1
|
Agrawal M, Chowhan AK. Paediatric renal tumors: An insight into molecular characteristics, histomorphology and syndromic association. World J Nephrol 2025; 14:99380. [DOI: 10.5527/wjn.v14.i2.99380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 12/19/2024] [Accepted: 01/14/2025] [Indexed: 04/09/2025] Open
Abstract
Paediatric renal tumors are rare and accounts for about 7% of all paediatric malignant tumors. The spectrum of paediatric renal tumors ranges from benign to malignant. Benign tumors include cystic nephroma, metanephric tumors and ossifying renal tumor of infancy. Tumor with low grade malignancy includes mesoblastic nephroma. Malignant tumors are nephroblastoma, clear cell sarcoma, malignant rhabdoid tumor, anaplastic sarcoma and Ewing sarcoma. Additionally, there are molecularly defined renal tumors, which includes renal cell carcinoma (RCC) with MiT translocations, ALK-rearranged RCC, eosinophilic solid and cystic RCC and SMARCB1- deficient renal medullary carcinoma. These tumors apart from having characteristic clinical presentation and histomorphology, also carry typical molecular mutations and translocations. Certain renal tumors have association with various genetic syndromes such as Beckwith-Weidmann syndrome, Wilm’s tumor, aniridia, genitourinary anomalies and mental retardation syndrome, Denys-Drash syndrome, rhabdoid tumor predisposition syndrome and DICER syndrome. This review article focusses on molecular characteristics, histomorphology and syndromic association of pediatric renal tumors, their immunohistochemical approach to diagnosis with recent updates in molecularly defined renal tumors.
Collapse
Affiliation(s)
- Mousmi Agrawal
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences (AIIMS), Raipur 492099, Chhattisgarh, India
| | - Amit K Chowhan
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences (AIIMS), Raipur 492099, Chhattisgarh, India
| |
Collapse
|
2
|
Huang M, Shu M, Xu Z, Wang L, Liu L, Liu J, Zhang H, Yang S, Wang C, Gao P. Pathological Insights into Non-Neoplastic Renal Parenchyma in Wilms Tumor: Implications for Nephron-Sparing Surgery. Eur J Pediatr Surg 2025; 35:187-194. [PMID: 39424346 DOI: 10.1055/s-0044-1791846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
INTRODUCTION This study aimed to evaluate the non-neoplastic renal parenchyma in Wilms tumor (WT) and investigate its impact on nephron-sparing surgery (NSS). MATERIALS AND METHODS The non-neoplastic renal parenchyma of WT patients was prospectively collected for pathological examination. The histology of non-neoplastic renal parenchyma was assessed from two perspectives: nephrogenic rests (NRs) and nephrons. RESULTS A total of 46 non-neoplastic renal parenchyma specimens were collected from 42 WT patients. The surgeons assessed the median proportion of non-neoplastic renal parenchyma as 30%, whereas using ellipsoid volume, it was calculated to be 27%. The Youden index of surgeons' assessment peaked at a 15% proportion of non-neoplastic renal parenchyma. The bilateral WT (BWT) group and NSS group exhibited significant differences compared with the unilateral WT group and radical nephrectomy group, respectively, with the BWT group showing a tendency toward thickened basement membrane. CONCLUSION The presence of NRs and endogenous nephron alternations should be given due attention in WT. The probability of abnormalities is low when the proportion of non-neoplastic renal parenchyma exceeds 15%, providing pathological support for expanding the adaptation of NSS.
Collapse
Affiliation(s)
- Mingchuan Huang
- Department of Pediatric Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, People's Republic of China
| | - Man Shu
- Department of Pathology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, People's Republic of China
| | - Zhe Xu
- Department of Pediatric Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, People's Republic of China
| | - Lin Wang
- Guangzhou KingMed Diagnostics Laboratory Group Co Ltd, Guangzhou, People's Republic of China
| | - Longshan Liu
- Organ Transplant Center, Sun Yat-sen University First Affiliated Hospital, Guangzhou, People's Republic of China
| | - Juncheng Liu
- Department of Pediatric Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, People's Republic of China
| | - Huanxi Zhang
- Organ Transplant Center, Sun Yat-sen University First Affiliated Hospital, Guangzhou, People's Republic of China
| | - Shicong Yang
- Department of Pathology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, People's Republic of China
| | - Changxi Wang
- Organ Transplant Center, Sun Yat-sen University First Affiliated Hospital, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, People's Republic of China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Sun Yat-sen University First Affiliated Hospital, Guangzhou, People's Republic of China
| | - Pengfei Gao
- Department of Pediatric Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, People's Republic of China
- Organ Transplant Center, Sun Yat-sen University First Affiliated Hospital, Guangzhou, People's Republic of China
| |
Collapse
|
3
|
Parsons LN, Treece AL. Emerging, Uncommon, and Aggressive Pediatric Kidney Tumors: An Update and Diagnostic Pitfalls. Surg Pathol Clin 2025; 18:269-279. [PMID: 40412826 DOI: 10.1016/j.path.2024.10.004] [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] [Indexed: 05/27/2025]
Abstract
Although favorable histology Wilms tumors are the most prevalent pediatric renal tumor, the remainder includes a small but diverse and important group of malignant neoplasms. Among these are diffuse anaplastic Wilms tumor, blastemal Wilms tumor, anaplastic sarcoma of kidney, and clear cell sarcoma of kidney. Given their rarity, these tumors may pose diagnostic challenges for pathologists, but accurate diagnosis is essential to ensuring these aggressive tumors are managed appropriately. This article summarizes the salient clinical, histopathological, and molecular features of these uncommon but important tumors.
Collapse
Affiliation(s)
- Lauren N Parsons
- Division of Pediatric Pathology, Medical College of Wisconsin, Children's Wisconsin, 9000 West Wisconsin Avenue, MS#701, Milwaukee, WI 53226, USA.
| | - Amy L Treece
- Department of Pathology and Laboratory Medicine, Children's of Alabama, Birmingham, AL, USA
| |
Collapse
|
4
|
Sutton KS, Walz AL, Groenendijk A, Murphy AJ, Pater L, Janssens GO, Brzezinski J, Mullen EA, Spreafico F, Godzinski J, van der Beek J, Hwa Wijnen M, van den Heuvel-Eibrink MM, Pachl M, Graf N, Ehrlich PF, Furtwangler R, Brok J, Geller J. Remaining Challenges in the Treatment of Relapsed Wilms Tumor: Children's Oncology Group and International Society of Paediatric Oncology Perspectives. Pediatr Blood Cancer 2025:e31790. [PMID: 40369656 DOI: 10.1002/pbc.31790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 04/10/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
Wilms tumor (WT) is the most common pediatric renal tumor, and with multidisciplinary treatment overall outcomes are excellent. However, a small subset of patients with WT will relapse. The ideal treatment of relapsed WT is yet to be defined. Ongoing studies through the Children's Oncology Group Renal Tumors Committee (COG-RTC) and the International Society of Paediatric Oncology Renal Tumor Study Group (SIOP-RTSG) aim to improve risk stratification and treatment strategies. Members met at the SIOP 55th Annual Congress 2023 to outline available data and knowledge gaps and develop future research priorities.
Collapse
Affiliation(s)
- Kathryn S Sutton
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Amy L Walz
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | - Andrew J Murphy
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Luke Pater
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Geert O Janssens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Radiation Oncology, University Medical Center, Utrecht, The Netherlands
| | - Jack Brzezinski
- Division of Paediatric Haematology/Oncology, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Elizabeth A Mullen
- Department of Pediatric Hematology/Oncology, Dana-Farber Cancer Institute/Boston Children's Hospital, Boston, Massachusetts, USA
| | - Filippo Spreafico
- Department of Oncology and Hematology, Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Jan Godzinski
- Department of Pediatric Surgery, Marciniak Hospital, Wroclaw, Poland
- Department of Pediatric Traumatology and Emergency Medicine, Medical University of Wroclaw, Wroclaw, Poland
| | - Justine 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
| | - Marc Hwa Wijnen
- Department of Surgery, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Max Pachl
- Department of Paediatric Surgery and Urology, Birmingham Children's Hospital, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Norbert Graf
- Department of Pediatric Hematology and Oncology, Saarland University, Homburg, Saarland, Germany
| | - Peter F Ehrlich
- Section of Pediatric Surgery, CS Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan, USA
| | - Rhoikos Furtwangler
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Inselspital University Hospital, Bern, Switzerland
| | - Jesper Brok
- Department of Paediatric Oncology and Haematology, Rigshospitalet, Copenhagen, Denmark
| | - James Geller
- Division of Pediatric Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
5
|
Meyerheim M, Panagiotidou F, Georgiadi E, Soudris D, Stamatakos G, Graf N. Exploring the in silico adaptation of the Nephroblastoma Oncosimulator to MRI scans, treatment data, and histological profiles of patients from different risk groups. Front Physiol 2025; 16:1465631. [PMID: 40313874 PMCID: PMC12043452 DOI: 10.3389/fphys.2025.1465631] [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: 07/16/2024] [Accepted: 03/20/2025] [Indexed: 05/03/2025] Open
Abstract
Introduction Nephroblastoma or Wilms' tumor is the most prevalent type of renal tumor in pediatric oncology. Although the overall survival rate for this condition is excellent today (∼90%), there have been no significant improvements over the past two decades. In silico models aim to simulate tumor progression and treatment responses over time; they hold immense potential for enhancing the predictive accuracy and optimizing treatment protocols as they are inspired by the digital twin paradigm. Methods The present study uses T2-weighted magnetic resonance images, chemotherapy treatment plans, and post-surgical histological profiles from three patients enrolled in the SIOP 2001/GPOH clinical trial, where each patient represents a distinct clinically assessed risk group. We investigated the clinical adaptation of the Nephroblastoma Oncosimulator to the datasets from these patients with the goal of deriving appropriate value distributions of the model input parameters that enable accurate prediction of tumor volume reduction in response to preoperative chemotherapy. Results Our primary focus was on the total cell kill ratio as a parameter reflecting treatment effectiveness. We derived the distribution of this parameter for one patient from each risk group: low (Mdn = 0.875, IQR [0.750, 0.875], n = 178), intermediate (Mdn = 0.875, IQR [0.750, 0.875], n = 175), and high (Mdn = 0.485, IQR [0.438, 0.532], n = 103). Statistically significant differences were observed between the high-risk group and both the low- and intermediate-risk groups (p < 0.001). Discussion The present work establishes a foundation for further studies using available retrospective datasets and additional patients per risk group. These efforts are expected to help validate the findings, advance model development, and extend this mechanistic multiscale discretized cancer model. However, clinical validation is ultimately required to assess the potential uses of the model in clinical decision-support systems.
Collapse
Affiliation(s)
- Marcel Meyerheim
- Pediatric Oncology and Hematology, Faculty of Medicine, Saarland University, Homburg, Germany
| | - Foteini Panagiotidou
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Eleni Georgiadi
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
- Biomedical Engineering Department, University of West Attica, Athens, Greece
| | - Dimitrios Soudris
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Georgios Stamatakos
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Norbert Graf
- Pediatric Oncology and Hematology, Faculty of Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
6
|
Perlman EJ, Webster F, Chang KTE, Coulomb A, Galluzzo L, Graf NS, Mullen EA, Okita H, O'Sullivan MJ, Somers GR, Treece A, Cohen M, Reyes-Múgica M. Data set for reporting paediatric renal tumours: recommendations from the international collaboration on cancer reporting (ICCR). Histopathology 2025. [PMID: 40231423 DOI: 10.1111/his.15450] [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: 04/16/2025]
Abstract
Tumours arising within the developing kidney of children vary widely in their histological appearance and outcome; optimal therapy requires accurate classification and staging. The two major paediatric cooperative groups provide different therapeutic protocols based on different staging and classification, initially developed to serve patients in North America and Europe, but also used in many other parts/regions of the world. The International Collaboration on Cancer Reporting (ICCR) has developed a structure whereby such complex information may be harmonised, and able to be applied to patients globally. An international expert panel consisting of paediatric pathologists and oncologists produced a set of items critical to cancer reporting and subjected these to review and discussion using the structured processes provided by the ICCR. A formal ICCR structure was assembled, and consensus surrounding elements and their application to different therapeutic protocols was developed. The data set underwent open international consultation. This resulted in the first international data set for Wilms tumour (WT) and other paediatric renal tumours, provided herein. The use of ICCR methods enables a full understanding of highly complex and often overlapping reporting elements by international experts, and the potential of developing a set of commonly applied data elements that are fully defined. This sets the groundwork for future consolidation of definitions and harmonisation of therapies for WT and other paediatric renal tumour patients. It also allows institutions outside the major paediatric cooperative groups to provide therapy based on known elements.
Collapse
Affiliation(s)
- E J Perlman
- Department of Pathology and Laboratory Medicine, The Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago, IL, USA
| | - F Webster
- International Collaboration on Cancer Reporting, Surry Hills, NSW, Australia
| | - K T E Chang
- Department of Pathology and Laboratory Medicine, Duke-NUS Medical School, K. K. Women's and Children's Hospital, Singapore, Singapore
| | - A Coulomb
- APHP-Sorbonne University, Paris, France
- Department of Pathology, Armand-Trousseau Children's Hospital, Paris, France
| | - L Galluzzo
- Pathology Department, Hospital Nacional de Pediatría Dr Professor J. P. Garrahan, Buenos Aires, Argentina
| | - N S Graf
- Histopathology Department, The Children's Hospital at Westmead, Westmead and University of Sydney, Sydney, NSW, Australia
| | - E A Mullen
- Division of Pediatric Hematology/Oncology, Dana-Farber Cancer Institute/Boston Children's Hospital, Boston, MA, USA
| | - H Okita
- Division of Diagnostic Pathology, Keio University School of Medicine, Tokyo, Japan
| | - M J O'Sullivan
- Department of Histopathology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - G R Somers
- Department of Paediatric Laboratory Medicine, Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - A Treece
- Department of Pathology, Children's Hospital of Alabama, Birmingham, AL, USA
| | - M Cohen
- Department of Pathology, Sheffield Children's Hospital NHS Foundation Trust, University of Sheffield, Western Bank, Sheffield, UK
| | - M Reyes-Múgica
- Department of Pathology and Laboratory Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
7
|
Glick RD, Romao RLP, Pachl M, Kotagal M, Buchanan AF, Murphy AJ, Tracy ET, Pio L, Cost NG, Godzinski J, Ehrlich PF. Current surgical approaches to pediatric renal tumors. Pediatr Blood Cancer 2025; 72 Suppl 2:e31118. [PMID: 38809413 PMCID: PMC11604818 DOI: 10.1002/pbc.31118] [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: 04/29/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Pediatric renal tumors are among the most common pediatric solid malignancies. Surgical resection is a key component in the multidisciplinary therapy for children with kidney tumors. Therefore, it is imperative that surgeons caring for children with renal tumors fully understand the current standards of care in order to provide appropriate surgical expertise within this multimodal framework. Fortunately, the last 60 years of international, multidisciplinary pediatric cancer cooperative group studies have enabled high rates of cure for these patients. This review will highlight the international surgical approaches to pediatric patients with kidney cancer to help surgeons understand the key differences and similarities between the European (International Society of Pediatric Oncology) and North American (Children's Oncology Group) recommendations.
Collapse
Affiliation(s)
- Richard D. Glick
- Division of Pediatric Surgery, Cohen Children’s Medical Center, Zucker School of Medicine at Northwell/Hofstra, New Hyde Park, NY
| | - Rodrigo LP Romao
- Divisions of Pediatric General Surgery and Pediatric Urology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Max Pachl
- Department of Paediatric Surgery and Urology, Birmingham Children’s Hospital, Birmingham Women’s and Children’s NHS Foundation Trust, Birmingham, UK
| | - Meera Kotagal
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children’s Hospital Medical Center; Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Amanda F. Buchanan
- University of Kentucky, Departments of Urology and Pediatrics, Lexington, KY
| | - Andrew J. Murphy
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN
| | - Elisabeth T. Tracy
- Division of Pediatric Surgery, Duke Children’s Hospital and Health Center, Durham, North Carolina
| | - Luca Pio
- Paediatric Surgery Unit, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Nicholas G. Cost
- Division of Urology, Department of Surgery, University of Colorado School of Medicine and the Surgical Oncology Program at Children’s Hospital Colorado, Aurora, CO
| | - Jan Godzinski
- Department of Pediatric Surgery, Marciniak Hospital, Wroclaw, Poland; Department of Pediatric Traumatology and Emergency Medicine, Medical University, Wroclaw, Poland
| | - Peter F. Ehrlich
- University of Michigan Department of Surgery, Section of Pediatric Surgery, Ann Arbor Michigan 48104, USA
| |
Collapse
|
8
|
Pachl M, Lautz TB, Aldrink JH, Abdelhafeez H, Irtan S. Minimally invasive and robotic-assisted approaches applied to pediatric surgical oncology. Pediatr Blood Cancer 2025; 72 Suppl 2:e31162. [PMID: 38987997 DOI: 10.1002/pbc.31162] [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: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024]
Abstract
The management of pediatric tumors is complex, with surgery, chemotherapy, and radiotherapy being cornerstones in their treatment. Tumor removal is increasingly performed by a minimally invasive approach, which allows for quicker postoperative recovery and less postoperative pain. The goal of this report is to give an overview of minimally invasive surgical approaches for common pediatric tumors, with a focus on technical considerations and postoperative outcomes.
Collapse
Affiliation(s)
- Max Pachl
- Department of Pediatric Surgery and Urology, Birmingham Children's Hospital, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Timothy B Lautz
- Division of Pediatric Surgery, Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern University, Chicago, Illinois, USA
| | - Jennifer H Aldrink
- Division of Pediatric Surgery, Department of Surgery, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Hafeez Abdelhafeez
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Sabine Irtan
- Department of Visceral and Neonatal Pediatric Surgery, Hôpital Armand Trousseau - APHP, Sorbonne University, Paris, France
| |
Collapse
|
9
|
Yang R, Xie L, Wang R, Li Y, Lu Y, Liu B, Dai S, Zheng S, Dong K, Dong R. Integration of single-nuclei and spatial transcriptomics to decipher tumor phenotype predictive of relapse-free survival in Wilms tumor. Front Immunol 2025; 16:1539897. [PMID: 40098972 PMCID: PMC11911335 DOI: 10.3389/fimmu.2025.1539897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Background Wilms tumor (WT) is the most common childhood renal malignancy, with recurrence linked to poor prognosis. Identifying the molecular features of tumor phenotypes that drive recurrence and discovering novel targets are crucial for improving treatment strategies and enhancing patient outcomes. Methods Single-nuclei RNA sequencing (snRNA-seq), spatial transcriptomics (ST), bulk RNA-seq, and mutation/copy number data were curated from public databases. The Seurat package was used to process snRNA-seq and ST data. Scissor analysis was applied to identify tumor subpopulations associated with poor relapse-free survival (RFS). Univariate Cox and LASSO analyses were utilized to reduce features. A prognostic ensemble machine learning model was developed. Immunohistochemistry was used to validate the expression of key features in tumor tissues. The CellChat and Commot package was utilized to infer cellular interactions. The PERCEPTION computational pipeline was used to predict the response of tumor cells to chemotherapy and targeted therapies. Results By integrating snRNA-seq and bulk RNA-seq data, we identified a subtype of Scissor+ tumor cells associated with poor RFS, predominantly derived from cap mesenchyme-like blastemal and fibroblast-like tumor subgroups. These cells displayed nephron progenitor signatures and cancer stem cell markers. A prognostic ensemble machine learning model was constructed based on the Scissor+ tumor signature to accurately predict patient RFS. TGFA was identified as the most significant feature in this model and validated by immunohistochemistry. Cellular communication analysis revealed strong associations between Scissor+ tumor cells and cancer-associated fibroblasts (CAFs) through IGF, SLIT, FGF, and PDGF pathways. ST data revealed that Scissor+ tumor cells were primarily located in immune-desert niche surrounded by CAFs. Despite reduced responsiveness to conventional chemotherapy, Scissor+ tumor cells were sensitive to EGFR inhibitors, providing insights into clinical intervention strategies for WT patients at high risk of recurrence. Conclusion This study identified a relapse-associated tumor subtype resembling nephron progenitor cells, residing in immune-desert niches through interactions with CAFs. The proposed prognostic model could accurately identify patients at high risk of relapse, offering a promising method for clinical risk stratification. Targeting these cells with EGFR inhibitors, in combination with conventional chemotherapy, may provide a potential therapeutic strategy for WT patients.
Collapse
Affiliation(s)
- Ran Yang
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
- Children’s Hospital of Fudan University (Xiamen Branch), Xiamen Children’s Hospital, Xiamen Key Laboratory of Pediatric General Surgery Diseases, Xiamen, China
| | - Lulu Xie
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Rui Wang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Li
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Yifei Lu
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Baihui Liu
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Shuyang Dai
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Shan Zheng
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
- Children’s Hospital of Fudan University (Xiamen Branch), Xiamen Children’s Hospital, Xiamen Key Laboratory of Pediatric General Surgery Diseases, Xiamen, China
| | - Kuiran Dong
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
- Children’s Hospital of Fudan University (Xiamen Branch), Xiamen Children’s Hospital, Xiamen Key Laboratory of Pediatric General Surgery Diseases, Xiamen, China
| | - Rui Dong
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
- Children’s Hospital of Fudan University (Xiamen Branch), Xiamen Children’s Hospital, Xiamen Key Laboratory of Pediatric General Surgery Diseases, Xiamen, China
| |
Collapse
|
10
|
Puchertova M, Rychly B, Kolenova A, Vujanić GM. A Unique Case of Cystic Partially Differentiated Nephroblastoma Associated With Botryoid Intralobar Nephrogenic Rests. Pediatr Dev Pathol 2025; 28:121-125. [PMID: 39648335 DOI: 10.1177/10935266241304691] [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] [Indexed: 12/10/2024]
Abstract
Cystic partially differentiated nephroblastoma (CPDN) is a rare pediatric renal tumor composed of multiple cystic spaces divided by septa containing immature nephrogenic elements. The presence of expansile solid areas in the septa of such a lesion indicates an alternative diagnosis of Wilms tumor (WT). We present a unique case of CPDN associated with grossly visible polypoid proliferations, which histologically correspond to botryoid growth of intralobar nephrogenic rests. Correct pathological diagnosis of CPDN and its differentiation from cystic WT can be challenging, but is critical, because of the distinct treatment approaches and prognoses of these entities, since CPDN is a low-risk tumor not requiring further postoperative therapy.
Collapse
Affiliation(s)
- Magdalena Puchertova
- Unilabs Slovakia Ltd., Diagnostic Center of Pathology, Bratislava, Slovakia
- Department of Pathological Anatomy, Faculty of Medicine, Slovak Medical University, Bratislava, Slovakia
| | - Boris Rychly
- Unilabs Slovakia Ltd., Diagnostic Center of Pathology, Bratislava, Slovakia
| | - Alexandra Kolenova
- Department of Pediatric Hematology and Oncology, National Institute of Children's Diseases and Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | | |
Collapse
|
11
|
Gretser S, Kinzler MN, Theilen TM, Wild PJ, Vogler M, Gradhand E. Fluorescence confocal microscopy for evaluation of fresh surgical specimens and consecutive tumor cell isolation in rare pediatric tumors. Virchows Arch 2025; 486:585-593. [PMID: 38980338 PMCID: PMC11950019 DOI: 10.1007/s00428-024-03861-1] [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] [Received: 04/17/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
Fluorescence confocal microscopy (FCM) is an optical technique that uses laser light sources of different wavelengths to generate real-time images of fresh, unfixed tissue specimens. FCM allows histological evaluation of fresh tissue samples without the associated cryo artifacts after frozen sectioning. The aim of this study was to prospectively evaluate pediatric tumor specimens and assess their suitability for fresh tumor sampling. In addition, we aimed to determine whether tumor cell isolation for stable cell culture is still feasible after FCM imaging. Pediatric tumor specimens were imaged using FCM. Tumor viability and suitability for tissue sampling were evaluated and compared with H&E staining after paraffin embedding. In addition, FCM-processed and non-FCM-processed tissue samples were sent for tumor cell isolation to evaluate possible effects after FCM processing. When comparing estimated tumor cell viability using FCM and H&E, we found good to excellent correlating estimates (intraclass correlation coefficient = 0.891, p < 0.001), as well as substantial agreement in whether the tissue appeared adequate for fresh tissue collection (κ = 0.762, p < 0.001). After FCM, seven out of eight samples yielded passable cell cultures, compared to eight out of eight for non-FCM processed samples. Our study suggests that the use of FCM in tumor sampling can increase the yield of suitable fresh tumor samples by identifying viable tumor areas and ensuring that sufficient tissue remains for diagnosis. Our study also provides first evidence that the isolation and growth of tumor cells in culture are not compromised by the FCM technique.
Collapse
Affiliation(s)
- S Gretser
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Pathology, Theodor-Stern-Kai 6, 60590, Frankfurt Am Main, Germany.
| | - M N Kinzler
- Goethe University Frankfurt, University Hospital, Medical Clinic 1, Frankfurt Am Main, Germany
| | - T M Theilen
- Goethe University Frankfurt, University Hospital, Department of Pediatric Surgery and Pediatric Urology, Frankfurt Am Main, Germany
| | - P J Wild
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Pathology, Theodor-Stern-Kai 6, 60590, Frankfurt Am Main, Germany
| | - M Vogler
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt Am Main, Germany
| | - E Gradhand
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute of Pathology, Theodor-Stern-Kai 6, 60590, Frankfurt Am Main, Germany
| |
Collapse
|
12
|
Salgado CM, Gestrich CK, Reyes-Múgica M. Pediatric Genitourinary Tumors: The Developmental Angle. Surg Pathol Clin 2025; 18:191-207. [PMID: 39890304 DOI: 10.1016/j.path.2024.09.002] [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] [Indexed: 02/03/2025]
Abstract
Pediatric cancer is relatively rare compared to cancer in adults. Most pediatric neoplasms affect the hemopoietic and central nervous systems. Of the solid extracranial tumors, renal and genitourinary lesions are among the most frequent pediatric neoplasms. Wilms tumors (nephroblastomas) and their variants predominate. Others are less frequent, and their rarity leads to significant diagnostic challenges. This review presents the most important points for diagnosis using histopathological, immunophenotypical, and molecular novel information on the most important renal and genitourinary pediatric neoplasms.
Collapse
Affiliation(s)
- Claudia M Salgado
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Jackson Memorial Hospital Children's Holtz, 1611 Northwest 12th Avenue, Suite 2153 A, Miami, FL 33136, USA. https://twitter.com/clamsalgado
| | - Catherine K Gestrich
- Division of Pediatric Pathology, Department of Pathology, University of Pittsburgh, Children's Hospital of Pittsburgh, One Children's Hospital Drive, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Miguel Reyes-Múgica
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Jackson Memorial Hospital Children's Holtz, 1611 Northwest 12th Avenue, Suite 2153 B, Miami, FL 33136, USA.
| |
Collapse
|
13
|
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
|
14
|
Treger TD, Wegert J, Wenger A, Coorens THH, Al-Saadi R, Kemps PG, Kennedy J, Parks C, Anderson ND, Hodder A, Letunovska A, Jung H, Ogbonnah T, Trinh MK, Lee-Six H, Morcrette G, van den Heuvel-Eibrink MM, Drost J, van Boxtel R, Bertrums EJM, Goemans BF, Antoniou E, Reinhardt D, Streitenberger H, Ziegler B, Bartram J, Hutchinson JC, Vujanic GM, Vokuhl C, Chowdhury T, Furtwängler R, Graf N, Pritchard-Jones K, Gessler M, Behjati S. Predisposition Footprints in the Somatic Genome of Wilms Tumors. Cancer Discov 2025; 15:286-298. [PMID: 39665570 PMCID: PMC7617291 DOI: 10.1158/2159-8290.cd-24-0878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/02/2024] [Accepted: 12/10/2024] [Indexed: 12/13/2024]
Abstract
Approximately 10% of children with cancer harbor a mutation in a predisposition gene. In children with the kidney cancer Wilms tumor, the prevalence is as high as 30%. Certain predispositions are associated with defined histological and clinical features, suggesting differences in tumorigenesis. To investigate this, we assembled a cohort of 137 children with Wilms tumor, of whom 71 had a pathogenic germline or mosaic variant. We examined 237 neoplasms (including two secondary leukemias), utilizing whole-genome sequencing, RNA sequencing, and genome-wide methylation, validating our findings in an independent cohort. Tumor development differed in children harboring a predisposition, depending on the variant gene and its developmental timing. Differences pervaded the repertoire of driver events, including high-risk mutations, the clonal architecture of normal kidneys, and the relatedness of neoplasms from the same individual. Our findings indicate that predisposition may preordain Wilms tumorigenesis, suggesting a variant-specific approach to managing children merits consideration. Significance: Tumors that arise in children with a cancer predisposition may develop through the same mutational pathways as sporadic tumors. We examined this question in the childhood kidney cancer, Wilms tumor. We found that certain predispositions dictate the genetic development of tumors, with clinical implications for these children. See related commentary by Brzezinski and Malkin, p. 258.
Collapse
Affiliation(s)
- Taryn D Treger
- Wellcome Sanger Institute, Hinxton, United Kingdom
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Jenny Wegert
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Anna Wenger
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Tim H H Coorens
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Reem Al-Saadi
- Great Ormond Street Hospital for Children, London, United Kingdom
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Paul G Kemps
- Wellcome Sanger Institute, Hinxton, United Kingdom
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jonathan Kennedy
- Wellcome Sanger Institute, Hinxton, United Kingdom
- Great Ormond Street Hospital for Children, London, United Kingdom
| | - Conor Parks
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | | | - Angus Hodder
- Wellcome Sanger Institute, Hinxton, United Kingdom
- Great Ormond Street Hospital for Children, London, United Kingdom
| | | | | | | | - Mi K Trinh
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Henry Lee-Six
- Wellcome Sanger Institute, Hinxton, United Kingdom
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Guillaume Morcrette
- Department of Fetopathology, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | | | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Ruben van Boxtel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | | | - Bianca F Goemans
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Evangelia Antoniou
- Clinic of Pediatrics III, University Hospital of Essen, Essen, Germany
- AML-BFM Study Group, Essen, Germany
| | - Dirk Reinhardt
- Clinic of Pediatrics III, University Hospital of Essen, Essen, Germany
- AML-BFM Study Group, Essen, Germany
| | - Heike Streitenberger
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Barbara Ziegler
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Jack Bartram
- Great Ormond Street Hospital for Children, London, United Kingdom
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | | | | | - Christian Vokuhl
- Department of Pathology, Section of Pediatric Pathology, University of Bonn, Bonn, Germany
| | - Tanzina Chowdhury
- Great Ormond Street Hospital for Children, London, United Kingdom
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Rhoikos Furtwängler
- Department of Paediatric Haematology and Oncology, Saarland University, Homburg, Germany
- Pediatric Hematology and Oncology, Inselspital Children's Hospital, University Bern, Bern, Switzerland
| | - Norbert Graf
- Department of Paediatric Haematology and Oncology, Saarland University, Homburg, Germany
| | | | - Manfred Gessler
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, University of Wuerzburg, Wuerzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Wuerzburg, Germany
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, United Kingdom
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Great Ormond Street Hospital for Children, London, United Kingdom
| |
Collapse
|
15
|
Götz L, Wegert J, Paikari A, Appenzeller S, Bausenwein S, Vokuhl C, Treger TD, Drost J, Linderkamp C, Schneider DT, Ernestus K, Warman SW, Fuchs J, Welter N, Graf N, Behjati S, Furtwängler R, Gessler M. Wilms tumor primary cultures capture phenotypic heterogeneity and facilitate preclinical screening. Transl Oncol 2025; 52:102263. [PMID: 39740515 PMCID: PMC11750297 DOI: 10.1016/j.tranon.2024.102263] [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: 09/15/2024] [Revised: 11/26/2024] [Accepted: 12/23/2024] [Indexed: 01/02/2025] Open
Abstract
Wilms tumors (WT) are characterized by variable contributions of blastemal, epithelial and stromal elements, reflecting their diverse cellular origins and genetic drivers. In vitro models remain rare, despite a growing need to better characterize tumor biology and evaluate new treatments. Using three approaches, we have now established a large collection of long-term cultures that represent this diversity. Adherent WT cultures are predominated by stromal cells, 3D spheroids model blastema, and patient-derived organoid cultures of both tumor and healthy kidney tissue result in the preferential growth of epithelial cells. Adherent, spheroid and organoid cultures are also clearly distinguishable by their transcriptome. Preclinical drug screening experiments revealed sensitivity to a range of inhibitors, that are highly effective in other childhood solid tumors. Sensitivity was related to MYCN status, a marker associated with adverse outcome across human cancers including WT. The combination of the three culture techniques represents a promising tool to both explore tumor heterogeneity in vitro and to facilitate characterization of candidate driver genes, in order to improve treatment regimens in the future.
Collapse
Affiliation(s)
- Lisa Götz
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Jenny Wegert
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Alireza Paikari
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Silke Appenzeller
- Comprehensive Cancer Center Mainfranken, University Hospital of Würzburg, Würzburg, Germany
| | - Sabrina Bausenwein
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Christian Vokuhl
- Section of Pediatric Pathology, Department of Pathology, University Hospital Bonn, Bonn, Germany
| | - Taryn D Treger
- Wellcome Sanger Institute, Hinxton, UK; Department of Pediatrics, University of Cambridge, Cambridge CB2 0QQ, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Christin Linderkamp
- Department of Pediatric Hematology and Oncology, Hannover Medical School (MHH), Hannover, Germany
| | - Dominik T Schneider
- Clinic of Pediatrics, Klinikum Dortmund, University Witten/Herdecke, Germany
| | - Karen Ernestus
- Comprehensive Cancer Center Mainfranken, University Hospital of Würzburg, Würzburg, Germany; Department of Pathology, University of Würzburg, Würzburg, Germany
| | - Steven W Warman
- Clinic of Pediatric Surgery, Charité - University Hospital Berlin, Berlin, Germany; Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, Tuebingen, Germany
| | - Jörg Fuchs
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, Tuebingen, Germany
| | - Nils Welter
- Department of Pediatric Hematology and Oncology, Saarland University Hospital, Homburg, Germany
| | - Norbert Graf
- Department of Pediatric Hematology and Oncology, Saarland University Hospital, Homburg, Germany
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, UK; Department of Pediatrics, University of Cambridge, Cambridge CB2 0QQ, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Rhoikos Furtwängler
- Department of Pediatric Hematology and Oncology, Saarland University Hospital, Homburg, Germany; Pediatric Hematology and Oncology, Dep. of Pediatrics, Bern University Hospital, University of Bern, Inselspital, Switzerland
| | - Manfred Gessler
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany; Comprehensive Cancer Center Mainfranken, University Hospital of Würzburg, Würzburg, Germany.
| |
Collapse
|
16
|
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: 0] [Impact Index Per Article: 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
|
17
|
Huang M, Fei Y, Zhong Z, Jiang H, Liu L, Liu J, Zhang H, Li J, Xu Z, Gao P, Wang C. A refined kidney tumor nephrometry system employed to screen pediatric patients who are eligible for nephron sparing surgery. Front Pediatr 2025; 12:1501560. [PMID: 39845455 PMCID: PMC11752886 DOI: 10.3389/fped.2024.1501560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
Purpose Comprehension of the anatomical characteristics of pediatric kidney tumors is crucial for making surgical decisions. Previous kidney tumor nephrometry systems failed to incorporate two significant factors: tumor thrombus and multifocality. We develop a refined nephrometry system based on a comprehensive understanding of the characteristics exhibited by pediatric kidney tumors. Methods The TUMORS nephrometry scoring system comprises 6 indicators, including tumor (T)hrombus, (U)rinary collecting system involvement, (M)ultiple tumors, (O)utward property, (R)adius, and (S)ite relative to the polar lines. Each renal unit was assessed and scored independently. The complexity characteristics of kidney tumors were summarized, and the correlation was compared with RENAL nephrometry system. Furthermore, the complexity of kidney tumors was compared across different surgical procedures. Results A total of 43 patients were enrolled, involving 70 kidney units. Radical nephrectomy (RN) was performed on 13 kidneys, while the remaining 57 kidneys underwent nephron sparing surgery. In the NSS group, tumors in 37 kidneys were resected in vivo, whereas 20 kidneys underwent tumor resection ex vivo followed by kidney autotransplantation. According to the TUMORS nephrometry scoring system, there were 13, 34 and 23 kidney units classified as low, moderate and high complexity, respectively. Tumors that underwent RN or ex vivo removal exhibited higher complexity. The complications and positive margins of NSS were not statistically significant in relation to tumor complexity. Conclusion The TUMORS nephrometry scoring system holds significant guidance for the decision of surgical protocol and can be applied to the preoperative evaluation.
Collapse
Affiliation(s)
- Mingchuan Huang
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingchun Fei
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihai Zhong
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong Jiang
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Longshan Liu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Juncheng Liu
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huanxi Zhang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Li
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhe Xu
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Pengfei Gao
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Changxi Wang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Somers GR, L'Herminé-Coulomb A, Matoso A, O'Sullivan MJ. Paediatric renal tumours: an update on challenges and recent developments. Virchows Arch 2025; 486:49-64. [PMID: 39786574 DOI: 10.1007/s00428-024-04017-x] [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: 07/22/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/12/2025]
Abstract
Paediatric renal tumours include a broad range of neoplasms which largely differ, but also overlap to a smaller extent, with adult kidney cancer. These include the embryonal tumour nephroblastoma, which accounts for the majority of cases of kidney cancer in the first decade of life and, despite boasting a cure in ~ 90% cases, still presents clinical challenges in a small proportion of cases. A variety of less common mesenchymal tumours, including the mostly indolent congenital mesoblastic nephroma, clear cell sarcoma of kidney which continues to be associated with poor outcomes for higher stage disease, and the typically lethal malignant rhabdoid tumour, form the bulk of the remaining presentations in the first decade. All three of these represent the intrarenal form of a wider 'family' of genetically related and histologically overlapping entities occurring in soft tissue and other anatomical locations. The latter two are examples of aggressive 'epigenetic' tumours driven by dysregulation of chromatin. In the second decade of life, renal cell carcinoma dominates, and with molecular characterisation many distinct subtypes are now described. Herein we discuss the developments in relation to diagnostic categorisation of paediatric renal cancers and how deeper understanding of the underlying biology is already providing therapeutic opportunity, while also focussing on the challenges that remain for the pathologist.
Collapse
Affiliation(s)
- Gino R Somers
- Department of Paediatric Laboratory Medicine, Hospital for Sick Children, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Aurore L'Herminé-Coulomb
- Pathology Department, Hôpital Armand Trousseau-Sorbonne Université-Assistance Publique Hôpitaux de Paris, Paris, France
| | - Andres Matoso
- Genitourinary Pathology Division, The Johns Hopkins Hospital, Baltimore, MD, 21231-2410, USA
| | - Maureen J O'Sullivan
- Histology Laboratory, Children's Health Ireland, Dublin, Ireland.
- Histopathology Department, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
19
|
Zeng Q, Tao J, Qin L, Zeng Y, Liu Z, Xu M, Zeng L. Comprehensive prognostic gene identification and functional characterization of GRAMD1A in Wilms tumor: development of risk prediction models and therapeutic implications. Front Oncol 2024; 14:1501718. [PMID: 39659787 PMCID: PMC11628387 DOI: 10.3389/fonc.2024.1501718] [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: 09/25/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Background Wilms tumor (WT) is the most common pediatric kidney cancer, with survival rates exceeding 90% in localized cases. However, advanced or recurrent WT remains difficult to treat due to poor prognosis and limited knowledge of its molecular mechanisms. Gene expression profiling has shown promise in identifying prognostic markers and therapeutic targets. This study aimed to identify key prognostic genes and pathways in WT, construct risk prediction models, and validate their role in tumor progression. Methods RNA sequencing and clinical data from 136 WT patients were obtained from the TARGET database. Differential gene expression analysis was conducted using GEO datasets GSE11024 and GSE66405 to compare WT and normal kidney tissues. Identified differentially expressed genes (DEGs) underwent Gene Ontology (GO) and KEGG pathway enrichment analysis to explore biological functions and pathways associated with WT progression. Univariate Cox regression was used to assess the association between DEGs and overall survival (OS) and progression-free survival (PFS). LASSO regression models were developed for risk stratification, and model accuracy was evaluated using time-dependent ROC curves. External validation confirmed key hub genes, while functional assays in WT cell lines (WiT-49) assessed the role of GRAMD1A in tumor behavior. Results A total of 3,395 DEGs were identified, with 1,564 upregulated and 1,831 downregulated genes. Enrichment analyses revealed significant pathways involved in cell cycle regulation and metabolic reprogramming. Six key genes (GRAMD1A, PLXNA3, SPR, EBAG9, RBM47, and RIDA) were associated with both OS and PFS. LASSO models demonstrated strong predictive performance, with GRAMD1A identified as a major risk factor. External validation confirmed differential expression, and functional assays showed that GRAMD1A silencing significantly inhibited WT cell viability, proliferation, migration, and invasion. Conclusions This study identifies novel prognostic genes and potential therapeutic targets in WT. GRAMD1A, SPR, EBAG9, RBM47, and RIDA play critical roles in WT progression, with GRAMD1A as a key oncogenic factor, offering potential for risk stratification and future therapeutic intervention.
Collapse
Affiliation(s)
- Qiang Zeng
- Department of Pediatric Surgery, Jiangxi Maternal and Child Health Hospital, Jiangxi Children’s Medical Center, Nanchang, Jiangxi, China
| | - Junfeng Tao
- Department of Pediatric Surgery, Jiangxi Maternal and Child Health Hospital, Jiangxi Children’s Medical Center, Nanchang, Jiangxi, China
| | - Lilu Qin
- Department of Pediatric Surgery, Jiangxi Maternal and Child Health Hospital, Jiangxi Children’s Medical Center, Nanchang, Jiangxi, China
| | - Yong Zeng
- Department of Pediatric Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zhong Liu
- Department of Pediatric Surgery, Jiangxi Maternal and Child Health Hospital, Jiangxi Children’s Medical Center, Nanchang, Jiangxi, China
| | - Mingxian Xu
- Department of Musculoskeletal Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Linshan Zeng
- Department of Pediatric Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
20
|
Benlhachemi S, Khattab M, Hattoufi K, Abouqal R, Boutayeb S, El Fahime E. Analysis of Wilms Tumour Epidemiology, Clinicopathological Features and Treatment Outcomes in 84 Moroccan Patients. Cancer Rep (Hoboken) 2024; 7:e2158. [PMID: 39506861 PMCID: PMC11541055 DOI: 10.1002/cnr2.2158] [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: 01/05/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Wilms tumour (WT), the second most reported childhood cancer in Morocco, is a malignant kidney tumour that affects children under 15 years old. Prognosis has improved with the adoption of multimodal treatment. However, data on WT in Morocco remain limited. AIMS This study aims to comprehensively describe and analyse the epidemiological, clinicopathological features and treatment outcomes of WT in Moroccan patients, including treatment response and recurrence rates. METHODS AND RESULTS A retrospective study involved 84 children under 15 years with WT, treated according to the SIOP protocol and followed at the Paediatric Haematology and Oncology Centre at Children's hospital of Rabat, between January 2014 and February 2018. The median age of participants was 36 months, with a male/female sex ratio of 0.79. Abdominal mass was the primary concern in 55 cases (66%). Five patients (6%) had bilateral WT. Metastatic WT occurred in 21 cases (25%). Stage III was predominant in 33 cases (43%). Twenty cases (26%) had high-risk WT, and IVC tumour thrombus was observed in 12 cases (14%). WT histotype correlated significantly with both sex and tumour localisation (p values of 0.040 and 0.013, respectively). Age correlated significantly with WT extension, overall stage and SIOP histology risk grades (p values of 0.003, 0.003 and 0.045, respectively). Overall stage was statistically related to the occurrence of IVC tumour thrombus (p = 0.002). Over a 5-year span post-nephrectomy, complete remission was achieved in 63 patients (75%), partial remission in one patient (1%), while 19 patients (23%) died and one patient (1%) relapsed. CONCLUSION These findings are encouraging for a developing country. However, the elevated rates of Stages III and IVC thrombus in this series are still high, primarily attributed to delays in diagnosis and treatment and the limited number of paediatric haematology and oncology units at the time of the study. Nevertheless, further multicentric research is warranted to enrich Moroccan data and establish a national register for WT cases.
Collapse
Affiliation(s)
- Sara Benlhachemi
- Neuroscience and Neurogenetics Research Team (ERNN), Faculty of Medicine and PharmacyMohammed V UniversityRabatMorocco
- Molecular Biology and Functional Genomics PlatformNational Centre for Scientific and Technical ResearchRabatMorocco
| | - Mohammed Khattab
- Department of Paediatric Haematology and OncologyAbulcasis International University of Health SciencesRabatMorocco
| | - Kenza Hattoufi
- Department of Neonatology and NutritionUniversity Hospital Centre IBN SINARabatMorocco
- Research Team on Health and Nutrition of Mother and Child, Faculty of Medicine and PharmacyMohammed V UniversityRabatMorocco
| | - Redouane Abouqal
- Biostatistics Laboratory, Clinical Epidemiology Research, Faculty of Medicine and PharmacyMohammed V UniversityRabatMorocco
| | - Saber Boutayeb
- Medical Oncology DepartmentNational Institute of Oncology, Mohammed V UniversityRabatMorocco
- Mohamed VI Centre for Research and Innovation (CM6RI)RabatMorocco
| | - Elmostafa El Fahime
- Neuroscience and Neurogenetics Research Team (ERNN), Faculty of Medicine and PharmacyMohammed V UniversityRabatMorocco
- Molecular Biology and Functional Genomics PlatformNational Centre for Scientific and Technical ResearchRabatMorocco
- Mohamed VI Centre for Research and Innovation (CM6RI)RabatMorocco
| |
Collapse
|
21
|
Sudour-Bonnange H, van Tinteren H, Ramírez-Villar GL, Godzinski J, Irtan S, Gessler M, Chowdhury T, Audry G, Fuchs J, Powis M, van de Ven CP, Okoye B, Smeulders N, Vujanic GM, Verschuur A, L'Herminé-Coulomb A, de Camargo B, de Aguirre Neto JC, Schenk JP, van den Heuvel-Eibrink MM, Pritchard-Jones K, Graf N, Bergeron C, Furtwängler R. Characteristics and outcome of synchronous bilateral Wilms tumour in the SIOP WT 2001 Study: Report from the SIOP Renal Tumour Study Group (SIOP-RTSG). Br J Cancer 2024; 131:972-981. [PMID: 39080350 PMCID: PMC11405756 DOI: 10.1038/s41416-024-02799-0] [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: 03/04/2024] [Revised: 07/02/2024] [Accepted: 07/09/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Among patients with nephroblastoma, those with bilateral disease are a unique population where maximising tumour control must be balanced with preserving renal parenchyma. METHODS The SIOP 2001 protocol recommended surgery after neoadjuvant cycle(s) of Dactinomycin and Vincristine (AV) with response-adapted intensification, if needed. Adjuvant treatment was given based on the lesion with the worst histology. RESULTS Three hundred and twenty seven patients with stage V disease were evaluable: 174 had bilateral Wilms tumour (BWT), 101 unilateral WT and contralateral nephroblastomatosis (NB) and 52 bilateral nephroblastomatosis. In these three groups, the estimated 5y-EFS was 76.1%, 84.6%, and 74.9%, respectively. AV chemotherapy alone was the successful chemotherapy for 58.7% of all the patients and 65.6% of the non-metastatic patients. Among the 174 patients with BWT, 149 (88.2%) had at least one nephron-sparing surgery. Twenty of 61 bilateral stage I patients were treated with four-week AV postoperatively achieving 94.4% 5y-EFS. At last follow-up, 87% of patients had normal renal function. CONCLUSIONS This study demonstrates that AV without anthracyclines is sufficient to achieve NSS and good survival in the majority of patients. For patients with bilateral stage I WT and intermediate risk histology, only four weeks adjuvant AV seems to be sufficient. CLINICAL TRIAL REGISTRATION NCT00047138.
Collapse
Affiliation(s)
| | - Harm van Tinteren
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Gema L Ramírez-Villar
- Department of Pediatric Oncology, Hospital Universitario Virgen Del Rocio, Sevilla, Spain
| | - Jan Godzinski
- Department of Pediatric Surgery, Marciniak Hospital, Fieldorfa 2 and Department of Pediatric Traumatology and Emergency Medicine, Wroclaw Medical university, Wrocław, Poland
| | - Sabine Irtan
- Department of Pediatric Surgery, Hospital Trousseau, Paris, France
| | - Manfred Gessler
- Theosor-Boveri Institute/Biocenter and Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Würzburg, Germany
| | - Tanzina Chowdhury
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Georges Audry
- Department of Pediatric Surgery, Hospital Trousseau, Paris, France
| | - Joerg Fuchs
- Department of Pediatric Surgery and Urology, University Hospital Tübingen, 72076, Tuebingen, Germany
| | - Mark Powis
- Department of Pediatric Surgery, Leeds Teaching Hospital NHS Trust, Leeds, UK
| | | | - Bruce Okoye
- Department of Paediatric Surgery, St George's University Hospital, London, UK
| | - Naima Smeulders
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK
| | - Gordan M Vujanic
- Department of Pathology, Sidra Medicine and weill Cornell Medicine- Qatar, Doha, Qatar
| | - Arnaud Verschuur
- Department of Pediatric Hemato-Oncology, Hospital La Timone, Marseille, France
| | | | - Beatriz de Camargo
- Pediatric Hematology and Oncology program, research Center, Instituto National de Cancer, Rio de Janeiro, Brazil
| | | | - Jens Peter Schenk
- Sektion Pädiatrische Radiologie, Klinik für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - Mary M van den Heuvel-Eibrink
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Division of Childhealth, Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | - Katy Pritchard-Jones
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | | | - Christophe Bergeron
- Department of Pediatric Oncology and Hematology, Centre Léon Berard, Lyon, France
| | - Rhoikos Furtwängler
- Saarland University, Homburg, Germany
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Inselspital, University Bern, Bern, Switzerland
| |
Collapse
|
22
|
Chen X, Bao Y, Sun G, Wang X, Zhu J. UNC13B regulates the sensitivity of Wilms' tumor cells to doxorubicin by modulating lysosomes. Oncol Lett 2024; 28:446. [PMID: 39091580 PMCID: PMC11292464 DOI: 10.3892/ol.2024.14579] [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: 02/15/2024] [Accepted: 06/14/2024] [Indexed: 08/04/2024] Open
Abstract
Wilms' tumor is a malignant neoplasm where current medical advancements have significantly improved survival rates; however, challenges persist such as the resistance of the tumor to chemotherapy drugs like doxorubicin. This necessitates higher dosages, leading to decreased sensitivity. However, using high doses of doxorubicin can have late effects on the heart. Unc-13 homolog B (UNC13B) may be involved in the drug resistance in several tumors, yet its role in modulating drug sensitivity in Wilms' tumor remains unexplored. UNC13B levels were quantified using reverse transcription-qPCR and Western blotting. The half-maximal inhibitory concentration for doxorubicin, vincristine, and actinomycin-D was determined using CCK-8 assays. Cell cycle and apoptosis were analyzed using flow cytometry, and lysosomal changes were observed using Lyso-Tracker staining. The present study initially evaluated UNC13B expression levels in the Wilms' tumor 17.94 cell line. Additionally, through short hairpin RNA-mediated knockdown, changes in doxorubicin sensitivity in 17.94 Wilms' tumor cells were assessed. Concurrently, preliminary investigations into the role of UNC13B in regulating lysosomes was performed, revealing a significant positive association between UNC13B levels and lysosome formation in the 17.94 cell line. Lysosomes likely serve a role in the sensitivity of Wilms' tumor cell lines to drugs. Elevated UNC13B expression was observed in the 17.94 Wilms' tumor cell line compared to normal kidney cells. UNC13B knockdown also resulted in increased apoptosis levels upon doxorubicin treatment. Immunofluorescence revealed UNC13B localization within cellular vesicles, and its knockdown significantly decreased lysosome levels. Overall, the findings of the present study demonstrate that UNC13B regulates the sensitivity of the Wilms' tumor 17.94 cell line to doxorubicin by modulating lysosome formation within cells. The results suggest that UNC13B is likely an enriched target involved in lysosomal regulation in certain tumors, offering a new approach for optimizing chemotherapy in Wilms' tumor and other cancers with high UNC13B expression.
Collapse
Affiliation(s)
- Xi Chen
- Department of Neonatology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yingying Bao
- Department of Neonatology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Ge Sun
- Department of Neonatology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xiaobo Wang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Jiajun Zhu
- Department of Neonatology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
- Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
23
|
Kiss R, Micsik T, Bedics G, Papp G, Csóka M, Jenővári Z, Szabó S, Tornóczki T, Vujanic G, Kuthi L. Pediatric thyroid-like follicular renal cell carcinoma-a post-neuroblastoma case with comprehensive genomic profiling data. Virchows Arch 2024; 485:583-588. [PMID: 38990362 DOI: 10.1007/s00428-024-03867-9] [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] [Received: 06/11/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Thyroid-like follicular renal cell carcinoma (TLFRCC), an emerging subtype of renal cell carcinoma, presents diagnostic challenges due to its resemblance to normal thyroid tissue. Here, we report a rare case of TLFRCC in a pediatric patient, a demographic rarely affected by this subtype. Histologically resembling a typical TLFRCC, our case exhibited unique features including post-neuroblastoma development, occurrence in a male teenager, and diffuse MelanA expression, which has not been previously reported in TLFRCC. Comprehensive genomic profiling revealed the EWSR1::PATZ1 fusion, confirming its genetic basis. Due to the advanced tumor stage, the patient received combined immunotherapy, and after a 9-month follow-up, remains tumor-free. Our case broadens the diagnostic spectrum of pediatric renal cell carcinomas, highlighting the importance of comprehensive molecular profiling in rare subtypes such as TLFRCC. Further research is needed to better understand TLFRCC's genetic landscape and optimize therapeutic strategies, especially in pediatric populations with evolving treatment protocols.
Collapse
Affiliation(s)
- Richárd Kiss
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Tamás Micsik
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gábor Bedics
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gergő Papp
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Monika Csóka
- Tűzoltó Street Department, Pediatric Center, Semmelweis University, Budapest, Hungary
| | - Zoltán Jenővári
- Tűzoltó Street Department, Pediatric Center, Semmelweis University, Budapest, Hungary
| | - Sándor Szabó
- Tűzoltó Street Department, Pediatric Center, Semmelweis University, Budapest, Hungary
| | - Tamás Tornóczki
- Department of Pathology, Faculty of Medicine and Clinical Center, University of Pécs, Pécs, Hungary
| | | | - Levente Kuthi
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.
- Department of Surgical and Molecular Pathology, Center of Tumor Pathology, National Institute of Oncology, Budapest, Hungary.
| |
Collapse
|
24
|
Koska IO, Ozcan HN, Tan AA, Beydogan B, Ozer G, Oguz B, Haliloglu M. Radiomics in differential diagnosis of Wilms tumor and neuroblastoma with adrenal location in children. Eur Radiol 2024; 34:5016-5027. [PMID: 38311701 PMCID: PMC11255001 DOI: 10.1007/s00330-024-10589-8] [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] [Received: 11/13/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 02/06/2024]
Abstract
OBJECTIVES Machine learning methods can be applied successfully to various medical imaging tasks. Our aim with this study was to build a robust classifier using radiomics and clinical data for preoperative diagnosis of Wilms tumor (WT) or neuroblastoma (NB) in pediatric abdominal CT. MATERIAL AND METHODS This is a single-center retrospective study approved by the Institutional Ethical Board. CT scans of consecutive patients diagnosed with WT or NB admitted to our hospital from January 2005 to December 2021 were evaluated. Three distinct datasets based on clinical centers and CT machines were curated. Robust, non-redundant, high variance, and relevant radiomics features were selected using data science methods. Clinically relevant variables were integrated into the final model. Dice score for similarity of tumor ROI, Cohen's kappa for interobserver agreement among observers, and AUC for model selection were used. RESULTS A total of 147 patients, including 90 WT (mean age 34.78 SD: 22.06 months; 43 male) and 57 NB (mean age 23.77 SD:22.56 months; 31 male), were analyzed. After binarization at 24 months cut-off, there was no statistically significant difference between the two groups for age (p = .07) and gender (p = .54). CT clinic radiomics combined model achieved an F1 score of 0.94, 0.93 accuracy, and an AUC 0.96. CONCLUSION In conclusion, the CT-based clinic-radiologic-radiomics combined model could noninvasively predict WT or NB preoperatively. Notably, that model correctly predicted two patients, which none of the radiologists could correctly predict. This model may serve as a noninvasive preoperative predictor of NB/WT differentiation in CT, which should be further validated in large prospective models. CLINICAL RELEVANCE STATEMENT CT-based clinic-radiologic-radiomics combined model could noninvasively predict Wilms tumor or neuroblastoma preoperatively. KEY POINTS • CT radiomics features can predict Wilms tumor or neuroblastoma from abdominal CT preoperatively. • Integrating clinic variables may further improve the performance of the model. • The performance of the combined model is equal to or greater than human readers, depending on the lesion size.
Collapse
Affiliation(s)
- Ilker Ozgur Koska
- Department of Radiology, Behcet Uz Children's Hospital, Konak İzmir, Turkey.
| | - H Nursun Ozcan
- Department of Radiology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Aziz Anil Tan
- Department of Radiology, Hacettepe University School of Medicine, Ankara, Turkey
- Department of Radiology, Sincan Training and Research Hospital, Ankara, Turkey
| | - Beyza Beydogan
- Department of Radiology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Gozde Ozer
- Department of Radiology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Berna Oguz
- Department of Radiology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Mithat Haliloglu
- Department of Radiology, Hacettepe University School of Medicine, Ankara, Turkey
| |
Collapse
|
25
|
Gehle DB, Morrison ZD, Halepota HF, Kumar A, Gwaltney C, Krasin MJ, Graetz DE, Santiago T, Boston US, Davidoff AM, Murphy AJ. Wilms Tumor with Vena Caval Intravascular Extension: A Surgical Perspective. CHILDREN (BASEL, SWITZERLAND) 2024; 11:896. [PMID: 39201831 PMCID: PMC11353173 DOI: 10.3390/children11080896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 09/03/2024]
Abstract
Wilms tumor (WT) is the most common kidney tumor in pediatric patients. Intravascular extension of WT above the level of the renal veins is a rare manifestation that complicates surgical management. Patients with intravascular extension are frequently asymptomatic at diagnosis, and tumor thrombus extension is usually diagnosed by imaging. Neoadjuvant chemotherapy is indicated for thrombus extension above the level of the hepatic veins and often leads to thrombus regression, obviating the need for cardiopulmonary bypass in cases of cardiac thrombus at diagnosis. In cases of tumor extension to the retrohepatic cava, neoadjuvant therapy is not strictly indicated, but it may facilitate the regression of tumor thrombi, making resection safer. Hepatic vascular isolation and cardiopulmonary bypass increase the risk of bleeding and other complications when utilized for tumor thrombectomy. Fortunately, WT patients with vena caval with or with intracardiac extension have similar overall and event-free survival when compared to patients with WT without intravascular extension when thrombectomy is successfully performed. Still, patients with metastatic disease at presentation or unfavorable histology suffer relatively poor outcomes. Dedicated pediatric surgical oncology and pediatric cardiothoracic surgery teams, in conjunction with multimodal therapy directed by a multidisciplinary team, are preferred for optimized outcomes in this patient population.
Collapse
Affiliation(s)
- Daniel B. Gehle
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (D.B.G.); (Z.D.M.); (H.F.H.); (C.G.); (A.M.D.)
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zachary D. Morrison
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (D.B.G.); (Z.D.M.); (H.F.H.); (C.G.); (A.M.D.)
| | - Huma F. Halepota
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (D.B.G.); (Z.D.M.); (H.F.H.); (C.G.); (A.M.D.)
| | - Akshita Kumar
- Department of Surgery, Division of Pediatric Surgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Clark Gwaltney
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (D.B.G.); (Z.D.M.); (H.F.H.); (C.G.); (A.M.D.)
| | - Matthew J. Krasin
- Department of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Dylan E. Graetz
- Department of Oncology, Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Teresa Santiago
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Umar S. Boston
- Department of Surgery, Division of Pediatric Cardiothoracic Surgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Andrew M. Davidoff
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (D.B.G.); (Z.D.M.); (H.F.H.); (C.G.); (A.M.D.)
- Department of Surgery, Division of Pediatric Surgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Andrew J. Murphy
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (D.B.G.); (Z.D.M.); (H.F.H.); (C.G.); (A.M.D.)
- Department of Surgery, Division of Pediatric Surgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| |
Collapse
|
26
|
Kroll MR, Au C, Slostad J, Christ TN, Papas SG, Tan A. Case report: Metastatic BRAF V600E-mutated adult Wilms' tumor with robust response to BRAF/MEK inhibitor therapy. Front Oncol 2024; 14:1376270. [PMID: 39234402 PMCID: PMC11373342 DOI: 10.3389/fonc.2024.1376270] [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: 01/25/2024] [Accepted: 06/13/2024] [Indexed: 09/06/2024] Open
Abstract
Nephroblastoma or Wilms' tumor (WT) is the most common pediatric renal malignancy but rare in adults. Treatment protocols for adults are typically extrapolated from pediatric guidelines, but there are no standard guidelines for adults due to the rarity of the disease. However, next-generation sequencing has led to new therapeutic options for adult WT patients. We present the first case to our knowledge of a recurrent adult WT treated with dual BRAF/MEK-targeted therapy, which showed initial robust clinical response and was well tolerated.
Collapse
Affiliation(s)
- Matthew R. Kroll
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Cherry Au
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Jessica Slostad
- Division of Hematology, Oncology, and Cellular Therapies, Rush University Medical Center, Chicago, IL, United States
| | - Trevor N. Christ
- Department of Pharmacy, Rush University Medical Center, Chicago, IL, United States
| | - Sam G. Papas
- Division of Surgical Oncology, Rush University Medical Center, Chicago, IL, United States
| | - Alan Tan
- Division of Hematology, Oncology, and Cellular Therapies, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
27
|
de Faria LL, Ponich Clementino C, Véras FASE, Khalil DDC, Otto DY, Oranges Filho M, Suzuki L, Bedoya MA. Staging and Restaging Pediatric Abdominal and Pelvic Tumors: A Practical Guide. Radiographics 2024; 44:e230175. [PMID: 38722785 DOI: 10.1148/rg.230175] [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: 06/06/2024]
Abstract
The most common abdominal malignancies diagnosed in the pediatric population include neuroblastoma, Wilms tumor, hepatoblastoma, lymphoma, germ cell tumor, and rhabdomyosarcoma. There are distinctive imaging findings and patterns of spread for each of these tumors that radiologists must know for diagnosis and staging and for monitoring the patient's response to treatment. The multidisciplinary treatment group that includes oncologists, surgeons, and radiation oncologists relies heavily on imaging evaluation to identify the best treatment course and prognostication of imaging findings, such as the image-defined risk factors for neuroblastomas, the PRETreatment EXtent of Disease staging system for hepatoblastoma, and the Ann Arbor staging system for lymphomas. It is imperative for radiologists to be able to correctly indicate the best imaging methods for diagnosis, staging, and restaging of each of these most prevalent tumors to avoid inconclusive or unnecessary examinations. The authors review in a practical manner the most updated key points in diagnosing and staging disease and assessing response to treatment of the most common pediatric abdominal tumors. ©RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Luisa Leitão de Faria
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Carolina Ponich Clementino
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Felippe Augusto Silvestre E Véras
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Douglas da Cunha Khalil
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Deborah Yukiko Otto
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Marcelo Oranges Filho
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Lisa Suzuki
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - M Alejandra Bedoya
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| |
Collapse
|
28
|
Motta M, Barresi S, Pizzi S, Bifano D, Lopez Marti J, Garrido-Pontnou M, Flex E, Bruselles A, Giovannoni I, Rotundo G, Fragale A, Tirelli V, Vallese S, Ciolfi A, Bisogno G, Alaggio R, Tartaglia M. RAF1 gene fusions are recurrent driver events in infantile fibrosarcoma-like mesenchymal tumors. J Pathol 2024; 263:166-177. [PMID: 38629245 DOI: 10.1002/path.6272] [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] [Received: 11/08/2023] [Revised: 12/21/2023] [Accepted: 02/13/2024] [Indexed: 05/12/2024]
Abstract
Infantile fibrosarcomas (IFS) and congenital mesoblastic nephroma (CMN) are rare myofibroblastic tumors of infancy and early childhood commonly harboring the ETV6::NTRK3 gene fusion. IFS/CMN are considered as tumors with an 'intermediate prognosis' as they are locally aggressive, but rarely metastasize, and generally have a favorable outcome. A fraction of IFS/CMN-related neoplasms are negative for the ETV6::NTRK3 gene rearrangement and are characterized by other chimeric proteins promoting MAPK signaling upregulation. In a large proportion of these tumors, which are classified as IFS-like mesenchymal neoplasms, the contributing molecular events remain to be identified. Here, we report three distinct rearrangements involving RAF1 among eight ETV6::NTRK3 gene fusion-negative tumors with an original histological diagnosis of IFS/CMN. The three fusion proteins retain the entire catalytic domain of the kinase. Two chimeric products, GOLGA4::RAF1 and LRRFIP2::RAF1, had previously been reported as driver events in different cancers, whereas the third, CLIP1::RAF1, represents a novel fusion protein. We demonstrate that CLIP1::RAF1 acts as a bona fide oncoprotein promoting cell proliferation and migration through constitutive upregulation of MAPK signaling. We show that the CLIP1::RAF1 hyperactive behavior does not require RAS activation and is mediated by constitutive 14-3-3 protein-independent dimerization of the chimeric protein. As previously reported for the ETV6::NTRK3 fusion protein, CLIP1::RAF1 similarly upregulates PI3K-AKT signaling. Our findings document that RAF1 gene rearrangements represent a recurrent event in ETV6::NTRK3-negative IFS/CMN and provide a rationale for the use of inhibitors directed to suppress MAPK and PI3K-AKT signaling in these cancers. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Marialetizia Motta
- Molecular Genetics and Functional Genomics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sabina Barresi
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Simone Pizzi
- Molecular Genetics and Functional Genomics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Delfina Bifano
- Pathology Unit, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Jennifer Lopez Marti
- Department of Pathology, Hospital Nacional de Pediatria Juan P. Garrahan, Buenos Aires, Argentina
| | | | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Giovannina Rotundo
- Molecular Genetics and Functional Genomics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Alessandra Fragale
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Silvia Vallese
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Ciolfi
- Molecular Genetics and Functional Genomics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gianni Bisogno
- Pediatric Hematology-Oncology Division, University Hospital, Padova, Italy
| | - Rita Alaggio
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University, Latina, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
29
|
Fu Y, Zhong J, Tan Y, Zheng T, Liu M, Wang G. Contrast-enhanced ultrasound for differentiating benign from malignant focal solid renal lesions in pediatric patients. Sci Rep 2024; 14:11409. [PMID: 38762673 PMCID: PMC11102535 DOI: 10.1038/s41598-024-62496-z] [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: 01/30/2024] [Accepted: 05/17/2024] [Indexed: 05/20/2024] Open
Abstract
The contrast-enhanced ultrasound (CEUS) has been mainly applied to adults to differentiate benign and malignant renal lesions, however, the characteristics of CEUS in pediatric has not been as well studied as in adults. In the present work, the eligible pediatric patients who underwent renal CEUS between March 2016 and February 2023 were retrospectively analyzed. It included 20 lesions (median diameter, 8.4 cm; range, 1.8-18.0 cm) from 20 patients (median age, 28.0 months; range, 3.0-212.0 months; 9 boys) in malignant group and 5 lesions (median diameter, 3.8 cm; range, 1.3-7.5 cm) from 5 patients (median age, 25.0 months; range, 0.7-216.0 months; 2 boys) in benign group. The diagnostic performance was assessed. Nonparametric and Chi-square tests were performed. With hyperenhancement plus wash-out, CEUS showed a sensitivity of 95.0% [95% confidence interval (CI): 75.1%, 99.9%], a specificity of 80.0% (CI: 28.4%, 99.5%), a positive predictive value of 95.0% (CI: 75.1%, 99.9%) and a negative predictive value of 80.0% (CI: 28.4%, 99.5%). It suggested that CEUS is a valuable technique for identifying between malignant and benign renal lesions in children.
Collapse
Affiliation(s)
- Yusi Fu
- Department of Ultrasound Diagnosis, The Second Xiang ya Hospital, Central South University, No 139, Renmin Middle Road, Changsha, 410011, Hunan, People's Republic of China
| | - Jia Zhong
- Department of Ultrasound, Mawangdui District of Hunan Provincial People's Hospital, Hunan Normal University, No 89, Guhan Road, Changsha, 410000, Hunan, People's Republic of China
| | - Yan Tan
- Department of Ultrasound Diagnosis, The Second Xiang ya Hospital, Central South University, No 139, Renmin Middle Road, Changsha, 410011, Hunan, People's Republic of China
| | - Taiqing Zheng
- Department of Pathology, Hunan Children's Hospital, No 86, Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Minghui Liu
- Department of Ultrasound Diagnosis, The Second Xiang ya Hospital, Central South University, No 139, Renmin Middle Road, Changsha, 410011, Hunan, People's Republic of China
| | - Guotao Wang
- Department of Ultrasound Diagnosis, The Second Xiang ya Hospital, Central South University, No 139, Renmin Middle Road, Changsha, 410011, Hunan, People's Republic of China.
| |
Collapse
|
30
|
Song YH, Li WL, Yang Z, Gao Y, Feng ZP. Loss of heterozygosity for chromosomes 16q in Wilms tumors predicts outcomes: A meta-analysis. World J Gastrointest Oncol 2024; 16:2159-2167. [PMID: 38764827 PMCID: PMC11099455 DOI: 10.4251/wjgo.v16.i5.2159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/06/2024] [Accepted: 03/12/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND The research findings suggest that the prognosis of children with Wilms tumor (WT) is affected by various factors. Some scholars have indicated that loss of heterozygosity (LOH) on chromosome 16q is associated with a poor prognosis in patients with WT. AIM To further elucidate this relationship, we conducted a meta-analysis. METHODS This meta-analysis was registered in INPLASY (INPLASY2023100060). We systematically searched databases including Embase, PubMed, Web of Science, Cochrane, and Google Scholar up to May 31, 2020, for randomized trials reporting any intrapartum fetal surveillance approach. The meta-analysis was performed within a frequentist framework, and the quality and network inconsistency of trials were assessed. Odds ratios and 95%CIs were calculated to report the relationship between event-free survival and 16q LOH in patients with WT. RESULTS Eleven cohort studies were included in this meta-analysis to estimate the relationship between event-free survival and 16q LOH in patients with WT (I2 = 25%, P < 0.001). As expected, 16q LOH can serve as an effective predictor of event-free survival in patients with WT (risk ratio = 1.95, 95%CI: 1.52-2.49, P < 0.001). CONCLUSION In pediatric patients with WT, there exists a partial correlation between 16q LOH and an unfavorable treatment prognosis. Clinical detection of 16q chromosome LOH warrants increased attention to the patient's prognosis.
Collapse
Affiliation(s)
- Yuan-Hua Song
- Department of Oncology, Kunming Children's Hospital, Kunming 650103, Yunnan Province, China
| | - Wen-Ling Li
- Department of Oncology, Kunming Children's Hospital, Kunming 650103, Yunnan Province, China
| | - Zhen Yang
- Department of Oncology, Kunming Children's Hospital, Kunming 650103, Yunnan Province, China
| | - Yan Gao
- Department of Oncology, Kunming Children's Hospital, Kunming 650103, Yunnan Province, China
| | - Zhi-Ping Feng
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming 650118, Yunnan Province, China
| |
Collapse
|
31
|
Li W, Sun Y, Zhang G, Yang Q, Wang B, Ma X, Zhang H. Automated segmentation and volume prediction in pediatric Wilms' tumor CT using nnu-net. BMC Pediatr 2024; 24:321. [PMID: 38724944 PMCID: PMC11080230 DOI: 10.1186/s12887-024-04775-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 04/18/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Radiologic volumetric evaluation of Wilms' tumor (WT) is an important indicator to guide treatment decisions. However, due to the heterogeneity of the tumors, radiologists have main-guard differences in diagnosis that can lead to misdiagnosis and poor treatment. The aim of this study was to explore whether CT-based outlining of WT foci can be automated using deep learning. METHODS We included CT intravenous phase images of 105 patients with WT and double-blind outlining of lesions by two radiologists. Then, we trained an automatic segmentation model using nnUnet. The Dice similarity coefficient (DSC) and 95th percentile Hausdorff distance (HD95) were used to assess the performance. Next, we optimized the automatic segmentation results based on the ratio of the three-dimensional diameter of the lesion to improve the performance of volumetric assessment. RESULTS The DSC and HD95 was 0.83 ± 0.22 and 10.50 ± 8.98 mm. The absolute difference and percentage difference in tumor size was 72.27 ± 134.84 cm3 and 21.08% ± 30.46%. After optimization according to our method, it decreased to 40.22 ± 96.06 cm3 and 10.16% ± 9.70%. CONCLUSION We introduce a novel method that enhances the accuracy of predicting WT volume by integrating AI automated outlining and 3D tumor diameters. This approach surpasses the accuracy of using AI outcomes alone and has the potential to enhance the clinical evaluation of pediatric patients with WT. By intertwining AI outcomes with clinical data, this method becomes more interpretive and offers promising applications beyond Wilms tumor, extending to other pediatric diseases.
Collapse
Affiliation(s)
- Weikang Li
- Department of Radiology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binshneg Rd, Hangzhou, China
| | - Yiran Sun
- Wenzhou Medical University, Wenzhou, China
| | - Guoxun Zhang
- Department of Radiology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binshneg Rd, Hangzhou, China
| | - Qing Yang
- Department of Radiology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binshneg Rd, Hangzhou, China
| | - Bo Wang
- Department of Radiology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binshneg Rd, Hangzhou, China
| | - Xiaohui Ma
- Department of Radiology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binshneg Rd, Hangzhou, China.
| | - Hongxi Zhang
- Department of Radiology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333, Binshneg Rd, Hangzhou, China.
| |
Collapse
|
32
|
Green R, Ahmed A, Fleming B, Long AM, Behjati S, Trotman J, Tarpey P, Nicholson JC, Coleman N, Elizabeth Hook C, Murray MJ. Wilms Tumor With Raised Serum Alpha-Fetoprotein: Highlighting the Need for Novel Circulating Biomarkers. Pediatr Dev Pathol 2024; 27:260-265. [PMID: 38098239 PMCID: PMC11088205 DOI: 10.1177/10935266231213467] [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] [Indexed: 05/12/2024]
Abstract
Wilms tumor (WT) is the commonest cause of renal cancer in children. In Europe, a diagnosis is made for most cases on typical clinical and radiological findings, prior to pre-operative chemotherapy. Here, we describe a case of a young boy presenting with a large abdominal tumor, associated with raised serum alpha-fetoprotein (AFP) levels at diagnosis. Given the atypical features present, a biopsy was taken, and histology was consistent with WT, showing triphasic WT, with epithelial, stromal, and blastemal elements present, and positive WT1 and CD56 immunohistochemical staining. During pre-operative chemotherapy, serial serum AFP measurements showed further increases, despite a radiological response, before a subsequent fall to normal following nephrectomy. The resection specimen was comprised of ~55% and ~45% stromal and epithelial elements, respectively, with no anaplasia, but immunohistochemistry using AFP staining revealed positive mucinous intestinal epithelium, consistent with the serum AFP observations. The lack of correlation between tumor response and serum AFP levels in this case highlights a more general clinical unmet need to identify WT-specific circulating tumor markers.
Collapse
Affiliation(s)
- Rebecca Green
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Adeeb Ahmed
- Department of Paediatrics, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, Norfolk, UK
| | - Ben Fleming
- Department of Radiology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Anna-May Long
- Department of Paediatric Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sam Behjati
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Jamie Trotman
- East Genomics Laboratory Hub (GLH) Genetics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Patrick Tarpey
- East Genomics Laboratory Hub (GLH) Genetics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - James C. Nicholson
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, Level 8, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Nicholas Coleman
- Department of Paediatric Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - C. Elizabeth Hook
- Department of Paediatric Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Matthew J. Murray
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
33
|
Banerjee A, Babu R, Jayaraman D, Chilukuri S. Preoperative three-dimensional modelling and virtual reality planning aids nephron sparing surgery in a child with bilateral Wilms tumour. BMJ Case Rep 2024; 17:e260600. [PMID: 38642931 PMCID: PMC11033631 DOI: 10.1136/bcr-2024-260600] [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] [Indexed: 04/22/2024] Open
Abstract
Bilateral Wilms tumour (BWT) is a surgically challenging condition. Virtual reality (VR) reconstruction aids surgeons to foresee the anatomy ahead of Nephron Sparing Surgery (NSS). Three-dimensional (3D) visualisation improves the anatomical orientation of surgeons performing NSS. We herewith report a case of BWT where VR planning and 3D printing were used to aid NSS. Conventional imaging is often found to be inadequate while assessing the tumour-organ-vascular anatomy. Advances like VR and 3D printing help surgeons plan better for complex surgeries like bilateral NSS. Next-generation extended reality tools will likely aid robotic-assisted precision NSS and improve patient outcomes.
Collapse
Affiliation(s)
- Avijit Banerjee
- Urology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Ramesh Babu
- Pediatric Urology, Sri Ramachandra University Medical College, Chennai, India
| | - Dhaarani Jayaraman
- Paediatric Hematology and Oncology, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | | |
Collapse
|
34
|
Glembocki AI, Somers GR. Prognostic and predictive biomarkers in paediatric solid tumours. Pathology 2024; 56:283-296. [PMID: 38216399 DOI: 10.1016/j.pathol.2023.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 01/14/2024]
Abstract
Characterisation of histological, immunohistochemical and molecular prognostic and predictive biomarkers has contributed significantly to precision medicine and better outcomes in the management of paediatric solid tumours. Prognostic biomarkers allow predictions to be made regarding a tumour's aggressiveness and clinical course, whereas predictive biomarkers help determine responses to a specific treatment. This review summarises prognostic biomarkers currently used in the more common paediatric solid tumours, with a brief commentary on the most relevant less common predictive biomarkers. MYCN amplification is the most important genetic alteration in neuroblastoma prognosis, and the histological classification devised by Shimada in 1999 is still used in routine diagnosis. Moreover, a new subgrouping of unfavourable histology neuroblastoma enables immunohistochemical characterisation of tumours with markedly different genetic features and prognosis. The predominant histology and commonly observed cytogenetic abnormalities are recognised outcome predictors in Wilms tumour. Evaluation for anaplasia, which is tightly associated with TP53 gene mutations and poor outcomes, is central in both the International Society of Paediatric Oncology and the Children's Oncology Group approaches to disease classification. Characterisation of distinct genotype-phenotype subclasses and critical mutations has expanded overall understanding of hepatoblastoma outcomes. The C1 subclass hepatoblastoma and CTNNB1 mutations are associated with good prognosis. In contrast, the C2 subclass, NFE2L2 mutations, TERT promoter mutations and high expression of oncofetal proteins and stem cell markers are associated with poor outcomes. Risk stratification in sarcomas is highly variable depending on the entity. The prognosis of rhabdomyosarcoma, for example, primarily depends on histological and molecular characteristics. Advances in our understanding of clinically significant biomarkers will translate into more precise diagnoses, improved risk stratification and more effective and less toxic treatment in this challenging group of patients.
Collapse
Affiliation(s)
- Aida I Glembocki
- Division of Pathology, Department of Paediatric Laboratory Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Gino R Somers
- Division of Pathology, Department of Paediatric Laboratory Medicine, Hospital for Sick Children, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
35
|
Vujanić GM, Graf N, D'Hooghe E, Pritchard-Jones K, Bergeron C, Tinteren HV, Furtwängler R. Omission of adjuvant chemotherapy in patients with completely necrotic Wilms tumor stage I and radiotherapy in stage III: The 30-year SIOP-RTSG experience. Pediatr Blood Cancer 2024; 71:e30852. [PMID: 38185745 DOI: 10.1002/pbc.30852] [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: 09/19/2023] [Revised: 11/27/2023] [Accepted: 12/21/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Completely necrotic Wilms tumor (CN-WT) following preoperative chemotherapy has been regarded as low-risk WT since the International Society of Paediatric Oncology (SIOP) 93-01 study, and patients have been treated with reduced postoperative therapy. The aim of the study was to evaluate whether the omission of adjuvant chemotherapy in patients with localized CN-WT stage I and radiotherapy in stage III was safe. PATIENTS AND METHODS The retrospective observational study of outcomes of patients diagnosed with localized CN-WT on central pathology review and treated according to the SIOP 93-01 and SIOP-WT-2001 protocols (1993-2022). RESULTS There were 125 patients with localized CN-WT: 90 with stage I, 10 with stage II, and 25 with stage III. Sixty-two of 125 (49.6%) patients had a discrepant diagnosis and/or staging between the institutional pathologist and central pathology review. In the group of 90 patients with stage I, postoperative chemotherapy was not given to 41 (46%) patients, whereas 49 patients received postoperative chemotherapy-in the latter group, two patients relapsed, and one of them died. One stage I and one stage II patient developed chemotherapy-induced toxicity and died. Nineteen of 25 patients with stage III received no flank radiotherapy. No stage III patient relapsed or died. The overall 5-year event-free survival (EFS) estimate for the entire cohort (stages I-III) was 96.8% [95% confidence interval, CI: 93.6%-99.6%] and the overall survival (OS) was 97.6% [95% CI: 95.0-100%]. The EFS and OS were 97% and 98%, respectively, for stage I, and 100% for stage III. CONCLUSION Omission of postoperative chemotherapy for patients with CN-WT stage I, and radiotherapy for stage III is safe. Rapid central pathology review is required to assign appropriate treatment and avoid treatment-related side effects.
Collapse
Affiliation(s)
- Gordan M Vujanić
- Department of Pathology, Sidra Medicine, Doha, Qatar
- Weill Cornell Medicine - Qatar, Doha, Qatar
| | - Norbert Graf
- Department of Hematology and Oncology, Saarland University Hospital, Homburg, Germany
| | - Ellen D'Hooghe
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | | | - Christophe Bergeron
- Institut d'Hematologie et d'Oncologie Pédiatrique, Centre Léon Bérard, Lyon, France
| | - Harm van Tinteren
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Rhoikos Furtwängler
- Department of Hematology and Oncology, Saarland University Hospital, Homburg, Germany
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Inselspital University Hospital, Bern, Switzerland
| |
Collapse
|
36
|
Perotti D, Williams RD, Wegert J, Brzezinski J, Maschietto M, Ciceri S, Gisselsson D, Gadd S, Walz AL, Furtwaengler R, Drost J, Al-Saadi R, Evageliou N, Gooskens SL, Hong AL, Murphy AJ, Ortiz MV, O'Sullivan MJ, Mullen EA, van den Heuvel-Eibrink MM, Fernandez CV, Graf N, Grundy PE, Geller JI, Dome JS, Perlman EJ, Gessler M, Huff V, Pritchard-Jones K. Hallmark discoveries in the biology of Wilms tumour. Nat Rev Urol 2024; 21:158-180. [PMID: 37848532 DOI: 10.1038/s41585-023-00824-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2023] [Indexed: 10/19/2023]
Abstract
The modern study of Wilms tumour was prompted nearly 50 years ago, when Alfred Knudson proposed the 'two-hit' model of tumour development. Since then, the efforts of researchers worldwide have substantially expanded our knowledge of Wilms tumour biology, including major advances in genetics - from cloning the first Wilms tumour gene to high-throughput studies that have revealed the genetic landscape of this tumour. These discoveries improve understanding of the embryonal origin of Wilms tumour, familial occurrences and associated syndromic conditions. Many efforts have been made to find and clinically apply prognostic biomarkers to Wilms tumour, for which outcomes are generally favourable, but treatment of some affected individuals remains challenging. Challenges are also posed by the intratumoural heterogeneity of biomarkers. Furthermore, preclinical models of Wilms tumour, from cell lines to organoid cultures, have evolved. Despite these many achievements, much still remains to be discovered: further molecular understanding of relapse in Wilms tumour and of the multiple origins of bilateral Wilms tumour are two examples of areas under active investigation. International collaboration, especially when large tumour series are required to obtain robust data, will help to answer some of the remaining unresolved questions.
Collapse
Affiliation(s)
- Daniela Perotti
- Predictive Medicine: Molecular Bases of Genetic Risk, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - 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
| | - Jenny Wegert
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Wuerzburg University, Wuerzburg, Germany
| | - Jack Brzezinski
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Mariana Maschietto
- Research Center, Boldrini Children's Hospital, Campinas, São Paulo, Brazil
| | - Sara Ciceri
- Predictive Medicine: Molecular Bases of Genetic Risk, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - David Gisselsson
- Cancer Cell Evolution Unit, Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Clinical Genetics, Pathology and Molecular Diagnostics, Office of Medical Services, Skåne, Sweden
| | - Samantha Gadd
- Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Amy L Walz
- Division of Hematology,Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Rhoikos Furtwaengler
- Division of Pediatric Oncology and Hematology, Department of Pediatrics, Inselspital Bern University, Bern, Switzerland
| | - Jarno Drost
- Princess Máxima Center for Paediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - 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
| | - Nicholas Evageliou
- Divisions of Hematology and Oncology, Children's Hospital of Philadelphia, CHOP Specialty Care Center, Vorhees, NJ, USA
| | - Saskia L Gooskens
- Princess Máxima Center for Paediatric Oncology, Utrecht, Netherlands
| | - Andrew L Hong
- Aflac Cancer and Blood Disorders Center, Emory University and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Andrew J Murphy
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Michael V Ortiz
- Department of Paediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maureen J O'Sullivan
- Histology Laboratory, Children's Health Ireland at Crumlin, Dublin, Ireland
- Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland
| | - Elizabeth A Mullen
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | | | - Conrad V Fernandez
- Division of Paediatric Hematology Oncology, IWK Health Centre and Dalhousie University, Halifax, Nova Scotia, Canada
| | - Norbert Graf
- Department of Paediatric Oncology and Hematology, Saarland University Hospital, Homburg, Germany
| | - Paul E Grundy
- Department of Paediatrics Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - James I Geller
- Division of Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Jeffrey S Dome
- Division of Oncology, Center for Cancer and Blood Disorders, Children's National Hospital and the Department of Paediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Elizabeth J Perlman
- Department of Pathology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Manfred Gessler
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Wuerzburg University, Wuerzburg, Germany
- Comprehensive Cancer Center Mainfranken, Wuerzburg, Germany
| | - Vicki Huff
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 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
|
37
|
Pire A, Hirsch TZ, Morcrette G, Imbeaud S, Gupta B, Pilet J, Cornet M, Fabre M, Guettier C, Branchereau S, Brugières L, Guerin F, Laithier V, Coze C, Nagae G, Hiyama E, Laurent-Puig P, Rebouissou S, Sarnacki S, Chardot C, Capito C, Faure-Conter C, Aerts I, Taque S, Fresneau B, Zucman-Rossi J. Mutational signature, cancer driver genes mutations and transcriptomic subgroups predict hepatoblastoma survival. Eur J Cancer 2024; 200:113583. [PMID: 38330765 DOI: 10.1016/j.ejca.2024.113583] [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: 08/16/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Hepatoblastoma is the most frequent pediatric liver cancer. The current treatments lead to 80% of survival rate at 5 years. In this study, we evaluated the clinical relevance of molecular features to identify patients at risk of chemoresistance, relapse and death of disease. METHODS All the clinical data of 86 children with hepatoblastoma were retrospectively collected. Pathological slides were reviewed, tumor DNA sequencing (by whole exome, whole genome or target) and transcriptomic profiling with RNAseq or 300-genes panel were performed. Associations between the clinical, pathological, mutational and transcriptomic data were investigated. RESULTS High-risk patients represented 44% of our series and the median age at diagnosis was 21.9 months (range: 0-208). Alterations of the WNT/ß-catenin pathway and of the 11p15.5 imprinted locus were identified in 98% and 74% of the tumors, respectively. Other cancer driver genes mutations were only found in less than 11% of tumors. After neoadjuvant chemotherapy, disease-specific survival and poor response to neoadjuvant chemotherapy were associated with 'Liver Progenitor' (p = 0.00049, p < 0.0001) and 'Immune Cold' (p = 0.0011, p < 0.0001) transcriptomic tumor subtypes, SBS35 cisplatin mutational signature (p = 0.018, p = 0.001), mutations in rare cancer driver genes (p = 0.0039, p = 0.0017) and embryonal predominant histological type (p = 0.0013, p = 0.0077), respectively. Integration of the clinical and molecular features revealed a cluster of molecular markers associated with resistance to chemotherapy and survival, enlightening transcriptomic 'Immune Cold' and Liver Progenitor' as a predictor of survival independent of the clinical features. CONCLUSIONS Response to neoadjuvant chemotherapy and survival in children treated for hepatoblastoma are associated with genomic and pathological features independently of the clinical features.
Collapse
Affiliation(s)
- Aurore Pire
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, F-75006 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer, Labex Onco-Immunology, Institute du Cancer Paris CARPEM, AP-HP, F-75015 Paris, France; Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Bruxelles, Belgium
| | - Theo Z Hirsch
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, F-75006 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer, Labex Onco-Immunology, Institute du Cancer Paris CARPEM, AP-HP, F-75015 Paris, France
| | - Guillaume Morcrette
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, F-75006 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer, Labex Onco-Immunology, Institute du Cancer Paris CARPEM, AP-HP, F-75015 Paris, France; Pathology Department, AP-HP Necker Enfants Malades Hospital, F-75015 Paris, France
| | - Sandrine Imbeaud
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, F-75006 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer, Labex Onco-Immunology, Institute du Cancer Paris CARPEM, AP-HP, F-75015 Paris, France
| | - Barkha Gupta
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, F-75006 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer, Labex Onco-Immunology, Institute du Cancer Paris CARPEM, AP-HP, F-75015 Paris, France
| | - Jill Pilet
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, F-75006 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer, Labex Onco-Immunology, Institute du Cancer Paris CARPEM, AP-HP, F-75015 Paris, France
| | - Marianna Cornet
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, F-75006 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer, Labex Onco-Immunology, Institute du Cancer Paris CARPEM, AP-HP, F-75015 Paris, France
| | - Monique Fabre
- Pathology Department, AP-HP Necker Enfants Malades Hospital, F-75015 Paris, France
| | - Catherine Guettier
- Department of Pathology, AP-HP Bicêtre Hospital, F-94270 Le Kremlin-Bicêtre, France
| | - Sophie Branchereau
- Department of Pediatric Surgery, AP-HP Bicêtre Hospital, F-94270 Le Kremlin-Bicêtre, France
| | - Laurence Brugières
- Gustave Roussy, Université Paris-Saclay, Department of Children and Adolescents Oncology, Villejuif F-94805, France
| | - Florent Guerin
- Department of Pediatric Surgery, AP-HP Bicêtre Hospital, F-94270 Le Kremlin-Bicêtre, France
| | | | - Carole Coze
- Department of Pediatric and Oncology, Hopital de La Timone, Aix Marseille University, F-13005 Marseille, France
| | - Genta Nagae
- Genome Science Laboratory, Research Center for Advanced Science and Technology (RCAST), the University of Tokyo, Tokyo, Japan
| | - Eiso Hiyama
- Department of Pediatric Surgery, Hiroshima University Hospital, Hiroshima, Japan; Department of Biomedical Science, Natural Science Center for Basic Research and Development (N-BARD), Hiroshima University, Hiroshima, Japan
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, F-75006 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer, Labex Onco-Immunology, Institute du Cancer Paris CARPEM, AP-HP, F-75015 Paris, France
| | - Sandra Rebouissou
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, F-75006 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer, Labex Onco-Immunology, Institute du Cancer Paris CARPEM, AP-HP, F-75015 Paris, France
| | - Sabine Sarnacki
- Department of Pediatric Surgery, AP-HP Necker Enfants Malades Hospital, F-75015 Paris, France
| | - Christophe Chardot
- Department of Pediatric Surgery, AP-HP Necker Enfants Malades Hospital, F-75015 Paris, France
| | - Carmen Capito
- Department of Pediatric Surgery, AP-HP Necker Enfants Malades Hospital, F-75015 Paris, France
| | - Cécile Faure-Conter
- Institut d'hématologie et d'oncologie pédiatrique de Lyon, F-69008 Lyon, France
| | - Isabelle Aerts
- Institut Curie, Oncology Center SIREDO, F-75005 Paris, France
| | - Sophie Taque
- Pediatric Department hemato-oncology, CHU Rennes, F-35033 Rennes, France
| | - Brice Fresneau
- Gustave Roussy, Université Paris-Saclay, Department of Children and Adolescents Oncology, Villejuif F-94805, France; Université Paris-Saclay, Université Paris-Sud, UVSQ, CESP, Cancer and Radiation Team, F-94805 Villejuif, France
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, Inserm, F-75006 Paris, France; Equipe Labellisée Ligue Nationale Contre le Cancer, Labex Onco-Immunology, Institute du Cancer Paris CARPEM, AP-HP, F-75015 Paris, France; AP-HP, Department of Oncology, Hopital Européen Georges Pompidou, F-75015 Paris, France.
| |
Collapse
|
38
|
Fasler-Kan E, Milošević M, Ruggiero S, Aliu N, Cholewa D, Häcker FM, Dekany G, Bartenstein A, Berger SM. Cytokine Signaling in Pediatric Kidney Tumor Cell Lines WT-CLS1, WT-3ab and G-401. Int J Mol Sci 2024; 25:2281. [PMID: 38396958 PMCID: PMC10889092 DOI: 10.3390/ijms25042281] [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] [Received: 12/20/2023] [Revised: 02/01/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Renal tumors comprise ~7% of all malignant pediatric tumors. Approximately 90% of pediatric kidney tumors comprise Wilms tumors, and the remaining 10% include clear cell sarcoma of the kidney, malignant rhabdoid tumor of the kidney, renal cell carcinoma and other rare renal tumors. Over the last 30 years, the role of cytokines and their receptors has been considerably investigated in both cancer progression and anti-cancer therapy. However, more effective immunotherapies require the cytokine profiling of each tumor type and comprehensive understanding of tumor biology. In this study, we aimed to investigate the activation of signaling pathways in response to cytokines in three pediatric kidney tumor cell lines, in WT-CLS1 and WT-3ab cells (both are Wilms tumors), and in G-401 cells (a rhabdoid kidney tumor, formerly classified as Wilms tumor). We observed that interferon-alpha (IFN-α) and interferon-gamma (IFN-γ) very strongly induced the activation of the STAT1 protein, whereas IL-6 and IFN-α activated STAT3 and IL-4 activated STAT6 in all examined tumor cell lines. STAT protein activation was examined by flow cytometry and Western blot using phospho-specific anti-STAT antibodies which recognize only activated (phosphorylated) STAT proteins. Nuclear translocation of phospho-STAT proteins upon activation with specific cytokines was furthermore confirmed by immunofluorescence. Our results also showed that both IFN-α and IFN-γ caused upregulation of major histocompatibility complex (MHC) class I proteins, however, these cytokines did not have any effect on the expression of MHC class II proteins. We also observed that pediatric kidney tumor cell lines exhibit the functional expression of an additional cytokine signaling pathway, the tumor necrosis factor (TNF)-α-mediated activation of nuclear factor kappa B (NF-κB). In summary, our data show that human pediatric renal tumor cell lines are responsive to stimulation with various human cytokines and could be used as in vitro models for profiling cytokine signaling pathways.
Collapse
Affiliation(s)
- Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, CH-3010 Bern, Switzerland; (M.M.); (S.R.); (D.C.); (G.D.); (A.B.)
| | - Milan Milošević
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, CH-3010 Bern, Switzerland; (M.M.); (S.R.); (D.C.); (G.D.); (A.B.)
| | - Sabrina Ruggiero
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, CH-3010 Bern, Switzerland; (M.M.); (S.R.); (D.C.); (G.D.); (A.B.)
| | - Nijas Aliu
- Department of Human Genetics, Inselspital Bern, University of Bern, CH-3010 Bern, Switzerland;
| | - Dietmar Cholewa
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, CH-3010 Bern, Switzerland; (M.M.); (S.R.); (D.C.); (G.D.); (A.B.)
| | - Frank-Martin Häcker
- Department of Pediatric Surgery, Children’s Hospital of Eastern Switzerland, CH-9000 St. Gallen, Switzerland;
- Faculty of Medicine, University of Basel, CH-4031 Basel, Switzerland
| | - Gabriela Dekany
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, CH-3010 Bern, Switzerland; (M.M.); (S.R.); (D.C.); (G.D.); (A.B.)
| | - Andreas Bartenstein
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, CH-3010 Bern, Switzerland; (M.M.); (S.R.); (D.C.); (G.D.); (A.B.)
| | - Steffen M. Berger
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, CH-3010 Bern, Switzerland; (M.M.); (S.R.); (D.C.); (G.D.); (A.B.)
| |
Collapse
|
39
|
Li P, Tao Y, Zhao Y, Lyu X, Zhou X, Zhuo R, Ma L, Tao T, Zhou H. Robotic-assisted laparoscopic surgery for the treatment of Wilms' tumor in children: single-center experience and medium-term outcomes. J Robot Surg 2024; 18:3. [PMID: 38175361 DOI: 10.1007/s11701-023-01759-9] [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] [Received: 08/31/2023] [Accepted: 10/28/2023] [Indexed: 01/05/2024]
Abstract
To report our institutional experience and the medium-term outcomes of utilizing robotic-assisted laparoscopic surgery (RALS) in patients with Wilms' tumor (WT). The robotic surgical interventions include nephron-sparing surgery (RAL-NSS), radical nephrectomy (RAL-RN), and nephrectomy with inferior vena cava thrombectomy (RAL-N-IVCT). We retrospectively collected medical records of WT patients who underwent RALS in our center between August 2019 and February 2022. Patients' baseline demographics, preoperative parameters, and perioperative/postoperative data were recorded and analyzed. Follow-up results were collected to evaluate the oncological outcomes. A total of 12 patients (13 sides) with a median age of 30 (IQR: 19.5-45.5) months were included. All operations were successfully completed without conversion. Seven patients received preoperative chemotherapy. Distribution of surgical interventions was as follows: five patients underwent RAL-RN, five received RAL-NSS, one with bilateral WT underwent concurrent RAL-RN and RAL-NSS, and one received RAL-RN-IVCT post preoperative chemotherapy. Postoperative chemotherapy was conducted in ten patients. The estimated intraoperative blood loss was 27 ± 4.0 ml for the RAL-NSS group, 41.67 ± 12.13 ml for the RAL-RN group, and 350 ml for the RAL-RN-IVCT groups, respectively. The median perioperative serum creatinine levels were 32.5 (IQR: 30.75-39.5) μmol/l preoperatively and 35 (IQR: 31.75-38.5) μmol/l postoperatively, which showed no significant difference. No positive lymph nodes were detected. Postoperative chemotherapy was performed according to the tumor volume and pathological findings. The median follow-up time was 17.5 (15.8-22.3) months. During this interval, neither distant metastasis nor recurrence was identified. Based on our medium-term follow-up observations, RAL-NSS, RAL-RN, and RAL-RN-IVCT exhibit promising feasibility and safety profiles in the therapeutic landscape of WT.
Collapse
Affiliation(s)
- Pin Li
- Department of Urology, Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, No. 5 Nan Mencang, Dongcheng District, Beijing, 100700, People's Republic of China
- Department of Urology, Bayi Children's Hospital, Affiliated to The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, People's Republic of China
| | - Yuandong Tao
- Department of Urology, Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, No. 5 Nan Mencang, Dongcheng District, Beijing, 100700, People's Republic of China
- Department of Urology, Bayi Children's Hospital, Affiliated to The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, People's Republic of China
| | - Yang Zhao
- Department of Urology, Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, No. 5 Nan Mencang, Dongcheng District, Beijing, 100700, People's Republic of China
- Department of Urology, Bayi Children's Hospital, Affiliated to The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, People's Republic of China
- Medical School of Chinese PLA, Beijing, 100853, People's Republic of China
| | - Xuexue Lyu
- Department of Urology, Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, No. 5 Nan Mencang, Dongcheng District, Beijing, 100700, People's Republic of China
- Department of Urology, Bayi Children's Hospital, Affiliated to The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, People's Republic of China
- Medical School of Chinese PLA, Beijing, 100853, People's Republic of China
| | - Xiaoguang Zhou
- Department of Urology, Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, No. 5 Nan Mencang, Dongcheng District, Beijing, 100700, People's Republic of China
- Department of Urology, Bayi Children's Hospital, Affiliated to The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, People's Republic of China
| | - Ran Zhuo
- Department of Urology, Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, No. 5 Nan Mencang, Dongcheng District, Beijing, 100700, People's Republic of China
- Department of Urology, Bayi Children's Hospital, Affiliated to The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, People's Republic of China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Lifei Ma
- Department of Urology, Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, No. 5 Nan Mencang, Dongcheng District, Beijing, 100700, People's Republic of China
- Department of Urology, Bayi Children's Hospital, Affiliated to The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, People's Republic of China
| | - Tian Tao
- Department of Urology, Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, No. 5 Nan Mencang, Dongcheng District, Beijing, 100700, People's Republic of China
- Department of Urology, Bayi Children's Hospital, Affiliated to The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, People's Republic of China
| | - Huixia Zhou
- Department of Urology, Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, No. 5 Nan Mencang, Dongcheng District, Beijing, 100700, People's Republic of China.
- Department of Urology, Bayi Children's Hospital, Affiliated to The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, People's Republic of China.
- Medical School of Chinese PLA, Beijing, 100853, People's Republic of China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
40
|
Pasam MK, Rao BV, Chaganty SK, Sharma RM, Patil V, Kodandapani S, Challa S, Thammineedi SR. Grossing to reporting of Wilms tumor with emphasis on proper sampling in treatment-naive and postchemotherapy specimens and their clinicopathological correlation with outcome. Urol Ann 2024; 16:87-93. [PMID: 38415234 PMCID: PMC10896324 DOI: 10.4103/ua.ua_60_23] [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: 06/07/2023] [Accepted: 10/31/2023] [Indexed: 02/29/2024] Open
Abstract
Context Emphasis on grossing to reporting for the assessment of histopathological parameters predicting outcomes in Wilms tumor. Aims To analyze various clinicopathological parameters that effect outcomes in treatment naïve and post chemotherapy Wilms tumor specimens. Settings and Design This was a retrospective observational study. Subjects and Methods All patients diagnosed with Wilms tumor between 2012 and 2018 at our institute will be included with their clinical findings, laboratory reports, and radiological findings. The patients will be categorized into two groups based on treatment protocol (Society of Pediatric Oncology (SIOP) or the National Wilms Tumor Study Group/Children's Oncology Group (COG) guidelines) used. Details of Grossing and reporting protocols used for the in pre treatment and post treatment specimens will be analyzed. Follow-up till December 2020 will be analyzed. Statistical Analysis Used Chi-square and Fisher's exact tests were used for statistical analysis. Results A total of 36 patients with the diagnosis of Wilms tumor were included in the present study. The mean age of presentation was 3.9 ± 0.7 years, and males were more common than females. Most of them presented as abdominal mass and few with isolated hematuria. Twenty-six (72%) patients were treated under SIOP protocol with preoperative neoadjuvant chemotherapy. Ten patients underwent upfront surgery as per COG protocol. In SIOP group patients, the mean tumor size was 9.3cm. Forty percent (n = 10) we mixed histological type followed by blastemal type constituting (32%, n = 8). Regressive and epithelial histological types constituted 16% (n = 4) and 12% (n = 3), respectively. In the SIOP group 72% (n = 19) had no anaplasia and 28% (n = 7) had anaplasia. Fifty seven percent (n = 15) cases were Stage I, followed by 26.9% n = 7) and 11.5% (n = 3) being Stage II and Stage III, respectively. Ten patients underwent upfront surgery as per COG protocol. The mean tumor size among this group was 8 cm ranging from 7 cm to 11 cm. Eight (80%) cases had favorable histology and two cases showed focal anaplasia. Heterologous differentiation is seen in 3 (70%). Out of the 10 cases, one case was Stage I, six were Stage 2, one was Stage III, and two were clinical Stage IV. None of the cases showed either vessel or lymph node metastasis. All the patients received adjuvant chemotherapy postsurgery and were followed up till December 2020 for (at least 3 years). Of 25 patients in the SIOP group, 18 (72%) had complete remission with no radiological evidence of residual disease. Of the 10 patients in the COG group, 6 (70%) had complete remission. Conclusions Histopathological evaluation of Wilms tumor is a critical aspect in the management of Wilms tumor, as tumor characteristics are different in the tumors treated under SIOP and COG protocols, which will ultimately affect the prognostic risk stratification. This necessitates the knowledge of the important grossing and reporting of these tumors under the two protocols.
Collapse
Affiliation(s)
- Mohan Krishna Pasam
- Department of Pathology and Lab Medicine, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, Telangana, India
| | - B Vishal Rao
- Department of Pathology and Lab Medicine, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, Telangana, India
| | - Sai Kiran Chaganty
- Department of Pathology and Lab Medicine, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, Telangana, India
| | - Rakesh Manilal Sharma
- Department of Surgical Oncology, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, Telangana, India
| | - Veerendra Patil
- Department of Medical Oncology, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, Telangana, India
| | - Suseela Kodandapani
- Department of Pathology and Lab Medicine, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, Telangana, India
| | - Sundaram Challa
- Department of Pathology and Lab Medicine, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, Telangana, India
| | - Subramanyeshwar Rao Thammineedi
- Department of Surgical Oncology, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, Telangana, India
| |
Collapse
|
41
|
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
|
42
|
Alhashim M, Anan N, Tamal M, Altarrah H, Alshaibani S, Hill R. A review on optimization of Wilms tumour management using radiomics. BJR Open 2024; 6:tzae034. [PMID: 39483333 PMCID: PMC11525052 DOI: 10.1093/bjro/tzae034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 08/26/2024] [Accepted: 10/03/2024] [Indexed: 11/03/2024] Open
Abstract
Background Wilms tumour, a common paediatric cancer, is difficult to treat in low- and middle-income countries due to limited access to imaging. Artificial intelligence (AI) has been introduced for staging, detecting, and classifying tumours, aiding physicians in decision-making. However, challenges include algorithm accuracy, translation into conventional diagnosis, reproducibility, and reliability. As AI technology advances, radiomics, an AI tool, emerges to extract tumour morphology and stage information. Objectives This review explores the application of radiomics in Wilms tumour management, including its potential in diagnosis, prognosis, and treatment. Additionally, it discusses the future prospects of AI in this field and potential directions for automation-aided Wilms tumour treatment. Methods The review analyses various research studies and articles on the use of radiomics in Wilms tumour management. This includes studies on automated deep learning-based classification, interobserver variability in histopathological analysis, and the application of AI in staging, detecting, and classifying Wilms tumours. Results The review finds that radiomics offers several promising applications in Wilms tumour management, including improved diagnosis: it helps in classifying Wilms tumours from other paediatric kidney tumours, prognosis prediction: radiomic features can be used to predict both staging and response to preoperative chemotherapy, Treatment response assessment: Radiomics can be used to monitor the response of Wilms and to predict the feasibility of nephron-sparing surgery. Conclusions This review concludes that radiomics has the potential to significantly improve the diagnosis, prognosis, and treatment of Wilms tumours. Despite some challenges, such as the need for further research and validation, AI integration in Wilms tumour management offers promising opportunities for improved patient care. Advances in knowledge This review provides a comprehensive overview of the potential applications of radiomics in Wilms tumour management and highlights the significant role AI can play in improving patient outcomes. It contributes to the growing body of knowledge on AI-assisted diagnosis and treatment of paediatric cancers.
Collapse
Affiliation(s)
- Maryam Alhashim
- Radiology Department, College of Medicine, Imam Abdulrahman Bin Faisal University, King Faisal Ibn Abd Al Aziz Rd, Dammam 34212, Saudi Arabia
- Medical Imaging Services Center, King Fahad Specialist Hospital Dammam, Dammam 32253, Saudi Arabia
| | - Noushin Anan
- Department of Biomedical Imaging, Advanced Medical and Dental Institute, Universiti Sains Malaysia, SAINS@BERTAM, 13200, Kepala Batas, Pulau Pinang, Malaysia
| | - Mahbubunnabi Tamal
- College of Engineering, Imam Abdulrahman Bin Faisal University, Dammam 31451, Saudi Arabia
| | - Hibah Altarrah
- Oncology center, King Fahad Specialist Hospital Dammam, Dammam 32253, Saudi Arabia
| | - Sarah Alshaibani
- Medical Imaging Services Center, King Fahad Specialist Hospital Dammam, Dammam 32253, Saudi Arabia
| | - Robin Hill
- Department of Radiation Oncology, Chris O'Brien Lifehouse, Sydney 2050, Australia
| |
Collapse
|
43
|
Alhassoun A, Bara AH, Ibrahim MN, Berro SM, Khalil MG. Cystic partially differentiated nephroblastoma in an 18-month-old girl: a case report. Ann Med Surg (Lond) 2023; 85:6148-6151. [PMID: 38098581 PMCID: PMC10718359 DOI: 10.1097/ms9.0000000000001245] [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: 07/07/2023] [Accepted: 08/20/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction and importance Cystic partially differentiated nephroblastoma (CPDN) is a rare cystic tumor that affects the kidney. It has a low potential for malignancy. It usually presents as an abdominal mass. It may be difficult to confirm the diagnosis of CPDN without a histopathological study. Case presentation The authors report a case of an 18-month-old girl with abdominal distention, which was noticed by her parents. An abdominal computed tomography scan showed a large multilocular cystic mass arising from the lower pole of the left kidney. A left total nephrectomy was performed. Immature blastemal elements without evidence of malignant cells were observed on histological analysis. Conclusion The authors report a case of an 18-month-old girl with CPDN managed by total nephrectomy. CPDN should be considered in the differential diagnosis of patients with cystic renal lesions. The authors would also like to affirm that partial or total nephrectomy should be done in all cases of CPDN and other cystic renal tumors.
Collapse
Affiliation(s)
| | - Albaraa H. Bara
- Department of Cardiothoracic Surgery, Damascus University Cardiovascular Surgical Center, Damascus
| | | | - Selman M. Berro
- Department of Pediatric Surgery, Children’s Hospital
- Peoples’ Friendship University of Russia: Rossijskij universitet druzby narodov, Moscow, Russia
| | - Mays G. Khalil
- Department of Otolaryngology, Al-Bassel Hospital, Tartus, Syrian Arab Rebublic
| |
Collapse
|
44
|
de Souza FKM, Fanelli MCA, Duarte AAB, Alves MTDS, Lederman HM, Cypriano MDS, Abib SDCV. Surgery in Bilateral Wilms Tumor-A Single-Center Experience. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1790. [PMID: 38002881 PMCID: PMC10670692 DOI: 10.3390/children10111790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023]
Abstract
The treatment of bilateral Wilms tumors (BWT) involves curing the cancer, preserving long-term renal function, and maintaining a good quality of life. Established methods for achieving these goals include preoperative chemotherapy and nephron-sparing surgery (NSS). This study aimed to evaluate the experience of a single institution in treating patients with BWT. We analyzed cases of BWT treated at the Pediatric Oncology Institute-GRAACC-Federal University of São Paulo over a period of 35 years. Bleeding control was performed with manual compression of the renal parenchyma. Thirty-three patients were included in the study. Thirty cases were synchronous tumors. The mean age at diagnosis was 30.4 months (±22 m) and 66.7% were girls. The median follow-up period was 83 months. Neoadjuvant chemotherapy was the primary approach in most patients (87.9%), with a simultaneous upfront surgical approach performed in 84.8%. Most patients underwent bilateral NSS (70.4%). There were no early complications in this series, but 39.4% had clinical complications. The five-year survival rate was 76%. Therefore, it is clear that the surgical approach to BWT plays a crucial role in achieving good outcomes. However, it is difficult to standardize surgical techniques and technology may have the potential to enhance safety.
Collapse
Affiliation(s)
- Fernanda Kelly Marques de Souza
- Department of Pediatric Surgery, Pediatric Oncology Institute, GRAACC, Federal University of São Paulo, São Paulo 04039-001, Brazil; (M.C.A.F.); (A.A.B.D.); (S.d.C.V.A.)
| | - Mayara Caroline Amorim Fanelli
- Department of Pediatric Surgery, Pediatric Oncology Institute, GRAACC, Federal University of São Paulo, São Paulo 04039-001, Brazil; (M.C.A.F.); (A.A.B.D.); (S.d.C.V.A.)
| | - Alexandre Alberto Barros Duarte
- Department of Pediatric Surgery, Pediatric Oncology Institute, GRAACC, Federal University of São Paulo, São Paulo 04039-001, Brazil; (M.C.A.F.); (A.A.B.D.); (S.d.C.V.A.)
- Department of Pediatric Surgery, Foundation Regional Faculty of Medicine of São José do Rio Preto, Children’s and Maternity Hospital, São José do Rio Preto 15091-240, Brazil
| | | | - Henrique Manoel Lederman
- Department of Radiology, Pediatric Oncology Institute, GRAACC, Federal University of São Paulo, São Paulo 04039-001, Brazil;
| | - Monica dos Santos Cypriano
- Department of Pediatric Oncology, Pediatric Oncology Institute, GRAACC, Federal University of São Paulo, São Paulo 04039-001, Brazil;
| | - Simone de Campos Vieira Abib
- Department of Pediatric Surgery, Pediatric Oncology Institute, GRAACC, Federal University of São Paulo, São Paulo 04039-001, Brazil; (M.C.A.F.); (A.A.B.D.); (S.d.C.V.A.)
| |
Collapse
|
45
|
Chen E, Hackney L, VanHeyst K, Miyasaka EA. Wilms Tumor in Child With Trisomy 18 and Horseshoe Kidney. J Pediatr Hematol Oncol 2023; 45:e1018-e1022. [PMID: 37749779 DOI: 10.1097/mph.0000000000002756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/16/2023] [Indexed: 09/27/2023]
Abstract
Trisomy 18 is associated with several congenital malformations, including horseshoe kidney. It can be full, partial, or mosaic, and mosaicism is often associated with lesser severity and longer life expectancy, placing patients at greater risk of developing neoplasms or malignancies. One common tumor among children with Trisomy 18 is Wilms tumor, which is also associated with renal congenital abnormalities such as horseshoe kidney. We present a case describing the occurrence of these three characteristics: development of Wilms tumor in a patient with Trisomy 18 and a horseshoe kidney and discuss treatment with regards to these conditions.
Collapse
Affiliation(s)
| | - Lisa Hackney
- Department of Pediatrics, Division of Pediatric Hematology Oncology
| | - Kristen VanHeyst
- Department of Pediatrics, Division of Pediatric Hematology Oncology
| | - Eiichi A Miyasaka
- Division of Pediatric Surgery, Rainbow Babies and Children's Hospital at University Hospitals Cleveland Medical Center, Cleveland, OH
| |
Collapse
|
46
|
Xiang B, Chen ML, Gao ZQ, Mi T, Shi QL, Dong JJ, Tian XM, Liu F, Wei GH. CCNB1 is a novel prognostic biomarker and promotes proliferation, migration and invasion in Wilms tumor. BMC Med Genomics 2023; 16:189. [PMID: 37592341 PMCID: PMC10433552 DOI: 10.1186/s12920-023-01627-3] [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: 03/03/2023] [Accepted: 08/05/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Wilms tumour (WT) is a mixed type of embryonal tumour that usually occurs in early childhood. However, our knowledge of the pathogenesis or progression mechanism of WT is inadequate, and there is a scarcity of beneficial therapeutic strategies. METHODS High-throughput RNA sequencing was employed in this study to identify differentially expressed genes (DEGs) in clinical tumor samples and matching normal tissues. The STRING database was utilized to build a protein-protein interaction (PPI) network, and the Cytohubba method was used to identify the top 10 highly related HUB genes. Then, the key genes were further screened by univariate COX survival analysis. Subsequently, the XCELL algorithm was used to evaluate the tumour immune infiltration. RT-PCR, WB, and IF were used to verify the expression level of key genes in clinical tissues and tumour cell lines. Finally, the function of the key gene was further verified by loss-of-function experiments. RESULTS We initially screened 1612 DEGs, of which 1030 were up-regulated and 582 were down-regulated. The GO and KEGG enrichment analysis suggested these genes were associated with 'cell cycle', 'DNA replication'. Subsequently, we identified 10 key HUB genes, among them CCNB1 was strongly related to WT patients' overall survival. Multiple survival analyses showed that CCNB1 was an independent indicator of WT prognosis. Thus, we constructed a nomogram of CCNB1 combined with other clinical indicators. Single gene GSEA and immune infiltration analysis revealed that CCNB1 was associated with the degree of infiltration or activation status of multiple immune cells. TIDE analysis indicated that this gene was correlated with multiple key immune checkpoint molecules and TIDE scores. Finally, we validated the differential expression level of CCNB1 in an external gene set, the pan-cancer, clinical samples, and cell lines. CCNB1 silencing significantly inhibited the proliferation, migration, and invasive capabilities of WIT-49 cells, also, promoted apoptosis, and in turn induced G2 phase cell cycle arrest in loss-of-function assays. CONCLUSION Our study suggests that CCNB1 is closely related to WT progression and prognosis, and serves as a potential target.
Collapse
Affiliation(s)
- Bin Xiang
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Mei-Lin Chen
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Zhi-Qiang Gao
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Tao Mi
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Qin-Lin Shi
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Jun-Jun Dong
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Xiao-Mao Tian
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China.
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China.
| | - Feng Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China.
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China.
| | - Guang-Hui Wei
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
47
|
Sarkany B, Kuthi L, Kovacs G. Novel concept of Wilms' tumor development: involvement of pluripotential cells of ureteric bud. Hum Pathol 2023; 138:34-40. [PMID: 37209922 DOI: 10.1016/j.humpath.2023.05.004] [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: 04/14/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
It is acknowledged that nephron develops after bilateral induction of the metanephric mesenchyma and branching ureteric bud (UB), and that nephrogenic rest and Wilms' tumor (nephroblastoma) arises from impaired differentiation of metanephric blastema. The aim of this study was to obtain more information on the involvement of UB derivatives in nephrogenic rest and Wilms' tumor. We applied immunohistochemistry to analyze nephrogenic rests and Wilms' tumors with mixed histology, including regressive and blastemal types. We used antibodies recognizing UB tip cells (ROBO1, SLIT2, RET), principal cells (AQP2), α- and β-intercalated cells (SLC26A4, SLC4A1, ATP6V1B1, ATP6V0D2), and their precursors (CA2). Tubules surrounded by tumorous blastemal cells resembling UB tip were positive for RET, ROBO1, and SLIT2 in Wilms' tumor. Moreover, CA2-positive tubular structures and ATP6V1B1- and ATP6V0D2-positive immature non-α- and non-β-intercalated cells were detected in nephrogenic rest and Wilms' tumor. We suggest that Wilms' tumor is more than nephroblastoma and propose a definition that Wilms tumor is a malignant embryonal neoplasm derived from pluripotential cells of nephrogenic blastema and of ureteric bud tip.
Collapse
Affiliation(s)
- Beatrix Sarkany
- Department of Urology, Medical School, University of Pecs, 7621, Pecs, Hungary
| | - Levente Kuthi
- Department of Pathology, University of Szeged, 6725, Szeged, Hungary
| | - Gyula Kovacs
- Department of Urology, Medical School, University of Pecs, 7621, Pecs, Hungary; Medical Faculty, Ruprecht-Karls-University, 69120, Heidelberg, Germany.
| |
Collapse
|
48
|
Roy P, van Peer SE, Dandis R, Duncan C, de Aguirre‐Neto JC, Verschuur A, de Camargo B, Karim‐Kos HE, Boschetti L, Spreafico F, Ramirez‐Villar GL, Graf N, van Tinteren H, Pritchard‐Jones K, van den Heuvel‐Eibrink MM. Impact of the COVID-19 pandemic on paediatric renal tumour presentation and management, a SIOP renal tumour study group study. Cancer Med 2023; 12:17098-17111. [PMID: 37496317 PMCID: PMC10501283 DOI: 10.1002/cam4.6358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/26/2023] [Accepted: 07/09/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND The COVID-19 pandemic had global catastrophic effects on the management of non-communicable diseases including paediatric cancers. Restrictions during the start of 2020 complicated timely referrals of patients to specialized centres. We aimed to evaluate the pandemic's impact on the number of new diagnoses, disease characteristics and management delay for paediatric renal tumour patients included in the SIOP-RTSG-UMBRELLA study, as compared with data from a historical SIOP-RTSG trial (2005-2009). METHODS The number of intensive care admissions, population mobility rates and national lockdown periods/restrictions were used as proxies of the pandemic's severity and impact on societies. Clinical and tumour data were extracted from the SIOP-RTSG-UMBRELLA study and from historical SIOP-RTSG trials. RESULTS During the first lockdown in Europe, the number of newly diagnosed patients decreased following restrictions and population immobilisation. Additionally, there was a higher proportion of advanced disease (37% vs. 17% before and after COVID-9, p < 0.001) and larger median tumour volume (559 cm3 vs. 328 and 434 cm3 before and after, p < 0.0001). Also in Brazil, the proportion of advanced disease was higher during the national decrease in mobilisation and start of restrictions (50% and 24% vs. 11% and 18% before and after, p < 0.01). Tumour volume in Brazil was also higher during the first months of COVID-19 (599 cm3 vs. 459 and 514 cm3 ), although not significant (p = 0.17). We did not observe any delays in referral time nor in time to start treatment, even though COVID-19 restrictions may have caused children to reach care later. CONCLUSION The COVID-19 pandemic briefly changed the tumour characteristics of children presenting with renal tumours. The longer-term impact on clinical outcomes will be kept under review.
Collapse
Affiliation(s)
- Prakriti Roy
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | - Rana Dandis
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | | | - Arnauld Verschuur
- Department of Paediatric Oncology & HaematologyLa Timone Children's HospitalMarseilleFrance
| | - Beatriz de Camargo
- Grupo Brasileiro de Tumores Renais (Brazilian Renal Tumor Group)São PauloBrazil
| | - Henrike E. Karim‐Kos
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Department of ResearchNetherlands Comprehensive Cancer Organisation (IKNL)UtrechtThe Netherlands
| | - Luna Boschetti
- Department of Medical Oncology and Hematology, Pediatric Oncology UnitFondazione IRCCS Istituto Nazionale dei Tumori di MilanMilanItaly
| | - Filippo Spreafico
- Department of Medical Oncology and Hematology, Pediatric Oncology UnitFondazione IRCCS Istituto Nazionale dei Tumori di MilanMilanItaly
| | | | - Norbert Graf
- Department of Paediatric Oncology & HaematologySaarland UniversityHomburgGermany
| | | | - Kathy Pritchard‐Jones
- UCL Great Ormond Street Institute of Child Health, University College LondonLondonUK
| | - Marry M. van den Heuvel‐Eibrink
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Division of Child Health, Wilhelmina Children's HospitalUniversity Medical Center UtrechtThe Netherlands
| |
Collapse
|
49
|
Meier CM, Fuchs J, von Schweinitz D, Stein R, Wagenpfeil S, Kager L, Schenk JP, Vokuhl C, Melchior P, Welter N, Furtwängler R, Graf N. Surgical Factors Influencing Local Relapse and Outcome in the Treatment of Unilateral Nephroblastoma. Ann Surg 2023; 278:e360-e367. [PMID: 36017935 DOI: 10.1097/sla.0000000000005690] [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: 11/25/2022]
Abstract
OBJECTIVE This study aims to identify factors associated with the occurrence of local relapse (LR) after treatment for unilateral nephroblastoma. BACKGROUND Despite the fact that LR is rare (~5%) its adverse impact on the need for relapse treatment and outcome (40%-80% overall survival) cannot be neglected. Identifying the causative factors may improve initial treatment to achieve better local control. METHODS Altogether 2386 patients with unilateral nephroblastoma prospectively enrolled over a period of 32 years (1989-2020) by the German Society for Pediatric Oncology and Hematology (SIOP-9/GPOH, SIOP-93-01/GPOH and SIOP-2001/GPOH) were retrospectively analyzed. Hazard ratios (HR) of LR were calculated for sex, age, size, local staging, histology, type of removal, rupture, lymph node (LN) removal using univariate and multivariate Cox models. RESULTS Age >48 months, tumor volume >500 mL, histology and LN extent of removal were identified as significant risk factors for LR [HR: 1.68, P =0.018, confidence interval (CI): 1.09-2.58; HR: 1.84, P =0.015, CI: 1.13-3.00; HR: 3.19, P <0.001, CI: 2.03-5.00; HR: 2.26, P =0.002, CI: 1.36-3.576]. LR occur significantly more often in Stage I and II, even if no LN are removed. The risk of metastases is significantly increased after local recurrence (HR: 11.5, P <0.001, CI: 7.11-18.60). LR is associated with a subsequent 18.79-fold increased risk of death (HR: 18.79, P <0.001, CI: 2.07-5.28). CONCLUSIONS Several factors are responsible for the occurrence of LR. Surgical ones, like LN sampling allow further reduction of LR and consequently a better outcome of patients with unilateral nephroblastoma.
Collapse
Affiliation(s)
- Clemens M Meier
- Department of General Surgery, Visceral, Vascular and Pediatric Surgery, Saarland University, Medical Center, Homburg, Germany
| | - Jörg Fuchs
- Department of Pediatric Surgery and Urology, University Hospital Tübingen, Tübingen, Germany
| | - Dietrich von Schweinitz
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital of the University of Munich, Munich, Germany
| | - Raimund Stein
- Center for Pediatric, Adolescent and Reconstructive Urology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan Wagenpfeil
- Institute for Medical Biometry, Epidemiology and Medical Informatics, Saarland University, Campus Homburg, Germany
| | - Leo Kager
- Department of Pediatrics, St Anna Children's Hospital, Medical University Vienna, Vienna, Austria
| | - Jens-Peter Schenk
- Pediatric Radiology Section, Department for Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Patrick Melchior
- Department of Radiation Oncology, Saarland University, Medical Center, Homburg, Germany
| | - Nils Welter
- Department of Pediatric Oncology and Hematology, Saarland University, Medical Center, Homburg, Germany
| | - Rhoikos Furtwängler
- Department of Pediatric Oncology and Hematology, Saarland University, Medical Center, Homburg, Germany
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Saarland University, Medical Center, Homburg, Germany
| |
Collapse
|
50
|
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
|