1
|
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
|
2
|
Walz AL, Maschietto M, Crompton B, Evageliou N, Dix D, Tytgat G, Gessler M, Gisselsson D, Daw NC, Wegert J. Tumor biology, biomarkers, and liquid biopsy in pediatric renal tumors. Pediatr Blood Cancer 2023; 70 Suppl 2:e30130. [PMID: 36592003 DOI: 10.1002/pbc.30130] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 01/03/2023]
Abstract
The expansion of knowledge regarding driver mutations for Wilms tumor (WT) and malignant rhabdoid tumor of the kidney (MRT) and various translocations for other pediatric renal tumors opens up new possibilities for diagnosis and treatment. In addition, there are growing data surrounding prognostic factors that can be used to stratify WT treatment to improve outcomes. Here, we review the molecular landscape of WT and other pediatric renal tumors as well as WT prognostic factors. We also review incorporation of circulating tumor DNA/liquid biopsies to leverage this molecular landscape, with potential use in the future for distinguishing renal tumors at the time of diagnosis and elucidating intratumor heterogeneity, which is not well evaluated with standard biopsies. Incorporation of liquid biopsies will require longitudinal collection of multiple biospecimens. Further preclinical research, identification and validation of biomarkers, molecular studies, and data sharing among investigators are crucial to inform therapeutic strategies that improve patient outcomes.
Collapse
Affiliation(s)
- Amy L Walz
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Mariana Maschietto
- Research Center, Boldrini Children's Hospital, Campinas, São Paulo, Brazil
| | - Brian Crompton
- Department of Pediatric Oncology, Dana-Farber/Harvard Cancer Center, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Nicholas Evageliou
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - David Dix
- British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| | - Godelieve Tytgat
- Princess Máxima Center for Pediatric Oncology, CS Utrecht, The Netherlands
| | - Manfred Gessler
- Comprehensive Cancer Center Mainfranken, Wuerzburg, Germany.,Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - David Gisselsson
- Cancer Cell Evolution Unit, Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Najat C Daw
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jenny Wegert
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
3
|
Differential expression profiling of onco and tumor-suppressor genes from major-signaling pathways in Wilms' tumor. Pediatr Surg Int 2022; 38:1601-1617. [PMID: 36107237 DOI: 10.1007/s00383-022-05202-2] [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] [Accepted: 08/22/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE Wilms' tumor is the most-frequent malignant-kidney tumor in children under 3-4 years of age and is caused by genetic alterations of oncogenes (OG) and tumor-suppressor genes (TG). Wilms' tumor has been linked to many OG-&-TG. However, only WT1 has a proven role in the development of this embryonic-tumor. METHODS The study investigates the level of mRNA expression of 16 OGs and 20 TGs involved in key-signaling pathways, including chromatin modification; RAS; APC; Cell Cycle/Apoptosis; Transcriptional Regulation; PI3K; NOTCH-&-HH; PI3K & RAS of 24-fresh Wilms'-tumor cases by capture-and-reporter probe Code-Sets chemistry, as CNVs in these pathway genes have been reported. RESULTS Upon extensively investigating, MEN1, MLL2, MLL3, PBRM1, PRDM1, SMARCB1, SETD2, WT1, PTPN11, KRAS, HRAS, NF1, APC, RB1, FUBP1, BCOR, U2AF1, PIK3CA, PTEN, EBXW7, SMO, ALK, CBL, EP300-and-GATA1 were found to be significantly up-regulated in 58.34, 62.5, 79.17, 91.67, 58, 66.66,54, 58.34, 66.67, 75, 62.5, 62.5, 58, 79.17, 79.17, 75, 70.84, 50, 50, 75, 66.66, 62.50, 61.66, 58.34-and-62.50% of cases respectively, whereas BRAF, NF2, CDH1, BCL2, FGFR3, ERBB2, MET, RET, EGFR-and-GATA2 were significantly down regulated in 58, 87.50, 79.16, 54.16, 79.17, 91.66, 66.66, 58.33, 91.66-and-62.50% of cases, respectively. Interestingly, the WT1 gene was five-fold down regulated in 41.66% of cases only. CONCLUSION Hence, extensive profiling of OGs and TGs association of major-signaling pathways in Wilms' tumor cases may aid in disease diagnosis. PBRM1 (up-regulated in 91.67% of cases), ERBB2 and EGFR (down-regulated in 91.66 and 91.66% of cases, respectively) could be marker genes. However, validation of all relevant results in a larger number of samples is required.
Collapse
|
4
|
How Genetics and Genomics Advances Are Rewriting Pediatric Cancer Research and Clinical Care. Medicina (B Aires) 2022; 58:medicina58101386. [PMID: 36295546 PMCID: PMC9610804 DOI: 10.3390/medicina58101386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
In the last two decades, thanks to the data that have been obtained from the Human Genome Project and the development of next-generation sequencing (NGS) technologies, research in oncology has produced extremely important results in understanding the genomic landscape of pediatric cancers, which are the main cause of death during childhood. NGS has provided significant advances in medicine by detecting germline and somatic driver variants that determine the development and progression of many types of cancers, allowing a distinction between hereditary and non-hereditary cancers, characterizing resistance mechanisms that are also related to alterations of the epigenetic apparatus, and quantifying the mutational burden of tumor cells. A combined approach of next-generation technologies allows us to investigate the numerous molecular features of the cancer cell and the effects of the environment on it, discovering and following the path of personalized therapy to defeat an "ancient" disease that has had victories and defeats. In this paper, we provide an overview of the results that have been obtained in the last decade from genomic studies that were carried out on pediatric cancer and their contribution to the more accurate and faster diagnosis in the stratification of patients and the development of new precision therapies.
Collapse
|
5
|
Theilen TM, Braun Y, Bochennek K, Rolle U, Fiegel HC, Friedmacher F. Multidisciplinary Treatment Strategies for Wilms Tumor: Recent Advances, Technical Innovations and Future Directions. Front Pediatr 2022; 10:852185. [PMID: 35911825 PMCID: PMC9333359 DOI: 10.3389/fped.2022.852185] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Significant progress has been made in the management of Wilms tumor (WT) in recent years, mostly as a result of collaborative efforts and the implementation of protocol-driven, multimodal therapy. This article offers a comprehensive overview of current multidisciplinary treatment strategies for WT, whilst also addressing recent technical innovations including nephron-sparing surgery (NSS) and minimally invasive approaches. In addition, surgical concepts for the treatment of metastatic disease, advances in tumor imaging technology and potentially prognostic biomarkers will be discussed. Current evidence suggests that, in experienced hands and selected cases, laparoscopic radical nephrectomy and laparoscopic-assisted partial nephrectomy for WT may offer the same outcome as the traditional open approach. While NSS is the standard procedure for bilateral WT, NSS has evolved as an alternative technique in patients with smaller unilateral WT and in cases with imminent renal failure. Metastatic disease of the lung or liver that is associated with WT is preferably treated with a three-drug chemotherapy and local radiation therapy. However, surgical sampling of lung nodules may be advisable in persistent nodules before whole lung irradiation is commenced. Several tumor markers such as loss of heterozygosity of chromosomes 1p/16q, 11p15 and gain of function at 1q are associated with an increased risk of recurrence or a decreased risk of overall survival in patients with WT. In summary, complete resection with tumor-free margins remains the primary surgical aim in WT, while NSS and minimally invasive approaches are only suitable in a subset of patients with smaller WT and low-risk disease. In the future, advances in tumor imaging technology may assist the surgeon in defining surgical resection margins and additional biomarkers may emerge as targets for development of new diagnostic tests and potential therapies.
Collapse
Affiliation(s)
- Till-Martin Theilen
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Yannick Braun
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Konrad Bochennek
- Division of Pediatric Hematology and Pediatric Oncology, Hospital for Children and Adolescents, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Udo Rolle
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Henning C. Fiegel
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Florian Friedmacher
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
6
|
Molecular Markers of Pediatric Solid Tumors—Diagnosis, Optimizing Treatments, and Determining Susceptibility: Current State and Future Directions. Cells 2022; 11:cells11071238. [PMID: 35406801 PMCID: PMC8997439 DOI: 10.3390/cells11071238] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Advances in molecular technologies, from genomics and transcriptomics to epigenetics, are providing unprecedented insight into the molecular landscape of pediatric tumors. Multi-omics approaches provide an opportunity to identify a wide spectrum of molecular alterations that account for the initiation of the neoplastic process in children, response to treatment and disease progression. The detection of molecular markers is crucial to assist clinicians in accurate tumor diagnosis, risk stratification, disease subtyping, prediction of treatment response, and surveillance, allowing also for personalized cancer management. This review summarizes the most recent developments in genomics research and their relevance to the field of pediatric oncology with the aim of generating an overview of the most important, from the clinical perspective, molecular markers for pediatric solid tumors. We present an overview of the molecular markers selected based on therapeutic protocols, guidelines from international committees and scientific societies, and published data.
Collapse
|
7
|
Spreafico F, Fernandez CV, Brok J, Nakata K, Vujanic G, Geller JI, Gessler M, Maschietto M, Behjati S, Polanco A, Paintsil V, Luna-Fineman S, Pritchard-Jones K. Wilms tumour. Nat Rev Dis Primers 2021; 7:75. [PMID: 34650095 DOI: 10.1038/s41572-021-00308-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2021] [Indexed: 02/08/2023]
Abstract
Wilms tumour (WT) is a childhood embryonal tumour that is paradigmatic of the intersection between disrupted organogenesis and tumorigenesis. Many WT genes play a critical (non-redundant) role in early nephrogenesis. Improving patient outcomes requires advances in understanding and targeting of the multiple genes and cellular control pathways now identified as active in WT development. Decades of clinical and basic research have helped to gradually optimize clinical care. Curative therapy is achievable in 90% of affected children, even those with disseminated disease, yet survival disparities within and between countries exist and deserve commitment to change. Updated epidemiological studies have also provided novel insights into global incidence variations. Introduction of biology-driven approaches to risk stratification and new drug development has been slower in WT than in other childhood tumours. Current prognostic classification for children with WT is grounded in clinical and pathological findings and in dedicated protocols on molecular alterations. Treatment includes conventional cytotoxic chemotherapy and surgery, and radiation therapy in some cases. Advanced imaging to capture tumour composition, optimizing irradiation techniques to reduce target volumes, and evaluation of newer surgical procedures are key areas for future research.
Collapse
Affiliation(s)
- Filippo Spreafico
- Department of Medical Oncology and Hematology, Paediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Conrad V Fernandez
- Department of Paediatrics, IWK Health, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jesper Brok
- Department of Paediatric Haematology and Oncology, Rigshospitalet, Copenhagen, Denmark
| | - Kayo Nakata
- Cancer Control Center, Osaka International Cancer Institute, Osaka, Japan
| | | | - James I Geller
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Cincinnati, OH, USA
| | - Manfred Gessler
- Theodor-Boveri-Institute, Developmental Biochemistry, and Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Wuerzburg, Germany
| | - Mariana Maschietto
- Research Center, Boldrini Children's Hospital, Genetics and Molecular Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Angela Polanco
- National Cancer Research Institute Children's Group Consumer Representative, London, UK
| | - Vivian Paintsil
- Department of Child Health, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Sandra Luna-Fineman
- Division of Hematology, Oncology and Bone Marrow Transplantation, Department of Paediatrics, University of Colorado, Aurora, CO, USA
| | - Kathy Pritchard-Jones
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
8
|
Quarello P, Perotti D, Carli D, Giorgio E, Sirchia F, Brusco A, Ferrero GB, Mussa A, Spadea M, Ciceri S, Spreafico F, Fagioli F. Wilms tumour occurring in a patient with osteopathia striata with cranial sclerosis: A still unsolved biological question. Pediatr Blood Cancer 2021; 68:e29132. [PMID: 34028980 DOI: 10.1002/pbc.29132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 11/10/2022]
Affiliation(s)
- Paola Quarello
- Paediatric Onco-Haematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Turin, Italy.,Department of Public Health and Paediatric Sciences, University of Torino, Turin, Italy
| | - Daniela Perotti
- Molecular Bases of Genetic Risk and Genetic Testing Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Diana Carli
- Paediatric Onco-Haematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Turin, Italy.,Department of Public Health and Paediatric Sciences, University of Torino, Turin, Italy
| | - Elisa Giorgio
- Department of Medical Sciences, University of Torino, Turin, Italy.,Department of Molecular Medicine, University of Pavia, Pavia, Italy.,IRCCS Mondino Foundation, Pavia, Italy
| | - Fabio Sirchia
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,IRCCS Mondino Foundation, Pavia, Italy.,IRCCS Materno Infantile 'Burlo Garofolo', Trieste, Italy
| | - Alfredo Brusco
- Department of Medical Sciences, University of Torino, Turin, Italy.,Medical Genetics Unit, 'Città della Salute e della Scienza' Hospital, Turin, Italy
| | - Giovanni Battista Ferrero
- Department of Clinical and Biological Sciences, School of Medicine, University of Torino, Orbassano, Turin, Italy
| | - Alessandro Mussa
- Department of Public Health and Paediatric Sciences, University of Torino, Turin, Italy.,Pediatric Clinical Genetics Unit, Regina Margherita Children's Hospital, Turin, Italy
| | - Manuela Spadea
- Paediatric Onco-Haematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Turin, Italy
| | - Sara Ciceri
- Molecular Bases of Genetic Risk and Genetic Testing Unit, Department of Research, 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
| | - Franca Fagioli
- Paediatric Onco-Haematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Turin, Italy.,Department of Public Health and Paediatric Sciences, University of Torino, Turin, Italy
| |
Collapse
|
9
|
de Sá Pereira BM, Azevedo RMD, Aguirre Neto JCD, Menezes CF, Rodrigues KE, Faria PA, Camargo BD, Maschietto M. Intra-tumor genetic heterogeneity in Wilms tumor samples. ACTA ACUST UNITED AC 2020; 65:1496-1501. [PMID: 31994632 DOI: 10.1590/1806-9282.65.12.1496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022]
Abstract
Childhood renal tumors account for ~7% of all childhood cancers, and most cases are embryonic Wilms' tumors (WT). Children with WT are usually treated by either COG or SIOP. The later treats the children using preoperative chemotherapy, but both have around 90% of overall survival in five years. WT is a genetically heterogeneous group with a low prevalence of known somatic alterations. Only around 30% of the cases present mutation in known genes, and there is a relatively high degree of intra-tumor genetic heterogeneity (ITGH). Besides potentially having an impact on the clinical outcome of patients, ITGH may interfere with the search for molecular markers that are prospectively being tested by COG and SIOP. In this review, we present the proposal of the current UMBRELLA SIOP Study 2017/Brazilian Renal Tumor Group that requires the multi-sampling collection of each tumor to better evaluate possible molecular markers, as well as to understand WT biology.
Collapse
Affiliation(s)
- Bruna M de Sá Pereira
- . Centro de Pesquisas (CPQ), Instituto Nacional de Câncer (Inca), Rio de Janeiro, RJ, Brasil
| | | | | | | | | | - Paulo A Faria
- . Divisão de Patologia (Dipat), Instituto Nacional de Câncer (Inca), Rio de Janeiro, RJ, Brasil
| | - Beatriz de Camargo
- . Centro de Pesquisas (CPQ), Instituto Nacional de Câncer (Inca), Rio de Janeiro, RJ, Brasil
| | - Mariana Maschietto
- . Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP, Brasil
| |
Collapse
|
10
|
Chong WC, Cain JE. Lessons learned from the developmental origins of childhood renal cancer. Anat Rec (Hoboken) 2019; 303:2561-2577. [DOI: 10.1002/ar.24315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 08/14/2019] [Accepted: 10/05/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Wai Chin Chong
- Centre for Cancer ResearchHudson Institute of Medical Research Clayton Victoria Australia
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health SciencesMonash University Clayton Victoria Australia
| | - Jason E. Cain
- Centre for Cancer ResearchHudson Institute of Medical Research Clayton Victoria Australia
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health SciencesMonash University Clayton Victoria Australia
| |
Collapse
|
11
|
Große A, Perner B, Naumann U, Englert C. Zebrafish Wtx is a negative regulator of Wnt signaling but is dispensable for embryonic development and organ homeostasis. Dev Dyn 2019; 248:866-881. [PMID: 31290212 DOI: 10.1002/dvdy.84] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 06/20/2019] [Accepted: 06/28/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The X-chromosomally linked gene WTX is a human disease gene and a member of the AMER family. Mutations in WTX are found in Wilms tumor, a form of pediatric kidney cancer and in patients suffering from OSCS (Osteopathia striata with cranial sclerosis), a sclerosing bone disorder. Functional data suggest WTX to be an inhibitor of the Wnt/β-catenin signaling pathway. Deletion of Wtx in mouse leads to perinatal death, impeding the analysis of its physiological role. RESULTS To gain insights into the function of Wtx in development and homeostasis we have used zebrafish as a model and performed both knockdown and knockout studies using morpholinos and transcription activator-like effector nucleases (TALENs), respectively. Wtx knockdown led to increased Wnt activity and embryonic dorsalization. Also, wtx mutants showed a transient upregulation of Wnt target genes in the context of caudal fin regeneration. Surprisingly, however, wtx as well as wtx/amer2/amer3 triple mutants developed normally, were fertile and did not show any anomalies in organ maintenance. CONCLUSIONS Our data show that members of the zebrafish wtx/amer gene family, while sharing a partially overlapping expression pattern do not compensate for each other. This observation demonstrates a remarkable robustness during development and regeneration in zebrafish.
Collapse
Affiliation(s)
- Andreas Große
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Birgit Perner
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Uta Naumann
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Christoph Englert
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany.,Institute of Biochemistry and Biophysics, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
12
|
Abstract
Wilms tumour is the most common renal malignancy of childhood. The disease is curable in the majority of cases, albeit at considerable cost in terms of late treatment-related effects in some children. However, one in ten children with Wilms tumour will die of their disease despite modern treatment approaches. The genetic changes that underpin Wilms tumour have been defined by studies of familial cases and by unbiased DNA sequencing of tumour genomes. Together, these approaches have defined the landscape of cancer genes that are operative in Wilms tumour, many of which are intricately linked to the control of fetal nephrogenesis. Advances in our understanding of the germline and somatic genetic changes that underlie Wilms tumour may translate into better patient outcomes. Improvements in risk stratification have already been seen through the introduction of molecular biomarkers into clinical practice. A host of additional biomarkers are due to undergo clinical validation. Identifying actionable mutations has led to potential new targets, with some novel compounds undergoing testing in early phase trials. Avenues that warrant further exploration include targeting Wilms tumour cancer genes with a non-redundant role in nephrogenesis and targeting the fetal renal transcriptome.
Collapse
Affiliation(s)
- Taryn Dora Treger
- Wellcome Sanger Institute, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Tanzina Chowdhury
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Kathy Pritchard-Jones
- UCL Great Ormond Street Institute of Child Health, London, UK.
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| | - Sam Behjati
- Wellcome Sanger Institute, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| |
Collapse
|
13
|
Zhu GF, Xu YW, Li J, Niu HL, Ma WX, Xu J, Zhou PR, Liu X, Ye DL, Liu XR, Yan T, Zhai WK, Xu ZJ, Liu C, Wang L, Wang H, Luo JM, Liu L, Li XQ, Guo S, Jiang HP, Shen P, Lin HK, Yu DH, Ding YQ, Zhang QL. Mir20a/106a-WTX axis regulates RhoGDIa/CDC42 signaling and colon cancer progression. Nat Commun 2019; 10:112. [PMID: 30631060 PMCID: PMC6328557 DOI: 10.1038/s41467-018-07998-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/12/2018] [Indexed: 02/07/2023] Open
Abstract
Wilms tumor gene on the X chromosome (WTX) is a putative tumor suppressor gene in Wilms tumor, but its expression and functions in other tumors are unclear. Colorectal cancer (CRC) is the third leading cause of cancer-related deaths in women and the second leading cause in men in the United States. We demonstrated that WTX frequently lost in CRC which was highly correlated with cell proliferation, tumor invasion and metastasis. Mechanistically, WTX loss disrupts the interaction between RhoGDIα and CDC42 by losing of the binding with RhoGDIα and triggers the activation of CDC42 and its downstream cascades, which promotes CRC development and liver metastasis. The aberrant upregulation of miR-20a/miR-106a were identified as the reason of WTX loss in CRC both in vivo and in vitro. These study defined the mechanism how miR-20a/miR-106a-mediated WTX loss regulates CRC progression and metastasis, and provided a potential therapeutic target for preventing CRC progression.
Collapse
Affiliation(s)
- Gui-Fang Zhu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Key Laboratory of Molecular Tumor Pathology of Guangdong Province, Guangzhou, GuangDong, 510515, China
| | - Yang-Wei Xu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Key Laboratory of Molecular Tumor Pathology of Guangdong Province, Guangzhou, GuangDong, 510515, China
| | - Jian Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Key Laboratory of Molecular Tumor Pathology of Guangdong Province, Guangzhou, GuangDong, 510515, China
| | - Hui-Lin Niu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Key Laboratory of Molecular Tumor Pathology of Guangdong Province, Guangzhou, GuangDong, 510515, China
| | - Wen-Xia Ma
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Key Laboratory of Molecular Tumor Pathology of Guangdong Province, Guangzhou, GuangDong, 510515, China
| | - Jia Xu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Pei-Rong Zhou
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Nanfang Hospital/First clinical Medical School, Southern Medical University, Guangzhou, GuangDong, 510515, China
| | - Xia Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Key Laboratory of Molecular Tumor Pathology of Guangdong Province, Guangzhou, GuangDong, 510515, China
| | - Dan-Li Ye
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Key Laboratory of Molecular Tumor Pathology of Guangdong Province, Guangzhou, GuangDong, 510515, China
| | - Xiao-Rong Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Key Laboratory of Molecular Tumor Pathology of Guangdong Province, Guangzhou, GuangDong, 510515, China
| | - Tao Yan
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Nanfang Hospital/First clinical Medical School, Southern Medical University, Guangzhou, GuangDong, 510515, China
| | - Wei-Ke Zhai
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Key Laboratory of Molecular Tumor Pathology of Guangdong Province, Guangzhou, GuangDong, 510515, China
| | - Zhi-Jun Xu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Nanfang Hospital/First clinical Medical School, Southern Medical University, Guangzhou, GuangDong, 510515, China
| | - Chun Liu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Nanfang Hospital/First clinical Medical School, Southern Medical University, Guangzhou, GuangDong, 510515, China
| | - Lei Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
| | - Hao Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Nanfang Hospital/First clinical Medical School, Southern Medical University, Guangzhou, GuangDong, 510515, China
| | - Jia-Mao Luo
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
- Nanfang Hospital/First clinical Medical School, Southern Medical University, Guangzhou, GuangDong, 510515, China
| | - Li Liu
- Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China
| | - Xuan-Qi Li
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China
| | - Suiqun Guo
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, GuangDong, 510630, China
| | - Hui-Ping Jiang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, GuangDong, 510630, China
| | - Peng Shen
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China
| | - Hui-Kuan Lin
- Cancer Biology Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Di-Hua Yu
- Department of Molecular & Cellular Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yan-Qing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China.
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China.
- Key Laboratory of Molecular Tumor Pathology of Guangdong Province, Guangzhou, GuangDong, 510515, China.
| | - Qing-Ling Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, GuangDong, 510515, China.
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, GuangDong, 510515, China.
- Key Laboratory of Molecular Tumor Pathology of Guangdong Province, Guangzhou, GuangDong, 510515, China.
| |
Collapse
|
14
|
Halliday BJ, Fukuzawa R, Markie DM, Grundy RG, Ludgate JL, Black MA, Skeen JE, Weeks RJ, Catchpoole DR, Roberts AGK, Reeve AE, Morison IM. Germline mutations and somatic inactivation of TRIM28 in Wilms tumour. PLoS Genet 2018; 14:e1007399. [PMID: 29912901 PMCID: PMC6005459 DOI: 10.1371/journal.pgen.1007399] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 05/08/2018] [Indexed: 12/21/2022] Open
Abstract
Wilms tumour is a childhood tumour that arises as a consequence of somatic and rare germline mutations, the characterisation of which has refined our understanding of nephrogenesis and carcinogenesis. Here we report that germline loss of function mutations in TRIM28 predispose children to Wilms tumour. Loss of function of this transcriptional co-repressor, which has a role in nephrogenesis, has not previously been associated with cancer. Inactivation of TRIM28, either germline or somatic, occurred through inactivating mutations, loss of heterozygosity or epigenetic silencing. TRIM28-mutated tumours had a monomorphic epithelial histology that is uncommon for Wilms tumour. Critically, these tumours were negative for TRIM28 immunohistochemical staining whereas the epithelial component in normal tissue and other Wilms tumours stained positively. These data, together with a characteristic gene expression profile, suggest that inactivation of TRIM28 provides the molecular basis for defining a previously described subtype of Wilms tumour, that has early age of onset and excellent prognosis. The germline and somatic molecular events associated with Wilms tumour, a childhood kidney cancer, have been progressively defined over the past three decades. Among the uncharacterised tumours are a group of tumours that have monomorphic epithelial histology, familial association, distinctively clustered gene-expression patterns, early age of diagnosis, and excellent prognosis. Here, we describe germline mutations and loss of function of TRIM28 in familial Wilms tumours, along with somatic loss of function in a non-familial Wilms tumour. All TRIM28-mutant tumours showed the rare monomorphic epithelial histology, suggesting that loss of TRIM28 expression could be a useful marker to define a group of tumours with excellent prognosis. Future studies could lead to identification and reassurance of families that carry TRIM28 mutations, and to the use of reduced intensity of treatment for children who develop TRIM28-null tumours.
Collapse
Affiliation(s)
- Benjamin J. Halliday
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Ryuji Fukuzawa
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Department of Pathology, International University of Health and Welfare, School of Medicine, Narita, Japan
| | - David M. Markie
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Richard G. Grundy
- Children’s Brain Tumour Research Centre, University of Nottingham, Nottingham, United Kingdom
| | - Jackie L. Ludgate
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Michael A. Black
- Cancer Genetics Laboratory, Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | | | - Robert J. Weeks
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Daniel R. Catchpoole
- Tumour Bank, Children’s Cancer Research Unit, The Children’s Hospital at Westmead, Westmead, NSW, Australia
| | - Aedan G. K. Roberts
- Tumour Bank, Children’s Cancer Research Unit, The Children’s Hospital at Westmead, Westmead, NSW, Australia
| | - Anthony E. Reeve
- Cancer Genetics Laboratory, Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Ian M. Morison
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- * E-mail:
| |
Collapse
|
15
|
Boutet A, Comai G, Charlet A, Jian Motamedi F, Dhib H, Bandiera R, Schedl A. A knock-in mouse line conditionally expressing the tumor suppressor WTX/AMER1. Genesis 2017; 55. [PMID: 28960679 DOI: 10.1002/dvg.23074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/13/2017] [Accepted: 09/23/2017] [Indexed: 01/02/2023]
Abstract
WTX/AMER1 is an important developmental regulator, mutations in which have been identified in a proportion of patients suffering from the renal neoplasm Wilms' tumor and in the bone malformation syndrome Osteopathia Striata with Cranial Sclerosis (OSCS). Its cellular functions appear complex and the protein can be found at the membrane, within the cytoplasm and the nucleus. To understand its developmental and cellular function an allelic series for Wtx in the mouse is crucial. Whereas mice carrying a conditional knock out allele for Wtx have been previously reported, a gain-of-function mouse model that would allow studying the molecular, cellular and developmental role of Wtx is still missing. Here we describe the generation of a novel mouse strain that permits the conditional activation of WTX expression. Wtx fused to GFP was introduced downstream a stop cassette flanked by loxP sites into the Rosa26 locus by gene targeting. Ectopic WTX expression is reported after crosses with several Cre transgenic mice in different embryonic tissues. Further, functionality of the fusion protein was demonstrated in the context of a Wtx null allele.
Collapse
Affiliation(s)
- Agnès Boutet
- Université Côte d'Azur, Inserm U1091, CNRS UMR 7277, iBV, France
| | - Glenda Comai
- Université Côte d'Azur, Inserm U1091, CNRS UMR 7277, iBV, France
| | - Aurélie Charlet
- Université Côte d'Azur, Inserm U1091, CNRS UMR 7277, iBV, France
| | | | - Haroun Dhib
- Université Côte d'Azur, Inserm U1091, CNRS UMR 7277, iBV, France
| | - Roberto Bandiera
- Université Côte d'Azur, Inserm U1091, CNRS UMR 7277, iBV, France
| | - Andreas Schedl
- Université Côte d'Azur, Inserm U1091, CNRS UMR 7277, iBV, France
| |
Collapse
|
16
|
Wegert J, Vokuhl C, Ziegler B, Ernestus K, Leuschner I, Furtwängler R, Graf N, Gessler M. TP53 alterations in Wilms tumour represent progression events with strong intratumour heterogeneity that are closely linked but not limited to anaplasia. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2017; 3:234-248. [PMID: 29085664 PMCID: PMC5653929 DOI: 10.1002/cjp2.77] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/20/2017] [Accepted: 06/24/2017] [Indexed: 12/20/2022]
Abstract
TP53 mutations have been associated with anaplasia in Wilms tumour, which conveys a high risk for relapse and fatal outcome. Nevertheless, TP53 alterations have been reported in no more than 60% of anaplastic tumours, and recent data have suggested their presence in tumours that do not fulfil the criteria for anaplasia, questioning the clinical utility of TP53 analysis. Therefore, we characterized the TP53 status in 84 fatal cases of Wilms tumour, irrespective of histological subtype. We identified TP53 alterations in at least 90% of fatal cases of anaplastic Wilms tumour, and even more when diffuse anaplasia was present, indicating a very strong if not absolute coupling between anaplasia and deregulation of p53 function. Unfortunately, TP53 mutations do not provide additional predictive value in anaplastic tumours since the same mutation rate was found in a cohort of non-fatal anaplastic tumours. When classified according to tumour stage, patients with stage I diffuse anaplastic tumours still had a high chance of survival (87%), but this rate dropped to 26% for stages II-IV. Thus, volume of anaplasia or possible spread may turn out to be critical parameters. Importantly, among non-anaplastic fatal tumours, 26% had TP53 alterations, indicating that TP53 screening may identify additional cases at risk. Several of these non-anaplastic tumours fulfilled some criteria for anaplasia, for example nuclear unrest, suggesting that such partial phenotypes should be under special scrutiny to enhance detection of high-risk tumours via TP53 screening. A major drawback is that these alterations are secondary changes that occur only later in tumour development, leading to striking intratumour heterogeneity that requires multiple biopsies and analysis guided by histological criteria. In conclusion, we found a very close correlation between histological signs of anaplasia and TP53 alterations. The latter may precede development of anaplasia and thereby provide diagnostic value pointing towards aggressive disease.
Collapse
Affiliation(s)
- Jenny Wegert
- Theodor-Boveri-Institute/Biocenter, Developmental BiochemistryWuerzburg UniversityWuerzburgGermany
| | - Christian Vokuhl
- Department of Pathology, Kiel Paediatric Cancer RegistryChristian Albrechts UniversityKielGermany
| | - Barbara Ziegler
- Theodor-Boveri-Institute/Biocenter, Developmental BiochemistryWuerzburg UniversityWuerzburgGermany
| | - Karen Ernestus
- Institute for PathologyWuerzburg UniversityWuerzburgGermany
| | - Ivo Leuschner
- Department of Pathology, Kiel Paediatric Cancer RegistryChristian Albrechts UniversityKielGermany
| | - Rhoikos Furtwängler
- Department of Pediatric Oncology and HematologySaarland University HospitalHomburgGermany
| | - Norbert Graf
- Department of Pediatric Oncology and HematologySaarland University HospitalHomburgGermany
| | - Manfred Gessler
- Theodor-Boveri-Institute/Biocenter, Developmental BiochemistryWuerzburg UniversityWuerzburgGermany.,Comprehensive Cancer Center MainfrankenWuerzburg UniversityWuerzburgGermany
| |
Collapse
|
17
|
Bahrami A, Joodi M, Maftooh M, Ferns GA, M. Ahmadi M, Hassanian SM, Avan A. The genetic factors contributing to the development of Wilm's tumor and their clinical utility in its diagnosis and prognosis. J Cell Physiol 2017; 233:2882-2888. [DOI: 10.1002/jcp.26021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/19/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Afsane Bahrami
- Department of Modern Sciences and Technologies; Faculty of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
- Student Research Committee; Faculty of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Marjan Joodi
- Department of Pediatric Surgery; Faculty of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
- Endoscopic and Minimally Invasive Surgery Research Center; Sarvar Children's Hospital; Mashhad Iran
| | - Mina Maftooh
- Metabolic Syndrome Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| | - Gordon A. Ferns
- Division of Medical Education, Falmer, Brighton; Brighton and Sussex Medical School; Sussex UK
| | - Mehrdad M. Ahmadi
- Metabolic Syndrome Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| | - Seyed M. Hassanian
- Metabolic Syndrome Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| | - Amir Avan
- Metabolic Syndrome Research Center; Mashhad University of Medical Sciences; Mashhad Iran
- Cancer Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
18
|
Brok J, Treger TD, Gooskens SL, van den Heuvel-Eibrink MM, Pritchard-Jones K. Biology and treatment of renal tumours in childhood. Eur J Cancer 2016; 68:179-195. [PMID: 27969569 DOI: 10.1016/j.ejca.2016.09.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/25/2016] [Accepted: 09/01/2016] [Indexed: 02/08/2023]
Abstract
In Europe, almost 1000 children are diagnosed with a malignant renal tumour each year. The vast majority of cases are nephroblastoma, also known as Wilms' tumour (WT). Most children are treated according to Société Internationale d'Oncologie Pédiatrique Renal Tumour Study Group (SIOP-RTSG) protocols with pre-operative chemotherapy, surgery, and post-operative treatment dependent on stage and histology. Overall survival approaches 90%, but a subgroup of WT, with high-risk histology and/or relapsed disease, still have a much poorer prognosis. Outcome is similarly poor for the rare non-WT, particularly for malignant rhabdoid tumour of the kidney, metastatic clear cell sarcoma of the kidney (CCSK), and metastatic renal cell carcinoma (RCC). Improving outcome and long-term quality of life requires more accurate risk stratification through biological insights. Biomarkers are also needed to signpost potential targeted therapies for high-risk subgroups. Our understanding of Wilms' tumourigenesis is evolving and several signalling pathways, microRNA processing and epigenetics are now known to play pivotal roles. Most rhabdoid tumours display somatic and/or germline mutations in the SMARCB1 gene, whereas CCSK and paediatric RCC reveal a more varied genetic basis, including characteristic translocations. Conducting early-phase trials of targeted therapies is challenging due to the scarcity of patients with refractory or relapsed disease, the rapid progression of relapse and the genetic heterogeneity of the tumours with a low prevalence of individual somatic mutations. A further consideration in improving population survival rates is the geographical variation in outcomes across Europe. This review provides a comprehensive overview of the current biological knowledge of childhood renal tumours alongside the progress achieved through international collaboration. Ongoing collaboration is needed to ensure consistency of outcomes through standardised diagnostics and treatment and incorporation of biomarker research. Together, these objectives constitute the rationale for the forthcoming SIOP-RTSG 'UMBRELLA' study.
Collapse
Affiliation(s)
- Jesper Brok
- Cancer Section, University College London, Institute of Child Health, UK; Department of Paediatric Haematology and Oncology, Rigshospitalet, Copenhagen University Hospital, Denmark.
| | - Taryn D Treger
- Cancer Section, University College London, Institute of Child Health, UK
| | - Saskia L Gooskens
- Department of Paediatric Oncology, Princess Máxima Center for Pediatric Oncology and University of Utrecht, The Netherlands; Department of Paediatric Haematology and Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Marry M van den Heuvel-Eibrink
- Department of Paediatric Oncology, Princess Máxima Center for Pediatric Oncology and University of Utrecht, The Netherlands
| | | |
Collapse
|
19
|
Cresswell GD, Apps JR, Chagtai T, Mifsud B, Bentley CC, Maschietto M, Popov SD, Weeks ME, Olsen ØE, Sebire NJ, Pritchard-Jones K, Luscombe NM, Williams RD, Mifsud W. Intra-Tumor Genetic Heterogeneity in Wilms Tumor: Clonal Evolution and Clinical Implications. EBioMedicine 2016; 9:120-129. [PMID: 27333041 PMCID: PMC4972528 DOI: 10.1016/j.ebiom.2016.05.029] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/23/2016] [Accepted: 05/25/2016] [Indexed: 11/30/2022] Open
Abstract
The evolution of pediatric solid tumors is poorly understood. There is conflicting evidence of intra-tumor genetic homogeneity vs. heterogeneity (ITGH) in a small number of studies in pediatric solid tumors. A number of copy number aberrations (CNA) are proposed as prognostic biomarkers to stratify patients, for example 1q+ in Wilms tumor (WT); current clinical trials use only one sample per tumor to profile this genetic biomarker. We multisampled 20 WT cases and assessed genome-wide allele-specific CNA and loss of heterozygosity, and inferred tumor evolution, using Illumina CytoSNP12v2.1 arrays, a custom analysis pipeline, and the MEDICC algorithm. We found remarkable diversity of ITGH and evolutionary trajectories in WT. 1q+ is heterogeneous in the majority of tumors with this change, with variable evolutionary timing. We estimate that at least three samples per tumor are needed to detect >95% of cases with 1q+. In contrast, somatic 11p15 LOH is uniformly an early event in WT development. We find evidence of two separate tumor origins in unilateral disease with divergent histology, and in bilateral WT. We also show subclonal changes related to differential response to chemotherapy. Rational trial design to include biomarkers in risk stratification requires tumor multisampling and reliable delineation of ITGH and tumor evolution.
Collapse
Affiliation(s)
| | - John R Apps
- UCL Institute of Child Health, London, United Kingdom; Department of Paediatric Haematology and Oncology, Great Ormond Street Hospital, London, United Kingdom
| | | | | | - Christopher C Bentley
- The Francis Crick Institute, London, United Kingdom; UCL Genetics Institute, Department of Genetics, Evolution & Environment, University College London, United Kingdom
| | | | - Sergey D Popov
- Divisions of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, London, United Kingdom
| | - Mark E Weeks
- UCL Institute of Child Health, London, United Kingdom
| | - Øystein E Olsen
- Department of Radiology, Great Ormond Street Hospital, London, United Kingdom
| | - Neil J Sebire
- UCL Institute of Child Health, London, United Kingdom; Department of Histopathology, Great Ormond Street Hospital, London, United Kingdom
| | - Kathy Pritchard-Jones
- UCL Institute of Child Health, London, United Kingdom; Department of Paediatric Haematology and Oncology, Great Ormond Street Hospital, London, United Kingdom
| | - Nicholas M Luscombe
- The Francis Crick Institute, London, United Kingdom; UCL Genetics Institute, Department of Genetics, Evolution & Environment, University College London, United Kingdom; Okinawa Institute of Science & Technology, Okinawa, Japan
| | | | - William Mifsud
- UCL Institute of Child Health, London, United Kingdom; Department of Histopathology, Great Ormond Street Hospital, London, United Kingdom.
| |
Collapse
|
20
|
Deng C, Dai R, Li X, Liu F. Genetic variation frequencies in Wilms' tumor: A meta-analysis and systematic review. Cancer Sci 2016; 107:690-9. [PMID: 26892980 PMCID: PMC4970837 DOI: 10.1111/cas.12910] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 02/11/2016] [Accepted: 02/12/2016] [Indexed: 12/11/2022] Open
Abstract
Over the last few decades, numerous biomarkers in Wilms' tumor have been confirmed and shown variations in prevalence. Most of these studies were based on small sample sizes. We carried out a meta-analysis of the research published from 1992 to 2015 to obtain more precise and comprehensive outcomes for genetic tests. In the present study, 70 out of 5175 published reports were eligible for the meta-analysis, which was carried out using Stata 12.0 software. Pooled prevalence for gene mutations WT1, WTX, CTNNB1, TP53, MYCN, DROSHA, and DGCR8 was 0.141 (0.104, 0.178), 0.147 (0.110, 0.184), 0.140 (0.100, 0.190), 0.410 (0.214, 0.605), 0.071 (0.041, 0.100), 0.082 (0.048, 0.116), and 0.036 (0.026, 0.046), respectively. Pooled prevalence of loss of heterozygosity at 1p, 11p, 11q, 16q, and 22q was 0.109 (0.084, 0.133), 0.334 (0.295, 0.373), 0.199 (0.146, 0.252), 0.151 (0.129, 0.172), and 0.148 (0.108, 0.189), respectively. Pooled prevalence of 1q and chromosome 12 gain was 0.218 (0.161, 0.275) and 0.273 (0.195, 0.350), respectively. The limited prevalence of currently known genetic alterations in Wilms' tumors indicates that significant drivers of initiation and progression remain to be discovered. Subgroup analyses indicated that ethnicity may be one of the sources of heterogeneity. However, in meta-regression analyses, no study-level characteristics of indicators were found to be significant. In addition, the findings of our sensitivity analysis and possible publication bias remind us to interpret results with caution.
Collapse
Affiliation(s)
- Changkai Deng
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China.,Chengdu Women and Children's Central Hospital, Chengdu, China
| | - Rong Dai
- Chengdu Center for Disease Control and Prevention, Chengdu, China
| | - Xuliang Li
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Feng Liu
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| |
Collapse
|
21
|
Berry RL, Ozdemir DD, Aronow B, Lindström NO, Dudnakova T, Thornburn A, Perry P, Baldock R, Armit C, Joshi A, Jeanpierre C, Shan J, Vainio S, Baily J, Brownstein D, Davies J, Hastie ND, Hohenstein P. Deducing the stage of origin of Wilms' tumours from a developmental series of Wt1-mutant mice. Dis Model Mech 2015; 8:903-17. [PMID: 26035382 PMCID: PMC4527280 DOI: 10.1242/dmm.018523] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 05/07/2015] [Indexed: 12/28/2022] Open
Abstract
Wilms' tumours, paediatric kidney cancers, are the archetypal example of tumours caused through the disruption of normal development. The genetically best-defined subgroup of Wilms' tumours is the group caused by biallelic loss of the WT1 tumour suppressor gene. Here, we describe a developmental series of mouse models with conditional loss of Wt1 in different stages of nephron development before and after the mesenchymal-to-epithelial transition (MET). We demonstrate that Wt1 is essential for normal development at all kidney developmental stages under study. Comparison of genome-wide expression data from the mutant mouse models with human tumour material of mutant or wild-type WT1 datasets identified the stage of origin of human WT1-mutant tumours, and emphasizes fundamental differences between the two human tumour groups due to different developmental stages of origin. Summary: The comparison of different nephron-specific Wt1-knockout mouse models identifies the stage of origin of human WT1-mutant Wilms' tumours.
Collapse
Affiliation(s)
- Rachel L Berry
- MRC Human Genetics Unit, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Derya D Ozdemir
- MRC Human Genetics Unit, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Bruce Aronow
- Department of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Nils O Lindström
- MRC Human Genetics Unit, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Tatiana Dudnakova
- MRC Human Genetics Unit, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Anna Thornburn
- MRC Human Genetics Unit, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Paul Perry
- MRC Human Genetics Unit, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Richard Baldock
- MRC Human Genetics Unit, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Chris Armit
- MRC Human Genetics Unit, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Anagha Joshi
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Cécile Jeanpierre
- INSERM, UMR 1163, Laboratory of Inherited Kidney Diseases, Paris 75015, France Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris 75015, France
| | - Jingdong Shan
- Biocenter Oulu, InfoTech Oulu, Faculty of Biochemistry and Molecular Medicine, Aapistie 5A, University of Oulu, PO Box 5000, Oulu 90014, Finland
| | - Seppo Vainio
- Biocenter Oulu, InfoTech Oulu, Faculty of Biochemistry and Molecular Medicine, Aapistie 5A, University of Oulu, PO Box 5000, Oulu 90014, Finland
| | - James Baily
- Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - David Brownstein
- Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Jamie Davies
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, 15 George Square, Edinburgh, EH8 9XD, UK
| | - Nicholas D Hastie
- MRC Human Genetics Unit, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Peter Hohenstein
- MRC Human Genetics Unit, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| |
Collapse
|
22
|
Kim WJ, Wittner BS, Amzallag A, Brannigan BW, Ting DT, Ramaswamy S, Maheswaran S, Haber DA. The WTX Tumor Suppressor Interacts with the Transcriptional Corepressor TRIM28. J Biol Chem 2015; 290:14381-90. [PMID: 25882849 DOI: 10.1074/jbc.m114.631945] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Indexed: 02/05/2023] Open
Abstract
WTX encodes a tumor suppressor implicated in the pediatric kidney cancer Wilms tumor and in mesenchymal differentiation with potentially distinct functions in the cytoplasm, at the plasma membrane, and in the nucleus. Although modulating components of the WNT signaling pathway is a proposed function for cytoplasmic and membrane-bound WTX, its nuclear properties are not well understood. Here we report that the transcriptional corepressor TRIM28 is the major binding partner for nuclear WTX. WTX interacted with the coiled coil domain of TRIM28 required for its binding to Krüppel-associated box domains of transcription factors and for its chromatin recruitment through its own coiled coil and proline-rich domains. Knockdown of endogenous WTX reduced the recruitment of TRIM28 to a chromatinized reporter sequence and its ability to repress a target transcript. In mouse embryonic stem cells where TRIM28 plays a major role in repressing endogenous retroviruses and long interspersed elements, knockdown of either TRIM28 or WTX combined with single molecule RNA sequencing revealed a highly significant shared set of differentially regulated transcripts, including derepression of non-coding repetitive sequences and their neighboring protein encoding genes (p < 1e-20). In mesenchymal precursor cells, depletion of WTX and TRIM28 resulted in analogous β-catenin-independent defects in adipogenic and osteogenic differentiation, and knockdown of WTX reduced TRIM28 binding to Pparγ promoter. Together, the physical and functional interaction between WTX and TRIM28 suggests that the nuclear fraction of WTX plays a role in epigenetic silencing, an effect that may contribute to its function as a regulator of cellular differentiation and tumorigenesis.
Collapse
Affiliation(s)
- Woo Jae Kim
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Ben S Wittner
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Arnaud Amzallag
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - Brian W Brannigan
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and
| | - David T Ting
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and From the Departments of Medicine and
| | - Sridhar Ramaswamy
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and From the Departments of Medicine and
| | - Shyamala Maheswaran
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and Surgery
| | - Daniel A Haber
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, Massachusetts 02129 and From the Departments of Medicine and Howard Hughes Medical Institute, Chevy Chase, Maryland 20815
| |
Collapse
|
23
|
Abstract
Wilms' tumor, or nephroblastoma, is the most common pediatric renal cancer. The tumors morphologically resemble embryonic kidneys with a disrupted architecture and are associated with undifferentiated metanephric precursors. Here, we discuss genetic and epigenetic findings in Wilms' tumor in the context of renal development. Many of the genes implicated in Wilms' tumorigenesis are involved in the control of nephron progenitors or the microRNA (miRNA) processing pathway. Whereas the first group of genes has been extensively studied in normal development, the second finding suggests important roles for miRNAs in general-and specific miRNAs in particular-in normal kidney development that still await further analysis. The recent identification of Wilms' tumor cancer stem cells could provide a framework to integrate these pathways and translate them into new or improved therapeutic interventions.
Collapse
Affiliation(s)
- Peter Hohenstein
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom;
| | - Kathy Pritchard-Jones
- UCL Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Jocelyn Charlton
- UCL Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| |
Collapse
|
24
|
Wegert J, Ishaque N, Vardapour R, Geörg C, Gu Z, Bieg M, Ziegler B, Bausenwein S, Nourkami N, Ludwig N, Keller A, Grimm C, Kneitz S, Williams RD, Chagtai T, Pritchard-Jones K, van Sluis P, Volckmann R, Koster J, Versteeg R, Acha T, O'Sullivan MJ, Bode PK, Niggli F, Tytgat GA, van Tinteren H, van den Heuvel-Eibrink MM, Meese E, Vokuhl C, Leuschner I, Graf N, Eils R, Pfister SM, Kool M, Gessler M. Mutations in the SIX1/2 pathway and the DROSHA/DGCR8 miRNA microprocessor complex underlie high-risk blastemal type Wilms tumors. Cancer Cell 2015; 27:298-311. [PMID: 25670083 DOI: 10.1016/j.ccell.2015.01.002] [Citation(s) in RCA: 224] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 11/24/2014] [Accepted: 01/09/2015] [Indexed: 10/24/2022]
Abstract
Blastemal histology in chemotherapy-treated pediatric Wilms tumors (nephroblastoma) is associated with adverse prognosis. To uncover the underlying tumor biology and find therapeutic leads for this subgroup, we analyzed 58 blastemal type Wilms tumors by exome and transcriptome sequencing and validated our findings in a large replication cohort. Recurrent mutations included a hotspot mutation (Q177R) in the homeo-domain of SIX1 and SIX2 in tumors with high proliferative potential (18.1% of blastemal cases); mutations in the DROSHA/DGCR8 microprocessor genes (18.2% of blastemal cases); mutations in DICER1 and DIS3L2; and alterations in IGF2, MYCN, and TP53, the latter being strongly associated with dismal outcome. DROSHA and DGCR8 mutations strongly altered miRNA expression patterns in tumors, which was functionally validated in cell lines expressing mutant DROSHA.
Collapse
Affiliation(s)
- Jenny Wegert
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, and Comprehensive Cancer Center Mainfranken, Wuerzburg University, 97074 Wuerzburg, Germany
| | - Naveed Ishaque
- Division of Theoretical Bioinformatics (B080), German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany; Heidelberg Center for Personalised Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany
| | - Romina Vardapour
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, and Comprehensive Cancer Center Mainfranken, Wuerzburg University, 97074 Wuerzburg, Germany
| | - Christina Geörg
- Heidelberg Center for Personalised Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany
| | - Zuguang Gu
- Division of Theoretical Bioinformatics (B080), German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany; Heidelberg Center for Personalised Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany
| | - Matthias Bieg
- Division of Theoretical Bioinformatics (B080), German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany; Heidelberg Center for Personalised Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany
| | - Barbara Ziegler
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, and Comprehensive Cancer Center Mainfranken, Wuerzburg University, 97074 Wuerzburg, Germany
| | - Sabrina Bausenwein
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, and Comprehensive Cancer Center Mainfranken, Wuerzburg University, 97074 Wuerzburg, Germany
| | - Nasenien Nourkami
- Department of Pediatric Oncology and Hematology, Saarland University Hospital, 66421 Homburg, Germany
| | - Nicole Ludwig
- Department of Human Genetics, Saarland University, 66421 Homburg, Germany
| | - Andreas Keller
- Department of Human Genetics, Saarland University, 66421 Homburg, Germany
| | - Clemens Grimm
- Theodor-Boveri-Institute/Biocenter, Biochemistry, Wuerzburg University, 97074 Wuerzburg, Germany
| | - Susanne Kneitz
- Theodor-Boveri-Institute/Biocenter, Physiological Chemistry, Wuerzburg University, 97074 Wuerzburg, Germany
| | | | - Tas Chagtai
- UCL Institute of Child Health, London WC1N 1EH, UK
| | | | - Peter van Sluis
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, 1007 MB Amsterdam, the Netherlands
| | - Richard Volckmann
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, 1007 MB Amsterdam, the Netherlands
| | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, 1007 MB Amsterdam, the Netherlands
| | - Rogier Versteeg
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, 1007 MB Amsterdam, the Netherlands
| | - Tomas Acha
- Unidad de Oncología Pediátrica, Hospital Materno-Infantil de Málaga, 29011 Malaga, Spain
| | - Maureen J O'Sullivan
- National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, and Trinity College, Dublin 12, Ireland
| | - Peter K Bode
- Department of Pediatric Hematology and Oncology, Children's University Hospital, 8032 Zurich, Switzerland
| | - Felix Niggli
- Department of Pediatric Hematology and Oncology, Children's University Hospital, 8032 Zurich, Switzerland
| | - Godelieve A Tytgat
- Department of Pediatric Oncology, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, 1007 MB Amsterdam, the Netherlands
| | - Harm van Tinteren
- Department of Pediatric Oncology, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, 1007 MB Amsterdam, the Netherlands
| | | | - Eckart Meese
- Department of Human Genetics, Saarland University, 66421 Homburg, Germany
| | - Christian Vokuhl
- Kiel Paediatric Cancer Registry, Christian Albrechts University, 24105 Kiel, Germany
| | - Ivo Leuschner
- Kiel Paediatric Cancer Registry, Christian Albrechts University, 24105 Kiel, Germany
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Saarland University Hospital, 66421 Homburg, Germany
| | - Roland Eils
- Division of Theoretical Bioinformatics (B080), German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany; Heidelberg Center for Personalised Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany; Department for Bioinformatics and Functional Genomics, Institute for Pharmacy and Molecular Biotechnology (IPMB) and BioQuant, Heidelberg University, 69121 Heidelberg, Germany
| | - Stefan M Pfister
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Marcel Kool
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Manfred Gessler
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, and Comprehensive Cancer Center Mainfranken, Wuerzburg University, 97074 Wuerzburg, Germany.
| |
Collapse
|
25
|
Cattaneo E, Ciceri S, Liberati N, Radice P, Tarani L, Selicorni A, Perotti D. Osteopathia striata with cranial sclerosis, Wilms’ tumor and the WTX gene. World J Med Genet 2014; 4:34-38. [DOI: 10.5496/wjmg.v4.i2.34] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/10/2014] [Accepted: 02/18/2014] [Indexed: 02/06/2023] Open
Abstract
Osteopathia striata with cranial sclerosis (OSCS, OMIM#300373) is an X-linked dominant sclerosing bone dysplasia that shows a distinct phenotype in females and males. In 2009, Zandra Jenkins et al found that germline mutations in the FAM123B/WTX/AMER1 gene, mapped to chromosome Xq11.2, cause both the familial and sporadic forms of OSCS. Intriguingly, the WTX gene was already known as a putative tumor suppressor gene, since in 2007 Rivera et al had reported inactivating WTX mutations in Wilms’ tumor (WT), the most frequent renal tumor of childhood. Here we review the heterogeneous clinical presentation of OSCS patients and the involvement of WTX anomalies in OSCS and in WT.
Collapse
|
26
|
Fujita A, Ochi N, Fujimaki H, Muramatsu H, Takahashi Y, Natsume J, Kojima S, Nakashima M, Tsurusaki Y, Saitsu H, Matsumoto N, Miyake N. A novelWTXmutation in a female patient with osteopathia striata with cranial sclerosis and hepatoblastoma. Am J Med Genet A 2014; 164A:998-1002. [DOI: 10.1002/ajmg.a.36369] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Accepted: 11/01/2013] [Indexed: 01/06/2023]
Affiliation(s)
- Atsushi Fujita
- Department of Human Genetics; Yokohama City University Graduate School of Medicine; Yokohama Japan
| | - Nobuhiko Ochi
- Department of Pediatrics; Aichi Prefectural Rehabilitation Center for Children with Disabilities Daini Aoitori Gakuen; Okazaki Japan
| | - Hidehiko Fujimaki
- Division of Neonatology, Center for Maternal-Neonatal Care; Nagoya University Hospital; Nagoya Japan
| | - Hideki Muramatsu
- Department of Pediatrics; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Jun Natsume
- Department of Pediatrics; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Seiji Kojima
- Department of Pediatrics; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Mitsuko Nakashima
- Department of Human Genetics; Yokohama City University Graduate School of Medicine; Yokohama Japan
| | - Yoshinori Tsurusaki
- Department of Human Genetics; Yokohama City University Graduate School of Medicine; Yokohama Japan
| | - Hirotomo Saitsu
- Department of Human Genetics; Yokohama City University Graduate School of Medicine; Yokohama Japan
| | - Naomichi Matsumoto
- Department of Human Genetics; Yokohama City University Graduate School of Medicine; Yokohama Japan
| | - Noriko Miyake
- Department of Human Genetics; Yokohama City University Graduate School of Medicine; Yokohama Japan
| |
Collapse
|
27
|
Szychot E, Apps J, Pritchard-Jones K. Wilms' tumor: biology, diagnosis and treatment. Transl Pediatr 2014; 3:12-24. [PMID: 26835318 PMCID: PMC4728859 DOI: 10.3978/j.issn.2224-4336.2014.01.09] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 01/22/2014] [Indexed: 11/14/2022] Open
Abstract
Wilms' tumor is the commonest renal tumor of childhood affecting one in 10,000 children. It is also one of the successes of paediatric oncology with long term survival above 90% for localised disease and 75% for metastatic disease. Successful management of Wilms' tumor necessitates meticulous attention to correct staging of the tumor and a collaborative effort between paediatric oncologists, specialist surgeons, radiologists, pathologists, and radiation oncologists. Although current treatment protocols are based on risk assignment to minimise toxicity for low risk patients and improve outcomes for those with high risk disease, challenges remain in identifying novel molecular, histological and clinical risk factors for stratification of treatment intensity. Knowledge about Wilms' tumor biology and treatment is evolving rapidly and remains a paradigm for multimodal malignancy treatment. Future efforts will focus on the use of biomarkers to improve risk stratification and the introduction of newer molecularly targeted therapies that will minimise toxicity and improve the outcomes for patients with unfavourable histology and recurrent disease. The aim of this article is to summarise advances in our understanding of the biology of Wilms' tumor and to describe the current approaches to clinical management of patients.
Collapse
|
28
|
Perotti D, Hohenstein P, Bongarzone I, Maschietto M, Weeks M, Radice P, Pritchard-Jones K. Is Wilms tumor a candidate neoplasia for treatment with WNT/β-catenin pathway modulators?--A report from the renal tumors biology-driven drug development workshop. Mol Cancer Ther 2013; 12:2619-27. [PMID: 24258344 DOI: 10.1158/1535-7163.mct-13-0335] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The European Network for Cancer Research in Children and Adolescents consortium organized a workshop in Rome, in June 2012, on "Biology-Driven Drug Development Renal Tumors Workshop" to discuss the current knowledge in pediatric renal cancers and to recommend directions for further research. Wilms tumor is the most common renal tumor of childhood and represents a success of pediatric oncology, with cure rates of more than 85% of cases. However, a substantial minority (∼25%) responds poorly to current therapies and requires "high-risk" treatment or relapse. Moreover, the successfully treated majority are vulnerable to the late effects of treatment, with nearly one quarter reporting severe chronic health conditions by 25 years of follow-up. Main purposes of this meeting were to advance our understanding on the molecular drivers in Wilms tumor, their heterogeneity and interdependencies; to provide updates on the clinical-pathologic associations with biomarkers; to identify eligible populations for targeted drugs; and to model opportunities to use preclinical model systems and prioritize targeted agents for early phase clinical trials. At least three different pathways are involved in Wilms tumor; this review represents the outcome of the workshop discussion on the WNT/β-catenin pathway in Wilms tumorigenesis.
Collapse
Affiliation(s)
- Daniela Perotti
- Corresponding Author: Kathy Pritchard-Jones, Hugh and Catherine Stevenson Professor of Pediatric Oncology, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
29
|
Cardoso LCA, De Souza KRL, De O Reis AH, Andrade RC, Britto AC, De Lima MAFD, Dos Santos ACE, De Faria PS, Ferman S, Seuánez HN, Vargas FR. WT1, WTX and CTNNB1 mutation analysis in 43 patients with sporadic Wilms' tumor. Oncol Rep 2012; 29:315-20. [PMID: 23117548 DOI: 10.3892/or.2012.2096] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 08/28/2012] [Indexed: 11/05/2022] Open
Abstract
Wilms' tumor (WT) is a heterogeneous neoplasia characterized by a number of genetic abnormalities, involving tumor suppressor genes, oncogenes and genes related to the Wnt signaling pathway. Somatic biallelic inactivation of WT1 is observed in 5-10% of sporadic WT. Somatic mutations in exon 3 of CTNNB1, which encodes β-catenin, were initially observed in 15% of WT. WTX encodes a protein that negatively regulates the Wnt/β-catenin signaling pathway and mediates the binding of WT1. In this study, we screened germline and somatic mutations in selected regions of WT1, WTX and CTNNB1 in 43 WT patients. Mutation analysis of WT1 identified two single-nucleotide polymorphisms, one recurrent nonsense mutation (p.R458X) in a patient with proteinuria but without genitourinary findings of Denys-Drash syndrome (DDS) and one novel missense mutation, p.C428Y, in a patient with Denys-Drash syndrome phenotype. WT1 SNP rs16754A>G (R369R) was observed in 17/43 patients, and was not associated with significant difference in age at diagnosis distribution, or with 60-month overall survival rate. WTX mutation analysis identified five sequence variations, two synonymous substitutions (p.Q1019Q and p.D379D), a non-synonymous mutation (p.F159L), one frameshift mutation (p.157X) and a novel missense mutation, p.R560W. Two sequence variations in CTNNB1 were identified, p.T41A and p.S45C. Overall survival of bilateral cases was significantly lower (p=0.005). No difference was observed when survival was analyzed among patients with WT1 or with WTX mutations. On the other hand, the survival of two patients with the CTNNB1 p.T41A mutation was significantly lower (p=0.000517) than the average.
Collapse
Affiliation(s)
- Leila C A Cardoso
- Department of Genetics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21944‑970, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Scott RH, Murray A, Baskcomb L, Turnbull C, Loveday C, Al-Saadi R, Williams R, Breatnach F, Gerrard M, Hale J, Kohler J, Lapunzina P, Levitt GA, Picton S, Pizer B, Ronghe MD, Traunecker H, Williams D, Kelsey A, Vujanic GM, Sebire NJ, Grundy P, Stiller CA, Pritchard-Jones K, Douglas J, Rahman N. Stratification of Wilms tumor by genetic and epigenetic analysis. Oncotarget 2012; 3:327-35. [PMID: 22470196 PMCID: PMC3359888 DOI: 10.18632/oncotarget.468] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Somatic defects at five loci, WT1, CTNNB1, WTX, TP53 and the imprinted 11p15 region, are implicated in Wilms tumor, the commonest childhood kidney cancer. In this study we analysed all five loci in 120 Wilms tumors. We identified epigenetic 11p15 abnormalities in 69% of tumors, 37% were H19 epimutations and 32% were paternal uniparental disomy (pUPD). We identified mutations of WTX in 32%, CTNNB1 in 15%, WT1 in 12% and TP53 in 5% of tumors. We identified several significant associations: between 11p15 and WTX (P=0.007), between WT1 and CTNNB1 (P less than 0.001), between WT1 and pUPD 11p15 (P=0.01), and a strong negative association between WT1 and H19 epimutation (P less than 0.001). We next used these data to stratify Wilms tumor into three molecular Groups, based on the status at 11p15 and WT1. Group 1 tumors (63%) were defined as 11p15-mutant and WT1-normal; a third also had WTX mutations. Group 2 tumors (13%) were WT1-mutant. They either had 11p15 pUPD or were 11p15-normal. Almost all had CTNNB1 mutations but none had H19 epimutation. Group 3 tumors (25%) were defined as 11p15-normal and WT1-normal and were typically normal at all five loci (P less than 0.001). We also identified a novel clinical association between H19 epimutation and bilateral disease (P less than 0.001). These data provide new insights into the pattern, order, interactions and clinical associations of molecular events in Wilms tumor.
Collapse
Affiliation(s)
- Richard H Scott
- Division of Genetics and Epidemiology, Institute of Cancer Research and Royal Marsden Hospital, Sutton, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Rakheja D, Khokhar S, Mitui M, Cost NG. Immunohistochemical expression of GLUT1 and its correlation with unfavorable histology and TP53 codon 72 polymorphism in Wilms tumors. Pediatr Dev Pathol 2012; 15:286-92. [PMID: 22483234 DOI: 10.2350/12-01-1151-oa.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Reprogramming of energy metabolism, such as increased glycolysis, is a hallmark of cancer cells. One mechanism by which cancer cells fuel glycolysis is through increased uptake of glucose across cell membranes via the glucose transporter GLUT1. One of the transcriptional repressors of GLUT1 is wild-type TP53, and cancer-associated loss of function mutations within the DNA-binding domain of TP53 impairs the repressive effect of TP53 on transcriptional activity of the GLUT1 gene promoter. Because TP53 mutations are associated with unfavorable histology (diffuse anaplasia) in Wilms tumors, we hypothesized increased expression of GLUT1 in these tumors. To evaluate this hypothesis, we performed tissue microarray-based immunohistochemistry for GLUT1 in a set of 50 Wilms tumors, including 5 with unfavorable histology. In a subset of 16 favorable histology Wilms tumors, we compared the GLUT1 immunoexpression with TP53 codon 72 polymorphism status. We found consistently stronger immunoexpression of GLUT1 in unfavorable histology Wilms tumors compared to favorable histology Wilms tumors (P = 0.04). We noted that the favorable histology Wilms tumors with a proline residue at position 72 of TP53 tended to have higher immunoexpression of GLUT1, although this immunoexpression did not reach statistical significance in this small set of cases. In summary, our finding of strong GLUT1 immunoexpression in unfavorable histology Wilms tumors indicates that these tumors are likely to be 2-deoxy-2-((18)F)fluoro-d-glucose avid and that GLUT1 should be evaluated as a therapeutic target for these tumors that otherwise show resistance to conventional therapy.
Collapse
Affiliation(s)
- Dinesh Rakheja
- Department of Pathology, Children's Medical Center, Dallas, TX, USA.
| | | | | | | |
Collapse
|
32
|
Haruta M, Arai Y, Watanabe N, Fujiwara Y, Honda S, Ohshima J, Kasai F, Nakadate H, Horie H, Okita H, Hata JI, Fukuzawa M, Kaneko Y. Different incidences of epigenetic but not genetic abnormalities between Wilms tumors in Japanese and Caucasian children. Cancer Sci 2012; 103:1129-35. [PMID: 22409817 DOI: 10.1111/j.1349-7006.2012.02269.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 02/28/2012] [Accepted: 03/07/2012] [Indexed: 11/27/2022] Open
Abstract
Epidemiological studies show that the incidence of Wilms tumor (WT) in East-Asian children is half of that in Caucasian children. Abnormalities of WT1, CTNNB1, WTX, and IGF2 were reported to be involved in Wilms tumorigenesis in Caucasians, although none of the studies simultaneously evaluated the four genes. WTX forms the β-catenin degradation complex; however, the relationship between WTX abnormality and CTNNB1 mutation was uncertain in WTs. We examined abnormalities of the four genes in 114 Japanese with WTs to clarify the relationship between genetic and epigenetic factors and the incidence of WTs. We found that abnormalities of WTX and CTNNB1 were mutually exclusive, and that although CTNNB1 mutation was frequent in WTs with WT1 abnormality, but rare in WTs without, the incidences of WTX abnormality were similar between WTs with or without WT1 abnormality. These findings were consistent with those reported in Caucasian populations, and indicate multiple roles of WTX abnormality. Abnormalities of WT1, WTX and CTNNB1, and loss of IGF2 imprinting (LOI) were detected in 31.6%, 22.8%, 26.3%, and 21.1% of the 114 WTs, respectively. When we selected 101 sporadic WTs, the incidences of WT1, CTNNB1, or WTX abnormality were generally comparable between the two populations, whereas the incidence of IGF2 LOI was lower in Japanese than that of IGF2 LOI reported in Caucasians (P = 0.04). This is the first comprehensive study of the four genes, and the results supported the hypothesis that the lower incidence of IGF2 LOI contributes to the lower incidence of WTs in Japanese children.
Collapse
Affiliation(s)
- Masayuki Haruta
- Department of Cancer Diagnosis, Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Cancer cells silence autosomal tumor suppressor genes by Knudson's two-hit mechanism in which loss-of-function mutations and then loss of heterozygosity occur at the tumor suppressor gene loci. However, the identification of X-linked tumor suppressor genes has challenged the traditional theory of 'two-hit inactivation' in tumor suppressor genes, introducing the novel concept that a single genetic hit can cause loss of tumor suppressor function. The mechanism through which these genes are silenced in human cancer is unclear, but elucidating the details will greatly enhance our understanding of the pathogenesis of human cancer. Here, we review the identification of X-linked tumor suppressor genes and discuss the potential mechanisms of their inactivation. In addition, we also discuss how the identification of X-linked tumor suppressor genes can potentially lead to new approaches in cancer therapy.
Collapse
Affiliation(s)
- Runhua Liu
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Department of Genetics, School of Medicine, University of Alabama at Birmingham and Comprehensive Cancer Center, Birmingham, AL, USA
| | - Mandy Kain
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Lizhong Wang
- Division of Immunotherapy, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Department of Genetics, School of Medicine, University of Alabama at Birmingham and Comprehensive Cancer Center, Birmingham, AL, USA
| |
Collapse
|
34
|
Zin R, Pham K, Ashleigh M, Ravine D, Waring P, Charles A. SNP-based arrays complement classic cytogenetics in the detection of chromosomal aberrations in Wilms’ tumor. Cancer Genet 2012; 205:80-93. [DOI: 10.1016/j.cancergen.2011.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Revised: 12/09/2011] [Accepted: 12/16/2011] [Indexed: 12/11/2022]
|
35
|
Wegert J, Bausenwein S, Roth S, Graf N, Geissinger E, Gessler M. Characterization of primary Wilms tumor cultures as an in vitro model. Genes Chromosomes Cancer 2011; 51:92-104. [PMID: 22034155 DOI: 10.1002/gcc.20936] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 09/07/2011] [Indexed: 01/31/2023] Open
Abstract
Functional analysis of gene candidates and testing of novel therapeutics in Wilms tumors (WT) has been hampered by the lack of in vitro model systems. WT are characterized by a spectrum of histological appearances, but published cell lines are mostly derived from rare anaplastic variants or even non-WT. There has been some success in establishing primary cultures, but these are often poorly characterized or only derived from less frequent WT1 mutant tumors. We report the generation of a set of primary WT-cell cultures using a simple cultivation protocol. Our cultures could be established after preoperative chemotherapy and irrespective of histological subtypes or genetic alterations. The presence of tumor-specific genetic alterations validates these cultures as being tumor-derived. Genetic characterization is of utmost importance as some cultures with similar morphological appearance lacked such alterations and either represent clonal variants or normal cells. By immunohistochemistry, the cells are either epithelial or more mesenchymal, and the latter exhibiting a longer life span with 30 or more passages before undergoing senescence. This may be related to WT being embryonal tumors with a strong differentiation potential that may prevail in vitro. Telomeres progressively shorten with cultivation, but their length does not predict lifespan. hTERT transfection may partly allow establishment of immortalized lines, because 2/7 cultures avoid senescence even in later passages. Importantly, these cells can be efficiently manipulated by transfection, making them a useful model system for in vitro testing.
Collapse
Affiliation(s)
- Jenny Wegert
- Developmental Biochemistry, Biocenter, University of Wuerzburg, Wuerzburg, Germany
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Wilms' tumour (WT) is an embryonal cancer of childhood and is thought to be derived from embryonic kidney precursor cells. The Knudson two hit model was initially thought to occur in WT, but findings emerging from genetic and cytogenetic studies in the past two decades have implicated several genetic events. Recent techniques in genetic analysis have improved our ability to characterise changes in genes involved in WT which include WT1, CTNNB1, IGF2 and WTX. These genetic events have not only provided insight into the pathobiology of this malignancy, but the recognition of these candidate genes may offer potential targets for novel therapies. In this review, we will provide an overview of the pathological, genetic and cytogenetic characteristics of WT.
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW To review the 2009/2010 literature on pediatric genitourinary tumors and highlight the most significant publications. RECENT FINDINGS New techniques such as gene expression profiling, PET, nephron-sparing surgery, and stem cell transplantation are being incorporated into contemporary treatments for pediatric patients with genitourinary tumors. Biologic markers are increasingly being used to help with risk stratification of patients. WT1 mutation and 11p15 loss of heterozygosity have been associated with relapse in very low-risk Wilms tumors treated with surgery alone and may help reduce the use of chemotherapy in some children. Discussion continues on the use of fusion gene status to risk stratify alveolar rhabdomyosarcoma. Meta-analysis of the use of high-dose chemotherapy with autologous hematopoetic stem cell rescue in patients with relapsed Wilms tumor and rhabdomyosarcoma suggests that some patients may benefit more from conventional salvage chemotherapy. New agents are needed for patients with high-risk and relapsed disease to improve outcomes. SUMMARY In general, the prognosis for patients with pediatric genitourinary tumors is favorable. The elucidation of the molecular abnormalities in these tumors is determining risk stratification, treatment strategies, and candidates for new drug development.
Collapse
|
38
|
Pode-Shakked N, Dekel B. Wilms tumor--a renal stem cell malignancy? Pediatr Nephrol 2011; 26:1535-43. [PMID: 21499773 DOI: 10.1007/s00467-011-1858-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 02/03/2011] [Accepted: 02/22/2011] [Indexed: 12/11/2022]
Abstract
Wilms' tumor (WT; nephroblastoma) is the most common pediatric renal malignancy and rated fourth in overall incidence among childhood cancers. It is viewed as a prototype of differentiation failure in human neoplasia as it recapitulates the histology of the nephrogenic zone of the growing fetal kidney. The cellular origin of WT is unclear. However, recent genomic, genetic and epigenetic studies point to an early renal stem/progenitor cell that undergoes malignant transformation as the source for WT. In this context, classical WT shares genes and pathways activated in progenitors committed to the renal lineage. However, direct proof and characterization of the WT initiating cell have remained elusive. Novel methodologies recently adopted from the cancer stem cell scientific field, including the analysis of sorted single human tumor cells, have been applied to WT. These have enabled the identification of cell sub-populations that show similarities-in terms of molecular marker expression-to human fetal kidney progenitors and are, therefore, likely to be derivatives of the same lineage. Further elucidation of the WT cancer stem cell or the cell of origin in human tumors and in transgenic mouse models that generate murine tumors may not only provide novel therapeutic targets but also shed light on the normal kidney stem cell.
Collapse
Affiliation(s)
- Naomi Pode-Shakked
- Pediatric Stem Cell Research Institute, Edmond & Lili Safra Children's Hospital, Sheba Center for Regenerative Medicine, Chaim Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
39
|
Abstract
Genes identified as being mutated in Wilms' tumour include TP53, a classic tumour suppressor gene (TSG); CTNNB1 (encoding β-catenin), a classic oncogene; WTX, which accumulating data indicate is a TSG; and WT1, which is inactivated in some Wilms' tumours, similar to a TSG. However, WT1 does not always conform to the TSG label, and some data indicate that WT1 enhances cell survival and proliferation, like an oncogene. Is WT1 a chameleon, functioning as either a TSG or an oncogene, depending on cellular context? Are these labels even appropriate for describing and understanding the function of WT1?
Collapse
Affiliation(s)
- Vicki Huff
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| |
Collapse
|
40
|
Functional characterization of Wilms tumor-suppressor WTX and tumor-associated mutants. Oncogene 2010; 30:832-42. [PMID: 20956941 DOI: 10.1038/onc.2010.452] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The WTX, Wilms tumor-associated tumor-suppressor gene, is present on the X chromosome and a single WTX mutation may be sufficient to promote carcinogenesis. Unlike the WT1 tumor suppressor, a transcription factor, WTX lacks conserved functional protein domains. To study the function of WTX, we constructed inducible cell lines expressing WTX and tumor-associated WTX mutants. Induction of WTX inhibited cell growth and caused G(1)/G(0) arrest. In contrast, a short, tumor-associated truncation mutant of WTX358 only slightly inhibited cell growth without a significant cell-cycle arrest, although expression of a longer truncation mutant WTX565 led to the growth inhibition and cell-cycle arrest to a similar extent as wild-type WTX. Like WT1, WTX slowed growth and caused cell-cycle arrest through p21 induction. Gene expression profiling showed that these two tumor-suppressors regulated genes in similar pathways, including those implicated in control of the cellular growth, cell cycle, cell death, cancer and cardiovascular system development. When gene expression changes mediated by wild-type WTX were compared with those affected by mutant forms, WTX565 showed a 55% overlap (228 genes) in differentially regulated genes, whereas WTX358 regulated only two genes affected by wild-type WTX, implying that amino-acid residues 358-561 are critical for WTX function.
Collapse
|
41
|
Mengelbier LH, Karlsson J, Lindgren D, Øra I, Isaksson M, Frigyesi I, Frigyesi A, Bras J, Sandstedt B, Gisselsson D. Deletions of 16q in Wilms tumors localize to blastemal-anaplastic cells and are associated with reduced expression of the IRXB renal tubulogenesis gene cluster. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:2609-21. [PMID: 20847289 DOI: 10.2353/ajpath.2010.100130] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Wilms tumor is the most common pediatric renal neoplasm, but few molecular prognostic markers have been identified for this tumor. Somatic deletion in the long arm of chromosome 16 (16q) is known to predict a less favorable outcome in Wilms tumor, but the underlying molecular mechanisms are not known. We show that 16q deletions are typically confined to immature anaplastic-blastic tumor elements, while deletions are absent in maturing tumor components. The smallest region of deletion overlap mapped to a 1.8-Mb segment containing the IRXB gene cluster including IRX3, IRX5, and IRX6, of which IRX3 is a recently identified regulator of tubular maturation during nephrogenesis. Tumors with 16q deletion showed a lower overall mRNA expression of IRXB genes, and 16q-deleted tumor cells failed to express IRX3 while it was expressed in differentiating tubular tumor elements with intact 16q. Consistent with a role for IRX3 in tubular differentiation, gene sets linked to Notch signaling, Rho signaling, and ion channel activity were enriched in tumors with high IRX3 expression, while WTs with low expression were enriched for gene sets linked to cell cycle progression. Low mRNA levels of IRXB genes were associated with diffuse anaplasia, high-stage disease, and death. A disturbed balance between tubular differentiation and self-renewal of anaplastic-blastic elements may thus be one mechanism linking 16q deletion to adverse outcome in Wilms tumor.
Collapse
Affiliation(s)
- Linda Holmquist Mengelbier
- Department of Clinical Genetics, University and Regional Laboratories, Lund University, Skåne University Hospital, SE 221 85 Lund, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW To review the 2008-2009 literature on pediatric genitourinary tumors and highlight the most significant publications. RECENT FINDINGS New techniques such as gene expression profiling, PET, nephron-sparing surgery, and stem cell transplantation are being incorporated into contemporary treatments for pediatric patients with genitourinary tumors. The WTX gene is the most commonly mutated gene in Wilms tumor, and its product enhances Wilms tumor gene 1-mediated transcription. Germline WTX mutations cause an X-linked sclerosing bone dysplasia but do not appear to predispose to Wilms tumor formation. Protocadherin gene clusters on chromosome 5q31 may act as tumor suppressors. In rhabdomyosarcoma, ILK and platelet-derived growth factor receptor-A join the paired box gene 7 and 3-forkhead box O1 fusions as potential therapeutic targets, and muscle-specific microRNAs offer promise as adjuvant therapy. Despite the high cure rate of Wilms tumor, long-term survivors remain at risk of death from various causes. SUMMARY In general, the prognosis for patients with pediatric genitourinary tumors is favorable. The elucidation of the molecular abnormalities in these tumors is determining risk stratification, treatment strategies, and candidates for new drug development.
Collapse
|
43
|
Liu Y, Wang L, Zheng P. X-linked tumor suppressors: perplexing inheritance, a unique therapeutic opportunity. Trends Genet 2010; 26:260-5. [PMID: 20434787 PMCID: PMC2901104 DOI: 10.1016/j.tig.2010.03.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 03/29/2010] [Accepted: 03/30/2010] [Indexed: 01/03/2023]
Abstract
Unlike autosomal genes, the majority of X-linked genes are subject to dosage compensation. As a result, female tissues comprise cells exclusively expressing X-linked genes from one or other parent. The implication of having only one allele of active X-linked genes in cancer pathogenesis, i.e. somatic single-hit inactivation and dominant inheritance, has not been extensively explored. Recent studies have identified FOXP3 and WTX as two X-linked tumor suppressor genes that are somatically inactivated by single genetic hits. Because the predicted dominant inheritance of cancer risk has not been demonstrated in humans, we will discuss the possible conditions that might prevent such dominant inheritance. We also argue that the existence of a genetically intact allele in cancer cells in women, together with apparent abnormal X inactivation in cancer cells, might provide an opportunity to selectively reactivate tumor suppressor genes for cancer therapy.
Collapse
Affiliation(s)
- Yang Liu
- Divisions of Immunotherapy, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48105, USA.
| | | | | |
Collapse
|