• All
  • Article Title
  • Abstract
  • Keywords
  • Author
  • Institution
  • Open Access
    Original Article

    High-risk neuroblastoma stage 4 (NBS4): multi-target inhibitors for c-Src kinases (Csk) and retinoic acid (RA) signalling pathways

    Amgad Gerges 1*
    Una Canning 2

    Explor Drug Sci. 2025;3:1008109 DOI: https://doi.org/10.37349/eds.2025.1008109

    Received: January 09, 2025 Accepted: April 15, 2025 Published: May 09, 2025

    Academic Editor: Fernando Albericio, University of KwaZulu-Natal, South Africa, Universidad de Barcelona, Spain

    Abstract

    Aim:

    This paper investigates two possible treatment targets for neuroblastoma (NB) stage 4 (NBS4), c-Src kinase (Csk) and retinoic acid (RA) signalling pathways as potential candidates for a multi-target drug. Research has demonstrated that many cancer cells overexpress and/or hyperactivate c-Src, a tyrosine that is a member of the Src-family kinases. In the case of NBS4, there are indications that successful inhibition of c-Src could inhibit disease progression. Research into the altered signalling of RA, which preserves the differentiated state of adult neurons, neural stem cells, and NB cells (SH-SY5Y), is also investigated as a potential multi-target drug.

    Methods:

    Using computer-aided technology, including OpenEye Scientific suite, Molegro Virtual docking, Samson suite, and Discovery Studio Visualiser, the results revealed that the receptors for both targets, Csk and RA, share similar amino acid sequencing that ranges from 80–100%, offering the possibility of further testing for multi-target drug use. Work was done to explore possible synthesis routes for each of the four compounds using the retrosynthesis program Spaya. Predictive toxicology was done using the Toxicity Estimation Software Tool (T.E.S.T.).

    Results:

    Four compounds (inhibitors) targeting the Csk tyrosine kinase and RA pathways were identified as potential inhibitors.

    Conclusions:

    Currently, no effective therapeutic agents for NBS4 exist. Immunotherapy which has proven effective in treating various cancers, is currently used to treat NBS4 and has a 40% to 50% survival rate. This paper investigates two possible treatment targets for NBS4, Csk and RA signalling pathways as possible candidates for a multi-target drug. Four potential inhibitors have been identified.

    Keywords

    Neuroblastoma stage 4 (NBS4), receptors, multi-target drugs, docking, cross-docking, binding interaction

    Introduction

    Neuroblastoma

    There are five stages in the clinical neuroblastoma (NB) staging system: 1, 2A, 2B, 3, 4S, and 4 [1]. For those diagnosed with NB stage 4 (NBS4) the treatment options are limited with a survival rate of between 40% to 50% [1]. In a review of drug therapies aimed at treating NBS4, Gerges and Canning [2] (2024) examined the potential for creating multi-targeted drugs to treat high-risk NBS4 using computational methods available to medicinal chemistry. Finding novel therapeutic drug targets by this approach usually involves computational techniques like molecular docking, homology modelling, molecular dynamics, and quantitative structure-activity relationships (QSAR) [3]. The work reported here follows on from that review and investigates two different types of inhibitors for the signalling pathways c-Src kinase (Csk) and retinoic acid (RA) signalling pathways[2]. Six targets were originally identified and investigated by the authors [2] and the results of the Protein Aligner revealed that the receptors for four of the six targets shared similar amino acid sequencing that ranged from 80–100%. The four targets that share similar sequencing included histone deacetylase (HDAC), bromodomain (BRD), hedgehog (HH), and tropomyosin receptor kinase (TRK) and are the focus of a separate paper. The remaining two targets of the original six, Csk and RA signalling pathways showed similarity across their receptors of 80–100% are the focus of this paper. Their potential for multi-targeted drug use was investigated further with four compounds (inhibitors) targeting the Csk and RA pathways identified.

    Current treatment for NBS4

    With therapeutic agents finding little success in the treatment of NBS4, immunotherapy which has been effective in treating a number of cancers is currently used. This treatment uses monoclonal antibodies and has increased the 5-year survival rate of NBS4 to 40% to 50% [46]. Dinutuximab (UNITUXIN), a monoclonal antibody, is an example of cancer immunotherapy and for patients with NBS4, it is used in conjunction with other therapeutic agents. It was approved by the FDA in 2015 for use in the United States [6].

    c-Src kinase inhibitors

    Proliferation, differentiation, migration, and membrane trafficking are just a few of the cellular functions that are critical to the Src family kinases (SFKs) of non-receptor tyrosine kinases (TK) [7]. TK deregulation has been linked to cancer, with pharmaceutical companies and academic institutions now targeting small molecule TK inhibitors (TKIs) as one of the largest drug families for the treatment of cancer (Figure 1) [8]. TKIs may be associated with fewer harmful side effects during anti-tumor treatments because they target particular molecular targets [8]. As a result, numerous TKIs have undergone testing for their anti-proliferative and anti-cancer properties in vitro and in vivo, with some approved for clinical trials and others currently used in cancer treatment [9]. One subclass of non-receptor TKs targeted in the treatment of human cancers is the SFKs. Of these, c-Src promotes angiogenesis, invasion, migration, and cell proliferation (Figure 1) [10]. c-Src hyperactivation results in abnormal cell activity, which aids in cancer development. High expression levels of c-Src, which have been found in several cancers, are typically associated with a poor overall survival prognosis [11]. More than one study has shown that c-Src inhibitors play an important role in antiproliferative activity in SH-SY5Y cells (NBS4 cell line) [1223].

    c-Src in cancer. VEGF: vascular endothelial growth factor; VEGFR2: VEGF receptor 2; LATS: large tumor suppressor; YAP: Yes-associated protein; TAZ: transcriptional co-activator with PDZ-binding motif; TEAD: transcriptional enhanced associate domain; GAPs: GTPase-activating proteins. Created in BioRender. Gerges, A. (2024) https://BioRender.com/t65n288

    Retinoic acid inhibitors

    The small lipophilic molecule RA, derived from vitamin A, is essential for axonal growth, neuronal differentiation, neuronal patterning, and embryonic development (Figure 2) [24]. Research on the effect of RA on neuronal development has improved our knowledge of neurodegenerative diseases [25]. It has been noted that the symptoms of neurodegenerative diseases are caused by RA altered signalling, which maintains the differentiated state of adult neurons, neural stem cells, and NB cells in particular (cell line SH-SY5Y) [2636].

    Retinoic Acid receptors and their actions. The three retinoic acid receptor subtypes (RARα, RARβ and RARγ) act as ligand-inducible transcription factors binding to DNA regulatory elements in the promoter regions of target genes by forming heterodimers with the retinoid X receptors (RXRα, RXRβ and RXRγ). RDH: retinol dehydrogenase; PALDH: retinaldehyde dehydrogenase; CRABP: cellular retinoic acid-binding protein; FABP5: fatty acid binding protein 5; PPARβ/δ: peroxisome proliferator-activated receptor beta/delta; ERα: estrogen receptor alpha; ERE: estrogen response elements; E2: estradiol; RARα: retinoic acid receptor-alpha; RXR: retinoid X receptor; PDK-1: phosphoinositide-dependent kinase-1; RA: retinoic acid; RARE: RA responsive element. Created in BioRender. Gerges, A. (2025) https://BioRender.com/v19a099

    Medicinal chemistry approach to drug development

    In the search for a multi-target drug for NBS4 using advanced medicinal chemistry software, the approach is to assesses the possibility of different targets (Figures 1 and 2) by assessing receptor similarities and selecting two lead compounds for each target (four compounds in total), see Figure 3 [8] and Figure 4 [29], that have demonstrated an inhibitory effect on NBS4 cells to form the basis of a search for a multi-targeted drug [29].

    Chemical structure of the lead c-Src inhibitors, compounds 18 and 29

    Chemical structure of the lead RA pathway inhibitors, compounds 8A and 8E

    For the two targets (Csk and RA) two receptors from each target were selected (Table 1) and their amino acid sequence similarity tested using Protein Aligner [37] (see Figure 5), which reported 80–100% similarity. Protein Aligner checks for amino acid sequence similarity between receptors with high similarity indicating their suitability for use in cross-docking. For the purposes of this work docking involves docking a compound to a receptor known for that target (i.e. Csk 4O2P [8] and 5D10 [38]) whereas cross-docking involves docking a compound to a receptor that belongs to a different target (i.e. RA 1CBQ and 1CBS [39]) and vice versa. The aim of cross-docking is to explore the suitability of selected compounds belonging to different targets to determine their suitability for use as a multi-target drug. This is a process that involves selecting known lead compounds to produce hitlists of compounds, and was done using BROOD [40] as part of the OpenEye suite.

    Drug targets and their selected receptors

    Drug target (n = 2)Proteins selected from PDB (n = 4)
    c-Src kinase (Csk)4O2P [8] and 5D10 [38]
    Retinoic acid (RA)1CBQ and 1CBS [39]
    Display full size

    PDB: Protein Data Bank

    Several BROOD rounds (Figure 6) were completed on the two lead compounds for each target to produce hitlists using “Shape and Color” and “Shape and Electrostatics”. On completion, BROOD ranked each of the hitlists according to BROOD hitlist parameters (see list below) and the top 25 of each hitlist was selected to run on OEDocking, using FRED (Figure 7) [41], ROCS (Figure 8) [42], AFITT (Figure 9) [43], and VIDA [44]. In addition, the Samson docking suite, including AutoDock Vina Extended (Figures 10 and 11) [45, 46] and Fitted (Molecular Forecaster) [47], with another docking program, Molegro [48], acted as confirmation for both the docking and cross-docking procedures. From this process, eight compounds for the Csk target were identified and six from the RA target, making a total of 14 compounds. All 14 compounds showed improved parameters compared to the original lead compounds (see Table 2). The compounds for each individual target were then cross-docked with the receptors identified for the other targets [43]. From the 14 compounds run, four showed potential suitability for multi-target use (two from each list) see Table 2.

    Docking results from AFITT with the top clusters from the hitlists (n = 14)

    TargetClustersAFFITFREDAutoDock Vina ExtendedMolegroFitted
    RA (n = 6)Clusters1CBS1CBS1CBS1CBS1CBS
    1Cluster 18, 1 of 10.6619–6.652–8.355106.873–25.3637
    2Cluster 3, 1 of 300.6612–7.42–9.36432.95–28.3973
    3Cluster 23, 1 of 250.6207–7.27–9.222113.34–22.8749
    4Cluster 20, 1 of 70.5474–9.061–9.47214.94–23.4724
    5Cluster 21, 1 of 50.5448–4.6477–8.95276.28–20.8754
    6Cluster 22, 1 of 40.5448–4.67–8.843–10.53–25.0819
    78E (lead compound)0.4543–4.156–8.589–94.37–23.6039
    c-Src (n = 8)Clusters402P402P402P402P402P
    1Cluster 16, 1 of 10.7654–6.03–8.384–58.58–25.413
    2Cluster 20, 1 of 50.7471–6.51–8.361–45.59–26.526
    3Cluster 7, 1 of 10.5397–6.43–8.856–60–28.785
    4Cluster 19, 1 of 50.5065–5.7–8.386–58.99–22.472
    5Cluster 1, 1 of 430.503–6.34–8.931–58.72–24.192
    6Cluster 17, 1 of 30.489–5.65–8.823–39.86–26.285
    7Cluster 4, 1 of 10.3898–6.88–8.39258.04–28.788
    8Cluster 3, 1 of 10.3717–3.63–8.655–44.07–27.369
    9Compound 18 (lead compound)0.7432–6.34–8.108–45.94–24.476
    Display full size

    Selected compounds in bold. RA: retinoic acid

    To check and compare the compounds (clusters) for toxicity and mutagenicity, the Toxicity Estimation Software Tool (T.E.S.T.) was used to provide the prediction mechanisms of the toxic action of the clusters [49]. The results from T.E.S.T. showed some similarity with the original lead compounds (Table 3), which also confirmed their suitability for use. Further work was done to explore possible synthesis routes for each of the four compounds using the retrosynthesis program Spaya [50].

    Results from the T.E.S.T. compared to the lead compounds

    ClustersBioconcentration factor1Ames mutagenicity2Oral rat LD503 [–Log10(moL/kg)]48-hour T. pyriformis IGC504 (mg/L)
    18, 1 of 119.08Negative373.49N/A
    23, 1 of 2518.78Negative833.23N/A
    19, 1 of 539.65N/A839.23N/A
    4, 1 of 121.54N/A1,714.20N/A
    Compound 8A12.25Negative881.07N/A
    Compound 8E15.68Negative1,193.20N/A
    Compound 1815.64Positive1,010.18N/A
    Compound 2928.91Positive457.49N/A
    Display full size

    1 Bioconcentration factor: ratio of the chemical concentration in fish as a result of absorption via the respiratory surface to that in water at a steady state; 2 Ames mutagenicity: a compound is positive for mutagenicity if it induces revertant colony growth in any strain of Salmonella typhimurium; 3 Oral rat LD50: amount of chemical (mg/kg body weight) that causes 50% of rats to die after oral ingestion; 4 48-hour T. pyriformis IGC50: Concentration of the test chemical in water (mg/L) that causes 50% growth inhibition to Tetrahymena pyriformis after 48 hours. N/A: not applicable

    Materials and methods

    Materials

    Computer programs:

    • OpenEye Scientific programs, which include various applications. The suite comprises BROOD, VIDA, MakeReceptor, FRED, and AFITT;

    • Molegro Virtual Docker;

    • The Samson suite includes Protein Aligner, AutoDock Vina Extended, and the Fitted suite by Molecular Forecaster;

    • T.E.S.T.;

    • BIOVIA Discovery Studio Visualizer [51];

    • Spaya-retrosynthesis software.

    Methods

    • Identifying drug targets;

    • Selection of two proteins (receptors) for each target;

    • Download the Protein Data Bank (PDB) files and their electron density map from the PDB database;

    • Comparing the similarities of the receptors. Run protein similarity on Samson (Protein Aligner) to determine suitability;

    • Selection of two lead compounds for each type;

    • Run the lead compounds on BROOD (from the OpenEye suite) and produce hitlists using Shape and Colour and Shape and Electrostatics;

    • Receptor preparations using MakeReceptor from the OpenEye suite;

    • Docking the hit compounds with OpenEye suite (FRED), Molegro, and Samson suite (AutoDockVina and Fitted);

    • Run cross-docking: each hitlist clusters from one target to the other target (using their protein/receptor);

    • Run hits with AFITT to rank the compounds according to their fitting probabilities;

    • Run selected clusters on ROCS;

    • Run selected clusters on T.E.S.T.;

    • Run clusters on Spaya to find the best synthesis route.

    Results

    Figure 5 shows the Protein alignment [37] (similarities) between the selected proteins ranging from 80% to 100%.

    Protein alignment of the selected receptors from two targets. The similarity (in grey) was obtained using Protein Alignment by Samson. Zero indicates 100% similarity, and 100% means no similarity

    Figure 6 below shows some of the hitlist from BROOD [40].

    Print screen of BROOD (Shape and Color) outcome when using compound 18 as a lead compound, only showing the first 11 clusters

    BROOD Hitlist parameters (Figure 6) include:

    • AroRingCt: number of aromatic rings in the molecule;

    • ClusterID/IdeaGroup: cluster ID of the molecule;

    • Colour: the replacement fragment's colour Tanimoto score in comparison to the query fragment;

    • Combo: Tanimoto combo score for the replacement fragment's shape and colour in comparison to the query fragment;

    • Egan: the Boolean indicates if the molecule satisfies the Egan bioavailability model;

    • Fragment: SMILES string of the replacement fragment;

    • Freq: the replacement fragment’s frequency;

    • fsp3C: the molecule’s fraction of sp3 hybridized carbon atoms;

    • HvyAtoms: number of heavy atoms in the molecule;

    • LipinskiDon: number of Lipinski donors in the molecule;

    • LipinskiAcc: number of Lipinski acceptors in the molecule;

    • LipinskiFail: Boolean specifying whether the molecule fails Lipinski’s rule of five;

    • Local strain: calculated local strain of the molecule;

    • Molecular TanimotoCombo: shape + colour Tanimoto combo score of the molecule against the query molecule;

    • MolWt: molecular weight of the molecule;

    • p(active): belief score of the molecule;

    • RingCt: number of ring atoms;

    • RingRatio: ratio of the number of ring atoms to the total number of heavy atoms;

    • Rotors: number of rotatable bonds in the molecule;

    • Shape: compare the replacement fragment’s Shape Tanimoto score to that of the query fragment;

    • Source Mols: SMILES strings of the molecules the replacement fragment is part of;

    • Source Mol Labels: labels of the molecules the replacement fragment is part of;

    • tPSA: calculated topological polar surface area of the molecule;

    • Veber: Boolean specifying whether the molecule passes the Veber bioavailability model;

    • XlogP: calculated LogP of the molecule [52].

    The next stage was to dock the hitlists with their relevant receptors: Figure 7 shows one of the docking outcomes using FRED (OpenEye suite).

    FRED docking result: 4O2P with some of the selected clusters

    The chosen compounds were also subjected to ROCS, a program that scores and aligns a database of molecules with a query. The score assigns a number to molecules according to their likelihood of having biological characteristics in common with the query molecule (see Figure 8).

    An example from ROCS outcome

    AFITT is another crystallographic tool from the OpenEye suite (Figure 9). By combining the shape and MMFF technologies of OpenEye, AFITT creates a new combined forcefield that fits small molecules into crystallographic density while preserving superior chemistry. It also offers an interface to external refinement programs, such as real space correlation coefficient (RSCC) calculation and interactive Ramachandran plots to verify the refinement [43].

    Docking receptor 5D10 with all clusters from c-Src, and RA with AFITT

    AutoDock Vina Extended (Figures 10 and 11) showing the docking of clusters to receptors 402P and 1CBS.

    Docking receptor 4O2P with all the clusters from c-Src and RA, including the lead compounds, using AutoDock Vina Extended from Samson suite

    Docking receptor 1CBS with all clusters from c-Src, and RA, including the lead compounds, using AutoDock Vina Extended

    Retrosynthesis results using Spaya

    Synthesis of cluster 4, 1 of 1, c-Src inhibitor (Figure 12).

    Synthesis of cluster 4, 1 of 1, c-Src inhibitor

    Synthesis of cluster 19, 1 of 5, c-Src inhibitor (Figure 13).

    Synthesis of cluster 19, 1 of 5, c-Src inhibitor

    Synthesis of clusters 18, 1 of 1, RA pathway inhibitor (Figure 14).

    Synthesis of clusters 18, 1 of 1, RA pathway inhibitor

    Synthesis of cluster 23, 1 of 25, RA pathway inhibitor (Figure 15).

    Synthesis of cluster 23, 1 of 25, RA pathway inhibitor

    Discussion

    The process of developing a multi-target drug is described as taking “a well-established primary target for a particular disease [and adding], secondary activities […] to enhance efficacy and reduce the effect” [53]. Creating such a drug involves taking the inhibitory effects of two or more drugs and combining them into a single molecule [54]. Previous methods used for drug development include “Pharmacophore” and “Screening”. This article employs a computer-aided virtual screening (VS) strategy with some modifications in the pursuit of multi-target drug discovery. Having identified two targets, two receptors from each target were selected (see Table 1) and a Protein Aligner tool from Samson was used to check for their suitability. Protein Aligner checks for amino acid sequence similarity between receptors with high similarity indicating their suitability for use in cross-docking. The aim of cross-docking is to explore the selected compounds for use as a multi-target drug. This is a process that involves selecting known lead compounds to produce hitlists of compounds and was done using BROOD [40] as part of the OpenEye suite.

    As part of the OpenEye suite, several rounds from BROOD were completed on the two lead compounds for each target to produce hitlists using “Shape and Color” and “Shape and Electrostatics” with the top 25 of each hitlist selected to run on OEDocking [41], ROCS [42], AFITT [43], and VIDA [44]. The hitlists of compounds (clusters) were docked, and the selected compounds cross-docked. The clusters were docked with more than one docking program as a means to validating the result. The final ranking of the selected clusters was done on AFITT as the receptor preparation with the tool MakeReceptor, giving the user more control over the receptor-creating process. AFITT also has the advantage of real-space fitting of ligands in density, integrated with REFMAC, PHENIX, BUSTER, CNX, and COOT and fragment and cocktail fitting [43]. In AFFIT, it is possible to select more than one ligand to fit generation of high-quality refinement dictionaries for use. This can be done during reciprocal space refinement that includes the use of forcefield (MMFF); semi-empirical (AM1, PM3) methods during reciprocal space refinement for BUSTER and Phenix; real space fitting of protein residues; proper handling of covalently bonded ligands, and proper handling of multiple occupancy ligands.

    The ranking is based not only on the best results but also on the ability of the identified compounds to cross-dock on the receptors of other targets, and it was this process that led to the selection of the four compounds. Using the tool ROCS provided cluster validation, which is based on a large database search [42]. The toxicity and mutagenicity of the four compounds were tested using T.E.S.T. to provide the prediction mechanisms of the toxic action of the clusters [49]. The results from T.E.S.T. showed some similarity with the original lead compounds (Table 3), which also provided data on their suitability for use. Further work was done to explore possible synthesis routes for each of the four compounds using the retrosynthesis program Spaya [50].

    In conclusion using a computer-aided modified VS approach, four compounds were identified demonstrating the possibility of inhibiting two targets simultaneously: the Csk and the RA signalling pathways. Going forward, future research will concentrate on preparing and evaluating the identified compounds both in vitro and in vivo [55].

    Abbreviations

    Csk:

    c-Src kinase

    NB:

    neuroblastoma

    NBS4:

    neuroblastoma stage 4

    PDB:

    Protein Data Bank

    RA:

    retinoic acid

    SFKs:

    Src family kinases

    T.E.S.T:

    Toxicity Estimation Software Tool

    TK:

    tyrosine kinase

    TKIs:

    tyrosine kinase inhibitors

    VS:

    virtual screening

    Declarations

    Acknowledgements

    Thanks to Dr. Corinne Kay for her advice and support over the years and to Mr. Colin Gaudion for testing some of the programs used for this work.

    Author contributions

    AG: Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Resources, Validation, Writing—original draft. UC: Writing—review & editing. Both authors read and approved the submitted version.

    Conflicts of interest

    The authors’ daughter, Isabella, was diagnosed with NBS4 in January 2003. Isabella relapsed in March 2005 and died in July 2005, a week after her seventh birthday.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2025.

    Publisher’s note

    Open Exploration maintains a neutral stance on jurisdictional claims in published institutional affiliations and maps. All opinions expressed in this article are the personal views of the author(s) and do not represent the stance of the editorial team or the publisher.

    References

    Paraboschi I, Privitera L, Kramer-Marek G, Anderson J, Giuliani S. Novel Treatments and Technologies Applied to the Cure of Neuroblastoma. Children (Basel). 2021;8:482. [DOI] [PubMed] [PMC]
    Gerges A, Canning U. Neuroblastoma and its Target Therapies: A Medicinal Chemistry Review. ChemMedChem. 2024;19:e202300535. [DOI] [PubMed]
    Kumalo HM, Bhakat S, Soliman MES. Theory and applications of covalent docking in drug discovery: merits and pitfalls. Molecules. 2015;20:19842000. [DOI] [PubMed] [PMC]
    Mora J. Dinutuximab for the treatment of pediatric patients with high-risk neuroblastoma. Expert Rev Clin Pharmacol. 2016;9:64753. [DOI] [PubMed]
    Bhoopathi P, Mannangatti P, Emdad L, Das SK, Fisher PB. The quest to develop an effective therapy for neuroblastoma. J Cell Physiol. 2021;236:777591. [DOI] [PubMed]
    Pennington B, Ren S, Barton S, Bacelar M, Edwards SJ. Dinutuximab Beta for Treating Neuroblastoma: An Evidence Review Group and Decision Support Unit Perspective of a NICE Single Technology Appraisal. Pharmacoeconomics. 2019;37:98593. [DOI] [PubMed]
    Roskoski R Jr. Src protein-tyrosine kinase structure, mechanism, and small molecule inhibitors. Pharmacol Res. 2015;94:925. [DOI] [PubMed]
    Tintori C, Fallacara AL, Radi M, Zamperini C, Dreassi E, Crespan E, et al. Combining X-ray crystallography and molecular modeling toward the optimization of pyrazolo[3,4-d]pyrimidines as potent c-Src inhibitors active in vivo against neuroblastoma. J Med Chem. 2015;58:34761. [DOI] [PubMed]
    Pytel D, Sliwinski T, Poplawski T, Ferriola D, Majsterek I. Tyrosine kinase blockers: new hope for successful cancer therapy. Anticancer Agents Med Chem. 2009;9:6676. [DOI] [PubMed]
    Brunton VG, Frame MC. Src and focal adhesion kinase as therapeutic targets in cancer. Curr Opin Pharmacol. 2008;8:42732. [DOI] [PubMed]
    Grant S, Dent P. Kinase inhibitors and cytotoxic drug resistance. Clin Cancer Res. 2004;10:22057. [DOI] [PubMed]
    Radi M, Brullo C, Crespan E, Tintori C, Musumeci F, Biava M, et al. Identification of potent c-Src inhibitors strongly affecting the proliferation of human neuroblastoma cells. Bioorg Med Chem Lett. 2011;21:592833. [DOI] [PubMed]
    Schenone S, Zanoli S, Brullo C, Crespan E, Maga G. Current Advances in the Development of Anticancer Drugs Targeting Tyrosine Kinases of the Src Family. Current Drug Therapy. 2008;3:15876. [DOI]
    Vitali R, Mancini C, Cesi V, Tanno B, Piscitelli M, Mancuso M, et al. Activity of tyrosine kinase inhibitor Dasatinib in neuroblastoma cells in vitro and in orthotopic mouse model. Int J Cancer. 2009;125:254755. [DOI] [PubMed] [PMC]
    Wheeler DL, Iida M, Dunn EF. The role of Src in solid tumors. Oncologist. 2009;14:66778. [DOI] [PubMed] [PMC]
    Kruewel T, Schenone S, Radi M, Maga G, Rohrbeck A, Botta M, et al. Molecular characterization of c-Abl/c-Src kinase inhibitors targeted against murine tumour progenitor cells that express stem cell markers. PLoS One. 2010;5:e14143. [DOI] [PubMed] [PMC]
    Lovera S, Sutto L, Boubeva R, Scapozza L, Dölker N, Gervasio FL. The different flexibility of c-Src and c-Abl kinases regulates the accessibility of a druggable inactive conformation. J Am Chem Soc. 2012;134:24969. [DOI] [PubMed]
    Lin Y, Roux B. Computational analysis of the binding specificity of Gleevec to Abl, c-Kit, Lck, and c-Src tyrosine kinases. J Am Chem Soc. 2013;135:1474153. [DOI] [PubMed] [PMC]
    Lin Y, Meng Y, Huang L, Roux B. Computational study of Gleevec and G6G reveals molecular determinants of kinase inhibitor selectivity. J Am Chem Soc. 2014;136:1475362. [DOI] [PubMed] [PMC]
    Meng Y, Lin Y, Roux B. Computational study of the "DFG-flip" conformational transition in c-Abl and c-Src tyrosine kinases. J Phys Chem B. 2015;119:144356. [DOI] [PubMed] [PMC]
    Redavide E. Role of an N2-Src-COPII pathway in neuroblastoma differentiation [dissertation]. University of York; 2018.
    Molinari A, Fallacara AL, Maria SD, Zamperini C, Poggialini F, Musumeci F, et al. Efficient optimization of pyrazolo[3,4-d]pyrimidines derivatives as c-Src kinase inhibitors in neuroblastoma treatment. Bioorg Med Chem Lett. 2018;28:34547. [DOI] [PubMed]
    Jha V, Macchia M, Tuccinardi T, Poli G. Three-Dimensional Interactions Analysis of the Anticancer Target c-Src Kinase with Its Inhibitors. Cancers (Basel). 2020;12:2327. [DOI] [PubMed] [PMC]
    le Maire A, Germain P, Bourguet W. Protein-protein interactions in the regulation of RAR-RXR heterodimers transcriptional activity. Methods Enzymol. 2020;637:175207. [DOI] [PubMed]
    Yin L, Shen H, Diaz-Ruiz O, Bäckman CM, Bae E, Yu S, et al. Early post-treatment with 9-cis retinoic acid reduces neurodegeneration of dopaminergic neurons in a rat model of Parkinson's disease. BMC Neurosci. 2012;13:120. [DOI] [PubMed] [PMC]
    Mandili G, Marini C, Carta F, Zanini C, Prato M, Khadjavi A, et al. Identification of phosphoproteins as possible differentiation markers in all-trans-retinoic acid-treated neuroblastoma cells. PLoS One. 2011;6:e18254. [DOI] [PubMed] [PMC]
    Chen M, Hsu S, Lin H, Yang T. Retinoic acid and cancer treatment. Biomedicine (Taipei). 2014;4:22. [DOI] [PubMed] [PMC]
    Janesick A, Wu SC, Blumberg B. Retinoic acid signaling and neuronal differentiation. Cell Mol Life Sci. 2015;72:155976. [DOI] [PubMed] [PMC]
    Lone AM, Dar NJ, Hamid A, Shah WA, Ahmad M, Bhat BA. Promise of Retinoic Acid-Triazolyl Derivatives in Promoting Differentiation of Neuroblastoma Cells. ACS Chem Neurosci. 2016;7:829. [DOI] [PubMed]
    Lascio SD, Saba E, Belperio D, Raimondi A, Lucchetti H, Fornasari D, et al. PHOX2A and PHOX2B are differentially regulated during retinoic acid-driven differentiation of SK-N-BE(2)C neuroblastoma cell line. Exp Cell Res. 2016;342:6271. [DOI] [PubMed] [PMC]
    Zage PE. Novel Therapies for Relapsed and Refractory Neuroblastoma. Children (Basel). 2018;5:148. [DOI] [PubMed] [PMC]
    Oral Liquid 13-cis-retinoic Acid (13-CRA) (My-CRA) [Internet]. [Cited 2023 Jul 12]. Available from: https://clinicaltrials.gov/study/NCT03291080
    Cui S, Wang Y, Wang Y, Tang X, Ren X, Zhang L, et al. Design, synthesis and biological evaluation of 3-(imidazo[1,2-a]pyrazin-3-ylethynyl)-2-methylbenzamides as potent and selective pan-tropomyosin receptor kinase (TRK) inhibitors. Eur J Med Chem. 2019;179:47082. [DOI] [PubMed]
    Magalingam KB, Radhakrishnan AK, Somanath SD, Md S, Haleagrahara N. Influence of serum concentration in retinoic acid and phorbol ester induced differentiation of SH-SY5Y human neuroblastoma cell line. Mol Biol Rep. 2020;47:877588. [DOI] [PubMed]
    Bayeva N, Coll E, Piskareva O. Differentiating Neuroblastoma: A Systematic Review of the Retinoic Acid, Its Derivatives, and Synergistic Interactions. J Pers Med. 2021;11:211. [DOI] [PubMed] [PMC]
    Hunsu VO, Facey COB, Fields JZ, Boman BM. Retinoids as Chemo-Preventive and Molecular-Targeted Anti-Cancer Therapies. Int J Mol Sci. 2021;22:7731. [DOI] [PubMed] [PMC]
    Protein Aligner [Internet]. OneAngstrom; [cited 2024 Jul 4]. Available from: https://www.samson-connect.net/extensions/0bee42d5-181b-b366-39a5-36607f0b5731
    Engel J, Richters A, Getlik M, Tomassi S, Keul M, Termathe M, et al. Targeting Drug Resistance in EGFR with Covalent Inhibitors: A Structure-Based Design Approach. J Med Chem. 2015;58:684463. [DOI] [PubMed]
    Kleywegt GJ, Bergfors T, Senn H, Motte PL, Gsell B, Shudo K, et al. Crystal structures of cellular retinoic acid binding proteins I and II in complex with all-trans-retinoic acid and a synthetic retinoid. Structure. 1994;2:124158. [DOI] [PubMed]
    Wang L-h, Evers A, Monecke P, Naumann T. Ligand based lead generation - considering chemical accessibility in rescaffolding approaches via BROOD. J Cheminf. 2012;4:O20. [DOI] [PMC]
    McGann M. FRED and HYBRID docking performance on standardized datasets. J Comput Aided Mol Des. 2012;26:897906. [DOI] [PubMed]
    López-Ramos M, Perruccio F. HPPD: ligand- and target-based virtual screening on a herbicide target. J Chem Inf Model. 2010;50:80114. [DOI] [PubMed]
    Janowski PA, Moriarty NW, Kelley BP, Case DA, York DM, Adams PD, et al. Improved ligand geometries in crystallographic refinement using AFITT in PHENIX. Acta Crystallogr D Struct Biol. 2016;72:106272. [DOI] [PubMed] [PMC]
    VIDA v4.4.0 released [Internet]. OpenEye, Cadence Molecular Sciences; c2025 [cited 2023 Jun 6]. Available from: https://www.eyesopen.com/news/vidav4.4.0-released
    Eberhardt J, Santos-Martins D, Tillack AF, Forli S. AutoDock Vina 1.2.0: New Docking Methods, Expanded Force Field, and Python Bindings. J Chem Inf Model. 2021;61:38918. [DOI] [PubMed] [PMC]
    Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010;31:45561. [DOI] [PubMed] [PMC]
    Moitessier N, Pottel J, Therrien E, Englebienne P, Liu Z, Tomberg A, et al. Medicinal Chemistry Projects Requiring Imaginative Structure-Based Drug Design Methods. Acc Chem Res. 2016;49:164657. [DOI] [PubMed]
    Thomsen R, Christensen MH. MolDock: a new technique for high-accuracy molecular docking. J Med Chem. 2006;49:331521. [DOI] [PubMed]
    Toxicity Estimation Software Tool (TEST) [Internet]. [Cited 2024 Sep 9]. Available from: https://www.epa.gov/comptox-tools/toxicity-estimation-software-tool-test
    Parrot M, Tajmouati H, Silva VBRd, Atwood BR, Fourcade R, Gaston-Mathé Y, et al. Integrating synthetic accessibility with AI-based generative drug design. J Cheminform. 2023;15:83. [DOI] [PubMed] [PMC]
    BIOVIA Discovery Studio Visualizer [Internet]. Dassault Systèmes; c2002-2025 [cited 2023 May 4]. Available from: https://discover.3ds.com/discovery-studio-visualizer-download
    BROOD [Internet]. OpenEye, Cadence Molecular Sciences; c2025 [cited 2023 Apr 10]. Available from: https://www.eyesopen.com/BROOD
    Morphy R, Kay C, Rankovic Z. From magic bullets to designed multiple ligands. Drug Discov Today. 2004;9:64151. [DOI] [PubMed]
    Li X, Li X, Liu F, Li S, Shi D. Rational Multitargeted Drug Design Strategy from the Perspective of a Medicinal Chemist. J Med Chem. 2021;64:10581605. [DOI] [PubMed]
    Klebe G. From In Vitro to In Vivo: Optimization of ADME and Toxicology Properties. In: Klebe G, editor. Drug Design. Springer, Berlin: Heidelberg; 2024. pp. 291–308. [DOI]