1
|
Fitski M, Bökkerink GMJ, Davidoff AM, Murphy AJ, Abdelhafeez AH, Krauel L, Fuchs J, Pachl M, Abib SDCV, Fernandez-Pineda I, Sarnacki S, van de Ven CP, Wijnen MHWA, Klijn AJ, Godziński J, van der Steeg AFW. Nephron-Sparing Surgery for Patients With Wilms Tumor, a Surgical Delphi Study Consensus Statement. Pediatr Blood Cancer 2025; 72:e31636. [PMID: 40059283 DOI: 10.1002/pbc.31636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND AND AIM Current pediatric renal tumor treatment protocols allow for nephron-sparing surgery (NSS) for unilateral disease only under strict conditions. Oncological guidelines do not account for surgical feasibility, however, possibly reducing the utilization of NSS. To potentially change this, a definition of surgical feasibility is required. This study aimed to define surgical consensus statements for the assessment of patients with Wilms tumor (WT) for NSS. METHODS A Delphi study was performed inviting 34 potential experts. Surgeons were asked to answer the questionnaires without considering their current treatment protocol. The first questionnaire contained five open-ended questions regarding surgery, oncology, contraindications for NSS, technique, and organization. Follow-up questionnaires contained closed-ended statements based on previous answers. RESULTS Eleven surgeons were included in the expert panel and continued with three follow-up questionnaires containing 72 statements in total. A median of seven (3 minimum to 10 maximum) NSS procedures were performed per year in the hospitals of the experts. NSS for nonsyndromic unilateral WT patients can be surgically considered if they receive 4 weeks of neoadjuvant chemotherapy, have a preoperative tumor volume of less than 200 mL, and if partial nephrectomy with wide resection margin (>5 mm) can be performed. In keeping with organizational guidelines, among COG surgeons, NSS was not advocated for patients with nonsyndromic unilateral WT. CONCLUSIONS Using a Delphi method, surgical experts defined consensus statements regarding NSS for patients with WT to define surgical feasibility in future treatment protocols and expand the utilization of oncologically appropriate NSS.
Collapse
Affiliation(s)
- Matthijs Fitski
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Andrew Jackson Murphy
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Lucas Krauel
- Department of Pediatric Surgery, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Joerg Fuchs
- Department of Pediatric Surgery and Pediatric Urology, Tuebingen University Hospital, Tuebingen, Germany
| | - Maximilian Pachl
- Department of Pediatric Surgery and Urology, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - S de Campos Vieira Abib
- Department of Pediatric Surgery, Pediatric Oncology Institute - GRAACC - Federal University of São Paulo, São Paulo, Brazil
| | - Israel Fernandez-Pineda
- Department of Pediatric Surgery, Children's Health Ireland at Our Lady's Children's Hospital, Dublin, UK
| | - Sabine Sarnacki
- Department of Pediatric Surgery, Necker-Enfants Malades Hospital, APHP, Paris Cité University, Paris, France
| | - Cees P van de Ven
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Marc H W A Wijnen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - A J Klijn
- Department of Pediatric Urology, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - J Godziński
- Department of Pediatric Surgery, Marciniak Hospital, Wroclaw, Poland
- Department of Pediatric Traumatology and Emergency Medicine, Medical University, Wroclaw, Poland
| | | |
Collapse
|
2
|
Wang H, Yu C, Li Z, Zhang J, Xiao H, Wen S, Hua Y. Incidence and prognosis of pediatric kidney malignant tumors diagnosed from 2010 to 2021: a Surveillance, Epidemiology, and End Results-based study. Eur J Cancer Prev 2025:00008469-990000000-00226. [PMID: 40402532 DOI: 10.1097/cej.0000000000000976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Pediatric kidney parenchymal malignant tumor (KPMT) is a common primary tumor. We conducted this study to characterize the incidence proportions and prognosis of Wilms tumor and non-Wilms tumors (NWTs). We collected 1739 patients under the age of 18 who were diagnosed with KPMT from the Surveillance, Epidemiology, and End Results database between 2010 and 2021, and divided them into two groups to analyze. The incidence rate of Wilms tumor (85.1%) is significantly higher than NWT (14.9%). The proportion of Wilms tumor decreases with age [odds ratio (OR) = 0.81, 95% confidence interval (CI) = 0.78-0.84], and is gradually replaced by NWT. Male (OR = 0.65, 95% CI = 0.47-0.90), Asian (vs. White, OR = 0.46, 95% CI = 0.26-0.81) had significantly lower odds of Wilms tumor. In addition, Wilms tumor had the lower odds of initial diagnosis at the regional stage (vs. localized, OR = 0.68, 95% CI = 0.47-0.98), bone metastasis (OR = 0.09, 95% CI = 0.03-0.24), and brain metastasis (OR = 0.007, 95% CI = 0.001-0.068) than NWT, but the higher odds of lung metastasis (OR = 3.07, 95% CI = 1.42-6.62) and larger size of tumor (OR = 1.009, 95% CI = 1.005-1.012). NWT is associated with a significantly higher mortality than Wilms tumor (P < 0.01). Patients with later stage and major organ involvement are significantly associated with a higher mortality (P < 0.01). Surgery significantly improved the prognosis of patients with Wilms tumor and NWT.
Collapse
Affiliation(s)
- Huyu Wang
- Department of Urology Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Chengjun Yu
- Department of Urology Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Zhengbai Li
- Department of Urology Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Jie Zhang
- Department of Urology Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Hanyu Xiao
- Department of Urology Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Sheng Wen
- Department of Urology Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Yi Hua
- Department of Urology Surgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| |
Collapse
|
3
|
Sun H, Yang Z, Jin H, Cui Y, Yin Y, Chen S, Fu C, Wang H, Cheng W. 18F-Fluorodeoxyglucose positron emission tomography/computed tomography in distinguishing pediatric clear cell sarcoma of the kidney from Wilms' tumor. Pediatr Radiol 2025; 55:1289-1297. [PMID: 40323439 DOI: 10.1007/s00247-025-06239-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 04/03/2025] [Accepted: 04/05/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND Clear cell sarcoma of the kidney is a rare primary renal malignancy in children and its differentiation from other renal tumors, particularly Wilms' tumor, poses significant diagnostic challenges. Notably, clear cell sarcoma of the kidney is associated with a poorer prognosis than Wilms' tumor, making the preoperative presumptive diagnosis critical for optimizing surgical resection and appropriate chemotherapy regimens. OBJECTIVE We conducted a feasibility study to evaluate the diagnostic utility of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) in distinguishing clear cell sarcoma of the kidney from Wilms' tumor. MATERIALS AND METHODS This study included 19 patients diagnosed with clear cell sarcoma of the kidney and 84 patients with Wilms' tumor. Among these, 11/19 patients with clear cell sarcoma of the kidney and 28/84 patients of Wilms' tumor displayed positive findings on 18F-FDG PET/CT imaging, which were subsequently validated as tumor lesions by histologic examination, conventional imaging, or clinical follow-up. A retrospective analysis of the imaging features of clear cell sarcoma of the kidney and Wilms' tumor lesions on 18F-FDG PET/CT scans was conducted. Receiver operating characteristic (ROC) curve analyses were performed to determine the optimal cutoff of the maximum standardized uptake value (SUVmax) for diagnostic prediction. RESULTS The mean SUVmax for all lesions of clear cell sarcoma of the kidney was significantly lower than that of Wilms' tumors (3.9 vs. 7.4; P<0.001). When analyzed separately, primary lesions (3.6 vs. 6.6; P=0.042) and metastatic lesions (4.0 vs. 7.8; P=0.011) both had lower SUVmax in clear cell sarcomas of the kidney compared with Wilms' tumors. ROC curve analysis revealed a cutoff SUVmax of 4.3, which effectively supported the diagnosis of clear cell sarcoma of the kidney over Wilms' tumor, with a sensitivity of 75.0% and a specificity of 87.5%. CONCLUSIONS Clear cell sarcomas of the kidney exhibited a significantly lower SUVmax on 18F-FDG PET/CT when compared to Wilms' tumor lesions, with a proposed threshold SUVmax of 4.3 serving as an effective criterion for differentiation. Consequently, 18F-FDG PET/CT demonstrated potential in distinguishing clear cell sarcoma of the kidney from Wilms' tumor in cases involving large renal masses.
Collapse
Affiliation(s)
- Hui Sun
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 1665 Kongjiang Road, 200092, China
| | - Zhiyong Yang
- Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Hongfu Jin
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 1665 Kongjiang Road, 200092, China
| | - Yanfen Cui
- Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Yafu Yin
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 1665 Kongjiang Road, 200092, China
| | - Suyun Chen
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 1665 Kongjiang Road, 200092, China
| | - Chenglai Fu
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 1665 Kongjiang Road, 200092, China
| | - Hui Wang
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 1665 Kongjiang Road, 200092, China.
| | - Weiwei Cheng
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 1665 Kongjiang Road, 200092, China.
| |
Collapse
|
4
|
Welter N, Al-Saadi R, Gravier-Dumonceau R, Furtwängler R, Graf N, Wegert J, Gessler M, Williams RD, Pritchard-Jones K, Coulomb-L'Hermine A, van den Heuvel-Eibrink MM, Verschuur AC. The Clinical Impact of Somatic Copy Number Variations in Patients With Stage IV Wilms Tumor Enrolled in the SIOP 2001 Trial and Study. Pediatr Blood Cancer 2025; 72:e31580. [PMID: 39895484 DOI: 10.1002/pbc.31580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Recent research elucidated the prognostic significance of molecular biology in Wilms tumor (WT) by linking somatic genomic variants (such as gain of chromosome 1q) to unfavorable patient outcomes. This analysis describes the clinical impact of copy number variations (CNV) in tumor samples of WT patients with stage IV disease. METHODS Tumor samples of 55 WT patients with stage IV disease from the United Kingdom, France, and Germany enrolled in the SIOP 2001 study and treated with preoperative chemotherapy (pCHT) were examined for their CNVs of chromosome 1q and other regions of interest using multiplex ligation-dependent probe amplification (MLPA). The identified CNV were analyzed regarding their prognostic impact. RESULTS Chromosome 1q gain (1q+) and TP53 loss occurred in 38.2% and 16.4% of tumors and were associated with older patient age at diagnosis (median [months]: 65 and 64 vs. 49 each, p = 0.03 and 0.02, respectively) and poorer 5-year event-free survival (40.0% and 11.1% vs. 67.7% and 82.6%, p = 0.04 and <0.01, respectively) compared to their specific control group of tumors without the respective CNV. In patients with pulmonary-only metastasis, 1q+ was an adverse prognostic marker irrespective of remission status after pCHT with or without metastasectomy. A simultaneous MYCN gain occurred more frequently in tumors with 1q+ than in tumors without 1q+ (p = 0.03). TP53 loss was linked to high-risk histology and inferior 5-year overall survival (p < 0.001). CONCLUSIONS We confirm the prognostic relevance of 1q+ and TP53 loss in stage IV WTs and emphasize their potential utility for future treatment stratification.
Collapse
Affiliation(s)
- Nils Welter
- Department of Pediatric Oncology and Hematology, Saarland University, Homburg, Germany
| | - Reem Al-Saadi
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Histopathology, Great Ormond Street Hospital for Children, London, UK
| | - Robinson Gravier-Dumonceau
- APHM, Hop Timone, BioSTIC, Biostatistique et Technologies de l'Information et de la Communication, Marseille, France
| | - Rhoikos Furtwängler
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Inselspital University Hospital, Bern, Switzerland
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Saarland University, Homburg, Germany
| | - Jenny Wegert
- Developmental Biochemistry and Comprehensive Cancer Centre Mainfranken, Theodor-Boveri-Institute/Biocenter, University of Würzburg, Würzburg, Germany
| | - Manfred Gessler
- Developmental Biochemistry and Comprehensive Cancer Centre Mainfranken, Theodor-Boveri-Institute/Biocenter, University of Würzburg, Würzburg, Germany
| | - Richard D Williams
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Section of Genetics and Genomics, Faculty of Medicine, Imperial College London, London, UK
| | - Kathy Pritchard-Jones
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | | | | | - Arnauld C Verschuur
- Department of Pediatric Haematology-Oncology, La Timone Children's Hospital, AP-HM, Marseille, France
| |
Collapse
|
5
|
Neagu MC, David VL, Iacob ER, Chiriac SD, Muntean FL, Boia ES. Wilms' Tumor: A Review of Clinical Characteristics, Treatment Advances, and Research Opportunities. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:491. [PMID: 40142302 PMCID: PMC11943957 DOI: 10.3390/medicina61030491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025]
Abstract
Nephroblastoma is a complex childhood cancer with a generally favorable prognosis, well-defined incidence, and demographic profile but with significant challenges in terms of recurrence and long-term health outcomes. Although the management of this pathology has evolved, leading to improved survival rates, continued research into the long-term effects of treatment and the genetic factors influencing its development is still required. The survival landscape for Wilms tumor is evolving, with emerging research focusing on therapeutic biomarkers and genetic predispositions that influence treatment efficacy and survival rates. Identifying predictors for treatment response, such as specific genetic markers and histologic features, emerges as a critical area of study that could refine future interventions. The management of Wilms tumor is complex, taking into account the stage of the disease, histological classification, and individual patient factors, including age and the presence of syndromic associations. As treatment paradigms evolve, the integration of precision medicine approaches may enhance the ability of clinicians to personalize treatment to improve long-term survival outcomes for a broader range of patients. Recent advances in technology, including machine-learning approaches, have facilitated the identification of therapeutic biomarkers that correlate with clinical outcomes. This innovative method enhances the ability to integrate clinical and genetic data to predict disease trajectory and therapeutic response.
Collapse
Affiliation(s)
- Mihai Cristian Neagu
- Department of Pediatric Surgery and Orthopedics, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (M.C.N.); (V.L.D.); (E.R.I.); (E.S.B.)
| | - Vlad Laurenţiu David
- Department of Pediatric Surgery and Orthopedics, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (M.C.N.); (V.L.D.); (E.R.I.); (E.S.B.)
| | - Emil Radu Iacob
- Department of Pediatric Surgery and Orthopedics, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (M.C.N.); (V.L.D.); (E.R.I.); (E.S.B.)
| | - Sorin Dan Chiriac
- Department X—Surgery II, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Florin Lucian Muntean
- Department X—Surgery II, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Eugen Sorin Boia
- Department of Pediatric Surgery and Orthopedics, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (M.C.N.); (V.L.D.); (E.R.I.); (E.S.B.)
| |
Collapse
|
6
|
Salzillo C, Cazzato G, Serio G, Marzullo A. Paediatric Renal Tumors: A State-of-the-Art Review. Curr Oncol Rep 2025; 27:211-224. [PMID: 39918792 PMCID: PMC11958499 DOI: 10.1007/s11912-025-01644-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 04/01/2025]
Abstract
PURPOSE OF REVIEW Pediatric renal tumors comprise a wide range of conditions, both malignant and benign, that affect children and require a multidisciplinary approach for optimal diagnosis and treatment. This review offers an in-depth analysis of the epidemiology, diagnosis, treatment options, outcomes, and survival of major pediatric renal tumors. RECENT FINDINGS Wilms tumor, or nephroblastoma, is the most common form of renal tumor in children, characterized by growth from primitive renal cells. Standard treatment involves a combination of surgery, chemotherapy and, in some cases, radiation therapy, with the aim of removing the tumor, preventing recurrence and maximizing the chances of long-term recovery. Less common pediatric renal tumors, such as renal clear cell sarcoma, renal cell carcinoma, mesoblastic nephroma, and malignant rhabdoid tumor, require similarly careful and individualized management. Therapeutic strategies, which depend on the characteristics of the tumor, the stage of the disease and the individual response to therapy, may include surgery, chemotherapy, radiotherapy and, in some cases, molecular targeted therapies, immunotherapies and genetic and epigenetic therapies. The management of pediatric kidney tumors requires the involvement of a multidisciplinary team of specialists to ensure accurate evaluation, optimal treatments and long-term follow-up. The aim is to maximize the prospects for recovery and improve the quality of life of patients and their families. Advances in innovative, personalized therapies represent an important opportunity to further improve clinical outcomes in these patients.
Collapse
Affiliation(s)
- Cecilia Salzillo
- Department of Precision and Regenerative Medicine and Ionian Area, Pathology Unit, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70121, Bari, Italy.
- Department of Experimental Medicine, PhD Course in Public Health, University of Campania "Luigi Vanvitelli", Luciano Armanni 5, 80138, Naples, Italy.
| | - Gerardo Cazzato
- Department of Precision and Regenerative Medicine and Ionian Area, Pathology Unit, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70121, Bari, Italy
| | - Gabriella Serio
- Department of Precision and Regenerative Medicine and Ionian Area, Pathology Unit, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70121, Bari, Italy
| | - Andrea Marzullo
- Department of Precision and Regenerative Medicine and Ionian Area, Pathology Unit, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70121, Bari, Italy
| |
Collapse
|
7
|
van der Beek JN, Fitski M, de Krijger RR, Vermeulen MA, Nikkels PGJ, Maat A, Buser MAD, Wijnen MHWA, Hendrikse J, van den Heuvel-Eibrink MM, van der Steeg AFW, Littooij AS. Direct correlation of MR-DWI and histopathology of Wilms' tumours through a patient-specific 3D-printed cutting guide. Eur Radiol 2025; 35:652-663. [PMID: 39115585 PMCID: PMC11782413 DOI: 10.1007/s00330-024-10959-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/21/2024] [Accepted: 06/26/2024] [Indexed: 02/01/2025]
Abstract
OBJECTIVES The International Society of Paediatric Oncology-Renal Tumour Study Group (SIOP-RTSG) discourages invasive procedures to determine the histology of paediatric renal neoplasms at diagnosis. Therefore, the histological subtype of Wilms' tumours (WT) is unknown at the start of neoadjuvant chemotherapy. MR-DWI shows potential value as a non-invasive biomarker through apparent diffusion coefficients (ADCs). This study aimed to describe MR characteristics and ADC values of paediatric renal tumours to differentiate subtypes. MATERIALS AND METHODS Children with a renal tumour undergoing surgery within the SIOP-RTSG 2016-UMBRELLA protocol were prospectively included between May 2021 and 2023. In the case of a total nephrectomy, a patient-specific cutting guide based on the neoadjuvant MR was 3D-printed, allowing a correlation between imaging and histopathology. Whole-tumour volumes and ADC values were statistically compared with the Mann-Whitney U-test. Direct correlation on the microscopic slide level was analysed through mixed model analysis. RESULTS Fifty-nine lesions of 54 patients (58% male, median age 3.0 years (range 0-17.7 years)) were included. Forty-four lesions involved a WT. Stromal type WT showed the lowest median decrease in volume after neoadjuvant chemotherapy (48.1 cm3, range 561.5-(+)332.7 cm3, p = 0.035). On a microscopic slide level (n = 240 slides) after direct correlation through the cutting guide, stromal areas showed a significantly higher median ADC value compared to epithelial and blastemal foci (p < 0.001). With a cut-off value of 1.195 * 10-3 mm2/s, sensitivity, and specificity were 95.2% (95% confidence interval 87.6-98.4%) and 90.5% (95% confidence interval 68.2-98.3%), respectively. CONCLUSION Correlation between histopathology and MR-DWI through a patient-specific 3D-printed cutting guide resulted in significant discrimination of stromal type WT from epithelial and blastemal subtypes. CLINICAL RELEVANCE STATEMENT Stromal Wilms' tumours could be discriminated from epithelial- and blastemal lesions based on high apparent diffusion coefficient values and limited decrease in volume after neoadjuvant chemotherapy. This may aid in future decision-making, especially concerning discrimination between low- and high-risk neoplasms. KEY POINTS MR-DWI shows potential value as a non-invasive biomarker in paediatric renal tumours. The patient-specific cutting guide leads to a correlation between apparent diffusion coefficient values and Wilms' tumour subtype. Stromal areas could be discriminated from epithelial and blastemal foci in Wilms' tumours based on apparent diffusion coefficient values.
Collapse
Affiliation(s)
- Justine N van der Beek
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands.
| | - Matthijs Fitski
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marijn A Vermeulen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter G J Nikkels
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Arie Maat
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Myrthe A D Buser
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Marc H W A Wijnen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jeroen Hendrikse
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Division of Child Health, Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | | | - Annemieke S Littooij
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| |
Collapse
|
8
|
Yagasaki H, Katsumi Y, Nozaki M, Hamanoue S, Fukuzawa H, Fukumoto K, Mochizuki S, Uehara S, Oue T, Koshinaga T. Long-Term Survey of Japanese Children with Recurrent Nephroblastoma: A Report from Japan Children's Cancer Group. Pediatr Hematol Oncol 2025; 42:14-26. [PMID: 39501628 DOI: 10.1080/08880018.2024.2423207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 09/30/2024] [Accepted: 10/25/2024] [Indexed: 01/21/2025]
Abstract
In prospective Japanese studies of pediatric renal tumors, 5-year event-free survival and overall survival (OS) for patients with nephroblastoma ranges from 75-90% and 89-97%, respectively. However, treatments strategies for recurrent nephroblastoma in Japanese patients remain unclear. This retrospective study aimed to inform the development of treatment strategies by analyzing the long-term results and side effects of salvage therapies for recurrent nephroblastoma in Japan. A questionnaire survey involving 41 institutions (74 patients) collected clinical data on recurrent cases reported to the Renal Tumor Committee of the Japan Children's Cancer Group. Survey forms from 54 cases were evaluated. Median time to recurrence was 9.5 months among 51 patients without underlying disorders. Recurrence occurred at lung-only in 18 patients and at other sites in 33. The 5-year OS for all 51 patients was 70.6%, with recurrent disease causing death in 15 patients and one patient dying from treatment-related complications. Patients with lung-only recurrence had higher 5-year OS rates than those with other-site recurrence. Initial chemotherapy intensity also affected prognosis, with lower intensity associated with higher 5-year OS. In 17 survivors with lung-only recurrence, the most frequent treatment approach combined chemotherapy, surgery and radiotherapy. Conventional chemotherapy included platinum-containing regimens and/or Regimen I-based treatment containing cyclophosphamide and etoposide. Salvage therapies showed remarkable effectiveness for patients with lung-only recurrence or low intensity of the initial chemotherapy, highlighting the need to standardize prospective studies for post-recurrence treatment and identify risks of late complications for long-term survivors.
Collapse
Affiliation(s)
| | - Yoshiki Katsumi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Miwako Nozaki
- Department of Radiology, Dokkyo Medical University Saitama Medical Center, Koshigaya, Japan
| | - Satoshi Hamanoue
- Division of Hematology/Oncology, Kanagawa Children's Medical Center, Kanagawa, Japan
| | - Hiroaki Fukuzawa
- Department of Pediatric Surgery, Japanese Red Cross Society Himeji Hospital, Hyogo, Japan
| | - Koji Fukumoto
- Department of Pediatric Surgery, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Shinji Mochizuki
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
- Department of Pediatrics, Hiroshima University Hospital, Hiroshima, Japan
| | - Shuichiro Uehara
- Division of Pediatric Surgery, Department of Surgery, Nihon University Itabashi Hospital, Tokyo, Japan
| | - Takaharu Oue
- Department of Pediatric Surgery, Hyogo College of Medicine, Hyogo, Japan
| | - Tsugumichi Koshinaga
- Division of Pediatric Surgery, Department of Surgery, Nihon University Itabashi Hospital, Tokyo, Japan
| |
Collapse
|
9
|
Al-Jilaihawi S, Spreafico F, Mavinkurve-Groothuis A, Drost J, Perotti D, Koenig C, Brok J. Bevacizumab-containing treatment for relapsed or refractory Wilms tumor. Expert Rev Anticancer Ther 2024; 24:837-843. [PMID: 39016020 DOI: 10.1080/14737140.2024.2381537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/15/2024] [Indexed: 07/18/2024]
Abstract
INTRODUCTION Angiogenesis is critical for tumor growth and metastasis. Bevacizumab is an antiangiogenic drug used to treat various adult and childhood solid tumors. Its potential efficacy in Wilms tumor (WT) with poor prognosis is not established. AREAS COVERED The response to bevacizumab-containing regimens in relapsed or refractory WT was reviewed in available literature. Searches were conducted using PubMed, Scopus, and ClinicalTrials.gov databases. Eight papers were identified, published between 2007 and 2020, including six treatment regimens, predominantly vincristine, irinotecan, and bevacizumab (VIB) ± temozolomide (VITB). Among 16 evaluable patients, there were two complete responses, seven partial responses, five patients achieved stable disease (SD), and two patients had progressive disease. Objective responses (OR) were observed in 56% of all cases. OR or SD was observed in 89% (8/9) patients who received VIB/VITB. Bevacizumab was generally well tolerated. Related toxicities included hypertension, proteinuria, and delayed wound healing. EXPERT OPINION This review suggests potential effectiveness and good tolerability of bevacizumab in the setting of relapsed/refractory WT when used in combination with other drugs. Such combination therapies may serve as a bridging treatment option to other interventions and more personalized treatment options in the future; however, focused trials are needed to obtain additional evidence.
Collapse
Affiliation(s)
- Sarah Al-Jilaihawi
- Department of Paediatric Oncology, Bristol Royal Hospital for Children, Bristol, UK
| | - Filippo Spreafico
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | | | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Daniela Perotti
- Predictive Medicine: Molecular Bases of Genetic Risk and Genetic Testing Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Christa Koenig
- Pediatric Hematology/Oncology, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jesper Brok
- Department of Pediatric Hematology and Oncology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
10
|
Love JR, Karthaus WR. Next-Generation Modeling of Cancer Using Organoids. Cold Spring Harb Perspect Med 2024; 14:a041380. [PMID: 37734867 PMCID: PMC11146310 DOI: 10.1101/cshperspect.a041380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
In the last decade, organoid technology has become a cornerstone in cancer research. Organoids are long-term primary cell cultures, usually of epithelial origin, grown in a three-dimensional (3D) protein matrix and a fully defined medium. Organoids can be derived from many organs and cancer types and sites, encompassing both murine and human tissues. Importantly, they can be established from various stages during tumor evolution and recapitulate with high accuracy patient genomics and phenotypes in vitro, offering a platform for personalized medicine. Additionally, organoids are remarkably amendable for experimental manipulation. Taken together, these features make organoids a powerful tool with applications in basic cancer research and personalized medicine. Here, we will discuss the origins of organoid culture, applications in cancer research, and how cancer organoids can synergize with other models of cancer to drive basic discoveries as well as to translate these toward clinical solutions.
Collapse
Affiliation(s)
- Jillian R Love
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Wouter R Karthaus
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| |
Collapse
|
11
|
Andres S, Huang K, Shatara M, Abdelbaki MS, Ranalli M, Finlay J, Gupta A. Rhabdoid tumor predisposition syndrome: A historical review of treatments and outcomes for associated pediatric malignancies. Pediatr Blood Cancer 2024; 71:e30979. [PMID: 38553892 PMCID: PMC11039352 DOI: 10.1002/pbc.30979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/12/2024] [Accepted: 03/14/2024] [Indexed: 04/24/2024]
Abstract
Rhabdoid tumor predisposition syndrome (RTPS) is a rare disorder associated with malignant rhabdoid tumor of the kidney (RTK), atypical teratoid rhabdoid tumor (ATRT), and/or other extracranial, extrarenal rhabdoid tumors (EERT), and these pediatric malignancies are difficult to treat. Presently, most of the information regarding clinical manifestations, treatment, and outcomes of rhabdoid tumors comes from large data registries and case series. Our current understanding of treatments for patients with rhabdoid tumors may inform how we approach patients with RTPS. In this manuscript, we review the genetic and clinical features of RTPS and, using known registry data and clinical reports, review associated tumor types ATRT, RTK, and EERT, closing with potential new approaches to treatment. We propose collaborative international efforts to study the use of SMARC (SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin)-targeting agents, high-dose consolidative therapy, and age-based irradiation of disease sites in RTPS.
Collapse
Affiliation(s)
- Sarah Andres
- Jacobs School of Medicine and Biomedical Sciences at the University at Buffalo, Buffalo, New York
| | - Karen Huang
- Jacobs School of Medicine and Biomedical Sciences at the University at Buffalo, Buffalo, New York
| | - Margaret Shatara
- The Division of Hematology and Oncology, St. Louis Children’s Hospital, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Mohamed S. Abdelbaki
- The Division of Hematology and Oncology, St. Louis Children’s Hospital, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Mark Ranalli
- Division of Hematology/Oncology/Bone Marrow Transplant, Nationwide Children’s Hospital, Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Jonathan Finlay
- Departments of Pediatrics and Radiation Oncology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Ajay Gupta
- Division of Pediatric Oncology, Roswell Park Comprehensive Cancer Center, Department of Pediatrics, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York
| |
Collapse
|
12
|
Jin H, Yin Y, Wang H, Cheng W. 18 F-FDG PET/CT in a Case of Clear Cell Sarcoma of the Kidney. Clin Nucl Med 2024; 49:468-469. [PMID: 38377388 DOI: 10.1097/rlu.0000000000005071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
ABSTRACT 18 F-FDG PET/CT was performed in a 1-year-old girl who had a heterogeneous mass in the right abdominal cavity revealed by abdominal ultrasound. A heterogeneous mass with internal necrosis, cystic changes, and hemorrhage in the right kidney, accompanied by a slight increase of FDG uptake, was observed in FDG PET/CT. Malignant renal tumor was considered, and Wilms tumor was preferentially suspected. However, the mass was demonstrated as clear cell sarcoma of the kidney by histopathological examination.
Collapse
Affiliation(s)
- Hongfu Jin
- From the Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | |
Collapse
|
13
|
Choochuen P, Nokchan N, Khongcharoen N, Laochareonsuk W, Surachat K, Chotsampancharoen T, Sila T, Consortium SS. Discovery of Novel Potential Prognostic Markers and Targeted Therapy to Overcome Chemotherapy Resistance in an Advanced-Stage Wilms Tumor. Cancers (Basel) 2024; 16:1567. [PMID: 38672648 PMCID: PMC11049388 DOI: 10.3390/cancers16081567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Wilms tumor (WT), the most prevalent type of renal cancer in children, exhibits overall survival rates exceeding 90%. However, chemotherapy resistance, which occurs in approximately 10% of WT cases, is a major challenge for the treatment of WT, particularly for advanced-stage patients. In this study, we aimed to discover potential mutation markers and drug targets associated with chemotherapy resistance in advanced-stage WT. We performed exome sequencing to detect somatic mutations and molecular targets in 43 WT samples, comprising 26 advanced-stage WTs, of which 7 cases were chemotherapy-resistant. Our analysis revealed four genes (ALPK2, C16orf96, PRKDC, and SVIL) that correlated with chemotherapy resistance and reduced disease-free survival in advanced-stage WT. Additionally, we identified driver mutations in 55 genes within the chemotherapy-resistant group, including 14 druggable cancer driver genes. Based on the mutation profiles of the resistant WT samples, we propose potential therapeutic strategies involving platinum-based agents, PARP inhibitors, and antibiotic/antineoplastic agents. Our findings provide insights into the genetic landscape of WT and offer potential avenues for targeted treatment, particularly for patients with chemotherapy resistance.
Collapse
Affiliation(s)
- Pongsakorn Choochuen
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.C.); (N.N.); (N.K.); (W.L.); (K.S.)
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Natakorn Nokchan
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.C.); (N.N.); (N.K.); (W.L.); (K.S.)
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Natthapon Khongcharoen
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.C.); (N.N.); (N.K.); (W.L.); (K.S.)
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Wison Laochareonsuk
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.C.); (N.N.); (N.K.); (W.L.); (K.S.)
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Komwit Surachat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.C.); (N.N.); (N.K.); (W.L.); (K.S.)
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | | | - Thanit Sila
- Department of Pathology, Facualty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand;
| | - Surasak Sangkhathat Consortium
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.C.); (N.N.); (N.K.); (W.L.); (K.S.)
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| |
Collapse
|
14
|
Alfaifi J. miRNAs Role in Wilms tumor pathogenesis: Signaling pathways interplay. Pathol Res Pract 2024; 256:155254. [PMID: 38460245 DOI: 10.1016/j.prp.2024.155254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Wilms' tumors (WTs) are the most common type of kidney tumor in children, and a negative outlook is generally associated with widespread anaplastic. MicroRNAs (miRNAs) are crucial in the development of WT by regulating the expression of specific genes. There is an increasing amount of research that connects the dysregulation of miRNAs to the development of various renal illnesses. The conditions encompassed are renal fibrosis, renal cancers, and chronic and polycystic kidney disease. Dysregulation of several important miRNAs, either oncogenic or tumor-suppressing, has been found in WT. The present state of knowledge on the involvement of dysregulated miRNAs in the progression of WT is summarized in this review.
Collapse
Affiliation(s)
- Jaber Alfaifi
- Department of Child Health, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| |
Collapse
|
15
|
Uno K, Rastegar B, Jansson C, Durand G, Valind A, Chattopadhyay S, Bertolotti A, Ciceri S, Spreafico F, Collini P, Perotti D, Mengelbier LH, Gisselsson D. A Gradual Transition Toward Anaplasia in Wilms Tumor Through Tolerance to Genetic Damage. Mod Pathol 2024; 37:100382. [PMID: 37951357 DOI: 10.1016/j.modpat.2023.100382] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/23/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
Patients with Wilms tumor (WT) in general have excellent survival, but the prognosis of patients belonging to the subgroup of WT with diffuse anaplasia (DA) is poor due to frequent resistance to chemotherapy. We hypothesized that DA WT cells might undergo changes, such as acquiring a persistent tolerance to DNA damage and copy number aberrations (CNAs), which could eventually lead to their resistance to chemotherapy treatment. Tissue sections from chemotherapy-treated DA WTs (n = 12) were compared with chemotherapy-treated nonanaplastic WTs (n = 15) in a tissue microarray system, enabling analysis of 769 tumor regions. All regions were scored for anaplastic features and immunohistochemistry was used to quantify p53 expression, proliferation index (Ki67), and DNA double-strand breaks (γH2AX). CNAs were assessed by array-based genotyping and TP53 mutations using targeted sequencing. Proliferation index and the frequency of DNA double-strand breaks (γH2AX dot expression) increased with higher anaplasia scores. Almost all (95.6%) areas with full-scale anaplasia had TP53 mutations or loss of heterozygosity, along with an increased amount of CNAs. Interestingly, areas with wild-type TP53 with loss of heterozygosity and only one feature of anaplasia (anaplasia score 1) also had significantly higher proliferation indices, more DNA double-strand breaks, and more CNAs than regions without any anaplastic features (score 0); such areas may be preanaplastic cell populations under selective pressure for TP53 mutations. In conclusion, we suggest that chemoresistance of DA WTs may be partly explained by a high proliferative capability of anaplastic cells, which also have a high burden of double-stranded DNA breaks and CNAs, and that there is a gradual emergence of anaplasia in WT.
Collapse
Affiliation(s)
- Kaname Uno
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden; Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Bahar Rastegar
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Caroline Jansson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Geoffroy Durand
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Anders Valind
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden; Now with Childhood Cancer Center, Skåne University Hospital, Lund, Sweden
| | - Subhayan Chattopadhyay
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Alessia Bertolotti
- Diagnostic and Molecular Research Lab, Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Ciceri
- Molecular Bases of Genetic Risk and Genetic Testing Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Now with Predictive Medicine: Molecular Bases of Genetic Risk, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Spreafico
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paola Collini
- Soft Tissue Tumor Pathology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Daniela Perotti
- Molecular Bases of Genetic Risk and Genetic Testing Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Now with Predictive Medicine: Molecular Bases of Genetic Risk, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - David Gisselsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden; Division of Oncology-Pathology, Department of Clinical Science, Lund University, Lund, Sweden; Division of Clinical Genetics and Pathology, Department of Laboratory Medicine, Lund University Hospital, Skåne Healthcare Region, Lund, Sweden
| |
Collapse
|
16
|
Tsiflikas I. [Renal tumors in children and adolescents]. RADIOLOGIE (HEIDELBERG, GERMANY) 2024; 64:18-25. [PMID: 37947863 DOI: 10.1007/s00117-023-01238-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
CLINICAL/METHODOLOGICAL ISSUE Diagnosis and therapy of renal tumors in children and adolescents are standardized by study protocols from major international societies. Imaging plays a central role, and in Europe patients between the ages of 6 months and 14 years with renal tumors are referred to neoadjuvant chemotherapy without histological confirmation according to the study protocol due to the frequency of Wilms tumor. STANDARD RADIOLOGIC METHODS Ultrasound is used worldwide as the primary investigative procedure for suspected renal tumors. In Europe, magnetic resonance imaging (MRI) has become established for more advanced diagnosis. In addition to differential diagnosis, staging is crucial for therapy. According to current protocol, this includes computed tomography (CT) of the thorax for the evaluation of pulmonary metastases. METHODOLOGICAL INNOVATIONS Diffusion-weighted MRI provides promising results for the differentiation of nephroblastoma subtypes in addition to improved detectability of tumor foci. However, sufficient evidence is lacking. PERFORMANCE Differentiation of Wilms tumor from the highly malignant non-Wilm tumors, such as malignant rhabdoid tumor and clear cell sarcoma of the kidney, remains inconclusive based on imaging alone. Differential diagnosis is, therefore, based on morphologic and epidemiologic criteria. ASSESSMENT The high degree of standardization in the diagnosis and treatment of renal tumors in children and adolescents has led to a significant improvement in prognosis. Overall survival of patients with Wilms tumor is currently over 90%.
Collapse
Affiliation(s)
- Ilias Tsiflikas
- Abteilung für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Deutschland.
| |
Collapse
|
17
|
Jia C, Gao H, Ma W, Liu X, Chang M, Sun F. Identification of the expression patterns and potential prognostic role of m6A-RNA methylation regulators in Wilms Tumor. BMC Med Genomics 2023; 16:222. [PMID: 37735424 PMCID: PMC10514975 DOI: 10.1186/s12920-023-01660-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND To explore the potential role of m6A methylation modification in Wilms Tumor (WT) by m6A-RNA Methylation (m6A) regulators. METHODOLOGY The association of m6A modification patterns with immune and prognostic characteristics of tumors was systematically evaluated using 19 m6A regulators extracted from Wilms Tumor's samples in public databases. A comprehensive model of "m6Ascore" was constructed using principal component analysis, and its prognostic value was evaluated. RESULTS Almost all m6A regulators were differentially expressed between WT and normal tissues. Unsupervised clustering identified three distinct m6A clusters that differed in both immune cell infiltration and biological pathways. The m6Ascore was constructed to quantify m6A modifications in individual patients. Our analysis suggests that m6Ascore is an independent prognostic factor for WT and can be used as a novel predictor of WT prognosis. CONCLUSIONS This study comprehensively explored and systematically characterized m6A modifications in WT. m6A modification patterns play a critical role in the tumor immune microenvironment (TIME) and WT prognosis. m6Ascore provides a more comprehensive understanding of m6A modifications in WT and offers a practical tool for predicting WT prognosis. This study will help clinicians to identify valid indicators of WT to improve the poor prognosis of this disease. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at https://www.aliyundrive.com/drive/folder/64be739cd6956a741fb24670baeea53422be6024 .
Collapse
Affiliation(s)
- Changlin Jia
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hongjie Gao
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wenyue Ma
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoya Liu
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Mengmeng Chang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Fengyin Sun
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
18
|
Benlhachemi S, Abouqal R, Coleman N, Murray MJ, Khattab M, El fahime E. Circulating microRNA profiles in Wilms tumour (WT): A systematic review and meta-analysis of diagnostic test accuracy. Noncoding RNA Res 2023; 8:413-425. [PMID: 37305178 PMCID: PMC10247954 DOI: 10.1016/j.ncrna.2023.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/13/2023] Open
Abstract
Background Wilms tumour (WT) is caused by aberrant embryonic kidney development and associated with dysregulated expression of short, non-protein-coding RNAs termed microRNAs (miRNAs). At present, there is no reliable circulating biomarker of WT, and this remains an urgent unmet clinical need. Such biomarkers may assist diagnosis, subtyping/prognostication, and disease-monitoring. Here, we established the list of dysregulated circulating miRNAs in WT from the existing published literature. Methods Regardless of publication date, PubMed, Scopus, Web-of-Science, and Wiley online library databases were searched for English/French studies on WT circulating miRNAs. The PRISMA-compliant search was registered in PROSPERO. The QUADAS tool measured retained article quality. The meta-analysis assessed the sensitivity and specificity of miRNAs for WT diagnosis. Results Qualitative analysis included 280 samples (172 WT patients; 108 healthy controls) from five of 450 published articles. The study uncovered 301 dysregulated miRNAs (144 up-regulated, 143 down-regulated, 14 conflicting). The pooled sensitivity, specificity, and AUC of the 49 significantly dysregulated microRNAs from two studies was 0.67 [0.62; 0.73], 0.95 [0.92; 0.96] and 0.77 [0.73; 0.81] respectively, indicating a stronger diagnostic potential for WT. Conclusions Circulating miRNAs show promise for WT diagnosis and prognosis. More research is needed to confirm these findings and determine associations with tumour stage/subtype. Prospero registration number CRD42022301597.
Collapse
Affiliation(s)
- Sara Benlhachemi
- Laboratory of Genomics and Molecular Epidemiology of Genetic Diseases (GE2MG). Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
- Molecular Biology and Functional Genomics Platform, National Center for Scientific and Technical Research, Rabat, Morocco
| | - Redouane Abouqal
- Biostatistics Laboratory, Clinical Epidemiology Research. Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Nicholas Coleman
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Matthew Jonathan Murray
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Mohammed Khattab
- Department of Paediatric Haematology and Oncology, Abulcasis International University of Health Sciences, Rabat, Morocco
| | - Elmostafa El fahime
- Molecular Biology and Functional Genomics Platform, National Center for Scientific and Technical Research, Rabat, Morocco
| |
Collapse
|
19
|
Stoltze UK, Hildonen M, Hansen TVO, Foss-Skiftesvik J, Byrjalsen A, Lundsgaard M, Pignata L, Grønskov K, Tumer Z, Schmiegelow K, Brok JS, Wadt KAW. Germline (epi)genetics reveals high predisposition in females: a 5-year, nationwide, prospective Wilms tumour cohort. J Med Genet 2023; 60:842-849. [PMID: 37019617 PMCID: PMC10447365 DOI: 10.1136/jmg-2022-108982] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/10/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Studies suggest that Wilms tumours (WT) are caused by underlying genetic (5%-10%) and epigenetic (2%-29%) mechanisms, yet studies covering both aspects are sparse. METHODS We performed prospective whole-genome sequencing of germline DNA in Danish children diagnosed with WT from 2016 to 2021, and linked genotypes to deep phenotypes. RESULTS Of 24 patients (58% female), 3 (13%, all female) harboured pathogenic germline variants in WT risk genes (FBXW7, WT1 and REST). Only one patient had a family history of WT (3 cases), segregating with the REST variant. Epigenetic testing revealed one (4%) additional patient (female) with uniparental disomy of chromosome 11 and Beckwith-Wiedemann syndrome (BWS). We observed a tendency of higher methylation of the BWS-related imprinting centre 1 in patients with WT than in healthy controls. Three patients (13%, all female) with bilateral tumours and/or features of BWS had higher birth weights (4780 g vs 3575 g; p=0.002). We observed more patients with macrosomia (>4250 g, n=5, all female) than expected (OR 9.98 (95% CI 2.56 to 34.66)). Genes involved in early kidney development were enriched in our constrained gene analysis, including both known (WT1, FBXW7) and candidate (CTNND1, FRMD4A) WT predisposition genes. WT predisposing variants, BWS and/or macrosomia (n=8, all female) were more common in female patients than male patients (p=0.01). CONCLUSION We find that most females (57%) and 33% of all patients with WT had either a genetic or another indicator of WT predisposition. This emphasises the need for scrutiny when diagnosing patients with WT, as early detection of underlying predisposition may impact treatment, follow-up and genetic counselling.
Collapse
Affiliation(s)
- Ulrik Kristoffer Stoltze
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
- Department of Pediatrics, Rigshospitalet, Copenhagen, Denmark
| | - Mathis Hildonen
- Department of Genetics, Kennedy Center-National Research Center on Rare Genetic Diseases, Glostrup, Denmark
| | | | | | - Anna Byrjalsen
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Malene Lundsgaard
- Department of Clinical Genetics, Aalborg University Hospital, Aalborg, North Denmark Region, Denmark
| | - Laura Pignata
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università Degli Studi Della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Karen Grønskov
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Zeynep Tumer
- Applied Human Molecular Genetics, Kennedy Center, Copenhagen University Hospital, Rigshospitalet, Glostrup, Denmark
| | | | - Jesper Sune Brok
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark
| | - Karin A W Wadt
- Department of Clinical Genetics, University Hospital of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
20
|
Avčin SL, Črepinšek K, Jenko Bizjan B, Šket R, Kovač J, Vrhovšek B, Blazina J, Blatnik O, Kordič R, Kitanovski L, Jazbec J, Debeljak M, Tesovnik T. Integrative Transcriptomic Profiling of the Wilms Tumor. Cancers (Basel) 2023; 15:3846. [PMID: 37568662 PMCID: PMC10416970 DOI: 10.3390/cancers15153846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Our study aimed to identify relevant transcriptomic biomarkers for the Wilms tumor, the most common pediatric kidney cancer, independent of the histological type and stage. Using next-generation sequencing, we analyzed the miRNA profiles of 74 kidney samples, which were divided into two independent groups: fresh frozen tissue and formalin-fixed paraffin-embedded tissue samples. Subsequent mRNA expression profiling and pathway analysis were performed to establish the interplay and potential involvement of miRNAs and mRNA in the Wilms tumor. Comparative analysis, irrespective of post-dissection tissue processing, revealed 41 differentially expressed miRNAs, with 27 miRNAs having decreased expression and 14 miRNAs having increased expression in the Wilms tumor tissue compared to healthy kidney tissue. Among global mRNA transcriptomic profile differences, cross-sectional analysis suggested a limited list of genes potentially regulated by differentially expressed miRNAs in the Wilms tumor. This study identified the comprehensive miRNA and mRNA profile of the Wilms tumor using next-generation sequencing and bioinformatics approach, providing better insights into the pathogenesis of the Wilms tumor. The identified Wilms tumor miRNAs have potential as biomarkers for the diagnosis and treatment of the Wilms tumor, regardless of histological subtype and disease stage.
Collapse
Affiliation(s)
- Simona Lucija Avčin
- Department of Haematology and Oncology, University Children’s Hospital, University Medical Centre Ljubljana (UMC), 1000 Ljubljana, Slovenia; (S.L.A.)
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Klementina Črepinšek
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Institute of Special Laboratory Diagnostic, University Children’s Hospital, University Medical Centre Ljubljana (UMC), 1000 Ljubljana, Slovenia
| | - Barbara Jenko Bizjan
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Institute of Special Laboratory Diagnostic, University Children’s Hospital, University Medical Centre Ljubljana (UMC), 1000 Ljubljana, Slovenia
| | - Robert Šket
- Institute of Special Laboratory Diagnostic, University Children’s Hospital, University Medical Centre Ljubljana (UMC), 1000 Ljubljana, Slovenia
| | - Jernej Kovač
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Institute of Special Laboratory Diagnostic, University Children’s Hospital, University Medical Centre Ljubljana (UMC), 1000 Ljubljana, Slovenia
| | - Blaž Vrhovšek
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Institute of Special Laboratory Diagnostic, University Children’s Hospital, University Medical Centre Ljubljana (UMC), 1000 Ljubljana, Slovenia
| | - Jerca Blazina
- Department of Pathology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia
| | - Olga Blatnik
- Department of Pathology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia
| | - Robert Kordič
- Department of Pediatric Surgery, University Children’s Hospital, University Medical Centre Ljubljana (UMC), 1000 Ljubljana, Slovenia
| | - Lidija Kitanovski
- Department of Haematology and Oncology, University Children’s Hospital, University Medical Centre Ljubljana (UMC), 1000 Ljubljana, Slovenia; (S.L.A.)
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Janez Jazbec
- Department of Haematology and Oncology, University Children’s Hospital, University Medical Centre Ljubljana (UMC), 1000 Ljubljana, Slovenia; (S.L.A.)
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Maruša Debeljak
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Institute of Special Laboratory Diagnostic, University Children’s Hospital, University Medical Centre Ljubljana (UMC), 1000 Ljubljana, Slovenia
| | - Tine Tesovnik
- Institute of Special Laboratory Diagnostic, University Children’s Hospital, University Medical Centre Ljubljana (UMC), 1000 Ljubljana, Slovenia
| |
Collapse
|
21
|
Zheng H, Liu J, Pan X, Cui X. Biomarkers for patients with Wilms tumor: a review. Front Oncol 2023; 13:1137346. [PMID: 37554168 PMCID: PMC10405734 DOI: 10.3389/fonc.2023.1137346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/27/2023] [Indexed: 08/10/2023] Open
Abstract
Wilms tumor, originating from aberrant fetal nephrogenesis, is the most common renal malignancy in childhood. The overall survival of children is approximately 90%. Although existing risk-stratification systems are helpful in identifying patients with poor prognosis, the recurrence rate of Wilms tumors remains as high as 15%. To resolve this clinical problem, diverse studies on the occurrence and progression of the disease have been conducted, and the results are encouraging. A series of molecular biomarkers have been identified with further studies on the mechanism of tumorigenesis. Some of these show prognostic value and have been introduced into clinical practice. Identification of these biomarkers can supplement the existing risk-stratification systems. In the future, more biomarkers will be discovered, and more studies are required to validate their roles in improving the detection rate of occurrence or recurrence of Wilms tumor and to enhance clinical outcomes.
Collapse
Affiliation(s)
| | | | - Xiuwu Pan
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xingang Cui
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
22
|
Ortiz MV, Koenig C, Armstrong AE, Brok J, de Camargo B, Mavinkurve-Groothuis AMC, Herrera TBV, Venkatramani R, Woods AD, Dome JS, Spreafico F. Advances in the clinical management of high-risk Wilms tumors. Pediatr Blood Cancer 2023; 70 Suppl 2:e30342. [PMID: 37096797 PMCID: PMC10857813 DOI: 10.1002/pbc.30342] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/24/2022] [Indexed: 04/26/2023]
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.
Collapse
Affiliation(s)
- Michael V Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Christa Koenig
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Amy E Armstrong
- Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jesper Brok
- Developmental Biology and Cancer Research and Teaching Department, University College London Great Ormond Street Institute of Child Health, London, UK
- Department of Pediatric Oncology and Hematology, Rigshospitalet, Copenhagen, Denmark
| | - Beatriz de Camargo
- Pediatric Hematology and Oncology Program, Research Center, Instituto Nacional de Cancer, Rio de Janeiro, Brazil
| | | | | | - Rajkumar Venkatramani
- Department of Pediatrics, Division of Hematology/Oncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Andrew D Woods
- Children's Cancer Therapy Development Institute, Beaverton, Oregon, USA
| | - Jeffrey S Dome
- Division of Oncology, Children's National Hospital and Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Filippo Spreafico
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
23
|
Ortiz MV, Koenig C, Armstrong AE, Brok J, de Camargo B, Mavinkurve-Groothuis AMC, Herrera TBV, Venkatramani R, Woods AD, Dome JS, Spreafico F. Advances in the clinical management of high-risk Wilms tumors. Pediatr Blood Cancer 2023; 70:e30153. [PMID: 36625399 DOI: 10.1002/pbc.30153] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/24/2022] [Indexed: 01/11/2023]
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.
Collapse
Affiliation(s)
- Michael V Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Christa Koenig
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Amy E Armstrong
- Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jesper Brok
- Developmental Biology and Cancer Research and Teaching Department, University College London Great Ormond Street Institute of Child Health, London, UK.,Department of Pediatric Oncology and Hematology, Rigshospitalet, Copenhagen, Denmark
| | - Beatriz de Camargo
- Pediatric Hematology and Oncology Program, Research Center, Instituto Nacional de Cancer, Rio de Janeiro, Brazil
| | | | | | - Rajkumar Venkatramani
- Department of Pediatrics, Division of Hematology/Oncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Andrew D Woods
- Children's Cancer Therapy Development Institute, Beaverton, Oregon, USA
| | - Jeffrey S Dome
- Division of Oncology, Children's National Hospital and Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Filippo Spreafico
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
24
|
Development of a Machine Learning-Based Prediction Model for Chemotherapy-Induced Myelosuppression in Children with Wilms' Tumor. Cancers (Basel) 2023; 15:cancers15041078. [PMID: 36831423 PMCID: PMC9954251 DOI: 10.3390/cancers15041078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
Purpose: Develop and validate an accessible prediction model using machine learning (ML) to predict the risk of chemotherapy-induced myelosuppression (CIM) in children with Wilms' tumor (WT) before chemotherapy is administered, enabling early preventive management. Methods: A total of 1433 chemotherapy cycles in 437 children with WT who received chemotherapy in our hospital from January 2009 to March 2022 were retrospectively analyzed. Demographic data, clinicopathological characteristics, hematology and blood biochemistry baseline results, and medication information were collected. Six ML algorithms were used to construct prediction models, and the predictive efficacy of these models was evaluated to select the best model to predict the risk of grade ≥ 2 CIM in children with WT. A series of methods, such as the area under the receiver operating characteristic curve (AUROC), the calibration curve, and the decision curve analysis (DCA) were used to test the model's accuracy, discrimination, and clinical practicability. Results: Grade ≥ 2 CIM occurred in 58.5% (839/1433) of chemotherapy cycles. Based on the results of the training and validation cohorts, we finally identified that the extreme gradient boosting (XGB) model has the best predictive efficiency and stability, with an AUROC of up to 0.981 in the training set and up to 0.896 in the test set. In addition, the calibration curve and the DCA showed that the XGB model had the best discrimination and clinical practicability. The variables were ranked according to the feature importance, and the five variables contributing the most to the model were hemoglobin (Hgb), white blood cell count (WBC), alkaline phosphatase, coadministration of highly toxic chemotherapy drugs, and albumin. Conclusions: The incidence of grade ≥ 2 CIM was not low in children with WT, which needs attention. The XGB model was developed to predict the risk of grade ≥ 2 CIM in children with WT for the first time. The model has good predictive performance and stability and has the potential to be translated into clinical applications. Based on this modeling and application approach, the extension of CIM prediction models to other pediatric malignancies could be expected.
Collapse
|
25
|
Gharehzadehshirazi A, Zarejousheghani M, Falahi S, Joseph Y, Rahimi P. Biomarkers and Corresponding Biosensors for Childhood Cancer Diagnostics. SENSORS (BASEL, SWITZERLAND) 2023; 23:1482. [PMID: 36772521 PMCID: PMC9919359 DOI: 10.3390/s23031482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/26/2023] [Accepted: 01/26/2023] [Indexed: 05/11/2023]
Abstract
Although tremendous progress has been made in treating childhood cancer, it is still one of the leading causes of death in children worldwide. Because cancer symptoms overlap with those of other diseases, it is difficult to predict a tumor early enough, which causes cancers in children to be more aggressive and progress more rapidly than in adults. Therefore, early and accurate detection methods are urgently needed to effectively treat children with cancer therapy. Identification and detection of cancer biomarkers serve as non-invasive tools for early cancer screening, prevention, and treatment. Biosensors have emerged as a potential technology for rapid, sensitive, and cost-effective biomarker detection and monitoring. In this review, we provide an overview of important biomarkers for several common childhood cancers. Accordingly, we have enumerated the developed biosensors for early detection of pediatric cancer or related biomarkers. This review offers a restructured platform for ongoing research in pediatric cancer diagnostics that can contribute to the development of rapid biosensing techniques for early-stage diagnosis, monitoring, and treatment of children with cancer and reduce the mortality rate.
Collapse
Affiliation(s)
- Azadeh Gharehzadehshirazi
- Institute of Electronic and Sensor Materials, Faculty of Materials Science and Materials Technology, Technische Universität Bergakademie Freiberg, 09599 Freiberg, Germany
| | - Mashaalah Zarejousheghani
- Freiberg Center for Water Research—ZeWaF, Technische Universität Bergakademie Freiberg, 09599 Freiberg, Germany
| | - Sedigheh Falahi
- Institute of Electronic and Sensor Materials, Faculty of Materials Science and Materials Technology, Technische Universität Bergakademie Freiberg, 09599 Freiberg, Germany
| | - Yvonne Joseph
- Institute of Electronic and Sensor Materials, Faculty of Materials Science and Materials Technology, Technische Universität Bergakademie Freiberg, 09599 Freiberg, Germany
- Freiberg Center for Water Research—ZeWaF, Technische Universität Bergakademie Freiberg, 09599 Freiberg, Germany
| | - Parvaneh Rahimi
- Institute of Electronic and Sensor Materials, Faculty of Materials Science and Materials Technology, Technische Universität Bergakademie Freiberg, 09599 Freiberg, Germany
- Freiberg Center for Water Research—ZeWaF, Technische Universität Bergakademie Freiberg, 09599 Freiberg, Germany
| |
Collapse
|
26
|
Koh KN, Han JW, Choi HS, Kang HJ, Lee JW, Yoo KH, Sung KW, Koo HH, Hong KT, Choi JY, Kang SH, Kim H, Im HJ, Hahn SM, Lyu CJ, Baek HJ, Kook H, Park KM, Yang EJ, Lim YT, Kim S, Lee JW, Chung NG, Cho B, Park M, Park HJ, Park BK, Lee JA, Park JE, Kim SK, Kim JY, Kim HS, Ma Y, Park KD, Park SK, Park ES, Shim YJ, Yoo ES, Ryu KH, Yoo JW, Lim YJ, Yoon HS, Lee MJ, Lee JM, Jeon IS, Jung HL, Chueh HW, Won S, The Korean Pediatric Hematology and Oncology Group (KPHOG). Epidemiologic and Clinical Outcomes of Pediatric Renal Tumors in Korea: A Retrospective Analysis of The Korean Pediatric Hematology and Oncology Group (KPHOG) Data. Cancer Res Treat 2023; 55:279-290. [PMID: 35952715 PMCID: PMC9873342 DOI: 10.4143/crt.2022.073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 08/07/2022] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Renal tumors account for approximately 7% of all childhood cancers. These include Wilms tumor (WT), clear cell sarcoma of the kidney (CCSK), malignant rhabdoid tumor of the kidney (MRTK), renal cell carcinoma (RCC), congenital mesoblastic nephroma (CMN) and other rare tumors. We investigated the epidemiology of pediatric renal tumors in Korea. MATERIALS AND METHODS From January 2001 to December 2015, data of pediatric patients (0-18 years) newly-diagnosed with renal tumors at 26 hospitals were retrospectively analyzed. RESULTS Among 439 patients (male, 240), the most common tumor was WT (n=342, 77.9%), followed by RCC (n=36, 8.2%), CCSK (n=24, 5.5%), MRTK (n=16, 3.6%), CMN (n=12, 2.7%), and others (n=9, 2.1%). Median age at diagnosis was 27.1 months (range 0-225.5) and median follow-up duration was 88.5 months (range 0-211.6). Overall, 32 patients died, of whom 17, 11, 1, and 3 died of relapse, progressive disease, second malignant neoplasm, and treatment-related mortality. Five-year overall survival and event free survival were 97.2% and 84.8% in WT, 90.6% and 82.1% in RCC, 81.1% and 63.6% in CCSK, 60.3% and 56.2% in MRTK, and 100% and 91.7% in CMN, respectively (p < 0.001). CONCLUSION The pediatric renal tumor types in Korea are similar to those previously reported in other countries. WT accounted for a large proportion and survival was excellent. Non-Wilms renal tumors included a variety of tumors and showed inferior outcome, especially MRTK. Further efforts are necessary to optimize the treatment and analyze the genetic characteristics of pediatric renal tumors in Korea.
Collapse
Affiliation(s)
- Kyung-Nam Koh
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul,
Korea
| | - Jung Woo Han
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Yonsei University College of Medicine, Seoul,
Korea
| | - Hyoung Soo Choi
- Department of Pediatrics, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam,
Korea
| | - Hyoung Jin Kang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul,
Korea,Seoul National University Cancer Institute, Seoul,
Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Kyung Taek Hong
- Department of Pediatrics, Seoul National University College of Medicine, Seoul,
Korea,Seoul National University Cancer Institute, Seoul,
Korea
| | - Jung Yoon Choi
- Department of Pediatrics, Seoul National University College of Medicine, Seoul,
Korea,Seoul National University Cancer Institute, Seoul,
Korea
| | - Sung Han Kang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul,
Korea
| | - Hyery Kim
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul,
Korea
| | - Ho Joon Im
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul,
Korea
| | - Seung Min Hahn
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Yonsei University College of Medicine, Seoul,
Korea
| | - Chuhl Joo Lyu
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Yonsei University College of Medicine, Seoul,
Korea
| | - Hee-Jo Baek
- Department of Pediatrics, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Gwangju,
Korea
| | - Hoon Kook
- Department of Pediatrics, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Gwangju,
Korea
| | - Kyung Mi Park
- Department of Pediatrics, Pusan National University School of Medicine, Yangsan,
Korea
| | - Eu Jeen Yang
- Department of Pediatrics, Pusan National University School of Medicine, Yangsan,
Korea
| | - Young Tak Lim
- Department of Pediatrics, Pusan National University School of Medicine, Yangsan,
Korea
| | - Seongkoo Kim
- Department of Pediatrics, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Jae Wook Lee
- Department of Pediatrics, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Nack-Gyun Chung
- Department of Pediatrics, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Bin Cho
- Department of Pediatrics, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Meerim Park
- Center for Pediatric Cancer, Department of Pediatrics, National Cancer Center, Goyang,
Korea
| | - Hyeon Jin Park
- Center for Pediatric Cancer, Department of Pediatrics, National Cancer Center, Goyang,
Korea
| | - Byung-Kiu Park
- Center for Pediatric Cancer, Department of Pediatrics, National Cancer Center, Goyang,
Korea
| | - Jun Ah Lee
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul,
Korea
| | - Jun Eun Park
- Department of Pediatrics, Korea University School of Medicine, Seoul,
Korea
| | - Soon Ki Kim
- Department of Pediatrics, Inha University Hospital, Inha University College of Medicine, Incheon,
Korea
| | - Ji Yoon Kim
- Department of Pediatrics, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu,
Korea
| | - Hyo Sun Kim
- Department of Pediatrics, Inje University Haeundae Paik Hospital, Busan,
Korea
| | - Youngeun Ma
- Department of Pediatrics, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam,
Korea
| | - Kyung Duk Park
- Department of Pediatrics and Research Institute of Clinical Medicine of Jeonbuk National University-Jeonbuk National University Hospital, Jeonbuk National University Medical School, Jeonju,
Korea
| | - Sang Kyu Park
- Department of Pediatrics, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan,
Korea
| | - Eun Sil Park
- Department of Pediatrics, Gyeongsang National University School of Medicine, Jinju,
Korea
| | - Ye Jee Shim
- Department of Pediatrics, Keimyung University Dongsan Hospital, Keimyung University School of Medicine, Daegu,
Korea
| | - Eun Sun Yoo
- Department of Pediatrics, Ewha Womans University Seoul Hospital, Ewha Womans University College of Medicine, Seoul,
Korea
| | - Kyung Ha Ryu
- Department of Pediatrics, Ewha Womans University Seoul Hospital, Ewha Womans University College of Medicine, Seoul,
Korea
| | - Jae Won Yoo
- Department of Pediatrics, Chungnam National University College of Medicine, Daejeon,
Korea
| | - Yeon Jung Lim
- Department of Pediatrics, Chungnam National University College of Medicine, Daejeon,
Korea
| | - Hoi Soo Yoon
- Department of Pediatrics, Kyung Hee University College of Medicine, Seoul,
Korea
| | - Mee Jeong Lee
- Department of Pediatrics, Dankook University College of Medicine, Cheonan,
Korea
| | - Jae Min Lee
- Department of Pediatrics, Yeungnam University College of Medicine, Daegu,
Korea
| | - In-Sang Jeon
- Department of Pediatrics, Gachon University Gil Medical Center, Incheon,
Korea
| | - Hye Lim Jung
- Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Hee Won Chueh
- Department of Pediatrics, Dong-A University College of Medicine, Busan,
Korea
| | - Seunghyun Won
- Medical Research Collaborating Center, Seoul National University Bundang Hospital, Seongnam,
Korea
| | | |
Collapse
|
27
|
Zhong Z, Jiang H, Chen H, Wu C, Wang Y, Zhang Z, Li J, Liu J. Ex vivo tumor dissection followed by kidney autotransplantation in bilateral wilms tumor. Front Pediatr 2023; 11:1120797. [PMID: 36816368 PMCID: PMC9936068 DOI: 10.3389/fped.2023.1120797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 01/12/2023] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Successful management of bilateral Wilm's tumor (BWT) involves a radical resection while preserving enough normal kidney tissue. Nephron-sparing surgery often results in an R1/R2 resection with a high recurrence rate in children with huge or multiple tumors, or tumors proximity to the renal hilum. In contrast, kidney autotransplantation can completely resect the tumor while maintaining homeostasis and preserving the patient's healthy kidney tissues. METHODS We summarized the clinical data of 8 synchronous BWT patients who underwent kidney autotransplantation at the First Affiliated Hospital of Sun Yat-sen University from 2018 to 2020. Ex vivo tumor resection and kidney autotransplantions were performed on 11 kidneys. The baseline characteristics, perioperative management, and survival status were reported. RESULTS Nephron-sparing surgeries were performed on 5 kidneys in vivo. Among all the 8 patients, six of them (75%) received staged operation and the other 2 patients (25%) received single-stage operation. No residual tumors were found on the postoperative imaging in all the 8 patients. In total, 6 (75%) patients occurred complications after the autotransplantation, among which, 2 (33.3%) patients had complication of Clavien-Dindo grade IIIa, and 4 (66.7%) patients had complication of grade < 3. During the 38 months of follow-up, 87.5% (7/8) of patients were tumor-free survival with normal renal function. One patient died from renal failure without tumor recurrence. DISCUSSION Therefore, our study indicated that autologous kidney transplantation can be an option for patients with complex BWT if the hospital's surgical technique and perioperative management conditions are feasible.
Collapse
Affiliation(s)
- Zhihai Zhong
- Department of Pediatric Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong Jiang
- Department of Pediatric Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huadong Chen
- Department of Pediatric Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chenglin Wu
- Department of Organ Transplantation, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuanqi Wang
- Department of Pediatric Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhichong Zhang
- Department of Pediatric Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Li
- Department of Organ Transplantation, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Juncheng Liu
- Department of Pediatric Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Tao J, Yang H, Hao Z, Liang C, Du Y, Zhang C, Yin Y, Zhou J. Positive response of a recurrent clear cell sarcoma to anlotinib combined with chemotherapy: A case report. Medicine (Baltimore) 2022; 101:e32109. [PMID: 36482604 PMCID: PMC9726286 DOI: 10.1097/md.0000000000032109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
RATIONALE Renal clear cell sarcoma is a rare and highly invasive malignant renal tumor that easily relapses after treatment. Recurrent recurrent clear cell carcinoma (CCSK) responds poorly to chemotherapy and has no established standardized treatment, and need to be explored potentially useful treatments. PATIENT CONCERNS A 18-years-old patient with renal clear cell sarcoma recurrence after open radical nephrectomy. DIAGNOSIS Recurrent clear cell sarcoma. INTERVENTIONS After chemotherapy alone failed, the patient received 6 courses of anlotinib combined with chemotherapy. The tumor had significantly reduced in size and the recurrent tumor and part of the liver were resected. OUTCOMES No tumor recurrence or metastasis was detected during the follow-up 8 months after the operation. LESSONS This is the first report describing the use of anlotinib in treating CCSK. We believe that anlotinib combined with chemotherapy may be a useful treatment option for patients with recurrent CCSK.
Collapse
Affiliation(s)
- Junyue Tao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Hao Yang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Zongyao Hao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Yingying Du
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Chao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Yu Yin
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
- * Correspondence: Jun Zhou, Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, People’s Republic of China (e-mail: )
| |
Collapse
|
29
|
Brok JS, Shelmerdine S, Damsgaard F, Smets A, Irtan S, Swinson S, Hedayati V, Jacob J, Nair A, Oostveen M, Pritchard-Jones K, Olsen Ø. The clinical impact of observer variability in lung nodule classification in children with Wilms tumour. Pediatr Blood Cancer 2022; 69:e29759. [PMID: 35652617 PMCID: PMC7615195 DOI: 10.1002/pbc.29759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/10/2022] [Accepted: 04/18/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVES To investigate the extent to which observer variability of computed tomography (CT) lung nodule assessment may affect clinical treatment stratification in Wilms tumour (WT) patients, according to the recent Société Internationale d'Oncologie Pédiatrique Renal Tumour Study Group (SIOP-RTSG) UMBRELLA protocol. METHODS I: CT thoraces of children with WT submitted for central review were used to estimate size distribution of lung metastases. II: Scans were selected for blinded review by five radiologists to determine intra- and inter-observer variability. They assessed identical scans on two occasions 6 months apart. III: Monte Carlo simulation (MCMC) was used to predict the clinical impact of observer variation when applying the UMBRELLA protocol size criteria. RESULTS Lung nodules were found in 84 out of 360 (23%) children with WT. For 21 identified lung nodules, inter-observer limits of agreement (LOA) for the five readers were ±2.4 and ±1.4 mm (AP diameter), ±1.9 and ±1.8 mm (TS diameter) and ±2.0 and ±2.4 mm (LS diameter) at assessments 1 and 2. Intra-observer LOA across the three dimensions were ±1.5, ±2.2, ±3.5, ±3.1 and ±2.6 mm (readers 1-5). MCMC demonstrated that 17% of the patients with a 'true' nodule size of ≥3 mm will be scored as <3 mm, and 21% of the patients with a 'true' nodule size of <3 mm will be scored as being ≥3 mm. CONCLUSION A significant intra-inter observer variation was found when measuring lung nodules on CT for patients with WT. This may have significant implications on treatment stratification, and thereby outcome, when applying a threshold of ≥3 mm for a lung nodule to dictate metastatic status.
Collapse
Affiliation(s)
- Jesper Sune Brok
- Department of Paediatric Haematology and Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Susan Shelmerdine
- Department of Clinical Radiology, Great Ormond Street Hospital for Children NHS foundation Trust, London, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Frederikke Damsgaard
- Department of Paediatric Haematology and Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Anne Smets
- Department of Radiology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Sabine Irtan
- Department of Visceral and Neonatal Paediatric Surgery, Sorbonne University, Armand Trousseau Hospital - APHP, Paris, France
| | | | - Venus Hedayati
- King’s College Hospital NHS Foundation Trust, London, UK
| | - Joseph Jacob
- Centre for Medical Image Computing, University College London, London, UK
- Department of Respiratory Medicine, University College London, London, UK
| | - Arjun Nair
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Minou Oostveen
- UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Øystein Olsen
- Department of Clinical Radiology, Great Ormond Street Hospital for Children NHS foundation Trust, London, UK
| |
Collapse
|
30
|
Zhang Z, Gao X, Deng L, Jia W, Zhang J, Cheng J, Zhou H, Liu G, Fu W. Association between LIN28B gene polymorphisms and Wilms' tumor susceptibility. Biomark Med 2022; 16:1113-1120. [PMID: 36606447 DOI: 10.2217/bmm-2022-0291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Aim: To survey the association between LIN28B gene polymorphisms and the increased risk of Wilms' tumor (WT). Methods: Five LIN28B polymorphisms (rs314276 C>A, rs221634 A>T, rs221635 T>C, the rs4145418 A>C and rs9404590 T>G) were genotyped in 355 WT patients and 1070 healthy controls to assess the association. Result: The rs314276 CA/AA genotype was a protective factor against WT (corrected odds ratio [OR]: 0.71; p = 0.006). Individuals older than 18 months (corrected OR: 0.60; p = 0.001), males (corrected OR: 0.65; p = 0.011) and in clinical stage I + II patients (corrected OR: 0.60; p = 0.0008) with this genotype were less susceptible to WT. Conclusion: The rs314276 CA/AA genotype may protect against WT.
Collapse
Affiliation(s)
- Zhengtao Zhang
- Department of Pediatric Surgery, Guangzhou Women & Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, China
| | - Xiaofeng Gao
- Department of Pediatric Surgery, Guangzhou Women & Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, China
| | - Linqing Deng
- Department of Pediatric Surgery, Guangzhou Women & Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, China
| | - Wei Jia
- Department of Pediatric Surgery, Guangzhou Women & Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, China
| | - Jiao Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Jiwen Cheng
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Haixia Zhou
- Department of Hematology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Guochang Liu
- Department of Pediatric Surgery, Guangzhou Women & Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, China
| | - Wen Fu
- Department of Pediatric Surgery, Guangzhou Women & Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, China
| |
Collapse
|
31
|
Schulpen M, Roy P, Wijnen MHWA, Tytgat GAM, van den Heuvel-Eibrink MM, van Tinteren H, Karim-Kos HE. Incidence and survival of paediatric renal tumours in the Netherlands between 1990 and 2014. Eur J Cancer 2022; 175:282-290. [PMID: 36174300 DOI: 10.1016/j.ejca.2022.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/09/2022] [Accepted: 08/17/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND This population-based study is the first to provide a detailed analysis of trends in incidence and survival of children and adolescents diagnosed with renal malignancies in the Netherlands. METHODS Data on all renal malignancies diagnosed in paediatric patients (0-18 years) between 1990 and 2014 [N = 648, 92% Wilms tumour (WT)] were extracted from the Netherlands Cancer Registry. Five-year overall survival (OS) was estimated using the actuarial method. Time trends in incidence were assessed by calculating average annual percentage change. A parametric survival model was used to compare the multivariable-adjusted risk of dying from WT between two diagnostic periods. RESULTS The incidence was 8 per million person-years and was constant over time (average annual percentage change -0.8%, p = 0.29). Patients with WT had a favourable outcome in both time periods; 5-year OS was 88% in 1990-2001 and 91% in 2002-2014. Multivariable analysis showed that the risk of dying from WT was not significantly decreased in the latest period (hazard ratio, 95% CI: 0.7, 0.4-1.3). Five-year OS decreased with increasing disease stage, ranging from 95 to 100% for stage I-II and about 80% for stage III-IV to 74% for bilateral disease. Five-year OS were 81% for renal cell carcinoma, 77% for clear cell sarcoma of the kidney and 20% for malignant rhabdoid tumour of the kidney. CONCLUSIONS Incidence of paediatric renal malignancies in the Netherlands has been stable since the 1990s. Five-year OS of WT reached 91% and was similar to findings for other developed countries. Contrary to the excellent outcome for WT, the outcome of malignant rhabdoid tumour of the kidney remained inferior.
Collapse
Affiliation(s)
- Maya Schulpen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Prakriti Roy
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Marc H W A Wijnen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | | | - Harm van Tinteren
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Henrike E Karim-Kos
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands.
| |
Collapse
|
32
|
Tian XM, Xiang B, Jin LM, Mi T, Wang JK, Zhanghuang C, Zhang ZX, Chen ML, Shi QL, Liu F, Lin T, Wei GH. Immune-related gene signature associates with immune landscape and predicts prognosis accurately in patients with Wilms tumour. Front Immunol 2022; 13:920666. [PMID: 36172369 PMCID: PMC9510599 DOI: 10.3389/fimmu.2022.920666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Wilms tumour (WT) is the most common kidney malignancy in children. Chemoresistance is the leading cause of tumour recurrence and poses a substantial therapeutic challenge. Increasing evidence has underscored the role of the tumour immune microenvironment (TIM) in cancers and the potential for immunotherapy to improve prognosis. There remain no reliable molecular markers for reflecting the immune landscape and predicting patient survival in WT. Here, we examine differences in gene expression by high-throughput RNA sequencing, focused on differentially expressed immune-related genes (IRGs) based on the ImmPort database. Via univariate Cox regression analysis and Lasso-penalized Cox regression analysis, IRGs were screened out to establish an immune signature. Kaplan-Meier curves, time-related ROC analysis, univariate and multivariate Cox regression studies, and nomograms were used to evaluate the accuracy and prognostic significance of this signature. Furthermore, we found that the immune signature could reflect the immune status and the immune cell infiltration character played in the tumour microenvironment (TME) and showed significant association with immune checkpoint molecules, suggesting that the poor outcome may be partially explained by its immunosuppressive TME. Remarkably, TIDE, a computational method to model tumour immune evasion mechanisms, showed that this signature holds great potential for predicting immunotherapy responses in the TARGET-wt cohort. To decipher the underlying mechanism, GSEA was applied to explore enriched pathways and biological processes associated with immunophenotyping and Connectivity map (CMap) along with DeSigN analysis for drug exploration. Finally, four candidate immune genes were selected, and their expression levels in WT cell lines were monitored via qRT-PCR. Meanwhile, we validated the function of a critical gene, NRP2. Taken together, we established a novel immune signature that may serve as an effective prognostic signature and predictive biomarker for immunotherapy response in WT patients. This study may give light on therapeutic strategies for WT patients from an immunological viewpoint.
Collapse
Affiliation(s)
- Xiao-Mao Tian
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Bin Xiang
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Li-Ming Jin
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Tao Mi
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Jin-Kui Wang
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Chenghao Zhanghuang
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Zhao-Xia Zhang
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Mei-Ling Chen
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Qin-Lin Shi
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Feng Liu
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
- *Correspondence: Feng Liu,
| | - Tao Lin
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Guang-Hui Wei
- Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| |
Collapse
|
33
|
Habachi G, Sahli S, Ammar SB, Jouini R. Rhabdoid renal tumor: an aggressive embryonal tumor in an infant — a case report. ANNALS OF PEDIATRIC SURGERY 2022. [DOI: 10.1186/s43159-022-00200-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Malignant rhabdoid tumor of the kidney is the most aggressive childhood renal tumor. A preoperative diagnosis is critical in order to correctly establish a therapeutic strategy and a full metastatic workup.
Case presentation
We report on a 3-month-old case with fever, diarrhea, and abdominal distension treated surgically with adjuvant chemotherapy. The diagnosis was confirmed postoperatively. Relapse was quick, and the child died 5 months after surgical resection.
Conclusions
Rhabdoid renal tumor in young age is associated with a high mortality rate even with invasive strategies. Case reports and research are critical for evaluating existing protocols and improving prognosis. Diverse clinical trials are being conducted in the hopes of improving the prognosis of rhabdoid renal tumors.
Collapse
|
34
|
Behfar M, Muhammadnejad S, Abdolahi S, Mohseni R, Shoae-Hassani A, Monzavi SM, Hamidieh AA. Adoptive NK-cell transfer as a potential treatment paradigm for Wilms tumor: A preclinical study. Pediatr Blood Cancer 2022; 69:e29676. [PMID: 35441789 DOI: 10.1002/pbc.29676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 02/15/2022] [Accepted: 03/05/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Natural killer (NK) cell therapy has been shown to be effective in the treatment of some cancers. However, the effects of this adoptive immunotherapy have not been investigated for Wilms tumor (WT). In this study, the effects of adoptive NK-cell transfer on a patient-derived xenograft (PDX) model of anaplastic WT were evaluated, and the impacts of cell source and ex vivo activation strategy on the therapeutic efficacy of NK-cell product were appraised. METHODS NK cells were isolated from human peripheral blood mononuclear cells (NKPB ) and human cord blood (NKCB ), and were expanded and activated using a cytokine cocktail. Another group of NK cells (NKET ) was produced through activation with the exosomes extracted from previously challenged NKPB cells with WT. PDX-bearing mice were treated with clinically relevant doses of NKPB , NKCB , NKET , standard chemotherapy, and placebo (phosphate-buffered saline). RESULTS PDX models treated with NKCB showed a better survival rate, though the difference among the study groups was not significant. Compared with the placebo control group, NKCB significantly improved the histopathologic response, NKPB significantly inhibited the proliferation of neoplastic cells, and NKET led to a significant decrease in the metastasis score (all p-values <.05). Standard chemotherapy provided the greatest tumor growth inhibition and the lowest mitotic count, though it did not show any significant advantage over NK-cell therapies in any of the outcome parameters in two-by-two comparisons. CONCLUSIONS This study spotlights the efficacy of adoptive NK-cell transfer as a potential treatment candidate for high-risk WT.
Collapse
Affiliation(s)
- Maryam Behfar
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Samad Muhammadnejad
- Gene Therapy Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahrokh Abdolahi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rashin Mohseni
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Shoae-Hassani
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mostafa Monzavi
- Gene Therapy Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Theilen TM, Braun Y, Bochennek K, Rolle U, Fiegel HC, Friedmacher F. Multidisciplinary Treatment Strategies for Wilms Tumor: Recent Advances, Technical Innovations and Future Directions. Front Pediatr 2022; 10:852185. [PMID: 35911825 PMCID: PMC9333359 DOI: 10.3389/fped.2022.852185] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Significant progress has been made in the management of Wilms tumor (WT) in recent years, mostly as a result of collaborative efforts and the implementation of protocol-driven, multimodal therapy. This article offers a comprehensive overview of current multidisciplinary treatment strategies for WT, whilst also addressing recent technical innovations including nephron-sparing surgery (NSS) and minimally invasive approaches. In addition, surgical concepts for the treatment of metastatic disease, advances in tumor imaging technology and potentially prognostic biomarkers will be discussed. Current evidence suggests that, in experienced hands and selected cases, laparoscopic radical nephrectomy and laparoscopic-assisted partial nephrectomy for WT may offer the same outcome as the traditional open approach. While NSS is the standard procedure for bilateral WT, NSS has evolved as an alternative technique in patients with smaller unilateral WT and in cases with imminent renal failure. Metastatic disease of the lung or liver that is associated with WT is preferably treated with a three-drug chemotherapy and local radiation therapy. However, surgical sampling of lung nodules may be advisable in persistent nodules before whole lung irradiation is commenced. Several tumor markers such as loss of heterozygosity of chromosomes 1p/16q, 11p15 and gain of function at 1q are associated with an increased risk of recurrence or a decreased risk of overall survival in patients with WT. In summary, complete resection with tumor-free margins remains the primary surgical aim in WT, while NSS and minimally invasive approaches are only suitable in a subset of patients with smaller WT and low-risk disease. In the future, advances in tumor imaging technology may assist the surgeon in defining surgical resection margins and additional biomarkers may emerge as targets for development of new diagnostic tests and potential therapies.
Collapse
Affiliation(s)
- Till-Martin Theilen
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Yannick Braun
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Konrad Bochennek
- Division of Pediatric Hematology and Pediatric Oncology, Hospital for Children and Adolescents, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Udo Rolle
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Henning C. Fiegel
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Florian Friedmacher
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
36
|
Li Z, Xu H, Yu L, Wang J, Meng Q, Mei H, Cai Z, Chen W, Huang W. Patient-derived renal cell carcinoma organoids for personalized cancer therapy. Clin Transl Med 2022; 12:e970. [PMID: 35802820 PMCID: PMC9270001 DOI: 10.1002/ctm2.970] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/02/2022] [Accepted: 06/23/2022] [Indexed: 12/11/2022] Open
Abstract
Background Kidney cancer is one of the most common solid tumors. The advancement of human kidney cancer research and treatment has been hindered by a lack of research models that faithfully recapitulate the diversity of the disease. Methods We established an effective three‐dimensional culture system for generating kidney cancer organoids from clinical renal cell carcinoma samples. Renal cell carcinoma (RCC) organoids were characterized by H&E staining, immunofluorescence, whole‐exome sequencing, RNA sequencing and single‐cell RNA sequencing. The use of RCC organoids in personalized cancer therapy was assessed by testing their responses to treatment drugs and chimeric antigen receptor T cells. Results Using this organoid culture system, 33 kidney cancer organoid lines from common kidney cancer subtypes, including clear cell renal cell carcinoma (ccRCC), papillary renal cell carcinoma (pRCC), and chromophobe renal cell carcinoma (chRCC), were generated. RCC organoids preserved the histological architectures, mutational landscapes, and transcriptional profile of the parental tumor tissues. Single‐cell RNA‐sequencing revealed inter‐ and intra‐tumoral heterogeneity in RCC organoids. RCC organoids allowed for in vitro drug screening and provided a tool for assessing the efficacy of chimeric antigen receptor T cells. Conclusions Patient‐derived RCC organoids are valuable pre‐clinical models for academic research and personalized medicine.
Collapse
Affiliation(s)
- Zhichao Li
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Haibo Xu
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Lei Yu
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jia Wang
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qian Meng
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Hongbing Mei
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhiming Cai
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Wei Chen
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Weiren Huang
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
37
|
Doganis D, Karalexi MA, Panagopoulou P, Bouka P, Bouka E, Markozannes G, Ntzani EE, Steliarova-Foucher E, Petridou ET. Incidence patterns of childhood non-Wilms renal tumors: Comparing data of the Nationwide Registry of Childhood Hematological Malignancies and Solid Tumors (NARECHEM-ST), Greece, and the Surveillance, Epidemiology, and End Results Program (SEER), USA. Cancer Epidemiol 2022; 78:102153. [PMID: 35390585 DOI: 10.1016/j.canep.2022.102153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/21/2022] [Accepted: 03/26/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND We used, for the first time, data registered in the Nationwide Registry for Childhood Hematological Malignancies and Solid Tumors (NARECHEM-ST)-Greece to estimate incidence/time trends of the rare childhood (0-14 years) non-Wilms tumors (non-WT), and compared the results of malignant non-WT to those from the Surveillance, Epidemiology, and End Results Program (SEER)-USA. METHODS Fifty-five cases (n = 33 malignant-only) were extracted from NARECHEM-ST (2001-2020) and 332 malignant cases from SEER (1990-2017). To allow between-country comparisons, age-standardized incidence rates (AIR) of malignant-only non-WT were calculated, and temporal trends were evaluated using Poisson and joinpoint regressions. RESULTS In NARECHEM-ST, malignant and non-malignant non-WT accounted for 22.6% of all renal tumors. Among malignant tumors, the AIR was 1.0/106 children in Greece, similar to that calculated for SEER, USA (AIR=0.9/106). The proportion of infant malignant and non-malignant non-WT was 27% (20% before 6 months) in NARECHEM-ST. Most common non-WT in Greece were congenital mesoblastic nephromas (CMN) diagnosed mainly in infancy (CIR=7.2/106). The proportion of infant malignant non-WT was 20% in SEER (AIRinfancy=2.5/106), mainly attributed to rhabdoid tumors (CIR=1.6/106). The male-to-female (M:F) ratio of malignant non-WT was 0.9 in NARECHEM-ST vs. 1.2 in SEER, whereas boys outnumbered girls with clear cell sarcoma in NARECHEM-ST (M:F=4.0). Lastly, significantly increasing trends in incidence rates were noted in NARECHEM-ST [+ 6.8%, 95% confidence intervals (CI): 0.5, 13.3] and in SEER (+7.3%, 95%CI: 5.6, 9.0). CONCLUSIONS Observed incidence, time trends and sociodemographic variations of non-WT may reflect differential registration practices and healthcare delivery patterns including differences regarding surveillance, coding and treatment practices.
Collapse
Affiliation(s)
- Dimitrios Doganis
- Hellenic Society for Social Pediatrics and Health Promotion, Athens, Greece; Department of Pediatric Hematology-Oncology, "P&A Kyriakou" Children's Hospital, Athens, Greece
| | - Maria A Karalexi
- Hellenic Society for Social Pediatrics and Health Promotion, Athens, Greece; Department of Hygiene and Epidemiology, School of Medicine, University of Ioannina, Ioannina, Greece
| | | | - Panagiota Bouka
- Hellenic Society for Social Pediatrics and Health Promotion, Athens, Greece
| | - Evdoxia Bouka
- Hellenic Society for Social Pediatrics and Health Promotion, Athens, Greece
| | - Georgios Markozannes
- Department of Hygiene and Epidemiology, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Evangelia E Ntzani
- Department of Hygiene and Epidemiology, School of Medicine, University of Ioannina, Ioannina, Greece; Center for Evidence Synthesis in Health, Brown University School of Public Health, Providence, RI, USA
| | - Eva Steliarova-Foucher
- Section of Cancer Surveillance, International Agency for Research on Cancer (IARC), Lyon, France
| | - Eleni Th Petridou
- Hellenic Society for Social Pediatrics and Health Promotion, Athens, Greece; Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
38
|
Ali S, Latif T, Sheikh MA, Shafiq MB, Zahra DEZ, Abu Bakar M. Review of Perioperative Care Pathway for Children With Renal Tumors. Cureus 2022; 14:e24928. [PMID: 35706749 PMCID: PMC9187842 DOI: 10.7759/cureus.24928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2022] [Indexed: 11/05/2022] Open
|
39
|
Atwa AM, Hafez AT, Abdelhameed M, Dawaba M, Nabeeh A, Helmy TE. Does immunohistochemical staining of P53, Ki 67 and cyclin A accurately predict Wilms tumor recurrence and survival? Arab J Urol 2022; 20:107-114. [PMID: 35935912 PMCID: PMC9354635 DOI: 10.1080/2090598x.2022.2058240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/12/2022] [Indexed: 11/02/2022] Open
Abstract
Objective To evaluate whether p53, cyclin A and ki67 immunohistochemical (IHC) assay can be used as predictors for Wilms' tumor (WT) unfavorable outcomes. Methods It is a non-concurrent cohort study including patients who underwent nephrectomy for WT from January 2000 to December 2015 in a tertiary referral center. Over a 5- year follow-up, unfavorable events, including relapse and cancer-specific mortality (CSM), were recorded. P53, cyclin A, and ki67 IHC assay were carried out for formalin-fixed paraffin-embedded WT samples. Results After excluding those who did not meet the inclusion criteria, 75 patients were enrolled. Of the patients, 15/75 (20%) experienced WT relapse while 11/75 (14.6%) died of WT over five years. Unfavorable histology (UFH), including prominent blastemal components and anaplasia, was found in 15/75 (20%) children.Cyclin A immunopositivity was associated with high rates of relapse and CSM. P53 and ki67 positive IHC assay did not show any statistically significant association with unfavorable outcomes. Other risk factors e.g. advanced staging, UFH, extracapsular extension, tumor rupture, lymphadenopathy, and venous thrombosis were not associated with poor prognosis. However, the presence of residual tumors was accompanied by lower survival rates. Conclusion Cyclin A IHC assay can be used as a predictor of WT recurrence and CSM. Further studies with prospective patterns and a larger sample size are needed.Abbreviations: WT: Wilms' tumor, UFH: unfavorable histology, IHC: immunohistochemical, PI: proliferation index, RFS: relapse-free survival, CSS: cancer-specific survival, FH: favorable histology, CSM: cancer-specific mortality, CDK: cyclin-dependent kinase.
Collapse
Affiliation(s)
- Ahmed M Atwa
- Urology department, Urology and Nephrology Center, Mansoura University, Egypt
| | - Ashraf T Hafez
- Urology department, Urology and Nephrology Center, Mansoura University, Egypt
| | - Mohamed Abdelhameed
- Pathology department, Urology and Nephrology Center, Mansoura University, Egypt
| | - Mohamed Dawaba
- Urology department, Urology and Nephrology Center, Mansoura University, Egypt
| | - Adel Nabeeh
- Urology department, Urology and Nephrology Center, Mansoura University, Egypt
| | - Tamer E Helmy
- Urology department, Urology and Nephrology Center, Mansoura University, Egypt
| |
Collapse
|
40
|
van der Perk MEM, Cost NG, Bos AME, Brannigan R, Chowdhury T, Davidoff AM, Daw NC, Dome JS, Ehrlich P, Graf N, Geller J, Kalapurakal J, Kieran K, Malek M, McAleer MF, Mullen E, Pater L, Polanco A, Romao R, Saltzman AF, Walz AL, Woods AD, van den Heuvel-Eibrink MM, Fernandez CV. White paper: Onco-fertility in pediatric patients with Wilms tumor. Int J Cancer 2022; 151:843-858. [PMID: 35342935 PMCID: PMC9541948 DOI: 10.1002/ijc.34006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/11/2022] [Accepted: 02/17/2022] [Indexed: 11/18/2022]
Abstract
The survival of childhood Wilms tumor is currently around 90%, with many survivors reaching reproductive age. Chemotherapy and radiotherapy are established risk factors for gonadal damage and are used in both COG and SIOP Wilms tumor treatment protocols. The risk of infertility in Wilms tumor patients is low but increases with intensification of treatment including the use of alkylating agents, whole abdominal radiation or radiotherapy to the pelvis. Both COG and SIOP protocols aim to limit the use of gonadotoxic treatment, but unfortunately this cannot be avoided in all patients. Infertility is considered one of the most important late effects of childhood cancer treatment by patients and their families. Thus, timely discussion of gonadal damage risk and fertility preservation options is important. Additionally, irrespective of the choice for preservation, consultation with a fertility preservation (FP) team is associated with decreased patient and family regret and better quality of life. Current guidelines recommend early discussion of the impact of therapy on potential fertility. Since most patients with Wilms tumors are prepubertal, potential FP methods for this group are still considered experimental. There are no proven methods for FP for prepubertal males (testicular biopsy for cryopreservation is experimental), and there is just a single option for prepubertal females (ovarian tissue cryopreservation), posing both technical and ethical challenges. Identification of genetic markers of susceptibility to gonadotoxic therapy may help to stratify patient risk of gonadal damage and identify patients most likely to benefit from FP methods.
Collapse
Affiliation(s)
| | - Nicholas G Cost
- Department of Surgery, Division of Urology, University of Colorado School of Medicine and the Surgical Oncology Program of the Children's Hospital Colorado, Aurora, CO, USA
| | - Annelies M E Bos
- University Medical Center Utrecht, Reproductive Medicine and Gynaecology, Utrecht, Netherlands
| | - Robert Brannigan
- Department of Urology, Northwestern University, Chicago, Illinois, USA
| | - Tanzina Chowdhury
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, USA
| | - Najat C Daw
- Department of Pediatrics - Patient Care, MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey S Dome
- Division of Oncology at Children's National Hospital, Washington, DC, USA
| | - Peter Ehrlich
- University of Michigan, C.S. Mott Children's Hospital Section of Pediatric Surgery, Ann Arbor, MI, USA
| | - Norbert Graf
- Department for Pediatric Oncology and Hematology, Saarland University Medical Center, Homburg, Germany
| | - James Geller
- Division of Pediatric Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - John Kalapurakal
- Department of Radiation Oncology, Northwestern University, Chicago, Illinois, USA
| | - Kathleen Kieran
- Department of Urology, University of Washington, and Division of Urology, Seattle Children's Hospital, Seattle, USA
| | - Marcus Malek
- Division of Pediatric General and Thoracic Surgery, UPMC Children's Hospital of Pittsburgh, Pittsburgh, USA
| | - Mary F McAleer
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth Mullen
- Department of Pediatric Oncology, Children's Hospital Boston/Dana-Farber Cancer Institute, Boston, MA, USA
| | - Luke Pater
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Angela Polanco
- National Cancer Research Institute Children's Group Consumer Representative, London, UK
| | - Rodrigo Romao
- Departments of Surgery and Urology, IWK Health Centre, Dalhousie University, Halifax, Canada
| | | | - Amy L Walz
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, USA
| | - Andrew D Woods
- Children's Cancer Therapy Development Institute, Beaverton, Oregon, USA
| | | | - Conrad V Fernandez
- Department of Pediatric Hematology/Oncology, IWK Health Centre and Dalhousie University, Halifax, Canada
| |
Collapse
|
41
|
Virtual Resection: A New Tool for Preparing for Nephron-Sparing Surgery in Wilms Tumor Patients. Curr Oncol 2022; 29:777-784. [PMID: 35200565 PMCID: PMC8870999 DOI: 10.3390/curroncol29020066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/26/2022] [Accepted: 01/29/2022] [Indexed: 12/03/2022] Open
Abstract
Nephron-sparing surgery (NSS) in Wilms tumor (WT) patients is a surgically challenging procedure used in highly selective cases only. Virtual resections can be used for preoperative planning of NSS to estimate the remnant renal volume (RRV) and to virtually mimic radical tumor resection. In this single-center evaluation study, virtual resection for NSS planning and the user experience were evaluated. Virtual resection was performed in nine WT patient cases by two pediatric surgeons and one pediatric urologist. Pre- and postoperative MRI scans were used for 3D visualization. The virtual RRV was acquired after performing virtual resection and a questionnaire was used to assess the ease of use. The actual RRV was derived from the postoperative 3D visualization and compared with the derived virtual RRV. Virtual resection resulted in virtual RRVs that matched nearly perfectly with the actual RRVs. According to the questionnaire, virtual resection appeared to be straightforward and was not considered to be difficult. This study demonstrated the potential of virtual resection as a new planning tool to estimate the RRV after NSS in WT patients. Future research should further evaluate the clinical relevance of virtual resection by relating it to surgical outcome.
Collapse
|
42
|
van Peer SE, Hol JA, van der Steeg AFW, van Grotel M, Tytgat GAM, Mavinkurve-Groothuis AMC, Janssens GOR, Littooij AS, de Krijger RR, Jongmans MCJ, Lilien MR, Drost J, Kuiper RP, van Tinteren H, Wijnen MHWA, van den Heuvel-Eibrink MM. Bilateral Renal Tumors in Children: The First 5 Years' Experience of National Centralization in The Netherlands and a Narrative Review of the Literature. J Clin Med 2021; 10:jcm10235558. [PMID: 34884260 PMCID: PMC8658527 DOI: 10.3390/jcm10235558] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/17/2021] [Accepted: 11/24/2021] [Indexed: 02/07/2023] Open
Abstract
Survival of unilateral Wilms tumors (WTs) is exceeding 90%, whereas bilateral WTs have an inferior outcome. We evaluated all Dutch patients with bilateral kidney tumors, treated in the first five years of national centralization and reviewed relevant literature. We identified 24 patients in our center (2015–2020), 23 patients had WT/nephroblastomatosis and one renal cell carcinoma. Patients were treated according to SIOP-RTSG protocols. Chemotherapy response was observed in 26/34 WTs. Nephroblastomatosis lesions were stable (n = 7) or showed response (n = 18). Nephron-sparing surgery was performed in 11/22 patients undergoing surgery (n = 2 kidneys positive margins). Local stage in 20 patients with ≥1 WT revealed stage I (n = 7), II (n = 4) and III (n = 9). Histology was intermediate risk in 15 patients and high risk in 5. Three patients developed a WT in a treated nephroblastomatosis lesion. Two of 24 patients died following toxicity and renal failure, i.e., respectively dialysis-related invasive fungal infection and septic shock. Genetic predisposition was confirmed in 18/24 patients. Our literature review revealed that knowledge is scarce on bilateral renal tumor patients with metastases and that radiotherapy seems important for local stage III patients. Bilateral renal tumors are a therapeutic challenge. We describe management and outcome in a national expert center and summarized available literature, serving as baseline for further improvement of care.
Collapse
Affiliation(s)
- Sophie E. van Peer
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
- Correspondence:
| | - Janna A. Hol
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
| | - Alida F. W. van der Steeg
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
| | - Martine van Grotel
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
| | - Godelieve A. M. Tytgat
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
| | - Annelies M. C. Mavinkurve-Groothuis
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
| | - Geert O. R. Janssens
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
- Department of Radiation Oncology, University Medical Center Utrecht (UMCU), Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Annemieke S. Littooij
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
- Department of Radiology and Nuclear Medicine, Wilhelmina’s Children Hospital, University Medical Center Utrecht (UMCU), Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Ronald R. de Krijger
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
- Department of Pathology, University Medical Center Utrecht (UMCU), Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Marjolijn C. J. Jongmans
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
- Department of Genetics, University Medical Center Utrecht (UMCU), Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Marc R. Lilien
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
- Department of Pediatric Nephrology, Wilhelmina’s Children Hospital, University Medical Center Utrecht (UMCU), Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Jarno Drost
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
- Oncode Institute, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - Roland P. Kuiper
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
- Department of Genetics, University Medical Center Utrecht (UMCU), Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Harm van Tinteren
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
| | - Marc H. W. A. Wijnen
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
| | - Marry M. van den Heuvel-Eibrink
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (J.A.H.); (A.F.W.v.d.S.); (M.v.G.); (G.A.M.T.); (A.M.C.M.-G.); (G.O.R.J.); (A.S.L.); (R.R.d.K.); (M.C.J.J.); (M.R.L.); (J.D.); (R.P.K.); (H.v.T.); (M.H.W.A.W.); (M.M.v.d.H.-E.)
| |
Collapse
|
43
|
Spreafico F, Fernandez CV, Brok J, Nakata K, Vujanic G, Geller JI, Gessler M, Maschietto M, Behjati S, Polanco A, Paintsil V, Luna-Fineman S, Pritchard-Jones K. Wilms tumour. Nat Rev Dis Primers 2021; 7:75. [PMID: 34650095 DOI: 10.1038/s41572-021-00308-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2021] [Indexed: 02/08/2023]
Abstract
Wilms tumour (WT) is a childhood embryonal tumour that is paradigmatic of the intersection between disrupted organogenesis and tumorigenesis. Many WT genes play a critical (non-redundant) role in early nephrogenesis. Improving patient outcomes requires advances in understanding and targeting of the multiple genes and cellular control pathways now identified as active in WT development. Decades of clinical and basic research have helped to gradually optimize clinical care. Curative therapy is achievable in 90% of affected children, even those with disseminated disease, yet survival disparities within and between countries exist and deserve commitment to change. Updated epidemiological studies have also provided novel insights into global incidence variations. Introduction of biology-driven approaches to risk stratification and new drug development has been slower in WT than in other childhood tumours. Current prognostic classification for children with WT is grounded in clinical and pathological findings and in dedicated protocols on molecular alterations. Treatment includes conventional cytotoxic chemotherapy and surgery, and radiation therapy in some cases. Advanced imaging to capture tumour composition, optimizing irradiation techniques to reduce target volumes, and evaluation of newer surgical procedures are key areas for future research.
Collapse
Affiliation(s)
- Filippo Spreafico
- Department of Medical Oncology and Hematology, Paediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Conrad V Fernandez
- Department of Paediatrics, IWK Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jesper Brok
- Department of Paediatric Haematology and Oncology, Rigshospitalet, Copenhagen, Denmark
| | - Kayo Nakata
- Cancer Control Center, Osaka International Cancer Institute, Osaka, Japan
| | | | - James I Geller
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Cincinnati, OH, USA
| | - Manfred Gessler
- Theodor-Boveri-Institute, Developmental Biochemistry, and Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Wuerzburg, Germany
| | - Mariana Maschietto
- Research Center, Boldrini Children's Hospital, Genetics and Molecular Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Angela Polanco
- National Cancer Research Institute Children's Group Consumer Representative, London, UK
| | - Vivian Paintsil
- Department of Child Health, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Sandra Luna-Fineman
- Division of Hematology, Oncology and Bone Marrow Transplantation, Department of Paediatrics, University of Colorado, Aurora, CO, USA
| | - Kathy Pritchard-Jones
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
44
|
Li W, Lingdi L, Xiqiang D, Jiheng L, Xin T, Qin H, Haisha L. MicroRNA-215-5p Inhibits the Proliferation and Migration of Wilm's Tumor Cells by Targeting CRK. Technol Cancer Res Treat 2021; 20:15330338211036523. [PMID: 34384283 PMCID: PMC8366128 DOI: 10.1177/15330338211036523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective: Wilm’s tumor is a common renal malignancy in childhood with unsatisfactory prognosis. microRNA-215-5p (miR-215-5p) has been reported as a tumor-suppressive miRNA in different types of human cancers, but rarely in the Wilm’s tumor. In light of this, we tried to investigate the regulatory role and underlying mechanism of miR-215-5p in the Wilm’s tumor. Methods: After sample collection and cell culture, the expression of miR-215-5p and CT10 Regulator of Kinase (CRK) was detected. Then rhabdoid tumor cell lines (formerly classified as Wilms’ tumor cell lines), G401 and WT-CLS1 cells were transfected with pcDNA3.1, pcDNA3.1-CRK, sh-NC, sh-CRK, agomir NC, miR-215-5p agomir, antagomir NC or miR-215-5p antagomir to explore the function of miR-215-5p and CRK in the Wilm’s tumor cell proliferation and migration. Moreover, the relationship between miR-215-5p and CRK was analyzed by dual luciferase reporter gene assay. Results: Lowly-expressed miR-215-5p and highly-expressed CRK were observed in the Wilm’s tumor tissues and cells. Transfection of pcDNA3.1-CRK or miR-215-5p antagomir could promote G401 and WT-CLS1 cell proliferation and enhance migration ability, while transfection of sh-CRK or miR-215-5p agomir led to opposite results. Additionally, miR-215-5p may bind to CRK. Moreover, transfection of pcDNA3.1-CRK in G401 and WT-CLS1 cells could partially reverse the inhibitory effect of miR-215-5p agomir on the proliferation and migration of Wilm’s tumor cells. Conclusion: Our study highlighted that miR-215-5p could suppress the proliferation and migration of Wilm’s tumor cells by regulating the expression of CRK, providing new ideas for molecular targeted therapy for Wilm’s tumor.
Collapse
Affiliation(s)
- Wang Li
- Children's Medical Center of The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Li Lingdi
- Children's Medical Center of The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Dang Xiqiang
- Children's Medical Center of The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Liu Jiheng
- Department of Hematology and Oncology, The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Tan Xin
- Children's Medical Center of The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Huang Qin
- Children's Medical Center of The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| | - Li Haisha
- Cardiac Function Department of The First Hospital of Changsha, Changsha, Hunan, People's Republic of China
| |
Collapse
|
45
|
van der Beek JN, Watson TA, Nievelstein RAJ, Brisse HJ, Morosi C, Lederman HM, Coma A, Gavra MM, Vult von Steyern K, Lakatos K, Breysem L, Varga E, Ducou Le Pointe H, Lequin MH, Schäfer JF, Mentzel HJ, Hötker AM, Calareso G, Swinson S, Kyncl M, Granata C, Aertsen M, Di Paolo PL, de Krijger RR, Graf N, Olsen ØE, Schenk JP, van den Heuvel-Eibrink MM, Littooij AS. MRI Characteristics of Pediatric Renal Tumors: A SIOP-RTSG Radiology Panel Delphi Study. J Magn Reson Imaging 2021; 55:543-552. [PMID: 34363274 PMCID: PMC9291546 DOI: 10.1002/jmri.27878] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The SIOP-Renal Tumor Study Group (RTSG) does not advocate invasive procedures to determine histology before the start of therapy. This may induce misdiagnosis-based treatment initiation, but only for a relatively small percentage of approximately 10% of non-Wilms tumors (non-WTs). MRI could be useful for reducing misdiagnosis, but there is no global consensus on differentiating characteristics. PURPOSE To identify MRI characteristics that may be used for discrimination of newly diagnosed pediatric renal tumors. STUDY TYPE Consensus process using a Delphi method. POPULATION Not applicable. FIELD STRENGTH/SEQUENCE Abdominal MRI including T1- and T2-weighted imaging, contrast-enhanced MRI, and diffusion-weighted imaging at 1.5 or 3 T. ASSESSMENT Twenty-three radiologists from the SIOP-RTSG radiology panel with ≥5 years of experience in MRI of pediatric renal tumors and/or who had assessed ≥50 MRI scans of pediatric renal tumors in the past 5 years identified potentially discriminatory characteristics in the first questionnaire. These characteristics were scored in the subsequent second round, consisting of 5-point Likert scales, ranking- and multiple choice questions. STATISTICAL TESTS The cut-off value for consensus and agreement among the majority was ≥75% and ≥60%, respectively, with a median of ≥4 on the Likert scale. RESULTS Consensus on specific characteristics mainly concerned the discrimination between WTs and non-WTs, and WTs and nephrogenic rest(s) (NR)/nephroblastomatosis. The presence of bilateral lesions (75.0%) and NR/nephroblastomatosis (65.0%) were MRI characteristics indicated as specific for the diagnosis of a WT, and 91.3% of the participants agreed that MRI is useful to distinguish NR/nephroblastomatosis from WT. Furthermore, all participants agreed that age influenced their prediction in the discrimination of pediatric renal tumors. DATA CONCLUSION Although the discrimination of pediatric renal tumors based on MRI remains challenging, this study identified some specific characteristics for tumor subtypes, based on the shared opinion of experts. These results may guide future validation studies and innovative efforts. LEVEL OF EVIDENCE 3 Technical Efficacy Stage: 3.
Collapse
Affiliation(s)
- Justine N van der Beek
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Tom A Watson
- Department of Paediatric Radiology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Rutger A J Nievelstein
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Carlo Morosi
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Henrique M Lederman
- Department of Diagnostic Imaging, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Ana Coma
- Department of Pediatric Radiology, Hospital Vall d'Hebron, Barcelona, Spain
| | - Maria M Gavra
- Department of Pediatric Radiology and Nuclear Medicine, 'Aghia Sophia' Children's Hospital, Athens, Greece
| | | | - Karoly Lakatos
- Department of Radiology, St. Anna Children's Hospital, University Clinic of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Luc Breysem
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Edit Varga
- Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | | | - Maarten H Lequin
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jürgen F Schäfer
- Division of Pediatric Radiology, Department of Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Hans-Joachim Mentzel
- Section of Pediatric Radiology, Institute of Diagnostic and Interventional Radiology, University Hospital Jena, Jena, Germany
| | - Andreas M Hötker
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Giuseppina Calareso
- Radiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sophie Swinson
- Department of Paediatric Radiology, Leeds Teaching Hospitals, Leeds, UK
| | - Martin Kyncl
- Department of Radiology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Claudio Granata
- Department of Paediatric Radiology, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Michael Aertsen
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | | | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Norbert Graf
- Department of Pediatric Oncology & Hematology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Øystein E Olsen
- Department of Paediatric Radiology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Jens-Peter Schenk
- Clinic of Diagnostic and Interventional Radiology, Division of Pediatric Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Annemieke S Littooij
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
46
|
Bellantoni AJ, Wagner LM. Pursuing Precision: Receptor Tyrosine Kinase Inhibitors for Treatment of Pediatric Solid Tumors. Cancers (Basel) 2021; 13:3531. [PMID: 34298746 PMCID: PMC8303693 DOI: 10.3390/cancers13143531] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/05/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Receptor tyrosine kinases are critical for the growth and proliferation of many different cancers and therefore represent a potential vulnerability that can be therapeutically exploited with small molecule inhibitors. Over forty small molecule inhibitors are currently approved for the treatment of adult solid tumors. Their use has been more limited in pediatric solid tumors, although an increasing number of single-agent and combination studies are now being performed. These agents have been quite successful in certain clinical contexts, such as the treatment of pediatric tumors driven by kinase fusions or activating mutations. By contrast, only modest activity has been observed when inhibitors are used as single agents for solid tumors that do not have genetically defined alterations in the target genes. The absence of predictive biomarkers has limited the wider applicability of these drugs and much work remains to define the appropriate patient population and clinical situation in which receptor tyrosine kinase inhibitors are most beneficial. In this manuscript, we discuss these issues by highlighting past trials and identifying future strategies that may help add precision to the use of these agents for pediatric extracranial solid tumors.
Collapse
Affiliation(s)
| | - Lars M. Wagner
- Division of Pediatric Hematology/Oncology, Duke University, Durham, NC 27710, USA;
| |
Collapse
|
47
|
Groenendijk A, Spreafico F, de Krijger RR, Drost J, Brok J, Perotti D, van Tinteren H, Venkatramani R, Godziński J, Rübe C, Geller JI, Graf N, van den Heuvel-Eibrink MM, Mavinkurve-Groothuis AMC. Prognostic Factors for Wilms Tumor Recurrence: A Review of the Literature. Cancers (Basel) 2021; 13:cancers13133142. [PMID: 34201787 PMCID: PMC8268923 DOI: 10.3390/cancers13133142] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 01/10/2023] Open
Abstract
Simple Summary A Wilms tumor is a childhood kidney tumor. In high-income countries, 90% of patients with this tumor survive. However, the tumor recurs in 15% of patients. It is important to identify the patients at risk of recurrence in order to adjust treatment in such a way that recurrence may potentially be prevented. However, we are currently unable to determine precisely which patients are at risk of recurrence. Therefore, we present an overview of factors that influence the risk of recurrence, also known as prognostic factors. These factors range from patient-, tumor- and treatment-related characteristics to geographic and socioeconomic factors. In addition to these factors, biological markers, such as genetic alterations, should be studied more intensively as these markers may be able to better identify patients at risk of tumor recurrence. Abstract In high-income countries, the overall survival of children with Wilms tumors (WT) is ~90%. However, overall, 15% of patients experience tumor recurrence. The adverse prognostic factors currently used for risk stratification (advanced stage, high risk histology, and combined loss of heterozygosity at 1p and 16q in chemotherapy-naïve WTs) are present in only one third of these cases, and the significance of these factors is prone to change with advancing knowledge and improved treatment regimens. Therefore, we present a comprehensive, updated overview of the published prognostic variables for WT recurrence, ranging from patient-, tumor- and treatment-related characteristics to geographic and socioeconomic factors. Improved first-line treatment regimens based on clinicopathological characteristics and advancing knowledge on copy number variations unveil the importance of further investigating the significance of biological markers for WT recurrence in international collaborations.
Collapse
Affiliation(s)
- Alissa Groenendijk
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (H.v.T.); (M.M.v.d.H.-E.); (A.M.C.M.-G.)
- Correspondence:
| | - Filippo Spreafico
- Department of Medical Oncology and Hematology, Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Ronald R. de Krijger
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (H.v.T.); (M.M.v.d.H.-E.); (A.M.C.M.-G.)
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (H.v.T.); (M.M.v.d.H.-E.); (A.M.C.M.-G.)
- Oncode Institute, 3584 CS Utrecht, The Netherlands
| | - Jesper Brok
- Department of Pediatric Oncology and Hematology, Rigshospitalet, 2100 Copenhagen, Denmark;
- Developmental Biology and Cancer Research and Teaching Department, University College London Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Daniela Perotti
- Molecular Bases of Genetic Risk and Genetic Testing Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Harm van Tinteren
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (H.v.T.); (M.M.v.d.H.-E.); (A.M.C.M.-G.)
| | | | - Jan Godziński
- Department of Pediatric Surgery, Marciniak Hospital, Fieldorfa 2, 54-049 Wroclaw, Poland;
- Department of Pediatric Traumatology and Emergency Medicine, Wroclaw Medical University, Bujwida 44a, 50-345 Wroclaw, Poland
| | - Christian Rübe
- Department of Radiation Oncology, Saarland University Medical Center and Saarland University Faculty of Medicine, D-66421 Homburg, Germany;
| | - James I. Geller
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA;
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Saarland University Medical Center and Saarland University Faculty of Medicine, D-66421 Homburg, Germany;
| | - Marry M. van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (H.v.T.); (M.M.v.d.H.-E.); (A.M.C.M.-G.)
| | - Annelies M. C. Mavinkurve-Groothuis
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (H.v.T.); (M.M.v.d.H.-E.); (A.M.C.M.-G.)
| |
Collapse
|
48
|
Mul J, van Grotel M, Seravalli E, Bosman ME, van Tinteren H, Roy P, Dávila Fajardo R, Tytgat GAM, Mavinkurve-Groothuis AMC, van de Ven CP, Wijnen MHWA, de Krijger RR, Littooij AS, van den Heuvel-Eibrink MM, Janssens GO. Locoregional control using highly conformal flank target volumes and volumetric-modulated arc therapy in pediatric renal tumors: Results from the Dutch national cohort. Radiother Oncol 2021; 159:249-254. [PMID: 33845042 DOI: 10.1016/j.radonc.2021.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/24/2021] [Accepted: 04/01/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE In pediatric renal tumors, conventional two opposing photon beams have been used to cover the postoperative flank target volume for decades. This single center study describes the locoregional outcome using highly conformal flank target volumes adjusted for postoperative changes and intra-fraction motion combined with Volumetric-Modulated Arc Therapy (VMAT). MATERIALS AND METHODS Between 01-2015 and 12-2019, 36/161 newly diagnosed patients with renal tumors underwent flank only irradiation (n = 30) or flank + whole lung irradiation (n = 6) using highly conformal target volumes in line with the SIOP-RTSG consensus statement. VMAT consisted of full-arc 10MV photon beams optimized for constraints of the organs at risk. In case of locoregional relapses, image co-registration and dose reconstruction was performed. Each relapse was classified as either 'infield' (V95%relapse: ≥99.0%), 'marginal' (V95%relapse: 20.0-98.9%) or 'outfield' (V95%relapse: 0-19.9%). RESULTS At a median follow-up from diagnosis of 3.1 years (range:0.4-5.7), the estimated 2-year Locoregional Control Rate, Disease-Free Interval and Overall Survival were 94%, 91% and 94%, respectively. Locoregional relapse was observed in two patients. One patient had a combined tumor bed and regional recurrence, classified as infield (V95%relapse: 100%) and outfield (V95%relapse: 1.2%). The second patient had a regional relapse in the inferior vena cava classified as marginal recurrence (V95%relapse: 93%). Relapses would not have been adequately covered by conventional beams. CONCLUSIONS This single center analysis provides encouraging evidence that excellent locoregional control can be obtained by using highly conformal flank target volumes with VMAT in pediatric renal tumors. The safety of this approach will be validated in a prospective multicenter study.
Collapse
Affiliation(s)
- Joeri Mul
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Department of Radiation Oncology, University Medical Center Utrecht, The Netherlands
| | | | - Enrica Seravalli
- Department of Radiation Oncology, University Medical Center Utrecht, The Netherlands
| | - Mirjam E Bosman
- Department of Radiation Oncology, University Medical Center Utrecht, The Netherlands
| | - Harm van Tinteren
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Prakriti Roy
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Raquel Dávila Fajardo
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Department of Radiation Oncology, University Medical Center Utrecht, The Netherlands
| | | | | | | | - Marc H W A Wijnen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Department of Pathology, University Medical Center Utrecht, The Netherlands
| | - Annemieke S Littooij
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Department of Radiology, University Medical Center Utrecht, The Netherlands
| | | | - Geert O Janssens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Department of Radiation Oncology, University Medical Center Utrecht, The Netherlands.
| |
Collapse
|
49
|
Liu L, Song Z, Gao XD, Chen X, Wu XB, Wang M, Hong YD. Identification of the potential novel biomarkers as susceptibility gene for Wilms tumor. BMC Cancer 2021; 21:316. [PMID: 33765954 PMCID: PMC7992941 DOI: 10.1186/s12885-021-08034-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/15/2021] [Indexed: 11/10/2022] Open
Abstract
Background Wilms tumor (WT) is the most common malignant renal tumor in children. The aim of this study was to identify potential susceptibility gene of WT for better prognosis. Methods Weighted gene coexpression network analysis is used for the detection of clinically important biomarkers associated with WT. Results In the study, 59 tissue samples from National Cancer Institute were pretreated for constructing gene co-expression network, while 224 samples also downloaded from National Cancer Institute were used for hub gene validation and module preservation analysis. Three modules were found to be highly correlated with WT, and 44 top hub genes were identified in these key modules eventually. In addition, both the module preservation analysis and gene validation showed ideal results based on other dataset with 224 samples. Meanwhile, Functional enrichment analysis showed that genes in module were enriched to sister chromatid cohesion, cell cycle, oocyte meiosis. Conclusion In summary, we established a gene co-expression network to identify 44 hub genes are closely to recurrence and staging of WT, and 6 of these hub genes was closely related to the poor prognosis of patients. Our findings revealed that those hub genes may be used as potential susceptibility gene for clinical diagnosis and prognosis of this tumor. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08034-w.
Collapse
Affiliation(s)
- Li Liu
- Department of Urology, The Second Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zhe Song
- Department of Urology, The Second Hospital, University of South China, Hengyang, 421001, Hunan, China.
| | - Xu-Dong Gao
- College of Health Science and Nursing, Wuhan Polytechnic University, Wuhan, 420000, China
| | - Xian Chen
- Department of Urology, The Second Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Xiao-Bin Wu
- Department of Urology, The Second Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Mi Wang
- Department of Urology, The Second Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Yu-De Hong
- Department of Urology, The Second Hospital, University of South China, Hengyang, 421001, Hunan, China
| |
Collapse
|
50
|
Guerreiro F, Seravalli E, Janssens G, Maduro J, Knopf A, Langendijk J, Raaymakers B, Kontaxis C. Deep learning prediction of proton and photon dose distributions for paediatric abdominal tumours. Radiother Oncol 2021; 156:36-42. [DOI: 10.1016/j.radonc.2020.11.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/09/2020] [Accepted: 11/23/2020] [Indexed: 11/16/2022]
|