sábado, 18 de enero de 2020

Wilms Tumor and Other Childhood Kidney Tumors Treatment (PDQ®)–Health Professional Version - National Cancer Institute

Wilms Tumor and Other Childhood Kidney Tumors Treatment (PDQ®)–Health Professional Version - National Cancer Institute

National Cancer Institute

Wilms Tumor and Other Childhood Kidney Tumors Treatment (PDQ®)–Health Professional Version

Stage Information for Wilms Tumor

Both the results of the imaging studies and the surgical and pathologic findings at nephrectomy are used to determine the stage of disease. The stage is the same for tumors with FH or anaplastic histology. Thus, the stage information is characterized by a statement of both criteria (for example, stage II, FH or stage II, anaplastic histology).[161,173]
The staging system was originally developed by the NWTS Group and is still used by the COG. The staging system used in North America and incidence by stage are outlined below.[161]

Stage I

In stage I Wilms tumor (43% of patients), all of the following criteria must be met:
  • Tumor is limited to the kidney and is completely resected.
  • The renal capsule is intact.
  • The tumor is not ruptured or biopsied before being removed.
  • No involvement of renal sinus vessels.
  • No evidence of the tumor at or beyond the margins of resection.
  • All lymph nodes sampled are negative.
For a tumor to qualify for certain therapeutic protocols such as very low-risk stage I, regional lymph nodes must be examined microscopically. Lymph node sampling is strongly recommended for all patients, even in the absence of clinical abnormal nodes, to achieve the most accurate stage.

Stage II

In stage II Wilms tumor (20% of patients), the tumor is completely resected, and there is no evidence of tumor at or beyond the margins of resection. The tumor extends beyond the kidney as evidenced by any one of the following criteria:
  • There is regional extension of the tumor (i.e., penetration of the renal capsule, or extensive invasion of the soft tissue of the renal sinus, as discussed below).
  • Blood vessels in the nephrectomy specimen outside the renal parenchyma, including those of the renal sinus, contain tumor cells. Margins are clear.
  • Vascular extension of tumor is considered stage II only if it is completely removed en bloc in the nephrectomy specimen.
All lymph nodes sampled are negative.
Rupture or spillage confined to the flank, including biopsy of the tumor, is now included in stage III by the COG Renal Tumor Committee (COG RTC); however, data to support this approach are controversial.[140,174]

Stage III

In stage III Wilms tumor (21% of patients), there is postsurgical residual nonhematogenous tumor that is confined to the abdomen. Any one of the following may occur:
  • Lymph nodes in the abdomen or pelvis are involved by tumor. (Lymph node involvement in the thorax or other extra-abdominal sites is a criterion for stage IV.)
  • The tumor has penetrated through the peritoneal surface.
  • Tumor implants are found on the peritoneal surface.
  • Gross or microscopic tumor remains postoperatively (e.g., tumor cells are found at the margin of surgical resection on microscopic examination).
  • The tumor is not completely resectable because of local infiltration into vital structures.
  • Tumor rupture before surgery or any spill during surgery is considered stage III.
  • Any biopsy is performed, regardless of type—Tru-cut biopsy, open biopsy, or fine-needle aspiration—before the tumor is removed.
  • The tumor is removed in more than one piece (e.g., tumor cells are found in a separately excised adrenal gland; a tumor thrombus in the renal vein is removed separately from the nephrectomy specimen). Extension of the primary tumor in the vena cava into the thoracic vena cava and heart is considered stage III, rather than stage IV, even though outside the abdomen—and it can even be stage II if completely resected en bloc with the nephrectomy specimen.
Lymph node involvement and microscopic residual disease are reported as highly predictive of outcome in patients with stage III FH Wilms tumor.[175]

Stage IV

In stage IV Wilms tumor (11% of patients), one of the following is present:
  • Hematogenous metastases (lung, liver, bone, brain).
  • Lymph node metastases outside the abdominopelvic region.
The presence of tumor within the adrenal gland is not interpreted as metastasis and staging depends on all other staging parameters present. According to the criteria described above, the primary tumor is assigned a local stage, which determines local therapy. For example, a patient may have stage IV, local stage III disease.

Stage V

In stage V Wilms tumor (5% of patients), bilateral involvement by tumor is present at diagnosis. The current paradigm treats all patients with bilateral Wilms tumor the same for the first 6 or 12 weeks. After definitive surgery, the treatment is based on the highest stage of the remaining kidneys and the posttreatment pathology.[141]

Treatment of Wilms Tumor

Treatment option overview for Wilms tumor

Because of the relative rarity of Wilms tumor, all patients with this tumor should be considered for entry into a clinical trial. Treatment planning by a multidisciplinary team of cancer specialists (pediatric surgeon and/or pediatric urologist, pediatric radiation oncologist, and pediatric oncologist) who have experience treating children with Wilms tumor is necessary to determine and implement optimal treatment.
Most randomized clinical studies for treatment of children with Wilms tumor have been conducted by two large clinical groups (COG RTC and SIOP). Differences between the two groups affect staging and classification. There are two standard approaches to Wilms tumor treatment: the COG RTC uses immediate surgery for all unilateral tumors and the SIOP uses preoperative chemotherapy as the first step in treatment. Both groups use postoperative chemotherapy, except for selected cases who do not receive chemotherapy, and in advanced stages, radiation therapy is used in a risk-adapted approach.
  • COG RTC (includes the previous NWTS Group): The NWTS Group established standard treatment for Wilms tumor in North America, consisting of initial nephrectomy (when feasible) followed by chemotherapy and, in some patients, radiation therapy.[176-178] This approach allows for early and accurate histologic diagnosis, collection of biologic materials unaltered by therapy, and staging information, such as the presence of tumor spill or tumor involvement in lymph nodes, before chemotherapy is administered.
  • SIOP: SIOP is a European consortium whose trials provide preoperative chemotherapy before definitive resection for patients with renal tumors. This results in fewer tumor spills during surgery and lower postoperative stage.[179] When the histological features of Wilms tumors from patients who underwent immediate surgery were compared with the histological features of those who received preoperative chemotherapy, preoperative chemotherapy was shown to significantly alter the histology, with fewer blastemal and mixed histology types in the tumors. Additionally, there were fewer stage III tumors in the preoperative chemotherapy group.[180]
  • Both SIOP and COG treat infants younger than 6 months with a primary nephrectomy.[181]
This summary focuses on the NWTS (now COG RTC) results and studies.
The major treatment and study conclusions of NWTS-1, NWTS-2, NWTS-3, NWTS-4, and NWTS-5 are as follows:
  1. Routine, postoperative radiation therapy of the flank is not necessary for children with stage I tumors or stage II tumors with FH when postnephrectomy combination chemotherapy consisting of vincristine and dactinomycin is administered.[178]
  2. The prognosis for patients with stage III FH is best when treatment includes either (a) dactinomycin, vincristine, doxorubicin, and 10.8 Gy of radiation therapy to the flank; or (b) dactinomycin, vincristine, and 20 Gy of radiation therapy to the flank. Whole abdominal radiation is indicated for extensive intraperitoneal disease or widespread intraperitoneal tumor spill with possible boost to gross residual disease.[178]
  3. The addition of cyclophosphamide at the protocol dose (10 mg/kg/d for 3 days every 6 weeks) to the combination of vincristine, dactinomycin, and doxorubicin does not improve prognosis for patients with stage IV FH tumors.[178]
  4. A single dose of dactinomycin per course (stages I–II FH, stage I anaplastic histology, stage III FH, stages III–IV, or stages I–IV clear cell sarcoma of the kidney) is equivalent to the divided-dose courses, results in the same EFS, achieves greater dose intensity, and is associated with less toxicity and expense.[182]
  5. Eighteen weeks of therapy is adequate for patients with stage I and stage II FH, and stage III and IV patients can be treated with 6 months of therapy instead of 15 months.[148,176,182-184]
  6. Gain of 1q is associated with inferior survival in unilateral FH Wilms tumor. It is the single most powerful predictor of outcome, and in the presence of 1q gain, neither 1p nor 16q loss is significant. In the absence of 1q gain in unilateral FH Wilms tumor, 1p and/or 16q loss retain some prognostic significance and are associated with a higher risk of recurrence.[97,99]
Surgery
The following operative principles have also evolved from NWTS trials:
  1. The most important role for the surgeon is to ensure complete tumor removal without rupture and assess the extent of disease. Radical nephrectomy and lymph node sampling via a transabdominal or thoracoabdominal incision is the procedure of choice.[185] A flank incision is not performed because it provides limited exposure to the kidney.
    For patients with resectable tumors, preoperative biopsy or intraoperative biopsy is not performed because either would upstage the tumor in the current COG staging system.[185]
  2. Routine exploration of the contralateral kidney is not necessary if technically adequate imaging studies do not suggest a bilateral process. If the initial imaging studies suggest bilateral kidney involvement, treatment approaches should facilitate renal-sparing surgery.[136]
  3. About 2% of Wilms tumor cases have ureteral involvement. The presence of gross hematuria, nonfunctioning kidney, or hydronephrosis suggests the tumor may extend into the ureter, and cystoscopy is recommended. En bloc resection to avoid tumor spill is recommended.[186]
  4. The surgeon needs to be aware of the risk of intraoperative spill, especially in patients who have right-sided and large tumors, as noted in a review of cases of intraoperative spill among 1,131 patients registered on COG study AREN03B2 (NCT00898365).[187]
  5. Even if stage IV disease (e.g., pulmonary metastases) is evident on imaging, resection of the renal tumor should be considered. Treatment of local stage I or II Wilms tumor in the setting of distant metastasis does not require local radiation therapy.
Renal-sparing surgery remains controversial and is not recommended, except for children with the following:[188,189]; [190][Level of evidence: 3iiB]
  • A solitary kidney.
  • Predisposition to bilateral tumors. Some children who are predisposed to bilateral tumors and who have very small tumors detected by screening ultrasonography may be considered for renal-sparing surgery to preserve renal tissue.[188]
  • Horseshoe kidney. Wilms tumor arising in a horseshoe kidney is rare, and accurate preoperative diagnosis is important for planning the operative approach. Primary resection is possible in most cases. Inoperable cases can usually be resected after chemotherapy.[191]
  • Wilms tumor in infants with Denys-Drash or Frasier syndrome (to delay the need for dialysis).
Renal-sparing surgery does not appear to be feasible for most patients at the time of diagnosis because of the location of the tumor within the kidney, even in patients with very low-risk tumors.[192] In North America, renal-sparing surgery (partial nephrectomy) of unilateral Wilms tumor after administration of chemotherapy to shrink the tumor mass is considered investigational.[193,194]
Hilar and periaortic lymph node sampling is appropriate even if the nodes appear normal.[185,195] Furthermore, any suspicious node basin is sampled. Margins of resection, residual tumor, and any suspicious node basins are marked with titanium clips.
Wilms tumor rarely invades adjacent organs; therefore, resection of contiguous organs is seldom indicated. There is an increased incidence of complications occurring in more extensive resections that involve removal of additional organs beyond the diaphragm and adrenal gland. This finding has led to the recommendation in current COG protocols that patients in whom nephrectomy will require removal of additional organs should be considered for initial biopsy, neoadjuvant chemotherapy, and then secondary resection.[196] Primary resection of liver metastasis is not recommended.[197]
Chemotherapy
Preoperative chemotherapy before nephrectomy is indicated in the following situations, which have been listed previously under situations requiring a biopsy (refer to the Diagnostic and Staging Evaluation for Wilms Tumor section of this summary for more information):[185,196,198-201]
  • Wilms tumor in a solitary kidney.
  • Synchronous bilateral Wilms tumor.
  • Extension of tumor thrombus in the inferior vena cava above the level of the hepatic veins. About 4% of Wilms tumor patients present with inferior vena cava or atrial involvement, and 11% of patients present with renal vein involvement. Embolization of a caval thrombus to the pulmonary artery is rare but can be lethal, and the presence of a thrombus must be identified preoperatively to prevent this occurrence and guide treatment.[137,145]
  • Tumor involves contiguous structures whereby the only means of removing the kidney tumor requires removal of the other structures (e.g., spleen, pancreas, or colon but excluding the adrenal gland).
  • Inoperable Wilms tumor.
  • Pulmonary compromise resulting from extensive pulmonary metastases.
Preoperative chemotherapy follows a biopsy. The biopsy may be performed through a flank approach.[145,202-206] Adequate tissue is essential for accurate histological assessment and molecular studies. Preoperative chemotherapy includes doxorubicin in addition to vincristine and dactinomycin unless anaplastic histology is present; in such cases, chemotherapy then includes treatment with regimen I (refer to Table 2). The chemotherapy generally makes tumor removal easier by decreasing the size and vascular supply of the tumor; it may also reduce the frequency of surgical complications.[140,145,196,198,207,208]
In North America, the use of preoperative chemotherapy in patients with evidence of a contained preoperative rupture has been suggested to avoid intraoperative spill, but this is controversial.[209,210] The preoperative diagnosis of a contained retroperitoneal rupture on CT is difficult, even for experienced pediatric radiologists.[134]
Newborns and all infants younger than 12 months who will be treated with chemotherapy require a 50% reduction in chemotherapy dose compared with the dose given to older children.[211] Dosing for infants (younger than 12 months) will be calculated per kilogram of weight, not body surface area. This reduction diminishes the toxic effects reported in children in this age group enrolled in NWTS studies while maintaining an excellent overall outcome.[212]
Liver function tests in children with Wilms tumor are monitored closely during the early course of therapy because hepatic toxic effects (sinusoidal obstructive syndrome, previously called veno-occlusive disease) have been reported in these patients.[213,214] Dactinomycin or doxorubicin should not be administered during radiation therapy. Patients who develop renal failure while undergoing therapy can continue receiving chemotherapy with vincristine, dactinomycin, and doxorubicin. Vincristine and doxorubicin can be given at full doses; however, dactinomycin is associated with severe neutropenia. Reductions in dosing these agents may not be necessary, but accurate pharmacologic and pharmacokinetic studies are needed while the patient is receiving therapy.[215,216]
Augmentation of therapy improves EFS for patients with FH Wilms tumor and loss of heterozygosity of 1p/16q. In the AREN0532 (NCT00352534) and AREN0533 (NCT00379340) trials, patients with stage I and stage II FH Wilms tumor who were treated with the DD-4A regimen (dactinomycin, vincristine, and doxorubicin) demonstrated a 4-year EFS rate of 87.3%, compared with the 4-year EFS rate of 68.8% (P = .042) for stage I and stage II patients treated on the NWTS-5 trial. Patients with stage III and stage IV disease had a 4-year EFS rate of 90.2% when treated with regimen M, compared with a 61.3% 4-year EFS rate (P = .001) for stage III and stage IV patients treated on the NWTS-5 trial. Trends toward improved 4-year survival rates were seen in stage I and II patients and in stage III and IV patients.[217][Level of evidence: 3iiiDi]
Postoperative radiation therapy to the tumor bed is required when a biopsy is performed or in the setting of local tumor stage III. In a study of 1,488 patients with Wilms tumors who underwent surgery and radiation therapy, delay in starting radiation therapy after surgery of greater than 14 days was associated with an increased risk of mortality for patients with nonmetastatic Wilms tumor.[218][Level of evidence: 3iiiA]
Table 2 describes the accepted chemotherapy regimens used to treat Wilms tumor.
Table 2. Accepted Chemotherapy Regimens for Wilms Tumor
Regimen NameRegimen Description
Regimen EE-4A [99]Vincristine, dactinomycin × 18 weeks postnephrectomy
Regimen DD-4A [99]Vincristine, dactinomycin, doxorubicin × 24 weeks; baseline nephrectomy or biopsy with subsequent nephrectomy
Regimen I [151]Vincristine, doxorubicin, cyclophosphamide, etoposide × 24 weeks postnephrectomy
Regimen M [219]Vincristine, dactinomycin, doxorubicin, cyclophosphamide, and etoposide with subsequent radiation therapy
Radiation therapy
Radiation therapy is used to improve local control and treat sites of metastatic disease. Radiation therapy has historically been dependent on stage and histology, but more recently is also guided by the tumor molecular signature.[220]
  1. COG approach:
    Upfront surgery provides histologic confirmation and tumor extent, providing the rationale for adjuvant therapy, including radiation therapy. Besides histology, postoperative risk factors for worse local control include: (1) incomplete resection, (2) positive margins, and (3) nodal involvement. Radiation therapy is not used in patients with stage I or stage II FH Wilms tumor. For patients with FH Wilms tumor, flank or abdominal radiation therapy is used for treatment in stage III tumors. (Refer to Table 3 for more information.) In cases of unfavorable histology (focal or diffuse anaplasia), flank or abdominal radiation therapy is indicated for all patients.
    • Flank radiation therapy covers the tumor bed, involved nodal region, and entire adjacent vertebral bodies at 10.8 Gy in six fractions. The dose of radiation therapy was based on the results of the NWTS-3 study where there was no increase in abdominal relapse for stage III FH patients receiving 10 Gy versus 20 Gy with DD-4A chemotherapy.[221]
    • Whole-abdominal radiation therapy of 10.5 Gy in seven fractions is used to treat diffuse spill or peritoneal metastasis.
    • In the closed COG AREN0321 (NCT00335556) study, the radiation therapy dose to the tumor bed was 10.8 Gy in six fractions, with the exception of patients with stage III diffuse anaplasia, where the dose of 19.8 Gy in 11 fractions was used.
    • Results of the early NWTS studies (1 and 2) suggest that a radiation therapy delay of more than 10 days from surgery results in worse local control, particularly in unfavorable histology Wilms tumor.[222,223] However, no difference in local control was found if radiation therapy was delayed more than 10 days from surgery for patients with stages II to IV FH tumors treated on NWTS-3 or NWTS-4.[93] More recent data from the National Cancer Database confirms improved survival in patients with nonmetastatic Wilms tumor who received adjuvant radiation therapy less than or equal to 14 days postoperatively.[218]
    • Results from the NWTS-3 and NWTS-4 trials indicate that there is no survival benefit of whole-lung irradiation in the setting of lung metastases seen on CT scan only.[224] Current COG guidelines allow for whole-lung irradiation omission in cases of FH disease without extrapulmonary metastases, loss of heterozygosity at 1p and 16q, and complete response at 6 weeks after vincristine, dactinomycin, and doxorubicin.[220] When whole-lung irradiation is given, a dose of 12 Gy in eight fractions is indicated for children older than 18 months and 9 Gy in six fractions for patients younger than 18 months with pulmonary metastasis.
    • Other sites of metastatic disease in Wilms tumor are uncommon and may include liver, extra-abdominal nodes, brain, and bone. In the COG AREN0533 (NCT00379340) study, the radiation therapy dose recommendations for patients younger than 16 years are 19.8 Gy in 11 fractions to liver and gross residual nodes, 21.6 Gy in 17 fractions to the whole brain with a boost of 10.8 Gy in six fractions to gross metastatic disease in the brain, and 25.2 Gy in 14 fractions for bone metastasis. For patients older than 16 years, the radiation therapy dose to the whole brain and bone is increased to 30.6 Gy in 17 fractions.
Table 3. Radiation Therapy Guidelines in Children’s Oncology Group AREN0532, AREN0533, and AREN0321 Protocols
Local/Locoregional Disease
XRT = radiation therapy.
aRequires whole-abdominal XRT at 1.5 Gy daily fraction size. Patients with diffuse unresectable peritoneal implants receive 21 Gy.
bWhole-lung irradiation is given at 1.5 Gy daily fraction size.
cNot all patients receive radiation therapy.
dA boost is given for macroscopic disease.
 Stage IStage IIStage IIIStage III (diffuse spill, peritoneal metastasis, preoperative rupture)a
Favorable histologyNo XRTNo XRT10.8 Gy10.5 Gy
Focal anaplasia10.8 Gy10.8 Gy10.8 Gy10.5 Gy
Diffuse anaplasia10.8 Gy10.8 Gy19.8 Gy10.5 Gy + 9 Gy boost
 Metastatic Disease
 Stage IV LungStage IV LiverStage IV BrainStage IV Bone
Favorable histology10.5 Gy for age <12 monthsb,c; 12 Gy for age >12 monthsb,c19.8 Gy +/- 5.4 to 10.8 Gy boostd21.6 Gy + 10.8 Gy boost for age <16 years; 30.6 Gy for age >16 years25.2 Gy for age <16 years; 30.6 Gy for age >16 years
Focal or diffuse anaplasia10.5 Gy for age <12 monthsb; 12 Gy for age >12 monthsb19.8 Gy +/- 5.4 to 10.8 Gy boostd21.6 Gy + 10.8 Gy boost for age <16 years; 30.6 Gy for age >16 years25.2 Gy for age <16 years; 30.6 Gy for age >16 years

Treatment of stage I Wilms tumor

Table 4 provides an overview of the standard treatment options and survival data for patients with stage I Wilms tumor, based on published results.
Table 4. Overview of Standard Treatment Options for Stage I Wilms Tumora
Histology4-Year RFS or EFS4-Year OSTreatment (refer to Table 2 for chemotherapy regimen descriptions)
DA = diffuse anaplastic; EFS = event-free survival; FA = focal anaplastic; FH = favorable histology; LOH = loss of heterozygosity; OS = overall survival; RFS = relapse-free survival; XRT = radiation therapy.
aSource: Grundy et al.,[99] Shamberger et al.,[152] Fernandez et al.,[220] Dix et al.,[217] and Daw et al.[225]
bOne patient with a pulmonary relapse 4.12 years after diagnosis.
FH <24 mo/tumor weight <550g90%100%Surgery, including lymph node biopsy only
FH >24 mo/tumor weight >550g94% RFS98%Nephrectomy + lymph node sampling followed by regimen EE-4A
FH with LOH 1p/16q (n = 8)100% EFS100%Nephrectomy + lymph node sampling followed by regimen DD-4A
FA100%100% (n = 8)Nephrectomy + lymph node sampling followed by regimen DD-4A and XRT
DA100%b100% (n = 10)Nephrectomy + lymph node sampling followed by regimen DD-4A and XRT
The COG validated the hypothesis that nephrectomy only is appropriate therapy for patients younger than 2 years at diagnosis with stage I FH Wilms tumor that weighed less than 550 g in the AREN0532 (NCT00352534) trial. The NWTS-5 trial investigated this approach for children younger than 2 years at diagnosis with stage I FH Wilms tumor that weighed less than 550 g.
Evidence (surgery only for children younger than 2 years at diagnosis with stage I FH tumor that weighed <550 g):
  1. The AREN0532 (NCT00352534) trial was designed to confirm the findings from NWTS-5 that adjuvant chemotherapy could be omitted for children younger than 2 years at diagnosis with stage I FH Wilms tumor that weighed less than 550 g. A total of 116 patients met the criteria for very low-risk Wilms tumor and were enrolled on the study.[152,220,226]
    • Twelve patients relapsed.
    • The estimated 4-year EFS rate was 89.7%, and the OS rate was 100%.
    • 11p15 methylation status was associated with relapse (20% relapse with loss of heterozygosity, 25% relapse with loss of imprinting, and 3.3% relapse with retention of the normal imprinting [P = .011]).
    • Risk of developing metachronous Wilms tumor is very low in patients with very low-risk Wilms tumor who lack evidence of an underlying syndrome.
  2. The AREN0321 (NCT00335556) study demonstrated that outcomes for patients with stage I anaplastic Wilms tumor were improved with the addition of doxorubicin and flank radiation therapy to vincristine/dactinomycin therapy.[225]
    • Four-year EFS and OS rate estimates were 100% in AREN0321, compared with 70% and 81.5%, respectively, in an updated analysis of 27 patients from NWTS-5 (median follow up, 13.3 years). One patient with diffuse anaplasia relapsed 4.12 years after diagnosis on the AREN0321 trial.
    • The addition of doxorubicin and radiation therapy to AREN0321 was on the basis of the pattern of relapse observed in stage I anaplastic Wilms tumor in the abdomen and distant sites in the NWTS-5 trial.
    • Retrospective analysis of all patients with stage I anaplastic Wilms tumor treated on NWTS-1 through NWTS-5 and AREN0321 showed a significant improvement in EFS for patients treated with doxorubicin (4-year EFS rate, 97.2% vs. 77.5%; P = .01), but no difference in EFS according to flank radiation therapy was shown (4-year EFS rate, 91.7% vs. 80.2%; P = .15).
    • The rate of local recurrence was low (3.6%) and appeared to be similar for patients who received flank radiation therapy (4%) and patients who did not receive flank radiation therapy (6.2%). Local relapse occurred only in patients with diffuse anaplasia.
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.

Treatment of stage II Wilms tumor

Table 5 provides an overview of the standard treatment options and survival data for patients with stage II Wilms tumor, based on published results.
Table 5. Overview of Standard Treatment Options for Stage II Wilms Tumora
Histology4-Year RFS or EFS4-Year OSTreatment (refer to Table 2 for chemotherapy regimen descriptions)
DA = diffuse anaplastic; EFS = event-free survival; FA = focal anaplastic; FH = favorable histology; LOH = loss of heterozygosity; OS = overall survival; RFS = relapse-free survival; XRT = radiation therapy.
aSource: Grundy et al.,[99] Dome et al.,[151] and Dix et al.[217]
FH86% RFS98%Nephrectomy + lymph node sampling followed by regimen EE-4A
FH LOH 1p/16q (n = 24)83% EFS100%Nephrectomy + lymph node sampling followed by regimen DD-4A
FA80% EFS80% (n = 5)Nephrectomy + lymph node sampling followed by abdominal XRT and regimen DD-4A
DA83% EFS82%Nephrectomy + lymph node sampling followed by abdominal XRT and regimen I
On NWTS-3, NWTS-4, and NWTS-5, patients with intraoperative spill were divided into two groups: (1) those with diffuse spillage involving the whole abdominal cavity; and (2) those with local spillage confined to the flank. Patients with diffuse spillage were treated with radiation therapy to the entire abdomen and three-drug chemotherapy (vincristine, dactinomycin, and doxorubicin), whereas patients with local spillage were treated with vincristine and dactinomycin only. On the basis of an analysis of patients treated on NWTS-3 and NWTS-4 indicating that patients with stage II disease and local spillage had inferior OS compared with patients with stage II disease without local spillage, COG studies treat patients with local spillage with doxorubicin and flank radiation.[227] This approach is controversial and has not been tested; therefore, it should not be considered standard.
In a review of 499 patients from NWTS-4 with stage II FH Wilms tumor, 95 of the patients experienced tumor spill. The 8-year RFS and OS rates for patients who experienced intraoperative tumor spill and were treated with vincristine and dactinomycin without flank radiation therapy were lower, at 75.7% and 90.3%, than the 85% and 95.6% rates for those who did not experience tumor spill. None of these differences achieved statistical significance.[174]
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.

Treatment of stage III Wilms tumor

Table 6 provides an overview of the standard treatment options and survival data for patients with stage III Wilms tumor, based on published results.
Table 6. Overview of Standard Treatment Options for Stage III Wilms Tumora
Histology4-Year RFS or EFS4-Year OSTreatment (refer to Table 2 for chemotherapy regimen descriptions)
DA = diffuse anaplastic; EFS = event-free survival; FA = focal anaplastic; FH = favorable histology; LOH = loss of heterozygosity; OS = overall survival; RFS = relapse-free survival; XRT = radiation therapy.
aSource: Grundy et al.,[99] Dome et al.,[151] Fernandez et al.,[228] and Dix et al.[217]
FH (all patients)88% EFS97%Nephrectomy + lymph node sampling followed by abdominal XRT and regimen DD-4A
FH (without LOH of 1p and/or 16q) and positive lymph nodes85% EFS97%Nephrectomy + lymph node sampling followed by abdominal XRT and regimen DD-4A
FH (without LOH of 1p and/or 16q) and negative lymph nodes97% EFS99%Nephrectomy + lymph node sampling followed by abdominal XRT and regimen DD-4A
FH (with LOH of 1p and 16q) (n = 31)87% EFS94%Nephrectomy + lymph node sampling followed by abdominal XRT and regimen M
FA88% RFS100% (n = 8)Nephrectomy + lymph node sampling followed by abdominal XRT and regimen DD-4A
FA (preoperative treatment)71% RFS71% (n = 7)Preoperative treatment with regimen DD-4A followed by nephrectomy + lymph node sampling and abdominal XRT
DA46% EFS53% (n = 16)Preoperative treatment with regimen I followed by nephrectomy + lymph node sampling and abdominal XRT
DA65% EFS67%Immediate nephrectomy + lymph node sampling followed by abdominal XRT and regimen I
Loss of heterozygosity of 1p or 16q was shown to influence EFS but not OS in 588 patients with stage III FH Wilms tumor treated on the COG AREN0532 protocol. When combined, lymph node status and loss of heterozygosity status provided a strong predictor of excellent EFS and OS when both were absent, with a 4-year EFS rate of 97%, and an OS rate of 99%.[228][Level of evidence: 2Di] The outcome was poorer for patients having both positive lymph nodes and loss of heterozygosity of 1p or 16q, with a 4-year EFS rate of 74%. However, the 4-year OS rate was not influenced, at 92%.[228] On the basis of these results, therapy was augmented for patients with loss of heterozygosity of 1p/16q for patients enrolled on the AREN0533 trial. Patients with stage III and stage IV Wilms tumor with loss of heterozygosity were treated with regimen M. The 4-year EFS rate was 90.2%, and the OS rate was 96.1%, compared with a 4-year EFS rate of 61.3% (P = .001) and a 4-year OS rate of 86.0% (P = .087) for patients in the NWTS-5 trial. There was a suggestion of improvement in survival; however, the study was not powered to detect differences in survival.[217][Level of evidence: 3iiiDi]
Early initiation of radiation therapy is a critical component of multimodal therapy for patients with nonmetastatic Wilms tumor. In a review of 1,488 patients with Wilms tumor who underwent surgery and radiation therapy, a surgery-to-radiation therapy interval of greater than 14 days was associated with an increased risk of mortality (hazard ratio, 2.13; P = .013). This underscores the importance of initiating radiation therapy within 14 days of surgery, which is specified in Wilms tumor treatment protocols.[218][Level of evidence: 3iiiA]
For patients classified as stage III purely on the basis of local spill, refer to the Treatment of stage II Wilms tumor section of this summary.
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.

Treatment of stage IV Wilms tumor

Table 7 provides an overview of the standard treatment options and survival data for patients with stage IV Wilms tumor, based on published results.
Table 7. Overview of Standard Treatment Options for Stage IV Wilms Tumora
Histology4-Year RFS or EFS4-Year OSTreatment (refer to Table 2 for chemotherapy regimen descriptions)
CR = complete response; DA = diffuse anaplasia; EFS = event-free survival; FA = focal anaplasia; FH = favorable histology; LOH = loss of heterozygosity; OS = overall survival; RFS = relapse-free survival; XRT = radiation therapy.
aSource: Grundy et al.,[99] Dome et al.,[151] Dix et al.,[219] and Dix et al.[217]
bAbdominal XRT is planned according to local stage of renal tumor.
cPulmonary XRT is reserved for patients with chest x-ray/chest computed tomography evidence of pulmonary metastases.
dRefer to the AREN0533 (NCT00379340) study for more information.
FH76% RFS86%Nephrectomy + lymph node sampling, followed by abdominal XRT,b radiation to sites of metastases, bilateral pulmonary XRT,c and regimen DD-4A
FH (with isolated lung nodules)85% EFS96%Nephrectomy + lymph node sampling, followed by abdominal XRT,b +/- bilateral pulmonary XRT,c and regimen DD-4A or regimen Md
FH (with isolated lung nodules with CR to DD-4A)83% EFS94%Nephrectomy + lymph node sampling, followed by abdominal XRTb and regimen DD-4A
FH (with isolated lung nodules with incomplete response to DD-4A)92% EFS96%Nephrectomy + lymph node sampling, followed by abdominal XRTb and bilateral pulmonary XRTc and regimen M
FH (with LOH of 1p and/or 16q) (n = 20)95% EFS100%Nephrectomy + lymph node sampling, abdominal XRTb radiation to sites of metastasesb, and regimen M
FA61% EFS72% (n = 11)Nephrectomy + lymph node sampling, followed by abdominal XRT,b radiation to sites of metastases, bilateral pulmonary XRT,c and regimen DD-4A
DA33% EFS33% (n = 15)Immediate nephrectomy + lymph node sampling followed by abdominal XRT,b radiation to sites of metastases, whole-lung XRT,c and regimen I
DA (preoperative treatment)31% EFS44% (n = 13)Preoperative treatment with regimen I followed by nephrectomy + lymph node sampling, followed by abdominal XRT,b radiation to sites of metastases, and whole-lung XRTc
Stage IV disease is defined by the presence of hematogenous metastases to the lung, liver, bone, brain, or other sites, with the lung being the most common site. Historically, chest x-rays were used to detect pulmonary metastases. The introduction of CT created controversy because many patients had lung nodules detected by chest CT scans that were not seen on chest x-rays. Management of newly diagnosed patients with FH Wilms tumor who have lung nodules detected only by CT scans (with negative chest x-ray) has elicited controversy as to whether they need to be treated with additional intensive treatment that is accompanied by acute and late toxicities.
Evidence (treatment of pulmonary nodules detected by chest CT scan only):
  1. A retrospective review of 186 patients from NWTS-4 and NWTS-5 with CT-only–detected lung nodules reported on the use of doxorubicin, vincristine, and dactinomycin versus the use of two drugs.[229]
    • Patients who received doxorubicin, vincristine, and dactinomycin with or without lung irradiation had a 5-year EFS rate of 80% versus an EFS rate of 56% for patients receiving only two drugs (P = .004).
    • There was no difference in EFS according to whether the lung was irradiated.
    • There was no difference in the 5-year OS rate (87% vs. 86%).
Retrospective studies from Europe have examined the impact of omitting pulmonary radiation in patients with pulmonary metastases diagnosed by chest x-ray. European investigators omitted radiation from the treatment of most patients with Wilms tumor and pulmonary metastases as identified on chest x-ray who were treated on the SIOP-93-01 (NCT00003804) trial. The European approach to renal tumors differs from the approach used in North America. All patients who were shown to have a renal tumor by imaging underwent 9 weeks of prenephrectomy chemotherapy consisting of vincristine, dactinomycin, and doxorubicin.
Evidence (omission of pulmonary irradiation):
  1. In a retrospective SIOP study, 234 newly diagnosed patients with Wilms tumor presenting with pulmonary metastases were treated according to the response of the pulmonary metastases to the prenephrectomy chemotherapy.[230]
    1. Patients who were in complete remission (67%) after 6 weeks of therapy continued with the same chemotherapy and did not require radiation to their lungs.
      • The 5-year EFS rate was 77%, and the OS rate was 88%.
    2. Patients who had residual pulmonary metastases were evaluated for metastasectomy.
      • Thirty-seven patients (17%) obtained complete remission with surgery, and their outcome was similar to that of the group of patients who were treated with chemotherapy. Tumor viability in the resected pulmonary metastases was not a factor for omitting radiation therapy.
      • The 5-year EFS rate was 84%, and the OS rate was 92%.
    3. Patients with residual pulmonary metastases that were incompletely resected or inoperable received more aggressive chemotherapy consisting of ifosfamide/anthracycline alternating with carboplatin/etoposide for 9 weeks.
      • Patients showing a complete remission at that time were spared pulmonary radiation and continued with chemotherapy, whereas patients with residual pulmonary metastases continued with additional chemotherapy (to complete 34 weeks) and pulmonary irradiation. The 5-year OS rate was 48%, compared with the OS rates for patients who responded to chemotherapy alone (88%) and those who underwent metastasectomy (92%) (P < .001).
      • Patients with high-risk histologies, such as anaplastic Wilms tumor, were treated with more aggressive chemotherapy but had a poorer outcome, compared with that of patients with nonanaplastic histologies (5-year OS rate, 87% vs. 33%; P < .001).
  2. The COG AREN0533 (NCT00379340) study applied a new strategy for patients with FH Wilms tumor and isolated lung metastases to improve EFS while reducing exposure to lung irradiation on the basis of the European experience. Therapy was adjusted on the basis of lung nodule response and tumor-specific loss of heterozygosity at 1p and 16q.[219][Level of evidence: 3iiiDi]
    • Of the 292 patients enrolled in the study, 133 patients (42%) showed a complete lung nodule response after 6 weeks of DD-4A (vincristine, dactinomycin, doxorubicin) and continued receiving the same chemotherapy without lung radiation therapy. The 4-year EFS rate was 80%, and OS rate was 96%.
    • Patients who had an incomplete lung nodule response (n = 145) or loss of heterozygosity at 1p/16q (n = 18) received lung radiation therapy and four cycles of cyclophosphamide/etoposide in addition to the DD-4A drugs (regimen M). The 4-year EFS rate was 89%, and the OS rate was 95% for the incomplete lung nodule response group without loss of heterozygosity. Of the patients with pulmonary metastases only and loss of heterozygosity, the 4-year EFS and OS rates were 100%.
    • In a post hoc analysis of 1q gain in 212 patients enrolled in AREN0533 who had DNA available, patients with lung nodule complete remission with 1q gain had a significantly worse 4-year EFS rate (86% vs. 57%, P = .001) and trend toward inferior OS rates (97% vs. 89%). Relapses were predominantly pulmonary. There was no difference in outcome for patients with incomplete lung nodule response on the basis of 1q gain.
    • Regimen M has a higher potential for late effects (increased risk of secondary leukemias and risk of infertility related to a cumulative dose of cyclophosphamide of 8.8 g/m2).
    • COG showed that initial lung radiation therapy could be avoided in approximately 40% of patients. OS was excellent; however, there was a trend toward more events than expected (expected, 15% and observed, 20%; P = .052).
Although fewer patients were spared pulmonary radiation when treated in the COG trial than in the European trials, it is important to note several differences between the studies and why the studies cannot be directly compared.[219,230] Patients in Europe receive a more dose-dense regimen of dactinomycin and doxorubicin before their pulmonary metastases are reevaluated than do patients in North America (135 ug/kg dactinomycin and 100 mg/m2 doxorubicin in Europe, compared with 45 ug/kg dactinomycin and 45 mg/m2 of doxorubicin in North America). European studies allow lung radiation therapy to be omitted for patients with a complete remission achieved by chemotherapy or pulmonary metastasectomy, whereas radiation therapy was only omitted in the United States for patients with a complete remission with chemotherapy alone. Imaging studies were not centrally reviewed in the European studies, whereas they were in the United States, and the definition of complete remission may have been more stringent in the AREN0533 (NCT00379340) trial.
The presence of liver metastases at diagnosis is not an independent adverse prognostic factor in patients with stage IV Wilms tumor.[197]
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.

Treatment of stage V Wilms tumor and those predisposed to developing bilateral Wilms tumor

Currently, there is not a standard approach for the treatment of stage V Wilms tumor (bilateral Wilms tumor at diagnosis) and those predisposed to developing Wilms tumor; however, for the first time, a prospective study of the treatment of patients with bilateral Wilms tumor has been completed and provides guidance for the approach.[141]
Management of a child with bilateral Wilms tumor is very challenging. The goals of therapy are to eradicate all tumor and to preserve as much normal renal tissue as possible, with the hope of decreasing the risk of chronic renal failure among these children.[231]
Historically, based on the NWTS-4 and NWTS-5 trials and trials performed in Europe, patients with bilateral Wilms tumor have had a lower EFS and OS than have patients with localized Wilms tumor. The NWTS-4 study reported that the 8-year EFS rate for patients with bilateral FH Wilms tumor was 74%, and the OS rate was 89%; for patients with anaplastic histology, the EFS rate was 40%, and the OS rate was 45%.[164] The NWTS-5 study reported that the 4-year EFS rate for all bilateral Wilms tumor patients was 56%, and the OS rate was 81%; the 4-year EFS rates for patients with FH (65%), focal anaplastic histology (76%), and diffuse anaplastic histology (25%) were also reported.[99,151] Similar outcomes for patients with bilateral Wilms tumor have been reported in Europe.[163,232] In a single-institution experience in the Netherlands (N = 41), there was significant morbidity in terms of renal failure (32%) and secondary tumors (20%).[232] The incidence of end-stage renal failure in the Dutch study may be a reflection of a longer follow-up period.
Treatment options for stage V Wilms tumor include the following:
Preoperative chemotherapy and resection for bilateral Wilms tumor
For patients with bilateral Wilms tumor, the goal of therapy is to preserve as much renal tissue as possible without compromising overall outcome. This approach is used to avoid the late effect of end-stage renal disease, which can be caused by underlying germline genetic aberrations and treatment-related loss of functional renal tissue. End-stage renal disease occurs more frequently in patients with bilateral Wilms tumor (12% nonsyndromic) than in patients with unilateral Wilms tumor (<1%). Functional renal outcome is considerably better after bilateral nephron-sparing surgery than after other types of surgery.[141]
Traditionally, patients have undergone bilateral renal biopsies, with staging of each kidney followed by preoperative chemotherapy. In the first prospective multi-institutional treatment trial (COG AREN0534 [NCT00945009]), pretreatment biopsies were not required if results of imaging tests were consistent with Wilms tumor.[141] This approach was taken because the bilateral occurrence of non-Wilms renal tumors is very low. Also, core-needle and wedge biopsies are not highly successful in identifying anaplasia in Wilms tumor.[142] In the setting of an unusual clinical situation, such as age older than 10 years or atypical imaging features, when a diagnosis other than Wilms should be considered, a tissue diagnosis is obtained.[141]
For patients who are treated with preoperative chemotherapy, the tumor pathology needs to be evaluated after 4 to 8 weeks. For patients not treated in a clinical trial, the ideal time to perform a biopsy or resection is unknown because minimal shrinkage may reflect chemotherapy-induced differentiation or anaplastic histology. A planned attempt at resection or biopsy of apparently unresectable tumor is undertaken no later than 12 weeks from diagnosis. Continuing therapy without evaluating tumor pathology in a patient with bilateral Wilms tumor may miss anaplastic histology or chemotherapy-induced differentiation (including rhabdomyomatous differentiation) and thus increase toxicity for the patient without providing additional benefit for tumor control. Anaplastic histology occurs in 10% of patients with bilateral Wilms tumor, and these tumors respond poorly to chemotherapy.[164]
Once the diagnosis is confirmed, a complete resection is performed. Histologic confirmation of the diagnosis is not straightforward. In a series of 27 patients from NWTS-4, discordant pathology (unilateral anaplastic tumor) was seen in 20 cases (74%), which highlights the need to obtain tissue from both kidneys. Seven children who were later diagnosed with diffuse anaplastic tumors had core biopsies performed to establish the diagnosis; however, anaplasia was not found. Anaplasia was identified in only three of the nine patients when an open-wedge biopsy was performed and in seven of nine patients who had a partial or complete nephrectomy.[164]
The decision to administer chemotherapy and/or radiation therapy after biopsy or a second-look operation is dependent on the tumor's response to initial therapy. More aggressive therapy is required for patients with inadequate response to initial therapy observed at the second procedure or in the setting of anaplasia.[173,233,234]
End-stage renal disease is the most clinically significant morbidity in patients with bilateral Wilms tumor and can be caused by underlying germline genetic aberrations, as well as treatment-related loss of functional renal tissue. Long-term monitoring of renal function is required after treatment for bilateral disease.
Evidence (preoperative chemotherapy and resection for bilateral Wilms tumor):
  1. The first prospective study in bilateral Wilms tumor (AREN0534 [NCT00945009]) aimed to improve EFS and OS while preserving renal tissue by intensifying preoperative chemotherapy (utilizing three drugs—vincristine, dactinomycin, and doxorubicin), completing definitive surgery by 12 weeks from diagnosis, and modifying postoperative chemotherapy on the basis of histologic response.[141]
    • For the arm that treated children with bilateral Wilms tumor, results showed that central review of imaging, surgical resection within 12 weeks of diagnosis, and response-based and histology-based postoperative therapy improved EFS and OS, when compared with the historical outcomes of children with bilateral Wilms tumor.
    • For the 189 patients with bilateral Wilms tumor, 4-year EFS rate was 82.1% (95% confidence interval [CI], 73.5%–90.8%), and the OS rate was 94.9% (95% CI, 90.1%–99.7%). Because biopsy was not performed before treatment in this series, some of the patients enrolled may have had only nephrogenic rests and not a true Wilms tumor. This finding may have improved these survival figures over historical controls.
    • One of the aims of the study was that 75% of patients undergo definitive surgery by 12 weeks. After induction chemotherapy, 163 of 189 patients (84%) underwent definitive surgical treatment in at least one kidney by 12 weeks, and 39% of patients retained parts of both kidneys.
    • Chemotherapy after surgery was tailored according to histologic response. The 4-year EFS rate was 84.1% for FH tumors, 58.2% for anaplastic histology tumors, and 82% for blastemal-type tumors.
    • Because of the higher risk of renal failure in patients with bilateral Wilms tumor than in patients with unilateral Wilms tumor, one of the goals of the study was that 50% of the patients undergo bilateral nephron-sparing surgery. This threshold was not met, with only 39% of patients successfully treated with bilateral nephron-sparing surgery.
    • Based on the above study, the recommendation was to continue with three-drug preoperative chemotherapy for 6 to 12 weeks followed by nephron-sparing surgery whenever possible. After resection, postoperative therapy is based on the histology of the resected specimen. The disappointing use of nephron-sparing surgery in this study may have been because of the level of experience of the surgeons in this multi-institutional study.
  2. In a retrospective review of 93 children with bilateral Wilms tumor registered at Associazione Italiana di Ematologia e Oncologia Pediatrica (AIEOP) centers over a 21-year period, 43 patients were treated with vincristine and dactinomycin preoperatively and 37 patients were treated with vincristine, dactinomycin, and doxorubicin. The duration of preoperative chemotherapy ranged from 1 week to 40 weeks (median, 12 weeks).[163]
    • The 4-year DFS rate was 67%, and the OS rate was 80%.
    • There was a trend toward better EFS in nonmetastatic patients receiving vincristine, dactinomycin, and doxorubicin preoperatively (4-year EFS rate, 84%) than for patients receiving vincristine and dactinomycin (4-year EFS rate, 65%), but this was not significant.
    • The bilateral renal parenchyma was preserved in 48% of patients.
  3. In a retrospective review of 49 patients with Wilms tumor who received preoperative therapy according to the SIOP-93-01 (NCT00003804) guidelines, the timing of surgery was determined when there was no longer imaging evidence of tumor regression. The mean treatment duration was 80 days before renal-sparing surgery.[235]
    • The 5-year EFS rate was 83.4%, and the OS rate was 89.5%.
    • All but one of the patients had renal-sparing surgery in at least one kidney.
    • Despite the good survival, 14% of the patients developed end-stage renal disease.
  4. In a retrospective review from St. Jude Children's Research Hospital, investigators described their experience with preoperative chemotherapy followed by renal-sparing procedures in children with bilateral FH Wilms tumor.[236]
    • In a series, 39 of 42 patients with bilateral FH Wilms tumor underwent successful bilateral renal-sparing procedures after receiving preoperative chemotherapy. Three patients underwent unilateral nephrectomy with contralateral nephron-sparing surgery. Three patients required early (within 4 months) repeat nephron-sparing surgery for residual tumor. In the long term, seven patients had local tumor recurrence, and three patients had intestinal obstruction.
    • The OS rate was 86% (mean follow-up, 4.1 years). Of the six patients who died, five had diffuse anaplastic histology.
    • All of the patients had an estimated glomerular filtration rate of more than 60 mL/min/1.73m2 at the last follow-up; none of the patients developed end-stage renal disease.
    • The authors concluded that bilateral renal-sparing surgery is almost always feasible and can be done safely with good oncologic outcomes in patients with synchronous, bilateral Wilms tumor. It should be considered even if preoperative imaging studies suggest that the lesions are unresectable. Sparing of renal parenchyma is likely to help preserve renal function in children who are at significant risk of chronic renal insufficiency. Careful long-term follow-up is required to fully assess the potential progression of renal dysfunction.
    • A follow-up review of these patients revealed the following: 8 of 36 patients underwent repeat nephron-sparing surgery, and an additional two patients required a third nephron-sparing surgery. Six of these patients were alive without disease at the 4.5-year follow-up. The two patients who died had blastemal-predominant histology.[237]
Renal transplant
Renal transplant for children with stage V Wilms tumor is usually delayed until 1 to 2 years have passed without evidence of malignancy because most relapses occur within 2 years of diagnosis.[238] Similarly, renal transplant for children with Denys-Drash syndrome and Wilms tumor, all of whom require bilateral nephrectomy, is generally delayed 1 to 2 years after completion of initial treatment.[238]
Treatment options under clinical evaluation
Information about National Cancer Institute (NCI)–supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, refer to the ClinicalTrials.gov website.
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
(Refer to the Treatment of Recurrent Childhood Kidney Tumors section of this summary for information about recurrent disease.)

Follow-up after treatment

For patients who have completed therapy for Wilms tumor and exhibit features consistent with genetic predisposition, such as bilateral Wilms tumor, screening involves renal ultrasonography examination every 3 months for metachronous tumors during the risk period for that particular syndrome (5 years for WT1-related syndromes; 8 years for Beckwith-Wiedemann syndrome).

Late effects after Wilms tumor therapy

Children treated for Wilms tumor are at increased risk of developing the following:
  • Premature mortality after Wilms tumor diagnosis. In 1,441 5-year survivors of Wilms tumor, a substantial increase in cumulative mortality from 5.4% to 22.7% was noted from 30 to 50 years after Wilms tumor diagnosis. Excess deaths after 30 years were attributed to subsequent malignant neoplasms (50%) and cardiac-related causes (25%).[239] Radiation therapy was a risk factor for both outcomes.
  • Subsequent malignant neoplasms.[239-241] Digestive cancers and breast cancer are the most frequent subsequent neoplasms, and radiation therapy is a risk factor. Women treated with lower doses of radiation to large volumes of breast tissue for a childhood cancer have a risk of breast cancer that is higher than previously recognized. The cumulative incidence of invasive breast cancer in Wilms tumor survivors who had received pulmonary radiation for metastatic Wilms tumor is nearly 15% by age 40 years.[242]
  • Congestive heart failure. The risk of congestive heart failure is influenced by dose of doxorubicin received, radiation to the heart, and female sex.[241,243]
  • Complications of pregnancy.[244]
  • End-stage renal disease. The cumulative incidence of end-stage renal disease caused by chronic renal failure at 20 years from diagnosis of Wilms tumor is low, at 3.1% for patients with bilateral Wilms tumor and less than 1% for those with unilateral Wilms tumor.[81] Efforts, therefore, have been aimed toward reducing the intensity of therapy when possible.
(Refer to the PDQ summary on Late Effects of Treatment for Childhood Cancer for a full discussion of the late effects of cancer treatment in children and adolescents.)
References
  1. Howlader N, Noone AM, Krapcho M, et al.: SEER Cancer Statistics Review (CSR) 1975-2016. Bethesda, Md: National Cancer Institute, 2019. Available online. Last accessed November 04, 2019.
  2. Breslow N, Olshan A, Beckwith JB, et al.: Epidemiology of Wilms tumor. Med Pediatr Oncol 21 (3): 172-81, 1993. [PUBMED Abstract]
  3. Horner MJ, Ries LA, Krapcho M, et al.: SEER Cancer Statistics Review, 1975-2006. Bethesda, Md: National Cancer Institute, 2009. Also available online. Last accessed June 04, 2019.
  4. Scott RH, Stiller CA, Walker L, et al.: Syndromes and constitutional chromosomal abnormalities associated with Wilms tumour. J Med Genet 43 (9): 705-15, 2006. [PUBMED Abstract]
  5. Narod SA, Hawkins MM, Robertson CM, et al.: Congenital anomalies and childhood cancer in Great Britain. Am J Hum Genet 60 (3): 474-85, 1997. [PUBMED Abstract]
  6. Vujanić GM, Apps JR, Moroz V, et al.: Nephrogenic rests in Wilms tumors treated with preoperative chemotherapy: The UK SIOP Wilms Tumor 2001 Trial experience. Pediatr Blood Cancer 64 (11): , 2017. [PUBMED Abstract]
  7. Dumoucel S, Gauthier-Villars M, Stoppa-Lyonnet D, et al.: Malformations, genetic abnormalities, and Wilms tumor. Pediatr Blood Cancer 61 (1): 140-4, 2014. [PUBMED Abstract]
  8. Gracia Bouthelier R, Lapunzina P: Follow-up and risk of tumors in overgrowth syndromes. J Pediatr Endocrinol Metab 18 (Suppl 1): 1227-35, 2005. [PUBMED Abstract]
  9. Lapunzina P: Risk of tumorigenesis in overgrowth syndromes: a comprehensive review. Am J Med Genet C Semin Med Genet 137 (1): 53-71, 2005. [PUBMED Abstract]
  10. Treger TD, Chowdhury T, Pritchard-Jones K, et al.: The genetic changes of Wilms tumour. Nat Rev Nephrol 15 (4): 240-251, 2019. [PUBMED Abstract]
  11. Clericuzio CL: Clinical phenotypes and Wilms tumor. Med Pediatr Oncol 21 (3): 182-7, 1993. [PUBMED Abstract]
  12. Fischbach BV, Trout KL, Lewis J, et al.: WAGR syndrome: a clinical review of 54 cases. Pediatrics 116 (4): 984-8, 2005. [PUBMED Abstract]
  13. Breslow NE, Norris R, Norkool PA, 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 21 (24): 4579-85, 2003. [PUBMED Abstract]
  14. Barbosa AS, Hadjiathanasiou CG, Theodoridis C, et al.: The same mutation affecting the splicing of WT1 gene is present on Frasier syndrome patients with or without Wilms' tumor. Hum Mutat 13 (2): 146-53, 1999. [PUBMED Abstract]
  15. Koziell AB, Grundy R, Barratt TM, et al.: Evidence for the genetic heterogeneity of nephropathic phenotypes associated with Denys-Drash and Frasier syndromes. Am J Hum Genet 64 (6): 1778-81, 1999. [PUBMED Abstract]
  16. Royer-Pokora B, Beier M, Henzler M, et al.: Twenty-four new cases of WT1 germline mutations and review of the literature: genotype/phenotype correlations for Wilms tumor development. Am J Med Genet A 127 (3): 249-57, 2004. [PUBMED Abstract]
  17. Pelletier J, Bruening W, Kashtan CE, et al.: Germline mutations in the Wilms' tumor suppressor gene are associated with abnormal urogenital development in Denys-Drash syndrome. Cell 67 (2): 437-47, 1991. [PUBMED Abstract]
  18. Mueller RF: The Denys-Drash syndrome. J Med Genet 31 (6): 471-7, 1994. [PUBMED Abstract]
  19. Barbaux S, Niaudet P, Gubler MC, et al.: Donor splice-site mutations in WT1 are responsible for Frasier syndrome. Nat Genet 17 (4): 467-70, 1997. [PUBMED Abstract]
  20. Porteus MH, Narkool P, Neuberg D, et al.: Characteristics and outcome of children with Beckwith-Wiedemann syndrome and Wilms' tumor: a report from the National Wilms Tumor Study Group. J Clin Oncol 18 (10): 2026-31, 2000. [PUBMED Abstract]
  21. Rump P, Zeegers MP, van Essen AJ: Tumor risk in Beckwith-Wiedemann syndrome: A review and meta-analysis. Am J Med Genet A 136 (1): 95-104, 2005. [PUBMED Abstract]
  22. Choufani S, Shuman C, Weksberg R: Molecular findings in Beckwith-Wiedemann syndrome. Am J Med Genet C Semin Med Genet 163C (2): 131-40, 2013. [PUBMED Abstract]
  23. Eggermann T, Algar E, Lapunzina P, et al.: Clinical utility gene card for: Beckwith-Wiedemann Syndrome. Eur J Hum Genet 22 (3): , 2014. [PUBMED Abstract]
  24. Ibrahim A, Kirby G, Hardy C, et al.: Methylation analysis and diagnostics of Beckwith-Wiedemann syndrome in 1,000 subjects. Clin Epigenetics 6 (1): 11, 2014. [PUBMED Abstract]
  25. Brioude F, Lacoste A, Netchine I, et al.: Beckwith-Wiedemann syndrome: growth pattern and tumor risk according to molecular mechanism, and guidelines for tumor surveillance. Horm Res Paediatr 80 (6): 457-65, 2013. [PUBMED Abstract]
  26. Mussa A, Russo S, Larizza L, et al.: (Epi)genotype-phenotype correlations in Beckwith-Wiedemann syndrome: a paradigm for genomic medicine. Clin Genet 89 (4): 403-415, 2016. [PUBMED Abstract]
  27. Green DM, Breslow NE, Beckwith JB, et al.: Screening of children with hemihypertrophy, aniridia, and Beckwith-Wiedemann syndrome in patients with Wilms tumor: a report from the National Wilms Tumor Study. Med Pediatr Oncol 21 (3): 188-92, 1993. [PUBMED Abstract]
  28. DeBaun MR, Siegel MJ, Choyke PL: Nephromegaly in infancy and early childhood: a risk factor for Wilms tumor in Beckwith-Wiedemann syndrome. J Pediatr 132 (3 Pt 1): 401-4, 1998. [PUBMED Abstract]
  29. DeBaun MR, Tucker MA: Risk of cancer during the first four years of life in children from The Beckwith-Wiedemann Syndrome Registry. J Pediatr 132 (3 Pt 1): 398-400, 1998. [PUBMED Abstract]
  30. Milani D, Pezzani L, Tabano S, et al.: Beckwith-Wiedemann and IMAGe syndromes: two very different diseases caused by mutations on the same gene. Appl Clin Genet 7: 169-75, 2014. [PUBMED Abstract]
  31. Morris MR, Astuti D, Maher ER: Perlman syndrome: overgrowth, Wilms tumor predisposition and DIS3L2. Am J Med Genet C Semin Med Genet 163C (2): 106-13, 2013. [PUBMED Abstract]
  32. Astuti D, Morris MR, Cooper WN, et al.: Germline mutations in DIS3L2 cause the Perlman syndrome of overgrowth and Wilms tumor susceptibility. Nat Genet 44 (3): 277-84, 2012. [PUBMED Abstract]
  33. Golabi M, Leung A, Lopez C: Simpson-Golabi-Behmel Syndrome Type 1. In: Pagon RA, Adam MP, Bird TD, et al., eds.: GeneReviews. Seattle, Wash: University of Washington, 1993-2018, pp. Available online. Last accessed April 17, 2019.
  34. Peterman CM, Fevurly RD, Alomari AI, et al.: Sonographic screening for Wilms tumor in children with CLOVES syndrome. Pediatr Blood Cancer 64 (12): , 2017. [PUBMED Abstract]
  35. Fagali C, Kok F, Nicola P, et al.: MLPA analysis in 30 Sotos syndrome patients revealed one total NSD1 deletion and two partial deletions not previously reported. Eur J Med Genet 52 (5): 333-6, 2009 Sep-Oct. [PUBMED Abstract]
  36. Isidor B, Bourdeaut F, Lafon D, et al.: Wilms' tumor in patients with 9q22.3 microdeletion syndrome suggests a role for PTCH1 in nephroblastomas. Eur J Hum Genet 21 (7): 784-7, 2013. [PUBMED Abstract]
  37. Cairney AE, Andrews M, Greenberg M, et al.: Wilms tumor in three patients with Bloom syndrome. J Pediatr 111 (3): 414-6, 1987. [PUBMED Abstract]
  38. Hartley AL, Birch JM, Tricker K, et al.: Wilms' tumor in the Li-Fraumeni cancer family syndrome. Cancer Genet Cytogenet 67 (2): 133-5, 1993. [PUBMED Abstract]
  39. Bourdeaut F, Guiochon-Mantel A, Fabre M, et al.: Alagille syndrome and nephroblastoma: Unusual coincidence of two rare disorders. Pediatr Blood Cancer 50 (4): 908-11, 2008. [PUBMED Abstract]
  40. Russell B, Johnston JJ, Biesecker LG, et al.: Clinical management of patients with ASXL1 mutations and Bohring-Opitz syndrome, emphasizing the need for Wilms tumor surveillance. Am J Med Genet A 167A (9): 2122-31, 2015. [PUBMED Abstract]
  41. Bonaïti-Pellié C, Chompret A, Tournade MF, et al.: Genetics and epidemiology of Wilms' tumor: the French Wilms' tumor study. Med Pediatr Oncol 20 (4): 284-91, 1992. [PUBMED Abstract]
  42. Winther JF, Sankila R, Boice JD, et al.: Cancer in siblings of children with cancer in the Nordic countries: a population-based cohort study. Lancet 358 (9283): 711-7, 2001. [PUBMED Abstract]
  43. Breslow NE, Olson J, Moksness J, et al.: Familial Wilms' tumor: a descriptive study. Med Pediatr Oncol 27 (5): 398-403, 1996. [PUBMED Abstract]
  44. Li FP, Williams WR, Gimbrere K, et al.: Heritable fraction of unilateral Wilms tumor. Pediatrics 81 (1): 147-9, 1988. [PUBMED Abstract]
  45. Ruteshouser EC, Huff V: Familial Wilms tumor. Am J Med Genet C Semin Med Genet 129 (1): 29-34, 2004. [PUBMED Abstract]
  46. Paulino AC, Thakkar B, Henderson WG: Metachronous bilateral Wilms' tumor: the importance of time interval to the development of a second tumor. Cancer 82 (2): 415-20, 1998. [PUBMED Abstract]
  47. Coppes MJ, Arnold M, Beckwith JB, et al.: Factors affecting the risk of contralateral Wilms tumor development: a report from the National Wilms Tumor Study Group. Cancer 85 (7): 1616-25, 1999. [PUBMED Abstract]
  48. Grundy P, Koufos A, Morgan K, et al.: Familial predisposition to Wilms' tumour does not map to the short arm of chromosome 11. Nature 336 (6197): 374-6, 1988. [PUBMED Abstract]
  49. Little SE, Hanks SP, King-Underwood L, et al.: Frequency and heritability of WT1 mutations in nonsyndromic Wilms' tumor patients: a UK Children's Cancer Study Group Study. J Clin Oncol 22 (20): 4140-6, 2004. [PUBMED Abstract]
  50. Hanks S, Perdeaux ER, Seal S, et al.: Germline mutations in the PAF1 complex gene CTR9 predispose to Wilms tumour. Nat Commun 5: 4398, 2014. [PUBMED Abstract]
  51. Scott RH, Douglas J, Baskcomb L, et al.: Constitutional 11p15 abnormalities, including heritable imprinting center mutations, cause nonsyndromic Wilms tumor. Nat Genet 40 (11): 1329-34, 2008. [PUBMED Abstract]
  52. Grønskov K, Olsen JH, Sand A, et al.: Population-based risk estimates of Wilms tumor in sporadic aniridia. A comprehensive mutation screening procedure of PAX6 identifies 80% of mutations in aniridia. Hum Genet 109 (1): 11-8, 2001. [PUBMED Abstract]
  53. Clericuzio C, Hingorani M, Crolla JA, et al.: Clinical utility gene card for: WAGR syndrome. Eur J Hum Genet 19 (4): , 2011. [PUBMED Abstract]
  54. Hoyme HE, Seaver LH, Jones KL, et al.: Isolated hemihyperplasia (hemihypertrophy): report of a prospective multicenter study of the incidence of neoplasia and review. Am J Med Genet 79 (4): 274-8, 1998. [PUBMED Abstract]
  55. Shanske AL: Trisomy 18 in a second 20-year-old woman. Am J Med Genet A 140 (9): 966-7, 2006. [PUBMED Abstract]
  56. Reid S, Renwick A, Seal S, et al.: Biallelic BRCA2 mutations are associated with multiple malignancies in childhood including familial Wilms tumour. J Med Genet 42 (2): 147-51, 2005. [PUBMED Abstract]
  57. Hirsch B, Shimamura A, Moreau L, et al.: Association of biallelic BRCA2/FANCD1 mutations with spontaneous chromosomal instability and solid tumors of childhood. Blood 103 (7): 2554-9, 2004. [PUBMED Abstract]
  58. Reid S, Schindler D, Hanenberg H, et al.: Biallelic mutations in PALB2 cause Fanconi anemia subtype FA-N and predispose to childhood cancer. Nat Genet 39 (2): 162-4, 2007. [PUBMED Abstract]
  59. Gadd S, Huff V, Walz AL, et al.: A Children's Oncology Group and TARGET initiative exploring the genetic landscape of Wilms tumor. Nat Genet 49 (10): 1487-1494, 2017. [PUBMED Abstract]
  60. Wegert J, Wittmann S, Leuschner I, et al.: WTX inactivation is a frequent, but late event in Wilms tumors without apparent clinical impact. Genes Chromosomes Cancer 48 (12): 1102-11, 2009. [PUBMED Abstract]
  61. Ruteshouser EC, Robinson SM, Huff V: Wilms tumor genetics: mutations in WT1, WTX, and CTNNB1 account for only about one-third of tumors. Genes Chromosomes Cancer 47 (6): 461-70, 2008. [PUBMED Abstract]
  62. Walz AL, Ooms A, Gadd S, et al.: Recurrent DGCR8, DROSHA, and SIX homeodomain mutations in favorable histology Wilms tumors. Cancer Cell 27 (2): 286-97, 2015. [PUBMED Abstract]
  63. Wegert J, Ishaque N, Vardapour R, et al.: Mutations in the SIX1/2 pathway and the DROSHA/DGCR8 miRNA microprocessor complex underlie high-risk blastemal type Wilms tumors. Cancer Cell 27 (2): 298-311, 2015. [PUBMED Abstract]
  64. Rakheja D, Chen KS, Liu Y, et al.: Somatic mutations in DROSHA and DICER1 impair microRNA biogenesis through distinct mechanisms in Wilms tumours. Nat Commun 2: 4802, 2014. [PUBMED Abstract]
  65. Torrezan GT, Ferreira EN, Nakahata AM, et al.: Recurrent somatic mutation in DROSHA induces microRNA profile changes in Wilms tumour. Nat Commun 5: 4039, 2014. [PUBMED Abstract]
  66. Dome JS, Huff V: Wilms Tumor Predisposition. In: Pagon RA, Adam MP, Bird TD, et al., eds.: GeneReviews. Seattle, Wash: University of Washington, 1993-2018, pp. Available online. Last accessed October 25, 2019.
  67. Mahamdallie SS, Hanks S, Karlin KL, et al.: Mutations in the transcriptional repressor REST predispose to Wilms tumor. Nat Genet 47 (12): 1471-4, 2015. [PUBMED Abstract]
  68. Huff V: Wilms tumor genetics. Am J Med Genet 79 (4): 260-7, 1998. [PUBMED Abstract]
  69. Scott RH, Murray A, Baskcomb L, et al.: Stratification of Wilms tumor by genetic and epigenetic analysis. Oncotarget 3 (3): 327-35, 2012. [PUBMED Abstract]
  70. Corbin M, de Reyniès A, Rickman DS, et al.: WNT/beta-catenin pathway activation in Wilms tumors: a unifying mechanism with multiple entries? Genes Chromosomes Cancer 48 (9): 816-27, 2009. [PUBMED Abstract]
  71. Maiti S, Alam R, Amos CI, et al.: Frequent association of beta-catenin and WT1 mutations in Wilms tumors. Cancer Res 60 (22): 6288-92, 2000. [PUBMED Abstract]
  72. Gadd S, Huff V, Huang CC, et al.: Clinically relevant subsets identified by gene expression patterns support a revised ontogenic model of Wilms tumor: a Children's Oncology Group Study. Neoplasia 14 (8): 742-56, 2012. [PUBMED Abstract]
  73. Breslow NE, Beckwith JB, Perlman EJ, et al.: Age distributions, birth weights, nephrogenic rests, and heterogeneity in the pathogenesis of Wilms tumor. Pediatr Blood Cancer 47 (3): 260-7, 2006. [PUBMED Abstract]
  74. Fukuzawa R, Heathcott RW, More HE, et al.: Sequential WT1 and CTNNB1 mutations and alterations of beta-catenin localisation in intralobar nephrogenic rests and associated Wilms tumours: two case studies. J Clin Pathol 60 (9): 1013-6, 2007. [PUBMED Abstract]
  75. Perlman EJ, Gadd S, Arold ST, et al.: MLLT1 YEATS domain mutations in clinically distinctive Favourable Histology Wilms tumours. Nat Commun 6: 10013, 2015. [PUBMED Abstract]
  76. Diller L, Ghahremani M, Morgan J, et al.: Constitutional WT1 mutations in Wilms' tumor patients. J Clin Oncol 16 (11): 3634-40, 1998. [PUBMED Abstract]
  77. Perlman EJ, Grundy PE, Anderson JR, 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 29 (6): 698-703, 2011. [PUBMED Abstract]
  78. Scott RH, Walker L, Olsen ØE, et al.: Surveillance for Wilms tumour in at-risk children: pragmatic recommendations for best practice. Arch Dis Child 91 (12): 995-9, 2006. [PUBMED Abstract]
  79. Lipska BS, Ranchin B, Iatropoulos P, et al.: Genotype-phenotype associations in WT1 glomerulopathy. Kidney Int 85 (5): 1169-78, 2014. [PUBMED Abstract]
  80. Lehnhardt A, Karnatz C, Ahlenstiel-Grunow T, et al.: Clinical and molecular characterization of patients with heterozygous mutations in wilms tumor suppressor gene 1. Clin J Am Soc Nephrol 10 (5): 825-31, 2015. [PUBMED Abstract]
  81. Lange J, Peterson SM, Takashima JR, et al.: Risk factors for end stage renal disease in non-WT1-syndromic Wilms tumor. J Urol 186 (2): 378-86, 2011. [PUBMED Abstract]
  82. Breslow NE, Takashima JR, Ritchey ML, et al.: Renal failure in the Denys-Drash and Wilms' tumor-aniridia syndromes. Cancer Res 60 (15): 4030-2, 2000. [PUBMED Abstract]
  83. Koesters R, Ridder R, Kopp-Schneider A, et al.: Mutational activation of the beta-catenin proto-oncogene is a common event in the development of Wilms' tumors. Cancer Res 59 (16): 3880-2, 1999. [PUBMED Abstract]
  84. Koesters R, Niggli F, von Knebel Doeberitz M, et al.: Nuclear accumulation of beta-catenin protein in Wilms' tumours. J Pathol 199 (1): 68-76, 2003. [PUBMED Abstract]
  85. Major MB, Camp ND, Berndt JD, et al.: Wilms tumor suppressor WTX negatively regulates WNT/beta-catenin signaling. Science 316 (5827): 1043-6, 2007. [PUBMED Abstract]
  86. Rivera MN, Kim WJ, Wells J, et al.: An X chromosome gene, WTX, is commonly inactivated in Wilms tumor. Science 315 (5812): 642-5, 2007. [PUBMED Abstract]
  87. Fukuzawa R, Anaka MR, Weeks RJ, et al.: Canonical WNT signalling determines lineage specificity in Wilms tumour. Oncogene 28 (8): 1063-75, 2009. [PUBMED Abstract]
  88. Jenkins ZA, van Kogelenberg M, Morgan T, et al.: Germline mutations in WTX cause a sclerosing skeletal dysplasia but do not predispose to tumorigenesis. Nat Genet 41 (1): 95-100, 2009. [PUBMED Abstract]
  89. Grohmann A, Tanneberger K, Alzner A, et al.: AMER1 regulates the distribution of the tumor suppressor APC between microtubules and the plasma membrane. J Cell Sci 120 (Pt 21): 3738-47, 2007. [PUBMED Abstract]
  90. Satoh Y, Nakadate H, Nakagawachi T, et al.: Genetic and epigenetic alterations on the short arm of chromosome 11 are involved in a majority of sporadic Wilms' tumours. Br J Cancer 95 (4): 541-7, 2006. [PUBMED Abstract]
  91. Algar EM, St Heaps L, Darmanian A, et al.: Paternally inherited submicroscopic duplication at 11p15.5 implicates insulin-like growth factor II in overgrowth and Wilms' tumorigenesis. Cancer Res 67 (5): 2360-5, 2007. [PUBMED Abstract]
  92. Lennerz JK, Timmerman RJ, Grange DK, et al.: Addition of H19 'loss of methylation testing' for Beckwith-Wiedemann syndrome (BWS) increases the diagnostic yield. J Mol Diagn 12 (5): 576-88, 2010. [PUBMED Abstract]
  93. Mussa A, Molinatto C, Baldassarre G, et al.: Cancer Risk in Beckwith-Wiedemann Syndrome: A Systematic Review and Meta-Analysis Outlining a Novel (Epi)Genotype Specific Histotype Targeted Screening Protocol. J Pediatr 176: 142-149.e1, 2016. [PUBMED Abstract]
  94. Bliek J, Gicquel C, Maas S, et al.: Epigenotyping as a tool for the prediction of tumor risk and tumor type in patients with Beckwith-Wiedemann syndrome (BWS). J Pediatr 145 (6): 796-9, 2004. [PUBMED Abstract]
  95. Bjornsson HT, Brown LJ, Fallin MD, et al.: Epigenetic specificity of loss of imprinting of the IGF2 gene in Wilms tumors. J Natl Cancer Inst 99 (16): 1270-3, 2007. [PUBMED Abstract]
  96. Fukuzawa R, Breslow NE, Morison IM, et al.: Epigenetic differences between Wilms' tumours in white and east-Asian children. Lancet 363 (9407): 446-51, 2004. [PUBMED Abstract]
  97. Gratias EJ, Dome JS, Jennings LJ, et al.: Association of Chromosome 1q Gain With Inferior Survival in Favorable-Histology Wilms Tumor: A Report From the Children's Oncology Group. J Clin Oncol 34 (26): 3189-94, 2016. [PUBMED Abstract]
  98. Chagtai T, Zill C, Dainese L, et al.: Gain of 1q As a Prognostic Biomarker in Wilms Tumors (WTs) Treated With Preoperative Chemotherapy in the International Society of Paediatric Oncology (SIOP) WT 2001 Trial: A SIOP Renal Tumours Biology Consortium Study. J Clin Oncol 34 (26): 3195-203, 2016. [PUBMED Abstract]
  99. Grundy PE, Breslow NE, Li S, 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 23 (29): 7312-21, 2005. [PUBMED Abstract]
  100. Messahel B, Williams R, Ridolfi A, 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 45 (5): 819-26, 2009. [PUBMED Abstract]
  101. Spreafico F, Gamba B, Mariani L, et al.: Loss of heterozygosity analysis at different chromosome regions in Wilms tumor confirms 1p allelic loss as a marker of worse prognosis: a study from the Italian Association of Pediatric Hematology and Oncology. J Urol 189 (1): 260-6, 2013. [PUBMED Abstract]
  102. Gratias EJ, Jennings LJ, Anderson JR, et al.: Gain of 1q is associated with inferior event-free and overall survival in patients with favorable histology Wilms tumor: a report from the Children's Oncology Group. Cancer 119 (21): 3887-94, 2013. [PUBMED Abstract]
  103. Hohenstein P, Pritchard-Jones K, Charlton J: The yin and yang of kidney development and Wilms' tumors. Genes Dev 29 (5): 467-82, 2015. [PUBMED Abstract]
  104. Foulkes WD, Priest JR, Duchaine TF: DICER1: mutations, microRNAs and mechanisms. Nat Rev Cancer 14 (10): 662-72, 2014. [PUBMED Abstract]
  105. Wu MK, Sabbaghian N, Xu B, et al.: Biallelic DICER1 mutations occur in Wilms tumours. J Pathol 230 (2): 154-64, 2013. [PUBMED Abstract]
  106. Palculict TB, Ruteshouser EC, Fan Y, et al.: Identification of germline DICER1 mutations and loss of heterozygosity in familial Wilms tumour. J Med Genet 53 (6): 385-8, 2016. [PUBMED Abstract]
  107. Chang HM, Triboulet R, Thornton JE, et al.: A role for the Perlman syndrome exonuclease Dis3l2 in the Lin28-let-7 pathway. Nature 497 (7448): 244-8, 2013. [PUBMED Abstract]
  108. Alessandri JL, Cuillier F, Ramful D, et al.: Perlman syndrome: report, prenatal findings and review. Am J Med Genet A 146A (19): 2532-7, 2008. [PUBMED Abstract]
  109. Bardeesy N, Falkoff D, Petruzzi MJ, et al.: Anaplastic Wilms' tumour, a subtype displaying poor prognosis, harbours p53 gene mutations. Nat Genet 7 (1): 91-7, 1994. [PUBMED Abstract]
  110. el Bahtimi R, Hazen-Martin DJ, Re GG, et al.: Immunophenotype, mRNA expression, and gene structure of p53 in Wilms' tumors. Mod Pathol 9 (3): 238-44, 1996. [PUBMED Abstract]
  111. Wallkamm V, Dörlich R, Rahm K, et al.: Live imaging of Xwnt5A-ROR2 complexes. PLoS One 9 (10): e109428, 2014. [PUBMED Abstract]
  112. Ooms AH, Gadd S, Gerhard DS, et al.: Significance of TP53 Mutation in Wilms Tumors with Diffuse Anaplasia: A Report from the Children's Oncology Group. Clin Cancer Res 22 (22): 5582-5591, 2016. [PUBMED Abstract]
  113. Williams RD, Al-Saadi R, Chagtai T, et al.: Subtype-specific FBXW7 mutation and MYCN copy number gain in Wilms' tumor. Clin Cancer Res 16 (7): 2036-45, 2010. [PUBMED Abstract]
  114. Muller E, Hudgins L: 9q22.3 Microdeletion. In: Pagon RA, Adam MP, Bird TD, et al., eds.: GeneReviews. Seattle, Wash: University of Washington, 1993-2018, pp. Available online. Last accessed October 25, 2019.
  115. Garavelli L, Piemontese MR, Cavazza A, et al.: Multiple tumor types including leiomyoma and Wilms tumor in a patient with Gorlin syndrome due to 9q22.3 microdeletion encompassing the PTCH1 and FANC-C loci. Am J Med Genet A 161A (11): 2894-901, 2013. [PUBMED Abstract]
  116. Cajaiba MM, Bale AE, Alvarez-Franco M, et al.: Rhabdomyosarcoma, Wilms tumor, and deletion of the patched gene in Gorlin syndrome. Nat Clin Pract Oncol 3 (10): 575-80, 2006. [PUBMED Abstract]
  117. Williams RD, Chagtai T, Alcaide-German M, et al.: Multiple mechanisms of MYCN dysregulation in Wilms tumour. Oncotarget 6 (9): 7232-43, 2015. [PUBMED Abstract]
  118. Fievet A, Belaud-Rotureau MA, Dugay F, et al.: Involvement of germline DDX1-MYCN duplication in inherited nephroblastoma. Eur J Med Genet 56 (12): 643-7, 2013. [PUBMED Abstract]
  119. Martins AG, Pinto AT, Domingues R, et al.: Identification of a novel CTR9 germline mutation in a family with Wilms tumor. Eur J Med Genet 61 (5): 294-299, 2018. [PUBMED Abstract]
  120. Charlton J, Irtan S, Bergeron C, et al.: Bilateral Wilms tumour: a review of clinical and molecular features. Expert Rev Mol Med 19: e8, 2017. [PUBMED Abstract]
  121. Hu M, Fletcher J, McCahon E, et al.: Bilateral Wilms tumor and early presentation in pediatric patients is associated with the truncation of the Wilms tumor 1 protein. J Pediatr 163 (1): 224-9, 2013. [PUBMED Abstract]
  122. Murphy AJ, Davidoff AM: Bilateral Wilms Tumor: A Surgical Perspective. Children (Basel) 5 (10): , 2018. [PUBMED Abstract]
  123. Kalish JM, Doros L, Helman LJ, et al.: Surveillance Recommendations for Children with Overgrowth Syndromes and Predisposition to Wilms Tumors and Hepatoblastoma. Clin Cancer Res 23 (13): e115-e122, 2017. [PUBMED Abstract]
  124. Teplick A, Kowalski M, Biegel JA, et al.: Educational paper: screening in cancer predisposition syndromes: guidelines for the general pediatrician. Eur J Pediatr 170 (3): 285-94, 2011. [PUBMED Abstract]
  125. Mussa A, Duffy KA, Carli D, et al.: The effectiveness of Wilms tumor screening in Beckwith-Wiedemann spectrum. J Cancer Res Clin Oncol 145 (12): 3115-3123, 2019. [PUBMED Abstract]
  126. Hingorani M, Hanson I, van Heyningen V: Aniridia. Eur J Hum Genet 20 (10): 1011-7, 2012. [PUBMED Abstract]
  127. van Heyningen V, Hoovers JM, de Kraker J, et al.: Raised risk of Wilms tumour in patients with aniridia and submicroscopic WT1 deletion. J Med Genet 44 (12): 787-90, 2007. [PUBMED Abstract]
  128. Greene AK, Kieran M, Burrows PE, et al.: Wilms tumor screening is unnecessary in Klippel-Trenaunay syndrome. Pediatrics 113 (4): e326-9, 2004. [PUBMED Abstract]
  129. Schultz KAP, Rednam SP, Kamihara J, et al.: PTEN, DICER1, FH, and Their Associated Tumor Susceptibility Syndromes: Clinical Features, Genetics, and Surveillance Recommendations in Childhood. Clin Cancer Res 23 (12): e76-e82, 2017. [PUBMED Abstract]
  130. Schultz KAP, Williams GM, Kamihara J, et al.: DICER1 and Associated Conditions: Identification of At-risk Individuals and Recommended Surveillance Strategies. Clin Cancer Res 24 (10): 2251-2261, 2018. [PUBMED Abstract]
  131. Mitchell SG, Pencheva B, Porter CC: Germline Genetics and Childhood Cancer: Emerging Cancer Predisposition Syndromes and Psychosocial Impacts. Curr Oncol Rep 21 (10): 85, 2019. [PUBMED Abstract]
  132. Green DM: Wilms' tumor. In: Greem DM: Diagnosis and Management of Malignant Solid Tumors in Infants and Children. Boston, Ma: Martinus Nijhoff Publishing, 1985, pp 129-86.
  133. Servaes S, Khanna G, Naranjo A, et al.: Comparison of diagnostic performance of CT and MRI for abdominal staging of pediatric renal tumors: a report from the Children's Oncology Group. Pediatr Radiol 45 (2): 166-72, 2015. [PUBMED Abstract]
  134. Khanna G, Naranjo A, Hoffer F, et al.: Detection of preoperative wilms tumor rupture with CT: a report from the Children's Oncology Group. Radiology 266 (2): 610-7, 2013. [PUBMED Abstract]
  135. McDonald K, Duffy P, Chowdhury T, et al.: Added value of abdominal cross-sectional imaging (CT or MRI) in staging of Wilms' tumours. Clin Radiol 68 (1): 16-20, 2013. [PUBMED Abstract]
  136. Ritchey ML, Shamberger RC, Hamilton T, et al.: Fate of bilateral renal lesions missed on preoperative imaging: a report from the National Wilms Tumor Study Group. J Urol 174 (4 Pt 2): 1519-21; discussion 1521, 2005. [PUBMED Abstract]
  137. Khanna G, Rosen N, Anderson JR, et al.: Evaluation of diagnostic performance of CT for detection of tumor thrombus in children with Wilms tumor: a report from the Children's Oncology Group. Pediatr Blood Cancer 58 (4): 551-5, 2012. [PUBMED Abstract]
  138. Begent J, Sebire NJ, Levitt G, et al.: Pilot study of F(18)-Fluorodeoxyglucose Positron Emission Tomography/computerised tomography in Wilms' tumour: correlation with conventional imaging, pathology and immunohistochemistry. Eur J Cancer 47 (3): 389-96, 2011. [PUBMED Abstract]
  139. Callaghan MU, Wong TE, Federici AB: Treatment of acquired von Willebrand syndrome in childhood. Blood 122 (12): 2019-22, 2013. [PUBMED Abstract]
  140. Shamberger RC, Guthrie KA, Ritchey ML, et al.: Surgery-related factors and local recurrence of Wilms tumor in National Wilms Tumor Study 4. Ann Surg 229 (2): 292-7, 1999. [PUBMED Abstract]
  141. Ehrlich P, Chi YY, Chintagumpala MM, et al.: Results of the First Prospective Multi-institutional Treatment Study in Children With Bilateral Wilms Tumor (AREN0534): A Report From the Children's Oncology Group. Ann Surg 266 (3): 470-478, 2017. [PUBMED Abstract]
  142. Hamilton TE, Green DM, Perlman EJ, et al.: Bilateral Wilms' tumor with anaplasia: lessons from the National Wilms' Tumor Study. J Pediatr Surg 41 (10): 1641-4, 2006. [PUBMED Abstract]
  143. Othersen HB, DeLorimer A, Hrabovsky E, et al.: Surgical evaluation of lymph node metastases in Wilms' tumor. J Pediatr Surg 25 (3): 330-1, 1990. [PUBMED Abstract]
  144. Green DM: Controversies in the management of Wilms tumour - immediate nephrectomy or delayed nephrectomy? Eur J Cancer 43 (17): 2453-6, 2007. [PUBMED Abstract]
  145. Shamberger RC, Ritchey ML, Haase GM, et al.: Intravascular extension of Wilms tumor. Ann Surg 234 (1): 116-21, 2001. [PUBMED Abstract]
  146. Servaes SE, Hoffer FA, Smith EA, et al.: Imaging of Wilms tumor: an update. Pediatr Radiol 49 (11): 1441-1452, 2019. [PUBMED Abstract]
  147. Smith MA, Altekruse SF, Adamson PC, et al.: Declining childhood and adolescent cancer mortality. Cancer 120 (16): 2497-506, 2014. [PUBMED Abstract]
  148. Green DM, Breslow NE, Beckwith JB, 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 16 (12): 3744-51, 1998. [PUBMED Abstract]
  149. Kalapurakal JA, Dome JS, Perlman EJ, et al.: Management of Wilms' tumour: current practice and future goals. Lancet Oncol 5 (1): 37-46, 2004. [PUBMED Abstract]
  150. Ehrlich PF: Wilms tumor: progress and considerations for the surgeon. Surg Oncol 16 (3): 157-71, 2007. [PUBMED Abstract]
  151. Dome JS, Cotton CA, Perlman EJ, et al.: Treatment of anaplastic histology Wilms' tumor: results from the fifth National Wilms' Tumor Study. J Clin Oncol 24 (15): 2352-8, 2006. [PUBMED Abstract]
  152. Shamberger RC, Anderson JR, Breslow NE, et al.: Long-term outcomes for infants with very low risk Wilms tumor treated with surgery alone in National Wilms Tumor Study-5. Ann Surg 251 (3): 555-8, 2010. [PUBMED Abstract]
  153. Hol JA, Lopez-Yurda MI, Van Tinteren H, et al.: Prognostic significance of age in 5631 patients with Wilms tumour prospectively registered in International Society of Paediatric Oncology (SIOP) 93-01 and 2001. PLoS One 14 (8): e0221373, 2019. [PUBMED Abstract]
  154. Mitry E, Ciccolallo L, Coleman MP, et al.: Incidence of and survival from Wilms' tumour in adults in Europe: data from the EUROCARE study. Eur J Cancer 42 (14): 2363-8, 2006. [PUBMED Abstract]
  155. Chen I, Pasalic D, Fischer-Valuck B, et al.: Disparity in Outcomes for Adolescent and Young Adult Patients Diagnosed With Pediatric Solid Tumors Across 4 Decades. Am J Clin Oncol 41 (5): 471-475, 2018. [PUBMED Abstract]
  156. Kalapurakal JA, Nan B, Norkool P, et al.: Treatment outcomes in adults with favorable histologic type Wilms tumor-an update from the National Wilms Tumor Study Group. Int J Radiat Oncol Biol Phys 60 (5): 1379-84, 2004. [PUBMED Abstract]
  157. Arrigo S, Beckwith JB, Sharples K, et al.: Better survival after combined modality care for adults with Wilms' tumor. A report from the National Wilms' Tumor Study. Cancer 66 (5): 827-30, 1990. [PUBMED Abstract]
  158. Byrd RL, Evans AE, D'Angio GJ: Adult Wilms tumor: effect of combined therapy on survival. J Urol 127 (4): 648-51, 1982. [PUBMED Abstract]
  159. de Vries-Brilland M, Sionneau B, Dutriaux C, et al.: Successful Treatment of Metastatic Adult Wilms Tumor With Anti-BRAF Treatment: A Case Report and a Brief Review of the Literature. Clin Genitourin Cancer 17 (4): e721-e723, 2019. [PUBMED Abstract]
  160. Segers H, van den Heuvel-Eibrink MM, Pritchard-Jones K, et al.: Management of adults with Wilms' tumor: recommendations based on international consensus. Expert Rev Anticancer Ther 11 (7): 1105-13, 2011. [PUBMED Abstract]
  161. Perlman EJ: Pediatric renal tumors: practical updates for the pathologist. Pediatr Dev Pathol 8 (3): 320-38, 2005 May-Jun. [PUBMED Abstract]
  162. Popov SD, Sebire NJ, Pritchard-Jones K, et al.: Renal tumors in children aged 10-16 Years: a report from the United Kingdom Children's Cancer and Leukaemia Group. Pediatr Dev Pathol 14 (3): 189-93, 2011 May-Jun. [PUBMED Abstract]
  163. Indolfi P, Jenkner A, Terenziani M, et al.: Synchronous bilateral Wilms tumor: a report from the Associazione Italiana Ematologia Oncologia Pediatrica (AIEOP). Cancer 119 (8): 1586-92, 2013. [PUBMED Abstract]
  164. Hamilton TE, Ritchey ML, Haase GM, et al.: The management of synchronous bilateral Wilms tumor: a report from the National Wilms Tumor Study Group. Ann Surg 253 (5): 1004-10, 2011. [PUBMED Abstract]
  165. Williams RD, Al-Saadi R, Natrajan R, et al.: Molecular profiling reveals frequent gain of MYCN and anaplasia-specific loss of 4q and 14q in Wilms tumor. Genes Chromosomes Cancer 50 (12): 982-95, 2011. [PUBMED Abstract]
  166. Vujanić GM, Harms D, Sandstedt B, et al.: New definitions of focal and diffuse anaplasia in Wilms tumor: the International Society of Paediatric Oncology (SIOP) experience. Med Pediatr Oncol 32 (5): 317-23, 1999. [PUBMED Abstract]
  167. Faria P, Beckwith JB, Mishra K, 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 20 (8): 909-20, 1996. [PUBMED Abstract]
  168. Beckwith JB: New developments in the pathology of Wilms tumor. Cancer Invest 15 (2): 153-62, 1997. [PUBMED Abstract]
  169. Beckwith JB: Precursor lesions of Wilms tumor: clinical and biological implications. Med Pediatr Oncol 21 (3): 158-68, 1993. [PUBMED Abstract]
  170. Perlman EJ, Faria P, Soares A, et al.: Hyperplastic perilobar nephroblastomatosis: long-term survival of 52 patients. Pediatr Blood Cancer 46 (2): 203-21, 2006. [PUBMED Abstract]
  171. Furtwängler R, Schmolze M, Gräber S, et al.: Pretreatment for bilateral nephroblastomatosis is an independent risk factor for progressive disease in patients with stage V nephroblastoma. Klin Padiatr 226 (3): 175-81, 2014. [PUBMED Abstract]
  172. Cooke A, Deshpande AV, La Hei ER, et al.: Ectopic nephrogenic rests in children: the clinicosurgical implications. J Pediatr Surg 44 (12): e13-6, 2009. [PUBMED Abstract]
  173. Wilms' tumor: status report, 1990. By the National Wilms' Tumor Study Committee. J Clin Oncol 9 (5): 877-87, 1991. [PUBMED Abstract]
  174. Green DM, Breslow NE, D'Angio GJ, et al.: Outcome of patients with Stage II/favorable histology Wilms tumor with and without local tumor spill: a report from the National Wilms Tumor Study Group. Pediatr Blood Cancer 61 (1): 134-9, 2014. [PUBMED Abstract]
  175. Ehrlich PF, Anderson JR, Ritchey ML, et al.: Clinicopathologic findings predictive of relapse in children with stage III favorable-histology Wilms tumor. J Clin Oncol 31 (9): 1196-201, 2013. [PUBMED Abstract]
  176. D'Angio GJ, Breslow N, Beckwith JB, et al.: Treatment of Wilms' tumor. Results of the Third National Wilms' Tumor Study. Cancer 64 (2): 349-60, 1989. [PUBMED Abstract]
  177. Jereb B, Burgers JM, Tournade MF, et al.: Radiotherapy in the SIOP (International Society of Pediatric Oncology) nephroblastoma studies: a review. Med Pediatr Oncol 22 (4): 221-7, 1994. [PUBMED Abstract]
  178. Green DM: The treatment of stages I-IV favorable histology Wilms' tumor. J Clin Oncol 22 (8): 1366-72, 2004. [PUBMED Abstract]
  179. Graf N, Tournade MF, de Kraker J: The role of preoperative chemotherapy in the management of Wilms' tumor. The SIOP studies. International Society of Pediatric Oncology. Urol Clin North Am 27 (3): 443-54, 2000. [PUBMED Abstract]
  180. Vujanić GM, D'Hooghe E, Popov SD, et al.: The effect of preoperative chemotherapy on histological subtyping and staging of Wilms tumors: The United Kingdom Children's Cancer Study Group (UKCCSG) Wilms tumor trial 3 (UKW3) experience. Pediatr Blood Cancer 66 (3): e27549, 2019. [PUBMED Abstract]
  181. van den Heuvel-Eibrink MM, Hol JA, Pritchard-Jones K, et al.: Position paper: Rationale for the treatment of Wilms tumour in the UMBRELLA SIOP-RTSG 2016 protocol. Nat Rev Urol 14 (12): 743-752, 2017. [PUBMED Abstract]
  182. Green DM, Breslow NE, Beckwith JB, et al.: Comparison between single-dose and divided-dose administration of dactinomycin and doxorubicin for patients with Wilms' tumor: a report from the National Wilms' Tumor Study Group. J Clin Oncol 16 (1): 237-45, 1998. [PUBMED Abstract]
  183. D'Angio GJ, Evans AE, Breslow N, et al.: The treatment of Wilms' tumor: Results of the national Wilms' tumor study. Cancer 38 (2): 633-46, 1976. [PUBMED Abstract]
  184. D'Angio GJ, Evans A, Breslow N, et al.: The treatment of Wilms' tumor: results of the Second National Wilms' Tumor Study. Cancer 47 (9): 2302-11, 1981. [PUBMED Abstract]
  185. Kieran K, Anderson JR, Dome JS, et al.: Lymph node involvement in Wilms tumor: results from National Wilms Tumor Studies 4 and 5. J Pediatr Surg 47 (4): 700-6, 2012. [PUBMED Abstract]
  186. Ritchey M, Daley S, Shamberger RC, et al.: Ureteral extension in Wilms' tumor: a report from the National Wilms' Tumor Study Group (NWTSG). J Pediatr Surg 43 (9): 1625-9, 2008. [PUBMED Abstract]
  187. Gow KW, Barnhart DC, Hamilton TE, et al.: Primary nephrectomy and intraoperative tumor spill: report from the Children's Oncology Group (COG) renal tumors committee. J Pediatr Surg 48 (1): 34-8, 2013. [PUBMED Abstract]
  188. McNeil DE, Langer JC, Choyke P, et al.: Feasibility of partial nephrectomy for Wilms' tumor in children with Beckwith-Wiedemann syndrome who have been screened with abdominal ultrasonography. J Pediatr Surg 37 (1): 57-60, 2002. [PUBMED Abstract]
  189. Scalabre A, Bergeron C, Brioude F, et al.: Is Nephron Sparing Surgery Justified in Wilms Tumor With Beckwith-Wiedemann Syndrome or Isolated Hemihypertrophy? Pediatr Blood Cancer 63 (9): 1571-7, 2016. [PUBMED Abstract]
  190. Auber F, Jeanpierre C, Denamur E, et al.: Management of Wilms tumors in Drash and Frasier syndromes. Pediatr Blood Cancer 52 (1): 55-9, 2009. [PUBMED Abstract]
  191. Neville H, Ritchey ML, Shamberger RC, et al.: The occurrence of Wilms tumor in horseshoe kidneys: a report from the National Wilms Tumor Study Group (NWTSG). J Pediatr Surg 37 (8): 1134-7, 2002. [PUBMED Abstract]
  192. Ferrer FA, Rosen N, Herbst K, et al.: Image based feasibility of renal sparing surgery for very low risk unilateral Wilms tumors: a report from the Children's Oncology Group. J Urol 190 (5): 1846-51, 2013. [PUBMED Abstract]
  193. Ritchey ML: Renal sparing surgery for Wilms tumor. J Urol 174 (4 Pt 1): 1172-3, 2005. [PUBMED Abstract]
  194. Cozzi DA, Zani A: Nephron-sparing surgery in children with primary renal tumor: indications and results. Semin Pediatr Surg 15 (1): 3-9, 2006. [PUBMED Abstract]
  195. Zhuge Y, Cheung MC, Yang R, et al.: Improved survival with lymph node sampling in Wilms tumor. J Surg Res 167 (2): e199-203, 2011. [PUBMED Abstract]
  196. Ritchey ML, Kelalis PP, Breslow N, et al.: Surgical complications after nephrectomy for Wilms' tumor. Surg Gynecol Obstet 175 (6): 507-14, 1992. [PUBMED Abstract]
  197. Ehrlich PF, Ferrer FA, Ritchey ML, et al.: Hepatic metastasis at diagnosis in patients with Wilms tumor is not an independent adverse prognostic factor for stage IV Wilms tumor: a report from the Children's Oncology Group/National Wilms Tumor Study Group. Ann Surg 250 (4): 642-8, 2009. [PUBMED Abstract]
  198. Ritchey ML: Primary nephrectomy for Wilms' tumor: approach of the National Wilms' Tumor Study Group. Urology 47 (6): 787-91, 1996. [PUBMED Abstract]
  199. Lall A, Pritchard-Jones K, Walker J, et al.: Wilms' tumor with intracaval thrombus in the UK Children's Cancer Study Group UKW3 trial. J Pediatr Surg 41 (2): 382-7, 2006. [PUBMED Abstract]
  200. Ritchey ML, Pringle KC, Breslow NE, et al.: Management and outcome of inoperable Wilms tumor. A report of National Wilms Tumor Study-3. Ann Surg 220 (5): 683-90, 1994. [PUBMED Abstract]
  201. Ritchey ML, Shamberger RC, Haase G, et al.: Surgical complications after primary nephrectomy for Wilms' tumor: report from the National Wilms' Tumor Study Group. J Am Coll Surg 192 (1): 63-8; quiz 146, 2001. [PUBMED Abstract]
  202. Tournade MF, Com-Nougué C, Voûte PA, et al.: Results of the Sixth International Society of Pediatric Oncology Wilms' Tumor Trial and Study: a risk-adapted therapeutic approach in Wilms' tumor. J Clin Oncol 11 (6): 1014-23, 1993. [PUBMED Abstract]
  203. Oberholzer HF, Falkson G, De Jager LC: Successful management of inferior vena cava and right atrial nephroblastoma tumor thrombus with preoperative chemotherapy. Med Pediatr Oncol 20 (1): 61-3, 1992. [PUBMED Abstract]
  204. Saarinen UM, Wikström S, Koskimies O, et al.: Percutaneous needle biopsy preceding preoperative chemotherapy in the management of massive renal tumors in children. J Clin Oncol 9 (3): 406-15, 1991. [PUBMED Abstract]
  205. Dykes EH, Marwaha RK, Dicks-Mireaux C, et al.: Risks and benefits of percutaneous biopsy and primary chemotherapy in advanced Wilms' tumour. J Pediatr Surg 26 (5): 610-2, 1991. [PUBMED Abstract]
  206. Thompson WR, Newman K, Seibel N, et al.: A strategy for resection of Wilms' tumor with vena cava or atrial extension. J Pediatr Surg 27 (7): 912-5, 1992. [PUBMED Abstract]
  207. Szavay P, Luithle T, Semler O, et al.: Surgery of cavoatrial tumor thrombus in nephroblastoma: a report of the SIOP/GPOH study. Pediatr Blood Cancer 43 (1): 40-5, 2004. [PUBMED Abstract]
  208. Powis M, Messahel B, Hobson R, et al.: Surgical complications after immediate nephrectomy versus preoperative chemotherapy in non-metastatic Wilms' tumour: findings from the 1991-2001 United Kingdom Children's Cancer Study Group UKW3 Trial. J Pediatr Surg 48 (11): 2181-6, 2013. [PUBMED Abstract]
  209. Rutigliano DN, Kayton ML, Steinherz P, et al.: The use of preoperative chemotherapy in Wilms' tumor with contained retroperitoneal rupture. J Pediatr Surg 42 (9): 1595-9, 2007. [PUBMED Abstract]
  210. Brisse HJ, Schleiermacher G, Sarnacki S, et al.: Preoperative Wilms tumor rupture: a retrospective study of 57 patients. Cancer 113 (1): 202-13, 2008. [PUBMED Abstract]
  211. Corn BW, Goldwein JW, Evans I, et al.: Outcomes in low-risk babies treated with half-dose chemotherapy according to the Third National Wilms' Tumor Study. J Clin Oncol 10 (8): 1305-9, 1992. [PUBMED Abstract]
  212. Morgan E, Baum E, Breslow N, et al.: Chemotherapy-related toxicity in infants treated according to the Second National Wilms' Tumor Study. J Clin Oncol 6 (1): 51-5, 1988. [PUBMED Abstract]
  213. Green DM, Norkool P, Breslow NE, et al.: Severe hepatic toxicity after treatment with vincristine and dactinomycin using single-dose or divided-dose schedules: a report from the National Wilms' Tumor Study. J Clin Oncol 8 (9): 1525-30, 1990. [PUBMED Abstract]
  214. Raine J, Bowman A, Wallendszus K, et al.: Hepatopathy-thrombocytopenia syndrome--a complication of dactinomycin therapy for Wilms' tumor: a report from the United Kingdom Childrens Cancer Study Group. J Clin Oncol 9 (2): 268-73, 1991. [PUBMED Abstract]
  215. Feusner JH, Ritchey ML, Norkool PA, et al.: Renal failure does not preclude cure in children receiving chemotherapy for Wilms tumor: a report from the National Wilms Tumor Study Group. Pediatr Blood Cancer 50 (2): 242-5, 2008. [PUBMED Abstract]
  216. Veal GJ, English MW, Grundy RG, et al.: Pharmacokinetically guided dosing of carboplatin in paediatric cancer patients with bilateral nephrectomy. Cancer Chemother Pharmacol 54 (4): 295-300, 2004. [PUBMED Abstract]
  217. Dix DB, Fernandez CV, Chi YY, et al.: Augmentation of Therapy for Combined Loss of Heterozygosity 1p and 16q in Favorable Histology Wilms Tumor: A Children's Oncology Group AREN0532 and AREN0533 Study Report. J Clin Oncol 37 (30): 2769-2777, 2019. [PUBMED Abstract]
  218. Stokes CL, Stokes WA, Kalapurakal JA, et al.: Timing of Radiation Therapy in Pediatric Wilms Tumor: A Report From the National Cancer Database. Int J Radiat Oncol Biol Phys 101 (2): 453-461, 2018. [PUBMED Abstract]
  219. Dix DB, Seibel NL, Chi YY, et al.: Treatment of Stage IV Favorable Histology Wilms Tumor With Lung Metastases: A Report From the Children's Oncology Group AREN0533 Study. J Clin Oncol 36 (16): 1564-1570, 2018. [PUBMED Abstract]
  220. Fernandez CV, Perlman EJ, Mullen EA, et al.: Clinical Outcome and Biological Predictors of Relapse After Nephrectomy Only for Very Low-risk Wilms Tumor: A Report From Children's Oncology Group AREN0532. Ann Surg 265 (4): 835-840, 2017. [PUBMED Abstract]
  221. Thomas PR, Tefft M, Compaan PJ, et al.: Results of two radiation therapy randomizations in the third National Wilms' Tumor Study. Cancer 68 (8): 1703-7, 1991. [PUBMED Abstract]
  222. Tefft M, D'Angio GJ, Beckwith B, et al.: Patterns of intra-abdominal relapse (IAR) in patients with Wilms' tumor who received radiation: analysis by histopathology. A report of National Wilms' Tumor Studies 1 and 2 (NWTS-1 & 2). Int J Radiat Oncol Biol Phys 6 (6): 663-7, 1980. [PUBMED Abstract]
  223. Thomas PR, Tefft M, Farewell VT, et al.: Abdominal relapses in irradiated second National Wilms' Tumor Study patients. J Clin Oncol 2 (10): 1098-101, 1984. [PUBMED Abstract]
  224. Meisel JA, Guthrie KA, Breslow NE, 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 44 (3): 579-85, 1999. [PUBMED Abstract]
  225. Daw NC, Chi YY, Kim Y, et al.: Treatment of stage I anaplastic Wilms' tumour: a report from the Children's Oncology Group AREN0321 study. Eur J Cancer 118: 58-66, 2019. [PUBMED Abstract]
  226. Green DM, Breslow NE, Beckwith JB, et al.: Treatment with nephrectomy only for small, stage I/favorable histology Wilms' tumor: a report from the National Wilms' Tumor Study Group. J Clin Oncol 19 (17): 3719-24, 2001. [PUBMED Abstract]
  227. Kalapurakal JA, Li SM, Breslow NE, et al.: Intraoperative spillage of favorable histology wilms tumor cells: influence of irradiation and chemotherapy regimens on abdominal recurrence. A report from the National Wilms Tumor Study Group. Int J Radiat Oncol Biol Phys 76 (1): 201-6, 2010. [PUBMED Abstract]
  228. Fernandez CV, Mullen EA, Chi YY, et al.: Outcome and Prognostic Factors in Stage III Favorable-Histology Wilms Tumor: A Report From the Children's Oncology Group Study AREN0532. J Clin Oncol 36 (3): 254-261, 2018. [PUBMED Abstract]
  229. Grundy PE, Green DM, Dirks AC, et al.: Clinical significance of pulmonary nodules detected by CT and Not CXR in patients treated for favorable histology Wilms tumor on national Wilms tumor studies-4 and -5: a report from the Children's Oncology Group. Pediatr Blood Cancer 59 (4): 631-5, 2012. [PUBMED Abstract]
  230. Verschuur A, Van Tinteren H, Graf N, et al.: Treatment of pulmonary metastases in children with stage IV nephroblastoma with risk-based use of pulmonary radiotherapy. J Clin Oncol 30 (28): 3533-9, 2012. [PUBMED Abstract]
  231. Breslow NE, Collins AJ, Ritchey ML, 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 174 (5): 1972-5, 2005. [PUBMED Abstract]
  232. Aronson DC, Slaar A, Heinen RC, et al.: Long-term outcome of bilateral Wilms tumors (BWT). Pediatr Blood Cancer 56 (7): 1110-3, 2011. [PUBMED Abstract]
  233. Zuppan CW, Beckwith JB, Weeks DA, et al.: The effect of preoperative therapy on the histologic features of Wilms' tumor. An analysis of cases from the Third National Wilms' Tumor Study. Cancer 68 (2): 385-94, 1991. [PUBMED Abstract]
  234. Ehrlich PF: Bilateral Wilms' tumor: the need to improve outcomes. Expert Rev Anticancer Ther 9 (7): 963-73, 2009. [PUBMED Abstract]
  235. Sudour H, Audry G, Schleimacher G, et al.: Bilateral Wilms tumors (WT) treated with the SIOP 93 protocol in France: epidemiological survey and patient outcome. Pediatr Blood Cancer 59 (1): 57-61, 2012. [PUBMED Abstract]
  236. Davidoff AM, Interiano RB, Wynn L, et al.: Overall Survival and Renal Function of Patients With Synchronous Bilateral Wilms Tumor Undergoing Surgery at a Single Institution. Ann Surg 262 (4): 570-6, 2015. [PUBMED Abstract]
  237. Kieran K, Williams MA, McGregor LM, et al.: Repeat nephron-sparing surgery for children with bilateral Wilms tumor. J Pediatr Surg 49 (1): 149-53, 2014. [PUBMED Abstract]
  238. Kist-van Holthe JE, Ho PL, Stablein D, et al.: Outcome of renal transplantation for Wilms' tumor and Denys-Drash syndrome: a report of the North American Pediatric Renal Transplant Cooperative Study. Pediatr Transplant 9 (3): 305-10, 2005. [PUBMED Abstract]
  239. Wong KF, Reulen RC, Winter DL, et al.: Risk of Adverse Health and Social Outcomes Up to 50 Years After Wilms Tumor: The British Childhood Cancer Survivor Study. J Clin Oncol 34 (15): 1772-9, 2016. [PUBMED Abstract]
  240. Breslow NE, Lange JM, Friedman DL, et al.: Secondary malignant neoplasms after Wilms tumor: an international collaborative study. Int J Cancer 127 (3): 657-66, 2010. [PUBMED Abstract]
  241. Termuhlen AM, Tersak JM, Liu Q, et al.: Twenty-five year follow-up of childhood Wilms tumor: a report from the Childhood Cancer Survivor Study. Pediatr Blood Cancer 57 (7): 1210-6, 2011. [PUBMED Abstract]
  242. Lange JM, Takashima JR, Peterson SM, et al.: Breast cancer in female survivors of Wilms tumor: a report from the national Wilms tumor late effects study. Cancer 120 (23): 3722-30, 2014. [PUBMED Abstract]
  243. Green DM, Grigoriev YA, Nan B, et al.: Congestive heart failure after treatment for Wilms' tumor: a report from the National Wilms' Tumor Study group. J Clin Oncol 19 (7): 1926-34, 2001. [PUBMED Abstract]
  244. Green DM, Lange JM, Peabody EM, et al.: Pregnancy outcome after treatment for Wilms tumor: a report from the national Wilms tumor long-term follow-up study. J Clin Oncol 28 (17): 2824-30, 2010. [PUBMED Abstract]

No hay comentarios:

Publicar un comentario