ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Epidemiology, pathogenesis, and pathology of neuroblastoma

Epidemiology, pathogenesis, and pathology of neuroblastoma
Literature review current through: Jan 2024.
This topic last updated: Apr 01, 2022.

INTRODUCTION — The term neuroblastoma is commonly used to refer to a spectrum of neuroblastic tumors (including neuroblastomas, ganglioneuroblastomas, and ganglioneuromas) that arise from primitive sympathetic ganglion cells. The neuroectodermal cells that comprise neuroblastic tumors originate from the neural crest during fetal development, and are destined for the adrenal medulla and sympathetic nervous system. By contrast, pheochromocytomas and paragangliomas arise from a different type of cell, the chromaffin cell, which also migrates from the neural crest to the adrenal gland [1-3]. Together, both types of cells make up the adrenal medulla, a component of the sympathetic nervous system. (See "Pheochromocytoma and paraganglioma in children" and "Paragangliomas: Epidemiology, clinical presentation, diagnosis, and histology".)

Neuroblastomas, which account for 97 percent of all neuroblastic tumors, are heterogeneous, varying in terms of location, histopathologic appearance, and biologic characteristics [4]. They are most remarkable for their broad spectrum of clinical behavior, which can range from spontaneous regression, to maturation to a benign ganglioneuroma, or aggressive disease with metastatic dissemination leading to death [5]. Clinical diversity correlates closely with numerous clinical and biological factors (including patient age, tumor stage and histology, and genetic and chromosomal abnormalities), although its molecular basis remains largely unknown. For example, most infants with disseminated disease have a favorable outcome following treatment with chemotherapy and surgery, while the majority of children over the age of 18 months with advanced-stage disease die from progressive disease despite intensive multimodality therapy.

The epidemiology, embryogenesis, molecular pathogenesis, and pathology of neuroblastoma will be presented here. The clinical presentation, diagnosis, evaluation, treatment, and prognosis of neuroblastoma are presented separately. Neuroblastomas arising in the olfactory epithelium, which have a different cell of origin, presentation, and treatment than neuroblastoma, also are discussed separately.

(See "Clinical presentation, diagnosis, and staging evaluation of neuroblastoma".)

(See "Treatment and prognosis of neuroblastoma".)

(See "Olfactory neuroblastoma (esthesioneuroblastoma)".)

EPIDEMIOLOGY — Neuroblastoma is almost exclusively a disease of children. It is the third most common childhood cancer, after leukemia and brain tumors, and is the most common solid extracranial tumor in children. More than 600 cases are diagnosed in the United States each year [4], and neuroblastoma accounts for approximately 15 percent of all pediatric cancer fatalities.

Incidence rates are age-dependent (figure 1). The median age at diagnosis is 17.3 months, and 40 percent of patients are diagnosed before one year of age [4,5]. Neuroblastomas are the most common extracranial solid malignant tumor diagnosed during the first two years of life, and the most common cancer among infants younger than 12 months, in whom the incidence rate is almost twice that of leukemia (58 versus 37 per one million infants) [6]. The incidence of neuroblastoma is greater among White than Black infants (ratio of 1.7 and 1.9 to 1 for males and females, respectively), but little if any racial difference is apparent among older children [4]. Neuroblastoma is slightly more common among boys compared with girls [4].

RISK FACTORS — Little is known about the etiology of sympathetic nervous system tumors. The early age of onset suggests that preconceptual or gestational environmental events (eg, exposure to drugs, hormones, toxins, or viruses) may play a role.

Maternal and fetal factors — Studies have suggested a number of maternal factors that may be associated with the subsequent development of neuroblastoma. These include the following:

Opiate consumption – At least one case control study has linked maternal consumption of opiates (particularly codeine, odds ratio 3.4) while pregnant or nursing with an increased risk of neuroblastoma in children [7].

Folate deficiency – Maternal folate consumption has been associated with a decreased risk of neuroblastoma. In a population-based study investigating the effect of fortification of flour with folic acid to prevent neural tube defects, the incidence of neuroblastoma declined from 1.6 to 0.6 cases per 10,000 births before and after fortification, respectively [8]. This finding was consistent with other studies suggesting an association between maternal vitamin use and decreased risk of neuroblastoma [9,10].

Toxic exposures – Epidemiologic studies provide little convincing evidence for toxic or infectious environmental exposure as an etiologic factor for the development of neuroblastoma [4,11-21]. However, given the identification of maternal folate deficiency as a risk factor, it is possible that case control studies in a folate deficient population might uncover an environmental factor.

Congenital abnormalities – An association between the presence of major congenital abnormalities and the subsequent development of neuroblastoma has been reported in some [10,22-26], but not all [16,27-29], studies.

Size for gestational age – A population-based case control study of 357 patients with neuroblastoma found an association between small or large for gestational age and increased neuroblastoma risk [10].

Gestational diabetes mellitus – A case control study of 240 children with neuroblastoma showed a correlation with the presence of maternal gestational diabetes mellitus [25]. The effect was greatest in those children diagnosed prior to one year of age.

Genetic factors — The majority of neuroblastomas are sporadic and not correlated with any specific constitutional germline chromosomal abnormality, inherited predisposition, or associated congenital anomalies. However, there are at least some exceptions as follows:

Single-nucleotide polymorphisms at LIM domain only 1 (LMO1) have been observed in approximately 12 percent of patients with neuroblastoma, with evidence suggesting that these genetic variations play a causal role in neuroblastoma tumorigenesis [30,31].

Another study examining genetic determinants of pediatric cancer did not observe LMO1 mutations, but reported germline mutations in succinate dehydrogenase complex, subunit B (SDHB), adenomatous polyposis coli (APC), anaplastic lymphoma kinase (ALK), and breast cancer susceptibility gene 2 (BRCA2) in 1 out of 100 patients with neuroblastoma [32].

A higher incidence of neuroblastoma has been suggested in girls with Turner syndrome [33]. (See "Clinical manifestations and diagnosis of Turner syndrome".)

Somatic and germline mutations in the paired-like homeobox 2B (PHOX2B) gene are in both sporadic and familial neuroblastoma [34-37]. Germline PHOX2B mutations are also observed in congenital central hypoventilation syndrome and in some cases of congenital aganglionic megacolon (Hirschsprung disease) [35,38]. (See "Disorders of ventilatory control", section on 'Congenital central hypoventilation syndrome' and "Congenital aganglionic megacolon (Hirschsprung disease)" and "Congenital central hypoventilation syndrome and other causes of sleep-related hypoventilation in children", section on 'Congenital central hypoventilation syndrome'.)

Familial neuroblastoma — Although the majority of neuroblastomas are sporadic, in 1 to 2 percent of cases, there can be a familial (ie, inherited) cases of neuroblastoma [39-41]. Inherited cases usually present at an earlier age than sporadic cases (mean age 9 versus 17 months), and a large proportion have bilateral adrenal or multifocal disease.

Most cases of familial neuroblastoma are due to germline mutations in the ALK receptor tyrosine kinase [42,43]. These cases appear to be inherited in an autosomal dominant pattern with incomplete penetrance and a broad spectrum of clinical behavior [44]. In addition, germline PHOX2B mutations are noted in approximately 6 percent of familial cases [34-36]. (See 'Genetic factors' above.)

Some reports suggest that familial predisposition may be conferred through disruption of a locus at 16p12-13 [45,46]. Others have identified rare germline variants at the TP53 locus (17p13.1) that are associated with neuroblastoma susceptibility [47]. Germline mutations in TP53 are the cause of Li-Fraumeni syndrome, and individuals who inherit these mutations are at increased risk of developing a wide variety of cancers at an early age. (See "Li-Fraumeni syndrome".)

Implications for siblings and future offspring — In families without a history of multiple affected individuals, it is unlikely that a sibling of a patient with neuroblastoma will also be affected. The risk for children of survivors of neuroblastoma is difficult to determine because of the small number of cases and treatment-related infertility. (See "Overview of infertility and pregnancy outcome in cancer survivors".)

PATHOGENESIS

Embryology — Neuroblastoma arises from early neural crest precursors that undergo transformation secondary to genetic or epigenetic events that lead to blocked or aberrant developmental differentiation [48]. The neural crest is a transient multipotent embryologic tissue which migrates out of the neural chord during development. Neuroblasts undergo an epithelial to mesenchymal transition and migrate both ventrally and caudally to form components of many tissues including the branchial arches, cardiac and thoracic vessels, and the sympathetic nervous system, which includes the adrenal glands.

While a great deal has been discovered about the genetic and transcriptional regulation of neural crest development over the past decade [49-51], the events that induce neuroblastoma tumorigenesis remain poorly defined. It is clear, however, that multiple different changes can induce tumor formation and the timing and character of driving oncogenic events may well define the phenotype of the resulting cancer (figure 2). As an example, cases of localized neuroblastoma arising under the age of one year likely represent tumors that develop at a different stage of neural crest differentiation than does high-stage disease [52,53]. Spontaneous regression of neuroblastomas found in newborn infants through screening likely occurs due to differentiation or age-dependent changes in growth factors in these infants. (See "Clinical presentation, diagnosis, and staging evaluation of neuroblastoma", section on 'Is there a role for neuroblastoma screening?'.)

Placing the oncogenic factors driving neuroblastoma in the context of neural crest development helps to explain the heterogeneity of this complex tumor type (figure 2). The pathology of neuroblastoma also varies considerably between patients, with age, location of tumor, and host/tumor immune interactions likely playing major roles in biology behavior. (See 'Pathology' below.)

Molecular abnormalities (prognostic impact) — Various molecular and cytogenetic factors have been implicated in the pathogenesis of neuroblastoma [54-57]. The molecular and cytogenetic characterization of neuroblastomas is a routine part of the clinical evaluation because of the influence of these findings on clinical outcome. The selection of treatment based upon molecular and genetic factors (ie, risk-adapted therapy) is described separately. (See "Treatment and prognosis of neuroblastoma".)

While an extensive discussion of the oncogenic drivers, mutations, and cytogenetic alterations found in neuroblastoma is beyond the scope of this review [48,58], we detail some of the most well-characterized factors here.

Segmental chromosome abberrations — Chromosomal deletions (as detected by loss of heterozygosity [LOH]) and segmental chromosome aberrations (SCA) are found in approximately 50 percent of neuroblastomas, localized to chromosomes 1p, 11q, and 14q [59-63], among others [64-66].

Deletion of a part of chromosome 1p is one of the most common chromosomal changes observed in neuroblastoma and is associated with a poor prognosis [67-70]. As an example, in a study of cytogenetic factors in 89 neuroblastomas among patients with stage 1, 2, or 4S disease, mean three-year event-free survival was greater among those without than with allelic loss of chromosome 1p (100 versus 34 percent for stage 1, 2, or 4S disease and 53 versus 0 percent for stage 3 or 4 disease) [70]. The staging used in neuroblastoma is the International Neuroblastoma Risk Group (INRG) Staging System (table 1) [71,72]. However, previous staging systems were the basis of some earlier studies cited in this topic and have been included where relevant. (See "Clinical presentation, diagnosis, and staging evaluation of neuroblastoma".)

MYCN status — Deletions of 1p are highly associated with amplification (increased copy number) and overexpression of the oncogene MYCN (also called N-myc), a close relative of the oncogene c-myc that resides on chromosome 2p24-25 [67,73]. Gene overexpression results in persistently high levels of the MYCN protein, a DNA binding transcription factor known to cause malignant transformation in both in vitro and in vivo tumor models [74,75]. Additionally, a subgroup of patients with neuroblastoma overexpress a separate gene FOXR2, which stabilizes and increases MYCN protein levels [76]. A 50- to 400-fold amplification of MYCN is found in approximately 25 percent of neuroblastomas and is an indicator of poor prognosis [77-83].

The prognostic significance of MYCN amplification can be illustrated by the following data:

In one study of 2660 patients from the INRG database with stage 1 or stage 2 neuroblastoma, according to an earlier staging system (table 2), patients with MYCN-amplified tumors had significantly worse event-free and overall survival (53 versus 90, and 72 versus 98 percent, respectively) compared with those without MYCN amplification [82].

A similar impact of MYCN amplification was observed in a study of 110 infants with stage 4S neuroblastoma in whom survival was significantly worse for those with MYCN amplification compared with those without amplification (<50 versus >90 percent) [81].

In contrast, among children with stage 4 neuroblastoma without amplification of MYCN, prognosis depends upon age [84]. In a study from the Children's Cancer Group, the six-year event-free survival rates for those under 12 months, 12 to 18 months, 18 to 24 months, and over 24 months was 92, 74, 31, and 23 percent, respectively.

The absence of MYCN amplification and the absence of other structural abnormalities, such as in 11q or 17q, can define low-risk tumors [71]. Deletions of 11q and/or 14q are detected in 25 to 50 percent of neuroblastomas [62,63,71]. Neuroblastomas that are characterized by these changes generally lack 1p deletions and MYCN amplification, and they appear to represent a distinct tumor subtype [85].

A gain of chromosome 17q material (trisomy 17q) occurs in over one-half of neuroblastomas and appears to be associated with a particularly aggressive phenotype [86,87]. As an example, in one report, overall survival was significantly worse in children with trisomy 17q compared with those whose tumors had a normal 17q number (31 versus 86 percent) [86]. In contrast, in another report, whole chromosome 17 gain was associated with increased survival [87].

Alterations in total DNA content — In additional to structural chromosomal changes, alterations in total DNA content, which presumably result from mitotic dysfunction, are an important indicator of both outcome and response to therapy. Neuroblastomas with a higher DNA content (hyperdiploid, with a DNA index [DI] >1) are associated with lower tumor stage, better response to initial therapy, and an overall better prognosis than diploid tumors (ie, DI = 1), particularly if they lack MYCN amplification [88-95]. As an example, in one study, the two-year disease free-survival rate was 94 percent for patients with near-triploid neuroblastoma compared with 45 percent for patients with diploid tumors without MYCN amplification, and 11 percent for patients with diploid or near-diploid tumors and MYCN amplification [92]. The influence of ploidy on outcome of neuroblastoma seems to be lost in children over the age of two, possibly because hyperdiploid tumors in older children typically have a number of structural rearrangements as well.

Other molecular alterations — Other factors that have not been incorporated into risk stratification schemes [66] but may affect prognosis include the following:

Neurotrophic factors – Expression of neurotrophic factors, such as nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF) along with their receptors (tyrosine kinases that are encoded by three tropomyosin receptor kinase [TRK] genes, TRK-A, B, and C) has been implicated in the pathogenesis of neuroblastoma, although their precise role is unclear [96]. Expression of TRK-A is inversely correlated with MYCN amplification, and high TRK-A and C expression appears to identify a biologically favorable subgroup of neuroblastomas, while expression of TRK-B is prognostically unfavorable [97-103].

Alterations in ALK – In contrast to adult cancers, there is a relative paucity of mutations in neuroblastomas. However, variations in the anaplastic lymphoma kinase (ALK) gene have been identified in several studies as being an important contributor to the development of both familial and sporadic neuroblastomas [42,43,58,104-106]. Somatic mutations in the tyrosine kinase region of this gene appear to have a significant role in subsequent tumor development, and ALK inhibitors are being studied as a possible therapeutic intervention. (See "Treatment and prognosis of neuroblastoma", section on 'Investigational induction therapies'.)

Telomeres – Telomeres are repeated nucleotide sequences that stabilize chromosomes, thereby preventing cell senescence. Telomerase is the enzyme that compensates for telomere shortening during cell division by synthesizing telomeric DNA, thereby maintaining telomere length. Preserved length of telomeres has been identified as a possible independent poor prognostic sign in children with neuroblastoma [107]. In addition to activation of telomerase, alternative lengthening of telomeres (ALT) has been shown to correlate with worse prognosis in neuroblastoma patients [108,109]. Specifically, telomerase activation and ALT are associated with a protracted clinical course and worse overall survival. One mechanism of ALT is associated with loss of function mutations or decreased expression of the alpha-thalassemia/mental retardation syndrome X-linked (ATRX) gene [58], which are seen in a substantial number of sporadic neuroblastomas (particularly those arising in older children) [58]. In children older than 12, the presence of ATRX mutations has been associated with a poor prognosis [110]. ATRX mutations have not been identified in any tumors with MYCN amplification.

ATRX mutations are associated with X-linked intellectual disability and alpha-thalassemia, suggesting that ATRX functions in various developmental processes; however, little is known about how ATRX contributes to the development or differentiation of the sympathoadrenal lineage. Children with X-linked intellectual disability do not have a higher incidence of neuroblastoma, suggesting that ATRX mutations alone are not sufficient to promote tumorigenesis. (See "Intellectual disability in children: Evaluation for a cause", section on 'X-linked disorders'.)

Gene expression profiling (GEP) may offer additional information to distinguish between patients with favorable and unfavorable prognoses [111-113]. This was illustrated by a study in which a 59 gene microarray expression was developed in a series of 579 patients and then validated in an independent cohort of 236 cases [111]. When clinical outcomes in the validation cohort were compared with prognosis using standard classification systems, the gene signature was an independent risk predictor, identifying patients with an increased risk of poor outcome in the current clinical risk groups.

PATHOLOGY — Neuroblastoma is a highly heterogeneous disease, and the pathology varies according to the degree of neural crest differentiation and possibly with the specific cells of origin within the neural crest. (See 'Pathogenesis' above.)

The International Neuroblastoma Pathology Classification classifies tumors of neuroblastic origin according to the balance between neural-type cells (primitive neuroblasts, maturing neuroblasts, and ganglion cells) and Schwann-type cells (Schwannian-blasts and mature Schwann cells) into one of three types: neuroblastoma, ganglioneuroblastoma, or ganglioneuroma. Neuroblastomas are the most undifferentiated-appearing and aggressive of this family of tumors, and they in turn may be classified as undifferentiated, poorly differentiated, or differentiating [114].

The degree of differentiation and stromal component of neuroblastoma tumors can be predictive of outcome and is used in the determination of Children's Oncology Group risk category for treatment (table 3):

According to this system, favorable tumors include those that are:

Poorly differentiated or differentiating neuroblastoma, with low or intermediate mitosis-karyorrhexis index (MKI), patient age ≤1.5 years

Differentiating neuroblastoma and low-MKI tumors in patients 1.5 to 5 years of age

Ganglioneuroblastoma, intermixed, regardless of age

Ganglioneuroma, regardless of age

Unfavorable tumors include those that are:

Undifferentiated or high-MKI tumors in patients of any age

Poorly differentiated and/or intermediate-MKI tumors in patients 1.5 to 5 years of age

Any grade of differentiation and any MKI class in patients ≥5 years of age

Nodular ganglioneuroblastoma, regardless of age

This topic is discussed in detail separately. (See "Treatment and prognosis of neuroblastoma", section on 'Histology'.)

Neuroblastoma — The most undifferentiated neuroblastomas are composed almost entirely of neuroblasts, with very few Schwannian (or stromal) cells. Because of the lack of Schwannian cells, these tumors are called "stroma-poor" [59]. Under light microscopy, they appear as a monotonous collection of small, round, blue cells. Morphologically, the appearance is similar to that of other small round blue cell tumors involving bone and soft tissue, including lymphoma, small cell osteosarcoma, mesenchymal chondrosarcoma, the Ewing sarcoma family of tumors, primitive neuroectodermal tumors (PNETs), and undifferentiated soft tissue sarcomas such as rhabdomyosarcoma [115]. (See "Epidemiology, pathology, and molecular genetics of Ewing sarcoma".)

Because of their morphologic similarity, these tumors are difficult to distinguish on the basis of light microscopic findings. Electron microscopy or panels of tissue-specific monoclonal antibodies can be used to help with the differentiation. Neuroblastomas typically react with antibodies that distinguish neural tissue (eg, neuron-specific enolase [NSE], synaptophysin, chromogranin, and S100). While NSE may be focally positive in other tumors (eg, rhabdomyosarcoma), the staining pattern is characteristically diffuse and strongly positive in neuroblastomas.

In contrast to the undifferentiated neuroblastomas, some evidence of neural differentiation (eg, primitive neuroblasts) can be seen in the poorly-differentiated and differentiating types of neuroblastoma. These cells are approximately 7 to 10 microns in diameter, have hyperchromatic nuclei and scanty cytoplasm, and may form Homer-Wright rosettes (picture 1). The density of the neuroblasts, rate of mitosis or MKI, and neuroblastic differentiation can vary between neuroblastomas and even within the tumor itself.

Ganglioneuroblastoma — Ganglioneuroblastoma is called an "intermixed stroma-rich" or "stroma-rich" tumor because of the increased proportion of Schwannian cells. The neuroblasts, which generally have a more mature appearance, are clustered together in foci or nests surrounded by the Schwannian cells (picture 2). These tumors generally have intermediate malignant potential, between that of neuroblastomas and ganglioneuromas.

Ganglioneuroma — Ganglioneuroma (Schwannian cell dominant) is predominantly composed of Schwannian cells studded with maturing or fully mature ganglion cells (picture 3) [114,116,117]. These tumors tend to occur in older children (five to seven years of age) rather than the more aggressive neuroblastomas. They are considered to be benign [118,119], although they can metastasize [120]. Nevertheless, the prognosis is excellent, even when complete tumor removal is not possible [121].

SUMMARY

Epidemiology – Neuroblastomas are neuroblastic tumors of children, with a median age at diagnosis of about 17 months. The causative factors are not well defined, although various genetic disorders are occasionally associated with the development of neuroblastomas. (See 'Epidemiology' above.)

Molecular pathogenesis – The molecular pathogenesis of neuroblastomas has been extensively studied, and information about specific abnormalities is an important component of the definition of prognostic risk groups (see 'Molecular abnormalities (prognostic impact)' above):

SCAs – Segmental chromosomal aberrations (SCAs), particularly segmental deletions of chromosome 1p, are associated with a poor prognosis. (See 'Segmental chromosome abberrations' above.)

MYCN status – Deletions of chromosome 1p are associated with amplification of the MYCN oncogene (also called N-myc), the most common focal genetic lesion in sporadic neuroblastoma, which is associated with poor prognosis. (See 'MYCN status' above.)

Alterations in total DNA content – The presence or absence of alterations in total DNA content and the amplification of MYCN have been incorporated into the Children's Oncology Group (COG) Neuroblastoma Risk Stratification System (table 3), which is important in determining the appropriate therapy for newly diagnosed patients. (See 'Alterations in total DNA content' above and "Treatment and prognosis of neuroblastoma".)

Pathology – The International Neuroblastoma Pathology Classification classifies tumors of neuroblastic origin according to the balance between neural-type cells (primitive neuroblasts, maturing neuroblasts, and ganglion cells) and Schwann-type cells (Schwannian-blasts and mature Schwann cells) into one of three types: neuroblastoma, ganglioneuroblastoma, or ganglioneuroma. (See 'Pathology' above.)

Neuroblastomas are the most undifferentiated-appearing, and they in turn may be classified as undifferentiated, poorly differentiated, or differentiating. (See 'Neuroblastoma' above.)

The histologic subtype and degree of differentiation can be predictive of outcome and is used in the determination of COG risk category for treatment (table 3).

  1. Tomolonis JA, Agarwal S, Shohet JM. Neuroblastoma pathogenesis: deregulation of embryonic neural crest development. Cell Tissue Res 2018; 372:245.
  2. Mayanil CS. Transcriptional and epigenetic regulation of neural crest induction during neurulation. Dev Neurosci 2013; 35:361.
  3. Rogers CD, Nie S. Specifying neural crest cells: From chromatin to morphogens and factors in between. Wiley Interdiscip Rev Dev Biol 2018; 7:e322.
  4. Goodman MT, Gurney JG, Smith MA, Olshan AF. Sympathetic nervous system tumors. In: Cancer Incidence and Survival among Children and Adolescents: United States SEER Program, 1975-1995, Ries LA, Smith,MA, Gurney JG, et al (Eds), National Cancer Institute, Bethesda, MD 1999. p.35.
  5. Brodeur GM, Hogarty MD, Mosse YP, Maris JM. Neuroblastoma. In: Principles and Practice of Pediatric Oncology, Pizzo PA, Poplack DG (Eds), Lippincott Williams & Wilkins, Philadelphia 2011. p.886.
  6. Gurney JG, Ross JA, Wall DA, et al. Infant cancer in the U.S.: histology-specific incidence and trends, 1973 to 1992. J Pediatr Hematol Oncol 1997; 19:428.
  7. Cook MN, Olshan AF, Guess HA, et al. Maternal medication use and neuroblastoma in offspring. Am J Epidemiol 2004; 159:721.
  8. French AE, Grant R, Weitzman S, et al. Folic acid food fortification is associated with a decline in neuroblastoma. Clin Pharmacol Ther 2003; 74:288.
  9. Olshan AF, Smith JC, Bondy ML, et al. Maternal vitamin use and reduced risk of neuroblastoma. Epidemiology 2002; 13:575.
  10. Rios P, Bailey HD, Orsi L, et al. Risk of neuroblastoma, birth-related characteristics, congenital malformations and perinatal exposures: A pooled analysis of the ESCALE and ESTELLE French studies (SFCE). Int J Cancer 2016; 139:1936.
  11. Kramer S, Ward E, Meadows AT, Malone KE. Medical and drug risk factors associated with neuroblastoma: a case-control study. J Natl Cancer Inst 1987; 78:797.
  12. Schwartzbaum JA. Influence of the mother's prenatal drug consumption on risk of neuroblastoma in the child. Am J Epidemiol 1992; 135:1358.
  13. Michalek AM, Buck GM, Nasca PC, et al. Gravid health status, medication use, and risk of neuroblastoma. Am J Epidemiol 1996; 143:996.
  14. Satgé D, Sasco AJ, Little J. Antenatal therapeutic drug exposure and fetal/neonatal tumours: review of 89 cases. Paediatr Perinat Epidemiol 1998; 12:84.
  15. Johnson CC, Spitz MR. Neuroblastoma: case-control analysis of birth characteristics. J Natl Cancer Inst 1985; 74:789.
  16. Neglia JP, Smithson WA, Gunderson P, et al. Prenatal and perinatal risk factors for neuroblastoma. A case-control study. Cancer 1988; 61:2202.
  17. Bunin GR, Ward E, Kramer S, et al. Neuroblastoma and parental occupation. Am J Epidemiol 1990; 131:776.
  18. Spitz MR, Johnson CC. Neuroblastoma and paternal occupation. A case-control analysis. Am J Epidemiol 1985; 121:924.
  19. Wilkins JR 3rd, Hundley VD. Paternal occupational exposure to electromagnetic fields and neuroblastoma in offspring. Am J Epidemiol 1990; 131:995.
  20. Flaegstad T, Andresen PA, Johnsen JI, et al. A possible contributory role of BK virus infection in neuroblastoma development. Cancer Res 1999; 59:1160.
  21. Menegaux F, Olshan AF, Neglia JP, et al. Day care, childhood infections, and risk of neuroblastoma. Am J Epidemiol 2004; 159:843.
  22. Mili F, Khoury MJ, Flanders WD, Greenberg RS. Risk of childhood cancer for infants with birth defects. I. A record-linkage study, Atlanta, Georgia, 1968-1988. Am J Epidemiol 1993; 137:629.
  23. Foulkes WD, Buu PN, Filiatrault D, et al. Excess of congenital abnormalities in French-Canadian children with neuroblastoma: a case series study from Montréal. Med Pediatr Oncol 1997; 29:272.
  24. Menegaux F, Olshan AF, Reitnauer PJ, et al. Positive association between congenital anomalies and risk of neuroblastoma. Pediatr Blood Cancer 2005; 45:649.
  25. Chow EJ, Friedman DL, Mueller BA. Maternal and perinatal characteristics in relation to neuroblastoma. Cancer 2007; 109:983.
  26. Munzer C, Menegaux F, Lacour B, et al. Birth-related characteristics, congenital malformation, maternal reproductive history and neuroblastoma: the ESCALE study (SFCE). Int J Cancer 2008; 122:2315.
  27. Buck GM, Michalek AM, Chen CJ, et al. Perinatal factors and risk of neuroblastoma. Paediatr Perinat Epidemiol 2001; 15:47.
  28. Windham GC, Bjerkedal T, Langmark F. A population-based study of cancer incidence in twins and in children with congenital malformations or low birth weight, Norway, 1967-1980. Am J Epidemiol 1985; 121:49.
  29. Mili F, Lynch CF, Khoury MJ, et al. Risk of childhood cancer for infants with birth defects. II. A record-linkage study, Iowa, 1983-1989. Am J Epidemiol 1993; 137:639.
  30. Oldridge DA, Wood AC, Weichert-Leahey N, et al. Genetic predisposition to neuroblastoma mediated by a LMO1 super-enhancer polymorphism. Nature 2015; 528:418.
  31. Wang K, Diskin SJ, Zhang H, et al. Integrative genomics identifies LMO1 as a neuroblastoma oncogene. Nature 2011; 469:216.
  32. Zhang J, Walsh MF, Wu G, et al. Germline Mutations in Predisposition Genes in Pediatric Cancer. N Engl J Med 2015; 373:2336.
  33. Blatt J, Olshan AF, Lee PA, Ross JL. Neuroblastoma and related tumors in Turner's syndrome. J Pediatr 1997; 131:666.
  34. van Limpt V, Schramm A, van Lakeman A, et al. The Phox2B homeobox gene is mutated in sporadic neuroblastomas. Oncogene 2004; 23:9280.
  35. Trochet D, Bourdeaut F, Janoueix-Lerosey I, et al. Germline mutations of the paired-like homeobox 2B (PHOX2B) gene in neuroblastoma. Am J Hum Genet 2004; 74:761.
  36. Mosse YP, Laudenslager M, Khazi D, et al. Germline PHOX2B mutation in hereditary neuroblastoma. Am J Hum Genet 2004; 75:727.
  37. Raabe EH, Laudenslager M, Winter C, et al. Prevalence and functional consequence of PHOX2B mutations in neuroblastoma. Oncogene 2008; 27:469.
  38. Amiel J, Laudier B, Attié-Bitach T, et al. Polyalanine expansion and frameshift mutations of the paired-like homeobox gene PHOX2B in congenital central hypoventilation syndrome. Nat Genet 2003; 33:459.
  39. Arenson EB Jr, Hutter JJ Jr, Restuccia RD, Holton CP. Neuroblastoma in father and son. JAMA 1976; 235:727.
  40. Maris JM, Chatten J, Meadows AT, et al. Familial neuroblastoma: a three-generation pedigree and a further association with Hirschsprung disease. Med Pediatr Oncol 1997; 28:1.
  41. Kushner BH, Gilbert F, Helson L. Familial neuroblastoma. Case reports, literature review, and etiologic considerations. Cancer 1986; 57:1887.
  42. Janoueix-Lerosey I, Lequin D, Brugières L, et al. Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature 2008; 455:967.
  43. Mossé YP, Laudenslager M, Longo L, et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 2008; 455:930.
  44. Perri P, Longo L, McConville C, et al. Linkage analysis in families with recurrent neuroblastoma. Ann N Y Acad Sci 2002; 963:74.
  45. Maris JM, Weiss MJ, Mosse Y, et al. Evidence for a hereditary neuroblastoma predisposition locus at chromosome 16p12-13. Cancer Res 2002; 62:6651.
  46. Weiss MJ, Guo C, Shusterman S, et al. Localization of a hereditary neuroblastoma predisposition gene to 16p12-p13. Med Pediatr Oncol 2000; 35:526.
  47. Diskin SJ, Capasso M, Diamond M, et al. Rare variants in TP53 and susceptibility to neuroblastoma. J Natl Cancer Inst 2014; 106:dju047.
  48. Louis CU, Shohet JM. Neuroblastoma: molecular pathogenesis and therapy. Annu Rev Med 2015; 66:49.
  49. Weston JA, Thiery JP. Pentimento: Neural Crest and the origin of mesectoderm. Dev Biol 2015; 401:37.
  50. Baggiolini A, Varum S, Mateos JM, et al. Premigratory and migratory neural crest cells are multipotent in vivo. Cell Stem Cell 2015; 16:314.
  51. Plouhinec JL, Roche DD, Pegoraro C, et al. Pax3 and Zic1 trigger the early neural crest gene regulatory network by the direct activation of multiple key neural crest specifiers. Dev Biol 2014; 386:461.
  52. Woods WG, Gao RN, Shuster JJ, et al. Screening of infants and mortality due to neuroblastoma. N Engl J Med 2002; 346:1041.
  53. Bessho F. Comparison of the incidences of neuroblastoma for screened and unscreened cohorts. Acta Paediatr 1999; 88:404.
  54. Brodeur GM. Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer 2003; 3:203.
  55. Perez CA, Matthay KK, Atkinson JB, et al. Biologic variables in the outcome of stages I and II neuroblastoma treated with surgery as primary therapy: a children's cancer group study. J Clin Oncol 2000; 18:18.
  56. Riley RD, Heney D, Jones DR, et al. A systematic review of molecular and biological tumor markers in neuroblastoma. Clin Cancer Res 2004; 10:4.
  57. Viprey VF, Gregory WM, Corrias MV, et al. Neuroblastoma mRNAs predict outcome in children with stage 4 neuroblastoma: a European HR-NBL1/SIOPEN study. J Clin Oncol 2014; 32:1074.
  58. Cheung NK, Dyer MA. Neuroblastoma: developmental biology, cancer genomics and immunotherapy. Nat Rev Cancer 2013; 13:397.
  59. Schwab M, Shimada H, Joshi V, Brodeur GM. Neuroblastic tumours of adrenal gland and sympathetic nervous system. In: Pathology and Genetics of Tumours of the Nervous System, Kleihues P, Cavenee WK (Eds), Lyon 2000. p.153.
  60. Westermann F, Schwab M. Genetic parameters of neuroblastomas. Cancer Lett 2002; 184:127.
  61. Guo C, White PS, Hogarty MD, et al. Deletion of 11q23 is a frequent event in the evolution of MYCN single-copy high-risk neuroblastomas. Med Pediatr Oncol 2000; 35:544.
  62. Srivatsan ES, Ying KL, Seeger RC. Deletion of chromosome 11 and of 14q sequences in neuroblastoma. Genes Chromosomes Cancer 1993; 7:32.
  63. Attiyeh EF, London WB, Mossé YP, et al. Chromosome 1p and 11q deletions and outcome in neuroblastoma. N Engl J Med 2005; 353:2243.
  64. Schleiermacher G, Janoueix-Lerosey I, Ribeiro A, et al. Accumulation of segmental alterations determines progression in neuroblastoma. J Clin Oncol 2010; 28:3122.
  65. Schleiermacher G, Mosseri V, London WB, et al. Segmental chromosomal alterations have prognostic impact in neuroblastoma: a report from the INRG project. Br J Cancer 2012; 107:1418.
  66. Irwin MS, Naranjo A, Zhang FF, et al. Revised Neuroblastoma Risk Classification System: A Report From the Children's Oncology Group. J Clin Oncol 2021; 39:3229.
  67. Maris JM, Weiss MJ, Guo C, et al. Loss of heterozygosity at 1p36 independently predicts for disease progression but not decreased overall survival probability in neuroblastoma patients: a Children's Cancer Group study. J Clin Oncol 2000; 18:1888.
  68. Krona C, Ejeskär K, Abel F, et al. Screening for gene mutations in a 500 kb neuroblastoma tumor suppressor candidate region in chromosome 1p; mutation and stage-specific expression in UBE4B/UFD2. Oncogene 2003; 22:2343.
  69. Roberts T, Chernova O, Cowell JK. NB4S, a member of the TBC1 domain family of genes, is truncated as a result of a constitutional t(1;10)(p22;q21) chromosome translocation in a patient with stage 4S neuroblastoma. Hum Mol Genet 1998; 7:1169.
  70. Caron H, van Sluis P, de Kraker J, et al. Allelic loss of chromosome 1p as a predictor of unfavorable outcome in patients with neuroblastoma. N Engl J Med 1996; 334:225.
  71. Cohn SL, Pearson AD, London WB, et al. The International Neuroblastoma Risk Group (INRG) classification system: an INRG Task Force report. J Clin Oncol 2009; 27:289.
  72. Monclair T, Brodeur GM, Ambros PF, et al. The International Neuroblastoma Risk Group (INRG) staging system: an INRG Task Force report. J Clin Oncol 2009; 27:298.
  73. Komuro H, Valentine MB, Rowe ST, et al. Fluorescence in situ hybridization analysis of chromosome 1p36 deletions in human MYCN amplified neuroblastoma. J Pediatr Surg 1998; 33:1695.
  74. Cole MD, McMahon SB. The Myc oncoprotein: a critical evaluation of transactivation and target gene regulation. Oncogene 1999; 18:2916.
  75. Lutz W, Stöhr M, Schürmann J, et al. Conditional expression of N-myc in human neuroblastoma cells increases expression of alpha-prothymosin and ornithine decarboxylase and accelerates progression into S-phase early after mitogenic stimulation of quiescent cells. Oncogene 1996; 13:803.
  76. Schmitt-Hoffner F, van Rijn S, Toprak UH, et al. FOXR2 Stabilizes MYCN Protein and Identifies Non-MYCN-Amplified Neuroblastoma Patients With Unfavorable Outcome. J Clin Oncol 2021; 39:3217.
  77. Schwab M. Oncogene amplification in solid tumors. Semin Cancer Biol 1999; 9:319.
  78. Matthay KK, Villablanca JG, Seeger RC, et al. Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplantation, and 13-cis-retinoic acid. Children's Cancer Group. N Engl J Med 1999; 341:1165.
  79. Brodeur GM, Seeger RC, Schwab M, et al. Amplification of N-myc in untreated human neuroblastomas correlates with advanced disease stage. Science 1984; 224:1121.
  80. Seeger RC, Brodeur GM, Sather H, et al. Association of multiple copies of the N-myc oncogene with rapid progression of neuroblastomas. N Engl J Med 1985; 313:1111.
  81. Katzenstein HM, Bowman LC, Brodeur GM, et al. Prognostic significance of age, MYCN oncogene amplification, tumor cell ploidy, and histology in 110 infants with stage D(S) neuroblastoma: the pediatric oncology group experience--a pediatric oncology group study. J Clin Oncol 1998; 16:2007.
  82. Bagatell R, Beck-Popovic M, London WB, et al. Significance of MYCN amplification in international neuroblastoma staging system stage 1 and 2 neuroblastoma: a report from the International Neuroblastoma Risk Group database. J Clin Oncol 2009; 27:365.
  83. Campbell K, Naranjo A, Hibbitts E, et al. Association of heterogeneous MYCN amplification with clinical features, biological characteristics and outcomes in neuroblastoma: A report from the Children's Oncology Group. Eur J Cancer 2020; 133:112.
  84. Schmidt ML, Lal A, Seeger RC, et al. Favorable prognosis for patients 12 to 18 months of age with stage 4 nonamplified MYCN neuroblastoma: a Children's Cancer Group Study. J Clin Oncol 2005; 23:6474.
  85. Stallings RL, Howard J, Dunlop A, et al. Are gains of chromosomal regions 7q and 11p important abnormalities in neuroblastoma? Cancer Genet Cytogenet 2003; 140:133.
  86. Bown N, Cotterill S, Lastowska M, et al. Gain of chromosome arm 17q and adverse outcome in patients with neuroblastoma. N Engl J Med 1999; 340:1954.
  87. Vandesompele J, Baudis M, De Preter K, et al. Unequivocal delineation of clinicogenetic subgroups and development of a new model for improved outcome prediction in neuroblastoma. J Clin Oncol 2005; 23:2280.
  88. Look AT, Hayes FA, Shuster JJ, et al. Clinical relevance of tumor cell ploidy and N-myc gene amplification in childhood neuroblastoma: a Pediatric Oncology Group study. J Clin Oncol 1991; 9:581.
  89. Look AT, Hayes FA, Nitschke R, et al. Cellular DNA content as a predictor of response to chemotherapy in infants with unresectable neuroblastoma. N Engl J Med 1984; 311:231.
  90. Christiansen H, Lampert F. Tumour karyotype discriminates between good and bad prognostic outcome in neuroblastoma. Br J Cancer 1988; 57:121.
  91. Kaneko Y, Kanda N, Maseki N, et al. Different karyotypic patterns in early and advanced stage neuroblastomas. Cancer Res 1987; 47:311.
  92. Bourhis J, De Vathaire F, Wilson GD, et al. Combined analysis of DNA ploidy index and N-myc genomic content in neuroblastoma. Cancer Res 1991; 51:33.
  93. Oppedal BR, Storm-Mathisen I, Lie SO, Brandtzaeg P. Prognostic factors in neuroblastoma. Clinical, histopathologic, and immunohistochemical features and DNA ploidy in relation to prognosis. Cancer 1988; 62:772.
  94. Cohn SL, Rademaker AW, Salwen HR, et al. Analysis of DNA ploidy and proliferative activity in relation to histology and N-myc amplification in neuroblastoma. Am J Pathol 1990; 136:1043.
  95. Mora J, Cheung NK, Chen L, et al. Survival analysis of clinical, pathologic, and genetic features in neuroblastoma presenting as locoregional disease. Cancer 2001; 91:435.
  96. Brodeur GM, Nakagawara A, Yamashiro DJ, et al. Expression of TrkA, TrkB and TrkC in human neuroblastomas. J Neurooncol 1997; 31:49.
  97. Brodeur GM, Minturn JE, Ho R, et al. Trk receptor expression and inhibition in neuroblastomas. Clin Cancer Res 2009; 15:3244.
  98. 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.
  99. Brodeur GM. Spontaneous regression of neuroblastoma. Cell Tissue Res 2018; 372:277.
  100. Eggert A, Grotzer MA, Ikegaki N, et al. Expression of neurotrophin receptor TrkA inhibits angiogenesis in neuroblastoma. Med Pediatr Oncol 2000; 35:569.
  101. Nakagawara A, Arima M, Azar CG, et al. Inverse relationship between trk expression and N-myc amplification in human neuroblastomas. Cancer Res 1992; 52:1364.
  102. Nakagawara A, Arima-Nakagawara M, Scavarda NJ, et al. Association between high levels of expression of the TRK gene and favorable outcome in human neuroblastoma. N Engl J Med 1993; 328:847.
  103. Suzuki T, Bogenmann E, Shimada H, et al. Lack of high-affinity nerve growth factor receptors in aggressive neuroblastomas. J Natl Cancer Inst 1993; 85:377.
  104. Chen Y, Takita J, Choi YL, et al. Oncogenic mutations of ALK kinase in neuroblastoma. Nature 2008; 455:971.
  105. George RE, Sanda T, Hanna M, et al. Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature 2008; 455:975.
  106. Cazes A, Louis-Brennetot C, Mazot P, et al. Characterization of rearrangements involving the ALK gene reveals a novel truncated form associated with tumor aggressiveness in neuroblastoma. Cancer Res 2013; 73:195.
  107. Ohali A, Avigad S, Ash S, et al. Telomere length is a prognostic factor in neuroblastoma. Cancer 2006; 107:1391.
  108. Koneru B, Lopez G, Farooqi A, et al. Telomere Maintenance Mechanisms Define Clinical Outcome in High-Risk Neuroblastoma. Cancer Res 2020; 80:2663.
  109. Hartlieb SA, Sieverling L, Nadler-Holly M, et al. Alternative lengthening of telomeres in childhood neuroblastoma from genome to proteome. Nat Commun 2021; 12:1269.
  110. Cheung NK, Zhang J, Lu C, et al. Association of age at diagnosis and genetic mutations in patients with neuroblastoma. JAMA 2012; 307:1062.
  111. Vermeulen J, De Preter K, Naranjo A, et al. Predicting outcomes for children with neuroblastoma using a multigene-expression signature: a retrospective SIOPEN/COG/GPOH study. Lancet Oncol 2009; 10:663.
  112. Oberthuer A, Berthold F, Warnat P, et al. Customized oligonucleotide microarray gene expression-based classification of neuroblastoma patients outperforms current clinical risk stratification. J Clin Oncol 2006; 24:5070.
  113. Fischer M, Oberthuer A, Brors B, et al. Differential expression of neuronal genes defines subtypes of disseminated neuroblastoma with favorable and unfavorable outcome. Clin Cancer Res 2006; 12:5118.
  114. Shimada H, Ambros IM, Dehner LP, et al. The International Neuroblastoma Pathology Classification (the Shimada system). Cancer 1999; 86:364.
  115. Golden CB, Feusner JH. Malignant abdominal masses in children: quick guide to evaluation and diagnosis. Pediatr Clin North Am 2002; 49:1369.
  116. Origone P, Defferrari R, Mazzocco K, et al. Homozygous inactivation of NF1 gene in a patient with familial NF1 and disseminated neuroblastoma. Am J Med Genet A 2003; 118A:309.
  117. Shimada H, Chatten J, Newton WA Jr, et al. Histopathologic prognostic factors in neuroblastic tumors: definition of subtypes of ganglioneuroblastoma and an age-linked classification of neuroblastomas. J Natl Cancer Inst 1984; 73:405.
  118. Koch CA, Brouwers FM, Rosenblatt K, et al. Adrenal ganglioneuroma in a patient presenting with severe hypertension and diarrhea. Endocr Relat Cancer 2003; 10:99.
  119. Meyer S, Reinhard H, Ziegler K, et al. Ganglianeuroma: radiological and metabolic features in 4 children. Pediatr Hematol Oncol 2002; 19:501.
  120. Geoerger B, Hero B, Harms D, et al. Metabolic activity and clinical features of primary ganglioneuromas. Cancer 2001; 91:1905.
  121. De Bernardi B, Gambini C, Haupt R, et al. Retrospective study of childhood ganglioneuroma. J Clin Oncol 2008; 26:1710.
Topic 5204 Version 28.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟