Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 May 1.
Published in final edited form as: Cell Tissue Res. 2018 Jan 5;372(2):277–286. doi: 10.1007/s00441-017-2761-2

Spontaneous regression of neuroblastoma

Garrett M Brodeur 1,2,
PMCID: PMC5920563  NIHMSID: NIHMS932716  PMID: 29305654

Abstract

Neuroblastomas are characterized by heterogeneous clinical behavior, from spontaneous regression or differentiation into a benign ganglioneuroma, to relentless progression despite aggressive, multimodality therapy. Indeed, neuroblastoma is unique among human cancers in terms of its propensity to undergo spontaneous regression. The strongest evidence for this comes from the mass screening studies conducted in Japan, North America and Europe and it is most evident in infants with stage 4S disease. This propensity is associated with a pattern of genomic change characterized by whole chromosome gains rather than segmental chromosome changes but the mechanism(s) underlying spontaneous regression are currently a matter of speculation. There is evidence to support several possible mechanisms of spontaneous regression in neuroblastomas: (1) neurotrophin deprivation, (2) loss of telomerase activity, (3) humoral or cellular immunity and (4) alterations in epigenetic regulation and possibly other mechanisms. It is likely that a better understanding of the mechanisms of spontaneous regression will help to identify targeted therapeutic approaches for these tumors. The most easily targeted mechanism is the delayed activation of developmentally programmed cell death regulated by the tropomyosin receptor kinase A (TrkA) pathway. Pan-Trk inhibitors are currently in clinical trials and so Trk inhibition might be used as the first line of therapy in infants with biologically favorable tumors that require treatment. Alternative approaches consist of breaking immune tolerance to tumor antigens but approaches to telomere shortening or epigenetic regulation are not easily druggable. The different mechanisms of spontaneous neuroblastoma regression are reviewed here, along with possible therapeutic approaches.

Keywords: Neuroblastoma, Regression, Spontaneous, TrkA, Telomerase

Introduction

Most infants with neuroblastoma, even with metastatic disease, can be cured with moderate intensity chemotherapy and some patients with a special pattern of metastasis have a high likelihood of undergoing spontaneous regression without chemotherapy (Diede 2014; Matthay 1998; Nakagawara 1998; Nickerson et al. 2000; Pritchard and Hickman 1994). Indeed, the prevalence of spontaneous regression has been well documented by mass screening programs undertaken in Japan, North America and Europe. Furthermore, children (and adults) can present with localized, benign ganglioneuromas, which presumably represent neuroblastic tumors that differentiated (Brodeur 2003; Garvin et al. 1984; Haas et al. 1988; Hoehner et al. 1995; Shimada et al. 1999a). The exact mechanisms responsible for spontaneous regression (or differentiation) are uncertain but several plausible mechanisms have been proposed to explain these phenomena (Brodeur and Bagatell 2014; Diede 2014; Matthay 1998; Nakagawara 1998; Nickerson et al. 2000; Pritchard and Hickman 1994). Here, the current understanding of the genomic, biological and immunological mechanisms that underlie spontaneous regression is reviewed and possible approaches to therapy are explored.

Historical perspective on neuroblastoma regression

Spontaneous regression of cancer has been documented in different cancer types for decades. Spontaneous regression is defined as the shrinking or disappearance of primary or metastatic disease without therapeutic intervention. Regression has been observed in renal cell carcinoma, malignant melanoma, choriocarcinoma and lymphoid malignancies (Challis and Stam 1990; Everson 1964; Everson and Cole 1966; Papac 1998). However, neuroblastoma is generally considered the disease in which this phenomenon is most prevalent. The actual prevalence of neuroblastoma regression is not unknown but recent studies have provided evidence that regression may be at least as common as clinically detected neuroblastoma.

Beckwith and Perrin (1963) studied the adrenal glands from autopsies of infants under 3 months old who had died for reasons other than neuroblastoma. Interestingly, they discovered foci of neuroblasts in the adrenal glands of 1 out of every 40 infants studied and they proposed that these neuroblastic nodules might eventually evolve into clinically detectable neuroblastoma, so they called them insipient or “in situ” neuroblastoma (Beckwith and Perrin 1963). If the prevalence of clinically detected neuroblastoma is about 1 in 8000 live births, then the prevalence of spontaneous regression of neuroblastoma would be 200 times more common than clinically detected disease (i.e., 2%), which seems very high.

Later, other investigators studied the adrenal glands of fetuses that died prenatally at various gestational ages. They found similar neuroblastic foci in all the fetal adrenal glands they studied, with a peak between 16 and 20 weeks of gestation, after that these foci gradually disappeared (Ikeda et al. 1981; Turkel and Itabashi 1974). Based on this information, the neuroblastic rests seen by Beckwith and Perrin were likely the residual elements of normal sympathoadrenal development rather than insipient neuroblastoma. Nevertheless, these normal neuroblastic nodules may contain the cells from which adrenal neuroblastomas arise.

The phenomenon of neuroblastoma regression was already known but Evans and D’Angio identified a specific pattern of metastatic spread in infants less than a year of age that they called stage IVS (D'Angio et al. 1971; Evans et al. 1971). Infants with stage IVS generally had small abdominal primary tumors with dissemination limited to the liver and skin, with or without minimal marrow involvement. These patients generally had a very good prognosis and some even underwent spontaneous regression in the absence of tumor-specific therapy (D'Angio et al. 1971; Evans et al. 1971). In contrast, patients over 12–18 months of age with metastatic disease generally had a different pattern of dissemination that included bone lesions and extensive marrow involvement and patients with this pattern generally had a very poor prognosis and never regressed (George et al. 2006; Matthay et al. 1999).

The definition of stage IVS was refined by the International Neuroblastoma Staging System (INSS) as stage 4S (Brodeur et al. 1988, 1993) and by the International Neuroblastoma Risk Group Staging System as stage MS (M for metastatic, S for special) (Monclair et al. 2009); this stage will be referred to as 4S in this review. The main difference now is that children up to 18 months of age can be considered to have stage 4S (George et al. 2005; London et al. 2005; Monclair et al. 2009). However, it is clear that spontaneous regression is not restricted to stage 4S, because regression can be seen in infants with virtually any stage of disease if they have biologically favorable tumors (Cozzi et al. 2013; Fisher and Tweddle 2012; Kushner et al. 1996; Taggart et al. 2011).

Genomic and biological features of regressing neuroblastomas

It is difficult to know for certain which neuroblastomas will regress based on age and stage alone. Therefore, investigators have focused on stage 4S tumors as a surrogate for neuroblastomas that are likely to regress. Lavarino et al. (2009) conducted a study on 35 infants with metastatic neuroblastoma—25 with stage 4 and 10 with stage 4S. The tumors from patients with stage 4 disease were characterized by segmental chromosomal aberrations, whereas 90% of stage 4S tumors were near-triploid with whole chromosome gains (Lavarino et al. 2009). These investigators found differential expression of certain genes (such as CHD5, GNB1 and RERE) in stage 4 and stage 4S neuroblastoma, with higher expression of genes mapping to the short arm of chromosome 1 in stage 4S tumors and to chromosome 11 for stage 4 tumors. Benard et al. (2008) studied 29 cases of metastatic neuroblastoma lacking MYCN amplification. They developed a genetic signature of 45 genes that was significantly associated with stage 4S (12 cases) versus stage 4 tumors (17 cases) and this was validated in an independent set of 22 tumors. A smaller proteomic study was performed on eight tumors from infants with stage 4 and 4S that identified another set of differentially expressed proteins between the two stages (Yu et al. 2011). There was essentially no overlap of genes (or proteins) that were differentially expressed by regressing 4S versus non-regressing infant tumors among these studies, so more studies are needed.

Insights from mass screening for neuroblastoma

Mass screening studies for neuroblastoma were undertaken in Japan, North America and Europe to identify neuroblastomas early, because the outcome of infants with neuroblastoma is substantially better than that of older patients. Almost all neuroblastomas produce catecholamines and their metabolites, so mass screening was conducted by measuring urinary catecholamine metabolites of infants at specific times between 3 weeks and 6 months of age. Mass screening of infants for neuroblastoma was initiated in Japan and initial results were promising (Bessho 1999; Sawada et al. 1984; Yamamoto et al. 2002), so similar efforts were initiated in North America and in Europe (Erttmann et al. 1998; Woods et al. 1996). However, mass screening for neuroblastoma resulted in a substantial increase in the prevalence of neuroblastoma in screened compared with unscreened populations (~1:2000 vs. 1:8000 respectively) and the overall mortality from neuroblastoma was unchanged (Bessho 1999; Schilling et al. 2002; Woods et al. 2002; Yamamoto et al. 2002). Thus, mass screening did not reduce neuroblastoma mortality and screening efforts have essentially stopped worldwide.

Nevertheless, these mass-screening studies provided valuable insights into the pathogenesis and clinical behavior of biologically favorable tumors. The increased prevalence of neuroblastoma observed in the screened populations indicates that spontaneous regression of neuroblastoma (without clinical detection) occurs at least as frequently as clinically detected neuroblastoma. In addition, genomic analyses performed on screened tumors showed that most of them, regardless of their stage, were biologically favorable with respect to MYCN status and tumor cell ploidy (Brodeur et al. 1998; Hayashi et al. 1992; Kaneko et al. 1990). This is in contrast to the unfavorable biological features generally found in clinically detected tumors from older children. Importantly, these studies also suggested that biologically favorable tumors rarely evolve into biologically unfavorable tumors. There have also been reports of incidental prenatal detection of neuroblastoma by maternal ultrasound (Acharya et al. 1997; Ho et al. 1993; Saylors et al. 1994). These cases are similar both clinically and biologically to those identified by screening and the vast majority does well with little or no therapy.

Mechanisms of spontaneous regression

Neurotrophin receptors and regression

Neuroblastomas are derived from sympathetic neuronal precursors and many more precursor cells are produced during normal development than are necessary to form the sympathetic nervous system. Those that make a proper connection to a target organ or tissue will survive and those that do not are destined to undergo developmentally programmed cell death (Estus et al. 1994; Ham et al. 1995). This process is regulated primarily by the TrkA neurotrophin receptor and the limiting availability of its cognate ligand, nerve growth factor (NGF) at their target site. Nevertheless, these neuronal precursors survive, migrate and proliferate in the absence of NGF during early embryogenesis, so there must be a developmental switch from an NGF-independent to an NGF-dependent state.

TrkA (encoded by NTRK1), a neurotrophin receptor, is a member of a family of receptor tyrosine kinases that includes TrkB (encoded by NTRK2) and TrkC (encoded by NTRK3). Each of these receptors plays a critical role in the development and maintenance of the central and peripheral nervous systems. These receptors also have important roles in neuroblastoma pathogenesis (Brodeur et al. 1997, 2009; Thiele et al. 2009). High TrkA expression is associated with favorable clinical and biological features, such as younger age, lower stage and absence of MYCN amplification and these patients have an excellent outcome (Kogner et al. 1993; Nakagawara et al. 1992, 1993; Suzuki et al. 1993). In contrast, TrkB is coexpressed at high levels along with its ligand, BDNF, in clinically and biologically unfavorable tumors, especially those with MYCN amplification (Nakagawara et al. 1994). Autocine activation of TrkB by BDNF leads to invasion, metastasis, angiogenesis and drug resistance (Acheson et al. 1995; Jaboin et al. 2002; Matsumoto et al. 1995; Nakagawara et al. 1994; Nakamura et al. 2006). TrkA and TrkC are dependence receptors, because the absence of ligand activation leads to apoptotic signaling and cell death, whereas TrkB is not (Goldschneider and Mehlen 2010; Nikoletopoulou et al. 2010; Rabizadeh et al. 1999). Co-expression of the P75/NGFR receptor can increase the sensitivity and specificity of all three Trk receptors for their cognate ligands (Hantzopoulos et al. 1994; Ho et al. 2011) but activation of P75/NGFR in the absence of TrkA signaling leads to apoptosis (Bamji et al. 1998; Rabizadeh et al. 1999).

Tumors from infants, especially with low-stage or 4S disease, generally have high levels of TrkA expression (Kogner et al. 1993; Nakagawara et al. 1992 1993; Suzuki et al. 1993). When cells derived from these tumors are put in primary culture with exogenous NGF, they undergo neuronal differentiation and survive for months. In contrast, the same cells undergo apoptosis within a week if cultured in the absence of exogenous NGF (Nakagawara et al. 1993; Nakagawara and Brodeur 1997) (Fig. 1a). Taken together with the pivotal role of TrkA and NGF in development of the sympathetic nervous system, these results mimic the behavior of TrkA-expressing neuroblastomas in patients: tumors undergo neuronal differentiation to a ganglioneuroma, or they undergo spontaneous regression (apoptosis), depending on the presence or absence (respectively) of NGF in their microenvironment.

Fig. 1.

Fig. 1

Mechanisms of spontaneous regression. a The TrkA-NGF pathway and apoptosis. Neurotrophin deprivation (TrkA without NGF) leads to activation of developmentally programmed apoptosis and regression of neuroblastomas. b Telomere shortening and apoptosis. Telomere shortening results from low/absent levels of telomerase and triggers apoptosis and regression of neuroblastomas. c Immune-mediated destruction. Immune-mediated cell killing by anti-neuroblastoma antibodies (and antibody-dependent cellular toxicity) or by NK cells leads to death of neuroblastoma cells and tumor regression. d Epigenetic modification. Alterations of gene expression can occur by promoter methylation, histone modification or chromatin remodeling, leading to neuroblastoma regression

It is unclear why migrating neural crest precursors and favorable TrkA-expressing neuroblastomas survive (at least initially), despite a lack of available NGF. Initially, TrkA expression on these precursor cells is low, as is P75/NGFR and these cells are not dependent on NGF (Nikoletopoulou et al. 2010). NGF-independent neuronal precursors could depend on an alternative receptor or pathway for survival (such as TrkC or RET) (Kahane and Kalcheim 1994; Pachnis et al. 1993; Tsuzuki et al. 1995) and then switch dependence to TrkA, only to undergo apoptosis and regress in the absence of NGF. Another possibility is that these migrating cells express TrkAIII, a TrkA(I) isoform that is expressed in normal sympathoadrenal progenitors as well as in some neuroblastomas (Tacconelli et al. 2004, 2005). This isoform results from alternative splicing and maintains the reading frame but removes the ligand-binding site, leading to constitutive kinase activation. Thus, the conversion from a TrkA-expressing, NGF-independent neuroblastoma to a NGF-dependent tumor could result simply from a developmentally programmed isoform switch from TrkAIII to TrkAI. Thus, a switch in TrkA dependence could explain spontaneous regression of neuroblastomas but there are additional mechanisms to explain this phenomenon as well.

Telomerase, telomeres and regression

High telomerase activity is generally associated with more-aggressive behavior and poor prognosis in patients with neuroblastoma (Krams et al. 2003; Ohali et al. 2006 Peifer et al. 2015; Streutker et al. 2001). However, telomere shortening has been proposed as another possible explanation for spontaneous regression of neuroblastoma. Telomeres are specialized structures at the ends of chromosomes that are important for the replication and stability of chromosomes and regulation of the telomere length is controlled in part by the enzyme telomerase. Telomerase expression is frequently high in cancer and immortalized cells but generally low in most normal and senescent cells Kim et al. 1994). Hiyama et al. (1995) examined the telomere length and telomerase activity of 100 neuroblastoma samples. Most tumors with high telomerase activity had a poor prognosis and all tumors with MYCN amplification had high telomerase activity. However, most of the 4S neuroblastoma samples had low telomerase activity or short telomeres, a pattern usually associated with senescent cells (Fig. 1b) (Hiyama et al. 1995). Interestingly, Samy et al. (2012) transfected a neuroblastoma cell line with a dominant negative form of telomerase and tumors formed by these cells showed increased apoptosis and reduced tumorigenicity compared to untransfected cells in a mouse xenograft model. Together, these data suggest that loss of telomerase activity and telomere shortening is a plausible mechanism to explain spontaneous regression of neuroblastoma and possibly of other tumors.

Immunological mechanisms and regression

Spontaneous regression of primary and metastatic cancers (including neuroblastoma) has occurred in association with acute infection (Everson 1964; Everson and Cole 1966). Furthermore, tumor-infiltrating lymphocytes have been observed in neuroblastomas and there is evidence for both tumor-targeted T-cells and antineural antibodies in patients with neuroblastoma (Antunes et al. 2000; Kataoka et al. 1993; Valteau et al. 1996). Thus, another potential explanation of spontaneous regression is tumor destruction mediated by an anti-tumor immune response (Fig. 1c). Interestingly, the paraneoplastic opsomyoclonus syndrome (OMS) is associated with the presence of antineural antibodies and a favorable outcome in patients with neuroblastoma (Antunes et al. 2000; Cooper et al. 2001; Pranzatelli et al. 2004; Rudnick et al. 2001; Russo et al. 1997). About 50% of patients with OMS have neuroblastoma, which suggests that the other 50% either had a neuroblastoma that regressed or they have a de novo autoimmune disease. However, it is still unclear if spontaneous regression can be mediated by a humoral or cellular immune response.

Neuroblastoma cells from patients with high-risk disease may evade immune destruction by downregulating human leukocyte antigen (HLA) class I molecules (Raffaghello et al. 2005). However, most tumors from patients with stage 4S neuroblastoma express normal levels of HLA class I antigens (Squire et al. 1990). In vitro exposure to interferon-γ can induce upregulation of the expression of class I antigens in neuroblastoma cells (Raffaghello et al. 2005). Thus, upregulation of HLA class I might be a strategy to augment immune surveillance and promote tumor regression. Although in vitro exposure to interferon-γ can enhance the recognition of neuroblastoma cells by cytotoxic T cells, it can also reduce their susceptibility to killing by natural killer (NK) cells (Raffaghello et al. 2005). Only a few low stage and 4S neuroblastomas have been studied, so further study of the role of cytokines in mediating regression is needed.

Asgharzadeh et al. (2012) studied tumor-associated macrophage (TAM) infiltration in tumors from patients with various stages of MYCN non-amplified disease. However, this study reported that the number of TAM in INSS 4S tumors is similar to locoregional neuroblastomas. Metastatic tumors (stages 4 and 4S) from young patients (<18 months old) had significantly higher expression of genes representing TAMs than did metastatic tumors from older patients. This suggests that the inflammatory response and the tumor microenvironment might contribute to the behavior of neuroblastoma in patients based on age at diagnosis or stage of disease (Asgharzadeh et al. 2012). This study did not specifically evaluate regressing tumors, so further investigation is needed to understand the contribution of the immune system and tumor microenvironment to neuroblastoma regression.

Epigenetic regulation and other mechanisms

Gene expression is affected by promoter methylation, histone modification or chromatin remodeling, which in turn may effect neuroblastoma cell survival or differentiation (Fig. 1d). Epigenetic changes affecting expression of genes relevant to neuroblastoma development were initially reported over a decade ago (Astuti et al. 2001; Takita et al. 2000) and several studies have suggested that alterations in gene methylation or histone modification are related to patient outcome (Barbieri et al. 2014; Decock et al. 2011; Grau et al. 2010; Yang et al. 2007). Diskin et al. (2014) identified global differences in the methylomes of 22 stage 4S neuroblastomas compared to low-risk tumors, high-risk tumors and normal brain tissues. Reduced promoter methylation in the 4S samples was observed in 97% of the genes for which differential methylation was detected. Others have also reported different patterns of methylation in tumors from patients with stage 4S compared to other stages (Decock et al. 2011). However, additional studies are needed to validate these findings and develop more specific information regarding DNA methylation, histone modification, or chromatin modification during differentiation and regression (Baylin 2005; Gros et al. 2012; McCabe and Creasy 2014).

Therapy to induce neuroblastoma regression

TrkA inhibitors

Inhibition of the TrkA receptor pathway is a promising approach to induce tumor regression in biologically favorable neuroblastomas. TrkA-expressing neuroblastoma cells placed in culture survive and differentiate in the presence of NGF but they undergo apoptosis in its absence (Nakagawara and Brodeur 1997). Thus, depriving cells of NGF or inhibiting TrkA signaling may be an effective approach to induce regression. Lestaurtinib (CEP-701) is a small molecule inhibitor of TRK receptors (TrkA, TrkB and TrkC) and it has shown preclinical activity against TrkB-expressing neuroblastoma xenografts (Evans et al. 1999, 2001; Ho et al. 2002; Iyer et al. 2010). Furthermore, lestaurtinib showed significant clinical activity in a phase I trial for recurrent and/or refractory neuroblastoma (Minturn et al. 2011). These studies provide evidence that Trk-selective inhibitors could be effective in the treatment of Trk-driven neuroblastomas. Indeed, several second-generation Trk inhibitors are currently in phase 1 clinical trials or in preclinical development (Croucher et al. 2015, Doebele et al. 2015, Drilon et al. 2017; Iyer et al. 2012, 2016; Nagasubramanian et al. 2016). These agents are potent inhibitors of all three Trk family neurotrophin receptors, so the same agent could be used to target TrkA in biologically favorable tumors and TrkB in unfavorable tumors. If second-generation Trk inhibitors prove to be safe and effective against TrkB-expressing, high-risk disease, these agents could be used to treat patients with TrkA-expressing 4S neuroblastoma who have massive liver involvement in lieu of chemotherapy or radiation therapy. We hypothesize that a Trk inhibitor would initiate apoptosis and regression in these tumors, obviating the need to wait for spontaneous regression to occur.

Immunological therapy

Immunotherapy using a chimeric antibody (ch14.18) directed against the disialoganglioside GD2 has been incorporated into frontline treatment of patients with high-risk neuroblastoma (Yu et al. 2010) and preliminary studies have been carried out using adoptive immunotherapy in patients with recurrent/refractory disease (Louis et al. 2011). Future studies of the immunology of neuroblastoma regression should influence the evolution of current immunotherapeutic approaches and lead to new strategies to accelerate regression in young infants with life-threatening but biologically favorable disease. Immune modulation to induce regression could be advantageous for patients with 4S or locoregional disease, because there is a trend to reduce conventional cytotoxic therapy and avoid aggressive surgery in patients with a favorable prognosis (Baker et al. 2010; Hero et al. 2008; Nuchtern et al. 2012). Treatment with interferon-γ may upregulate HLA class I expression and increase immune cell recognition. However, strategies to enhance one component of the immune system may diminish the anti-tumor effects of another key component. A better understanding is needed of the complex interactions between neuroblastoma cells, the tumor microenvironment and the immune system, as well as the implications of immune modulation in very young children.

Other approaches

At the present time, there are no therapeutic approaches to influence telomere length in neuroblastomas. However, there are other approaches that might be considered for these patients. For example, retinoids are a class of compounds that have been shown to induce cellular differentiation and decrease proliferation of neuroblastoma cells in vitro, presumably mediated by the upregulation of neural differentiation genes (Yuza et al. 2003). Indeed, 13-cis-retinoic acid has been incorporated into frontline therapy for children with high-risk neuroblastoma to induce differentiation in states of minimal residual disease following intensive, multimodality therapy (Shimada et al. 1999b). The mechanisms by which isotretinoin induces differentiation are unclear but retinoids are associated with increased expression of Trk receptors, which may explain the induction of differentiation (Rodriguez-Tebar and Rohrer 1991; Yuza et al. 2003). Vorinostat, a histone deacetylase inhibitor, has been given to children with relapsed or refractory disease. Further studies of epigenetic modifiers may alter gene expression in neuroblastomas and consequently induce tumor regression.

Conclusions

Neuroblastomas show a remarkable capacity to undergo spontaneous regression. The prevalence of this phenomenon is hard to determine precisely but the experience from mass screening programs suggests that there are at least as many children who have tumors undergoing spontaneous regression without clinical detection as there are patients with neuroblastoma detected clinically. Further exploration of this issue and a greater understanding of the normal mechanism(s) of spontaneous regression might allow the identification of tumors that have the capacity to undergo spontaneous regression and to induce regression in susceptible tumors using pharmacological, biological or immunological approaches. To this end, we would need to study samples from regressing tumors, such as skin nodules from stage 4S patients. These go through three phases of growth (proliferation), a plateau of growth and then disappearance. The potential mechanisms of regression described above could each lead to these successive changes, once the mechanism was activated (TrkA dependence, telomere shortening, immune response, or epigenetic modification).

At the present time, the most promising therapeutic approach would be aimed at inhibiting the TrkA receptor pathway. However, most Trk inhibitors are potent inhibitors of TrkA, TrkB and TrkC. Before these agents are used to treat infants with stage 4S disease, clinical trials of second-generation Trk inhibitors would need to demonstrate safety and efficacy against TrkB-expressing recurrent and/or refractory neuroblastomas, especially in young children. Currently, there are no opportunities to selectively promote telomere shortening, initiate a targeted immune response, or preferentially induce epigenetic modifications in tumor tissue.

Acknowledgments

Some of the information in this review has been presented previously (Brodeur and Bagatell 2014). This work was supported in part by a grant from the National Cancer Institute, Alex’s Lemonade Stand Foundation and the Audrey E. Evans endowed chair (GMB).

Abbreviations

HLA

Human leukocyte antigen

INSS

International Neuroblastoma Staging System

NK

Natural killer

NGF

Nerve growth factor

OMS

Opsomyoclonus syndrome

TAM

Tumor-associated macrophages

TRK

Tropomyosin receptor kinase

References

  1. Acharya S, Jayabose S, Kogan SJ, Tugal O, Beneck D, Leslie D, Slim M. Prenatally diagnosed neuroblastoma. Cancer. 1997;80:304–310. doi: 10.1002/(sici)1097-0142(19970715)80:2<304::aid-cncr19>3.0.co;2-y. [DOI] [PubMed] [Google Scholar]
  2. Acheson A, Conover JC, Fandi JP, DeChiara TM, Russell M, Thadani A, Squinto SP, Yancopoulos GD, Lindsay RM. A BDNF autocrine loop in adult sensory neurons prevents cell death. Nature. 1995;374:450–453. doi: 10.1038/374450a0. [DOI] [PubMed] [Google Scholar]
  3. Antunes NL, Khakoo Y, Matthay KK, Seeger RC, Stram DO, Gerstner E, Abrey LE, Dalmau J. Antineuronal antibodies in patients with neuroblastoma and paraneoplastic opsoclonus-myoclonus. J Pediatr Hematol Oncol. 2000;22:315–320. doi: 10.1097/00043426-200007000-00007. [DOI] [PubMed] [Google Scholar]
  4. Asgharzadeh S, Salo JA, Ji L, Oberthuer A, Fischer M, Berthold F, Hadjidaniel M, Liu CW, Metelitsa LS, Pique-Regi R, Wakamatsu P, Villablanca JG, Kreissman SG, Matthay KK, Shimada H, London WB, Sposto R, Seeger RC. Clinical significance of tumor-associated inflammatory cells in metastatic neuroblastoma. J Clin Oncol. 2012;30:3525–3532. doi: 10.1200/JCO.2011.40.9169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Astuti D, Agathanggelou A, Honorio S, Dallol A, Martinsson T, Kogner P, Cummins C, Neumann HP, Voutilainen R, Dahia P, Eng C, Maher ER, Latif F. RASSF1A promoter region CpG island hypermethylation in phaeochromocytomas and neuroblastoma tumours. Oncogene. 2001;20:7573–7577. doi: 10.1038/sj.onc.1204968. [DOI] [PubMed] [Google Scholar]
  6. Baker DL, Schmidt ML, Cohn SL, Maris JM, London WB, Buxton A, Stram D, Castleberry RP, Shimada H, Sandler A, Shamberger RC, Look AT, Reynolds CP, Seeger RC, Matthay KK, Children's Oncology G Outcome after reduced chemotherapy for intermediate-risk neuroblastoma. N Engl J Med. 2010;363:1313–1323. doi: 10.1056/NEJMoa1001527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bamji SX, Majdan M, Pozniak CD, Belliveau DJ, Aloyz R, Kohn J, Causing CG, Miller FD. The p75 neurotrophin receptor mediates neuronal apoptosis and is essential for naturally occurring sympathetic neuron death. J Cell Biol. 1998;140:911–923. doi: 10.1083/jcb.140.4.911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Barbieri E, De Preter K, Capasso M, Chen Z, Hsu DM, Tonini GP, Lefever S, Hicks J, Versteeg R, Pession A, Speleman F, Kim ES, Shohet JM. Histone chaperone CHAF1A inhibits differentiation and promotes aggressive neuroblastoma. Cancer Res. 2014;74:765–774. doi: 10.1158/0008-5472.CAN-13-1315. [DOI] [PubMed] [Google Scholar]
  9. Baylin SB. DNA methylation and gene silencing in cancer. Nat Clin Pract Oncol. 2005;2(Suppl 1):S4–11. doi: 10.1038/ncponc0354. [DOI] [PubMed] [Google Scholar]
  10. Beckwith JB, Perrin EV. In situ neuroblastomas: a contribution to the natural history of neural crest tumors. Am J Pathol. 1963;43:1089–1104. [PMC free article] [PubMed] [Google Scholar]
  11. Benard J, Raguenez G, Kauffmann A, Valent A, Ripoche H, Joulin V, Job B, Danglot G, Cantais S, Robert T, Terrier-Lacombe MJ, Chassevent A, Koscielny S, Fischer M, Berthold F, Lipinski M, Tursz T, Dessen P, Lazar V, Valteau-Couanet D. MYCN-non-amplified metastatic neuroblastoma with good prognosis and spontaneous regression: a molecular portrait of stage 4S. Mol Oncol. 2008;2:261–271. doi: 10.1016/j.molonc.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bessho F. Comparison of the incidences of neuroblastoma for screened and unscreened cohorts. Acta Paediatr. 1999;88:404–406. doi: 10.1080/08035259950169774. [DOI] [PubMed] [Google Scholar]
  13. Brodeur GM. Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer. 2003;3:203–216. doi: 10.1038/nrc1014. [DOI] [PubMed] [Google Scholar]
  14. Brodeur GM, Ambros PF, Favrot MC. Biological aspects of neuroblastoma screening. Med Pediatr Oncol. 1998;31:394–400. [Google Scholar]
  15. Brodeur GM, Bagatell R. Mechanisms of neuroblastoma regression. Nat Rev Clin Oncol. 2014;11:704–713. doi: 10.1038/nrclinonc.2014.168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Brodeur GM, Minturn JE, Ho R, Simpson AM, Iyer R, Varela CR, Light JE, Kolla V, Evans AE. Trk receptor expression and inhibition in neuroblastomas. Clin Cancer Res. 2009;15:3244–3250. doi: 10.1158/1078-0432.CCR-08-1815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Brodeur GM, Nakagawara A, Yamashiro DJ, Ikegaki N, Liu XG, Azar CG, Lee CP, Evans AE. Expression of TrkA, TrkB and TrkC in human neuroblastomas. J Neuro-Oncol. 1997;31:49–55. doi: 10.1023/a:1005729329526. [DOI] [PubMed] [Google Scholar]
  18. Brodeur GM, Pritchard J, Berthold F, Carlsen NL, Castel V, Castelberry RP, De Bernardi B, Evans AE, Favrot M, Hedborg F, et al. Revisions of the international criteria for neuroblastoma diagnosis, staging, and response to treatment. J Clin Oncolo. 1993;11:1466–1477. doi: 10.1200/JCO.1993.11.8.1466. [DOI] [PubMed] [Google Scholar]
  19. Brodeur GM, Seeger RC, Barrett A, Berthold F, Castleberry RP, D'Angio G, De Bernardi B, Evans AE, Favrot M, Freeman AI, et al. International criteria for diagnosis, staging, and response to treatment in patients with neuroblastoma. J Clin Oncol. 1988;6:1874–1881. doi: 10.1200/JCO.1988.6.12.1874. [DOI] [PubMed] [Google Scholar]
  20. Challis GB, Stam HJ. The spontaneous regression of cancer. A review of cases from 1900 to 1987. Acta Oncol. 1990;29:545–550. doi: 10.3109/02841869009090048. [DOI] [PubMed] [Google Scholar]
  21. Cooper R, Khakoo Y, Matthay KK, Lukens JN, Seeger RC, Stram DO, Gerbing RB, Nakagawa A, Shimada H. Opsoclonus-myoclonus-ataxia syndrome in neuroblastoma: histopathologic features-a report from the Children's cancer group. Med Pediatr Oncol. 2001;36:623–629. doi: 10.1002/mpo.1139. [DOI] [PubMed] [Google Scholar]
  22. Cozzi DA, Mele E, Ceccanti S, Natale F, Clerico A, Schiavetti A, Dominici C. Long-term follow-up of the "wait and see" approach to localized perinatal adrenal neuroblastoma. World J Surg. 2013;37:459–465. doi: 10.1007/s00268-012-1837-0. [DOI] [PubMed] [Google Scholar]
  23. Croucher JL, Iyer R, Li N, Molteni V, Loren J, Gordon WP, Tuntland T, Liu B, Brodeur GM. TrkB inhibition by GNF-4256 slows growth and enhances chemotherapeutic efficacy in neuroblastoma xenografts. Cancer Chemother Pharmacol. 2015;75:131–141. doi: 10.1007/s00280-014-2627-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. D'Angio GJ, Evans AE, Koop CE. Special pattern of widespread neuroblastoma with a favourable prognosis. Lancet. 1971;1:1046–1049. doi: 10.1016/s0140-6736(71)91606-0. [DOI] [PubMed] [Google Scholar]
  25. Decock A, Ongenaert M, Vandesompele J, Speleman F. Neuroblastoma epigenetics: from candidate gene approaches to genome-wide screenings. Epigenetics. 2011;6:962–970. doi: 10.4161/epi.6.8.16516. [DOI] [PubMed] [Google Scholar]
  26. Diede SJ. Spontaneous regression of metastatic cancer: learning from neuroblastoma. Nat Rev Cancer. 2014;14:71–72. doi: 10.1038/nrc3656. [DOI] [PubMed] [Google Scholar]
  27. Diskin SJ, McDaniel L, Oldridge DA, Attiyeh E, Asgharzadeh S, Weisenberger DJ, Shen H, Diamond M, Auvil AG, Smith MA, Gerhard DS, Hogarty MD, London WB, Khan J, Seeger RC, Laird PW, Maris JM. Integrative genomic and epigenomic characterization of stage4S neuroblastoma. Advances in neuroblastoma research–2014, Cologne, Germany 2014 [Google Scholar]
  28. Doebele RC, Davis LE, Vaishnavi A, Le AT, Estrada-Bernal A, Keysar S, Jimeno A, Varella-Garcia M, Aisner DL, Li Y, Stephens PJ, Morosini D, Tuch BB, Fernandes M, Nanda N, Low JA. An oncogenic NTRK fusion in a patient with soft-tissue sarcoma with response to the tropomyosin-related kinase inhibitor LOXO-101. Cancer Discov. 2015;5:1049–1057. doi: 10.1158/2159-8290.CD-15-0443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Drilon A, Siena S, Ou SI, Patel M, Ahn MJ, Lee J, Bauer TM, Farago AF, Wheler JJ, Liu SV, Doebele R, Giannetta L, Cerea G, Marrapese G, Schirru M, Amatu A, Bencardino K, Palmeri L, Sartore-Bianchi A, Vanzulli A, Cresta S, Damian S, Duca M, Ardini E, Li G, Christiansen J, Kowalski K, Johnson AD, Patel R, Luo D, Chow-Maneval E, Hornby Z, Multani PS, Shaw AT, De Braud FG. Safety and antitumor activity of the multitargeted pan-TRK, ROS1, and ALK inhibitor Entrectinib: combined results from two phase I trials (ALKA-372-001 and STARTRK-1) Cancer Discov. 2017;7:400–409. doi: 10.1158/2159-8290.CD-16-1237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Erttmann R, Tafese T, Berthold F, Kerbl R, Mann J, Parker L, Schilling F, Ambros P, Christiansen H, Favrot M, Kabisch H, Hero B, Philip T. 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) Eur J Cancer. 1998;34:1391–1397. doi: 10.1016/s0959-8049(98)00135-x. [DOI] [PubMed] [Google Scholar]
  31. Estus S, Zaks WJ, Freeman RS, Gruda M, Bravo R, Johnson EM., Jr Altered gene expression in neurons during programmed cell death: identification of c-jun as necessary for neuronal apoptosis. J Cell Biol. 1994;127:1717–1727. doi: 10.1083/jcb.127.6.1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Evans AE, D'Angio GJ, Randolph J. A proposed staging for children with neuroblastoma. Child Cancer Stud Group A. 1971;27:374–378. doi: 10.1002/1097-0142(197102)27:2<374::aid-cncr2820270221>3.0.co;2-g. [DOI] [PubMed] [Google Scholar]
  33. Evans AE, Kisselbach KD, Liu X, Eggert A, Ikegaki N, Camoratto AM, Dionne C, Brodeur GM. Effect of CEP-751 (KT-6587) on neuroblastoma xenografts expressing TrkB. Med Pediat Oncol. 2001;36:181–184. doi: 10.1002/1096-911X(20010101)36:1<181::AID-MPO1043>3.0.CO;2-Q. [DOI] [PubMed] [Google Scholar]
  34. Evans AE, Kisselbach KD, Yamashiro DJ, Ikegaki N, Camoratto AM, Dionne CA, Brodeur GM. Antitumor activity of CEP-751 (KT-6587) on human neuroblastoma and medulloblastoma xenografts. Clin Cancer Res. 1999;5:3594–3602. [PubMed] [Google Scholar]
  35. Everson TC. Spontaneous regression of cancer. Ann N Y Acad Sci. 1964;114:721–735. [PubMed] [Google Scholar]
  36. Everson TC, Cole WH. Spontaneous regression of cancer. Saunders; Philadelphia: 1966. [Google Scholar]
  37. Fisher JP, Tweddle DA. Neonatal neuroblastoma. Sem Fetal Neon Med. 2012;17:207–215. doi: 10.1016/j.siny.2012.05.002. [DOI] [PubMed] [Google Scholar]
  38. Garvin JH, Jr, Lack EE, Berenberg W, Frantz CN. Ganglioneuroma presenting with differentiated skeletal metastases. Rep Case Cancer. 1984;54:357–360. doi: 10.1002/1097-0142(19840715)54:2<357::aid-cncr2820540230>3.0.co;2-4. [DOI] [PubMed] [Google Scholar]
  39. George RE, Li S, Medeiros-Nancarrow C, Neuberg D, Marcus K, Shamberger RC, Pulsipher M, Grupp SA, Diller L. High-risk neuroblastoma treated with tandem autologous peripheral-blood stem cell-supported transplantation: long-term survival update. J Clin Oncol. 2006;24:2891–2896. doi: 10.1200/JCO.2006.05.6986. [DOI] [PubMed] [Google Scholar]
  40. George RE, London WB, Cohn SL, Maris JM, Kretschmar C, Diller L, Brodeur GM, Castleberry RP, Look AT. Hyperdiploidy plus nonamplified MYCN confers a favorable prognosis in children 12 to 18 months old with disseminated neuroblastoma: a pediatric oncology group study. J Clin Oncol. 2005;23:6466–6473. doi: 10.1200/JCO.2005.05.582. [DOI] [PubMed] [Google Scholar]
  41. Goldschneider D, Mehlen P. Dependence receptors: a new paradigm in cell signaling and cancer therapy. Oncogene. 2010;29:1865–1882. doi: 10.1038/onc.2010.13. [DOI] [PubMed] [Google Scholar]
  42. Grau E, Martinez F, Orellana C, Canete A, Yanez Y, Oltra S, Noguera R, Hernandez M, Bermudez JD, Castel V. Epigenetic alterations in disseminated neuroblastoma tumour cells: influence of TMS1 gene hypermethylation in relapse risk in NB patients. J Cancer Res Clin Oncol. 2010;136:1415–1421. doi: 10.1007/s00432-010-0796-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Gros C, Fahy J, Halby L, Dufau I, Erdmann A, Gregoire JM, Ausseil F, Vispe S, Arimondo PB. DNA methylation inhibitors in cancer: recent and future approaches. Biochimie. 2012;94:2280–2296. doi: 10.1016/j.biochi.2012.07.025. [DOI] [PubMed] [Google Scholar]
  44. Haas D, Ablin AR, Miller C, Zoger S, Matthay KK. Complete pathologic maturation and regression of stage IVS neuroblastoma without treatment. Cancer. 1988;62:818–825. doi: 10.1002/1097-0142(19880815)62:4<818::aid-cncr2820620430>3.0.co;2-k. [DOI] [PubMed] [Google Scholar]
  45. Ham J, Babij C, Whitfield J, Pfarr CM, Lallemand D, Yaniv M, Rubin LL. A c-Jun dominant negative mutant protects sympathetic neurons against programmed cell death. Neuron. 1995;14:927–939. doi: 10.1016/0896-6273(95)90331-3. [DOI] [PubMed] [Google Scholar]
  46. Hantzopoulos PA, Suri C, Glass DJ, Goldfarb MP, Yancopoulos GD. The low affinity NGF receptor, p75, can collaborate with each of the Trks to potentiate functional responses to the neurotrophins. Neuron. 1994;13:187–201. doi: 10.1016/0896-6273(94)90469-3. [DOI] [PubMed] [Google Scholar]
  47. Hayashi Y, Hanada R, Yamamoto K. Biology of neuroblastomas in Japan found by screening. Am J Pediatr Hematol Oncol. 1992;14:342–347. [PubMed] [Google Scholar]
  48. Hero B, Simon T, Spitz R, Ernestus K, Gnekow AK, Scheel-Walter HG, Schwabe D, Schilling FH, Benz-Bohm G, Berthold F. Localized infant neuroblastomas often show spontaneous regression: results of the prospective trials NB95-S and NB97. J Clin Oncol. 2008;26:1504–1510. doi: 10.1200/JCO.2007.12.3349. [DOI] [PubMed] [Google Scholar]
  49. Hiyama E, Hiyama K, Yokoyama T, Matsuura Y, Piatyszek MA, Shay JW. Correlating telomerase activity levels with human neuroblastoma outcomes. Nat Med. 1995;1:249–255. doi: 10.1038/nm0395-249. [DOI] [PubMed] [Google Scholar]
  50. Ho PT, Estroff JA, Kozakewich H, Shamberger RC, Lillehei CW, Grier HE, Diller L. Prenatal detection of neuroblastoma: a ten-year experience from the Dana-Farber Cancer Institute and Children's hospital. Pediatrics. 1993;92:358–364. [PubMed] [Google Scholar]
  51. Ho R, Eggert A, Hishiki T, Minturn JE, Ikegaki N, Foster P, Camoratto AM, Evans AE, Brodeur GM. Resistance to chemotherapy mediated by TrkB in neuroblastomas. Cancer Res. 2002;62:6462–6466. [PubMed] [Google Scholar]
  52. Ho R, Minturn JE, Simpson AM, Iyer R, Light JE, Evans AE, Brodeur GM. The effect of P75 on Trk receptors in neuroblastomas. Cancer Lett. 2011;305:76–85. doi: 10.1016/j.canlet.2011.02.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Hoehner JC, Olsen L, Sandstedt B, Kaplan DR, Pahlman S. Association of neurotrophin receptor expression and differentiation in human neuroblastoma. Am J Pathol. 1995;147:102–113. [PMC free article] [PubMed] [Google Scholar]
  54. Ikeda Y, Lister J, Bouton JM, Buyukpamukcu M. Congenital neuroblastoma, neuroblastoma in situ, and the normal fetal development of the adrenal. J Pediatr Surg. 1981;16:636–644. doi: 10.1016/0022-3468(81)90019-1. [DOI] [PubMed] [Google Scholar]
  55. Iyer R, Evans AE, Qi X, Ho R, Minturn JE, Zhao H, Balamuth N, Maris JM, Brodeur GM. Lestaurtinib enhances the antitumor efficacy of chemotherapy in murine xenograft models of neuroblastoma. Clin Cancer Res. 2010;16:1478–1485. doi: 10.1158/1078-0432.CCR-09-1531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Iyer R, Varela CR, Minturn JE, Ho R, Simpson AM, Light JE, Evans AE, Zhao H, Thress K, Brown JL, Brodeur GM. AZ64 inhibits TrkB and enhances the efficacy of chemotherapy and local radiation in neuroblastoma xenografts. Cancer Chemother Pharmacol. 2012;70:477–486. doi: 10.1007/s00280-012-1879-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Iyer R, Wehrmann L, Golden RL, Naraparaju K, Croucher JL, MacFarland SP, Guan P, Kolla V, Wei G, Cam N, Li G, Hornby Z, Brodeur GM. Entrectinib is a potent inhibitor of Trk-driven neuroblastomas in a xenograft mouse model. Cancer Lett. 2016;372:179–186. doi: 10.1016/j.canlet.2016.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Jaboin J, Kim CJ, Kaplan DR, Thiele CJ. Brain-derived neurotrophic factor activation of TrkB protects neuroblastoma cells from chemotherapy-induced apoptosis via phosphatidylinositol 3′-kinase pathway. Cancer Res. 2002;62:6756–6763. [PubMed] [Google Scholar]
  59. Kahane N, Kalcheim C. Expression of trkC receptor mRNA during development of the avian nervous system. J Neurobiol. 1994;25:571–584. doi: 10.1002/neu.480250509. [DOI] [PubMed] [Google Scholar]
  60. Kaneko Y, Kanda N, Maseki N, Nakachi K, Takeda T, Okabe I, Sakurai M. Current urinary mass screening for catecholamine metabolites at 6 months of age may be detecting only a small portion of high-risk neuroblastomas: a chromosome and N-myc amplification study. J Clin Oncol. 1990;8:2005–2013. doi: 10.1200/JCO.1990.8.12.2005. [DOI] [PubMed] [Google Scholar]
  61. Kataoka Y, Matsumura T, Yamamoto S, Sugimoto T, Sawada T. Distinct cytotoxicity against neuroblastoma cells of peripheral blood and tumor-infiltrating lymphocytes from patients with neuroblastoma. Cancer Lett. 1993;73:11–21. doi: 10.1016/0304-3835(93)90182-9. [DOI] [PubMed] [Google Scholar]
  62. Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, Coviello GM, Wright WE, Weinrich SL, Shay JW. Specific association of human telomerase activity with immortal cells and cancer. Science. 1994;266:2011–2015. doi: 10.1126/science.7605428. [DOI] [PubMed] [Google Scholar]
  63. Kogner P, Barbany G, Dominici C, Castello MA, Raschella G, Persson H. Coexpression of messenger RNA for TRK protooncogene and low affinity nerve growth factor receptor in neuroblastoma with favorable prognosis. Cancer Res. 1993;53:2044–2050. [PubMed] [Google Scholar]
  64. Krams M, Hero B, Berthold F, Parwaresch R, Harms D, Rudolph P. Full-length telomerase reverse transcriptase messenger RNA is an independent prognostic factor in neuroblastoma. Am J Pathol. 2003;162:1019–1026. doi: 10.1016/S0002-9440(10)63896-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Kushner BH, Cheung NK, LaQuaglia MP, Ambros PF, Ambros IM, Bonilla MA, Gerald WL, Ladanyi M, Gilbert F, Rosenfield NS, Yeh SD. Survival from locally invasive or widespread neuroblastoma without cytotoxic therapy. J Clin Oncol. 1996;14:373–381. doi: 10.1200/JCO.1996.14.2.373. [DOI] [PubMed] [Google Scholar]
  66. Lavarino C, Cheung NK, Garcia I, Domenech G, de Torres C, Alaminos M, Rios J, Gerald WL, Kushner B, LaQuaglia M, Mora J. Specific gene expression profiles and chromosomal abnormalities are associated with infant disseminated neuroblastoma. BMC Cancer. 2009;9:44. doi: 10.1186/1471-2407-9-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. London WB, Castleberry RP, Matthay KK, Look AT, Seeger RC, Shimada H, Thorner P, Brodeur G, Maris JM, Reynolds CP, Cohn SL. Evidence for an age cutoff greater than 365 days for neuroblastoma risk group stratification in the Children's oncology group. J Clin Oncol. 2005;23:6459–6465. doi: 10.1200/JCO.2005.05.571. [DOI] [PubMed] [Google Scholar]
  68. Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, Rossig C, Russell HV, Diouf O, Liu E, Liu H, Wu MF, Gee AP, Mei Z, Rooney CM, Heslop HE, Brenner MK. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118:6050–6056. doi: 10.1182/blood-2011-05-354449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Matsumoto K, Wada RK, Yamashiro JM, Kaplan DR, Thiele CJ. Expression of brain-derived neurotrophic factor and p145TrkB affects survival, differentiation, and invasiveness of human neuroblastoma cells. Cancer Res. 1995;55:1798–1806. [PubMed] [Google Scholar]
  70. Matthay KK. Stage 4S neuroblastoma: what makes it special? J Clin Oncol. 1998;16:2003–2006. doi: 10.1200/JCO.1998.16.6.2003. [DOI] [PubMed] [Google Scholar]
  71. Matthay KK, Villablanca JG, Seeger RC, Stram DO, Harris RE, Ramsay NK, Swift P, Shimada H, Black CT, Brodeur GM, Gerbing RB, Reynolds CP. Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. Child Cancer Group New Eng J Med. 1999;341:1165–1173. doi: 10.1056/NEJM199910143411601. [DOI] [PubMed] [Google Scholar]
  72. McCabe MT, Creasy CL. EZH2 as a potential target in cancer therapy. Epigenomics. 2014;6:341–351. doi: 10.2217/epi.14.23. [DOI] [PubMed] [Google Scholar]
  73. Minturn JE, Evans AE, Villablanca JG, Yanik GA, Park JR, Shusterman S, Groshen S, Hellriegel ET, Bensen-Kennedy D, Matthay KK, Brodeur GM, Maris JM. Phase I trial of lestaurtinib for children with refractory neuroblastoma: a new approaches to neuroblastoma therapy consortium study. Cancer Chemother Pharmacol. 2011;68:1057–1065. doi: 10.1007/s00280-011-1581-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Monclair T, Brodeur GM, Ambros PF, Brisse HJ, Cecchetto G, Holmes K, Kaneko M, London WB, Matthay KK, Nuchtern JG, von Schweinitz D, Simon T, Cohn SL, Pearson AD, Force IT. The international neuroblastoma risk group (INRG) staging system: an INRG task force report. J Clin Oncol. 2009;27:298–303. doi: 10.1200/JCO.2008.16.6876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Nagasubramanian R, Wei J, Gordon P, Rastatter JC, Cox MC, Pappo A. Infantile Fibrosarcoma with NTRK3-ETV6 fusion successfully treated with the tropomyosin-related kinase inhibitor LOXO-101. Pediatr Blood Cancer. 2016;63:1468–1470. doi: 10.1002/pbc.26026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Nakagawara A. Molecular basis of spontaneous regression of neuroblastoma: role of neurotrophic signals and genetic abnormalities. Hum Cell. 1998;11:115–124. [PubMed] [Google Scholar]
  77. Nakagawara A, Arima M, Azar CG, Scavarda NJ, Brodeur GM. Inverse relationship between trk expression and N-myc amplification in human neuroblastomas. Cancer Res. 1992;52:1364–1368. [PubMed] [Google Scholar]
  78. Nakagawara A, Arima-Nakagawara M, Scavarda NJ, Azar CG, Cantor AB, Brodeur GM. Association between high levels of expression of the TRK gene and favorable outcome in human neuroblastoma. N Engl J Med. 1993;328:847–854. doi: 10.1056/NEJM199303253281205. [DOI] [PubMed] [Google Scholar]
  79. Nakagawara A, Azar CG, Scavarda NJ, Brodeur GM. Expression and function of TRK-B and BDNF in human neuroblastomas. Mol Cell Biol. 1994;14:759–767. doi: 10.1128/mcb.14.1.759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Nakagawara A, Brodeur GM. Role of neurotrophins and their receptors in human neuroblastomas: a primary culture study. Eur J Cancer. 1997;33:2050–2053. doi: 10.1016/s0959-8049(97)00280-3. [DOI] [PubMed] [Google Scholar]
  81. Nakamura K, Martin KC, Jackson JK, Beppu K, Woo CW, Thiele CJ. Brain-derived neurotrophic factor activation of TrkB induces vascular endothelial growth factor expression via hypoxia-inducible factor-1alpha in neuroblastoma cells. Cancer Res. 2006;66:4249–4255. doi: 10.1158/0008-5472.CAN-05-2789. [DOI] [PubMed] [Google Scholar]
  82. Nickerson HJ, Matthay KK, Seeger RC, Brodeur GM, Shimada H, Perez C, Atkinson JB, Selch M, Gerbing RB, Stram DO, Lukens J. Favorable biology and outcome of stage IV-S neuroblastoma with supportive care or minimal therapy: a Children's cancer group study. J Clin Oncol. 2000;18:477–486. doi: 10.1200/JCO.2000.18.3.477. [DOI] [PubMed] [Google Scholar]
  83. Nikoletopoulou V, Lickert H, Frade JM, Rencurel C, Giallonardo P, Zhang L, Bibel M, Barde YA. Neurotrophin receptors TrkA and TrkC cause neuronal death whereas TrkB does not. Nature. 2010;467:59–63. doi: 10.1038/nature09336. [DOI] [PubMed] [Google Scholar]
  84. Nuchtern JG, London WB, Barnewolt CE, Naranjo A, McGrady PW, Geiger JD, Diller L, Schmidt ML, Maris JM, Cohn SL, Shamberger RC. A prospective study of expectant observation as primary therapy for neuroblastoma in young infants: a Children's oncology group study. Ann Surg. 2012;256:573–580. doi: 10.1097/SLA.0b013e31826cbbbd. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Ohali A, Avigad S, Ash S, Goshen Y, Luria D, Feinmesser M, Zaizov R, Yaniv I. Telomere length is a prognostic factor in neuroblastoma. Cancer. 2006;107:1391–1399. doi: 10.1002/cncr.22132. [DOI] [PubMed] [Google Scholar]
  86. Pachnis V, Mankoo B, Costantini F. Expression of the c-ret proto-oncogene during mouse embryogenesis. Development. 1993;119:1005–1017. doi: 10.1242/dev.119.4.1005. [DOI] [PubMed] [Google Scholar]
  87. Papac RJ. Spontaneous regression of cancer: possible mechanisms. In vivo. 1998;12:571–578. [PubMed] [Google Scholar]
  88. Peifer M, Hertwig F, Roels F, Dreidax D, Gartlgruber M, Menon R, Kramer A, Roncaioli JL, Sand F, Heuckmann JM, Ikram F, Schmidt R, Ackermann S, Engesser A, Kahlert Y, Vogel W, Altmuller J, Nurnberg P, Thierry-Mieg J, Thierry-Mieg D, Mariappan A, Heynck S, Mariotti E, Henrich KO, Gloeckner C, Bosco G, Leuschner I, Schweiger MR, Savelyeva L, Watkins SC, Shao C, Bell E, Hofer T, Achter V, Lang U, Theissen J, Volland R, Saadati M, Eggert A, de Wilde B, Berthold F, Peng Z, Zhao C, Shi L, Ortmann M, Buttner R, Perner S, Hero B, Schramm A, Schulte JH, Herrmann C, O'Sullivan RJ, Westermann F, Thomas RK, Fischer M. Telomerase activation by genomic rearrangements in high-risk neuroblastoma. Nature. 2015;526:700–704. doi: 10.1038/nature14980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Pranzatelli MR, Travelstead AL, Tate ED, Allison TJ, Moticka EJ, Franz DN, Nigro MA, Parke JT, Stumpf DA, Verhulst SJ. B- and T-cell markers in opsoclonus-myocl onus syndrome: immunophenotyping of CSF lymphocytes. Neurology. 2004;62:1526–1532. doi: 10.1212/wnl.62.9.1526. [DOI] [PubMed] [Google Scholar]
  90. Pritchard J, Hickman JA. Why does stage 4s neuroblastoma regress spontaneously? Lancet. 1994;344:869–870. doi: 10.1016/s0140-6736(94)92834-7. [DOI] [PubMed] [Google Scholar]
  91. Rabizadeh S, Ye X, Wang JJ, Bredesen DE. Neurotrophin dependence mediated by p75NTR: contrast between rescue by BDNF and NGF. Cell Death Dif. 1999;6:1222–1227. doi: 10.1038/sj.cdd.4400614. [DOI] [PubMed] [Google Scholar]
  92. Raffaghello L, Prigione I, Bocca P, Morandi F, Camoriano M, Gambini C, Wang X, Ferrone S, Pistoia V. Multiple defects of the antigen-processing machinery components in human neuroblastoma: immunotherapeutic implications. Oncogene. 2005;24:4634–4644. doi: 10.1038/sj.onc.1208594. [DOI] [PubMed] [Google Scholar]
  93. Rodriguez-Tebar A, Rohrer H. Retinoic acid induces NGF-dependent survival response and high-affinity NGF receptors in immature chick sympathetic neurons. Development. 1991;112:813–820. doi: 10.1242/dev.112.3.813. [DOI] [PubMed] [Google Scholar]
  94. Rudnick E, Khakoo Y, Antunes NL, Seeger RC, Brodeur GM, Shimada H, Gerbing RB, Stram DO, Matthay KK. Opsoclonus-myoclonus-ataxia syndrome in neuroblastoma: clinical outcome and antineuronal antibodies-a report from the Children's cancer group study. Med Pediatr Oncol. 2001;36:612–622. doi: 10.1002/mpo.1138. [DOI] [PubMed] [Google Scholar]
  95. Russo C, Cohn SL, Petruzzi MJ, de Alarcon PA. Long-term neurologic outcome in children with opsoclonus-myoclonus associated with neuroblastoma: a report from the pediatric oncology group. Med Pediatr Oncol. 1997;28:284–288. doi: 10.1002/(sici)1096-911x(199704)28:4<284::aid-mpo7>3.0.co;2-e. [DOI] [PubMed] [Google Scholar]
  96. Samy M, Gattolliat CH, Pendino F, Hillion J, Nguyen E, Bombard S, Douc-Rasy S, Benard J, Segal-Bendirdjian E. Loss of the malignant phenotype of human neuroblastoma cells by a catalytically inactive dominant-negative hTERT mutant. Mol Cancer Ther. 2012;11:2384–2393. doi: 10.1158/1535-7163.MCT-12-0281. [DOI] [PubMed] [Google Scholar]
  97. Sawada T, Kidowaki T, Sakamoto I, Hashida T, Matsumura T, Nakagawa M, Kusunoki T. Neuroblastoma. Mass Screen Detect Prognos Cancer. 1984;53:2731–2735. doi: 10.1002/1097-0142(19840615)53:12<2731::aid-cncr2820531232>3.0.co;2-d. [DOI] [PubMed] [Google Scholar]
  98. Saylors RL, 3rd, Cohn SL, Morgan ER, Brodeur GM. Prenatal detection of neuroblastoma by fetal ultrasonography. Am J Pediat Hematol/Oncol. 1994;16:356–360. [PubMed] [Google Scholar]
  99. Schilling FH, Spix C, Berthold F, Erttmann R, Fehse N, Hero B, Klein G, Sander J, Schwarz K, Treuner J, Zorn U, Michaelis J. Neuroblastoma screening at one year of age. N Engl J Med. 2002;346:1047–1053. doi: 10.1056/NEJMoa012277. [DOI] [PubMed] [Google Scholar]
  100. Shimada H, Ambros IM, Dehner LP, Hata J, Joshi VV, Roald B. Terminology and morphologic criteria of neuroblastic tumors: recommendations by the international neuroblastoma pathology committee. Cancer. 1999a;86:349–363. [PubMed] [Google Scholar]
  101. Shimada H, Ambros IM, Dehner LP, Hata J, Joshi VV, Roald B, Stram DO, Gerbing RB, Lukens JN, Matthay KK, Castleberry RP. The international neuroblastoma pathology classification (the Shimada system) Cancer. 1999b;86:364–372. [PubMed] [Google Scholar]
  102. Squire R, Fowler CL, Brooks SP, Rich GA, Cooney DR. The relationship of class I MHC antigen expression to stage IV-S disease and survival in neuroblastoma. J Pediatr Surg. 1990;25:381–386. doi: 10.1016/0022-3468(90)90375-j. [DOI] [PubMed] [Google Scholar]
  103. Streutker CJ, Thorner P, Fabricius N, Weitzman S, Zielenska M. Telomerase activity as a prognostic factor in neuroblastomas. Pediatr Dev Pathol. 2001;4:62–67. doi: 10.1007/s100240010108. [DOI] [PubMed] [Google Scholar]
  104. Suzuki T, Bogenmann E, Shimada H, Stram D, Seeger RC. Lack of high-affinity nerve growth factor receptors in aggressive neuroblastomas. J Natl Cancer Inst. 1993;85:377–384. doi: 10.1093/jnci/85.5.377. [DOI] [PubMed] [Google Scholar]
  105. Tacconelli A, Farina AR, Cappabianca L, Desantis G, Tessitore A, Vetuschi A, Sferra R, Rucci N, Argenti B, Screpanti I, Gulino A, Mackay AR. TrkA alternative splicing: a regulated tumor-promoting switch in human neuroblastoma. Cancer Cell. 2004;6:347–360. doi: 10.1016/j.ccr.2004.09.011. [DOI] [PubMed] [Google Scholar]
  106. Tacconelli A, Farina AR, Cappabianca L, Gulino A, Mackay AR. Alternative TrkAIII splicing: a potential regulated tumor-promoting switch and therapeutic target in neuroblastoma. Future Oncol. 2005;1:689–698. doi: 10.2217/14796694.1.5.689. [DOI] [PubMed] [Google Scholar]
  107. Taggart DR, London WB, Schmidt ML, DuBois SG, Monclair TF, Nakagawara A, De Bernardi B, Ambros PF, Pearson AD, Cohn SL, Matthay KK. Prognostic value of the stage 4S metastatic pattern and tumor biology in patients with metastatic neuroblastoma diagnosed between birth and 18 months of age. J Clin Oncol. 2011;29:4358–4364. doi: 10.1200/JCO.2011.35.9570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Takita J, Yang HW, Bessho F, Hanada R, Yamamoto K, Kidd V, Teitz T, Wei T, Hayashi Y. Absent or reduced expression of the caspase 8 gene occurs frequently in neuroblastoma, but not commonly in Ewing sarcoma or rhabdomyosarcoma. Med Pediatr Oncol. 2000;35:541–543. doi: 10.1002/1096-911x(20001201)35:6<541::aid-mpo9>3.0.co;2-t. [DOI] [PubMed] [Google Scholar]
  109. Thiele CJ, Li Z, McKee AE. On Trk–the TrkB signal transduction pathway is an increasingly important target in cancer biology. Clin Cancer Res. 2009;15:5962–5967. doi: 10.1158/1078-0432.CCR-08-0651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Tsuzuki T, Takahashi M, Asai N, Iwashita T, Matsuyama M, Asai J. Spatial and temporal expression of the ret proto-oncogene product in embryonic, infant and adult rat tissues. Oncogene. 1995;10:191–198. [PubMed] [Google Scholar]
  111. Turkel SB, Itabashi HH. The natural history of neuroblastic cells in the fetal adrenal gland. Am J Pathol. 1974;76:225–244. [PMC free article] [PubMed] [Google Scholar]
  112. Valteau D, Scott V, Carcelain G, Hartmann O, Escudier B, Hercend T, Triebel F. T-cell receptor repertoire in neuroblastoma patients. Cancer Res. 1996;56:362–369. [PubMed] [Google Scholar]
  113. Woods WG, Gao RN, Shuster JJ, Robison LL, Bernstein M, Weitzman S, Bunin G, Levy I, Brossard J, Dougherty G, Tuchman M, Lemieux B. Screening of infants and mortality due to neuroblastoma. N Engl J Med. 2002;346:1041–1046. doi: 10.1056/NEJMoa012387. [DOI] [PubMed] [Google Scholar]
  114. Woods WG, Tuchman M, Robison LL, Bernstein M, Leclerc J-M, Brisson LC, Brossard J, Hill G, Shuster J, Luepker R, Weitzman S, Bunin G, Lemieux B. A population-based study of the usefulness of screening for neuroblastoma. Lancet. 1996;348:1682–1687. doi: 10.1016/S0140-6736(96)06020-5. [DOI] [PubMed] [Google Scholar]
  115. Yamamoto K, Ohta S, Ito E, Hayashi Y, Asami T, Mabuchi O, Higashigawa M, Tanimura M. Marginal decrease in mortality and marked increase in incidence as a result of neuroblastoma screening at 6 months of age: cohort study in seven prefectures in Japan. J Clin Oncol. 2002;20:1209–1214. doi: 10.1200/JCO.2002.20.5.1209. [DOI] [PubMed] [Google Scholar]
  116. Yang Q, Kiernan CM, Tian Y, Salwen HR, Chlenski A, Brumback BA, London WB, Cohn SL. Methylation of CASP8, DCR2, and HIN-1 in neuroblastoma is associated with poor outcome. Clin Cancer Res. 2007;13:3191–3197. doi: 10.1158/1078-0432.CCR-06-2846. [DOI] [PubMed] [Google Scholar]
  117. Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, Smith M, Anderson B, Villablanca JG, Matthay KK, Shimada H, Grupp SA, Seeger R, Reynolds CP, Buxton A, Reisfeld RA, Gillies SD, Cohn SL, Maris JM, Sondel PM, Children's Oncology G Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 2010;363:1324–1334. doi: 10.1056/NEJMoa0911123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Yu F, Zhu X, Feng C, Wang T, Hong Q, Liu Z, Tang S. Proteomics-based identification of spontaneous regression-associated proteins in neuroblastoma. J Pediatr Surg. 2011;46:1948–1955. doi: 10.1016/j.jpedsurg.2011.06.024. [DOI] [PubMed] [Google Scholar]
  119. Yuza Y, Agawa M, Matsuzaki M, Yamada H, Urashima M. Gene and protein expression profiling during differentiation of neuroblastoma cells triggered by 13-cis retinoic acid. J Pediatr Hematol Oncol. 2003;25:715–720. doi: 10.1097/00043426-200309000-00008. [DOI] [PubMed] [Google Scholar]

RESOURCES