1
|
Sprokkerieft J, van der Beek JN, Spreafico F, Selle B, Chowdhury T, Graf N, Verschuur AC, Dandis R, Bex A, Geller JI, Tytgat GAM, van den Heuvel-Eibrink MM. Outcome after treatment with axitinib in children, young adults, and adults with renal cell carcinoma: a narrative review. Crit Rev Oncol Hematol 2024; 204:104523. [PMID: 39326645 DOI: 10.1016/j.critrevonc.2024.104523] [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: 05/03/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024] Open
Abstract
Renal cell carcinoma (RCC) is a very rare type of renal cancer in children and young adults. When metastasized or recurrent, no standards of care are available, and outcome is still poor. The tyrosine kinase inhibitor axitinib is approved for treatment of RCC in adults, but its effects in children and young adults with RCC remains unclear. Due to the histological and biological differences between children and adults, it is difficult to extrapolate knowledge on treatments from the adult to the pediatric and young adult setting. This paper summarizes the clinical characteristics and outcomes of patients with RCC who were treated with axitinib, with the aim to gain insight in the clinical efficacy of this compound in this young patient group.
Collapse
Affiliation(s)
- Julia Sprokkerieft
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| | - Justine N van der Beek
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Filippo Spreafico
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - 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
| | - Tanzina Chowdhury
- Pediatric Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Saarland University, Homburg, Germany
| | - Arnauld C Verschuur
- Department of Pediatric Hematology-Oncology, Hôpital d'Enfants de la Timone, APHM, Marseille, France
| | - Rana Dandis
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Axel Bex
- Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam 1066CX, the Netherlands; Division of Surgical and Interventional Science, The Royal Free London NHS Foundation Trust and UCL, London, UK
| | - James I Geller
- Division of Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, USA
| | - Godelieve A M Tytgat
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | |
Collapse
|
2
|
Holmes L, Masire P, Eaton A, Mason R, Holmes M, William J, Poleon M, Enwere M. Pediatric Renal Cell Carcinoma (pRCC) Subpopulation Environmental Differentials in Survival Disadvantage of Black/African American Children in the United States: Large-Cohort Evidence. Cancers (Basel) 2024; 16:3975. [PMID: 39682162 DOI: 10.3390/cancers16233975] [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: 10/08/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
OBJECTIVE Renal cell carcinoma (RCC) is a rare but severe and aggressive pediatric malignancy. While incidence is uncommon, survival is relatively low with respect to acute lymphocytic leukemia (ALL), AML, lymphoma, ependymoma, glioblastoma, and Wilms Tumor. The pediatric renal cell carcinoma (pRCC) incidence, cumulative incidence (period prevalence), and mortality vary by health disparities' indicators, namely sex, race, ethnicity, age at tumor diagnosis, and social determinants of health (SDHs) as well as Epigenomic Determinants of Health (EDHs). However, studies are unavailable on some pRCC risk determinants, such as area of residence and socio-economic status (SES). The current study aimed at assessing the temporal trends, cumulative incidence, household median income, urbanity, mortality, and pRCC survival differentials. MATERIALS AND METHODS A retrospective cohort design was utilized to examine the event-free survival of children (0-19) with RCC using the Surveillance Epidemiology and End Result Data, 1973-2015. While the time-dependent variable, namely survival months, was utilized, we assessed the predictors of pRCC survival, mainly sex, age at diagnosis, education, insurance status, income, and tumor grade, as prognostic factors. In examining the joint effect of area of residence and race, as an exposure function with time in survival, we utilized the Cox proportional hazard model, while the annual percent change was assessed using a generalized linear model, implying a weighted average. RESULTS Between 1973 and 2015, there were 174 cases of pRCC, of whom 49 experienced mortality (28.2%). The pRCC cumulative incidence tends to increase with advancing age. A significant survival differential was observed between black/AA children with RCC and their white counterparts. Compared with white children, black/AA children were almost three times as likely to die, hazard ratio (HR) = 2.90, 95% CI = 1.56-5.31, p = 0.001. A survival differential was observed in sex, with males presenting with a 21% increased likelihood of dying, HR = 1.21; 95% CI, 0.69-2.11. In the metropolitan area, the risk of dying was almost three times as likely among black/AA children compared to their white counterparts, HR = 2.78; 95% CI, 1.45-5.43, while in the urban area, the risk of dying was almost four times as likely among black/AA children compared to their white counterparts, HR = 4.18; 95% CI, 0.84-20.80. After controlling for age, sex, education, and insurance, the risk of dying increased amongst black/AA children in metropolitan areas, adjusted HR (aHR) = 3.37, 99% CI = 1.35-8.44. In the urban area, after adjustment for age, sex, and insurance, there was an increased risk of dying for black/AA children, compared with their white counterparts with pRCC, aHR = 8.87, 99% CI = 2.77-28.10. CONCLUSION pRCC indicates an increased trend in males and age at diagnosis between 10 and 14, as well as a survival disadvantage among black/AA children, compared with their white counterparts. Additionally, urbanity significantly influences the racial differences in survival. These data are suggestive of the conjoined effect of environment and race in pRCC survival, indicative of further assessment of gene-environment interaction (epigenomics) in incidence, mortality, and survival in pRCC.
Collapse
Affiliation(s)
- Laurens Holmes
- Public Health & Allied Health Science Department, Wesley College, Delaware State University, Dover, DE 19901, USA
- Biological Sciences Department, University of Delaware, Newark, DE 19716, USA
- Medical College of Wisconsin, Milwaukee, WI 53226, USA
- College of Population Health, Thomas Jefferson University, Philadelphia, PA 19144, USA
- Global Health Equity Foundation, Bear, DE 19701, USA
| | - Phatismo Masire
- College of Population Health, Thomas Jefferson University, Philadelphia, PA 19144, USA
| | - Arieanna Eaton
- Department of Integrated Physiology and Health Sciences, Alma College, Alma, MI 48801, USA
| | - Robert Mason
- Public Health & Allied Health Science Department, Wesley College, Delaware State University, Dover, DE 19901, USA
| | - Mackenzie Holmes
- Global Health Equity Foundation, Bear, DE 19701, USA
- Texas A & M University, College Station, TX 77843, USA
| | | | - Maura Poleon
- Global Health Equity Foundation, Bear, DE 19701, USA
| | - Michael Enwere
- Global Health Equity Foundation, Bear, DE 19701, USA
- PrimeLife360, Wellness 20 Wenlock Road, London N1 7GU, UK
| |
Collapse
|
3
|
Hu L, Li L, Li A, Tong J. Four years of natural progressive course: A rare case report of juvenile Xp11.2 translocations renal cell carcinoma with TFE3 gene fusion. Open Med (Wars) 2024; 19:20240985. [PMID: 38953008 PMCID: PMC11215303 DOI: 10.1515/med-2024-0985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/02/2024] [Accepted: 06/02/2024] [Indexed: 07/03/2024] Open
Abstract
Background Renal cell carcinoma (RCC) with TFE3 gene fusion caused by Xp11.2 translocations is a rare RCC subtype. This tumor is typically seen in children, comprising 20‒40% of overall RCC cases compared to 1‒1.6% observed in adults. Xp11.2 RCC is associated with a poor prognosis due to both the progression of local lesions and early distant and lymphatic metastasis. Case presentation A case of RCC with Xp11.2 RCC translocations and TFE3 gene fusion was found in a pediatric patient, illustrating the catastrophic effects of ignoring the condition. The tumor developed from a local lesion to lymph metastasis (3.2-12 cm) within 4 years. Despite ongoing controversy, surgical resection remains the most common and productive approach. In this patient, renal retroperitoneal lymph node dissection and radical nephrectomy of the left kidney were performed via laparoscopic surgery. The RCC-associated Xp11.2 translocation/TFE3 gene fusions were identified by postoperative pathology. Microscopic analysis showed the presence of intravascular cancer thrombus, renal sinus invasion, and cancer necrosis. The pathological stages were confirmed as PT3aN1M0 with a negative margin. Follow-up at 5 months showed that the patient recovered without the use of any adjuvant treatments. Conclusion Our study highlights the natural course, diagnosis, and treatment of RCC-associated Xp11.2 translocation/TFE3 gene fusions, especially the necessity of early surgery. This case may be a helpful reference for urologists in the treatment of similar cases. It also serves as a precautionary signal for patients who neglect the renal neoplasm.
Collapse
Affiliation(s)
- Liang Hu
- Department of Urology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Lina Li
- College of Medicine, Jinhua Polytechnic, Jinhua, Zhejiang, China
| | - Angcheng Li
- Department of Pathology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jianyong Tong
- Department of Urology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
4
|
van der Beek JN, Schenk JP, Morosi C, Watson TA, Coma A, Graf N, Chowdhury T, Ramírez-Villar GL, Spreafico F, Welter N, Dzhuma K, van Tinteren H, de Krijger RR, van den Heuvel-Eibrink MM, Littooij AS. Diagnostic magnetic resonance imaging characteristics of congenital mesoblastic nephroma: a retrospective multi-center International Society of Pediatric Oncology-Renal Tumor Study Group (SIOP-RTSG) radiology panel study. Pediatr Radiol 2024; 54:965-976. [PMID: 38609702 PMCID: PMC11111520 DOI: 10.1007/s00247-024-05918-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Congenital mesoblastic nephroma is the most common solid renal tumor in neonates. Therefore, patients <3 months of age are advised to undergo upfront nephrectomy, whereas invasive procedures at diagnosis in patients ≥3 months of age are discouraged by the International Society of Pediatric Oncology-Renal Tumor Study Group (SIOP-RTSG). Nevertheless, discriminating congenital mesoblastic nephroma, especially from the more common Wilms tumor, solely based on imaging remains difficult. Recently, magnetic resonance imaging (MRI) has become the preferred modality. Studies focusing on MRI characteristics of congenital mesoblastic nephroma are limited. OBJECTIVE This study aims to identify diagnostic MRI characteristics of congenital mesoblastic nephroma in the largest series of patients to date. MATERIALS AND METHODS In this retrospective multicenter study, five SIOP-RTSG national review radiologists identified 52 diagnostic MRIs of histologically proven congenital mesoblastic nephromas. MRI was performed following SIOP-RTSG protocols, while radiologists assessed their national cases using a validated case report form. RESULTS Patients (24/52 classic, 11/52 cellular, and 15/52 mixed type congenital mesoblastic nephroma, 2/52 unknown) had a median age of 1 month (range 1 day-3 months). Classic type congenital mesoblastic nephroma appeared homogeneous with a lack of hemorrhage, necrosis and/or cysts, showing a concentric ring sign in 14 (58.3%) patients. Cellular and mixed type congenital mesoblastic nephroma appeared more heterogeneous and were larger (311.6 and 174.2 cm3, respectively, versus 41.0 cm3 for the classic type (P<0.001)). All cases were predominantly T2-weighted isointense and T1-weighted hypointense, and mean overall apparent diffusion coefficient values ranged from 1.05-1.10×10-3 mm2/s. CONCLUSION This retrospective international collaborative study showed classic type congenital mesoblastic nephroma predominantly presented as a homogeneous T2-weighted isointense mass with a typical concentric ring sign, whereas the cellular type appeared more heterogeneous. Future studies may use identified MRI characteristic of congenital mesoblastic nephroma for validation and for exploring the discriminative non-invasive value of MRI, especially from Wilms tumor.
Collapse
Affiliation(s)
- Justine N van der Beek
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| | - Jens-Peter Schenk
- Clinic of Diagnostic and Interventional Radiology, Division of Pediatric Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Carlo Morosi
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Tom A Watson
- Department of Paediatric Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Ana Coma
- Department of Pediatric Radiology, Hospital Vall d'Hebron, Barcelona, Spain
| | - Norbert Graf
- Department of Pediatric Oncology & Hematology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Tanzina Chowdhury
- Department of Haematology and Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Gema L Ramírez-Villar
- Department of Paediatric Oncology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Filippo Spreafico
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Nils Welter
- Department of Pediatric Oncology & Hematology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Kristina Dzhuma
- Developmental Biology and Cancer Department, University College London Great Ormond Street Institute of Child Health, London, UK
- Department of Paediatric Urology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Harm van Tinteren
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Division of Child Health, Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Annemieke S Littooij
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children's Hospital, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
5
|
Yu L, He L. Aberrant expression of TFE3 in granular cell tumor: Four cases in pediatrics. Pediatr Blood Cancer 2024; 71:e30811. [PMID: 38073015 DOI: 10.1002/pbc.30811] [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: 11/22/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 01/24/2024]
Affiliation(s)
- Lianyuan Yu
- Department of Pathology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Lejian He
- Department of Pathology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
6
|
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
|
7
|
Huang W, Peng Y, Zhang Y, Qiu Y, Liu Y, Wang A, Kang L. Multimodality imaging of Xp11.2 translocation/TFE3 gene fusion associated with renal cell carcinoma: a case report. Front Med (Lausanne) 2023; 10:1266630. [PMID: 37795411 PMCID: PMC10546202 DOI: 10.3389/fmed.2023.1266630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 08/31/2023] [Indexed: 10/06/2023] Open
Abstract
Background Xp11.2 translocation/TFE3 gene fusion associated with renal cell carcinoma (Xp11.2 RCC) exhibits unique biological characteristics and is associated with an increased incidence of tumor thrombosis, lymph node metastasis, and advanced disease stages. Multimodality imaging, including US, contrast-enhanced CT, multi-parametric MRI, and 18F-FDG PET/CT plays a crucial role in the preoperative diagnosis and differentiation of renal tumors. Case report A 15-year-old female presented with lumbar pain worsened, and developed persistent painless hematuria. The CT attenuation values of the scan without contrast, corticomedullary phase, nephrographic phase, and delayed phases were 35 HU, 83 HU, 82 HU, and 75 HU, respectively. The solid component of the mass displayed heterogeneous marked enhancement. Furthermore, MRU indicated that the lesion involved the cortical medulla and infringed on the renal sinus fat. The lesion appeared isosignal in T1WI, slightly low signal in T2WI, and slightly high signal in DWI. The degree of enhancement in the three phases of enhancement scan was lower than that in the renal parenchyma, and hemorrhage and necrosis were observed within the internal part of the lesion. To further clarify the staging, the patient underwent 18F-FDG PET/CT. PET/CT images showed multiple irregular occupancies in the right kidney with unclear borders, showing a heterogeneous increase in 18F-FDG uptake, with SUVmax values ranging from 2.3 to 5.2 in the routine imaging phase (60 min post-injection), compared to SUVmax values ranging from 2.8 to 6.9 in the delayed imaging phase (160 min post-injection). Additionally, multiple enlarged and fused lymph nodes were observed in the medial part of the right kidney and the retroperitoneum, exhibiting a heterogeneous increase in 18F-FDG uptake, with SUVmax values ranging from 4.1 to 8.7 in the routine imaging phase, compared to SUVmax values ranging from 4.4 to 9.1 in the delayed imaging phase. The postoperative pathology, immunohistochemistry, and molecular analysis of histiocytes were consistent with a diagnosis of Xp11.2 RCC. One month after surgery, enhanced-CT examination of the patient revealed lung metastasis, peritoneal metastasis, and multiple lymph node metastases throughout the body, with an overall survival of 16 months. Conclusion Xp11.2 RCC exhibits unique biological characteristics and is associated with an increased incidence of tumor thrombosis, lymph node metastasis, and advanced disease stages. Long-term follow-up is essential to monitor the likelihood of recurrence and metastasis. 18F-FDG PET/CT examination can comprehensively visualize the lesion's location and extent, providing a basis for clinical tumor staging and aiding in treatment monitoring and follow-up. To address the limitations of FDG, the utilization of specific tracers designed for RCC or tracers that are not excreted via the urinary system would be ideal. Further advancements in molecular imaging technologies and the development of novel tracers hold great promise in advancing the diagnosis and management of RCC, ultimately contributing to better patient outcomes and overall disease management.
Collapse
Affiliation(s)
- Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Yushuo Peng
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Yongbai Zhang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Yongkang Qiu
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Yi Liu
- Department of Urology, Peking University First Hospital, Beijing, China
- Institute of Urology, Peking University, Beijing, China
- National Urological Cancer Center, Beijing, China
| | - Aixiang Wang
- Department of Urology, Peking University First Hospital, Beijing, China
- Institute of Urology, Peking University, Beijing, China
- National Urological Cancer Center, Beijing, China
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| |
Collapse
|
8
|
Naviwala MS, Dawood T, Uddin Z, Chundriger Q, Jalbani IK. The Transition From Localized to Metastatic: A Case Report of Adult TFE3-Positive Xp11.2 Translocation Renal Cell Carcinoma. Cureus 2023; 15:e43378. [PMID: 37700986 PMCID: PMC10494990 DOI: 10.7759/cureus.43378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2023] [Indexed: 09/14/2023] Open
Abstract
Xp11.2 translocation renal cell carcinoma (Xp11.2 RCC) is a rare tumor, occurring more frequently in childhood than in adulthood. It results from Xp11.2 chromosome translocations and the fusion of the transcription factor E3 (TFE3) gene. In this context, we present a case report of an 18-year-old female who was diagnosed with Xp11.2 RCC following open radical nephrectomy and lymph node dissection on the left side. The histopathological analysis indicated stage T3aN1Mx disease, which was confirmed through immunohistochemistry (IHC) and fluorescent in situ hybridization (FISH). The patient remained under observation until March 2023 when systemic scans uncovered the presence of ascites, peritoneal carcinomatosis, and left supraclavicular lymphadenopathy. A subsequent biopsy reaffirmed the primary disease, leading to the planning of systemic treatment involving tyrosine kinase inhibitors (TKIs) and immunotherapy. However, due to financial constraints, the patient's treatment options were limited to sunitinib initially. The current plan involves reevaluation after three months using scans to determine the subsequent course of treatment. Our case report offers crucial insights into the clinical presentation, diagnosis, and treatment of this rare malignancy. This enhances medical understanding, guides research, and improves the management of similar cases. Case reports like this share practical experiences, shaping future studies and patient care.
Collapse
Affiliation(s)
| | | | - Zeeshan Uddin
- Pathology and Laboratory Medicine, Aga Khan University Hospital, Karachi, PAK
| | | | | |
Collapse
|
9
|
Tawk A, Abou Zahr R, Chalhoub K, Danaf S, Kamareddine MH, Nohra J. Renal cell carcinoma in the pediatric population: A case report and review of the literature. Urol Case Rep 2023; 49:102453. [PMID: 37323771 PMCID: PMC10267517 DOI: 10.1016/j.eucr.2023.102453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/17/2023] Open
Abstract
Renal cell carcinoma is a cancer thought to originate from renal epithelial cells. Commonly seen in patients older than 60 years of age, renal cell carcinoma presents as rare pathological entity seen in urological cancers among the pediatric population. A 17-year-old female patient presented with complaints of intermittency, dysuria, and gross hematuria. Radiological imaging was in favor of a left renal mass. Under general anesthesia, the left kidney was completely laparoscopically resected and sent to pathology, which along with correlating the age group of the patient and the morphology on pathological analysis, was suggestive of microphthalmia family translocation renal cell carcinoma.
Collapse
Affiliation(s)
- Antonios Tawk
- Faculty of Medicine and Medical Sciences, University of Balamand, Aschrafieh, Beirut, Lebanon
| | - Rawad Abou Zahr
- Department of Urology, Saint George Hospital University Medical Center, University of Balamand, Beirut, 1100 2807, Lebanon
| | - Khalil Chalhoub
- Department of Urology, Saint George Hospital University Medical Center, University of Balamand, Beirut, 1100 2807, Lebanon
| | - Samer Danaf
- Department of Urology, Saint George Hospital University Medical Center, University of Balamand, Beirut, 1100 2807, Lebanon
| | | | - Joe Nohra
- Department of Urology, Saint George Hospital University Medical Center, University of Balamand, Beirut, 1100 2807, Lebanon
| |
Collapse
|
10
|
Okamoto T, Sasaki T, Takahashi Y, Takamatsu M, Kanda H, Hiratsuka M, Matsuyama M, Ozaka M, Sasahira N. Perivascular epithelioid cell tumor (PEComa) of the cystic duct. Clin J Gastroenterol 2023; 16:87-95. [PMID: 36309916 DOI: 10.1007/s12328-022-01730-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/23/2022] [Indexed: 02/03/2023]
Abstract
Perivascular epithelioid cell tumors, also known as PEComas, are rare mesenchymal tumors composed mainly of epithelioid cells found in perivascular tissue. PEComas occur most frequently in the kidney, uterus, the gastrointestinal tract, liver, and retroperitoneum; those originating in the biliary tree are extremely rare. We report a case of benign PEComa of the cystic duct with positive TFE3 staining on immunohistochemistry.A 66-year-old woman was referred for a 20 mm mass adjacent to the common bile duct discovered incidentally on abdominal ultrasound. Laboratory data including tumor markers were unremarkable. The tumor appeared to arise from the cystic duct, showed early enhancement, and compressed the common bile duct on imaging studies. Endoscopic ultrasound-guided fine-needle aspiration revealed round- and spindle-shaped atypical cells with eosinophilic cytoplasm and brown deposits suggestive of melanin granules. Histological examination of the resected specimen revealed a tumor consisting of epithelioid cells forming an alveolar structure, with melanin pigmentation. Immunohistochemistry was positive for HMB-45 and TFE3, consistent with benign pigmented PEComa of the cystic duct. Melanotic, myogenic, and TFE3 staining are helpful when diagnosing PEComas arising in unusual locations.
Collapse
Affiliation(s)
- Takeshi Okamoto
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Takashi Sasaki
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan.
| | - Yu Takahashi
- Department of Hepato-Biliary-Pancreatic Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Koto-Ku, AriakeTokyo, 135-8550, Japan
| | - Manabu Takamatsu
- Department of Pathology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Hiroaki Kanda
- Department of Pathology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Makiko Hiratsuka
- Department of Diagnostic Imaging, Diagnostic Imaging Center, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Masato Matsuyama
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Naoki Sasahira
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| |
Collapse
|
11
|
Qu Y, Wu X, Anwaier A, Feng J, Xu W, Pei X, Zhu Y, Liu Y, Bai L, Yang G, Tian X, Su J, Shi GH, Cao DL, Xu F, Wang Y, Gan HL, Ni S, Sun MH, Zhao JY, Zhang H, Ye D, Ding C. Proteogenomic characterization of MiT family translocation renal cell carcinoma. Nat Commun 2022; 13:7494. [PMID: 36470859 PMCID: PMC9722939 DOI: 10.1038/s41467-022-34460-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/26/2022] [Indexed: 12/12/2022] Open
Abstract
Microphthalmia transcription factor (MiT) family translocation renal cell carcinoma (tRCC) is a rare type of kidney cancer, which is not well characterized. Here we show the comprehensive proteogenomic analysis of tRCC tumors and normal adjacent tissues to elucidate the molecular landscape of this disease. Our study reveals that defective DNA repair plays an important role in tRCC carcinogenesis and progression. Metabolic processes are markedly dysregulated at both the mRNA and protein levels. Proteomic and phosphoproteome data identify mTOR signaling pathway as a potential therapeutic target. Moreover, molecular subtyping and immune infiltration analysis characterize the inter-tumoral heterogeneity of tRCC. Multi-omic integration reveals the dysregulation of cellular processes affected by genomic alterations, including oxidative phosphorylation, autophagy, transcription factor activity, and proteasome function. This study represents a comprehensive proteogenomic analysis of tRCC, providing valuable insights into its biological mechanisms, disease diagnosis, and prognostication.
Collapse
Affiliation(s)
- Yuanyuan Qu
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Xiaohui Wu
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Aihetaimujiang Anwaier
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Jinwen Feng
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Xiaoru Pei
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Yu Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Yang Liu
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Lin Bai
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Guojian Yang
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Xi Tian
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Jiaqi Su
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Guo-Hai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Da-Long Cao
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Fujiang Xu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yue Wang
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Hua-Lei Gan
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
- Tissue Bank & Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Shujuan Ni
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
- Tissue Bank & Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Meng-Hong Sun
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
- Tissue Bank & Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China.
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China.
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| | - Chen Ding
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
12
|
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
|
13
|
Zhao J, Dai K, Xie J, Fang C, Chen N, Dai J, Xu D. Case Report: Clinical complete response of advanced renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene fusion by treated by camrelizumab and axitinib: A rare case report. Front Pharmacol 2022; 13:927299. [PMID: 36034832 PMCID: PMC9403306 DOI: 10.3389/fphar.2022.927299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Renal cell carcinoma (RCC) associated with Xp11.2 translocation/TFE3 gene fusions is a rare subtype of renal tumor. This entity predominantly occurs in juveniles, but rarely in adults. Xp11.2 translocation RCC (tRCC) patients with lymph node or organ metastasis are associated with poor prognosis, and the strategy remains controversial. Herein, we presented our experience with the diagnosis and treatment of an adult case of Xp11.2 tRCC. In our clinical practice, a 32-year-old male manifested fever and right flank paroxysmal blunt pain, and computed tomography showed an inhomogeneous mass, 6 cm in diameter, in the right kidney. Then right partial nephrectomy (PN) and renal hilar lymph node dissection by laparoscopic surgery were performed. Pathology revealed that the tumor cells were positive for TFE3 immunohistologically and positive for TFE3 break-apart fluorescence in situ hybridization assay. A splice site mutation c.1544-1G>T of protein tyrosine phosphatase receptor delta (PTPRD) was detected by next-generation sequencing and weak PTPRD expression was confirmed in tumor tissues compared to tumor periphery. This patient was diagnosed with stage III RCC and received immune checkpoint inhibitor (camrelizumab) in combination with tyrosine kinase inhibitor (axitinib) treatment for 1 year. He achieved a clinical complete response with no sign of recurrence or metastasis. PTPRD mutation might be a favorable indicator for Xp11.2 tRCC patients managed by PN and followed by the adjuvant therapy of immune checkpoint inhibitor and tyrosine kinase inhibitor.
Collapse
Affiliation(s)
- Juping Zhao
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Juping Zhao, ; Jun Dai, ; Danfeng Xu,
| | - Kun Dai
- Hangzhou Jichenjunchuang Medical Laboratory Co.Ltd, Hangzhou, China
| | - Jialing Xie
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Fang
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Na Chen
- Hangzhou Jichenjunchuang Medical Laboratory Co.Ltd, Hangzhou, China
| | - Jun Dai
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Juping Zhao, ; Jun Dai, ; Danfeng Xu,
| | - Danfeng Xu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Juping Zhao, ; Jun Dai, ; Danfeng Xu,
| |
Collapse
|
14
|
Tretiakova MS. Chameleon TFE3-translocation RCC and How Gene Partners Can Change Morphology: Accurate Diagnosis Using Contemporary Modalities. Adv Anat Pathol 2022; 29:131-140. [PMID: 35180736 DOI: 10.1097/pap.0000000000000332] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Translocation renal cell carcinoma (tRCC) with TFE3 gene rearrangements has been born as a distinct entity 20 years ago. These relatively rare tumors were notable among other RCC subtypes because of their disproportionally high incidence among children and young adults. Initial reports were focused on describing unifying morphologic criteria and typical clinical presentation. Follow-up studies of ancillary immunohistochemical and hybridization techniques provided additional diagnostic tools allowing recognition of tRCC tumors in practice. However, a growing body of literature also expanded the clinicomorphologic spectrum of tRCCs, to include a significant morphologic overlap with other RCC variants thus blurring the diagnostic clarity of this entity. More recent molecular studies utilizing next-generation sequencing technology accelerated recognition of numerous novel gene partners fusing at different breakpoints with the TFE3 gene. Accumulating data indicates that morphologic and clinical heterogeneity of tRCC could be explained by fusion subtypes, and knowledge of TFE3 partnering genes may be important in predicting tumor behavior. Herein we provided a comprehensive analysis of ∼400 tRCC cases with known TFE3 fusion partners, estimated their relative incidence and summarized clinicomorphologic features associated with most common fusion subtypes. Our data was based on an extensive literature review and had a special focus on comparing immunohistochemistry, fluorescent in situ hybridization and contemporary molecular studies for the accurate diagnosis of tRCC.
Collapse
Affiliation(s)
- Maria S Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| |
Collapse
|
15
|
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
|
16
|
Roy P, van Peer SE, de Witte MM, Tytgat GAM, Karim-Kos HE, van Grotel M, van de Ven CP, Mavinkurve-Groothuis AMC, Merks JHM, Kuiper RP, Hol JA, Janssens GOR, de Krijger RR, Jongmans MCJ, Drost J, van der Steeg AFW, Littooij AS, Wijnen MHWA, van Tinteren H, van den Heuvel-Eibrink MM. Characteristics and outcome of children with renal tumors in the Netherlands: The first five-year's experience of national centralization. PLoS One 2022; 17:e0261729. [PMID: 35025887 PMCID: PMC8757983 DOI: 10.1371/journal.pone.0261729] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/08/2021] [Indexed: 01/20/2023] Open
Abstract
Around 6% of all childhood malignancies represent renal tumors, of which a majority includes Wilms tumor (WT). Although survival rates have improved over the last decades, specific patients are still at risk for adverse outcome. In the Netherlands, since 2015, pediatric oncology care for renal tumors has been centralized in the Princess Máxima Center for Pediatric Oncology. Here, we describe experiences of the first 5 years of centralized care and explore whether this influences the epidemiological landscape by comparing data with the Netherlands Cancer Registry (NCR). We identified all patients <19 years with a renal mass diagnosed between 01-01-2015 and 31-12-2019 in the Princess Máxima Center. Epidemiology, characteristics and management were analyzed. We identified 164 patients (including 1 patient who refused consent for registration), in our center with a suspicion of a renal tumor. The remaining 163 cases included WT (n = 118)/cystic partially differentiated nephroblastoma (n = 2)/nephrogenic rests only (n = 6) and non-WT (n = 37). In this period, the NCR included 138 children, 1 17-year-old patient was not referred to the Princess Máxima Center. Central radiology review (before starting treatment) was performed in 121/163 patients, and central pathology review in 148/152 patients that underwent surgery. Treatment stratification, according to SIOP/EpSSG protocols was pursued based on multidisciplinary consensus. Preoperative chemotherapy was administered in 133 patients, whereas 19 patients underwent upfront surgery. Surgery was performed in 152 patients, and from 133 biomaterial was stored. Centralization of care for children with renal tumors led to referral of all but 1 new renal tumor cases in the Netherlands, and leads to referral of very rare subtypes not registered in the NCR, that benefit from high quality diagnostics and multidisciplinary decision making. National centralization of care led to enhanced development of molecular diagnostics and other innovation-based treatments for the future.
Collapse
Affiliation(s)
- Prakriti Roy
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- * E-mail:
| | | | | | | | - Henrike E. Karim-Kos
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, The Netherlands
| | | | | | | | | | - Roland P. Kuiper
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Clinical Genetics, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Janna A. Hol
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Geert O. R. Janssens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Radiation Oncology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Ronald R. de Krijger
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pathology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Marjolijn C. J. Jongmans
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Clinical Genetics, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
| | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | | | - Annemieke S. Littooij
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children’s Hospital (UMCU), Utrecht, The Netherlands
| | | | - Harm van Tinteren
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | |
Collapse
|
17
|
González-Arboleda AA, Fernandez N, García-Perdomo HA. Genitourinary Tract Tumors in Children: An Update. Curr Pediatr Rev 2022; 18:166-178. [PMID: 35021978 DOI: 10.2174/1573396318666220111143902] [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: 07/09/2021] [Revised: 09/17/2021] [Accepted: 10/11/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Genitourinary tract tumors in children are less common than in adults. Most of these tumors have different genetic backgrounds, clinical presentation, and oncologic behavior than their adult counterpart. As a result of low prevalence in children, some of the treatment approaches and recommendations are based on treatment experience in adult patients. However, thanks to scientific and technological development, survival rates have risen considerably. OBJECTIVE This paper presents a review of the principal features of the tumors involving the genitourinary tract in children and an update in genetic background, diagnosis, and treatment. METHODS A narrative review was performed on published literature about genitourinary tract tumors in pediatric patients. Papers presented in English and Spanish literature were reviewed. PubMed, Science Direct, and SciELO databases were used to collect information and present this article. RESULTS Kidney tumors are the most common type of genitourinary tumors in children. Among those, Wilms tumor represents the majority of cases and shows the successful work of clinical trial groups studying this tumor type. Other tumors involving the genitourinary tract in children include Rhabdomyosarcoma, Transitional cell carcinoma, Testicular, and Adrenal tumors. CONCLUSION Genitourinary tract tumors in children represent significant morbidity and economic burden, so awareness in early diagnosis represents improvement in treatment, clinical, and oncological outcomes.
Collapse
Affiliation(s)
| | - Nicolás Fernandez
- Division of Urology, Seattle Children´s Hospital, The University of Washington, Seattle, WA, USA
| | - Herney Andrés García-Perdomo
- UROGIV Research Group, School of Medicine, Universidad del Valle, Cali, Colombia.,Division of Urology, Department of Surgery, School of Medicine, Universidad del Valle, Cali, Colombia
| |
Collapse
|
18
|
Integrated exome and RNA sequencing of TFE3-translocation renal cell carcinoma. Nat Commun 2021; 12:5262. [PMID: 34489456 PMCID: PMC8421377 DOI: 10.1038/s41467-021-25618-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/21/2021] [Indexed: 02/05/2023] Open
Abstract
TFE3-translocation renal cell carcinoma (TFE3-tRCC) is a rare and heterogeneous subtype of kidney cancer with no standard treatment for advanced disease. We describe comprehensive molecular characteristics of 63 untreated primary TFE3-tRCCs based on whole-exome and RNA sequencing. TFE3-tRCC is highly heterogeneous, both clinicopathologically and genotypically. ASPSCR1-TFE3 fusion and several somatic copy number alterations, including the loss of 22q, are associated with aggressive features and poor outcomes. Apart from tumors with MED15-TFE3 fusion, most TFE3-tRCCs exhibit low PD-L1 expression and low T-cell infiltration. Unsupervised transcriptomic analysis reveals five molecular clusters with distinct angiogenesis, stroma, proliferation and KRAS down signatures, which show association with fusion patterns and prognosis. In line with the aggressive nature, the high angiogenesis/stroma/proliferation cluster exclusively consists of tumors with ASPSCR1-TFE3 fusion. Here, we describe the genomic and transcriptomic features of TFE3-tRCC and provide insights into precision medicine for this disease. TFE3-translocation renal cell carcinoma (TFE3-tRCC) is a rare subtype of kidney cancer with no standard treatment options for the advanced disease. Here, the authors perform genomic and transcriptomic profiling of 63 untreated primary TFE3-tRCC tumours and reveal potential therapeutic targets.
Collapse
|
19
|
Pediatric onco-nephrology: time to spread the word : Part I: early kidney involvement in children with malignancy. Pediatr Nephrol 2021; 36:2227-2255. [PMID: 33245421 DOI: 10.1007/s00467-020-04800-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/28/2020] [Accepted: 09/25/2020] [Indexed: 12/29/2022]
Abstract
Onco-nephrology has been a growing field within the adult nephrology scope of practice. Even though pediatric nephrologists have been increasingly involved in the care of children with different forms of malignancy, there has not been an emphasis on developing special expertise in this area. The fast pace of discovery in this field, including the development of new therapy protocols with their own kidney side effects and the introduction of the CD19-targeted chimeric antigen receptor T cell (CAR-T) therapy, has introduced new challenges for general pediatric nephrologists because of the unique effects of these treatments on the kidney. Moreover, with the improved outcomes in children receiving cancer therapy come an increased number of survivors at risk for chronic kidney disease related to both their cancer diagnosis and therapy. Therefore, it is time for pediatric onco-nephrology to take its spot on the expanding subspecialties map in pediatric nephrology.
Collapse
|
20
|
Baniak N, Barletta JA, Hirsch MS. Key Renal Neoplasms With a Female Predominance. Adv Anat Pathol 2021; 28:228-250. [PMID: 34009777 DOI: 10.1097/pap.0000000000000301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Renal neoplasms largely favor male patients; however, there is a growing list of tumors that are more frequently diagnosed in females. These tumors include metanephric adenoma, mixed epithelial and stromal tumor, juxtaglomerular cell tumor, mucinous tubular and spindle cell carcinoma, Xp11.2 (TFE3) translocation-associated renal cell carcinoma, and tuberous sclerosis complex (somatic or germline) associated renal neoplasms. The latter category is a heterogenous group with entities still being delineated. Eosinophilic solid and cystic renal cell carcinoma is the best-described entity, whereas, eosinophilic vacuolated tumor is a proposed entity, and the remaining tumors are currently grouped together under the umbrella of tuberous sclerosis complex/mammalian target of rapamycin-related renal neoplasms. The entities described in this review are often diagnostic considerations when evaluating renal mass tissue on biopsy or resection. For example, Xp11.2 translocation renal cell carcinoma is in the differential when a tumor has clear cell cytology and papillary architecture and occurs in a young or middle-aged patient. In contrast, tuberous sclerosis complex-related neoplasms often enter the differential for tumors with eosinophilic cytology. This review provides an overview of the clinical, gross, microscopic, immunohistochemical, genetic, and molecular alterations in key renal neoplasms occurring more commonly in females; differential diagnoses are also discussed regardless of sex predilection.
Collapse
Affiliation(s)
- Nicholas Baniak
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Justine A Barletta
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
21
|
Uncommon Localization of Extrarenal Xp11.2 Translocation-associated Renal Cell Carcinoma (RCC): Case Report. Appl Immunohistochem Mol Morphol 2021; 28:e33-e35. [PMID: 28877071 DOI: 10.1097/pai.0000000000000576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The World Health Organization has recognized Xp11.2 translocation-associated renal cell carcinoma (RCC) as a distinct neoplasm that arises within the kidney. Although many reports of extrarenal carcinoma may be found in the literature, to the best of our knowledge, Xp11 translocation-associated RCC with intact kidneys has not been documented. This report describes a multilobulated right retroperitoneal soft tissue mass (7.9×5.3×12.6 cm) of a 37-year-old man complaining of abdominal pain in the right side. The patient underwent a computed tomography-guided biopsy. Microscopic evaluation reveals a tumor with papillary and sheaths architectures with cells revealing clear to eosinophilic cytoplasm. Immunohistochemical evaluation on the biopsy reveals that the tumor is positive for PAX-8, CD10, and TFE3. It is negative for CK7, EMA, Vimentin, RCC, CK8/18, D20, CD3, PLAP, OCT4, CD30, MART-1, Inhibin, S-100, HMB-45, Desmin, SMA, and DOG-1. The diagnosis was malignant epithelioid neoplasm and the diagnosis of translocation RCC was suggested. Excision was recommended. The patient underwent right radical nephrectomy with removal of this large mass. Pathologic examination showed a large cystic and solid, nonhomogenous mass with some necrotic areas, originating from the perirenal fat between the adrenal gland and the kidney. Microscopic features showed a tumor with papillary, rhabdoid, and clear cell features. Immunohistochemical stains showed that the tumor cells positively expressed AMACR, PAX-8, CD10, RCC, and TFE3, but were negative for cytokeratins, vimentin, HMB-45, desmin, SMA, EMA, and MSA. Cytogenetic studies confirmed the diagnosis of Xp11.2 translocation-associated RCC with positive TFE3 gene rearrangement. To the best of our knowledge, this type of extrarenal tumor has never been reported.
Collapse
|
22
|
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: 3.0] [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
|
23
|
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: 1.8] [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
|
24
|
Prachi, Aiyer HM. MiT family translocation renal cell carcinoma in an elderly male. INDIAN J PATHOL MICR 2021; 64:553-555. [PMID: 34341271 DOI: 10.4103/ijpm.ijpm_365_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Translocation-associated renal cell carcinoma (t-RCC) is a relatively uncommon subtype of renal cell carcinoma characterized by recurrent gene rearrangements involving the TFE3 or TFEB loci. TFE3 and TFEB are members of the microphthalmia transcription factor (MiT) family, which regulate differentiation in melanocytes and osteoclasts. Renal cell carcinomas (RCCs) associated with Xp11 translocations have gene fusions involving TFE3, which has multiple gene partners; RCCs with t(6:11) translocations have MALAT1-TFEB gene fusions. These tumors are histologically diverse, often have papillary, alveolar, and nested growth pattern with clear and eosinophilic cells and psammoma bodies and are seen commonly in children and young adults, accounting to 40% of pediatric RCCs and 1.6%-4% of adult RCCs. The mean and median patient age is 31 years. Thus, distinguishing t-RCC from its morphologic, immunophenotypic, and molecular mimics has important clinical implications. Directed ancillary testing is an essential aspect to t-RCC cases and may include a panel of immunohistochemical stains, such as PAX8, pancytokeratins, AMACR, CD10, and TFE-3. We, hereby report a case of TFE3 positiveXp11 translocation renal cell carcinoma in a 52-year-old male which is unusual.
Collapse
Affiliation(s)
- Prachi
- Department of Pathology, Dharamshila Narayana Superspeciality Hospital, New Delhi, India
| | - Hema Mailini Aiyer
- Department of Pathology, Dharamshila Narayana Superspeciality Hospital, New Delhi, India
| |
Collapse
|
25
|
Ray S, Jones R, Pritchard-Jones K, Dzhuma K, van den Heuvel-Eibrink M, Tytgat G, van der Beek J, Oades G, Murphy D. Pediatric and young adult renal cell carcinoma. Pediatr Blood Cancer 2020; 67:e28675. [PMID: 32869954 DOI: 10.1002/pbc.28675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 12/11/2022]
Abstract
Renal cell carcinoma (RCC) is rare in children but is the most common renal tumor in adults. Pediatric RCC has different clinical characteristics, histopathology, and treatment compared with adult disease. Databases were reviewed from inception to February 2020, identifying 32 publications pertaining to 350 patients under 27 years. Surgery is the cornerstone for cure in localized RCC. Lymph node dissection remains controversial. Conventional radiotherapy has no curative role in RCC; similarly, conventional chemotherapy has not proven to be effective in large cohorts. Pediatric metastatic RCC has a poor outlook. There are no published prospective studies demonstrating which adjuvant therapy could improve outcome. Sunitinib, a tyrosine kinase inhibitor, is recommended in this group despite limited evidence. This review provides an overview for pediatric RCC, including the evolving role of precision medicine.
Collapse
Affiliation(s)
- Satyajit Ray
- Department of Paediatric Oncology, Royal Hospital for Children, Glasgow, Scotland, UK
| | - Robert Jones
- Beatson West of Scotland Cancer Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | | | - Kristina Dzhuma
- University College London Institute of Child Health, London, UK
| | | | - Godelieve Tytgat
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Grenville Oades
- Department of Uro-Oncology, Queen Elizabeth University Hospital, Glasgow, Scotland, UK
| | - Dermot Murphy
- Department of Paediatric Oncology, Royal Hospital for Children, Glasgow, Scotland, UK
| |
Collapse
|
26
|
Catic A, Kurtovic-Kozaric A, Sophian A, Mazur L, Skenderi F, Hes O, Rohan S, Rakheja D, Kogan J, Pins MR. KANK1-NTRK3 fusions define a subset of BRAF mutation negative renal metanephric adenomas. BMC MEDICAL GENETICS 2020; 21:202. [PMID: 33046021 PMCID: PMC7552490 DOI: 10.1186/s12881-020-01143-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/09/2020] [Indexed: 12/27/2022]
Abstract
Background Metanephric adenoma (MA) is a rare benign renal neoplasm. On occasion, MA can be difficult to differentiate from renal malignancies such as papillary renal cell carcinoma in adults and Wilms̕ tumor in children. Despite recent advancements in tumor genomics, there is limited data available regarding the genetic alterations characteristic of MA. The purpose of this study is to determine the frequency of metanephric adenoma cases exhibiting cytogenetic aberration t (9;15)(p24;q24), and to investigate the association between t (9,15) and BRAF mutation in metanephric adenoma. Methods This study was conducted on 28 archival formalin fixed paraffin-embedded (FFPE) specimens from patients with pathologically confirmed MA. Tissue blocks were selected for BRAF sequencing and fluorescent in situ hybridization (FISH) analysis for chromosomal rearrangement between KANK1 on chromosome 9 (9p24.3) and NTRK3 on chromosome 15 (15q25.3), which was previously characterized and described in two MA cases. Results BRAFV600E mutation was identified in 62% of our cases, 9 (38%) cases were BRAFWT, and 4 cases were uninformative. Of the 20 tumors with FISH results, two (10%) were positive for KANK1-NTRK3 fusion. Both cases were BRAFWT suggesting mutual exclusivity of BRAFV600E and KANK1-NTRK3 fusion, the first such observation in the literature. Conclusions Our data shows that BRAF mutation in MA may not be as frequent as suggested in the literature and KANK-NTRK3 fusions may account for a subset of BRAFWT cases in younger patients. FISH analysis for KANK1-NTRK3 fusion or conventional cytogenetic analysis may be warranted to establish the diagnosis of MA in morphologically and immunohistochemically ambiguous MA cases lacking BRAF mutations.
Collapse
Affiliation(s)
- Aida Catic
- Department of Cytogenetics, ACL Laboratories, Rosemont, IL, USA. .,Department of Genetics and Bioengineering, International Burch University, Francuske revolucije bb, Ilidza, 71000, Sarajevo, Bosnia and Herzegovina.
| | - Amina Kurtovic-Kozaric
- Department of Genetics and Bioengineering, International Burch University, Francuske revolucije bb, Ilidza, 71000, Sarajevo, Bosnia and Herzegovina.,Department of Clinical Pathology, Cytology and Human Genetics, Clinical Center of the University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Ardis Sophian
- Department of Cytogenetics, ACL Laboratories, Rosemont, IL, USA
| | - Lech Mazur
- Department of Cytogenetics, ACL Laboratories, Rosemont, IL, USA
| | - Faruk Skenderi
- Department of Clinical Pathology, Cytology and Human Genetics, Clinical Center of the University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Ondrej Hes
- Department of Pathology, Charles University Hospital Pilsen, Pilsen, Czech Republic
| | - Stephen Rohan
- Department of Pathology, Saint Joseph Hospital, Denver, CO, USA
| | - Dinesh Rakheja
- Department of Pathology and Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Departments of Pathology and Laboratory Medicine, Children's Health, Dallas, TX, USA
| | - Jillene Kogan
- Department of Cytogenetics, ACL Laboratories, Rosemont, IL, USA.,Department of Pathology, Advocate Lutheran General Hospital, Park Ridge, IL, USA.,Department of Pathology, Chicago Medical School of Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.,Advocate Medical Group Genetics, Park Ridge, IL, USA
| | - Michael R Pins
- Department of Pathology, Advocate Lutheran General Hospital, Park Ridge, IL, USA.,Department of Pathology, Chicago Medical School of Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
27
|
Xp11.2 translocation renal cell carcinoma with TFE3 gene fusion in the elderly: case report and literature review. Int Cancer Conf J 2020; 9:182-186. [PMID: 32903928 DOI: 10.1007/s13691-020-00430-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/26/2020] [Indexed: 10/23/2022] Open
Abstract
A 68-year-old man was followed up with chronic kidney disease. Follow-up CT incidentally detected a tumor at the left kidney and multiple small nodular shadows in the lungs bilaterally. The patient underwent needle biopsy and was diagnosed with Xp11.2 translocation renal cell carcinoma (RCC) pathologically. Hence, laparoscopic nephrectomy was performed. Fluorescence in situ hybridization analysis revealed a break-apart of the transcription factor E3 (TFE3) genes in the left tumor. After 2 months postoperatively, nivolumab and ipilimumab were administered thrice intravenously, considering the intermediate risk by the IMDC risk classification. However, pleural effusion occurred but was removed adequately. Lung metastasis decreased, but new metastasis occurred at the left iliopsoas muscle. Target therapy was performed with axitinib. Unfortunately, he died 6 months later postoperatively. These tumors commonly occur in children than in adults, and very rare in elderly patients. Xp11.2 translocation RCC in the elderly has a poorer prognosis than that in children. To date, no effective treatment for Xp11.2 translocation RCC has been established.
Collapse
|
28
|
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: 2.4] [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
|
29
|
Abdulfatah E, Kennedy JM, Hafez K, Davenport MS, Xiao H, Weizer AZ, Palapattu GS, Morgan TM, Mannan R, Wang XM, Dhanasekaran SM, Kaffenberger SD, Spratt DE, Kunju L, Wu A, Lew M, Udager AM, Chinnaiyan AM, Mehra R. Clinicopathological characterisation of renal cell carcinoma in young adults: a contemporary update and review of literature. Histopathology 2020; 76:875-887. [PMID: 31872452 DOI: 10.1111/his.14051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/21/2019] [Indexed: 11/28/2022]
Abstract
AIMS Renal cell carcinomas are relatively rare in children and young adults. While well characterised in adults, the morphological and molecular characterisation of these tumours in young patients is relatively lacking. The objective of this study was to explore the spectrum of renal cell carcinoma (RCC) subtypes in children and young adults and to determine their clinico-pathological, immunohistochemical and molecular characteristics by evaluating a large retrospective cohort of renal cell carcinoma patients age 30 years or younger. METHODS AND RESULTS Sixty-eight cases with confirmed diagnosis of renal cell carcinoma at age 30 years or younger were identified at our institution. Clear cell carcinoma accounted for the most common subtype seen in this age group. Translocation renal cell carcinoma and rare familial syndrome subtypes such as succinate dehydrogenase deficient renal cell carcinoma and tuberous sclerosis complex-associated renal cell carcinoma were found relatively more frequently in this cohort. Despite applying the 2016 WHO classification criteria, a high proportion of the tumours in our series remained unclassified. CONCLUSIONS Our results suggest that renal cell carcinoma in children and young adults is a relatively rare disease that shares many histological similarities to renal cell carcinoma occurring in adults and yet demonstrate some unique clinical-pathological differences. Microphthalmia-associated transcription (MiT) family translocation RCC and rare familial syndrome subtypes are relatively more frequent in the paediatric and adolescent age groups than in adults. Clear cell RCC still accounted for the most common subtype seen in this age group. MiT family translocation RCC patients presented with advanced stage disease and had poor clinical outcomes. The large and heterogeneous subgroup of unclassified renal cell carcinoma contains phenotypically distinct tumours with further potential for future subcategories in the renal cell carcinoma classification.
Collapse
Affiliation(s)
- Eman Abdulfatah
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - John M Kennedy
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Khaled Hafez
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Matthew S Davenport
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Radiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hong Xiao
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alon Z Weizer
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ganesh S Palapattu
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Todd M Morgan
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Rahul Mannan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.,Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Xiao-Ming Wang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.,Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Saravana M Dhanasekaran
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.,Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | | | - Daniel E Spratt
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lakshmi Kunju
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Angela Wu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Madelyn Lew
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Aaron M Udager
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.,Michigan Center for Translational Pathology, Ann Arbor, MI, USA.,Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA.,Michigan Center for Translational Pathology, Ann Arbor, MI, USA.,Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA.,Howard Hughes Medical Institute, Ann Arbor, MI, USA
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.,Michigan Center for Translational Pathology, Ann Arbor, MI, USA.,Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Liao X, Abu-Farsakh SH, Zhang D. Sarcomatoid Renal Cell Carcinoma With Unusual Metastasis to the Small Intestine Manifesting as Perforated Appendicitis. In Vivo 2020; 33:2225-2228. [PMID: 31662560 DOI: 10.21873/invivo.11726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/09/2019] [Accepted: 09/19/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Sarcomatoid renal cell carcinoma is a rare form of dedifferentiated carcinoma with a high metastatic rate and adverse prognosis. Common sites of metastasis include lymph nodes, lung, liver and bone. We report a case of sarcomatoid renal cell carcinoma with unusual metastasis to the small intestine in a 65-year-old female with a history of clear-cell renal cell carcinoma with focal sarcomatoid transformation. CASE REPORT The patient presented to the Emergency Department with worsening abdominal pain. Imaging showed perforated acute appendicitis, however, diagnostic laparoscopy found no evidence of appendicitis, but a small punctate perforation in the small intestine. Gross examination of the small intestine showed a 2 cm tan-white lobular firm lesion at the perforation site involving the full thickness of the wall. Histological examination revealed a high-grade spindle-cell neoplasm with hyperchromatic and pleomorphic nuclei, frequent mitotic figures, and necrosis. Immunohistochemically, the tumor cells were positive for CD10 and carbonic anhydrase 9, but negative for pan-cytokeratin, epithelial membrane antigen, paired box gene 8, renal cell carcinoma, desmin, smooth-muscle actin, c-KIT, discovered on gastrointestinal stromal tumor protein 1, CD34, and S100. Molecular studies showed that the tumor cells were microsatellite stable but harbored mutations in polybromo-1, telomerase reverse transcriptase, and von Hippel-Lindau genes, supporting renal cell carcinoma in nature. The patient received radiation therapy but unfortunately died after one month due to rapid disease progression. CONCLUSION This was a rare and challenging case of sarcomatoid renal cell carcinoma metastasis to the small intestine with loss of some renal cell carcinoma markers, reinforcing the aggressive nature of this entity and the importance of correlating findings with the prior history for reaching correct diagnosis.
Collapse
Affiliation(s)
- Xiaoyan Liao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, U.S.A
| | - Sohaib H Abu-Farsakh
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, U.S.A
| | - Dongwei Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, U.S.A.
| |
Collapse
|
31
|
Major Subtypes of Renal Cell Carcinoma. KIDNEY CANCER 2020. [DOI: 10.1007/978-3-030-28333-9_5] [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]
|
32
|
MacLennan GT, Cheng L. Five decades of urologic pathology: the accelerating expansion of knowledge in renal cell neoplasia. Hum Pathol 2019; 95:24-45. [PMID: 31655169 DOI: 10.1016/j.humpath.2019.09.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023]
Abstract
Those who are knowledgeable in cosmology inform us that the expansion of the universe is such that the velocity at which a distant galaxy is receding from the observer is continually increasing with time. We humbly paraphrase that as "The bigger the universe gets, the faster it gets bigger." This is an interesting analogy for the expansion of knowledge in the field of renal tumor pathology over the past 30 to 50 years. It is clear that a multitude of dedicated investigators have devoted incalculable amounts of time and effort to the pursuit of knowledge about renal epithelial neoplasms. As a consequence of the contributions of numerous investigators over many decades, the most recent World Health Organization classification of renal neoplasms includes about 50 well defined and distinctive renal tumors, as well as various miscellaneous and metastatic tumors. In addition, a number of emerging or provisional new entities are under active investigation and may be included in future classifications. In this review, we will focus on a number of these tumors, tracing as accurately as we can the origins of their discovery, relating relevant additions to the overall knowledge base surrounding them, and in some instances addressing changes in nomenclature.
Collapse
Affiliation(s)
- Gregory T MacLennan
- Department of Pathology and Laboratory Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH.
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
33
|
Gandhi JS, Malik F, Amin MB, Argani P, Bahrami A. MiT family translocation renal cell carcinomas: A 15th anniversary update. Histol Histopathol 2019; 35:125-136. [PMID: 31489603 DOI: 10.14670/hh-18-159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Microphthalmia (MiT) family translocation renal cell carcinomas (RCCs) are a heterogeneous category of renal tumors which all express MiT transcription factors, typically from chromosomal translocation and rarely from gene amplification. This tumor family has two major subtypes [i.e., Xp11 translocation RCC and t(6;11) RCC] and several related neoplasms (i.e., TFEB amplification RCC and melanotic Xp11 translocation renal cancers). Increased understanding of the clinical, pathological, molecular and prognostic heterogeneity of these tumors, since their official recognition in 2004, provides the opportunity to identify prognostic biomarkers and to understand the reasons for tumor aggression. We will review the literature from the past 15 years and highlight the need for a greater understanding of the molecular mechanisms underpinning heterogeneous tumor behavior.
Collapse
Affiliation(s)
- Jatin S Gandhi
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Faizan Malik
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mahul B Amin
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Pedram Argani
- Department of Pathology, John Hopkins University, Baltimore, MD, USA
| | - Armita Bahrami
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN, USA.,Departments of Pathology and Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
34
|
Kato I, Furuya M, Baba M, Kameda Y, Yasuda M, Nishimoto K, Oyama M, Yamasaki T, Ogawa O, Niino H, Nakaigawa N, Yano Y, Sakamoto K, Urata Y, Mikami K, Yamasaki S, Tanaka R, Takagi T, Kondo T, Nagashima Y. RBM10-TFE3 renal cell carcinoma characterised by paracentric inversion with consistent closely split signals in break-apart fluorescence in-situ hybridisation: study of 10 cases and a literature review. Histopathology 2019; 75:254-265. [PMID: 30908700 DOI: 10.1111/his.13866] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/20/2019] [Indexed: 01/25/2023]
Abstract
AIMS Xp11 rearrangement in renal cell carcinoma (RCC) typically involves gene fusion to the gene encoding transcription factor E3 (TFE3), a member of the microphthalmia-associated transcription factor family on chromosome Xp11.2. Dual-colour break-apart fluorescence in-situ hybridisation (FISH) is recommended to confirm histological diagnoses. Recently, RNA-binding motif protein 10 (RBM10), encoded by a gene on chromosome Xp11.3, was identified as a chimeric partner of TFE3; thus, RBM10-TFE3 fusion results from paracentric inversion. RBM10-TFE3 RCC may yield a false-negative result in FISH analysis of TFE3 expression. The aim of the present study was to investigate the clinicopathological features of RBM10-TFE3 RCC. METHODS AND RESULTS Ten patients with RBM10-TFE3 RCC aged 31-71 years were investigated. Histological analysis, immunostaining, dual-colour break-apart FISH for TFE3, reverse transcription polymerase chain reaction and sequencing analysis were performed. No patient had a history of exposure to chemotherapy. Two of these patients died of RCC, and three were alive but developed metastases. Microscopically, the tumours were composed of a mixed architecture of tubulocystic and papillary patterns with scattered psammoma bodies. The tumours showed strong nuclear immunoreactivity for TFE3. FISH showed consistent closely spaced split signals in the RCCs of four patients, and polysomic signals with occasional closely spaced split signals in the RCCs of six patients. Of the latter six patients, five had renal failure, and four developed tumours in kidneys subjected to haemodialysis. CONCLUSIONS The present study suggests that the carcinogenesis of RBM10-TFE3 RCC in some, but not all, patients may be associated with chronic kidney disease. The aggressive nature of RBM10-TFE3 RCC should be considered, as five patients experienced metastases.
Collapse
Affiliation(s)
- Ikuma Kato
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mitsuko Furuya
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masaya Baba
- International Research Centre for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoichi Kameda
- Department of Pathology, Ashigarakami Hospital, Kanagawa, Japan
| | - Masanori Yasuda
- Department of Pathology, Saitama Medical University International Medical Centre, Saitama, Japan
| | - Koshiro Nishimoto
- Department of Urological Oncology, Saitama Medical University International Medical Centre, Saitama, Japan
| | - Masafumi Oyama
- Department of Urological Oncology, Saitama Medical University International Medical Centre, Saitama, Japan
| | - Toshinari Yamasaki
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hitoshi Niino
- Department of Pathology, National Hospital Organization Yokohama Medical Centre, Yokohama, Japan
| | - Noboru Nakaigawa
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuta Yano
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Centre Komagome Hospital, Tokyo, Japan
| | - Kazumasa Sakamoto
- Department of Urology, Tokyo Metropolitan Cancer and Infectious Diseases Centre Komagome Hospital, Tokyo, Japan
| | - Yoji Urata
- Department of Pathology, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Kazuya Mikami
- Department of Urology, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | | | - Reiko Tanaka
- Medical Mycology Research Centre, Chiba University, Chiba, Japan
| | - Toshio Takagi
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Tsunenori Kondo
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Yoji Nagashima
- Department of Surgical Pathology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
35
|
Perera RM, Di Malta C, Ballabio A. MiT/TFE Family of Transcription Factors, Lysosomes, and Cancer. ANNUAL REVIEW OF CANCER BIOLOGY 2019; 3:203-222. [PMID: 31650096 PMCID: PMC6812561 DOI: 10.1146/annurev-cancerbio-030518-055835] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancer cells have an increased demand for energy sources to support accelerated rates of growth. When nutrients become limiting, cancer cells may switch to nonconventional energy sources that are mobilized through nutrient scavenging pathways involving autophagy and the lysosome. Thus, several cancers are highly reliant on constitutive activation of these pathways to degrade and recycle cellular materials. Here, we focus on the MiT/TFE family of transcription factors, which control transcriptional programs for autophagy and lysosome biogenesis and have emerged as regulators of energy metabolism in cancer. These new findings complement earlier reports that chromosomal translocations and amplifications involving the MiT/TFE genes contribute to the etiology and pathophysiology of renal cell carcinoma, melanoma, and sarcoma, suggesting pleiotropic roles for these factors in a wider array of cancers. Understanding the interplay between the oncogenic and stress-adaptive roles of MiT/TFE factors could shed light on fundamental mechanisms of cellular homeostasis and point to new strategies for cancer treatment.
Collapse
Affiliation(s)
- Rushika M Perera
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143, USA
| | - Chiara Di Malta
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, 80138Naples, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, 80138Naples, Italy
- Department of Molecular and Human Genetics and Neurological Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
36
|
Schneider M, Dinkelborg K, Xiao X, Chan-Smutko G, Hruska K, Huang D, Sagar P, Harisinghani M, Iliopoulos O. Early onset renal cell carcinoma in an adolescent girl with germline FLCN exon 5 deletion. Fam Cancer 2019. [PMID: 28623476 DOI: 10.1007/s10689-017-0008-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Birt-Hogg-Dube (BHD) disease is an autosomal dominant cancer syndrome characterized by benign skin tumors, renal cancer and spontaneous pneumothorax and is caused by mutations in the Folliculin (FLCN) gene. Benign skin tumors and pneumothorax occur in the majority of patients affected by BHD syndrome, but only 30-45% of them develop renal cell carcinoma (RCC) with a median age of diagnosis at 48. The earliest onset of RCC in a BHD patient has been reported at age 20. Here we report a case of a 14 year-old patient with germline FLCN mutation leading to an early-onset bulky RCC that could not be classified strictly according to existing histological types. Germline genetic testing revealed a deletion at FLCN exon 5. The father of the patient was identified as the asymptomatic carrier. We report the youngest patient with BHD-related RCC. This early onset presentation supports genetic testing of at-risk patients and initiation of imaging surveillance for RCC in early adolescence. In addition, future studies are necessary to understand the determinants of reduced penetrance in BHD disease.
Collapse
Affiliation(s)
- Meike Schneider
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA.,Harvard Medical School, Boston, MA, 02115, USA.,MGH Center for Cancer Research, 149 13th Street, Charlestown, MA, 02129, USA
| | - Katja Dinkelborg
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA.,Harvard Medical School, Boston, MA, 02115, USA.,MGH Center for Cancer Research, 149 13th Street, Charlestown, MA, 02129, USA
| | - Xiuli Xiao
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Gayun Chan-Smutko
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
| | | | | | - Pallavi Sagar
- Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Mukesh Harisinghani
- Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Othon Iliopoulos
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA. .,Harvard Medical School, Boston, MA, 02115, USA. .,MGH Center for Cancer Research, 149 13th Street, Charlestown, MA, 02129, USA.
| |
Collapse
|
37
|
Concomitant presentation and surgical management of an abdominal aortic aneurysm and translocation XP11.2 associated renal cell carcinoma in a female infant. JOURNAL OF PEDIATRIC SURGERY CASE REPORTS 2018. [DOI: 10.1016/j.epsc.2018.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
38
|
Cajaiba MM, Dyer LM, Geller JI, Jennings LJ, George D, Kirschmann D, Rohan SM, Cost NG, Khanna G, Mullen EA, Dome JS, Fernandez CV, Perlman EJ. The classification of pediatric and young adult renal cell carcinomas registered on the children's oncology group (COG) protocol AREN03B2 after focused genetic testing. Cancer 2018; 124:3381-3389. [PMID: 29905933 DOI: 10.1002/cncr.31578] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/30/2018] [Accepted: 05/07/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Renal cell carcinomas (RCCs) are rare in young patients. Knowledge of their pathologic and molecular spectrum remains limited, and no prospective studies have been performed to date in this population. This study analyzes patients diagnosed with RCC who were prospectively enrolled in the AREN03B2 Children's Oncology Group (COG). The objective was to classify these tumors with the aid of focused genetic testing and to characterize their features. METHODS All tumors registered as RCC by central review were retrospectively re-reviewed and underwent additional ancillary studies. Tumors were classified according to the 2016 World Health Organization classification system when possible. RESULTS In total, 212 tumors were identified, and these were classified as microphthalmia transcription factor (MiT) translocation RCC (MiT-RCC) (41.5%), papillary RCC (16.5%), renal medullary carcinoma (12.3%), chromophobe RCC (6.6%), clear cell RCC (3.3%), fumarate hydratase-deficient RCC (1.4%), and succinate dehydrogenase-deficient RCC (0.5%). Other subtypes included tuberous sclerosis-associated RCC (4.2%), anaplastic lymphoma kinase (ALK)-rearranged RCC (3.8%), thyroid-like RCC (1.4%), myoepithelial carcinoma (0.9%), and unclassified (7.5%). MiT-RCCs were classified as either transcription factor E3 (TFE3) (93.2%) or EB (TFEB) (6.8%) translocations, and characterization of fusion partners was possible in most tumors. CONCLUSIONS The current study delineates the frequency of distinct RCC subtypes in a large prospective series of young patients and contributes knowledge to the diagnostic, clinical, and genetic features of MiT-RCC, the most common subtype among this age group. The identification of rare subtypes expands the spectrum of RCC in young patients, supporting the need for a thorough diagnostic workup. These studies may aid in the introduction of specific therapies for different RCC subtypes in the future. Cancer 2018. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Mariana M Cajaiba
- Department of Pathology and Laboratory Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago,`, Chicago, Illinois
| | - Lisa M Dyer
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - James I Geller
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Lawrence J Jennings
- Department of Pathology and Laboratory Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago,`, Chicago, Illinois
| | - David George
- Department of Pathology and Laboratory Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago,`, Chicago, Illinois
| | - Dawn Kirschmann
- Department of Pathology and Laboratory Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago,`, Chicago, Illinois
| | - Stephen M Rohan
- Department of Pathology, Colorado Pathology Consultants, Denver, Colorado
| | - Nicholas G Cost
- Department of Surgery, University of Colorado, Denver, Colorado
| | - Geetika Khanna
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Elizabeth A Mullen
- Department of Pediatric Oncology, Children's Hospital, Boston, Massachusetts
| | - Jeffrey S Dome
- Division of Oncology, Children's National Medical Center, Washington, District of Columbia
| | | | - Elizabeth J Perlman
- Department of Pathology and Laboratory Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago,`, Chicago, Illinois
| |
Collapse
|
39
|
Gong P, Zhuang Q, Wang K, Xu R, Chen Y, Wang X, Yin S. Adult-onset renal cell carcinoma associated with Xp11.2 translocation/TFE3 gene fusion: 3 case reports and review of literature. Medicine (Baltimore) 2018; 97:e11023. [PMID: 29901594 PMCID: PMC6024875 DOI: 10.1097/md.0000000000011023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
RATIONALE Renal cell carcinoma associated with Xp11.2 translocations/TFE3 gene fusions is a rare subtype of renal cell carcinoma. This predominantly occurs in juveniles, but rarely seen in adults with lymph node or organic metastasis and a worsened prognosis. PATIENTS CONCERNS Herein, we presented 3 adult cases of Xp11-RCC. Two patients were in early stage and good condition, and the third patient had lymph node metastasis but showed no recurrence after a 3-month follow-up. DIAGNOSES Case 1: A 50-year-old female without any lumbago and gross hematuria was incidentally detected by left renal mass by ultrasonography. Case 2: A 31-year-old female with 2-year hemodialysis was detected with right renal carcinoma during preoperative examination of renal transplant. Case 3: A 45-year-old male with right lumbago for 1 month was detected with a mass in the lower pole of right kidney by ultrasonography. INTERVENTION The characteristics of these 3 images are not consistent with each other, and showed some differences with the previous ones. OUTCOMES All these 3 patients underwent laparoscopic radical nephrectomy, and case 1 patient underwent renal hilar lymphnode dissection at the same time. Immunohistochemistry was performed on all the 3 tumors, revealing that the tumor cells were positive for TFE3 and Melan-A. Case 1 showed lymph node metastasis, and received mTOR inhibitors. The 3 patients had no recurrent and new metastasis in other organs after follow-up for 3 months, 2 months, and 11 months, respectively. LESSONS Whether the adult-onset Xp-RCC has an aggressive clinical course still remains controversial. Characteristics of the images of the 3 adult cases showed some uniformity but still have some differences. Immunohistochemistry results revealed tumor cell positive for TFE3, but have no consistency in carbonic anhydrase IX, CD117, Ki67, CK8/18 AE1/AE3 and so on. Therefore, the uniform and definitive diagnostic standards of the tumors are uncertain. Hence, more cases and findings are required to elaborate the standards of all the tumor subtypes. Vascular endothelial growth factor-targeted therapy showed some efficacious results in patients with metastasis, but more useful treatments are warranted.
Collapse
|
40
|
Li Y, Reuter VE, Matoso A, Netto GJ, Epstein JI, Argani P. Re-evaluation of 33 'unclassified' eosinophilic renal cell carcinomas in young patients. Histopathology 2018; 72:588-600. [PMID: 28898443 PMCID: PMC7582203 DOI: 10.1111/his.13395] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 09/07/2017] [Indexed: 12/16/2022]
Abstract
AIMS We sought to determine if some unclassified renal cell carcinomas (RCCs) in children and young adults that are characterised by predominantly eosinophilic cytoplasm are related to the recently described succinate dehydrogenase (SDH)-deficient RCC, fumarate hydratase (FH)-deficient RCC or eosinophilic solid and cystic (ESC) RCC. METHODS AND RESULTS We reviewed 33 unclassified RCCs with predominantly eosinophilic cytoplasm in patients aged 35 years or younger. Immunohistochemistry (IHC) for SDHB, FH and CK20 (a marker of ESC) was performed in all cases. IHC for 2-succinocysteine (2SC) was performed on RCC with loss of FH labelling. Four RCC (12%) (median age 18 years) demonstrated loss of FH labelling as well as aberrant 2SC labelling, and were thus classified as FH-deficient RCCs. Importantly, none of these cases demonstrated the characteristic macronucleoli typical of FH-deficient RCC. Eight RCC (24%) (median age 20.5 years) demonstrated loss of SDHB and were reclassified as SDH-deficient RCCs. Importantly, only four of eight SDH-deficient RCC demonstrated the characteristic cytoplasmic vacuoles and inclusions of typical SDH-deficient RCC. Ten RCC (30%) (median age 27 years) were reclassified as ESC RCCs. Four of 10 ESC RCC were multifocal (one bilateral), four of 10 ESC RCC occurred in males and one patient presented with liver and lung metastases, all not described previously in ESC. Eleven RCC (33%) remained unclassified. CONCLUSIONS Pathologists should have a low threshold for performing FH, SDHB and CK20 IHC when confronted with unclassified eosinophilic RCC or 'oncocytoma' in young patients.
Collapse
Affiliation(s)
- Yunjie Li
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - Victor E Reuter
- Memorial Sloan Kettering Cancer Center, Pathology, New York, NY, United States
| | - Andres Matoso
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - George J Netto
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
- University of Alabama at Birmingham School of Medicine, Birmingham,AL, USA
| | - Jonathan I Epstein
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - Pedram Argani
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| |
Collapse
|
41
|
Rupp NJ, Montironi R, Mischo A, Moch H. Clinical Trials for Specific Renal Cancer Subtypes—The Time Will Come! ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.eursup.2017.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
42
|
Pan X, Quan J, Zhao L, Li W, Wei B, Yang S, Lai Y. Xp11.2 translocation renal cell carcinoma with TFE3 gene fusion: A case report. Mol Clin Oncol 2017; 8:83-85. [PMID: 29399348 DOI: 10.3892/mco.2017.1497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/02/2017] [Indexed: 11/05/2022] Open
Abstract
Xp11.2 translocation renal cell carcinoma (RCC) with transcription factor E3 (TFE3) gene fusion is a rare tumor, and the prognosis of this tumor is poorer compared with that of other subtypes of RCC. The patient presented herein was a 70-year-old man who presented with a solid mass sized ~8.2×6.1 cm in the right kidney and underwent radical right nephrectomy. Following pathological and immunohistochemical (IHC) examination and fluorescent in situ hybridization (FISH), the patient was diagnosed with Xp11.2 translocation RCC with TFE3 gene fusion. These tumors are more commonly encountered in children rather than in adults, and adult Xp11.2 translocation RCC is associated with a poorer prognosis compared with its pediatric counterpart. IHC assay and FISH are important diagnostic methods. However, there is currently no established effective treatment for Xp11.2 RCC.
Collapse
Affiliation(s)
- Xiang Pan
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China.,The First Clinical College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Jing Quan
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China.,The First Clinical College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Liwen Zhao
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China.,The First Clinical College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Wenhua Li
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Benlin Wei
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Shangqi Yang
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Yongqing Lai
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
43
|
Perrino CM, Grignon DJ, Williamson SR, Idrees MT, Eble JN, Cheng L. Morphological spectrum of renal cell carcinoma, unclassified: an analysis of 136 cases. Histopathology 2017; 72:305-319. [PMID: 28833389 DOI: 10.1111/his.13362] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 08/16/2017] [Indexed: 12/28/2022]
Abstract
AIMS Renal cell carcinoma, unclassified (RCCU) is a category that includes a morphologically and biologically heterogeneous group of tumours that are unable to be diagnosed as other well-defined entities. We aim to describe the morphological findings of tumours within this category and to determine the most frequent morphological features leading to classification difficulty. METHODS AND RESULTS One hundred and thirty-six cases of RCCU were examined. Patients ranged in age from 23 to 87 years. Seventy-seven patients were men and 59 were women. International Society of Urological Pathology (ISUP) grade was most commonly 3 (n = 66), followed by 2 (n = 42) and 4 (n = 28). Tumour size ranged from 0.6 to 24.9 cm. The American Joint Committee on Cancer (AJCC) pathological T categories included pT1a (n = 50), pT1b (n = 14), pT2a (n = 7), pT2b (n = 4), pT3a (n = 50) and pT4 (n = 9). Forty-four cases included lymph node(s), 41% of which (n = 18) had metastases. Tumours were assessed for a variety of histological features and assigned to the following morphological groups: predominantly oncocytoma/chromophobe RCC-like; clear cell RCC-like; papillary RCC-like; collecting duct-like; and pure sarcomatoid differentiation. The majority of the oncocytoma/chromophobe and clear cell RCC-like phenotypes were low stage (pT1 or pT2). The papillary RCC-like, collecting duct-like and pure sarcomatoid phenotypes were mainly high stage (pT3 or pT4). CONCLUSIONS Renal cell carcinoma, unclassified is a term that encompasses tumours with a variety of morphological features and a wide biological spectrum. The most common source of diagnostic difficulty was tumours composed of predominantly eosinophilic cells.
Collapse
Affiliation(s)
- Carmen M Perrino
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David J Grignon
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sean R Williamson
- Department of Pathology and Laboratory Medicine, Henry Ford Health System, Detroit, MI, USA.,Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, USA.,Department of Pathology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Muhammad T Idrees
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John N Eble
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Liang Cheng
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Urology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
44
|
Hirokawa M, Miyauchi A, Kihara M, Kudo T, Hashimoto Y, Suzuki S, Daa T, Vuong HG, Mitsutake N. Chromophobe renal cell carcinoma-like thyroid carcinoma: A novel clinicopathologic entity possibly associated with tuberous sclerosis complex. Endocr J 2017; 64:843-850. [PMID: 28680002 DOI: 10.1507/endocrj.ej17-0096] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
We report three cases of chromophobe renal cell carcinoma-like thyroid carcinoma as a novel clinicopathologic entity possibly associated with tuberous sclerosis complex. A 15-year-old female, a 19-year-old male, and a 21-year-old male presented with primary thyroid carcinoma. Two of the patients had associated tuberous sclerosis complex. Macroscopically, the carcinomas showed invasive growth. Histologically, the carcinoma cells showed a trabecular pattern with thin vascular stroma, and were characterized by abundant eosinophilic cytoplasm with perinuclear clearing, a prominent cell border, a wrinkled nuclear membrane, and binucleation, which are all features of chromophobe renal cell carcinoma. Immunohistochemically, the carcinoma cells were positive for thyroglobulin, TTF1, and PAX8, and negative for CD10, calcitonin, and carcinoembryonic antigen. Vascular invasion was visible in all cases, but distant metastasis was not detected during follow-up. The original pathological diagnoses of the three cases were widely invasive follicular thyroid carcinoma, poorly differentiated thyroid carcinoma, and oxyphilic variant of papillary thyroid carcinoma. Thus, the cases were similar to chromophobe renal cell carcinoma associated with tuberous sclerosis complex as they were characterized by histologic findings consistent with chromophobe renal cell carcinoma, occurrence in an adolescent or young adult, and favorable prognosis regardless of the presence of vascular invasion and an infiltrating growth pattern resembling poorly differentiated carcinoma. The etiopathogenesis also seemed to suggest the presence of the tuberous sclerosis complex genetic abnormality.
Collapse
Affiliation(s)
- Mitsuyoshi Hirokawa
- Department of Diagnostic Pathology and Cytology, Kuma Hospital, Kobe 650-0011, Japan
| | - Akira Miyauchi
- Department of Surgery, Kuma Hospital, Kobe 650-0011, Japan
| | - Minoru Kihara
- Department of Surgery, Kuma Hospital, Kobe 650-0011, Japan
| | - Takumi Kudo
- Department of Internal Medicine, Kuma Hospital, Kobe 650-0011, Japan
| | - Yuko Hashimoto
- Department of Diagnostic Pathology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Shinichi Suzuki
- Department of Thyroid and Endocrinology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Tsutomu Daa
- Department of Diagnostic Pathology, Faculty of Medicine, Oita University, Yufu 879-5593, Japan
| | - Huy Gia Vuong
- Department of Pathology, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Norisato Mitsutake
- Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| |
Collapse
|
45
|
Chang K, Qu Y, Dai B, Zhao JY, Gan H, Shi G, Zhu Y, Shen Y, Zhu Y, Zhang H, Ye D. PD-L1 expression in Xp11.2 translocation renal cell carcinoma: Indicator of tumor aggressiveness. Sci Rep 2017; 7:2074. [PMID: 28522811 PMCID: PMC5437001 DOI: 10.1038/s41598-017-02005-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/03/2017] [Indexed: 01/05/2023] Open
Abstract
Programmed death ligand-1 (PD-L1), a promising antitumor target, has proven clinical value against many malignancies. However, the PD-L1 content of Xp11.2 translocation renal cell carcinoma (Xp11.2 RCC) and its correlation with clinical outcomes remain unclear. This study aimed to investigate PD-L1 expression in Xp11.2 RCC and to assess its prognostic value. Formalin-fixed paraffin-embedded specimens from 36 adult patients that were histologically confirmed (by fluorescence in situ hybridization) were subjected to immunohistochemical analysis. Of the 36 Xp11.2 RCC patients, 9 (25.0%) had tumors with positive PD-L1 expression and 27 (75.0%) had tumors with negative PD-L1 expression. Positive PD-L1 expression correlated with advanced tumor stage (P = 0.001), regional lymph node metastasis (P < 0.001), and distant metastasis (P < 0.001). A multivariate analysis identified positive PD-L1 expression was an independent adverse prognostic factor for both progression free survival (hazard ratio: 3.7, P = 0.018) and overall survival (hazard ratio: 4.5, P = 0.034). The median PFS and OS for the whole cohort were 13.0 months (95% confidence interval [CI], 9.4-16.6 months) and 36.0 months (95% CI, 23.9-48.1 months), respectively. Our findings suggest that positive PD-L1 expression is indicative of worse clinical outcome in Xp11.2 RCC. Further studies are needed to explore the potential efficacy of targeting PD-L1 in Xp11.2 RCC.
Collapse
Affiliation(s)
- Kun Chang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Qu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jian-Yuan Zhao
- The State Key Laboratory of Genetic Engineering and Collaborative Innovation Center of Genetics & Development, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Hualei Gan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yiping Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yijun Shen
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
46
|
Choo MS, Jeong CW, Song C, Jeon HG, Seo SI, Hong SK, Byun SS, Chung JS, Hong SH, Hwang EC, Kim HH, Kwak C. Clinicopathologic Characteristics and Prognosis of Xp11.2 Translocation Renal Cell Carcinoma: Multicenter, Propensity Score Matching Analysis. Clin Genitourin Cancer 2017; 15:e819-e825. [PMID: 28549862 DOI: 10.1016/j.clgc.2017.04.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/08/2017] [Accepted: 04/14/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND We evaluated the clinicopathologic characteristics and prognosis of Xp11.2 translocation (Xp11.2t) renal cell carcinoma (RCC) from a multicenter study and compare them with clear-cell RCC using a propensity score matching analysis. PATIENTS AND METHODS Between 2004 and 2013, 8384 consecutive patients from 7 institutions who were diagnosed with RCC were reviewed, and the pathologically confirmed Xp11.2t cases were enrolled. The oncological outcomes of Xp11.2t were compared with those of clear-cell RCC by selecting matched cases using 1:3 propensity score matching methods in a precollected clear-cell RCC data set from our hospital. The patients were divided into 2 subgroups on the basis of age of onset, either before (early) or after (late) 45 years old. RESULTS Xp11.2t was found in 61 cases, corresponding to 0.72% of RCC cases for the 10 years. The mean age was 38.2 ± 19.4 years, and the mean tumor size was 6.2 ± 3.9 cm. The Xp11.2t cases were at more advanced stages and showed tendencies to involve lymph nodes at diagnosis. After the matching, there were no significant differences in recurrence-free and overall survival compared with clear-cell RCC. The age of incidence for Xp11.2t had a bimodal distribution, which was most common in the 30s and smaller peak in the 60s. Xp11.2t corresponded to a significantly worse prognosis for overall survival in late onset (after 45 years) subgroup (P = .038; hazard ratio, 3.199; 95% confidence interval, 1.065-9.609). CONCLUSION This neoplasm has more aggressive clinicopathologic features at diagnosis. In older patients with onset age > 45 years, Xp11.2t showed a significantly worse prognosis than clear-cell RCC.
Collapse
Affiliation(s)
- Min Soo Choo
- Department of Urology, Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Chang Wook Jeong
- Department of Urology, Seoul National University Hospital, Seoul, Korea
| | - Cheryn Song
- Department of Urology, Asan Medical Center, Seoul, Korea
| | - Hwang Gyun Jeon
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seong Il Seo
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung Kyu Hong
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Seok-Soo Byun
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jin Soo Chung
- Department of Urology, National Cancer Center, Goyang, Korea
| | - Sung-Hoo Hong
- Department of Urology, Kangnam St Mary's Hospital, Seoul, Korea
| | - Eu Chang Hwang
- Department of Urology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Hyeon Hoe Kim
- Department of Urology, Seoul National University Hospital, Seoul, Korea
| | - Cheol Kwak
- Department of Urology, Seoul National University Hospital, Seoul, Korea.
| | | |
Collapse
|
47
|
Catic A, Kurtovic-Kozaric A, Johnson SH, Vasmatzis G, Pins MR, Kogan J. A novel cytogenetic and molecular characterization of renal metanephric adenoma: Identification of partner genes involved in translocation t(9;15)(p24;q24). Cancer Genet 2017; 214-215:9-15. [PMID: 28595733 DOI: 10.1016/j.cancergen.2017.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/25/2017] [Accepted: 03/02/2017] [Indexed: 10/20/2022]
Abstract
Renal metanephric adenoma (MA) is a rare benign tumor frequently misclassified when microscopic features alone are applied. The correct classification of a renal tumor is critical for diagnostic, prognostic, and therapeutic purposes. Despite the advancements in cancer genomics, up until recently relatively few genetic alterations critical to MA development have been recognized. Recent data suggest that 90% of MA have BRAFV600E mutations; the genetics of the remaining 10% are unclear. To date, only one case of a chromosomal translocation, t(9;15)(p24;q24) associated with MA has been reported. However, the potential role of the KANK1 gene, which lies near the breakpoint of the short arm of chromosome 9p24, in the etiology of MA was not examined. We identified the same cytogenetic aberration utilizing molecular cytogenetic techniques in a 22-year-old female patient, and further investigated the genes involved in the translocation that might have contributed to tumorigenesis. A series of fluorescence in situ hybridization (FISH) probes identified the rearranged genes to be KANK1 on chromosome 9 (9p24.3) and NTRK3 on chromosome 15 (15q25.3). Mate-Pair genome sequencing validated the balanced translocation between 9p24.3 and 15q25.3, involving genes KANK1 and NTRK3, respectively. BRAFV600E mutational analysis was normal. Our findings indicate that gene fusions may be one mechanism by which functionally relevant genes are altered in the development of MA. Molecular and cytogenetic analyses have elucidated a novel genetic aberration, which helps to provide a better understanding of this genomic change and assist in diagnosis and classification of new subgroups/entities in metanephric adenomas.
Collapse
Affiliation(s)
- Aida Catic
- Department of Cytogenetics, ACL Laboratories, Rosemont, Illinois, USA; International Burch University, Department of Genetics and Bioengineering, Sarajevo, Bosnia and Herzegovina
| | - Amina Kurtovic-Kozaric
- International Burch University, Department of Genetics and Bioengineering, Sarajevo, Bosnia and Herzegovina; Department of Clinical Pathology, Cytology and Human Genetics, Clinical Center of the University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Sarah H Johnson
- Center for Individualized Medicine (CIM), Mayo Clinic, Rochester, Minnesota, USA
| | - George Vasmatzis
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael R Pins
- Department of Pathology, Advocate Lutheran General Hospital, Park Ridge, Illinois, USA; Department of Pathology, Chicago Medical School of Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Jillene Kogan
- Department of Cytogenetics, ACL Laboratories, Rosemont, Illinois, USA; Department of Pathology, Advocate Lutheran General Hospital, Park Ridge, Illinois, USA; Advocate Medical Group Genetics, Park Ridge, Illinois, USA.
| |
Collapse
|
48
|
Udager AM, Mehra R. Morphologic, Molecular, and Taxonomic Evolution of Renal Cell Carcinoma: A Conceptual Perspective With Emphasis on Updates to the 2016 World Health Organization Classification. Arch Pathol Lab Med 2017; 140:1026-37. [PMID: 27684973 DOI: 10.5858/arpa.2016-0218-ra] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Molecular and morphologic interrogation has driven a much-needed reexamination of renal cell carcinoma (RCC). Indeed, the recently released 2016 World Health Organization classification now recognizes 12 distinct RCC subtypes, as well as several other emerging/provisional RCC entities. From a clinical perspective, accurate RCC classification may have important implications for patients and their families, including prognostic risk stratification, targeted therapeutics selection, and identification for genetic testing. In this review, we provide a conceptual framework for approaching RCC diagnosis and classification by categorizing RCCs as tumors with clear cytoplasm, papillary architecture, and eosinophilic (oncocytic) cytoplasm. The currently recognized 2016 World Health Organization classification for RCC subtypes is briefly discussed, including new diagnostic entities (clear cell papillary RCC, hereditary leiomyomatosis and RCC-associated RCC, succinate dehydrogenase-deficient RCC, tubulocystic RCC, and acquired cystic disease-associated RCC) and areas of evolving RCC classification, such as transcription elongation factor B subunit 1 (TCEB1)-mutated RCC/RCC with angioleiomyoma-like stroma/RCC with leiomyomatous stroma, RCC associated with anaplastic lymphoma receptor tyrosine kinase (ALK) gene rearrangement, thyroidlike follicular RCC, and RCC in neuroblastoma survivors. For each RCC subtype, relevant clinical, molecular, gross, and microscopic findings are reviewed, and ancillary studies helpful for its differential diagnosis are presented, providing a practical approach to modern RCC classification.
Collapse
Affiliation(s)
- Aaron M Udager
- From the Department of Pathology (Drs Udager and Mehra) and the Comprehensive Cancer Center (Dr Mehra), University of Michigan Health System, Ann Arbor; and the Michigan Center for Translational Pathology, Ann Arbor (Dr Mehra)
| | | |
Collapse
|
49
|
Ellati RT, Abukhiran I, Alqasem K, Jasser J, Khzouz J, Bisharat T, Al-saidi I, Al-Daghmin A. Clinicopathologic Features of Translocation Renal Cell Carcinoma. Clin Genitourin Cancer 2017; 15:112-116. [DOI: 10.1016/j.clgc.2016.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 05/04/2016] [Accepted: 05/18/2016] [Indexed: 01/28/2023]
|
50
|
Abstract
The MiT family of translocation-associated renal cell carcinomas comprise approximately 40 % of renal cell carcinomas in young patients but only up to 4 % of renal cell carcinomas in adult patients. The Xp11.2 translocation-associated tumors are the most frequent and were included in the 2004 World Health Organization (WHO) classification. They contain a fusion of the TFE3 gene with ASPSCR1, PRCC, NONO, SPFQ or CLTC resulting in an immunohistochemically detectable nuclear overexpression of TFE3. The Xp11.2 translocation-associated renal cell carcinomas are characterized by ample clear cytoplasm, papillary architecture and abundant psammoma bodies. The TFEB translocation-associated renal cell carcinomas are much rarer and show a biphasic architecture. Fluorescence in situ hybridization permits the detection of a translocation by means of a break apart probe for the TFE3 and TFEB genes and is recommended for the diagnosis of renal cell carcinomas in patients under 30 years of age. The TFE3 and TFEB translocation-associated tumors are classified as MiT family translocation carcinomas in the new WHO classification.The rare renal cell carcinomas harboring an ALK rearrangement with fusion to VCL in young patients with sickle cell trait show a characteristic morphology and are listed in the new WHO classification as a provisional entity.
Collapse
|