Open access peer-reviewed chapter

Molecular Target Therapy against Neuroblastoma

Written By

Hidemi Toyoda, Dong-Qing Xu, Lei Qi and Masahiro Hirayama

Submitted: 06 June 2018 Reviewed: 27 September 2018 Published: 12 November 2018

DOI: 10.5772/intechopen.81706

From the Edited Volume

Biophysical Chemistry - Advance Applications

Edited by Mohammed A. A. Khalid

Chapter metrics overview

881 Chapter Downloads

View Full Metrics

Abstract

Neuroblastoma, originated from neural crest cells, is the most common extracranial solid tumor in childhood. Treatment is of limited utility for high-risk neuroblastoma and prognosis is poor. The high incidence of resistance of advanced-stage neuroblastoma to conventional therapies has prompt investigators to search for novel therapeutic approaches. Activation of IGF-R/PI3K/Akt/mTOR signaling pathway correlates with oncogenesis, poor prognosis, and chemotherapy resistance in neuroblastoma. Therefore, we investigated the effect of IGF-R/PI3K/Akt/mTOR signaling inhibitors in neuroblastoma. Significantly, IGF-R/PI3K/Akt/mTOR signaling inhibitors effectively inhibited cell growth and induced cell cycle arrest, autophagy, and apoptosis in neuroblastoma cells. Moreover, IGF-R/PI3K/Akt/mTOR signaling inhibitors significantly reduced tumor growth in mice xenograft model without apparent toxicity. Therefore, these results highlight the potential of IGF-R/PI3K/Akt/mTOR signaling pathway as a promising target for neuroblastoma treatment. Therefore, IGF-1R/PI3K/Akt/mTOR signaling inhibitors should be further investigated for treatment in clinical trials for high-risk neuroblastoma.

Keywords

  • neuroblastoma
  • insulin-like growth factor (IGF)
  • phosphatidylinositol 3-kinase (PI3K)
  • protein kinase-B (Akt)
  • mammalian target of rapamycin (mTOR)

1. Introduction

Neuroblastoma (NB) is one of the most common extracranial solid tumors of early childhood [1, 2]. Prognosis of patients with NB depends on tumor stage, patient’s age, and biologic feature of the tumor cells [3]. In patients under 1 year of age, NB is curable and sometimes spontaneously regress [4]. However, in older children with advanced stage, often the tumor is very aggressive, and patients have poor prognosis despite treatment with high-dose chemotherapy combined with autologous hematopoietic stem cell transplantation. Although immunotherapeutic therapy such as anti-GD2 monoclonal antibody has improved outcomes of advanced stage of NB, a number of patients still relapse and eventually die of disease [5]. The high incidence of resistance of high-risk NB to conventional therapies has prompted us to search for novel therapeutic approaches.

It was reported that phosphorylated protein kinase-B (Akt) correlates with poor patients’ prognosis in NB [6], and the phosphatidylinositol-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway has subsequently been linked to augmented cell survival [7] and increased resistance to chemotherapy in NB [8]. Therefore, targeting the PI3K/Akt/mTOR signaling pathway by appropriate inhibitors appears to be a promising strategy for overcoming therapy resistance [9].

Here, we demonstrate that NB cell lines are heterogeneous in their insulin growth factor-1 (IGF-1) receptor-mediated signaling [10]. The pattern of IGF-1 receptor/PI3K/Akt/mTOR pathway-mediated proliferation is an important determinant of the response to IGF-1 receptor antagonistic therapy in human NB [10]. Furthermore, our results highlight the potential of IGF-1 receptor/PI3K/Akt/mTOR signaling pathway as a promising target for NB treatment [10, 11, 12, 13].

Advertisement

2. Heterogeneity of IGF-1 receptor/Akt pathway-mediated proliferation in NB

IGF-1, IGF-2, and insulin belong to a family of mitogenic growth factors and are involved in normal growth and differentiation of most tissues. The biological actions of both IGFs and insulin can be mediated by the IGF-1 receptor which is involved in mitogenic, anti-apoptotic, and oncogenic transforming responses [14, 15]. The IGF-1 receptor has two extracellular α-subunits and two intracellular β-subunits that form a heterotetrameric complex. Ligand interaction with α-subunits triggers the autophosphorylation of tyrosine kinase domains within the β-subunit [16, 17, 18]. The tyrosine kinase domains are connected to several intracellular pathways such as PI3K/Akt [19, 20]. Dysregulation of the IGF-1 receptor pathway is involved in promoting oncogenic transformation, cell proliferation, metastasis, angiogenesis, and resistance in numerous malignant diseases, such as multiple myeloma [21], carcinomas [22], and NB [23]. Since high cellular heterogeneity is a hallmark of NB, which may account for the wide range of clinical presentations and nonuniform response to treatment, we hypothesized that NB cells are heterogeneous in their IGF-1 receptor signaling-mediated cell proliferation. Thirty-one NB cell lines were cultured in three different conditions, insulin-containing serum-free medium (SFM), RPMI1640 without FBS (serum starvation medium), and RPMI1640 with 10% FBS (serum-containing medium). Based on the response patterns, 31 cell lines were subdivided into three groups [10]. Group 1, which consisted of three NB cell lines, could proliferate for more than 3 days in SFM, RPMI1640 without FBS, and RPMI1640 with 10% FBS [10]. Group 2, which consisted of 10 cell lines, could proliferate in SFM and RPMI1640 with 10% FBS but not in RPMI1640 [10]. Group 3, which consisted of 18 NB cell lines, proliferated only in RPMI1640 with 10% FBS [10]. NB cell proliferation in RPMI1640 in the presence of exogenous IGF (IGF-1, IGF-2) and insulin was examined. These IGF and insulin accelerated cell proliferation in group 1 and group 2 NB cell lines but not in group 3 NB cell lines [10]. Group 1 NB cell lines were able to proliferate in RPMI1640 in the absence of exogenous IGF and insulin [10].

Advertisement

3. Impairment of Akt activation and cell proliferation in NB by IGF-1 receptor inhibitor, picropodophyllin (PPP)

The IGF-1 receptor inhibitors, such as IGF-1 receptor-neutralizing antibodies and IGF-1 receptor antisense/siRNA, have been shown to block cancer cell proliferation [24]. Selective IGF-1 receptor inhibitor, picropodophyllin (PPP), lacks inhibitory activity on tyrosine phosphorylation of insulin receptor tyrosine kinase (RTK) [25]. Inhibition of the IGF-1 RTK with PPP is noncompetitive with respect to ATP, suggesting interference with the IGF-1 receptor at substrate level [26]. It is reported that PPP specifically blocks phosphorylation of the Tyr1136 residue in the activation loop of IGF-1 receptor kinase [27]. Inhibition of IGF-1 receptor with PPP has been demonstrated in a lot of cancers such as multiple myeloma [26], melanoma [28], breast cancer [29], and glioblastoma cells [30]. PPP inhibited Akt activation and suppressed cell proliferation in group 1 and group 2 NB cell lines but less in group 3 NB cell lines [10]. Elevation of ERK phosphorylation was only observed in group 1 NB cell lines [10]. U0126 is a MEK inhibitor effectively suppressed ERK phosphorylation, U0126 (2.5 μM) did not suppress cell proliferation induced by IGF-1, IGF-2, or insulin in group 1 and group 2 NB cell lines [10]. In groups 1 and 2 NB cell lines, IGF-1 receptor/PI3K/Akt pathway is critical for cell proliferation. Although IGF-1, IGF-2, and insulin activated Akt in group 3 NB cell line, cell proliferation was not increased in RPMI1640. This suggests that the activation of IGF-1R/Akt pathway is insufficient for cell proliferation [10]. Since IGF-1 and IGF-2 regulate apoptosis [31] and cell cycle progression [32], activation of caspase 3 and PARP was examined in NB cell lines. In group 1 NB cell lines, caspase 3 and PARP were not cleaved in RPMI1640 [10]. In group 2 NB cell lines, caspase 3 and PARP were cleaved in RPMI1640, and the cleavage of caspase 3 was suppressed by addition of IGF-1, IGF-2, and insulin [10]. In group 3 NB cell lines, cleavages of caspase 3 and PARP were observed in RPMI1640 [10]. However, they were not suppressed by IGF-1, IGF-2, and insulin, even though Akt activation was upregulated by IGF-1, IGF-2, and insulin [10]. Since PPP induced G2/M arrest and apoptosis by inhibiting IGF-1 receptor [26, 33], cell cycle phase distribution in NB cells was examined in the presence of PPP. PPP treatment (2.5 μM) for 12 hours increased the G2/M fraction and shifted cell cycle profile from G0-G1 dominant to G2/M dominant in group 1 and group 2 NB cell lines [10]. However, group 3 NB cell lines did not show G2/M arrest and the G0/G1 fraction was not affected [10]. Furthermore, cyclin B1, a marker protein of G2/M phase of cell cycle, was upregulated, and cyclin D1, a marker protein of G0/G1, synchronously declined in group 1 and group 2 NB cell lines [10]. However, accumulation of cyclin B1 and decline of cyclin D1 were not observed in group 3 NB cell lines [10]. This may be explained by the insensitiveness of group 3 cell lines to PPP-induced G2/M arrest.

Advertisement

4. Impairment of Akt activation and cell proliferation in NB by Akt inhibitor, MK2006

MK2206 selectively inhibits AKT and has potency against Akt1 and Akt2 isoforms than Akt3. In pediatric solid tumors, MK2206 is effective in vitro and in vivo [34, 35]. In clinical trials, stable disease was observed in different kinds of cancers [36, 37]. Our results also suggested that MK2206 (2.5 μM) completely inhibited Akt phosphorylation and cell proliferation in NB cell lines [10]. Furthermore, MK2206 also impaired the cell proliferation induced by exogenous IGF and insulin in NB cell lines [10].

Advertisement

5. Inhibition of NB cell proliferation by mTOR inhibitor

The PI3K/Akt/mTOR signaling cascade is one of the most important intracellular pathways, which is frequently activated in diverse cancers [38, 39]. The PI3K/Akt/mTOR pathway can be activated by transmembrane tyrosine kinase growth factor receptors, such as IGF-1 receptor, fibroblast growth factor receptors, ErbB family receptors, and others [40, 41]. Both mTOR S2448 and mTOR S2481 were extensively phosphorylated in NB cell lines [13]. Cell proliferation of NB cell lines was inhibited by AZD8055, a potent dual mTORC1-mTORC2 inhibitor [13]. According to the IC50, the NB cell lines were divided into two groups, sensitive to AZD8055 group (IC50 < 0.5 μM) and insensitive to AZD8055 group (IC50 > 0.5 μM) [13]. We also found that insensitive group showed lower mTOR (p < 0.001) expression and lower activity of mTOR complex 1 (p = 0.013) and mTOR complex 2 (p = 0.023) [13]. Cell cycle distribution analysis of NB cell lines was performed by flow cytometry. Cell cycle was affected with an increase in G0/G1 phase in dose-dependent manner of AZD8055 [13]. Western blotting analysis revealed that Cyclin D1 and Cyclin D3 were downregulated in AZD8055-treated NB cells [13]. AZD8055 inhibited both mTOR S2448 and mTOR S2481 phosphorylation significantly in a concentration-dependent manner [13]. Phosphorylation of downstream targets of mTOR complex 1, P70S6K T389 and 4E-BP1 S65, was also inhibited by AZD8055 treatment [13]. AZD8055 inhibited mTOR complex 2 substrates, Akt S473 and Akt T308 [13]. Although phosphorylation of Akt S473 was persistently inhibited in response to AZD8055 treatment, phosphorylation of Akt at the T308 site was inhibited for only 3–6 hours. These results indicate that AZD8055 inhibits mTOR activity and its downstream proteins in vitro in NB cells. Interestingly, NB cell lines were induced autophagy by AZD8055 treatment via downregulation of Akt/mTOR signaling pathway [13]. Autophagy inhibitor, 3-methyladenine, treatment resulted in a significant decrease of the AZD8055-induced apoptosis [13]. These results suggest that AZD8055 inhibited cell growth and induced cell cycle arrest, autophagy, and apoptosis in NB cells. Moreover, NB tumor growth in athymic nude mice was significantly inhibited by AZD8055 without toxicity [13]. Taken together, our results highlight that mTOR is a promising target for NB treatment. AZD8055 might be investigated for treatment of patients with advanced and refractory NB.

Advertisement

6. AZD8055 treatment in combination with MEK/ERK inhibitor

The problem that will plague single-target drugs is the cancer’s ability to activate alternative survival pathways leading to drug resistance and toxicity even in the multimodality setting. In addition, activation of multiple signaling pathways effectively causes cancer cell proliferation and survival. For example, RAS and PI3KCA are concurrently activated in melanoma, lung, and colorectal cancers [42, 43, 44]. These results suggested that combination therapies targeted on multiple signaling pathways could be more effective than targeting either pathway alone. Although AZD8055 is a promising drug in NB treatment [13], AZD8055-resistant NB sublines were acquired by prolonged stepwise exposure. After incubation with AZD8055 for 4–12 weeks, two acquired AZD8055-resistant sublines proliferated stably in RPMI1640 plus 10% FBS medium in the presence of AZD8055 (3 μM) [12]. The AZD8055-resistant sublines exhibited marked resistance to AZD8055 compared to the parent cells, and IC50 of the resistant sublines was 60–100 times higher than the parent NB cell lines [12]. By cell cycle analysis, accumulation of S phase was observed, and cyclin D3 and CDK4 were upregulated in AZD8055-resistant sublines [12]. Although AZD8055 treatment inhibited MEK/ERK activation in parent cells, MEK/ERK phosphorylation was continued despite AZD8055 treatment in resistant cells [12]. The combination therapy of AZD8055 and MEK/ERK inhibitor U0126 significantly inhibited cell proliferation compared to U0126 monotherapy [12], suggesting that combination therapy can overcome AZD8055 resistance. Furthermore, in athymic mice model, AZD8055 and U0126 co-treatment was more efficient to suppress resistant NB tumor growth compared to U0126 monotherapy [12].

Advertisement

7. MK2206 treatment in combination with PDK1 inhibitor

MK2206 treatment induced a dose-dependent inhibition of cell proliferation, with IC50 ranging from 1.22 to 4.35 μM in NB cell lines [11]. MK2206-resistant cells were induced by stepwise escalation of MK-2206 exposure (4–12 weeks) [11]. These cells proliferated in RPMI1640 plus 10% FBS medium in the presence of MK2206 (5 μM), while cell death was induced in parent cells [11]. IC50 of the resistant cell lines was 6–7 times higher than the parent NB cell lines [11]. Small-molecule GSK2334470 selectively inhibits 3-phosphoinositide-dependent protein kinase 1 (PDK1) with low concentration but does not suppress the activity of other protein kinases at higher concentrations [45]. Although GSK2334470 attenuated cell proliferation in both parent cells and MK2206-resistant sublines, IC50 of GSK2334470 in resistant sublines was lower than that of parent cell lines [11]. GSK2334470 induced G0-G1 accumulation of cell cycle phase distribution in parent cell lines [11]. In MK2206-resistant sublines, G0-G1 accumulation induced by GSK2334470 was higher than parent cell lines [11].

Advertisement

8. Effect of NB MYCN status on susceptibility to AZD8055

Amplification of the MYCN oncogene is the most powerful single predictor of adverse outcome of NB [46]. MYCN amplification is observed in about 20% of all NB patients and is usually associated with fatal outcome of the disease [47]. Schramm et al. demonstrated transcriptomal upregulation of mTOR-related genes by MYCN [48]. MYCN-driven NB in mice displayed activation of the mTOR pathway on the protein level, and activation of MYCN in NB cells resulted in high sensitivity toward mTOR inhibition [48]. Therefore, it is examined whether MYCN status of NB cell lines affects susceptibility to AZD8055. Using fluorescence in situ hybridization (FISH), we observed MYCN amplification in all investigated nuclei of NB19, IMR32, TGW, OZAWA, LAN-1, SMS-KAN, and SCMC-N4 cell lines (Figure 1A). As shown in Figure 1A, MYCN amplification was not observed in INDEN (loss/imbalance), SK-N-SH (gain), KP-N-SI (loss/imbalance), KP-N-SIFA (gain), and SJ-N-KP (loss/imbalance). As shown in Figure 1B, the relationship between MYCN status and susceptibility to AZD8055 was not found.

Figure 1.

Effect of MYCN status on susceptibility to AZD8055. (A) MYCN gene in tested 12 NB cell lines. MYCN status was defined as previously [53]. No alteration: cells with two MYCN signals and two CEP2 signals; amplification: the number of MYCN signals is at least 10 copies greater than the control probe signals; loss/imbalance: presence of at least two MYCN signals and increased CEP2 signals; gain: the number of MYCN signals is 1–9 copies more than CEP2 signals. (B) Fifteen NB cell lines were treated with AZD8055 at different concentrations in RPMI1640 + 10% FBS. Cell growth was evaluated as cell numbers at 72 hours. Data are expressed as the mean ± SD. IC50 (half maximal inhibitory concentration) of the 15 NB cell lines was calculated depending on the MTT results.

Advertisement

9. ALK as a therapeutic target in NB

Constitutive activation of the ALK receptor tyrosine kinase by mutation or translocation appears to contribute to the malignant phenotype of several cancers, including NB, making it a potentially therapeutic target [49]. Both wild type ALK and the F1174L-mutated ALK upregulate MYCN expression [50], and these two genes lead to constitutive phosphorylation of ALK and of downstream signaling molecules, PI3K/AKT/mTOR, that are critical for cell proliferation and survival [51]. ALKF1174L cosegregates with MYCN amplification in patients, and this combination is associated with a particularly poor prognosis, as shown by the fatal outcome of 9 of 10 children with ALKF1174L/MYCN-amplified tumors [52]. Targeting ALK with PI3K/Akt/mTOR inhibitors may inhibit cell growth in tumor lines with concomitant MYCN amplification.

Advertisement

10. Conclusions

NB cell lines can be categorized into three groups by the patterns of IGF-1R/Akt pathway response. PPP, MK2206, and AZD8055 showed significant antitumor effect in NB cells. Our current results highlight the potential of IGF-1R/PI3K/Akt/mTOR pathway as a promising target for NB treatment. PPP, MK2206, and AZD8055 should be further investigated for NB treatment in clinical trials.

Conflict of interest

The authors declare no potential conflicts of interest.

References

  1. 1. Louis CU, Shohet JM. Neuroblastoma: Molecular pathogenesis and therapy. Annual Review of Medicine. 2015;66:49-63
  2. 2. Maris JM. Recent advances in neuroblastoma. The New England Journal of Medicine. 2010;362(23):2202-2211
  3. 3. Erttmann R, Tafese T, Berthold F, Kerbl R, Mann J, Parker L, et al. 10 years’ neuroblastoma screening in Europe: Preliminary results of a clinical and biological review from the Study Group for Evaluation of Neuroblastoma Screening in Europe (SENSE). European Journal of Cancer. 1998;34(9):1391-1397
  4. 4. D’Angio GJ, Evans AE, Koop CE. Special pattern of widespread neuroblastoma with a favourable prognosis. Lancet. 1971;1(7708):1046-1049
  5. 5. Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. The New England Journal of Medicine. 2010;363(14):1324-1334
  6. 6. Opel D, Poremba C, Simon T, Debatin KM, Fulda S. Activation of Akt predicts poor outcome in neuroblastoma. Cancer Research. 2007;67(2):735-745
  7. 7. Boller D, Schramm A, Doepfner KT, Shalaby T, von Bueren AO, Eggert A, et al. Targeting the phosphoinositide 3-kinase isoform p110 delta impairs growth and survival in neuroblastoma cells. Clinical Cancer Research. 2008;14(4):1172-1181
  8. 8. Li Z, Thiele CJ. Targeting Akt to increase the sensitivity of neuroblastoma to chemotherapy: Lessons learned from the brain-derived neurotrophic factor/TrkB signal transduction pathway. Expert Opinion on Therapeutic Targets. 2007;11(12):1611-1621
  9. 9. Paz-Ares L, Blanco-Aparicio C, Garcia-Carbonero R, Carnero A. Inhibiting PI3K as a therapeutic strategy against cancer. Clinical & Translational Oncology. 2009;11(9):572-579
  10. 10. Qi L, Toyoda H, Shankar V, Sakurai N, Amano K, Kihira K, et al. Heterogeneity of neuroblastoma cell lines in insulin-like growth factor 1 receptor/Akt pathway-mediated cell proliferative responses. Cancer Science. 2013;104(9):1162-1171
  11. 11. Qi L, Toyoda H, Xu DQ, Zhou Y, Sakurai N, Amano K, et al. PDK1-mTOR signaling pathway inhibitors reduce cell proliferation in MK2206 resistant neuroblastoma cells. Cancer Cell International. 2015;15:91
  12. 12. Xu DQ, Toyoda H, Qi L, Morimoto M, Hanaki R, Iwamoto S, et al. Induction of MEK/ERK activity by AZD8055 confers acquired resistance in neuroblastoma. Biochemical and Biophysical Research Communications. 2018;499(3):425-432
  13. 13. Xu DQ, Toyoda H, Yuan XJ, Qi L, Chelakkot VS, Morimoto M, et al. Anti-tumor effect of AZD8055 against neuroblastoma cells in vitro and in vivo. Experimental Cell Research. 2018;365(2):177-184
  14. 14. Bentov I, Werner H. IGF, IGF receptor and overgrowth syndromes. Pediatric Endocrinology Reviews. 2004;1(4):352-360
  15. 15. Werner H, Bruchim I. The insulin-like growth factor-I receptor as an oncogene. Archives of Physiology and Biochemistry. 2009;115(2):58-71
  16. 16. Brodt P, Samani A, Navab R. Inhibition of the type I insulin-like growth factor receptor expression and signaling: Novel strategies for antimetastatic therapy. Biochemical Pharmacology. 2000;60(8):1101-1107
  17. 17. Gronborg M, Wulff BS, Rasmussen JS, Kjeldsen T, Gammeltoft S. Structure-function relationship of the insulin-like growth factor-I receptor tyrosine kinase. The Journal of Biological Chemistry. 1993;268(31):23435-23440
  18. 18. Laviola L, Natalicchio A, Giorgino F. The IGF-I signaling pathway. Current Pharmaceutical Design. 2007;13(7):663-669
  19. 19. Pollak M. Insulin and insulin-like growth factor signalling in neoplasia. Nature Reviews Cancer. 2008;8(12):915-928
  20. 20. Sartelet H, Oligny LL, Vassal G. AKT pathway in neuroblastoma and its therapeutic implication. Expert Review of Anticancer Therapy. 2008;8(5):757-769
  21. 21. Mahindra A, Cirstea D, Raje N. Novel therapeutic targets for multiple myeloma. Future Oncology. 2010;6(3):407-418
  22. 22. Barlaskar FM, Spalding AC, Heaton JH, Kuick R, Kim AC, Thomas DG, et al. Preclinical targeting of the type I insulin-like growth factor receptor in adrenocortical carcinoma. The Journal of Clinical Endocrinology and Metabolism. 2009;94(1):204-212
  23. 23. Zumkeller W, Schwab M. Insulin-like growth factor system in neuroblastoma tumorigenesis and apoptosis: Potential diagnostic and therapeutic perspectives. Hormone and Metabolic Research. 1999;31(2-3):138-141
  24. 24. Surmacz E. Growth factor receptors as therapeutic targets: Strategies to inhibit the insulin-like growth factor I receptor. Oncogene. 2003;22(42):6589-6597
  25. 25. Girnita A, Girnita L, del Prete F, Bartolazzi A, Larsson O, Axelson M. Cyclolignans as inhibitors of the insulin-like growth factor-1 receptor and malignant cell growth. Cancer Research. 2004;64(1):236-242
  26. 26. Stromberg T, Ekman S, Girnita L, Dimberg LY, Larsson O, Axelson M, et al. IGF-1 receptor tyrosine kinase inhibition by the cyclolignan PPP induces G2/M-phase accumulation and apoptosis in multiple myeloma cells. Blood. 2006;107(2):669-678
  27. 27. Vasilcanu D, Girnita A, Girnita L, Vasilcanu R, Axelson M, Larsson O. The cyclolignan PPP induces activation loop-specific inhibition of tyrosine phosphorylation of the insulin-like growth factor-1 receptor. Link to the phosphatidyl inositol-3 kinase/Akt apoptotic pathway. Oncogene. 2004;23(47):7854-7862
  28. 28. Karasic TB, Hei TK, Ivanov VN. Disruption of IGF-1R signaling increases TRAIL-induced apoptosis: A new potential therapy for the treatment of melanoma. Experimental Cell Research. 2010;316(12):1994-2007
  29. 29. Klinakis A, Szabolcs M, Chen G, Xuan S, Hibshoosh H, Efstratiadis A. Igf1r as a therapeutic target in a mouse model of basal-like breast cancer. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(7):2359-2364
  30. 30. Yin S, Girnita A, Stromberg T, Khan Z, Andersson S, Zheng H, et al. Targeting the insulin-like growth factor-1 receptor by picropodophyllin as a treatment option for glioblastoma. Neuro-Oncology. 2010;12(1):19-27
  31. 31. Varela-Nieto I, Hartl M, Gorospe I, Leon Y. Anti-apoptotic actions of insulin-like growth factors: Lessons from development and implications in neoplastic cell transformation. Current Pharmaceutical Design. 2007;13(7):687-703
  32. 32. Dupont J, Pierre A, Froment P, Moreau C. The insulin-like growth factor axis in cell cycle progression. Hormone and Metabolic Research. 2003;35(11-12):740-750
  33. 33. Tazzari PL, Tabellini G, Bortul R, Papa V, Evangelisti C, Grafone T, et al. The insulin-like growth factor-I receptor kinase inhibitor NVP-AEW541 induces apoptosis in acute myeloid leukemia cells exhibiting autocrine insulin-like growth factor-I secretion. Leukemia. 2007;21(5):886-896
  34. 34. Gorlick R, Maris JM, Houghton PJ, Lock R, Carol H, Kurmasheva RT, et al. Testing of the Akt/PKB inhibitor MK-2206 by the pediatric preclinical testing program. Pediatric Blood & Cancer. 2012;59(3):518-524
  35. 35. Li Z, Yan S, Attayan N, Ramalingam S, Thiele CJ. Combination of an allosteric Akt Inhibitor MK-2206 with etoposide or rapamycin enhances the antitumor growth effect in neuroblastoma. Clinical Cancer Research. 2012;18(13):3603-3615
  36. 36. Fouladi M, Perentesis JP, Phillips CL, Leary S, Reid JM, McGovern RM, et al. A phase I trial of MK-2206 in children with refractory malignancies: A children’s oncology group study. Pediatric Blood & Cancer. 2014;61(7):1246-1251
  37. 37. Yap TA, Yan L, Patnaik A, Fearen I, Olmos D, Papadopoulos K, et al. First-in-man clinical trial of the oral pan-AKT inhibitor MK-2206 in patients with advanced solid tumors. Journal of Clinical Oncology. 2011;29(35):4688-4695
  38. 38. Liu P, Cheng H, Roberts TM, Zhao JJ. Targeting the phosphoinositide 3-kinase pathway in cancer. Nature Reviews Drug Discovery. 2009;8(8):627-644
  39. 39. Polivka J Jr, Janku F. Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway. Pharmacology & Therapeutics. 2014;142(2):164-175
  40. 40. Knuefermann C, Lu Y, Liu B, Jin W, Liang K, Wu L, et al. HER2/PI-3K/Akt activation leads to a multidrug resistance in human breast adenocarcinoma cells. Oncogene. 2003;22(21):3205-3212
  41. 41. Stern DF. ERBB3/HER3 and ERBB2/HER2 duet in mammary development and breast cancer. Journal of Mammary Gland Biology and Neoplasia. 2008;13(2):215-223
  42. 42. Hoeflich KP, Merchant M, Orr C, Chan J, Den Otter D, Berry L, et al. Intermittent administration of MEK inhibitor GDC-0973 plus PI3K inhibitor GDC-0941 triggers robust apoptosis and tumor growth inhibition. Cancer Research. 2012;72(1):210-219
  43. 43. Smalley KS, Haass NK, Brafford PA, Lioni M, Flaherty KT, Herlyn M. Multiple signaling pathways must be targeted to overcome drug resistance in cell lines derived from melanoma metastases. Molecular Cancer Therapeutics. 2006;5(5):1136-1144
  44. 44. Sos ML, Fischer S, Ullrich R, Peifer M, Heuckmann JM, Koker M, et al. Identifying genotype-dependent efficacy of single and combined PI3K- and MAPK-pathway inhibition in cancer. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(43):18351-18356
  45. 45. Najafov A, Sommer EM, Axten JM, Deyoung MP, Alessi DR. Characterization of GSK2334470, a novel and highly specific inhibitor of PDK1. The Biochemical Journal. 2011;433(2):357-369
  46. 46. Goto S, Umehara S, Gerbing RB, Stram DO, Brodeur GM, Seeger RC, et al. Histopathology (International Neuroblastoma Pathology Classification) and MYCN status in patients with peripheral neuroblastic tumors: A report from the children’s cancer group. Cancer. 2001;92(10):2699-2708
  47. 47. Seeger RC, Brodeur GM, Sather H, Dalton A, Siegel SE, Wong KY, et al. Association of multiple copies of the N-myc oncogene with rapid progression of neuroblastomas. The New England Journal of Medicine. 1985;313(18):1111-1116
  48. 48. Schramm A, Koster J, Marschall T, Martin M, Schwermer M, Fielitz K, 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. International Journal of Cancer. 2013;132(3):E106-E115
  49. 49. Davidoff AM. Neuroblastoma. Seminars in Pediatric Surgery. 2012;21(1):2-14
  50. 50. Reiff T, Huber L, Kramer M, Delattre O, Janoueix-Lerosey I, Rohrer H. Midkine and Alk signaling in sympathetic neuron proliferation and neuroblastoma predisposition. Development (Cambridge, England). 2011;138(21):4699-4708
  51. 51. Matthay KK, George RE, Yu AL. Promising therapeutic targets in neuroblastoma. Clinical Cancer Research. 2012;18(10):2740-2753
  52. 52. De Brouwer S, De Preter K, Kumps C, Zabrocki P, Porcu M, Westerhout EM, et al. Meta-analysis of neuroblastomas reveals a skewed ALK mutation spectrum in tumors with MYCN amplification. Clinical Cancer Research. 2010;16(17):4353-4362
  53. 53. Ambros PF, Ambros IM. Pathology and biology guidelines for resectable and unresectable neuroblastic tumors and bone marrow examination guidelines. Medical and Pediatric Oncology. 2001;37(6):492-504

Written By

Hidemi Toyoda, Dong-Qing Xu, Lei Qi and Masahiro Hirayama

Submitted: 06 June 2018 Reviewed: 27 September 2018 Published: 12 November 2018