Abstract
Outcomes are excellent for the majority of patients with Wilms tumors (WT). However, there remain WT subgroups for which the survival rate is approximately 50% or lower. Acknowledging that the composition of this high-risk group has changed over time reflecting improvements in therapy, we introduce the authors’ view of the historical and current approach to the classification and treatment of high-risk WT. For this review we consider high-risk WT to include patients with newly diagnosed metastatic blastemal-type or diffuse anaplastic histology, those who relapse after having been initially treated with three or more different chemotherapeutics, or those who relapse more than once. In certain low or low-middle income settings, socio-economic factors expand the definition of what constitutes a high-risk WT. As conventional therapies are inadequate to cure the majority of high-risk WT patients, advancement of laboratory and early phase clinical investigations to identify active agents is urgently needed.
Keywords: Wilms tumor, nephroblastoma, high-risk, relapsed, SIOP, COG
Defining high-risk Wilms tumor
Risk-stratified approaches using either the Société Internationale d’Oncologie Pédiatrique (SIOP) Renal Tumor Study Group (RTSG) or Children’s Oncology Group (COG) Renal Tumor Committee (RTC) strategies have led to survival rates over 90% for children with Wilms tumors (WT), in aggregate1. However, there remain subgroups of WT for which the risk of treatment failure and subsequent mortality are unacceptably high.
In this article, we define “high-risk” as those patients with expected overall survival (OS) of approximately 50% or lower. This “high-risk” category has evolved as we have iteratively improved clinical management through the addition of effective therapies and supportive care, as well as refined risk stratification. For example, stage I-III diffuse anaplastic WT (DAWT) and stage III/IV non-anaplastic WT with specific adverse genetic features (combined loss of heterozygosity (LOH) at chromosomes 1p and 16q) previously had poor OS, but clinical trials using augmented therapies have substantially improved outcomes (Table 1)2. Likewise, survival after relapse has improved over time and patients with WT relapse after receiving only vincristine and actinomycin-D up-front now surpass post-relapse OS of 80% (Table 1 and 2).
TABLE 1:
Diffuse anaplastic Wilms tumor | ||||||
---|---|---|---|---|---|---|
NWTS-3 and 4; Regimen DD-RT | NWTS-3 and 4; Regimen J | NWTS-5; Regimen I | AREN0321; Regimen UH-1 or UH-2 (original or revised) | |||
Stage II | 4-year RFS 40.0% 4-year OS 46.9% |
4-year RFS 71.6% 4-year OS 70.1% |
4-year EFS 79.2% (95% CI, 60.9–97.5%) 4-year OS 78.4% (95% CI, 60.0–96.9%) |
4-year EFS 86.7% (95% CI, 68.8–100%) 4-year OS 86.2% ( |
||
Stage III | 4-year RFS 33.3% 4-year OS 20.8% |
4-year RFS 58.7% 4-year OS 56.3% |
4-year EFS 61.3% (95% CI, 47.8–74.7%) 4-year OS 64.7% (95% CI, 51.6–77.8%) |
4-year EFS 80.9% (95% CI, 65.8–96.0%) 4-year OS 88.6% (95% CI, 76.4–100%) |
||
Stage IV | 4-year RFS 0% 4-year OS 0% |
4-year RFS 16.7% 4-year OS 16.7% |
4-year EFS 32.1% (95% CI, 14.8–49.4%) 4-year OS 32.1% (95% CI, 14.8–49.4%) |
4-year EFS 41.7% (95% CI, 19.6–63.7%) 4-year OS 49.2% (95% CI, 27.5–71.0%) |
||
Favorable histology Wilms tumor with LOH of 1p and 16q | ||||||
NWTS-5; EE4A | NWTS-5; DD4A | AREN0532; DD4A | AREN0533; Regimen M | |||
Stage I-II | 4-year EFS 68.8% (95% CI, 55.2–82.3%) 4-year OS 91.6% (95% CI, 83.6–99.6%) |
NA | 4-year EFS 87.3% (95% CI, 75.1–99.5%) 4-year OS 100% |
NA | ||
Stage III-IV | NA | 4-year EFS 61.3% (95% CI, 44.9–77.6%) 4-year OS 86.0% (95% CI 90.5–100%) |
NA | 4-year EFS 90.2% (95% CI, 81.8–98.6%) 4-year OS 96.1% (95% CI, 90.5–100%) |
Relapsed favorable histology Wilms tumor | |||
---|---|---|---|
NWTS-2 and −3 (varied, see below) | NWTS-5; Stratum B/Regimen I | NWTS-5; Stratum C | |
2-drug pretreated | Stage I: 3-year OS 56.6% Stage II/III: 3-year OS 42% |
4-year EFS 71.1% 4-year OS 81.8% |
NA |
3-drug pretreated | Stage II/III: 3-year OS 26% Stage IV: 3-year OS 17.3% |
NA | 4-year EFS 42.3%* 4-year OS 48%* |
Note: adapted from Green, 19945, Daw, 20204, Gundy, 20057, Dix, 20198, Green, 200710, Malogolowkin, 200811
mainly included FHWT, but also included small portion of patients with focal anaplastic WT
RFS: relapse-free survival; OS: overall survival; EFS: event-free survival;
DD-RT: Vincristine, Actinomycin-D, Doxorubicin
Regimen J: Vincristine, Actinomycin-D, Doxorubicin, Cyclophosphamide
Regimen I: Vincristine, Doxorubicin, Cyclophosphamide alternating with Cyclophosphamide, Etoposide
Regimen UH-1: Cyclophosphamide, Carboplatin, Etoposide alternating with Vincristine, Doxorubicin, Cyclophosphamide
Regimen UH-2: Cyclophosphamide, Carboplatin, Etoposide alternating with Vincristine, Doxorubicin, Cyclophosphamide plus Vincristine, Irinotecan
EE4A: Vincristine, Actinomycin-D
DD4A: Vincristine, Actinomycin-D, Doxorubicin
Regimen M: Vincristine, Actinomycin-D, Doxorubicin alternating with Cyclophosphamide, Etoposide
NWTS-2/−3 relapse regimens: patients were retreated with different regimens, most commonly containing Vincristine, Actinomycin-D, Doxorubicin and Cyclophosphamide; Cisplatin and Etoposide were used occasionally
Stratum B/Regimen I: Vincristine, Doxorubicin, Cyclophosphamide alternating with Etoposide, Cyclophosphamide
Stratum C: Cyclophosphamide, Etoposide alternating with Carboplatin, Etoposide
TABLE 2.
SIOP 2001 | |||||
---|---|---|---|---|---|
Stage | Histology | N | |||
II/III | Blastemal-type | 153 | 5-year EFS 77% (95% CI 69–86%)* | 5-year OS 82% (95% CI 74–91%) | |
III | All high-risk histology | 141 | 2-year EFS 68% | 5-year OS 70% | |
IV | All high-risk histology | 75 | 2-year EFS 31% | 5-year OS 35% | |
IV | Blastemal-type | 34 | 5-year EFS 44% (95% CI 27–61) | 5-year OS 53% (95% CI 36–70%) | |
IV | Diffuse anaplastic | 40 | 5-year EFS 28% (95% CI 13–43%) | 5-year OS 29% (95% CI 13–45%) | |
Relapse | |||||
Initial stage | Histology | SIOP 93–01 | |||
I | Excluding blastemal-type and diffuse anaplastic | 33 | 5-year EFS 55% (95% CI 38–70) | 5-year OS 64% (95% CI 47–78) | |
Relapse | SIOP 2001 | ||||
I/II + III (no RT) | Excluding blastemal-type and diffuse anaplastic | 76 | 5-year EFS 83% (95% CI 73–90) | 5-year OS 88% (95% CI 79–94) | |
All stages | Relapse (all histology types) | 538 | NA | 5-Year OS 56% (95% CI 51–61%) |
WT subgroups that continue to have poor outcomes include a) newly diagnosed metastatic WT with post-chemotherapy blastemal-type and/or diffuse anaplastic histology, b) first relapse of WT after initially three or more prior systemic agents, and c) multiply relapsed WT. Survival for these patients is 50% at best3,4. Historical, current, and future approaches to managing these high-risk WT patients are the focus of this manuscript. Additionally, we note that this definition of high-risk is setting dependent. In low- and middle-income countries (LMIC), additional factors influenced by socio-economic status, including malnutrition, infections, shortage of drugs and delayed access to sufficient care may significantly contribute to treatment failure, thereby broadening the groups with OS estimates less than 50%.
High-risk Wilms tumor in the COG context
The National Wilms Tumor Study (NWTS) Group and successor COG approach to the treatment of high-risk WT including DAWT, favorable histology (FH) WT with LOH of 1p and 16q, and relapsed FHWT has evolved over the past 40 years with improvements in OS across all groups (Table 1). The NWTS-3 and 4 studies demonstrated increased OS with the addition of cyclophosphamide to vincristine, actinomycin-D and doxorubicin for stages II to IV DAWT5. NWTS-5 further improved OS with a regimen alternating vincristine, doxorubicin, and cyclophosphamide with cyclophosphamide and etoposide (Regimen I)6. AREN0321 added carboplatin for stage II-III DAWT patients employing the combinations of cyclophosphamide, carboplatin, and etoposide alternating with vincristine, doxorubicin, and cyclophosphamide (Regimen UH-1) as well as vincristine and irinotecan for stage IV DAWT (Regimen UH-2). The up-front vincristine/irinotecan combination revealed promising objective responses in 11 of 14 patients with metastatic DAWT4. Regimens UH-1/UH-2 led to an apparent improvement in outcomes for stages II-IV DAWT, albeit at the expense of greater toxicity compared to the historical Regimen I4. A revised regimen UH-1/UH-2 with lower cumulative doses of doxorubicin and cyclophosphamide to limit toxicity showed equivalent efficacy to the original AREN0321 regimens4.
The combination of LOH of chromosomes 1p and 16q in FHWT is an adverse prognostic factor and augmentation of therapy has benefitted this population (Table 1)7. Compared to the NWTS-5, the addition of doxorubicin to vincristine and actinomycin-D in COG study AREN0532 increased both 4-year EFS and OS in patients with stage I and II FHWT with LOH of 1p and 16q. For patients with stage III and IV FHWT with LOH of 1p and 16q, addition of cyclophosphamide/etoposide to vincristine, actinomycin-D, and doxorubicin on AREN0533 (Regimen M) likewise significantly improved 4-year EFS and OS8.
Outcomes for patients with relapsed FHWT who were treated on NWTS-2 or NWTS-3 were poor using non-standardized salvage therapy including actinomycin-D, vincristine, doxorubicin and cyclophosphamide with occasional cisplatin and etoposide (Table 1)9. NWTS-5 specified treatment recommendations for patients with WT who relapsed after initial therapy with 2- or 3-drug therapy respectively, and mainly included stages I-IV FHWT with a small subset of patients with anaplastic WT. For those who relapsed after 2-drug therapy, treatment recommendations were vincristine, doxorubicin and cyclophosphamide alternating with cyclophosphamide and etoposide (Stratum B/Regimen I) which led to a 4-year OS of 81.8%10. For those who relapsed after 3-drug therapy, treatment with alternating courses of cyclophosphamide/etoposide with carboplatin/etoposide (Stratum C) led to a 4-year OS of 48%11. Outcomes for both groups were substantially improved compared to NWTS-2 and NWTS-39. However, a significant limitation to Stratum C was hematologic toxicities11.
Based on the activity of vincristine/irinotecan on AREN0321, the current COG AREN1921 trial is assessing the benefit and harms of vincristine/irinotecan in addition to the Regimen UH-1/2 for stage II-IV DAWT (new regimen, UH-3). AREN1921 also includes patients with relapsed FHWT: those treated initially with 2-drug therapy receive Regimen UH-3, and those treated initially with three or more drugs receive ifosfamide/carboplatin/etoposide alternating with cyclophosphamide/topotecan. The rationale for using topotecan is that in a phase II study 13 of 36 relapsed WT demonstrated an objective response on topotecan monotherapy12 and activity of topotecan in combination with cyclophosphamide has been observed13.
High-risk Wilms tumor in the SIOP context
Using the SIOP approach, most renal tumors in patients aged ≥ 6 months are treated with pre-operative chemotherapy (vincristine and actinomycin-D for localized and additional doxorubicin for metastatic disease)14. Tumor histology and stage after surgery dictate risk classification. In the SIOP 6 trial, response to preoperative chemotherapy was identified as an important stratification parameter, and the SIOP 93–01 study showed inferior outcomes for patients with blastemal-type tumors (5-year EFS 67%)15. Therefore, SIOP regards blastemal-type tumors as high-risk histology, similar to DAWT. The SIOP 2001 protocol was the first study to increase therapy for blastemal-type histology and that study improved EFS for patients with stage I-III (and OS for stage I) compared to the historical 93–01 study15. However, 5-year OS for stage IV WT with high-risk histology was disappointingly low despite increased therapy (blastemal-type 53%, DAWT 29%, Table 2)14. For patients with stage III and IV tumors with high-risk histology, the SIOP-RTSG 2016 UMBRELLA protocol recommends cyclophosphamide/doxorubicin alternating with etoposide/carboplatin for 34 weeks (HR-1) and higher doses of local flank radiotherapy (RT) (25.2 Gy, with or without 10.8 Gy boost to remaining tumor tissue), with additive lung RT (15 Gy) for lung metastases. Given the very poor outcomes, patients with stage IV blastemal-type or DAWT have alternative treatment options such as following the COG approach with a more intensive irinotecan-based regimen or considering consolidation with high-dose melphalan with autologous hematopoietic stem cell transplant (HSCT), but this is an individualized decision4,16.
Similar to the COG experience, in SIOP standardized treatment of relapse has improved outcome significantly for WT that relapsed after only two drugs up-front. In the SIOP 93–01 study, 5-year OS was 64% compared to 88% in the SIOP 2001 for this group1727. In the SIOP-RTSG 2016 UMBRELLA protocol, a risk-stratified approach is integrated in the standard of care registration study.
Relapsed WT in the SIOP context is now classified into three risk groups (AA, BB, CC), analogous to COG (Table 3) and primarily based upon the up-front treatment, as this was a strong prognostic factor in retrospective studies15,18. Group AA includes patients who relapse after treatment with only vincristine and actinomycin-D (standard risk, post-relapse survival rate about 80%) and are treated with alternating cyclophosphamide/doxorubicin and etoposide/carboplatin (similar to HR-1)10. Group BB includes patients who relapse after at least three drugs including doxorubicin (high-risk, survival rate about 40–50%)11 and are treated with four cycles of carboplatin, etoposide, with alternating additional either cyclophosphamide or ifosfamide followed by high dose chemotherapy (HDT) with melphalan and autologous HSCT to consolidate previous chemotherapy response3,14. Group CC includes patients who relapse with initial high-risk histology (advanced-stage DAWT or blastemal-type tumors), or multiple relapses of any histology type, which all have a dismal prognosis (very high-risk, survival rate about 10%)19–23. For CC patients, the UMBRELLA protocol encourages administration of a camptothecin-containing regimen such as vincristine/irinotecan (VI), vincristine/irinotecan/temozolomide (VIT) or topotecan/temozolomide because they usually are naïve to these agents in the context of SIOP protocols. The rationale for this is based on a few relapsed cases that demonstrated objective responses, however outcomes data for these regimens are still limited4,24. Additionally, the UMBRELLA protocol endorses initiatives dedicated to performing thorough molecular analyses collaboratively with national or international precision medicine programs, using organoids or xenografts, and the potential enrollment onto relevant early phase clinical trials25.
TABLE 3.
COG definition (COG-RTC AREN1921) | SIOP definition (SIOP-RTSG 2016 UMBRELLA) | |||
---|---|---|---|---|
Standard risk relapse | Initial therapy with two chemotherapy agents; generally vincristine and actinomycin-D | AA | Relapse after treatment with vincristine and actinomycin-D | |
High risk relapse | Initial therapy with three chemotherapy agents; primarly vincristine, actinomycin-D and doxorubicin OR vincristine, actinomycin-D and irinotecan | BB | Relapse after treatment with at least three drugs including doxorubicin | |
Very high risk relapse | Initial therapy with four or more chemotherapy agents.* | CC | Relapse with initial high-risk histology (advanced-stage diffuse anaplasia or blastemal-type tumors) |
COG AREN1921 includes patients with very high-risk FHWT relapses; patients with relapsed anaplastic histology WT are also considered in a very high-risk category but are not eligible for the treatment regimens proposed because there is too much overlap with up-front therapy.
Local control measures for high-risk Wilms tumor
Surgery and RT have well-established roles in the treatment of newly diagnosed high-risk WT. While surgical approaches and pulmonary RT doses are generally similar between high-risk WT and non-high-risk WT, abdominal RT is often administered at augmented doses in high-risk cases. For example, in the current COG approach, patients with stage III favorable histology WT requiring flank radiation are given 1080 cGy whereas those with stage III DAWT receive 1980 cGy.
For relapsed WT, while surgery and RT with dosing similar to that used in the up-front setting are widely used, there has been limited evidence on how and when to perform local control17,18. There is a consensus that patients with relapsed WT who show at least a minimal response to induction chemotherapy should have surgical resection of the recurrent tumor(s), followed by RT to all sites of disease11,26,27. Surgical resection of relapsed disease in a chemo-responsive disease setting seems to be associated with improved survival20,27. Dome et al. showed that patients with complete surgical resection of relapsed disease had a higher probability of survival than patients who had partial resection or no resection20. Similarly, the administration of RT in patients with relapsed WT who were not previously irradiated was associated with improved survival20,28. The SIOP UMBRELLA and COG 1921 studies aim to collect more data on local control of relapsed WT.
Role of high-dose therapy and hematopoietic stem cell transplant
A clear role of HDT followed by HSCT has not been definitively established in either the relapsed or upfront setting in high-risk WT. The available evidence is limited by small case numbers, selection bias and lack of adequate control arms. Ha et al. reviewed and meta-analyzed20 non-randomized studies that overall included 1,226 patients with relapsed WT, treated with or without HDT3. Within the caveats of such an analysis, the investigators demonstrated a potential but not statistically significant EFS benefit in patients treated with HDT with high-risk relapse (HR = 0.90, 95% CI 0.62–1.31) and significant advantage for patients with very high-risk relapse (HR = 0.50, CI 0.31–0.82), but not for lower-risk patients initially treated with only two drugs. Malogolowkin et al. reviewed 253 patients with relapsed WT who underwent HDT in the Center for International Blood and Marrow Transplantation Research database. The 5-year EFS and OS rates were 36% and 45% respectively, comparable to salvage regimens using standard dose chemotherapy29. Others have attempted to evaluate the efficacy of HDT as part of upfront therapy in addition to relapse setting. Spreafico et al. reviewed 69 patients with relapsed WT who received HDT after achieving first or subsequent remission in the European Blood and Marrow Transplantation Registry and revealed a 5-year EFS and OS of 63% and 67%, respectively30. The authors provided initial data to further explore the benefit of HDT as frontline consolidation in high-risk patients (DAWT or blastemal-type metastatic cases). The limited data seems to support the possibility that HDT may overcome the intrinsic resistance to cytotoxic chemotherapy inherent to TP53 mutations observed in anaplastic WT. In summary, the evidence for use of HDT in patients with high-risk WT is inconclusive. Although randomized trials would be ideal, such a trial even through international cooperation is unlikely given the small patient numbers. The currently open SIOP UMBRELLA protocol will study the use of HDT with melphalan in some patients with relapsed WT that is responsive to re-induction chemotherapy14, or as an option for consolidation therapy in patients with initially metastatic tumors with high-risk histology.
Development of novel agents for Wilms tumor
Current treatment regimens with conventional cytotoxic therapies are reaching the limit of tolerated drug doses 1,3,4,14,18. This is the case even for non-relapsed WT patients, where regimens UH-1 and UH-2 ultimately had to be dose reduced due to unacceptably high toxicity4. Accordingly, with a diminishing therapeutic window for further augmentation of conventional chemotherapy, there is a need for identification of agents with different mechanisms of action to improve survival and minimize adverse effects for patients with high-risk WT25.
Beyond the established effective systemic agents, taxanes and vascular endothelial growth factor receptor (VEGFR)-directed kinase inhibitors represent the next most common classes of systemic agents used in the treatment high-risk WT patients. Paclitaxel given as a 24-hour continuous intravenous infusion on POG9262 revealed single agent activity in a minority of patients with relapsed WT31. Case reports have described single agent activity of paclitaxel as well as in combination with platinum chemotherapies32–34. Bevacizumab, a monoclonal antibody directed against VEGFR has shown activity when combined with irinotecan, vincristine, and temozolomide in multiply relapsed WT24,35. However, outside of this combination, the best responses to monotherapy or combinations including bevacizumab have been stable disease36–38. The multi-kinase inhibitors sorafenib and cabozantinib have shown only minimal activity in high-risk WT. Stable disease was the best response observed with sorafenib both in monotherapy and combination39. Cabozantinib responses were limited to prolonged stable disease in the phase I and a partial response lasting nearly two years in a case report but no responses were observed in the phase II study setting40–42. When used to treat high-risk WT, taxanes and VEGFR/multi-directed kinase inhibitors are generally limited to palliation of patients with multiply relapsed disease who are not eligible for therapeutic clinical trials.
Since conventional therapies are inadequate to cure many patients with high-risk WT, such patients may be more promptly directed onto early phase clinical trials. Historically, early phase clinical trials were predominantly tumor type agnostic and have not included sufficient numbers of patients with WT to definitively assess activity. Two recent reviews identified 257 WT patients across 79 early phase trials from 2000–2020 where patients with predominantly relapsed, occasionally refractory, disease were enrolled. Only nine of these trials had enrolled 10 or more WT patients (ATRA/IFN-α2A, Irinotecan, Topotecan, rTNFα/actinomycin-D, Ixabepilone, Cixutumumab, Sorafenib, Alisertib, Atezolizumab)25,39. Excluding studies involving irinotecan, topotecan, or actinomycin-D, there were only three patients with WT enrolled onto these studies with objective responses25,39. As such, our collective experience in leveraging novel agents in the treatment of relapsed or refractory WT is limited and generally underwhelming.
Current investigations of targeted and immune-based therapies for high-risk WT attempt to exploit established specific WT vulnerabilities. Given the dependency of WT on canonical Wnt-Beta-catenin signaling, NCT04851119 trial (PEPN2011) is investigating the utility of TBL1 inhibitor Tegavivint43. Surface proteins WT144 and GPC345 are potential therapeutic immune targets in WT and are currently being explored in immunotherapy studies NCT02789228/NCT05238792 and NCT04928677, respectively. DS-8201a, a HER2 antibody conjugated to a topoisomerase 1 payload, and Selinexor46–48, an inhibitor of the nuclear pore XPO1, are two agents with promising laboratory data which are undergoing clinical trials in other pediatric solid tumors and thus may be amenable to clinical investigations in WT. The heterogeneous genomic landscape of WT makes it challenging to identify selective inhibitors that are effective across all high-risk WT cases however therapeutic vulnerabilities have been identified that could benefit particular subsets of patients. For example, CDK9 inhibitors in MLL1/ENL mutant tumors49, BRD4 inhibitors in MYCN driven tumors50, as well as WT with specific DNA damage response defects such as deleterious mutations in ATM via the ATR inhibitor Elimusertib on NCT05071209 (PEPN2112).
Clinical studies of novel agents for high-risk WT are advanced in large part based upon WT-specific preclinical data. This has been challenged by limited robust WT model systems as WT cell lines and mouse models have failed to capture the profound phenotypic and genetic heterogeneity of these tumors. Only a small number of cell lines have been described in the literature, such as the Wit4951 and 17.9452 cell lines representing high-risk anaplastic disease and, most recently, a small series of WT1-mutant WT cell cultures53. Wegert et al. propagated WT spheroid cell cultures, providing three-dimensional (3D) in vitro models that can even recapitulate the difficult-to-culture blastemal WT cells54. A limited number of Genetically Engineered Mouse Models (GEMMs) have been developed by exploiting mutations observed in human WT such as WT1 loss and IGF2 activation55, or LIN28 overexpression56. Patient-derived xenograft (PDX) models of WT have developed rather well, with groups reporting high rates of WT engraftment compared to other tumor types57. Notably, kidney capsule implantation protocols have been well developed, greatly facilitating the use of anatomically appropriate orthotopic PDX WT models. Finally, a relatively new model system for studying WT is the use of organoid technology, which can be derived with high efficiency from WT and expands rapidly25,58. With these more efficient model designs, future studies could potentially assess in real time the best treatment for a specific patient, but now there is a dearth of sufficiently promising therapeutic approaches.
High-risk Wilms tumor in low and low-middle income countries
Although the aforementioned laboratory investigations and early phase clinical trials are attempting to improve survival in patients with high-risk WT in high income countries (HIC), the challenges and strategies to overcome poor survival for WT patients in LMIC are inherently different. Successful treatment of patients with WT in this context requires an integrated multidisciplinary approach involving imaging, surgery, pathology, and RT services59. In this view, the definition of high-risk tumors in LMIC is largely influenced by non-clinical factors limiting timely access to integrated - when available - care (Table 4). Compared to HIC, patients with WT in LMIC are diagnosed later, with higher tumor volume and stage60 and an older age61–63. Malnutrition and poor clinical conditions due to advanced illness are common64 and favor a higher incidence of severe treatment-related toxicities and deaths61,65–67. The combination of poor clinical status at time of diagnosis, shortage of essential medicines, high cost of treatment and transportation resulting in treatment abandonment or refusal65,68–70, low treatment compliance, and utilization of inadequately intensive treatment including omission of RT negatively impact survival62,68.
TABLE 4:
Characteristic | Sub-Saharan Africa* 65 | AHOPCA** 70 |
---|---|---|
Year (s) | 2014–2018 | 2012–2015 |
No. Patients. | 201 | 182 |
Age (median) | 3.6 y | 3.5 y |
Diagnostic approach | Clinical, abdominal US, chest x-ray | Abdominal/chest CT if available; otherwise, clinical, abdominal US, chest x-ray |
Tumor volume | Median Size: 14 cm | Median Volume: 579 cc |
% Advanced disease | Stage IV: 62 (31%) | Stage III: 116 (63%) Stage IV: 37 (20%) |
Radiotherapy | Available in Ghana but not in Malawi or Cameroon. | Available, with late delivery |
Chemotherapy (drugs used) | SIOP-Adapted. (VAD) | COG-Adapted. (VAD and CE) |
Abandonment | 24/201 (12%) | 19/182 (10%) |
Deaths (First event) | 30/201 (15%) | 5/182 (3%) |
Survival | 49% | 68% |
Sub-Saharan Africa: Includes centers from Malawi (1), Cameroon (3), and Ghana (2).
AHOPCA: Includes centers from Guatemala (1), El Salvador (1), Honduras (2), Nicaragua (1), and Dominican Republic (1).
VAD: Vincristine + actinomycin D +/− Doxorubicin.
CE: Cyclophosphamide and Etoposide (Note: intensified for high-risk cases).
LMICs report a higher proportion of patients with anaplasia and advanced disease, which correlate with poor prognosis66,68,71,72. However, the prevalence of high-risk factors may be underestimated in LMICs due to difficult access to standardized diagnostic studies like CT scans, which reduce the accuracy of staging and surgical planning73. There also is limited training of pathologists to recognize anaplasia71, correctly define local stage, and to evaluate chemotherapy-induced changes in pre-treated tumors67,68,74. The lack of referral centers with high surgical expertise correlates with a higher incidence of tumor rupture and suboptimal surgical staging72,75 The limited access to supportive care, RT, and certain chemotherapy medications (i.e., carboplatin, alkylating agents) limit the ability to intensify therapy in high-risk tumors66,76,77. The combination of underdiagnosis of metastatic disease, later detection of tumors, and lack of central pathology review could explain the lower survival for middle income countries (MIC) compared with HIC, as was seen in the international comparison of outcomes in the SIOP WT 2001 trial for the Brazilian group67. We also need to acknowledge that the lack of cancer registries with all information limits the capacity of LMIC to determine the actual incidence of high-risk WT.
Local research initiatives to study and validate adverse prognostic indicators specific to LMICs are expected to help better stratify patients according to realistic cure estimates and administer more reasonably deliverable adapted therapy regimens. The primary interventions that could minimize the impact of high-risk non-clinical factors that reduce the survival of WT in LMIC are (1) universal coverage to avoid late diagnosis, abandonment, and poor compliance with therapy78, (2) ensure access to standard diagnostic procedures, supportive therapy, and essential medicines, and (3) development of twinning programs (HIC-LMIC) to train the multidisciplinary team and standardize the approach to perform accurate diagnosis, surgical planning, and risk-stratify postoperative therapy61,64.
The parent and patient advocate perspective
Recent years have seen increased patient/family and advocate involvement in the research process, leading to faster clinical translation, improvement in the transparency of research, and enhanced trust and rapport between all stakeholders79–81. Despite strong curative intent, aggressive and lengthy treatment strategies for high-risk WT have so far demonstrated only partial success and can leave survivors to deal with life-long sequelae. Recently, patients, families, advocates, and medical teams have pointed out the need for more-effective and less-toxic treatments for children with high-risk WT25,63. Inclusive stakeholder involvement in the design and implementation of new research/protocols and clinical trials allows for improved therapeutic strategies and ultimately, safer and more-efficacious treatments for children with high-risk WT.
Conclusion
Iterative prospective clinical trials of progressively augmented therapies have systematically improved survival in the vast majority of WT patients and narrowed our definition of high-risk WT. Nonetheless, survival is less than 50% in patients with newly diagnosed metastatic blastemal-type and/or DAWT as well as relapsed WT patients excluding those treated with only two drugs in the up-front setting. Such cases of high-risk WT remain a challenge and focused efforts, both preclinically and clinically, are needed to establish better treatment approaches.
ACKNOWLEDGMENTS
Michael Ortiz acknowledges active support from Team Walk with Eloise, Kids Walk for MSK Kids led by the Freiberger Family; the Serra Family and the Bianco Family Foundation; Cannonball Kids’ cancer; Cookies for Kids’ Cancer; the Starr Cancer Consortium; Conquer Cancer, the ASCO Foundation; and the National Cancer Institute (NCI) of the National Institutes of Health (NIH) via U01 CA263967 and P30 CA008748. Filippo Spreafico acknowledges constant support from the Parents’ Association “Associazione Bianca Garavaglia Onlus”. Jeffrey Dome acknowledges support from the NCI (grants U01 CA180886 and R01CA219013).
Abbreviations Key
- COG
Children’s Oncology Group
- DAWT
diffuse anaplastic Wilms tumor
- EFS
event free survival
- FH
favorable histology
- GEMM
genetically engineered mouse models
- HDT
high-dose chemotherapy
- HIC
high income countries
- HSCT
hematopoietic stem cell transplant
- LMIC
low and middle income countries
- LOH
loss of heterozygosity
- MIC
middle income countries
- NWTS
National Wilms Tumor Study
- OS
overall survival
- PDX
patient-derived xenograft
- RT
radiotherapy
- RTC
Renal Tumor Committee
- RTSG
Renal Tumor Study Group
- SIOP
Société Internationale D’oncologie Pédiatrique
- VEGFR
vascular endothelial growth factor receptor
- WT
Wilms tumor
Footnotes
DATA SHARING AND DATA ACCESSIBILITY
Not applicable
CONFLICT OF INTEREST STATEMENT
Michael Ortiz has previously served as an expert advisor for Guidepoint Global and has received research funds from Amgen, none of which is relevant to anything discussed in this manuscript. Amy Armstrong has previously served as a consultant for Springworks and EM Partners, none of which is relevant to the content of this manuscript. All other authors declare no competing interests.
REFERENCES
- 1.Dome JS, Graf N, Geller JI, et al. Advances in Wilms Tumor Treatment and Biology: Progress Through International Collaboration. J Clin Oncol. Sep 20 2015;33(27):2999–3007. doi: 10.1200/jco.2015.62.1888 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Dome JS, Mullen EA, Dix DB, et al. Impact of the First Generation of Children’s Oncology Group Clinical Trials on Clinical Practice for Wilms Tumor. J Natl Compr Canc Netw. Aug 1 2021;19(8):978–985. doi: 10.6004/jnccn.2021.7070 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ha TC, Spreafico F, Graf N, et al. An international strategy to determine the role of high dose therapy in recurrent Wilms’ tumour. Eur J Cancer. Jan 2013;49(1):194–210. doi: 10.1016/j.ejca.2012.07.010 [DOI] [PubMed] [Google Scholar]
- 4.Daw NC, Chi YY, Kalapurakal JA, et al. Activity of Vincristine and Irinotecan in Diffuse Anaplastic Wilms Tumor and Therapy Outcomes of Stage II to IV Disease: Results of the Children’s Oncology Group AREN0321 Study. J Clin Oncol. May 10 2020;38(14):1558–1568. doi: 10.1200/JCO.19.01265 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Green DM, Beckwith JB, Breslow NE, et al. Treatment of children with stages II to IV anaplastic Wilms’ tumor: a report from the National Wilms’ Tumor Study Group. J Clin Oncol. Oct 1994;12(10):2126–31. [DOI] [PubMed] [Google Scholar]
- 6.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. May 20 2006;24(15):2352–8. doi: 10.1200/jco.2005.04.7852 [DOI] [PubMed] [Google Scholar]
- 7.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. Oct 10 2005;23(29):7312–21. doi: 10.1200/JCO.2005.01.2799 [DOI] [PubMed] [Google Scholar]
- 8.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. Oct 20 2019;37(30):2769–2777. doi: 10.1200/JCO.18.01972 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Grundy P, Breslow N, Green DM, Sharples K, Evans A, D’Angio GJ. Prognostic factors for children with recurrent Wilms’ tumor: results from the Second and Third National Wilms’ Tumor Study. J Clin Oncol. May 1989;7(5):638–47. doi: 10.1200/JCO.1989.7.5.638 [DOI] [PubMed] [Google Scholar]
- 10.Green DM, Cotton CA, Malogolowkin M, et al. Treatment of Wilms tumor relapsing after initial treatment with vincristine and actinomycin D: a report from the National Wilms Tumor Study Group. Pediatr Blood Cancer. May 2007;48(5):493–9. doi: 10.1002/pbc.20822 [DOI] [PubMed] [Google Scholar]
- 11.Malogolowkin M, Cotton CA, Green DM, et al. Treatment of Wilms tumor relapsing after initial treatment with vincristine, actinomycin D, and doxorubicin. A report from the National Wilms Tumor Study Group. Pediatr Blood Cancer. Feb 2008;50(2):236–41. doi: 10.1002/pbc.21267 [DOI] [PubMed] [Google Scholar]
- 12.Metzger ML, Stewart CF, Freeman BB 3rd, et al. Topotecan is active against Wilms’ tumor: results of a multi-institutional phase II study. J Clin Oncol. Jul 20 2007;25(21):3130–6. doi: 10.1200/JCO.2007.10.9298 [DOI] [PubMed] [Google Scholar]
- 13.Saylors RL 3rd, Stewart CF, Zamboni WC, et al. Phase I study of topotecan in combination with cyclophosphamide in pediatric patients with malignant solid tumors: a Pediatric Oncology Group Study. J Clin Oncol. Mar 1998;16(3):945–52. doi: 10.1200/jco.1998.16.3.945 [DOI] [PubMed] [Google Scholar]
- 14.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. Dec 2017;14(12):743–752. doi: 10.1038/nrurol.2017.163 [DOI] [PubMed] [Google Scholar]
- 15.van den Heuvel-Eibrink MM, van Tinteren H, Bergeron C, et al. Outcome of localised blastemal-type Wilms tumour patients treated according to intensified treatment in the SIOP WT 2001 protocol, a report of the SIOP Renal Tumour Study Group (SIOP-RTSG). Eur J Cancer. Mar 2015;51(4):498–506. doi: 10.1016/j.ejca.2014.12.011 [DOI] [PubMed] [Google Scholar]
- 16.Pasqualini C, Furtwangler R, van Tinteren H, et al. Outcome of patients with stage IV high-risk Wilms tumour treated according to the SIOP2001 protocol: A report of the SIOP Renal Tumour Study Group. Eur J Cancer. Mar 2020;128:38–46. doi: 10.1016/j.ejca.2020.01.001 [DOI] [PubMed] [Google Scholar]
- 17.Groenendijk A, van Tinteren H, Jiang Y, et al. Outcome of SIOP patients with low- or intermediate-risk Wilms tumour relapsing after initial vincristine and actinomycin-D therapy only - the SIOP 93–01 and 2001 protocols. Eur J Cancer. Mar 2022;163:88–97. doi: 10.1016/j.ejca.2021.12.014 [DOI] [PubMed] [Google Scholar]
- 18.Spreafico F, Pritchard Jones K, Malogolowkin MH, et al. Treatment of relapsed Wilms tumors: lessons learned. Expert Rev Anticancer Ther. Dec 2009;9(12):1807–15. doi: 10.1586/era.09.159 [doi] [DOI] [PubMed] [Google Scholar]
- 19.Reinhard H, Schmidt A, Furtwangler R, et al. Outcome of relapses of nephroblastoma in patients registered in the SIOP/GPOH trials and studies. Oncol Rep. Aug 2008;20(2):463–7. [PubMed] [Google Scholar]
- 20.Dome JS, Liu T, Krasin M, et al. Improved survival for patients with recurrent Wilms tumor: the experience at St. Jude Children’s Research Hospital. J Pediatr Hematol Oncol. Mar-Apr 2002;24(3):192–8. doi: 10.1097/00043426-200203000-00007 [DOI] [PubMed] [Google Scholar]
- 21.Tournade MF, Lemerle J, Brunat-Mentigny M, et al. Ifosfamide is an active drug in Wilms’ tumor: a phase II study conducted by the French Society of Pediatric Oncology. J Clin Oncol. May 1988;6(5):793–6. doi: 10.1200/JCO.1988.6.5.793 [DOI] [PubMed] [Google Scholar]
- 22.Pein F, Pinkerton R, Tournade MF, et al. Etoposide in relapsed or refractory Wilms’ tumor: a phase II study by the French Society of Pediatric Oncology and the United Kingdom Children’s Cancer Study Group. J Clin Oncol. Aug 1993;11(8):1478–81. doi: 10.1200/JCO.1993.11.8.1478 [DOI] [PubMed] [Google Scholar]
- 23.de Camargo B, Melaragno R, Saba e Silva N, et al. Phase II study of carboplatin as a single drug for relapsed Wilms’ tumor: experience of the Brazilian Wilms’ Tumor Study Group. Med Pediatr Oncol. 1994;22(4):258–60. doi: 10.1002/mpo.2950220409 [DOI] [PubMed] [Google Scholar]
- 24.Venkatramani R, Malogolowkin MH, Mascarenhas L. Treatment of multiply relapsed wilms tumor with vincristine, irinotecan, temozolomide and bevacizumab. Pediatr Blood Cancer. Apr 2014;61(4):756–9. doi: 10.1002/pbc.24785 [DOI] [PubMed] [Google Scholar]
- 25.Brok J, Mavinkurve-Groothuis AMC, Drost J, et al. Unmet needs for relapsed or refractory Wilms tumour: Mapping the molecular features, exploring organoids and designing early phase trials - A collaborative SIOP-RTSG, COG and ITCC session at the first SIOPE meeting. Eur J Cancer. Feb 2021;144:113–122. doi: 10.1016/j.ejca.2020.11.012 [DOI] [PubMed] [Google Scholar]
- 26.Gooskens SL, Graf N, Furtwangler R, et al. Position paper: Rationale for the treatment of children with CCSK in the UMBRELLA SIOP-RTSG 2016 protocol. Nat Rev Urol. May 2018;15(5):309–319. doi: 10.1038/nrurol.2018.14 [DOI] [PubMed] [Google Scholar]
- 27.Spreafico F, Bisogno G, Collini P, et al. Treatment of high-risk relapsed Wilms tumor with dose-intensive chemotherapy, marrow-ablative chemotherapy, and autologous hematopoietic stem cell support: experience by the Italian Association of Pediatric Hematology and Oncology. Pediatr Blood Cancer. Jul 2008;51(1):23–8. doi: 10.1002/pbc.21524 [DOI] [PubMed] [Google Scholar]
- 28.Paulino AC. Relapsed Wilms tumor: is there a role for radiation therapy? Am J Clin Oncol. Aug 2001;24(4):408–13. doi: 10.1097/00000421-200108000-00022 [DOI] [PubMed] [Google Scholar]
- 29.Malogolowkin MH, Hemmer MT, Le-Rademacher J, et al. Outcomes following autologous hematopoietic stem cell transplant for patients with relapsed Wilms’ tumor: a CIBMTR retrospective analysis. Bone Marrow Transplant. Nov 2017;52(11):1549–1555. doi: 10.1038/bmt.2017.178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Spreafico F, Dalissier A, Potschger U, et al. High dose chemotherapy and autologous hematopoietic cell transplantation for Wilms tumor: a study of the European Society for Blood and Marrow Transplantation. Bone Marrow Transplant. Feb 2020;55(2):376–383. doi: 10.1038/s41409-019-0661-7 [DOI] [PubMed] [Google Scholar]
- 31.Harris MBHCSJ, et al. Taxol in pediatric solid tumors: a Pediatric Oncology Group (POG) phase II study. Proc Annu Meet Am Soc Clin Oncol. 1999.;18:2170a. [Google Scholar]
- 32.Italiano A, Sirvent N, Michiels JF, Peyrade F, Otto J, Thyss A. Tumour response to paclitaxel in an adult with relapsed nephroblastoma. Lancet Oncol. Apr 2005;6(4):252–3. doi: 10.1016/S1470-2045(05)70098-X [DOI] [PubMed] [Google Scholar]
- 33.Ramanathan RK, Rubin JT, Ohori NP, Belani CP. Dramatic response of adult wilms tumor to paclitaxel and cisplatin. Med Pediatr Oncol. Apr 2000;34(4):296–8. doi: [DOI] [PubMed] [Google Scholar]
- 34.Ozaki S, Takigawa N, Ichihara E, et al. Favorable response of heavily treated Wilms’ tumor to paclitaxel and carboplatin. Onkologie. 2012;35(5):283–6. doi: 10.1159/000338532 [DOI] [PubMed] [Google Scholar]
- 35.Venkatramani R, Malogolowkin M, Davidson TB, May W, Sposto R, Mascarenhas L. A phase I study of vincristine, irinotecan, temozolomide and bevacizumab (vitb) in pediatric patients with relapsed solid tumors. PLoS One. 2013;8(7):e68416. doi: 10.1371/journal.pone.0068416 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Benesch M, Windelberg M, Sauseng W, et al. Compassionate use of bevacizumab (Avastin) in children and young adults with refractory or recurrent solid tumors. Ann Oncol. Apr 2008;19(4):807–13. doi: 10.1093/annonc/mdm510 [DOI] [PubMed] [Google Scholar]
- 37.Glade Bender JL, Adamson PC, Reid JM, et al. Phase I trial and pharmacokinetic study of bevacizumab in pediatric patients with refractory solid tumors: a Children’s Oncology Group Study. J Clin Oncol. Jan 20 2008;26(3):399–405. doi: 10.1200/JCO.2007.11.9230 [DOI] [PubMed] [Google Scholar]
- 38.Navid F, Baker SD, McCarville MB, et al. Phase I and clinical pharmacology study of bevacizumab, sorafenib, and low-dose cyclophosphamide in children and young adults with refractory/recurrent solid tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. Jan 1 2013;19(1):236–46. doi: 10.1158/1078-0432.CCR-12-1897 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Brok J, Pritchard-Jones K, Geller JI, Spreafico F. Review of phase I and II trials for Wilms’ tumour - Can we optimise the search for novel agents? Eur J Cancer. Jul 2017;79:205–213. doi: 10.1016/j.ejca.2017.04.005 [DOI] [PubMed] [Google Scholar]
- 40.Chuk MK, Widemann BC, Minard CG, et al. A phase 1 study of cabozantinib in children and adolescents with recurrent or refractory solid tumors, including CNS tumors: Trial ADVL1211, a report from the Children’s Oncology Group. Pediatr Blood Cancer. Aug 2018;65(8):e27077. doi: 10.1002/pbc.27077 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Anderson B, Jasty-Rao R, Wu YM, Paul T, Robinson D, Mody RJ. Exceptional Response to Cabozantinib in a Patient With Multiply Relapsed Wilms Tumor. JCO Precis Oncol. 2018;2doi: 10.1200/po.18.00021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Akshintala S, Widemann BC, Barkauskas DA, et al. Phase 2 trial of cabozantinib in children and young adults with refractory sarcomas, Wilms tumor, and rare tumors: Children’s Oncology Group Study (ADVL1622). Journal of Clinical Oncology. 2021;39(15_suppl):10010–10010. doi: 10.1200/JCO.2021.39.15_suppl.10010 [DOI] [Google Scholar]
- 43.Nomura M, Rainusso N, Lee YC, et al. Tegavivint and the beta-Catenin/ALDH Axis in Chemotherapy-Resistant and Metastatic Osteosarcoma. J Natl Cancer Inst. Nov 1 2019;111(11):1216–1227. doi: 10.1093/jnci/djz026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hont AB, Cruz CR, Ulrey R, et al. Immunotherapy of Relapsed and Refractory Solid Tumors With Ex Vivo Expanded Multi-Tumor Associated Antigen Specific Cytotoxic T Lymphocytes: A Phase I Study. J Clin Oncol. Sep 10 2019;37(26):2349–2359. doi: 10.1200/JCO.19.00177 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ortiz MV, Roberts SS, Glade Bender J, Shukla N, Wexler LH. Immunotherapeutic Targeting of GPC3 in Pediatric Solid Embryonal Tumors. Frontiers in oncology. 2019;9:108. doi: 10.3389/fonc.2019.00108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ortiz MV, Siddiquee A, You D, et al. Preclinical evaluation of XPO1 inhibition in Wilms tumors. Journal of Clinical Oncology. 2020;38(15_suppl):3580–3580. doi: 10.1200/JCO.2020.38.15_suppl.3580 [DOI] [Google Scholar]
- 47.Mittal K, Lee BP, Cooper GW, et al. Targeting TRIP13 in Wilms Tumor with Nuclear Export Inhibitors. bioRxiv. 2022:2022.02.23.481521. doi: 10.1101/2022.02.23.481521 [DOI] [Google Scholar]
- 48.Attiyeh EF, Maris JM, Lock R, et al. Pharmacodynamic and genomic markers associated with response to the XPO1/CRM1 inhibitor selinexor (KPT-330): A report from the pediatric preclinical testing program. Pediatr Blood Cancer. Feb 2016;63(2):276–86. doi: 10.1002/pbc.25727 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Wan L, Chong S, Xuan F, et al. Impaired cell fate through gain-of-function mutations in a chromatin reader. Nature. Jan 2020;577(7788):121–126. doi: 10.1038/s41586-019-1842-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Woods AD, Berlow NE, Ortiz MV, et al. Bromodomain 4 inhibition leads to MYCN downregulation in Wilms tumor. Pediatr Blood Cancer. Feb 2022;69(2):e29401. doi: 10.1002/pbc.29401 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Alami J, Williams BR, Yeger H. Derivation and characterization of a Wilms’ tumour cell line, WiT 49. Int J Cancer. Nov 10 2003;107(3):365–74. doi: 10.1002/ijc.11429 [DOI] [PubMed] [Google Scholar]
- 52.Brown KW, Charles A, Dallosso A, et al. Characterization of 17.94, a novel anaplastic Wilms’ tumor cell line. Cancer Genet. Jun 2012;205(6):319–26. doi: 10.1016/j.cancergen.2012.04.009 [DOI] [PubMed] [Google Scholar]
- 53.Royer-Pokora B, Busch MA, Tenbusch S, et al. Comprehensive Biology and Genetics Compendium of Wilms Tumor Cell Lines with Different WT1 Mutations. Cancers. Dec 28 2020;13(1)doi: 10.3390/cancers13010060 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wegert J, Zauter L, Appenzeller S, et al. High-risk blastemal Wilms tumor can be modeled by 3D spheroid cultures in vitro. Oncogene. Jan 2020;39(4):849–861. doi: 10.1038/s41388-019-1027-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hu Q, Gao F, Tian W, et al. Wt1 ablation and Igf2 upregulation in mice result in Wilms tumors with elevated ERK1/2 phosphorylation. J Clin Invest. Jan 2011;121(1):174–83. doi: 10.1172/jci43772 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Atala A. Re: Lin28 sustains early renal progenitors and induces Wilms tumor. The Journal of urology. Feb 2015;193(2):730–1. doi: 10.1016/j.juro.2014.11.021 [DOI] [PubMed] [Google Scholar]
- 57.Murphy AJ, Chen X, Pinto EM, et al. Forty-five patient-derived xenografts capture the clinical and biological heterogeneity of Wilms tumor. Nat Commun. Dec 20 2019;10(1):5806. doi: 10.1038/s41467-019-13646-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Calandrini C, Schutgens F, Oka R, et al. An organoid biobank for childhood kidney cancers that captures disease and tissue heterogeneity. Nature communications. Mar 11 2020;11(1):1310. doi: 10.1038/s41467-020-15155-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Atun R, Bhakta N, Denburg A, et al. Sustainable care for children with cancer: a Lancet Oncology Commission. Lancet Oncol. Apr 2020;21(4):e185–e224. doi: 10.1016/s1470-2045(20)30022-x [DOI] [PubMed] [Google Scholar]
- 60.John R, Kurian JJ, Sen S, et al. Clinical outcomes of children with Wilms tumor treated on a SIOP WT 2001 protocol in a tertiary care hospital in south India. J Pediatr Urol. Dec 2018;14(6):547.e1–547.e7. doi: 10.1016/j.jpurol.2018.05.020 [DOI] [PubMed] [Google Scholar]
- 61.Ford K, Gunawardana S, Manirambona E, et al. Investigating Wilms’ Tumours Worldwide: A Report of the OxPLORE Collaboration-A Cross-Sectional Observational Study. World J Surg. Jan 2020;44(1):295–302. doi: 10.1007/s00268-019-05213-6 [DOI] [PubMed] [Google Scholar]
- 62.Drysdale H, Fawkner-Corbett D, Solomon Z, et al. Bilateral Wilms’ tumour: An international comparison of treatments and outcomes. J Pediatr Surg. Sep 2021;56(9):1487–1493. doi: 10.1016/j.jpedsurg.2021.01.040 [DOI] [PubMed] [Google Scholar]
- 63.Spreafico F, Fernandez CV, Brok J, et al. Wilms tumour. Nat Rev Dis Primers. Oct 14 2021;7(1):75. doi: 10.1038/s41572-021-00308-8 [DOI] [PubMed] [Google Scholar]
- 64.Israels T, Borgstein E, Jamali M, de Kraker J, Caron HN, Molyneux EM. Acute malnutrition is common in Malawian patients with a Wilms tumour: A role for peanut butter. Pediatr Blood Cancer. Dec 15 2009;53(7):1221–6. doi: 10.1002/pbc.22158 [DOI] [PubMed] [Google Scholar]
- 65.Chagaluka G, Paintsil V, Renner L, et al. Improvement of overall survival in the Collaborative Wilms Tumour Africa Project. Pediatr Blood Cancer. Sep 2020;67(9):e28383. doi: 10.1002/pbc.28383 [DOI] [PubMed] [Google Scholar]
- 66.Israels T, Borgstein E, Pidini D, et al. Management of children with a Wilms tumor in Malawi, sub-Saharan Africa. J Pediatr Hematol Oncol. Nov 2012;34(8):606–10. doi: 10.1097/MPH.0b013e3182580921 [DOI] [PubMed] [Google Scholar]
- 67.de Aguirre-Neto JC, de Camargo B, van Tinteren H, et al. International Comparisons of Clinical Demographics and Outcomes in the International Society of Pediatric Oncology Wilms Tumor 2001 Trial and Study. JCO global oncology. May 2022;8:e2100425. doi: 10.1200/GO.21.00425 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ekenze SO, Nwangwu EI, Ezomike UO, Orji EI, Okafor OO. Continuing barriers to care of Wilms tumor in a low-income country. Pediatr Blood Cancer. Jan 2019;66(1):e27416. doi: 10.1002/pbc.27416 [DOI] [PubMed] [Google Scholar]
- 69.Abuidris DO, Elimam ME, Nugud FM, Elgaili EM, Ahmed ME, Arora RS. Wilms tumour in Sudan. Pediatr Blood Cancer. Jun 2008;50(6):1135–7. doi: 10.1002/pbc.21547 [DOI] [PubMed] [Google Scholar]
- 70.Valverde P, Ortiz R, Fuentes-Alabi S, et al. An Analysis of Treatment Failure in Wilms Tumor (WT): A Report from the Central American Association of Pediatric Hematology/Oncology (AHOPCA). Journal of Global Oncology. 2016;2(3_suppl):2s–2s. doi: 10.1200/jgo.2016.004416 [DOI] [Google Scholar]
- 71.Davidson A, Hartley P, Desai F, Daubenton J, Rode H, Millar A. Wilms tumour experience in a South African centre. Pediatr Blood Cancer. Apr 2006;46(4):465–71. doi: 10.1002/pbc.20388 [DOI] [PubMed] [Google Scholar]
- 72.Asfour HY, Khalil SA, Zakaria AS, Ashraf ES, Zekri W. Localized Wilms’ tumor in low-middle-income countries (LMIC): how can we get better? J Egypt Natl Canc Inst. Aug 14 2020;32(1):32. doi: 10.1186/s43046-020-00043-3 [DOI] [PubMed] [Google Scholar]
- 73.Israels T, Molyneux EM, Caron HN, et al. Preoperative chemotherapy for patients with Wilms tumor in Malawi is feasible and efficacious. Pediatr Blood Cancer. Oct 2009;53(4):584–9. doi: 10.1002/pbc.22138 [DOI] [PubMed] [Google Scholar]
- 74.Borgstein E, Kamiza S, Vujanic G, et al. Wilms tumour in Malawi: surgical staging to stratify postoperative chemotherapy? Pediatr Blood Cancer. Dec 2014;61(12):2180–4. doi: 10.1002/pbc.25138 [DOI] [PubMed] [Google Scholar]
- 75.Fuchs J, Kienecker K, Furtwangler R, et al. Surgical aspects in the treatment of patients with unilateral wilms tumor: a report from the SIOP 93–01/German Society of Pediatric Oncology and Hematology. Annals of surgery. Apr 2009;249(4):666–71. doi: 10.1097/SLA.0b013e31819ed92b [DOI] [PubMed] [Google Scholar]
- 76.Ghafoor T, Bashir F, Ahmed S, Khalil S, Farah T. Predictors of treatment outcome of Wilms Tumour in low-income country; single centre experience from Pakistan. J Pediatr Urol. Jun 2020;16(3):375 e1–375 e7. doi: 10.1016/j.jpurol.2020.03.001 [DOI] [PubMed] [Google Scholar]
- 77.Hadley GP, Govender D, Landers G. Wilms tumour with unfavourable histology: implications for clinicians in the Third World. Med Pediatr Oncol. Jun 2001;36(6):652–3. doi: 10.1002/mpo.1145 [DOI] [PubMed] [Google Scholar]
- 78.Israels T, Harif M, Pritchard-Jones K. Treatment of Wilms tumor in low-income countries: challenges and potential solutions. Future Oncol. Aug 2013;9(8):1057–9. doi: 10.2217/fon.13.81 [DOI] [PubMed] [Google Scholar]
- 79.Polanco A, Al-Saadi R, Tugnait S, Scobie N, Pritchard-Jones K. Setting international standards for patient and parent involvement and engagement in childhood, adolescent and young adult cancer research: A report from a European Collaborative Workshop. Cancer Rep (Hoboken). Aug 12 2021:e1523. doi: 10.1002/cnr2.1523 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ciccarella A, Staley AC, Franco AT. Transforming research: engaging patient advocates at all stages of cancer research. Ann Transl Med. May 2018;6(9):167. doi: 10.21037/atm.2018.04.46 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Esmail L, Moore E, Rein A. Evaluating patient and stakeholder engagement in research: moving from theory to practice. J Comp Eff Res. Mar 2015;4(2):133–45. doi: 10.2217/cer.14.79 [DOI] [PubMed] [Google Scholar]
- 82.Brok J, Treger TD, Gooskens SL, van den Heuvel-Eibrink MM, Pritchard-Jones K. Biology and treatment of renal tumours in childhood. Eur J Cancer. Nov 2016;68:179–195. doi: 10.1016/j.ejca.2016.09.005 [DOI] [PubMed] [Google Scholar]
- 83.Brok J, Lopez-Yurda M, Tinteren HV, et al. Relapse of Wilms’ tumour and detection methods: a retrospective analysis of the 2001 Renal Tumour Study Group-International Society of Paediatric Oncology Wilms’ tumour protocol database. Lancet Oncol. Aug 2018;19(8):1072–1081. doi: 10.1016/S1470-2045(18)30293-6 [DOI] [PubMed] [Google Scholar]