1
|
Allen A, McAbee K, Kryger JV. Chromophobe Renal Cell Carcinoma in a Pediatric Patient With Neurofibromatosis Type 1: A Case Report and Review of the Literature. Urology 2024; 193:e92-e95. [PMID: 39173933 DOI: 10.1016/j.urology.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
Renal Cell Carcinoma is rare in the pediatric population, making up only 2%-6% of all pediatric renal tumors. Literature on pediatric Chromophobe Renal Cell Carcinoma (chRCC) is exceptionally limited. In this report, we describe the case of a 12-year-old patient with Neurofibromatosis Type 1 (NF1), incidentally found to have a kidney lesion with pathology revealing chRCC. Treatment included open partial nephrectomy with lymph node dissection and current follow-up is nearly 1 year. To our knowledge, this is the first case of chRCC in the setting of NF1.
Collapse
Affiliation(s)
| | - Kara McAbee
- Medical College of Wisconsin, Milwaukee, WI; Children's Wisconsin, Milwaukee, WI
| | - John V Kryger
- Medical College of Wisconsin, Milwaukee, WI; Children's Wisconsin, Milwaukee, WI
| |
Collapse
|
2
|
Pacyna CN, Anandapadamanaban M, Loudon KW, Hay IM, Perisic O, Li R, Byrne M, Allen L, Roberts K, Hooks Y, Warren AY, Stewart GD, Clatworthy MR, Teichmann SA, Behjati S, Campbell PJ, Williams RL, Mitchell TJ. Multifocal, multiphenotypic tumours arising from an MTOR mutation acquired in early embryogenesis. Oncogene 2024; 43:3268-3276. [PMID: 39271965 PMCID: PMC11518995 DOI: 10.1038/s41388-024-03137-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/05/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024]
Abstract
Embryogenesis is a vulnerable time. Mutations in developmental cells can result in the wide dissemination of cells predisposed to disease within mature organs. We characterised the evolutionary history of four synchronous renal tumours from a 14-year-old girl using whole genome sequencing alongside single cell and bulk transcriptomic sequencing. Phylogenetic reconstruction timed the origin of all tumours to a multipotent embryonic cell committed to the right kidney, around 4 weeks post-conception. Biochemical and structural analysis of their shared MTOR mutation, absent from normal tissues, demonstrates enhanced protein flexibility, enabling a FAT domain hinge to dramatically increase activity of mTORC1 and mTORC2. Developmental mutations, not usually detected in traditional genetic screening, have vital clinical importance in guiding prognosis, targeted treatment, and family screening decisions for paediatric tumours.
Collapse
Affiliation(s)
- Clarissa N Pacyna
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | | | - Kevin W Loudon
- Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Iain M Hay
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Olga Perisic
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Ruoyan Li
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Matthew Byrne
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Laura Allen
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Kirsty Roberts
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Yvette Hooks
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Anne Y Warren
- Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
| | - Grant D Stewart
- Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
- Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Menna R Clatworthy
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Sam Behjati
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
- Department of Paediatrics, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Peter J Campbell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | | | - Thomas J Mitchell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
- Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK.
- Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
3
|
Zou H, Liu C, Ruan Y, Fang L, Wu T, Han S, Dang T, Meng H, Zhang Y. Colorectal medullary carcinoma: a pathological subtype with intense immune response and potential to benefit from immune checkpoint inhibitors. Expert Rev Clin Immunol 2024; 20:997-1008. [PMID: 38459764 DOI: 10.1080/1744666x.2024.2328746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 03/06/2024] [Indexed: 03/10/2024]
Abstract
INTRODUCTION Different pathological types of colorectal cancer have distinguished immune landscape, and the efficacy of immunotherapy will be completely different. Colorectal medullary carcinoma, accounting for 2.2-3.2%, is characterized by massive lymphocyte infiltration. However, the attention to the immune characteristics of colorectal medullary carcinoma is insufficient. AREA COVERED We searched the literature about colorectal medullary carcinoma on PubMed through November 2023to investigate the hallmarks of colorectal medullary carcinoma's immune landscape, compare medullary carcinoma originating from different organs and provide theoretical evidence for precise treatment, including applying immunotherapy and BRAF inhibitors. EXPERT OPINION Colorectal medullary carcinoma is a pathological subtype with intense immune response, with six immune characteristics and has the potential to benefit from immunotherapy. Mismatch repair deficiency, ARID1A missing and BRAF V600E mutation often occurs. IFN-γ pathway is activated and PD-L1 expression is increased. Abundant lymphocyte infiltration performs tumor killing function. In addition, BRAF mutation plays an important role in the occurrence and development, and we can consider the combination of BRAF inhibitors and immunotherapy in patients with BRAF mutant. The exploration of colorectal medullary carcinoma will arouse researchers' attention to the correlation between pathological subtypes and immune response, and promote the process of precise immunotherapy.
Collapse
Affiliation(s)
- Haoyi Zou
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chao Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuli Ruan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lin Fang
- Phase I Clinical Research Center, The Affiliated Hospital of Qingdao University in Shandong, Qingdao, China
| | - Tong Wu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuling Han
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tianjiao Dang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin Medical University Cancer Hospital, Harbin, China
- Clinical Research Center for Colorectal Cancer in Heilongjiang, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
4
|
Glick RD, Romao RLP, Pachl M, Kotagal M, Buchanan AF, Murphy AJ, Tracy ET, Pio L, Cost NG, Godzinski J, Ehrlich PF. Current surgical approaches to pediatric renal tumors. Pediatr Blood Cancer 2024:e31118. [PMID: 38809413 DOI: 10.1002/pbc.31118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Pediatric renal tumors are among the most common pediatric solid malignancies. Surgical resection is a key component in the multidisciplinary therapy for children with kidney tumors. Therefore, it is imperative that surgeons caring for children with renal tumors fully understand the current standards of care in order to provide appropriate surgical expertise within this multimodal framework. Fortunately, the last 60 years of international, multidisciplinary pediatric cancer cooperative group studies have enabled high rates of cure for these patients. This review will highlight the international surgical approaches to pediatric patients with kidney cancer to help surgeons understand the key differences and similarities between the European (International Society of Pediatric Oncology) and North American (Children's Oncology Group) recommendations.
Collapse
Affiliation(s)
- Richard D Glick
- Division of Pediatric Surgery, Cohen Children's Medical Center, Zucker School of Medicine at Northwell/Hofstra, New Hyde Park, New York, USA
| | - Rodrigo L P Romao
- Divisions of Pediatric General Surgery and Pediatric Urology, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Max Pachl
- Department of Paediatric Surgery and Urology, Birmingham Children's Hospital, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Meera Kotagal
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Amanda F Buchanan
- Departments of Urology and Pediatrics, University of Kentucky, Lexington, Kentucky, USA
| | - Andrew J Murphy
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Elisabeth T Tracy
- Division of Pediatric Surgery, Duke Children's Hospital and Health Center, Durham, North Carolina, USA
| | - Luca Pio
- Paediatric Surgery Unit, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Nicholas G Cost
- Division of Urology, Department of Surgery, University of Colorado School of Medicine and the Surgical Oncology Program at Children's Hospital Colorado, Aurora, Colorado, USA
| | - Jan Godzinski
- Department of Pediatric Surgery, Marciniak Hospital, Wroclaw, Poland
- Department of Pediatric Traumatology and Emergency Medicine, Medical University, Wroclaw, Poland
| | - Peter F Ehrlich
- Section of Pediatric Surgery, University of Michigan Department of Surgery, Ann Arbor, Michigan, USA
| |
Collapse
|
5
|
Zhang Y, Li C, Deng X, Urabe F, Burotto M, Buti S, Giudice GC, Zhao Z, Yang C, Sun J, Du Y, Wang S. Treatment of metastatic TFE3 microphthalmia transcription factor translocation renal cell carcinoma: a case report. Transl Pediatr 2024; 13:499-507. [PMID: 38590368 PMCID: PMC10998990 DOI: 10.21037/tp-24-35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 03/12/2024] [Indexed: 04/10/2024] Open
Abstract
Background Microphthalmia-associated transcription factor/transcription factor E (MiTF/TFE) translocation renal cell carcinoma (RCC) is a rare type of non-clear cell RCC (nccRCC), which is more common in females. Currently, there is no standardized treatment for advanced metastatic microphthalmia translocation RCC (MiT-RCC). The main treatment modalities include surgery, chemotherapy, immunotherapy, anti-vascular endothelial growth factor or vascular endothelial growth factor receptor (VEGFR) inhibitors, mammalian target of rapamycin (mTOR) inhibitors, and targeted therapy against the mesenchymal-epithelial transition (MET) factor signaling pathway. Case Description We present the case of an 8-year-old male patient with hematuria and paroxysmal urinary pain. Based on tumor genetic testing results and targeted drug matching analysis, the patient underwent tumor biopsy, tumor radical surgery with vascular osteotomy, and cervicothoracic lymph node dissection. The patient was then treated with a combination of immunotherapy [sintilimab, a drug directed against programmed cell death receptor-1 (PD-1)] and VEGFR tyrosine kinase inhibitor (TKI) (from pazopanib to sunitinib). Throughout the 10 cycles of conventional chemotherapy (seven courses of sintilimab since the start of the third chemotherapy treatment), the patient's condition remained stable, with no tumor recurrence at the primary site. However, in the later stages, the patient developed a large amount of ascites, and the family requested discontinuation of treatment, ultimately leading to the patient's death. Conclusions In this case report, we summarize the therapeutic strategy of a young patient with metastatic transcription factor E3 (TFE3) MiT-RCC. For this disease, early immunotherapy and the use of precision-targeted drugs may have a favorable impact on the survival prognosis of the patient but may still be of less benefit in children with advanced multiple metastases. Therefore, further research on tumor driver genes, among other treatment components, is urgently needed to improve precision therapy.
Collapse
Affiliation(s)
- Yunlong Zhang
- Department of Pediatric Surgical Oncology, Children’s Hospital of Chongqing Medical University, The National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Changchun Li
- Department of Pediatric Surgical Oncology, Children’s Hospital of Chongqing Medical University, The National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiaobin Deng
- Department of Pediatric Surgical Oncology, Children’s Hospital of Chongqing Medical University, The National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Fumihiko Urabe
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Sebastiano Buti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Giulia Claire Giudice
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Zhenzhen Zhao
- Department of Pediatric Surgical Oncology, Children’s Hospital of Chongqing Medical University, The National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Chao Yang
- Department of Pediatric Surgical Oncology, Children’s Hospital of Chongqing Medical University, The National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jian Sun
- Department of Pediatric Surgical Oncology, Children’s Hospital of Chongqing Medical University, The National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yifei Du
- Department of Pediatric Surgical Oncology, Children’s Hospital of Chongqing Medical University, The National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Shan Wang
- Department of Pediatric Surgical Oncology, Children’s Hospital of Chongqing Medical University, The National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
6
|
Kamanda S, Huanca-Amesquita L, Milla E, Argani P, Epstein JI. Clinicopathologic Classification of Renal Cell Carcinoma in Patients ≤40 Years Old From Peru. Int J Surg Pathol 2024; 32:35-45. [PMID: 37062985 PMCID: PMC10577151 DOI: 10.1177/10668969231167539] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
INTRODUCTION There are scant data on renal cell carcinoma (RCC) from relatively younger patients in South America using contemporary classification. METHODS Fifty-nine consecutively treated patients with RCC (≤40 years old) were assessed from the National Institute of Neoplastic Diseases in Peru from 2008 to 2020 (34 males; 25 females), age range of 13 to 40 years. RESULTS Most common presenting symptoms were flank pain (n = 40), hematuria (n = 19), and weight loss (n = 12). Associated conditions included 4 patients with proven or presumed tuberous sclerosis and 1 patient with von Hippel Lindau syndrome, all with clear cell RCC. Tumor histopathology was clear cell RCC in 32 of 59 (54%), chromophobe RCC in 6 of 59 (10%), and 5 of 59 (8%) each of papillary RCC and MiT family translocation-associated RCC. Four of 59 (7%) were FH-deficient RCC and 2 of 59 (3%) remained unclassified. The remaining tumors were isolated examples of clear cell papillary renal cell tumor, eosinophilic solid and cystic RCC (ESC RCC), RCC with fibromyomatous stroma, sarcomatoid RCC, and sarcomatoid clear cell RCC. Of the 4 FH-deficient RCCs, none had the classic morphology. The 5 MiT family translocation RCCs had variable morphology. There were 41 tumors without recurrence or metastases, 3 tumors with local recurrence only, 8 tumors with metastases only, and 7 tumors with both local recurrence and metastases. CONCLUSIONS The current study demonstrates the importance of special studies in accurately classifying RCC in younger individuals. The distribution of RCC subtypes in younger individuals is similar between 2 representative large institutions of the United States and Peru.
Collapse
Affiliation(s)
- Sonia Kamanda
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | - Esperanza Milla
- Department of Pathology, National Institute of Neoplastic Diseases, Lima, Peru
| | - Pedram Argani
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Jonathan I Epstein
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Departments of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Departments of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|
7
|
Sprokkerieft J, van der Beek JN, Spreafico F, Selle B, Thebaud E, Chowdhury T, Brok J, Ottóffy G, Sun X, Ramírez Villar GL, Sagoyan G, Segers H, Doganis D, Serra A, Lemelle L, Graf N, Verschuur AC, Tytgat GAM, van den Heuvel‐Eibrink MM. Targeted therapies in children with renal cell carcinoma (RCC): An International Society of Pediatric Oncology-Renal Tumor Study Group (SIOP-RTSG)-related retrospective descriptive study. Cancer Med 2024; 13:e6782. [PMID: 39102694 PMCID: PMC10807685 DOI: 10.1002/cam4.6782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 11/10/2023] [Accepted: 11/21/2023] [Indexed: 08/07/2024] Open
Abstract
INTRODUCTION Introduction: Renal cell carcinoma (RCC) is a very rare pediatric renal tumor. Robust evidence to guide treatment is lacking and knowledge on targeted therapies and immunotherapy is mainly based on adult studies. Currently, the International Society of Pediatric Oncology-Renal Tumor Study Group (SIOP-RTSG) 2016 UMBRELLA protocol recommends sunitinib for metastatic or unresectable RCC. METHODS This retrospective study describes the effects of tyrosine kinase inhibitors (TKI), anti-programmed cell death 1 (PD-(L)1) monoclonal antibodies, and immunotherapeutic regimens in advanced-stage and relapsed pediatric RCC. RESULTS Of the 31 identified patients (0-18 years) with histologically proven RCC, 3/31 presented with TNM stage I/II, 8/31 with TNM stage III, and 20/31 with TNM stage IV at diagnosis. The majority were diagnosed with translocation type RCC (MiT-RCC) (21/31) and the remaining patients mainly presented with papillary or clear-cell RCC. Treatment in a neoadjuvant or adjuvant setting, or upon relapse or progression, included mono- or combination therapy with a large variety of drugs, illustrating center specific choices in most patients. Sunitinib was often administered as first choice and predominantly resulted in stable disease (53%). Other frequently used drugs included axitinib, cabozantinib, sorafenib, and nivolumab; however, no treatment seemed more promising than sunitinib. Overall, 15/31 patients died of disease, 12/31 are alive with active disease, and only four patients had a complete response. The sample size and heterogeneity of this cohort only allowed descriptive statistical analysis. CONCLUSION This study provides an overview of a unique series of clinical and treatment characteristics of pediatric patients with RCC treated with targeted therapies.
Collapse
Affiliation(s)
- Julia Sprokkerieft
- Department of Pediatric OncologyPrincess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Justine N. van der Beek
- Department of Pediatric OncologyPrincess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Filippo Spreafico
- Pediatric Oncology Unit, Department of Medical Oncology and HematologyFondazione IRCCS Istituto Nazionale dei TumoriMilanoItaly
| | - Barbara Selle
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric NeurooncologyGerman Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany
| | | | - Tanzina Chowdhury
- Pediatric Oncology, Great Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Jesper Brok
- Department of Pediatric Hematology and Oncology, RigshospitaletCopenhagen University HospitalCopenhagenDenmark
| | - Gábor Ottóffy
- Department of Pediatrics, Medical SchoolUniversity of PécsPécsHungary
| | - Xiaofei Sun
- Department of Pediatric OncologySun Yat‐sen University Cancer CenterGuanghzouChina
| | | | - Garik Sagoyan
- N.N. Blokhin National Medical Research Center of OncologyMoscowRussia
| | - Heidi Segers
- Department of Pediatric Hemato‐OncologyUniversity Hospitals Leuven and Catholic University LeuvenLeuvenBelgium
| | | | - Annalisa Serra
- Dipartimento di Onco‐Ematologia e Medicina TrasfusionaleIRCCS Ospedale Bambino GesùRomaItaly
| | - Lauriane Lemelle
- SIREDO Oncology Center (Care, Innovation and Research for Children and AYA with Cancer)Institut CurieParisFrance
| | - Norbert Graf
- Department of Pediatric Oncology and HematologySaarland UniversityHomburgGermany
| | - Arnauld C. Verschuur
- Department of Pediatric Hematology‐Oncology, Hôpital d'Enfants de la Timone, APHMMarseilleFrance
| | - Godelieve A. M. Tytgat
- Department of Pediatric OncologyPrincess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | |
Collapse
|
8
|
Zhao L, Liu W, Chu L, Luo L. Factors associated with survival in paediatric and adolescent renal cell carcinoma: a population-based study. ANZ J Surg 2023; 93:2710-2715. [PMID: 37458221 DOI: 10.1111/ans.18614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/12/2023] [Accepted: 07/06/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND The purpose of this study was to conduct a population-based study to determine the prognosis of renal cell carcinoma (RCC) in children and adolescents. METHODS Patients with RCC who were registered in the Surveillance, Epidemiology, and End Results (SEER) program between 2000 and 2018 had their demographic and clinical characteristics evaluated retrospectively. The log-rank test was used to compare survival curves. Kaplan-Meier estimates were used to generate survival curves based on various factors. To identify factors associated with overall survival, Cox proportional-hazards regression was used. RESULTS A total of 251 patients were enrolled in the study. For all patients, the overall survival (OS) rates at 3- and 5- year were 93.5% and 92.0%, respectively. A multivariable study revealed that the following factors were independently associated with overall survival: sex, race, histologic type, SEER stage, AJCC stage, and type of surgery. Cox analysis showed that white patients had the lowest risk of mortality (hazard ratio (HR) 2.58, 95% confidence interval (CI), 1.33-4.99; P = 0.005), compared with black patients. Patients having metastatic disease had significantly higher mortality risk (HR 43, 95% CI, 14.8-125; P < 0.001) than the patients with localized tumour. CONCLUSIONS Our study emphasizes the importance of race, SEER stage, and surgery in the prognosis of paediatric RCC, providing valuable epidemiological evidence for clinical practice. Economic studies assessing a race/ethnic group specific strategy are also required.
Collapse
Affiliation(s)
- Lingling Zhao
- Department of Pathology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Wenyuan Liu
- Department of Pediatrics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Likai Chu
- Department of Ultrasound, Children's Hospital of Soochow University, Suzhou, China
| | - Laiyue Luo
- Department of Nephrology, Anji Branch of the First Affiliated Hospital of Zhejiang University, Anji County People's Hospital, Huzhou, China
| |
Collapse
|
9
|
Drobner J, Portal D, Runcie K, Yang Y, Singer EA. Systemic Treatment for Advanced and Metastatic Non-Clear Cell Renal Cell Carcinoma: Examining Modern Therapeutic Strategies for a Notoriously Challenging Malignancy. J Kidney Cancer VHL 2023; 10:37-60. [PMID: 37789902 PMCID: PMC10542704 DOI: 10.15586/jkcvhl.v10i3.295] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/09/2023] [Indexed: 10/05/2023] Open
Abstract
Non-clear cell renal cell carcinoma (nccRCC) is a heterogeneous group of malignancies that represents 25% of renal cell carcinoma (RCC) cases. Treatment for non-clear cell histologies is mostly based on evidence from small phase II clinical trials or extrapolated from successful therapies in clear cell RCC because of the low incidence of non-clear cell pathology. Advances in genomic profiling have improved clinicians' understanding of molecular targets for nccRCC, such as altered mesenchymal epithelial transition (MET) gene status and fumarate hydratase (FH) gene inactivation, but patient outcomes remain poor and optimal management of this disease remains unclear. This review assesses outcomes by histologic subtype from 27 prospective and 13 ongoing clinical trials to identify therapeutic strategies for advanced or metastatic nccRCC. Vascular endothelial growth factor tyrosine kinase inhibitors (TKI), such as sunitinib, and mammalian target of rapamycin (mTOR) inhibitors, such as everolimus, have demonstrated efficacy and remain viable treatment options, with a preference for sunitinib. However, everolimus is preferred in patients with chromophobe RCC because folliculin (FLCN) gene mutations upregulate the mTOR pathway. Novel TKIs, such as cabozantinib, show improved outcomes in patients with papillary RCC because of targeted MET inhibition. Platinum-based chemotherapy continues to be the recommended treatment strategy for collecting duct and medullary RCC. Clinically meaningful antitumor activity has been observed across all non-clear cell histologies for immune checkpoint inhibitors, such as nivolumab, pembrolizumab, and ipilimumab. Ongoing trials are evaluating novel tyrosine kinase inhibitor and immunotherapy combination regimens, with an emphasis on the promising MET-inhibitor cabozantinib and pembrolizumab plus lenvatinib.
Collapse
Affiliation(s)
- Jake Drobner
- Division of Urology, Rutgers Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ, USA
| | - Daniella Portal
- Division of Urology, Rutgers Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, NJ, USA
| | - Karie Runcie
- Division of Hematology/Oncology, New York-Presbyterian/Columbia University Medical Center, New York, NY, USA
| | - Yuanquan Yang
- Genitourinary Oncology Section, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center-James Cancer Hospital, Columbus, OH, USA
| | - Eric A. Singer
- Division of Urologic Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
10
|
Grubliauskaite M, van der Perk MEM, Bos AME, Meijer AJM, Gudleviciene Z, van den Heuvel-Eibrink MM, Rascon J. Minimal Infiltrative Disease Identification in Cryopreserved Ovarian Tissue of Girls with Cancer for Future Use: A Systematic Review. Cancers (Basel) 2023; 15:4199. [PMID: 37686475 PMCID: PMC10486797 DOI: 10.3390/cancers15174199] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Ovarian tissue cryopreservation and transplantation are the only available fertility techniques for prepubertal girls with cancer. Though autotransplantation carries a risk of reintroducing malignant cells, it can be avoided by identifying minimal infiltrative disease (MID) within ovarian tissue. METHODS A broad search for peer-reviewed articles in the PubMed database was conducted in accordance with PRISMA guidelines up to March 2023. Search terms included 'minimal residual disease', 'cryopreservation', 'ovarian', 'cancer' and synonyms. RESULTS Out of 542 identified records, 17 were included. Ovarian tissues of at least 115 girls were evaluated and categorized as: hematological malignancies (n = 56; 48.7%), solid tumors (n = 42; 36.5%) and tumors of the central nervous system (n = 17; 14.8%). In ovarian tissue of 25 patients (21.7%), MID was detected using RT-qPCR, FISH or multicolor flow cytometry: 16 of them (64%) being ALL (IgH rearrangements with/without TRG, BCL-ABL1, EA2-PBX1, TEL-AML1 fusion transcripts), 3 (12%) Ewing sarcoma (EWS-FLI1 fusion transcript, EWSR1 rearrangements), 3 (12%) CML (BCR-ABL1 fusion transcript, FLT3) and 3 (12%) AML (leukemia-associated immunophenotypes, BCR-ABL1 fusion transcript) patients. CONCLUSION While the majority of malignancies were found to have a low risk of containing malignant cells in ovarian tissue, further studies are needed to ensure safe implementation of future fertility restoration in clinical practice.
Collapse
Affiliation(s)
- Monika Grubliauskaite
- Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Santariskiu Str. 4, LT-08406 Vilnius, Lithuania
- Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
- Department of Biobank, National Cancer Institute, Santariskiu Str. 1, LT-08406 Vilnius, Lithuania
| | | | - Annelies M. E. Bos
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Department of Reproductive Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | | | - Zivile Gudleviciene
- Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21/27, LT-03101 Vilnius, Lithuania
| | - Marry M. van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Division of Child Health, UMCU-Wilhelmina Children’s Hospital, 3584 EA Utrecht, The Netherlands
| | - Jelena Rascon
- Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Santariskiu Str. 4, LT-08406 Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21/27, LT-03101 Vilnius, Lithuania
| |
Collapse
|
11
|
Nze C, Msaouel P, Derbala MH, Stephen B, Abonofal A, Meric-Bernstam F, Tannir NM, Naing A. A Phase II Clinical Trial of Pembrolizumab Efficacy and Safety in Advanced Renal Medullary Carcinoma. Cancers (Basel) 2023; 15:3806. [PMID: 37568622 PMCID: PMC10417298 DOI: 10.3390/cancers15153806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Renal medullary carcinoma (RMC) is one of most aggressive renal cell carcinomas and novel therapeutic strategies are therefore needed. Recent comprehensive molecular and immune profiling of RMC tissues revealed a highly inflamed phenotype, suggesting the potential therapeutic role for immune checkpoint therapies. We present the first prospective evaluation of an immune checkpoint inhibitor in a cohort of patients with RMC. METHODS A cohort of patients with locally advanced or metastatic RMC was treated with pembrolizumab 200 mg intravenously every 21 days in a phase II basket trial (ClinicalTrials.gov: NCT02721732). Responses were assessed by irRECIST. Tumor tissues were evaluated for PD-L1 expression and for tumor-infiltrating lymphocyte (TIL) levels. Somatic mutations were assessed by targeted next-generation sequencing. RESULTS A total of five patients were treated. All patients had advanced disease, with the majority of patients (60%) having metastatic disease at diagnosis. All patients had rapid disease progression despite pembrolizumab treatment, with a median time to progression of 8.7 weeks. One patient (patient 5) experienced sudden clinical progression immediately after treatment initiation and was thus taken off trial less than one week after receiving pembrolizumab. CONCLUSIONS This prospective evaluation showed no evidence of clinical activity for pembrolizumab in patients with RMC, irrespective of PD-L1 or TIL levels.
Collapse
Affiliation(s)
- Chijioke Nze
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mohamed H. Derbala
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.H.D.); (B.S.); (F.M.-B.)
| | - Bettzy Stephen
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.H.D.); (B.S.); (F.M.-B.)
| | - Abdulrahman Abonofal
- Department of Medicine, Section of Hematology/Oncology, West Virginia University, Morgantown, WV 26506, USA;
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.H.D.); (B.S.); (F.M.-B.)
| | - Nizar M. Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.H.D.); (B.S.); (F.M.-B.)
| |
Collapse
|
12
|
Yang K, Ma Y, Dai S, Dong R. MiT family translocation renal cell carcinoma with retroperitoneal metastasis in childhood: a case report. Front Pediatr 2023; 11:1141223. [PMID: 37528880 PMCID: PMC10388245 DOI: 10.3389/fped.2023.1141223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/30/2023] [Indexed: 08/03/2023] Open
Abstract
RCC accounts for only 0.1%-0.3% of all kidney tumors and 2%-6% of malignant kidney tumors in children. Accounting for approximately one-third of the total number of cases in children and adolescents with RCC, Xp11.2 tRCC is the most common subtype of the MiT family translocation renal cell carcinoma, which is a group of rare childhood and adult tumors, characterized by recurrent gene rearrangements of TFE3. Here we report a rare case of a 6-year-old male patient with MiT family translocation renal cell carcinoma (MiTF tRCC) where the patient developed retroperitoneal metastasis. The patient underwent partial nephrectomy (PN), radical nephrectomy (RN), abdominal lymph node resection, and intestinal adhesion lysis. Microscopically, we detected focal and nest clump-shaped clusters of tumor cells whose cytoplasm was bright and clear. Immunohistochemistry (IHC) showed tumor cells diffusely expressed TFE3, and fluorescence in situ hybridization (FISH) demonstrated disruption of the TFE3 locus, confirming the diagnosis of Xp11.2 tRCC, the most common subtype of MiTF tRCC. Eventually, the patient obtained a good therapeutic result. This case can provide a good reference and guidance for pediatric urologists and oncologists to recognize and diagnose rare renal cell carcinoma in children.
Collapse
|
13
|
Roebuck DJ, Chippington S, Berry BD, Gibson C. Core needle biopsy and embolization of fat-poor renal tumors in children with tuberous sclerosis complex. World J Pediatr 2023; 19:614-618. [PMID: 36795318 DOI: 10.1007/s12519-023-00692-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/19/2023] [Indexed: 02/17/2023]
Affiliation(s)
- Derek J Roebuck
- Department of Medical Imaging, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, 6009, Australia.
- Division of Paediatrics, Medical School, University of Western Australia, Crawley, 6009, Australia.
| | - Samantha Chippington
- Department of Radiology, Great Ormond Street Hospital for Children, London, WC1N 3JH, UK
| | - Bligh D Berry
- PathWest Laboratory Medicine WA, Murdoch, 6150, Australia
| | - Craig Gibson
- Department of Medical Imaging, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, 6009, Australia
| |
Collapse
|
14
|
Vokshi BH, Davidson G, Tawanaie Pour Sedehi N, Helleux A, Rippinger M, Haller AR, Gantzer J, Thouvenin J, Baltzinger P, Bouarich R, Manriquez V, Zaidi S, Rao P, Msaouel P, Su X, Lang H, Tricard T, Lindner V, Surdez D, Kurtz JE, Bourdeaut F, Tannir NM, Davidson I, Malouf GG. SMARCB1 regulates a TFCP2L1-MYC transcriptional switch promoting renal medullary carcinoma transformation and ferroptosis resistance. Nat Commun 2023; 14:3034. [PMID: 37236926 DOI: 10.1038/s41467-023-38472-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Renal medullary carcinoma (RMC) is an aggressive tumour driven by bi-allelic loss of SMARCB1 and tightly associated with sickle cell trait. However, the cell-of-origin and oncogenic mechanism remain poorly understood. Using single-cell sequencing of human RMC, we defined transformation of thick ascending limb (TAL) cells into an epithelial-mesenchymal gradient of RMC cells associated with loss of renal epithelial transcription factors TFCP2L1, HOXB9 and MITF and gain of MYC and NFE2L2-associated oncogenic and ferroptosis resistance programs. We describe the molecular basis for this transcriptional switch that is reversed by SMARCB1 re-expression repressing the oncogenic and ferroptosis resistance programs leading to ferroptotic cell death. Ferroptosis resistance links TAL cell survival with the high extracellular medullar iron concentrations associated with sickle cell trait, an environment propitious to the mutagenic events associated with RMC development. This unique environment may explain why RMC is the only SMARCB1-deficient tumour arising from epithelial cells, differentiating RMC from rhabdoid tumours arising from neural crest cells.
Collapse
Affiliation(s)
- Bujamin H Vokshi
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400, Illkirch, France
| | - Guillaume Davidson
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400, Illkirch, France
| | - Nassim Tawanaie Pour Sedehi
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400, Illkirch, France
| | - Alexandra Helleux
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400, Illkirch, France
| | - Marc Rippinger
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400, Illkirch, France
| | - Alexandre R Haller
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400, Illkirch, France
| | - Justine Gantzer
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400, Illkirch, France
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, 67200, Strasbourg, France
| | - Jonathan Thouvenin
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400, Illkirch, France
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, 67200, Strasbourg, France
| | - Philippe Baltzinger
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400, Illkirch, France
| | - Rachida Bouarich
- INSERM U830, Équipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, Institut Curie Research Centre, 75005, Paris, France
| | - Valeria Manriquez
- INSERM U830, Équipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, Institut Curie Research Centre, 75005, Paris, France
| | - Sakina Zaidi
- INSERM U830, Équipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, Institut Curie Research Centre, 75005, Paris, France
| | - Priya Rao
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, Division of Quantitative Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hervé Lang
- Department of Urology, CHRU Strasbourg, Strasbourg University, 67000, Strasbourg, France
| | - Thibault Tricard
- Department of Urology, CHRU Strasbourg, Strasbourg University, 67000, Strasbourg, France
| | - Véronique Lindner
- Department of Pathology, CHRU Strasbourg, Strasbourg University, 67200, Strasbourg, France
| | - Didier Surdez
- Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- INSERM, U830, Pediatric Translational Research, PSL Research University, SIREDO Oncology Center, Institut Curie, Paris, France
| | - Jean-Emmanuel Kurtz
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, 67200, Strasbourg, France
| | - Franck Bourdeaut
- INSERM U830, Équipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, Institut Curie Research Centre, 75005, Paris, France
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Irwin Davidson
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400, Illkirch, France.
- 'Équipe Labellisée' Ligue National contre le Cancer, Paris, France.
| | - Gabriel G Malouf
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400, Illkirch, France.
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, 67200, Strasbourg, France.
- 'Équipe Labellisée' Ligue National contre le Cancer, Paris, France.
| |
Collapse
|
15
|
MRI Characteristics of Pediatric and Young-Adult Renal Cell Carcinoma: A Single-Center Retrospective Study and Literature Review. Cancers (Basel) 2023; 15:cancers15051401. [PMID: 36900194 PMCID: PMC10000563 DOI: 10.3390/cancers15051401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Pediatric renal cell carcinoma (RCC) is a rare malignancy. Magnetic resonance imaging (MRI) is the preferred imaging modality for assessment of these tumors. The previous literature has suggested that cross-sectional-imaging findings differ between RCC and other pediatric renal tumors and between RCC subtypes. However, studies focusing on MRI characteristics are limited. Therefore, this study aims to identify MRI characteristics of pediatric and young-adult RCC, through a single-center case series and literature review. Six identified diagnostic MRI scans were retrospectively assessed, and an extensive literature review was conducted. The included patients had a median age of 12 years (63-193 months). Among other subtypes, 2/6 (33%) were translocation-type RCC (MiT-RCC) and 2/6 (33%) were clear-cell RCC. Median tumor volume was 393 cm3 (29-2191 cm3). Five tumors had a hypo-intense appearance on T2-weighted imaging, whereas 4/6 were iso-intense on T1-weighted imaging. Four/six tumors showed well-defined margins. The median apparent diffusion coefficient (ADC) values ranged from 0.70 to 1.20 × 10-3 mm2/s. In thirteen identified articles focusing on MRI characteristics of MiT-RCC, the majority of the patients also showed T2-weighted hypo-intensity. T1-weighted hyper-intensity, irregular growth pattern and limited diffusion-restriction were also often described. Discrimination of RCC subtypes and differentiation from other pediatric renal tumors based on MRI remains difficult. Nevertheless, T2-weighted hypo-intensity of the tumor seems a potential distinctive characteristic.
Collapse
|
16
|
Hont AB, Dumont B, Sutton KS, Anderson J, Kentsis A, Drost J, Hong AL, Verschuur A. The tumor microenvironment and immune targeting therapy in pediatric renal tumors. Pediatr Blood Cancer 2022; 70 Suppl 2:e30110. [PMID: 36451260 DOI: 10.1002/pbc.30110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 12/04/2022]
Abstract
This review highlights the role of several immunomodulating elements contributing to the tumor microenvironment of various pediatric renal tumors including Wilms tumor. The roles of innate and adaptive immune cells in renal tumors are summarized as well as immunomodulatory cytokines and other proteins. The expression and the predictive role of checkpoint modulators like PD-L1 and immunomodulating proteins like glypican-3, B7-H3, COX-2 are highlighted with a translational view toward potential therapeutic innovations. We further discuss the current state of preclinical models in advancing this field of study. Finally, examples of clinical trials of immunomodulating strategies such as monoclonal antibodies and chimeric antigen receptor T (CAR-T) cells for relapsed/refractory/progressive pediatric renal tumors are described.
Collapse
Affiliation(s)
- Amy B Hont
- Department of Hematology/Oncology, Children's National Hospital, George Washington University, Washington, District of Columbia, USA
| | - Benoit Dumont
- Pediatric Hematology and Oncology Institute, Léon Bérard Cancer Center, Lyon, France
| | - Kathryn S Sutton
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - John Anderson
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alex Kentsis
- Tow Center for Developmental Oncology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center and Weill Medical College of Cornell University, New York, New York, USA
| | - Jarno Drost
- Princess Máxima Center and Oncode Institute, Utrecht, The Netherlands
| | - Andrew L Hong
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Arnauld Verschuur
- Department of Pediatric Hematology and Oncology, Hôpital d'Enfants de la Timone, APHM, Marseille, France
| |
Collapse
|
17
|
van de Beek I, Glykofridis IE, Wagner A, den Toom DT, Bongers EMHF, van Leenders GJLH, Johannesma PC, Meijers‐Heijboer HEJ, Wolthuis RMF, van Steensel MAM, Dubbink HJ, Houweling AC. Combined germline pathogenic variants in FLCN and TP53 are associated with early onset renal cell carcinoma and brain tumors. Mol Genet Genomic Med 2022; 11:e2098. [PMID: 36382415 PMCID: PMC9938753 DOI: 10.1002/mgg3.2098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND We present a family consisting of a father and his two children with an exceptional phenotype of childhood renal cell carcinoma and brain tumors. Extensive genetic testing revealed two inherited tumor predisposition syndromes in all three family members: Birt-Hogg-Dubé syndrome and Li-Fraumeni syndrome. The corresponding genes (FLCN and TP53) are both located on the short arm of chromosome 17. METHODS We describe the phenotype and performed single nucleotide polymorphism (SNP)-based loss of heterozygosity (LOH) analysis of the tumors. RESULTS All examined tumors showed somatic loss of the wild-type alleles of both FLCN and TP53. CONCLUSIONS We hypothesize that a synergistic effect of both mutations caused the unusual phenotype of childhood renal cell carcinoma in this family. This family emphasizes the importance of further genetic testing if a tumor develops at an unexpected young age in an inherited cancer predisposition syndrome.
Collapse
Affiliation(s)
- Irma van de Beek
- Department of Human GeneticsAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| | - Iris E. Glykofridis
- Department of Human Genetics, Cancer Center AmsterdamAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| | - Anja Wagner
- Department of Clinical GeneticsUniversity Medical Center Rotterdam, Erasmus MC Cancer InstituteRotterdamthe Netherlands
| | - Dorine T. den Toom
- Department of PathologyUniversity Medical Center Rotterdam, Erasmus MC Cancer InstituteRotterdamthe Netherlands
| | | | - Geert J. L. H. van Leenders
- Department of PathologyUniversity Medical Center Rotterdam, Erasmus MC Cancer InstituteRotterdamthe Netherlands
| | - Paul C. Johannesma
- Department of PulmonologyAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| | | | - Rob M. F. Wolthuis
- Department of Human Genetics, Cancer Center AmsterdamAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| | - Maurice A. M. van Steensel
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore,Singapore Skin Research Institute, Agency for Science, Technology and ResearchSingaporeSingapore
| | - Hendrikus J. Dubbink
- Department of PathologyUniversity Medical Center Rotterdam, Erasmus MC Cancer InstituteRotterdamthe Netherlands
| | - Arjan C. Houweling
- Department of Human GeneticsAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| |
Collapse
|
18
|
Yu Q, Wu Y, Yao L, Jiang R. Upper ureteral calculi complicated with severe hydronephrosis and renal medullary carcinoma in elderly Chinese woman: A case report. Asian J Surg 2022; 46:2166-2167. [PMID: 36450622 DOI: 10.1016/j.asjsur.2022.11.078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Qiwei Yu
- Department of Urology, Kunshan Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Kunshan, Jiangsu Province, 215300, China
| | - Yufan Wu
- Department of Urology, Kunshan Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Kunshan, Jiangsu Province, 215300, China
| | - Linya Yao
- Department of Urology, Kunshan Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Kunshan, Jiangsu Province, 215300, China
| | - Rilei Jiang
- School of Basic Medicine Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
19
|
Fincke VE, Krulik ME, Joshi P, Frühwald MC, Chen YB, Johann PD. Renal Medullary Carcinomas Harbor a Distinct Methylation Phenotype and Display Aberrant Methylation of Genes Related to Early Nephrogenesis. Cancers (Basel) 2022; 14:cancers14205044. [PMID: 36291828 PMCID: PMC9599670 DOI: 10.3390/cancers14205044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
Renal medullary carcinomas (RMC) are rare aggressive tumors of the kidneys, characterized by a loss of SMARCB1. Characteristically, these tumors arise in patients with sickle cell trait or other hemoglobinopathies. Recent characterization efforts have unraveled oncogenic pathways that drive tumorigenesis. Among these, gene sets that characterize replicative stress and the innate immune response are upregulated in RMCs. Despite comprehensive genetic and transcriptomic characterizations, commonalities or differences to other SMARCB1 deficient entities so far have not been investigated. We analyzed the methylome of seven primary RMC and compared it to other SMARCB1 deficient entities such as rhabdoid tumors (RT) and epithelioid sarcomas using 850 K methylation arrays. Moreover, we evaluated the differential gene expression of RMC using RNA-sequencing in comparison to other rhabdoid tumors. In accordance with previous gene expression data, we found that RMCs separate from other SMARCB1 deficient entities, pointing to a potentially different cell of origin and a role of additional genetic aberrations that may drive tumorigenesis and thus alter the methylome when compared to rhabdoid tumors. In a focused analysis of genes that are important for nephrogenesis, we particularly detected genes that govern early nephrogenesis such as FOXI1 to be hypomethylated and expressed at high levels in RMC. Overall, our analyses underscore the fact that RMCs represent a separate entity with limited similarities to rhabdoid tumors, warranting specific treatment tailored to the aggressiveness of the disease.
Collapse
Affiliation(s)
- Victoria E. Fincke
- Swabian Children’s Cancer Center, University Hospital Augsburg, 86156 Augsburg, Germany
- Correspondence: (V.E.F.); (P.D.J.)
| | - Mateja E. Krulik
- Swabian Children’s Cancer Center, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Piyush Joshi
- Hopp Children’s Cancer Center (KiTZ) Heidelberg, Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Michael C. Frühwald
- Swabian Children’s Cancer Center, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Ying-Bei Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Pascal D. Johann
- Swabian Children’s Cancer Center, University Hospital Augsburg, 86156 Augsburg, Germany
- Hopp Children’s Cancer Center (KiTZ) Heidelberg, Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence: (V.E.F.); (P.D.J.)
| |
Collapse
|
20
|
Madanat-Harjuoja LM, Renfro LA, Klega K, Tornwall B, Thorner AR, Nag A, Dix D, Dome JS, Diller LR, Fernandez CV, Mullen EA, Crompton BD. Circulating Tumor DNA as a Biomarker in Patients With Stage III and IV Wilms Tumor: Analysis From a Children's Oncology Group Trial, AREN0533. J Clin Oncol 2022; 40:3047-3056. [PMID: 35580298 PMCID: PMC9462535 DOI: 10.1200/jco.22.00098] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/09/2022] [Accepted: 04/01/2022] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The utility of circulating tumor DNA (ctDNA) analyses has not been established in the risk stratification of Wilms tumor (WT). We evaluated the detection of ctDNA and selected risk markers in the serum and urine of patients with WT and compared findings with those of matched diagnostic tumor samples. PATIENTS AND METHODS Fifty of 395 children with stage III or IV WT enrolled on Children's Oncology Group trial AREN0533 had banked pretreatment serum, urine, and tumor available. Next-generation sequencing was used to detect ctDNA. Copy-number changes in 1q, 16q, and 1p, and single-nucleotide variants in serum and urine were compared with tumor biopsy data. Event-free survival (EFS) was compared between patients with and without ctDNA detection. RESULTS ctDNA was detected in the serum of 41/50 (82%) and in the urine in 13/50 (26%) patients. Agreement between serum ctDNA detection and tumor sequencing results was as follows: 77% for 1q gain, 88% for 16q deletions, and 70% for 1p deletions, with ĸ-coefficients of 0.56, 0.74, and 0.29, respectively. Sequencing also demonstrated that single-nucleotide variants detected in tumors could be identified in the ctDNA. There was a trend toward worse EFS in patients with ctDNA detected in the serum (4-year EFS 80% v 100%, P = .14). CONCLUSION ctDNA demonstrates promise as an easily accessible prognostic biomarker with potential to detect tumor heterogeneity. The observed trend toward more favorable outcome in patients with undetectable ctDNA requires validation. ctDNA profiling should be further explored as a noninvasive diagnostic and prognostic tool in the risk-adapted treatment of patients with WT.
Collapse
Affiliation(s)
| | | | - Kelly Klega
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | - Brett Tornwall
- Children's Oncology Group Statistics and Data Center, Monrovia, CA
| | - Aaron R. Thorner
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA
| | - Anwesha Nag
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA
| | - David Dix
- BC Children's Hospital, Vancouver, BC, Canada
| | - Jeffrey S. Dome
- Children's National Hospital and the George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Lisa R. Diller
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | | | | | - Brian D. Crompton
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| |
Collapse
|
21
|
Onal B, Gultekin MH, Simsekoglu MF, Selcuk B, Gurbuz A. Biomarkers in Urological Cancers. Biomark Med 2022. [DOI: 10.2174/9789815040463122010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Urological tumours have become one of the most common cancers in the
last decade. It is important to apply an approach that evaluates many factors related to
the patient and the disease carefully to minimize cancer-associated morbidity and
mortality. The clinical use of cancer biomarkers is a valuable part of the clinical
management of urological cancers. These biomarkers may lead to optimized detection,
treatment, and follow-up of urological cancers. With the development of molecular
research, newly developed biomarkers and next-generation sequencing have also
contributed to patient management. In this chapter, we will present biomarkers in the
most common urological cancers under subheadings of bladder cancer, prostate cancer,
kidney cancer, and testicular cancer. Additionally, due to the development that
occurred in the next-generation sequencing (NGS), all the above-mentioned
malignancies are evaluated with regard to NGS.
Collapse
Affiliation(s)
- Bulent Onal
- Department of Urology, Cerrahpasa School of Medicine, Istanbul University - Cerrahpasa,
Istanbul, Turkey
| | - Mehmet Hamza Gultekin
- Department of Urology, Cerrahpasa School of Medicine, Istanbul University - Cerrahpasa,
Istanbul, Turkey
| | - Muhammed Fatih Simsekoglu
- Department of Urology, Cerrahpasa School of Medicine, Istanbul University - Cerrahpasa,
Istanbul, Turkey
| | - Berin Selcuk
- Department of Urology, Cerrahpasa School of Medicine, Istanbul University - Cerrahpasa,
Istanbul, Turkey
| | - Ahmet Gurbuz
- Department of Urology, Cerrahpasa School of Medicine, Istanbul University - Cerrahpasa,
Istanbul, Turkey
| |
Collapse
|
22
|
Li Q, Zhang Z, Yin M, Cui C, Zhang Y, Wang Y, Liu F. What do we actually know about exosomal microRNAs in kidney diseases? Front Physiol 2022; 13:941143. [PMID: 36105281 PMCID: PMC9464820 DOI: 10.3389/fphys.2022.941143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
There are several types of kidney diseases with complex causes. If left untreated, these diseases irreversibly progress to end-stage renal disease. Thus, their early diagnosis and targeted treatment are important. Exosomes-extracellular vesicles released by a variety of cells-are ideal carriers for DNA, RNA, proteins, and other metabolites owing to their bilayer membranes. Studies have shown that almost all renal cells can secrete exosomes. While research on exosomal microRNAs in the context of renal diseases begun only recently, rapid progress has been achieved. This review summarizes the changes in exosomal microRNA expression in different kidney diseases. Thus, it highlights the diagnostic and prognostic value of these exosomal microRNAs. Further, this review analyzes their roles in the development of different kidney diseases, guiding research on molecular mechanisms and therapeutic strategies.
Collapse
Affiliation(s)
- Qianyu Li
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, China
| | - Zhiping Zhang
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, China
| | - Min Yin
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, China
| | - Cancan Cui
- Clinical Laboratory, China–Japan Union Hospital of Jilin University, Changchun, China
| | - Yucheng Zhang
- Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, China
| | - Yali Wang
- Department of Blood Transfusion, China–Japan Union Hospital of Jilin University, Changchun, China
| | - Feng Liu
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Funasaki S, Mehanna S, Ma W, Nishizawa H, Kamikubo Y, Sugiyama H, Ikeda S, Motoshima T, Hasumi H, Linehan WM, Schmidt LS, Ricketts C, Suda T, Oike Y, Kamba T, Baba M. Targeting chemoresistance in Xp11.2 translocation renal cell carcinoma using a novel polyamide-chlorambucil conjugate. Cancer Sci 2022; 113:2352-2367. [PMID: 35396773 PMCID: PMC9277412 DOI: 10.1111/cas.15364] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/26/2022] [Accepted: 04/02/2022] [Indexed: 11/29/2022] Open
Abstract
Renal cell carcinoma with Xp11.2 translocation involving the TFE3 gene (TFE3-RCC) is a recently identified subset of RCC with unique morphology and clinical presentation. The chimeric PRCC-TFE3 protein produced by Xp11.2 translocation has been shown to transcriptionally activate its downstream target genes that play important roles in carcinogenesis and tumor development of TFE3-RCC. However, the underlying molecular mechanisms remain poorly understood. Here we show that in TFE3-RCC cells, PRCC-TFE3 controls heme oxygenase 1 (HMOX1) expression to confer chemoresistance. Inhibition of HMOX1 sensitized the PRCC-TFE3 expressing cells to genotoxic reagents. We screened for a novel chlorambucil-polyamide conjugate (Chb) to target PRCC-TFE3-dependent transcription, and identified Chb16 as a PRCC-TFE3-dependent transcriptional inhibitor of HMOX1 expression. Treatment of the patient-derived cancer cells with Chb16 exhibited senescence and growth arrest, and increased sensitivity of the TFE3-RCC cells to the genotoxic reagent etoposide. Thus, our data showed that the TFE3-RCC cells acquired chemoresistance through HMOX1 expression and that inhibition of HMOX1 by Chb16 may be an effective therapeutic strategy for TFE3-RCC.
Collapse
Affiliation(s)
- Shintaro Funasaki
- Laboratory of Cancer MetabolismInternational Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
| | - Sally Mehanna
- Laboratory of Cancer MetabolismInternational Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
| | - Wenjuan Ma
- Laboratory of Cancer MetabolismInternational Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
| | - Hidekazu Nishizawa
- Laboratory of Cancer MetabolismInternational Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
- Department of UrologyGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yasuhiko Kamikubo
- Department of Human Health ScienceGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Hiroshi Sugiyama
- Department of ChemistryGraduate School of ScienceKyoto UniversityKyotoJapan
| | - Shuji Ikeda
- Department of ChemistryGraduate School of ScienceKyoto UniversityKyotoJapan
| | - Takanobu Motoshima
- Department of UrologyGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Hisashi Hasumi
- Department of UrologyGraduate School of MedicineYokohama City UniversityYokohamaJapan
| | - W. Marston Linehan
- Urologic Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Laura S. Schmidt
- Urologic Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
- Basic Science ProgramFrederick National Laboratory for Cancer ResearchNational Cancer InstituteFrederickMarylandUSA
| | - Chris Ricketts
- Urologic Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Toshio Suda
- Laboratory of Stem Cell RegulationInternational Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
- Cancer Science Institute of SingaporeCentre for Translational MedicineNational University of SingaporeSingapore CitySingapore
| | - Yuichi Oike
- Department of Molecular GeneticsGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Tomomi Kamba
- Department of UrologyGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Masaya Baba
- Laboratory of Cancer MetabolismInternational Research Center for Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
| |
Collapse
|
24
|
Iqbal M. Renal Cell Carcinoma: A Complex Therapeutic Challenge in the Elderly. Cureus 2022; 14:e26346. [PMID: 35903561 PMCID: PMC9321336 DOI: 10.7759/cureus.26346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2022] [Indexed: 01/20/2023] Open
Abstract
An increase in age and the occurrence of renal cell carcinoma have been positively correlated. A strict therapeutic protocol for early diagnosis, screening, prevention, and population awareness needs to be well-established as a rationale to approach the morbidity at a treatment-eligible phase in the aged. Genetic predisposition appears to have a minor role in the disease pathology. Imaging modalities, providing high-resolution images of the tumor, have undoubtedly benefitted the diseased subset in aiding the diagnosis, however, a preliminary guideline protocol for its early implication in concordance with the initial symptoms needs to be adopted. Burdening of the geriatric age group by concomitant co-morbidities further deteriorates the devastating effects of the primary tumor, which, in total, appear to evolve as a final, complex stage of the illness in the majority, leading to an eventual high mortality rate. Despite being a challenging task for managing the tumor, age should not be considered the sole treatment barrier for approaching the disease.
Collapse
|
25
|
Beck P, Selle B, Madenach L, Jones DTW, Vokuhl C, Gopisetty A, Nabbi A, Brecht IB, Ebinger M, Wegert J, Graf N, Gessler M, Pfister SM, Jäger N. The genomic landscape of pediatric renal cell carcinomas. iScience 2022; 25:104167. [PMID: 35445187 PMCID: PMC9014386 DOI: 10.1016/j.isci.2022.104167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/03/2022] [Accepted: 03/24/2022] [Indexed: 12/08/2022] Open
Abstract
Pediatric renal cell carcinomas (RCC) differ from their adult counterparts not only in histologic subtypes but also in clinical characteristics and outcome. However, the underlying biology is still largely unclear. For this reason, we performed whole-exome and transcriptome sequencing analyses on a cohort of 25 pediatric RCC patients with various histologic subtypes, including 10 MiT family translocation (MiT) and 10 papillary RCCs. In this cohort of pediatric RCC, we find only limited genomic overlap with adult RCC, even within the same histologic subtype. Recurrent somatic mutations in genes not previously reported in RCC were detected, such as in CCDC168, PLEKHA1, VWF, and MAP3K9. Our papillary pediatric RCCs, which represent the largest cohort to date with comprehensive molecular profiling in this age group, appeared as a distinct genomic subtype differing in terms of gene mutations and gene expression patterns not only from MiT-RCC but also from their adult counterparts. WES and RNA-seq of 25 pediatric RCCs with various histologic subtypes Detected only limited genomic overlap with adult RCC Revealed recurrent somatic mutations in genes not previously reported in RCC Discovery of a CRK-PITPNA fusion gene in a pediatric papillary RCC
Collapse
Affiliation(s)
- Pengbo Beck
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Barbara Selle
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Lukas Madenach
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Pediatric Glioma Research Group, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Vokuhl
- Section of Pediatric Pathology, Department of Pathology, University Hospital Bonn, Bonn, Germany
| | - Apurva Gopisetty
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Arash Nabbi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Ines B Brecht
- Department of Pediatric Oncology and Hematology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Martin Ebinger
- Department of Pediatric Oncology and Hematology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Jenny Wegert
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Würzburg University & Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Saarland University, Homburg, Germany
| | - Manfred Gessler
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Würzburg University & Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Natalie Jäger
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
26
|
Denize T, Massa S, Valent A, Militti L, Bertolotti A, Barisella M, Rioux-Leclercq N, Malouf GG, Spreafico F, Verschuur A, van der Beek J, Tytgat L, van den Heuvel-Eibrink MM, Vujanic G, Collini P, Coulomb A. Renal cell carcinoma in children and adolescents: A retrospective study of a French-Italian series of 93 cases. Histopathology 2022; 80:928-945. [PMID: 35238063 DOI: 10.1111/his.14634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/06/2022] [Accepted: 02/23/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Renal cell carcinomas represent 2 to 5% of kidney malignancies in children and adolescents. Appropriate diagnostic and classification are crucial for the correct management of the patients and in order to avoid inappropriate preoperative chemotherapy, which is usually recommended if a Wilms tumor is suspected. METHODS a French-Italian series of 93 renal cell carcinomas collected from 1990 to 2019 in patients aged less than 18 years old was reclassified according to the 2016 WHO classification and the latest literature. TFE3 and TFEB FISH analyses and a panel of immunohistochemical stains were applied. RESULTS The median age at diagnosis was 11 years (range: 9 months - 17 years). MiT family (MiTF) translocation renal cell carcinomas accounted for 52% of the tumors, followed by papillary renal cell carcinomas (20%) and unclassified renal cell carcinomas (13%). Other subtypes, such as SDHB-deficient and Fumarate hydratase-deficient renal cell carcinomas, represented 1 to 3% of the cases. We also described a case of ALK-rearranged renal cell carcinoma with a metanephric adenoma-like morphology. CONCLUSION A precise histological diagnosis is mandatory as targeted therapy could be applied for some RCC subtypes, i.e., MiTF-translocation and ALK-translocation renal cell carcinomas. Moreover, some RCC subtypes may be associated with a predisposition syndrome that will impact patients' and family's management and genetic counseling. A precise RCC subtype is also mandatory for the clinical management of the patients and the inclusion in new prospective clinical trials.
Collapse
Affiliation(s)
- Thomas Denize
- Department of Pathology, Sorbonne Université, Assistance Publique Hôpitaux de Paris - Hôpital Armand Trousseau, Paris, France
| | - Simona Massa
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,present address: Unit of Pathology, Azienda Ospedaliera Specialistica dei Colli Monaldi-Cotugno-CTO, Naples, Italy
| | - Alexander Valent
- Service de Génétique des tumeurs, Département de Pathologie, Institut Gustave Roussy, Villejuif, France
| | - Lucia Militti
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessia Bertolotti
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marta Barisella
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Gabriel G Malouf
- Service d'Oncologie Médicale, Institut de Cancérologie de Strasbourg, Strasbourg, France
| | - Filippo Spreafico
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Arnauld Verschuur
- Department of Pediatric Oncology, Hôpital d'enfants de la Timone, Marseille, France
| | - Justine van der Beek
- Princess Máxima Center for Pediatric Oncology, and Utrecht University, Utrecht, The Netherlands
| | - Lieve Tytgat
- Princess Máxima Center for Pediatric Oncology, and Utrecht University, Utrecht, The Netherlands
| | | | - Gordan Vujanic
- Department of Pathology, Sidra Medicine / Weill Cornell Medicine, Doha, Qatar.,Sorbonne Université, Assistance Publique Hôpitaux de Paris - Hôpital Armand Trousseau, Paris, France
| | - Paola Collini
- Soft Tissue and Bone Pathology and Pediatric Pathology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Aurore Coulomb
- Department of Pathology, Sorbonne Université, Assistance Publique Hôpitaux de Paris - Hôpital Armand Trousseau, Paris, France
| |
Collapse
|
27
|
Extracellular Vesicles: New Tools for Early Diagnosis of Breast and Genitourinary Cancers. Int J Mol Sci 2021; 22:ijms22168430. [PMID: 34445131 PMCID: PMC8395117 DOI: 10.3390/ijms22168430] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancers and cancers of the genitourinary tract are the most common malignancies among men and women and are still characterized by high mortality rates. In order to improve the outcomes, early diagnosis is crucial, ideally by applying non-invasive and specific biomarkers. A key role in this field is played by extracellular vesicles (EVs), lipid bilayer-delimited structures shed from the surface of almost all cell types, including cancer cells. Subcellular structures contained in EVs such as nucleic acids, proteins, and lipids can be isolated and exploited as biomarkers, since they directly stem from parental cells. Furthermore, it is becoming even more evident that different body fluids can also serve as sources of EVs for diagnostic purposes. In this review, EV isolation and characterization methods are described. Moreover, the potential contribution of EV cargo for diagnostic discovery purposes is described for each tumor.
Collapse
|
28
|
van der Beek JN, Hol JA, Coulomb‐l'Hermine A, Graf N, van Tinteren H, Pritchard‐Jones K, Houwing ME, de Krijger RR, Vujanic GM, Dzhuma K, Schenk J, Littooij AS, Ramírez‐Villar GL, Murphy D, Ray S, Al‐Saadi R, Gessler M, Godzinski J, Ruebe C, Collini P, Verschuur AC, Frisk T, Vokuhl C, Hulsbergen‐van de Kaa CA, de Camargo B, Sandstedt B, Selle B, Tytgat GAM, van den Heuvel‐Eibrink MM. Characteristics and outcome of pediatric renal cell carcinoma patients registered in the International Society of Pediatric Oncology (SIOP) 93-01, 2001 and UK-IMPORT database: A report of the SIOP-Renal Tumor Study Group. Int J Cancer 2021; 148:2724-2735. [PMID: 33460450 PMCID: PMC8048605 DOI: 10.1002/ijc.33476] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 01/16/2023]
Abstract
In children, renal cell carcinoma (RCC) is rare. This study is the first report of pediatric patients with RCC registered by the International Society of Pediatric Oncology-Renal Tumor Study Group (SIOP-RTSG). Pediatric patients with histologically confirmed RCC, registered in SIOP 93-01, 2001 and UK-IMPORT databases, were included. Event-free survival (EFS) and overall survival (OS) were analyzed using the Kaplan-Meier method. Between 1993 and 2019, 122 pediatric patients with RCC were registered. Available detailed data (n = 111) revealed 56 localized, 30 regionally advanced, 25 metastatic and no bilateral cases. Histological classification according to World Health Organization 2004, including immunohistochemical and molecular testing for transcription factor E3 (TFE3) and/or EB (TFEB) translocation, was available for 65/122 patients. In this group, the most common histological subtypes were translocation type RCC (MiT-RCC) (36/64, 56.3%), papillary type (19/64, 29.7%) and clear cell type (4/64, 6.3%). One histological subtype was not reported. In the remaining 57 patients, translocation testing could not be performed, or TFE-cytogenetics and/or immunohistochemistry results were missing. In this group, the most common RCC histological subtypes were papillary type (21/47, 44.7%) and clear cell type (11/47, 23.4%). Ten histological subtypes were not reported. Estimated 5-year (5y) EFS and 5y OS of the total group was 70.5% (95% CI = 61.7%-80.6%) and 84.5% (95% CI = 77.5%-92.2%), respectively. Estimated 5y OS for localized, regionally advanced, and metastatic disease was 96.8%, 92.3%, and 45.6%, respectively. In conclusion, the registered pediatric patients with RCC showed a reasonable outcome. Survival was substantially lower for patients with metastatic disease. This descriptive study stresses the importance of full, prospective registration including TFE-testing.
Collapse
Affiliation(s)
- Justine N. van der Beek
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Department of Radiology and Nuclear MedicineUniversity Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht UniversityUtrechtThe Netherlands
| | - Janna A. Hol
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | - Norbert Graf
- Department of Pediatric Oncology and HematologySaarland University Medical Center and Saarland University Faculty of MedicineHomburgGermany
| | | | | | - Maite E. Houwing
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Ronald R. de Krijger
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Kristina Dzhuma
- UCL Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Jens‐Peter Schenk
- Clinic of Diagnostic and Interventional Radiology, Division of Pediatric RadiologyHeidelberg University HospitalHeidelbergGermany
| | - Annemieke S. Littooij
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Department of Radiology and Nuclear MedicineUniversity Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht UniversityUtrechtThe Netherlands
| | | | - Dermot Murphy
- Department of Paediatric OncologyRoyal Hospital for ChildrenGlasgowScotland
| | - Satyajit Ray
- Department of Paediatric OncologyRoyal Hospital for ChildrenGlasgowScotland
| | - Reem Al‐Saadi
- UCL Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
- Department of HistopathologyGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Manfred Gessler
- Theodor‐Boveri‐Institute/BiocenterUniversity of WuerzburgWuerzburgGermany
| | - Jan Godzinski
- Department of Paediatric SurgeryMarciniak HospitalWroclawPoland
- Department of Paediatric Traumatology and Emergency MedicineMarciniak HospitalWroclawPoland
| | - Christian Ruebe
- Department of Radiation OncologySaarland University Medical Center and Saarland University Faculty of MedicineHomburgGermany
| | - Paola Collini
- Department of PathologyFondazione IRCCS Istituto Nazionale dei TumoriMilanoItaly
| | - Arnaud C. Verschuur
- Department of Pediatric OncologyHôpital d'Enfants de la TimoneMarseilleFrance
| | - Tony Frisk
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
| | - Christian Vokuhl
- Section of Pediatric PathologyUniversity Hospital BonnBonnGermany
| | | | | | - Bengt Sandstedt
- Childhood Cancer Research UnitAstrid Lindgren's Children's Hospital, Karolinska InstitutetStockholmSweden
| | - Barbara Selle
- Department of Pediatric Hematology and OncologySt. Annastift Children's HospitalLudwigshafenGermany
| | | | | |
Collapse
|
29
|
Zoumpourlis P, Genovese G, Tannir NM, Msaouel P. Systemic Therapies for the Management of Non-Clear Cell Renal Cell Carcinoma: What Works, What Doesn't, and What the Future Holds. Clin Genitourin Cancer 2021; 19:103-116. [PMID: 33358151 PMCID: PMC8169717 DOI: 10.1016/j.clgc.2020.11.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/31/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023]
Abstract
Non-clear cell renal cell carcinoma (nccRCC) is a broad term that refers to a diverse group of tumors, each with its own distinct biologic and therapeutic profile. The management of nccRCCs is often based on extrapolating data from clinical trials in the more common clear cell renal cell carcinoma, but our emerging prospective and retrospective clinical experience in nccRCC allows us to make more precise recommendations tailored to each histology. The systemic therapy options for metastatic nccRCC include targeted therapies such as tyrosine kinase inhibitors, immune checkpoint inhibitors, and, for specific rare subtypes, cytotoxic chemotherapy. Each nccRCC histology may respond differently to these regimens, which makes accurate pathologic diagnosis imperative. In the present review, we discuss the available clinical and biological data that can help guide systemic therapy recommendations for specific nccRCC subtypes.
Collapse
Affiliation(s)
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX; Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
30
|
Lang M, Vocke CD, Ricketts CJ, Metwalli AR, Ball MW, Schmidt LS, Linehan WM. Clinical and Molecular Characterization of Microphthalmia-associated Transcription Factor (MITF)-related Renal Cell Carcinoma. Urology 2021; 149:89-97. [PMID: 33242557 PMCID: PMC8728951 DOI: 10.1016/j.urology.2020.11.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVES To characterize the clinical presentation, genomic alterations, pathologic phenotype and clinical management of microphthalmia-associated transcription factor (MITF) familial renal cell carcinoma (RCC), caused by a member of the TFE3, TFEB, and MITF family of transcription factor genes. METHODS The clinical presentation, family history, tumor histopathology, and surgical management were evaluated and reported herein. DNA sequencing was performed on blood DNA, tumor DNA and DNA extracted from adjacent normal kidney tissue. Copy number and gene expression analyses on tumor and normal tissues were performed by Real-Time Polymerase chain reaction. TCGA gene expression data were used for comparative analysis. Protein expression and subcellular localization were evaluated by immunohistochemistry. RESULTS Germline genomic analysis identified the MITF p.E318K variant in a patient with bilateral, multifocal type 1 papillary RCC and a family history of RCC. All tumors displayed the MITF variant and were characterized by amplification of chromosomes 7 and 17, hallmarks of type 1 papillary RCC. We demonstrated that MITF p.E318K variant results in altered transcriptional activity and that downstream targets of MiT family members, such as GPNMB, are dysregulated in the tumors. CONCLUSION Association of the pathogenic MITF variant with bilateral and multifocal type 1 papillary RCC in this family supports its role as a risk allele for the development of RCC and emphasizes the importance of screening for MITF variants irrelevant of the RCC histologic subtype. This study identifies potential biomarkers for the disease, such as GPNMB expression, that may facilitate the development of targeted therapies for patients affected with MITF-associated RCC.
Collapse
Affiliation(s)
- Martin Lang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Cathy D Vocke
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christopher J Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Adam R Metwalli
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mark W Ball
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Laura S Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD; Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - William M Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Pediatric renal tumors account for 7% of new cancer diagnoses in children. Here, we will review results from recently completed clinical trials informing the current standard of care and discuss targeted and immune therapies being explored for the treatment of high risk or relapsed/refractory pediatric renal malignancies. RECENT FINDINGS Cooperative group trials have continued to make improvements in the care of children with pediatric tumors. In particular, trials that standardize treatment of rare cancers (e.g., bilateral Wilms tumor) have improved outcomes significantly. We have seen improvements in event free and overall survival in recently completed clinical trials for many pediatric renal tumors. Still, there are subsets of rarer cancers where outcomes remain poor and new therapeutic strategies are needed. Future trials aim to balance treatment toxicity with treatment efficacy for those with excellent outcomes while identifying novel therapeutics for those with poor outcomes.
Collapse
Affiliation(s)
- Juhi Jain
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, USA.,Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA.,Emory Children's Center, 2015 Uppergate Drive NE , 400, Atlanta, GA, 30322, USA
| | - Kathryn S Sutton
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, USA.,Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA.,Emory Children's Center, 2015 Uppergate Drive NE, 434B, Atlanta, GA, 30322, USA
| | - Andrew L Hong
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, USA. .,Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA. .,Winship Cancer Institute, Atlanta, GA, USA. .,Health Sciences Research Building, 1760 Haygood Drive NE, E-370, Atlanta, GA, 30322, USA.
| |
Collapse
|
32
|
Hol JA, Jongmans MCJ, Littooij AS, de Krijger RR, Kuiper RP, van Harssel JJT, Mensenkamp A, Simons M, Tytgat GAM, van den Heuvel-Eibrink MM, van Grotel M. Renal cell carcinoma in young FH mutation carriers: case series and review of the literature. Fam Cancer 2021; 19:55-63. [PMID: 31792767 PMCID: PMC7026215 DOI: 10.1007/s10689-019-00155-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hereditary Leiomyomatosis and Renal Cell Cancer (HLRCC) is an autosomal dominant syndrome caused by heterozygous pathogenic germline variants in the fumarate hydratase (FH) gene. It is characterized by cutaneous and uterine leiomyomas and an increased risk of developing renal cell carcinoma (RCC), which is usually adult-onset. HLRCC-related RCC tends to be aggressive and can metastasize even when the primary tumor is small. Data on children and adolescents are scarce. Herein, we report two patients from unrelated Dutch families, with HLRCC-related RCC at the ages of 15 and 18 years, and a third patient with an FH mutation and complex renal cysts at the age of 13. Both RCC’s were localized and successfully resected, and careful MRI surveillance was initiated to monitor the renal cysts. One of the patients with RCC subsequently developed an ovarian Leydig cell tumor. A review of the literature identified 10 previously reported cases of HLRCC-related RCC in patients aged younger than 20 years, five of them presenting with metastatic disease. These data emphasize the importance of recognizing HLRCC in young patients to enable early detection of RCC, albeit rare. They support the recommendations from the 2014 consensus guideline, in which genetic testing for FH mutations, and renal MRI surveillance, is advised for HLRCC family members from the age of 8–10 years onwards.
Collapse
Affiliation(s)
- J A Hol
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.
| | - M C J Jongmans
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.,Department of Genetics, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - A S Littooij
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.,Department of Radiology, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - R R de Krijger
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.,Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R P Kuiper
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - J J T van Harssel
- Department of Genetics, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - A Mensenkamp
- Department of Genetics, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - M Simons
- Department of Pathology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - G A M Tytgat
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | | | - M van Grotel
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| |
Collapse
|
33
|
Akgul M, Williamson SR, Ertoy D, Argani P, Gupta S, Caliò A, Reuter V, Tickoo S, Al-Ahmadie HA, Netto GJ, Hes O, Hirsch MS, Delahunt B, Mehra R, Skala S, Osunkoya AO, Harik L, Rao P, Sangoi AR, Nourieh M, Zynger DL, Smith SC, Nazeer T, Gumuskaya B, Kulac I, Khani F, Tretiakova MS, Vakar-Lopez F, Barkan G, Molinié V, Verkarre V, Rao Q, Kis L, Panizo A, Farzaneh T, Magers MJ, Sanfrancesco J, Perrino C, Gondim D, Araneta R, So JS, Ro JY, Wasco M, Hameed O, Lopez-Beltran A, Samaratunga H, Wobker SE, Melamed J, Cheng L, Idrees MT. Diagnostic approach in TFE3-rearranged renal cell carcinoma: a multi-institutional international survey. J Clin Pathol 2021; 74:291-299. [PMID: 33514585 DOI: 10.1136/jclinpath-2020-207372] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 01/07/2021] [Indexed: 12/19/2022]
Abstract
Transcription factor E3-rearranged renal cell carcinoma (TFE3-RCC) has heterogenous morphologic and immunohistochemical (IHC) features.131 pathologists with genitourinary expertise were invited in an online survey containing 23 questions assessing their experience on TFE3-RCC diagnostic work-up.Fifty (38%) participants completed the survey. 46 of 50 participants reported multiple patterns, most commonly papillary pattern (almost always 9/46, 19.5%; frequently 29/46, 63%). Large epithelioid cells with abundant cytoplasm were the most encountered cytologic feature, with either clear (almost always 10/50, 20%; frequently 34/50, 68%) or eosinophilic (almost always 4/49, 8%; frequently 28/49, 57%) cytology. Strong (3+) or diffuse (>75% of tumour cells) nuclear TFE3 IHC expression was considered diagnostic by 13/46 (28%) and 12/47 (26%) participants, respectively. Main TFE3 IHC issues were the low specificity (16/42, 38%), unreliable staining performance (15/42, 36%) and background staining (12/42, 29%). Most preferred IHC assays other than TFE3, cathepsin K and pancytokeratin were melan A (44/50, 88%), HMB45 (43/50, 86%), carbonic anhydrase IX (41/50, 82%) and CK7 (32/50, 64%). Cut-off for positive TFE3 fluorescent in situ hybridisation (FISH) was preferably 10% (9/50, 18%), although significant variation in cut-off values was present. 23/48 (48%) participants required TFE3 FISH testing to confirm TFE3-RCC regardless of the histomorphologic and IHC assessment. 28/50 (56%) participants would request additional molecular studies other than FISH assay in selected cases, whereas 3/50 participants use additional molecular cases in all cases when TFE3-RCC is in the differential.Optimal diagnostic approach on TFE3-RCC is impacted by IHC and/or FISH assay preferences as well as their conflicting interpretation methods.
Collapse
Affiliation(s)
- Mahmut Akgul
- Pathology, Albany Medical Center, Albany, New York, USA
| | - Sean R Williamson
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Dilek Ertoy
- Department of Pathology, Koc University School of Medicine, Istanbul, Turkey
| | - Pedram Argani
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Sounak Gupta
- Department of Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Anna Caliò
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Veneto, Italy
| | - Victor Reuter
- Pathology, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Satish Tickoo
- Pathology, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Hikmat A Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - George J Netto
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ondrej Hes
- Faculty of Medicine in Plzen, Charles University, Plzen, Czech Republic.,Medical Teaching School, University Hospital, Charles University, Plzen, Czech Republic
| | - Michelle S Hirsch
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Brett Delahunt
- Pathology and Molecular Medicine, Wellington School of Medicine and Health Sciences, Wellington South, New Zealand
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephanie Skala
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Adeboye O Osunkoya
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lara Harik
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Priya Rao
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ankur R Sangoi
- Department of Pathology, El Camino Hospital, Mountain View, California, USA
| | - Maya Nourieh
- Department of Pathology, Institut Curie, Paris, France
| | - Debra L Zynger
- Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio, USA
| | - Steven Cristopher Smith
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Tipu Nazeer
- Pathology, Albany Medical Center, Albany, New York, USA
| | - Berrak Gumuskaya
- Department of Pathology, Yildirim Beyazit University School of Medicine, Ankara, Turkey
| | - Ibrahim Kulac
- Department of Pathology, Koc University School of Medicine, Istanbul, Turkey
| | - Francesca Khani
- Department of Pathology, Cornell University Joan and Sanford I Weill Medical College, New York City, New York, USA
| | - Maria S Tretiakova
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Funda Vakar-Lopez
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Guliz Barkan
- Department of Pathology, Loyola University Health System, Maywood, Illinois, USA
| | - Vincent Molinié
- Pathology, University Hospital Center of Martinique, Fort-de-France, Martinique
| | - Virginie Verkarre
- Department of Pathology, Hôpital Européen Georges Pompidou Anatomie Pathologie, Paris, Île-de-France, France
| | - Qiu Rao
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Lorand Kis
- Department of Pathology, Karolinska Institute, Stockholm, Sweden
| | - Angel Panizo
- Department of Pathology, Complejo Hospitalario de Navarra Servicio de Cardiologia, Pamplona, Navarra, Spain
| | - Ted Farzaneh
- Department of Pathology, Univer Irvine Healthcare, Orange County, California, USA
| | - Martin J Magers
- IHA Pathology and Laboratory Medicine, Ann Arbor, Michigan, USA
| | - Joseph Sanfrancesco
- Department of Pathology, Charleston Area Medical Center, Charleston, South Carolina, USA
| | - Carmen Perrino
- Department of Pathology, Mount Auburn Hospital, Cambridge, Massachusetts, USA
| | - Dibson Gondim
- Department of Pathology, University of Louisville, Louisville, Kentucky, USA
| | - Ronald Araneta
- Department of Pathology, Hartford Hospital, Hartford, Connecticut, USA
| | - Jeffrey S So
- Department of Pathology, St Luke's Hospital, Manila, Philippines
| | - Jae Y Ro
- Department of Pathology, Houston Methodist Hospital, Houston, Texas, USA
| | - Matthew Wasco
- Department of Pathology, St Joseph Mercy Hospital, Ann Arbor, Michigan, USA
| | - Omar Hameed
- Forward Pathology Solutions, Vanderbilt University, Kansas City, Montana, USA
| | - Antonio Lopez-Beltran
- Department of Pathology and Surgery, Cordoba University Medical School, Cordoba, Spain
| | | | - Sara E Wobker
- Department of Pathology, University of North Carolina System, Chapel Hill, North Carolina, USA
| | - Jonathan Melamed
- Department of Pathology, New York University Langone Medical Center, New York City, New York, USA
| | - Liang Cheng
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Muhammad T Idrees
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
34
|
He M, Cai J, Zhu K, Gu W, Li M, Xiong J, Guan Z, Wang J, Shu Q. Renal cell carcinoma in children and adolescents: Single-center experience and literature review. Medicine (Baltimore) 2021; 100:e23717. [PMID: 33466124 PMCID: PMC7808530 DOI: 10.1097/md.0000000000023717] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 11/16/2020] [Indexed: 01/05/2023] Open
Abstract
Renal cell carcinoma (RCC) is infrequent in the pediatric population. In addition, till date, only a few reports have summarized the characteristics of pediatric RCC and differences between pediatric and adult RCC. Therefore, the current study aimed to investigate the clinical characteristics of RCC in children and adolescents, and identify the differences between children and adolescent patients and adult patients through literature retrieval.The data of 13 pediatric patients diagnosed with RCC at the Children's Hospital of Zhejiang University School of Medicine between 2005 and 2019 were retrospectively analyzed.Three patients were aged <5 years, 2 were aged 6 to 10 years, and 8 were aged 11 to 18 years. Among the 13 patients, common clinical manifestations included abdominal pain in 5 patients, gross hematuria in 4, and an abdominal mass in 1, while the other 3 patients were incidentally detected after an abdominal contusion. The pathological types were microphthalmia family translocation RCC in 9 patients, clear-cell RCC in 2, papillary RCC in 1, and unclassified in 1. All the children underwent radical nephrectomy, including 2 patients with advanced disease who underwent preoperative transcatheter arterial chemoembolization. The mean follow-up time was 58.6 months. Two patients died after 4 and 17 months of follow-up, respectively.In conclusion, microphthalmia family translocation renal cell carcinoma is the predominant type of pediatric RCC associated with advanced tumor stage. The early diagnosis and treatment of pediatric patients is important for improving prognosis. Nevertheless, future studies are urgently needed to determine the treatment for pediatric advanced RCC to increase the survival rate.
Collapse
Affiliation(s)
- Min He
- Department of Surgical Oncology
| | | | - Kun Zhu
- Department of Pathology, The Children's Hospital, Zhejiang University School Of Medicine, National Clinical Research Center For Child Health, Hangzhou, China
| | - Weizhong Gu
- Department of Pathology, The Children's Hospital, Zhejiang University School Of Medicine, National Clinical Research Center For Child Health, Hangzhou, China
| | | | | | | | | | | |
Collapse
|
35
|
Koetter P, Martin K. Management of renal cell carcinoma presenting during teenage pregnancy. JOURNAL OF PEDIATRIC SURGERY CASE REPORTS 2020. [DOI: 10.1016/j.epsc.2020.101664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
36
|
Zhu Y, Liu N, Guo W, Pu X, Guo H, Gan W, Li D. ALK rearrangement in TFE3-positive renal cell carcinoma: Alternative diagnostic option to exclude Xp11.2 translocation carcinoma. Pathol Res Pract 2020; 216:153286. [PMID: 33197836 DOI: 10.1016/j.prp.2020.153286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/18/2022]
Abstract
Anaplastic lymphoma kinase (ALK)-rearranged renal cell carcinoma (RCC) is a rare subtype of RCC with gene fusion involving ALK at 2p23. It was first included in the renal tumor classification system by WorldHealth organization (WHO) as a distinct emerging/provisional renal entity in 2016. To date, only a few cases of ALK-RCC have been reported. Here, we report an exceptional case of ALK-RCC in a 15-year-old girl and review the literature. The patient presented with gross hematuria and a tumor measured 7 cm × 6 cm was found in the left kidney by imaging examination. Then a laparoscopic radical nephrectomy combined with local lymph node dissection was performed. The pathologic stage of the tumor was pT1bN1Mx and postoperative pathology showed that the tumor corresponded to WHO/ISUP grade 3-4. Immunohistochemistry (IHC) demonstrated moderate nuclear expression of TFE3 protein. Interestingly, ALK gene rearrangement rather than TFE3 gene rearrangement was observed by fluorescence in situ hybridization (FISH). Now the girl is still alive without evidence of recurrence for 10 months follow-up. In conclusion, the positive expression of nuclear TFE3 in immunohistochemistry may be deceptive, the detection of ALK could be a diagnostic option if TFE3 was negative in FISH study. Large-scale and long-term studies are still needed to explore the biological behavior and molecular characteristic of ALK-RCC.
Collapse
Affiliation(s)
- Yiqi Zhu
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Ning Liu
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wei Guo
- Department of Urology, Drum Tower Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China; Department of Urology, Jiangsu Taizhou People's Hospital, Taizhou, Jiangsu, China
| | - Xiaohong Pu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Weidong Gan
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.
| | - Dongmei Li
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
37
|
Abstract
Molecular characterization has led to advances in the understanding of pediatric renal tumors, including the association of pediatric cystic nephromas with DICER1 tumor syndrome, the metanephric family of tumors with somatic BRAF mutations, the characterization of ETV6-NTRK3-negative congenital mesoblastic nephromas, the expanded spectrum of gene fusions in translocation renal cell carcinoma, the relationship of clear cell sarcoma of the kidney with other BCOR-altered tumors, and the pathways affected by SMARCB1 alterations in rhabdoid tumors of the kidney. These advances have implications for diagnosis, classification, and treatment of pediatric renal tumors.
Collapse
|
38
|
Geller JI, Cost NG, Chi YY, Tornwall B, Cajaiba M, Perlman EJ, Kim Y, Mullen EA, Glick RD, Khanna G, Daw NC, Ehrlich P, Fernandez CV, Dome JS. A prospective study of pediatric and adolescent renal cell carcinoma: A report from the Children's Oncology Group AREN0321 study. Cancer 2020; 126:5156-5164. [PMID: 32926409 DOI: 10.1002/cncr.33173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/06/2020] [Accepted: 07/17/2020] [Indexed: 11/07/2022]
Abstract
BACKGROUND To the authors' knowledge, AREN0321 is the first prospective clinical study of pediatric and adolescent renal cell carcinoma (RCC). Goals of the study included establishing epidemiological, treatment, and outcome data and confirming that patients with completely resected pediatric RCC, including lymph node-positive disease (N1), have a favorable prognosis without adjuvant therapy. METHODS From 2006 to 2012, patients aged <30 years with centrally reviewed pathology of RCC were enrolled prospectively. RESULTS A total of 68 patients were enrolled (39 of whom were male; median age of 13 years [range, 0.17-22.1 years]). Stage was classified according to the American Joint Committee on Cancer TNM stage seventh edition as stage I in 26 patients, stage II in 7 patients, stage III in 26 patients, and stage IV in 8 patients, and was not available in 1 patient. Sixty patients underwent resection of all known sites of disease, including 2 patients with stage IV disease. Surgery included radical nephrectomy (53 patients [81.5%]), partial nephrectomy (12 patients [18.5%]), and unknown (3 patients [4.4%]). Histology was TFE-associated RCC (translocation-type RCC; tRCC) in 40 patients, RCC not otherwise specified and/or other in 13 patients, papillary RCC in 9 patients, and renal medullary carcinoma (RMC) in 6 patients. Lymph node status was N0 in 21 patients, N1 in 21 patients (tRCC in 15 patients, RMC in 3 patients, papillary RCC in 2 patients, and not otherwise specified and/or other in 1 patient), and Nx in 26 patients. The 4-year event-free survival and overall survival rates were 80.2% (95% CI, 69.6%-90.9%) and 84.8% (95% CI, 75.2%-94.5%), respectively, overall and 87.5% (95% CI, 68.3%-100%) and 87.1% (95% CI, 67.6%-100%), respectively, for the 16 patients with N1M0 disease. Among patients presenting with metastases, 2 of 8 patients (2 of 5 patients with RMC) were alive (1 with disease) at the time of last follow-up, including 1 patient who was lost to follow-up (succinate dehydrogenase deficiency). The predominant RCC subtypes associated with mortality were tRCC and RMC. CONCLUSIONS Favorable short-term outcomes can be achieved without adjuvant therapy in children and adolescents with completely resected RCC, independent of lymph node status. A prospective study of patients with tRCC and RMC with M1 or recurrent disease is needed to optimize treatment.
Collapse
Affiliation(s)
- James I Geller
- Division of Pediatric Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Nicholas G Cost
- Division of Urology, Department of Surgery, University of Colorado School of Medicine, the Children's Hospital Colorado, Aurora, Colorado
| | - Yueh-Yun Chi
- Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, Florida
| | - Brett Tornwall
- Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, Florida
| | - Mariana Cajaiba
- Department of Pathology, Anne and Robert H. Lurie Children's Hospital, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Elizabeth J Perlman
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Yeonil Kim
- Biostatistics and Research Decision Sciences, Merck Research Laboratories, Merck & Company Inc, Rahway, New Jersey
| | - Elizabeth A Mullen
- Department of Pediatric Oncology, Dana-Farber Cancer Institute/Boston Children's Hospital, Boston, Massachusetts
| | - Richard D Glick
- Division of Pediatric Surgery, Steven and Alexandra Cohen Medical Center of New York, New York, New York
| | - Geetika Khanna
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Najat C Daw
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Peter Ehrlich
- Section of Pediatric Surgery, Department of Surgery, C.S. Mott Children's Hospital, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Conrad V Fernandez
- Division of Pediatric Oncology, IWK Health Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jeffrey S Dome
- Division of Pediatric Oncology, Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC
| | | |
Collapse
|
39
|
Marcon J, DiNatale RG, Sanchez A, Kotecha RR, Gupta S, Kuo F, Makarov V, Sandhu A, Mano R, Silagy AW, Blum KA, Nassau DE, Benfante NE, Ortiz MV, Carlo MI, Chan TA, Motzer RJ, Voss MH, Coleman J, Russo P, Reuter V, Hakimi AA, Reznik E. Comprehensive Genomic Analysis of Translocation Renal Cell Carcinoma Reveals Copy-Number Variations as Drivers of Disease Progression. Clin Cancer Res 2020; 26:3629-3640. [PMID: 32220885 PMCID: PMC7367714 DOI: 10.1158/1078-0432.ccr-19-3283] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/02/2020] [Accepted: 03/24/2020] [Indexed: 12/30/2022]
Abstract
PURPOSE Translocation renal cell carcinoma (tRCC) is a rare, aggressive renal cell carcinoma (RCC) subtype. There is currently limited understanding on the role of molecular alterations in the pathogenesis and progression of these tumors. We investigated the association between somatic alterations and clinical outcomes in two independent cohorts profiled using DNA sequencing. EXPERIMENTAL DESIGN Twenty-two tRCCs underwent targeted sequencing [Memorial Sloan Kettering Cancer Center (MSK)-IMPACT]; a subset was profiled using exome-sequencing and combined with exome data from The Cancer Genome Atlas (TCGA) for analysis. The prognostic value of specific somatic aberrations, tumor mutation burden (TMB), and fraction of copy-number-altered genome (FCNAg) was explored. In TCGA cases, neoantigen prediction and immune cell deconvolution were performed using RNA-sequencing and exome data. Overall survival estimates were computed using the Kaplan-Meier method; time-on-treatment was calculated for 14 MSK-IMPACT patients who underwent systemic therapy. Associations between molecular features and outcomes were evaluated using nonparametric testing. RESULTS Copy-number aberrant tRCCs were associated with poor overall survival (P = 0.03). Pediatric patients had tumors with lower FCNAg (P = 0.01). In one adult case with two chronologically distinct tumor samples sequenced, we confirmed that copy-number events occurred early during evolution. TERT promoter mutations were found exclusively in high-stage tumors. We found that tRCCs displayed distinct angiogenesis and PD-L1 gene expression profiles compared with other RCC subtypes. CONCLUSIONS Tumors molecularly defined by increased copy-number variations were associated with aggressive disease in tRCC. A higher burden of genomic events in adults compared with pediatric cases likely reflects a more aggressive clinical course. The unique immunophenotypic characteristics of tRCC merit further exploration.
Collapse
Affiliation(s)
- Julian Marcon
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Renzo G DiNatale
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alejandro Sanchez
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ritesh R Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sounak Gupta
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fengshen Kuo
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vladimir Makarov
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amar Sandhu
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Roy Mano
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew W Silagy
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kyle A Blum
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniel E Nassau
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nicole E Benfante
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael V Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria I Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jonathan Coleman
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paul Russo
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Victor Reuter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - A Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Ed Reznik
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York.
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
40
|
van der Beek JN, Geller JI, de Krijger RR, Graf N, Pritchard-Jones K, Drost J, Verschuur AC, Murphy D, Ray S, Spreafico F, Dzhuma K, Littooij AS, Selle B, Tytgat GAM, van den Heuvel-Eibrink MM. Characteristics and Outcome of Children with Renal Cell Carcinoma: A Narrative Review. Cancers (Basel) 2020; 12:E1776. [PMID: 32635225 PMCID: PMC7407101 DOI: 10.3390/cancers12071776] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/20/2022] Open
Abstract
Pediatric renal cell carcinoma (RCC) is a rare type of kidney cancer, most commonly occurring in teenagers and young adolescents. Few relatively large series of pediatric RCC have been reported. Knowledge of clinical characteristics, outcome and treatment strategies are often based on the more frequently occurring adult types of RCC. However, published pediatric data suggest that clinical, molecular and histological characteristics of pediatric RCC differ from adult RCC. This paper summarizes reported series consisting of ≥10 RCC pediatric patients in order to create an up-to-date overview of the clinical and histopathological characteristics, treatment and outcome of pediatric RCC patients.
Collapse
Affiliation(s)
- Justine N. van der Beek
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (A.S.L.); (G.A.M.T.); (M.M.v.d.H.-E.)
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children’s Hospital, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - James I. Geller
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA;
| | - Ronald R. de Krijger
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (A.S.L.); (G.A.M.T.); (M.M.v.d.H.-E.)
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Norbert Graf
- Department of Pediatric Oncology & Hematology, Saarland University Medical Center and Saarland University Faculty of Medicine, D-66421 Homburg, Germany;
| | - Kathy Pritchard-Jones
- UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (K.P.-J.); (K.D.)
| | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (A.S.L.); (G.A.M.T.); (M.M.v.d.H.-E.)
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Arnauld C. Verschuur
- Department of Pediatric Oncology, Hôpital d’Enfants de la Timone, APHM, 13005 Marseille, France;
| | - Dermot Murphy
- Department of Paediatric Oncology, Royal Hospital for Children, Glasgow G51 4TF, Scotland; (D.M.); (S.R.)
| | - Satyajit Ray
- Department of Paediatric Oncology, Royal Hospital for Children, Glasgow G51 4TF, Scotland; (D.M.); (S.R.)
| | - Filippo Spreafico
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy;
| | - Kristina Dzhuma
- UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (K.P.-J.); (K.D.)
| | - Annemieke S. Littooij
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (A.S.L.); (G.A.M.T.); (M.M.v.d.H.-E.)
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children’s Hospital, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Barbara Selle
- Department of Pediatric Hematology and Oncology, St. Annastift Children’s Hospital, 67065 Ludwigshafen, Germany;
| | - Godelieve A. M. Tytgat
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (A.S.L.); (G.A.M.T.); (M.M.v.d.H.-E.)
| | - Marry M. van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (A.S.L.); (G.A.M.T.); (M.M.v.d.H.-E.)
| |
Collapse
|
41
|
Breen KE, Carlo MI, Kemel Y, Maio A, Chen YB, Zhang L, Ceyhan-Birsoy O, Mandelker D. Fumarate hydratase c.914T > C (p.Phe305Ser) is a pathogenic variant associated with hereditary leiomyomatosis and renal cell cancer syndrome. Mol Genet Genomic Med 2020; 8:e1293. [PMID: 32463173 PMCID: PMC7434728 DOI: 10.1002/mgg3.1293] [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: 11/11/2019] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/22/2022] Open
Abstract
Background Hereditary leiomyomatosis and renal cell cancer syndrome (HLRCC), caused by heterozygous germline pathogenic variants in the FH, confers an increased risk for cutaneous and uterine leiomyomas and renal cancer. Methods About 13,722 advanced cancer patients, including 560 with renal cell carcinoma, had germline analysis performed in the context of tumor‐normal sequencing under an IRB approved protocol. Results We report two unrelated individuals with early onset kidney cancer who both carried the c.914C > T (p.Phe305Ser) germline variant in the FH. Both tumors exhibited loss of FH staining by immunohistochemistry and/or positive 2SC staining. Subsequent familial testing discovered that a daughter of a proband who carried the variant had both cutaneous and uterine leiomyomas. Conclusion This combination of evidence suggests that the FH c.914C > T (p.Phe305Ser) is pathogenic for HLRCC.
Collapse
Affiliation(s)
- Kelsey E Breen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Maria I Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Yelena Kemel
- Niehaus Center for Inherited Cancer Genomics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Anna Maio
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Liying Zhang
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Ozge Ceyhan-Birsoy
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Diana Mandelker
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| |
Collapse
|
42
|
Msaouel P, Malouf GG, Su X, Yao H, Tripathi DN, Soeung M, Gao J, Rao P, Coarfa C, Creighton CJ, Bertocchio JP, Kunnimalaiyaan S, Multani AS, Blando J, He R, Shapiro DD, Perelli L, Srinivasan S, Carbone F, Pilié PG, Karki M, Seervai RNH, Vokshi BH, Lopez-Terrada D, Cheng EH, Tang X, Lu W, Wistuba II, Thompson TC, Davidson I, Giuliani V, Schlacher K, Carugo A, Heffernan TP, Sharma P, Karam JA, Wood CG, Walker CL, Genovese G, Tannir NM. Comprehensive Molecular Characterization Identifies Distinct Genomic and Immune Hallmarks of Renal Medullary Carcinoma. Cancer Cell 2020; 37:720-734.e13. [PMID: 32359397 PMCID: PMC7288373 DOI: 10.1016/j.ccell.2020.04.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 02/02/2020] [Accepted: 04/01/2020] [Indexed: 12/26/2022]
Abstract
Renal medullary carcinoma (RMC) is a highly lethal malignancy that mainly afflicts young individuals of African descent and is resistant to all targeted agents used to treat other renal cell carcinomas. Comprehensive genomic and transcriptomic profiling of untreated primary RMC tissues was performed to elucidate the molecular landscape of these tumors. We found that RMC was characterized by high replication stress and an abundance of focal copy-number alterations associated with activation of the stimulator of the cyclic GMP-AMP synthase interferon genes (cGAS-STING) innate immune pathway. Replication stress conferred a therapeutic vulnerability to drugs targeting DNA-damage repair pathways. Elucidation of these previously unknown RMC hallmarks paves the way to new clinical trials for this rare but highly lethal malignancy.
Collapse
MESH Headings
- Adult
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Medullary/genetics
- Carcinoma, Medullary/immunology
- Carcinoma, Medullary/pathology
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/pathology
- Cell Proliferation
- Chromosome Aberrations
- Cohort Studies
- DNA Copy Number Variations
- DNA Replication
- Female
- Gene Expression Regulation, Neoplastic
- Genomics
- High-Throughput Nucleotide Sequencing
- Humans
- Kidney Neoplasms/genetics
- Kidney Neoplasms/immunology
- Kidney Neoplasms/pathology
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Nude
- Nucleotidyltransferases/genetics
- Nucleotidyltransferases/metabolism
- Prognosis
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- SMARCB1 Protein/genetics
- SMARCB1 Protein/metabolism
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Pavlos Msaouel
- Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX 77030-3721, USA; Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA.
| | - Gabriel G Malouf
- Department of Hematology and Oncology, Strasbourg University Hospitals, Strasbourg University, Strasbourg, France; Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UNISTRA, Illkirch Cedex, France
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hui Yao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Durga N Tripathi
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA
| | - Melinda Soeung
- Department of Genomic Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX 77030-3721, USA
| | - Priya Rao
- Department of Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Cristian Coarfa
- Department of Medicine and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chad J Creighton
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Medicine and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jean-Philippe Bertocchio
- Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX 77030-3721, USA; Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA
| | - Selvi Kunnimalaiyaan
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Asha S Multani
- Department of Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Jorge Blando
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Rong He
- Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX 77030-3721, USA
| | - Daniel D Shapiro
- Department of Urology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Luigi Perelli
- Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX 77030-3721, USA
| | - Sanjana Srinivasan
- Department of Genomic Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Federica Carbone
- Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX 77030-3721, USA
| | - Patrick G Pilié
- Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX 77030-3721, USA
| | - Menuka Karki
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA
| | - Riyad N H Seervai
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA; Molecular & Cellular Biology Graduate Program, Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bujamin H Vokshi
- Department of Hematology and Oncology, Strasbourg University Hospitals, Strasbourg University, Strasbourg, France; Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UNISTRA, Illkirch Cedex, France
| | | | - Emily H Cheng
- Human Oncology & Pathogenesis Program and Department of Pathology, Memorial Sloan Kettering Cancer Institute, New York City, NY 10065, USA
| | - Ximing Tang
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Wei Lu
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Timothy C Thompson
- Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX 77030-3721, USA
| | - Irwin Davidson
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UNISTRA, Illkirch Cedex, France
| | - Virginia Giuliani
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Translational Research to Advance Therapeutics and Innovation in Oncology (TRACTION), The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Katharina Schlacher
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alessandro Carugo
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Translational Research to Advance Therapeutics and Innovation in Oncology (TRACTION), The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Timothy P Heffernan
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Translational Research to Advance Therapeutics and Innovation in Oncology (TRACTION), The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX 77030-3721, USA; Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Jose A Karam
- Department of Urology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Christopher G Wood
- Department of Urology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Cheryl L Walker
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA; Department of Medicine, Baylor College of Medicine, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX 77030-3721, USA; Department of Genomic Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, Unit 1374, The University of Texas MD Anderson Cancer Center, 1155 Pressler Street, Houston, TX 77030-3721, USA.
| |
Collapse
|
43
|
Abstract
TFEB is overexpressed in TFEB-rearranged renal cell carcinomas as well as in renal tumors with amplifications of TFEB at 6p21.1. As recent literature suggests that renal tumors with 6p21.1 amplification behave more aggressively than those with rearrangements of TFEB, we compared relative TFEB gene expression in these tumors. This study included 37 TFEB-altered tumors: 15 6p21.1-amplified and 22 TFEB-rearranged (including 5 cases from The Cancer Genome Atlas data set). TFEB status was verified using a combination of fluorescent in situ hybridization (n=27) or comprehensive molecular profiling (n=13) and digital droplet polymerase chain reaction was used to quantify TFEB mRNA expression in 6p21.1-amplified (n=9) and TFEB-rearranged renal tumors (n=19). These results were correlated with TFEB immunohistochemistry. TFEB-altered tumors had higher TFEB expression when normalized to B2M (mean: 168.9%, n=28), compared with non-TFEB-altered controls (mean: 7%, n=18, P=0.005). Interestingly, TFEB expression in tumors with rearrangements (mean: 224.7%, n=19) was higher compared with 6p21.1-amplified tumors (mean: 51.2%, n=9; P=0.06). Of note, classic biphasic morphology was only seen in TFEB-rearranged tumors and when present correlated with 6.8-fold higher TFEB expression (P=0.00004). Our results suggest that 6p21.1 amplified renal tumors show increased TFEB gene expression but not as much as t(6;11) renal tumors. These findings correlate with the less consistent/diffuse expression of downstream markers of TFEB activation (cathepsin K, melan A, HMB45) seen in the amplified neoplasms. This suggests that the aggressive biological behavior of 6p21.1 amplified renal tumors might be secondary to other genes at the 6p21.1 locus that are co-amplified, such as VEGFA and CCND3, or other genetic alterations.
Collapse
|
44
|
Ooms AH, Vujanić GM, D’Hooghe E, Collini P, L’Herminé-Coulomb A, Vokuhl C, Graf N, van den Heuvel-Eibrink MM, de Krijger RR. Renal Tumors of Childhood-A Histopathologic Pattern-Based Diagnostic Approach. Cancers (Basel) 2020; 12:cancers12030729. [PMID: 32204536 PMCID: PMC7140051 DOI: 10.3390/cancers12030729] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/04/2020] [Accepted: 03/07/2020] [Indexed: 12/15/2022] Open
Abstract
Renal tumors comprise approximately 7% of all malignant pediatric tumors. This is a highly heterogeneous group of tumors, each with its own therapeutic management, outcome, and association with germline predispositions. Histopathology is the key in establishing the correct diagnosis, and therefore pathologists with expertise in pediatric oncology are needed for dealing with these rare tumors. While each tumor shows different histologic features, they do have considerable overlap in cell type and histologic pattern, making the diagnosis difficult to establish, if based on routine histology alone. To this end, ancillary techniques, such as immunohistochemistry and molecular analysis, can be of great importance for the correct diagnosis, resulting in appropriate treatment. To use ancillary techniques cost-effectively, we propose a pattern-based approach and provide recommendations to aid in deciding which panel of antibodies, supplemented by molecular characterization of a subset of genes, are required.
Collapse
Affiliation(s)
- Ariadne H.A.G. Ooms
- Princess Máxima Center for pediatric oncology, 3584 CS Utrecht, The Netherlands (M.M.v.d.H.-E.)
- Pathan B.V., 3045 PM Rotterdam, The Netherlands
| | | | - Ellen D’Hooghe
- Department of Pathology, Oslo University Hospital, Rikshospitalet, 0372 Oslo, Norway;
| | - Paola Collini
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Aurore L’Herminé-Coulomb
- Sorbonne Université, Department of Pathology, Hôpital Armand Trousseau, Hopitaux Universitaires Est Parisien, 75012 Paris, France;
| | - Christian Vokuhl
- Section of Pediatric Pathology, Department of Pathology, University Hospital Bonn, 53127 Bonn, Germany;
| | - Norbert Graf
- Department of Pediatric Oncology & Hematology, Saarland University, D-66421 Homburg, Germany;
| | | | - Ronald R. de Krijger
- Princess Máxima Center for pediatric oncology, 3584 CS Utrecht, The Netherlands (M.M.v.d.H.-E.)
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Correspondence: ; Tel.: +31-088-9727272
| |
Collapse
|
45
|
Campbell K, Mullen E. 50 Years Ago in TheJournal ofPediatrics: 50 years of progress in renal cell carcinoma: From case reports to targeted therapy. J Pediatr 2020; 218:120. [PMID: 32089177 DOI: 10.1016/j.jpeds.2019.09.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Kevin Campbell
- Dana-Farber/Boston Children's Blood Disorders and Cancer Center, Boston, Massachusetts
| | - Elizabeth Mullen
- Dana-Farber/Boston Children's Blood Disorders and Cancer Center, Boston, Massachusetts
| |
Collapse
|
46
|
Woo CG, Yun SJ, Son SM, Lim YH, Lee OJ. Characteristics of Renal Cell Carcinoma Harboring TPM3-ALK Fusion. Yonsei Med J 2020; 61:262-266. [PMID: 32102128 PMCID: PMC7044692 DOI: 10.3349/ymj.2020.61.3.262] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022] Open
Abstract
The World Health Organization 2016 edition assigned anaplastic lymphoma kinase (ALK) rearrangement-associated renal cell carcinoma (ALK-RCC) as an emerging renal tumor entity. Identifying ALK-RCC is important because ALK inhibitors have been shown to be effective in treatment. Here, we report the case of a 14-year-old young man with ALK-RCC. Computed tomography revealed a well-demarcated 5.3-cm enhancing mass at the upper pole of the left kidney. There was no further history or symptoms of the sickle-cell trait. The patient underwent left radical nephrectomy. Pathologically, the mass was diagnosed as an unclassified RCC. Targeted next-generation sequencing identified a TPM3-ALK fusion gene. The present report and literature review demonstrate that TPM3-ALK RCC may be associated with distinct clinicopathological features. Microscopically, the tumors showed diffuse growth and tubulocystic changes with inflammatory cell infiltration. Tumor cells were dis-cohesive and epithelioid with abundant eosinophilic cytoplasm and cytoplasmic vacuoles. If morphological features and TFE3 expression are present in adolescent and young patients, molecular tests for ALK translocation should be performed. This awareness is critically important, because ALK rearrangement confers sensitivity to ALK inhibitors.
Collapse
Affiliation(s)
- Chang Gok Woo
- Department of Pathology, Chungbuk National University Hospital, Cheongju, Korea
- Department of Pathology, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Seok Jung Yun
- Department of Urology, Chungbuk National University Hospital, Cheongju, Korea
- Department of Urology, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Seung Myoung Son
- Department of Pathology, Chungbuk National University Hospital, Cheongju, Korea
- Department of Pathology, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Young Hyun Lim
- Department of Pathology, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Ok Jun Lee
- Department of Pathology, Chungbuk National University Hospital, Cheongju, Korea
- Department of Pathology, Chungbuk National University College of Medicine, Cheongju, Korea.
| |
Collapse
|
47
|
Ünal E, Yilmaz E, Özcan A, Işik B, Karakükcü M, Turan C, Akgün H, Öztürk F, Coşkun A, Özdemir MA, Patiroğlu T. Twenty children with non-Wilms renal tumors from a reference center in Central Anatolia, Turkey. Turk J Med Sci 2020; 50:18-24. [PMID: 31655501 PMCID: PMC7080372 DOI: 10.3906/sag-1902-106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/17/2019] [Indexed: 12/21/2022] Open
Abstract
Background/aim Non-Wilms renal tumors (NWRTs) are rarely encountered in children. The aim of this study is to determine the treatment strategies, prognosis, outcomes, and survival of children with NWRTs at Erciyes University in Kayseri, Turkey. Materials and methods Medical records of all patients (n = 20) treated for NWRTs over a 23-year period (1995–2018) were reviewed retrospectively. Results There was male predominance (female/male: 7/13); the median age at diagnosis was 3.2 years old (0.1–13.5 years old). The major histological groups included mesoblastic nephroma (MBN), (n: 5, 25%), malignant rhabdoid tumor (MRT), (n: 5, 25%), renal cell carcinoma, (n: 3, 15%), inflammatory myofibroblastic tumor (n: 2, 10%), multilocular cystic renal tumors (n: 2, 10%), metanephric adenoma (n: 1, 5%), renal neuroblastoma (n: 1, 5%), and bilateral renal Ewing sarcoma/primitive neuroectodermal tumor (ES/PNET) (n: 1, 5%). All of the patients with NWRTs had radical nephrectomy except the child with bilateral renal ES/PNET. Six children died because of progressive disease; the mortality rate was 30% (n: 6). Conclusion We have made the first report of bilateral renal involvement of ES/PNET in the English medical literature. Physicians dealing with pediatric renal masses should be alert to the high mortality rate in children with MRT, MBN, and ES/PNET and they should design substantial management plans for NWRTs.
Collapse
Affiliation(s)
- Ekrem Ünal
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey,Molecular Biology and Genetic Department, Gevher Nesibe Genom and Stem Cell Institution, Genome and Stem Cell Center (GENKÖK), Erciyes University, Kayseri, Turkey
| | - Ebru Yilmaz
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Alper Özcan
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Bilgen Işik
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Musa Karakükcü
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Cüneyt Turan
- Department of Pediatric Surgery, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Hülya Akgün
- Department of Pathology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Figen Öztürk
- Department of Pathology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Abdulhakim Coşkun
- Division of Pediatrics Radiology, Department of Radiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Mehmet Akif Özdemir
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Türkan Patiroğlu
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
48
|
Fuller MY. Pediatric Renal Tumors: Diagnostic Updates. KIDNEY CANCER 2020. [DOI: 10.1007/978-3-030-28333-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
49
|
|
50
|
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: 0.6] [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
|