Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

SECTION G Oncology

53 Pediatric Urologic Oncology: Renal and Adrenal


Michael L. Ritchey, MD, Nicholas G. Cost, MD, and Robert C. Shamberger, MD

NEUROBLASTOMA changes occur in the form of chromosomal deletions, translocations,


and cytogenetic evidence of gene amplification. Aneuploidy of the
Neuroblastoma is the most common extracranial solid tumor of tumor DNA, an abnormal number of copies of the normal 23
childhood. Unfortunately, more than half of the children are seen chromosomes, occurs in a significant number of cases (55%) and
initially with metastatic disease. Neuroblastoma is known to arise from is a favorable prognostic indicator when compared with tumors that
cells of the neural crest that form the adrenal medulla and sympathetic have a normal or tetraploid number (Kaneko et al., 1987). Amplifica-
ganglia. Tumors may occur anywhere along the sympathetic chain tion of the MYCN oncogene at 2p24 defined as greater than 10
within the neck, thorax, retroperitoneum, or pelvis or in the adrenal copies of this gene is seen in roughly 20% to 25% of primary
gland: 75% arise in the retroperitoneum, 50% in the adrenal, and tumors and is an adverse prognostic indicator (Look et al., 1991;
25% in the paravertebral ganglia. The variety of locations where Muraji et al., 1993). It is present in 40% of patients with advanced
these tumors arise and the spectrum of their differentiation result in stage disease but in only 5% to 10% of children with low-stage
a wide range of clinical presentations and behaviors (Brodeur, 1991). disease (Brodeur et al., 1984). It is associated with rapid tumor
These tumors can undergo spontaneous regression (Brodeur, 1991), progression and poor outcome from treatment. These findings have
differentiate to benign neoplasms, or exhibit extremely malignant been so striking that neuroblastoma was the first tumor in which
behavior. Biologic factors have been defined that predict and explain the intensity of chemotherapy for a patient was determined not only
much of the variance in behavior between tumors. by the stage and histology of the tumor but also by its “biologic
markers,” which were primarily chromosomal (Matthay et al., 1998).
Deletion of the short arm of chromosome 1 (1p) is found in
Epidemiology and Genetics 25% to 35% of neuroblastomas and is an adverse prognostic marker
Incidence (Brodeur et al., 1992; Caron et al., 1996). The deletions are of various
lengths, but, in a series of eight cases, a consensus deletion included
Neuroblastoma accounts for 8% to 10% of all childhood cancers. the segment 1p36.1-2, suggesting that a tumor suppressor may be
In the United States, the annual incidence is 10 cases per 1 million present in this region; although the involved genes have not been
live births. It is the most common malignant tumor of infancy. A identified conclusively, CHD5 is a strong candidate for this role
review of 3666 children enrolled in the cooperative group trials of (Fujita et al., 2008; Maris et al., 2007; Weith et al., 1989). This deletion
the Pediatric Oncology Group and the Children’s Cancer Group is present in 70% of advanced stage neuroblastoma, and it has been
showed a median age at diagnosis of 19 months (Brodeur and Maris, demonstrated to be an independent prognostic factor (Attiyeh et al.,
2006): 36% were infants, 89% were younger than 5 years, and 98% 2005). There have been reports of constitutional abnormalities
were diagnosed by 10 years of age. involving the short arm of chromosome 1 (Laureys et al., 1990). It
has been demonstrated that loss of heterozygosity (LOH) at 1p36
Genetics and Unb11q is independently associated with a worse outcome in
patients with neuroblastoma (Attiyeh et al., 2005). (Unbalanced
There have been a number of familial cases reported that are pos- LOH implies LOH at markers on 11q with retention of 11p material,
tulated to represent an autosomal dominant pattern of inheritance in contrast with “whole-chromosome” 11 LOH, where there is LOH
(Knudson and Strong, 1972; Robertson et al., 1991). Although the at every marker along the chromosome.) Although the 1p deletions
median age at diagnosis of neuroblastoma is 19 months, in familial are seen in conjunction with advanced stage and MYCN amplification,
cases it is 9 months (Kushner et al., 1986). At least 20% of patients the 11q deletions are rarely seen in tumors with MYCN amplification
with familial neuroblastoma have bilateral adrenal or multifocal but are associated with other high-risk features and are predictive
primary tumors, which are unusual in spontaneous cases. The risk for of poor outcomes in the subset of patients with low-stage disease
development of neuroblastoma in a sibling or offspring of a patient without MYCN amplification (Spitz et al., 2006). An additional
with neuroblastoma is less than 6% (Kushner et al., 1986). Linkage genetic abnormality, gain of one to three copies of 17q, often the
analysis in seven families with two or more first-degree relatives result of translocation with chromosomes 1 or 11, has been dem-
affected with neuroblastoma identified a single interval at chromo- onstrated to correlate with more aggressive tumors (Bown et al.,
some 16p12-13 with consistent linkage (Maris et al., 2002). This 1999). The break points vary, but the addition of a region from
suggested that a hereditary neuroblastoma predisposition gene may 17q22-qter is common, suggesting that genes replicated in this region
be located at this site and may explain the familial cases. Subsequent provide an advantage (Schleiermacher et al., 2004). In multivariate
studies in these familial cases, which account for less than 1% of analysis, gain of 17q was the most powerful prognostic factor, followed
patients with neuroblastoma, have identified PHOX2B and ALK as by the presence of stage IV disease and deletion of 1p.
hereditary predisposition genes (Mosse et al., 2004, 2008).
Embryology and Spontaneous Regression
Constitutional Chromosome Abnormalities
In 1963 Beckwith and Perrin coined the term in situ neuroblastoma
Numerous karyotypic abnormalities have been found in neuroblas- for small nodules of neuroblasts found incidentally within the adrenal
toma, and these are recognized to have prognostic significance. These gland that are histologically indistinguishable from neuroblastoma.

1087
1088 PART III Pediatric Urology

In situ neuroblastoma was found during postmortem examination International Neuroblastoma Pathology Classification in 1999, has
in 1 of 224 infants younger than 3 months. This represents an helped to define risk-based subtypes of ganglioneuroblastoma and
incidence of approximately 40 to 45 times greater than that of clinical neuroblastoma (Shimada et al., 1984, 1999a, 1999b). This revised
tumors, suggesting that these small tumors regress spontaneously system has been demonstrated to add independent prognostic
in most cases. Subsequent studies have shown that these nodules information beyond the contribution of age that is one of the factors
of neuroblasts are found in all fetuses studied and generally regress contained in the system (Sano et al., 2006). Ganglioneuroma is a
(Ikeda et al., 1981). Neuroblastoma identified by prenatal ultraso- histologically benign, fully differentiated counterpart of neuroblas-
nography (US) has also been shown to have a clinically favorable toma. It is unclear whether ganglioneuroma arises de novo or by
course (Ho et al., 1993). maturation of a preexisting neuroblastoma or ganglioneuroblastoma.
The concept of in situ neuroblastoma has been used to support Metastatic lesions from neuroblastoma have been observed to develop
the argument that many neuroblastomas arise and regress spontane- the histology of mature ganglioneuroma, supporting the latter theory
ously. This concept has been further supported by population-based (Hayes et al., 1989).
studies in Quebec province and in Japan, where prospective screening The Shimada classification is an age-linked histopathologic
of infants for neuroblastoma has been performed based on urinary classification. One of its important aspects is determining whether
catecholamine excretion. An increased number of children were the tumor is stroma poor or stroma rich. Patients with stroma-poor
identified with low-stage neuroblastoma, a higher frequency than tumors with unfavorable histopathologic features have a very
present clinically, but resection of those tumors did not result in a poor prognosis (less than 10% survival) (Shimada et al., 1984).
decrease in the incidence of advanced-stage tumors seen at an older Stroma-rich tumors can be separated into three subgroups: nodular,
age (Hayashi et al., 1995; Woods et al., 1996). A subsequent German intermixed, and well differentiated. Tumors in the latter two categories
Neuroblastoma Screening Study postponed screening until 10 to 19 more closely resemble ganglioneuroblastoma or immature ganglio-
months of age. Fewer early stage cases were identified, and a greater neuroma and carry a higher rate of survival. The stroma-poor tumors
frequency of patients with unfavorable clinical and biologic features can be divided into favorable and unfavorable subgroups based on
were identified, but there was no decrease in the mortality of these the patient’s age at diagnosis, the degree of histologic maturation,
patients (Schilling et al., 2002). Evaluation of adrenal tumors resected and the mitotic rate. When compared with other clinical features,
in the neonatal period, whether cystic or solid, showed that in most, these histologic patterns were independently predictive of outcome
the “biologic markers” were favorable (Kozakewich et al., 1998). (Shimada et al., 1984).
The highly favorable outcome of infants diagnosed with neuroblas- In contrast to neuroblastomas, ganglioneuromas are most often
toma in the population screening studies led to attempts at expectant diagnosed in older children and are usually located in the posterior
observation. These trials demonstrated a high rate of spontaneous mediastinum and retroperitoneum, with only a small number arising
resolution (Yamamoto et al., 1998; Yoneda et al., 2001). Spontaneous in the adrenal glands (Enzinger and Weiss, 1988). Ganglioneuromas
regression of these perinatally identified lesions also has been often grow to a very large size before they cause symptoms as a
demonstrated radiographically (Holgerson et al., 1996). These findings result of compression of adjacent structures or extension into the
led to a prospective study by the Children’s Oncology Group (COG) spinal canal (Benjamin et al., 1972; Fig. 53.2). Because survival is
evaluating treatment of infants with adrenal masses identified in not influenced by extent of resection, it should be used only in those
the perinatal or neonatal period in which expectant observation for patients in whom significant symptoms are present, and aggressive
infants with small lesions with favorable catecholamine ratios was attempts at resection should be avoided (DeBernardi et al., 2008).
encouraged and is discussed later.
Clinical Presentation and Pattern of Spread
Pathology
The clinical manifestations of neuroblastoma vary widely. Most
Neuroblastoma, ganglioneuroblastoma, and ganglioneuroma display children have abdominal pain or a palpable mass, but others are
a spectrum of histologic maturation and differentiation (Fig. 53.1). identified as a result of manifestations of their metastatic disease,
A grading classification of neuroblastoma, introduced in 1984 by including bone or joint pain and periorbital ecchymosis. Thoracic
Shimada and subsequently modified by him and others as the lesions may produce respiratory symptoms of cough or dyspnea.

A B
Fig. 53.1. (A) Poorly differentiated neuroblastoma with a minimal to moderate amount of neuropil. (B)
Ganglioneuroblastoma with more than 50% of surface area occupied by Schwannian stroma on left.
Lobule contains ganglion cells, immature ganglion cells, and poorly differentiated neuroblasts; only minimal
neuropil is present within lobule. (Photo courtesy Dr. Harry Kozakewich.)
Chapter 53 Pediatric Urologic Oncology: Renal and Adrenal 1089

patients develop myoclonus, rapid multidirectional eye movements


(opsoclonus), and ataxia. It is thought to result from an interaction
of antibodies produced against the neuroblastoma to normal neural
tissues (Connolly et al., 1997; Farrelly et al, 1984). Although this
syndrome is associated with a favorable outcome from an oncologic
perspective (Altman and Baehner, 1976), prolonged neurologic impair-
ment, including learning disabilities and developmental delay, is the
rule in 70% to 80% of these children, and symptomatic therapy is
often required (Mitchell et al., 2002; Rudnick et al., 2001; Russo
et al., 1997). Adrenocorticotropic hormone (ACTH) or steroids are
the basis of most therapy, but other treatments include high-dose
intravenous gamma globulin and cyclophosphamide. A recently
completed protocol of the Children’s Oncology Group evaluated
the efficacy of cyclophosphamide plus steroids randomized to
receive intravenous gamma globulin or not. The addition of IVIG
to prednisone and risk-adapted chemotherapy improved opsoclonus/
myoclonus response (de Alarcon et al., 2018).
Fig. 53.2. MRI of child with thoracic ganglioneuroma extending into the
spinal canal (solid arrow).
Diagnosis
Laboratory Evaluation
When sensitive techniques are used, increased levels of urinary
metabolites of catecholamines, vanillylmandelic acid (VMA) and
homovanillic acid (HVA), are found in 90% to 95% of patients
(Williams and Greer, 1963). Therapy with various modalities produces
a reduction in catecholamine metabolite excretion in most patients
(Gerson and Koop, 1974). These metabolites can be monitored to
detect tumor relapse and response to therapy.
Anemia is noted in children with widespread bone marrow
involvement. Studies suggest that marrow biopsies add substantially
to the detection of marrow involvement by tumor, compared with
marrow aspirates alone (Franklin and Pritchard, 1983). It is recom-
mended that two marrow aspirates and two biopsies be performed
for staging at presentation. In the future, neuroblastoma-specific
immunocytology of marrow aspirates may obviate the need for
marrow biopsies in most patients (Hsiao et al., 1990).

Imaging
Imaging studies play an important role in the evaluation of a child
with neuroblastoma. Plain radiographs may demonstrate a calcified
abdominal or posterior mediastinal mass. Computed tomography
(CT) and magnetic resonance imaging (MRI) provide more infor-
mation about the local extent of the primary tumors and vascular
involvement. Invasion of the renal parenchyma is not common,
but it can be detected radiographically by CT (Albregts et al., 1994).
MRI has advantages over CT in the evaluation of intraspinal tumor
Fig. 53.3. MRI demonstrating compression of bowel and bladder by a pelvic
extension, which is common in the paravertebral lesions (see Fig.
neuroblastoma.
53.2), and in demonstrating the relationship between the major vessels
and the tumor (Azizkhan and Haase, 1993; Fig. 53.4). The finding
of intratumoral calcifications, vascular encasement, or both on
Direct extension of the tumor into the spinal canal may produce preoperative CT may help distinguish neuroblastoma from Wilms
neurologic deficits as a result of cord compression. tumor (Dickson et al., 2008). Current Children’s Oncology Group
Most primary tumors arise within the abdomen (65%); the protocols require a radionuclide meta-iodobenzylguanidine (MIBG)
frequency of adrenal tumors is slightly higher in children than in scan for staging but no longer require the classic skeletal survey or
infants. Physical examination often reveals a fixed, hard abdominal bone scans. MIBG scans use 123I-MIBG (Geatti et al., 1985), which
mass. Pelvic neuroblastoma arising from the organ of Zuckerkandl is taken up by the adrenergic secretory vesicles of the tumor cells in
accounts for 4% of tumors (Haase et al., 1995). Extrinsic compression primary and metastatic sites. MIBG scintigraphy can determine the
of the bowel and bladder can produce symptoms of urinary retention extent of disease and detect tumor recurrence after completion of
and constipation (Fig. 53.3). therapy (Geatti et al., 1985). 123I-MIBG has been shown to be more
Metastases are present in 70% of patients with neuroblastoma at sensitive than either 131I-MIBG or bone scans in detecting tumor
diagnosis and can be responsible for a variety of the clinical signs relapse (Kushner et al., 2009). For patients who have non-MIBG
and symptoms at presentation. A number of unique paraneoplastic avid tumors, positron emission tomography (PET) scans are used.
syndromes have been associated with localized and disseminated Osseous metastatic lesions occur most commonly in the long bones
neuroblastoma. Symptoms produced by catecholamine release may and skull.
mimic those seen in pheochromocytoma: paroxysmal hypertension,
palpitations, flushing, and headache. Secretion of vasoactive intestinal Screening
peptide (VIP) by the tumor can produce severe watery diarrhea
and hypokalemia (Cooney et al., 1982). Another unusual presenta- Mass population screening for neuroblastoma has been widely used
tion of neuroblastoma is acute myoclonic encephalopathy, in which in Japan for more than 20 years (Nishi et al., 1987). The goal of
1090 PART III Pediatric Urology

screening programs is to detect disease at an earlier stage and to There are biologic differences between tumors diagnosed by
decrease the number of older children with advanced-stage disease, screening and those detected clinically (Hayashi et al., 1992). In
thereby improving survival. In fact, the children diagnosed as a result one review of 48 cases discovered by screening, no tumors were
of screening studies have had almost uniformly favorable survival observed to have amplified MYCN oncogene expression (Ishimoto
(>97%) (Suita, 2002). An increased number of infants younger than et al., 1991), and in a second review of 20 infants, only one who
1 year of age have been diagnosed through the mass screening program did poorly had amplification (Hase et al., 2002). Furthermore, 80%
(Ishimoto et al., 1990), and most of these patients have lower-stage had a diploid chromosome pattern, which is also associated with a
tumors (Sawada, 1992). Before mass screening, 20% of neuroblastoma favorable prognosis. On follow-up, all 48 patients were still alive
cases were diagnosed before 1 year of age, compared with 55% after without tumor. In another series of 357 patients whose tumors were
its implementation. However, the number of children older than 1 diagnosed by mass screening, the overall survival rate was 97%
year of age diagnosed with advanced-stage disease has not decreased. (Sawada, 1992). Given the favorable biologic characteristics of tumors
These results suggest that the aggressive advanced-stage tumors in discovered by screening, it is possible that many would spontaneously
older children did not arise from the low-risk tumors seen in the resolve without therapy, particularly given the increased incidence
infants less than 1 year of age. of neuroblastomas seen in the screened population.
Two large prospective population-based studies were conducted
in the Province of Quebec and in Germany. These studies demon-
strated that urine screening at various ages was successful in identifica-
tion of neuroblastoma, but there was no decrease in the occurrence
of neuroblastoma in older children and its subsequent mortality
(Schilling et al., 2002; Woods et al., 2002).

Staging
Staging of neuroblastoma is an important aspect of management.
The stage of the disease is a significant prognostic variable that
determines adjuvant therapy in the cooperative group trials. The
International Neuroblastoma Staging System (INSS) is based on
clinical, radiographic, and surgical evaluation of children with
neuroblastoma (Brodeur et al., 1993; Table 53.1). Earlier staging
systems provided generally comparable results in terms of distinguish-
ing low-stage, good-prognosis disease from high-stage, poor-prognosis
disease. Use of a uniform international system, however, makes
comparison of results from various studies much easier. The biggest
differences arise when the various systems are applied to those with
intermediate-stage disease.
An effort has been made to identify, preoperatively, those tumors
that are resectable without significant surgical risks. The Localized
Neuroblastoma European Study Group (LNESG) defined the
radiographic criteria associated with higher surgical morbidity. They
validated that the criteria’s presence was associated with a lower rate
of complete resection and a greater risk of surgical complications
(Cecchetto et al., 2005). This finding has been confirmed by others
(Simon et al., 2008). The International Neuroblastoma Risk Group
(INRG) Task Force proposed a staging system based on tumor imaging
rather than the extent of surgical resection to address this problem
Fig. 53.4. MRI of pelvic neuroblastoma. The bifurcation of the aorta and iliac (Monclair et al., 2009). In this system, localized tumors are graded
vessels are well delineated, showing the relationship to the mass. as L1 or L2 based on the absence or presence of 1 or more of 20

TABLE 53.1 International Neuroblastoma Staging System

STAGE DEFINITION

1 Localized tumor with complete gross excision with or without microscopic residual disease; representative ipsilateral lymph
nodes negative for tumor microscopically (nodes attached to and removed with the primary tumor may be positive).
2A Localized tumor with incomplete gross excision; representative ipsilateral nonadherent lymph nodes negative for tumor
microscopically.
2B Localized tumor with or without complete gross excision, with ipsilateral nonadherent lymph nodes positive for tumor.
Enlarged contralateral lymph nodes must be negative microscopically.
3 Unresectable unilateral tumor infiltrating across the midlinea, with or without regional lymph node involvement; or localized
unilateral tumor with contralateral regional lymph node involvement; or midline tumor with bilateral extension by
infiltration (unresectable) or by lymph node involvement.
4 Any primary tumor with dissemination to distant lymph nodes, bone, bone marrow, liver, skin and/or other organs.
4S Localized primary tumor (as defined for stage I, IIA, or IIB), with dissemination limited to skin, liver, and/or bone marrow
(less than 10% tumor) in infants < 1 year of age.

a
The midline is defined as the vertebral column. Tumors originating on one side and crossing the midline must infiltrate to or beyond the opposite
side of the vertebral column.
Chapter 53 Pediatric Urologic Oncology: Renal and Adrenal 1091

TABLE 53.2 International Neuroblastoma Risk Group (INRG) Classification System

INRG AGE HISTOLOGIC GRADE OF TUMOR 11Q PRETREATMENT


STAGE (MONTHS) CATEGORY DIFFERENTIATION MYCN ABERRATION PLOIDY RISK GROUP
L1/L2 GN maturing A. Very low
GNB intermixed
L1 Any, except NA B. Very low
the above Amp K. High
L2 <18 Any, except NA No D. Low
the above Yes G. Intermediate
≥18 GNB nodular Differen NA No E. Low
neuroblastoma Yes H. Intermediate
Poorly differen NA H. Intermediate
or undifferen Amp N. High
M <18 NA Hyperdiploid F. Low
<12 NA Diploid I. Intermediate
12 to <18 NA Diploid J. Intermediate
<18 Amp O. High
≥18 P. High
MS <18 NA No C. Very low
NA Yes Q. High
Amp R. High

Blank field = any; Diploid – DNA index ≤ 1.0; Hyperdiploid – DNA index > 1.0 and includes near-triploid and near-tetraploid; very-low–risk 5-year EFS
>85%; low-risk 5-year EFS >75% to ≤85%; intermediate-risk 5-year EFS ≥50% to ≤75%; high-risk 5-year EFS <50%; L1 localized tumor confined to
one body compartment and with absence of image-defined risk factors (IDRFs); L2 locoregional tumor with presence of one or more IDRFs; M distant
metastatic disease except stage MS; MS metastatic disease confined to skin, liver, and/or bone marrow in children <18 months of age.
Amp, Amplified; Differen, differentiating; GN, ganglioneuroma; GNB, ganglioneuroblastoma; NA, not amplified; Undifferen, undifferentiating.

image-defined risk factors (IDRFs), which are related primarily to Clinical Variables
encasement of vessels or nerves. Metastatic tumors are termed stage
M, and MS refers to INSS stage IV-(S) in children younger than Age remains an important indicator of outcome, as originally reported
18 months of age, whereas in INSS the age cutoff was 12 months. by Breslow (Breslow and McCann, 1971). Children age 1 year or
These factors were found to be prognostic, and in 661 patients, those younger have been recognized historically to have a better survival
with L1 disease had a greater 5-year event-free survival (EFS) (90 than older children (Nitschke et al., 1988). This may be attributed
± 3%) compared with those with L2 disease (78 ± 4%, P = 0.001). to the presence of favorable biologic parameters in tumors
A further description of these guidelines has been reported by this diagnosed at this age. Retrospective reviews have suggested that age
group (Brisse et al., 2011). Although a close correlation between is a more “continuous” prognostic variable and that a cutoff of 460
presence of IDRFs and resectability of the tumor has been reported, days maximized the outcome difference between the younger and
others have not supported this finding (Günther et al., 2011; Rich older patients (London et al., 2005). More recent data from the
et al., 2011). A follow-up from the LNESG of patients with localized INRG suggest that an age at diagnosis cutoff of greater than 18
disease demonstrated that the presence of IDRFs is associated with months is associated with a higher risk of disease recurrence (Moroz
worse survival rates and higher rates of operative complications et al., 2011). Teenagers and adults with neuroblastoma have a par-
(Monclair et al., 2015). A SIOPEN study of children with unresectable ticularly indolent and relentless disease (Gaspar et al., 2003; Kushner
neuroblastoma demonstrated that the IDRFs remained unchanged et al., 2003). The site of origin is also of significance, with better
in 50% of patients receiving chemotherapy and that in fact 20% survival noted for nonadrenal primary tumors (Haase et al., 1995).
suffered the appearance of new IDRFs (Avanzini et al., 2017). They Most children with thoracic neuroblastoma are seen at a younger
also identified that children with midline perivascular IDRFs had age with localized disease and have improved survival even when
an increased incidence of operative complications but also lower corrected for age and stage (Adams et al., 1993). Tumors at this site
EFS and overall survival (OS). are less likely to have MYCN amplification and more likely to have
The International Neuroblastoma Risk Group Pretreatment Clas- a DNA index greater than 1.0, both favorable prognostic indicators
sification uses the Risk Group Classification (INRGSS), pathological (Morris et al., 1995).
findings, stage, age, and several of the biologic markers to best define Stage of the disease is a powerful independent prognostic
the child’s risk for progressive disease (Table 53.2; Cohn et al., 2009). indicator. All stage I patients with complete resection of the
Although the classification appears complex, it provides the most primary tumor survive. Stage II patients also have a favorable
accurate assessment of how intense the chemotherapy and radio- survival prospect, even though there may be incomplete excision
therapy must be to cure the child. The challenge with any staging (Matthay et al., 1989). Children with advanced regional disease,
system that uses extent of resection as a component, however, is stages III or IV, fare less well and require more intensive treatment.
that a more aggressive approach to a tumor at one institution may The proportion of patients with localized, regional, or metastatic
result in a different stage for the child than if treated at another. disease is age dependent (Nitschke et al., 1988). The OS for stages
I, II, or IV-S was a range of 75% to 90%, whereas those children
Prognostic Factors with stage IV disease had a historical 2-year disease-free survival
range of 19% to 30% despite intensive therapy, including bone
Many variables affect the prognosis of neuroblastoma. In addition marrow transplantation. The outcome for infants younger than 1
to the clinical features, there are now many biologic factors that can year of age is substantially better than for older patients with the
be used to stratify patients for treatment. same stage of disease.
1092 PART III Pediatric Urology

Stage IV-S (S meaning special) is a distinct category referring predictors and are inversely related to amplification of MYCN
to infants with small primary tumors and liver, skin, and bone oncogene (Nakagawara et al., 1993). Lack of expression of CD44
marrow metastases without radiographic evidence of bone glycoprotein on the tumor cell surface and increased levels of serum
metastases. This group of patients has a good prognosis, with OS ferritin, serum neuron-specific enolase, and serum lactate dehydro-
ranging from 80% to 88%. The INSS criteria later restricted this genase are adverse prognostic factors (Chan et al., 1991; Silber et al.,
stage to children with less than 10% bone marrow involvement 1991). They have not, however, been shown by multivariate analysis
(Brodeur et al., 1993). This stage accounts for 7% to 10% of all cases to be independently predictive from age, stage, ploidy, and MYCN
of neuroblastoma. Many of these tumors undergo spontaneous status. Telomere length has also been described as a significant
regression (Evans et al., 1987; Haas et al., 1990). In general, the prognostic parameter, and in a cohort of high-risk patients, it was
tumors in infants with stage IV-S neuroblastoma have favorable the sole significant parameter that identified a group of patients
prognostic findings not typically seen in children with stage IV disease with a favorable prognosis (Ohali et al., 2006).
(Hachitanda et al., 1991; Nickerson et al., 2000). Poor outcome in
stage IV-S patients is associated with elevated serum neuron-specific Treatment
enolase (>100 nmol/mL), ferritin (>280 ng/mL), urinary dopamine
levels (>2500 nmol/mmol creatinine), as well as MYCN amplification The treatment modalities primarily used in the management of
and chromosome 1p deletion (Schleiermacher et al., 2003). Most neuroblastoma are surgery, chemotherapy, and radiation therapy.
deaths in this group are in infants under 2 months of age with The role of each in individual patients varies depending on tumor
extensive abdominal involvement and respiratory compromise or stage, age, and biologic prognostic factors. These can be used to
disseminated intravascular coagulation (Nickerson et al., 2000). stratify patients into favorable and unfavorable categories by risk
group (see Table 53.2).
Biologic Variables
Surgery
The presence of homogeneously staining regions and double-minute
chromosomes was noted in approximately one-third of neuroblastoma The goals of surgery are to establish the diagnosis, stage the
tumors. These abnormalities are cytogenetic manifestations of gene tumor, excise the tumor (if localized), and provide tissue for
amplification, and it was subsequently found that the MYCN oncogene biologic studies. Resectability of the primary tumor should take
was mapped to these regions. The association of MYCN amplification into consideration tumor location, mobility, relationship to major
with the pathogenesis of neuroblastoma is unclear, but MYCN vessels and nerves (IDRFs), and overall prognosis of the patient.
amplification is almost always present at the time of diagnosis Neoadjuvant chemotherapy, given the efficacy of modern agents,
(Brodeur, 1991). Seeger et al. (1985, 1988) showed that MYCN is successful in reducing the size of primary tumors and enhancing
amplification is associated with rapid tumor progression and a their resectability. Sacrifice of vital structures to achieve resection
poor prognosis. Amplification is found in 5% to 10% of patients at diagnosis should be avoided, particularly in young children in
with low-stage or stage IV-S (Hachitanda et al., 1991) but in 30 whom prognosis is excellent.
to 40% of those with advanced-stage disease (Brodeur, 1990; Brodeur Low-Risk Disease (Stages I, II, and IV-S). Children with stage
and Fong, 1989). The poor prognosis associated with MYCN I neuroblastoma have a disease-free survival rate of greater than
amplification is independent of patient age or stage of disease 90% with surgical excision alone (DeBernardi et al., 1995; Nitschke
at presentation (Iehara et al., 2006). However, not all patients with et al., 1988; O’Neill et al., 1985). Chemotherapy is indicated only
a poor outcome have MYCN amplification. Many advanced-stage in the event of recurrence unless the child has MYCN amplification
tumors lack MYCN at diagnosis, and recurrence or progression of and unfavorable histology. The Pediatric Oncology Group reviewed
disease develops in most of these patients. 101 children with localized neuroblastoma who had complete gross
DNA content of tumor cells and ploidy number has been reported excision of the primary tumor (Nitschke et al., 1988). Nine patients
to have prognostic value in patients with neuroblastoma (Cohn experienced relapse, but 6 were salvaged with chemotherapy. Radiation
et al., 1990). Studies of DNA content measured by flow cytometry therapy has no role in this subset of patients. With current use of
showed that a “hyperdiploid” karyotype (or increased DNA content) the risk factor grading, those children with recurrence in the past
was associated with a favorable outcome (Kusafuka et al., 1994; may be identified now as the small number with adverse biologic
Look et al., 1984). DNA diploidy and tetraploidy were associated markers. In a comparable study from the Children’s Cancer Group,
with decreased survival. A review of 2660 children with localized 374 stage I and II patients were treated primarily by resection (Perez
neuroblastoma registered with the International Neuroblastoma Risk et al., 2000). EFS and OS for stage I patients were 93 ± 3% and 99
Group database evaluated tumor ploidy in children with MYCN– ± 1%, respectively, compared with 81 ± 4% and 98 ± 2% for stage
amplified tumors (Bagatell et al., 2009). They reported an improved II patients, respectively. Supplemental treatment was required in
prognosis in children with hyperdiploid tumors compared with only 10% of stage I and 20% of stage II patients, and excellent OS
diploid tumors. This could be used to identify children for reduction was achieved in the stage II patients. MYCN amplification, unfavorable
in the intensity of their therapy. A full INRG analytic cohort of 8800 histology, age greater than 2 years, and positive lymph nodes predicted
patients from North America, Europe, and Japan provided an extensive a lower OS. The Children’s Oncology Group (COG) conducted a
analysis of the survival implications of multiple genetic characteristics trial of asymptomatic INSS stages 2a and 2b patients treated with
(Cohn et al., 2009). Again, MYCN status was the most powerful surgery alone (Strother et al., 2012). Chemotherapy on this study
predictor of EFS and OS along with ploidy, 11q and 1p status, and was reserved for patients with symptomatic disease or in those
to a lesser extent 17q gain. considered at risk for its development, or those with less than 50%
Deletions of the short arm of chromosome 1 have been found resection with unresectable progressive disease after surgery. For all
in 70% to 80% of the near-diploid tumors that have been karyotyped 915 enrolled patients, the 5-year EFS and OS were 89% ± 1% and
(Brodeur, 1990; Brodeur and Fong, 1989). Preliminary studies suggest 97 ± 1%. In the cohort with IIb disease, the EFS and OS were sig-
a correlation between 1p deletion and poor survival (Brodeur and nificantly lower for those with unfavorable histology or diploid
Fong, 1989; Hayashi et al., 1989). Children currently treated on tumors, and OS was significantly lower for those 18 months and
protocols of the COG are assigned to a risk group that is determined older. The 5-year OS for patients with stage I and IV-S were 99% ±
by age, stage of disease, MYCN status, histologic grade, and DNA 1% and 91% ± 1%, respectively; 11.1% of patients observed after
ploidy (Katzenstein et al., 1998). Other factors that have been surgery experienced recurrence or progressive disease.
demonstrated to have prognostic significance, although they are often Radical resection resulting in removal of normal organs, particularly
associated with these genetic abnormalities, include expression of the kidney, is not justified in this group of patients. Radiation of
the gene encoding the high-affinity nerve growth factor receptor the local tumor bed has been advocated for treatment of residual
(termed TRKA proto-oncogene) and the low-affinity nerve growth disease in stage II cases. However, a review of 156 patients with stage
factor receptor (Tanaka et al., 1995). Both are favorable prognostic II neuroblastoma found a 90%, 6-year, progression-free survival rate
Chapter 53 Pediatric Urologic Oncology: Renal and Adrenal 1093

A B
Fig. 53.5. MRI before and after chemotherapy showing marked reduction in size of right suprarenal
neuroblastoma. (A) Before chemotherapy. (B) After chemotherapy.

regardless of whether radiation therapy was used (Matthay et al., tumor (Nuchtern et al., 2012) was found. Serial abdominal sonograms
1989). Therefore radiation should be reserved for those relapsed and urinary catecholamine levels were obtained during a 90-week
patients who fail to respond to either primary or secondary che- interval. A 50% increase in the volume of the mass, urine catecholamine
motherapy. In stage III disease, or in stage II with extensive tumor values, or the homovanillic acid–to–vanillylmandelic acid ratio greater
around the kidney and renal vessels, preoperative treatment with than 2 were referred for surgical resection: 87 infants were enrolled,
chemotherapy significantly decreases the risk of nephrectomy as a 83 families elected observation, and 4 chose immediate surgery.
result of resection of the tumor (Shamberger et al., 1998; Fig. 53.5). Sixteen infants on the observation arm ultimately had surgery; 8
The generally favorable behavior of IV-S disease has been explained had INSS stage I neuroblastoma, 2 had higher-stage neuroblastoma
by our current knowledge of the significance of biologic markers. (IIB and IV-S), 2 had a low-grade adrenocortical neoplasm, 2 had
The majority of these infants have tumors with entirely favorable adrenal hemorrhage, and 2 had extralobar pulmonary sequestration.
markers, explaining their favorable behavior. However, a small The 2 adrenal cortical tumors were resected because of a greater than
fraction have adverse markers; these children have progressive 50% increase in tumor volume. The 3-year EFS for a neuroblastoma
disease that often is fatal. Resection of the primary tumor is not event was 97.7% ± 2.2% within the entire cohort of infants. The
mandatory (Evans et al., 1987; Nickerson et al., 1985). Although 3-year OS was 100% with a median follow-up of 3.2 years. Following
excellent survival has been reported after surgery (Martinez et al., this protocol, 81% of the infants avoided resection, supporting the
1992), information regarding histologic prognostic factors was not role of expectant observation in this selected population. Based on
available for all of these patients. In a review of 110 infants with stage the success of the first trial with fairly restrictive entry criteria, a
IV-S disease, the entire cohort had an estimated 3-year survival rate follow-up study was developed in which infants can be enrolled
of 85% ± 4% (Katzenstein et al., 1998). This rate was significantly who are under 12 months of age at presentation with suspected
decreased to 68% ± 12% for infants whose tumors were diploid; lesions at any site that are under 5 cm in greatest diameter. For
to 44% ± 33% for those with MYCN amplification; and to 33% ± non-adrenal primary tumors, MIBG update in the tumor or elevated
19% for those with unfavorable histology. There was no statistical catecholamine levels must be present. Similar indications for surgical
difference in survival rate for infants who underwent complete intervention are used from the initial study. Until this approach is
resection of their primary tumor compared with those who had established as safe, infants should be managed in this manner only
partial resection or only biopsy (Katzenstein et al., 1998; Nickerson if enrolled in a therapeutic study.
et al., 2000; von Schweinitz et al., 2002). Patients with extensive Intermediate and High-Risk Disease (Stages III and IV). There
metastatic disease and MYCN amplification represent a high-risk is debate regarding the extent of surgical resection required for stage
group (Martinez et al., 1992). These patients should be considered III lesions. A report from the Children’s Cancer Group of 58 patients
for a more intensive treatment with multimodal therapy, according with stage III disease found that 8 of 12 patients with initial complete
to the risk group classification (see Table 53.2) (Schleiermacher et al., excision, and 12 of 14 with subsequent resection of the primary
2003). Those with favorable biologic markers and no symptoms tumor, were long-term survivors (Haase et al., 1989). This result
can be followed with supportive care and limited chemotherapy. contrasts with only 9 of 32 survivors among patients in whom
Intensive chemotherapy is reserved for those with adverse markers, complete tumor excision could not be accomplished. Significant
although these infants do poorly even with therapy. morbidity was reported in association with the surgical procedures,
Perinatal Neuroblastoma. Based on the favorable outcomes of including 21 major complications. The Italian Cooperative Group
infants identified with neuroblastoma from the screening studies for Neuroblastoma found that complete resection after chemotherapy
discussed earlier and the favorable biology of these tumors, a prospec- of extensive unresectable neuroblastoma was associated with improved
tive study was performed by the COG to assess the outcome of survival, compared with partial resection only (Garaventa et al.,
expectant observation for neuroblastoma in young infants. Infants 1993). Similar results have been noted by others (Fischer et al.,
were enrolled who were under 6 months of age with small adrenal 2017; LeTourneau et al., 1985; O’Neill et al., 1985; Powis et al.,
masses (≤16 mL in volume if solid or ≤65 mL if the mass was at 1996). It has been suggested by some that even children with stage
least 25% cystic) and no evidence of spread beyond the primary III disease do not need cytotoxic therapy if the biologic marker
1094 PART III Pediatric Urology

MYCN amplification is not present (Kushner et al., 1996). These local radiotherapy provided excellent local tumor control with no
results are not widely accepted, however, and confirmatory studies isolated local relapse in patients who had greater than 90% resection
are required before this policy can be widely adopted. (60 of 76 children).
Children with bulky pelvic tumors generally do well even with Subsequently, von Allmen et al. (2017) reviewed 220 patients
limited residual disease (LeClair et al., 2004). Extensive surgery at treated on the COG study A3973 with high-risk neuroblastoma. In
this site has been associated with long-term neurologic sequelae; this cohort the blinded radiographic central imaging review for
thus the extent of resection must be balanced against this morbidity completeness of resection was compared with the surgeon’s assess-
(Cruccetti et al., 2000). ment. Concordance between surgeon’s assessments and the radio-
A study from the COG of intermediate-risk patients attempted graphic assessment was low, 63%. Nonetheless, the clinical assessment
to decrease the intensity of therapy while maintaining outcomes of resection extent of greater than or equal to 90% (in 154 patients)
(Baker et al., 2010). Criteria for enrollment into this study included compared with resection of less than 90% (in 66 patients) after
intermediate-risk neuroblastoma without MYCN amplification, adjustment for MYCN amplification or diploidy was associated with
including infants younger than 365 days of age with stage III or IV longer EFS and lower clinical incidence of local progression but had
disease, children older than 365 days of age who had stage III tumors no effect on OS. In these nonrandomized studies, it has been difficult
with favorable histopathological features, and infants who had stage to determine whether improved survival in those with complete
IV-S disease with a diploid DNA index or unfavorable histopathologi- resection has been due to the more favorable intrinsic biology of
cal features. Children received four active agents in this study: the tumor, allowing resection or truly because of the completeness
cyclophosphamide, doxorubicin, carboplatin, and etoposide. They of resection. LaQuaglia has assessed the results in 33 non-redundant
received 4 cycles if they had favorable biology or 8 cycles if they studies, which provided adequate surgical data for analysis of survival
had unfavorable biology. Radiotherapy was used only for progressive (LaQuaglia et al., 2014). The analysis included 2599 patients for the
disease or in patients with an unresectable primary tumor with survival analysis and 412 in the analysis of local progression. Meta-
unfavorable prognostic features. The 3-year estimate of OS for the analysis revealed that the relative risk of mortality for stage III and
entire group was 96% ± 1%; 98% ± 1% for those with favorable IV patients who had more than 90% resection was 0.67 (95% CI
biologic features, and 93% ± 2% for those with unfavorable biologic 0.59–0.77) compared with those with a lesser resection. For stage
features. These findings supported the reduction in therapy for this IV patients alone the relative risk was 0.75 (95% CI 0.62–0.92). The
cohort of patients. Complete data regarding surgical intervention relative risk of local recurrence or progression in patients with stage
were available in 235 patients. Gross total resection was achieved IV disease was 0.38 (95% CI 0.27–0.53) for those undergoing extensive
in 89 patients, near total resection (>90% of the tumor) in 51, major primary resection compared with those who did not.
resection (>50% of the tumor) in 26, and limited resection (≤50% As the intensity of the therapy increases (including the use of
of the tumor) in 54. No significant difference was demonstrated autologous bone marrow transplantation) and control of distant
in OS according to the extent of resection (complete vs. incomplete; metastasis improves, the impact of maximal local control may become
P = 0.37); 28% of patients had one or more complications. apparent. The combination of gross total resection and external
A report from Memorial Sloan Kettering Cancer Center of children beam irradiation has achieved local control in 84% to 90% of children
with stage III, non-MYCN–amplified disease with gross residual (Kushner et al., 2001; Wolden et al., 2000). In another series of
disease after initial surgery demonstrated that these patients could be reports, intensive chemotherapy followed by double autologous bone
safely observed and maintained excellent OS without use of cytotoxic marrow transplantation, aggressive surgical resection, and radiotherapy
therapy (Kushner et al., 1996). Similar results were found in the has achieved an OS of 56% and a local recurrence rate of only 2.6%
German neuroblastoma 97 study, in which infants with stage II or (Marcus et al., 2003; von Allmen et al., 2005).
III MYCN non-amplified tumors who had incomplete resections Usually the safest approach for advanced tumors is to defer
were observed after surgery without chemotherapy. Regression of resection until after initial chemotherapy (Berthold et al., 1989;
the residual masses without additional treatment was seen in 32 of Black et al., 1996; LaQuaglia et al., 1994; Shamberger et al., 1991;
55 patients. Tumors that recurred or progressed were responsive to Shochat, 1992). The tumors are smaller and firmer, with less risk
therapy with a 3-year OS rate of 98% (Hero et al., 2008). of rupture and hemorrhage after chemotherapy, resulting in a
Unfortunately, more than half of all newly diagnosed patients decreased rate of complications, particularly nephrectomy (Sham-
with neuroblastoma have high-risk disease. There is conflicting berger et al., 1998; see Fig. 53.5). One specific complication
evidence regarding the benefit of extensive resection in children with encountered after resection of extensive tumor surrounding the celiac
stage IV disease between studies that support (Adkins et al., 2004; axis and the superior mesenteric artery is diarrhea (Kiely, 1994). It
Chamberlain et al., 1995; DeCou et al., 1995; Haase et al., 1991; is thought to result from resection of the autonomic nerves to the
Koh et al., 2005; Kuroda et al., 2003; LaQuaglia et al., 1994, 2004; gut found anterior to the aorta at the base of the superior mesenteric
LeTourneau et al., 1985; McGregor et al., 2005; Tsuchida et al., 1992; artery and the celiac axis (Rees et al., 1998). The second frequent
Vollmer et al., 2017; von Allmen et al., 2005; Yokoyama et al., 1995) complication after extensive resection is chylous ascities resulting
and those that refute that approach (Adams et al., 1993; Castel et al., from resection of the involved retroperitoneal lymph nodes. This is
2002; Kaneko et al., 1997; Kiely, 1994; Losty et al., 1993; Matsumura generally a self-limited problem, but initial extensive abdominal
et al., 1988; McGregor et al., 2005; Simon et al., 2013; Sitarz et al., distention may require intervention (Qureshi et al., 2016). Preopera-
1983; von Schweinitz et al., 2002). In a retrospective review, Kiely tive chemotherapy does appear to increase the proportion of children
(1994) compared the results of radical tumor resection with those able to achieve a complete resection (Adkins et al., 2004).
of more conventional surgery in patients with stages III and IV disease. Surgery usually is performed 13 to 18 weeks after initiation of
He found no difference in survival between 46 patients treated with chemotherapy, allowing three to four courses of treatment (Azizkhan
radical surgical procedures and 34 patients treated with more and Haase, 1993). Some tumors remain inoperable even after
conventional surgery. Shorter et al. (1995) also did not find any chemotherapy.
evidence that the extent of surgical resection had an impact on the Infants under 1 year of age with extensive local disease or stage
survival of stage IV patients. Simon et al. (2013) reported the results IV disease make up a special subset of patients. They have historically
of the German cooperative trial NB97 that evaluated the role of fared much better than children older than 1 year of age with
surgery (upfront and post-neoadjuvant therapy) in patients older comparable disease, but not as well as infants with stage IV-S disease.
than 18 months with stage IV disease: 278 patients were evaluated. It is now recognized that biologic markers can be used to identify
Their data suggested that neither upfront surgery nor operations which infants have high-risk disease and require intensive therapy
performed post neoadjuvant therapy produced a positive impact on and which will have intermediate-risk disease requiring less intensive
EFS, local progression-free survival, or OS. In contrast, von Allmen therapy. The SIOPEN Infant Neuroblastoma European Study reported
et al. (2005) reported on the local control of 76 patients treated at 120 infants with localized tumors and no MYCN amplification who
two institutions with the same treatment protocol and a consistent were deemed unresectable based on IDRFs (Rubie et al., 2011). These
surgical approach. They found that aggressive surgery followed by infants were treated with low-dose cyclophosphamide and vincristine
Chapter 53 Pediatric Urologic Oncology: Renal and Adrenal 1095

(VCR) repeated once to three times every 2 weeks until surgical et al., 2013). This study was terminated when interim analysis showed
excision could be safely performed. Infants with either one threatening no difference in outcome between the two groups. A further study
symptom or an insufficient response were given carboplatin and addressed the benefit between a single versus a paired autologous
etoposide (CaE) sometimes followed by vincristine, cyclophospha- transplant to see if additional intensification would improve outcome.
mide, and doxorubicin. Seventy-nine received the two-drug therapy It was demonstrated that tandem versus single myeloablative therapy
with VCR and 49 received CaE because of insufficient response. improves 3-year EFS in children with high-risk neuroblastoma (Park
Thirty-two infants had threatening symptoms and of these 30 received et al., 2016).
CaE. Anthracyclines were used in 46 infants. Surgery was performed Recent trials in COG have piloted a strategy of using non–cross-
in 102 infants leading to gross surgical excision in 93. Relapse occurred resistant chemotherapy to further improve induction response rates.
in 12 patients (9 local and 3 distant). The 5-year OS and EFS were Topotecan, a topoisomerase-1 inhibitor has shown activity against
99% ± 1% and 90% ± 3%, respectively. The low-dose chemotherapy neuroblastoma in phase I and II trials. In a COG feasibility trial
without an anthracycline was effective in 62% of these infants with high-dose topotecan was added to the Memorial Sloan-Kettering
an unresectable tumor and no MYCN amplification. induction backbone (Park et al., 2011). Thirty-one patients were
In a large cohort (134 infants), MYCN amplification, serum ferritin, enrolled and the combination was well tolerated without unexpected
Shimada histopathologic classification, and bone marrow involvement toxicity. Fifteen of 31 patients (48%) achieved a complete or very
by immunocytology were analyzed. Although each factor had good partial response. This regimen was added in a non-randomized
prognostic significance by univariate analysis, only MYCN amplifica- fashion in the large phase III trial between single and tandem
tion was significant by multivariate analysis; the EFS for infants transplantation.
without MYCN amplification was 93% ± 4% versus 10% ± 7% in The presence of osseous disease has long been recognized as an
those with amplification despite intensive therapy (Schmidt et al., adverse prognostic finding, particularly after induction therapy. A
2000). multivariate analysis of 549 patients with high-risk neuroblastoma
Clinical trials in Europe evaluated these infants without MYCN in the European Bone Marrow Transplantation Solid Tumor Registry
amplification and achieved excellent survival (OS of 97.6% at 2 demonstrated that persistent cortical bone lesions (P = 0.004) and
years) without chemotherapy in those with primaries extending across bone marrow involvement (P = 0.03) were the only independent
the midline or a positive skeletal survey (DeBernardi et al., 2009). adverse prognostic factors (Ladenstein et al., 1998). More recent
Infants with overt metastases to the skeleton, lung, and central nervous retrospective studies of patients treated on COG A3973 assessed a
system (CNS) were treated with a minimum of four chemotherapy “semiquantitative” MIBG score using the Curie scoring system. Patients
courses and achieved a 2-year OS of 95.6%. who had a Curie score greater than 2 at the end of induction (n =
52) had a significantly decreased EFS compared with patients who
Chemotherapy had Curie scores of 2 or less (3-year EFS 15.4% ± 5.3% vs. 44.9%
± 3,8%, P < 0.001) (Yanik et al., 2013). This finding was particularly
A variety of multiagent treatment regimens have been developed to stark for patients with MYCN amplification (6 patients), in whom
treat high-risk patients with neuroblastoma that use a combination 3-year EFS was 0.
of intensive induction therapy, myeloablative consolidation therapy The presence of bulky disease results in increased failure. Tumor
with stem cell support, and biologic therapy to control minimal debulking with surgery or radiation therapy is warranted before
residual disease. The goal of this treatment intensification in a autologous bone marrow transplantation. There are many questions
sequence of protocols is better disease control, and 5-year survival yet to be resolved about this modality of treatment. Toxicity of bone
rates have gone from less than 15% to 30% to 40% (Philip et al., marrow transplantation can be lethal, and the long-term complications
1997). Although initial response rates are improving, with a prolonged in patients with successful transplantation are unknown. However,
time to progression of disease, relapse continues to be a major these risks are acceptable given that long-term survival is difficult
problem. A meta-analysis of 44 trials showed a correlation between to achieve without such intensive therapy.
dose intensity and treatment efficacy including improved response Additional new agents in phase I and II trials for relapsed neu-
and survival (Cheung et al., 1991). roblastoma include temozolomide and irinotecan (Kushner et al.,
An induction regimen developed at Memorial Sloan-Kettering 2006; Rubie et al., 2006; Simon et al., 2007). As efficacy is established,
Cancer Center has been widely used in subsequent protocols. It used they will be advanced into clinical trials for high-risk tumors with
cycles of cyclophosphamide, vincristine, and doxorubicin alternating a goal to improve the overall outcome.
with cycles of cisplatin and etoposide. In their single institution
study Kushner et al. (1994) reported complete response rate of 63% New Innovative Biologic Therapies
and a complete plus very good partial response rate of 87% in 24
patients. The dose intensification of chemotherapy required for local Double autologous bone marrow transplantation has allowed the
tumor control results in significant myelosuppression, limiting the use of myeloablative therapies, but the maximum benefit to that
amount of therapy that can be given. This has prompted the use of approach has been reached. 13-cis-retinoic acid produces differentia-
autologous bone marrow transplantation after sublethal chemotherapy tion of neuroblastoma in cell cultures including cultures of recurrent
or total-body irradiation. The use of marrow-ablative chemoradio- tumors. It was given for a 6-month period after cytotoxic therapy
therapy followed by autologous marrow reinfusion has resulted in in children with advanced-stage disease and significantly decreased
complete remission in up to 50% of patients with stage IV disease the frequency of relapse (Matthay et al., 1999). Long-term follow-up
(Dinndorf et al., 1992; Grupp et al., 2000; Matthay et al., 1995; has shown that the 5-year OS was improved (Matthay et al., 2009).
Moss et al., 1987; Mugishima et al., 1994; Seeger et al., 1991). Antibody therapy against the GD2 disialoganglioside cell surface
However, a significant problem is the risk for late relapse. The random- marker uniformly expressed in neuroblastoma has been shown to
ized clinical trial of the German Society of Paediatric Oncology and be an effective adjuvant therapy in combination with granulocyte-
Hematology of high-risk neuroblastoma showed improved EFS at macrophage colony-stimulating factor and interleukin-2 along with
3 years with myeloablative therapy and autologous stem cell rescue 13-cis-retinoic acid after autologous bone marrow transplant compared
compared with maintenance chemotherapy (Berthold et al., 2005). with 13-cis-retinoic acid alone (Yu et al., 2010). More recent studies
These results were similar to those of the COG study (Matthay et al., have combined antibody therapy with a “backbone” of irinotecan
1999). Unfortunately, the OS rates were not statistically different in and temozolomide, a pair of newer agents not included in front-line
either study at initial report. An update of the latter study, however, therapy. In one study these agents were combined with either antibody
reveals that the 5-year OS of the patients receiving the autologous to GD2 or temsirolimus, an mTOR inhibitor that has a role in regula-
bone marrow transplant was improved (Matthay et al., 2009). tion of protein synthesis and cell proliferation (Mody et al., 2017).
A subsequent study by the COG addressed the issue of the need In this randomized study of relapsed patients the combination with
for “purging” of the bone marrow before return to the patient to the antibody to GD2 was effective but the mTOR inhibitor had a
remove any potential remaining neuroblastoma cells (Kreissman very low response rate. Newer “humanized” antibodies are also being
1096 PART III Pediatric Urology

tested to see if they are more effective and if they can minimize the
pain produced by the current antibody, which can limit the extent
KEY POINTS: NEUROBLASTOMA
of treatment. Studies are also in preparation to evaluate the use of • Neuroblastoma is the most common e tracranial solid
the GD2 antibody in front-line therapy. neoplasm in children.
A new synthetic retinoid, fenretinide, which has produced apoptosis • hildren 1 months of age or younger have a better
rather than differentiation in neuroblastoma cell lines, is also in survival rate than older children. This may be attributed to
clinical trials for maintenance therapy. It has been shown to be more favorable biologic parameters in tumors diagnosed
effective against some neuroblastoma cell lines that are resistant to at this age.
13-cis-retinoic acid. • omplete surgical resection is curative for low-stage
Other avenues of treatment in current phase I and phase II trials disease.
include vaccine and antiangiogenic therapy. Vaccines have been • MYCN amplification, found in 20% of patients with
developed against the GD2 antigen, but they are in early stages of primary tumors, is associated with tumor progression. This
testing. Inhibition of angiogenesis is another appealing avenue of poor prognosis is independent of patient age and tumor
therapy in this very vascular tumor, particularly once there is minimal stage. Additional biologic markers of poor prognosis have
residual disease, although efficacy has not yet been established in been defined and treatment protocols are currently based
phase 1 or 2 studies. on these factors.
Another modality in the treatment of metastatic neuroblastoma
is the use of 131I-MIBG (Hutchinson et al., 1992). The finding that
the primary tumor and metastatic areas take up this radiotracer
suggested the possibility that therapeutic doses can be delivered to of large randomized clinical trials in North America and Europe.
the tumor. Preliminary analysis indicates that objective responses The focus of the most recently completed trials was on reducing the
do occur in terms of reduction of tumor volume, even in previously morbidity of treatment for low-risk patients, reserving more intensive
heavily treated patients (Howard et al., 2005; Matthay et al., 2007). treatment for high-risk patients for whom survival remains poor. This
Significant myelosuppression is seen with dose escalation, however, section outlines current recommendations for treatment and reviews
and stem cell support is required. It has not yet been tested as an the latest developments in the biology of Wilms tumor.
“up-front” therapy.
Epidemiology
Radiotherapy
Wilms tumor accounts for approximately 6% to 7% of all childhood
Radiotherapy is effective for local control in neuroblastoma, and cancers. It is the most common renal tumor of childhood, accounting
risk of local relapse can be correlated with the biologic markers. for 95% of all kidney cancers in children under the age of 15 in the
Although irradiation has not provided a benefit in low-stage tumors, United States (Ali et al., 2012; Howlader et al., 2013). According to
it has increased local control in children with advanced stage IV or the Surveillance, Epidemiology, and End Results (SEER) registry, the
bulky stage III tumors (Castleberry et al., 1991; Evans et al., 1996; average annual age-adjusted incidence rate of Wilms tumor is 8.0
Matthay et al., 1989). A randomized control trial to evaluate the per million (Bernstein et al., 1999; Breslow et al., 1993). More than
efficacy of local control between radiotherapy alone and surgery has 80% of cases are diagnosed before 5 years of age, with a median
not been performed. Doses of external beam irradiation used have age of 3.5 years. Nevertheless, older children and occasionally even
ranged between 15 and 30 Gy, depending on the patient’s age, location adults can be affected (Ali et al., 2012; Arrigo et al., 1990; Kalapurakal
of the tumor, and extent of residual disease. et al., 2004a). The median age at diagnosis is lower in children with
Intraoperative radiation therapy has been used for patients with bilateral Wilms tumor (Breslow et al., 1993) and those with a
unresectable disease. This technique has the advantage of delivering syndromic predisposition to Wilms tumor (Kalish et al., 2017). Wilms
a higher dose of radiation to the operative field while sparing normal tumor presents at an earlier age among males with both unilateral
adjacent tissues (Leavey et al., 1997). Although its use has been and bilateral tumors.
promoted, it has not been convincingly demonstrated to improve In North America, Wilms tumor is slightly more frequent in
control when compared with external beam irradiation (Haas-Kogan females than in males. With regard to ethnicity, the incidence of
et al., 2000). Wilms tumor is lower in East Asian populations and higher in black
populations compared with the incidence reported for North American
Spinal Cord Compression and European Caucasians (Axt et al., 2011; Breslow et al., 1994;
Fukuzawa et al., 2004). The fact that such variations exist more
Extension of tumor into the spinal canal produces symptoms of closely associated with race than geography suggests that environ-
spinal cord compression in up to 5% of patients presenting with mental risk factors likely play a minor etiologic role, certainly in
neuroblastoma (DeBernardi et al., 2001), whereas up to 13% of comparison with adult epithelial cancers (Breslow et al., 1993). Black
patients have radiographic evidence of extension into the spinal children often have a more advanced stage of disease at presentation,
canal (Plantaz et al., 1996). These children have been treated by but it is not clear if this is related to tumor biology or reflects delays
decompressive laminectomy, radiotherapy, or chemotherapy. Neu- in diagnosis resulting from impaired access to health care (Axt et al.,
rologic outcome has been similar by all modalities and, unfortunately, 2011). Data also exist on the very poor survival outcomes of children
patients presenting with severe motor deficits generally recover little in Africa with Wilms tumor; however, as with the data on black
function (DeBernardi et al., 2001; Katzenstein et al., 2001). Because children in North America it is still unclear what contribution tumor
of the delayed complications of scoliosis after laminectomy, current biology plays relative to other known barriers to care (Paintsil et al.,
recommendations are to initiate treatment with chemotherapy and 2015). Several epidemiologic studies have investigated occupational,
reserve laminectomy for children with progressive neurologic deteriora- environmental, and lifestyle factors as risk factors for Wilms tumor.
tion (Katzenstein et al., 2001). Radiotherapy is now generally avoided, Although some studies have suggested that a number of parental
because of its adverse effect on growth of the spine. and prenatal exposures may be associated with an increased risk of
Wilms tumor, very few have been established conclusively (Breslow
et al., 1993; Shrestha et al., 2014).
WILMS TUMOR
Biology/Genetics
Wilms tumor, or nephroblastoma, is the most common primary
malignant renal tumor of childhood. It is an embryonal tumor that Children with Wilms tumor have been extensively studied to determine
develops from remnants of immature kidney. Treatment of patients the role of genetic alterations in tumor development (Gadd et al., 2012).
with nephroblastoma has been extensively investigated in a number We now recognize that the majority of Wilms tumors arise from somatic
Chapter 53 Pediatric Urologic Oncology: Renal and Adrenal 1097

TABLE 53.3 Gene Mutations in Wilms Tumors The WT1 gene is important for normal kidney and gonadal
development. WT1 encodes a zinc finger transcription regulating
SOMATIC OR mesenchymal to epithelial transition in kidney development (Dressler,
GENE (CHROMOSOME) FREQUENCY GERMLINE 1995; Pritchard-Jones, 1990). WT1 is necessary for ureteric bud
outgrowth and is also important in nephrogenesis. Targeted mutation
IGF2 (11p15) 70% Both of the WT1 gene in mice results in failure of kidney and gonadal
WTX (Xq11) 20% Somatic only development (Kreidberg et al., 1997). The Denys-Drash syndrome
WT1 (11p13) 20% Both (DDS) is the specific association of male pseudohermaphroditism,
CTNNB1 (3p21) 15% Somatic only renal mesangial sclerosis, and nephroblastoma (Drash et al., 1970).
TP53 (17p13) 4% all tumors Both The syndrome is caused by point mutations in the zinc-finger DNA
70% anaplastic binding region of WT1 (Coppes et al., 1993). More than 90% of
DDS patients harbor germline point mutations in only one WT1
allele (Coppes et al., 1992; Pelletier et al., 1991). Therefore the WT1
mutation acts dominantly with respect to genitourinary abnormalities.
The fact that the phenotype resulting from these heterozygous muta-
mutations restricted to tumor tissue and a much smaller percentage tions is far more severe than that resulting from constitutional deletion
originate from germline mutations (Dome and Huff, 2016; Ruteshouser of one WT1 allele (i.e., WAGR patients) suggests that the Denys-Drash
et al., 2008; Scott et al., 2012). It is apparent that several genetic events mutations do not result in inactivation of the WT1 protein, but
result in Wilms tumorigenesis and that the Knudson two-hit model of rather in the production of a dysfunctional WT1 protein. It is
cancer formation does not explain most cases (Knudson and Strong, postulated that this abnormally expressed protein alters regulation
1972). In 10% to 15% of individuals with Wilms tumor, the cause of transcription and urogenital development. The majority of these
is considered to be a germline pathogenic variant or an epigenetic patients progress to end-stage renal disease. Nephropathy usually
alteration occurring early during embryogenesis (see 11p15-Related occurs early in life and renal biopsy demonstrates mesangial sclerosis.
Wilms Tumor). These may or may not be associated with a known Some WT1 mutations can lead to an incomplete DDS phenotype,
congenital malformation syndrome or hereditary cancer syndrome. with varying degrees of genitourinary anomalies, renal pathologies,
Approximately 1% to 2% of individuals with Wilms tumor have at and penetrance of Wilms tumor (McTaggart et al., 2001). Although
least one relative also diagnosed with Wilms tumor (familial Wilms XY individuals have been reported most often, the DDS has been
tumor); however, although germline variants that are likely pathogenic reported in genotypic/phenotypic females. WAGR and DDS patients
have been identified in some families, they are still not known for are more likely to have bilateral tumors and are diagnosed at a
the majority of individuals. The most commonly reported germline younger age (Breslow et al., 2003, 2006).
genetic and epigenetic variants in individuals with Wilms tumor This risk for renal insufficiency in DDS is shared by children with
involve WT1 and the 11p15.5 locus. A growing number of variants WAGR and children with genitourinary abnormalities, all associated
in other genes have been reported. In some cases, the estimated of with a WT1 mutation (Diller et al., 1998). WAGR patients and those
risk of Wilms tumor with these variants is high, but the variant or with genitourinary abnormalities have an increased risk of renal
syndrome is so rare in the general population that only a handful of failure if they survive into puberty (Breslow et al., 2000, 2005).
individuals with Wilms tumor have ever been reported (Dome and Genitourinary anomalies (renal fusion anomalies, cryptorchidism,
Huff, 2016). A number of genes have been identified that play a role hypospadias) are present in 4.5% of patients with Wilms tumor
in the development of Wilms tumor (Table 53.3). (Breslow et al., 1993). The incidence of WT1 mutations in patients
with Wilms tumor not associated with any genitourinary abnormality
WT1 is approximately 2% (Little et al., 2004). Studies of non-tumoral
renal tissue in children with WAGR and DDS have demonstrated
The first gene mutation described in Wilms tumor was WT1 with smaller glomerular diameters than controls (Dahan et al., 2007). This
somatic and germ-line mutations noted. The identification and suggests a specific defect of WT1 function during renal development
subsequent cloning of WT1 resulted from cytogenetic observations that leads to subsequent deterioration in renal function with age.
of gross deletions at chromosome 11p13 in patients with the WT1 was originally considered to be a classic tumor suppressor
WAGR (Wilms tumor, Aniridia, Genital anomalies, mental Retarda- gene, and the loss of both copies or mutations of this gene would
tion) syndrome. These children were shown to have heterozygous lead to Wilms tumor development (Rauscher, 1993). Although this
germline deletions at 11p13 (Riccardi et al., 1978). Subsequent may be the case for some tumors, only 20% of patients with Wilms
molecular analysis of the DNA mapping of this specific region led tumor have a mutation in the germline or in tumor tissue (Diller
to the identification of the WT1 in 1990 (Bonetta et al., 1990; Call et al., 1998; Ruteshouser et al., 2008). It is well established that
et al., 1990). WT1 mutations are often associated with β-catenin most constitutional cancer syndrome mutations, including WT1,
(CTNNB1) mutations, defining a specific genetic subset of Wilms are associated with younger age of onset. The median age of onset
tumor (Li et al., 2004; Royer-Pokora et al., 2008). This subset has in children with somatic WT1 mutations was 14 months. It is
been called the ideal type I Wilms tumor (Breslow et al., 2006). It postulated that WT1 mutations are associated with a more rapid
is characterized by a stromal predominant favorable histology, progression to Wilms tumor than other molecular abnormalities
intralobar nephrogenic rests, early onset of Wilms tumor, and geni- (Scott et al., 2012).
tourinary abnormalities in males. Wilms tumors from patients with
Wilms tumor-aniridia-urogenital abnormalities-mental retardation WTX
(WAGR) and Denys–Drash syndrome (DDS) belong to this subtype
of tumors. Another tumor suppressor gene, WTX, is targeted by somatic mutations
Aniridia, found in 1.1% of Wilms tumor patients, arises from in up to 30% of cases of Wilms tumor, the most common type of
deletion of PAX6, which lies within approximately 0.6 Mb of WT1 pediatric kidney cancer (Rivera et al., 2007). Most tumors have
(Vasilyeva et al., 2017). Aniridia patients with visible (microscopic) deletions of the entire WTX gene, but one-third of WTX-mutated
chromosomal deletions that include WT1 were noted to be prone Wilms tumors carry truncating mutations or missense mutations.
to the development of Wilms tumor (Muto et al., 2002). FISH analysis The functional importance of the missense alterations is unclear,
of aniridia patients has been used to identify submicroscopic deletions because the missense alteration can be present in normal tissue from
of WT1 and is also a useful predictor of Wilms tumor development the same patient, whereas the deletion and truncation mutations
(van Heyningen et al., 2007). Wilms tumor will develop in approxi- are always specific to the tumor. The location of WTX on the X
mately 40% to 70% of aniridia patients with deletions of WT1. chromosome is of particular interest because it is inactivated by a
Conversely, aniridia patients with normal WT1 do not develop Wilms monoallelic, or “single-hit” event rather than by the classical biallelic
tumor (Grenskov et al., 2001; van Heyningen et al., 2007). inactivation of autosomal tumor suppressor genes. It targets the
1098 PART III Pediatric Urology

single X chromosome in males and the active X chromosome in are detected in 50% to 75% of patients with anaplastic histology Wilms
females with tumors and occurs at comparable frequencies in males tumor (Bardeesy et al., 1994, Ooms et al., 2016). It has been suggested
and females alike. Initially it was reported that tumors caused by that a favorable histology tumor may progress to an anaplastic tumor
WTX mutations lack WT1 mutations. Subsequently, it has been by acquiring a disruption of TP53 function in a group of selected cells
observed that WTX mutations occur with equivalent frequency in (Natrajan et al., 2007). The TP53 mutations also have been correlated
tumors with or without mutations in WT1 and CTNNB1 (Ruteshouser with advanced stage disease (Malkin et al., 1990; Sredni et al., 2001). A
et al., 2008). WTX, like WT1, appears to play a role in the Wnt/ recent report from COG on TP53 mutations in diffuse anaplastic Wilms
beta-catenin signaling pathway. Mutations in WT1, WTX, and CTNNB1 tumor noted improved relapse-free survival in those with advanced
provide the genetic basis for about one-third of Wilms tumors stage disease who had wild-type TP53 (Ooms et al., 2016).
(Bahrami et al., 2018). Loss of the long arm of chromosome 16 has been found in
approximately 20% of Wilms tumors (Maw et al., 1992), suggesting
11p15 the presence of a gene at 16q involved in the biology of Wilms
tumor. Similarly, loss of the short arm of chromosome 1p has been
Additional molecular alterations are observed at the chromosome found in approximately 10% of cases (Grundy et al., 1994). LOH
11p15 locus, which contains a cluster of imprinted genes (Koufos at 1p and 16q are associated with an increased risk of tumor
et al., 1989; Mannens et al., 1990). LOH or loss of imprinting (LOI) relapse and death (Grundy et al., 1994, 2005; Wittman et al., 2007).
on 11p15 occurs in up to 70% of tumors (Scott et al., 2012). A WT2 Confirming the utility of LOH of 16q and 1p to predict outcome
locus has been postulated in 11p15, but so far, the respective gene was one of the major objectives of the fifth National Wilms Tumor
has not been identified. This region is known to harbor genes for Study (NWTS) (see discussion later). LOH of chromosome 11q is
the Beckwith-Wiedemann syndrome (BWS) and other syndromes noted in approximately 20% of tumors. LOH of 11q is 3 to 4 times
with overgrowth features (Koufos et al., 1989). These include more frequent in anaplastic tumors (Wittman et al., 2007). There
hemihypertrophy, which may occur alone or as part of the BWS, is a correlation with tumor recurrence and death but only for tumors
Perlman, Soto, and the Simpson-Golabi-Behmel syndromes (Neri that lost the entire long arm of chromosome 11. Combined LOH
et al., 1998; Perlman et al., 1975). Most cases of BWS are sporadic, but (at 1p and 16q) was used to risk-stratify patients with favorable-
up to 15% exhibit heritable characteristics with apparent autosomal histology Wilms tumor (FHWT) on the most recently completed
dominant inheritance. The BWS critical region on 11p15 includes two COG renal tumor studies. COG Study AREN 0533 intensified therapy
domains of imprinted genes located in proximity in band 11p15. for those stage III and IV FHWT and demonstrated that therapeutic
Imprinting center region 1 (ICR1) regulates the expression of IGF2 and intensification improved EFS and OS (Dix et al., 2015; Fernandez
H19; and center 2 (ICR2), which regulates the CDKN1C, KCNQ10T1, et al., 2018). However, this is of limited clinical importance because
and KCNQ1. These imprinting centers are differentially methylated only approximately 5% of all FHWT patients have combined LOH.
on the paternal and maternal allele, leading to expression of only Gain of 1q has also been identified to be another genetic change
one parent-specific allele (Choufani et al., 2013). associated with outcome (Gratias et al., 2013, 2016; Segers et al.,
Wilms tumor associated with epigenetic alterations in the 11p15 2013). 1q gain has the potential to be a more clinically significant
region has been termed the ideal type II Wilms tumor (Breslow factor because it is consistently noted in one-fourth of FHWTs. It is
et al., 2006). A type II Wilms tumor is characterized by a limited important to note that 1q gain and LOH at 1p and 16q are not
nephrogenic differentiation and a mostly blastemal or epithelial-type independent events. LOH at 1p and 16q often arises through
histology; patients develop tumors at a later age and have a heavier chromosomal translocations that also result in 1q gain. After stratifica-
birth weight. The risk of nephroblastoma in children with BWS tion for stage of disease, 1q gain was associated with a significantly
and hemihypertrophy is estimated to be 4% to 10%; 21% of increased risk of disease recurrence and worse OS (Gratias et al.,
children have bilateral disease (Beckwith, 1996; Debaun and Tucker, 2016). Future studies will likely investigate the ability of 1q gain to
1998; Porteus et al., 2000). The mean age at diagnosis of Wilms be used to risk-stratify patients for therapeutic intensification.
tumor in BWS and hemihypertrophy patients is similar to that of COG investigators have evaluated a large cohort of FHWTs for
the general Wilms tumor population (Breslow et al., 1993). Adre- global gene expression patterns; WT1, CTTNNB1, and WTX mutation;
nocortical neoplasms and hepatoblastoma also occur with increased and 11p15 copy number and methylation patterns. They identified
frequency in BWS. Children with BWS found to have nephromegaly five subsets of tumors showing distinct differences in their clinical
(kidneys greater than or equal to the 95th percentile of age adjusted and pathological features. One unique subset was epithelial Wilms
renal length) are at the greatest risk for the development of Wilms tumor in infants that lacked WT1, CTTNNB1, and WTX mutations
tumor (Debaun et al., 1998). Investigators have been able to correlate and nephrogenic rests, and none had recurrence of tumor. COG
genetic changes to identify which BWS patients are at risk for Wilms investigators hope to use this type of information to develop specific
tumor development (Bliek et al., 2004; Brioude et al., 2013; DeBaun treatment strategies for different subsets of patients based on biology
et al., 2002). An increased Wilms tumor risk is observed only in (Gadd et al., 2012; Sredni et al., 2009).
BWS patients with ICR1 gain of methylation and 11p15 uniparental
disomy. In contrast, the tumor risk is lower in BWS with ICR2 loss Screening
of methylation and CDKN1C mutations.
Screening with serial renal sonograms has been recommended
Familial Wilms Tumor in children at high risk for development of Wilms tumor. Review
of most studies suggests that 3 to 4 months is the appropriate
As noted earlier, 1% to 2% of Wilms tumor patients have a family screening interval. The frequency of screening does not change as
history of Wilms tumor (Breslow et al., 1996; Ruteshouser and Huff, the patient ages; the rate of tumor growth is expected to be the same
2004). Familial cases have an earlier age of onset and an increased in older children. Tumors detected by screening will usually be at
frequency of bilateral disease. Two familial Wilms tumor genes have a lower stage (Choyke et al., 1999; Green et al., 1993; Kalish et al.,
been localized (Ruteshouser and Huff, 2004). FWT1 is located at 2017). No studies have demonstrated that early detection has
17q12-q21 and FWT2 at 19q13.4 (McDonald et al., 1998; Rahman improved patient survival. Early detection can provide an opportunity
et al., 1996). Penetrance of these genes appears to be moderate, and for nephron-sparing surgery, because these children are at an increased
these genes do not follow the typical tumor suppressor gene pattern risk for bilateral disease. The smaller tumors found on screening
of loss of heterozygosity (Strong, 2003). studies are more amenable to renal-sparing surgery (Romao et al.,
2012; Fig. 53.6). A recent report from the American Association for
Other Chromosomal Abnormalities Cancer Research Childhood Cancer Predisposition Workshop recom-
mended that screening be performed when a condition has a Wilms
Mutations of the tumor suppressor gene TP53 are frequently encountered tumor incidence of greater than 1% (Table 53.4; Kalish et al., 2017).
genetic events in human cancer (Hollstein et al., 1991). TP53 mutations Others have recommended screening when the Wilms tumor incidence
Chapter 53 Pediatric Urologic Oncology: Renal and Adrenal 1099

A B
Fig. 53.6. Small renal tumor found on serial screening. (A) Ultrasound shows solid lesion just pushing
out from the cortex. (B) CT demonstrates that lesion is amenable to renal-sparing surgery.

TABLE 53.4 Syndromes Associated With Development of Wilms Tumor

SYNDROME GENES LOCUS WILMS TUMOR RISK


HIGH RISK
WAGR WT1 11p13 50%
Denys-Drash WT1 11p13 50%
Fanconi anemia D1 BRCA2 13q12.3 >20%
Perlman Unknown >20%
Mosaic variegated aneuploidy BUB1B 15q15 >20%

MODERATE RISK
Beckwith-Wiedemann WT2 IGF2, H19 5%–10%
Hemihypertrophy WT2 IGF2, H19 5%
Frasier WT1 11p13’ 5%–10%
Simpson-Golabi-Behmel GPC3 Xq26 10% (in males)

LOW RISK
Li-Fraumeni p53 17p13 Low
Neurofibromatosis NF1 17q11 Low
Sotos NSD1 5q35 Low
Trisomy 18 Unknown 18 Low
Bloom BLM 15q26 Low

is greater than 5% (Scott et al., 2006a,b). Although there is emerging CT or MRI should be performed if ultrasonography demonstrates
evidence of different cancer risks based on genetic or epigenetic a suspicious lesion. Nonmalignant renal lesions—for example, renal
subgroups for certain syndromes, and several European countries cysts—do occur at an increased rate in children with BWS, and recogni-
now use subgroup-specific recommendations for WT screening tion of these is important to avoid unnecessary nephrectomy when
particularly in BWS, these practices have not yet been adopted in new lesions are identified on screening ultrasonography (Borer et al.,
North America (Mussa et al., 2016). Ultrasound surveillance is recom- 1999; Choyke et al., 1999).
mended until 7 years of age. For patients with BWS/IHH, trisomy
18, and SGBS, screening for hepatoblastoma is also required. They Pathology
need full abdominal ultrasound and simultaneous serum AFP
screening every 3 months starting at birth (or at the time of diagnosis) Pathologists have made important contributions to the study of the
and continuing through the child’s fourth birthday. clinical behavior and biology of Wilms tumor (Beckwith and Palmer,
Screening of the contralateral kidney after nephrectomy for 1978; Gadd et al., 2012, 2017; Perlman et al., 2011; Ravenel et al.,
unilateral Wilms tumor is also recommended (D’Angio et al., 1993). 2001; Schmidt and Beckwith, 1995; Vujanic et al., 2002, 2010; Weeks
Infants younger than 12 months found to have nephrogenic rests et al., 1987; Zuppan et al., 1991). Wilms tumor is characterized by
(NRs) in the resected kidney are at the greatest risk for metachronous wide histologic diversity, and thus the classification of these childhood
tumors (Coppes et al., 1999). Ultrasound surveillance is performed tumors can be difficult. As noted in the discussion earlier, correlation
from time of diagnosis until 5 years of age, with a frequency of every of the pathologic findings with genetic events is improving our
3 to 4 months. understanding of the development of Wilms tumor.
1100 PART III Pediatric Urology

et al., 1985; Green et al., 1994). Anaplasia is associated with


resistance to chemotherapy. The presence of anaplasia has clearly
been demonstrated to carry a poor prognosis even when the
tumor is apparently confined to the kidney, stage I (Dome et al.,
2006). Anaplasia has been further divided into focal and diffuse
patterns to reflect further the different prognosis of anaplasia that
is present throughout the kidney or in an extrarenal location (Faria
et al., 1996). The later age at diagnosis and the general absence of
anaplasia from nephrogenic rests suggests that anaplasia develops
from Wilms tumor cells that acquire additional genetic lesions
(Williams et al., 2011). As mentioned earlier, approximately 50% of
anaplastic Wilms tumors harbor TP53 mutations.

Pathology After Preoperative Chemotherapy


The International Society of Paediatric Oncology (SIOP) studies
have made some important observations on the histology of Wilms
Fig. 53.7. Typical Wilms tumor with blastemal, epithelial, and stromal components. tumor after preoperative chemotherapy. They have performed an
assessment of the tumor response after preoperative chemotherapy
in terms of tumor volume and histology. The relative proportions of
histologic subtypes of WT differ following preoperative chemotherapy
Favorable Histology Wilms Tumor when compared with those reported after primary surgical resection
(Weirich et al., 2001). Stromal and epithelial predominant tumors
Wilms tumor usually compresses the adjacent normal renal paren- are found more often after chemotherapy. These histologic subtypes
chyma, forming a pseudocapsule composed of compressed, atrophic may demonstrate a poor clinical response to therapy but have an
renal tissues. This intrarenal pseudocapsule can be helpful to dis- excellent prognosis if the tumor is completely excised (Verschuur
tinguish Wilms tumor from NRs and other renal tumors. The texture et al., 2010). The proportion of blastemal predominant tumors is
of the tumors varies depending on the predominant histologic pattern. decreased after chemotherapy, indicating some response of this
Many are soft and friable with necrotic or hemorrhagic areas frequently tumor type to the preoperative chemotherapy. However, patients
noted. This consistency increases the risk of intraoperative tumor with blastemal predominant tumors after chemotherapy have a
rupture during primary nephrectomy. Most Wilms tumors are high rate of relapse (Reinhard et al., 2004b). Tumor progression
unicentric, but 12% are multicentric unilateral tumors (Breslow et al., during therapy occurred in 5% of patients enrolled in SIOP 93-01 (Ora
1988a). Extrarenal Wilms tumor arising in the retroperitoneum and et al., 2007). These patients were documented to have decreased OS.
elsewhere are rare and are thought to arise from displaced metanephric As an embryonal tumor, nephroblastoma may differentiate into
elements or mesonephric remnants. Wilms tumor is derived from more mature mesenchymal tissue types, such as skeletal muscle,
primitive metanephric blastema (Beckwith and Palmer, 1978). In after chemotherapy. Chemotherapy and radiation may induce
addition to expressing a variety of cell types found in a normal cytodifferentiation of Wilms tumor cells or select for the survival
developing kidney, Wilms tumor often contains tissues such as skeletal of less mitotically active cells. In follow-up biopsies, the presence
muscle, cartilage, and squamous epithelium. These heterotopic cell of rhabdomyomatous differentiation can confound the histologic
types likely reflect the primitive developmental potential of meta- diagnosis. Furthermore, these differentiated tumors appear to be
nephric blastema that is not expressed in normal nephrogenesis. more resistant to chemotherapy, thus surgical excision is considered
“Classic” Wilms tumor is characterized by islands of compact the treatment of choice (Seifert et al., 2012).
undifferentiated blastema and the presence of variable epithelial SIOP classifies tumors with complete tumor necrosis after preopera-
differentiation in the form of embryonic tubules, rosettes, and tive chemotherapy as “low risk.” The blastemal element commonly
glomeruloid structures separated by a significant stromal component responds very dramatically to chemotherapy. The degree of tumor
(Fig. 53.7). The proportion of each of these components varies from necrosis after chemotherapy has a positive correlation with the
infrequent to abundant within and among individual tumors. proportion of blastemal component and a negative correlation
However, some Wilms tumors are not triphasic but have only biphasic with proportion of epithelial component in pre-chemotherapy
or even monomorphous patterns, and the latter can present diagnostic biopsy samples (Taskinen et al., 2017). Unfortunately, if not
difficulty (Schmidt and Beckwith, 1995). Wilms tumors with responsive to therapy and there is blastemal predominance in
predominantly epithelial differentiation have a low degree of residual tumor specimens after pre-surgical chemotherapy, this
aggressiveness, and the majority are stage I tumors (Beckwith predicts worse survival outcomes (Reinhard et al., 2004b). Children
et al., 1996; Gadd et al., 2012; Vujanic and Sandstedt, 2010). However, with stage I “low-risk” tumors after post-chemotherapy nephrectomy
these tumors may be more resistant to therapy if they are seen receive no further chemotherapy (Boccon-Gibod et al., 2000). Tumors
initially as advanced stage disease. with diffuse anaplasia and blastemal predominance after chemo-
therapy are classified as “high risk.” SIOP “intermediate risk” tumors
Anaplastic Wilms Tumor comprise all other histologies.

Identification of tumors with unfavorable histologic features such Nephrogenic Rests


as anaplasia was an important milestone accomplished by the inclu-
sion of the central pathological review of the National Wilms Tumor More than one-third of kidneys resected for Wilms tumor (regardless
Study Group protocols (Beckwith and Palmer, 1978; Bonadio et al., of those receiving preoperative chemotherapy or not) contain precur-
1985; Zuppan et al., 1991). It has allowed progressive use of adjuvant sor lesions known as NRs (Beckwith, 1993; Beckwith et al., 1990;
therapies in the sequential studies based on the risk and response Vujanić et al., 2017). Nephrogenic rests have a varied natural history
of the various pathological tumor types. Anaplasia is characterized and most do not form Wilms tumor. A rest can undergo maturation,
by the presence of three abnormalities: nuclear enlargement to three sclerosis, involution, or complete disappearance. NRs have also been
or more times the diameter of the adjacent cells, hyperchromasia detected in 1% of kidneys in infants on postmortem examination;
of enlarged nuclei, and abnormal mitotic figures. Anaplasia is rarely a much higher incidence than that of Wilms tumor. Therefore most
seen in tumors of patients younger than 2 years of age at diagnosis apparently undergo involution (Beckwith, 1998). It has been recently
(incidence about 2%), but its presence increases to a relatively stable postulated that epigenetic changes and relative methylation affect
incidence of about 13% in those older than 5 years of age (Bonadio Wilms tumor evolution from its precursor NRs, which may be useful
Chapter 53 Pediatric Urologic Oncology: Renal and Adrenal 1101

to differentiate between these tissues and potentially predict which


NRs will become Wilms tumors (Charlton et al., 2015).
NRs can be separated into two fundamentally distinct catego-
ries, perilobar nephrogenic rests (PLNRs) and intralobar nephrogenic
rests (ILNRs) (Beckwith et al., 1990). These two types of NRs are
distinguished by their location within the renal lobe. Relative posi-
tion within the lobe is a direct reflection of the chronology of the
embryologic development of the kidney. PLNRs are found only in
the lobar periphery, which is elaborated late in embryogenesis,
whereas ILNRs are found anywhere within the lobe, as well as
the renal sinus and the wall of the pelvicaliceal system. Therefore
ILNRs are generally believed to be the result of earlier gestational
aberrations (Beckwith, 1998). ILNRs are commonly stroma rich and
intermingle with the adjacent renal parenchyma. PLNRs are usually
subcortical, sharply demarcated, and contain predominantly blastema
and tubules. Of particular interest is the observation that PLNRs are
usually found in children with BWS, linked to 11p15, whereas ILNRs
are typically seen in children with aniridia, WAGR, and DDS or other
features associated with WT1. The age at diagnosis is lower for Wilms
tumor associated with WT1 mutations and those arising in association Fig. 53.8. Typical appearance of diffuse hyperplastic perilobar nephrogenic
with ILNR. These tumors have a stromal-predominant histology rest with thick rind compressing the normal renal tissue centrally.
with varying degrees of rhabdomyogenesis (Fukuzawa et al., 2008).
Multiple rests in one kidney usually implies that NRs are present
in the other kidney (Beckwith et al. 1990). Children younger than
12 months of age diagnosed with Wilms tumor who also have NRs, children with Wilms tumor have hematuria at diagnosis, and 25%
in particular PLNRs, have a markedly increased risk of developing will have hypertension at diagnosis that can be caused by elevated
contralateral disease and require frequent and regular surveillance plasma renin levels (Maas et al., 2007). Gross hematuria warrants
for several years (Coppes et al., 1999). Surveillance is also recom- further evaluation including a cystoscopy and retrograde pyelogram
mended for those diagnosed after 12 months of age who have NRs at the time of nephrectomy to rule out tumor extension into the
(D’Angio et al., 1993). The occurrence of metachronous Wilms tumor collecting system (Ritchey et al., 2008). Other symptoms include
in patients previously treated with conventional chemotherapeutic fever, anorexia, and weight loss in 10% of patients. Rarely, children
regimens suggests that nephrogenic rests are not always eradicated. may have acute abdominal pain from tumor rupture into the
NRs display a spectrum of appearances. Hyperplastic NRs can peritoneal cavity or bleeding within the tumor. There is limited
produce a renal mass that can be mistaken for a small Wilms tumor evidence that patterns of diagnosis may differ by medical specialty
(Beckwith, 1998). Incisional biopsy of a hyperplastic rest is of little and that delays in diagnosis can adversely affect survival outcomes
value in distinguishing this lesion from a Wilms tumor, unless the (Pritchard-Jones et al., 2016).
interface between the rest and normal kidney is included. Wilms tumor Compression or invasion of adjacent structures may result in an
has a pseudocapsule at the interface with the normal parenchyma atypical presentation. A persistent varicocele in the supine position
compressing the normal elements. The appearance of the lesion can or hepatomegaly may be reflective of inferior vena cava (IVC) obstruc-
provide some help in distinguishing between NR and Wilms tumor. tion from tumor thrombus. Atrial thrombus may manifest as
The Wilms tumor will have a spheric shape, whereas hyperplasic rests hypertension or congestive heart failure. Such symptoms are found
retain the appearance of the original rest and are more elliptical or in less than 10% of patients with intracaval or atrial tumor extension
lenticular in shape. MRI may be of some value in distinguishing (Ritchey et al., 1988; Shamberger et al., 2001). During the physical
between the two lesions, but this must be confirmed prospectively in examination, it is important to assess for signs of associated Wilms
large numbers of patients (Hoffer, 2005; Rohrschneider et al., 1998). tumor syndromes such as aniridia, hemihypertrophy, and genito-
Nephroblastomatosis refers to the presence of multiple neph- urinary anomalies.
rogenic rests. Diffuse overgrowth of PLNRs may produce a thick Emergent operation is not necessary unless there is evidence of
rind that enlarges the kidney but preserves its original shape active bleeding or tumor rupture. The preoperative laboratory evalu-
(Fig. 53.8). Patients with nephroblastomatosis are prone to Wilms ation of a child with an abdominal mass should include a complete
tumor development, and bilateral lesions are common. Perlman blood count, liver enzymes, and serum electrolytes, including blood
et al. (2005) have reviewed 52 cases of diffuse hyperplastic perilobar urea nitrogen, creatinine, and calcium. Because as many as 8% of
nephrogenic rests reported to the National Wilms Tumor Study Group newly diagnosed patients with Wilms tumor will have acquired von
(NWTSG) pathology center. Wilms tumor developed in 23 patients Willebrand disease, coagulation studies should be considered (Baxter
at a median of 30 months. Of children receiving adjuvant therapy et al., 2009). This includes prothrombin time and partial thrombo-
at diagnosis, 17/33 (52%) developed a Wilms tumor. There was an plastin time, which may be normal in the presence of von Willebrand
increased incidence of anaplasia noted in tumors that developed disease. This defect can generally be corrected preoperatively with
after chemotherapy in patients with nephroblastomatosis (Perlman the administration of 1-desamino-8-d-arginine-vasopressin (DDAVP)
et al., 2005). but may require IVIG or plasmapheresis (Callaghan et al., 2013).
Generally, this acquired defect and coagulopathy will be reversed
Preoperative Evaluation and Staging with surgical removal of the tumor or with preoperative chemotherapy
(Baxter et al., 2009; Blanchette and Coppes 2009).
The majority of children (>85%) are symptomatic at presentation
with either an abdominal mass, abdominal pain or hematuria Imaging
(Pritchard-Jones et al., 2016). Less commonly an asymptomatic
abdominal mass is discovered incidentally by a family member or All solid renal tumors of childhood have some common radio-
primary care physician. A small percentage of tumors are diagnosed graphic features (Miniati et al., 2008; Smets and de Kraker, 2010).
by screening. A higher proportion of stage I and II tumors (72.7%) Therefore Wilms tumors cannot be reliably distinguished from other
are noted in those who are asymptomatic at diagnosis compared renal tumors (e.g., renal cell carcinoma [RCC], or clear cell sarcoma
with those who are symptomatic (52.9%). of the kidney [CCSK]). In the SIOP-9 study, 5.4% of patients in
The mass may be extremely large relative to the size of the child whom preoperative chemotherapy for Wilms tumor was commenced
and is not necessarily confined to one side. Approximately 20% of before diagnostic biopsy were found on nephrectomy to have renal
1102 PART III Pediatric Urology

malignancies other than Wilms tumor or benign renal conditions Routine exploration of the contralateral kidney at the time of
(Tournade et al., 2001). In the United Kingdom Children’s Cancer nephrectomy is not necessary when preoperative imaging with
Study Group (UKCCSG), 12% of renal tumors clinically and radio- CT or MRI demonstrates a normal contralateral kidney (Ritchey
graphically consistent with Wilms tumor were found to have some et al., 2005).
other diagnosis on biopsy (Vujanic et al., 2003). Other clinical The role of imaging studies in renal tumor staging continues to
parameters can provide some clues to the diagnosis. The development be defined. The COG found that CT has moderate specificity but
of a renal tumor in a child known to have aniridia, hemihypertrophy, poor sensitivity in the detection of preoperative tumor rupture
or other syndromes associated with an increased incidence of (Khanna et al., 2013). Ascites beyond the cul-de-sac was most strongly
nephroblastoma is most likely to be a Wilms tumor. Bilateral or associated with tumor rupture. If the preoperative imaging could
multicentric tumors are more typical of Wilms tumor, but renal accurately detect local extension of tumor beyond the renal capsule
lymphoma can manifest in this fashion. Congenital mesoblastic or into regional lymph nodes, it would obviate concerns regarding
nephroma (CMN) is the most likely diagnosis in a neonate with a staging in patients treated with preoperative therapy. The local tumor
renal mass. However, favorable-histology (FH) Wilms tumor and burden (e.g., regional lymph node involvement) determines the
rhabdoid tumor of the kidney (RTK) can also manifest in the first intensity of the chemotherapy regimen and whether a child receives
few months of life (LeClair et al., 2005; Ritchey et al., 1995). The abdominal irradiation. Regional adenopathy can be identified on
renal origin of the mass is usually apparent on CT, but it can be CT or MRI, but enlarged retroperitoneal benign lymph nodes are
mistaken for neuroblastoma. common in children, and correlation between pathologic findings
The clinical presentation—that is, as abdominal pain with tumor and lymph node evaluation at surgical exploration in Wilms tumor
rupture—can create confusion regarding the preoperative diagnosis. patients has found significant false-positive and false-negative error
In 2.5% of NWTS-3 patients, there was an erroneous diagnosis before rates (Lubahn et al., 2012; Othersen et al., 1990). Positron emission
surgical exploration (Ritchey et al., 1992). Most of these children tomography (PET) has not been shown to have any advantages over
did not have any preoperative imaging studies performed, and this conventional imaging modalities in preoperative assessment of Wilms
group of patients had an increased incidence of surgical complications. tumor (Misch et al., 2008). Detection of extrarenal tumor extension
This emphasizes that defining the exact histology is not as important into the perirenal fat and into adjacent structures is also problematic.
as establishing that the child has a solid renal tumor, allowing the Therefore determination of inoperability should be made at surgical
surgeon to plan for a major cancer operation. These events are now exploration.
rare with modern imaging and the almost universal use of CT and The lung is the most common site of distant metastasis in
MRI for preoperative evaluation. children with Wilms tumor. Preoperative chest CT with or without
Ultrasonography is recommended as first-line imaging in children contrast is performed to rule out pulmonary metastases. The clinical
with an abdominal mass. This demonstrates the solid nature of the significance of lung nodules detected on CT scan alone is controversial
lesion, and it can also help triage appropriate subsequent imaging (Green, 2016; Grundy et al., 2012; Meisel et al., 1999; Owens et al.,
such as chest CT at the same time as abdominal cross-sectional 2002; Smets et al., 2012). CT will clearly detect more lesions than
imaging and avoid unnecessary scans such as a noncontrasted CT a standard chest radiograph, but not all of these lesions represent
abdomen. Doppler ultrasonography may be able to exclude intracaval metastases (Ehrlich et al., 2006). Some reports have suggested that
tumor extension that occurs in 4% of Wilms tumor patients (Ritchey treatment of such patients with dactinomycin (actinomycin D [AMD])
et al., 1988; Shamberger et al., 2001). However, properly phased CT and vincristine (VCR) is sufficient without the need for DOX or
has been shown to more reliably identify all clinically significant pulmonary radiation (Grundy et al., 2012; Smets et al., 2012). Others
extension of tumor into the IVC when compared with ultrasonography have found an increased risk of pulmonary relapse if more intensive
(Khanna et al., 2012). treatment is not given (Owens et al., 2002). The most recent round
All patients should undergo either CT of the abdomen and of renal tumor studies from COG prospectively investigated this
pelvis with oral and intravenous contrast or MRI of the abdomen issue and found no statistical significant decrease in EFS or OS in
and pelvis with gadolinium. MRI avoids radiation but typically children with favorable histology Wilms tumor who lacked LOH at
requires anesthesia or sedation in young children. These imaging 1p and 16q and who had complete resolution of their lung nodules
modalities can further define the extent of the lesion (Fig. 53.9; after 6 weeks of VCR, AMD, and doxorubicin and thus then omitted
Hoffer, 2005; McDonald et al., 2012; Schenk et al., 2008). This allows lung radiation (Dix et al., 2014).
improved preoperative planning by evaluating for extrarenal spread Imaging surveillance after treatment of the primary tumor is
of disease, the relationship of the tumor to adjacent visceral structures, recommended to detect tumor recurrence. COG studies advocate
and the presence of synchronous tumors in the contralateral kidney. for periodic chest CT and either CT or MRI of the abdomen. However,
some have begun to question the value of these studies and whether
they will detect relapse early enough to improve survival (McHugh
and Roebuck, 2014). Many patients will have clinical symptoms
related to relapse before imaging detection. Omitting even the pelvic
portion of the abdominal CT can significantly reduce the radiation
exposure without compromising detection of relapse (Kaste et al.,
2013; Mirza et al., 2015).
CCSK and rhabdoid tumor of the kidney (RTK) have a propensity
to metastasize to the skeleton (D’Angio et al., 1993; Seibel et al.,
2018). Bone scans are recommended after the histologic diagnosis
is confirmed (Feusner et al., 1990). Cranial CT or MRI is performed
on all children with CCSK or with RTK, because both are associated
with intracranial metastases (Gooskens et al., 2014; Tomlinson et al.,
2005; Weeks et al., 1989).

Staging
The most important determinants of outcome in children with
Wilms tumor are the histopathology and tumor stage. Accurate
staging of Wilms tumor allows treatment results to be evaluated and
enables universal comparisons of outcomes. The current staging
Fig. 53.9. CT scan of a Wilms tumor that demonstrates preoperative rupture system used by the COG (Table 53.5) is based primarily on the
with perirenal hemorrhage. surgical and histopathological findings. Examination for extension
Chapter 53 Pediatric Urologic Oncology: Renal and Adrenal 1103

TABLE 53.5 Staging System of the Childrens III FHWT displaying LOH of 16q and 1p (Dix et al., 2015). Treatment
Oncology Group was likewise intensified for stage I and II patients, with DOX added
to VCR/ACT-D, although this did not significantly affect outcome.
STAGE As mentioned earlier, gain of 1q has also been identified to be a
related genetic change associated with oncologic outcome (Gratias
I Tumor confined to the kidney and completely et al., 2013, 2016; Segers et al., 2013). Future studies will likely expand
resected. The renal capsule is intact and the the investigative thread from the prior generation of COG renal
tumor was not ruptured prior to removal. No renal tumor studies to look at treatment intensification for those with
sinus extension. There is no residual tumor. FHWT and gain of 1q because only approximately 5% of FHWT
will have combined LOH, whereas at least 25% will have gain of 1q.
II Extracapsular penetration but is completely
A number of other markers have been evaluated to predict tumor
resected. Renal sinus extension or extrarenal
recurrence including several immunohistochemical markers. It appears
vessels may contain tumor thrombus or be that most of the markers that are predictive of tumor progression
infiltrated by tumor. are found in the blastemal component of the tumor (Ghanem et al.,
III Residual nonhematogenous tumor confined to the 2013; Routh et al., 2013). These markers will have to be evaluated
abdomen: lymph node involvement, any tumor in larger numbers of patients to determine their usefulness for risk
spillage, peritoneal implants, tumor beyond stratification.
surgical margin either grossly or microscopically,
or tumor not completely removed. Treatment
IV Hematogenous metastases to lung, liver, bone,
Surgical Considerations
brain, etc.
The initial therapy for most children with Wilms tumor is radical
V Bilateral renal involvement at diagnosis.
nephrectomy. Nephrectomy is generally performed via a trans-
peritoneal approach. The surgeon is responsible for determining
the extent of tumor. Accurate staging is essential for the subsequent
determination of the need for radiation therapy and the appropriate
chemotherapy regimen. Thorough exploration of the abdominal
through the capsule, residual disease, vascular involvement, and cavity is necessary to exclude local tumor extension, liver and nodal
lymph node involvement are essential to properly assess the extent metastases, or peritoneal seeding. Exploration of the contralateral
of the tumor at presentation. kidney is no longer mandated before nephrectomy if the preoperative
Stage I tumors are limited to the kidney and completely resected. CT or MRI demonstrates a normal kidney (Ritchey et al., 2005). The
However, evidence for tumor extension can be subtle. Tumor invasion renal vein and IVC are palpated to exclude intravascular tumor
of blood and lymphatic vessels in the renal sinus is the first sign of extension before vessel ligation. Wilms tumor extends into the IVC
spread outside the kidney in stage II tumors (Weeks et al., 1987). in approximately 6% of cases and may be clinically asymptomatic
Penetration through the renal capsule is the next most common in more than 50% (Ritchey et al., 1988; Shamberger et al., 2001).
finding of extrarenal spread. Clear demonstration of tumor cells in The adrenal gland can be spared without increasing the risk for
the perirenal fat is required to document capsular penetration. Any tumor spill or recurrence if it is not in proximity to the tumor (Kieran
tumor spill leads to a stage III designation because of the increased et al., 2013a). Selective sampling of regional lymph nodes is an
risk for local tumor recurrence (Ehrlich et al., 2013; Shamberger essential component of local tumor staging. Formal retroperitoneal
et al., 1999). On the COG biology and banking study AREN 03B2, lymph node dissection is not mandated (Othersen et al., 1990;
the distribution by stage for patients with FHWT tumors was stage Shamberger et al., 1999). En bloc resection yields a lower rate of
I, 19%; stage II, 20%; stage III, 35%; stage IV, 18%; and stage V, 8% positive lymph nodes compared with separate sampling (Stewart
(Mullen et al., 2014). Patients with anaplastic tumors are more likely and Bruny, 2015). In a review of NWTS-4 and -5 patients, 12.5% of
to present with stage III or IV disease than those with FHWT tumors patients did not have lymph node sampling performed (Kieran et al.,
(Dome et al., 2006). 2012). The likelihood of having a positive lymph nodes is greater
when more lymph nodes were sampled, emphasizing the importance
Prognostic Factors of the surgeon and surgical quality in accurately staging Wilms tumor
(Kieran et al., 2012; Saltzman et al., 2018). Future studies will likely
As the treatment regimens for children with Wilms tumor have mandate more specific nodal sampling patterns to standardize surgical
become more effective, the ability of retrospectively determined staging across institutions and individual surgeons.
prognostic factors to predict outcomes is diminished. Traditional The other major responsibility when performing a nephrectomy
staging factors (e.g., tumor size, histology, and lymph node metastases) for Wilms tumor is complete removal of the tumor without con-
relied upon in the past to predict risk for tumor progression or tamination of the operative field. Gentle handling of the tumor
relapse are now less able to stratify FH patients for treatment. Clinical throughout the procedure is mandatory to avoid tumor spillage. A
cancer trials incorporate biologic factors that predict tumor behavior COG study reported intraoperative tumor spillage in 9.7% of patients
to stratify patients for treatment. undergoing primary nephrectomy (Gow et al., 2013). Multivariate
Chromosomal Abnormalities. As noted earlier, LOH for a portion analysis demonstrated that spillage was more common with right-sided
of chromosome 16q and/or 1p has been noted in 20% of Wilms tumors and larger tumors. Avoiding tumor spillage has a real impact
tumors. This is associated with an increased risk for relapse (Grundy on patient outcomes because these patients have an increase in local
et al., 1994, 2005; Messahel et al., 2009; Wittman et al., 2007). This abdominal relapse (Shamberger et al., 1999). Shamberger et al.
difference in outcome is independent of histology and stage. NWTS-5 (1999) identified risk factors for local tumor recurrence as tumor
patients with stage I or II FH tumors with LOH of either 1p or 16q spillage, unfavorable histology, incomplete tumor removal, and
had an increased risk of relapse and death in comparison with patients absence of any lymph node sampling. This study included stage
lacking LOH at either locus (Grundy et al., 2005). The risks of relapse II and III disease. The risk of recurrence was highest in those patients
and death for patients with stage III or IV FH tumors were increased with stage II disease. More recent COG studies have treated all spill
only with LOH for both regions. patients as stage III. Review of these patients shows that the greatest
Patients enrolled in the recently published COG renal tumor risk of recurrence in stage III patients are positive lymph nodes,
protocols were selected for more intensive treatment if the tumor combined LOH, or residual disease (Ehrlich et al., 2013; Fernandez
demonstrated LOH for 1p and 16q. Compared with historical controls, et al., 2018). Tumor spillage was not predictive of recurrence likely
they found that intensification of treatment improved EFS for stage resulting from the increased therapy currently given to these patients.
1104 PART III Pediatric Urology

There have been several reports of laparoscopic or robotic increase in OS for stage II patients with tumor spillage. This was
nephrectomy for Wilms tumor. This is usually done in conjunction attributed to a lower post-recurrence mortality in unirradiated patients.
with preoperative chemotherapy and is likely more feasible after the The overall risk for relapse is low in stage II FH with spillage; the
tumor is reduced in size (Duarte et al., 2014; Romao et al., 2014; risks of late effects of intensified treatment must be weighed against
Varlet et al., 2014; Warmann et al., 2014). Experience with open benefit of decreased relapse (Green et al., 2014).
nephrectomy after chemotherapy has shown that these tumors are NWTS-5 (1995–2003) was a single-arm therapeutic trial. One of
less prone to tumor spillage (Powis et al., 2013). Although pre- the major aims of the trial was to confirm the utility of LOH for
chemotherapy laparoscopic nephrectomy has been reported, many chromosomes 16q and 1p to predict increased risk of tumor relapse
more cases will have to be performed to determine if there is an and death (Grundy et al., 2005). Another objective was to evaluate the
increased risk of tumor spillage, residual disease, or surgical complica- efficacy of treatment regimens for anaplastic histology Wilms tumor.
tions (Barber et al., 2009; Cost et al., 2015). Stage I patients with anaplastic tumors were treated with AMD and
Removing a large renal tumor in a small child is associated with VCR, but this resulted in a low 4-year EFS of 69.5% (Dome et al.,
some morbidity. NWTS-4 patients undergoing primary nephrectomy 2006). A new intensified chemotherapy regimen used for patients with
had an 11% incidence of surgical complications (Ritchey et al., 1999). stage II to IV diffuse anaplasia did not result in an improved survival.
The most common complications encountered are hemorrhage and In NWTS-5, children less than 2 years of age with stage I FH
small bowel obstruction (Ritchey et al., 1992, 1993a, 1999). Factors tumors weighing less than 550 g were defined as very low-risk Wilms
that have been associated with an increased risk for surgical complica- tumors (VLRWT) did not receive chemotherapy after nephrectomy.
tions are higher tumor stage, tumor size greater than 10 cm, incorrect This portion of the study was closed early when the number of
preoperative diagnosis, thoracoabdominal incision, intracaval tumor tumor relapses exceeded the limit allowed by the design of the study
extension, and resection of other visceral organs. (Green et al., 2001a). A long-term review of this cohort was completed
Preoperative chemotherapy may influence surgical complication comparing the outcomes to similar patients treated with postoperative
rates by producing tumor shrinkage. A report from the UKCCSG AMD and VCR (Shamberger et al., 2010). The 5-year EFS for surgery
compared the complication rate for patients undergoing immediate alone was 84% compared with 97% for the treated group, but the
nephrectomy compared with delayed nephrectomy performed after 5-year OS was equivalent between the two groups at 98% and 99%,
6 weeks of chemotherapy (Powis et al., 2013). They found significantly respectively (P = 0.70). There is a trade-off between more intensive
fewer complications in those undergoing delayed nephrectomy (1% therapy and its potential long-term sequelae required for the 16%
vs. 5.8%). They also noted a much higher rate of tumor rupture/ of children who relapse versus the avoidance of any postoperative
spill in those undergoing immediate nephrectomy (14.6% vs. 0). chemotherapy in the majority. As noted earlier, COG investigators
This is similar to the rate of intraoperative tumor spill after immediate hope to use biologic prognostic factors to select patients who do
nephrectomy reported by the COG (Gow et al., 2013). not require adjuvant therapy. All VLRWT registered on NWTS-5 who
did not receive adjuvant chemotherapy were analyzed for LOH at
Cooperative Group Trials 11p15 and for WT1 mutation. LOH, as determined by 11p15 methyla-
tion analysis, was significantly associated with relapse in VLRWT as
Multiple randomized clinical trials have been conducted by the were WT1 abnormalities (Perlman et al., 2011). If these results are
NWTSG, COG, SIOP, and the UKCCSG to determine the appropriate validated in an independent cohort of patients, it would be worthwhile
role for each of the therapeutic modalities available. Patients are to conduct a clinical trial that uses molecular genetic factors rather
stratified into different treatment groups based on stage and pathology. than the arbitrarily defined clinical factors of patient age and tumor
The goals of these trials are to decrease the intensity of therapy for weight to identify patients with stage I FHWT who do not require
most patients in an effort to prevent late sequelae of treatment while adjuvant therapy. It is anticipated that such a trial would expand
maintaining excellent OS. the number of patients who would be candidates to be treated with
National Wilms Tumor Study Group. The NWTSG was formed surgery only.
in 1969 to study Wilms tumor. The early NWTSG studies, NWTS-1 Children’s Oncology Group. The first generation of COG studies
(1969–1973) and NWTS-2 (1974–1978), showed that the combina- (2006–2013) grouped patients by risk for recurrence (very low, low,
tion of VCR and AMD was more effective than the use of either drug standard, and high). The COG again examined the role of surgery-only
alone. The addition of doxorubicin (DOX) was found to improve treatment for patients with stage I FHWT in which the tumor and
survival for stage III and IV patients, and postoperative flank irradiation kidney weighed less than 550 g and patient’s age was less than 2
was unnecessary for stage I patients (D’Angio et al., 1976, 1981). A years. This further demonstrated the success in a surgery-only approach
major achievement of the early trials was identification of prog- for those with very low risk FHWT. The 4-year EFS was 89.7% and
nostic factors that allowed stratification of patients into high-risk OS was 100% (Fernandez et al., 2017). This will likely prompt future
and low-risk treatment groups. Patients with positive lymph nodes study of expanding those approaches offered surgery-only.
and diffuse tumor spill were found to be at increased risk of Children with stage I or II FHWT and LOH of 1p and 16q were
abdominal relapse and therefore considered stage III and given treated with VCR, AMD, and DOX without radiotherapy. Patients
postoperative irradiation. One of the most important findings with stage III FHWT disease without LOH of 1p and 16q were treated
was the identification of the unfavorable histologic features that with VCR, AMD, and DOX and irradiation of the flank or abdomen.
have a very adverse impact on survival. These risk-stratified stage III patients enjoyed an excellent outcome
NWTS-3 (1979–1986) demonstrated that stage I and II patients with 4-year EFS of 88% and OS of 97% (Fernandez et al., 2018).
could be treated with 18 weeks of AMD and VCR without irradiation The COG evaluated a response-based approach for management of
(D’Angio et al., 1989). For stage III FH patients, 10.8 Gy of abdominal children with pulmonary metastases. Those with resolution of the
irradiation was shown to be as effective as 20 Gy in preventing pulmonary lesions on chest CT after 6 weeks of chemotherapy were
abdominal relapse if DOX was added to VCR and AMD. NWTS-4 continued on treatment with VCR, AMD, and DOX. These patients
(1987–1994) proved that treatment durations of 6 months produced who were termed “rapid complete responders” had a 4-year EFS of
comparable outcomes to 15 months of therapy for patients with 79.5% and OS of 96.1% that was not statistically inferior to historical
stages II to IV/FH tumors (Green et al., 1998). The NWTSG has standards; this suggests that these risk-adapted patients may be able
assessed the impact of postoperative irradiation on flank recurrence to avoid lung radiation (Dix et al., 2018). Patients who did not
and survival (Breslow et al., 2006; Green et al., 2014; Kalapurakal have resolution of the pulmonary lesions by week 6 received more
et al., 2010). The investigators found that abdominal recurrence rates intensive chemotherapy and pulmonary irradiation. These patients
after tumor spillage were significantly higher among patients treated were termed “slow incomplete responders” and intensification to
with two- or three-drug chemotherapy without RT. Irradiation with regimen M led to an improved outcome with 4-year EFS of 88.5%
10 Gy appeared to be successful in reducing tumor recurrence rates and OS of 95.4% (Dix et al., 2018).
after tumor spillage, and 20 Gy even more so. Although radiation A specific study was focused on those with “high-risk” renal tumors.
did decrease the incidence of flank recurrence, there was only an Children with stage I to III focal anaplastic histology Wilms tumor
Chapter 53 Pediatric Urologic Oncology: Renal and Adrenal 1105

A B
Fig. 53.10. (A) CT of a Wilms tumor that was pretreated with chemotherapy. (B) After 6 weeks of che-
motherapy, the tumor is much smaller in size.

(AHWT) and stage I diffuse AHWT were treated with AMD, VCR, postoperative chemotherapy with AMD and VCR. Two-year EFS was
DOX, and abdominal irradiation. Patients with stage II, III, or IV 91.4% after 4 weeks and 88.8% after 18 weeks of therapy, demonstrating
(no measurable disease) diffuse AHWT, stage IV focal AHWT, stage that survival can be maintained while shortening the duration of
IV clear cell sarcoma, and stage I to III malignant rhabdoid tumor post-nephrectomy therapy. Patients with stage II or III with low- or
were treated with a new chemotherapy regimen to try to improve intermediate-risk histology received 4 weeks of dactinomycin and
OS. Publication of these trial data is still pending. VCR before surgery (Graf et al., 2012). Postoperative chemotherapy
International Society of Paediatric Oncology. In the randomized consisted of dactinomycin, VCR, and epirubicin/doxorubicin for 27
clinical trials conducted by SIOP, preoperative therapy is given before weeks. Flank or whole-abdomen irradiation was given for stage III.
surgery. This approach usually results in tumor shrinkage (Fig. 53.10), With a median follow-up of 8 years, 5-year EFS was 90% and OS 95%.
reducing the risk of intraoperative rupture or spill (Lemerle et al., Patients with blastemal type histology had a worse prognosis with 62%
1976). A greater number of patients have “post-chemotherapy stage EFS at 5 years. They made up only 10% of patients but contributed to
I” tumors resulting from disappearance of micrometastases after one-third of the relapses and deaths. The other important prognostic
neoadjuvant therapy. This was thought to be a significant advantage factors were a large tumor volume at surgery and stage III disease.
in terms of decreasing morbidity of treatment, particularly the late The SIOP 2001 study asked the question whether patients with
effects of radiotherapy. stage II and III intermediate-risk histology Wilms tumors could be
Early SIOP studies evaluated pre-nephrectomy XRT (Lemerle et al., safely treated without an anthracycline. Data from SIOP 93-01 had
1976). SIOP-5 (1976–1980) showed that 4 weeks of AMD and VCR suggested that survival was not adversely affected when nonviable
were as effective as pre-nephrectomy XRT in avoiding surgical tumor tumor was identified in the renal sinus and/or perirenal fat after
rupture and increasing the proportion of patients with low stage preoperative chemotherapy (Vujanic et al., 2009). Between 2001 and
disease (Lemerle et al., 1983). SIOP-6 (1980–1987) demonstrated 2009, 583 patients were randomized (Pritchard-Jones et al., 2015).
that patients with “post-chemotherapy stage I” disease can safely For stage II or stage III intermediate-risk histology Wilms tumor,
be treated with 18 weeks of AMD and VCR (Tournade et al., 1993). 2-year EFS was 92.6% for patients treated with DOX and 88.2% for
However, patients with “post-chemotherapy stage II” tumors and treatment excluding DOX. OS was 96.5% and 95.8%, respectively.
negative lymph nodes were found to have a higher rate of abdominal The SIOP group uses the response of the tumor to preoperative
relapse if postoperative irradiation was omitted (Tournade et al., chemotherapy in their risk stratification (Vujanic et al., 2002). This
1993). An anthracycline was subsequently added for treatment of these includes not only percentage of necrosis but also on the predominant
children. SIOP-6 confirmed the need for a three-drug chemotherapy cell type in the residual viable tumor component. Patients with
regimen after nephrectomy for patients with “post-chemotherapy stage high-risk blastemal tumors continue to receive treatment with DOX,
II” lymph node positive and stage III tumors. SIOP-9 (1987–1993) including stage I tumors (van den Heuvel-Eibrink et al., 2015). The
demonstrated that the relapse rate for stage II patients with negative intensified treatment for the latter patients yielded improved OS in
lymph nodes without radiation therapy was reduced with epirubicin SIOP 2001 with EFS of 96% (OS 100%) compared with EFS of 71%
(Tournade et al., 2001). This study also demonstrated that treatment (OS 90%) in SIOP 93-01.
with VCR and AMD for 4 weeks versus 8 weeks had comparable rates United Kingdom Children’s Cancer Study Group. The UKCCSG
of stage distribution and tumor shrinkage in patients with stage I and has conducted several trials using prenephrectomy chemotherapy
III disease. The majority of tumor shrinkage was noted in the first but, unlike SIOP, they perform biopsy before treatment (Mitchell
4 weeks of therapy. Radiotherapy was limited to patients with stage et al., 2000; Pritchard et al., 1995; Pritchard-Jones et al., 2003). This
II node-positive and III disease resulting in 18% of patients being is done to avoid giving chemotherapy to infants and children with
irradiated (Graf et al., 2000). There were 59 children with stage I to IV benign tumors, which accounts for 1% of lesions thought to be
tumors who had complete tumor necrosis induced by chemotherapy, Wilms tumor on imaging studies (Tournade et al., 2001). The other
and 98% of these children had no evidence of disease at 5 years reason to perform biopsy is to avoid giving inappropriate chemo-
(Boccon-Gibod et al., 2000). therapy to non–Wilms tumors, which often require more intensive
The SIOP 93-01 study (1993–2001), evaluated a reduction in therapy. The UKW3 trial noted a 12% incidence of non–Wilms tumors
postoperative therapy for patients with stage I intermediate risk in patients with the typical features of Wilms tumor on imaging
and anaplastic Wilms tumor (deKraker et al., 2004; Reinhard et al., study (Vujanic et al., 2003). The UKW1 and UKW2 studies evaluated
2004b). Patients were randomized to receive either 4 or 18 weeks of the single-agent VCR for treatment of stage I FH tumors (Mitchell
1106 PART III Pediatric Urology

et al., 2000; Pritchard et al., 1995). The OS of 96% compares well Repeat imaging is performed after 6 weeks of chemotherapy.
with two-drug chemotherapy, but age greater than 4 years was Experience in SIOP has shown that the majority of reduction (48%) in
considered an adverse prognostic factor (Pritchard-Jones et al., 2003). tumor volume occurs in the first 4 weeks of therapy (Tournade et al.,
The UKW3 trial randomly assigned patients to either immediate 2001) but that reduction extends out through 8 weeks (62%). After
surgery or to 6 weeks preoperative chemotherapy and then delayed there has been adequate shrinkage of the tumor, definitive resection
surgery (Mitchell et al., 2006). EFS and OS at 5 years were similar can usually be completed. A clinically good response (by imaging)
in the two groups. Around 20% of survivors avoided treatment with is usually associated with a pathologically good response in terms
doxorubicin or radiotherapy because of favorable stage distribution of regressive histologic changes (Weirich et al., 2001; Zuppan et al.,
after preoperative therapy. They concluded, like the SIOP group, 1991). The converse is not always true. The distribution of histologic
that all children with non-metastatic Wilms tumor should receive subtypes is different after preoperative chemotherapy compared with
chemotherapy before tumor resection. primary surgery, with differentiation of the tumor occurring after
chemotherapy. Stromal- and epithelial-predominant tumors are found
Adult Wilms Tumor more often after treatment with preoperative chemotherapy. These
histologic subtypes may demonstrate a poor clinical response to
Wilms tumor occasionally occurs in adults. Earlier reports suggested therapy but have an excellent prognosis if the tumor is completely
that the outcome for adults with Wilms tumor was poor and that excised. Patients with progressive disease have a poor prognosis, and
they require more intensive therapy (Arrigo et al., 1990). More recent these patients require treatment with a more intensive chemothera-
reviews of adult patients with FH Wilms tumor have found improved peutic regimen (Ora et al., 2007; Ritchey et al., 1994).
survival compared with prior reports (Ali et al., 2012; Kalapurakal Bilateral Wilms Tumors. Synchronous bilateral Wilms tumors
et al., 2004a; Reinhard et al., 2004a). The recommendation is that occur in 5% to 7% of children with Wilms tumor (Blute et al.,
adult patients receive stage-appropriate combined modality therapy. 1987; Coppes et al., 1989; Montgomery et al., 1991). Children with
bilateral tumors should not undergo initial radical nephrectomy.
Relapse These children should receive preoperative chemotherapy with the
goal of tumor shrinkage and renal-sparing surgery (Blute et al., 1987;
A uniform approach for the treatment of tumor relapse was used in Coppes et al., 1989; Hamilton et al., 2011; Kumar et al., 1998).
NWTS-5. Patients with relapsed Wilms tumor may be divided into Preservation of renal tissue is important to decrease the incidence
risk groups according to OS rates after salvage therapy (Spreafico of renal failure, which approaches 15%, at 15 years after treatment
et al., 2009). Children with non-anaplastic Wilms tumor who relapse in patients with bilateral Wilms tumor (Breslow et al., 2005; Ritchey
after therapy with only VCR and/or AMD are considered standard et al., 1996). The most common cause for renal failure was the need
risk and have survival rates in the 70% to 80% range (Green et al., for bilateral nephrectomy for persistent or recurrent tumor in the
2007). Patients with non-anaplastic Wilms tumor who relapse after remaining kidney after initial nephrectomy. Complete nephrectomy
therapy with three or more agents are called high risk and have can be avoided in the majority of patients if a careful protocol is
survival rates in the 40% to 50% range (Malagolowkin et al., 2008). followed and the surgery is performed by surgeons experienced in
The very high-risk group includes recurrent anaplastic or blastemal- renal-sparing techniques (Davidoff et al., 2008; Fuchs et al., 2011).
type Wilms tumor and have survival rates in the 10% range (Reinhard The recently completed COG protocol (AREN0534) recommended
et al., 2008). Further studies are needed to determine the optimal that patients with bilateral Wilms tumor receive 6 weeks of chemo-
treatment strategies for the “high risk” and “very high-risk” groups therapy before surgery. Biopsy was not required if the radiographic
and to define the role of high-dose myeloablative chemotherapy picture was consistent with Wilms tumor. Tumor response was assessed
and stem cell rescue (Ha et al., 2013). after 6 weeks with CT or MRI to determine the reduction in tumor
volume and feasibility of partial resection. Patients with tumors
Preoperative Chemotherapy (COG Recommendations) amenable to renal-sparing procedures were allowed to proceed with
surgery. Tumors not responding to therapy required bilateral open
On the COG renal tumor protocols, treatment is dependent on biopsy to determine histology. Open biopsies are recommended
surgical and pathological staging after immediate nephrectomy. There because they are more accurate than percutaneous needle biopsies
are, however, some situations in which preoperative chemotherapy when assessing for anaplasia, and bilateral biopsies are recommend
is recommended. These include children for whom renal-sparing because anaplasia is found to be discordant between the two
surgery is planned (Blute et al., 1987), tumors inoperable at surgical kidneys in 83% of children (Hamilton et al., 2006). Biopsy is
exploration (Ritchey et al., 1994), and tumor extension into the IVC necessary because imaging cannot predict the histology of the tumor
above the hepatic veins (Ritchey et al., 1993b; Shamberger et al., based on changes in volume of the tumor after chemotherapy (Olsen
2001; Szavay et al., 2004). The last two conditions are associated et al., 2004; Weirich et al., 2001). Failure to achieve a reduction in
with an increased risk for surgical complications if primary nephrec- volume is likely a result of tumor differentiation (Fig. 53.11) (Ander-
tomy is performed (Ritchey et al., 1992). son et al., 2002; Shamberger et al., 2006; Weirich et al., 2001).
Inoperable Tumors. The surgeon, not the oncologist or radio- Differentiated tumors may show a poor clinical response to therapy,
therapist, must make the determination that a tumor is inoperable. but they have an excellent prognosis if the tumor is completely
This decision should not be based on preoperative imaging studies, excised. If renal-sparing surgery is not feasible, additional chemo-
which can overestimate local tumor extension. As noted earlier, therapy is then given based on the biopsy findings, but all patients
not all renal masses in children represent Wilms tumor (Vujanic were recommended to undergo surgical resection within 12 weeks
et al., 2003). If the tumor is found to be unresectable, pretreatment of starting therapy. Continuing treatment beyond 12 weeks will not
with chemotherapy almost always reduces the bulk of the tumor likely provide any additional reduction in tumor burden.
and renders it resectable (Grundy et al., 2004; Ritchey et al., 1994). At the time of second-look surgery, partial nephrectomy or wedge
Patients who are staged with imaging studies alone and receive excision of the tumor is preferred with an attempt to achieve negative
preoperative chemotherapy before nephrectomy are also at risk for margins. Radical nephrectomy may be needed in a kidney with
understaging (Tournade et al., 1993). Patients enrolled in COG extensive tumor involvement. The kidney with the lower tumor
studies determined to have an inoperable tumor are treated as burden is addressed first. Tumor enucleation may be considered in
stage III disease (Dome et al., 2015). The number of patients in lieu of a formal partial nephrectomy. This is often needed for large
North America treated with preoperative chemotherapy has increased centrally located tumors when removal of a margin of renal tissue
over time. On NWTS-3, only 5.4% of patients were treated as inoper- would compromise the vascular supply to the kidney (Cozzi et al.,
able (Ritchey et al., 1994). For the recently completed COG AREN0532 1996; Horwitz et al., 1996). The concern is that enucleation will be
and AREN0533 studies, approximately 20% of patients with overall more likely to result in positive surgical margins. For FH tumors,
stage III disease and 40% of patients with overall stage IV disease adjuvant therapy may still achieve a good outcome (Cozzi et al.,
underwent delayed nephrectomy (Dome et al., 2015). 1996; Davidoff et al., 2008; Horwitz et al., 1996). However, if there
Chapter 53 Pediatric Urologic Oncology: Renal and Adrenal 1107

A B
Fig. 53.11. Patient with bilateral tumors who was treated with chemotherapy. (A) CT before treatment. (B) CT
after 12 weeks of chemotherapy, revealing only minimal decrease in the size of the tumors. Bilateral partial
nephrectomies were performed, revealing mature tumor elements with rhabdomyoblastic differentiation.

total nephrectomy (10.5%), unilateral partial nephrectomy (4%),


and bilateral total nephrectomies (2.5%). The 4-year EFS and OS
were 82.1% and 94.9%, respectively. This is compared with 4-year
EFS and OS of 56% and 80.8% on NWTS-5.
Bilateral nephrectomies and dialysis are rarely required when the
tumors fail to respond to chemotherapy and radiation therapy. This
is the most common cause of renal failure in patients with bilateral
Wilms tumor (Ritchey et al., 1996). Fortunately, anephric patients
can still be administered chemotherapy with some modifications
(Feusner et al., 2008). The recommended interval between successful
completion of treatment of the Wilms tumor and renal transplantation
varies (Kist-van Holthe et al., 2005; Penn, 1979). Some advocate a
waiting period of 2 years to ensure that the patient does not develop
metastatic disease; others have found that a 1-year interval is sufficient
(Gregoriev et al., 2012). Patients who develop renal failure after
renal-sparing surgery should have removal of the remaining renal
tissue before transplant to prevent tumor recurrence after starting
immunosuppression (Kubiak et al., 2004).
Fig. 53.12. Postoperative image from patient shown in Fig. 53.11. Demon-
All patients treated for bilateral Wilms tumor require close long-
strates that the kidneys have pretty near normal volume after resection of
term follow-up. SIOP investigators noted that late relapses have
the bilateral tumors.
occurred in patients with bilateral Wilms tumor more than 4 years
after treatment and recommended long-term follow-up (Coppes
et al., 1989). These patients should also have frequent assessment
is anaplasia in the resected specimen, a positive margin will adversely of renal function, urine protein, and blood pressure.
affect survival and requires additional resection. Even when large Partial Nephrectomy for Unilateral Tumors. There have been a
bilateral masses remain after initial chemotherapy, a high percentage number of papers examining the role of parenchymal-sparing
of children can be successfully managed with renal-sparing surgery procedures in children with unilateral Wilms tumors (Cost et al.,
(Davidoff et al., 2008). It is easy to underestimate the amount of 2012; Haecker et al., 2003; McLorie et al., 1991; Wilde et al., 2014;
renal parenchyma that can be salvaged as a result of compression Zani et al., 2005). The primary motivation for this approach is concern
by the tumor; therefore nephron-sparing surgery should be considered about late occurrence of renal dysfunction after unilateral nephrec-
in all patients (Fig. 53.12). One concern regarding enucleation of tomy. However, the incidence of renal failure after nephrectomy
large centrally located tumors is the potential for positive surgical for most children with unilateral Wilms tumor is low, 0.6% at
margins. Kieran et al. (2013b) reported a 23% incidence of positive 20 years after treatment (Breslow et al., 2005; Lange et al., 2011).
surgical margins in a cohort of 21 patients with bilateral Wilms The risk of renal failure is higher for patients with genitourinary
tumor undergoing renal-sparing surgery. They did not demonstrate anomalies, DDS, and WAGR. As noted earlier, this is a result of
an increased risk of local recurrence, but this was a small number mutation of WT1, which is necessary for normal renal development.
of patients, with all receiving 10.5-Gy flank radiation. Syndromic patients are more likely to have smaller tumors identified
The COG AREN0534 study enrolled 195 patients with bilateral on screening studies that are more amenable to renal-sparing surgery
Wilms tumor (Ehrlich et al., 2017). Of the 189 evaluable patients, (Romao et al., 2012; Scalabre et al., 2016).
84.0% underwent definitive surgical treatment (partial or complete Most Wilms tumors are too large at diagnosis to allow partial
nephrectomy or wedge resection in at least one kidney) by 12 weeks nephrectomy. After preoperative chemotherapy, partial nephrectomy
after initiation of chemotherapy. Thirty percent of those patients can be performed in 10% to 15% of patients. However, this higher
who achieved definitive surgical resection within the 12-week specified percentage is only achieved if the clinician is more aggressive in
aim did so by the 6-week evaluation point. Surgical approaches proceeding with nephron sparing surgery (NSS). The SIOP 2001
included unilateral total nephrectomy with contralateral partial study restricted NSS for polar or peripherally non-infiltrating tumors
nephrectomy (48%), bilateral partial nephrectomy (35%), unilateral (Wilde et al., 2014). They reported that 3% of tumors met the criteria
1108 PART III Pediatric Urology

for NSS. Only the occasional case of Wilms tumor will involve a is radiation dose dependent. The Leydig cells are more radioresistant
lesion small enough to allow partial nephrectomy at diagnosis—for than the germ cells, but higher doses can produce damage, resulting
example, tumors detected on screening studies for Beckwith- in inadequate production of testosterone. This can result in delayed
Wiedemann syndrome and aniridia (see Fig. 53.6). Even in the young sexual maturation. Chemotherapeutic agents can also adversely affect
children with very-low–risk stage I tumors, it is estimated that less testicular function (Mustieles et al., 1995). Pelvic irradiation and
than 10% of children would be amenable to partial nephrectomy exposure to alkylating agents are risk factors for ovarian failure and
(Ferrer et al., 2013). As noted earlier, there are concerns regarding premature menopause in female Wilms tumor survivors (Green et al.,
staging after chemotherapy, requiring some patients to receive added 2009). Very few pregnancies have been reported in patients who
therapy to prevent local recurrence. Another concern is the increased received whole abdominal radiation therapy (Green et al., 2010).
risk for local recurrence after partial nephrectomy (Haecker et al., Pregnancy complications were evaluated extensively through the
2003; Horwitz et al., 1996; Wilde et al., 2014). Patients who develop NWTSG. The offspring of irradiated female patients are at risk for
intra-abdominal relapse have a markedly decreased survival (Sham- low birth weights and premature birth. Radiation portals that include
berger et al., 1999). the pelvis and doses exceeding 20 Gy increase the risk of miscarriage
The COG AREN0534 study also included a renal-sparing protocol (Kalapurakal et al., 2004b). Wilms tumor survivors treated with
for select patients with unilateral Wilms tumors known to be at risk abdominal radiotherapy are also at increased risk of hypertension
for bilateral disease or at increased risk for renal failure. These patients complicating pregnancy (Reulen et al., 2017).
were managed with a strict surgical protocol to minimize risk for
residual disease (Cozzi et al., 2004). The protocol required that the Second Malignancies
lesion be completely excised with a margin of normal renal paren-
chyma. These patients should not undergo partial nephrectomy if An increased incidence of second malignant neoplasms has been
the tumor cannot be removed at stage I. Patients with high-risk noted in children treated for Wilms tumor. There is a 1% cumulative
histologic patterns such as anaplasia or persistent blastemal- incidence at 10 years postdiagnosis, and a rising incidence thereafter
predominant tumor after chemotherapy should be treated with (Breslow et al., 1988b, 2010; Taylor et al., 2008). One of the greatest
complete nephrectomy because these tumors have resistance to risk factors is prior irradiation, and most tumors occur in the radiation
chemotherapy (Reinhard et al., 2004b). Results of AREN0534 for field (Bassal et al., 2006; Breslow et al., 1988b; Taylor et al., 2008).
patients with unilateral tumors have not been published. The incidence of leukemia is highest during the first 5 years after
Wilms tumor treatment. The incidence of solid tumors increases
Late Effects of Treatment with longer periods of follow-up (Lee et al., 2015).

The 5-year survival rate for childhood cancers has increased over Cardiac Effects
the past decades to more than 80%. Achieving these high cure rates
requires exposure of normal healthy tissue to potential harm with The risk of cardiotoxicity in Wilms tumor survivors has been carefully
many organ systems subject to the late sequelae of anticancer therapy. studied. In a review of patients entered in NWTS-1, NWTS-2, NWTS-3,
These survivors are at increased risk for numerous chronic health and NWTS-4, the frequency of congestive heart failure was 4.4%
conditions, hospitalizations, and reduced productivity because of among DOX-treated patients who received this drug as part of their
health problems (Phillips et al., 2015). Clinicians must be aware of initial chemotherapy regimen (Green et al., 2001b). The risk was
the spectrum of problems that face children as they grow into adult- increased if the patient received whole-lung or left-flank irradiation.
hood. Our understanding of late effects in Wilms tumor survivors Only one patient with congestive heart failure received a DOX
has been advanced through several large studies. The NWTSG Late cumulative dose below 150 mg/m2, which is used in contemporary
Effects Study followed patients treated in NWTS-1 to NWTS-5. The North American treatment regimens. However, subclinical cardiotoxic-
original Childhood Cancer Survivor Study (CCSS) was a retrospectively ity was not assessed, and it is possible that clinical effects with
ascertained cohort of childhood cancer survivors diagnosed from modern regimens will become apparent with longer follow-up. The
1970 to 1986. More than 14,000 survivors were surveyed and followed British CCSS has found that cardiac problems were the second most
for long-term health outcomes. Because of the significant changes common cause of mortality more than 30 years after Wilms tumor
in therapy for children with cancer over the past 30 years, a second diagnosis (Wong et al., 2016).
group of about 10,000 survivors diagnosed between 1987 and 1999
were also recruited for the study. Similar efforts are also conducted
in other countries, some of which are population-based cohort studies Other Renal Tumors
(Wong et al., 2016). Clear Cell Sarcoma of the Kidney
The CCSS reported a cumulative incidence of 65% for all chronic
health conditions in Wilms tumor survivors at 25 years after CCSK accounts for 3% of renal tumors reported to the NWTSG. The
completion of therapy (Termuhlen et al., 2011). These rates continue tumor derives its name from the clear cytoplasm of the predominant
to increase with longer periods of follow-up (Phillips et al., 2015). cell type (Schmidt and Beckwith, 1995). Important predictors of
The cumulative incidence of severe (grades 3 or 4) chronic health improved survival are lower stage, younger age at diagnosis, treat-
conditions was 24% (Termuhlen et al., 2011). ment with DOX, and absence of tumor necrosis (Argani et al., 2000).
The addition of doxorubicin improved OS and relapse-free survival
Mortality (Argani et al., 2000; D’Angio et al., 1989; Seibel et al., 2004). Patients
with stage I tumors (using the current criteria of absence of renal
The NWTSG Late Effects Study showed that survivors remain at sinus invasion) had a 98% survival rate. Unlike anaplastic Wilms
elevated risk for death compared with the general population for tumor, even stage I CCSK lesions are associated with increased rates
many years after their original diagnosis (Cotton et al., 2009). A of relapse and require postoperative irradiation. Long-term follow-up
more recent report from the CCSS has shown that mortality for of CCSK patients is needed because 30% of relapses occurred more
survivors of childhood cancer has decreased for those patients treated than 3 years after diagnosis and some as late as 10 years. Unlike Wilms
in the 1990s compared with the early 1970s (Armstrong et al., 2016). tumor, CCSK is associated with bone and brain metastases. Bilateral
This is attributed to changes in therapy (e.g., reduction the number involvement has thus far not been reported, nor has the presence of
of patients exposed to radiotherapy or anthracyclines). Wilms tumor associated congenital anomalies such as aniridia or
hemihypertrophy. Patients with CCSK were treated on NWTS-5 with
Fertility and Pregnancy a regimen combining VCR, DOX, cyclophosphamide, and etoposide
in an attempt to further improve the survival of this high-risk group.
Gonadal radiation can produce hypogonadism and temporary However, outcomes for patients with CCSK treated on NWTS-5 were
azoospermia in boys (Kinsella et al., 1989). The severity of damage similar to that seen on NWTS-4 (5-year RFS and OS of 79% and 89%,
Chapter 53 Pediatric Urologic Oncology: Renal and Adrenal 1109

respectively) (Seibel et al., 2018). Stage was found to be highly predictive cysts are found in young children, usually boys. The second peak
of outcome; 5-year RFS rates for stages I, II, III, and IV on NWTS-5 incidence occurs in young adult women (Eble and Bonsib, 1998;
were 100%, 88%, 73%, and 29%, respectively. Similar outcomes have Luithle et al., 2007). Although the majority of cases of multilocular
been reported by SIOP investigators (Furtwangler et al., 2013). cystic renal disease have been unilateral, there are rare reports of
bilateral cases (Ferrer and McKenna, 1994). The gross appearance
Rhabdoid Tumor of the Kidney of the tumor is its most distinguishing feature. The cut surfaces reveal
a well-encapsulated multilocular tumor composed of various-size
RTK is the most aggressive and lethal childhood renal tumor that cysts compressing the surrounding renal parenchyma. This tumor
accounts for 2% of renal tumors. RTK and CCSK occur in renal is distinguished by the finding of only mature cell types within the
and extrarenal locations, suggesting an origin from a non–organ- septa of the cyst wall. Multilocular cystic nephroma is cured by
specific mesenchymal cell. These tumors are characterized by loss nephrectomy, but recurrence has occurred after incomplete excision
of function of the SMARCB1/INI1/SNF5/BAF47 gene in chromosome by partial nephrectomy. If partial nephrectomy is considered, frozen
band 22q11.2 (Biegel et al., 1999). Consistent with its role as a section is indicated to exclude cystic partially differentiated nephro-
tumor suppressor gene, tumors arise after inactivation of both alleles blastoma (CPDN) or clear cell sarcoma, which can occasionally have
of SMARCB1. Germ-line alterations of the SMARCB1 gene are found a similar appearance.
in one-third of the patients (Eaton et al., 2011). Germ-line mutations Another entity reported in the literature with similar features is
of INI1 have been identified in renal rhabdoid tumors. Staining for CPDN. The majority of these lesions occur in the first 2 years of life
the products of the INI1 gene can be useful because rhabdoid tumor (Blakely et al., 2003; Joshi and Beckwith, 1989; Luithle et al., 2007).
of the kidney is consistently negative (Hoot et al., 2004). Eble and Bonsib (1998) recommend that multilocular cystic nephroma
Typical clinical features include early age of diagnosis (median and CPDN be considered the same entity. They are indistinguishable
age <16 months), advanced stage, resistance to chemotherapy, and radiographically. Histologic examination reveals that blastemal cells
high mortality (Amar et al., 2001; Tomlinson et al., 2005). Younger or nephrogenic rests may be found in the septa of both tumors.
age at diagnosis is an adverse prognostic factor. RTK is distinguished Surgery is curative in almost all patients with recurrence being the
by its propensity to metastasize to the brain (D’Angio et al., 1993). result of incomplete resection (Blakely et al., 2003; Eble and Bonsib,
There is no standard therapy for RTK. Intensification of therapy with 1998). In a review of 21 children with cystic partially differentiated
high-dose chemotherapy and autologous stem-cell transplantation nephroblastoma (CPDN) reported to the NWTSG, there was 100%
have not yielded improved survival (Furtwangler et al., 2018). survival. Eight were treated with surgery alone (Blakely et al., 2003).
Thirteen patients received postoperative chemotherapy, including
Congenital Mesoblastic Nephroma two patients with stage II disease.

CMN is the most common renal tumor in infants with a mean Metanephric Adenofibroma
age at diagnosis of 3½ months (Howell et al., 1982; van den
Heuvel-Eibrink et al., 2008). This is the most common renal tumor Another tumor with prominent stromal features that can resemble
diagnosed on antenatal ultrasound (LeClair et al., 2005). Congenital CMN is metanephric adenofibroma (Arroyo et al., 2000). The epi-
mesoblastic nephroma is a very firm tumor on gross examination, thelial component of these tumors can range from inactive meta-
and the cut surface has the yellowish-gray trabeculated appearance nephric adenoma to Wilms tumor. Other lesions contain areas
of a leiomyoma. There are three histologic subtypes: classic, cellular, morphologically identical to papillary renal cell carcinoma. This
and mixed (showing areas of classical and cellular). The classic uncommon entity may be derived from ILNR (Arroyo et al., 2000).
subtype, characterized by interlacing sheets of bland spindle cells, Metanephric adenofibromas with a composite Wilms tumor com-
resembles infantile fibromatosis. The cellular variant, with a solid ponent occur at a young age (mean of 12 months) similar to other
sheet-like growth pattern and frequent mitoses, is virtually identical ILNR-related Wilms tumors that develop in patients with DDS and
histologically to congenital fibrosarcoma (Beckwith, 1986; Gormley aniridia. None of these tumors have recurred after nephrectomy, but
et al., 1989; Joshi et al., 1986). Both tumors have a similar transloca- all have been treated with Wilms tumor chemotherapy.
tion that fuses the ETV6 (TEL) gene from 12p13 with the 15q25
neurotrophin-3 receptor gene, NTRK3 (Argani et al., 1998; Vokuhl Renal Cell Carcinoma
et al., 2017). A SIOP/GPOH showed that patients with cellular type
CMN who have translocation-positive tumors had a significantly Renal cell carcinoma (RCC) is the most common renal malignancy
superior RFS (5-year RFS: 100% vs. 73%). In CMN, tumor induction in the second decade of life. Only 5% of RCCs occur in children
is postulated to occur at a time when the multipotent blastema is (Broecker, 1991; Geller et al., 2015; Hartman et al., 1982). An
predominately stromagenic (Snyder et al., 1981; Tomlinson et al., abdominal mass is the most common presentation, but hematuria
1992). WT1 is not expressed in CMN (Tomlinson et al., 1992). is more common than in Wilms tumor (Broecker, 1991). Imaging
The most important aspect of CMN is the usually excellent studies cannot differentiate RCC from other solid renal tumors. There
outcome with radical surgery only (Howell et al., 1982). The tumor is a higher incidence of papillary RCC in children (Renshaw et al.,
can extend into the hilar or perirenal soft tissue; therefore complete 1999; Selle et al., 2006). However, the most common histology of
surgical resection is important (Beckwith, 1986). Local recurrence and RCC in children and adolescents, at approximately 50%, is
metastasis can occur particularly with the cellular variant of CMN translocation-associated RCC (Geller et al., 2015). These tumors,
(Fitchey et al., 2003; Gormley et al., 1989; Joshi et al., 1986). The typically seen in adolescence or young adults, are genetically
risk of recurrence is thought to be less in children under 3 months of unique in that they have chromosome translocations involving
age at diagnosis, but metastases have been reported in a few infants a common breakpoint in the TFE3 gene located at Xp11.2 (Bruder
(Heidelberger et al., 1993). Neither chemotherapy nor radiation therapy et al., 2004). These tumors differ from adult RCC in that immuno-
is routinely recommended (Howell et al., 1982), but consideration reactivity for epithelial markers is reduced or absent. There is also
for adjuvant treatment should be given to patients with cellular vari- an increased risk of lymph node involvement (40%–50%), and
ants that are incompletely resected (Gormley et al., 1989). There are presentation is at a higher stage than adult RCC (Geller et al., 2015).
reports demonstrating response of inoperable and recurrent tumors Another type of RCC more often seen in children is renal medullary
to chemotherapy (Loeb et al., 2002; McCahon et al., 2003). carcinoma found in patients with sickle cell hemoglobinopathy
(Swartz et al., 2002). The median age at presentation is 13 years,
Solitary Multilocular Cyst and Cystic Partially but it can be found in much younger children. It is a highly lethal
Differentiated Nephroblastoma tumor. A recent report from COG noted that 22 of 25 patients (88%)
with renal medullary carcinoma in their Biology and Banking Study
Solitary multilocular cyst, or multilocular cystic nephroma, is an (AREN03B2) were diagnosed with stage IV disease at diagnosis
uncommon, benign renal tumor. Fifty percent of multilocular (Sandberg et al., 2017).
1110 PART III Pediatric Urology

Complete tumor resection is the most important determinant


of outcome in RCC. Raney et al. (1983) found that all children
with stage I lesions survived, and others have reported 64% to 80%
survival for stage I and II tumors (Aronson et al., 1996; Castellanos
et al., 1974; Dehner et al., 1970). Partial nephrectomy has been
used in pediatric patients with RCC, but outcome data are limited
(Cook et al., 2006). Younger age at diagnosis is a favorable prognostic
factor (Raney et al., 1983). Regional lymph node involvement does
not portend the same poor prognosis as adult renal cell carcinoma
(Geller and Dome, 2004). Geller et al. (2008) reported that patients
with TFE+ translocation and positive nodes were found to have 93%
survival at a median follow-up of 4.3 years. However, others have
noted higher rates of relapse (Perlman, 2010). Like RCC in adults,
these tumors are generally not responsive to chemotherapy or radia-
tion therapy, although case reports of successful adjuvant treatment
have been published (Chowdhury et al., 2013).

Angiomyolipoma
Renal angiomyolipoma is a hamartomatous lesion that is only rarely Fig. 53.13. Angiomyolipoma of the right kidney in a patient with tuberous
seen in childhood. The majority of adult patients have sporadic sclerosis.
angiomyolipomas. In children, they are most commonly detected
in patients with the tuberous sclerosis complex (TSC), and presenta-
tion is more often bilateral in these patients (Bissler et al., 2016;
Ewalt et al., 1998; Warncke et al., 2017). Angiomyolipoma develops taken continuously. When discontinued after 12 months, the angio-
in 50% to 80% of patients with TSC. The lesions progressively enlarge myolipomata started to enlarge again, demonstrating that the mTOR
through childhood with an apparent growth spurt in teenagers and inhibitors must be continued (Bissler and Kingwood, 2016).
young adults. Warncke et al. (2017) noted increase annual growth A randomized placebo-controlled trial (EXIST-1) reported that
in those TSC patients older than 11 years and those with AMLs 42% of patients had at least a 50% reduction in total volume of
smaller than 2 cm in size. Annual ultrasounds are recommended angiomyolipoma after treatment with everolimus compared with 0
with consideration of MRI for those older than 11 years and having of placebo (Bissler et al., 2013). The patients were all age 18 years
lesions smaller than 2 cm. Other renal lesions found in the TSC or older. An extended follow-up study (EXIST-2) has shown that
include simple cysts, polycystic kidney disease, and RCC. Renal cysts 14% of patients will experience some progression of the angiomyo-
occur in up to 30% of patients with TSC. Mutations of one of two lipomas (Bissler et al., 2017). No renal bleeding or nephrectomies
genes on chromosome 9 (TSC1) and chromosome 16 (TSC2) are were reported.
found in 85% of TSC patients (Crino et al., 2006). It has been Some lesions are fat poor, and differentiation from other renal
postulated that these genes act as tumor suppressor genes and that tumors—for example, RCC—on imaging studies can be difficult
the LOH of TSC1 or TSC2 may explain the progressive growth pattern (Hindman et al., 2012). In some cases, biopsy of the lesion may be
of renal lesions seen in these patients (Henske, 2004). needed to confirm diagnosis of angiomyolipoma before proceeding
Patients with angiomyolipomas resulting from tuberous sclerosis with treatment. Nephron-sparing approaches are recommended in
are at risk for hemorrhage and impaired renal function (Bissler et al., children with TSC because of the presence of multiple, bilateral
2016). For many years, the threshold diameter for intervention has lesions and the risk of development of new lesions.
been more than 4 cm. Recent data in adult patients with sporadic
angiomyolipoma suggest that intervention can be delayed until the
diameter exceeds 6 cm (Kuusk et al., 2015). The International TSC KEY POINTS: RENAL TUMORS
Consensus Group recommends intervention for lesions larger than • eletions of WT1, located on chromosome 11p, are found
3 cm in diameter (Kingswood et al., 2016). Lesions in TSC patients in patients with aniridia and Wilms tumor. Mutations of
that are larger than 3 cm and enlarging appear to be at greatest risk WT1 occur in DDS.
for bleeding. The first-line treatment for children with growing • ocal recurrence is increased in ilms tumor patients with
lesions (Fig. 53.13) is with mTOR inhibitor, and embolization or local tumor spillage, and this is not classified stage III
surgery are second-line treatments. disease. The 2-year survival rate after local recurrence is
The main problems with embolization are risks of damage to 43%.
infarction of normal adjacent renal parenchyma and risk of short-term • Patients with bilateral ilms tumor should be treated with
complications and that it will not prevent other smaller lesions from preoperative chemotherapy. This will allow more patients
progressing. Repeat intervention is required in 35% of patients to undergo renal-sparing surgery in an attempt to decrease
undergoing embolization (Kuusk et al., 2015). Embolization is the risk of renal failure.
reserved for patients with active bleeding. • MN is the most common renal tumor in infants.
TSC1 or TSC2 mutations in patients with TSC give rise to hyper-
activation of the mTOR pathway. The first mTOR inhibitor studied
for TSC patients was sirolimus. The renal studies all showed that
sirolimus was very effective at preventing enlargement of the lesions REFERENCES
and also significantly reducing their size, while the medication was The complete reference list is available online at ExpertConsult.com.

You might also like