1
|
Mahmoud M, Trabulsi M. Eosinophilic solid and cystic renal cell carcinoma: A case study with pathological correlates. Int J Surg Case Rep 2025; 126:110738. [PMID: 39700587 PMCID: PMC11722899 DOI: 10.1016/j.ijscr.2024.110738] [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: 10/25/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
INTRODUCTION AND SIGNIFICANCE Eosinophilic solid and cystic renal cell carcinoma (ESC RCC) is a rare renal tumor primarily associated with female patients and those with tuberous sclerosis complex (TSC). Despite lacking distinct clinical or radiological features, its unique histological characteristics allow for differentiation from other renal neoplasms. While it often exhibits indolent growth, metastatic potential remains a concern. Surgical resection with pathological confirmation is the standard of care. CASE PRESENTATION We present a case of a patient who presented with mild flank pain. Successful treatment was achieved, followed by a 12-month uneventful follow-up. CLINICAL DISCUSSION ESC RCC typically demonstrates a favorable prognosis, often detected at an early stage. However, complete surgical resection with adjunct imaging is recommended to rule out metastatic disease. Despite its indolent nature, a vigilant approach is warranted due to the potential for incomplete tumor visualization. CONCLUSION This case report contributes to the growing body of literature on ESC RCC. By documenting our experience and reviewing the existing literature, we aim to enhance our understanding of this rare entity and optimize patient care.
Collapse
|
2
|
Abah MO, Ogenyi DO, Zhilenkova AV, Essogmo FE, Ngaha Tchawe YS, Uchendu IK, Pascal AM, Nikitina NM, Rusanov AS, Sanikovich VD, Pirogova YN, Boroda A, Moiseeva AV, Sekacheva MI. Innovative Therapies Targeting Drug-Resistant Biomarkers in Metastatic Clear Cell Renal Cell Carcinoma (ccRCC). Int J Mol Sci 2024; 26:265. [PMID: 39796121 PMCID: PMC11720203 DOI: 10.3390/ijms26010265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/09/2024] [Accepted: 12/14/2024] [Indexed: 01/13/2025] Open
Abstract
A thorough study of Clear Cell Renal Cell Carcinoma (ccRCC) shows that combining tyrosine kinase inhibitors (TKI) with immune checkpoint inhibitors (ICI) shows promising results in addressing the tumor-promoting influences of abnormal immunological and molecular biomarkers in metastatic Clear Cell Renal Cell Carcinoma (ccRCC). These abnormal biomarkers enhance drug resistance, support tumor growth, and trigger cancer-related genes. Ongoing clinical trials are testing new treatment options that appear more effective than earlier ones. However, more research is needed to confirm their long-term safety use and potential side effects. This study highlights vital molecular and immunological biomarkers associated with drug resistance in Clear Cell Renal Cell Carcinoma (ccRCC). Furthermore, this study identifies a number of promising drug candidates and biomarkers that serve as significant contributors to the enhancement of the overall survival of ccRCC patients. Consequently, this article offers pertinent insights on both recently completed and ongoing clinical trials, recommending further toxicity study for the prolonged use of this treatment strategy for patients with metastatic ccRCC, while equipping researchers with invaluable information for the progression of current treatment strategies.
Collapse
Affiliation(s)
- Moses Owoicho Abah
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
- Department of Cancer Bioinformatics and Molecular Biology, Royal Society of Clinical and Academic Researchers (ROSCAR) International, Abuja 900104, Nigeria
| | - Deborah Oganya Ogenyi
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Angelina V. Zhilenkova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Freddy Elad Essogmo
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Yvan Sinclair Ngaha Tchawe
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Ikenna Kingsley Uchendu
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
- Medical Laboratory Science Department, Faculty of Health Science and Technology, College of Medicine, University of Nigeria, Enugu Campus, Enugu 410001, Nigeria
| | - Akaye Madu Pascal
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Natalia M. Nikitina
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Alexander S. Rusanov
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Varvara D. Sanikovich
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Yuliya N. Pirogova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Alexander Boroda
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Aleksandra V. Moiseeva
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Marina I. Sekacheva
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| |
Collapse
|
3
|
Quinn AE, Bell SD, Marrah AJ, Wakefield MR, Fang Y. The Current State of the Diagnoses and Treatments for Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2024; 16:4034. [PMID: 39682220 DOI: 10.3390/cancers16234034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Clear cell renal cell carcinoma is the most common form of kidney cancer, accounting for 75% of malignant kidney tumors, and is generally associated with poor patient outcomes. With risk factors including smoking, obesity, and hypertension, all of which have a high prevalence in the United States and Europe, as well as genetic factors including tuberous sclerosis complex and Von Hippel-Lindau syndrome, there is an increasing need to expand our present understanding. The current clear cell renal cell carcinoma knowledge is outdated, with obsolete diagnostic criteria and moderately invasive surgical treatments still prevailing, partially ascribed to its resistance to chemotherapy and radiation therapy. The standard of treatment relies on surgical intervention, including radical nephrectomy and partial nephrectomy, while more recent treatments target neoplastic growth pathways and immune regulation checkpoints.
Collapse
Affiliation(s)
- Anthony E Quinn
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA
| | - Scott D Bell
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA
| | - Austin J Marrah
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
4
|
Akbari A, Villanueva CR, Hes O, Williamson SR, Kandukuri S, Sharma S, Aditi A, Pivovarcikova K, Argani P, Mohanty SK, Asrani K, Lotan TL. Genetic validation of a TSC2 immunohistochemistry assay in TSC/mTOR-pathway altered renal tumors. Hum Pathol 2024; 154:105693. [PMID: 39571694 DOI: 10.1016/j.humpath.2024.105693] [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: 08/18/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
Pathogenic mutations in the genes associated with tuberous sclerosis complex (TSC)/mTOR pathway are linked to histologically diverse renal cell neoplasms, including eosinophilic solid and cystic renal cell carcinoma (ESC RCC), low grade oncocytic tumor (LOT), eosinophilic vacuolated tumor (EVT), and xanthomatous giant cell renal cell carcinoma (XGC RCC). Here, we validate a TSC2 immunohistochemistry (IHC) assay by comparison to genomic data in these neoplasms. Automated TSC2 IHC was performed on formalin-fixed paraffin embedded (FFPE) tissues from 38 genetically-confirmed TSC/mTOR-associated renal tumors (6 ESCs, 16 EVTs, 13 LOTs, 2 XGC and 1 clear cell RCC) and visually scored in a semi-dichotomous fashion compared to internal control tissue. The positive predictive value (PPV) of TSC2 protein loss for underlying pathogenic mutation in TSC2 was 92% (11/12), while the negative predictive value (NPV) of intact TSC2 by IHC for lack of underlying pathogenic mutation in TSC2 was 81% (21/26). Intact TSC2 by IHC was 95% (21/22) specific for absence of underlying pathogenic TSC2 mutation. All the cases lacking TSC2 mutation with intact TSC2 protein had an underlying mutation in TSC1, MTOR or PIK3CA. Loss of TSC2 was 77% (10/13) sensitive for underlying TSC2 truncation mutations and 33% (1/3) sensitive for underlying TSC2 missense mutations. Overall, 73% (8/11) tumors with TSC2 IHC loss and underlying pathogenic alterations in TSC2 showed heterogeneous protein loss, with rare interspersed positively staining tumor cells. These data support TSC2 IHC as a potentially useful assay for the diagnostic workup of renal tumors suspected to belong to the TSC/mTOR-associated subgroups.
Collapse
Affiliation(s)
- Amir Akbari
- Department of Pathology, Johns Hopkins School of Medicine, USA; Department of Pathology, University of Southern California Keck School of Medicine, USA.
| | | | - Ondrej Hes
- Department of Pathology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | | | - Shivani Kandukuri
- Department of Pathology, University of Southern California Keck School of Medicine, USA
| | | | | | - Kristyna Pivovarcikova
- Department of Pathology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pedram Argani
- Department of Pathology, Johns Hopkins School of Medicine, USA
| | - Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, OR, India
| | - Kaushal Asrani
- Department of Pathology, Johns Hopkins School of Medicine, USA.
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins School of Medicine, USA; Department of Urology, Johns Hopkins School of Medicine, USA; Department of Oncology, Johns Hopkins School of Medicine, USA.
| |
Collapse
|
5
|
Gupta S, McCarthy MR, Tjota MY, Antic T, Cheville JC. Metastatic renal cell carcinoma with fibromyomatous stroma associated with tuberous sclerosis or MTOR, TSC1/TSC2-Mutations: A Series of 4 cases and a review of the literature. Hum Pathol 2024; 153:105680. [PMID: 39522702 DOI: 10.1016/j.humpath.2024.105680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Renal cell carcinoma with fibromyomatous stroma (RCCfms) are characterized by a constellation of morphologic findings that include elongated tubules lined by cells with clear to pale eosinophilic cytoplasm and intersecting bands of smooth muscle stroma. Consistent immunohistochemistry findings in RCCfms include diffuse positivity for carbonic anhydrase 9 and variable expression of keratin 7. Molecular profiling of such tumors show either pathogenic alterations of the ELOC (TCEB1) gene, or alterations of MTOR, TSC1, and TSC2. MTOR, TSC1/TSC2-altered RCCfms (M/TSC-RCCfms) has been reported both in the sporadic setting and in association with tuberous sclerosis complex (TSC). The importance of accurate diagnosis of M/TSC-RCCfms includes prompting germline testing in the appropriate clinical context. In addition, it can lead to patient management strategies that are focused on the preservation of renal function, as TSC patients often have multifocal and bilateral disease. As diagnostic criteria for M/TSC-RCCfms have only been recently established, additional data are needed to understand the natural history of this disease. Herein, we report 6 patients with metastatic M/TSC-RCCfms, including four patients from our institutional archives (four males, aged 36-58 years at nephrectomy), and two additional cases reported in the literature. Five patients had TSC, and the sixth had an MTOR-altered RCCfms. The majority of patients (5/6, 83%) presented with regional lymph node involvement and one patient developed metastases to the lung. All patients were alive at last follow up (median follow-up of 85 months). Our report is intended to raise awareness regarding rare instances of metastatic behavior for M/TSC-RCCfms.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Michael R McCarthy
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Melissa Y Tjota
- Department of Pathology, The University of Chicago, IL, 60637, USA.
| | - Tatjana Antic
- Department of Pathology, The University of Chicago, IL, 60637, USA.
| | - John C Cheville
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
6
|
Mohanty SK, Lobo A, Jha S, Sangoi AR, Akgul M, Trpkov K, Hes O, Mehra R, Hirsch MS, Moch H, Smith SC, Shah RB, Cheng L, Amin MB, Epstein JI, Parwani AV, Delahunt B, Desai S, Przybycin CG, Manini C, Luthringer DJ, Sirohi D, Jain D, Midha D, Jain E, Maclean F, Giannico GA, Paner GP, Martignoni G, Al-Ahmadie HA, McKenney J, Srigley JR, Lopez JI, Kunju LP, Browning L, Aron M, Picken MM, Tretiakova M, Zhou M, Sable M, Kuroda N, Pattnaik N, Gupta NS, Rao P, Fine SW, Mishra P, Adhya AK, Kulkarni BN, Dixit M, Baisakh MR, Arora S, Sancheti S, Menon S, Wobker SE, Tickoo SK, Kaushal S, Soni S, Kandukuri S, Sharma S, Mitra S, Reuter VE, Malik V, Rao V, Chen YB, Williamson SR. Acceptance of emerging renal oncocytic neoplasms: a survey of urologic pathologists. Virchows Arch 2024; 485:829-840. [PMID: 39287823 PMCID: PMC11564393 DOI: 10.1007/s00428-024-03909-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024]
Abstract
Oncocytic renal neoplasms are a major source of diagnostic challenge in genitourinary pathology; however, they are typically nonaggressive in general, raising the question of whether distinguishing different subtypes, including emerging entities, is necessary. Emerging entities recently described include eosinophilic solid and cystic renal cell carcinoma (ESC RCC), low-grade oncocytic tumor (LOT), eosinophilic vacuolated tumor (EVT), and papillary renal neoplasm with reverse polarity (PRNRP). A survey was shared among 65 urologic pathologists using SurveyMonkey.com (Survey Monkey, Santa Clara, CA, USA). De-identified and anonymized respondent data were analyzed. Sixty-three participants completed the survey and contributed to the study. Participants were from Asia (n = 21; 35%), North America (n = 31; 52%), Europe (n = 6; 10%), and Australia (n = 2; 3%). Half encounter oncocytic renal neoplasms that are difficult to classify monthly or more frequently. Most (70%) indicated that there is enough evidence to consider ESC RCC as a distinct entity now, whereas there was less certainty for LOT (27%), EVT (29%), and PRNRP (37%). However, when combining the responses for sufficient evidence currently and likely in the future, LOT and EVT yielded > 70% and > 60% for PRNRP. Most (60%) would not render an outright diagnosis of oncocytoma on needle core biopsy. There was a dichotomy in the routine use of immunohistochemistry (IHC) in the evaluation of oncocytoma (yes = 52%; no = 48%). The most utilized IHC markers included keratin 7 and 20, KIT, AMACR, PAX8, CA9, melan A, succinate dehydrogenase (SDH)B, and fumarate hydratase (FH). Genetic techniques used included TSC1/TSC2/MTOR (67%) or TFE3 (74%) genes and pathways; however, the majority reported using these very rarely. Only 40% have encountered low-grade oncocytic renal neoplasms that are deficient for FH. Increasing experience with the spectrum of oncocytic renal neoplasms will likely yield further insights into the most appropriate work-up, classification, and clinical management for these entities.
Collapse
Affiliation(s)
- Sambit K Mohanty
- Department of Pathology, Advanced Medical Research Institute Hospital, Kolkata, India
- Department of Pathology, Core Diagnostics, Gurgaon, India
| | - Anandi Lobo
- Department of Pathology, Kapoor Center of Urology and Pathology, Raipur, India
| | - Shilpy Jha
- Department of Pathology, Advanced Medical Research Institute Hospital, Kolkata, India
| | - Ankur R Sangoi
- Department of Pathology, El Camino Hospital, Mountain View, USA
| | - Mahmut Akgul
- Department of Pathology, Albany Medical Centre, Albany, USA
| | - Kiril Trpkov
- Department of Pathology, University of Calgary, Calgary, Canada
| | - Ondrej Hes
- Department of Pathology, Bioptika Laborator S.R.O, Pilsen, Czech Republic
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | | | - Holger Moch
- Department of Pathology, University Hospital, Zurich, Switzerland
| | - Steven C Smith
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, USA
| | - Rajal B Shah
- Department of Pathology, UT Southwestern Medical Center, Dallas, USA
| | - Liang Cheng
- Department of Pathology, Brown University, Providence, USA
| | - Mahul B Amin
- Department of Pathology, The University of Tennessee Health Science Center, Memphis, USA
| | | | - Anil V Parwani
- Department of Pathology, Ohio State University, Columbus, USA
| | - Brett Delahunt
- Department of Pathology, Wellington School/Medicine, Wellington, New Zealand
| | - Sangeeta Desai
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | | | - Claudia Manini
- Department of Pathology, University of Turin, Turin, Italy
| | | | - Deepika Sirohi
- Department of Pathology, University of Utah/ARUP, Salt Lake City, USA
| | - Deepika Jain
- Department of Pathology, Core Diagnostics, Gurgaon, India
| | - Divya Midha
- Department of Pathology, Tata Medical Center, Kolkata, India
| | - Ekta Jain
- Department of Pathology, Core Diagnostics, Gurgaon, India
| | - Fiona Maclean
- Department of Pathology, Douglass Hanly Moir Pathology, Sydney, Australia
| | - Giovanna A Giannico
- Department of Pathology, Vanderbilt University Medical Center, Nashville, USA
| | - Gladell P Paner
- Department of Pathology, University of Chicago, Chicago, USA
| | | | - Hikmat A Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Jesse McKenney
- Department of Pathology, Cleveland Clinic, Cleveland, USA
| | - John R Srigley
- Department of Pathology, Trillium Health Partners, Credit Valley Hospital, Mississauga, Canada
| | | | - L Priya Kunju
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Lisa Browning
- Department of Pathology, Oxford University Hospital NHS Foundation Trust, Oxford, UK
| | - Manju Aron
- Department of Pathology, Keck School of Medicine of USC, Los Angeles, USA
| | - Maria M Picken
- Department of Pathology, Loyola University Medical Center, Maywood, USA
| | | | - Ming Zhou
- Department of Pathology, Tufts University School of Medicine, Boston, USA
| | - Mukund Sable
- Department of Pathology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Naoto Kuroda
- Department of Pathology, Kochi Red Cross Hospital, Kochi City, Kochi, Japan
| | - Niharika Pattnaik
- Department of Pathology, Advanced Medical Research Institute Hospital, Kolkata, India
| | - Nilesh S Gupta
- Department of Pathology, Henry Ford Health System, Detroit, USA
| | - Priya Rao
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Samson W Fine
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Pritinanda Mishra
- Department of Pathology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Amit K Adhya
- Department of Pathology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Bijal N Kulkarni
- Department of Pathology, Kokilaben Ambani Hospital, Mumbai, India
| | - Mallika Dixit
- Department of Pathology, Core Diagnostics, Gurgaon, India
| | - Manas R Baisakh
- Department of Pathology, Apollo Hospitals Bhubaneshwar, Bhubaneswar, India
| | - Samriti Arora
- Department of Pathology, Core Diagnostics, Gurgaon, India
| | - Sankalp Sancheti
- Department of Pathology, Homi Bhabha Cancer Center, Visakhapatnam, India
| | - Santosh Menon
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Sara E Wobker
- Department of Pathology, The University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Satish K Tickoo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Seema Kaushal
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Shailesh Soni
- Department of Pathology, Muljibhai Patel Urological Hospital, Nadiad, India
| | - Shivani Kandukuri
- Department of Pathology, Keck School of Medicine of USC, Los Angeles, USA
| | - Shivani Sharma
- Department of Pathology, Core Diagnostics, Gurgaon, India
| | - Suvradeep Mitra
- Department of Pathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Victor E Reuter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Vipra Malik
- Department of Pathology, Core Diagnostics, Gurgaon, India
| | - Vishal Rao
- Department of Pathology, Basavatakaram Indo-American Cancer Hospital and Research Institute, Hyderabad, India
| | - Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | | |
Collapse
|
7
|
Hartung J, Müller C, Calkhoven CF. The dual role of the TSC complex in cancer. Trends Mol Med 2024:S1471-4914(24)00276-4. [PMID: 39488444 DOI: 10.1016/j.molmed.2024.10.009] [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: 08/19/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
The tuberous sclerosis complex (TSC1/TSC2/TBC1D7) primarily functions to inhibit the mechanistic target of rapamycin complex 1 (mTORC1), a crucial regulator of cell growth. Mutations in TSC1 or TSC2 cause tuberous sclerosis complex (TSC), a rare autosomal dominant genetic disorder marked by benign tumors in multiple organs that rarely progress to malignancy. Traditionally, TSC proteins are considered tumor suppressive due to their inhibition of mTORC1 and other mechanisms. However, more recent studies have shown that TSC proteins can also promote tumorigenesis in certain cancer types. In this review, we explore the composition and function of the TSC protein complex, the roles of its individual components in cancer biology, and potential future therapeutic targeting strategies.
Collapse
Affiliation(s)
- Josephine Hartung
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Christine Müller
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Cornelis F Calkhoven
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands.
| |
Collapse
|
8
|
Guo Q, Yao X, Yang B, Qi L, Wang F, Guo Y, Liu Y, Cao Z, Wang Y, Wang J, Li L, Huang Q, Liu C, Qu T, Zhao W, Ren D, Yang M, Yan C, Meng B, Wang C, Cao W. Eosinophilic Solid and Cystic Renal Cell Carcinoma: Morphologic and Immunohistochemical Study of 18 Cases and Review of the Literature. Arch Pathol Lab Med 2024; 148:1126-1134. [PMID: 38282571 DOI: 10.5858/arpa.2023-0122-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2023] [Indexed: 01/30/2024]
Abstract
CONTEXT.— Eosinophilic solid and cystic renal cell carcinoma is now defined in the 5th edition of the 2022 World Health Organization classification of urogenital tumors. OBJECTIVE.— To perform morphologic, immunohistochemical, and preliminary genetic studies about this new entity in China for the purpose of understanding it better. DESIGN.— The study includes 18 patients from a regional tertiary oncology center in northern China (Tianjin, China). We investigated the clinical and immunohistochemical features of these cases. RESULTS.— The mean age of patients was 49.6 years, and the male to female ratio was 11:7. Macroscopically, 1 case had the classic cystic and solid appearance, whereas the others appeared purely solid. Microscopically, all 18 tumors shared a similar solid and focal macrocystic or microcystic growth pattern, and the cells were characterized by voluminous and eosinophilic cytoplasm, along with coarse amphophilic stippling. Immunohistochemically, most of the tumors had a predominant cytokeratin (CK) 20-positive feature, ranging from focal cytoplasmic staining to diffuse membranous accentuation. Initially, we separated these cases into different immunohistochemical phenotypes. Group 1 (7 of 18; 38.5%) was characterized by positive phospho-4EBP1 and phospho-S6, which can imply hyperactive mechanistic target of rapamycin complex 1 (mTORC1) signaling. Group 2 (4 of 18; 23%) was negative for NF2, probably implying a germline mutation of NF2. Group 3 (7 of 18; 38.5%) consisted of the remaining cases. One case had metastatic spread and exhibited an aggressive clinical course, and we detected cyclin-dependent kinase inhibitor 2A (CDKN2A) mutation in this case; other patients were alive and without disease progression. CONCLUSIONS.— Our research proposes that eosinophilic solid and cystic renal cell carcinoma exhibits prototypical pathologic features with CK20 positivity and has aggressive potential.
Collapse
Affiliation(s)
- Qianru Guo
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xin Yao
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Bo Yang
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lisha Qi
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Frank Wang
- the Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada (F. Wang)
| | - Yuhong Guo
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yanxue Liu
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zi Cao
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yalei Wang
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jinpeng Wang
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lingmei Li
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Qiujuan Huang
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Changxu Liu
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tongyuan Qu
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wei Zhao
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Danyang Ren
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Manlin Yang
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chenhui Yan
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Bin Meng
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Cheng Wang
- the Department of Pathology and Laboratory Medicine, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada (C. Wang)
| | - Wenfeng Cao
- From the Departments of Pathology (Q. Guo, B. Yang, L. Qi, Y. Guo, Y. Liu, Z. Cao, Y. Wang, J Wang, L. Li, Q. Huang, C. Liu, T. Qu, W. Zhao, D. Ren, M. Yang, C. Yan, B. Meng, W. Cao) and Medical Oncology (X. Yao), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
9
|
Yoon AH, Tse JR. Hereditary renal mass syndromes: a pictorial review. Abdom Radiol (NY) 2024:10.1007/s00261-024-04534-y. [PMID: 39235599 DOI: 10.1007/s00261-024-04534-y] [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: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/06/2024]
Abstract
Hereditary renal mass syndromes, although rare, account for at least 3-5% of kidney cancers and significantly impact affected families. Accurate diagnosis and management by radiologists are crucial as these syndromes often present at imaging with both renal and extra-renal manifestations. The radiologist may be the first to recognize these stigmata at imaging, some of which satisfy major features for diagnosis and obviate the need for genetic testing. Furthermore, radiologists contribute to lifelong imaging surveillance and locoregional treatment. This pictorial review discusses the following major hereditary renal mass syndromes with their typical renal mass appearance, extrarenal manifestations, inheritance pattern, diagnosis, and management strategies based on the most recent National Comprehensive Cancer Network guidelines: Von Hippel-Lindau syndrome, tuberous sclerosis complex, Birt-Hogg-Dube syndrome, hereditary paraganglioma/pheochromocytoma (PGL/PCC) syndrome/succinate dehydrogenase deficiency, hereditary leiomyomatosis and renal cell cancer (HLRCC)/fumarate hydratase deficiency, PTEN hamartoma syndrome, BRCA1-associated protein 1 (BAP1) tumor disposition syndrome, hereditary papillary renal cell carcinoma, and familial clear cell renal cell cancer with chromosome 3 translocation.
Collapse
Affiliation(s)
- Acacia H Yoon
- Menlo-Atherton High School, Atherton, CA, 94027, USA
| | - Justin R Tse
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
10
|
Monich AG, Bissler JJ, Barreto FC. Tuberous Sclerosis Complex and the kidneys: what nephrologists need to know. J Bras Nefrol 2024; 46:e20240013. [PMID: 38991206 PMCID: PMC11239183 DOI: 10.1590/2175-8239-jbn-2024-0013en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/24/2024] [Indexed: 07/13/2024] Open
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant disease characterized by the development of hamartomas in the central nervous system, heart, skin, lungs, and kidneys and other manifestations including seizures, cortical tubers, radial migration lines, autism and cognitive disability. The disease is associated with pathogenic variants in the TSC1 or TSC2 genes, resulting in the hyperactivation of the mTOR pathway, a key regulator of cell growth and metabolism. Consequently, the hyperactivation of the mTOR pathway leads to abnormal tissue proliferation and the development of solid tumors. Kidney involvement in TSC is characterized by the development of cystic lesions, renal cell carcinoma and renal angiomyolipomas, which may progress and cause pain, bleeding, and loss of kidney function. Over the past years, there has been a notable shift in the therapeutic approach to TSC, particularly in addressing renal manifestations. mTOR inhibitors have emerged as the primary therapeutic option, whereas surgical interventions like nephrectomy and embolization being reserved primarily for complications unresponsive to clinical treatment, such as severe renal hemorrhage. This review focuses on the main clinical characteristics of TSC, the mechanisms underlying kidney involvement, the recent advances in therapy for kidney lesions, and the future perspectives.
Collapse
Affiliation(s)
- Aline Grosskopf Monich
- Universidade Federal do Paraná, Departamento de Clínica Médica, Programa de Pós-Graduação em Medicina Interna e Ciências da Saúde, Curitiba, PR, Brazil
- Hospital Universitário Evangélico Mackenzie, Serviço de Nefrologia, Curitiba, PR, Brazil
| | - John J. Bissler
- University of Tennessee, Health Science Center, Le Bonheur Children's Hospital, Department of Pediatrics, Memphis, TN, USA
- Le Bonheur Children's Hospital, Children's Foundation Research Institute, Memphis, TN, USA
- St. Jude Children’s Research Hospital, Pediatric Medicine Department, Memphis, TN, USA
| | - Fellype Carvalho Barreto
- Universidade Federal do Paraná, Departamento de Clínica Médica, Programa de Pós-Graduação em Medicina Interna e Ciências da Saúde, Curitiba, PR, Brazil
- Universidade Federal do Paraná, Departamento de Clínica Médica, Serviço de Nefrologia, Curitiba, PR, Brazil
| |
Collapse
|
11
|
Lin X. Characteristic morphology and immunohistochemical patterns of clear cell papillary renal cell tumours may be observed in renal cell carcinomas, a critical pitfall in renal biopsy cytopathology. Cytopathology 2024; 35:481-487. [PMID: 38751143 DOI: 10.1111/cyt.13384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/03/2024] [Accepted: 04/19/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND Clear cell papillary renal cell tumour (CCPRCT) was renamed from previous clear cell papillary renal cell carcinoma (CCPRCC) in the latest WHO Classification of Tumours. It is essential to differentiate RCC from CCPRCT in renal mass biopsies (RMB). DESIGN RMB cases with subsequent resections were reviewed. The pathology reports and pertinent clinical information were recorded. RESULTS Fifteen cases displaying either CCPRCT morphology (20% diffuse, 67% focal) or immunohistochemical patterns (cup-like CA9: 20% diffuse, 47% focal; CK7: 33% diffuse, 40% focal) were identified. One case was positive for TFE3. TSC mutation was identified in one case. Both cases exhibited both CCPRCT morphology and immunohistochemical patterns for CA9 and CK7, with focal high-grade nuclei. RMB diagnoses were as follows: 6 (40%) as CCRCC, 2 (13%) as CCPRCT, 2 (13%) as CCRCC versus CCPRCT, 2 (13%) as CCRCC versus PRCC, 1 (7%) as RCC with TSC mutation versus CCPRCT, 1 (7%) as TFE3-rearranged RCC versus PRCC, and 1 (7%) as cyst with low-grade atypia. 71% of patients underwent nephrectomy, 21% received systemic treatment for stage 4 RCCs, and 7% with ablation for small renal mass (1.6 cm) with low-grade CCRCC. CONCLUSIONS Our study highlights that morphologic and immunochemical features of CCPRCT may be present in RCCs, including RCC-TFE3 expression and TSC-associated RCC, a critical pitfall to misdiagnose aggressive RCC as indolent CCPRCT and result in undertreatment. Careful examination of morphology and immunostains for CA9, CK7, and TFE3, as well as molecular tests, is crucial for distinguishing aggressive RCC from indolent CCPRCT.
Collapse
Affiliation(s)
- Xiaoqi Lin
- Department of Pathology, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
12
|
Mekahli D, Müller RU, Marlais M, Wlodkowski T, Haeberle S, de Argumedo ML, Bergmann C, Breysem L, Fladrowski C, Henske EP, Janssens P, Jouret F, Kingswood JC, Lattouf JB, Lilien M, Maleux G, Rozenberg M, Siemer S, Devuyst O, Schaefer F, Kwiatkowski DJ, Rouvière O, Bissler J. Clinical practice recommendations for kidney involvement in tuberous sclerosis complex: a consensus statement by the ERKNet Working Group for Autosomal Dominant Structural Kidney Disorders and the ERA Genes & Kidney Working Group. Nat Rev Nephrol 2024; 20:402-420. [PMID: 38443710 DOI: 10.1038/s41581-024-00818-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2024] [Indexed: 03/07/2024]
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant disorder characterized by the presence of proliferative lesions throughout the body. Management of TSC is challenging because patients have a multifaceted systemic illness with prominent neurological and developmental impact as well as potentially severe kidney, heart and lung phenotypes; however, every organ system can be involved. Adequate care for patients with TSC requires a coordinated effort involving a multidisciplinary team of clinicians and support staff. This clinical practice recommendation was developed by nephrologists, urologists, paediatric radiologists, interventional radiologists, geneticists, pathologists, and patient and family group representatives, with a focus on TSC-associated kidney manifestations. Careful monitoring of kidney function and assessment of kidney structural lesions by imaging enable early interventions that can preserve kidney function through targeted approaches. Here, we summarize the current evidence and present recommendations for the multidisciplinary management of kidney involvement in TSC.
Collapse
Affiliation(s)
- Djalila Mekahli
- PKD Research Group, Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
- Department of Paediatric Nephrology, University Hospitals Leuven, Leuven, Belgium.
| | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- CECAD, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Center for Rare Diseases Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Matko Marlais
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Tanja Wlodkowski
- Division of Paediatric Nephrology, Center for Paediatrics and Adolescent Medicine, University Hospital, Heidelberg, Germany
| | - Stefanie Haeberle
- Division of Paediatric Nephrology, Center for Paediatrics and Adolescent Medicine, University Hospital, Heidelberg, Germany
| | - Marta López de Argumedo
- Basque Office for Health Technology Assessment, (OSTEBA), Basque Government, Vitoria-Gasteiz, Spain
| | - Carsten Bergmann
- Department of Medicine IV, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Medizinische Genetik Mainz, Limbach Genetics, Mainz, Germany
| | - Luc Breysem
- Department of Radiology, University Hospital of Leuven, Leuven, Belgium
| | - Carla Fladrowski
- Associazione Sclerosi Tuberosa ASP, Rome, Italy
- European Tuberous Sclerosis Complex Association (ETSC), Oestrich-Winkel, Germany
| | - Elizabeth P Henske
- Center for LAM Research and Clinical Care, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter Janssens
- Department of Nephrology and Arterial Hypertension, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel, Brussels, Belgium
| | - François Jouret
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital, Liège, Belgium
- Interdisciplinary Group of Applied Genoproteomics, Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - John Christopher Kingswood
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Centre, St Georges University of London, London, UK
| | - Jean-Baptiste Lattouf
- Department of Surgery-Urology, CHUM-Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Marc Lilien
- Department of Paediatric Nephrology, Wilhelmina Children´s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Geert Maleux
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Micaela Rozenberg
- European Tuberous Sclerosis Complex Association (ETSC), Oestrich-Winkel, Germany
- Associação de Esclerose Tuberosa em Portugal, Lisbon, Portugal
| | - Stefan Siemer
- Department of Urology and Paediatric Urology, Saarland University, Homburg, Germany
| | - Olivier Devuyst
- Department of Physiology, Mechanisms of Inherited Kidney Disorders, University of Zurich, Zurich, Switzerland
- Institute for Rare Diseases, Saint-Luc Academic Hospital, UC Louvain, Brussels, Belgium
| | - Franz Schaefer
- Division of Paediatric Nephrology, Center for Paediatrics and Adolescent Medicine, University Hospital, Heidelberg, Germany
| | - David J Kwiatkowski
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Olivier Rouvière
- Department of Radiology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
- Université Lyon 1, Lyon, France, Faculté de médecine Lyon Est, Lyon, France
| | - John Bissler
- Department of Paediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN, USA.
- Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN, USA.
- Paediatric Medicine Department, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
13
|
Chen Z, Zhang X. The role of metabolic reprogramming in kidney cancer. Front Oncol 2024; 14:1402351. [PMID: 38884097 PMCID: PMC11176489 DOI: 10.3389/fonc.2024.1402351] [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: 03/17/2024] [Accepted: 05/13/2024] [Indexed: 06/18/2024] Open
Abstract
Metabolic reprogramming is a cellular process in which cells modify their metabolic patterns to meet energy requirements, promote proliferation, and enhance resistance to external stressors. This process also introduces new functionalities to the cells. The 'Warburg effect' is a well-studied example of metabolic reprogramming observed during tumorigenesis. Recent studies have shown that kidney cells undergo various forms of metabolic reprogramming following injury. Moreover, metabolic reprogramming plays a crucial role in the progression, prognosis, and treatment of kidney cancer. This review offers a comprehensive examination of renal cancer, metabolic reprogramming, and its implications in kidney cancer. It also discusses recent advancements in the diagnosis and treatment of renal cancer.
Collapse
Affiliation(s)
- Ziyi Chen
- The First Clinical College of Fujian Medical University, Fuzhou, China
| | - Xiaohong Zhang
- Department of Nephrology, Blood Purification Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
14
|
Banda A, Naaldenberg J, Timen A, van Eeghen A, Leusink G, Cuypers M. Cancer risks related to intellectual disabilities: A systematic review. Cancer Med 2024; 13:e7210. [PMID: 38686623 PMCID: PMC11058689 DOI: 10.1002/cam4.7210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND People with intellectual disabilities (ID) face barriers in cancer care contributing to poorer oncological outcomes. Yet, understanding cancer risks in the ID population remains incomplete. AIM To provide an overview of cancer incidence and cancer risk assessments in the entire ID population as well as within ID-related disorders. METHODS This systematic review examined cancer risk in the entire ID population and ID-related disorders. We systematically searched PubMed (MEDLINE) and EMBASE for literature from January 1, 2000 to July 15, 2022 using a search strategy combining terms related to cancer, incidence, and ID. RESULTS We found 55 articles assessing cancer risks in the ID population at large groups or in subgroups with ID-related syndromes, indicating that overall cancer risk in the ID population is lower or comparable with that of the general population, while specific disorders (e.g., Down's syndrome) and certain genetic mutations may elevate the risk for particular cancers. DISCUSSION The heterogeneity within the ID population challenges precise cancer risk assessment at the population level. Nonetheless, within certain subgroups, such as individuals with specific ID-related disorders or certain genetic mutations, a more distinct pattern of varying cancer risks compared to the general population becomes apparent. CONCLUSION More awareness, and personalized approach in cancer screening within the ID population is necessary.
Collapse
Affiliation(s)
- Amina Banda
- Department of Primary and Community CareRadboud university medical centreNijmegenthe Netherlands
| | - Jenneken Naaldenberg
- Department of Primary and Community CareRadboud university medical centreNijmegenthe Netherlands
| | - Aura Timen
- Department of Primary and Community CareRadboud university medical centreNijmegenthe Netherlands
| | - Agnies van Eeghen
- Emma Children's HospitalAmsterdam University Medical CentersAmsterdamthe Netherlands
- 'S Heeren LooAmersfoortthe Netherlands
| | - Geraline Leusink
- Department of Primary and Community CareRadboud university medical centreNijmegenthe Netherlands
| | - Maarten Cuypers
- Department of Primary and Community CareRadboud university medical centreNijmegenthe Netherlands
| |
Collapse
|
15
|
Almuqbil M, Aldoohan W, Alhinti S, Almahmoud N, Abdulmajeed I, Alkhodair R, Kashgari A, Baarmah D, Altwaijri W, Alrumayyan A. Review of the spectrum of tuberous sclerosis complex: The Saudi Arabian Experience. NEUROSCIENCES (RIYADH, SAUDI ARABIA) 2024; 29:113-121. [PMID: 38740395 PMCID: PMC11305360 DOI: 10.17712/nsj.2024.2.20230061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/28/2023] [Indexed: 05/16/2024]
Abstract
OBJECTIVES To determine the prevalence of tuberous sclerosis complex (TSC) in the paediatric Saudi population and to characterise the range of clinical symptoms, neurocutaneous findings, neuroimaging results, and complications of the disease. METHODS A total of 61 genetically confirmed TSC patients from the National Guard Health Affairs (NGHA) in Saudi Arabia were the subject of this retrospective descriptive analysis. The data were presented using descriptive measures. RESULTS The mean age at diagnosis was found to be 4.9 years. Subependymal nodules (86.9%), numerous cortical tubers and/or radial migration lines (63.9%), and hypomelanotic macules (63.9%) were the 3 most common significant criteria. The vast majority (86.9%) of those diagnosed had epilepsy, of which 50% were considered medically intractable. Nearly half of our subjects underwent genetic testing, which revealed that TSC2 predominated over TSC1. Symptoms of Tuberous Sclerosis Complex-Associated Neuropsychiatric Disorders (TAND) were present in 66.7% of TSC1 patients and 73.9% of TSC2 patients. CONCLUSION The findings of this study demonstrate that the clinical spectrum of TSC among Saudi children is consistent with the body of existing literature. The TSC2 was more prevalent than TSC1. The most frequent signs were cutaneous and neurological. Monitoring TSC patients regularly is crucial to identify any issues as soon as possible.
Collapse
Affiliation(s)
- Mohammed Almuqbil
- From the College of Medicine (Almuqbil, Aldoohan, Alhinti, Alkhodair, Kashgari, Altwaijri, Alrumayyan), King Saud bin Abdulaziz University for Health Sciences, from King Abdullah International Medical Research Center (Almuqbil, Alkhodair), Ministry of National Guard, from the Division of Pediatric Neurology (Almuqbil Altwaijri, Alrumayyan), Department of Pediatrics, King Abdullah Specialized Children’s Hospital, National Guard Health Affairs, from the Department of Family Medicine and Primary Health Care (Abdulmajeed), King Abdulaziz Medical City, from the Division of Pediatric Dermatology (Alkhodair), Department of Pediatrics, King Abdullah Specialized Children’s Hospital (KASCH), National Guard Health Affairs, and from Department of Pediatric (Baarmah), King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia
| | - Waad Aldoohan
- From the College of Medicine (Almuqbil, Aldoohan, Alhinti, Alkhodair, Kashgari, Altwaijri, Alrumayyan), King Saud bin Abdulaziz University for Health Sciences, from King Abdullah International Medical Research Center (Almuqbil, Alkhodair), Ministry of National Guard, from the Division of Pediatric Neurology (Almuqbil Altwaijri, Alrumayyan), Department of Pediatrics, King Abdullah Specialized Children’s Hospital, National Guard Health Affairs, from the Department of Family Medicine and Primary Health Care (Abdulmajeed), King Abdulaziz Medical City, from the Division of Pediatric Dermatology (Alkhodair), Department of Pediatrics, King Abdullah Specialized Children’s Hospital (KASCH), National Guard Health Affairs, and from Department of Pediatric (Baarmah), King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia
| | - Sara Alhinti
- From the College of Medicine (Almuqbil, Aldoohan, Alhinti, Alkhodair, Kashgari, Altwaijri, Alrumayyan), King Saud bin Abdulaziz University for Health Sciences, from King Abdullah International Medical Research Center (Almuqbil, Alkhodair), Ministry of National Guard, from the Division of Pediatric Neurology (Almuqbil Altwaijri, Alrumayyan), Department of Pediatrics, King Abdullah Specialized Children’s Hospital, National Guard Health Affairs, from the Department of Family Medicine and Primary Health Care (Abdulmajeed), King Abdulaziz Medical City, from the Division of Pediatric Dermatology (Alkhodair), Department of Pediatrics, King Abdullah Specialized Children’s Hospital (KASCH), National Guard Health Affairs, and from Department of Pediatric (Baarmah), King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia
| | - Nora Almahmoud
- From the College of Medicine (Almuqbil, Aldoohan, Alhinti, Alkhodair, Kashgari, Altwaijri, Alrumayyan), King Saud bin Abdulaziz University for Health Sciences, from King Abdullah International Medical Research Center (Almuqbil, Alkhodair), Ministry of National Guard, from the Division of Pediatric Neurology (Almuqbil Altwaijri, Alrumayyan), Department of Pediatrics, King Abdullah Specialized Children’s Hospital, National Guard Health Affairs, from the Department of Family Medicine and Primary Health Care (Abdulmajeed), King Abdulaziz Medical City, from the Division of Pediatric Dermatology (Alkhodair), Department of Pediatrics, King Abdullah Specialized Children’s Hospital (KASCH), National Guard Health Affairs, and from Department of Pediatric (Baarmah), King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia
| | - Imad Abdulmajeed
- From the College of Medicine (Almuqbil, Aldoohan, Alhinti, Alkhodair, Kashgari, Altwaijri, Alrumayyan), King Saud bin Abdulaziz University for Health Sciences, from King Abdullah International Medical Research Center (Almuqbil, Alkhodair), Ministry of National Guard, from the Division of Pediatric Neurology (Almuqbil Altwaijri, Alrumayyan), Department of Pediatrics, King Abdullah Specialized Children’s Hospital, National Guard Health Affairs, from the Department of Family Medicine and Primary Health Care (Abdulmajeed), King Abdulaziz Medical City, from the Division of Pediatric Dermatology (Alkhodair), Department of Pediatrics, King Abdullah Specialized Children’s Hospital (KASCH), National Guard Health Affairs, and from Department of Pediatric (Baarmah), King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia
| | - Rayan Alkhodair
- From the College of Medicine (Almuqbil, Aldoohan, Alhinti, Alkhodair, Kashgari, Altwaijri, Alrumayyan), King Saud bin Abdulaziz University for Health Sciences, from King Abdullah International Medical Research Center (Almuqbil, Alkhodair), Ministry of National Guard, from the Division of Pediatric Neurology (Almuqbil Altwaijri, Alrumayyan), Department of Pediatrics, King Abdullah Specialized Children’s Hospital, National Guard Health Affairs, from the Department of Family Medicine and Primary Health Care (Abdulmajeed), King Abdulaziz Medical City, from the Division of Pediatric Dermatology (Alkhodair), Department of Pediatrics, King Abdullah Specialized Children’s Hospital (KASCH), National Guard Health Affairs, and from Department of Pediatric (Baarmah), King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia
| | - Amna Kashgari
- From the College of Medicine (Almuqbil, Aldoohan, Alhinti, Alkhodair, Kashgari, Altwaijri, Alrumayyan), King Saud bin Abdulaziz University for Health Sciences, from King Abdullah International Medical Research Center (Almuqbil, Alkhodair), Ministry of National Guard, from the Division of Pediatric Neurology (Almuqbil Altwaijri, Alrumayyan), Department of Pediatrics, King Abdullah Specialized Children’s Hospital, National Guard Health Affairs, from the Department of Family Medicine and Primary Health Care (Abdulmajeed), King Abdulaziz Medical City, from the Division of Pediatric Dermatology (Alkhodair), Department of Pediatrics, King Abdullah Specialized Children’s Hospital (KASCH), National Guard Health Affairs, and from Department of Pediatric (Baarmah), King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia
| | - Duaa Baarmah
- From the College of Medicine (Almuqbil, Aldoohan, Alhinti, Alkhodair, Kashgari, Altwaijri, Alrumayyan), King Saud bin Abdulaziz University for Health Sciences, from King Abdullah International Medical Research Center (Almuqbil, Alkhodair), Ministry of National Guard, from the Division of Pediatric Neurology (Almuqbil Altwaijri, Alrumayyan), Department of Pediatrics, King Abdullah Specialized Children’s Hospital, National Guard Health Affairs, from the Department of Family Medicine and Primary Health Care (Abdulmajeed), King Abdulaziz Medical City, from the Division of Pediatric Dermatology (Alkhodair), Department of Pediatrics, King Abdullah Specialized Children’s Hospital (KASCH), National Guard Health Affairs, and from Department of Pediatric (Baarmah), King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia
| | - Waleed Altwaijri
- From the College of Medicine (Almuqbil, Aldoohan, Alhinti, Alkhodair, Kashgari, Altwaijri, Alrumayyan), King Saud bin Abdulaziz University for Health Sciences, from King Abdullah International Medical Research Center (Almuqbil, Alkhodair), Ministry of National Guard, from the Division of Pediatric Neurology (Almuqbil Altwaijri, Alrumayyan), Department of Pediatrics, King Abdullah Specialized Children’s Hospital, National Guard Health Affairs, from the Department of Family Medicine and Primary Health Care (Abdulmajeed), King Abdulaziz Medical City, from the Division of Pediatric Dermatology (Alkhodair), Department of Pediatrics, King Abdullah Specialized Children’s Hospital (KASCH), National Guard Health Affairs, and from Department of Pediatric (Baarmah), King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia
| | - Ahmad Alrumayyan
- From the College of Medicine (Almuqbil, Aldoohan, Alhinti, Alkhodair, Kashgari, Altwaijri, Alrumayyan), King Saud bin Abdulaziz University for Health Sciences, from King Abdullah International Medical Research Center (Almuqbil, Alkhodair), Ministry of National Guard, from the Division of Pediatric Neurology (Almuqbil Altwaijri, Alrumayyan), Department of Pediatrics, King Abdullah Specialized Children’s Hospital, National Guard Health Affairs, from the Department of Family Medicine and Primary Health Care (Abdulmajeed), King Abdulaziz Medical City, from the Division of Pediatric Dermatology (Alkhodair), Department of Pediatrics, King Abdullah Specialized Children’s Hospital (KASCH), National Guard Health Affairs, and from Department of Pediatric (Baarmah), King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
16
|
Coffey NJ, Simon MC. Metabolic alterations in hereditary and sporadic renal cell carcinoma. Nat Rev Nephrol 2024; 20:233-250. [PMID: 38253811 PMCID: PMC11165401 DOI: 10.1038/s41581-023-00800-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/24/2024]
Abstract
Kidney cancer is the seventh leading cause of cancer in the world, and its incidence is on the rise. Renal cell carcinoma (RCC) is the most common form and is a heterogeneous disease comprising three major subtypes that vary in their histology, clinical course and driver mutations. These subtypes include clear cell RCC, papillary RCC and chromophobe RCC. Molecular analyses of hereditary and sporadic forms of RCC have revealed that this complex and deadly disease is characterized by metabolic pathway alterations in cancer cells that lead to deregulated oxygen and nutrient sensing, as well as impaired tricarboxylic acid cycle activity. These metabolic changes facilitate tumour growth and survival. Specifically, studies of the metabolic features of RCC have led to the discovery of oncometabolites - fumarate and succinate - that can promote tumorigenesis, moonlighting functions of enzymes, and substrate auxotrophy owing to the disruption of pathways that enable the production of arginine and cholesterol. These metabolic alterations within RCC can be exploited to identify new therapeutic targets and interventions, in combination with novel approaches that minimize the systemic toxicity of metabolic inhibitors and reduce the risk of drug resistance owing to metabolic plasticity.
Collapse
Affiliation(s)
- Nathan J Coffey
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Shah RB, Mehra R. Renal Cell Carcinoma Associated With TSC/MTOR Genomic Alterations: An Update on its Expanding Spectrum and an Approach to Clinicopathologic Work-up. Adv Anat Pathol 2024; 31:105-117. [PMID: 37899532 DOI: 10.1097/pap.0000000000000419] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
Renal cell carcinoma (RCC) with tuberous sclerosis complex (TSC)/mammalian target of rapamycin (MTOR) pathway-related genomic alterations have been classically described in hereditary TSC syndrome setting involving germline mutations, whereby cells with a bi-allelic inactivation of genes originate tumors in a classic tumor-suppressor "two-hit" Knudson paradigm. Initial studies of TSC-associated RCC categorized tumors into 3 broad heterogeneous morphologic groups: RCC with smooth muscle stroma, chromophobe-like, and eosinophilic-macrocytic. Recently, a similar morphologic spectrum has been increasingly recognized in novel and emerging entities characterized by somatic mutations in the TSC1/2 and MTOR in patients who do not suffer from the TSC. Correct recognition of RCC with TSC / MTOR mutations is critical for accurate prognostication because such tumors with aggressive behavior have the potential to be tailored to mTOR inhibitors. Whether TSC/MTOR mutated renal epithelial neoplasms represent a distinct molecular class has been confounded by the fact that TSC1/2 , and the gene encoding the downstream protein MTOR, are mutated secondarily in ∼5% of the more common subtypes of RCC, including the commonest subtype of clear cell RCC. This review summarizes the expanding morphologic spectrum of renal tumors with TSC/mTOR pathway alterations, specifically for sporadically occurring tumors where these genomic alterations likely are primary pathologic events. Finally, a practical surgical pathology approach to handling these tumors, and a conceptual framework of renal epithelial tumors with TSC/MTOR mutations as a "family of tumors", is presented.
Collapse
Affiliation(s)
- Rajal B Shah
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Rohit Mehra
- Department of Pathology and Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI
| |
Collapse
|
18
|
Garcia ABDM, Viola GD, Corrêa BDS, Fischer TDS, Pinho MCDF, Rodrigues GM, Ashton-Prolla P, Rosset C. An overview of actionable and potentially actionable TSC1 and TSC2 germline variants in an online Database. Genet Mol Biol 2024; 46:e20230132. [PMID: 38373162 PMCID: PMC10876083 DOI: 10.1590/1678-4685-gmb-2023-0132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/26/2023] [Indexed: 02/21/2024] Open
Abstract
Tuberous Sclerosis Complex (TSC) is caused by loss of function germline variants in the TSC1 or TSC2 tumor suppressor genes. Genetic testing for the detection of pathogenic variants in either TSC1 or TSC2 was implemented as a diagnostic criterion for TSC. However, TSC molecular diagnosis can be challenging due to the absence of variant hotspots and the high number of variants described. This review aimed to perform an overview of TSC1/2 variants submitted in the ClinVar database. Variants of uncertain significance (VUS), missense and single nucleotide variants were the most frequent in clinical significance (37-40%), molecular consequence (37%-39%) and variation type (82%-83%) categories in ClinVar in TSC1 and TSC2 variants, respectively. Frameshift and nonsense VUS have potential for pathogenic reclassification if further functional and segregation studies were performed. Indeed, there were few functional assays deposited in the database and literature. In addition, we did not observe hotspots for variation and many variants presented conflicting submissions regarding clinical significance. This study underscored the importance of disseminating molecular diagnostic results in a public database to render the information largely accessible and promote accurate diagnosis. We encourage the performance of functional studies evaluating the pathogenicity of TSC1/2 variants.
Collapse
Affiliation(s)
- Arthur Bandeira de Mello Garcia
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Departamento de Genética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
| | - Guilherme Danielski Viola
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Departamento de Genética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
| | - Bruno da Silveira Corrêa
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Departamento de Genética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
| | - Taís da Silveira Fischer
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
| | - Maria Clara de Freitas Pinho
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Centro Universitário CESUCA, Cachoeirinha, RS, Brazil
| | - Grazielle Motta Rodrigues
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| | - Patricia Ashton-Prolla
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Departamento de Genética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
- Hospital de Clínicas de Porto Alegre, Serviço de Genética Médica, Porto Alegre, RS, Brazil
| | - Clévia Rosset
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| |
Collapse
|
19
|
Sethi SK, Nataraj SA, Sankhyan N, Rana A, Nair A, Bansal SB. An Adolescent with Tuberous Sclerosis and Hypocalcemia and a Renal Mass. Indian J Nephrol 2024; 34:94-95. [PMID: 38645923 PMCID: PMC11003591 DOI: 10.4103/ijn.ijn_103_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/11/2023] [Indexed: 04/23/2024] Open
Affiliation(s)
| | | | - Naveen Sankhyan
- Pediatric Neurology, PGIMER Chandigarh, Chandigarh, Punjab, India
| | - Alka Rana
- Department of Pathology and Laboratory Medicine, Medanta, The Medicity, Gurgaon, Haryana, India
| | - Aishwarya Nair
- Pediatric Nephrology, Kidney Institute, Medanta, The Medicity, Gurgaon, India
| | | |
Collapse
|
20
|
Machacek ME, Wu CL, Cornejo KM. Pathology of hereditary renal cell carcinoma syndromes: Tuberous sclerosis complex (TSC). Semin Diagn Pathol 2024; 41:8-19. [PMID: 37993384 DOI: 10.1053/j.semdp.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 11/24/2023]
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant genetic disease characterized by hamartomatous tumors involving multiple organs such as the brain, skin, heart, lung and kidney. TSC is caused by inactivating mutations in TSC1/TSC2, which encodes hamartin and tuberin, respectively, and forms a complex that regulates mechanistic target of rapamycin complex 1 (mTORC1), resulting in cell overgrowth and oncogenesis. Since a leading cause of morbidity and mortality in TSC relates to chronic kidney disease and the ability to preserve renal function, this review describes the important pathologic findings in TSC-associated renal neoplasms and their correlating sporadic counterparts. The most common renal tumor in TSC patients are AMLs, followed by a heterogeneous spectrum of renal epithelial tumors, which may provide clues to establishing a diagnosis of TSC.
Collapse
Affiliation(s)
- Miranda E Machacek
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Chin-Lee Wu
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Kristine M Cornejo
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
21
|
Caliò A, Marletta S, Settanni G, Rizzo M, Gobbo S, Pedron S, Stefanizzi L, Munari E, Brunelli M, Marcolini L, Pesci A, Fratoni S, Pierconti F, Raspollini MR, Marchetti A, Doglioni C, Amin MB, Porta C, Martignoni G. mTOR eosinophilic renal cell carcinoma: a distinctive tumor characterized by mTOR mutation, loss of chromosome 1, cathepsin-K expression, and response to target therapy. Virchows Arch 2023; 483:821-833. [PMID: 37938323 PMCID: PMC10700445 DOI: 10.1007/s00428-023-03688-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/17/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023]
Abstract
In the spectrum of oncocytic renal neoplasms, a subset of tumors with high-grade-appearing histologic features harboring pathogenic mutations in mammalian target of rapamycin (mTOR) and hitherto clinical indolent behavior has been described. Three cases (2F,1 M) with histologically documented metastases (lymph node, skull, and liver) were retrieved and extensively investigated by immunohistochemistry, FISH, and next-generation sequencing. Tumors were composed of eosinophilic cells with prominent nucleoli (G3 by ISUP/WHO) arranged in solid to nested architecture. Additionally, there were larger cells with perinuclear cytoplasmic shrinkage and sparse basophilic Nissl-like granules, superficially resembling the so-called spider cells of cardiac rhabdomyomas. The renal tumors, including the skull and liver metastases, showed immunoexpression PAX8, CK8-18, and cathepsin-K, and negativity for vimentin. NGS identified mTOR genetic alterations in the three cases, including the skull and liver metastases. One patient was then treated with Everolimus (mTOR inhibitors) with clinical response (metastatic tumor shrinkage). We present a distinct renal tumor characterized by high-grade eosinophilic cells, cathepsin-K immunohistochemical expression, and harboring mTOR gene mutations demonstrating a malignant potential and showing responsiveness to mTOR inhibitors.
Collapse
Affiliation(s)
- Anna Caliò
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
| | - Stefano Marletta
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Giulio Settanni
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Italy
| | - Mimma Rizzo
- Division of Medical Oncology, A.O.U. Consorziale Policlinico Di Bari, Bari, Italy
| | - Stefano Gobbo
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Serena Pedron
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
| | | | - Enrico Munari
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Matteo Brunelli
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
| | - Lisa Marcolini
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Anna Pesci
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Italy
| | - Stefano Fratoni
- Division of Anatomic Pathology, S. Eugenio Hospital, Rome, Italy
| | - Francesco Pierconti
- Division of Anatomic Pathology and Histology, Foundation "A. Gemelli" University Hospital, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Rosaria Raspollini
- Histopathology and Molecular Diagnostics, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Antonio Marchetti
- Division of Anatomic Pathology and Histology, Ospedale Clinicizzato "SS. Annunziata" Università Di Chieti, Chieti, Italy
| | | | - Mahul B Amin
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science, Memphis, TN, USA
- Department of Urology, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Camillo Porta
- Interdisciplinary Department of Medicine, University of Bari "A. Moro, Bari, Italy
| | - Guido Martignoni
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy.
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy.
| |
Collapse
|
22
|
Du H, Yang YC, Liu HJ, Yuan M, Asara JM, Wong KK, Henske EP, Singh M, Kwiatkowski DJ. Bi-steric mTORC1 inhibitors induce apoptotic cell death in tumor models with hyperactivated mTORC1. J Clin Invest 2023; 133:e167861. [PMID: 37909334 PMCID: PMC10617776 DOI: 10.1172/jci167861] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 09/06/2023] [Indexed: 11/03/2023] Open
Abstract
The PI3K/AKT/mTOR pathway is commonly dysregulated in cancer. Rapalogs exhibit modest clinical benefit, likely owing to their lack of effects on 4EBP1. We hypothesized that bi-steric mTORC1-selective inhibitors would have greater potential for clinical benefit than rapalogs in tumors with mTORC1 dysfunction. We assessed this hypothesis in tumor models with high mTORC1 activity both in vitro and in vivo. Bi-steric inhibitors had strong growth inhibition, eliminated phosphorylated 4EBP1, and induced more apoptosis than rapamycin or MLN0128. Multiomics analysis showed extensive effects of the bi-steric inhibitors in comparison with rapamycin. De novo purine synthesis was selectively inhibited by bi-sterics through reduction in JUN and its downstream target PRPS1 and appeared to be the cause of apoptosis. Hence, bi-steric mTORC1-selective inhibitors are a therapeutic strategy to treat tumors driven by mTORC1 hyperactivation.
Collapse
Affiliation(s)
- Heng Du
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Yu Chi Yang
- Department of Biology, Revolution Medicines Inc., Redwood City, California, USA
| | - Heng-Jia Liu
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Min Yuan
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - John M. Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York University Langone Health, New York, New York, USA
| | - Elizabeth P. Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Mallika Singh
- Department of Biology, Revolution Medicines Inc., Redwood City, California, USA
| | - David J. Kwiatkowski
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Osawa T, Oya M, Okanishi T, Kuwatsuru R, Kawano H, Tomita Y, Niida Y, Nonomura N, Hatano T, Fujii Y, Mizuguchi M, Shinohara N. Clinical Practice Guidelines for tuberous sclerosis complex-associated renal angiomyolipoma by the Japanese Urological Association: Summary of the update. Int J Urol 2023; 30:808-817. [PMID: 37278492 DOI: 10.1111/iju.15213] [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: 03/09/2023] [Accepted: 05/14/2023] [Indexed: 06/07/2023]
Abstract
New clinical issues have been raised through an interval of 7 years from the previous version (2016). In this study, we update the "Clinical Practice Guidelines for tuberous sclerosis complex-associated renal angiomyolipoma" as a 2023 version under guidance by the Japanese Urological Association. The present guidelines were cooperatively prepared by the Japanese Urological Association and Japanese Society of Tuberous Sclerosis Complex; committee members belonging to one of the two societies or specializing in the treatment of this disease were selected to prepare the guidelines in accordance with the "Guidance for preparing treatment guidelines" published by Minds (2020 version). The "Introduction" consisted of four sections, "Background Questions (BQ)" consisted of four sections, "Clinical Questions (CQ)" consisted of three sections, and "Future Questions (FQ)" consisted of three sections (total: 14 sections). Concerning CQ, an agreement was confirmed through voting by the committee members based on the direction and strength of recommendation, accuracy of evidence, and recommendation comments. The present guidelines were updated based on the current evidence. We hope that the guidelines will provide guiding principles for the treatment of tuberous sclerosis complex-associated renal angiomyolipoma to many urologists, becoming a foundation for subsequent updating.
Collapse
Affiliation(s)
- Takahiro Osawa
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Tohru Okanishi
- Division of Child Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ryohei Kuwatsuru
- Department of Radiology, School of Medicine and Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Haruna Kawano
- Department of Urology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshihiko Tomita
- Department of Urology and Department of Molecular Oncology, Niigata University Graduate School of Medicine, Niigata, Japan
| | - Yo Niida
- Center for Clinical Genomics, Kanazawa Medical University Hospital, Uchinada, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takashi Hatano
- Department of Urology, Seirei Yokohama Hospital, Yokohama, Japan
| | - Yasuhisa Fujii
- Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masashi Mizuguchi
- Department of Pediatrics, National Rehabilitation Center for Children with Disabilities, Tokyo, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
24
|
Gallo-Bernal S, Kilcoyne A, Gee MS, Paul E. Cystic kidney disease in tuberous sclerosis complex: current knowledge and unresolved questions. Pediatr Nephrol 2023; 38:3253-3264. [PMID: 36445479 DOI: 10.1007/s00467-022-05820-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/02/2022] [Accepted: 11/02/2022] [Indexed: 12/02/2022]
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant disorder with an estimated incidence of one in 5000 to 10,000 live births worldwide. Two million people of all races and genders are estimated to have TSC secondary to mutations in one of two tumor suppressor genes, TSC1 or TSC2. The respective TSC1 and 2 gene products - hamartin and tuberin - form cytoplasmic heterodimers that inhibit mTOR-mediated cell growth and division. When mTOR inhibition is lost, people with TSC develop characteristic and usually benign tumors in various organ systems. Kidney tumors and cysts are common, particularly in the setting of TSC2 gene mutations. In most TSC patients, the number of kidney cysts is limited, their morphology is simple, their size is small, and their clinical significance is negligible. In some, cyst morphology progresses from simple to complex with the risk of malignant transformation. In others, aggressive accumulation and growth of kidney cysts can cause hypertension, impaired kidney function, and progression to kidney failure. This educational review summarizes current knowledge and remaining open questions regarding cystic kidney disease in TSC, emphasizing detection, classification, surveillance, and treatment options.
Collapse
Affiliation(s)
- Sebastian Gallo-Bernal
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Aoife Kilcoyne
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Michael S Gee
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Elahna Paul
- Department of Pediatric Nephrology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
25
|
Abstract
Up to 5% of renal cell carcinomas (RCCs) can be associated with a known hereditary RCC syndrome. In addition to the well-characterized RCC syndromes, there are also emerging syndromes associated with increased RCC risk. In the last few years, consensus guidelines have outlined recommendations for who should be referred for genetic evaluation, and what screening should be done for early detection of RCC. Although much progress has been made, work is still needed-guidelines are still mostly based on expert opinion and the role of emerging genetic associations will need to be clarified.
Collapse
Affiliation(s)
- Maria I Carlo
- Genitourinary Oncology Service, Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, 353 East 68th Street. New York, NY 10065, USA.
| |
Collapse
|
26
|
Kapur P, Brugarolas J, Trpkov K. Recent Advances in Renal Tumors with TSC/mTOR Pathway Abnormalities in Patients with Tuberous Sclerosis Complex and in the Sporadic Setting. Cancers (Basel) 2023; 15:4043. [PMID: 37627070 PMCID: PMC10452688 DOI: 10.3390/cancers15164043] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
A spectrum of renal tumors associated with frequent TSC/mTOR (tuberous sclerosis complex/mechanistic target of rapamycin) pathway gene alterations (in both the germline and sporadic settings) have recently been described. These include renal cell carcinoma with fibromyomatous stroma (RCC FMS), eosinophilic solid and cystic renal cell carcinoma (ESC RCC), eosinophilic vacuolated tumor (EVT), and low-grade oncocytic tumor (LOT). Most of these entities have characteristic morphologic and immunohistochemical features that enable their recognition without the need for molecular studies. In this report, we summarize recent advances and discuss their evolving complexity.
Collapse
Affiliation(s)
- Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - James Brugarolas
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hematology-Oncology Division of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kiril Trpkov
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2L 2K5, Canada
- Alberta Precision Labs, Rockyview General Hospital, 7007 14 St., Calgary, AB T2V 1P9, Canada
| |
Collapse
|
27
|
Alomar K, Alia L, Qatleesh S, Mardenly F, Orabi A, Alhussein AA. A rare case of eosinophilic solid and cystic renal cell carcinoma in a 48-year-old woman: Case report and literature review. Int J Surg Case Rep 2023; 108:108463. [PMID: 37423149 PMCID: PMC10382833 DOI: 10.1016/j.ijscr.2023.108463] [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: 06/05/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
INTRODUCTION AND SIGNIFICANCE This is (ESC RCC) a rare renal tumor that was thought to occur exclusively in female patients and almost exclusively in TSC patients. The tumor does not have distinctive clinical symptoms or radiological manifestations that are important for distinguishing it from other tumors or renal formations, but it has unique features and distinct histological characteristics that allow us to distinguish it from other tumors. Despite its slow growth, it sometimes metastasizes to other parts of the body. Surgical interventions are treated by examining tissue samples that show the characteristic features of the tumor. CASE PRESENTATION We present the case of a patient who complained of mild flank pain without other associated symptoms. She was successfully treated in our hospital and followed up for 8 months without any problems. CLINICAL DISCUSSION This tumor generally has slow growth and good prognosis and is often detected at an early stage. However, when confronted with this tumor, good surgical excision with a full body scan is necessary to rule out the presence of metastases, monitor the patient well, and act decisively despite the good warning of this tumor, as we have not yet achieved complete visualization of this formation. Neoplastic. CONCLUSION By studying the successive reports of this unique tumor, this manuscript will help document our case and review the literature on this tumor to try to understand this tumor formation in the hope of achieving the best medical care for these patients.
Collapse
Affiliation(s)
- Khaled Alomar
- Damascus University, University Pediatrics' Hospital, Syria.
| | - Louei Alia
- Damascus University, Al-Assad University Hospital, Syria
| | - Safaa Qatleesh
- Damascus University, Al-Assad University Hospital, Syria
| | - Fadel Mardenly
- Damascus University, Al-Assad University Hospital, Syria
| | - Alaa Orabi
- Damascus University, University Pediatrics' Hospital, Syria
| | | |
Collapse
|
28
|
Roebuck DJ, Chippington S, Berry BD, Gibson C. Core needle biopsy and embolization of fat-poor renal tumors in children with tuberous sclerosis complex. World J Pediatr 2023; 19:614-618. [PMID: 36795318 DOI: 10.1007/s12519-023-00692-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/19/2023] [Indexed: 02/17/2023]
Affiliation(s)
- Derek J Roebuck
- Department of Medical Imaging, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, 6009, Australia.
- Division of Paediatrics, Medical School, University of Western Australia, Crawley, 6009, Australia.
| | - Samantha Chippington
- Department of Radiology, Great Ormond Street Hospital for Children, London, WC1N 3JH, UK
| | - Bligh D Berry
- PathWest Laboratory Medicine WA, Murdoch, 6150, Australia
| | - Craig Gibson
- Department of Medical Imaging, Perth Children's Hospital, 15 Hospital Avenue, Nedlands, 6009, Australia
| |
Collapse
|
29
|
Badoiu SC, Greabu M, Miricescu D, Stanescu-Spinu II, Ilinca R, Balan DG, Balcangiu-Stroescu AE, Mihai DA, Vacaroiu IA, Stefani C, Jinga V. PI3K/AKT/mTOR Dysregulation and Reprogramming Metabolic Pathways in Renal Cancer: Crosstalk with the VHL/HIF Axis. Int J Mol Sci 2023; 24:8391. [PMID: 37176098 PMCID: PMC10179314 DOI: 10.3390/ijms24098391] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/26/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Renal cell carcinoma (RCC) represents 85-95% of kidney cancers and is the most frequent type of renal cancer in adult patients. It accounts for 3% of all cancer cases and is in 7th place among the most frequent histological types of cancer. Clear cell renal cell carcinoma (ccRCC), accounts for 75% of RCCs and has the most kidney cancer-related deaths. One-third of the patients with ccRCC develop metastases. Renal cancer presents cellular alterations in sugars, lipids, amino acids, and nucleic acid metabolism. RCC is characterized by several metabolic dysregulations including oxygen sensing (VHL/HIF pathway), glucose transporters (GLUT 1 and GLUT 4) energy sensing, and energy nutrient sensing cascade. Metabolic reprogramming represents an important characteristic of the cancer cells to survive in nutrient and oxygen-deprived environments, to proliferate and metastasize in different body sites. The phosphoinositide 3-kinase-AKT-mammalian target of the rapamycin (PI3K/AKT/mTOR) signaling pathway is usually dysregulated in various cancer types including renal cancer. This molecular pathway is frequently correlated with tumor growth and survival. The main aim of this review is to present renal cancer types, dysregulation of PI3K/AKT/mTOR signaling pathway members, crosstalk with VHL/HIF axis, and carbohydrates, lipids, and amino acid alterations.
Collapse
Affiliation(s)
- Silviu Constantin Badoiu
- Department of Anatomy and Embryology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania;
| | - Maria Greabu
- Department of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, Sector 5, 050474 Bucharest, Romania;
| | - Daniela Miricescu
- Department of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, Sector 5, 050474 Bucharest, Romania;
| | - Iulia-Ioana Stanescu-Spinu
- Department of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, Sector 5, 050474 Bucharest, Romania;
| | - Radu Ilinca
- Department of Medical Informatics and Biostatistics, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania;
| | - Daniela Gabriela Balan
- Department of Physiology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania; (D.G.B.); (A.-E.B.-S.)
| | - Andra-Elena Balcangiu-Stroescu
- Department of Physiology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania; (D.G.B.); (A.-E.B.-S.)
| | - Doina-Andrada Mihai
- Department of Diabetes, Nutrition and Metabolic Diseases, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania;
| | - Ileana Adela Vacaroiu
- Department of Nephrology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Constantin Stefani
- Department of Family Medicine and Clinical Base, Dr. Carol Davila Central Military Emergency University Hospital, 134 Calea Plevnei, 010825 Bucharest, Romania;
| | - Viorel Jinga
- Department of Urology, “Prof. Dr. Theodor Burghele” Hospital, 050653 Bucharest, Romania
- “Prof. Dr. Theodor Burghele” Clinical Hospital, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania
- Medical Sciences Section, Academy of Romanian Scientists, 050085 Bucharest, Romania
| |
Collapse
|
30
|
Reprint of: lessons from histopathologic examination of nephrectomy specimens in patients with tuberous sclerosis complex: cysts, angiomyolipomas & renal cell carcinoma. Hum Pathol 2023; 133:136-152. [PMID: 36894367 DOI: 10.1016/j.humpath.2023.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 09/08/2022] [Indexed: 03/09/2023]
Abstract
Renal manifestations in patients with tuberous sclerosis complex (TSC) include cysts, angiomyolipoma, and renal cell carcinoma. Unlike many hereditary predisposition syndromes, the spectrum of renal tumors in TSC patients (including both angiomyolipoma and renal cell carcinoma) is broad, with significant morphologic heterogeneity. An improved understanding of histopathologic findings in TSC patients and associated clinicopathologic correlates has significant implications not just in establishing a diagnosis of TSC, but also in the recognition of sporadic tumors occurring secondary to somatic alterations of TSC1/TSC2/MTOR pathway genes and accurate prognostication. In this review, we have discussed issues relevant to clinical management based on histopathologic findings in nephrectomy specimens from patients with TSC. This includes discussions related to screening for TSC, diagnosis of PKD1/TSC2 contiguous gene deletion syndrome, the morphologic spectrum of angiomyolipoma and renal epithelium-derived neoplasia, including the risk of disease progression.
Collapse
|
31
|
Orosz P, Kollák Z, Pethő Á, Fogarasi A, Reusz G, Hadzsiev K, Szabó T. The Importance of Genetic Testing in the Differential Diagnosis of Atypical TSC2-PKD1 Contiguous Gene Syndrome-Case Series. CHILDREN (BASEL, SWITZERLAND) 2023; 10:420. [PMID: 36979978 PMCID: PMC10047143 DOI: 10.3390/children10030420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND In clinical practice, the possible diagnosis of tuberous sclerosis or polycystic kidney disease is primarily based on clinical criteria, which can later be verified by genetic testing. But in the case of TSC2/PKD1 contiguous gene syndrome (TSC2/PKD1-CGS), the renal appearance of the disease is more serious. Therefore, early genetic analysis is recommended. METHODS Herein we present the report of four children with TSC2/PKD1-CGS, one involving the NTHL1 gene. We aim to emphasize the importance of genetic testing in this rare syndrome. RESULTS During the follow-up of tuberous sclerosis and polycystic kidney disease patients, it is essential to reappraise the diagnosis if the clinical symptoms' appearance or onset time is unusual. Targeted genetic testing is recommended. However, early tumor formation necessitates the extension of genetic analysis. CONCLUSIONS An appropriate evaluation of the phenotype is the cornerstone of diagnosing the rare TSC2/PKD1-CGS with the help of genetic results. In addition, malignant tumors could draw attention to an infrequent large deletion.
Collapse
Affiliation(s)
- Petronella Orosz
- Bethesda Children’s Hospital, 1146 Budapest, Hungary
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Zita Kollák
- Bethesda Children’s Hospital, 1146 Budapest, Hungary
| | - Ákos Pethő
- Department of Internal Medicine and Oncology Clinic, Faculty of Medicine, Semmelweis University, 1083 Budapest, Hungary
| | | | - György Reusz
- 1st Department of Pediatrics, Faculty of Medicine, Semmelweis University, 1083 Budapest, Hungary
| | - Kinga Hadzsiev
- Department of Medical Genetics, Medical School, Clinical Centre, University of Pécs, 7624 Pécs, Hungary
| | - Tamás Szabó
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
32
|
Bissler JJ, Batchelor D, Kingswood JC. Progress in Tuberous Sclerosis Complex Renal Disease. Crit Rev Oncog 2023; 27:35-49. [PMID: 36734871 DOI: 10.1615/critrevoncog.2022042857] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant disorder that affects both fetal development and postnatal tissue growth, resulting in altered brain structures and a tumor predisposition syndrome. Although every organ system is affected by the disease, kidney involvement is a leading cause of death in adults with TSC. Over the past decade, significant progress has been made in understanding the renal disease. This review focuses on the cystic and solid renal lesions in TSC, including their pathobiology and treatment.
Collapse
Affiliation(s)
- John J Bissler
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38105; Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105; Pediatric Medicine Department, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Dinah Batchelor
- Johns Hopkins All Children's Hospital, St. Petersburg, FL 33702
| | - J Christopher Kingswood
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Centre, St. Georges University of London, London, United Kingdom
| |
Collapse
|
33
|
Giannikou K, Martin KR, Abdel-Azim AG, Pamir KJ, Hougard TR, Bagwe S, Tang Y, MacKeigan JP, Kwiatkowski DJ, Henske EP, Lam HC. Spectrum of germline and somatic mitochondrial DNA variants in Tuberous Sclerosis Complex. Front Genet 2023; 13:917993. [PMID: 36793390 PMCID: PMC9923026 DOI: 10.3389/fgene.2022.917993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 11/23/2022] [Indexed: 02/03/2023] Open
Abstract
Tuberous Sclerosis Complex (TSC) is caused by loss of function variants in either TSC1 or TSC2 and is characterized by broad phenotypic heterogeneity. Currently, there is limited knowledge regarding the role of the mitochondrial genome (mtDNA) in TSC pathogenesis. In this study, we aimed to determine the prevalence and spectrum of germline and somatic mtDNA variants in TSC and identify potential disease modifiers. Analysis of mtDNA amplicon massively parallel sequencing (aMPS) data, off-target mtDNA from whole-exome sequencing (WES), and/or qPCR, revealed mtDNA alterations in 270 diverse tissues (139 TSC-associated tumors and 131 normal tissue samples) from 199 patients and six healthy individuals. Correlation of clinical features to mtDNA variants and haplogroup analysis was done in 102 buccal swabs (age: 20-71 years). No correlation was found between clinical features and either mtDNA variants or haplogroups. No pathogenic variants were identified in the buccal swab samples. Using in silico analysis, we identified three predicted pathogenic variants in tumor samples: MT-ND4 (m.11742G>A, p. Cys328Tyr, VAF: 43%, kidney angiomyolipoma), MT-CYB (m.14775T>C, p. Leu10Pro, VAF: 43%, LAM abdominal tumor) and MT-CYB (m.15555C>T, p. Pro270Leu, VAF: 7%, renal cell carcinoma). Large deletions of the mitochondrial genome were not detected. Analysis of tumors from 23 patients with corresponding normal tissue did not reveal any recurrent tumor-associated somatic variants. The mtDNA/gDNA ratio between tumors and corresponding normal tissue was also unchanged. Overall, our findings demonstrate that the mitochondrial genome is highly stable across tissues and within TSC-associated tumors.
Collapse
Affiliation(s)
- Krinio Giannikou
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Division of Hematology/Oncology, Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Katie R. Martin
- Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Ahmad G. Abdel-Azim
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Kaila J. Pamir
- Center for LAM Research and Clinical Care, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Thomas R. Hougard
- Center for LAM Research and Clinical Care, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Shefali Bagwe
- Center for LAM Research and Clinical Care, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Yan Tang
- Center for LAM Research and Clinical Care, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Jeffrey P. MacKeigan
- Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - David J. Kwiatkowski
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Elizabeth P. Henske
- Center for LAM Research and Clinical Care, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Hilaire C. Lam
- Center for LAM Research and Clinical Care, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
34
|
Are Renal Cell Carcinoma with Fibromyomatous Stroma (RCC-FMS) and Thyroid-like Follicular Carcinoma of the Kidney (TLFCK) Really Independent Variants? Diagnostics (Basel) 2022; 13:diagnostics13010086. [PMID: 36611378 PMCID: PMC9818596 DOI: 10.3390/diagnostics13010086] [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: 10/11/2022] [Revised: 11/16/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Renal cell carcinoma with fibromyomatous stroma (RCC-FMS) is a recent provisional entity already recognised in the 2016 WHO Classification of Cancer of the Urinary Tract and Male Genital Organs 4th Edition as renal cell carcinoma with (angio)leiomyomatous stroma, histologically defined as a tumour characterised by clear cells intertwined in a conspicuous vascular stroma. In the casuistry taken into consideration, another proposed variant, thyroid-like follicular carcinoma of the kidney (TLFCK), endowed with a morphology mimicking thyroid parenchyma, was examined. The aim of this work was to parse the theoretical system, experimental data and diagnostic impact of these new entities proposed in the field of renal neoplasms. MATERIALS AND METHODS An analysis of 120 cases of kidney tumours from the Department of Surgical, Medical, Molecular and Critical Area at the University of Pisa was run. Subsequently, all samples were reassessed by two pathologists with expertise in uropathology, whose revaluation provided a histomorphological study combined with subsequent and coherent immunohistochemical analyses of CK7, CD10, CAIX, CK34betaE12, CD117, vimentin, TTF-1 and thyroglobulin. These analyses were performed using the Ventana Benchmark Automated Staining System (Ventana Medical Systems, Tucson, AZ, USA) and Ventana reagents. RESULTS On the one hand, the data, thus brought to light, did not show an immunohistochemical profile consistent with that proposed for RCC-FMS. However, it should be emphasised that the morphological background also unearthed a poor specificity for RCC-FMS. This was specifically due to a stromal component which was, in any case, evident, although characterised by a wide range of presentation, in clear cell renal cell carcinoma (ccRCC). This latter is, indeed, the reference background for this theorised variant. On the other hand, a thyroid-like pattern was highlighted in 11 cases, more specifically in 10 ccRCCs and in one oncocytoma, presenting itself as a type of neoplastic appearance rather than as the peculiar morphological pattern of a standalone cancer. CONCLUSIONS In the light of these results, RCC-FMS and TLFCK appear to be more appropriately variants of already categorised neoplastic entities rather than new independent neoplasias.
Collapse
|
35
|
Kong J, Tao J, Wang Q, Zhang Q, Yin L. Rare renal cell carcinoma with haemangioblastoma-like features and leiomyomatous stroma: report of a unique case with TSC2 and SETD2 variations. World J Surg Oncol 2022; 20:395. [PMID: 36510186 PMCID: PMC9746168 DOI: 10.1186/s12957-022-02844-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/20/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) with haemangioblastoma-like characteristics is a type of RCC reported in recent years. RCC with (angio) leiomyomatous stroma (RCCLMS) was included as a provisional entity of the 2016 World Health Organization (WHO) classification. RCC with haemangioblastoma-like characteristics and leiomyomatous stroma is extremely rare. This is the first report of a rare tumour harbouring TSC2 and SETD2 variations. CASE PRESENTATION The patient was a 38-year-old woman who presented with discomfort in the area of her right kidney. Ultrasound and enhanced CT showed a right renal mass, and clear cell renal cell carcinoma (CCRCC) was suspected; hence, robot-assisted laparoscopic nephron-sparing partial nephrectomy was performed. Gross examination revealed a well-circumscribed tumour measuring 2.0 cm × 1 cm × 0.7 cm under the renal capsule adjacent to the stripping edge that was greyish yellow and greyish red in colour. Histologic examination showed that the tumour consisted of three different structures: a CCRCC-like region, a haemangioblastoma-like region, and a focal leiomyomatous stroma component. Based on immunohistochemistry, the CCRCC-like region was diffusely strongly positive for AE1/AE3, vimentin, CAIX, PAX8, PAX2, CK7, and CAM5.2, partly positive for HNF1α, and negative for CD10, α-inhibin, NSE, S-100, CD34, and TFE3. The haemangioblastoma-like area was diffusely positive for vimentin, CAIX; partly positive for PAX8, PAX2, α-inhibin, and S-100; mostly positive for NSE; and slightly positive for HNF1α; the CD34 staining highlighted the complex capillary network. The Ki67 index was approximately 1-2% in the two above areas, and the leiomyomatous stroma was strongly positive for SMA. The whole-exon sequencing (WES) showed TSC2 and SETD2 variations. There was no progression after 18 months of follow-up. CONCLUSION We report for the first time a unique case of RCC with haemangioblastoma-like features and leiomyomatous stroma accompanied by rare molecular abnormalities. Whether this is a new tumour entity or a variant of clear cell carcinoma remains to be determined. The biological behaviour and clinical characteristics need to be further examined.
Collapse
Affiliation(s)
- Jixia Kong
- grid.452828.10000 0004 7649 7439Department of Pathology, the Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 Liaoning China
| | - Juan Tao
- grid.452828.10000 0004 7649 7439Department of Pathology, the Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 Liaoning China
| | - Qimin Wang
- grid.452828.10000 0004 7649 7439Department of Pathology, the Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 Liaoning China
| | - Qingfu Zhang
- grid.412636.40000 0004 1757 9485Department of Pathology, the First Affiliated Hospital of China Medical University, Shenyang, 110001 Liaoning China
| | - Liying Yin
- grid.452828.10000 0004 7649 7439Department of Pathology, the Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 Liaoning China
| |
Collapse
|
36
|
Neves JB, Roberts K, Nguyen JS, El Sheikh S, Tran-Dang MA, Horsfield C, Mumtaz F, Campbell P, Stauss H, Tran MG, Mitchell T. Defining the origin, evolution, and immune composition of SDH-deficient renal cell carcinoma. iScience 2022; 25:105389. [PMID: 36345344 PMCID: PMC9636038 DOI: 10.1016/j.isci.2022.105389] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/05/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022] Open
Abstract
Succinate dehydrogenase (SDH)-deficient renal cell carcinoma represents a rare subtype of hereditary kidney cancer. Clinical diagnosis can be challenging and there is little evidence to guide systemic therapeutic options. We performed genomic profiling of a cohort of tumors through the analysis of whole genomes, transcriptomes, as well as flow cytometry and immunohistochemistry in order to gain a deeper understanding of their molecular biology. We find neutral evolution after early tumor activation with a lack of secondary driver events. We show that these tumors have epithelial derivation, possibly from the macula densa, a specialized paracrine cell of the renal juxtaglomerular apparatus. They subsequently develop into immune excluded tumors. We provide transcriptomic and protein expression evidence of a highly specific tumor marker, PAPPA2. These translational findings have implications for the diagnosis and treatment for this rare tumor subtype.
Collapse
Affiliation(s)
- Joana B. Neves
- UCL Division of Surgery and Interventional Science, Royal Free Hospital, London, UK
- Specialist Centre for Kidney Cancer, Royal Free Hospital, London, UK
- UCL Institute of Immunity & Transplantation, The Pears Building, Pond Street, London, UK
| | - Kirsty Roberts
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Soha El Sheikh
- Department of Histopathology, Royal Free Hospital, London, UK
| | | | - Catherine Horsfield
- Guy’s & St Thomas’ National Health Service Trust, Westminster Bridge Road, London, UK
| | - Faiz Mumtaz
- UCL Division of Surgery and Interventional Science, Royal Free Hospital, London, UK
- Specialist Centre for Kidney Cancer, Royal Free Hospital, London, UK
| | - Peter Campbell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Hans Stauss
- UCL Institute of Immunity & Transplantation, The Pears Building, Pond Street, London, UK
| | - Maxine G.B. Tran
- UCL Division of Surgery and Interventional Science, Royal Free Hospital, London, UK
- Specialist Centre for Kidney Cancer, Royal Free Hospital, London, UK
- UCL Institute of Immunity & Transplantation, The Pears Building, Pond Street, London, UK
| | - Thomas Mitchell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
| |
Collapse
|
37
|
Webster BR, Gopal N, Ball MW. Tumorigenesis Mechanisms Found in Hereditary Renal Cell Carcinoma: A Review. Genes (Basel) 2022; 13:2122. [PMID: 36421797 PMCID: PMC9690265 DOI: 10.3390/genes13112122] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 09/29/2023] Open
Abstract
Renal cell carcinoma is a heterogenous cancer composed of an increasing number of unique subtypes each with their own cellular and tumor behavior. The study of hereditary renal cell carcinoma, which composes just 5% of all types of tumor cases, has allowed for the elucidation of subtype-specific tumorigenesis mechanisms that can also be applied to their sporadic counterparts. This review will focus on the major forms of hereditary renal cell carcinoma and the genetic alterations contributing to their tumorigenesis, including von Hippel Lindau syndrome, Hereditary Papillary Renal Cell Carcinoma, Succinate Dehydrogenase-Deficient Renal Cell Carcinoma, Hereditary Leiomyomatosis and Renal Cell Carcinoma, BRCA Associated Protein 1 Tumor Predisposition Syndrome, Tuberous Sclerosis, Birt-Hogg-Dubé Syndrome and Translocation RCC. The mechanisms for tumorigenesis described in this review are beginning to be exploited via the utilization of novel targets to treat renal cell carcinoma in a subtype-specific fashion.
Collapse
Affiliation(s)
| | | | - Mark W. Ball
- Center for Cancer Research, Urologic Oncology Branch, National Cancer Institute/NIH, 10 Center Drive, CRC Room 2W-5940, Bethesda, MD 20892, USA
| |
Collapse
|
38
|
Argani P, Rose G, Matoso A, Gagan J, Palsgrove DN. Biphasic Hyalinizing Psammomatous Renal Cell Carcinoma (BHP RCC) in a Child With Neurofibromatosis Type 2 Syndrome. Am J Surg Pathol 2022; 46:1595-1598. [PMID: 35941722 DOI: 10.1097/pas.0000000000001942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Pedram Argani
- Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Gary Rose
- Department of Pathology, Saint Joseph's Hospital, BayCare Laboratories, Tampa FL
| | - Andres Matoso
- Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jeffrey Gagan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Doreen N Palsgrove
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
39
|
Xia QY, Wang XT, Zhao M, He HY, Fang R, Ye SB, Li R, Wang X, Zhang RS, Lu ZF, Ma HH, Wang ZY, Rao Q. TSC/MTOR -associated Eosinophilic Renal Tumors Exhibit a Heterogeneous Clinicopathologic Spectrum : A Targeted Next-generation Sequencing and Gene Expression Profiling Study. Am J Surg Pathol 2022; 46:1562-1576. [PMID: 35980750 DOI: 10.1097/pas.0000000000001955] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Several TSC1/2- or MTOR -mutated eosinophilic renal tumor subsets are emerging, including eosinophilic solid and cystic renal cell carcinoma (ESC RCC), eosinophilic vacuolated tumors (EVTs) and low-grade oncocytic tumors (LOTs). "Unclassified renal tumors with TSC/MTOR mutations" ( TSC -mt RCC-NOS) do not meet the criteria for other histomolecular subtypes. Whether these tumors represent a continuum of 1 TS C/ MTOR -mutation-associated disease is unknown. DESIGN We evaluated the clinicopathologic and IHC profiles of 39 eosinophilic renal tumors with targeted DNA sequencing-confirmed TSC/MTOR mutations. Twenty-eight of these, plus 6 ChRCC, 5 RO, 5 ccRCC, 7 MiT RCC and 6 normal renal tissues, were profiled transcriptionally by RNA-seq. RESULTS The 39 cases were reclassified based on morphological and IHC features as ESC RCC (12), EVT (9), LOT, (8) and TSC -mt RCC-NOS (10). The mutation profiles demonstrated consistency; ESC RCCs (12/12) had TSC mutations, and most LOTs (7/8) had MTOR mutations. Ten TSC -mt RCC-NOSs exhibited heterogeneous morphology, arising a differential diagnosis with other renal tumors, including MiT RCC, PRCC and epithelioid PEComa. RNA sequencing-based clustering segregated ESC RCC, EVT and LOT from each other and other renal tumors, indicating expression profile-level differences. Most TSC- mt RCC-NOSs (6/7) formed a mixed cluster with ESC RCC, indicating similar expression signatures; one TSC- mt RCC-NOS with unusual biphasic morphology clustered with EVT. CONCLUSIONS We expanded the TSC/MTOR -associated eosinophilic renal tumor morphologic spectrum, identified gene mutation characteristics, and highlighted differential diagnosis challenges, especially with MiT RCC. ESC RCC, EVT, and LOT having distinct expression profiles. TSC -mt RCC-NOS may cluster with recognized TSC/MTOR -associated entities.
Collapse
Affiliation(s)
- Qiu-Yuan Xia
- Department of Pathology, Affiliated Jinling Hospital, Medical School of Nanjing University, Jiangsu
| | - Xiao-Tong Wang
- Department of Pathology, Affiliated Jinling Hospital, Medical School of Nanjing University, Jiangsu
| | - Ming Zhao
- Cancer Center, Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou
| | - Hui-Ying He
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, China
| | - Ru Fang
- Department of Pathology, Affiliated Jinling Hospital, Medical School of Nanjing University, Jiangsu
| | - Sheng-Bing Ye
- Department of Pathology, Affiliated Jinling Hospital, Medical School of Nanjing University, Jiangsu
| | - Rui Li
- Department of Pathology, Affiliated Jinling Hospital, Medical School of Nanjing University, Jiangsu
| | - Xuan Wang
- Department of Pathology, Affiliated Jinling Hospital, Medical School of Nanjing University, Jiangsu
| | - Ru-Song Zhang
- Department of Pathology, Affiliated Jinling Hospital, Medical School of Nanjing University, Jiangsu
| | - Zhen-Feng Lu
- Department of Pathology, Affiliated Jinling Hospital, Medical School of Nanjing University, Jiangsu
| | - Heng-Hui Ma
- Department of Pathology, Affiliated Jinling Hospital, Medical School of Nanjing University, Jiangsu
| | - Zi-Yu Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine
| | - Qiu Rao
- Department of Pathology, Affiliated Jinling Hospital, Medical School of Nanjing University, Jiangsu
| |
Collapse
|
40
|
Gupta S, Stanton ML, Reynolds JP, Whaley RD, Herrera-Hernandez L, Jimenez RE, Cheville JC. Lessons from histopathologic examination of nephrectomy specimens in patients with tuberous sclerosis complex: cysts, angiomyolipomas, and renal cell carcinoma. Hum Pathol 2022; 129:123-139. [PMID: 36115585 DOI: 10.1016/j.humpath.2022.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 09/08/2022] [Indexed: 02/07/2023]
Abstract
Renal manifestations in patients with tuberous sclerosis complex (TSC) include cysts, angiomyolipoma, and renal cell carcinoma. Unlike many hereditary predisposition syndromes, the spectrum of renal tumors in TSC patients (including both angiomyolipoma and renal cell carcinoma) is broad, with significant morphologic heterogeneity. An improved understanding of histopathologic findings in TSC patients and associated clinicopathologic correlates has significant implications not just in establishing a diagnosis of TSC, but also in the recognition of sporadic tumors occurring secondary to somatic alterations of TSC1/TSC2/MTOR pathway genes and accurate prognostication. In this review, we have discussed issues relevant to clinical management based on histopathologic findings in nephrectomy specimens from patients with TSC. This includes discussions related to screening for TSC, diagnosis of PKD1/TSC2 contiguous gene deletion syndrome, the morphologic spectrum of angiomyolipoma and renal epithelium-derived neoplasia, including the risk of disease progression.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Melissa L Stanton
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ, 85054, USA.
| | - Jordan P Reynolds
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, 32224, USA.
| | - Rumeal D Whaley
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | | | - Rafael E Jimenez
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - John C Cheville
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
41
|
Napolitano G, Di Malta C, Ballabio A. Non-canonical mTORC1 signaling at the lysosome. Trends Cell Biol 2022; 32:920-931. [PMID: 35654731 DOI: 10.1016/j.tcb.2022.04.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 01/21/2023]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) signaling hub integrates multiple environmental cues to modulate cell growth and metabolism. Over the past decade considerable knowledge has been gained on the mechanisms modulating mTORC1 lysosomal recruitment and activation. However, whether and how mTORC1 is able to elicit selective responses to diverse signals has remained elusive until recently. We discuss emerging evidence for a 'non-canonical' mTORC1 signaling pathway that controls the function of microphthalmia/transcription factor E (MiT-TFE) transcription factors, key regulators of cell metabolism. This signaling pathway is mediated by a specific mechanism of substrate recruitment, and responds to stimuli that appear to converge on the lysosomal surface. We discuss the relevance of this pathway in physiological and disease conditions.
Collapse
Affiliation(s)
- Gennaro Napolitano
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy; Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Via Pansini 5, 80131 Naples, Italy.
| | - Chiara Di Malta
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy; Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Via Pansini 5, 80131 Naples, Italy.
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy; Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Via Pansini 5, 80131 Naples, Italy; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
42
|
Li J, Liu F, Liu X, Hu Y, Liu Z, Shen Y, Wan J. Heterozygous germline FLCN mutation in Birt-Hogg-Dubé syndrome with bilateral renal hybrid oncocytic/chromophobe tumor and unilateral renal chromophobe cell carcinoma: a case report. J Cancer Res Clin Oncol 2022; 149:2319-2325. [PMID: 36258004 DOI: 10.1007/s00432-022-04417-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 10/11/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Birt-Hogg-Dubé (BHD) syndrome is a rare autosomal dominant tumor syndrome caused by Folliculin (FLCN) gene mutation. The clinical manifestations are diverse, and the renal tumor is the most serious. CASE PRESENTATION We report the case of a 51-year-old female with multiple space-occupying lesions in both kidneys, as well as bilateral renal cysts and multiple lung cysts. The resected tumors were finally diagnosed as bilateral renal hybrid oncocytic/chromophobe tumor (HOCT) and unilateral chromophobe cell carcinoma. A history of pneumothorax surgery 4 years ago was convinced. All of these strongly suggested the possibility of BHD syndrome. Gene test confirmed a heterozygous germline FLCN nonsense mutation (c.1429C > T, p.Arg477Ter). The patient is still doing well after 20 months of follow-up (cut-off date July 2022). CONCLUSION This is a case of BHD syndrome presented with bilateral renal tumor, renal cysts, and multiple lung cysts, and confirmed by gene testing. Patients with any combination of one or more of the manifestations should remain alert to the BHD syndrome. Our report will help to deepen the understanding of the clinicopathological features and molecular changes of BHD syndrome.
Collapse
Affiliation(s)
- Jun Li
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fan Liu
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Pathology, The Eighth Hospital of Wuhan, Wuhan, 430010, China
| | - Xuguang Liu
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Hu
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zheng Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yaqi Shen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jie Wan
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
43
|
Amin MB, McKenney JK, Martignoni G, Campbell SC, Pal S, Tickoo SK. Low grade oncocytic tumors of the kidney: a clinically relevant approach for the workup and accurate diagnosis. Mod Pathol 2022; 35:1306-1316. [PMID: 35896615 DOI: 10.1038/s41379-022-01108-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/02/2022] [Accepted: 05/09/2022] [Indexed: 11/09/2022]
Abstract
Renal oncocytoma and chromophobe renal cell carcinoma were accepted as unique renal tumors in the late 1990s. Since their formal description, criteria for diagnosis have evolved and additional distinct tumor subtypes originally considered as one these two entities are now recognized. The last two decades have witnessed unprecedented interest in the spectrum of low grade oncocytic renal neoplasms in three specific areas: (1) histologic characterization of tumors with overlapping morphologic features between oncocytoma and chromophobe renal cell carcinoma; (2) description of potentially unique entities within this spectrum, such as eosinophilic vacuolated tumor and low-grade oncocytic tumor; and (3) better appreciation of the association between a subset of low grade oncocytic tumors and hereditary renal neoplasia. While this important work has been academically rewarding, the proposal of several histologic entities with overlapping morphologic and immunophenotypic features (which may require esoteric adjunctive immunohistochemical and/or molecular techniques for confirmation) has created frustration in the diagnostic pathology and urology community as information evolves regarding classification within this spectrum of renal neoplasia. Pathologists, including genitourinary subspecialists, are often uncertain as to the "best practice" diagnostic approach to such tumors. In this review, we present a practical clinically relevant algorithmic approach to classifying tumors within the low grade oncocytic family of renal neoplasia, including a proposal for compressing terminology for evolving categories where appropriate without sacrificing prognostic relevance.
Collapse
Affiliation(s)
- Mahul B Amin
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science, Memphis, TN, USA.
| | - Jesse K McKenney
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Guido Martignoni
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona, Italy.,Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Steven C Campbell
- Department of Urology, and Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sumanta Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Satish K Tickoo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
44
|
Mohanty SK, Lobo A, Cheng L. The 2022 revision of World Health Organization classification of tumors of the urinary system and male genital organs: advances and challenges. Hum Pathol 2022; 136:123-143. [PMID: 36084769 DOI: 10.1016/j.humpath.2022.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 01/07/2023]
Abstract
The fifth edition of the World Health Organization (WHO) classification of urinary and male genital organ tumors has been recently published in 2022. The application of molecular profiling has made a substantial impact on classification of urologic tumors. The new WHO classification introduces a group of molecularly well-defined renal tumor subtypes. The significant changes include addition of a category of "other oncocytic tumors" with oncocytoma/chromophobe renal cell carcinoma (chRCC)-like features, elimination of the subcategorization of type1/2 papillary RCC and inclusion of eosinophilic solid and cystic RCC as an independent tumor entity. The WHO/ISUP grading now has been recommended for all RCCs. Major nomenclature changes include replacement of histologic 'variants' by 'subtypes', 'clear cell papillary renal cell carcinoma' to 'clear cell renal cell tumor','TCEB1-mutated RCC' to 'ELOC-mutated RCC', 'hereditary leiomyomatosis and renal cell carcinoma' to 'fumarate hydratase-deficient RCC', 'RCC-Unclassified' to 'RCC-NOS', 'primitive neuroectodermal tumor' to 'embryonic neuroectodermal tumor', 'testicular carcinoid' to 'testicular neuroendocrine tumor', and 'basal cell carcinoma of the prostate' to 'adenoid-cystic (basal-cell) carcinoma of the prostate'. Metastatic, hematolymphoid, mesenchymal, melanocytic, soft tissue and neuroendocrine tumors are collectively discussed in separate chapters. It has been suggested that the morphological classification of urothelial cancer be replaced with a new molecular taxonomic classification system.
Collapse
Affiliation(s)
- Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute and CORE Diagnostics, Gurgaon, India (Zipcode:122016)
| | - Anandi Lobo
- Department of Pathology and Laboratory Medicine, Kapoor Center of Urology and Pathology, Raipur, India (Zipcode:490042)
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Lifespan Academic Medical Center, Providence, RI, USA (Zipcode: 02903).
| |
Collapse
|
45
|
Mapuranga H, Douglas-Jones B, du Plessis D, le Roux CE, du Buisson C, Moosa S. A rare case of tuberous sclerosis complex-associated renal cell carcinoma. SA J Radiol 2022; 26:2406. [PMID: 35747782 PMCID: PMC9210185 DOI: 10.4102/sajr.v26i1.2406] [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: 01/23/2022] [Accepted: 03/23/2022] [Indexed: 12/05/2022] Open
Abstract
Renal cell carcinoma is rarely described in paediatric patients with tuberous sclerosis complex. This report describes a case of an 11-year-old male with tuberous sclerosis-associated renal cell carcinoma.
Collapse
Affiliation(s)
- Humphrey Mapuranga
- Department of Radio-Diagnosis, Faculty of Medical Imaging and Clinical Oncology, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
| | - Bianca Douglas-Jones
- Department of Medical Genetics, Division of Molecular Biology and Human Genetics, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
| | - Danelo du Plessis
- Department of Surgery, Division of Urology, Faculty of Medicine and Health Sciences, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
| | - Camilla E le Roux
- Department of Radio-Diagnosis, Faculty of Medical Imaging and Clinical Oncology, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
| | - Christel du Buisson
- Department of Paediatrics and Child Health, Paediatric Nephrology, Faculty of Medicine and Health Sciences, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
| | - Shahida Moosa
- Department of Medical Genetics, Division of Molecular Biology and Human Genetics, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
46
|
Abstract
Renal cell carcinoma (RCC) with fibromyomatous stroma (FMS) was included as an "emerging/provisional" entity in the 2016 World Health Organization (WHO) classification as a "RCC with (angio) leiomyomatous stroma." It has been debated whether RCCFMS represents a separate entity or a group of RCCs with overlapping morphologies. Accordingly, various names have been used to refer to the RCCs that exhibited clear cells and prominent smooth muscle and fibromatous stroma. Recent studies have demonstrated that RCCFMS indeed represents a distinct entity with subtle but distinguishable features that can be separated from other RCCs that exhibit clear cells, as well as tubulopapillary morphology and smooth muscle/fibromatous stroma, such as clear cell RCC and clear cell papillary RCC. Microscopically, the epithelial component forms tumor nodules composed of elongated and frequently branching tubules, lined by clear or mildly eosinophilic cells containing voluminous cytoplasm. Focal papillary morphology is also frequently present. Diffuse CK7 positivity is typical and is required for the diagnosis. Molecular analysis of these tumors demonstrated recurrent mutations involving the TSC/mTOR pathway. A subset of tumors with similar morphology has shown mutations involving ELOC (previously referred to as TCEB1), typically associated with monosomy 8. Finally, in addition to the more common RCCFMS that are sporadic, essentially identical tumors have been found in patients with tuberous sclerosis complex, suggesting the existence of hereditary and sporadic counterparts of this tumor. It is currently debated whether TSC/mTOR and ELOC mutated RCCFMS should be grouped together, based on their shared and overlapping morphology and common CK7 reactivity, despite the differing molecular alterations. This review outlines evidence supporting the recognition of RCCFMS as a novel subtype of RCC with morphologic, immunohistochemical, and molecular characteristics distinct from clear cell RCC and clear cell papillary RCC.
Collapse
Affiliation(s)
- Rajal B Shah
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
47
|
Lobo J, Rechsteiner M, Helmchen BM, Rupp NJ, Weber A, Moch H. Eosinophilic solid and cystic renal cell carcinoma and renal cell carcinomas with TFEB alterations: a comparative study. Histopathology 2022; 81:32-43. [PMID: 35403742 DOI: 10.1111/his.14663] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/23/2022] [Accepted: 04/10/2022] [Indexed: 11/27/2022]
Abstract
AIMS Eosinophilic solid and cystic renal cell carcinoma (ESC RCC) is a recently described renal tumour entity with frequent CK20 positivity, commonly harbouring TSC mutations. In contrast, frequency of CK20 expression and presence of TSC mutations are unclear in TFEB-amplified RCC and TFEB-translocated RCC, which frequently express Melan A. Herein, we provide a comparative analysis of 6 ESC RCC with 4 TFEB-amplified/translocated RCC. METHODS AND RESULTS We assessed the frequency of CK20 and Melan A expression by immunohistochemistry, and of TSC mutations by next generation sequencing. TFEB alterations were confirmed by fluorescence in situ hybridization (FISH). All tumours showed voluminous eosinophilic cells with granular cytoplasm, prominent nucleoli, and most showed admixture of solid and cystic areas. CK20 expression was found in all 6 ESC RCC and in all RCCs with TFEB alterations. Melan A positivity was identified in 5/6 ESC RCC and 4/4 RCC with TFEB alterations. We found TSC mutations in 2 ESC RCCs, including in one case also harbouring a CIC fusion, and identified a TSC mutation in one TFEB-amplified RCC. CONCLUSIONS ESC RCC represents an emerging renal tumour entity with some histological, immunohistochemical and molecular overlap to TFEB-amplified/translocated RCC. FISH for TFEB aids in this differential diagnosis in challenging cases.
Collapse
Affiliation(s)
- João Lobo
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal.,Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513, Porto, Portugal
| | - Markus Rechsteiner
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH 8091, Zurich, Switzerland
| | - Birgit M Helmchen
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH 8091, Zurich, Switzerland
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH 8091, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Rämistrasse 71, 8006, Zurich, Switzerland
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH 8091, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH 8091, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Rämistrasse 71, 8006, Zurich, Switzerland
| |
Collapse
|
48
|
Estienne du Plessis D, Hofmeyr S, van der Merwe A. Clinical Challenge in Urology: Management of a 11-year-old boy with Tuberous Sclerosis associated Renal Cell Carcinoma. Urology 2022; 165:e9-e10. [DOI: 10.1016/j.urology.2022.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/15/2022] [Accepted: 03/26/2022] [Indexed: 10/18/2022]
|
49
|
Al-Obaidy KI, Alruwaii ZI, Williamson SR, Cheng L. The Pathologic and Molecular Genetic Landscape of the Hereditary Renal Cancer Predisposition Syndromes. Histopathology 2022; 81:15-31. [PMID: 35315118 DOI: 10.1111/his.14641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/18/2022] [Accepted: 03/02/2022] [Indexed: 11/30/2022]
Abstract
It is estimated that 5-8% of renal tumors are hereditary in nature with many inherited as autosomal dominant. These tumors carry a unique spectrum of pathologic and molecular alterations, the knowledge of which is expanding in the recent years. Indebted to this knowledge, many advances in treatment of these tumors have been achieved. In this review, we summarize the current understanding of the genetic renal neoplasia syndromes, the clinical and pathologic presentations, their molecular pathogenesis, the advances in therapeutic implications and targeted therapy.
Collapse
Affiliation(s)
- Khaleel I Al-Obaidy
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Zainab I Alruwaii
- Department of Pathology, Dammam Regional Laboratory and Blood Bank, Dammam, KSA
| | - Sean R Williamson
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Liang Cheng
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Urology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
50
|
Ritter DM, Fessler BK, Ebrahimi-Fakhari D, Wei J, Franz DN, Krueger DA, Trout AT, Towbin AJ. Prevalence of thoracoabdominal imaging findings in tuberous sclerosis complex. Orphanet J Rare Dis 2022; 17:124. [PMID: 35292049 PMCID: PMC8922878 DOI: 10.1186/s13023-022-02277-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/07/2022] [Indexed: 12/02/2022] Open
Abstract
Background Tuberous sclerosis complex (TSC) results in neurodevelopmental phenotypes, benign tumors, and cysts throughout the body. Recent studies show numerous rare findings in TSC. Guidelines suggest routine abdominal and chest imaging to monitor these thoracoabdominal findings, but imaging is not uniformly done across centers. Thus, the prevalence of many findings is unknown. To answer this, we categorized the clinical reads of 1398 thoracoabdominal scans from 649 patients of all ages in the Cincinnati Children’s Hospital TSC Repository Database. Results Typical TSC findings were present in many patients: kidney cysts (72%), kidney fat-containing angiomyolipomas (51%), kidney lipid-poor angiomyolipomas (27%), liver angiomyolipomas (19%), and lung nodules thought to represent multifocal micronodular pneumocyte hyperplasia (MMPH) (18%). While many features were more common in TSC2 patients, TSC1 patients had a higher prevalence of MMPH than TSC2 patients (24% versus 13%, p = 0.05). Many rare findings (e.g., lymphatic malformations and liver masses) are more common in TSC than in the general population. Additionally, most thoracoabdominal imaging findings increased with age except kidney cysts which decreased, with the 0–10 years age group having the highest percentage (69% 0–10 years, 49% 10–21 years, 48% 21 + years, p < 0.001). Finally, in our population, no patients had renal cell carcinoma found on abdominal imaging. Conclusions These results show that regular thoracoabdominal scans in TSC may show several findings that should not be ignored or, conversely, over-reacted to when found in patients with TSC. Female sex, TSC2 mutation, and age are risk factors for many thoracoabdominal findings. The data suggest novel interactions of genetic mutation with pulmonary nodules and age with renal cysts. Finally, in agreement with other works, these findings indicate that several rare thoracoabdominal imaging findings occur at higher rates in the TSC population than in the general population. This work supports obtaining detailed thoracoabdominal imaging in patients with TSC. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02277-x.
Collapse
Affiliation(s)
- David M Ritter
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Bailey K Fessler
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Daniel Ebrahimi-Fakhari
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster, Germany
| | - Jun Wei
- The First Hospital of Yichang, Yichang, China
| | - David N Franz
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Darcy A Krueger
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Andrew T Trout
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Alexander J Towbin
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|