Renal Tumors

Hsin-Hsiao Wang, MD, MPH; Jonathan Routh, MD, MPH; Richard Lee, MD

→ Enlace a la versión en español

 

Wilms’ Tumor
Introduction
Max Wilms’ description of a solid kidney tumor of triphasic pathology was first published in 1899.[1]   At the time, the pathological entity which would eventually come to be known as Wilms’ tumor (WT) was a near-universally lethal disease, despite aggressive surgical and chemotherapeutic interventions. 

Luckily, however, the world of pediatric renal tumors has dramatically changed since that time.  Beginning in the 1960s and 1970s, cooperative multi-institutional and international trials such as the National Wilms’ Tumor Study Group (NWTSG), the Children’s Oncology Group (COG), the United Kingdom Children’s Cancer Study Group (UKCCSG), and the Société Internationale d’Oncologie Pédiatrique (SIOP), among others, began to coordinate the care and study of children with renal tumors.  Currently, the overall five-year survival rates for WT are roughly 90 percent in the most recent trials of NWTS/COG and SIOP groups.[2-4]

Epidemiology
WT is the most common solid renal malignancy in children with an annual incidence rate of approximately 7 to 10 cases per million for children younger than 15 years; tumors occur in approximately 500 children per year in the United States.[5-8]  More than three-quarters of WT cases are diagnosed before age 5 with median age at 3.5 years.  WT is seen slightly more in females than males.  There are racial/ethnic differences in the development of WT; for example, African-Americans are at a slightly higher risk of developing WT, whereas Asian-Americans are found to have a lower risk of WT compared with Caucasians.[7, 9]  Bilateral WT are diagnosed at an earlier age than unilateral tumors, typically at a mean of 2.5 years.[7, 10]

Multi-disciplinary management of WT has revolutionized disease outcomes, although at an associated higher cost.  This translates into worse outcomes in developing countries, where factors such as an inability to afford chemotherapy or proper follow-up may lessen survival rates.[11, 12]  The total chemotherapy-related costs have been estimated at approximately USD 1,500 to 4,500 in a previous NWTSG report.[13]  Follow-up imaging costs have also been reported to be substantial.[14] Efforts to develop more cost-effective protocols without sacrificing patient survival are under investigation.[13, 15-17]

Etiology/Biology
Histopathology
The classic triphasic WT histologic pattern includes blastemal cells, stromal cells, and epithelial cells.  Blastemal cells, frequently referred to as “small round blue cells,” are undifferentiated cells characterized by a tightly packed pattern with basophilia and high nuclear-to-cytoplasmic ratio.  Stromal cells are most commonly observed as immature smooth and skeletal muscle, osseous, adipose, or cartilaginous cells. Epithelial cells frequently form tubular, glomerular, or papillary structures.[18]  The diagnosis of WT is complicated by the fact that there may only be one or two of the cell types found instead of the classic triphasic appearance.[19]  
Anaplasia, defined as large nuclear diameter, hyperchromatism, and abnormal mitotic figures,[20] is defined as an unfavorable histologic (UH) feature; anaplasia has an approximate 5% incidence across NWTS and SIOP.  Since it was first described, anaplasia has been consistently associated with a poor prognosis – specifically, with a significantly higher rate of death and tumor recurrence.[21-23]  Anaplasia is further categorized as a focal or diffuse pattern, with the latter found to be associated with an even worse prognosis[22].  In the absence of anaplasia, other WT histology types are categorized as favorable histology (FH). 
In FH WT, differentiated epithelial-predominant cell type was found to be associated with earlier stage but poor chemotherapy response; on the contrary, diffuse blastemal pattern was highly aggressive with a good response to chemotherapy.  Whether the predominant cell type plays an important role in FH WT prognosis with modern WT treatment strategies has been disputed, with some data suggesting equivalent outcomes,[24] while other reports suggest that epithelial- or blastemal-predominant tumors may have a worse event-free survival rate.[25]  In anaplastic WT, blastemal cell predominance has been identified to be an independent predictor for poor outcomes.[26
Nephrogenic rests (NR) are precursor lesions to WT that are found in more than a third of resected WT.[27, 28]  Most NR does not progress into WT cells as they are incidentally identified in around 1% of infant autopsy studies.[29]  Distinguishing between hyperplastic NRs and WT can be challenging.[29]  WT can present with a pseudocapsule composed of compressed atrophic renal tissue adjacent to normal renal parenchyma.  NR can be further categorized into perilobar nephrogenic rests (PLNRs) and intralobar nephrogenic rests (ILNRs).  ILNRs are associated with WT1 related condition such as aniridia, WAGR, and Deny-Drash syndrome.  PLNRs are seen in patients with Beckwith-Wiedemann syndrome related to the 11p15 locus.

NRs tend to occur bilaterally.[27] WT patients with NR, especially PLNRs, should have frequent and regular surveillance of their contralateral kidney due to an increased risk for metachronous tumor development.[15, 30]  Diffuse hyperplastic perilobar nephrogenic rests are associated with a higher risk of developing WT and with an increased incidence of anaplasia.[31]

Genetics
Like other pediatric malignancies, the genetic components of WT development are the subject of extensive research inquiry, including associated syndromes.  One of the first genetic alterations associated with WT identified was WT1(located at 11p13) deletion in patients with WAGR syndrome - a rare syndrome with features of WT, aniridia, genitourinary abnormalities, and mental retardation.[32PAX6 gene (highly associated with aniridia) is adjacent to WT1 gene; higher WT rates in aniridia patients with deletion of both WT1 and PAX6 have been reported.[33] Interestingly, further research has revealed that WT1 plays an essential role in differentiating genitourinary tissue[34-36]. 
Unlike WAGR syndrome, which is commonly associated with WT1 deletion, children with Denys-Drash syndrome (male pseudohermaphroditism, renal mesangial sclerosis, and nephroblastoma) have WT1 missense mutation.[37, 38]  WAGR syndrome and Denys-Drash syndrome patients were found to have higher risk of bilateral WT.[39]
Beckwith-Wiedermann syndrome (BWS) constitutes features including macroglossia, macrosomia, hypoglycemia, visceromegaly, omphalocele, and predisposition to several tumors, most commonly WT.  Genetic mutations with BWS affect the WT2 gene, located at the 11p15 region.[40, 41]  Imprinted genes expressing either maternally or paternally inherited gene are found at the 11p15 locus and alterations to those imprinted genes (such as loss of imprinting (LOI) of IGF-2) may be responsible for WT development in subsets of patients.[42]
Loss of heterozygosity (LOH) is defined as loss of one of two copies of a chromosomal region also plays an important role in WT tumorigenesis.  Approximately 20% of WT patients have been recently reported to have LOH at 16q,[43]  while LOH at chromosome 1p occurs in 10% of children with WT.[44]  Data from NWTS-5 suggested that tumor-specific LOH at chromosome 16q and 1p are associated with significantly worse outcomes, specifically with increased relapse and death.[45]  By contrast, LOH at 11q has been associated with diffuse anaplastic tumors, but is not independently associated with worse outcomes unless the entire long arm of chromosome 11 is lost.[46]
New techniques such as DNA microarray expression profiling and array comparative genomic hybridization are being utilized more to investigate the molecular genetics of WT.  They provided new perspectives on WT genetics by examining global gene expressions in specific tumor types/histologies.[47, 48]

Other genes reported to be associated with WT include: WTX (a tumor suppressor gene on X chromosome) ,[49] CTNNB1 (β-catenin gene in WNT/β-catenin signaling pathway associated with WT1 and WTX in WT),[50, 51] FWT1 (located at 17q12-q21) and FWT2 (located at 19q13.4).[52, 53]

Biomarkers
In order to avoid the twin extremes of over-treatment with excessive complications and under-treatment with compromised survival, prognostic biomarkers that can achieve better risk stratification are actively sought worldwide.  Multiple biomarkers have been reported to predict WT recurrence, including markers of genetic abnormalities,[54] chromosomal LOH,[55] disordered apoptosis,[56] abnormal cell/nuclear proliferation,[57] anomalous growth factor production,[58] and altered regulation of tumor suppressor genes and/or oncogenes.[59, 60]
Loss of apoptosis in tumor cells is identified as one of the important mechanisms in tumor progression.  Apoptosis-associated regulatory protein Bcl-2, Bax , and Bcl-X were found to be related to WT progression.[56]  Survivin, an inhibitor of apoptosis, was reported to be potentially associated with favorable outcome of WT with decreased cytoplasmic survivin expression.[61]  The tumor suppressor protein p53 has been reported to occur more frequently in anaplastic WT and in more advanced disease stages.[62, 63]  Interestingly, p53 expression has been reported to be altered after chemotherapy.[59] Tumor expression of B7-H1, a cell-surface glycoprotein of the B7 family of T-cell coregulatory molecules, is associated with an increased risk of tumor recurrence and of treatment failure in patients with both favorable histology and anaplastic tumors.[64, 65]

Table 1- Syndromes associated with WT

Syndromes

Gene

Characteristics

Overgrowth syndromes

Beckwith-Wiedemann syndrome 

WT2

macrosomia, macroglossia, omphalocele, prominent eyes, ear creases, large kidneys, pancreatic hyperplasia

Perlman syndrome

DIS3L2

fetal gigantism, visceromegaly, unusual face, bilateral renal hamartomas with nephroblastomatosis

Sotos syndrome

NSD1

facial, extremity, and cognitive abnormalities

Simpson-Golabi-Behmel syndrome

GPC3

organomegaly, bulldog appearance, congenital heart disease, polydactyly

Non-overgrowth syndromes

WAGR syndrome

WT1

aniridia, GU abnomalities, Renal impairement, intellectual disability

Deny-Drash syndrome

WT1

progressive renal disease, male pseudohermaphroditism

Frasier syndrome

WT1

progressive glomerulonephropathy, male pseudohermaphroditism, streak gonads, gonadoblastoma

 

 

 

 


 

Associated Syndomes
 There are a number of congenital anomalies and syndromes associated with development of WT (Table 1).  These can be broadly categorized as overgrowth and non-overgrowth syndromes.  Overgrowth syndromes include Beckwith-Wiedemann syndrome (BWS), Perlman syndrome, Sotos syndrome, and the Simpson-Golabi-Behmel syndromes.  The most studied non-overgrowth syndromes are WAGR syndrome and Deny-Drash syndrome.  Other reported syndromes that are associated with WT include Frasier syndrome, familial WT syndrome, Mosaic variegated aneuploidy, Fanconi syndrome, Li-Fraumeni syndrome, neurofibromatosis, and Bloom–Torre–Machacek syndrome.

Prognostic Factors

Current WT treatment strategies depend heavily on risk of tumor progression and recurrence.  Major prognostic factors include tumor staging, histology, and biology markers. 
Staging criteria are base upon anatomical extent of the tumor without considering other factors such as histology or biological prognostic markers.  Higher stage tumor is associated with worse prognosis.  The most commonly used staging systems are developed from COG and SIOP.  Unilateral tumors are classified as stage I to IV; bilateral tumors are classified as stage V.  The major difference is that NWTS/COG stages tumor before chemotherapy is administered and SIOP stages are determined after initial chemotherapy.  The details of staging are addressed in detail below. 
Tumor histology is perhaps the most critical factor affecting prognosis.  Anaplastic histology, especially diffuse type anaplasia, forebodes poor prognosis. Studies suggested the presence of anaplasia was associated with higher rate of recurrence, metastasis, and death.[21, 26]  NWTS/COG determines favorable or unfavorable histology based on presence of anaplasia.  SIOP has three histologic classifications: low-risk (completely necrotic or cystic partially differentiated nephroblastoma); intermediate-risk (epithelial, stromal, mixed, regressive, or focal anaplastic nephroblastoma); and high risk (blastemal or diffuse anaplastic nephroblastoma).[66
Biological factors provide additional information to further stratify the risk and depict prognosis for WT patients.  In NWTS-5, LOH of either chromosome 16q or 1p was found to have a significantly higher rate of relapse independent of tumor stage or histology.[45, 55, 67]  LOH of 11p15 was associated with more tumor relapse in very low risk WT patients who were not treated with chemotherapy, but rather were surgically excised and then observed.[60]  
Historically, other factors such as age were also heavily considered in prognostic evaluation.  Children less than 24 months old were found to have lower relapse rate.[68]  With the new advancement in WT regimens, the role of age greatly diminished.[21, 69]

Presentation
            The typical presentation of WT is a palpable, firm, smooth, non-tender abdominal mass.  Other symptoms include abdominal pain, hematuria, fever, and hypertension.  Subcapsular hemorrhage can cause rapidly increasing in abdominal size, anemia, hypertension, and fever.  Tumor rupture can present as an acute abdomen. Tumor extension to renal vein or IVC can lead to varicocele, hepatomegaly, ascites, and congestive heart failure.[70]  Due to the fact that WT is commonly associated with congenital syndromes, children should be assessed for associated anomalies including aniridia, hemihypertrophy, or genitourinary abnormalities. 

Differential Diagnosis
            The differential diagnosis of WT includes neuroblastoma and other renal tumors.  Their histologic and other major characteristics are listed in Table 2.


Table 2- WT differential diagnosis

Diagnosis

Histology and Major characteristics

Neuroblastoma

Homer-Wright pseudo-rosettes: small, round and blue, and rosette patterns

accumulation and excretion of the intermediates HVA, VMA, and dopaminem in urine

most common cancer in infancy

Clear cell sarcoma of the kidney

cords and nests of pale-stained tumor cells with abundant extracellular matrix separated by a network of fine capillary arcades

bone-metastasizing renal tumor of childhood 

typically presents between the ages of two and three years of life

Rhabdoid tumor of the kidney

presence of monomorphic cells with vesicular nuclei and prominent nucleoli

metastatic presentation

occurs most frequently in children less than two years of age and almost never in those older than five years of age

Congenital mesoblastic nephroma

classic CMN: locally-infiltrative fascicles of spindle cells; cellular CMN: hypercellular with sheets of closely-packed spindle cells

associated with hypertension and elevated concentrations of calcium and renin

common within the first year of life

Renal cell carcinoma

papillary or pseudopapillary architecture with or without psammoma bodies and clear cells with copious cytoplasm

frequently associated with genetic translocations

more common during the second decade of life and beyond

Renal medullary carcinoma

large nuclei with prominent vesicles and nucleoli as well as intensely eosinophilic cytoplasm with intense inflammatory response

almost exclusively in African-Americans with sickle cell trait or sickle cell disease

reported mostly in adolescent and young adults

                                                                                      
Workup
            Laboratory tests should include complete blood count, renal and liver function tests, serum calcium, urinalysis, and a coagulation panel.  Complete blood count can detect anemia. Serum creatinine and BUN can help evaluate pre-interventional renal function.  Liver function tests may be abnormal with liver metastasis.  Hypercalcemia can be found in patients with rhabdoid tumor of the kidney or congenital mesoblastic nephroma.[71] Urinalysis may reveal proteinuria which can be seen in patients with Denys-Drash syndrome.  Acquired von Willebrand's disease has been reported in around 8 percent of WT patients at presentation, which is obviously significant for operative planning.[72]

Abdominal ultrasound is the first-line imaging study for children with an abdominal mass.  Doppler can be used to further detect tumor extension to renal vein or inferior vena cava that occurs in 4 percent of WT patients.[73]  Once the mass has been confirmed on US, CT scan should be ordered in order to provide more detailed anatomy of the renal mass, contralateral kidney tumor evaluation, and metastasis detection.  In contrast to earlier recommendations, thin-sliced helical CT is recommended rather than routine surgical exploration of the contralateral kidney.[74, 75]  Controversy exists in the choice of imaging study between chest CT and chest X-ray for lung metastasis.[76, 77]  MRI is utilized when tumor infiltration of renal vein or IVC detection is equivocal by ultrasound or when the patient cannot tolerate CT contrast dyes.

Staging

Tumor staging and histology are the most potent predictors of WT outcomes.  The most commonly used staging systems are developed by NWTS/COG and SIOP.  Unilateral tumors are categorized as stage I to IV and bilateral tumors at presentation are categorized as stage V.  The central difference lies in the timing of the staging.  NWTS/COG system assigns staging at primary surgery.  The staging system by SIOP is performed after pre-operative chemotherapy.  The detailed staging criteria are listed in Table 3.

table 3- NWTS/COG & SIOP staging systems

Stage

Criteria

Stage I

Tumor confined to the kidney and completely resected. Intact renal capsule.  No tumor rupture or renal sinus extension.

Stage II

Complete resection with negative margins and lymph node involvement.  Regional tumor extension (extracapsular penetration, renal sinus extension, vascular involvement)

Stage III

Residual nonhematogenous tumor confined to the abdomen (lymph nodes , any tumor spillage, pre-operative biopsy, tumor removed more than one piece)

Stage IV

Hematogenous metastases (lung, liver, bone, brain), or lymph node metastases outside the abdominopelvic region

Stage V

Bilateral renal involvement at diagnosis

Treatment
Unless a tumor is deemed to be unresectable, biopsy prior to chemotherapy is not typically recommended due to risk of tumor spillage and seeding – a biopsied tumor will typically be upstaged based on the violation of the tumor capsule.  This is controversial, however, as some centers have adopted pre-chemotherapy biopsy to reduce inappropriate treatment such as giving chemotherapy to benign conditions or inadequate chemotherapy to non-WT.[78-80]  UKCCSG trials demonstrated no significant difference of 5-year survival between immediate nephrectomy compared with initial biopsy with chemotherapy and delayed nephrectomy.[81]  Open biopsy rather than percutaneous biopsy is indicated in bilateral WT with poor chemotherapy response in order to achieve accurate assessment of anaplasia.[82]
Radical nephrectomy with transperitoneal approach is the mainstay of WT management.  Proper surgical staging along with tumor extent evaluation and abdominal cavity exploration are essential for adjuvant therapy planning including chemotherapy and radiation therapy.  Sampling of suspicious nodes is recommended for tumor staging as well.  Gentle handling of the tumor is mandatory throughout the entire process to prevent tumor spillage; spillage is known to be a significant risk factor for local tumor relapse and requires more aggressive adjuvant therapy.[83]
NWTS/COG and SIOP differ in terms of timing of surgical excision relative to chemotherapy.  The NWTS/COG approach (pre-chemo surgical excision) provides accurate assessment of the disease extent and tumor histology.  It also identifies patients with benign disease and thus avoids unnecessary chemotherapy.  By contrast, the SIOP approach (post-chemo surgical excision) has the advantage that surgery is often technically easier with reduced tumor size and lower tumor spillage rate after initial chemotherapy.[84]  Currently both groups report an excellent 5-year overall survival rate of approximately 90%.[2-4]

            Concerning chemotherapy, the most significant recent advance has been the adoption of dactinomycin and vincristine; these drugs dramatically improved WT patient survival even in advanced disease.  NWTS/COG and SIOP conducted multiple randomized trials to adjust the combination of regimen in order to further improve survival with lower complication rates.  The recommended NWTS/COG treatment protocol is organized in Table 4.

Table 4 - COG treatment protocol

Stage/Histology/Biomarker

Chemothereapy

Radiation

Stage I FH <2y/o <550gm

none

none

Stage I FH >2y/o >550gm

EE-4A

none

Stage II FH

EE-4A

none

Stage I-II FH with 1p/16q LOH

DD-4A

none

Stage III FH without 1p/16q LOH

DD-4A

yes

Stage I-III FA

DD-4A

yes

Stage I DA

DD-4A

yes

Stage III-IV FH with 1p/16q LOH

M

yes

Stage IV FH non-pulmonary mets

M

yes

Stage IV FH pulmonary mets

 

 

Lesions persist after 6 wks of chemo

M

yes

Lesions resected at diagnosis

DD-4A

yes

Lesions resolve after 6 wks of chemo

DD-4A

none

Stage II-III DA

UH-1

yes

Stage IV FA

UH-1

yes

Stage IV DA (no measurable disease*)

UH-1

yes

Stage IV DA (measurable disease)

VCR+IRIN window

yes

Stage IV DA (CR/PR after VCR+IRIN window)

UH-2

yes

FH= favorable histology; FA= focal anaplasia; DA= diffuse anaplasia

EE-4A= Vincristine, dactinomycin

DD-4A= Vincristine, dactinomycin, doxorubicin

M= Vincristine, dactinomycin, doxorubicin, cyclophosphamide, etoposide

UH-1=Vincristine, dactinomycin, doxorubicin, cyclophosphamide, etoposide, carboplatin

UH-2=Vincristine, doxorubicin, cyclophosphamide, etoposide, carboplatin, Irinotecan

VCR= vincristine; IRIN= irinotecan; CR= complete response; PR= partial response

*measurable disease: presence of lesion with longest diameter at least 1cm on CT or MRI

There are some exceptions in which COG protocols recommend pre-operative chemotherapy.  These include WT patients who are undergoing a planned partial nephrectomy/nephron sparing surgery, who have inoperable tumor, and who have caval thrombus extending above the hepatic veins.[10, 70, 85-87]
            In the SIOP protocol, a 4-week course of preoperative vincristine and dactinomycin are given prior to surgery.  Vincristine, dactinomycin, and doxorubicin are administered for six weeks preoperatively if there is any extra-abdominal metastasis.  The post-operative regimen is organized in Table 5.

table 5 - SIOP treatment protocol

Stage

Chemothereapy

Radiation

Stage I

AMD+VCR x 4wks

none

Stage II

AMD+VCR+DOX x 27wks

15 Gy at regional LN

Stage III

AMD+VCR+DOX x 27wks

15 Gy

Stage IV

AMD+VCR+DOX x 27wks

12 Gy if persistent lung lesion at wk 9

add ifosfamide, carboplatin, etoposide at wk9 if complete response not achieved for 34 wks

AMD, dactinomycin; DOX, doxorubicin;  VCR, vincristine

 

 

            The COG recommended radiation regimen in detail is organized in Table 6.


table 6 - Radiation treatment recommendation by COG

Treatment site

Clinical presentation

 

Flank irradiation

Stage III FH, Stage I-III FA or DA

 

Whole abdomen irradiation

Abdominal stage III

 

1. preop tumor rupture

 

2. peritoneal mets

 

3. tumor spillage

 

Whole lung irradiation

Lung mets

 

1. 1p/16q LOH

 

2. No CR after chemo at wk6

 

3. Other sites of mets

 

Whoe brain irradiation

Brain mets

 

Liver irradiation

focal mets not resected at diagnosis or diffused mets

 

Bone irradiation

Bone mets

 

Lymph node irradiation

Lymph node mets

 

Laparoscopic nephrectomy techniques have a limited role in WT management due to the concern for tumor spillage and inadequate tumor staging.  Duarte described the initial experience of laparoscopic nephrectomy on WT patients treated with neoadjuvant chemotherapy.[88]  Since then, there have been multiple reported series of laparoscopic nephrectomy for WT, typically restricted to patients treated with neoadjuvant chemotherapy.[89, 90

           

Historically, nephron sparing surgery (NSS) was generally reserved for bilateral WT or children with a solitary kidney in whom preservation of renal function was mandatory.[3]  Recently, this technique has garnered increasing attention from investigators.  Data on NSS in preserving long-term renal function, cardiovascular function, and overall health provide an insight of the potential wider applicability of NSS on selected WT.[91, 92]  The AREN0534 trial of COG allows for NSS in children with bilateral WT, or for unilateral WT in children with syndromes which predispose to renal failure or bilateral WT.[93]  Although contemporary data on NSS seems quite promising,[89, 94-96] data on NSS outcomes in children with WT remain scarce.  As practice patterns continue to evolve, NSS will likely be primarily utilized mainly at high-volume referral centers. 

Bilateral WT
            Bilateral WT is found in roughly 5-7% of WT patients.[10, 97]  The major challenge in these children is to preserve renal function while adequately treating the tumors.  Historically, bilateral WT was managed by radical nephrectomy of the more involved side and NSS on the contralateral kidney.  However, these patients were found to have a significant risk of ESRD due to tumor recurrence and subsequent nephrectomy.[98, 99] Modern management of bilateral WT incorporates neoadjuvant chemotherapy along with NSS as first-line treatment.[100-102] The most current COG protocol (AREN0534, as noted above) recommends 6 weeks of vincristine, dactinomycin, and doxorubicin, followed by re-evaluation.  Depending on tumor response, the patient then receives either an attempt at NSS or an additional 6 weeks of chemotherapy. Following surgery, adjuvant chemotherapy and radiation therapy regimen depend on tumor histology and staging.  Open bilateral biopsy may be considered if the tumor has a poor response to adjuvant chemotherapy; poor chemotherapy response has been reported to be due to tumor differentiation, which has a good prognosis if completely resected.[82, 103]

Of note, true tumor enucleation is generally reserved for large, centrally located tumors due to the concern for positive margin.  Anaplasia with positive margin will require additional resection to avoid worse survival.[102, 104-106]

Late Effects of WT Treatments

            WT survivors may suffer from long-term complications depending on the therapy regimen.[107] The major reported sequelae include development of a second malignancy, cardiotoxicity, renal impairment, musculoskeletal problems, and reproductive issues. Patient who received more aggressive chemotherapy and radiation therapy are obviously at higher risk.
            Second malignancy is one of the concerning late effects from WT therapy.  WT survivors have been reported to have a 1% cumulative incidence at 10 years post-diagnosis and increasing incidence developing second tumor thereafter.[108-110] Radiation therapy is associated with a significantly higher risk of late tumor; most second tumors have been reported to occur within the radiation field.[109, 110] Additionally, doxorubicin may potentiate this radiation effect.[111]
            Cardiotoxicity is a well-established late effect of doxorubicin, specifically myocardial dysfunction and reduced contractility.  Common manifestations include congestive heart failure and increased left ventricular afterload.[112, 113] The risk of cardiac dysfunction is closely related to the total cumulative dose of doxorubicin.
            Renal impairment in WT survivors can be caused by loss of renal parenchyma from surgery or from nephrotoxicity related to chemotherapy and radiation.  Severe renal dysfunction is usually associated with bilateral WT involvement especially if they receive radiation therapy.[98, 114, 115] Other significant risk factors for late renal impairment include Denys-Drash and WAGR syndromes in addition to radiation nephritis.
            Musculoskeletal problems such as scoliosis and short stature were found to be associated with radiation treatment.[116-118] Females who received high-dose abdominal radiation therapy are at risk of abnormal ovary development, higher incidence of infertility, spontaneous miscarriages, premature birth and intrauterine growth retardation of offspring.[119-121]

Prognosis       
            Overall five-year survival rate of WT patient has dramatically improved to greater than 90% at present.[2-4] Event-free survival (EFS) rates rely heavily upon prognostic factors like tumor staging, histology, biologic markers, and to a lesser extent, age at diagnosis and tumor weight. 

           

In the past, only 30% of WT patients survived with tumor recurrence.  With salvage therapy, the five-year survival rate of patient who have recurrence improves to 50-60% [122, 123].  Lung is the most common site for recurrence.  Prognostic factors that are associated with favorable prognosis include favorable histology, lower tumor stage, lower number of pulmonary nodules, no prior radiotherapy, recurrence beyond 6-12 months after initial diagnosis, only dactinomycin and vincristine as initial treatment, and complete resection of original tumor.[2, 123, 124] The ideal salvage therapy strategy remains under investigation.[122, 125-129]

Non-Wilms’ Pediatric Renal Tumors
Congenital Mesoblastic Nephroma
Congenital Mesoblastic Nephroma(CMN) is the most common renal tumor in infants within the first several months of life, with 90% diagnosed within the first year of life.[130, 131]  In one large series, the median age at diagnosis was reported to be 19 days.[132]  Beyond three years of age, CMN is rarely encountered.[130-136]  CMN has been noted to be more common in males, with a roughly 2:1 male to female ratio.  CMN is thought to originate from the nephrogenic mesenchyme, with two distinct histologic subtypes: the classic and cellular varieties. 
            Classic CMN typically presents with a lower tumor stage, smaller tumor volume, and at a younger age than cellular CMN.[132]  Classic CMN was initially described in 1967 and noted to be composed of locally-infiltrative fascicles of spindle cells.  Vascular invasion at the tumor periphery has been reported.[137]  By contrast, cellular CMN is hypercellular, with sheets of closely-packed spindle cells.[138, 139]  Both types may coexist in a mixed pattern as well. 

Recent studies have shown that the classic and cellular variants have differing genetic features.  In the cellular variant, a t(12;15)(p13;q25) translocation has been shown to produce a ETV6-NTRK gene fusion product which over activates tyrosine kinase signaling; this translocation has not been demonstrated in classic CMN, however.[140, 141]  Interestingly, this same translocation is found in infantile fibrosarcoma, leading many authors to conclude that cellular CMN is in fact the renal manifestation of that disease process.[140, 141]
Prognosis for CMN is generally excellent, particularly in comparison to other renal tumors.[131]  Most patients can be successfully treated with radical nephrectomy alone; attempts to preserve renal parenchyma in these patients through the use of partial nephrectomy are often unsuccessful, perhaps due to the tumors’ locally invasive nature.[132]  For patients with Stage III tumors, particularly older infants with the cellular variant, some authors have suggested the use of adjuvant chemotherapy.[132]  Given the relative chemosensitivity of infantile fibrosarcoma, this approach would seem logical in high-risk patients.[142

Clear Cell Sarcoma of the Kidney
Clear Cell Sarcoma of the Kidney (CCSK) is one of the more common “high-risk” renal tumors of childhood, representing 3% of pediatric renal tumors in the United States.[143]  CCSK is typically described as a large, homogenous, and often multi-cystic mass arising from the renal medulla; necrosis and hemorrhage are common.  Histologically, these tumors are classically defined by cords of oval or spindle-shaped cells separated by regularly spaced fibrovascular septa.[20, 143, 144]  CCSK typically presents between the ages of two and three years of life (range 2 months to 14 years) with a 2:1 male to female ratio.  No cases of bilateral CCSK have been reported.[131, 143

Perhaps the most notable feature of CCSK is its propensity to metastasize to bone.   Among 351 tumors from the NWTSG, 4% of CCSK tumors presented with metastatic lesions; after lymph node involvement (20%), bone lesions were the most common metastatic site at presentation (1.4%), followed by lung and liver (1.1% each).  Importantly, bone was the most common site of tumor recurrence (14%), followed by lung (11%) and abdomen (7%).[143]  CCSK recurrences commonly occur years after the initial diagnosis; interestingly, this phenomenon is heightened among patients receiving doxorubicin, despite a reduced overall recurrence rate in patients receiving this drug.[143

Overall survival among children with CCSK is reduced as compared to other renal tumors.  Eight-year overall survival has been reported to be as high as 83% among children 1 to 3 years of age; however, this figure is reduced among both infants and children older than 4 years of age.[131, 143, 145]  Other negative prognostic factors include increased tumor stage and the presence of histologic tumor necrosis.  Therapy with doxorubicin is perhaps the most important modifiable risk factor for tumor recurrence, as its use reduced the risk of relapse by 68% in the largest series reported to date.[143]

Rhabdoid Tumor of the Kidney
            Rhabdoid Tumor of the Kidney (RTK) is a rare but lethal pediatric renal tumor, comprising less than 2% of all renal tumors in the combined European and US data.[131]  Initially described as a sarcomatoid variant of WT, RTK was subsequently recognized as a distinct pathologic entity.[20]  RTK is characterized histologically by the presence of monomorphic cells with vesicular nuclei and prominent nucleoli.  Immunohistochemical staining commonly shows perinuclear vimentin staining,[146] as well as complete lack of expression of the tumor suppressor gene product INI1.[147]  
RTK commonly presents with hematuria and metastatic disease.[131, 148]  Median age was 11 months (range 1 month to 5 years) with a slight male predominance (male to female ratio 1.4:1).  In a combined analysis of data from both NWTSG and European trials, 29% of infants with RTK presented with metastatic disease, far more than other tumor types.[131]    Importantly, RTK has been noted to have a predilection for concurrent central nervous system involvement, which may be either metastatic RTK or synchronous primary CNS malignancies.[148]  Among RTK patients with metastatic disease at presentation, the most common metastatic sites were lung (83%), brain (73%), liver (7%), and bone (5%).[131]Overall survival among patients with RTK in the NWTSG trials was 23%.   Survival has been shown to be decreased with higher stage tumors (42% vs. 16% 4-year survival), presence of CNS tumors (universal mortality in NWTS 1-5), and decreased patient age (41% 4-year survival above age 2 years vs. 9% survival below age 6 months).  By contrast, patient gender, chemotherapy regimen, and the use of radiotherapy have not been associated with survival.[148-150]  Recently, cases of long-term survival after autologous stem cell transplant have been reported.[151

Renal Cell Carcinoma/Translocation Renal Cell Carcinomas
Although WT is the most common primary renal neoplasm during the first decade of life, renal cell carcinoma (RCC) is more common during the second decade of life and beyond.[152, 153]  RCC constitutes 2-6% of pediatric renal tumors overall,[154-156] and with a roughly 2:1 female to male ratio is more common among girls.[157-160]  Pediatric RCC has recently been shown to significantly differ from adult RCC, in terms of both tumor biology and clinical behavior.[157, 161-163]
  In particular, pediatric RCC has been characterized by its unique genetic features, specifically by chromosome Xp11 or t(6;11) translocation and lack of chromosome 3p mutations.  This insight has led to the coining of the terms “translocation RCC” or “translocation carcinoma of the kidney”, now recognized as a distinct pathologic subtype under the 2004 WHO classification.[164-166]  Xp11.2 is the locus of the TFE3 gene, a member of the microphthalmia-associated transcription factor (MiTF) family; at least 6 specific TFE3-related cytogenetic translocations and gene fusion products have been reported, with the most common being ASPL-TFE3 (t(X;17)(p11.2;q25)) and PRCC-TFE3 (t(X;1)(p11.2;q21)).[167-169]  A second member of the MiTF family, TFEB, is also associated with a subset of pediatric RCC, characterized by the Alpha-TFEB gene fusion product and a t(6;11)(p21;q12) translocation.[164, 170] Histologically, Xp11.2 translocation RCC most commonly exhibits papillary or pseudopapillary architecture with or without psammoma bodies and clear cells with copious cytoplasm.[160] t(6;11) translocation RCC is morphologically similar, but tends to stain positive for HMB-45, while Xp11.2 translocation RCC typically does not.[164]  Importantly, the development of translocation RCC has been recently reported to be associated with a history of exposure to cytotoxic chemotherapy, particularly topoisomerase II inhibitors or alkylating agents such as cyclophosphamide.[166, 171]  The mechanism behind this association is at present unclear.
Just as translocation RCC can also occur in adults, so too can non-translocation RCC occur in the pediatric age range;[168] however, the average age of non-translocation RCC patients is between 60 and 65 years.[172]  Although non-translocation RCC may develop spontaneously even in young patients, multiple familial syndromes predispose to the development of RCC at young ages.  In particular, von Hippel-Lindau syndrome,[173, 174] tuberous sclerosis,[175] Birt-Hogg-Dube,[176] hereditary papillary renal carcinoma,[177] and hereditary leiomyomatosis renal cell carcinoma[178] have been reported to be associated with the development of RCC among children and adolescents.[175, 179, 180]   
In addition to genetic and etiologic differences, pediatric RCC differs from adult RCC in terms of clinical behavior.  While adult RCC patients with lymph node involvement tend to fare poorly, pediatric RCC patients with lymph node involvement have a surprisingly good prognosis.[160, 161]  Importantly, these tumors have been reported to occur more commonly than expected among children who have experienced prior chemotherapy.[159, 166, 181]  Depending on tumor characteristics (in particular tumor size), some authors have noted that some pediatric RCCs may be suitable for treatment with partial nephrectomy;[158] the role of retroperitoneal lymph node dissection in these patients has been debated as well.[182, 183]  Regardless of treatment strategy, the prognosis for RCC has been shown to be somewhat better than for other pediatric renal tumors, with five-year survival rates of 72-87%.[155, 161

Renal Medullary Carcinoma
Renal Medullary Carcinoma (RMC) is a rare and extremely aggressive primary renal tumor which has been reported to occur almost exclusively in African-American adolescents and young adults with sickle cell trait or sickle cell disease; indeed, RMC has been labeled as the “seventh nephropathy” of sickle cell disease.[184]  RMC was originally described in 1995, with fewer than 100 cases of RMC described since then.[184, 185]  The reported mean age of RMC patients is 21 years, ranging from 5 to 39 years, with a 2:1 male to female ratio.[186, 187
As their name implies, RMC tumors arise centrally within the kidney, perhaps from the transitional epithelium of the renal calyces; RMC is generally believed to be a variant of collecting duct RCC.[188]  Tumor growth patterns is often reticular, though a variety of growth patterns have been described.[189]  Microscopically, RMC is characterized by large nuclei with prominent vesicles and nucleoli as well as intensely eosinophilic cytoplasm; the tumors are often accompanied by an intense inflammatory response.[184]  Cytologically, RMC appears similar to other high-grade carcinomas.[190]   Similar to RTK, RMC specimens have also been shown to have complete loss of INI1 expression.[189

RMC often presents with gross hematuria and abdominal or flank pain, reflecting its propensity to invade the renal sinus and/or pelvis as well as other local structures.  Of 61 patients reported in the two largest case series, all presented at tumor Stage III or IV.[184, 187]  Despite a wide variety of attempted therapies,[185, 191] the prognosis of RMC patients is dismal.  With the exception of a single patient who was alive at 2 years’ follow-up (and whose tumor was discovered at 1.8 cm in size),[192] the longest reported survival of any RMC patient is 17 months.[193]

Renal Angiomyolipoma
Renal angiomyolipoma (AML) is a generally benign neoplasm of the kidney consisting of a varying proportion of dysplastic blood vessels, smooth muscle, and adipose tissue.[194]  These tumors are found much more frequently in women than men (female to male ratio 4:1) at a mean age of 48 years (range 1-86).  Though classically associated with tuberous sclerosis (TS), 80% of AML occur spontaneously.[195]  Among TS patients, however, up to 70% may develop AML, and up to 10% will have renal complications due to their AML.[196, 197]  AML in TS patients tend to develop at a young age, often during infancy; once present, AML is likely to grow over time.  This has prompted some authors to recommend annual renal ultrasound screening of TS patients.[196

AML is typically a solid tumor, though cystic variants have been described.[198]  The triphasic components of AML are classically present in roughly equal proportions, although any one can be predominant in individual tumors.[194]  On imaging studies, fat-poor AML in particular can be quite difficult to differentiate from RCC or other renal tumors.[199]  AML is generally located at the renal poles, may be bilateral or multifocal, and may extend into the perirenal fat.  Classic AML is most often cytologically bland, although the smooth muscle component may assume an atypical appearance with hyperchromatic nuclei and prominent nucleoli.[194]  Recently, a rare malignant AML variant known as epithelioid AML has been reported, with the possibility of widespread tumor metastasis.[200]
Although AML may be incidentally discovered on imaging studies, 60% of patients present symptomatically, most commonly with flank pain, a palpable mass, or hematuria.[195]  In 15% of patients, AML presents with retroperitoneal hemorrhage (Wunderlich syndrome), which can be life-threatening.[195, 201]  Previous studies have shown that tumor size of 4 cm or greater is the most significant predictor of tumor rupture; it is therefore recommended that asymptomatic AML larger than 4 cm be treated in order to prevent their rupture.[201]  Although rare, AML-related renal hemorrhage during pregnancy may result in fetal or maternal demise; thus this guideline is particularly recommended for women of child-bearing age who desire to become pregnant.[195, 202]

 

Given the generally benign nature of AML, and given the propensity for multiple or recurrent tumors in TS patients, preservation of renal function is a paramount concern in treatment planning.  Nephron-sparing techniques such as partial nephrectomy[199, 203, 204] or vascular embolization[205, 206] are thus preferred to radical nephrectomy in most patients, particularly children and young adults.  Although it is more invasive than vascular embolization, partial nephrectomy is less likely to result in the need for repeated treatments; up to 40% of embolized tumors require further therapy.[203-206]   

 

Metanephric tumors: Metanephric Adenoma, Metanephric Adenofibroma, & Metanephric Stromal Tumor
Metanephric tumors represent a spectrum of tumor pathology, varying based on the relative proportion of stroma and epithelium. Some authors have suggested that these tumors represent the most hyperdifferentiated, mature form of WT.[207]   These tumors arise from the renal medulla and are generally benign, although there have been rare (and controversial) reports of malignant behavior.[207, 208]

Metanephric adenoma (MA) is a purely epithelial lesion occurring across a wide range of ages (5 to 83 years in the largest reported series).  Most patients with MA are adults, though children may be affected as well; there is a roughly 2:1 female predominance.[209, 210]  The tumors are typically solid, though cysts may occur.  Microscopically, these tumors are unencapsulated but well demarcated, and are composed of embryonal epithelial cells forming regular, closely packed tubular structures.  Mitotic activity is rare.[209, 210]  On immunohistochemistry, MA stains positive for WT-1, lending support to its putative connection with WT;[211] further support comes from reported cases of MA occurring in patients with hemihypertrophy.[212]  Clinically, MA tends to be found incidentally or as a palpable mass; approximately 10% of MA patients also present with polycythemia, perhaps due to direct production of erythropoietin.[213]  Given their benign behavior, these tumors are generally best managed by partial nephrectomy.[199]

Metanephric stromal tumor (MST) is a purely stromal lesion occurring predominantly in children (median age 13 months, range 2 days to 11 years), though rare adult cases have been reported.[214, 215]  Both sexes are equally effected.  Like MA, MST are more commonly solid, though 10% demonstrate cystic components, and are typically unencapsulated.[214]  Unlike MA, these centrally-located lesions often infiltrate into the renal pelvis and occasionally into the renal sinus and ureter.[214, 216]  Microscopically, MST is also superficially infiltrative, enclosing native renal tubules and vessels in an “onion-skinning” pattern; these “collarettes” are typically surrounded by bland spindle cell fascicles.  Because of their tendency to entrap entire glomeruli, MST induce juxtaglomerular cell hyperplasia in up to 25% of affected kidneys, resulting in rennin-dependent hypertension. On immunohistochemistry, MST tends to stain positive for CD34 and vimentin.[214]  MST has been hypothesized to arise from intralobar nephrogenic rests, further supporting the connection between WT and metanephric tumors.[207, 214]  Clinically, these tumors may present with a palpable renal mass or with hematuria due to renal pelvis involvement; MST are generally treated with nephrectomy, though partial nephrectomy may be an adequate treatment if negative margins can be achieved.[214-216]  There are no reports of MST exhibiting malignant behavior.[207]

Metanephric adenofibroma (MAF) is a biphasic epithelial & stromal tumor, with a varying ratio of epithelium to stroma.[217, 218] Although the stromal component is identical to the bland spindle cell stroma seen in MST, the epithelial component may be more mitotically active than MA.[217, 218]   Importantly, either WT and papillary RCC may arise from the epithelial component of MAF,[217] and spindle-cell sarcomas (dubbed “metanephric adenosarcoma”) may arise from the stromal component.[219]  Similar to MA and MST, MAF is also unencapsulated, but with indistinct borders.  These tumors generally presents in children, though adults may be affected (median age 2.5 years, range 5 months to 36 years); males are more commonly affected (male to female ratio 2:1).[217]  As with MST, most patients have been treated with radical nephrectomy, though partial nephrectomy is likely a reasonable option provided that negative margins can be obtained.  Because most of the reported patients were initially diagnosed with WT, most received adjuvant chemotherapy; it is thus impossible to comment on the natural history of patients treated with surgery alone.  However, prognosis in patients with available follow-up has been universally excellent.[217]

Cystic Kidney Tumors: Cystic Nephroma, Cystic Partially Differentiated Nephroblastoma, Cystic Renal Cell Carcinoma, and Mixed Epithelial Stromal Tumors

Cystic renal tumors are relatively uncommon among young children, accounting for 0.5% of the renal tumors reported to both NWTSG and SIOP.[220, 221]  The first description of these tumors was published in 1892;[222] since then, however, the classification of these entities has proven controversial.[223-226]  Although they are considered as separate entities under the 2004 WHO classification, some authors have suggested that Cystic Nephroma (CN), Cystic Partially Differentiated Nephroblastoma (CPDN), and Mixed Epithelial Stromal Tumors (MEST) represent a pathologic spectrum which may be related to WT.[225, 226]  In general, these tumors follow a benign course, with surgical resection alone typically sufficient to ensure a cure.[220, 221, 224, 227, 228]

Like many renal tumors, CN can occur in adults or children.  While in adults CN occurs far more commonly in women (female to male ratio 8:1), the opposite is true in children (female to male ratio 1:2).[226]  In children, these tumors often present as a palpable abdominal mass at a median 18 months of age.[221, 224, 229]  Pathologically, CN is defined according to the criteria of Joshi and Beckwith[224] as a discrete cystic tumor, well-demarcated from the normal non-cystic renal parenchyma, without significant solid components.  The cysts are lined by flattened, cuboidal, or hobnail epithelium, while the septa contain only fibrous tissue or well-differentiated renal tubules.[224]  Owing to their benign clinical course and well-demarcated structure, these tumors can be removed using nephron-sparing surgery with no need for adjuvant chemotherapy.[227, 230
Similar to pediatric CN, CPDN occurs predominantly among boys, with a 3:1 male to female ratio.[220]  These tumors also present similarly to WT and CN, most commonly as a palpable abdominal mass at a median age of 16 months.[220, 224]  CPDN is pathologically quite similar to CN, with the primary difference between the two being the presence of immature or blastemal cells in the septa of CPDN.[223, 224]  Two pathologic subtypes of CPDN, conventional and papillonodular, have been described.[228]  Also similar to CN, CPDN follows a generally benevolent course with few reported tumor recurrences following surgical removal, which can be performed by either partial or radical nephrectomy.[220, 221

MEST is another tumor which is clinically and pathologically similar to CN; several authors have proposed that these tumors are in fact the same identity.[225, 226, 231]  However, unlike CN or CPDN, MEST occurs almost exclusively in adults, and as such will not be discussed in detail here.
Lastly, multilocular cystic RCC can also occur among children, though they are markedly more common among adults.[232, 233]  Cystic RCC is pathologically and taxonomically distinct from other pediatric cystic neoplasms such as CN or CPDN,[223, 233] and is generally considered to be a rare variant of clear cell RCC.[232, 234]  Cystic RCC can occur in the setting of genetic predisposition, such as VHL or TS, or in the setting of acquired renal conditions such as acquired cystic kidney disease.  Importantly, cystic RCC is characterized by a uniformly good prognosis following surgical resection by partial or radical nephrectomy.[232-234

Miscellaneous Rare Renal Tumors
Intrarenal Neuroblastoma
Although neuroblastoma has been previously reported to arise from within the kidney parenchyma,[235, 236] this entity is described in detail elsewhere in this text and we will therefore refrain from discussing it further here.  

Desmoplastic Small Round Cell Tumor
            Desmoplastic Small Round Cell Tumor (DSRCT) is an uncommon but aggressive tumor, with roughly 150 cases described since the original report in 1989.[237]  DSRCT most commonly arises from the peritoneal serosa, with roughly 25% of tumors found in genitourinary sites.[237, 238]  Rarely, these tumors may originate within the renal parenchyma.[239]  Regardless of site of origin, DSRCT is characterized by a single chromosomal translocation, t(11;22)(p13;q12).  This translocation results in the fusion of exon 7 of the Ewing sarcoma gene with exon 7 of Wilms’ tumor suppressor gene, leading to a unique gene fusion product, EWS-WT1.[240-242

As the name implies, DSRCT are microscopically characterized by sheets of abundant small undifferentiated cells with scant cytoplasm and hyperchromatic nuclei.  DSRCT of the kidney typically demonstrates abundant mitotic figures and prominent apoptosis and necrosis.  The tumors are unencapsulated, and tumor borders are uniformly infiltrative.[239, 241]  Because the histologic appearance of DSRCT can easily be confused with other tumor types, notably blastema-predominant WT and PNET,[243] the diagnosis of DSRCT should be applied to a primary renal tumor only in the presence of EWS-WT1staining.[239]
With the exception of a single case report in an adult,[244] DSRCT of the kidney occurs in young children (mean age 6.8 years), and, in contradistinction to extrarenal DSRCT, is slightly more common in girls.[239, 245]  Tumors tend to appear hypovascular, heterogeneous, and well-circumscribed by ultrasound and computed tomography.[246]  Although extrarenal DSRCT has a dismal prognosis, 2 of 4 reported patients had no evidence of disease at last follow-up, while the remaining 2 have had a stable clinical course following multi-modality therapy.[239

Primitive Neuroectodermal Tumor
            Primitive Neuroectodermal Tumor (PNET) is thought to be a part of the Ewing’s sarcoma family, most commonly found in bone or soft tissue.  Although Ewing’s is a relatively common tumor, primary intrarenal PNET is quite rare, with roughly 50 pediatric cases reported in the literature.[247-252]  PNET is frequently noted to have one of two balanced chromosomal translocations (t(11;22)(q24;q12) and t(21;22)(q22;q12)), which result in fusion of the EWS gene with FLI1 or ERG, respectively.[253]  These gene fusion products are expressed in >95% of tumors and can be readily detected by FISH or PCR for diagnostic purposes.[249, 250
            Intrarenal PNET typically occurs in adolescents or young adults at a median age of 24 years (range 4 to 66 years).[247]  Histologically, intrarenal PNET is identical to PNET in other locations, with sheets or rosettes of small ovoid cells and a high nuclear to cytoplasmic ratio.[251]  Strong membrane immunohistochemical staining for CD99 is a nearly universal feature.[248
PNET typically presents with abdominal or flank pain, hematuria, or the presence of a palpable mass; most tumors are detected at an advanced stage.[247, 248]  Inferior vena cava thrombi are not uncommonly present (33%) at the time of diagnosis, as are metastases to the lymph nodes (25%), lung (20%), and liver (14%).[247, 249]  Of patients with available follow-up data, 43% had died of disease within 2 years of their diagnosis, despite an aggressive multi-modality treatment regimen.

Anaplastic Sarcoma of the Kidney
            Anaplastic Sarcoma of the Kidney (ASK) has only recently been described as a distinct pathologic entity.[254]  These tumors are quite rare with only 21 reported cases,[254, 255]  and may be confused with anaplastic WT.  Grossly, ASK is typically a large, soft, and friable tumor, with mixed cystic and solid architecture in one-third of cases.  Histologically, these tumors consistently demonstrate a spindle cell component arranged in fascicles; as the name implies, anaplasia is universally present, with “bizarre, pleomorphic, multinucleated” cells commonly present.  Chondroid differentiation is commonly present, and ASK tumors often stain positive for vimentin and desmin.[254]

 ASK occurs at a median age of 5 years (range 10 months to 41 years), with a slight female predominance (female to male ratio 1.5 to 1).[254]  A palpable renal mass and/or flank pain are the most common presenting symptoms, with hematuria occurring less frequently.  The most common sites of metastases were lung (15%), liver (10%), and bone (10%). Of 14 patients with known follow-up, 3 (21%) had died at a mean 1.6 years after diagnosis; the remaining 11 (79%) were alive at a mean follow-up of 7.4 years.[254, 255]

Miscellaneous Rare Primary Renal Tumors
In addition to the non-WT pediatric primary renal tumors detailed above, multiple other neoplastic entities have been reported to rarely originate within the kidney parenchyma.  These include tumors which are typically extrarenal or typically occur in adults, such as synovial sarcoma,[256, 257] rhabdomyosarcoma,[258] lymphoma,[259, 260] leiomyosarcoma,[261] liposarcoma,[262] malignant fibrous histiocytoma,[263] and even melanoma.[264]  However, it should be noted that these tumors are prohibitively rare among children. 

REFERENCES
1.         Wilms, M., Die Mischgescwülste. 1899: Leipzig, Germany.
2.         Metzger, M.L. and J.S. Dome, Current therapy for Wilms' tumor. Oncologist, 2005. 10(10): p. 815-26.
3.         D'Angio, G.J., et al., Treatment of Wilms' tumor. Results of the Third National Wilms' Tumor Study. Cancer, 1989. 64(2): p. 349-60.
4.         Tournade, M.F., et al., Optimal duration of preoperative therapy in unilateral and nonmetastatic Wilms' tumor in children older than 6 months: results of the Ninth International Society of Pediatric Oncology Wilms' Tumor Trial and Study. J Clin Oncol, 2001. 19(2): p. 488-500.
5.         Gurney, J.G., et al., Incidence of cancer in children in the United States. Sex-, race-, and 1-year age-specific rates by histologic type. Cancer, 1995. 75(8): p. 2186-95.
6.         Gurney, J.G., et al., Trends in cancer incidence among children in the U.S. Cancer, 1996. 78(3): p. 532-41.
7.         Breslow, N., et al., Epidemiology of Wilms tumor. Med Pediatr Oncol, 1993. 21(3): p. 172-81.
8.         Bernstein L, L.M., Smith MA, et al. , ed. Cancer incidence and survival among children and adolescents: United States SEER Program 1975-1995. SEER Program. Bethesda, MD, National Cancer Institute 1999, SEER Program. Bethesda, MD, National Cancer Institute 79.
9.         Breslow, N., et al., Ethnic variation in the incidence, diagnosis, prognosis, and follow-up of children with Wilms' tumor. J Natl Cancer Inst, 1994. 86(1): p. 49-51.
10.       Blute, M.L., et al., Bilateral Wilms tumor. J Urol, 1987. 138(4 Pt 2): p. 968-73.
11.       Abdallah, F.K. and W.M. Macharia, Clinical presentation and treatment outcome in children with nephroblastoma in Kenya. East Afr Med J, 2001. 78(7 Suppl): p. S43-7.
12.       Uba, A.F. and L.B. Chirdan, Childhood Wilms' tumour: prognostic factors in North Central Nigeria. West Afr J Med, 2007. 26(3): p. 222-5.
13.       Green, D.M., et al., Effect of duration of treatment on treatment outcome and cost of treatment for Wilms' tumor: a report from the National Wilms' Tumor Study Group. J Clin Oncol, 1998. 16(12): p. 3744-51.
14.       Carrico, C.W., et al., Wilms tumor imaging: patient costs and protocol compliance. Radiology, 1997. 204(3): p. 627-33.
15.       D'Angio, G.J., et al., Position paper: imaging methods for primary renal tumors of childhood: costs versus benefits. Med Pediatr Oncol, 1993. 21(3): p. 205-12.
16.       Buxton, M.J., National Wilms' Tumor Study: economic perspective. J Natl Cancer Inst Monogr, 1995(19): p. 27-9.
17.       McNeil, D.E., et al., Screening for Wilms tumor and hepatoblastoma in children with Beckwith-Wiedemann syndromes: a cost-effective model. Med Pediatr Oncol, 2001. 37(4): p. 349-56.
18.       Hartman, D.J. and G.T. MacLennan, Wilms tumor. J Urol, 2005. 173(6): p. 2147.
19.       Schmidt, D. and J.B. Beckwith, Histopathology of childhood renal tumors. Hematol Oncol Clin North Am, 1995. 9(6): p. 1179-200.
20.       Beckwith, J.B. and N.F. Palmer, Histopathology and prognosis of Wilms tumors: results from the First National Wilms' Tumor Study. Cancer, 1978. 41(5): p. 1937-48.
21.       Breslow, N., et al., Prognosis for Wilms' tumor patients with nonmetastatic disease at diagnosis--results of the second National Wilms' Tumor Study. J Clin Oncol, 1985. 3(4): p. 521-31.
22.       Faria, P., et al., Focal versus diffuse anaplasia in Wilms tumor--new definitions with prognostic significance: a report from the National Wilms Tumor Study Group. Am J Surg Pathol, 1996. 20(8): p. 909-20.
23.       Dome, J.S., et al., Treatment of anaplastic histology Wilms' tumor: results from the fifth National Wilms' Tumor Study. J Clin Oncol, 2006. 24(15): p. 2352-8.
24.       Beckwith, J.B., et al., Histological analysis of aggressiveness and responsiveness in Wilms' tumor. Med Pediatr Oncol, 1996. 27(5): p. 422-8.
25.       Graf, N., et al., Characteristics and outcome of stage II and III non-anaplastic Wilms' tumour treated according to the SIOP trial and study 93-01. European Journal of Cancer, 2012. 48(17): p. 3240-8.
26.       Zuppan, C.W., J.B. Beckwith, and D.W. Luckey, Anaplasia in unilateral Wilms' tumor: a report from the National Wilms' Tumor Study Pathology Center. Hum Pathol, 1988. 19(10): p. 1199-209.
27.       Beckwith, J.B., N.B. Kiviat, and J.F. Bonadio, Nephrogenic rests, nephroblastomatosis, and the pathogenesis of Wilms' tumor. Pediatr Pathol, 1990. 10(1-2): p. 1-36.
28.       Beckwith, J.B., Precursor lesions of Wilms tumor: clinical and biological implications. Med Pediatr Oncol, 1993. 21(3): p. 158-68.
29.       Beckwith, J.B., Nephrogenic rests and the pathogenesis of Wilms tumor: developmental and clinical considerations. Am J Med Genet, 1998. 79(4): p. 268-73.
30.       Coppes, M.J., et al., Factors affecting the risk of contralateral Wilms tumor development: a report from the National Wilms Tumor Study Group. Cancer, 1999. 85(7): p. 1616-25.
31.       Perlman, E.J., et al., Hyperplastic perilobar nephroblastomatosis: long-term survival of 52 patients. Pediatr Blood Cancer, 2006. 46(2): p. 203-21.
32.       Riccardi, V.M., et al., Chromosomal imbalance in the Aniridia-Wilms' tumor association: 11p interstitial deletion. Pediatrics, 1978. 61(4): p. 604-10.
33.       Muto, R., et al., Prediction by FISH analysis of the occurrence of Wilms tumor in aniridia patients. Am J Med Genet, 2002. 108(4): p. 285-9.
34.       Kreidberg, J.A., et al., WT-1 is required for early kidney development. Cell, 1993. 74(4): p. 679-91.
35.       Scholz, H. and K.M. Kirschner, A role for the Wilms' tumor protein WT1 in organ development. Physiology (Bethesda), 2005. 20: p. 54-9.
36.       Pritchard-Jones, K., et al., The candidate Wilms' tumour gene is involved in genitourinary development. Nature, 1990. 346(6280): p. 194-7.
37.       Coppes, M.J., V. Huff, and J. Pelletier, Denys-Drash syndrome: relating a clinical disorder to genetic alterations in the tumor suppressor gene WT1. J Pediatr, 1993. 123(5): p. 673-8.
38.       Huff, V., Genotype/phenotype correlations in Wilms' tumor. Med Pediatr Oncol, 1996. 27(5): p. 408-14.
39.       Breslow, N.E., et al., Characteristics and outcomes of children with the Wilms tumor-Aniridia syndrome: a report from the National Wilms Tumor Study Group. J Clin Oncol, 2003. 21(24): p. 4579-85.
40.       Koufos, A., et al., Familial Wiedemann-Beckwith syndrome and a second Wilms tumor locus both map to 11p15.5. Am J Hum Genet, 1989. 44(5): p. 711-9.
41.       Shuman, C., et al., Constitutional UPD for chromosome 11p15 in individuals with isolated hemihyperplasia is associated with high tumor risk and occurs following assisted reproductive technologies. Am J Med Genet A, 2006. 140(14): p. 1497-503.
42.       Ravenel, J.D., et al., Loss of imprinting of insulin-like growth factor-II (IGF2) gene in distinguishing specific biologic subtypes of Wilms tumor. J Natl Cancer Inst, 2001. 93(22): p. 1698-703.
43.       Maw, M.A., et al., A third Wilms' tumor locus on chromosome 16q. Cancer Res, 1992. 52(11): p. 3094-8.
44.       Grundy, P., M.J. Coppes, and D. Haber, Molecular genetics of Wilms tumor. Hematol Oncol Clin North Am, 1995. 9(6): p. 1201-15.
45.       Grundy, P.E., et al., Loss of heterozygosity for chromosomes 16q and 1p in Wilms' tumors predicts an adverse outcome. Cancer Res, 1994. 54(9): p. 2331-3.
46.       Wittmann, S., et al., Loss of 11q and 16q in Wilms tumors is associated with anaplasia, tumor recurrence, and poor prognosis. Genes Chromosomes Cancer, 2007. 46(2): p. 163-70.
47.       Williams, R.D., et al., Molecular profiling reveals frequent gain of MYCN and anaplasia-specific loss of 4q and 14q in Wilms tumor. Genes Chromosomes Cancer, 2011. 50(12): p. 982-95.
48.       Huang, C.C., et al., Predicting relapse in favorable histology Wilms tumor using gene expression analysis: a report from the Renal Tumor Committee of the Children's Oncology Group. Clin Cancer Res, 2009. 15(5): p. 1770-8.
49.       Rivera, M.N., et al., An X chromosome gene, WTX, is commonly inactivated in Wilms tumor. Science, 2007. 315(5812): p. 642-5.
50.       Royer-Pokora, B., et al., Clinical relevance of mutations in the Wilms tumor suppressor 1 gene WT1 and the cadherin-associated protein beta1 gene CTNNB1 for patients with Wilms tumors: results of long-term surveillance of 71 patients from International Society of Pediatric Oncology Study 9/Society for Pediatric Oncology. Cancer, 2008. 113(5): p. 1080-9.
51.       Ruteshouser, E.C., S.M. Robinson, and V. Huff, Wilms tumor genetics: mutations in WT1, WTX, and CTNNB1 account for only about one-third of tumors. Genes Chromosomes Cancer, 2008. 47(6): p. 461-70.
52.       Rahman, N., et al., Evidence for a familial Wilms' tumour gene (FWT1) on chromosome 17q12-q21. Nat Genet, 1996. 13(4): p. 461-3.
53.       McDonald, J.M., et al., Linkage of familial Wilms' tumor predisposition to chromosome 19 and a two-locus model for the etiology of familial tumors. Cancer Res, 1998. 58(7): p. 1387-90.
54.       Ohshima, J., et al., Methylation of the RASSF1A promoter is predictive of poor outcome among patients with Wilms tumor. Pediatric blood & cancer, 2012. 59(3): p. 499-505.
55.       Grundy, P.E., et al., Loss of heterozygosity for chromosomes 1p and 16q is an adverse prognostic factor in favorable-histology Wilms tumor: a report from the National Wilms Tumor Study Group. J Clin Oncol, 2005. 23(29): p. 7312-21.
56.       Ghanem, M.A., et al., The prognostic significance of apoptosis-associated proteins BCL-2, BAX and BCL-X in clinical nephroblastoma. Br J Cancer, 2001. 85(10): p. 1557-63.
57.       Ghanem, M.A., et al., Expression and prognostic value of Wilms' tumor 1 and early growth response 1 proteins in nephroblastoma. Clinical cancer research : an official journal of the American Association for Cancer Research, 2000. 6(11): p. 4265-71.
58.       Bielen, A., et al., Dependence of Wilms tumor cells on signaling through insulin-like growth factor 1 in an orthotopic xenograft model targetable by specific receptor inhibition. Proceedings of the National Academy of Sciences of the United States of America, 2012. 109(20): p. E1267-76.
59.       Franken, J., et al., p53 Immunohistochemistry expression in wilms tumor: a prognostic tool in the detection of tumor aggressiveness. The Journal of urology, 2013. 189(2): p. 664-70.
60.       Perlman, E.J., et al., WT1 mutation and 11P15 loss of heterozygosity predict relapse in very low-risk wilms tumors treated with surgery alone: a children's oncology group study. J Clin Oncol, 2011. 29(6): p. 698-703.
61.       Basta-Jovanovic, G., et al., Prognostic value of survivin expression in Wilms tumor. J BUON, 2012. 17(1): p. 168-73.
62.       Bardeesy, N., et al., Anaplastic Wilms' tumour, a subtype displaying poor prognosis, harbours p53 gene mutations. Nat Genet, 1994. 7(1): p. 91-7.
63.       Malkin, D., et al., Mutations of the p53 tumor suppressor gene occur infrequently in Wilms' tumor. Cancer Res, 1994. 54(8): p. 2077-9.
64.       Routh, J.C., et al., B7-H1 expression in Wilms tumor: correlation with tumor biology and disease recurrence. J Urol, 2008. 179(5): p. 1954-9; discussion 1959-60.
65.       Routh, J.C., et al., B7-h1 as a biomarker for therapy failure in patients with favorable histology wilms tumor. J Urol, 2013. 189(4): p. 1487-92.
66.       Vujanic, G.M., et al., Revised International Society of Paediatric Oncology (SIOP) working classification of renal tumors of childhood. Med Pediatr Oncol, 2002. 38(2): p. 79-82.
67.       Messahel, B., et al., Allele loss at 16q defines poorer prognosis Wilms tumour irrespective of treatment approach in the UKW1-3 clinical trials: a Children's Cancer and Leukaemia Group (CCLG) Study. Eur J Cancer, 2009. 45(5): p. 819-26.
68.       Breslow, N.E., et al., Wilms' tumor: prognostic factors for patients without metastases at diagnosis: results of the National Wilms' Tumor Study. Cancer, 1978. 41(4): p. 1577-89.
69.       Breslow, N., et al., Prognostic factors in nonmetastatic, favorable histology Wilms' tumor. Results of the Third National Wilms' Tumor Study. Cancer, 1991. 68(11): p. 2345-53.
70.       Shamberger, R.C., et al., Intravascular extension of Wilms tumor. Ann Surg, 2001. 234(1): p. 116-21.
71.       Jayabose, S., et al., Hypercalcemia in childhood renal tumors. Cancer, 1988. 61(4): p. 788-91.
72.       Coppes, M.J., et al., Acquired von Willebrand disease in Wilms' tumor patients. J Clin Oncol, 1992. 10(3): p. 422-7.
73.       Ritchey, M.L., et al., Surgical complications after nephrectomy for Wilms' tumor. Surg Gynecol Obstet, 1992. 175(6): p. 507-14.
74.       Ritchey, M.L., et al., Fate of bilateral renal lesions missed on preoperative imaging: a report from the National Wilms Tumor Study Group. J Urol, 2005. 174(4 Pt 2): p. 1519-21; discussion 1521.
75.       Ritchey, M.L., et al., Accuracy of current imaging modalities in the diagnosis of synchronous bilateral Wilms' tumor. A report from the National Wilms Tumor Study Group. Cancer, 1995. 75(2): p. 600-4.
76.       Meisel, J.A., et al., Significance and management of computed tomography detected pulmonary nodules: a report from the National Wilms Tumor Study Group. Int J Radiat Oncol Biol Phys, 1999. 44(3): p. 579-85.
77.       Owens, C.M., et al., Role of chest computed tomography at diagnosis in the management of Wilms' tumor: a study by the United Kingdom Children's Cancer Study Group. J Clin Oncol, 2002. 20(12): p. 2768-73.
78.       Hall, G., et al., Predictors of surgical outcome in Wilms' tumor: a single-institution comparative experience. J Pediatr Surg, 2006. 41(5): p. 966-71.
79.       Saarinen, U.M., et al., Percutaneous needle biopsy preceding preoperative chemotherapy in the management of massive renal tumors in children. J Clin Oncol, 1991. 9(3): p. 406-15.
80.       McLorie, G.A., et al., Reduction in tumor burden allowing partial nephrectomy following preoperative chemotherapy in biopsy proved Wilms tumor. J Urol, 1991. 146(2 ( Pt 2)): p. 509-13.
81.       Mitchell, C., et al., Immediate nephrectomy versus preoperative chemotherapy in the management of non-metastatic Wilms' tumour: results of a randomised trial (UKW3) by the UK Children's Cancer Study Group. Eur J Cancer, 2006. 42(15): p. 2554-62.
82.       Shamberger, R.C., et al., Bilateral Wilms' tumors with progressive or nonresponsive disease. J Pediatr Surg, 2006. 41(4): p. 652-7; discussion 652-7.
83.       Shamberger, R.C., et al., Surgery-related factors and local recurrence of Wilms tumor in National Wilms Tumor Study 4. Ann Surg, 1999. 229(2): p. 292-7.
84.       Fuchs, J., et al., Surgical aspects in the treatment of patients with unilateral wilms tumor: a report from the SIOP 93-01/German Society of Pediatric Oncology and Hematology. Ann Surg, 2009. 249(4): p. 666-71.
85.       Ritchey, M.L., et al., Management and outcome of inoperable Wilms tumor. A report of National Wilms Tumor Study-3. Ann Surg, 1994. 220(5): p. 683-90.
86.       Ritchey, M.L., et al., Preoperative therapy for intracaval and atrial extension of Wilms tumor. Cancer, 1993. 71(12): p. 4104-10.
87.       Szavay, P., et al., Surgery of cavoatrial tumor thrombus in nephroblastoma: a report of the SIOP/GPOH study. Pediatr Blood Cancer, 2004. 43(1): p. 40-5.
88.       Duarte, R.J., et al., Laparoscopic nephrectomy for wilms tumor after chemotherapy: initial experience. J Urol, 2004. 172(4 Pt 1): p. 1438-40.
89.       Piche, N. and D. Barrieras, Minimally invasive nephron-sparing surgery for unilateral Wilms tumor. J Pediatr Surg, 2012. 47(7): p. E1-4.
90.       Javid, P.J., et al., Laparoscopic nephroureterectomy for Wilms' tumor: oncologic considerations. J Pediatr Surg, 2011. 46(5): p. 978-82.
91.       Miller, D.C., et al., Renal and cardiovascular morbidity after partial or radical nephrectomy. Cancer, 2008. 112(3): p. 511-20.
92.       Cozzi, F., et al., Renal function adaptation in children with unilateral renal tumors treated with nephron sparing surgery or nephrectomy. J Urol, 2005. 174(4 Pt 1): p. 1404-8.
93.       Breslow, N.E., et al., Renal failure in the Denys-Drash and Wilms' tumor-aniridia syndromes. Cancer Res, 2000. 60(15): p. 4030-2.
94.       Cost, N.G., et al., Pathological review of Wilms tumor nephrectomy specimens and potential implications for nephron sparing surgery in Wilms tumor. Journal of Urology, 2012. 188(4 Suppl): p. 1506-10.
95.       Cost, N.G., et al., Oncologic outcomes of partial versus radical nephrectomy for unilateral Wilms tumor. Pediatric Blood & Cancer, 2012. 58(6): p. 898-904.
96.       Sulkowski, J., T. Kolon, and P. Mattei, Nephron-sparing partial nephrectomy for bilateral Wilms' tumor. Journal of Pediatric Surgery, 2012. 47(6): p. 1234-8.
97.       Coppes, M.J., et al., Bilateral Wilms' tumor: long-term survival and some epidemiological features. J Clin Oncol, 1989. 7(3): p. 310-5.
98.       Ritchey, M.L., et al., Renal failure in Wilms' tumor patients: a report from the National Wilms' Tumor Study Group. Med Pediatr Oncol, 1996. 26(2): p. 75-80.
99.       Breslow, N.E., et al., End stage renal disease in patients with Wilms tumor: results from the National Wilms Tumor Study Group and the United States Renal Data System. J Urol, 2005. 174(5): p. 1972-5.
100.     Ritchey, M.L. and M.J. Coppes, The management of synchronous bilateral Wilms tumor. Hematol Oncol Clin North Am, 1995. 9(6): p. 1303-15.
101.     Hamilton, T.E., et al., The management of synchronous bilateral Wilms tumor: a report from the National Wilms Tumor Study Group. Ann Surg, 2011. 253(5): p. 1004-10.
102.     Horwitz, J.R., et al., Renal salvage procedures in patients with synchronous bilateral Wilms' tumors: a report from the National Wilms' Tumor Study Group. J Pediatr Surg, 1996. 31(8): p. 1020-5.
103.     Weirich, A., et al., Clinical impact of histologic subtypes in localized non-anaplastic nephroblastoma treated according to the trial and study SIOP-9/GPOH. Ann Oncol, 2001. 12(3): p. 311-9.
104.     Cozzi, F., et al., Enucleative surgery for stage I nephroblastoma with a normal contralateral kidney. J Urol, 1996. 156(5): p. 1788-91; discussion 1791-3.
105.     Ritchey, M., Renal sparing surgery for children with bilateral Wilms tumor. Cancer, 2008. 112(9): p. 1877-8.
106.     Davidoff, A.M., et al., The feasibility and outcome of nephron-sparing surgery for children with bilateral Wilms tumor. The St Jude Children's Research Hospital experience: 1999-2006. Cancer, 2008. 112(9): p. 2060-70.
107.     Termuhlen, A.M., et al., Twenty-five year follow-up of childhood Wilms tumor: a report from the Childhood Cancer Survivor Study. Pediatr Blood Cancer, 2011. 57(7): p. 1210-6.
108.     Li, F.P., et al., Second neoplasms after Wilms' tumor in childhood. J Natl Cancer Inst, 1983. 71(6): p. 1205-9.
109.     Taylor, A.J., et al., Second primary neoplasms in survivors of Wilms' tumour--a population-based cohort study from the British Childhood Cancer Survivor Study. Int J Cancer, 2008. 122(9): p. 2085-93.
110.     Breslow, N.E., et al., Second malignant neoplasms in survivors of Wilms' tumor: a report from the National Wilms' Tumor Study. J Natl Cancer Inst, 1988. 80(8): p. 592-5.
111.     Breslow, N.E., et al., Second malignant neoplasms following treatment for Wilm's tumor: a report from the National Wilms' Tumor Study Group. J Clin Oncol, 1995. 13(8): p. 1851-9.
112.     Green, D.M., et al., Congestive heart failure after treatment for Wilms' tumor: a report from the National Wilms' Tumor Study group. J Clin Oncol, 2001. 19(7): p. 1926-34.
113.     Sorensen, K., et al., Cardiac function in Wilms' tumor survivors. J Clin Oncol, 1995. 13(7): p. 1546-56.
114.     Lange, J., et al., Risk factors for end stage renal disease in non-WT1-syndromic Wilms tumor. J Urol, 2011. 186(2): p. 378-86.
115.     Montgomery, B.T., et al., Extended followup of bilateral Wilms tumor: results of the National Wilms Tumor Study. J Urol, 1991. 146(2 ( Pt 2)): p. 514-8.
116.     Evans, A.E., et al., Late effects of treatment for Wilms' tumor. A report from the National Wilms' Tumor Study Group. Cancer, 1991. 67(2): p. 331-6.
117.     Hogeboom, C.J., et al., Stature loss following treatment for Wilms tumor. Med Pediatr Oncol, 2001. 36(2): p. 295-304.
118.     Makipernaa, A., et al., Spinal deformity induced by radiotherapy for solid tumours in childhood: a long-term follow up study. Eur J Pediatr, 1993. 152(3): p. 197-200.
119.     Green, D.M., et al., Pregnancy outcome after treatment for Wilms tumor: a report from the National Wilms Tumor Study Group. J Clin Oncol, 2002. 20(10): p. 2506-13.
120.     Li, F.P., et al., Outcome of pregnancy in survivors of Wilms' tumor. JAMA, 1987. 257(2): p. 216-9.
121.     Kalapurakal, J.A., et al., Pregnancy outcomes after abdominal irradiation that included or excluded the pelvis in childhood Wilms tumor survivors: a report from the National Wilms Tumor Study. Int J Radiat Oncol Biol Phys, 2004. 58(5): p. 1364-8.
122.     Abu-Ghosh, A.M., et al., Ifosfamide, carboplatin and etoposide in children with poor-risk relapsed Wilms' tumor: a Children's Cancer Group report. Ann Oncol, 2002. 13(3): p. 460-9.
123.     Dome, J.S., et al., Improved survival for patients with recurrent Wilms tumor: the experience at St. Jude Children's Research Hospital. J Pediatr Hematol Oncol, 2002. 24(3): p. 192-8.
124.     Grundy, P., et al., Prognostic factors for children with recurrent Wilms' tumor: results from the Second and Third National Wilms' Tumor Study. J Clin Oncol, 1989. 7(5): p. 638-47.
125.     Saylors, R.L., 3rd, et al., Phase I study of topotecan in combination with cyclophosphamide in pediatric patients with malignant solid tumors: a Pediatric Oncology Group Study. J Clin Oncol, 1998. 16(3): p. 945-52.
126.     Campbell, A.D., et al., Treatment of relapsed Wilms' tumor with high-dose therapy and autologous hematopoietic stem-cell rescue: the experience at Children's Memorial Hospital. J Clin Oncol, 2004. 22(14): p. 2885-90.
127.     Pein, F., et al., Etoposide and carboplatin: a highly effective combination in relapsed or refractory Wilms' tumor--a phase II study by the French Society of Pediatric Oncology. J Clin Oncol, 1994. 12(5): p. 931-6.
128.     Pein, F., et al., Etoposide in relapsed or refractory Wilms' tumor: a phase II study by the French Society of Pediatric Oncology and the United Kingdom Children's Cancer Study Group. J Clin Oncol, 1993. 11(8): p. 1478-81.
129.     Kung, F.H., et al., Ifosfamide/carboplatin/etoposide (ICE) for recurrent malignant solid tumors of childhood: a Pediatric Oncology Group Phase I/II study. J Pediatr Hematol Oncol, 1995. 17(3): p. 265-9.
130.     Glick, R.D., et al., Renal tumors in infants less than 6 months of age. J Pediatr Surg, 2004. 39(4): p. 522-5.
131.     van den Heuvel-Eibrink, M.M., et al., Characteristics and survival of 750 children diagnosed with a renal tumor in the first seven months of life: A collaborative study by the SIOP/GPOH/SFOP, NWTSG, and UKCCSG Wilms tumor study groups. Pediatr Blood Cancer, 2008. 50(6): p. 1130-4.
132.     Furtwaengler, R., et al., Mesoblastic nephroma--a report from the Gesellschaft fur Padiatrische Onkologie und Hamatologie (GPOH). Cancer, 2006. 106(10): p. 2275-83.
133.     Ganick, D.J., et al., Congenital cystic mesoblastic nephroma. Hum Pathol, 1981. 12(11): p. 1039-43.
134.     Beckwith, J.B. and D.A. Weeks, Congenital mesoblastic nephroma. When should we worry? Arch Pathol Lab Med, 1986. 110(2): p. 98-9.
135.     Weeks, D.A., et al., Renal neoplasms mimicking rhabdoid tumor of kidney. A report from the National Wilms' Tumor Study Pathology Center. Am J Surg Pathol, 1991. 15(11): p. 1042-54.
136.     Bayindir, P., et al., Cellular mesoblastic nephroma (infantile renal fibrosarcoma): institutional review of the clinical, diagnostic imaging, and pathologic features of a distinctive neoplasm of infancy. Pediatr Radiol, 2009. 39(10): p. 1066-74.
137.     Bolande, R.P., A.J. Brough, and R.J. Izant, Jr., Congenital mesoblastic nephroma of infancy. A report of eight cases and the relationship to Wilms' tumor. Pediatrics, 1967. 40(2): p. 272-8.
138.     Joshi, V.V., J. Kasznica, and T.R. Walters, Atypical mesoblastic nephroma. Pathologic characterization of a potentially aggressive variant of conventional congenital mesoblastic nephroma. Arch Pathol Lab Med, 1986. 110(2): p. 100-6.
139.     O'Malley, D.P., et al., Ultrastructure of cellular congenital mesoblastic nephroma. Ultrastruct Pathol, 1996. 20(5): p. 417-27.
140.     Argani, P., et al., Detection of the ETV6-NTRK3 chimeric RNA of infantile fibrosarcoma/cellular congenital mesoblastic nephroma in paraffin-embedded tissue: application to challenging pediatric renal stromal tumors. Mod Pathol, 2000. 13(1): p. 29-36.
141.     Knezevich, S.R., et al., ETV6-NTRK3 gene fusions and trisomy 11 establish a histogenetic link between mesoblastic nephroma and congenital fibrosarcoma. Cancer Res, 1998. 58(22): p. 5046-8.
142.     Kurkchubasche, A.G., et al., The role of preoperative chemotherapy in the treatment of infantile fibrosarcoma. J Pediatr Surg, 2000. 35(6): p. 880-3.
143.     Argani, P., et al., Clear cell sarcoma of the kidney: a review of 351 cases from the National Wilms Tumor Study Group Pathology Center. Am J Surg Pathol, 2000. 24(1): p. 4-18.
144.     Morgan, E. and J.M. Kidd, Undifferentiated sarcoma of the kidney: a tumor of childhood with histopathologic and clinical characteristics distinct from Wilms' tumor. Cancer, 1978. 42(4): p. 1916-21.
145.     Seibel, N.L., et al., Effect of duration of treatment on treatment outcome for patients with clear-cell sarcoma of the kidney: a report from the National Wilms' Tumor Study Group. J Clin Oncol, 2004. 22(3): p. 468-73.
146.     Weeks, D.A., et al., Rhabdoid tumor of kidney. A report of 111 cases from the National Wilms' Tumor Study Pathology Center. Am J Surg Pathol, 1989. 13(6): p. 439-58.
147.     Hoot, A.C., et al., Immunohistochemical analysis of hSNF5/INI1 distinguishes renal and extra-renal malignant rhabdoid tumors from other pediatric soft tissue tumors. Am J Surg Pathol, 2004. 28(11): p. 1485-91.
148.     Tomlinson, G.E., et al., Rhabdoid tumor of the kidney in the National Wilms' Tumor Study: age at diagnosis as a prognostic factor. J Clin Oncol, 2005. 23(30): p. 7641-5.
149.     Reinhard, H., et al., Rhabdoid tumors in children: prognostic factors in 70 patients diagnosed in Germany. Oncol Rep, 2008. 19(3): p. 819-23.
150.     Sultan, I., et al., Age, stage, and radiotherapy, but not primary tumor site, affects the outcome of patients with malignant rhabdoid tumors. Pediatr Blood Cancer, 2010. 54(1): p. 35-40.
151.     Koga, Y., et al., Long-term survival after autologous peripheral blood stem cell transplantation in two patients with malignant rhabdoid tumor of the kidney. Pediatr Blood Cancer, 2009. 52(7): p. 888-90.
152.     Grabowski, J., et al., Renal tumors in the second decade of life: results from the California Cancer Registry. J Pediatr Surg, 2009. 44(6): p. 1148-51.
153.     Hartman, D.S., et al., Primary malignant renal tumors in the second decade of life: Wilms tumor versus renal cell carcinoma. J Urol, 1982. 127(5): p. 888-91.
154.     Chow, W.H., et al., Rising incidence of renal cell cancer in the United States. JAMA, 1999. 281(17): p. 1628-31.
155.     Pastore, G., et al., Malignant renal tumours incidence and survival in European children (1978-1997): report from the Automated Childhood Cancer Information System project. Eur J Cancer, 2006. 42(13): p. 2103-14.
156.     Silberstein, J., et al., Renal cell carcinoma in the pediatric population: Results from the California Cancer Registry. Pediatr Blood Cancer, 2009. 52(2): p. 237-41.
157.     Carcao, M.D., et al., Renal-cell carcinoma in children: a different disorder from its adult counterpart? Med Pediatr Oncol, 1998. 31(3): p. 153-8.
158.     Cook, A., et al., Pediatric renal cell carcinoma: single institution 25-year case series and initial experience with partial nephrectomy. J Urol, 2006. 175(4): p. 1456-60; discussion 1460.
159.     Estrada, C.R., et al., Renal cell carcinoma: Children's Hospital Boston experience. Urology, 2005. 66(6): p. 1296-300.
160.     Geller, J.I., et al., Translocation renal cell carcinoma: lack of negative impact due to lymph node spread. Cancer, 2008. 112(7): p. 1607-16.
161.     Geller, J.I. and J.S. Dome, Local lymph node involvement does not predict poor outcome in pediatric renal cell carcinoma. Cancer, 2004. 101(7): p. 1575-83.
162.     Selle, B., et al., Population-based study of renal cell carcinoma in children in Germany, 1980-2005: more frequently localized tumors and underlying disorders compared with adult counterparts. Cancer, 2006. 107(12): p. 2906-14.
163.     Tomlinson, G.E., et al., Cytogenetics of a renal cell carcinoma in a 17-month-old child. Evidence for Xp11.2 as a recurring breakpoint. Cancer Genet Cytogenet, 1991. 57(1): p. 11-7.
164.     Argani, P., et al., A distinctive pediatric renal neoplasm characterized by epithelioid morphology, basement membrane production, focal HMB45 immunoreactivity, and t(6;11)(p21.1;q12) chromosome translocation. Am J Pathol, 2001. 158(6): p. 2089-96.
165.     Argani, P. and M. Ladanyi, Distinctive neoplasms characterised by specific chromosomal translocations comprise a significant proportion of paediatric renal cell carcinomas. Pathology, 2003. 35(6): p. 492-8.
166.     Argani, P., et al., Translocation carcinomas of the kidney after chemotherapy in childhood. J Clin Oncol, 2006. 24(10): p. 1529-34.
167.     Argani, P., et al., Primary renal neoplasms with the ASPL-TFE3 gene fusion of alveolar soft part sarcoma: a distinctive tumor entity previously included among renal cell carcinomas of children and adolescents. Am J Pathol, 2001. 159(1): p. 179-92.
168.     Argani, P., et al., Xp11 translocation renal cell carcinoma in adults: expanded clinical, pathologic, and genetic spectrum. Am J Surg Pathol, 2007. 31(8): p. 1149-60.
169.     Rais-Bahrami, S., et al., Xp11 translocation renal cell carcinoma: delayed but massive and lethal metastases of a chemotherapy-associated secondary malignancy. Urology, 2007. 70(1): p. 178 e3-6.
170.     Argani, P., et al., Renal carcinomas with the t(6;11)(p21;q12): clinicopathologic features and demonstration of the specific alpha-TFEB gene fusion by immunohistochemistry, RT-PCR, and DNA PCR. Am J Surg Pathol, 2005. 29(2): p. 230-40.
171.     Hedgepeth, R.C., M. Zhou, and J. Ross, Rapid development of metastatic Xp11 translocation renal cell carcinoma in a girl treated for neuroblastoma. J Pediatr Hematol Oncol, 2009. 31(8): p. 602-4.
172.     Leibovich, B.C., et al., Histological subtype is an independent predictor of outcome for patients with renal cell carcinoma. J Urol, 2010. 183(4): p. 1309-15.
173.     Poston, C.D., et al., Characterization of the renal pathology of a familial form of renal cell carcinoma associated with von Hippel-Lindau disease: clinical and molecular genetic implications. J Urol, 1995. 153(1): p. 22-6.
174.     Walther, M.M., et al., Prevalence of microscopic lesions in grossly normal renal parenchyma from patients with von Hippel-Lindau disease, sporadic renal cell carcinoma and no renal disease: clinical implications. J Urol, 1995. 154(6): p. 2010-4; discussion 2014-5.
175.     Al-Saleem, T., et al., Malignant tumors of the kidney, brain, and soft tissues in children and young adults with the tuberous sclerosis complex. Cancer, 1998. 83(10): p. 2208-16.
176.     Pavlovich, C.P., et al., Evaluation and management of renal tumors in the Birt-Hogg-Dube syndrome. J Urol, 2005. 173(5): p. 1482-6.
177.     Zbar, B., et al., Hereditary papillary renal cell carcinoma. J Urol, 1994. 151(3): p. 561-6.
178.     Launonen, V., et al., Inherited susceptibility to uterine leiomyomas and renal cell cancer. Proc Natl Acad Sci U S A, 2001. 98(6): p. 3387-92.
179.     Alrashdi, I., et al., Hereditary leiomyomatosis and renal cell carcinoma: very early diagnosis of renal cancer in a paediatric patient. Fam Cancer, 2010. In press.
180.     Parast, M.M., et al., A unique case of renal carcinoma with Xp11.2 translocations/ TFE3 gene fusions in a 3-year-old child, with coexistent von Hippel-Lindau gene mutation. Pediatr Dev Pathol, 2004. 7(4): p. 403-6.
181.     Lazarus, J. and C. Moolman, Renal cell carcinoma as second malignancy in patient with previous Wilms tumor. Urology, 2009. 74(3): p. 598-600.
182.     Geller, J.I. and J.S. Dome, Retroperitoneal lymph node dissection for pediatric renal cell carcinoma. Pediatr Blood Cancer, 2009. 52(3): p. 430.
183.     Indolfi, P., et al., Local lymph node involvement in pediatric renal cell carcinoma: a report from the Italian TREP project. Pediatr Blood Cancer, 2008. 51(4): p. 475-8.
184.     Davis, C.J., Jr., F.K. Mostofi, and I.A. Sesterhenn, Renal medullary carcinoma. The seventh sickle cell nephropathy. Am J Surg Pathol, 1995. 19(1): p. 1-11.
185.     Strouse, J.J., et al., Significant responses to platinum-based chemotherapy in renal medullary carcinoma. Pediatr Blood Cancer, 2005. 44(4): p. 407-11.
186.     Dimashkieh, H., J. Choe, and G. Mutema, Renal medullary carcinoma: a report of 2 cases and review of the literature. Arch Pathol Lab Med, 2003. 127(3): p. e135-8.
187.     Swartz, M.A., et al., Renal medullary carcinoma: clinical, pathologic, immunohistochemical, and genetic analysis with pathogenetic implications. Urology, 2002. 60(6): p. 1083-9.
188.     Yang, X.J., et al., Gene expression profiling of renal medullary carcinoma: potential clinical relevance. Cancer, 2004. 100(5): p. 976-85.
189.     Cheng, J.X., et al., Renal medullary carcinoma: rhabdoid features and the absence of INI1 expression as markers of aggressive behavior. Mod Pathol, 2008. 21(6): p. 647-52.
190.     Assad, L., et al., Cytologic features of renal medullary carcinoma. Cancer, 2005. 105(1): p. 28-34.
191.     Warren, K.E., V. Gidvani-Diaz, and B. Duval-Arnould, Renal medullary carcinoma in an adolescent with sickle cell trait. Pediatrics, 1999. 103(2): p. E22.
192.     Selby, D.M., et al., Renal medullary carcinoma: can early diagnosis lead to long-term survival? J Urol, 2000. 163(4): p. 1238.
193.     Coogan, C.L., et al., Renal medullary carcinoma in patients with sickle cell trait. Urology, 1998. 51(6): p. 1049-50.
194.     L'Hostis, H., et al., Renal angiomyolipoma: a clinicopathologic, immunohistochemical, and follow-up study of 46 cases. Am J Surg Pathol, 1999. 23(9): p. 1011-20.
195.     Nelson, C.P. and M.G. Sanda, Contemporary diagnosis and management of renal angiomyolipoma. J Urol, 2002. 168(4 Pt 1): p. 1315-25.
196.     Ewalt, D.H., et al., Renal lesion growth in children with tuberous sclerosis complex. J Urol, 1998. 160(1): p. 141-5.
197.     O'Callaghan, F.J., et al., An epidemiological study of renal pathology in tuberous sclerosis complex. BJU Int, 2004. 94(6): p. 853-7.
198.     Fine, S.W., et al., Angiomyolipoma with epithelial cysts (AMLEC): a distinct cystic variant of angiomyolipoma. Am J Surg Pathol, 2006. 30(5): p. 593-9.
199.     Kutikov, A., et al., Incidence of benign pathologic findings at partial nephrectomy for solitary renal mass presumed to be renal cell carcinoma on preoperative imaging. Urology, 2006. 68(4): p. 737-40.
200.     Faraji, H., B.N. Nguyen, and K.T. Mai, Renal epithelioid angiomyolipoma: a study of six cases and a meta-analytic study. Development of criteria for screening the entity with prognostic significance. Histopathology, 2009. 55(5): p. 525-34.
201.     Oesterling, J.E., et al., The management of renal angiomyolipoma. J Urol, 1986. 135(6): p. 1121-4.
202.     Mitchell, A.L., M.A. Parisi, and V.P. Sybert, Effects of pregnancy on the renal and pulmonary manifestations in women with tuberous sclerosis complex. Genet Med, 2003. 5(3): p. 154-60.
203.     Boorjian, S.A., et al., The role of partial nephrectomy for the management of sporadic renal angiomyolipoma. Urology, 2007. 70(6): p. 1064-8.
204.     Heidenreich, A., et al., Nephron-sparing surgery for renal angiomyolipoma. Eur Urol, 2002. 41(3): p. 267-73.
205.     Ramon, J., et al., Renal angiomyolipoma: long-term results following selective arterial embolization. Eur Urol, 2009. 55(5): p. 1155-61.
206.     Williams, J.M., et al., Embolization of renal angiomyolipomata in patients with tuberous sclerosis complex. Am J Kidney Dis, 2006. 47(1): p. 95-102.
207.     Argani, P., Metanephric neoplasms: the hyperdifferentiated, benign end of the Wilms tumor spectrum? Clin Lab Med, 2005. 25(2): p. 379-92.
208.     Renshaw, A.A., D.R. Freyer, and Y.A. Hammers, Metastatic metanephric adenoma in a child. Am J Surg Pathol, 2000. 24(4): p. 570-4.
209.     Davis, C.J., Jr., et al., Metanephric adenoma. Clinicopathological study of fifty patients. Am J Surg Pathol, 1995. 19(10): p. 1101-14.
210.     Jones, E.C., et al., Metanephric adenoma of the kidney. A clinicopathological, immunohistochemical, flow cytometric, cytogenetic, and electron microscopic study of seven cases. Am J Surg Pathol, 1995. 19(6): p. 615-26.
211.     Muir, T.E., J.C. Cheville, and D.J. Lager, Metanephric adenoma, nephrogenic rests, and Wilms' tumor: a histologic and immunophenotypic comparison. Am J Surg Pathol, 2001. 25(10): p. 1290-6.
212.     Kupeli, S., et al., Metanephric adenoma in a 6-year-old child with hemihypertrophy. J Pediatr Hematol Oncol, 2009. 31(6): p. 453-5.
213.     Yoshioka, K., et al., Production of erythropoietin and multiple cytokines by metanephric adenoma results in erythrocytosis. Pathol Int, 2007. 57(8): p. 529-36.
214.     Argani, P. and J.B. Beckwith, Metanephric stromal tumor: report of 31 cases of a distinctive pediatric renal neoplasm. Am J Surg Pathol, 2000. 24(7): p. 917-26.
215.     Bluebond-Langner, R., et al., Adult presentation of metanephric stromal tumor. J Urol, 2002. 168(4 Pt 1): p. 1482-3.
216.     Lorenzo, A.J., et al., Metanephric stromal tumor with urothelial extension. J Urol, 2003. 169(3): p. 1095-7.
217.     Arroyo, M.R., et al., The spectrum of metanephric adenofibroma and related lesions: clinicopathologic study of 25 cases from the National Wilms Tumor Study Group Pathology Center. Am J Surg Pathol, 2001. 25(4): p. 433-44.
218.     Hennigar, R.A. and J.B. Beckwith, Nephrogenic adenofibroma. A novel kidney tumor of young people. Am J Surg Pathol, 1992. 16(4): p. 325-34.
219.     Picken, M.M., et al., Metanephric adenosarcoma in a young adult: morphologic, immunophenotypic, ultrastructural, and fluorescence in situ hybridization analyses: a case report and review of the literature. Am J Surg Pathol, 2001. 25(11): p. 1451-7.
220.     Blakely, M.L., et al., Outcome of children with cystic partially differentiated nephroblastoma treated with or without chemotherapy. J Pediatr Surg, 2003. 38(6): p. 897-900.
221.     Luithle, T., et al., Treatment of cystic nephroma and cystic partially differentiated nephroblastoma--a report from the SIOP/GPOH study group. J Urol, 2007. 177(1): p. 294-6.
222.     Edmunds, W., Cystic adenoma of the kidney. Trans Pathol Soc (London), 1892. 43: p. 89-90.
223.     Eble, J.N. and S.M. Bonsib, Extensively cystic renal neoplasms: cystic nephroma, cystic partially differentiated nephroblastoma, multilocular cystic renal cell carcinoma, and cystic hamartoma of renal pelvis. Semin Diagn Pathol, 1998. 15(1): p. 2-20.
224.     Joshi, V.V. and J.B. Beckwith, Multilocular cyst of the kidney (cystic nephroma) and cystic, partially differentiated nephroblastoma. Terminology and criteria for diagnosis. Cancer, 1989. 64(2): p. 466-79.
225.     Turbiner, J., et al., Cystic nephroma and mixed epithelial and stromal tumor of kidney: a detailed clinicopathologic analysis of 34 cases and proposal for renal epithelial and stromal tumor (REST) as a unifying term. Am J Surg Pathol, 2007. 31(4): p. 489-500.
226.     Zhou, M., et al., Adult cystic nephroma and mixed epithelial and stromal tumor of the kidney are the same disease entity: molecular and histologic evidence. Am J Surg Pathol, 2009. 33(1): p. 72-80.
227.     Ferrer, F.A. and P.H. McKenna, Partial nephrectomy in a metachronous multilocular cyst of the kidney (cystic nephroma). J Urol, 1994. 151(5): p. 1358-60.
228.     Joshi, V.V. and J.B. Beckwith, Pathologic delineation of the papillonodular type of cystic partially differentiated nephroblastoma. A review of 11 cases. Cancer, 1990. 66(7): p. 1568-77.
229.     Boybeyi, O., et al., Cystic nephroma and localized renal cystic disease in children: diagnostic clues and management. J Pediatr Surg, 2008. 43(11): p. 1985-9.
230.     Ashley, R.A. and Y.E. Reinberg, Familial multilocular cystic nephroma: a variant of a unique renal neoplasm. Urology, 2007. 70(1): p. 179 e9-10.
231.     Lane, B.R., et al., Adult cystic nephroma and mixed epithelial and stromal tumor of the kidney: clinical, radiographic, and pathologic characteristics. Urology, 2008. 71(6): p. 1142-8.
232.     Halat, S., et al., Multilocular cystic renal cell carcinoma is a subtype of clear cell renal cell carcinoma. Mod Pathol, 2010. In press.
233.     Menon, P., et al., Multilocular cystic renal cell carcinoma in a child. J Pediatr Surg, 2004. 39(10): p. e14-6.
234.     Webster, W.S., et al., Surgical resection provides excellent outcomes for patients with cystic clear cell renal cell carcinoma. Urology, 2007. 70(5): p. 900-4; discussion 904.
235.     Sellaturay, S.V., et al., Primary intrarenal neuroblastoma: a rare, aggressive tumour of childhood mimicking Wilms' tumour. J Pediatr Urol, 2006. 2(5): p. 522-4.
236.     Serrano, R., et al., Intrarenal neuroblastoma diagnosed by fine-needle aspiration: a report of two cases. Diagn Cytopathol, 2002. 27(5): p. 294-7.
237.     Gerald, W.L. and J. Rosai, Case 2. Desmoplastic small cell tumor with divergent differentiation. Pediatr Pathol, 1989. 9(2): p. 177-83.
238.     Furman, J., et al., Urogenital involvement by desmoplastic small round-cell tumor. J Urol, 1997. 158(4): p. 1506-9.
239.     Wang, L.L., et al., Desmoplastic small round cell tumor of the kidney in childhood. Am J Surg Pathol, 2007. 31(4): p. 576-84.
240.     Barnoud, R., et al., Immunohistochemical expression of WT1 by desmoplastic small round cell tumor: a comparative study with other small round cell tumors. Am J Surg Pathol, 2000. 24(6): p. 830-6.
241.     Lae, M.E., et al., Desmoplastic small round cell tumor: a clinicopathologic, immunohistochemical, and molecular study of 32 tumors. Am J Surg Pathol, 2002. 26(7): p. 823-35.
242.     Sawyer, J.R., A.F. Tryka, and J.M. Lewis, A novel reciprocal chromosome translocation t(11;22)(p13;q12) in an intraabdominal desmoplastic small round-cell tumor. Am J Surg Pathol, 1992. 16(4): p. 411-6.
243.     Valera, E.T., et al., Pitfalls in the differential diagnosis of renal tumor in an adolescent. Pediatr Blood Cancer, 2010. 54(2): p. 319-21.
244.     Su, M.C., Y.M. Jeng, and Y.C. Chu, Desmoplastic small round cell tumor of the kidney. Am J Surg Pathol, 2004. 28(10): p. 1379-83.
245.     Eaton, S.H. and M.A. Cendron, Primary desmoplastic small round cell tumor of the kidney in a 7-year-old girl. J Pediatr Urol, 2006. 2(1): p. 52-4.
246.     Egloff, A.M., et al., Desmoplastic small round cell tumor of the kidney in a pediatric patient: sonographic and multiphase CT findings. AJR Am J Roentgenol, 2005. 185(5): p. 1347-9.
247.     Ellinger, J., et al., Primitive neuroectodermal tumor: rare, highly aggressive differential diagnosis in urologic malignancies. Urology, 2006. 68(2): p. 257-62.
248.     Ellison, D.A., et al., Immunohistochemistry of primary malignant neuroepithelial tumors of the kidney: a potential source of confusion? A study of 30 cases from the National Wilms Tumor Study Pathology Center. Hum Pathol, 2007. 38(2): p. 205-11.
249.     Karnes, R.J., et al., Primitive neuroectodermal tumor (extraskeletal Ewing's sarcoma) of the kidney with vena caval tumor thrombus. J Urol, 2000. 164(3 Pt 1): p. 772.
250.     Marley, E.F., et al., Primitive neuroectodermal tumor of the kidney--another enigma: a pathologic, immunohistochemical, and molecular diagnostic study. Am J Surg Pathol, 1997. 21(3): p. 354-9.
251.     Parham, D.M., et al., Primary malignant neuroepithelial tumors of the kidney: a clinicopathologic analysis of 146 adult and pediatric cases from the National Wilms' Tumor Study Group Pathology Center. Am J Surg Pathol, 2001. 25(2): p. 133-46.
252.     Rodriguez-Galindo, C., et al., Is primitive neuroectodermal tumor of the kidney a distinct entity? Cancer, 1997. 79(11): p. 2243-50.
253.     Delattre, O., et al., The Ewing family of tumors--a subgroup of small-round-cell tumors defined by specific chimeric transcripts. N Engl J Med, 1994. 331(5): p. 294-9.
254.     Vujanic, G.M., et al., Anaplastic sarcoma of the kidney: a clinicopathologic study of 20 cases of a new entity with polyphenotypic features. Am J Surg Pathol, 2007. 31(10): p. 1459-68.
255.     Labanaris, A., et al., Anaplastic sarcoma of the kidney. ScientificWorldJournal, 2009. 9: p. 97-101.
256.     Argani, P., et al., Primary renal synovial sarcoma: molecular and morphologic delineation of an entity previously included among embryonal sarcomas of the kidney. Am J Surg Pathol, 2000. 24(8): p. 1087-96.
257.     Mirza, M., I. Zamilpa, and J. Bunning, Primary renal synovial sarcoma. Urology, 2008. 72(3): p. 716 e11-2.
258.     Raney, B., et al., Primary renal sarcomas in the Intergroup Rhabdomyosarcoma Study Group (IRSG) experience, 1972-2005: A report from the Children's Oncology Group. Pediatr Blood Cancer, 2008. 51(3): p. 339-43.
259.     Dobkin, S.F., A.S. Brem, and A.A. Caldamone, Primary renal lymphoma. J Urol, 1991. 146(6): p. 1588-90.
260.     Jindal, B., et al., Bilateral primary renal lymphoma with orbital metastasis in a child. Pediatr Blood Cancer, 2009. 52(4): p. 539-41.
261.     Norton, K.I., L.B. Godine, and C. Lempert, Leiomyosarcoma of the kidney in an HIV-infected child. Pediatr Radiol, 1997. 27(6): p. 557-8.
262.     Itzchak, Y., et al., Liposarcoma of the renal capsule in a 7 year old girl. Pediatr Radiol, 1975. 3(3): p. 182-3.
263.     Cole, C.H., et al., Malignant fibrous histiocytoma in childhood. Cancer, 1993. 71(12): p. 4077-83.
264.     Ehara, H., et al., Clear cell melanoma of the renal pelvis presenting as a primary tumor. J Urol, 1997. 157(2): p. 634.

 

 

 

 

 

 

 

 

 

 

Custom Search