on a combination approaches of laboratory tests, radiographic imaging, and pathological examination, considering clinical risk factors such as tumor spread, age at diagnosis, and molecular risk factors including MYCN oncogene amplification, diploid DNA contents, and specific segmental chromosomal aberrations, such as 11q deletion, 1p deletion and 17q gain [3,10]. The International Neuroblastoma Risk Group Staging System (INRGSS) was developed to stratify NB patients into four stages (extremely low-risk, low-risk, intermediate-risk, and high-risk groups) for the NB pretreatment risk classification, based on clinical criteria and image-defined risk factors (IDRFs) [11,12].
Recent research has focused on exploring the molecular mechanisms involved in the NB pathogenesis. The investigation of genetic/protein aberrations and signaling pathways is critical for developing potential biomarkers and therapeutic targets for NB treatment [2]. For example, the amplification of MYCN oncogene has been associated with malignant progression, poor prognosis and reduced survival rates, which is detected in approximately 20–30% of all NBs [10,13]. Additionally, segmental chromosome alterations and genetic mutation, such as ALK, PHOX2B, ATRX, PTPN11, ARID1A, or RAS–MAPK and p53 signaling pathway alterations have been confirmed to be associated with worse outcomes and offer potential for precision therapy [9,14]. Compared with genomic aberrations, proteomic alterations are more dynamic and complex, as well as more directly related to pathological phenotypes and response to external perturbations such as environmental changes and drug treatments [15,16]. Some important protein signatures in NB and their related signaling pathways have been proposed as therapeutic targets, such as anaplastic lymphoma kinase (ALK), receptor tyrosine kinase (RTK), and aurora kinases A [17]. For example, alisertib as a regular aurora kinase A inhibitor, has been confirmed to enhance therapeutic effect with 131I-MIBG therapy in high-risk NB [18]. Another recent study illustrated that the suppression of DNA excision repair protein ERCC-6, also known as cockayne syndrome B (CSB), a pleiotropic protein and essential DNA repair factor, could hamper the proliferative, clonogenic, and invasive capabilities in NB, making CSB ablation a promising anticancer strategy for NB therapy [19]. Collectively, the exploration of novel protein targets holds a promising future for novel NB therapy development.
Transcriptomics (RNA-seq) techniques, with their capability for deep coverage and quantitation of low-abundance transcripts, have been widely applied nowadays [20]. However, transcript levels do not always correlate with protein abundance due to post-transcriptional editing and modifications. In contrast to transcriptomics, proteomics-particularly mass spectrometry (MS)-based proteomics-not only play a critical role in characterizing the proteome, assessing protein expression levels, and studying protein degradation and stability, but also offer direct insights into the functional protein landscape, including post-translational modifications, interactions, and localization, thereby revealing important signaling or metabolic pathways associated with biological or pathological processes [21,22]. Despite the widespread utilization of proteomics in various diseases, a comprehensive review of multifaceted applications of proteomics in NB research over the past five years is still lacking. To fill this gap, this review focuses on the application of MS-proteomics in NB studies of tumorigenesis, drug treatment and resistance, as well as other clinical aspects of NB. Furthermore, it discusses recent advancements in proteomics methodologies that may contribute to future studies on NB.

2. Advances in Mass Spectrometry-Based Proteomics Technologies

Proteomics employs protein microarrays, electrophoresis, and mass spectrometry. Although protein microarrays provide a straightforward workflow for large-scale quantitative proteomics, they are limited by the availability of specific and sensitive antibodies. Gel-based electrophoresis, particularly the two-dimensional electrophoresis (2DE), has long been used as a conventional protein separation method, but its low sensitivity and low throughput have restricted its application in contemporary biomedical research [23,24]. In contrast, the MS-based proteomic workflow, which typically analyzes thousands of peptides and proteins in a short time from various types of samples and species, significantly broaden the application scope of proteomics [25,26]. Over the past decade, advancements in sample processing, instrumentation, identification and quantification algorithms, and bioinformatics tools have propelled remarkable progress in MS-based proteomics (Table 1).
Two analytical procedures commonly used in MS-based proteomics are top-down and bottom-up approaches [26]. Bottom-up workflows account for majority of proteomic applications, which measure digested peptides as surrogates for the proteins of interest [27]. Most advanced quantitative proteomics methods are performed using bottom-up proteomics. Untargeted proteomics and targeted proteomics are two main types of data acquisition approaches in MS-based proteomics [28]. In most untargeted studies aimed at providing in-depth and unbiased analysis of the global proteome, the MS quantifies an enormous quantity of mixed peptides that have been pre-separated by liquid chromatography (LC), utilizing either in data-dependent acquisition (DDA) or data-independent acquisition (DIA) modes. In conventional DDA method, the eluted peptides from LC are firstly detected by full scan (MS1), and then a number of peptides are selected sequentially for identification by tandem mass spectrometry (MS/MS or MS2) [29]. As the prevalence of co-eluted peptide precursors usually exceeding the available MS2 spectra and the semi-stochastic selection of the precursor ions, the DDA method is often limited in the proteome coverage and reproducibility [30]. Therefore, DIA proteomics has received more attention in recent years, as it employs predetermined isolation windows including all co-eluding peptides for identification and quantitation, thereby achieving broader proteome coverage with high reproducibility and accuracy [31]. Traditional liquid chromatograph mass spectrometer (LC-MS/MS) based bottom-up proteomics determines peptides based on three dimensions, including LC retention time (RT), mass-charge ratio (m/z) and MS signal intensity. In recent years, the ion mobility was introduced as the fourth separation dimension for peptide ions. This four-dimensional (4D) proteomics greatly improves detection sensitivity with significantly enhanced separation resolution and identification depth [32]. Notably, 4D-proteomics combined with DIA technology, with its precursor ion sampling efficiency and higher precursor identification specificity, can comprehensively improve protein identification ability, detection sensitivity, and data integrity, thus attracting significant attention on contemporary proteomics studies [33,34].
Despite the advancements in DIA-based proteomics, traditional DDA approaches remain prevalent. Label-free quantification (LFQ) proteomics represents a cost-effective method for comparing protein expression across different samples; however, it exhibits relatively lower reproducibility [35]. For sample-wise multiplexing experiments, labeled approaches also have been developed. Stable isotope labeling by amino acids in cell culture (SILAC) is one of the common labeled approaches utilized for in vivo labeling in cell culture system, which achieve dynamic proteomic studies with comprehensive proteome coverage [36]. While for samples from clinical sources or animal models, in vitro labeling approaches such as isobaric tags for relative and absolute quantitation (iTRAQ) and tandem mass tags (TMT) can be employed with their high sample-multiplexing capacity and superior proteome coverage [37,38]. Post-translational modifications (PTMs) encompass phosphorylation, ubiquitination, glycosylation, acetylation, methylation, and others. PTM-proteomics, has become an essential field in understanding protein function and various cellular processes. Due to the low abundance, proteins or peptides containing PTMs usually require to be enriched prior to MS analysis. The PTM-proteomics usually have their suitable enrichment methods before MS/MS detection, including the use of specific antibodies, chemical probes, and affinity chromatography, to enhance the detection of low-abundance PTMs [39,40].
Table 1. Overview of the advanced MS-based proteomics techniques.
Table 1. Overview of the advanced MS-based proteomics techniques.
The Classifications on MS-Based Proteomics TechniquesCharacteristics of MethodsAdvantages
Data acquisition methodsDDA [29]Selected numbers of fragmentation spectra of peptides are measuredWidely used, compatible with all quantification methods
DIA [31]All the precursor ions in the given range are acquired for the fragmentationHigh proteome coverage, high reproducibility
Targeted MS approaches [41]Including selected reaction monitoring, multiple reaction monitoring and parallel reaction monitoringMonitor the biologically important proteins and peptides in a complex mixture, high sensitivity
4D-proteomics with DIA [33,34]Ion mobility is introduced as the fourth separation dimension for peptide ionsHigh coverage, high reproducibility, capable of resolving PTM isoforms
Quantification methodsLabel-free [35]Measurement based on the intensity of peptide signals in MS without the use of labelsCost-effective
SILAC [36]Culturing cells in isotope-containing culture mediaHigh proteome coverage, high dynamic range and quantification accuracy
iTRAQ/TMTs [37,38]Add isobaric tags directly to enzyme-digested peptidesHigh proteome coverage, easy workflow with sample multiplexing
MS-based proteomics on PTMs [39,40]Affinity enrichment process in pretreatmentCapable to explore subtle alteration in PTM level
DDA: data-dependent acquisition; DIA: data-independent acquisition; SILAC: stable isotope labeling by amino acids in cell culture; iTRAQ: isobaric tags for relative and absolute quantitation; TMT: tandem mass tags; PTM: post-translational modifications.
Despite the capabilities of MS-proteomics, the intricate complexity and multidimensionality of the proteome, along with the increasingly abundant datasets it generates, present a formidable analytical challenge for its widespread adoption. In addition to the consistent advancements in algorithmic solutions over the years, artificial intelligence (AI), particularly machine learning (ML) and deep learning (DL), have introduced a novel perspective for tackling this challenge [42]. For instance, DL models have been employed to predict peptide measurements from amino acid sequences, enhancing accuracy and reliability [43]; AI and ML play an indispensable role in biomarker discovery [44,45], outperforming traditional methods in identifying disease-related proteins. However, challenges such as overfitting issues, the black-box nature of DL models that lack transparent traceability [46], as well as privacy and data sharing concerns, necessitate resolution. Nonetheless, the application on integration of multi-omics data and transfer learning techniques driven by AI holds great potential for future breakthroughs [42,47].

3. Proteomics and Neuroblastoma

The high-throughput proteomics techniques have been widely used to generate disease markers and unveil signaling pathways, which are related to critical biological processes and disease pathogenesis [48]. In this review, we provide an overview of the application of proteomics technologies in NB research. A literature search was conducted on the PubMed database from January 2019 to June 2024, using the search criteria encompassing the terms ‘neuroblastoma’ and ‘proteomics’. A total of 212 articles were retrieved, of which approximately 40 articles were relevant to the application of MS-based proteomics in NB. Only original research articles included while review articles, were excluded (2 articles). Articles utilizing NB cell lines as models for non-NB diseases, such as Parkinson’s disease or Alzheimer’s disease, were excluded (139 articles). Articles for NB study but without performing MS-based proteomic strategies during research were excluded (30 articles). These studies presented the advancements in proteomics approaches applied to NB, such as labeled-MS/MS methods and MS-based PTMs. The general workflows of MS-based quantitative proteomics in NB studies were outlined (Figure 1).
In this review, numerous significant protein signatures have been identified by MS-based proteomics over the past five years. Most of these surveyed literatures provided lists of significantly altered proteins and conducted notable analysis on these proteins’ mechanisms. Among these results, over 30 protein targets were emphasized, particularly those involved in established or potential mechanisms related to molecular risk factors and intricate molecular processes in NB. These protein findings significantly contributed to our current understanding of NB tumorigenesis mechanism, as detailed in Table 2. Furthermore, this review highlights the potential of proteomics technology in exploring drug treatment and resistance mechanisms in NB, which is shown in Table 3 and Table 4.

3.1. Proteomics Application in Neuroblastoma Tumorigenesis

NB is a complex and heterogeneous disease, with conventional risk stratification factors depending of the patient age, histological category, MYCN oncogene status, DNA ploidy, and specific segmental chromosomal aberrations, all of which demonstrate a strong correlation with molecular signatures in NB [3]. Genetic/proteomic aberrations such as MYCN, BIRC5, PHOX2B, and LIN28B, and molecular pathways alterations such as ALK signaling, MDM2, PI3K/Akt/mTOR and RAS-MAPK pathways, have potential to supplement traditional clinical risk stratification in NB [2]. Based on mechanisms associated with established molecular risk factors in NB, the proteomics technology has been extensively utilized in NB tumorigenesis investigation.
Table 2. NB study on tumorigenesis mechanisms identified by MS-proteomics and major discoveries in past five years.
Table 2. NB study on tumorigenesis mechanisms identified by MS-proteomics and major discoveries in past five years.
AuthorsSamplesMS-Proteomic MethodsProteins Surveyed by Proteomics Critical NB-Related Proteins Identified Molecular Functions and Mechanisms
Cheng et al. [49]SK-N-BE(2) and 293T cellsLabel-free proteomics;N-Myc interacting proteinsP300 Stabilizing N-Myc
Hsieh et al. [50]Th-MYCN mouse modeliTRAQ-labeled proteomicsProteins response to Aurora kinases inhibitionACADM (+)Inducing β-oxidation metabolism and reducing NB progression
Arlt et al. [51]49 NB biopsies and 13 NB cell linesLabel-free proteomics;Proteins correlated to MYCN-amplified levelPHGDH (+)Inducing serine synthesis and one-carbon metabolism and promoting NB proliferation.
Yang et al. [52]SK-N-BE(2), SK-N-DZ and SK-N-AS cells-lncRNA SNHG1 interacting proteinsMATR3Inducing RNA splicing and enhancing NB progression
Pedersen et al. [53]SH-SY5Y cellsSILAC-labeled proteomics, TMT-labeled phosphoproteomicsProteins response to Cbl proteins depletion IGF1R (+), SHP2 (+), CDK16 (+)Inducing ERK phosphorylation and promoting neurite outgrowth
Funke et al. [54].IMR5 cellLabel-free proteomics; phosphoproteomicsProteins response to NTRK1/TrkA activationLamin A/C/LMNA (+)Inducing stability of nuclear lamina and NB differentiation
Emdal et al. [55]NB1 cellSILAC-labeled, TMT-labeled and label-free proteomicsALK interacting proteinsIRS2Stimulating PI3K-Akt-FoxO3 signaling and promoting NB cell survival
Uckun et al. [56]SK-N-AS and SK-N-BE(2) cellsTMT-labeled proteomicsALK interacting proteinsSHP2Interacting with ALK and promoting NB proliferation
Li et al. [57]Medium of SK-N-SH cell-Secreted proteins from S-type cellsPAI1, SPARC, POSTN and LEG1Activating the STAT3 signaling and protecting cells from apoptosis
Hwang et al. [58]SH-SY5Y cells-Proteins response to LGR5 knockdownhnRNPH3, hnRNPA2B1 (−), and moreActivation of pre-mRNA processing and cell proliferation
Bugara et al. [59]IMR-32 cells-Proteins response to PHLDA1 activationmitochondrion related proteins (+) -
Dhamdhere et al. [60]EVs derived from M1 and 9464D cellsTMT-labeled proteomicsProteins correlated to IGF2BP1 levelSEMA3A (+), SHMT2 (+)Inducing PMN formation and promoting metastasis of NB
Tsakaneli et al. [61]EVs isolated from TET21-N NB cellsLabel-free proteomicsProteins correlated to MYCN-amplified levelPKM2 (+), hexokinase II/HK2 (+)Enhancing histone H3 phosphorylation and promoting the metabolic activity in NB
Fonseka et al. [62].Exosomes from SK-N-BE2 and SH-SY5Y cellsLabel-free proteomicsProteins correlated to MYCN-amplified levelAlix (+), TSG101 (+), FlOT1 (+) and VPS35 (+), and moreRegulating cell communication and signal transduction
Morini et al. [63]Plasma exosomes from HR-NB patients and LR-NB patients and healthy controls-Proteins correlated to NB risk levelNCAM1 (+), NCL (+), LGALS3BP (+), LUM (−), VASP (−), DCN (−), MYH9 (+), FN1 (+), CALR (−), AKAP12 (−) and LTBP1 (+),and more-
Garcia et al. [64].SH-Y5Y cellsLabel-free proteomicsMembrane proteinsNCAM1, L1CAM, EMMPRIN (CD147), ITGB1, ITGAV, CNTFR, and more-
Gangras et al. [65].SH-SY5Y cells -Membrane proteinsNRCAM-
(+): Up-regulation; (−): Down-regulation; ACADM: medium-chain specific acyl-CoA dehydrogenase; PHGDH: phosphoglycerate dehydrogenase; RBPs: RNA binding proteins; IGF1R: Insulin-like growth factor 1 receptor; SHP2: SH2 domain-containing protein tyrosine phosphatase-2; Cbl proteins: casitas B-lineage lymphoma proteins; ALK: anaplastic lymphoma kinase; IRS2: insulin receptor substrate 2; PAI1: plasminogen activator inhibitor 1; SPARC: secreted protein acidic and cysteine rich; POSTN: periostin; LEG1: galectin-1; LGR5: leucine-rich repeat-containing G-protein coupled receptor 5; IGF2BP1: insulin-like growth factor 2 mRNA-binding protein 1; PMN: pre-metastatic niche; SEMA3A: semaphorin 3A; SHMT2: mitochondrial serine hydroxymethyl transferase 2; PKM2: pyruvate kinase M2; HK2: hexokinase II; NCL: nucleolin; LGALS3BP: galectin-3-binding protein; NCAM: neural cell adhesion molecule; LUM: lumican; VASP: vasodilator stimulated phosphoprotein; DCN: decorin; MYH9: myosin-9; FN1: fibronectin; LTBP1: latent-transforming growth factor-beta-binding protein 1; CALR: calreticulin; AKAP12: A-kinase anchor protein 12; NRCAM: neuroglia related cell adhesion molecule.

3.1.1. MYCN-Related Mechanism

MYCN oncogene amplification has been integrated as a key risk factor into risk stratification and therapeutic strategies in NB research for several decades [10]. Its protein product N-Myc, as a transcription factor, was also associated with NB cell proliferation, invasion, angiogenesis and cell differentiation [66]. PTMs have been proven to play a vital role in regulating N-Myc oncoprotein, and corresponding regulatory enzymes could serve as potential targets for modulating N-Myc [67]. Cheng et al. performed endogenous and exogenous N-Myc coimmunoprecipitation (co-IP) followed by LC-MS/MS analysis in 293T cells and SK-N-BE(2) cells, to investigate the PTM residues and interaction proteins of N-Myc. Both acetylation and ubiquitination were identified on lysine 199 of N-Myc [49]. There were 14 potential N-Myc-interacting proteins identified, including several PTM regulatory enzymes. Subsequent investigation focused on histone acetyltransferase P300 as a potential N-Myc-interacting protein, which could acetylate N-Myc and regulate its ubiquitylation level. Further in vitro experiments confirmed that inhibition of P300 suppressed N-Myc level and correlated with favorable survival rates in NB.
Previous studies have shown aurora kinases A directly interacted with and stabilize N-Myc, leading to the NB development [68]. Hsieh et al. conducted iTRAQ-labeled quantitative proteomics in the Th-MYCN mouse model (MYCN overexpressed hemizygous mice with spontaneous NB) and identified 150 significantly differential expressed proteins, following treatment with the aurora kinase inhibitor tozasertib [50]. These proteins were mainly associated with metabolic processes, especially the carbohydrate and fatty acid metabolic pathways. Among these, the highly upregulated medium-chain specific acyl-CoA dehydrogenase (ACADM) was selected for further investigation due to its role in the β-oxidation metabolic pathway [69]. They further showed the high expression of ACADM was associated with a better survival in NB patients. Although the author suggested that inducing β-oxidation by inhibiting aurora kinases A may reduce NB progression mediated by ACADM, the exact mechanism and its relationship with N-Myc network still need to be clarified.
Arlt et al. employed LFQ proteomics analyses on 49 primary NB biopsies and 13 NB cell lines to investigate proteome alterations in various degrees of MYCN expression scores [51]. They identified the expression levels of 248 proteins in tumors and 38 proteins in cell lines were significantly correlated with MYCN expression. Spearman correlation analysis revealed that phosphoglycerate dehydrogenase (PHGDH) was positively correlated with MYCN level, exhibiting the highest affinity. The metabolic flux analyses and other existing databases demonstrated that PHGDH was involved in serine synthesis and one-carbon metabolism [70]. However, further functional experiments confirmed that PHGDH knockout and pharmaceutically inhibition slowed NB proliferation in the short term, but led to resistance to the standard treatment regimens. The chemoresistance was further verified in mouse models with patient-derived NB xenografts following treatment with PHGDH inhibitors and cisplatin.
Long non-coding RNA (lncRNA) SNHG1 is significantly upregulated in NB, and is associated with poor patient prognosis and MYCN status [71]. However, the molecular mechanisms of SNHG1 in NB are still unclear. Using RNA-protein pull-down assay combined with LC−MS/MS, 24 RNA binding proteins (RBPs) interacting with SNHG1 were identified in all three NB cell lines (SKN-BE(2), SK-N-DZ and SK-N-AS) [52]. Among these proteins, DeepBind motif screening and co-expression analysis confirmed the high binding affinity between MATR3 and SNHG1 in all three cell lines. The gene set enrichment analysis (GSEA) and further studies confirmed the binding of MATR3 to SNHG1 was involved in RNA splicing and cell cycle and was associated with poor patient survival. Although this paper provide a new clue that MATR3 could be a prognostic biomarker in high-risk NB, the more mechanism investigations on its relationship to MYCN network are still necessary in the future.

3.1.2. RTK Signaling

RTK signaling has been found involved in mediating NB cell differentiation [72]. Although several key RTK signaling players have been documented, such as ALK and tropomyosin-related kinase A (TrkA), our understanding of the relationship between the RTK superfamily and NB tumorigenesis remains incomplete [73]. Signaling from RTKs is tightly regulated by E3 ubiquitin ligases, such as the casitas B-lineage lymphoma protein-Cbl proteins (Cbl/Cbl-b). Many previous studies found that depletion of Cbl and Cbl-b was associated with induction of neurite outgrowth [74]. To elucidate the global signaling network of Cbl proteins in NB, including its regulated proteins and PTMs, Pedersen et al. employed SILAC-based quantitative proteomics along with TMT-based phosphoproteomics to compare the protein alteration in Cbl/Cbl-b -depleted SH-SY5Y cells in response to various drug treatments [53]. Insulin-like growth factor 1 receptor (IGF1R), as the only RTK protein with significant upregulation in response to Cbl/Cbl-b depletion, was considered to be the target of Cbl/Cbl-b to induce NB cell differentiation. The following phosphoproteomics revealed that both phosphorylation and expression level of SH2 domain-containing protein tyrosine phosphatase-2 (SHP2) responded to Cbl/Cbl-b depletion. The SHP2, which linked to neurite outgrowth according to previous clinical trials [75], along with CDK16, a kinase involved with various signaling pathways [76], as well as IGF1R, were further validated in combination to induce ERK phosphorylation and neurite outgrowth mediated by Cbl protein-depletion.
High expression of the neurotrophin receptor NTRK1/TrkA is associated with favorable outcomes in NB. NTRK1 and its activation by nerve growth factor (NGF) have been shown to induce differentiation in NB [77]. To better understand NTRK1 signaling and its link to MYCN, Funke et al. described global proteome and phosphoproteome alterations in IMR5 cells (a kind of MYCN-amplified cell line) under NGF treatment [54]. A total of 230 differentially regulated proteins and 147 significantly regulated phosphorylation sites were responsive to NTRK1 activation. Among these proteins, the nuclear laminar component Lamin A/C (LMNA) showed a consistent response to NTRK1 activation by NGF over a time-course. Moreover, the increased phosphorylation of LMNA-pS22 was identified as a prominent feature upon NTRK1 activation. Enrichment analysis and other in vitro experiments confirmed the functional relationship between stability of the nuclear lamina and NTRK1 signaling. Although the authors provide new insights into the regulation of NB differentiation mediated by NTRK1 [54], the interdependence of LMNA, NTRK1 and MYCN deserve more investigation.

3.1.3. ALK Signaling

ALK is a famous RTK in the insulin receptor superfamily, and its downstream signaling is related to the RAS/MAPK, PI3K/AKT, and JAK/STAT pathways [78]. ALK gene mutation is also a key genomic aberration, with high correlation to family history and recurrence of NB [79,80]. Oncogenic ALK is reported as druggable NB targets using tyrosine kinase inhibitors (TKIs) [81]. To comprehensively understand ALK signaling and its interactions in NB, Emdal et al. developed a novel integrated proximal proteomics strategy to simultaneously study the ALK interactome and phosphotyrosine interactome [55]. A list of 51 proteins that interacted with ALK was screened out with significant inhibition upon treatment with all three ALK-targeted TKIs. An additional 20 proteins were found directly bound to phosphorylated ALK tyrosine-containing peptides. In this newly constructed ALK interaction network, insulin receptor substrate 2 (IRS2) showing substantial changes in both expression level and tyrosine phosphorylation patterns, was proposed as a central node for ALK signaling transmission. The further abundant database analysis and functional experiments demonstrated the IRS2 as a proximal signaling adaptor promoting NB cell survival via PI3K-Akt-FoxO3 axis. Due to IRS2 being an adaptor protein that has been well characterized in the regulation of cellular glucose metabolism via insulin receptor (INSR) and IGF1R signaling [82], this paper may provide notable insight into the relationship between INSR/IGF1R and ALK signaling in NB.
In a similar study, Uckun et al. employed a biotin-based in vivo proximity labeling approach to identify intracellular partners of ALK [56]. In their study, ALK-BirA* (Escherichia coli biotin ligase) fusion protein were designed to be expressed in Tet-On NB cell lines (SK-N-AS and SK-N-BE(2)). ALK-concatenated BirA* can biotinylate proximal proteins, which can be purified by streptavidin and further analyzed by LC/MS-MS. They found 111 common candidates significantly enriched in ALK-BirA* NB cells compared to BirA* expressing cells. Many of these adaptor proteins have been previously reported to bind to ALK. The author further confirmed that SHP2 was an ALK interactor, and its interaction was enhanced by ALK ligand and was abrogated by lorlatinib (an ALK inhibitor). Further functional experiments confirmed that SHP2 could serve as a downstream target of ALK in NB cells and the combined inhibition of both ALK and SHP2 strongly decreases the proliferation of ALK-driven NB cells. The findings underscore the importance of proximal proteomics analysis in revealing ALK signaling networks, and these novel ALK interactors as potential therapeutic targets in ALK-driven NBs remain to be explored in future.
NB is highly heterogeneous and comprised of a mixture of neuroblastic cells and stromal cells. To test the sensitivity of these cell lines to ALK inhibition and related mechanism, the SK-N-SH cell line was developed as a cellular tool, which is composed of neuroblastic cells (N-type cells) and substrate-adherent cells (S-type cells), and both with ALK mutation in sequencing analysis [83]. To reveal the mechanism that secreted factors from S-type cell conditioned medium (CM) could protect N-type cells from apoptosis induced by the oncogenic ALK inhibitor TAE684., Li et al. used proteomics and identified the S-type CM harbored 74 specifically secreted proteins and most of them were related to biological adhesion [57]. Combined with RNA-Seq dataset and q-PCR verification, they selected the plasminogen activator inhibitor 1 (PAI1), secreted protein acidic and cysteine rich (SPARC), periostin (POSTN) and galectin-1 (LEG1) for further functional analysis. Notably, these four secreted factors were all able to activate STAT3 signaling via ALK-independent pathway, and combined inhibition on ALK and these significant factors could serve as a new direction for high-risk NB treatment.

3.1.4. WNT/β-Catenin Signaling Pathway

WNT/β-catenin signaling has been found to be responsible for NB tumorigenesis [2], and the investigation in Wnt signaling may offer promising targets for therapeutic interventions in NB. Previous study has reported that the activation of leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) promotes Wnt/β-catenin signaling, and plays a critical role in NB cell proliferation [84]. MALDI-TOF-MS-based proteomics identified 12 protein spots altered by LGR5 knockdown in SH-SY5Y cells [58]. Among them, decreased protein expressions of hnRNPA2B1 and hnRNPH3 (hnRNP family) were verified by western blotting. According to this result and other in vivo/vitro experiments, the authors suggested a clue that the LGR5-Wnt/β-catenin signaling-mediated proliferation may be achieved via stimulation of hnRNPs in NB.

3.1.5. Ganglioside GD2 Related Mechanism

Ganglioside GD2, is a glycolipid distributed on NB cell surface, and has been suggested as an immunotherapy target. Dinutuximab (ch14.18), a human/mouse chimeric monoclonal antibody (mAb) against GD2, has demonstrated efficacy to treat high-risk NB [85,86]. The anti-GD2 immunotherapy was proven to significantly increase survival of NB patients, but due to relatively high side effects and relapse, improvement is still needed for clinical implementation [87]. Proteomics profiling could offer comprehensive proteome alteration to investigate related mechanisms of anti-GD2 therapy. For instance, a recent study conducted a multi-dimensional proteomic, transcriptomic, and epigenetic analysis to evaluate GD2-targeting chimeric antigen receptor T cells (CAR-Ts) therapies in NB patient [88]. Additionally, considering that PHLDA1 has been shown with significant activation upon anti-GD2 treatment in NB patients [89], Bugara et al. conducted a study on GD2 therapy in NB utilizing transcriptomic and proteomic analyses of IMR-32 cells. Their findings indicated that mitochondrion related proteome was significantly regulated by PHLDA1 with the most pronounced alteration in expression level [59], thereby providing new protein therapeutic targets for combination strategies in GD2 immunotherapy.

3.1.6. Extracellular Vesicles (EVs)

The contribution of small EVs to NB pathogenesis and resistance to therapies has been demonstrated in recent years [90]. EVs are a class of heterogeneously sized vesicles released by cells for intercellular communication, which including microvesicles (100 to 1000 nm) and exosomes (<200 nm) [90,91]. Recent studies have shown EVs could have a role as signaling factors that impact cancer progression by regulating the tumor microenvironment, immune responses, drug resistance, and oncogenic properties [92].
Previous studies have reported EVs could induce permissive metastatic microenvironment by regulation of pre-metastatic niche (PMN) formation in various cancers, including NB [93]. In the previous study by Dhamdhere et al., Insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) could regulate cell proliferation and impart chemoresistance to human NB cell lines in culture [94]. However, as IGF2BP1 did not exert any discernible impact on the cellular phenotype of the highly aggressive M1 cells (a newly established NB mouse cell line) in culture, it suggests its potential role through secreted factors such as EVs in NB. Using TMT-labeling proteomics to analyze the EVs from IGF2BP1-modulated cells (shIGF2BP1/shNT-M1 and IGF2BP1OE/EGFP control-9464D cells), the authors found the EV-enriched Semaphorin 3A (SEMA3A) and mitochondrial serine hydroxymethyl transferase 2 (SHMT2) were highly correlated with IGF2BP1 level [60]. Further in vivo validation demonstrated stimulation of IGF2BP1-SEMA3A/SHMT2 axis via EVs could promote PMN formation and NB metastasis.
Previous studies also showed that cancer cells expressing MYC proteins could modify the tumor microenvironment, possibly by regulating the secretion or content of EVs [95]. Tsakaneli et al. used LFQ proteomic analysis on EVs isolated from TET21-N cells with or without MYCN expression [61]. They found 111 upregulated and 41 downregulated proteins, primarily consisting of glycolytic enzymes, ribosomal proteins, and extracellular matrix (ECM) interaction proteins in the EVs derived from MYCN-positive cells. To focus on the relevant mechanism of glycolytic enzymes in the EVs, pyruvate kinase M2 (PKM2) and hexokinase II (HK2) were selected for further investigation. The PKM2-enriched EVs were confirmed to induce histone H3 phosphorylation and contribute to the aggressive behavior of NB by promoting the metabolic activity.
Exosomes are released by many cell types and implicated as key mediators in cell-cell communication via transferring their molecular cargo to the target cells [96]. Using LFQ proteomics targeting exosomes from SK-N-BE2 and SH-SY5Y cells with varying MYCN status (amplified versus non-amplified) and western blotting verification, Fonseka et al. revealed 581 proteins higher expressed in SK-N-BE2, such as Alix, TSG101, FlOT1 and VPS35. GO-based analysis highlighted the significant enrichment of proteins secreted by SK-N-BE2 cells in cell communication and signal transduction, as well as ErbB1, mTOR and integrin cell surface interaction signaling pathways, which suggested that MYCN-amplified cell-derived exosomes could regulate various signaling pathways in the recipient cells [62].
A latest similar study conducted a proteomics survey on isolated plasma exosomes from 24 high-risk (HR-NB) patients, 24 low-risk (LR-NB) patients and 24 age-matched healthy controls (CTRL) [63]. Upregulated proteins, such as nucleolin (NCL), galectin-3-binding protein (LGALS3BP) and neural cell adhesion molecule (NCAM), and the downregulated proteins, such as lumican (LUM), vasodilator stimulated phosphoprotein (VASP) and decorin (DCN) were found to be able to discriminate the NB patients from controls; meanwhile, up-regulation of myosin-9 (MYH9), fibronectin (FN1) and latent-transforming growth factor-beta-binding protein 1 (LTBP1), and down-regulation of calreticulin (CALR) and A-kinase anchor protein 12 (AKAP12) were also identified in HR-NB patients as compared to LR-NB patients. All of these interesting proteins were verified by q-PCR detection. These findings suggest that exosomes may play a crucial role in MYCN-driven aggressive NB.

3.1.7. Membrane Proteomics

Comprehensive profilings of membrane proteins and their functions in NB have also gained increasing attention in recent years. The identification of prominent cell surface antigens in NB, such as glypican 2 (GPC2) and B7 homolog 3 (B7-H3, CD276), have paved the way for the advancement in novel targeted therapies for NB [97]. The initial investigation focusing on membrane proteins in NB employed cell surface biotinylation prior to LC-MS/MS analysis, and identified 2557 proteins in the cell surface-enriched fraction [64]. Based on label-free proteomic analyses, multiple cell surface proteins including NCAM1, L1CAM, EMMPRIN (CD147), ITGB1, ITGAV, and CNTFR, which were well characterized in other cancer types, were also considered as potential targets for NB treatment.
In a recent study conducted by Gangras et al., surface proteins were labeled on live SH-SY5Y cells with sulfo-NHS-SS-biotin for downstream enrichment prior to LC-MS/MS analysis [65]. By employing bioinformatic selection based on SurfaceGenie annotation [98] and GO cellular component terms, a total of 298 membrane-associated proteins expressed in the NB-derived SH-SY5Y cell were identified. Significant overlaps were observed between NB cell surfaceome and that of brain and dorsal root ganglion (DRG) neurons, but many protein isoforms were alternatively spliced in NB cells. Using RNA sequencing, the authors confirmed neuroglia related cell adhesion molecule (NRCAM) isoform 4 was absent in typical brain neurons but present in SH-SY5Y cell, making it a specific target candidate for NB therapeutics.

3.2. Proteomics Application in Drug Treatment

3.2.1. Inhibitors of NB Molecular Risk Factors

Proteomic analysis offers valuable insights into changes in protein expression and signaling pathways modulated by drug treatments, thereby significantly advancing the discovery of drug targets and pharmaceutical mechanisms associated with NB. In current research in NB drug treatment, molecular inhibitors of prominent molecular risk factors, are often considered to have new drug development potential. However, the specific mechanisms and action targets of these small molecule drugs require thorough investigation in NB. MS-based proteomics has proven to be an effective tool for comprehensive proteome profiling in pharmaceutical research.
Ataxia telangiectasia and Rad3-related protein (ATR) is an essential kinase that activates cell cycle checkpoint signaling in response to DNA stress and damage and plays an important role in cancer cell survival [99,100]. Szydzik et al. combined RNA-Seq, proteomics and phosphoproteomic analysis data to investigate the effects of ATR inhibition with BAY 1895344 treatment on NB cells (CLB-BAR or CLB-GE) [101]. They validated the target proteins associated with E2F transcription factors, as well as several transcription factors known to be involved in DNA damage response, such as RAD51 and BRCA1/2. Furthermore, phosphoproteomic analysis identified 444 differentially expressed phosphorylated proteins in response to BAY 1895344 treatment and highlighted the ATR target proteins were significantly enriched in DNA repair machinery in NB cells, such as E2F3 and DCK. In vivo/vitro experiments further confirmed its potential to inhibit the growth of ALK-driven NB cells and xenograft, suggesting ATR inhibition could be a promising therapeutic strategy to treat NB, particularly for ALK/MYCN-driven NB patients.
Previous study has proved combined inhibition of ATR (with elimusertib) and ALK (with lorlatinib) could lead to a complete ablation of tumors in ALK-driven NB mouse models [101]. In another research on ATR inhibitor in NB, Borenas et al. compared phosphoproteomics profiling of CLB-BAR cells treated with either ceralasertib or elimusertib, two ATR inhibitors, and showed both drugs led to compensation of ATR inhibition by activation of ataxia-telangiectasia mutated (ATM) and DNA-dependent protein kinase (DNAPK) [102]. Moreover, phosphorylation sites in different PI3-kinase-related protein kinase (PIKK) family members (ATM, ATR, DNAPK, or mTOR) were further focused. This study confirmed that elimusertib or ceralasertib, which leads to a highly similar reduction of ATR and mTOR signaling, may be driven by a compensatory activation response of both ATM and DNAPK, thus showing a clue that combination therapeutic strategy based on ATR inhibitor for the treatment of ALK-positive NB patients.
Various small-molecule TKIs that target ALK are currently used in clinical treatment for both pediatric and adult patient populations [103]. However, the limited success of ALK inhibitor monotherapy raised requirements for further investigation on molecular mechanisms of TKIs-ALK [104]. Phosphoproteomic and RNA-seq analysis was applied to NB cell lines treated with either crizotinib or lorlatinib, the first- and third-generation ALK inhibitors, identifying many differentially phosphorylated components of the ALK signaling pathway [105]. This result provided multiple analyses on protein targets and signaling pathways upon TKI–treatment. Among them, MAPK phosphatase DUSP4 (also known as MKP2) was regulated at the level of both phosphorylation and transcription, and further studies showed that high DUSP4 abundance may enable the maintenance of a delicate ERK signaling balance, suggesting that DUSP4 might be a phosphatase involved in negative feedback for ALK signaling. This in-depth investigation of downstream targets of ALK signaling upon TKI–treatment offers future avenues for ALK-driven NB treatment.
PI3K/Akt/mTOR pathway has been identified as a viable treatment target for aggressive NB [106,107]. However, the intrinsic resistance and acquired resistance to PI3K inhibitors pose significant challenges to treatment efficacy in NB. Increased expression of serine/threonine proviral insertion sites in murine leukemia virus (PIM) kinases has been associated with PI3K inhibitor resistance [108,109]. Mohlin et al. synthesized IBL-302, as a novel highly specific triple PIM, PI3K, and mTOR inhibitor, with the aim of enhancing the NB treatment efficacy [110]. Global RNAseq, proteome, and phosphoproteome analyses on PDX-derived cells revealed several cellular processes were influenced by IBL-302 treatment, including cell apoptosis, programmed cell death, and cell cycle. Notably, caspase-3 and CDK6 were the most significantly differentially upregulated proteins. The multi-omics analyses and sufficient in vivo/vitro experiments shed light on PIM/PI3K/mTOR inhibition as a promising combinatorial therapeutic strategy to improve clinical outcomes in NB.
Mitochondrial division inhibitor 1 (Mdivi-1) is a well-known synthetic compound that disrupts mitochondrial dynamics by targeting dynamin-related protein 1 (Drp1), and is reported to induce programmed cell death in many cancers [111,112]. Wang et al. utilized formaldehyde-H2 or formaldehyde-D2-labeled proteomic and phosphoproteomic analysis to provide comprehensive insight into the biological processes induced by Mdivi-1 in the SK-N-BE(2) [113]. Among the significant modulated proteins, three enzymes essential to serine synthesis were found to be activated upon Mdivi-1 treatment, namely PHGDH, PSAT1, and PSPH. The dephosphorylation of PSMA3-S250 was also focused on in this research with its function of curtailing proliferation.
MiR-204 is known as a tumor suppressor that targets multiple oncogenes, including MYCN, BCL2, NTRK2 and PHOX2B, which are associated with tumor progression and chemoresistance in NB [114,115,116]. However, the administration of miRNA mimics with liposomal nanoparticles as carriers may induce serious immune-mediated side effects in body [117]. Extracellular nanoparticles produced from red blood cells (REPs) can be used as biocompatible nanocarriers for RNA drug delivery [118]. To obtain mechanistic insights of miR-204-loaded REPs (REP-204) therapeutic effects on NB, Chiangjong et al. utilized SWATH-proteomics and bioinformatics analysis on REP-204 treated SK-N-BE2 and SH-SY5Y cells, comparing them to non-treatment conditions [119]. Based on their proteomic data and existing reports, interference with mRNA splicing machinery and SLIT/ROBO pathway were considered as main mechanisms upon REP-204 treatment. This study provided more evidence of anti-NB activity of REP-204, but further verification in in vivo model is warranted.
The nuclear-to-cytoplasmic transport protein Exportin-1 (XPO1) is an essential regulator for nuclear cargo proteins export [120], and is associated with poor prognosis in various adult cancers. Galinski et al. conducted LFQ based proteomic comparison on tumors from 50 patients and demonstrated that XPO1 was one of the most highly abundant proteins associated with the most aggressive diseases, suggesting that XPO1 is a potential prognostic biomarker and therapeutic target in NB [121]. Selinexor is a first class small molecule inhibitor of XPO1, which has shown therapeutic benefits in some adult cancers [122]. In Nguyen et al.’s study, TMT labeling based proteomics and phosphoproteomic analyses were conducted to identify critical target proteins and pathways influenced by selinexor [123]. NB cell line (KCNR, SH-SY5Y) and two MYCN-amplified patient-derived xenografts (PDX) lines with selinexor exposure were tested, and 11,174 unique proteins and 46,755 phosphopeptides were screened. The data revealed prominent increase in p53 protein expression. And the increased phosphorylation at site S315 in response to selinexor exposure was known as the initiating step for p53 degradation [124]. Combined with more in vivo/vitro experiments, it revealed a potential therapeutic benefit using selinexor to increase p53-mediated cytotoxicity in high-risk NB.
The MAPK pathway activity can lead to cell proliferation and tumorigenesis in NB. ERK is a major effector kinase in the MAPK pathway that activates various substrates through phosphorylation, possibly contributing to the chemoresistance and recurrence rate in NB [125,126]. In the work conducted by Yu et al., proteomic analysis was performed on NGP cells treated with ulixertinib, an ERK inhibitor [127]. A list of 72 upregulated and 85 downregulated proteins with ulixertinib treatment were found, which significantly related to the inhibition of cell cycle-related pathways and DNA replication/synthesis pathways. Among these proteins, ulixertinib enhanced the level of pro-apoptotic effector PRUNE2 [128] and cellular proliferation marker MK167 [129], providing new insights into the working mechanism of ulixertinib in NB. However, the author only listed the altered proteins identified by proteomics in this research, without employing enough functional experiments to investigate the mechanisms involving these specific targets with ulixertinib.
Table 3. NB study on drug therapy identified by MS-proteomics and major discoveries in past five years.
Table 3. NB study on drug therapy identified by MS-proteomics and major discoveries in past five years.
AuthorsSamplesMS-Proteomic
Methods
Drug NameInformation of DrugProtein Targets and the Response to DrugRelated Mechanisms of Drug
Szydzik et al. [101]CLB-BAR and CLB-GE cellsTMT-labeled proteomics, phosphoproteomicsBAY 1895344ATR inhibitorRAD51 (−), BRCA1/2 (−), E2F3 (−), DCK (−) and moreInhibiting E2F transcription and DNA repair machinery pathways
Borenas et al. [102].CLB-BAR cellsPhosphoproteomicsCeralasertib, ElimusertibATR inhibitorphospho-ATM (+),phospho-DNAPK (+), and moreDecreasing ATR and mTOR signaling
Van et al. [105]CLB-BAR, CLB-GE, SK-N-AS cellsLabel-free phosphoproteomicsCrizotinib or lorlatinibALK inhibitorDUSP4 (−) and moreInducing feedback loop of ERK and ALK signaling
Mohlin et al. [110]LU-NB-3 cellsLabel-free proteomics, phosphoproteomicsIBL-302PIM, PI3K, mTOR inhibitorCaspase3 (+) and CDK6 (+), and moreInducing programmed cell death and cell cycle signaling
Wang et al. [113].SK-N-BE(2) cellsformaldehyde-H2 and formaldehyde-D2-labled proteomics and phosphoproteomicsMdivi-1Mitochondrial division inhibitorPHGDH (+), PSAT1 (+), PSPH (+), PSMA3 (−), and moreInducing serine synthesis, curtailing proliferation, and more
Chiangjong et al. [119]SK-N-BE2 and SH-SY5Y cells-miR-204-loaded REPsMiR-204 mimic-Suppressing mRNA splicing and SLIT/ROBO pathway
Nguyen et al. [123]KCNR, SH-SY5Y cell lines and PDXs TMT-labeled proteomics, phosphoproteomicsSelinexorXPO1 inhibitorP53 (+), and moreIncreasing p53-mediated cytotoxicity
Yu et al. [127]NGP cellsLabel-free proteomicsUlixertinibERK inhibitorMK167 (+), PRUNE2 (+), and moreInhibiting Cycle-related and DNA replication/synthesis pathways
Chandel et al. [130]SH-SY5Y cellsLabel-free proteomicsEADLimonoidENO1 (−) and HSP90 (−),and morePreventing proliferation and triggering apoptosis in NB
Laghezza et al. [131]SH-SY5Y cellsLabel-free proteomicsHTyr-OLPolyphenolBAG3 (+), HMOX1 (+), CLU (+), HERPUD1 (+), and moreInducing apoptotic signaling pathway
Forbes et al. [132].SH-SY5Y, IMR-32, BE(2)-C, GI-M-EN, SK-N-AS cellsLFQ proteomics, phosphoproteomicL-GlyceraldehydeMonosaccharidePARVA (+), TP53 (+), DTD1 (+), and moreIncreasing oxidoreductase activity and inhibiting cell growth
Lee et al. [133]SH-SY5Y cells Label-free proteomicsMS13Curcumin analogENO1 (−), HSP90AA1 (+), HSP90AB1 (+), TUBB (−), and moreInducing glycolysis and PTM-modification pathways
Morretta et al. [134]HTLA-230 cellsLabel-free proteomicsSTIRUR 41Pyrazolyl-urea and dihydro-imidazo-pyrazolyl-urea compoundsUSP-7 (−)-
Chittavanich et al. [135]RB organoidsLabel-free proteomicsCeftriaxoneThird generation cephalosporin antibiotic for MYCN-driven tumors’ inhibitionDDX3X (−)Inhibiting MYCN translation
Halakos et al. [136]SK-N-SH cellsLabel-free proteomics13-cis RAVitamin A derivative used in the clinic post-chemotherapyICAM1 (+), NEFM (+), CRABP2 (+), PLAT (+) and moreReducing ECM and collagen metabolic process, and promoting neurofilament formation
Halakos et al. [137]SK-N-SH cellsLabel-free proteomicsK777 and 13-cis RAK777: cathepsin inhibitorARHGEF2 (+), B2M (+), CRABP2 (+), NEFL (+), TCF12 (+), APP protein family (+), and moreInducing neuronal differentiation
(+): Up-regulation; (−): Down-regulation; ATR: Ataxia telangiectasia and Rad3-related protein; ALK: anaplastic lymphoma kinase; XPO1: exportin-1; ATM: ataxia-telangiectasia mutated; DNAPK: DNA-dependent protein kinase; Mdivi-1: mitochondrial division inhibitor 1; EAD: epoxyazadiradionex; ENO1: enolase1; HTyr-OL: hydroxytyrosyl oleate; TUBB: tubulin beta chainx; USP-7: ubiquitin carboxyl-terminal hydrolase 7; RB: retinoblastoma; 13-cis RA: 13-cis retinoic acid; The table and corresponding text is organized in alphabetical order of the drug names during each section.

3.2.2. Natural or Synthesis Compounds for NB Treatment

Animal or plant-derived compounds have long been explored for their potential antitumor activities, yet the specific targets and action mechanisms in NB remain largely unknown. Epoxyazadiradione (EAD) is a limonoid, derived from Azadirachta indica [138], with anti-cancer potential against NB. To identify the specific EAD target, a LFQ proteomic approach was conducted on SH-SY5Y cells with or without treatment of EAD [130]. This study revealed that the expression of Enolase1 and HSP90 decreased in a concentration-dependent manner upon treatment with EAD. In vitro experiments further confirmed that EAD can prevent proliferation and trigger apoptosis in NB cells via Enolase1 and HSP90 pathways.
Hydroxytyrosyl oleate (HTyr-OL) has been structural optimized from a polyphenol, hydroxytyrosol (HTyr), derived from extra virgin olive oil, with high lipophilicity and bioavailability. By utilizing LFQ proteomic approach, Laghezza et al. identified 220 unique proteins in NB cells (SH-SY5Y) that responded to HTyr-OL treatment [131]. Among these upregulated proteins, BAG family molecular chaperone regulator 3 (BAG3) has been reported to be implicated in the extrinsic apoptotic signaling pathway, while Heme oxygenase 1 (HMOX1), Clusterin (CLU), and Homocysteine-responsive endoplasmic reticulum-resident ubiquitin-like domain member 1 protein (HERPUD1) are connected to the intrinsic apoptotic signaling pathway triggered by DNA damage. These proteins can be considered as the downstream targets of HTyr-OL treatment for further research.
Glyceraldehyde (GA) is a three-carbon monosaccharide, was reported to inhibit glycolysis and cell growth [139]. Previously published experiment has shown that NB cells were sensitive to GA in vivo, with inhibited glycolysis and cell proliferation [140]. Forbes et al. performed LFQ proteomic analysis of L-GA treated NB cells (SH-SY5Y, IMR-32, BE(2)-C, GI-M-EN, SK-N-AS), and protein signatures associated with oxidoreductase activity were focused [132]. Combined with further phosphoproteomic results, PARVA, TP53, DTD1, MAPT, and CDC25B were regarded with the strongest response following L-GA treatment. However, limitations in in vivo/vitro verification make it difficult to identify the actual molecular functions induced by L-GA in NB.
In the study by Lee et al., the pharmacological mechanism of curcumin analog MS13 was investigated in human glioblastoma U-87 MG and NB SH-SY5Y cells using LFQ proteomic analysis [133]. In both cells, exposure to MS13 resulted in significantly altered expression of heat shock protein HSP 90-alpha (HSP90AA1, HSP90AB1), tubulin beta chain (TUBB), and alphaenolase (ENO1). Reactome pathway database analysis showed “Glycolysis” and “Post-translational protein modification” were the two common pathways identified in both cells. Conclusively, MS13 demonstrates an anti-cancer effect that may indicate its potential use in the management of NB and brain malignancies.
Pyrazolyl-urea and dihydro-imidazo-pyrazolyl-urea compounds (STIRUR 13, STIRUR 41 and BUR 12) have been demonstrated to exert a potent inhibitory effect on interleukin 8 induced chemotaxis of human neutrophils. STIRUR 41 was also considered with ability to inhibit high-risk-NB recurrence, blocking inflammation and angiogenesis in NB [141]. Morretta et al. applied optimized drug affinity-responsive target stability (DARTS) based proteomics using in situ digestion and LC-MS/MS experiments [142,143] to investigate the effects of STIRUR 41 on HTLA-230, a stage-IV NB cell line with MYCN amplification [134]. Several putative targets from different biological pathways were identified, including ubiquitin carboxyl-terminal hydrolase 7 (USP-7), nuclear RNA export factor 1 (NXF1), and pescadillo homolog (PES1). USP-7 was chosen for further analysis due to its strong affinity for STIRUR 41, as confirmed by immunoblotting. The subsequent experiments demonstrated that treatment with STIRUR 41 could inhibit the both expression level and enzymatic activity of USP-7. In this research, authors conducted extensive analysis on the affinity of USP-7 to STIRUR 41, to verify USP-7 as a potential target, but did not further investigate the related mechanisms involved in NB.

3.2.3. Drug Repurposing

Drug repurposing presents an opportunity to develop existing drug molecules for new therapeutic indications [144]. Some clinical drugs that have been performed in other diseases have shown potential for treating NB and warrant further investigation regarding their function and mechanism. Ceftriaxone, an FDA-approved third generation cephalosporin antibiotic, has been demonstrated with the ability to reduce the volume of unexpected retinoblastoma (RB) with MYCN amplification [135,145]. Chittavanich et al. developed a novel therapeutic tool—drug target identification (DTI), which combines affinity-based proteomics and molecular docking approaches, to illustrate action mechanism of ceftriaxone [135]. The GO analysis showed enrichment of “nucleic acid/RNA/mRNA metabolic process”, and the RNA helicases DDX3X was considered as a potential target of ceftriaxone. Ceftriaxone-DDX3X binding was also confirmed by western blotting. Their further study revealed that ceftriaxone may repress MYCN translation via targeting DDX3X. Thereby, ceftriaxone holds promise as a novel strategy for treating MYCN amplified NB in high unmet needs.
Furthermore, deep investigation into downstream targets of existing NB clinical drugs contributes to the rational design of new indications. 13-cis retinoic acid (13-cis RA), is commonly used in the clinic post-chemotherapy due to its differentiating effects, and also has been used in the NB clinical treatment [146]. Halakos et al. used LFQ proteomics analysis in SK-N-SH cells treated with 13-cis RA and identified significantly altered proteins primarily involved in reduced extracellular matrix synthesis (ECM) and collagen metabolic process, as well as increased neurofilament formation, all contributing to development and/or differentiation in NB [136]. Nuclear transporter-CRABP2, adhesion protein-ICAM1 and cytoskeleton protein-NEFM were upregulated, which were considered as important markers of neuronal differentiation. Notably, PLAT, which facilitates neurite outgrowth [147], was also upregulated in 13-cis RA treated cells. This proteomic investigation offered an extensive overview of the proteomic network influenced by 13-cis RA and identified potential targets for more effective treatment of NB.
The use of 13-cis RA has improved outcomes in NB, but relapse is still common in many high-risk patients with multiple aberrant genes. Thereby, drug combinations targeting different aberrant pathways simultaneously were recommended to improve the treatment efficacy and reduce relapse [148,149]. Irreversible cathepsin inhibitor K777 could induce autophagy and reduce tumor volume in NB [150]. Halakos et al. explored the potential of combining 13-cis RA with a cathepsin inhibitor (K777) to enhance the therapeutic efficacy of NB [137]. LFQ proteomics was employed to identify proteins affected by treatment with K777, 13-cis RA, and their combination in SK-N-SH cells, respectively. Multiple analyses comparing these sample groups provided several key protein signatures, which were validated by western blotting. In the result, when K777 was combined with 13-cis RA, ARHGEF2, B2M, CRABP2, NEFL, and TCF12 were increased, suggesting the dual treatment provides a more robust effect on neuronal development; Additionally, the APP protein family, which exhibited the most significant differential alterations identified through proteomic analysis, demonstrated that the combined treatments synergistically enhanced neuronal differentiation in NB.

3.2.4. Proteomic Investigation of Drug Resistance

In current research in NB drug treatment, it is common for high-risk NB patients to develop drug resistance during chemotherapy, even leading to relapse [3]. Therefore, exploring more effective targets that can reduce drug resistance has been the focus in NB research for decades [151]. The application of MS-based proteomic analysis in NB provides deep insights into the molecular underpinnings of resistance mechanisms, which are essential for developing more effective treatments and improving the prognosis for NB patients.
In Wang et al.’s study, SILAC labeling based proteomics was used to compare two stable cell lines with different drug sensitivities [152]. SK-N-BE(1) and SK-N-BE(2) cells were isolated from the bone marrow of the same 2-year-old patient before and after several courses of chemotherapy, respectively. Among more than 460 significantly regulated proteins, Annexin A2 (ANXA2) was highlighted for over 12-fold upregulation in the chemoresistant NB cell line. Both in vitro and in vivo experiments revealed that ANXA2 inhibition induced more apoptosis with chemotherapeutic drugs and attenuated transcriptional activity of NF-κB, a key factor related to drug-resistance in NB [153]. These results suggested ANXA2 could be a prognostic biomarker and therapeutic target for multidrug-resistant NB patients. Furthermore, the authors presented an additional proteome dataset associated with drug resistance that may serve as potential targets for future investigation.
In another similar research, Tang et al. performed TMT-labeled quantitative proteomic analysis on NB tumor samples, comparing patients with a favorable prognosis to those with an unfavorable prognosis [154]. The result showed proteins involved in alternative splicing pathway, particularly the polypyrimidine tract binding protein 2 (PTBP2), were the most significantly upregulated in NB patients with favorable prognosis, which was also validated at mRNA level. Public microarray datasets showed the expression of PTBP2 was lower in NBs with relapse than in those without relapse. Further functional studies verified that PTBP2 induced the chemotactic activity and repolarization of tumor-associated monocytes/Mϕs in NB cells, thereby inhibiting tumor growth. The finding established PTBP2 as an independent and favorable prognostic factor for NB.
Tumor cells in the deeper regions are deprived of oxygen, nutrients, and growth factors. Therefore, cells grown in 2D cultures under serum starvation can serve as a good model system to study the behavior of tumor cells at the molecular level [155]. In the study by Chae et al., the dose-dependent effects of topotecan on human NB cells under various nutrient supply conditions were investigated, and serum-starved SK-N-SH cells showed unique resistance to topotecan [156]. They performed TMT proteomic and phosphoproteomic analysis on the model system to identify topotecan resistance factors. Functional enrichment and network analysis illustrated the increased DNA repair activity and cholesterol-mediated drug efflux, as well as the activated insulin/mTOR signaling may contribute to the resistance. The upregulation of the three representative proteins BLM, HCR24, and phosphorylated IRS-1 were further confirmed by immunoblotting. This study demonstrated the specific mechanism associated with topotecan resistance and provided a novel model for further investigation in drug resistance.
Cisplatin (CDDP) and/or carboplatin are the most common agents used in NB therapy but also plagued with chemoresistance [157]. Merlos Rodrigo et al. performed a proteomic comparison between UKF-NB-4 cells and their cisplatin-resistant counterpart, UKF-NB-4CDDP cells [158]. Among the significantly different proteomic signatures, functional analyses revealed that UKF-NB-4CDDP cells exhibited a marked increase in proteasome activity and demonstrated significant upregulation of various proteasomal complex subunits, including PSME2, PSMB2, PSMD7, PSMB1, PSMD12, PSMB7, PSMA3 and PSMB5. UKF-NB-4CDDP cells also exhibited up-regulation of proteins involved in various aspects of extracellular transport. These findings support further investigation into combination therapy with CDDP by targeting the lysosomal/proteasomal pathways to improve treatment efficacy in chemoresistant high-risk NB.
Table 4. NB study on drug resistance identified by MS-proteomics and major discoveries in past five years.
Table 4. NB study on drug resistance identified by MS-proteomics and major discoveries in past five years.
AuthorsSamplesMS Proteomic
Methods
Drugs Protein Targets and the Response to Chemoresistant StatusRelated Mechanisms Associated with Drug Resistance
Wang et al. [152]SK-N-BE(1) and SK-N-BE(2) cellsSILAC-labeled MS-ANXA2 (+)Inducing NF-κB signaling and drug resistance
Tang et al. [154]Tumors from NB patientsTMT-labled MS-PTBP2 (−)Inducing alternative splicing pathway and repolarization of monocytes
Chae et al. [156]SK-N-SH cellsTMT-labeled proteomics, phosphoproteomicsTopotecanBLM (+), HCR24 (+), and phospho-IRS1 (+)Activating DNA repair, cholesterol-mediated activity, and insulin/mTOR signaling
Merlos Rodrigo et al. [158]UKF-NB-4 cellsLabel-free proteomicsCisplatinProteasomal complex subunits (+)Activating lysosomal/proteasomal pathways
(+): Up-regulation; (−): Down-regulation; ANXA2: annexin A2; PTBP2: polypyrimidine tract binding protein 2.

4. Proteomics Application in Intratumor Heterogeneity of NB

4.1. Genetic and Proteomic Level Analysis in NB Intratumor Heterogeneity

Intratumor heterogeneity (ITH) refers to the existence of diverse subpopulations of cells within a single tumor, displaying variations at the genetic, epigenetic, and proteomic levels [159,160]. Multiple cellular subpopulations are easy to be found in high-risk NB, with genetic and epigenetic differences [161]. In early studies, the NB tumor cell phenotypes were described into three types: N-type (neuronal), S-type (substrate adherent), and I-type (intermediary) cells, with different cell culture behavior and protein expression pattern, such as membrane-GD2 and nuclear-calcyclin [83,162]. Recent study has regrouped NB into four distinct epigenetic subtypes based on the super-enhancer landscape and MYCN amplification status: MYCN-amplified high-risk, MYCN non-amplified high-risk, MYCN non-amplified low-risk, and mesenchymal-type (MES). The fourth type exhibiting mesenchymal characteristics, was induced by RAS activation and enriched in relapsed cases [163]. Significant efforts have been dedicated to characterizing genomic alterations and oncogenic pathways through comprehensive DNA and RNA sequencing analyses involving thousands of cases. There are many molecular signatures well-characterized in NB cellular subtype detection and risk definition, including MYCN amplification, mutations in ALK, PHOX2B, epigenetic factor such as ATRX, and TERT, and structural chromosomal changes [2,79,164]. Furthermore, the molecular mechanisms underlying trans-differentiation in NB may elucidate the observed plasticity and intratumoral heterogeneity, including activation of the NOTCH signaling pathway, RAS signaling, and inactivation of ARID1A [165,166,167].
MS-based profiling has revealed extensive proteomic heterogeneity within NB tumors. In a study on genetic ITH in NB, ten orthotopic xenograft (PDOX) models from a single primary tumor were subjected to comprehensive proteomic and phosphoproteomic analyses, revealing the spatial expression patterns and diverse molecular profiles involved in stromal contribution, neuronal differentiation, and axon guidance [168]. However, the limited availability of tissues hampers PDX models developing as an important tool for ITH assessment in NB. Furthermore, the differential protein expression patterns among various NB subclones, highlighting the functional diversity within the tumor microenvironment. For instance, recent studies have shown that exosomes could be one way by different clonal populations to interact with each other and shape cellular plasticity [62], and proteomics analysis compared the protein profiling of exosome may contribute to understanding of proteomic diversity in ITH. Moreover, MS-based proteomic approaches can be utilized to investigate distinct PTM patterns, including phosphorylation and ubiquitination, as well as protein stability and interactions, all of which contribute to the functional diversity of NB subclones

4.2. The Application of Emerging Proteomic Techniques in NB Intratumor Heterogeneity

The emerging instrumental and bioinformatic tools with cell-type or spatial resolving power have facilitated the exploration of spatial proteome profiles relevant to tumor heterogeneity. Compared to traditional bulk proteomics, which provides an average assessment of protein expression profile, the single cell proteomics (SCP) achieves cell specific proteome data revealing functional diversity in tissue microenvironment [169]. The primary challenge in SCP lies in separating the single cell from the tissue samples with low protein loss [170]. The current methodologies employed for obtaining single cells or cell groups prior to SCP analysis, include fluorescence-activated cell sorting (FACS), magnetic-activated cell sorting (MACS), laser capture microdissection (LCM), capillary electrophoresis and manual cell picking/micromanipulation [171]. Nevertheless, the utilization of SCP technology in NB research is presently restricted, thus requiring the further development.
On the other hand, proteomic patterns can be portrayed directly on tissue slides without isolating cells, therefore preserving critical spatial traits of the microenvironment. This is achieved by mass spectrometry imaging (MSI) using in situ ionization techniques, such as matrix-assisted laser desorption ionization (MALDI), to delineate the spatial distribution of analytes, including drugs, metabolites and peptides [172]. Ryu et al. used MALDI-MSI to analyze the distribution of alectinib (an ALK inhibitor) in NB1 (with ALK amplification) and SK-N-FI (ALK wild-type) NB xenograft tissues [173]. The intra-tumoral abundance of alectinib was quantified using ion signal intensities from MALDI-MSI and normalized by LC-MS/MS. Their findings demonstrated that the distribution pattern of alectinib within xenografted tumors is characterized by heterogeneity, indicating that the distribution of drugs in patient tumors is more complex than previously thought. Therefore, MSI can serve as a valuable tool for assessing drug exposure during early clinical trials and clinical practice, while also identifying regions with low penetration levels that may contribute to drug failure or resistance. In another study, Wu et al. performed MALDI-MSI to inquire spatial heterogeneity of peptides in NB tissues as factors for high and low/intermediate risk classification [174]. Peptides from AHNAK nucleoprotein and collapsin response mediator protein 1 (CRMP1) were identified, which were associated with divergent risks and were further validated immunohistochemically. This study highlights the capability of MALDI-MSI combined with univariate and multivariate analysis strategies to identify spatially risk-associated peptide signatures in NB tissues, thereby providing new biological insights into NB ITH.

5. Conclusions and Future Perspective

NB is the most common extracranial cancer affecting children. To improve clinical outcomes for children with high-risk NB, the MS-based proteomic approach, with the dramatic advance of application in recent years, has become a mainstream approach for exploring molecular mechanisms in NB. This article reviews the application of proteomics in NB over the past five years, focusing on diverse aspects such as tumorigenesis, drug treatment and resistance in NB and highlighting many promising protein signatures and molecular mechanisms that may promote clinical research in NB (Figure 2). Furthermore, based on the conclusions drawn from these referenced articles, Table 5 provides a detailed representation of the main protein signatures associated with potential molecular risk factors and risk-related mechanisms of NB. MYCN network and status, RTK/ALK signaling pathway, PI3K/Akt/mTOR pathway, WNT/β-catenin signaling, RAS/MAPK signaling pathway, and ATR signaling are highlighted as key risk factors in NB in our review.
Many of these protein findings are considered indispensable in NB tumorigenesis and prognosis, as they have been identified by multiple researchers and implicated in intersecting signaling pathways or drug mechanisms. For instance, SHP2 is an oncogenic tyrosine phosphatase encoded by PTPN11, exhibiting a significant mutation frequency (3.4% of NB cases) and extensively studied as a potential target for inhibiting NB tumor cell growth [175]. Hsp90 is indispensable for the stability and function of many client proteins, emerging as an important target in a variety of cancers [176], including NB growth [177]. PHGDH is an essential enzyme for de novo serine synthesis and has been found to correlate with poor prognosis and potential therapeutic option in many cancers, including MYCN-amplified NB [178]. NCAM is a neural cell adhesion molecule linked to metastatic capacity and aggressive cancer progression in NB. Not only NCAM but also other cell adhesion molecules (CAMs) play pivotal roles in multiple biological processes, necessitating research on CAMs to explore novel targeted therapies in NB. On the other hand, certain distinct protein findings identified across studies may be involved in common biological processes and signaling pathways. For example, IGF1R, IRS2, SHP2 and NTRK1 are prominent molecular signatures in RTK/ALK signaling associated with high-risk NB; PIKK family [179], PRUNE2 [128], E2F transcription factors [180], RAD51, BRCA1/2, BLM and HCR24 [156] are key components associated with DNA damage/repair machinery and cell cycle progression; PKM2, hexokinase II [61] and ENO1 [181] are glycolytic enzymes contributing to glycolysis metabolism; ICAM1, NEFM, CRABP2, PLAT are recognized as important components in neurofilament formation and neuronal differentiation [136,137].
Notably, MS-based proteomics methods are commonly customized for different sample types (cell lines, tissue from human or mouse model, biological fluids), and perturbations (gene overexpression or depletion, inhibitor or activator treatments). For some specific proteomics applications, special optimizations are needed. For instance, the co-IP and RNA pull-down assay combined with LC-MS/MS were conventional methods for investigating protein/RNA interacting proteins [49,52]. Integrated proximal proteomics (IPP) strategy [55] and proximity-dependent biotin identification (BioID)-Screening method [56] are novel proteomics strategies mentioned for interactome investigation. What is more, the protein extraction and purification methods in pre-treatment are critical for proteomics study, particular in PTMs identification, EVs or membrane proteome investigation.
Nonetheless, there are significant obstacles that need to be addressed for proteomics techniques to transition from limited experimental use to broader into more practical clinical applications in NB. The tumor heterogeneity poses a significant challenge. SCP and spatial proteomics are new approaches that deserve further development in the future. Moreover, the identification of low-abundance proteins remains a hurdle in deep proteomic investigations. Current techniques such as centrifugal ultrafiltration, organic solvent precipitation, electrophoresis, and chromatography can be employed to enhance the detection of low-abundance proteins, and more development in this field should be expected [182]. Furthermore, while proteomics has facilitated the discovery and evaluation of potential disease biomarkers using large-scale clinical sample cohorts in many other diseases, its application in NB diagnosis and prognosis research is still relatively nascent, potentially hampered by the limited availability of clinical NB samples. Encouragingly, advancements of new hardware with higher sensitivity, and novel sample preparation workflows designed for minimal sample requirement, are anticipated to drive more proteomics-driven studies in this area and eventually lead to exciting breakthroughs for NB treatment in the future.

Author Contributions

The first author, K.R., had the original idea of writing a review and wrote the first draft. Corresponding authors, T.T. and Z.S., read and reviewed all drafts of the article and assisted the first author to design the review work. Author Y.W. and M.Z. put together tables and figures on the various revised versions of the review and checked citations and references. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the National Key Research and Development Program (2022YFA1303801), National Natural Science Foundation of China (U20A200603, 82370612, 32270853), Independent Foundation of the State Key Laboratory for Diagnosis and Treatment of Infectious Diseases (zz202311), Shandong Provincial Laboratory Project (SYS202202), Research Project of Jinan Microecological Biomedicine Shandong Laboratory (JNL-2022030C), “Pioneer” and “Leading Goose” R&D Program of Zhejiang Province (2024C03181).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to acknowledge Dan Zhou of Zhejiang University for thorough review of the manuscript.

Conflicts of Interest

The authors declare they have no conflicting financial interests.

References

  1. Maris, J.M.; Hogarty, M.D.; Bagatell, R.; Cohn, S.L. Neuroblastoma. Lancet 2007, 369, 2106–2120. [Google Scholar] [CrossRef] [PubMed]
  2. Zafar, A.; Wang, W.; Liu, G.; Wang, X.; Xian, W.; McKeon, F.; Foster, J.; Zhou, J.; Zhang, R. Molecular targeting therapies for neuroblastoma: Progress and challenges. Med. Res. Rev. 2021, 41, 961–1021. [Google Scholar] [CrossRef] [PubMed]
  3. Mlakar, V.; Jurkovic Mlakar, S.; Lopez, G.; Maris, J.M.; Ansari, M.; Gumy-Pause, F. 11q deletion in neuroblastoma: A review of biological and clinical implications. Mol. Cancer 2017, 16, 114. [Google Scholar] [CrossRef] [PubMed]
  4. Maris, J.M. Recent advances in neuroblastoma. N. Engl. J. Med. 2010, 362, 2202–2211. [Google Scholar] [CrossRef]
  5. Whittle, S.B.; Smith, V.; Doherty, E.; Zhao, S.; McCarty, S.; Zage, P.E. Overview and recent advances in the treatment of neuroblastoma. Expert Rev. Anticancer Ther. 2017, 17, 369–386. [Google Scholar] [CrossRef] [PubMed]
  6. Baker, D.L.; Schmidt, M.L.; Cohn, S.L.; Maris, J.M.; London, W.B.; Buxton, A.; Stram, D.; Castleberry, R.P.; Shimada, H.; Sandler, A.; et al. Outcome after reduced chemotherapy for intermediate-risk neuroblastoma. N. Engl. J. Med. 2010, 363, 1313–1323. [Google Scholar] [CrossRef] [PubMed]
  7. Kholodenko, I.V.; Kalinovsky, D.V.; Doronin, I.I.; Deyev, S.M.; Kholodenko, R.V. Neuroblastoma Origin and Therapeutic Targets for Immunotherapy. J. Immunol. Res. 2018, 2018, 7394268. [Google Scholar] [CrossRef]
  8. Swift, C.C.; Eklund, M.J.; Kraveka, J.M.; Alazraki, A.L. Updates in Diagnosis, Management, and Treatment of Neuroblastoma. Radiographics 2018, 38, 566–580. [Google Scholar] [CrossRef]
  9. Qiu, B.; Matthay, K.K. Advancing therapy for neuroblastoma. Nat. Rev. Clin. Oncol. 2022, 19, 515–533. [Google Scholar] [CrossRef]
  10. Schleiermacher, G.; Janoueix-Lerosey, I.; Delattre, O. Recent insights into the biology of neuroblastoma. Int. J. Cancer 2014, 135, 2249–2261. [Google Scholar] [CrossRef]
  11. Shawraba, F.; Hammoud, H.; Mrad, Y.; Saker, Z.; Fares, Y.; Harati, H.; Bahmad, H.F.; Nabha, S. Biomarkers in Neuroblastoma: An Insight into Their Potential Diagnostic and Prognostic Utilities. Curr. Treat. Options Oncol. 2021, 22, 102. [Google Scholar] [CrossRef] [PubMed]
  12. Monclair, T.; Brodeur, G.M.; Ambros, P.F.; Brisse, H.J.; Cecchetto, G.; Holmes, K.; Kaneko, M.; London, W.B.; Matthay, K.K.; Nuchtern, J.G.; et al. The International Neuroblastoma Risk Group (INRG) staging system: An INRG Task Force report. J. Clin. Oncol. 2009, 27, 298–303. [Google Scholar] [CrossRef] [PubMed]
  13. Seeger, R.C.; Brodeur, G.M.; Sather, H.; Dalton, A.; Siegel, S.E.; Wong, K.Y.; Hammond, D. Association of multiple copies of the N-myc oncogene with rapid progression of neuroblastomas. N. Engl. J. Med. 1985, 313, 1111–1116. [Google Scholar] [CrossRef] [PubMed]
  14. Park, J.A.; Cheung, N.V. Targets and Antibody Formats for Immunotherapy of Neuroblastoma. J. Clin. Oncol. 2020, 38, 1836–1848. [Google Scholar] [CrossRef] [PubMed]
  15. Cheung, C.H.Y.; Juan, H.F. Quantitative proteomics in lung cancer. J. Biomed. Sci. 2017, 24, 37. [Google Scholar] [CrossRef]
  16. Pandey, A.; Mann, M. Proteomics to study genes and genomes. Nature 2000, 405, 837–846. [Google Scholar] [CrossRef]
  17. Smiles, W.J.; Catalano, L.; Stefan, V.E.; Weber, D.D.; Kofler, B. Metabolic protein kinase signalling in neuroblastoma. Mol. Metab. 2023, 75, 101771. [Google Scholar] [CrossRef]
  18. Kumar, P.; Koach, J.; Nekritz, E.; Mukherjee, S.; Braun, B.S.; DuBois, S.G.; Nasholm, N.; Haas-Kogan, D.; Matthay, K.K.; Weiss, W.A.; et al. Aurora Kinase A inhibition enhances DNA damage and tumor cell death with (131)I-MIBG therapy in high-risk neuroblastoma. EJNMMI Res. 2024, 14, 54. [Google Scholar] [CrossRef]
  19. Paccosi, E.; Costantino, M.; Balzerano, A.; Filippi, S.; Brancorsini, S.; Proietti-De-Santis, L. Neuroblastoma Cells Depend on CSB for Faithful Execution of Cytokinesis and Survival. Int. J. Mol. Sci. 2021, 22, 10070. [Google Scholar] [CrossRef]
  20. Wang, Z.; Gerstein, M.; Snyder, M. RNA-Seq: A revolutionary tool for transcriptomics. Nat. Rev. Genet. 2009, 10, 57–63. [Google Scholar] [CrossRef]
  21. Martinez-Rodriguez, F.; Limones-Gonzalez, J.E.; Mendoza-Almanza, B.; Esparza-Ibarra, E.L.; Gallegos-Flores, P.I.; Ayala-Lujan, J.L.; Godina-Gonzalez, S.; Salinas, E.; Mendoza-Almanza, G. Understanding Cervical Cancer through Proteomics. Cells 2021, 10, 1854. [Google Scholar] [CrossRef] [PubMed]
  22. Pascovici, D.; Wu, J.X.; McKay, M.J.; Joseph, C.; Noor, Z.; Kamath, K.; Wu, Y.; Ranganathan, S.; Gupta, V.; Mirzaei, M. Clinically Relevant Post-Translational Modification Analyses-Maturing Workflows and Bioinformatics Tools. Int. J. Mol. Sci. 2018, 20, 16. [Google Scholar] [CrossRef] [PubMed]
  23. Conrad, D.H.; Goyette, J.; Thomas, P.S. Proteomics as a method for early detection of cancer: A review of proteomics, exhaled breath condensate, and lung cancer screening. J. Gen. Intern. Med. 2008, 23 (Suppl. 1), 78–84. [Google Scholar] [CrossRef] [PubMed]
  24. Alessandro, R.; Fontana, S.; Kohn, E.; De Leo, G. Proteomic strategies and their application in cancer research. Tumori 2005, 91, 447–455. [Google Scholar] [CrossRef]
  25. Zhang, Z.; Wu, S.; Stenoien, D.L.; Pasa-Tolic, L. High-throughput proteomics. Annu. Rev. Anal. Chem. 2014, 7, 427–454. [Google Scholar] [CrossRef]
  26. Duong, V.A.; Lee, H. Bottom-Up Proteomics: Advancements in Sample Preparation. Int. J. Mol. Sci. 2023, 24, 5350. [Google Scholar] [CrossRef]
  27. Miller, R.M.; Smith, L.M. Overview and considerations in bottom-up proteomics. Analyst 2023, 148, 475–486. [Google Scholar] [CrossRef]
  28. Ryu, J.; Thomas, S.N. Quantitative Mass Spectrometry-Based Proteomics for Biomarker Development in Ovarian Cancer. Molecules 2021, 26, 2674. [Google Scholar] [CrossRef]
  29. Li, K.W.; Gonzalez-Lozano, M.A.; Koopmans, F.; Smit, A.B. Recent Developments in Data Independent Acquisition (DIA) Mass Spectrometry: Application of Quantitative Analysis of the Brain Proteome. Front. Mol. Neurosci. 2020, 13, 564446. [Google Scholar] [CrossRef]
  30. Elias, J.E.; Gygi, S.P. Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nat. Methods 2007, 4, 207–214. [Google Scholar] [CrossRef]
  31. Gillet, L.C.; Navarro, P.; Tate, S.; Rost, H.; Selevsek, N.; Reiter, L.; Bonner, R.; Aebersold, R. Targeted data extraction of the MS/MS spectra generated by data-independent acquisition: A new concept for consistent and accurate proteome analysis. Mol. Cell Proteom. 2012, 11, O111.016717. [Google Scholar] [CrossRef] [PubMed]
  32. Yang, J.; Liu, Q.; Yu, B.; Han, B.; Yang, B. 4D-quantitative proteomics signature of asthenozoospermia and identification of extracellular matrix protein 1 as a novel biomarker for sperm motility. Mol. Omics 2022, 18, 83–91. [Google Scholar] [CrossRef] [PubMed]
  33. Pei, Y.; Chen, S.; Zhang, Y.; Olga, V.; Li, Y.; Diao, X.; Zhou, H. Coral and it’s symbionts responses to the typical global marine pollutant BaP by 4D-Proteomics approach. Environ. Pollut. 2022, 307, 119440. [Google Scholar] [CrossRef] [PubMed]
  34. Meier, F.; Brunner, A.D.; Frank, M.; Ha, A.; Bludau, I.; Voytik, E.; Kaspar-Schoenefeld, S.; Lubeck, M.; Raether, O.; Bache, N.; et al. diaPASEF: Parallel accumulation-serial fragmentation combined with data-independent acquisition. Nat. Methods 2020, 17, 1229–1236. [Google Scholar] [CrossRef] [PubMed]
  35. Zhang, B.; VerBerkmoes, N.C.; Langston, M.A.; Uberbacher, E.; Hettich, R.L.; Samatova, N.F. Detecting differential and correlated protein expression in label-free shotgun proteomics. J. Proteome Res. 2006, 5, 2909–2918. [Google Scholar] [CrossRef]
  36. Ong, S.E.; Blagoev, B.; Kratchmarova, I.; Kristensen, D.B.; Steen, H.; Pandey, A.; Mann, M. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol. Cell Proteom. 2002, 1, 376–386. [Google Scholar] [CrossRef]
  37. Swiatly, A.; Horala, A.; Matysiak, J.; Hajduk, J.; Nowak-Markwitz, E.; Kokot, Z.J. Understanding Ovarian Cancer: iTRAQ-Based Proteomics for Biomarker Discovery. Int. J. Mol. Sci. 2018, 19, 2240. [Google Scholar] [CrossRef]
  38. Zhang, L.; Elias, J.E. Relative Protein Quantification Using Tandem Mass Tag Mass Spectrometry. Methods Mol. Biol. 2017, 1550, 185–198. [Google Scholar]
  39. Srinivasan, A.; Sing, J.C.; Gingras, A.C.; Rost, H.L. Improving Phosphoproteomics Profiling Using Data-Independent Mass Spectrometry. J. Proteome Res. 2022, 21, 1789–1799. [Google Scholar] [CrossRef]
  40. Sahu, I.; Zhu, H.; Buhrlage, S.J.; Marto, J.A. Proteomic approaches to study ubiquitinomics. Biochim. Biophys. Acta Gene Regul. Mech. 2023, 1866, 194940. [Google Scholar] [CrossRef]
  41. Peterson, A.C.; Russell, J.D.; Bailey, D.J.; Westphall, M.S.; Coon, J.J. Parallel reaction monitoring for high resolution and high mass accuracy quantitative, targeted proteomics. Mol. Cell Proteom. 2012, 11, 1475–1488. [Google Scholar] [CrossRef] [PubMed]
  42. Mann, M.; Kumar, C.; Zeng, W.F.; Strauss, M.T. Artificial intelligence for proteomics and biomarker discovery. Cell Syst. 2021, 12, 759–770. [Google Scholar] [CrossRef] [PubMed]
  43. Zhou, X.X.; Zeng, W.F.; Chi, H.; Luo, C.; Liu, C.; Zhan, J.; He, S.M.; Zhang, Z. pDeep: Predicting MS/MS Spectra of Peptides with Deep Learning. Anal. Chem. 2017, 89, 12690–12697. [Google Scholar] [CrossRef] [PubMed]
  44. Geyer, P.E.; Voytik, E.; Treit, P.V.; Doll, S.; Kleinhempel, A.; Niu, L.; Muller, J.B.; Buchholtz, M.L.; Bader, J.M.; Teupser, D.; et al. Plasma Proteome Profiling to detect and avoid sample-related biases in biomarker studies. EMBO Mol. Med. 2019, 11, e10427. [Google Scholar] [CrossRef] [PubMed]
  45. Ge, J.; Ge, J.; Tang, G.; Xiong, D.; Zhu, D.; Ding, X.; Zhou, X.; Sang, M. Machine learning-based identification of biomarkers and drugs in immunologically cold and hot pancreatic adenocarcinomas. J. Transl. Med. 2024, 22, 775. [Google Scholar] [CrossRef]
  46. Lee, T.; Natalwala, J.; Chapple, V.; Liu, Y. A brief history of artificial intelligence embryo selection: From black-box to glass-box. Hum. Reprod. 2024, 39, 285–292. [Google Scholar] [CrossRef]
  47. Sen, P.; Lamichhane, S.; Mathema, V.B.; McGlinchey, A.; Dickens, A.M.; Khoomrung, S.; Oresic, M. Deep learning meets metabolomics: A methodological perspective. Brief. Bioinform. 2021, 22, 1531–1542. [Google Scholar] [CrossRef]
  48. Cifani, P.; Kentsis, A. Towards comprehensive and quantitative proteomics for diagnosis and therapy of human disease. Proteomics 2017, 17, 1600079. [Google Scholar] [CrossRef]
  49. Cheng, C.; He, T.; Chen, K.; Cai, Y.; Gu, Y.; Pan, L.; Duan, P.; Wu, Y.; Wu, Z. P300 Interacted with N-Myc and Regulated Its Protein Stability via Altering Its Post-Translational Modifications in Neuroblastoma. Mol. Cell Proteom. 2023, 22, 100504. [Google Scholar] [CrossRef]
  50. Hsieh, C.H.; Cheung, C.H.Y.; Liu, Y.L.; Hou, C.L.; Hsu, C.L.; Huang, C.T.; Yang, T.S.; Chen, S.F.; Chen, C.N.; Hsu, W.M.; et al. Quantitative Proteomics of Th-MYCN Transgenic Mice Reveals Aurora Kinase Inhibitor Altered Metabolic Pathways and Enhanced ACADM To Suppress Neuroblastoma Progression. J. Proteome Res. 2019, 18, 3850–3866. [Google Scholar] [CrossRef]
  51. Arlt, B.; Zasada, C.; Baum, K.; Wuenschel, J.; Mastrobuoni, G.; Lodrini, M.; Astrahantseff, K.; Winkler, A.; Schulte, J.H.; Finkler, S.; et al. Inhibiting phosphoglycerate dehydrogenase counteracts chemotherapeutic efficacy against MYCN-amplified neuroblastoma. Int. J. Cancer 2021, 148, 1219–1232. [Google Scholar] [CrossRef] [PubMed]
  52. Yang, T.W.; Sahu, D.; Chang, Y.W.; Hsu, C.L.; Hsieh, C.H.; Huang, H.C.; Juan, H.F. RNA-Binding Proteomics Reveals MATR3 Interacting with lncRNA SNHG1 To Enhance Neuroblastoma Progression. J. Proteome Res. 2019, 18, 406–416. [Google Scholar] [CrossRef] [PubMed]
  53. Pedersen, A.K.; Pfeiffer, A.; Karemore, G.; Akimov, V.; Bekker-Jensen, D.B.; Blagoev, B.; Francavilla, C.; Olsen, J.V. Proteomic investigation of Cbl and Cbl-b in neuroblastoma cell differentiation highlights roles for SHP-2 and CDK16. iScience 2021, 24, 102321. [Google Scholar] [CrossRef] [PubMed]
  54. Funke, L.; Bracht, T.; Oeck, S.; Schork, K.; Stepath, M.; Dreesmann, S.; Eisenacher, M.; Sitek, B.; Schramm, A. NTRK1/TrkA Signaling in Neuroblastoma Cells Induces Nuclear Reorganization and Intra-Nuclear Aggregation of Lamin A/C. Cancers 2021, 13, 5293. [Google Scholar] [CrossRef]
  55. Emdal, K.B.; Pedersen, A.K.; Bekker-Jensen, D.B.; Lundby, A.; Claeys, S.; De Preter, K.; Speleman, F.; Francavilla, C.; Olsen, J.V. Integrated proximal proteomics reveals IRS2 as a determinant of cell survival in ALK-driven neuroblastoma. Sci. Signal 2018, 11, aap9752. [Google Scholar] [CrossRef]
  56. Uckun, E.; Siaw, J.T.; Guan, J.; Anthonydhason, V.; Fuchs, J.; Wolfstetter, G.; Hallberg, B.; Palmer, R.H. BioID-Screening Identifies PEAK1 and SHP2 as Components of the ALK Proximitome in Neuroblastoma Cells. J. Mol. Biol. 2021, 433, 167158. [Google Scholar] [CrossRef]
  57. Li, J.; Wang, Y.; Li, L.; Or, P.M.; Wai Wong, C.; Liu, T.; Ho, W.L.H.; Chan, A.M. Tumour-derived substrate-adherent cells promote neuroblastoma survival through secreted trophic factors. Mol. Oncol. 2021, 15, 2011–2025. [Google Scholar] [CrossRef]
  58. Hwang, M.; Han, M.H.; Park, H.H.; Choi, H.; Lee, K.Y.; Lee, Y.J.; Kim, J.M.; Cheong, J.H.; Ryu, J.I.; Min, K.W.; et al. LGR5 and Downstream Intracellular Signaling Proteins Play Critical Roles in the Cell Proliferation of Neuroblastoma, Meningioma and Pituitary Adenoma. Exp. Neurobiol. 2019, 28, 628–641. [Google Scholar] [CrossRef]
  59. Bugara, B.; Durbas, M.; Kudrycka, M.; Malinowska, A.; Horwacik, I.; Rokita, H. Silencing of the PHLDA1 leads to global proteome changes and differentiation pathways of human neuroblastoma cells. Front. Pharmacol. 2024, 15, 1351536. [Google Scholar] [CrossRef]
  60. Dhamdhere, M.R.; Gowda, C.P.; Singh, V.; Liu, Z.; Carruthers, N.; Grant, C.N.; Sharma, A.; Dovat, S.; Sundstrom, J.M.; Wang, H.G.; et al. IGF2BP1 regulates the cargo of extracellular vesicles and promotes neuroblastoma metastasis. Oncogene 2023, 42, 1558–1571. [Google Scholar] [CrossRef]
  61. Tsakaneli, A.; Carregari, V.C.; Morini, M.; Eva, A.; Cangemi, G.; Chayka, O.; Makarov, E.; Bibbo, S.; Capone, E.; Sala, G.; et al. MYC regulates metabolism through vesicular transfer of glycolytic kinases. Open Biol. 2021, 11, 210276. [Google Scholar] [CrossRef] [PubMed]
  62. Fonseka, P.; Liem, M.; Ozcitti, C.; Adda, C.G.; Ang, C.S.; Mathivanan, S. Exosomes from N-Myc amplified neuroblastoma cells induce migration and confer chemoresistance to non-N-Myc amplified cells: Implications of intra-tumour heterogeneity. J. Extracell. Vesicles 2019, 8, 1597614. [Google Scholar] [CrossRef] [PubMed]
  63. Morini, M.; Raggi, F.; Bartolucci, M.; Petretto, A.; Ardito, M.; Rossi, C.; Segalerba, D.; Garaventa, A.; Eva, A.; Cangelosi, D.; et al. Plasma-Derived Exosome Proteins as Novel Diagnostic and Prognostic Biomarkers in Neuroblastoma Patients. Cells 2023, 12, 2516. [Google Scholar] [CrossRef] [PubMed]
  64. Garcia, J.; Faca, V.; Jarzembowski, J.; Zhang, Q.; Park, J.; Hanash, S. Comprehensive profiling of the cell surface proteome of Sy5Y neuroblastoma cells yields a subset of proteins associated with tumor differentiation. J. Proteome Res. 2009, 8, 3791–3796. [Google Scholar] [CrossRef]
  65. Gangras, P.; Gelfanova, V.; Williams, G.D.; Handelman, S.K.; Smith, R.M.; Debets, M.F. Investigating SH-SY5Y Neuroblastoma Cell Surfaceome as a Model for Neuronal-Targeted Novel Therapeutic Modalities. Int. J. Mol. Sci. 2022, 23, 15062. [Google Scholar] [CrossRef]
  66. Misawa, A.; Hosoi, H.; Arimoto, A.; Shikata, T.; Akioka, S.; Matsumura, T.; Houghton, P.J.; Sawada, T. N-Myc induction stimulated by insulin-like growth factor I through mitogen-activated protein kinase signaling pathway in human neuroblastoma cells. Cancer Res. 2000, 60, 64–69. [Google Scholar]
  67. Marshall, G.M.; Liu, P.Y.; Gherardi, S.; Scarlett, C.J.; Bedalov, A.; Xu, N.; Iraci, N.; Valli, E.; Ling, D.; Thomas, W.; et al. SIRT1 promotes N-Myc oncogenesis through a positive feedback loop involving the effects of MKP3 and ERK on N-Myc protein stability. PLoS Genet. 2011, 7, e1002135. [Google Scholar] [CrossRef]
  68. Otto, T.; Horn, S.; Brockmann, M.; Eilers, U.; Schuttrumpf, L.; Popov, N.; Kenney, A.M.; Schulte, J.H.; Beijersbergen, R.; Christiansen, H.; et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell 2009, 15, 67–78. [Google Scholar] [CrossRef]
  69. Houten, S.M.; Wanders, R.J. A general introduction to the biochemistry of mitochondrial fatty acid beta-oxidation. J. Inherit. Metab. Dis. 2010, 33, 469–477. [Google Scholar] [CrossRef]
  70. Yang, M.; Vousden, K.H. Serine and one-carbon metabolism in cancer. Nat. Rev. Cancer 2016, 16, 650–662. [Google Scholar] [CrossRef]
  71. Sahu, D.; Hsu, C.L.; Lin, C.C.; Yang, T.W.; Hsu, W.M.; Ho, S.Y.; Juan, H.F.; Huang, H.C. Co-expression analysis identifies long noncoding RNA SNHG1 as a novel predictor for event-free survival in neuroblastoma. Oncotarget 2016, 7, 58022–58037. [Google Scholar] [CrossRef] [PubMed]
  72. Edsjo, A.; Holmquist, L.; Pahlman, S. Neuroblastoma as an experimental model for neuronal differentiation and hypoxia-induced tumor cell dedifferentiation. Semin. Cancer Biol. 2007, 17, 248–256. [Google Scholar] [CrossRef] [PubMed]
  73. Rozen, E.J.; Shohet, J.M. Systematic review of the receptor tyrosine kinase superfamily in neuroblastoma pathophysiology. Cancer Metastasis Rev. 2022, 41, 33–52. [Google Scholar] [CrossRef] [PubMed]
  74. Emdal, K.B.; Pedersen, A.K.; Bekker-Jensen, D.B.; Tsafou, K.P.; Horn, H.; Lindner, S.; Schulte, J.H.; Eggert, A.; Jensen, L.J.; Francavilla, C.; et al. Temporal proteomics of NGF-TrkA signaling identifies an inhibitory role for the E3 ligase Cbl-b in neuroblastoma cell differentiation. Sci. Signal 2015, 8, ra40. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, B.; Hammonds-Odie, L.; Perron, J.; Masters, B.A.; Bixby, J.L. SHP-2 mediates target-regulated axonal termination and NGF-dependent neurite growth in sympathetic neurons. Dev. Biol. 2002, 252, 170–187. [Google Scholar] [CrossRef]
  76. Dohmen, M.; Krieg, S.; Agalaridis, G.; Zhu, X.; Shehata, S.N.; Pfeiffenberger, E.; Amelang, J.; Butepage, M.; Buerova, E.; Pfaff, C.M.; et al. AMPK-dependent activation of the Cyclin Y/CDK16 complex controls autophagy. Nat. Commun. 2020, 11, 1032. [Google Scholar] [CrossRef]
  77. Pajtler, K.W.; Mahlow, E.; Odersky, A.; Lindner, S.; Stephan, H.; Bendix, I.; Eggert, A.; Schramm, A.; Schulte, J.H. Neuroblastoma in dialog with its stroma: NTRK1 is a regulator of cellular cross-talk with Schwann cells. Oncotarget 2014, 5, 11180–11192. [Google Scholar] [CrossRef]
  78. Pacenta, H.L.; Macy, M.E. Entrectinib and other ALK/TRK inhibitors for the treatment of neuroblastoma. Drug Des. Dev. Ther. 2018, 12, 3549–3561. [Google Scholar] [CrossRef]
  79. Pugh, T.J.; Morozova, O.; Attiyeh, E.F.; Asgharzadeh, S.; Wei, J.S.; Auclair, D.; Carter, S.L.; Cibulskis, K.; Hanna, M.; Kiezun, A.; et al. The genetic landscape of high-risk neuroblastoma. Nat. Genet. 2013, 45, 279–284. [Google Scholar] [CrossRef]
  80. Mosse, Y.P.; Laudenslager, M.; Longo, L.; Cole, K.A.; Wood, A.; Attiyeh, E.F.; Laquaglia, M.J.; Sennett, R.; Lynch, J.E.; Perri, P.; et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 2008, 455, 930–935. [Google Scholar] [CrossRef]
  81. Guan, J.; Tucker, E.R.; Wan, H.; Chand, D.; Danielson, L.S.; Ruuth, K.; El Wakil, A.; Witek, B.; Jamin, Y.; Umapathy, G.; et al. The ALK inhibitor PF-06463922 is effective as a single agent in neuroblastoma driven by expression of ALK and MYCN. Dis. Model. Mech. 2016, 9, 941–952. [Google Scholar] [CrossRef] [PubMed]
  82. Shaw, L.M. The insulin receptor substrate (IRS) proteins: At the intersection of metabolism and cancer. Cell Cycle 2011, 10, 1750–1756. [Google Scholar] [CrossRef] [PubMed]
  83. Ross, R.A.; Biedler, J.L.; Spengler, B.A. A role for distinct cell types in determining malignancy in human neuroblastoma cell lines and tumors. Cancer Lett. 2003, 197, 35–39. [Google Scholar] [CrossRef] [PubMed]
  84. Vieira, G.C.; Chockalingam, S.; Melegh, Z.; Greenhough, A.; Malik, S.; Szemes, M.; Park, J.H.; Kaidi, A.; Zhou, L.; Catchpoole, D.; et al. LGR5 regulates pro-survival MEK/ERK and proliferative Wnt/beta-catenin signalling in neuroblastoma. Oncotarget 2015, 6, 40053–40067. [Google Scholar] [CrossRef]
  85. Larrosa, C.; Mora, J.; Cheung, N.K. Global Impact of Monoclonal Antibodies (mAbs) in Children: A Focus on Anti-GD2. Cancers 2023, 15, 3729. [Google Scholar] [CrossRef]
  86. Ladenstein, R.; Potschger, U.; Valteau-Couanet, D.; Luksch, R.; Castel, V.; Yaniv, I.; Laureys, G.; Brock, P.; Michon, J.M.; Owens, C.; et al. Interleukin 2 with anti-GD2 antibody ch14.18/CHO (dinutuximab beta) in patients with high-risk neuroblastoma (HR-NBL1/SIOPEN): A multicentre, randomised, phase 3 trial. Lancet Oncol. 2018, 19, 1617–1629. [Google Scholar] [CrossRef]
  87. Philippova, J.; Shevchenko, J.; Sennikov, S. GD2-targeting therapy: A comparative analysis of approaches and promising directions. Front. Immunol. 2024, 15, 1371345. [Google Scholar] [CrossRef]
  88. Kaczanowska, S.; Murty, T.; Alimadadi, A.; Contreras, C.F.; Duault, C.; Subrahmanyam, P.B.; Reynolds, W.; Gutierrez, N.A.; Baskar, R.; Wu, C.J.; et al. Immune determinants of CAR-T cell expansion in solid tumor patients receiving GD2 CAR-T cell therapy. Cancer Cell 2024, 42, 35–51. [Google Scholar] [CrossRef]
  89. Horwacik, I.; Durbas, M.; Boratyn, E.; Sawicka, A.; Wegrzyn, P.; Krzanik, S.; Gorka, A.; Drozniak, J.; Augustyniak, E.; Kowalczyk, A.; et al. Analysis of genes involved in response to doxorubicin and a GD2 ganglioside-specific 14G2a monoclonal antibody in IMR-32 human neuroblastoma cells. Acta Biochim. Pol. 2015, 62, 423–433. [Google Scholar] [CrossRef]
  90. Dhamdhere, M.R.; Spiegelman, V.S. Extracellular vesicles in neuroblastoma: Role in progression, resistance to therapy and diagnostics. Front. Immunol. 2024, 15, 1385875. [Google Scholar] [CrossRef]
  91. van Niel, G.; Carter, D.R.F.; Clayton, A.; Lambert, D.W.; Raposo, G.; Vader, P. Challenges and directions in studying cell-cell communication by extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2022, 23, 369–382. [Google Scholar] [CrossRef] [PubMed]
  92. Azmi, A.S.; Bao, B.; Sarkar, F.H. Exosomes in cancer development, metastasis, and drug resistance: A comprehensive review. Cancer Metastasis Rev. 2013, 32, 623–642. [Google Scholar] [CrossRef] [PubMed]
  93. Guo, Y.; Ji, X.; Liu, J.; Fan, D.; Zhou, Q.; Chen, C.; Wang, W.; Wang, G.; Wang, H.; Yuan, W.; et al. Effects of exosomes on pre-metastatic niche formation in tumors. Mol. Cancer 2019, 18, 39. [Google Scholar] [CrossRef] [PubMed]
  94. Biegel, J.M.; Dhamdhere, M.; Gao, S.; Gowda, C.P.; Kawasawa, Y.I.; Spiegelman, V.S. Inhibition of the mRNA-Binding Protein IGF2BP1 Suppresses Proliferation and Sensitizes Neuroblastoma Cells to Chemotherapeutic Agents. Front. Oncol. 2021, 11, 608816. [Google Scholar] [CrossRef] [PubMed]
  95. Casey, S.C.; Baylot, V.; Felsher, D.W. The MYC oncogene is a global regulator of the immune response. Blood 2018, 131, 2007–2015. [Google Scholar] [CrossRef]
  96. Gangoda, L.; Boukouris, S.; Liem, M.; Kalra, H.; Mathivanan, S. Extracellular vesicles including exosomes are mediators of signal transduction: Are they protective or pathogenic? Proteomics 2015, 15, 260–271. [Google Scholar] [CrossRef]
  97. Li, N.; Spetz, M.R.; Li, D.; Ho, M. Advances in immunotherapeutic targets for childhood cancers: A focus on glypican-2 and B7-H3. Pharmacol. Ther. 2021, 223, 107892. [Google Scholar] [CrossRef]
  98. Waas, M.; Snarrenberg, S.T.; Littrell, J.; Jones Lipinski, R.A.; Hansen, P.A.; Corbett, J.A.; Gundry, R.L. SurfaceGenie: A web-based application for prioritizing cell-type-specific marker candidates. Bioinformatics 2020, 36, 3447–3456. [Google Scholar] [CrossRef]
  99. Saldivar, J.C.; Cortez, D.; Cimprich, K.A. Publisher correction: The essential kinase ATR: Ensuring faithful duplication of a challenging genome. Nat. Rev. Mol. Cell Biol. 2017, 18, 783. [Google Scholar] [CrossRef]
  100. Gilad, O.; Nabet, B.Y.; Ragland, R.L.; Schoppy, D.W.; Smith, K.D.; Durham, A.C.; Brown, E.J. Combining ATR suppression with oncogenic Ras synergistically increases genomic instability, causing synthetic lethality or tumorigenesis in a dosage-dependent manner. Cancer Res. 2010, 70, 9693–9702. [Google Scholar] [CrossRef]
  101. Szydzik, J.; Lind, D.E.; Arefin, B.; Kurhe, Y.; Umapathy, G.; Siaw, J.T.; Claeys, A.; Gabre, J.L.; Van den Eynden, J.; Hallberg, B.; et al. ATR inhibition enables complete tumour regression in ALK-driven NB mouse models. Nat. Commun. 2021, 12, 6813. [Google Scholar] [CrossRef] [PubMed]
  102. Borenas, M.; Umapathy, G.; Lind, D.E.; Lai, W.Y.; Guan, J.; Johansson, J.; Jennische, E.; Schmidt, A.; Kurhe, Y.; Gabre, J.L.; et al. ALK signaling primes the DNA damage response sensitizing ALK-driven neuroblastoma to therapeutic ATR inhibition. Proc. Natl. Acad. Sci. USA 2024, 121, e2315242121. [Google Scholar] [CrossRef] [PubMed]
  103. Mosse, Y.P. Anaplastic Lymphoma Kinase as a Cancer Target in Pediatric Malignancies. Clin. Cancer Res. 2016, 22, 546–552. [Google Scholar] [CrossRef] [PubMed]
  104. Lin, J.J.; Riely, G.J.; Shaw, A.T. Targeting ALK: Precision Medicine Takes on Drug Resistance. Cancer Discov. 2017, 7, 137–155. [Google Scholar] [CrossRef]
  105. Van den Eynden, J.; Umapathy, G.; Ashouri, A.; Cervantes-Madrid, D.; Szydzik, J.; Ruuth, K.; Koster, J.; Larsson, E.; Guan, J.; Palmer, R.H.; et al. Phosphoproteome and gene expression profiling of ALK inhibition in neuroblastoma cell lines reveals conserved oncogenic pathways. Sci. Signal 2018, 11, 557. [Google Scholar] [CrossRef]
  106. Chesler, L.; Schlieve, C.; Goldenberg, D.D.; Kenney, A.; Kim, G.; McMillan, A.; Matthay, K.K.; Rowitch, D.; Weiss, W.A. Inhibition of phosphatidylinositol 3-kinase destabilizes Mycn protein and blocks malignant progression in neuroblastoma. Cancer Res. 2006, 66, 8139–8146. [Google Scholar] [CrossRef]
  107. Segerstrom, L.; Baryawno, N.; Sveinbjornsson, B.; Wickstrom, M.; Elfman, L.; Kogner, P.; Johnsen, J.I. Effects of small molecule inhibitors of PI3K/Akt/mTOR signaling on neuroblastoma growth in vitro and in vivo. Int. J. Cancer 2011, 129, 2958–2965. [Google Scholar] [CrossRef]
  108. Le, X.; Antony, R.; Razavi, P.; Treacy, D.J.; Luo, F.; Ghandi, M.; Castel, P.; Scaltriti, M.; Baselga, J.; Garraway, L.A. Systematic Functional Characterization of Resistance to PI3K Inhibition in Breast Cancer. Cancer Discov. 2016, 6, 1134–1147. [Google Scholar] [CrossRef]
  109. Nawijn, M.C.; Alendar, A.; Berns, A. For better or for worse: The role of Pim oncogenes in tumorigenesis. Nat. Rev. Cancer 2011, 11, 23–34. [Google Scholar] [CrossRef]
  110. Mohlin, S.; Hansson, K.; Radke, K.; Martinez, S.; Blanco-Apiricio, C.; Garcia-Ruiz, C.; Welinder, C.; Esfandyari, J.; O’Neill, M.; Pastor, J.; et al. Anti-tumor effects of PIM/PI3K/mTOR triple kinase inhibitor IBL-302 in neuroblastoma. EMBO Mol. Med. 2019, 11, e10058. [Google Scholar] [CrossRef]
  111. Kim, H.; Lee, J.Y.; Park, K.J.; Kim, W.H.; Roh, G.S. A mitochondrial division inhibitor, Mdivi-1, inhibits mitochondrial fragmentation and attenuates kainic acid-induced hippocampal cell death. BMC Neurosci. 2016, 17, 33. [Google Scholar] [CrossRef] [PubMed]
  112. Dai, W.; Wang, G.; Chwa, J.; Oh, M.E.; Abeywardana, T.; Yang, Y.; Wang, Q.A.; Jiang, L. Mitochondrial division inhibitor (mdivi-1) decreases oxidative metabolism in cancer. Br. J. Cancer 2020, 122, 1288–1297. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, W.H.; Kao, Y.C.; Hsieh, C.H.; Tsai, S.Y.; Cheung, C.H.Y.; Huang, H.C.; Juan, H.F. Multiomics Reveals Induction of Neuroblastoma SK-N-BE(2)C Cell Death by Mitochondrial Division Inhibitor 1 through Multiple Effects. J. Proteome Res. 2024, 23, 301–315. [Google Scholar] [CrossRef] [PubMed]
  114. Ryan, J.; Tivnan, A.; Fay, J.; Bryan, K.; Meehan, M.; Creevey, L.; Lynch, J.; Bray, I.M.; O’Meara, A.; Tracey, L.; et al. MicroRNA-204 increases sensitivity of neuroblastoma cells to cisplatin and is associated with a favourable clinical outcome. Br. J. Cancer 2012, 107, 967–976. [Google Scholar] [CrossRef]
  115. Ooi, C.Y.; Carter, D.R.; Liu, B.; Mayoh, C.; Beckers, A.; Lalwani, A.; Nagy, Z.; De Brouwer, S.; Decaesteker, B.; Hung, T.T.; et al. Network Modeling of microRNA-mRNA Interactions in Neuroblastoma Tumorigenesis Identifies miR-204 as a Direct Inhibitor of MYCN. Cancer Res. 2018, 78, 3122–3134. [Google Scholar] [CrossRef]
  116. Bachetti, T.; Di Zanni, E.; Ravazzolo, R.; Ceccherini, I. miR-204 mediates post-transcriptional down-regulation of PHOX2B gene expression in neuroblastoma cells. Biochim. Biophys. Acta 2015, 1849, 1057–1065. [Google Scholar] [CrossRef]
  117. Hong, D.S.; Kang, Y.K.; Borad, M.; Sachdev, J.; Ejadi, S.; Lim, H.Y.; Brenner, A.J.; Park, K.; Lee, J.L.; Kim, T.Y.; et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br. J. Cancer 2020, 122, 1630–1637. [Google Scholar] [CrossRef]
  118. Usman, W.M.; Pham, T.C.; Kwok, Y.Y.; Vu, L.T.; Ma, V.; Peng, B.; Chan, Y.S.; Wei, L.; Chin, S.M.; Azad, A.; et al. Efficient RNA drug delivery using red blood cell extracellular vesicles. Nat. Commun. 2018, 9, 2359. [Google Scholar] [CrossRef]
  119. Chiangjong, W.; Panachan, J.; Keadsanti, S.; Newburg, D.S.; Morrow, A.L.; Hongeng, S.; Chutipongtanate, S. Development of red blood cell-derived extracellular particles as a biocompatible nanocarrier of microRNA-204 (REP-204) to harness anti-neuroblastoma effect. Nanomedicine 2024, 60, 102760. [Google Scholar] [CrossRef]
  120. Yang, Y.; Guo, L.; Chen, L.; Gong, B.; Jia, D.; Sun, Q. Nuclear transport proteins: Structure, function, and disease relevance. Signal Transduct. Target. Ther. 2023, 8, 425. [Google Scholar] [CrossRef]
  121. Galinski, B.; Luxemburg, M.; Landesman, Y.; Pawel, B.; Johnson, K.J.; Master, S.R.; Freeman, K.W.; Loeb, D.M.; Hebert, J.M.; Weiser, D.A. XPO1 inhibition with selinexor synergizes with proteasome inhibition in neuroblastoma by targeting nuclear export of IkB. Transl. Oncol. 2021, 14, 101114. [Google Scholar] [CrossRef] [PubMed]
  122. Chari, A.; Vogl, D.T.; Gavriatopoulou, M.; Nooka, A.K.; Yee, A.J.; Huff, C.A.; Moreau, P.; Dingli, D.; Cole, C.; Lonial, S.; et al. Oral Selinexor-Dexamethasone for Triple-Class Refractory Multiple Myeloma. N. Engl. J. Med. 2019, 381, 727–738. [Google Scholar] [CrossRef] [PubMed]
  123. Nguyen, R.; Wang, H.; Sun, M.; Lee, D.G.; Peng, J.; Thiele, C.J. Combining selinexor with alisertib to target the p53 pathway in neuroblastoma. Neoplasia 2022, 26, 100776. [Google Scholar] [CrossRef] [PubMed]
  124. Katayama, H.; Sasai, K.; Kawai, H.; Yuan, Z.M.; Bondaruk, J.; Suzuki, F.; Fujii, S.; Arlinghaus, R.B.; Czerniak, B.A.; Sen, S. Phosphorylation by aurora kinase A induces Mdm2-mediated destabilization and inhibition of p53. Nat. Genet. 2004, 36, 55–62. [Google Scholar] [CrossRef] [PubMed]
  125. Kohno, M.; Pouyssegur, J. Targeting the ERK signaling pathway in cancer therapy. Ann. Med. 2006, 38, 200–211. [Google Scholar] [CrossRef]
  126. Eleveld, T.F.; Oldridge, D.A.; Bernard, V.; Koster, J.; Colmet Daage, L.; Diskin, S.J.; Schild, L.; Bentahar, N.B.; Bellini, A.; Chicard, M.; et al. Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations. Nat. Genet. 2015, 47, 864–871. [Google Scholar] [CrossRef]
  127. Yu, Y.; Zhao, Y.; Choi, J.; Shi, Z.; Guo, L.; Elizarraras, J.; Gu, A.; Cheng, F.; Pei, Y.; Lu, D.; et al. ERK Inhibitor Ulixertinib Inhibits High-Risk Neuroblastoma Growth In Vitro and In Vivo. Cancers 2022, 14, 5534. [Google Scholar] [CrossRef]
  128. Islam, M.S.; Takano, R.; Yokochi, T.; Akter, J.; Nakamura, Y.; Nakagawara, A.; Tatsumi, Y. Programmed expression of pro-apoptotic BMCC1 during apoptosis, triggered by DNA damage in neuroblastoma cells. BMC Cancer 2019, 19, 542. [Google Scholar] [CrossRef]
  129. Scholzen, T.; Gerdes, J. The Ki-67 protein: From the known and the unknown. J. Cell Physiol. 2000, 182, 311–322. [Google Scholar] [CrossRef]
  130. Chandel, S.; Bhattacharya, A.; Gautam, A.; Zeng, W.; Alka, O.; Sachsenberg, T.; Gupta, G.D.; Narang, R.K.; Ravichandiran, V.; Singh, R. Investigation of the anti-cancer potential of epoxyazadiradione in neuroblastoma: Experimental assays and molecular analysis. J. Biomol. Struct. Dyn. 2023, 1–19. [Google Scholar] [CrossRef]
  131. Laghezza Masci, V.; Bernini, R.; Villanova, N.; Clemente, M.; Cicaloni, V.; Tinti, L.; Salvini, L.; Taddei, A.R.; Tiezzi, A.; Ovidi, E. In Vitro Anti-Proliferative and Apoptotic Effects of Hydroxytyrosyl Oleate on SH-SY5Y Human Neuroblastoma Cells. Int. J. Mol. Sci. 2022, 23, 12348. [Google Scholar] [CrossRef]
  132. Forbes, M.; Kempa, R.; Mastrobuoni, G.; Rayman, L.; Pietzke, M.; Bayram, S.; Arlt, B.; Spruessel, A.; Deubzer, H.E.; Kempa, S. L-Glyceraldehyde Inhibits Neuroblastoma Cell Growth via a Multi-Modal Mechanism on Metabolism and Signaling. Cancers 2024, 16, 1664. [Google Scholar] [CrossRef]
  133. Lee, Y.Q.; Rajadurai, P.; Abas, F.; Othman, I.; Naidu, R. Proteomic Analysis on Anti-Proliferative and Apoptosis Effects of Curcumin Analog, 1,5-bis(4-Hydroxy-3-Methyoxyphenyl)-1,4-Pentadiene-3-One-Treated Human Glioblastoma and Neuroblastoma Cells. Front. Mol. Biosci. 2021, 8, 645856. [Google Scholar] [CrossRef]
  134. Morretta, E.; Brullo, C.; Belvedere, R.; Petrella, A.; Spallarossa, A.; Monti, M.C. Targeting USP-7 by a Novel Fluorinated 5-Pyrazolyl-Urea Derivative. Int. J. Mol. Sci. 2023, 24, 9200. [Google Scholar] [CrossRef]
  135. Chittavanich, P.; Saengwimol, D.; Roytrakul, S.; Rojanaporn, D.; Chaitankar, V.; Srimongkol, A.; Anurathapan, U.; Hongeng, S.; Kaewkhaw, R. Ceftriaxone exerts antitumor effects in MYCN-driven retinoblastoma and neuroblastoma by targeting DDX3X for translation repression. Mol. Oncol. 2023, 18, 918–938. [Google Scholar] [CrossRef]
  136. Halakos, E.G.; Connell, A.J.; Glazewski, L.; Wei, S.; Mason, R.W. Bottom up proteomics reveals novel differentiation proteins in neuroblastoma cells treated with 13-cis retinoic acid. J. Proteom. 2019, 209, 103491. [Google Scholar] [CrossRef]
  137. Halakos, E.G.; Connell, A.J.; Glazewski, L.; Wei, S.; Mason, R.W. Bottom up proteomics identifies neuronal differentiation pathway networks activated by cathepsin inhibition treatment in neuroblastoma cells that are enhanced by concurrent 13-cis retinoic acid treatment. J. Proteom. 2021, 232, 104068. [Google Scholar] [CrossRef]
  138. Shilpa, G.; Renjitha, J.; Saranga, R.; Sajin, F.K.; Nair, M.S.; Joy, B.; Sasidhar, B.S.; Priya, S. Epoxyazadiradione Purified from the Azadirachta indica Seed Induced Mitochondrial Apoptosis and Inhibition of NFkappaB Nuclear Translocation in Human Cervical Cancer Cells. Phytother. Res. 2017, 31, 1892–1902. [Google Scholar] [CrossRef]
  139. Stickland, L.H. The inhibition of glucolysis by glyceraldehyde. Biochem. J. 1941, 35, 859–871. [Google Scholar] [CrossRef]
  140. Sakamoto, A.; Prasad, K.N. Effect of DL-glyceraldehyde on mouse neuroblastoma cells in culture. Cancer Res. 1972, 32, 532–534. [Google Scholar]
  141. Marengo, B.; Meta, E.; Brullo, C.; De Ciucis, C.; Colla, R.; Speciale, A.; Garbarino, O.; Bruno, O.; Domenicotti, C. Biological evaluation of pyrazolyl-urea and dihydro-imidazo-pyrazolyl-urea derivatives as potential anti-angiogenetic agents in the treatment of neuroblastoma. Oncotarget 2020, 11, 3459–3472. [Google Scholar] [CrossRef]
  142. Morretta, E.; Sidibe, A.; Spallarossa, A.; Petrella, A.; Meta, E.; Bruno, O.; Monti, M.C.; Brullo, C. Synthesis, functional proteomics and biological evaluation of new 5-pyrazolyl ureas as potential anti-angiogenic compounds. Eur. J. Med. Chem. 2021, 226, 113872. [Google Scholar] [CrossRef]
  143. Morretta, E.; Belvedere, R.; Petrella, A.; Spallarossa, A.; Rapetti, F.; Bruno, O.; Brullo, C.; Monti, M.C. Novel insights on the molecular mechanism of action of the anti-angiogenic pyrazolyl-urea GeGe-3 by functional proteomics. Bioorg Chem. 2021, 115, 105168. [Google Scholar] [CrossRef]
  144. Sardana, D.; Zhu, C.; Zhang, M.; Gudivada, R.C.; Yang, L.; Jegga, A.G. Drug repositioning for orphan diseases. Brief. Bioinform. 2011, 12, 346–356. [Google Scholar]
  145. Li, X.; Li, H.; Li, S.; Zhu, F.; Kim, D.J.; Xie, H.; Li, Y.; Nadas, J.; Oi, N.; Zykova, T.A.; et al. Ceftriaxone, an FDA-approved cephalosporin antibiotic, suppresses lung cancer growth by targeting Aurora B. Carcinogenesis 2012, 33, 2548–2557. [Google Scholar] [CrossRef]
  146. Matthay, K.K.; Villablanca, J.G.; Seeger, R.C.; Stram, D.O.; Harris, R.E.; Ramsay, N.K.; Swift, P.; Shimada, H.; Black, C.T.; Brodeur, G.M.; et al. Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. Children’s Cancer Group. N. Engl. J. Med. 1999, 341, 1165–1173. [Google Scholar] [CrossRef]
  147. Neuman, T.; Stephens, R.W.; Salonen, E.M.; Timmusk, T.; Vaheri, A. Induction of morphological differentiation of human neuroblastoma cells is accompanied by induction of tissue-type plasminogen activator. J. Neurosci. Res. 1989, 23, 274–281. [Google Scholar] [CrossRef]
  148. Moreno, L.; Caron, H.; Geoerger, B.; Eggert, A.; Schleiermacher, G.; Brock, P.; Valteau-Couanet, D.; Chesler, L.; Schulte, J.H.; De Preter, K.; et al. Accelerating drug development for neuroblastoma—New Drug Development Strategy: An Innovative Therapies for Children with Cancer, European Network for Cancer Research in Children and Adolescents and International Society of Paediatric Oncology Europe Neuroblastoma project. Expert Opin. Drug Discov. 2017, 12, 801–811. [Google Scholar]
  149. Johnsen, J.I.; Dyberg, C.; Fransson, S.; Wickstrom, M. Molecular mechanisms and therapeutic targets in neuroblastoma. Pharmacol. Res. 2018, 131, 164–176. [Google Scholar] [CrossRef]
  150. McKerrow, J.H. Update on drug development targeting parasite cysteine proteases. PLoS Negl. Trop. Dis. 2018, 12, e0005850. [Google Scholar] [CrossRef]
  151. Tucker, E.R.; Poon, E.; Chesler, L. Targeting MYCN and ALK in resistant and relapsing neuroblastoma. Cancer Drug Resist. 2019, 2, 803–812. [Google Scholar] [CrossRef]
  152. Wang, Y.; Chen, K.; Cai, Y.; Cai, Y.; Yuan, X.; Wang, L.; Wu, Z.; Wu, Y. Annexin A2 could enhance multidrug resistance by regulating NF-kappaB signaling pathway in pediatric neuroblastoma. J. Exp. Clin. Cancer Res. 2017, 36, 111. [Google Scholar] [CrossRef]
  153. Galenkamp, K.M.; Carriba, P.; Urresti, J.; Planells-Ferrer, L.; Coccia, E.; Lopez-Soriano, J.; Barneda-Zahonero, B.; Moubarak, R.S.; Segura, M.F.; Comella, J.X. TNFalpha sensitizes neuroblastoma cells to FasL-, cisplatin- and etoposide-induced cell death by NF-kappaB-mediated expression of Fas. Mol. Cancer 2015, 14, 62. [Google Scholar] [CrossRef]
  154. Tang, J.; He, J.; Guo, H.; Lin, H.; Li, M.; Yang, T.; Wang, H.Y.; Li, D.; Liu, J.; Li, L.; et al. PTBP2-Mediated Alternative Splicing of IRF9 Controls Tumor-Associated Monocyte/Macrophage Chemotaxis and Repolarization in Neuroblastoma Progression. Research 2023, 6, 0033. [Google Scholar] [CrossRef]
  155. Levin, V.A.; Panchabhai, S.C.; Shen, L.; Kornblau, S.M.; Qiu, Y.; Baggerly, K.A. Different changes in protein and phosphoprotein levels result from serum starvation of high-grade glioma and adenocarcinoma cell lines. J. Proteome Res. 2010, 9, 179–191. [Google Scholar] [CrossRef]
  156. Chae, S.Y.; Nam, D.; Hyeon, D.Y.; Hong, A.; Lee, T.D.; Kim, S.; Im, D.; Hong, J.; Kang, C.; Lee, J.W.; et al. DNA repair and cholesterol-mediated drug efflux induce dose-dependent chemoresistance in nutrient-deprived neuroblastoma cells. iScience 2021, 24, 102325. [Google Scholar] [CrossRef]
  157. Yang, C.; Tan, J.; Zhu, J.; Wang, S.; Wei, G. YAP promotes tumorigenesis and cisplatin resistance in neuroblastoma. Oncotarget 2017, 8, 37154–37163. [Google Scholar] [CrossRef]
  158. Merlos Rodrigo, M.A.; Buchtelova, H.; de Los Rios, V.; Casal, J.I.; Eckschlager, T.; Hrabeta, J.; Belhajova, M.; Heger, Z.; Adam, V. Proteomic Signature of Neuroblastoma Cells UKF-NB-4 Reveals Key Role of Lysosomal Sequestration and the Proteasome Complex in Acquiring Chemoresistance to Cisplatin. J. Proteome Res. 2019, 18, 1255–1263. [Google Scholar] [CrossRef]
  159. Marusyk, A.; Almendro, V.; Polyak, K. Intra-tumour heterogeneity: A looking glass for cancer? Nat. Rev. Cancer 2012, 12, 323–334. [Google Scholar] [CrossRef]
  160. McGranahan, N.; Swanton, C. Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future. Cell 2017, 168, 613–628. [Google Scholar] [CrossRef]
  161. Louis, C.U.; Shohet, J.M. Neuroblastoma: Molecular pathogenesis and therapy. Annu. Rev. Med. 2015, 66, 49–63. [Google Scholar] [CrossRef]
  162. Acosta, S.; Lavarino, C.; Paris, R.; Garcia, I.; de Torres, C.; Rodriguez, E.; Beleta, H.; Mora, J. Comprehensive characterization of neuroblastoma cell line subtypes reveals bilineage potential similar to neural crest stem cells. BMC Dev. Biol. 2009, 9, 12. [Google Scholar] [CrossRef]
  163. Gartlgruber, M.; Sharma, A.K.; Quintero, A.; Dreidax, D.; Jansky, S.; Park, Y.G.; Kreth, S.; Meder, J.; Doncevic, D.; Saary, P.; et al. Super enhancers define regulatory subtypes and cell identity in neuroblastoma. Nat. Cancer 2021, 2, 114–128. [Google Scholar] [CrossRef]
  164. Lopez-Carrasco, A.; Berbegall, A.P.; Martin-Vano, S.; Blanquer-Maceiras, M.; Castel, V.; Navarro, S.; Noguera, R. Intra-Tumour Genetic Heterogeneity and Prognosis in High-Risk Neuroblastoma. Cancers 2021, 13, 5173. [Google Scholar] [CrossRef]
  165. van Groningen, T.; Akogul, N.; Westerhout, E.M.; Chan, A.; Hasselt, N.E.; Zwijnenburg, D.A.; Broekmans, M.; Stroeken, P.; Haneveld, F.; Hooijer, G.K.J.; et al. A NOTCH feed-forward loop drives reprogramming from adrenergic to mesenchymal state in neuroblastoma. Nat. Commun. 2019, 10, 1530. [Google Scholar] [CrossRef]
  166. Shi, H.; Tao, T.; Abraham, B.J.; Durbin, A.D.; Zimmerman, M.W.; Kadoch, C.; Look, A.T. ARID1A loss in neuroblastoma promotes the adrenergic-to-mesenchymal transition by regulating enhancer-mediated gene expression. Sci. Adv. 2020, 6, eaaz3440. [Google Scholar] [CrossRef]
  167. Gomez, R.L.; Ibragimova, S.; Ramachandran, R.; Philpott, A.; Ali, F.R. Tumoral heterogeneity in neuroblastoma. Biochim. Biophys. Acta Rev. Cancer 2022, 1877, 188805. [Google Scholar] [CrossRef]
  168. Braekeveldt, N.; von Stedingk, K.; Fransson, S.; Martinez-Monleon, A.; Lindgren, D.; Axelson, H.; Levander, F.; Willforss, J.; Hansson, K.; Ora, I.; et al. Patient-Derived Xenograft Models Reveal Intratumor Heterogeneity and Temporal Stability in Neuroblastoma. Cancer Res. 2018, 78, 5958–5969. [Google Scholar] [CrossRef]
  169. Perkel, J.M. Single-cell proteomics takes centre stage. Nature 2021, 597, 580–582. [Google Scholar] [CrossRef]
  170. Arias-Hidalgo, C.; Juanes-Velasco, P.; Landeira-Vinuela, A.; Garcia-Vaquero, M.L.; Montalvillo, E.; Gongora, R.; Hernandez, A.P.; Fuentes, M. Single-Cell Proteomics: The Critical Role of Nanotechnology. Int. J. Mol. Sci. 2022, 23, 6707. [Google Scholar] [CrossRef]
  171. Lohani, V.; Akhiya, A.R.; Kundu, S.; Akhter, M.Q.; Bag, S. Single-Cell Proteomics with Spatial Attributes: Tools and Techniques. ACS Omega 2023, 8, 17499–17510. [Google Scholar] [CrossRef]
  172. Torok, S.; Vegvari, A.; Rezeli, M.; Fehniger, T.E.; Tovari, J.; Paku, S.; Laszlo, V.; Hegedus, B.; Rozsas, A.; Dome, B.; et al. Localization of sunitinib, its metabolites and its target receptors in tumour-bearing mice: A MALDI-MS imaging study. Br. J. Pharmacol. 2015, 172, 1148–1163. [Google Scholar] [CrossRef]
  173. Ryu, S.; Hayashi, M.; Aikawa, H.; Okamoto, I.; Fujiwara, Y.; Hamada, A. Heterogeneous distribution of alectinib in neuroblastoma xenografts revealed by matrix-assisted laser desorption ionization mass spectrometry imaging: A pilot study. Br. J. Pharmacol. 2018, 175, 29–37. [Google Scholar] [CrossRef]
  174. Wu, Z.; Hundsdoerfer, P.; Schulte, J.H.; Astrahantseff, K.; Boral, S.; Schmelz, K.; Eggert, A.; Klein, O. Discovery of Spatial Peptide Signatures for Neuroblastoma Risk Assessment by MALDI Mass Spectrometry Imaging. Cancers 2021, 13, 3184. [Google Scholar] [CrossRef]
  175. Chen, Y.N.; LaMarche, M.J.; Chan, H.M.; Fekkes, P.; Garcia-Fortanet, J.; Acker, M.G.; Antonakos, B.; Chen, C.H.; Chen, Z.; Cooke, V.G.; et al. Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases. Nature 2016, 535, 148–152. [Google Scholar] [CrossRef]
  176. Li, Y.; Dong, J.; Qin, J.J. Small molecule inhibitors targeting heat shock protein 90: An updated review. Eur. J. Med. Chem. 2024, 275, 116562. [Google Scholar] [CrossRef]
  177. Regan, P.L.; Jacobs, J.; Wang, G.; Torres, J.; Edo, R.; Friedmann, J.; Tang, X.X. Hsp90 inhibition increases p53 expression and destabilizes MYCN and MYC in neuroblastoma. Int. J. Oncol. 2011, 38, 105–112. [Google Scholar]
  178. Hsieh, C.H.; Huang, C.T.; Cheng, Y.S.; Hsu, C.H.; Hsu, W.M.; Chung, Y.H.; Liu, Y.L.; Yang, T.S.; Chien, C.Y.; Lee, Y.H.; et al. Homoharringtonine as a PHGDH inhibitor: Unraveling metabolic dependencies and developing a potent therapeutic strategy for high-risk neuroblastoma. Biomed. Pharmacother. 2023, 166, 115429. [Google Scholar] [CrossRef]
  179. Saldivar, J.C.; Cortez, D.; Cimprich, K.A. The essential kinase ATR: Ensuring faithful duplication of a challenging genome. Nat. Rev. Mol. Cell Biol. 2017, 18, 622–636. [Google Scholar] [CrossRef]
  180. DeGregori, J.; Johnson, D.G. Distinct and Overlapping Roles for E2F Family Members in Transcription, Proliferation and Apoptosis. Curr. Mol. Med. 2006, 6, 739–748. [Google Scholar]
  181. Ejeskar, K.; Krona, C.; Caren, H.; Zaibak, F.; Li, L.; Martinsson, T.; Ioannou, P.A. Introduction of in vitro transcribed ENO1 mRNA into neuroblastoma cells induces cell death. BMC Cancer 2005, 5, 161. [Google Scholar] [CrossRef]
  182. Omenn, G.S. Strategies for plasma proteomic profiling of cancers. Proteomics 2006, 6, 5662–5673. [Google Scholar] [CrossRef]
Figure 1. General Diagram of the workflow in proteomic studies in NB. Proteomic studies are applied to explore biomarkers and signaling pathways relevant to NB tumorigenesis, drug treatment and resistance, and they are carried out following with: (1) Collection of biological samples (tissues, cells, body fluid, EVs and other secreted proteins) from patients, animals, tissues or cell lines; (2) Collection of proteins by process of extraction and purification; (3) Collection of peptides after enzyme digestion; (4) MS/MS proteomics analysis with peptide samples based on different quantitative techniques: LFQ, TMT/iTRAQ, SILAC, DIA and others; (5) Data analysis with bioinformatics approaches, including expression difference analysis (exhibited with volcano plots, heatmaps and others), protein-protein interaction (PPI) analysis, gene ontology (GO) analysis, and additional approaches. EVs: extracellular vesicles; DDA: data-dependent acquisition; DIA: data-independent acquisition; SILAC: stable isotope labeling by amino acids in cell culture; iTRAQ: isobaric tags for relative and absolute quantitation; TMT: tandem mass tags; PPI: protein-protein interaction; GO: gene ontology.
Figure 1. General Diagram of the workflow in proteomic studies in NB. Proteomic studies are applied to explore biomarkers and signaling pathways relevant to NB tumorigenesis, drug treatment and resistance, and they are carried out following with: (1) Collection of biological samples (tissues, cells, body fluid, EVs and other secreted proteins) from patients, animals, tissues or cell lines; (2) Collection of proteins by process of extraction and purification; (3) Collection of peptides after enzyme digestion; (4) MS/MS proteomics analysis with peptide samples based on different quantitative techniques: LFQ, TMT/iTRAQ, SILAC, DIA and others; (5) Data analysis with bioinformatics approaches, including expression difference analysis (exhibited with volcano plots, heatmaps and others), protein-protein interaction (PPI) analysis, gene ontology (GO) analysis, and additional approaches. EVs: extracellular vesicles; DDA: data-dependent acquisition; DIA: data-independent acquisition; SILAC: stable isotope labeling by amino acids in cell culture; iTRAQ: isobaric tags for relative and absolute quantitation; TMT: tandem mass tags; PPI: protein-protein interaction; GO: gene ontology.
Children 11 01323 g001
Figure 2. The diagram on the MS-based proteomics application in key protein targets identification in recent five years for NB research. The columns “Tumorigenesis”, “drug treatment”, “drug resistance” are corresponding to protein/gene targets and related drugs listed in Table 1, Table 2 and Table 3, respectively. Protein targets were indicated in circle; drug names were indicated in square; the arrows indicated the potential relationship between the components. ACADM: medium-chain specific acyl-CoA dehydrogenase; PHGDH: phosphoglycerate dehydrogenase; IGF1R: Insulin-like growth factor 1 receptor; SHP2: SH2 domain-containing protein tyrosine phosphatase-2; Cbl: casitas B-lineage lymphoma proteins; IRS2: insulin receptor substrate 2; PAI1: plasminogen activator inhibitor 1; SPARC: secreted protein acidic and cysteine rich; POSTN: periostin; LEG1: galectin-1; LGR5: leucine-rich repeat-containing G-protein coupled receptor 5; IGF2BP1: Insulin-like growth factor 2 mRNA-binding protein 1; SEMA3A: semaphorin 3A; SHMT2: mitochondrial serine hydroxymethyl transferase 2; PKM2: pyruvate kinase M2; HK2: hexokinase II; NCAM: neural cell adhesion molecule; NCL: nucleolin; LGALS3BP: galectin-3-binding protein; LUM: lumican; VASP: vasodilator stimulated phosphoprotein; DCN: decorin; MYH9: myosin-9; FN1: fibronectin; LTBP1: latent-transforming growth factor-beta-binding protein 1; CALR: calreticulin; AKAP12: A-kinase anchor protein 12; NRCAM: neuroglia related cell adhesion molecule; ATR: ataxia telangiectasia and Rad3-related protein; PIKK: phosphoinositide three-kinase-related kinase; ALK: anaplastic lymphoma kinase; Mdivi-1: mitochondrial division inhibitor 1; Drp1: dynamin-related protein 1; XPO1: exportin-1; EAD: epoxyazadiradione; HTyr-OL: hydroxytyrosyl oleate; L-GA: L-glyceraldehyde; ENO1: enolase1; TUBB: tubulin beta chain; USP-7: ubiquitin carboxyl-terminal hydrolase 7; 13-cis RA: 13-cis retinoic acid; ANXA2: annexin A2; PTBP2: polypyrimidine tract binding protein 2.
Figure 2. The diagram on the MS-based proteomics application in key protein targets identification in recent five years for NB research. The columns “Tumorigenesis”, “drug treatment”, “drug resistance” are corresponding to protein/gene targets and related drugs listed in Table 1, Table 2 and Table 3, respectively. Protein targets were indicated in circle; drug names were indicated in square; the arrows indicated the potential relationship between the components. ACADM: medium-chain specific acyl-CoA dehydrogenase; PHGDH: phosphoglycerate dehydrogenase; IGF1R: Insulin-like growth factor 1 receptor; SHP2: SH2 domain-containing protein tyrosine phosphatase-2; Cbl: casitas B-lineage lymphoma proteins; IRS2: insulin receptor substrate 2; PAI1: plasminogen activator inhibitor 1; SPARC: secreted protein acidic and cysteine rich; POSTN: periostin; LEG1: galectin-1; LGR5: leucine-rich repeat-containing G-protein coupled receptor 5; IGF2BP1: Insulin-like growth factor 2 mRNA-binding protein 1; SEMA3A: semaphorin 3A; SHMT2: mitochondrial serine hydroxymethyl transferase 2; PKM2: pyruvate kinase M2; HK2: hexokinase II; NCAM: neural cell adhesion molecule; NCL: nucleolin; LGALS3BP: galectin-3-binding protein; LUM: lumican; VASP: vasodilator stimulated phosphoprotein; DCN: decorin; MYH9: myosin-9; FN1: fibronectin; LTBP1: latent-transforming growth factor-beta-binding protein 1; CALR: calreticulin; AKAP12: A-kinase anchor protein 12; NRCAM: neuroglia related cell adhesion molecule; ATR: ataxia telangiectasia and Rad3-related protein; PIKK: phosphoinositide three-kinase-related kinase; ALK: anaplastic lymphoma kinase; Mdivi-1: mitochondrial division inhibitor 1; Drp1: dynamin-related protein 1; XPO1: exportin-1; EAD: epoxyazadiradione; HTyr-OL: hydroxytyrosyl oleate; L-GA: L-glyceraldehyde; ENO1: enolase1; TUBB: tubulin beta chain; USP-7: ubiquitin carboxyl-terminal hydrolase 7; 13-cis RA: 13-cis retinoic acid; ANXA2: annexin A2; PTBP2: polypyrimidine tract binding protein 2.
Children 11 01323 g002
Table 5. a. Major protein findings related to molecular risk factors in NB. b. Major protein findings related to risk stratification in NB.
Table 5. a. Major protein findings related to molecular risk factors in NB. b. Major protein findings related to risk stratification in NB.
a. Molecular risk factors in NBRelevant protein targets in this review
MYCN network or MYCN statusP300 [49], PHGDH [51], PKM2 [61], HK2 [61], Alix [62], TSG101 [62], FLOT1 [62], VPS35 [62], DDX3X [135],
RTK/ALK signalingIGF1R [53], SHP2 [53,56], CDK16 [53], LMNA [54], IRS2 [55], DUSP4 [105]
Wnt/β-catenin signalinghnRNPH3, hnRNPA2B1 [58]
RAS/MAPK pathwaySHP2 [53,56], PRUNE2 [127], MK167 [127], DUSP4 [105]
ATR activityRAD51 [101], BRCA1/2 [101], E2F3 [101], DCK [101], ATM [102], DNAPK [102]
PI3K/Akt/mTOR pathwayIRS2 [55], Caspase3 [110], CDK6 [110], PIKK Family [102]
b. Risk stratification in NBRelevant protein targets in this review
High risk-relatedP300 [49], PHGDH [51,113], MATR3 [52], SHP2 [53,56], PAI1 [57], SPARC [57], POSTN [57], LEG1 [57], IRS2 [55], SEMA3A [60], SHMT2 [60], PKM2 [61], HK2 [61], NCAM [63,64], NCL [63], LGALS3BP [63], MYH9 [63], FN1 [63], LTBP1 [63], DDX3X [135], ANXA2 [152]
Favorable outcome-relatedACADM [50], LUM [63], VASP [63], DCN [63], CALR [63], AKAP12 [63], PRUNE2 [127], Caspase3 [110], PTBP2 [154]
PHGDH: phosphoglycerate dehydrogenase; PKM2: pyruvate kinase M2; HK2: hexokinase II; IGF1R: Insulin-like growth factor 1 receptor; SHP2: SH2 domain-containing protein tyrosine phosphatase-2; IRS2: insulin receptor substrate 2; ATM: ataxia-telangiectasia mutated; DNAPK: DNA-dependent protein kinase; PIKK: phosphoinositide three-kinase-related kinase; PAI1: plasminogen activator inhibitor 1; SPARC: secreted protein acidic and cysteine rich; POSTN: periostin; LEG1: galectin-1; SEMA3A: semaphorin 3A; SHMT2: mitochondrial serine hydroxymethyl transferase 2; NCAM: neural cell adhesion molecule; NCL: nucleolin; LGALS3BP: galectin-3-binding protein; MYH9: myosin-9; FN1: fibronectin; LTBP1: latent-transforming growth factor-beta-binding protein 1; ANXA2: annexin A2; CALR: calreticulin; AKAP12: A-kinase anchor protein 12; ACADM: medium-chain specific acyl-CoA dehydrogenase; LUM: lumican; VASP: vasodilator stimulated phosphoprotein; DCN: decorin; PTBP2: polypyrimidine tract binding protein 2.
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

Ren, K.; Wang, Y.; Zhang, M.; Tao, T.; Sun, Z. Unveiling Tumorigenesis Mechanisms and Drug Therapy in Neuroblastoma by Mass Spectrometry Based Proteomics. Children 2024, 11, 1323. https://doi.org/10.3390/children11111323

AMA Style

Ren K, Wang Y, Zhang M, Tao T, Sun Z. Unveiling Tumorigenesis Mechanisms and Drug Therapy in Neuroblastoma by Mass Spectrometry Based Proteomics. Children. 2024; 11(11):1323. https://doi.org/10.3390/children11111323

Chicago/Turabian Style

Ren, Keyi, Yu Wang, Minmin Zhang, Ting Tao, and Zeyu Sun. 2024. "Unveiling Tumorigenesis Mechanisms and Drug Therapy in Neuroblastoma by Mass Spectrometry Based Proteomics" Children 11, no. 11: 1323. https://doi.org/10.3390/children11111323

APA Style

Ren, K., Wang, Y., Zhang, M., Tao, T., & Sun, Z. (2024). Unveiling Tumorigenesis Mechanisms and Drug Therapy in Neuroblastoma by Mass Spectrometry Based Proteomics. Children, 11(11), 1323. https://doi.org/10.3390/children11111323

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