MYCN Impact on High-Risk Neuroblastoma: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , , , ,

Among childhood cancers, neuroblastoma is the most diffuse solid tumor and the deadliest in children. While to date, the pathology has become progressively manageable with a significant increase in 5-year survival for its less aggressive form, high-risk neuroblastoma (HR-NB) remains a major issue with poor outcome and little survivability of patients. The staging system has also been improved to better fit patient needs and to administer therapies in a more focused manner in consideration of pathology features. New and improved therapies have been developed; nevertheless, low efficacy and high toxicity remain a staple feature of current high-risk neuroblastoma treatment. For this reason, more specific procedures are required, and new therapeutic targets are also needed for a precise medicine approach. In this scenario, MYCN is certainly one of the most interesting targets. Indeed, MYCN is one of the most relevant hallmarks of HR-NB, and many studies has been carried out in recent years to discover potent and specific inhibitors to block its activities and any related oncogenic function. N-Myc protein has been considered an undruggable target for a long time. 

  • MYCN
  • high-risk neuroblastoma
  • pediatric tumor
  • neuroblastoma therapeutics
  • undruggable targets

1. Introduction

Neuroblastoma is one of the most diffuse neoplasia in children (<14 years), preceded only by leukemia, lymphomas and central nervous system neoplasms. In 2021, a total of 10,500 children in the USA were diagnosed with cancer, and 6% of these cases were neuroblastoma [1]. Notably, in the last fifteen years the 5-year survival rate has increased from 72% to 81%, highlighting an important advancement in the treatment and therapy of the pathology [1][2], but the problem still remains. In particular, high-risk neuroblastoma (HR-NB) patients, accounting for about half of the overall cases, can be considered more fragile and meaningful due to the poor outcome associated with this condition. In fact, patients affected by HR-NB normally show a strong reduction in 5-year survival with a mean of 50%, in comparison with low and intermediate-risk patients that reach about 90–85% [3]. HR-NB patients normally show mutations in major risk biomarkers such as MYCN amplification [4] and common segmental chromosomal aberration (SCA) [5]. The age of the patient at the pathology diagnosis is considered another important factor, and for this reason a proper evaluation of the tumor is fundamental not only for the treatment of the pathology, but also for better outcomes assessment.
The neuroblastoma risk classification system underwent modification in past decades, from the Evans staging system in 1971 to the most recent International Neuroblastoma Staging System (INSS) or International Neuroblastoma Risk Group Staging System (INRGSS), which was used for the first time in 2005 [6]. Recently, the Children’s Oncology Group revised this system, (Table 1) aiming to improve the correspondence between patient therapy and stage assignation and more highly considering the impact of the risk biomarkers [3]. The re-assignation of patients from INRGSS classes of risk [7] to newly identified classes highlighted that HR patients were well assigned using the previous version of INRGSS (only 3.4% of non-HR shifted to HR), and that MYCN is one of the most relevant factors occurring in a majority of case scenarios in the HR-NB patients [3].
Table 1. International Neuroblastoma Risk Group Staging System revised and updated by the Children’s Oncology Group in 2021.
Abbreviation: AMP, amplification; bx, biopsy; SCA, segmental chromosome aberration; DI, DNA index; INPC, International Neuroblastoma Pathology Classification; FH, favorable INPC histology; UH unfavorable INPC histology; LR, low-risk; IR, intermediate-risk; HR, high-risk; NA, not applicable. Note: Any included unknown.
MYCN amplification occurs in almost 25% of all neuroblastoma cases and correlates with HR-NB and poor prognosis [8][9][10]. Interestingly, despite the expectations, MYCN amplification does not always result in a higher expression of mRNA or protein [11][12][13], highlighting that more complex interaction should be considered in HR-NB disclosure [14][15]. While the relevance of the correlation between MYCN amplification and over-expression is still under discussion, the role of N-Myc oncoprotein as a potential target for therapy is well established [16][17][18]. In the same way, the prediction potential of MYCN amplification is considered a standard tool to distinguish tumors subtype and patient prognosis in medical practice [19][20]. For this reason, MYCN emerges as a key component of HR-NB in diagnostic, prognostic and medical procedures, such as in pharmaceutical research.

2. Diagnosis of High-Risk Neuroblastoma

A proper diagnostic evaluation can be relevant to perform the best patient assignation to a risk group and to select the best therapy available. The most common and used methods for NB assessment include a combination of histological observation with imaging features and multiple laboratory tests such as fluorescence in situ hybridization (FISH), polymerase chain reaction polymerase chain reaction (PCR), multiplex ligation-dependent probe amplification (MLPA) or array comparative genomic hybridization (aCGH) [4][21][22]. In a similar manner to laboratory tests, imaging also offers a broad spectrum of techniques that have been developed or improved over the years, in order to provide reliable tools to assess neuroblastoma staging and follow-up [23][24].
For example, anatomical imaging techniques such as computed tomography (CT) and magnetic resonance imaging (MRI) represent the diagnostic standard for the precise localization of primary tumor mass and provide anatomical details for consequent loco-regional staging [25][26]. Interestingly, the modern CT-based approach has shown the ability to predict MYCN amplification (MNA) status in neuroblastoma through the generation of radiomics profiles of tumors [27], highlighting the great potential of this technique. In contrast, functional imaging analysis, using positron emission tomography (PET) or the combined approach PET/CT, is less useful when approaching the primary tumor, but it is more reliable for distant metastasis disclosure and proper tumor evaluation after anatomical distortions induced by surgery or radiation [24][28]. Moreover, the use of different tracers in PET, such as 123I-MIBG, 18F-FDG, and 99mTc-MDP, may allow us to obtain the best imaging performance on neuroblastoma tumors, differing in biological and molecular characteristics and further improving the value of this tool [28].
With the advancement of new technologies, the cellular, molecular, genetic and anatomical features of NB become constantly more fast and accessible for analysis, allowing proper patient assignation [3][29]. In particular, MNA status is considered the strongest indicator for both HR-NB assignation and poor prognosis [29][30][31], and a fast diagnosis can be particularly relevant in consideration of the increase in risk with the age of the patient [3][32][33].

2.1. Circulating Free DNA and Circulating Free Cells

Most of the tests for NB assessment require bioptic material. The biopsy procedure needed for the analysis is an invasive procedure and the tumor mass is not always accessible for recovery and analysis. Moreover, the analysis of a tumor with an abundance of non-malignant cells [34] can be confounding and show MNA heterogeneous pattern results [35][36]. For this reason, a new approach was developed involving the use of circulating free DNA (cfDNA) that was isolated from plasma or serum [37]. Using this so called “liquid biopsy” approach, it is possible to overcome the problem related to the invasiveness of the surgical procedure and the genetic heterogeneity found in solid tissue [38]. The technique is fast and fully reliable for the assessment of MYCN copy number using a PCR based analysis [39][40][41]. Moreover, it was demonstrated that cfDNA can be used in combination with specifically quantitative PCR (q-PCR) to perform an MNA analysis with high sensitivity and specificity in patients with advanced disease [42][43].
Interestingly, circulating messenger RNA (mRNA), circulating tumor cells (CTCs) and circulating NB exosomes can also be found in biological fluids and used as biomarkers for diagnosis and prognosis assessment [44]. In particular, CTCs can provide comprehensive tumor profiling involving RNA, protein and/or metabolic information, while cfDNA only contains a genomic statement [45][46]. Of course, this method is more expensive and slower than cfDNA use [47], but it can be extensively considered as complementary in HR-NB evaluation, as suggested in other cancer studies [48][49].

3. Current Therapies of High-Risk Neuroblastoma

Current NB therapy in the majority of countries is constituted by three phases: induction, consolidation and maintenance therapies (Figure 1), and lasts approximately 18 months, varying by the patient risk [50][51]. Treatment strategies for each phase may include chemotherapy, surgical resection, high-dose chemotherapy with autologous stem cell rescue, radiation therapy, immunotherapy and isotretinoin, but the modality of the administration of the single procedures mostly depends on patient risk status [52]. Low- and intermediate-risk patients, for example, show high overall survival with minimal therapy approach, involving only surgical resection alone or combined with small chemotherapy administration in the induction phase [53][54][55]. On the other hand, the high-risk group need a more aggressive approach, and each phase is involved in the management of the pathology with the administration of any possible therapy available [52][56][57][58], being more challenging not only for the efficacy assessment, but also for the safety of the patient.
Figure 1. Neuroblastoma therapies are reported in relationship to risk group. Each treatment is collected in its own neuroblastoma therapy phase from induction to maintenance. Low- and intermediate-risk patients only receive the induction phase, while high-risk patients receive all treatments.

4. MYCN as Prognostic Indicator in High-Risk Neuroblastoma

Despite the advancement in medical standard therapy for the treatment of HR-NB, there is no specific therapy for MNA patients [59][60][61], but this aspect of the pathology becomes increasingly relevant in consideration of its prognostic effect. For example, the prognostic impact of MNA is particularly relevant in infants with stage M disease where both event-free survival (EFS) and overall survival (OS) are higher in an MYCN non-amplified tumor, that shows better outcomes in comparison with MNA patients (EFS and OS: 82.5% and 90.8% versus 36.9% and 44.8%) [3]. In a similar manner, the behavior of an L2 and MS stage tumor is mostly impacted by MYCN status. EFS and OS are lower at any age when MYCN is amplified, while the absence of amplification correlates with better survivability results for the patients [3]. Interestingly, MNA association with worse prognosis appears more pronounced in the context of other favorable prognostic features and can be considered as an indicator for aggressive intervention [62], highlighting how MYCN gene status impacts therapy selection and medical decision making.
While MNA is considered a staple in neuroblastoma diagnosis, as mentioned before, MYCN over-expression has a more controversial role; in a similar manner, MYCN expression has the same prognostic behavior. Some studies reported that MYCN expression is valuable for prognostic purpose, but only in a specific pathology context. In particular, a patient cohort showed that the lowest value in MNA cases has a more negative outcome when MYCN and mRNA levels are higher, in comparison to patients with higher MNA levels where mRNA expression cannot be considered as prognostic [11]. Otherwise N-Myc protein expression shows different prognostic value, being reported as an indicator of poor outcome regardless of MNA status [31]. These findings together suggest that deep investigation on how and when MNA or MYCN expression are predictors of patients’ outcomes needs to be improved, but their value as key elements of prognosis assessment is undeniable, remaining so in the establishment of HR-NB therapy administration.

5. MYCN Determines High-Risk Neuroblastoma

Despite MYCN amplification being the first discovered genetic mechanism in neuroblastoma, its role in driving the pathology is not fully understood [63][64][65]. In fact, MYCN amplification leads to deep remodeling of the cancer cell, influencing its apoptosis resistance, its undifferentiated status, its metabolic landscape and immune evasion.
Different studies showed that MYCN over-expression is an obstacle to neuronal differentiation. High-risk and MYCN amplified neuroblastoma present a different transcriptional profile, where different pathways and genes related to differentiation are particularly altered [66]. Indeed, MYCN amplified neuroblastoma cell lines fail to differentiate in response to 13-cis-retinoic acid [8]. The concomitant inhibition of MYCN and the administration of RA is able to reverse this block [66]. Moreover, the ectopic expression of MYCN in precursor cells blocks the differentiation in chromaffin cells.
In addition, MYCN controls both proliferation and apoptosis: many studies showed that over-expression disrupts the cell cycle, leading to maintained apoptosis inhibition and induced proliferation [67]. In fact, blocking MYCN leads to G1 phase cell accumulation and slows down the transition to S phase and PI3K repression (which is known to promote cell growth and proliferation) [68][69][70][71][72]. MYCN also positively affects the expression of other key cellular regulator such as E2 factor (E2F) and inhibitor of differentiation 2 (ID2), which are also involved in cell cycle progression [69][73][74]. As an additional mechanism, MYCN amplification is also associated to TERT expression and telomere anomalies [75][76].
Interestingly, MYCN can promote apoptosis and/or sensitizes cancer cells to cytotoxic drugs [77][78]. MYCN is able to promote the expression of phorbol-12-myristate-13-acetate-induced protein 1 (NOXA), which is a pro-apoptotic regulator. Moreover, E-box elements are present in the promoter of p53, which is the most known onco-suppressor able to stop cell proliferation and induce apoptosis (even if in a significant part of neuroblastoma p53 is found mutated) [78]. As it is known, the murine double minute 2 (MDM2) is a negative regulator of p53 and is over-expressed in different human malignant tumors [79]. In particular, MDM2 is able to reduce p53 levels using the mechanism of binding to p53 with consequent ubiquitination and proteosomal degradation [80][81]. However, MDM2 also promotes the stability of MYCN, while the latter induces MDM2 transcription [82][83][84]. Thus, MYCN can induce the transcription of p53 and MDM2, regulating the balance between proliferation and apoptosis. Over-expression is thought to alter this precarious equilibrium, inducing MDM2 expression and p53 blocking [82][83][84].
Early studies showed how metabolism is deeply altered in cancer cells. In fact, cancer cells are skewed towards rapid ATP production, which is generally obtained through the “Warburg Effect”, where cells rely on glycolysis and mitochondrial respiration is impaired [85][86][87][88]. Cancer cells then use the fatty acids and glutamine as a source for biosynthesis and ultimately sustain the cell growth and proliferation. Moreover, this alteration in the mitochondria leads to reactive oxygen species (ROS) production, while the fatty acid oxidation is used to replenish the NADPH pool in order to prevent excessive oxidative stress [89][90]. Neuroblastoma and, in particular, MYCN amplified tumors are heavily dependent in glutamine, and blocking MYCN leads to the arrest of glutamine transport [91][92]. In addition, MYCN promotes the glycolysis and fatty acid uptake and leads to mitochondria alteration [92][93]. MNA tumors also present different metabolic alterations, leading to an increase in iron uptake. For instance, it has recently been shown that MYCN induces massive lipid peroxidation and cysteine depletion. This leads MNA to be sensible to oxidative stress and especially to ferroptosis [94]. In this context, blocking MYCN in MNA neuroblastoma leads to ROS production (through TRAP1 decrease), which the cancer cell fails to handle, consequently undergoing apoptosis [95]. In addition, MYCN also blocks autophagy and mitophagy (an autophagy sub-pathway used by the cell to recycle damaged mitochondria), and it has been shown in inducible MYCN cell line (TET21N) that the MYCN blocking restores this pathway (OPTN transcription) [95]. All these studies show a strong rewiring of the metabolism by MYCN expression and the fine grain regulation of the redox equilibrium.
Different studies showed the role of the phosphatidylinositol 3-kinase (PI3K)/mTOR pathway in neuroblastoma [70][96][97]. In fact, the mTOR pathway is known to stabilize N-Myc, and its blocking affects cell growth. Moreover, retinoic acid has been described as being capable of mTOR inhibition. While the N-Myc protein is stabilized by the mTOR complex, it also regulates the expression of different MTOR genes in a positive loop [66][98]. Indeed, MNA cell lines have a higher expression of mTOR genes, and they are more resistant to mTOR inhibitors. Furthermore, it has been shown that mTOR is also negatively associated to the prognosis [66].
MYCN amplification impact is not limited to the cancer cell itself but also to the tumor microenvironment. In fact, MNA cancers remodel the external environment to sustain their growth and the immune evasion [99]. A significant portion of MNA tumors present PD-L1 expression and MHC I complex down-regulation, leading to a suppressive micro-environment [100][101]. In addition, MNAs are also enriched in M2 macrophages and CD4+ T helper 2 cells [99]. Macrophages are also responsible for maintaining a hypoxic environment and lead to the transcription of hypoxia inducible factor (HIF 2α), which ultimately leads to vascularization and metastasis spreading. Indeed, HR-NB that are fast growing are high in the immunostaining for HIF2 [102]. However, neuroblastoma also exploits other strategies such as expressing other immune-suppressive molecules (such as CD276), miRNAs and exosomes release [103]. Overall, both innate and adaptive immune systems seem to be down-regulated [99]. This complex landscape is probably at the origin of the fact that immune-therapy has shown modest results [99][104][105].
MYCN amplification also leads to extracellular matrix (ECM) modification. ECM is often altered in HR-NB with anomalous collagen I deposit and often correlates with bad prognosis [106][107][108][109]. These alterations are also promoted by the hypoxic and inflamed state of the tumor microenvironment [110][111][112]. Collagen I inhibition has shown a promising effect, allowing better chemotherapy delivery [113]. Moreover, different matrix metalloproteinases (MMPs) are altered in neuroblastoma which are linked to bad prognosis, angiogenesis and metastasis promotion [114][115][116].
HIF2 expression, ECM alteration and VEGF expression in HR-NB also lead to new vascularization. These tumors show more aggressive features such as more immature states and more easily spreading metastasis [117][118][119][120][121][122]. Moreover, there is evidence that PI3K kinases promote VEGF expression via MYCN [123][124]. Interestingly, blocking PI3K by SF1126 in neuroblastoma led to reduced MYCN expression, cell death and angiogenesis block, while temporarily increasing the macrophages’ M1 to M2 ratio, showing how all these mechanisms are interconnected [125][126][127][128].
Furthermore, the neuroblastoma tumor microenvironment presents an enrichment of cancer-associated fibroblasts (CAFs) and mesenchymal stromal cells (MSCs). This enrichment correlates with progression and it is important to sustain the tumor growth, micro-vessels formation and progression, showing correlation with poor outcome [129][130][131]. In fact, CAF produces TGFβ (a cytokine with immunosuppressive property) and CCL2, which recruit TAM to neuroblastoma [132][133][134]. In addition, studies have highlighted that the CAF area extension correlates with MYCN amplification [135][136][137]. MSCs are also involved in inducing an immune suppressive environment, leading to the recruitment of ulterior suppressive cells (T regulatory cells and macrophages).
Other than the direct action of MYCN on cellular process leading to neuroblastoma, in the past few years, many epigenetic mechanisms have been discovered regulating MYCN with a specific role in HR-NB development [138]. Micro RNAs (miRNAs) are small single-stranded RNA molecules that function as post-transcriptional RNA regulators [139]. MiRNAs targeting MYCN were found to be particularly important in regulating its expression in neuroblastoma in different ways [8][140]. Some miRNAs targeting MYCN, such as the let-7 family, work as inhibitors [141] and are down-regulated in neuroblastoma, inducing N-Myc protein expression [142][143][144]. These miRNA types are considered the most common, but other miRNAs were found to work with the opposite mechanism. For instance, miRNAs such as the family of miR-17-92 are substantially employed as MYCN up-regulators. Interestingly N-Myc is able to stimulate the expression of miR-17-92 cluster, suggesting the presence of a positive feedback mechanism of regulation between MYCN and the miR-17-92 cluster itself [144][145][146].
Moreover, the natural antisense transcript was found to be able to regulate MYCN expression. In particular, MYCN locus is able to generate an antisense transcript known as MYCNOS (or N-cym) [147]. This transcript originates from the opposite strand of the locus and regulates MYCN as either regulatory long non-coding RNA (lncRNA) or protein. In fact the lncRNA of MYCNOS regulates MYCN promoter through the recruiting of protein in this site [148], while the MYCNOS-encoded protein works as an inhibitor of glycogen synthase kinase 3 β (GSK3beta), stabilizing in this way the NMYC protein [149]. For this reason, high levels of MYCNOS can be found relative to MYCN over-expression and correlate with poor outcome in neuroblastoma [13][150]. Furthermore, MYCNOS itself was found to be regulated from some non-coding RNAs such as lncUSMycN [151] that are able to suppress its expression and indirectly regulate MYCN [148].
Methylation is another regulatory system used by cells to define gene expression and cellular function. While the correlation between MYCN and its methylation status in neuroblastoma is still unclear, many other genes involved in neuroblastoma transformation were well characterized in both MYCN amplified and non-amplified tumors [138][152]. Extensive methylation was found in many onco-suppressor genes with no particular correlation to MYCN status [153]. In particular, miRNAs were reported to be methylated in neuroblastoma cell lines, highlighting their role in tumor progression and poor prognosis [154].

6. MYCN as Therapeutic Target

Despite current therapeutic advances and ongoing clinical trials, NB remains a complex medical challenge, especially in the high-risk cases, and the discovery of new therapeutic approaches is needed to improve patient welfare and outcome (Table 2) [155].
Table 2. Summary of current therapies and the new therapeutic approach in the treatment of neuroblastoma and high-risk neuroblastoma.
In this scenario, MYCN certainly represents an ideal therapeutic target given its correlation with rapid tumor progression, poor prognosis and the limited expression in normal cells and tissue, suggesting high tolerability for an MYCN-specific approach [8][156]. Many attempts and investigations have been made to develop specific inhibitors for N-Myc protein, but both direct or indirect N-Myc modulators (Figure 2) failed to result in an efficient or reliable N-Myc-specific therapy [157]. Unfortunately, N-Myc targeting shows a different issue. Some of these issues can resemble any transcription factor, while some challenges are more peculiar and reside in the lack of specific interaction site on the protein [158][159] or in the homology with the MYC family oncogene, increasing the difficulty of preserving the physiological function of c-Myc protein in normal tissue [67][160]. For this reason, new strategies have been proposed in the hope of overcoming the failure of the precedent attempt to make MYCN a fully available target for HR-NB [8][18][67][161].
Figure 2. Schematic representation of drugs directly or indirectly targeting MYCN. Drugs are marked in red and reported as class, named on mechanism of action. Abbreviation: MDM2, murine double minute 2; PI3K, Phosphoinositide 3-kinases; PI3Ki, Phosphoinositide 3-kinases inhibitor; BETi, bromodomain and extra-terminal domain family inhibitor; BRD4, bromodomain-containing protein 4; mTORC1, mammalian target of rapamycin complex 1; mTORC2, mammalian target of rapamycin complex 2; mTORi, mammalian target of rapamycin inhibitor; PROTACs, proteolysis-targeting chimeras; MIZ1, MYC-interacting zinc-finger protein 1; HDAC, histone deacetylase; HDACi, histone deacetylase inhibitor; E3, E3 ubiquitin ligase; E2, E2 ubiquitin-conjugating enzyme; AURKA, Aurora kinase A; AURKAi, Aurora kinase A inhibitor; Ub, ubiquitin.

This entry is adapted from the peer-reviewed paper 10.3390/cancers14184421

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33.
  2. Siegel, R.; Ward, E.; Brawley, O.; Jemal, A. Cancer Statistics, 2011: The Impact of Eliminating Socioeconomic and Racial Disparities on Premature Cancer Deaths. CA Cancer J. Clin. 2011, 61, 212–236.
  3. Irwin, M.S.; Naranjo, A.; Zhang, F.F.; Cohn, S.L.; London, W.B.; Gastier-Foster, J.M.; Ramirez, N.C.; Pfau, R.; Reshmi, S.; Wagner, E.; et al. Revised Neuroblastoma Risk Classification System: A Report from the Children’s Oncology Group. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2021, 39, 3229–3241.
  4. Ambros, P.F.; Ambros, I.M.; Brodeur, G.M.; Haber, M.; Khan, J.; Nakagawara, A.; Schleiermacher, G.; Speleman, F.; Spitz, R.; London, W.B.; et al. International Consensus for Neuroblastoma Molecular Diagnostics: Report from the International Neuroblastoma Risk Group (INRG) Biology Committee. Br. J. Cancer 2009, 100, 1471–1482.
  5. Pinto, N.; Mayfield, J.R.; Raca, G.; Applebaum, M.A.; Chlenski, A.; Sukhanova, M.; Bagatell, R.; Irwin, M.S.; Little, A.; Rawwas, J.; et al. Segmental Chromosomal Aberrations in Localized Neuroblastoma Can Be Detected in Formalin-Fixed Paraffin-Embedded Tissue Samples and are Associated with Recurrence: Segmental Chromosomal Aberrations in Localized Neuroblastoma. Pediatr. Blood Cancer 2016, 63, 1019–1023.
  6. Sokol, E.; Desai, A. The Evolution of Risk Classification for Neuroblastoma. Children 2019, 6, 27.
  7. Cohn, S.L.; Pearson, A.D.J.; London, W.B.; Monclair, T.; Ambros, P.F.; Brodeur, G.M.; Faldum, A.; Hero, B.; Iehara, T.; Machin, D.; et al. The International Neuroblastoma Risk Group (INRG) Classification System: An INRG Task Force Report. J. Clin. Oncol. 2009, 27, 289–297.
  8. Huang, M.; Weiss, W.A. Neuroblastoma and MYCN. Cold Spring Harb. Perspect. Med. 2013, 3, a014415.
  9. Maris, J.M.; Hogarty, M.D.; Bagatell, R.; Cohn, S.L. Neuroblastoma. Lancet 2007, 369, 2106–2120.
  10. Rubie, H.; Hartmann, O.; Michon, J.; Frappaz, D.; Coze, C.; Chastagner, P.; Baranzelli, M.C.; Plantaz, D.; Avet-Loiseau, H.; Bénard, J.; et al. N-Myc Gene Amplification is a Major Prognostic Factor in Localized Neuroblastoma: Results of the French NBL 90 Study. Neuroblastoma Study Group of the Société Francaise d’Oncologie Pédiatrique. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 1997, 15, 1171–1182.
  11. Tang, X.X.; Zhao, H.; Kung, B.; Kim, D.Y.; Hicks, S.L.; Cohn, S.L.; Cheung, N.-K.; Seeger, R.C.; Evans, A.E.; Ikegaki, N. The MYCN Enigma: Significance of MYCN Expression in Neuroblastoma. Cancer Res. 2006, 66, 2826–2833.
  12. Nisen, P.D.; Waber, P.G.; Rich, M.A.; Pierce, S.; Garvin, J.R.; Gilbert, F.; Lanzkowsky, P. N-Myc Oncogene RNA Expression in Neuroblastoma. J. Natl. Cancer Inst. 1988, 80, 1633–1637.
  13. Jacobs, J.F.M.; van Bokhoven, H.; van Leeuwen, F.N.; Hulsbergen-van de Kaa, C.A.; de Vries, I.J.M.; Adema, G.J.; Hoogerbrugge, P.M.; de Brouwer, A.P.M. Regulation of MYCN Expression in Human Neuroblastoma Cells. BMC Cancer 2009, 9, 239.
  14. Scott, D.; Elsden, J.; Pearson, A.; Lunec, J. Genes Co-Amplified with MYCN in Neuroblastoma: Silent Passengers or Co-Determinants of Phenotype? Cancer Lett. 2003, 197, 81–86.
  15. Schwab, M. MYCN in Neuronal Tumours. Cancer Lett. 2004, 204, 179–187.
  16. Gherardi, S.; Valli, E.; Erriquez, D.; Perini, G. MYCN-Mediated Transcriptional Repression in Neuroblastoma: The Other Side of the Coin. Front. Oncol. 2013, 3, 42.
  17. Zafar, A.; Wang, W.; Liu, G.; Xian, W.; McKeon, F.; Zhou, J.; Zhang, R. Targeting the P53-MDM2 Pathway for Neuroblastoma Therapy: Rays of Hope. Cancer Lett. 2021, 496, 16–29.
  18. Wolpaw, A.J.; Bayliss, R.; Büchel, G.; Dang, C.V.; Eilers, M.; Gustafson, W.C.; Hansen, G.H.; Jura, N.; Knapp, S.; Lemmon, M.A.; et al. Drugging the “Undruggable” MYCN Oncogenic Transcription Factor: Overcoming Previous Obstacles to Impact Childhood Cancers. Cancer Res. 2021, 81, 1627–1632.
  19. Yue, Z.-X.; Huang, C.; Gao, C.; Xing, T.-Y.; Liu, S.-G.; Li, X.-J.; Zhao, Q.; Wang, X.-S.; Zhao, W.; Jin, M.; et al. MYCN Amplification Predicts Poor Prognosis Based on Interphase Fluorescence in Situ Hybridization Analysis of Bone Marrow Cells in Bone Marrow Metastases of Neuroblastoma. Cancer Cell Int. 2017, 17, 43.
  20. Campbell, K.; Gastier-Foster, J.M.; Mann, M.; Naranjo, A.H.; van Ryn, C.; Bagatell, R.; Matthay, K.K.; London, W.B.; Irwin, M.S.; Shimada, H.; et al. Association of MYCN Copy Number with Clinical Features, Tumor Biology, and Outcomes in Neuroblastoma: A Report from the Children’s Oncology Group. Cancer 2017, 123, 4224–4235.
  21. Swift, C.C.; Eklund, M.J.; Kraveka, J.M.; Alazraki, A.L. Updates in Diagnosis, Management, and Treatment of Neuroblastoma. Radiogr. Rev. Publ. Radiol. Soc. N. Am. Inc. 2018, 38, 566–580.
  22. Zhan, Y.; Shi, S.; Ehlerding, E.B.; Graves, S.A.; Goel, S.; Engle, J.W.; Liang, J.; Tian, J.; Cai, W. Radiolabeled, Antibody-Conjugated Manganese Oxide Nanoparticles for Tumor Vasculature Targeted Positron Emission Tomography and Magnetic Resonance Imaging. ACS Appl. Mater. Interfaces 2017, 9, 38304–38312.
  23. Bar-Sever, Z.; Biassoni, L.; Shulkin, B.; Kong, G.; Hofman, M.S.; Lopci, E.; Manea, I.; Koziorowski, J.; Castellani, R.; Boubaker, A.; et al. Guidelines on Nuclear Medicine Imaging in Neuroblastoma. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 2009–2024.
  24. Kroiss, A.S. Current Status of Functional Imaging in Neuroblastoma, Pheochromocytoma, and Paraganglioma Disease. Wien. Med. Wochenschr. 2019, 169, 25–32.
  25. Sarioglu, F.C.; Salman, M.; Guleryuz, H.; Ozer, E.; Cecen, E.; Ince, D.; Olgun, N. Radiological Staging in Neuroblastoma: Computed Tomography or Magnetic Resonance Imaging? Pol. J. Radiol. 2019, 84, e46–e53.
  26. Sofka, C.M.; Semelka, R.C.; Kelekis, N.L.; Worawattanakul, S.; Chung, C.J.; Gold, S.; Fordham, L.A. Magnetic Resonance Imaging of Neuroblastoma Using Current Techniques. Magn. Reson. Imaging 1999, 17, 193–198.
  27. Wu, H.; Wu, C.; Zheng, H.; Wang, L.; Guan, W.; Duan, S.; Wang, D. Radiogenomics of Neuroblastoma in Pediatric Patients: CT-Based Radiomics Signature in Predicting MYCN Amplification. Eur. Radiol. 2021, 31, 3080–3089.
  28. Sharp, S.E.; Parisi, M.T.; Gelfand, M.J.; Yanik, G.A.; Shulkin, B.L. Functional-Metabolic Imaging of Neuroblastoma. Q. J. Nucl. Med. Mol. Imaging 2013, 57, 6–20.
  29. Campbell, K.; Shyr, D.; Bagatell, R.; Fischer, M.; Nakagawara, A.; Nieto, A.C.; Brodeur, G.M.; Matthay, K.K.; London, W.B.; DuBois, S.G. Comprehensive Evaluation of Context Dependence of the Prognostic Impact of MYCN Amplification in Neuroblastoma: A Report from the International Neuroblastoma Risk Group (INRG) Project. Pediatr. Blood Cancer 2019, 66, e27819.
  30. Yanishevski, D.; McCarville, M.B.; Doubrovin, M.; Spiegl, H.R.; Zhao, X.; Lu, Z.; Federico, S.M.; Furman, W.L.; Murphy, A.J.; Davidoff, A.M. Impact of MYCN Status on Response of High-Risk Neuroblastoma to Neoadjuvant Chemotherapy. J. Pediatr. Surg. 2020, 55, 130–134.
  31. Chan, H.S.; Gallie, B.L.; DeBoer, G.; Haddad, G.; Ikegaki, N.; Dimitroulakos, J.; Yeger, H.; Ling, V. MYCN Protein Expression as a Predictor of Neuroblastoma Prognosis. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 1997, 3, 1699–1706.
  32. Van Heerden, J.; Esterhuizen, T.M.; Hendricks, M.; Poole, J.; Büchner, A.; Naidu, G.; du Plessis, J.; van Emmenes, B.; Uys, R.; Hadley, G.P.; et al. Age at Diagnosis as a Prognostic Factor in South African Children with Neuroblastoma. Pediatr. Blood Cancer 2021, 68, e28878.
  33. Sokol, E.; Desai, A.V.; Applebaum, M.A.; Valteau-Couanet, D.; Park, J.R.; Pearson, A.D.J.; Schleiermacher, G.; Irwin, M.S.; Hogarty, M.; Naranjo, A.; et al. Age, Diagnostic Category, Tumor Grade, and Mitosis-Karyorrhexis Index are Independently Prognostic in Neuroblastoma: An INRG Project. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2020, 38, 1906–1918.
  34. Mathew, P.; Valentine, M.B.; Bowman, L.C.; Rowe, S.T.; Nash, M.B.; Valentine, V.A.; Cohn, S.L.; Castleberry, R.P.; Brodeur, G.M.; Look, A.T. Detection of MYCN Gene Amplification in Neuroblastoma by Fluorescence in Situ Hybridization: A Pediatric Oncology Group Study. Neoplasia 2001, 3, 105–109.
  35. Squire, J.A.; Thorner, P.A.; Marrano, P.A.; Parkinson, D.I.; Ng, Y.K.; Gerrie, B.L.; Chilton-Macneill, S.; Zielenska, M. Identification of MYCN Copy Number Heterogeneity by Direct FISH Analysis of Neuroblastoma Preparations. Mol. Diagn. 1996, 1, 281–289.
  36. Marrano, P.; Irwin, M.S.; Thorner, P.S. Heterogeneity of MYCN Amplification in Neuroblastoma at Diagnosis, Treatment, Relapse, and Metastasis. Genes Chromosom. Cancer 2017, 56, 28–41.
  37. Marrugo-Ramírez, J.; Mir, M.; Samitier, J. Blood-Based Cancer Biomarkers in Liquid Biopsy: A Promising Non-Invasive Alternative to Tissue Biopsy. Int. J. Mol. Sci. 2018, 19, 2877.
  38. Namløs, H.M.; Boye, K.; Mishkin, S.J.; Barøy, T.; Lorenz, S.; Bjerkehagen, B.; Stratford, E.W.; Munthe, E.; Kudlow, B.A.; Myklebost, O.; et al. Noninvasive Detection of CtDNA Reveals Intratumor Heterogeneity and is Associated with Tumor Burden in Gastrointestinal Stromal Tumor. Mol. Cancer Ther. 2018, 17, 2473–2480.
  39. Combaret, V.; Audoynaud, C.; Iacono, I.; Favrot, M.-C.; Schell, M.; Bergeron, C.; Puisieux, A. Circulating MYCN DNA as a Tumor-Specific Marker in Neuroblastoma Patients. Cancer Res. 2002, 62, 3646–3648.
  40. Combaret, V.; Bergeron, C.; Noguera, R.; Iacono, I.; Puisieux, A. Circulating MYCN DNA Predicts MYCN-Amplification in Neuroblastoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2005, 23, 8919–8920; author reply 8920.
  41. Trigg, R.M.; Turner, S.D.; Shaw, J.A.; Jahangiri, L. Diagnostic Accuracy of Circulating-Free DNA for the Determination of MYCN Amplification Status in Advanced-Stage Neuroblastoma: A Systematic Review and Meta-Analysis. Br. J. Cancer 2020, 122, 1077–1084.
  42. Gotoh, T.; Hosoi, H.; Iehara, T.; Kuwahara, Y.; Osone, S.; Tsuchiya, K.; Ohira, M.; Nakagawara, A.; Kuroda, H.; Sugimoto, T. Prediction of MYCN Amplification in Neuroblastoma Using Serum DNA and Real-Time Quantitative Polymerase Chain Reaction. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2005, 23, 5205–5210.
  43. Iehara, T.; Yagyu, S.; Gotoh, T.; Ouchi, K.; Yoshida, H.; Miyachi, M.; Kikuchi, K.; Sugimoto, T.; Hosoi, H. A Prospective Evaluation of Liquid Biopsy for Detecting MYCN Amplification in Neuroblastoma Patients. Jpn. J. Clin. Oncol. 2019, 49, 743–748.
  44. Pinzani, P.; D’Argenio, V.; del Re, M.; Pellegrini, C.; Cucchiara, F.; Salvianti, F.; Galbiati, S. Updates on Liquid Biopsy: Current Trends and Future Perspectives for Clinical Application in Solid Tumors. Clin. Chem. Lab. Med. 2021, 59, 1181–1200.
  45. Rifatbegovic, F.; Frech, C.; Abbasi, M.R.; Taschner-Mandl, S.; Weiss, T.; Schmidt, W.M.; Schmidt, I.; Ladenstein, R.; Ambros, I.M.; Ambros, P.F. Neuroblastoma Cells Undergo Transcriptomic Alterations upon Dissemination into the Bone Marrow and Subsequent Tumor Progression. Int. J. Cancer 2018, 142, 297–307.
  46. Reza, K.K.; Dey, S.; Wuethrich, A.; Wang, J.; Behren, A.; Antaw, F.; Wang, Y.; Sina, A.A.I.; Trau, M. In Situ Single Cell Proteomics Reveals Circulating Tumor Cell Heterogeneity during Treatment. ACS Nano 2021, 15, 11231–11243.
  47. Lodrini, M.; Wünschel, J.; Thole-Kliesch, T.M.; Grimaldi, M.; Sprüssel, A.; Linke, R.B.; Hollander, J.F.; Tiburtius, D.; Künkele, A.; Schulte, J.H.; et al. Circulating Cell-Free DNA Assessment in Biofluids from Children with Neuroblastoma Demonstrates Feasibility and Potential for Minimally Invasive Molecular Diagnostics. Cancers 2022, 14, 2080.
  48. Beltran, H.; Jendrisak, A.; Landers, M.; Mosquera, J.M.; Kossai, M.; Louw, J.; Krupa, R.; Graf, R.P.; Schreiber, N.A.; Nanus, D.M.; et al. The Initial Detection and Partial Characterization of Circulating Tumor Cells in Neuroendocrine Prostate Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 1510–1519.
  49. Shaw, J.A.; Guttery, D.S.; Hills, A.; Fernandez-Garcia, D.; Page, K.; Rosales, B.M.; Goddard, K.S.; Hastings, R.K.; Luo, J.; Ogle, O.; et al. Mutation Analysis of Cell-Free DNA and Single Circulating Tumor Cells in Metastatic Breast Cancer Patients with High Circulating Tumor Cell Counts. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 88–96.
  50. Smith, V.; Foster, J. High-Risk Neuroblastoma Treatment Review. Children 2018, 5, 114.
  51. DuBois, S.G.; Macy, M.E.; Henderson, T.O. High-Risk and Relapsed Neuroblastoma: Toward More Cures and Better Outcomes. Am. Soc. Clin. Oncol. Educ. Book 2022, 42, 768–780.
  52. Tolbert, V.P.; Matthay, K.K. Neuroblastoma: Clinical and Biological Approach to Risk Stratification and Treatment. Cell Tissue Res. 2018, 372, 195–209.
  53. 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.
  54. Rubie, H.; de Bernardi, B.; Gerrard, M.; Canete, A.; Ladenstein, R.; Couturier, J.; Ambros, P.; Munzer, C.; Pearson, A.D.J.; Garaventa, A.; et al. Excellent Outcome with Reduced Treatment in Infants with Nonmetastatic and Unresectable Neuroblastoma without MYCN Amplification: Results of the Prospective INES 99.1. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2011, 29, 449–455.
  55. Strother, D.R.; London, W.B.; Schmidt, M.L.; Brodeur, G.M.; Shimada, H.; Thorner, P.; Collins, M.H.; Tagge, E.; Adkins, S.; Reynolds, C.P.; et al. Outcome after Surgery Alone or with Restricted Use of Chemotherapy for Patients with Low-Risk Neuroblastoma: Results of Children’s Oncology Group Study P9641. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2012, 30, 1842–1848.
  56. 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.
  57. Yanik, G.; Naranjo, A.; Parisi, M.T.; Shulkin, B.L.; Nadel, H.; Gelfand, M.J.; Ladenstein, R.; Boubaker, A.; Poetschger, U.; Valteau-Couanet, D.; et al. Impact of Post-Induction Curie Scores in High-Risk Neuroblastoma. Biol. Blood Marrow Transplant. 2015, 21, S107.
  58. Yanik, G.A.; Parisi, M.T.; Naranjo, A.; Nadel, H.; Gelfand, M.J.; Park, J.R.; Ladenstein, R.L.; Poetschger, U.; Boubaker, A.; Valteau-Couanet, D.; et al. Validation of Postinduction Curie Scores in High-Risk Neuroblastoma: A Children’s Oncology Group and SIOPEN Group Report on SIOPEN/HR-NBL1. J. Nucl. Med. Off. Publ. Soc. Nucl. Med. 2018, 59, 502–508.
  59. Park, J.R.; Kreissman, S.G.; London, W.B.; Naranjo, A.; Cohn, S.L.; Hogarty, M.D.; Tenney, S.C.; Haas-Kogan, D.; Shaw, P.J.; Kraveka, J.M.; et al. Effect of Tandem Autologous Stem Cell Transplant vs Single Transplant on Event-Free Survival in Patients with High-Risk Neuroblastoma: A Randomized Clinical Trial. JAMA 2019, 322, 746–755.
  60. 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.
  61. Ozkaynak, M.F.; Gilman, A.L.; London, W.B.; Naranjo, A.; Diccianni, M.B.; Tenney, S.C.; Smith, M.; Messer, K.S.; Seeger, R.; Reynolds, C.P.; et al. A Comprehensive Safety Trial of Chimeric Antibody 14.18 With GM-CSF, IL-2, and Isotretinoin in High-Risk Neuroblastoma Patients Following Myeloablative Therapy: Children’s Oncology Group Study ANBL0931. Front. Immunol. 2018, 9, 1355.
  62. Tonini, G.P.; Boni, L.; Pession, A.; Rogers, D.; Iolascon, A.; Basso, G.; Cordero di Montezemolo, L.; Casale, F.; Pession, A.; Perri, P.; et al. MYCN Oncogene Amplification in Neuroblastoma is Associated with Worse Prognosis, except in Stage 4s: The Italian Experience with 295 Children. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 1997, 15, 85–93.
  63. Brodeur, G.M. Neuroblastoma: Biological Insights into a Clinical Enigma. Nat. Rev. Cancer 2003, 3, 203–216.
  64. Brodeur, G.M.; Seeger, R.C.; Schwab, M.; Varmus, H.E.; Bishop, J.M. Amplification of N-Myc in Untreated Human Neuroblastomas Correlates with Advanced Disease Stage. Science 1984, 224, 1121–1124.
  65. 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.
  66. Lampis, S.; Raieli, S.; Montemurro, L.; Bartolucci, D.; Amadesi, C.; Bortolotti, S.; Angelucci, S.; Scardovi, A.L.; Nieddu, G.; Cerisoli, L.; et al. The MYCN Inhibitor BGA002 Restores the Retinoic Acid Response Leading to Differentiation or Apoptosis by the MTOR Block in MYCN-Amplified Neuroblastoma. J. Exp. Clin. Cancer Res. 2022, 41, 160.
  67. Rickman, D.S.; Schulte, J.H.; Eilers, M. The Expanding World of N-MYC–Driven Tumors. Cancer Discov. 2018, 8, 150–163.
  68. Harashima, H.; Dissmeyer, N.; Schnittger, A. Cell Cycle Control across the Eukaryotic Kingdom. Trends Cell Biol. 2013, 23, 345–356.
  69. Woo, C.-W.; Tan, F.; Cassano, H.; Lee, J.; Lee, K.C.; Thiele, C.J. Use of RNA Interference to Elucidate the Effect of MYCN on Cell Cycle in Neuroblastoma. Pediatr. Blood Cancer 2008, 50, 208–212.
  70. Cage, T.A.; Chanthery, Y.; Chesler, L.; Grimmer, M.; Knight, Z.; Shokat, K.; Weiss, W.A.; Gustafson, W.C. Downregulation of MYCN through PI3K Inhibition in Mouse Models of Pediatric Neural Cancer. Front. Oncol. 2015, 5, 111.
  71. Bosch, A.; Li, Z.; Bergamaschi, A.; Ellis, H.; Toska, E.; Prat, A.; Tao, J.J.; Spratt, D.E.; Viola-Villegas, N.T.; Castel, P.; et al. PI3K Inhibition Results in Enhanced Estrogen Receptor Function and Dependence in Hormone Receptor-Positive Breast Cancer. Sci. Transl. Med. 2015, 7, 283ra51.
  72. Bouchard, C.; Dittrich, O.; Kiermaier, A.; Dohmann, K.; Menkel, A.; Eilers, M.; Lüscher, B. Regulation of Cyclin D2 Gene Expression by the Myc/Max/Mad Network: Myc-Dependent TRRAP Recruitment and Histone Acetylation at the Cyclin D2 Promoter. Genes Dev. 2001, 15, 2042–2047.
  73. Ren, B.; Cam, H.; Takahashi, Y.; Volkert, T.; Terragni, J.; Young, R.A.; Dynlacht, B.D. E2F Integrates Cell Cycle Progression with DNA Repair, Replication, and G(2)/M Checkpoints. Genes Dev. 2002, 16, 245–256.
  74. Lasorella, A.; Stegmüller, J.; Guardavaccaro, D.; Liu, G.; Carro, M.S.; Rothschild, G.; de la Torre-Ubieta, L.; Pagano, M.; Bonni, A.; Iavarone, A. Degradation of Id2 by the Anaphase-Promoting Complex Couples Cell Cycle Exit and Axonal Growth. Nature 2006, 442, 471–474.
  75. Kuzyk, A.; Gartner, J.; Mai, S. Identification of Neuroblastoma Subgroups Based on Three-Dimensional Telomere Organization. Transl. Oncol. 2016, 9, 348–356.
  76. Valentijn, L.J.; Koster, J.; Zwijnenburg, D.A.; Hasselt, N.E.; van Sluis, P.; Volckmann, R.; van Noesel, M.M.; George, R.E.; Tytgat, G.A.M.; Molenaar, J.J.; et al. TERT Rearrangements are Frequent in Neuroblastoma and Identify Aggressive Tumors. Nat. Genet. 2015, 47, 1411–1414.
  77. Ham, J.; Costa, C.; Sano, R.; Lochmann, T.L.; Sennott, E.M.; Patel, N.U.; Dastur, A.; Gomez-Caraballo, M.; Krytska, K.; Hata, A.N.; et al. Exploitation of the Apoptosis-Primed State of MYCN-Amplified Neuroblastoma to Develop a Potent and Specific Targeted Therapy Combination. Cancer Cell 2016, 29, 159–172.
  78. Chen, L.; Iraci, N.; Gherardi, S.; Gamble, L.D.; Wood, K.M.; Perini, G.; Lunec, J.; Tweddle, D.A. P53 is a Direct Transcriptional Target of MYCN in Neuroblastoma. Cancer Res. 2010, 70, 1377–1388.
  79. Hou, H.; Sun, D.; Zhang, X. The Role of MDM2 Amplification and Overexpression in Therapeutic Resistance of Malignant Tumors. Cancer Cell Int. 2019, 19, 216.
  80. Kracikova, M.; Akiri, G.; George, A.; Sachidanandam, R.; Aaronson, S.A. A Threshold Mechanism Mediates P53 Cell Fate Decision between Growth Arrest and Apoptosis. Cell Death Differ. 2013, 20, 576–588.
  81. Muller, P.A.J.; Vousden, K.H. P53 Mutations in Cancer. Nat. Cell Biol. 2013, 15, 2–8.
  82. Barbieri, E.; Mehta, P.; Chen, Z.; Zhang, L.; Slack, A.; Berg, S.; Shohet, J.M. MDM2 Inhibition Sensitizes Neuroblastoma to Chemotherapy-Induced Apoptotic Cell Death. Mol. Cancer Ther. 2006, 5, 2358–2365.
  83. Yogev, O.; Barker, K.; Sikka, A.; Almeida, G.S.; Hallsworth, A.; Smith, L.M.; Jamin, Y.; Ruddle, R.; Koers, A.; Webber, H.T.; et al. P53 Loss in MYC-Driven Neuroblastoma Leads to Metabolic Adaptations Supporting Radioresistance. Cancer Res. 2016, 76, 3025–3035.
  84. Qi, D.-L.; Cobrinik, D. MDM2 but Not MDM4 Promotes Retinoblastoma Cell Proliferation through P53-Independent Regulation of MYCN Translation. Oncogene 2017, 36, 1760–1769.
  85. Cairns, R.A.; Harris, I.S.; Mak, T.W. Regulation of Cancer Cell Metabolism. Nat. Rev. Cancer 2011, 11, 85–95.
  86. Diers, A.R.; Broniowska, K.A.; Chang, C.-F.; Hogg, N. Pyruvate Fuels Mitochondrial Respiration and Proliferation of Breast Cancer Cells: Effect of Monocarboxylate Transporter Inhibition. Biochem. J. 2012, 444, 561–571.
  87. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science 2009, 324, 1029–1033.
  88. Carracedo, A.; Cantley, L.C.; Pandolfi, P.P. Cancer Metabolism: Fatty Acid Oxidation in the Limelight. Nat. Rev. Cancer 2013, 13, 227–232.
  89. Zirath, H.; Frenzel, A.; Oliynyk, G.; Segerström, L.; Westermark, U.K.; Larsson, K.; Munksgaard Persson, M.; Hultenby, K.; Lehtiö, J.; Einvik, C.; et al. MYC Inhibition Induces Metabolic Changes Leading to Accumulation of Lipid Droplets in Tumor Cells. Proc. Natl. Acad. Sci. USA 2013, 110, 10258–10263.
  90. DeBerardinis, R.J.; Mancuso, A.; Daikhin, E.; Nissim, I.; Yudkoff, M.; Wehrli, S.; Thompson, C.B. Beyond Aerobic Glycolysis: Transformed Cells Can Engage in Glutamine Metabolism That Exceeds the Requirement for Protein and Nucleotide Synthesis. Proc. Natl. Acad. Sci. USA 2007, 104, 19345–19350.
  91. Qing, G.; Li, B.; Vu, A.; Skuli, N.; Walton, Z.E.; Liu, X.; Mayes, P.A.; Wise, D.R.; Thompson, C.B.; Maris, J.M.; et al. ATF4 Regulates MYC-Mediated Neuroblastoma Cell Death upon Glutamine Deprivation. Cancer Cell 2012, 22, 631–644.
  92. Wahlström, T.; Henriksson, M.A. Impact of MYC in Regulation of Tumor Cell Metabolism. Biochim. Biophys. Acta 2015, 1849, 563–569.
  93. Tao, L.; Mohammad, M.A.; Milazzo, G.; Moreno-Smith, M.; Patel, T.D.; Zorman, B.; Badachhape, A.; Hernandez, B.E.; Wolf, A.B.; Zeng, Z.; et al. MYCN-Driven Fatty Acid Uptake is a Metabolic Vulnerability in Neuroblastoma. Nat. Commun. 2022, 13, 3728.
  94. Alborzinia, H.; Flórez, A.F.; Kreth, S.; Brückner, L.M.; Yildiz, U.; Gartlgruber, M.; Odoni, D.I.; Poschet, G.; Garbowicz, K.; Shao, C.; et al. MYCN Mediates Cysteine Addiction and Sensitizes Neuroblastoma to Ferroptosis. Nat. Cancer 2022, 3, 471–485.
  95. Montemurro, L.; Raieli, S.; Angelucci, S.; Bartolucci, D.; Amadesi, C.; Lampis, S.; Scardovi, A.L.; Venturelli, L.; Nieddu, G.; Cerisoli, L.; et al. A Novel MYCN-Specific Antigene Oligonucleotide Deregulates Mitochondria and Inhibits Tumor Growth in MYCN-Amplified Neuroblastoma. Cancer Res. 2019, 79, 6166–6177.
  96. Vaughan, L.; Clarke, P.A.; Barker, K.; Chanthery, Y.; Gustafson, C.W.; Tucker, E.; Renshaw, J.; Raynaud, F.; Li, X.; Burke, R.; et al. Inhibition of MTOR-Kinase Destabilizes MYCN and is a Potential Therapy for MYCN-Dependent Tumors. Oncotarget 2016, 7, 57525–57544.
  97. Schramm, A.; Köster, J.; Marschall, T.; Martin, M.; Schwermer, M.; Fielitz, K.; Büchel, G.; Barann, M.; Esser, D.; Rosenstiel, P.; et al. Next-Generation RNA Sequencing Reveals Differential Expression of MYCN Target Genes and Suggests the MTOR Pathway as a Promising Therapy Target in MYCN-Amplified Neuroblastoma. Int. J. Cancer 2013, 132, E106–E115.
  98. Yue, M.; Jiang, J.; Gao, P.; Liu, H.; Qing, G. Oncogenic MYC Activates a Feedforward Regulatory Loop Promoting Essential Amino Acid Metabolism and Tumorigenesis. Cell Rep. 2017, 21, 3819–3832.
  99. Raieli, S.; di Renzo, D.; Lampis, S.; Amadesi, C.; Montemurro, L.; Pession, A.; Hrelia, P.; Fischer, M.; Tonelli, R. MYCN Drives a Tumor Immunosuppressive Environment Which Impacts Survival in Neuroblastoma. Front. Oncol. 2021, 11, 625207.
  100. Nallasamy, P.; Chava, S.; Verma, S.S.; Mishra, S.; Gorantla, S.; Coulter, D.W.; Byrareddy, S.N.; Batra, S.K.; Gupta, S.C.; Challagundla, K.B. PD-L1, Inflammation, Non-Coding RNAs, and Neuroblastoma: Immuno-Oncology Perspective. Semin. Cancer Biol. 2018, 52, 53–65.
  101. Melaiu, O.; Mina, M.; Chierici, M.; Boldrini, R.; Jurman, G.; Romania, P.; D’Alicandro, V.; Benedetti, M.C.; Castellano, A.; Liu, T.; et al. PD-L1 is a Therapeutic Target of the Bromodomain Inhibitor JQ1 and, Combined with HLA Class I, a Promising Prognostic Biomarker in Neuroblastoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 4462–4472.
  102. Noguera, R.; Fredlund, E.; Piqueras, M.; Pietras, A.; Beckman, S.; Navarro, S.; Påhlman, S. HIF-1α and HIF-2α are Differentially Regulated In Vivo in Neuroblastoma: High HIF-1α Correlates Negatively to Advanced Clinical Stage and Tumor Vascularization. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2009, 15, 7130–7136.
  103. Burr, M.L.; Sparbier, C.E.; Chan, K.L.; Chan, Y.-C.; Kersbergen, A.; Lam, E.Y.N.; Azidis-Yates, E.; Vassiliadis, D.; Bell, C.C.; Gilan, O.; et al. An Evolutionarily Conserved Function of Polycomb Silences the MHC Class I Antigen Presentation Pathway and Enables Immune Evasion in Cancer. Cancer Cell 2019, 36, 385–401.e8.
  104. Merchant, M.S.; Wright, M.; Baird, K.; Wexler, L.H.; Rodriguez-Galindo, C.; Bernstein, D.; Delbrook, C.; Lodish, M.; Bishop, R.; Wolchok, J.D.; et al. Phase I Clinical Trial of Ipilimumab in Pediatric Patients with Advanced Solid Tumors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 1364–1370.
  105. Majzner, R.G.; Simon, J.S.; Grosso, J.F.; Martinez, D.; Pawel, B.R.; Santi, M.; Merchant, M.S.; Geoerger, B.; Hezam, I.; Marty, V.; et al. Assessment of Programmed Death-Ligand 1 Expression and Tumor-Associated Immune Cells in Pediatric Cancer Tissues. Cancer 2017, 123, 3807–3815.
  106. Hahn, M.; Glass, T.; Koke, J. Extracellular Matrix Effects on a Neuroblastoma Cell Line. Cytobios 2000, 102, 7–19.
  107. Meyer, A.; van Golen, C.M.; Kim, B.; van Golen, K.L.; Feldman, E.L. Integrin Expression Regulates Neuroblastoma Attachment and Migration. Neoplasia 2004, 6, 332–342.
  108. Young, S.A.; McCabe, K.E.; Bartakova, A.; Delaney, J.; Pizzo, D.P.; Newbury, R.O.; Varner, J.A.; Schlaepfer, D.D.; Stupack, D.G. Integrin A4 Enhances Metastasis and May Be Associated with Poor Prognosis in MYCN-Low Neuroblastoma. PLoS ONE 2015, 10, e0120815.
  109. Erdreich-Epstein, A.; Shimada, H.; Groshen, S.; Liu, M.; Metelitsa, L.S.; Kim, K.S.; Stins, M.F.; Seeger, R.C.; Durden, D.L. Integrins Alpha(v)Beta3 and Alpha(v)Beta5 are Expressed by Endothelium of High-Risk Neuroblastoma and Their Inhibition is Associated with Increased Endogenous Ceramide. Cancer Res. 2000, 60, 712–721.
  110. Pickup, M.W.; Mouw, J.K.; Weaver, V.M. The Extracellular Matrix Modulates the Hallmarks of Cancer. EMBO Rep. 2014, 15, 1243–1253.
  111. Willumsen, N.; Thomsen, L.B.; Bager, C.L.; Jensen, C.; Karsdal, M.A. Quantification of Altered Tissue Turnover in a Liquid Biopsy: A Proposed Precision Medicine Tool to Assess Chronic Inflammation and Desmoplasia Associated with a pro-Cancerous Niche and Response to Immuno-Therapeutic Anti-Tumor Modalities. Cancer Immunol. Immunother. 2018, 67, 1–12.
  112. Ng, M.R.; Brugge, J.S. A Stiff Blow from the Stroma: Collagen Crosslinking Drives Tumor Progression. Cancer Cell 2009, 16, 455–457.
  113. Diop-Frimpong, B.; Chauhan, V.P.; Krane, S.; Boucher, Y.; Jain, R.K. Losartan Inhibits Collagen I Synthesis and Improves the Distribution and Efficacy of Nanotherapeutics in Tumors. Proc. Natl. Acad. Sci. USA 2011, 108, 2909–2914.
  114. Sugiura, Y.; Shimada, H.; Seeger, R.C.; Laug, W.E.; DeClerck, Y.A. Matrix Metalloproteinases-2 and -9 are Expressed in Human Neuroblastoma: Contribution of Stromal Cells to Their Production and Correlation with Metastasis. Cancer Res. 1998, 58, 2209–2216.
  115. Sans-Fons, M.G.; Sole, S.; Sanfeliu, C.; Planas, A.M. Matrix Metalloproteinase-9 and Cell Division in Neuroblastoma Cells and Bone Marrow Macrophages. Am. J. Pathol. 2010, 177, 2870–2885.
  116. Ara, T.; Fukuzawa, M.; Kusafuka, T.; Komoto, Y.; Oue, T.; Inoue, M.; Okada, A. Immunohistochemical Expression of MMP-2, MMP-9, and TIMP-2 in Neuroblastoma: Association with Tumor Progression and Clinical Outcome. J. Pediatr. Surg. 1998, 33, 1272–1278.
  117. Bergers, G.; Benjamin, L.E. Tumorigenesis and the Angiogenic Switch. Nat. Rev. Cancer 2003, 3, 401–410.
  118. Ramani, P.; Nash, R.; Radevsky, L.; Patel, A.; Luckett, M.; Rogers, C. VEGF-C, VEGF-D and VEGFR-3 Expression in Peripheral Neuroblastic Tumours. Histopathology 2012, 61, 1006–1016.
  119. Chlenski, A.; Liu, S.; Cohn, S.L. The Regulation of Angiogenesis in Neuroblastoma. Cancer Lett. 2003, 197, 47–52.
  120. Meitar, D.; Crawford, S.E.; Rademaker, A.W.; Cohn, S.L. Tumor Angiogenesis Correlates with Metastatic Disease, N-Myc Amplification, and Poor Outcome in Human Neuroblastoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 1996, 14, 405–414.
  121. Ribatti, D.; Vacca, A.; Nico, B.; de Falco, G.; Giuseppe Montaldo, P.; Ponzoni, M. Angiogenesis and Anti-Angiogenesis in Neuroblastoma. Eur. J. Cancer 2002, 38, 750–757.
  122. Rössler, J.; Taylor, M.; Geoerger, B.; Farace, F.; Lagodny, J.; Peschka-Süss, R.; Niemeyer, C.M.; Vassal, G. Angiogenesis as a Target in Neuroblastoma. Eur. J. Cancer 2008, 44, 1645–1656.
  123. Chanthery, Y.H.; Gustafson, W.C.; Itsara, M.; Persson, A.; Hackett, C.S.; Grimmer, M.; Charron, E.; Yakovenko, S.; Kim, G.; Matthay, K.K.; et al. Paracrine Signaling through MYCN Enhances Tumor-Vascular Interactions in Neuroblastoma. Sci. Transl. Med. 2012, 4, 115ra3.
  124. Kang, J.; Rychahou, P.G.; Ishola, T.A.; Mourot, J.M.; Evers, B.M.; Chung, D.H. N-Myc is a Novel Regulator of PI3K-Mediated VEGF Expression in Neuroblastoma. Oncogene 2008, 27, 3999–4007.
  125. Singh, A.R.; Joshi, S.; Burgoyne, A.M.; Sicklick, J.K.; Ikeda, S.; Kono, Y.; Garlich, J.R.; Morales, G.A.; Durden, D.L. Single Agent and Synergistic Activity of the “First-in-Class” Dual PI3K/BRD4 Inhibitor SF1126 with Sorafenib in Hepatocellular Carcinoma. Mol. Cancer Ther. 2016, 15, 2553–2562.
  126. Joshi, S.; Singh, A.R.; Durden, D.L. Pan-PI-3 Kinase Inhibitor SF1126 Shows Antitumor and Antiangiogenic Activity in Renal Cell Carcinoma. Cancer Chemother. Pharmacol. 2015, 75, 595–608.
  127. Joshi, S.; Singh, A.R.; Zulcic, M.; Durden, D.L. A Macrophage-Dominant PI3K Isoform Controls Hypoxia-Induced HIF1α and HIF2α Stability and Tumor Growth, Angiogenesis, and Metastasis. Mol. Cancer Res. 2014, 12, 1520–1531.
  128. Singh, A.R.; Joshi, S.; George, E.; Durden, D.L. Anti-Tumor Effect of a Novel PI3-Kinase Inhibitor, SF1126, in (12) V-Ha-Ras Transgenic Mouse Glioma Model. Cancer Cell Int. 2014, 14, 105.
  129. Xing, F.; Saidou, J.; Watabe, K. Cancer Associated Fibroblasts (CAFs) in Tumor Microenvironment. Front. Biosci. Landmark Ed. 2010, 15, 166–179.
  130. Yazhou, C.; Wenlv, S.; Weidong, Z.; Licun, W. Clinicopathological Significance of Stromal Myofibroblasts in Invasive Ductal Carcinoma of the Breast. Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 2004, 25, 290–295.
  131. Tuxhorn, J.A.; Ayala, G.E.; Smith, M.J.; Smith, V.C.; Dang, T.D.; Rowley, D.R. Reactive Stroma in Human Prostate Cancer: Induction of Myofibroblast Phenotype and Extracellular Matrix Remodeling. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2002, 8, 2912–2923.
  132. Orimo, A.; Gupta, P.B.; Sgroi, D.C.; Arenzana-Seisdedos, F.; Delaunay, T.; Naeem, R.; Carey, V.J.; Richardson, A.L.; Weinberg, R.A. Stromal Fibroblasts Present in Invasive Human Breast Carcinomas Promote Tumor Growth and Angiogenesis through Elevated SDF-1/CXCL12 Secretion. Cell 2005, 121, 335–348.
  133. Silzle, T.; Kreutz, M.; Dobler, M.A.; Brockhoff, G.; Knuechel, R.; Kunz-Schughart, L.A. Tumor-Associated Fibroblasts Recruit Blood Monocytes into Tumor Tissue. Eur. J. Immunol. 2003, 33, 1311–1320.
  134. Yingling, J.M.; Blanchard, K.L.; Sawyer, J.S. Development of TGF-Beta Signalling Inhibitors for Cancer Therapy. Nat. Rev. Drug Discov. 2004, 3, 1011–1022.
  135. Hashimoto, O.; Yoshida, M.; Koma, Y.-I.; Yanai, T.; Hasegawa, D.; Kosaka, Y.; Nishimura, N.; Yokozaki, H. Collaboration of Cancer-Associated Fibroblasts and Tumour-Associated Macrophages for Neuroblastoma Development. J. Pathol. 2016, 240, 211–223.
  136. Kakarla, S.; Song, X.-T.; Gottschalk, S. Cancer-Associated Fibroblasts as Targets for Immunotherapy. Immunotherapy 2012, 4, 1129–1138.
  137. Fakhrai, H.; Dorigo, O.; Shawler, D.L.; Lin, H.; Mercola, D.; Black, K.L.; Royston, I.; Sobol, R.E. Eradication of Established Intracranial Rat Gliomas by Transforming Growth Factor Beta Antisense Gene Therapy. Proc. Natl. Acad. Sci. USA 1996, 93, 2909–2914.
  138. Braoudaki, M.; Hatziagapiou, K.; Zaravinos, A.; Lambrou, G.I. MYCN in Neuroblastoma: “Old Wine into New Wineskins”. Diseases 2021, 9, 78.
  139. Bartel, D.P. MicroRNAs: Target Recognition and Regulatory Functions. Cell 2009, 136, 215–233.
  140. Buechner, J.; Einvik, C. N-Myc and Noncoding RNAs in Neuroblastoma. Mol. Cancer Res. 2012, 10, 1243–1253.
  141. Beckers, A.; van Peer, G.; Carter, D.R.; Mets, E.; Althoff, K.; Cheung, B.B.; Schulte, J.H.; Mestdagh, P.; Vandesompele, J.; Marshall, G.M.; et al. MYCN-Targeting MiRNAs are Predominantly Downregulated during MYCN-driven Neuroblastoma Tumor Formation. Oncotarget 2015, 6, 5204–5216.
  142. Molenaar, J.J.; Domingo-Fernández, R.; Ebus, M.E.; Lindner, S.; Koster, J.; Drabek, K.; Mestdagh, P.; van Sluis, P.; Valentijn, L.J.; van Nes, J.; et al. LIN28B Induces Neuroblastoma and Enhances MYCN Levels via Let-7 Suppression. Nat. Genet. 2012, 44, 1199–1206.
  143. Powers, J.T.; Tsanov, K.M.; Pearson, D.S.; Roels, F.; Spina, C.S.; Ebright, R.; Seligson, M.; de Soysa, Y.; Cahan, P.; Theißen, J.; et al. Multiple Mechanisms Disrupt the Let-7 MicroRNA Family in Neuroblastoma. Nature 2016, 535, 246–251.
  144. Misiak, D.; Hagemann, S.; Bell, J.L.; Busch, B.; Lederer, M.; Bley, N.; Schulte, J.H.; Hüttelmaier, S. The MicroRNA Landscape of MYCN-Amplified Neuroblastoma. Front. Oncol. 2021, 11, 647737.
  145. Mogilyansky, E.; Rigoutsos, I. The MiR-17/92 Cluster: A Comprehensive Update on Its Genomics, Genetics, Functions and Increasingly Important and Numerous Roles in Health and Disease. Cell Death Differ. 2013, 20, 1603–1614.
  146. Mestdagh, P.; Boström, A.-K.; Impens, F.; Fredlund, E.; van Peer, G.; de Antonellis, P.; von Stedingk, K.; Ghesquière, B.; Schulte, S.; Dews, M.; et al. The MiR-17-92 MicroRNA Cluster Regulates Multiple Components of the TGF-β Pathway in Neuroblastoma. Mol. Cell 2010, 40, 762–773.
  147. Armstrong, B.C.; Krystal, G.W. Isolation and Characterization of Complementary DNA for N-Cym, a Gene Encoded by the DNA Strand Opposite to N-Myc. Cell Growth Differ. Mol. Biol. J. Am. Assoc. Cancer Res. 1992, 3, 385–390.
  148. Vadie, N.; Saayman, S.; Lenox, A.; Ackley, A.; Clemson, M.; Burdach, J.; Hart, J.; Vogt, P.K.; Morris, K.V. MYCNOS Functions as an Antisense RNA Regulating MYCN. RNA Biol. 2015, 12, 893–899.
  149. Suenaga, Y.; Islam, S.M.R.; Alagu, J.; Kaneko, Y.; Kato, M.; Tanaka, Y.; Kawana, H.; Hossain, S.; Matsumoto, D.; Yamamoto, M.; et al. NCYM, a Cis-Antisense Gene of MYCN, Encodes a de Novo Evolved Protein That Inhibits GSK3β Resulting in the Stabilization of MYCN in Human Neuroblastomas. PLoS Genet. 2014, 10, e1003996.
  150. O’Brien, E.M.; Selfe, J.L.; Martins, A.S.; Walters, Z.S.; Shipley, J.M. The Long Non-Coding RNA MYCNOS-01 Regulates MYCN Protein Levels and Affects Growth of MYCN-Amplified Rhabdomyosarcoma and Neuroblastoma Cells. BMC Cancer 2018, 18, 217.
  151. Liu, P.Y.; Atmadibrata, B.; Mondal, S.; Tee, A.E.; Liu, T. NCYM is Upregulated by LncUSMycN and Modulates N-Myc Expression. Int. J. Oncol. 2016, 49, 2464–2470.
  152. Decock, A.; Ongenaert, M.; Vandesompele, J.; Speleman, F. Neuroblastoma Epigenetics: From Candidate Gene Approaches to Genome-Wide Screenings. Epigenetics 2011, 6, 962–970.
  153. Westerlund, I.; Shi, Y.; Toskas, K.; Fell, S.M.; Li, S.; Surova, O.; Södersten, E.; Kogner, P.; Nyman, U.; Schlisio, S.; et al. Combined Epigenetic and Differentiation-Based Treatment Inhibits Neuroblastoma Tumor Growth and Links HIF2α to Tumor Suppression. Proc. Natl. Acad. Sci. USA 2017, 114, E6137–E6146.
  154. Parodi, F.; Carosio, R.; Ragusa, M.; di Pietro, C.; Maugeri, M.; Barbagallo, D.; Sallustio, F.; Allemanni, G.; Pistillo, M.P.; Casciano, I.; et al. Epigenetic Dysregulation in Neuroblastoma: A Tale of MiRNAs and DNA Methylation. Biochim. Biophys. Acta 2016, 1859, 1502–1514.
  155. Louis, C.U.; Shohet, J.M. Neuroblastoma: Molecular Pathogenesis and Therapy. Annu. Rev. Med. 2015, 66, 49–63.
  156. Pession, A.; Tonelli, R. The MYCN Oncogene as a Specific and Selective Drug Target for Peripheral and Central Nervous System Tumors. Curr. Cancer Drug Targets 2005, 5, 273–283.
  157. Fletcher, J.I.; Ziegler, D.S.; Trahair, T.N.; Marshall, G.M.; Haber, M.; Norris, M.D. Too Many Targets, Not Enough Patients: Rethinking Neuroblastoma Clinical Trials. Nat. Rev. Cancer 2018, 18, 389–400.
  158. Andresen, C.; Helander, S.; Lemak, A.; Farès, C.; Csizmok, V.; Carlsson, J.; Penn, L.Z.; Forman-Kay, J.D.; Arrowsmith, C.H.; Lundström, P.; et al. Transient Structure and Dynamics in the Disordered C-Myc Transactivation Domain Affect Bin1 Binding. Nucleic Acids Res. 2012, 40, 6353–6366.
  159. Bayliss, R.; Burgess, S.G.; Leen, E.; Richards, M.W. A Moving Target: Structure and Disorder in Pursuit of Myc Inhibitors. Biochem. Soc. Trans. 2017, 45, 709–717.
  160. Kohl, N.E.; Legouy, E.; DePinho, R.A.; Nisen, P.D.; Smith, R.K.; Gee, C.E.; Alt, F.W. Human N-Myc is Closely Related in Organization and Nucleotide Sequence to c-Myc. Nature 1986, 319, 73–77.
  161. Esposito, M.R.; Aveic, S.; Seydel, A.; Tonini, G.P. Neuroblastoma Treatment in the Post-Genomic Era. J. Biomed. Sci. 2017, 24, 14.
More
This entry is offline, you can click here to edit this entry!