1
|
Li H, Matoso A. Unlocking diagnostic potential: A retrospective analysis of GPNMB immunohistochemistry in nearly 1000 surgical pathology specimens. Hum Pathol 2025; 159:105799. [PMID: 40389121 DOI: 10.1016/j.humpath.2025.105799] [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: 05/04/2025] [Revised: 05/13/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Glycoprotein non-metastatic melanoma protein B (GPNMB) is a lysosomal transmembrane protein regulated by the TSC/mTOR-TFE pathway and has been proposed as a diagnostic immunohistochemical (IHC) marker for tumors associated with TSC/mTOR-TFE pathway alterations. However, its diagnostic performance in routine surgical pathology has not been systematically evaluated on a large scale. We retrospectively reviewed 934 cases from the Johns Hopkins pathology archives (2021-2025) in which GPNMB IHC was performed. Diagnoses were categorized into TSC/mTOR-TFE-related, non-TSC/mTOR/TFE-related, or undefined molecular groups. Correlation with fluorescence in situ hybridization (FISH) results and histologic features was performed to assess diagnostic utility. GPNMB was diffusely positive in 94.8 % (218/230) of TSC/mTOR-TFE-related neoplasms, including renal cell carcinomas with TFE3/TFEB rearrangements, perivascular epithelioid cell tumors (PECOMA/AML), and other related entities. In contrast, 83.3 % of non-TSC/mTOR-TFE-related tumors were negative for GPNMB. Discordant cases were seen in both groups, likely reflecting molecular heterogeneity, limitations of FISH, or the complex regulation of GPNMB expression. GPNMB outperformed cathepsin K in sensitivity in FISH-confirmed cases with TFE3 or TFEB alterations. Patchy or equivocal staining required careful histologic and ancillary correlation for interpretation. GPNMB IHC is a valuable ancillary tool in diagnosing TSC/mTOR-TFE-related neoplasms, particularly in renal and mesenchymal tumors. While not perfectly specific or sensitive, it offers a rapid and cost-effective alternative to molecular testing and can guide further diagnostic workup. Positive GPNMB staining in tumors without known TSC/mTOR-TFE alterations may suggest secondary pathway involvement, warranting additional molecular studies.
Collapse
Affiliation(s)
- Huili Li
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Andres Matoso
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Gupta A, Gupta P, Kaur A, Kumari S, Nalini G, Gainder S. Primary Uterine Alveolar Soft Part Sarcoma in a Postmenopausal Woman: Histopathologic and Immunohistochemical Characteristics of a Rare Case. Int J Surg Pathol 2024; 32:1165-1176. [PMID: 38086760 DOI: 10.1177/10668969231214810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
BACKGROUND Primary uterine alveolar soft part sarcoma (ASPS) is a rare, indolent mesenchymal malignancy with less than 40 patients documented in the literature. CASE We report an example of ASPS in a 61-year-old postmenopausal woman. Macroscopically, the uterus showed multiple nodular masses. Microscopic examination revealed tumor arranged in nests and alveolar pattern. The tumor cells were moderately to markedly pleomorphic, epithelioid to polygonal, with eccentrically placed nuclei, vesicular chromatin, prominent macro-nucleoli, and moderate to abundant eosinophilic cytoplasm. PAS-positive and diastase-resistant intracytoplasmic crystals were also seen in some tumor cells. On immunohistochemistry, the tumor cells showed diffuse positivity for vimentin and nuclear positivity for TFE3, a surrogate marker for ASPS. These were negative for SMA, desmin, CD10, h-caldesmon, cyclin D1, EMA, Melan A, and CD34. SMARCB1 expression was retained. Based on the histopathology and IHC, a final diagnosis of uterine ASPS was rendered. CONCLUSIONS Knowledge of the characteristic histopathologic and immunohistochemical features can help accurately diagnose such rare tumors. Knowledge of the characteristic histopathologic and immunohistochemical features can help accurately diagnose such rare sarcoma in an uncommon site with an unusual age.
Collapse
Affiliation(s)
- Anjali Gupta
- Department of Cytology and Gynecological Pathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Parikshaa Gupta
- Department of Cytology and Gynecological Pathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amarjot Kaur
- Department of Cytology and Gynecological Pathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Snigdha Kumari
- Department of Obstetrics and Gynecology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Gupta Nalini
- Department of Cytology and Gynecological Pathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Shalini Gainder
- Department of Obstetrics and Gynecology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
3
|
Collins K, Bridge JA, Mehra R, Mannan R, Dickson BC, Lotan TL, Idrees MT, Ulbright TM, Acosta AM. Renal epithelioid angiomyolipomas overexpress TFE3 and the TFE3-regulated gene TRIM63 in the absence of TFE3 rearrangement. Virchows Arch 2024; 485:471-478. [PMID: 38971946 DOI: 10.1007/s00428-024-03855-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/23/2024] [Accepted: 06/14/2024] [Indexed: 07/08/2024]
Abstract
Angiomyolipoma (AML) is a neoplasm within the perivascular epithelioid cell tumor family that occurs somewhat frequently in the kidney. Most are indolent and discovered incidentally, with rare tumors demonstrating malignant clinical behavior. A small subset of renal AMLs with epithelioid features are associated with aggressive behavior, and may demonstrate morphologic overlap with renal cell carcinomas (e.g., clear cell renal cell carcinoma (RCC), TFE3-rearranged RCC). Prior studies of spindle cell and epithelioid AMLs have identified rare examples with underlying TFE3 gene fusions. TFE3 protein expression (demonstrated by immunohistochemistry) with no evidence of concurrent TFE3 rearrangements has been reported previously in 4/24 AMLs (17%) (Argani et al. Am J Surg Pathol 34:1395-1406, 2010). Currently, the relationship between TFE3 protein expression, TFE3 fusions, and expression of TFE3-mediated genes remains incompletely understood in renal epithelioid AMLs. We sought to explore these relationships using TFE3 break-apart fluorescence in situ hybridization (FISH) and TRIM63 RNA in situ hybridization (ISH) on epithelioid AMLs with moderate to strong TFE3 expression by immunohistochemistry. RNA sequencing (fusion panel) was performed on two cases with negative FISH results to assess for FISH-cryptic gene fusions. The series comprised five epithelioid AMLs from four patients (three women, one man) aged 13 to 76 years. All were considered positive for TFE3 by immunohistochemistry (2 + /3 + expression). TRIM63 ISH was performed on four specimens from three patients, yielding positive results in 3/3 tumors (100%) that were successfully analyzed. TFE3 break-apart FISH was performed on all samples, demonstrating a TFE3 rearrangement in only 1/4 tumors (25%). RNA sequencing demonstrated the absence of productive TFE3 gene fusions in three tumors with negative break-apart TFE3 FISH results. This study demonstrates that renal epithelioid AMLs overexpress TFE3 and TFE3-mediated genes (TRIM63) even in the absence of TFE3 rearrangements. This finding could be explained by functional upregulation of TFE3 secondary to activation of the mammalian target of rapamycin complex 1 (mTORC1). Expression of TFE3 and TRIM63 in this tumor type represents a potential pitfall, given the morphologic and immunophenotypic overlap between epithelioid AML and TFE3-altered renal cell carcinoma.
Collapse
Affiliation(s)
- Katrina Collins
- Department of Pathology, Indiana University School of Medicine, 305 W 11 Street, Room 4080, Indianapolis, IN, USA
| | - Julia A Bridge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
- ProPath, Division of Molecular Pathology, Dallas, TX, USA
| | - Rohit Mehra
- Department of Pathology and Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Rahul Mannan
- Department of Pathology and Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Brendan C Dickson
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, University of Toronto, Toronto, Canada
| | - Tamara L Lotan
- Department of PathologyDepartment of OncologyDepartment of Urology, Johns Hopkins University School of MedicineJohns Hopkins University School of MedicineJohns Hopkins University School of Medicine, Baltimore, USA
| | - Muhammad T Idrees
- Department of Pathology, Indiana University School of Medicine, 305 W 11 Street, Room 4080, Indianapolis, IN, USA
| | - Thomas M Ulbright
- Department of Pathology, Indiana University School of Medicine, 305 W 11 Street, Room 4080, Indianapolis, IN, USA
| | - Andres M Acosta
- Department of Pathology, Indiana University School of Medicine, 305 W 11 Street, Room 4080, Indianapolis, IN, USA.
| |
Collapse
|
4
|
Repetto F, Sirohi D, Greipp P, Mahlow J. Incidental Detection of TFEB-Amplified Renal Cell Carcinoma by Colocated Gene Amplification of CCND3 (6p21): A Case Report and Review of the Literature. Int J Surg Pathol 2024; 32:551-555. [PMID: 37394760 DOI: 10.1177/10668969231185081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
TFEB-amplified renal cell carcinoma (RCC), which belongs to the MITF family of RCC, is characterized by genomic amplification at the 6p21.1 locus where the TFEB gene is located. The vascular endothelial growth factor A and cyclin D3 genes are also located at this same locus. When tumors lack classic morphologic features, they may be classified as "RCC not otherwise specified (NOS)." However, it is increasingly important to accurately diagnose the RCC subtype to define the patient's individual prognosis and select the subsequent therapeutic modalities, which now include targeted agents. Therefore, knowledge of the diagnostic features of TFEB-altered RCCs, such as t(6;11) RCCs and TFEB-amplified RCCs, is critical for identifying these tumors. Herein, we present an interesting case of TFEB-amplified RCC that was initially diagnosed as RCC NOS on biopsy of a renal tumor in a community practice setting with available molecular findings demonstrating CCND3 amplification. The genetic abnormality was "accidentally" detected due to the amplification of the colocated CCND3 gene at the 6p21 locus of the TFEB gene on a limited genetic sequencing panel. This case highlights the importance of molecular tests in accurately diagnosing RCC and carefully interpreting molecular findings in the context of histomorphologic features.
Collapse
Affiliation(s)
- Federico Repetto
- Favaloro University, School of Medicine, Buenos Aires, Argentina
| | - Deepika Sirohi
- University of Utah, Department of Pathology, Salt Lake City, UT, USA
| | - Patricia Greipp
- Mayo Clinic, Laboratory Medicine and Pathology, Rochester, MN, USA
| | - Jonathon Mahlow
- University of Utah, Department of Pathology, Salt Lake City, UT, USA
| |
Collapse
|
5
|
Mukherjee S, Basu K, Basu KS, Chatterjee U. MiT family translocation-associated renal cell carcinoma: A report of two cases in children. INDIAN J PATHOL MICR 2024; 67:469-471. [PMID: 38391376 DOI: 10.4103/ijpm.ijpm_385_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/16/2022] [Indexed: 02/24/2024] Open
Abstract
ABSTRACT Renal cell carcinoma is uncommon in children and must be distinguished from the more common Wilms' tumor. Here, we present two cases of renal cell carcinoma in children both of whom presented with hematuria. Accurate diagnosis is essential in order to differentiate it from epithelial predominant Wilms' tumor which has vastly different prognosis and treatment. Immunohistochemistry for TFE3 is useful in establishing the diagnosis.
Collapse
Affiliation(s)
| | - Keya Basu
- Department of Pathology, IPGME&R, Kolkata, West Bengal, India
| | - Kalyani S Basu
- Department of Pediatric Surgery, NRS Medical College, Kolkata, West Bengal, India
| | | |
Collapse
|
6
|
Wangsiricharoen S, Ingram DR, Morey RR, Wani K, Lazar AJ, Wang WL. Glycoprotein Nonmetastatic Melanoma Protein B (GPNMB) Immunohistochemistry Can Be a Useful Ancillary Tool to Identify Perivascular Epithelioid Cell Tumor. Mod Pathol 2024; 37:100426. [PMID: 38219952 DOI: 10.1016/j.modpat.2024.100426] [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: 06/22/2023] [Revised: 12/13/2023] [Accepted: 01/07/2024] [Indexed: 01/16/2024]
Abstract
Perivascular epithelioid cell tumors (PEComas) are rare mesenchymal tumors that express smooth muscle and melanocytic makers. Diagnosis of PEComas can be challenging due to focal or lost expression of traditional immunohistochemical markers, limited availability of molecular testing, and morphological overlap with much more common smooth muscle tumors. This study evaluates the use of glycoprotein nonmetastatic melanoma protein B (GPNMB) immunohistochemical staining as a surrogate marker for TSC1/2/MTOR alteration or TFE3 rearrangement to differentiate PEComas from other mesenchymal tumors. Cathepsin K was also assessed for comparison. A total of 399 tumors, including PEComas, alveolar soft part sarcomas, and other histologic PEComa mimics, were analyzed using GPNMB and cathepsin K immunohistochemistry. GPNMB expression was seen in all PEComas and alveolar soft part sarcomas with the majority showing diffuse and moderate-to-strong labeling, whereas other sarcomas were negative or showed focal labeling. When a cutoff of diffuse and at least moderate staining was used, GPNMB demonstrated 95% sensitivity and 97% specificity in distinguishing PEComas from leiomyosarcoma, well-differentiated/dedifferentiated liposarcomas, and undifferentiated pleomorphic sarcomas. Cathepsin K with a cutoff of any labeling had lower sensitivity (78%) and similar specificity (94%) to GPNMB. This study highlights GPNMB as a highly sensitive marker for PEComas and suggests its potential use as an ancillary tool within a panel of markers for accurate classification of these tumors.
Collapse
Affiliation(s)
- Sintawat Wangsiricharoen
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Pathology and Laboratory Medicine, Oregon Health & Science University, Portland, Oregon
| | - Davis R Ingram
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rohini R Morey
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Khalida Wani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexander J Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei-Lien Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
7
|
Taylor AS, Mannan R, Pantanowitz L, Chinnaiyan AM, Dhanasekaran SM, Hrycaj S, Cao X, Chan MP, Lucas D, Wang XM, Mehra R. Evaluation of TRIM63 RNA in situ hybridization (RNA-ISH) as a potential biomarker for alveolar soft-part sarcoma (ASPS). Med Oncol 2024; 41:76. [PMID: 38393424 PMCID: PMC10891236 DOI: 10.1007/s12032-024-02305-9] [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: 10/03/2023] [Accepted: 01/17/2024] [Indexed: 02/25/2024]
Abstract
Alveolar soft-part sarcoma (ASPS) is a rare soft tissue tumor with a broad morphologic differential diagnosis. While histology and immunohistochemistry can be suggestive, diagnosis often requires exclusion of other entities followed by confirmatory molecular analysis for its characteristic ASPSCR1-TFE3 fusion. Current stain-based biomarkers (such as immunohistochemistry for cathepsin K and TFE3) show relatively high sensitivity but may lack specificity, often showing staining in multiple other entities under diagnostic consideration. Given the discovery of RNA in situ hybridization (RNA-ISH) for TRIM63 as a sensitive and specific marker of MiTF-family aberration renal cell carcinomas, we sought to evaluate its utility in the workup of ASPS. TRIM63 RNA-ISH demonstrated high levels (H-score greater than 200) of expression in 19/20 (95%) cases of ASPS (average H-score 330) and was weak or negative in cases of paraganglioma, clear cell sarcoma, rhabdomyosarcoma, malignant epithelioid hemangioendothelioma, as well as hepatocellular and adrenal cortical carcinomas. Staining was also identified in tumors with known subsets characterized by TFE3 alterations such as perivascular epithelioid cell neoplasm (PEComa, average H-score 228), while tumors known to exhibit overexpression of TFE3 protein without cytogenetic alterations, such as melanoma and granular cell tumor, generally showed less TRIM63 ISH staining (average H-scores 147 and 96, respectively). Quantitative assessment of TRIM63 staining by RNA-ISH is potentially a helpful biomarker for tumors with molecular TFE3 alterations such as ASPS.
Collapse
Affiliation(s)
- Alexander S Taylor
- Department of Pathology, University of Michigan Medical School, 2800 Plymouth Road, Building 35, Ann Arbor, MI, 48109, USA
| | - Rahul Mannan
- Department of Pathology, University of Michigan Medical School, 2800 Plymouth Road, Building 35, Ann Arbor, MI, 48109, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Liron Pantanowitz
- Department of Pathology, University of Michigan Medical School, 2800 Plymouth Road, Building 35, Ann Arbor, MI, 48109, USA
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan Medical School, 2800 Plymouth Road, Building 35, Ann Arbor, MI, 48109, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
- Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, Ann Arbor, MI, USA
| | - Saravana M Dhanasekaran
- Department of Pathology, University of Michigan Medical School, 2800 Plymouth Road, Building 35, Ann Arbor, MI, 48109, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - Steven Hrycaj
- Department of Pathology, University of Michigan Medical School, 2800 Plymouth Road, Building 35, Ann Arbor, MI, 48109, USA
| | - Xuhong Cao
- Department of Pathology, University of Michigan Medical School, 2800 Plymouth Road, Building 35, Ann Arbor, MI, 48109, USA
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA
| | - May P Chan
- Department of Pathology, University of Michigan Medical School, 2800 Plymouth Road, Building 35, Ann Arbor, MI, 48109, USA
| | - David Lucas
- Department of Pathology, University of Michigan Medical School, 2800 Plymouth Road, Building 35, Ann Arbor, MI, 48109, USA
| | - Xiao-Ming Wang
- Department of Pathology, University of Michigan Medical School, 2800 Plymouth Road, Building 35, Ann Arbor, MI, 48109, USA.
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA.
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, 2800 Plymouth Road, Building 35, Ann Arbor, MI, 48109, USA.
- Michigan Center for Translational Pathology, Ann Arbor, MI, USA.
- Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Argani P, Medeiros LJ, Matoso A, Baraban E, Lotan T, Pawel BR, McKenney JK, Mehra R, Falzarano SM, Pallavajjalla A, Lin MT, Patel S, Rawwas J, Bendel AE, Gagan J, Palsgrove DN. "Oncocytoid Renal Cell Carcinomas After Neuroblastoma" Represent TSC -mutated Eosinophilic Solid and Cystic Renal Cell Carcinomas : Association With Prior Childhood Malignancy and Multifocality With Therapeutic Implications. Am J Surg Pathol 2023; 47:1335-1348. [PMID: 37522346 DOI: 10.1097/pas.0000000000002101] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
The concept of oncocytoid renal cell carcinoma in patients who have survived neuroblastoma as a distinct biologic entity has been controversial since its original description in 1999. This is in part because similar oncocytoid renal cell carcinomas have been described in association with other pediatric cancers, and also because other renal cell carcinoma subtypes (such as MiT family translocation renal cell carcinoma) have been described in children who have survived neuroblastoma. We identified an index case of a child who survived medulloblastoma and developed multifocal bilateral oncocytoid renal cell carcinomas with morphology and immunophenotype compatible with eosinophilic solid and cystic renal cell carcinoma (ESC RCC) and demonstrated that both neoplasms harbored distinctive mutations in the TSC1/TSC2 genes. Remarkably, the child's remaining bilateral multifocal renal neoplasms completely responded to MTOR inhibitor therapy without need for further surgery. To confirm our hypothesis that oncocytoid renal cell carcinomas after childhood cancer represent ESC RCC, we obtained formalin-fixed paraffin-embedded tissue blocks from 2 previously published cases of oncocytoid renal cell carcinoma after neuroblastoma, confirmed that the morphology and immunophenotype was consistent with ESC RCC, and demonstrated that both cases harbored somatic TSC gene mutations. Both expressed markers previously associated with neoplasms harboring TSC gene mutations, glycoprotein nonmetastatic B, and cathepsin K. Of note, one of these patients had 2 ESC RCC which harbored distinctive TSC2 mutations, while the background kidney of the other patient had multiple small cysts lined by similar oncocytoid cells which showed loss of TSC2 protein. We then reviewed 3 of 4 cases from the original 1999 report of oncocytoid renal cell carcinomas after neuroblastoma, found that all 3 demonstrated morphology (including basophilic cytoplasmic stippling) that is characteristic of ESC RCC, showed that all 3 overexpressed glycoprotein nonmetastatic B, and showed that both cases with adequate material demonstrated loss of TSC2 protein and expressed cytokeratin 20 and cathepsin K by immunohistochemistry. In summary, "oncocytoid renal cell carcinomas after neuroblastoma" represent ESC RCC which are often multifocal in patients who have survived childhood cancer, likely representing an incompletely characterized tumor predisposition syndrome. MTOR-targeted therapy represents an effective therapeutic option for such patients to preserve functional nephrons.
Collapse
Affiliation(s)
- Pedram Argani
- Departments of Pathology
- Urology
- Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - L Jeffrey Medeiros
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston
| | - Andres Matoso
- Departments of Pathology
- Urology
- Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ezra Baraban
- Departments of Pathology
- Urology
- Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Tamara Lotan
- Departments of Pathology
- Urology
- Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Bruce R Pawel
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles and Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Jesse K McKenney
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Rohit Mehra
- Department of Pathology and Michigan Center for Translational Pathology (MCTP), University of Michigan School of Medicine, Ann Arbor, MI
| | - Sara M Falzarano
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Aparna Pallavajjalla
- Departments of Pathology
- Urology
- Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ming-Tseh Lin
- Departments of Pathology
- Urology
- Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Jawhar Rawwas
- Hematology/Oncology, Children's Minnesota, Minneapolis, MN
| | - Anne E Bendel
- Hematology/Oncology, Children's Minnesota, Minneapolis, MN
| | - 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
|
9
|
Bai Z, Yan C, Chang D. Prediction and therapeutic targeting of the tumor microenvironment-associated gene CTSK in gastric cancer. Discov Oncol 2023; 14:200. [PMID: 37930479 PMCID: PMC10628060 DOI: 10.1007/s12672-023-00821-0] [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: 05/05/2023] [Accepted: 11/02/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Cathepsin-K (CTSK) is overexpressed in Gastric cancer (GC) and the mechanism of its overexpression in GC is still unclear. The present work found CTSK as a potential predictive biomarker and immunotherapeutic target for GC based on the tumor microenvironment (TME). METHODS From public databases, gene expression profiles and clinical data of GC were downloaded to analyze the distribution of stromal and immune cells and tumor abundance in TME. Differentially expressed genes (DEGs) associated with TME were obtained by differential analysis, followed by cross-screening to obtain CTSK as a gene associated with TME. Next, a series of methods and tools were employed to explore the relationships between clinicopathological features of GC and CTSK expression as well as prognosis, tumor immune microenvironment, immune checkpoints and drug sensitivity. And GSEA was used to investigate the potential role of CTSK in the tumor microenvironment of GC. RESULTS From the dataset, we obtained a total of 656 DEGs associated with TME and the stromal component of TME was found to be closely involved in GC prognosis. CTSK was cross-screened as the key gene associated with TME by the PPI network and univariate Cox regression analysis. Pan-cancer analysis revealed significant high expression of CTSK in a variety of cancers. Subsequently, we hypothesized that high-expressed CTSK was closely correlated with poor prognosis and lymph node metastasis of tumors, and that CTSK, a GC TME-related gene, was largely involved in a range of biological behaviors of tumors, with a significant correlation between several immune cells. CONCLUSION CTSK was validated as a potential prognostic biomarker related to TME of GC and could be a promising next-generation immunotherapeutic target for GC.
Collapse
Affiliation(s)
- Zilong Bai
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shanxi, China
| | - Chunyu Yan
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shanxi, China
| | - Dongmin Chang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shanxi, China.
| |
Collapse
|
10
|
Suneel R, Gupta P, Sekar A, Gupta N, Saxena A. Unravelling the mysteries of Xp11.2 translocation-associated renal cell carcinoma: A case report with a review of the literature. Cytopathology 2023; 34:367-376. [PMID: 37118914 DOI: 10.1111/cyt.13240] [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: 02/18/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/30/2023]
Abstract
Cytomorphologic and immunocytochemical features of TFE3 translocation-associated renal cell carcinoma.
Collapse
Affiliation(s)
- Rachagiri Suneel
- Department of Cytology and Gynecologic Pathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Parikshaa Gupta
- Department of Cytology and Gynecologic Pathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Aravind Sekar
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Nalini Gupta
- Department of Cytology and Gynecologic Pathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Akshay Saxena
- Department of Radiodiagnosis, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
11
|
Argani P, Matoso A, Baraban EG, Epstein JI, Antonescu CR. MED15::TFE3 Renal Cell Carcinomas: Report of Two New Cases and Review of the Literature Confirming Nearly Universal Multilocular Cystic Morphology. Int J Surg Pathol 2023; 31:409-414. [PMID: 36591911 PMCID: PMC10175165 DOI: 10.1177/10668969221143455] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
We report two novel cases of Xp11 translocation renal cell carcinomas with the MED15::TFE3 gene fusion in adult females aged 40 and 74 years. Both cases were extensively cystic and contained only minimal clear cells lining cysts and within septal walls, raising the differential diagnosis of multilocular cystic renal neoplasm of low malignant potential. By immunohistochemistry, both neoplasms labeled for PAX8, TFE3, cathepsin K and Melan A but not for HMB45. On review of the published literature and the two cases reported herein, over 90% of MED15::TFE3 renal cell carcinomas (RCCs) have been described as cystic. The correlation of the MED15::TFE3 fusion with extensively cystic morphology represents the strongest association of TFE3 fusion partner with clinicopathological features among TFE3-rearranged RCC reported to date.
Collapse
Affiliation(s)
- Pedram Argani
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Departments of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Andres Matoso
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Departments of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Departments of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Ezra G. Baraban
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Jonathan I. Epstein
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Departments of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Departments of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | |
Collapse
|
12
|
Lin J, Tang Z, Zhang C, Dong W, Liu Y, Huang H, Liu H, Huang J, Lin T, Chen X. TFE3 gene rearrangement and protein expression contribute to a poor prognosis of renal cell carcinoma. Heliyon 2023; 9:e16076. [PMID: 37215783 PMCID: PMC10196445 DOI: 10.1016/j.heliyon.2023.e16076] [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: 01/24/2023] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023] Open
Abstract
Background TFE3-rearranged renal cell carcinoma (TFE3-rearranged RCC) is a type of kidney cancer with a low incidence, with no consensus about whether it has a worse prognosis than clear cell renal cell carcinoma (ccRCC). This study attempted to elucidate the impact of TFE3-rearranged RCC by analyzing its clinical features and prognosis. Methods Patients treated in Sun Yat-sen Memorial Hospital (SYSMH) who were suspected to be diagnosed with TFE3-rearranged RCC were divided into two groups, TFE3-rearranged RCC and ccRCC with positive TFE3 protein expression on immunohistochemistry [TFE3(+) ccRCC], by dual-color, break-apart fluorescence in situ hybridization (FISH). After balancing the baseline characteristics with TFE3(+) ccRCC using the propensity score matching (PSM) method in a ratio of 2, we selected patients diagnosed with ccRCC with negative TFE3 protein expression on immunohistochemistry [TFE3(-) ccRCC]. The impact of TFE3 gene rearrangement and protein expression on renal cell carcinoma was determined by feature comparison with a nonparametric test and survival analysis with the Kaplan‒Meier method. Results Among 37 patients suspected of having TFE3-rearranged RCC, 13 patients were diagnosed with TFE3-rearranged RCC, and 24 patients had TFE3(+) ccRCC. The recurrence and new metastasis of TFE3-rearranged RCC was relatively common, even if the tumor stage was early at the first diagnosis. Through feature comparison and survival analysis, we found that TFE3-rearranged RCC was quite similar to TFE3(+) ccRCC. Compared with TFE3(-) ccRCC, TFE3(+) ccRCC tended to have a larger tumor diameter (P = 0.011), higher neutrophil/lymphocyte ratio (NLR) (P = 0.017) and metastatic potential (P = 0.022), and worse overall survival (OS) (P = 0.043) and PFS (P = 0.016). The survival analysis showed that TFE3-rearranged RCC had a worse PFS than ccRCC (P = 0.002), and TFE3(+) RCC had a worse PFS than TFE3(-) RCC (P = 0.001). According to the stratification system based on the combination of TFE3 and lymphovascular invasion (LVI), we further found that the prognosis from good to poor was TFE3(-) LVI(-), TFE3(+) LVI(-), TFE3(+) LVI(+) and TFE3(-) LVI(+), with statistically significant differences in both OS (P = 0.001) and PFS (P < 0.001). In addition, we also reported two cases with poor prognosis, of which one was TFE3-rearranged RCC and the other was TFE3(+) ccRCC. Conclusions This is a novel finding that both FISH confirmed TFE3 gene rearrangement-mediated TFE3-rearranged RCC and IHC confirmed positive TFE3 protein expression [TFE3(+)] contribute to a poor prognosis in RCC, suggesting more active treatment and careful follow-up for TFE3(+) RCC patients. The combination of TFE3 and LVI may be a new risk stratification system for RCC.
Collapse
Affiliation(s)
- Junyi Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, Guangzhou Urology Research Institute, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhuang Tang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Chengjunyu Zhang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Wen Dong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, China
| | - Yeqing Liu
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Hao Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Hao Liu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, China
| | - Xu Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, China
| |
Collapse
|
13
|
Choi JH, Ro JY. The Recent Advances in Molecular Diagnosis of Soft Tissue Tumors. Int J Mol Sci 2023; 24:ijms24065934. [PMID: 36983010 PMCID: PMC10051446 DOI: 10.3390/ijms24065934] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
Soft tissue tumors are rare mesenchymal tumors with divergent differentiation. The diagnosis of soft tissue tumors is challenging for pathologists owing to the diversity of tumor types and histological overlap among the tumor entities. Present-day understanding of the molecular pathogenesis of soft tissue tumors has rapidly increased with the development of molecular genetic techniques (e.g., next-generation sequencing). Additionally, immunohistochemical markers that serve as surrogate markers for recurrent translocations in soft tissue tumors have been developed. This review aims to provide an update on recently described molecular findings and relevant novel immunohistochemical markers in selected soft tissue tumors.
Collapse
Affiliation(s)
- Joon Hyuk Choi
- Department of Pathology, Yeungnam University College of Medicine, 170 Hyeonchung-ro, Namgu, Daegu 42415, Republic of Korea
| | - Jae Y Ro
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Medical College, Cornell University, Houston, TX 77030, USA
| |
Collapse
|
14
|
Takamatsu D, Kohashi K, Kiyozawa D, Kinoshita F, Ieiri K, Baba M, Eto M, Oda Y. TFE3-immunopositive papillary renal cell carcinoma: A clinicopathological, immunohistochemical, and genetic study. Pathol Res Pract 2023; 242:154313. [PMID: 36669395 DOI: 10.1016/j.prp.2023.154313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/14/2023] [Indexed: 01/18/2023]
Abstract
It is possible that PRCCs may still contain a variety of unknown histologic subtypes. Some PRCCs express high expression of TFE3 protein without TFE3 gene rearrangement, but no reports have investigated the significance of this. Here we attempted to examine clinicopathological and molecular significance of the TFE3-immunopositive PRCC. We reviewed the histology and immunohistochemistry in 58 PRCCs. TFE3 immunoexpression was recognized in 7 cases. Because TFE3 immunostaining shows false-positive, to ensure the integrity of TFE3 immunostaining, the immunostaining was performed under strict control of internal controls and western blotting was performed on 2 positive cases and 5 negative cases, and differences in protein expression between two groups were confirmed. Significant immunohistochemical expressions of autophagy/lysosome proteins were observed in TFE3-positive group. No TFE3 gene arrangement was detected in all positive cases by fluorescence in situ hybridization. Whole-exome sequencing was performed on 6 TFE3-positive and 2 TFE3-negative cases. Gain of chromosome 7 was found in five of 6 TFE3-positive cases (83%). TFE3-positive group was correlated significantly with higher pTstage, cNstage, WHO/ISUP nuclear grade, and decreased OS. TFE3-immunopositive PRCC group had a poorer prognosis than TFE3-negative PRCC group and showed correlation with expressions of autophagy/lysosome proteins, suggesting that enhancement of autophagy/lysosome function drives an environment of energy metabolism that is favorable for cancer. It is necessary to recognize that there is TFE3-immunopositive group without TFE3 gene rearrangement within PRCC. Because of its aggressive biological behaviour, TFE3 can act as a biomarker in PRCC; moreover, autophagy-inhibiting drugs may have therapeutic effects on TFE3-immunopositive PRCC.
Collapse
Affiliation(s)
- Dai Takamatsu
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, Japan
| | - Daisuke Kiyozawa
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, Japan
| | - Fumio Kinoshita
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, Japan
| | - Kosuke Ieiri
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, Japan
| | - Masaya Baba
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Higashi-Ku, Fukuoka, Japan.
| |
Collapse
|
15
|
Taylor AS, Skala SL. Tumors masquerading as type 2 papillary renal cell carcinoma: pathologists' ever-expanding differential diagnosis for a heterogeneous group of entities. Urol Oncol 2022; 40:499-511. [PMID: 34116938 DOI: 10.1016/j.urolonc.2021.04.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 03/22/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023]
Abstract
Although papillary renal cell carcinoma has historically been classified as either type 1 or type 2, data from The Cancer Genome Atlas (TCGA) has demonstrated significant genomic heterogeneity in tumors classified as "type 2 papillary renal cell carcinoma" (T2PRCC). Papillary renal cell carcinoma is expected to have a favorable clinical course compared to clear cell renal cell carcinoma (CCRCC). However, tumors with poor outcome more similar to CCRCC were included in the T2PRCC cohort studied by the TCGA. The differential diagnosis for T2PRCC includes a variety of other renal tumors, including aggressive entities such as TFE3 translocation-associated renal cell carcinoma, TFEB-amplified renal cell carcinoma, fumarate hydratase-deficient renal cell carcinoma, high-grade CCRCC, and collecting duct carcinoma. Accurate classification of these tumors is important for prognostication and selection of therapy.
Collapse
Affiliation(s)
- Alexander S Taylor
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Stephanie L Skala
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI.
| |
Collapse
|
16
|
MiT translocation renal cell carcinoma: A review of the literature from molecular characterization to clinical management. Biochim Biophys Acta Rev Cancer 2022; 1877:188823. [DOI: 10.1016/j.bbcan.2022.188823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/17/2022] [Accepted: 10/04/2022] [Indexed: 11/23/2022]
|
17
|
Cathepsin K: A Versatile Potential Biomarker and Therapeutic Target for Various Cancers. Curr Oncol 2022; 29:5963-5987. [PMID: 36005209 PMCID: PMC9406569 DOI: 10.3390/curroncol29080471] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 12/11/2022] Open
Abstract
Cancer, a common malignant disease, is one of the predominant causes of diseases that lead to death. Additionally, cancer is often detected in advanced stages and cannot be radically cured. Consequently, there is an urgent need for reliable and easily detectable markers to identify and monitor cancer onset and progression as early as possible. Our aim was to systematically review the relevant roles of cathepsin K (CTSK) in various possible cancers in existing studies. CTSK, a well-known key enzyme in the bone resorption process and most studied for its roles in the effective degradation of the bone extracellular matrix, is expressed in various organs. Nowadays, CTSK has been involved in various cancers such as prostate cancer, breast cancer, bone cancer, renal carcinoma, lung cancer and other cancers. In addition, CTSK can promote tumor cells proliferation, invasion and migration, and its mechanism may be related to RANK/RANKL, TGF-β, mTOR and the Wnt/β-catenin signaling pathway. Clinically, some progress has been made with the use of cathepsin K inhibitors in the treatment of certain cancers. This paper reviewed our current understanding of the possible roles of CTSK in various cancers and discussed its potential as a biomarker and/or novel molecular target for various cancers.
Collapse
|
18
|
Xanthomatous Giant Cell Renal Cell Carcinoma: Another Morphologic Form of TSC-associated Renal Cell Carcinoma. Am J Surg Pathol 2022; 46:1554-1561. [PMID: 35941720 DOI: 10.1097/pas.0000000000001940] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Over the past decade, several distinct novel renal epithelial neoplasms driven by underlying tuberous sclerosis comples (TSC)/mammalian target of rapamycin (MTOR) pathway mutations have been described. We report herein two distinctive TSC2-mutated renal cell carcinomas which do not fit any previously described entity. The two renal carcinomas occurred in young patients (ages 10 and 31 y), and were characterized by highly permeative growth within the kidney with metastases to perirenal lymph nodes. The neoplastic cells were predominantly large, multinucleated giant cells having variably eosinophilic to xanthomatous cytoplasm with basophilic stippling and frequent vacuolization. While the discohesive nature of the neoplastic cells, xanthomatous cytoplasm, immunoreactivity for histiocytic markers and minimal immunoreactivity for conventional epithelial markers raised the possibility of a histiocytic neoplasm, multifocal immunoreactivity for cytokeratin 20 helped establish their epithelial nature. Despite the aggressive growth pattern of these neoplasms and lymph node metastases, mitotic figures were rare and Ki-67 indices were low (<1%). One patient with follow-up shows no evidence of disease seven years after nephrectomy with no adjuvant therapy. Next-generation sequencing demonstrated TSC2 mutations in each case. By immunohistochemistry, downstream markers of mTOR pathway activation S6K1, 4EBP1, and glycoprotein nonmetastatic melanoma protein B were all highly expressed in these neoplasms, suggesting mTOR pathway activation as the neoplastic driver. While the cytokeratin 20 immunoreactivity and focal basophilic cytoplasmic stippling suggest a relationship to eosinophilic solid and cystic renal cell carcinoma, and cytoplasmic vacuolization suggests a relationship to eosinophilic vacuolated tumor, these neoplasms appear to be distinctive given their permeative growth patterns and predominant xanthomatous giant cell morphology. Addition of cytokeratin 20 to a panel of epithelial markers helps avoid misdiagnosis in such cases.
Collapse
|
19
|
Kmeid M, Akgul M. TFE3 Rearrangement and Expression in Renal Cell Carcinoma. Int J Surg Pathol 2022:10668969221108517. [PMID: 35912477 DOI: 10.1177/10668969221108517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
TFE3 rearranged Renal cell carcinoma (RCC) is not very common, and demonstrates unique heterogenous morphological features overlapping other recognized entities and distinct immunoprofile. It can be seen in any age group, therefore practicing pathologists should be aware of the distinctive clinical settings and histologic findings associated with these tumors and subsequently employ an adequate panel of ancillary studies in order to confirm the diagnosis. Recognizing these entities remains crucial for future clinical trials and development of novel therapies.
Collapse
Affiliation(s)
- Michel Kmeid
- Department of Pathology and Laboratory Medicine, 138207Albany Medical Center, Albany, NY, USA
| | - Mahmut Akgul
- Department of Pathology and Laboratory Medicine, 138207Albany Medical Center, Albany, NY, USA
| |
Collapse
|
20
|
Tretiakova MS. Chameleon TFE3-translocation RCC and How Gene Partners Can Change Morphology: Accurate Diagnosis Using Contemporary Modalities. Adv Anat Pathol 2022; 29:131-140. [PMID: 35180736 DOI: 10.1097/pap.0000000000000332] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Translocation renal cell carcinoma (tRCC) with TFE3 gene rearrangements has been born as a distinct entity 20 years ago. These relatively rare tumors were notable among other RCC subtypes because of their disproportionally high incidence among children and young adults. Initial reports were focused on describing unifying morphologic criteria and typical clinical presentation. Follow-up studies of ancillary immunohistochemical and hybridization techniques provided additional diagnostic tools allowing recognition of tRCC tumors in practice. However, a growing body of literature also expanded the clinicomorphologic spectrum of tRCCs, to include a significant morphologic overlap with other RCC variants thus blurring the diagnostic clarity of this entity. More recent molecular studies utilizing next-generation sequencing technology accelerated recognition of numerous novel gene partners fusing at different breakpoints with the TFE3 gene. Accumulating data indicates that morphologic and clinical heterogeneity of tRCC could be explained by fusion subtypes, and knowledge of TFE3 partnering genes may be important in predicting tumor behavior. Herein we provided a comprehensive analysis of ∼400 tRCC cases with known TFE3 fusion partners, estimated their relative incidence and summarized clinicomorphologic features associated with most common fusion subtypes. Our data was based on an extensive literature review and had a special focus on comparing immunohistochemistry, fluorescent in situ hybridization and contemporary molecular studies for the accurate diagnosis of tRCC.
Collapse
Affiliation(s)
- Maria S Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| |
Collapse
|
21
|
Munari E, Settanni G, Caliò A, Segala D, Lonardi S, Sandrini S, Vacca P, Tumino N, Marconi M, Brunelli M, Gobbo S, Netto GJ, Moretta L, Zamboni G, Martignoni G. TSC loss is a clonal event in eosinophilic solid and cystic renal cell carcinoma: a multiregional tumor sampling study. Mod Pathol 2022; 35:376-385. [PMID: 33990704 DOI: 10.1038/s41379-021-00816-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022]
Abstract
Eosinophilic, solid and cystic (ESC) renal cell carcinoma (RCC) is characterized by a solid and cystic architecture with cells showing abundant eosinophilic cytoplasm with hobnail arrangement and a cytokeratin 7-negative/cytokeratin 20-positive immunophenotype. Recent studies have suggested that bi-allelic events affecting TSC genes might play an important role for such tumors. However, only indirect evidence of the clonal origin of TSC mutation has been gathered so far. Therefore, in this paper we aimed to perform multi-regional tumor sampling molecular analysis in four ESC RCC cases that had been completely embedded, three sporadic and one occurring in a patient with tuberous sclerosis complex (TSC). Histologically, the 4 cases showed cystic and solid architecture and cells with abundant eosinophilic cytoplasm with cytoplasmic stippling and round to oval nuclei. Immunohistochemistry showed at least focal expression of cytokeratin 20 in all tissue samples and negative cytokeratin 7, as well as diffuse positivity for S100A1 and at least focal expression of cathepsin K in three out of four cases. The sporadic cases showed the same somatic TSC1 mutations in all tissue samples analyzed, while the TSC-associated case showed the same TSC1 alteration in both normal tissue and all tumor samples analyzed, proving the germline nature of the alteration. In conclusion, our data demonstrate that clonal TSC loss is a key event in ESC RCC and support considering ESC RCC as an entity given its distinct morphologic, immunophenotypical and molecular characteristics.
Collapse
Affiliation(s)
- Enrico Munari
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giulio Settanni
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy
| | - Anna Caliò
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Diego Segala
- Pathology Unit, ASST-Spedali Civili, Brescia, Italy
| | - Sara Lonardi
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy
| | - Silvia Sandrini
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy
| | - Paola Vacca
- Immunology Area, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Nicola Tumino
- Immunology Area, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Marcella Marconi
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy
| | - Matteo Brunelli
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Stefano Gobbo
- Pathology Unit, Pederzoli Hospital, Peschiera del Garda, Verona, Italy
| | - George J Netto
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lorenzo Moretta
- Immunology Area, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Giuseppe Zamboni
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy.,Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Guido Martignoni
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy. .,Pathology Unit, Pederzoli Hospital, Peschiera del Garda, Verona, Italy.
| |
Collapse
|
22
|
Caliò A, Marletta S, Brunelli M, Martignoni G. WHO 2022 Classification of Kidney Tumors: what is relevant? An update and future novelties for the pathologist. Pathologica 2022; 115:23-31. [PMID: 36645397 DOI: 10.32074/1591-951x-814] [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: 10/15/2022] [Accepted: 10/21/2022] [Indexed: 01/17/2023] Open
Abstract
Classification systems reflect our technical abilities in the investigation of tumors and our current theories on tumor development. Herein, by providing a historical perspective on the evolution of classifying renal tumors, we assess the current WHO classification highlighting the novelties and the implications of these changes in daily clinical practice.
Collapse
Affiliation(s)
- Anna Caliò
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Italy
| | - Stefano Marletta
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Italy.,Department of Pathology, Pederzoli Hospital, Peschiera, Italy
| | - Matteo Brunelli
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Italy
| | - Guido Martignoni
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Italy.,Department of Pathology, Pederzoli Hospital, Peschiera, Italy
| |
Collapse
|
23
|
TFE3 and TFEB-rearranged renal cell carcinomas: an immunohistochemical panel to differentiate from common renal cell neoplasms. Virchows Arch 2022; 481:877-891. [PMID: 35980471 PMCID: PMC9734233 DOI: 10.1007/s00428-022-03380-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/08/2022] [Accepted: 07/15/2022] [Indexed: 01/22/2023]
Abstract
TFE3/TFEB-rearranged renal cell carcinomas are characterized by translocations involving TFE3 and TFEB genes. Despite the initial description of typical morphology, their histological spectrum is wide, mimicking common subtypes of renal cell tumors. Thus, the diagnosis is challenging requiring the demonstration of the gene rearrangement, usually by FISH. However, this technique is limited in most laboratories and immunohistochemical TFE3/TFEB analysis is inconsistent. We sought to identify a useful immunohistochemical panel using the most common available markers to recognize those tumors. We performed an immunohistochemical panel comparing 27 TFE3-rearranged and 10 TFEB-rearranged renal cell carcinomas to the most common renal cell tumors (150 clear cell, 100 papillary, 50 chromophobe renal cell carcinomas, 18 clear cell papillary renal cell tumors, and 50 oncocytomas). When dealing with neoplasms characterized by cells with clear cytoplasm, CA9 is a helpful marker to exclude clear cell renal cell carcinoma. GATA3, AMACR, and CK7 are useful to rule out clear cell papillary renal cell tumor. CK7 is negative in TFE3/TFEB-rearranged renal cell carcinoma and positive in papillary renal cell carcinoma, being therefore useful in this setting. Parvalbumin and CK7/S100A1 respectively are of paramount importance when TFE3/TFEB-rearranged renal cell carcinoma resembles oncocytoma and chromophobe renal cell carcinoma. Moreover, in TFEB-rearranged renal cell carcinoma, cathepsin K and melanogenesis markers are constantly positive, whereas TFE3-rearranged renal cell carcinoma stains for cathepsin K in roughly half of the cases, HMB45 in 8% and Melan-A in 22%. In conclusion, since TFE3/TFEB-rearranged renal cell carcinoma may mimic several histotypes, an immunohistochemical panel to differentiate them from common renal cell tumors should include cathepsin K, CA9, CK7, and parvalbumin.
Collapse
|
24
|
Hirose K, Naniwa K, Usami Y, Kohara M, Uchihashi T, Tanaka S, Yamada S, Iwamoto Y, Murakami S, Oya K, Fukuda Y, Hori Y, Morii E, Toyosawa S. Primary Alveolar Soft Part Sarcoma of Cheek: Report of a Case and Review of the Literature. Head Neck Pathol 2021; 15:1426-1431. [PMID: 33840045 PMCID: PMC8633199 DOI: 10.1007/s12105-021-01324-6] [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: 02/21/2021] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Abstract
Alveolar soft part sarcoma (ASPS) is a rare soft tissue sarcoma characterized by an alveolar or organoid arrangement of polygonal tumour cells separated by fibrovascular septa. A specific fusion gene [ASPS critical region 1 (ASPSCR1)-TFE3] was detected in ASPS. Despite being a slow-growing tumour without pain and dysfunction, ASPS is characterized by early metastasis, which leads to poor prognosis. Herein, we report a rare case of primary ASPS of the cheek harbouring ASPSCR1 (exon 7)-TFE3 (exon 5) fusion gene in a 21 year-old woman. This tumour was a well-circumscribed, smooth, round mass that was clinically suspected as a benign tumour. However, histologically, it was observed that the polygonal tumour cells were arranged in solid and alveolar growth patterns. Post-operative examination of the whole body excluded the possibility of metastasis at other sites. Thus, careful immunohistochemical and genetic analyses, as well as whole-body examination, demonstrated that the tumour was a primary ASPS of the cheek.
Collapse
Affiliation(s)
- Katsutoshi Hirose
- Department of Oral Pathology, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Kohei Naniwa
- Department of Oral Pathology, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Yu Usami
- Department of Oral Pathology, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Masaharu Kohara
- Department of Pathology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Toshihiro Uchihashi
- Department of Oral and Maxillofacial Surgery 1, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
- Unit of Dentistry, Osaka University Hospital, 2-15 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Susumu Tanaka
- Department of Oral and Maxillofacial Surgery 1, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Saori Yamada
- Department of Oral and Maxillofacial Surgery 1, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Yuri Iwamoto
- Department of Oral and Maxillofacial Radiology, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Shumei Murakami
- Department of Oral and Maxillofacial Radiology, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Kaori Oya
- Clinical Laboratory, Osaka University Dental Hospital, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Yasuo Fukuda
- Clinical Laboratory, Osaka University Dental Hospital, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Yumiko Hori
- Department of Pathology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Satoru Toyosawa
- Department of Oral Pathology, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
| |
Collapse
|
25
|
Argani P. Translocation carcinomas of the kidney. Genes Chromosomes Cancer 2021; 61:219-227. [PMID: 34704642 DOI: 10.1002/gcc.23007] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/24/2021] [Indexed: 01/19/2023] Open
Abstract
The MiT subfamily of transcription factors includes TFE3, TFEB, TFEC, and MITF. Gene fusions involving two of these transcription factors have been well-characterized in renal cell carcinoma (RCC). The TFE3-rearranged RCC (also known as Xp11 translocation RCC) was first officially recognized in the 2004 World Health Organization (WHO) renal tumor classification. The TFEB-rearranged RCC, which typically harbor a t(6;11)(p21;q12) translocation which results in a MALAT1-TFEB gene fusion, were first officially recognized in the 2016 WHO renal tumor classification. These two subtypes of translocation RCC have many similarities. Both disproportionately involve young patients, although adult translocation RCC overall outnumber pediatric cases. Both often have unusual and distinctive morphologies; the TFE3-rearranged RCCs frequently have clear cells with papillary architecture and abundant psammoma bodies, while the TFEB-rearranged RCCs frequently have a biphasic appearance with both small and large epithelioid cells and nodules of basement membrane material. However, the morphology of these two neoplasms can overlap, with one mimicking the other or other more common renal neoplasms. Both of these RCC underexpress epithelial immunohistochemical markers, such as cytokeratin and epithelial membrane antigen, relative to most other RCC. Unlike other RCC, both frequently express the cysteine protease cathepsin k and often express melanocytic markers like HMB45 and Melan A. Finally, TFE3 and TFEB have overlapping functional activity as these two transcription factors frequently heterodimerize and bind to the same targets. Therefore, these two neoplasms are now grouped together under the heading of "MiT family translocation RCC." Approximately 50 renal cell carcinomas with gene fusions involving the anaplastic lymphoma kinase (ALK) gene have now been reported. While those with a Vinculin-ALK fusion have distinctive features (predilection to affect children with sickle cell trait and to show solid architecture with striking cytoplasmic vacuolization), other ALK-fusion RCCs have more varied clinical presentations and pathologic features. This review summarizes our current knowledge of these recently described RCC.
Collapse
Affiliation(s)
- Pedram Argani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Departments of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
26
|
Lobo J, Ohashi R, Helmchen BM, Rupp NJ, Rüschoff JH, Moch H. The Morphological Spectrum of Papillary Renal Cell Carcinoma and Prevalence of Provisional/Emerging Renal Tumor Entities with Papillary Growth. Biomedicines 2021; 9:1418. [PMID: 34680535 PMCID: PMC8533532 DOI: 10.3390/biomedicines9101418] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 12/11/2022] Open
Abstract
Renal cell carcinoma (RCC) represents a heterogeneous disease, encompassing an increasing number of tumor subtypes. Post-2016, the World Health Organization (WHO) classification recognized that the spectrum of papillary renal cell carcinoma is evolving and has long surpassed the dichotomic simplistic "type 1 versus type 2" classification. The differential diagnosis of pRCC includes several new provisional/emerging entities with papillary growth. Type 2 tumors have been cleared out of several confounding entities, now regarded as independent tumors with specific clinical and molecular backgrounds. In this work we describe the prevalence and characteristics of emerging papillary tumor entities in two renal tumor cohorts (one consisting of consecutive papillary tumors from a single institute, the other consisting of consultation cases from several centers). After a review of 154 consecutive pRCC cases, 58% remained type 1 pRCC, and 34% type 2 pRCC. Papillary renal neoplasm with reversed polarity (1.3%), biphasic hyalinizing psammomatous RCC (1.3%), and biphasic squamoid/alveolar RCC (4.5%) were rare. Among 281 consultation cases, 121 (43%) tumors had a dominant papillary growth (most frequently MiT family translocation RCCs, mucinous tubular and spindle cell carcinoma and clear cell papillary RCC). Our data confirm that the spectrum of RCCs with papillary growth represents a major diagnostical challenge, frequently requiring a second expert opinion. Papillary renal neoplasm with reversed polarity, biphasic hyalinizing psammomatous RCC, and biphasic squamoid/alveolar RCC are rarely sent out for a second opinion, but correct classification and knowledge of these variants will improve our understanding of the clinical behavior of renal tumors with papillary growth.
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, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS—School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Riuko Ohashi
- Histopathology Core Facility, Faculty of Medicine, Niigata University, 1-757 Asahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan;
- Division of Molecular and Diagnostic Pathology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan
| | - Birgit M. Helmchen
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland; (B.M.H.); (N.J.R.); (J.H.R.)
| | - Niels J. Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland; (B.M.H.); (N.J.R.); (J.H.R.)
| | - Jan H. Rüschoff
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland; (B.M.H.); (N.J.R.); (J.H.R.)
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland; (B.M.H.); (N.J.R.); (J.H.R.)
- Faculty of Medicine, University of Zurich, Rämistrasse 71, 8006 Zurich, Switzerland
| |
Collapse
|
27
|
Choi JH, Ro JY. Mesenchymal Tumors of the Mediastinum: An Update on Diagnostic Approach. Adv Anat Pathol 2021; 28:351-381. [PMID: 34050062 DOI: 10.1097/pap.0000000000000306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mesenchymal tumors of the mediastinum are a heterogenous group of rare tumors with divergent lineages. Mediastinal mesenchymal tumors are diagnostically challenging due to their diversity and morphologic overlap with nonmesenchymal lesions arising in the mediastinum. Accurate histologic diagnosis is critical for appropriate patient management and prognostication. Many mediastinal mesenchymal tumors affect distinct age groups or occur at specific mediastinal compartments. Neurogenic tumors, liposarcoma, solitary fibrous tumor, and synovial sarcoma are common mesenchymal tumors in the mediastinum. Herein, we provide an update on the diagnostic approach to mediastinal mesenchymal tumors and a review of the histologic features and differential diagnosis of common benign and malignant mesenchymal tumors of the mediastinum.
Collapse
Affiliation(s)
- Joon Hyuk Choi
- Department of Pathology, Yeungnam University College of Medicine, Daegu, South Korea
| | - Jae Y Ro
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Medical College of Cornell University, Houston, TX
| |
Collapse
|
28
|
Wang XM, Zhang Y, Mannan R, Skala SL, Rangaswamy R, Chinnaiyan A, Su F, Cao X, Zelenka-Wang S, McMurry L, Xiao H, Spratt DE, Sangoi A, Shao L, Betz BL, Brown N, Tickoo SK, McKenney JK, Argani P, Gupta S, Reuter VE, Chinnaiyan AM, Dhanasekaran SM, Mehra R. TRIM63 is a sensitive and specific biomarker for MiT family aberration-associated renal cell carcinoma. Mod Pathol 2021; 34:1596-1607. [PMID: 33854184 PMCID: PMC8298271 DOI: 10.1038/s41379-021-00803-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 12/20/2022]
Abstract
Microphthalmia-associated transcription factor (MiT) family aberration-associated renal cell carcinoma (MiTF-RCC) is a subtype of renal cell carcinoma harboring recurrent chromosomal rearrangements involving TFE3 or TFEB genes. MiTF-RCC is morphologically diverse, can histologically resemble common RCC subtypes like clear cell RCC and papillary RCC, and often poses a diagnostic challenge in genitourinary clinical and pathology practice. To characterize the MiTF-RCC at the molecular level and identify biomarker signatures associated with MiTF-RCC, we analyzed RNAseq data from MiTF-RCC, other RCC subtypes and benign kidney. Upon identifying TRIM63 as a cancer-specific biomarker in MiTF-RCC, we evaluated its expression independently by RNA in situ hybridization (RNA-ISH) in whole tissue sections from 177 RCC cases. We specifically included 31 cytogenetically confirmed MiTF-RCC cases and 70 RCC cases suspicious for MiTF-RCC in terms of clinical and morphological features, to evaluate and compare TRIM63 RNA-ISH results with the results from TFE3/TFEB fluorescence in situ hybridization (FISH), which is the current clinical standard. We confirmed that TRIM63 mRNA was highly expressed in all classes of MiTF-RCC compared to other renal tumor categories, where it was mostly absent to low. While the TRIM63 RNA-ISH and TFE3/TFEB FISH results were largely concordant, importantly, TRIM63 RNA-ISH was strongly positive in TFE3 FISH false-negative cases with RBM10-TFE3 inversion. In conclusion, TRIM63 can serve as a diagnostic marker to distinguish MiTF-RCC from other renal tumor subtypes with overlapping morphology. We suggest a combination of TFE3/TFEB FISH and TRIM63 RNA-ISH assays to improve the accuracy and efficiency of MiTF-RCC diagnosis. Accurate diagnosis of MiTF-RCC and other RCC subtypes would enable effective targeted therapy and avoid poor therapeutic response due to tumor misclassification.
Collapse
Affiliation(s)
- Xiao-Ming Wang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI,Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Yuping Zhang
- Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Rahul Mannan
- Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Stephanie L. Skala
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | | | | | - Fengyun Su
- Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Xuhong Cao
- Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Sylvia Zelenka-Wang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI,Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Lisa McMurry
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Hong Xiao
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Daniel E. Spratt
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI
| | - Ankur Sangoi
- Department of Pathology, El Camino Hospital, Mountain View, CA
| | - Lina Shao
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Bryan L. Betz
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Noah Brown
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Satish K. Tickoo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jesse K. McKenney
- Robert J Tomsich Pathology and Laboratory Medicine Institute, Anatomic Pathology, Cleveland Clinic, Cleveland, OH
| | - Pedram Argani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Victor E. Reuter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Arul M. Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI,Michigan Center for Translational Pathology, Ann Arbor, MI,Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI,Department of Urology, University of Michigan Medical School, Ann Arbor, MI,Howard Hughes Medical Institute, Ann Arbor, MI
| | - Saravana M. Dhanasekaran
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI,Michigan Center for Translational Pathology, Ann Arbor, MI
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA. .,Michigan Center for Translational Pathology, Ann Arbor, MI, USA. .,Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
29
|
Webster BR, Rompre-Brodeur A, Daneshvar M, Pahwa R, Srinivasan R. Kidney cancer: from genes to therapy. Curr Probl Cancer 2021; 45:100773. [PMID: 34261604 DOI: 10.1016/j.currproblcancer.2021.100773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 11/30/2022]
Abstract
Renal cell carcinoma incidence is rising worldwide with increasing subtype stratification by the World Health Organization. Each subtype has unique genetic alterations, cell biology changes and clinical findings. Such genetic alterations offer the potential for individualized therapeutic approaches that are rapidly progressing. This review highlights the most common subtypes of renal cell carcinoma, including both hereditary and sporadic forms, with a focus on genetic changes, clinical findings and ongoing clinical trials.
Collapse
Affiliation(s)
- Bradley R Webster
- Center for Cancer Research, Urologic Oncology Branch, National Cancer Institute/NIH, 10 Center Drive, CRC Room 2W-5940, Bethesda, MD 20892, USA
| | - Alexis Rompre-Brodeur
- Center for Cancer Research, Urologic Oncology Branch, National Cancer Institute/NIH, 10 Center Drive, CRC Room 2W-5940, Bethesda, MD 20892, USA
| | - Michael Daneshvar
- Center for Cancer Research, Urologic Oncology Branch, National Cancer Institute/NIH, 10 Center Drive, CRC Room 2W-5940, Bethesda, MD 20892, USA
| | - Roma Pahwa
- Center for Cancer Research, Urologic Oncology Branch, National Cancer Institute/NIH, 10 Center Drive, CRC Room 2W-5940, Bethesda, MD 20892, USA
| | - Ramaprasad Srinivasan
- Center for Cancer Research, Urologic Oncology Branch, National Cancer Institute/NIH, 10 Center Drive, CRC Room 2W-5940, Bethesda, MD 20892, USA.
| |
Collapse
|
30
|
Baniak N, Barletta JA, Hirsch MS. Key Renal Neoplasms With a Female Predominance. Adv Anat Pathol 2021; 28:228-250. [PMID: 34009777 DOI: 10.1097/pap.0000000000000301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Renal neoplasms largely favor male patients; however, there is a growing list of tumors that are more frequently diagnosed in females. These tumors include metanephric adenoma, mixed epithelial and stromal tumor, juxtaglomerular cell tumor, mucinous tubular and spindle cell carcinoma, Xp11.2 (TFE3) translocation-associated renal cell carcinoma, and tuberous sclerosis complex (somatic or germline) associated renal neoplasms. The latter category is a heterogenous group with entities still being delineated. Eosinophilic solid and cystic renal cell carcinoma is the best-described entity, whereas, eosinophilic vacuolated tumor is a proposed entity, and the remaining tumors are currently grouped together under the umbrella of tuberous sclerosis complex/mammalian target of rapamycin-related renal neoplasms. The entities described in this review are often diagnostic considerations when evaluating renal mass tissue on biopsy or resection. For example, Xp11.2 translocation renal cell carcinoma is in the differential when a tumor has clear cell cytology and papillary architecture and occurs in a young or middle-aged patient. In contrast, tuberous sclerosis complex-related neoplasms often enter the differential for tumors with eosinophilic cytology. This review provides an overview of the clinical, gross, microscopic, immunohistochemical, genetic, and molecular alterations in key renal neoplasms occurring more commonly in females; differential diagnoses are also discussed regardless of sex predilection.
Collapse
Affiliation(s)
- Nicholas Baniak
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Justine A Barletta
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
31
|
New developments in existing WHO entities and evolving molecular concepts: The Genitourinary Pathology Society (GUPS) update on renal neoplasia. Mod Pathol 2021; 34:1392-1424. [PMID: 33664427 DOI: 10.1038/s41379-021-00779-w] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/11/2021] [Accepted: 02/11/2021] [Indexed: 12/28/2022]
Abstract
The Genitourinary Pathology Society (GUPS) reviewed recent advances in renal neoplasia, particularly post-2016 World Health Organization (WHO) classification, to provide an update on existing entities, including diagnostic criteria, molecular correlates, and updated nomenclature. Key prognostic features for clear cell renal cell carcinoma (RCC) remain WHO/ISUP grade, AJCC/pTNM stage, coagulative necrosis, and rhabdoid and sarcomatoid differentiation. Accrual of subclonal genetic alterations in clear cell RCC including SETD2, PBRM1, BAP1, loss of chromosome 14q and 9p are associated with variable prognosis, patterns of metastasis, and vulnerability to therapies. Recent National Comprehensive Cancer Network (NCCN) guidelines increasingly adopt immunotherapeutic agents in advanced RCC, including RCC with rhabdoid and sarcomatoid changes. Papillary RCC subtyping is no longer recommended, as WHO/ISUP grade and tumor architecture better predict outcome. New papillary RCC variants/patterns include biphasic, solid, Warthin-like, and papillary renal neoplasm with reverse polarity. For tumors with 'borderline' features between oncocytoma and chromophobe RCC, a term "oncocytic renal neoplasm of low malignant potential, not further classified" is proposed. Clear cell papillary RCC may warrant reclassification as a tumor of low malignant potential. Tubulocystic RCC should only be diagnosed when morphologically pure. MiTF family translocation RCCs exhibit varied morphologic patterns and fusion partners. TFEB-amplified RCC occurs in older patients and is associated with more aggressive behavior. Acquired cystic disease (ACD) RCC-like cysts are likely precursors of ACD-RCC. The diagnosis of renal medullary carcinoma requires a negative SMARCB1 (INI-1) expression and sickle cell trait/disease. Mucinous tubular and spindle cell carcinoma (MTSCC) can be distinguished from papillary RCC with overlapping morphology by losses of chromosomes 1, 4, 6, 8, 9, 13, 14, 15, and 22. MTSCC with adverse histologic features shows frequent CDKN2A/2B (9p) deletions. BRAF mutations unify the metanephric family of tumors. The term "fumarate hydratase deficient RCC" ("FH-deficient RCC") is preferred over "hereditary leiomyomatosis and RCC syndrome-associated RCC". A low threshold for FH, 2SC, and SDHB immunohistochemistry is recommended in difficult to classify RCCs, particularly those with eosinophilic morphology, occurring in younger patients. Current evidence does not support existence of a unique tumor subtype occurring after chemotherapy/radiation in early childhood.
Collapse
|
32
|
Uncommon Localization of Extrarenal Xp11.2 Translocation-associated Renal Cell Carcinoma (RCC): Case Report. Appl Immunohistochem Mol Morphol 2021; 28:e33-e35. [PMID: 28877071 DOI: 10.1097/pai.0000000000000576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The World Health Organization has recognized Xp11.2 translocation-associated renal cell carcinoma (RCC) as a distinct neoplasm that arises within the kidney. Although many reports of extrarenal carcinoma may be found in the literature, to the best of our knowledge, Xp11 translocation-associated RCC with intact kidneys has not been documented. This report describes a multilobulated right retroperitoneal soft tissue mass (7.9×5.3×12.6 cm) of a 37-year-old man complaining of abdominal pain in the right side. The patient underwent a computed tomography-guided biopsy. Microscopic evaluation reveals a tumor with papillary and sheaths architectures with cells revealing clear to eosinophilic cytoplasm. Immunohistochemical evaluation on the biopsy reveals that the tumor is positive for PAX-8, CD10, and TFE3. It is negative for CK7, EMA, Vimentin, RCC, CK8/18, D20, CD3, PLAP, OCT4, CD30, MART-1, Inhibin, S-100, HMB-45, Desmin, SMA, and DOG-1. The diagnosis was malignant epithelioid neoplasm and the diagnosis of translocation RCC was suggested. Excision was recommended. The patient underwent right radical nephrectomy with removal of this large mass. Pathologic examination showed a large cystic and solid, nonhomogenous mass with some necrotic areas, originating from the perirenal fat between the adrenal gland and the kidney. Microscopic features showed a tumor with papillary, rhabdoid, and clear cell features. Immunohistochemical stains showed that the tumor cells positively expressed AMACR, PAX-8, CD10, RCC, and TFE3, but were negative for cytokeratins, vimentin, HMB-45, desmin, SMA, EMA, and MSA. Cytogenetic studies confirmed the diagnosis of Xp11.2 translocation-associated RCC with positive TFE3 gene rearrangement. To the best of our knowledge, this type of extrarenal tumor has never been reported.
Collapse
|
33
|
Cathepsin K: A Novel Diagnostic and Predictive Biomarker for Renal Tumors. Cancers (Basel) 2021; 13:cancers13102441. [PMID: 34069976 PMCID: PMC8157838 DOI: 10.3390/cancers13102441] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/07/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Our understanding of renal tumors has increased in the last years with the description of several novel entities. The expanding morphological spectrum complicates the pathologist’s diagnosis, often requiring immunohistochemical analysis. The role of cathepsin K immunoexpression is widened as a diagnostic tool in several renal tumors. This review describes the usefulness of cathepsin K in the differential diagnosis of renal neoplasms, highlighting the biological knowledge underpinning its expression. Moreover, cathepsin K seems to be a downstream marker of different genetic alterations, with a possible role as a predictive marker that may prospectively guide the development of therapeutic approaches as a molecular target. Abstract Cathepsin K is a papain-like cysteine protease with high matrix-degrading activity. Among several cathepsins, cathepsin K is the most potent mammalian collagenase, mainly expressed by osteoclasts. This review summarizes most of the recent findings of cathepsin K expression, highlighting its role in renal tumors for diagnostic purposes and as a potential molecular target. Indeed, cathepsin K is a recognized diagnostic tool for the identification of TFE3/TFEB-rearranged renal cell carcinoma, TFEB-amplified renal cell carcinoma, and pure epithelioid PEComa/epithelioid angiomyolipoma. More recently, its expression has been observed in a subgroup of eosinophilic renal neoplasms molecularly characterized by TSC/mTOR gene mutations. Interestingly, both TSC mutations or TFE3 rearrangement have been reported in pure epithelioid PEComa/epithelioid angiomyolipoma. Therefore, cathepsin K seems to be a downstream marker of TFE3/TFEB rearrangement, TFEB amplification, and mTOR pathway activation. Given the established role of mTOR inhibitors as a pharmacological option in renal cancers, cathepsin K could be of use as a predictive marker of therapy response and as a potential target. In the future, uropathologists may implement the use of cathepsin K to establish a diagnosis among renal tumors with clear cells, papillary architecture, and oncocytic features.
Collapse
|
34
|
Contemporary Characterization and Recategorization of Adult Unclassified Renal Cell Carcinoma. Am J Surg Pathol 2021; 45:450-462. [PMID: 33239504 DOI: 10.1097/pas.0000000000001629] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Our recent study of early-onset unclassified eosinophilic renal cell carcinoma (RCC) demonstrated that two third of cases could be reclassified by performing a limited number of immunohistochemistry stains. Following the same approach, we aimed to investigate what proportion of adult unclassified RCC could be reclassified. We identified 79 cases. The mean age at presentation was 58 years (range, 29 to 84 y). Tumors were grouped based on their predominant morphologic features as oncocytic (n=23); papillary (n=22); clear cell (n=22); mucinous tubular and spindle cell (MTSC; n=5); rhabdoid (n=4); or lacking a dominant pattern (n=3). By reviewing the morphologic features and performing ancillary studies, we were able to reclassify 10 cases (13%). Four cases were positive for CK20 and showed morphologic features consistent with eosinophilic solid and cystic RCC. Four cases were reclassified as MTSC based on VSTM2A expression by RNA in situ hybridization. One case was negative for SDHB and reclassified as succinate dehydrogenase-deficient RCC. None of the cases showed loss of expression of fumarate hydratase. One case was diffusely positive for CK7 and negative for CD117 and reclassified as a low-grade oncocytic tumor. Four cases were positive for both cathepsin-K and TFE3 by immunohistochemistry, although fluorescence in situ hybridization failed to identify rearrangement in either TFE3 or TFEB genes. Of the tumors that remained unclassified, those with oncocytic features were less likely to be a high grade (odds ratio [OR]=0.22, P=0.013) or advanced stage (OR=0.19, P=0.039) and were more common in women (OR=3.4, P=0.05) compared with those without oncocytic features. Tumors with rhabdoid morphology were associated with advanced stage (relative risk=3.6, P=0.009), while tumors with clear cell or papillary features had a wide range of grades and stages at presentation. In summary, the most frequent reclassified entity is eosinophilic solid and cystic RCC. Investigation of expression of succinate dehydrogenase or fumarate hydratase in individuals older than 35 years with unclassifiable tumors is low yield in the absence of specific morphologic features. A subset of MTSC without well-developed morphologic features can be reclassified by using RNA-ISH for VSTM2A. Recognition of more-recently described RCC subtypes allows for their distinction from the unclassified subtype and improves the prognostic information provided.
Collapse
|
35
|
Alveolar Soft Part Sarcoma of Uterine Corpus in a Young Female: A Case Report With Review of Literature. Int J Gynecol Pathol 2021; 40:272-277. [PMID: 33323848 DOI: 10.1097/pgp.0000000000000700] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Alveolar soft part sarcoma (ASPS) is a rare soft tissue sarcoma, often occurs in adolescents and young adults with a particular predilection for the deep soft tissue of extremities. Occurrence of ASPS in the female genital tract is very uncommon and poses a significant diagnostic challenge. A case of ASPS of the uterine corpus is described in a young unmarried female, who presented to the out-patient clinic of our Institute with complaints of abnormal uterine bleeding for the past 9 mo. She was being treated with oral contraceptive pills, progesterone and tranexamic acid. Following radiological imaging and hysteroscopy, a therapeutic curettage of the lesion was done. The histopathologic features raised a differential diagnosis of a myriad of morphologic mimickers. The diagnosis was clinched by exclusion of mimickers by relevant immunohistochemical markers and strong nuclear expression of TFE3 on immunohistochemistry. The patient is on regular follow-up with oral contraceptives and antifibrinolytic medication. Despite being infrequent at this location, ASPS should be kept in differential diagnosis in young females presenting with abnormal uterine bleeding.
Collapse
|
36
|
Iakymenko OA, Delma KS, Jorda M, Kryvenko ON. Cathepsin K (Clone EPR19992) Demonstrates Uniformly Positive Immunoreactivity in Renal Oncocytoma, Chromophobe Renal Cell Carcinoma, and Distal Tubules. Int J Surg Pathol 2021; 29:600-605. [PMID: 33764165 DOI: 10.1177/1066896921991588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction. Cathepsin K is overexpressed in several tumors associated with microphthalmia transcription factor (MiTF) family or mechanistic target of rapamycin (mTOR) upregulation. Among renal neoplasms, MiTF translocation renal cell carcinoma (RCC), perivascular epithelioid cell neoplasms (PEComa), and eosinophilic solid and cystic RCC have demonstrated Cathepsin K immunoreactivity. In this study, we demonstrate a uniform Cathepsin K expression in oncocytoma, chromophobe RCC (CHRCC), and distal tubules. Design. We stained 13 oncocytomas, 13 CHRCC, 14 clear cell RCC (CCRCC), 9 papillary RCC (PRCC), 9 PEComas, and 5 MiTF RCC. Additionally, we assessed immunoreactivity for Cathepsin K in non-neoplastic renal parenchyma. Immunolabeling was performed on regularly charged slides from formalin-fixed paraffin-embedded tissue with monoclonal anti-rabbit antibodies to human Cathepsin K (clone EPR19992, Abcam). Results. All oncocytomas demonstrated diffuse strong cytoplasmic immunolabeling. CHRCC demonstrated uniform less intense immunolabeling in all cases with membranous accentuation. The assessment of the non-neoplastic renal parenchyma in all cases showed strong cytoplasmic immunoreaction in distal tubules and proximal tubules stained faintly. Mesangial cells were not immunoreactive. All MiTF RCC and PEComas were immunoreactive for Cathepsin K, whereas CCRCC and PRCC were negative in all cases. Conclusions. In this study, we expand the spectrum of renal neoplasms reactive with a particular clone of Cathepsin K (EPR19992). Distal tubules are strongly immunoreactive for Cathepsin K. Our conclusions need to be taken into consideration when differential diagnosis includes MiTF RCC or PEComa and this Cathepsin K clone is included in the immunohistochemical panel. This newer antibody clone was not tested in prior publications, potentially explaining the difference in conclusions.
Collapse
Affiliation(s)
| | - Katiana S Delma
- 12235University of Miami Miller School of Medicine, Miami, FL, USA
| | - Merce Jorda
- 12235University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, 12235University of Miami Miller School of Medicine, Miami, FL, USA
| | - Oleksandr N Kryvenko
- 12235University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, 12235University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
37
|
[Molecular pathology of urogenital tumors : Recommendations from the 2019 International Society of Urological Pathology (ISUP) Consensus Conference]. DER PATHOLOGE 2021; 42:310-318. [PMID: 33398501 PMCID: PMC8084837 DOI: 10.1007/s00292-020-00888-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 12/07/2020] [Indexed: 11/21/2022]
Abstract
Das zunehmende Verständnis molekularer Grundlagen von Tumoren sowie der Fortschritt in der Diversifizierung der onkologischen Therapien versprechen individualisierte Therapieoptionen, welche bislang jedoch nur ansatzweise in die Therapieplanung von urologischen Tumoren eingegangen sind. Daher hat die Internationale Gesellschaft für Urologische Pathologie (ISUP) im März 2019 eine Konsenskonferenz zur Erarbeitung evidenzbasierter Handlungsempfehlungen zur molekularpathologischen Diagnostik beim Urothelkarzinom, Nierenzellkarzinom, Prostatakarzinom, Peniskarzinom und testikulären Keimzelltumoren durchgeführt. Die auf dieser Konsenskonferenz erarbeiteten Empfehlungen sind kürzlich in 5 separaten Manuskripten veröffentlich worden und werden in der vorliegenden Arbeit zusammengefasst. Im Rahmen der Konferenzvorbereitung wurde eine umfassende Umfrage zur derzeitigen Praxis molekularer Testungen bei urogenitalen Tumoren unter den Mitgliedern der ISUP durchgeführt. Auf der Konferenz wurden die Ergebnisse und die entsprechenden Hintergrundinformationen durch 5 Arbeitsgruppen präsentiert und Handlungsempfehlungen für die Diagnostik erarbeitet. Eine Übereinstimmung von 66 % der Konferenzteilnehmer wurde als Konsens definiert.
Collapse
|
38
|
Kuthi L, Somorácz Á, Micsik T, Jenei A, Hajdu A, Sejben I, Imre D, Pósfai B, Kóczián K, Semjén D, Bajory Z, Kulka J, Iványi B. Clinicopathological Findings on 28 Cases with XP11.2 Renal Cell Carcinoma. Pathol Oncol Res 2020; 26:2123-2133. [PMID: 31955345 PMCID: PMC7471254 DOI: 10.1007/s12253-019-00792-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
Abstract
Xp11.2 translocation carcinoma is a distinct subtype of renal cell carcinoma characterized by translocations involving the TFE3 gene. Our study included the morphological, immunohistochemical and clinicopathological examination of 28 Xp11.2 RCCs. The immunophenotype has been assessed by using CA9, CK7, CD10, AMACR, MelanA, HMB45, Cathepsin K and TFE3 immunostainings. The diagnosis was confirmed by TFE3 break-apart FISH in 25 cases. The ages of 13 male and 15 female patients, without underlying renal disease or having undergone chemotherapy ranged from 8 to 72. The mean size of the tumors was 78.5 mm. Forty-three percent of patients were diagnosed in the pT3/pT4 stage with distant metastasis in 6 cases. Histological appearance was branching-papillary composed of clear cells with voluminous cytoplasm in 13 and variable in 15 cases, including one tumor with anaplastic carcinoma and another with rhabdoid morphology. Three tumors were labeled with CA9, while CK7 was negative in all cases. Diffuse CD10 reaction was observed in 17 tumors and diffuse AMACR positivity was described in 14 tumors. The expression of melanocytic markers and Cathepsin K were seen only in 7 and 6 cases, respectively. TFE3 immunohistochemistry displayed a positive reaction in 26/28 samples. TFE3 rearrangement was detected in all the analyzed cases (25/25), including one with the loss of the entire labeled break-point region. The follow-up time ranged from 2 to 300 months, with 7 cancer-related deaths. In summary, Xp11.2 carcinoma is an uncommon form of renal cell carcinoma with a variable histomorphology and rather aggressive clinical course.
Collapse
Affiliation(s)
- Levente Kuthi
- Department of Pathology, University of Szeged, 1 Állomás Street, Szeged, H-6725, Hungary.
| | - Áron Somorácz
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Tamás Micsik
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Alex Jenei
- Department of Pathology, University of Szeged, 1 Állomás Street, Szeged, H-6725, Hungary
| | - Adrienn Hajdu
- Department of Pathology, University of Szeged, 1 Állomás Street, Szeged, H-6725, Hungary
| | - István Sejben
- Department of Pathology, Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary
| | - Dániel Imre
- Department of Pathology, Hetényi Géza County Hospital, Szolnok, Hungary
| | - Boglárka Pósfai
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - Katalin Kóczián
- Surgical and Molecular Tumor Pathology Centre, National Institute of Oncology, Budapest, Hungary
| | - Dávid Semjén
- Department of Pathology, Clinical Center and Medical School, University of Pécs, Pécs, Hungary
| | - Zoltán Bajory
- Department of Urology, University of Szeged, Szeged, Hungary
| | - Janina Kulka
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Béla Iványi
- Department of Pathology, University of Szeged, 1 Állomás Street, Szeged, H-6725, Hungary
| |
Collapse
|
39
|
Montella M, Franco R, Aquino G, Ronchi A, Zito Marino F, Di Napoli M, Pignata S, Cozzolino I. Cytological diagnosis of Xp11 translocation renal cell carcinoma: An unusual suspect in bone metastases from unknown primary malignancies. Diagn Cytopathol 2020; 49:11-17. [PMID: 32809251 DOI: 10.1002/dc.24587] [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: 04/30/2020] [Revised: 07/14/2020] [Accepted: 08/03/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Renal cell carcinoma (RCC) constitutes 3% of all cancers, with a higher incidence in patients with age between 60 and 70 years. RCC frequently present as a metastatic tumor at diagnosis, and bones represent one of the most frequent sites. Many cases, mainly in young patients, includes the Xp11 translocation RCC. The cytological diagnosis of Xp11 translocation RCC in adult population it is rarely performed, likely for the morphological overlap with other adult renal cell carcinoma subtypes. METHODS We retrospectively analyze a series of 92 adult patients with metastatic bone tumors, diagnosed on fine-needle aspiration cytology (FNAC) samples, focusing mainly on the cytological, immunophenotypic and molecular features of Xp11 translocation RCC. RESULTS In our series 6 of 92 (6.5%) cases were metastatic RCC (mRCC), among them 2 cases were metastasis from Xp11translocation RCC. Those cases showed a bloody background, with several groups of atypical cells arranged in syncytial groups or in papillary groups composed by atypical cells with abundant cytoplasm, with scattered clear cells. TFE3 was positive on immunocytochemical analysis and specific translocation t(Xp11.23) was detected by FISH analysis. CONCLUSIONS In adult patients with mRCC, it is necessary to consider also Xp11 translocation RCC among the diagnostic hypotheses. FNAC represents a valid tool to investigate bone lesions but cytological features of Xp11 translocation RCC are still poorly described and must necessarily be better defined.
Collapse
Affiliation(s)
- Marco Montella
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Gabriella Aquino
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale," IRCCS, Naples, Nepal
| | - Andrea Ronchi
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Federica Zito Marino
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Marilena Di Napoli
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Sandro Pignata
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Immacolata Cozzolino
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università della Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
40
|
Xp11.2 translocation renal cell carcinoma with TFE3 gene fusion in the elderly: case report and literature review. Int Cancer Conf J 2020; 9:182-186. [PMID: 32903928 DOI: 10.1007/s13691-020-00430-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/26/2020] [Indexed: 10/23/2022] Open
Abstract
A 68-year-old man was followed up with chronic kidney disease. Follow-up CT incidentally detected a tumor at the left kidney and multiple small nodular shadows in the lungs bilaterally. The patient underwent needle biopsy and was diagnosed with Xp11.2 translocation renal cell carcinoma (RCC) pathologically. Hence, laparoscopic nephrectomy was performed. Fluorescence in situ hybridization analysis revealed a break-apart of the transcription factor E3 (TFE3) genes in the left tumor. After 2 months postoperatively, nivolumab and ipilimumab were administered thrice intravenously, considering the intermediate risk by the IMDC risk classification. However, pleural effusion occurred but was removed adequately. Lung metastasis decreased, but new metastasis occurred at the left iliopsoas muscle. Target therapy was performed with axitinib. Unfortunately, he died 6 months later postoperatively. These tumors commonly occur in children than in adults, and very rare in elderly patients. Xp11.2 translocation RCC in the elderly has a poorer prognosis than that in children. To date, no effective treatment for Xp11.2 translocation RCC has been established.
Collapse
|
41
|
Williamson SR, Gill AJ, Argani P, Chen YB, Egevad L, Kristiansen G, Grignon DJ, Hes O. Report From the International Society of Urological Pathology (ISUP) Consultation Conference on Molecular Pathology of Urogenital Cancers: III: Molecular Pathology of Kidney Cancer. Am J Surg Pathol 2020; 44:e47-e65. [PMID: 32251007 PMCID: PMC7289677 DOI: 10.1097/pas.0000000000001476] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Renal cell carcinoma (RCC) subtypes are increasingly being discerned via their molecular underpinnings. Frequently this can be correlated to histologic and immunohistochemical surrogates, such that only simple targeted molecular assays, or none at all, are needed for diagnostic confirmation. In clear cell RCC, VHL mutation and 3p loss are well known; however, other genes with emerging important roles include SETD2, BAP1, and PBRM1, among others. Papillary RCC type 2 is now known to include likely several different molecular entities, such as fumarate hydratase (FH) deficient RCC. In MIT family translocation RCC, an increasing number of gene fusions are now described. Some TFE3 fusion partners, such as NONO, GRIPAP1, RBMX, and RBM10 may show a deceptive fluorescence in situ hybridization result due to the proximity of the genes on the same chromosome. FH and succinate dehydrogenase deficient RCC have implications for patient counseling due to heritable syndromes and the aggressiveness of FH-deficient RCC. Immunohistochemistry is increasingly available and helpful for recognizing both. Emerging tumor types with strong evidence for distinct diagnostic entities include eosinophilic solid and cystic RCC and TFEB/VEGFA/6p21 amplified RCC. Other emerging entities that are less clearly understood include TCEB1 mutated RCC, RCC with ALK rearrangement, renal neoplasms with mutations of TSC2 or MTOR, and RCC with fibromuscular stroma. In metastatic RCC, the role of molecular studies is not entirely defined at present, although there may be an increasing role for genomic analysis related to specific therapy pathways, such as for tyrosine kinase or MTOR inhibitors.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Renal Cell/diagnosis
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Kidney Neoplasms/diagnosis
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Mutation
- Neoplasm Metastasis
- Neoplastic Syndromes, Hereditary/diagnosis
- Neoplastic Syndromes, Hereditary/genetics
- Neoplastic Syndromes, Hereditary/metabolism
- Neoplastic Syndromes, Hereditary/pathology
- Pathology, Clinical
- Pathology, Molecular
- Prognosis
- Societies, Medical
- Urology
Collapse
Affiliation(s)
- Sean R Williamson
- Department of Pathology and Laboratory Medicine and Henry Ford Cancer Institute, Henry Ford Health System
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI
| | - Anthony J Gill
- NSW Health Pathology, Department of Anatomical Pathology
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Pedram Argani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lars Egevad
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - David J Grignon
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN
| | - Ondrej Hes
- Department of Pathology, Charles University, Medical Faculty and Charles University Hospital Plzen, Pilsen, Czechia
| |
Collapse
|
42
|
Comprehensive analysis of 34 MiT family translocation renal cell carcinomas and review of the literature: investigating prognostic markers and therapy targets. Pathology 2020; 52:297-309. [PMID: 32107074 DOI: 10.1016/j.pathol.2019.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/21/2019] [Accepted: 11/28/2019] [Indexed: 12/14/2022]
Abstract
Recently cabozantinib, a tyrosine kinase inhibitor with activity against VEGF, MET, AXL, and downregulating cathepsin K in vitro, has been proposed for the treatment of advanced clear and non-clear renal cell carcinomas. Since it is well known that cathepsin K is expressed in the majority of MiT family translocation renal cell carcinomas, we investigated cathepsin K, MET, AXL, and VEGF in a large series of those tumours, looking for possible predictive markers. We collected the clinicopathological features of 34 genetically confirmed MiT family translocation renal cell carcinomas [26 Xp11 and 8 t(6;11) renal cell carcinomas] and studied them using an immunohistochemical panel including PAX8, cathepsin K, HMB45, Melan-A, CD68 (PG-M1), CK7, CA9, MET, AXL and by FISH for VEGFA and MET. Cathepsin K was expressed in 14 of 26, HMB45 in 8 of 25, and Melan-A in 4 of 23 Xp11 renal cell carcinomas, whereas labelling for CK7 and CA9 was minimal. In t(6;11) renal cell carcinoma, cathepsin K and melanogenesis markers were constantly positive, whereas CK7 and CA9 were negative. None of the 34 carcinomas showed CD68 (PG-M1) and AXL expression. One aggressive Xp11 renal cell carcinoma showed increased VEGFA gene copy number (4-5 copies) with concurrent gains of TFE3 and TFEB. None of the 34 carcinomas showed MET gene amplification, whereas staining for MET was found in 7 of 8 t(6;11) and in 16 of 24 Xp11 renal cell carcinomas, and in the latter cases, when the expression was >50%, correlated with aggressiveness (p=0.0049). In Xp11 renal cell carcinomas, the aggressiveness was also correlated with larger tumour size (p=0.0008) and the presence of necrosis (p=0.027) but not nucleolar grading (p=1). Interestingly, in patients with tumours exhibiting two of three parameters (necrosis, larger tumour size and MET immunolabelling >50%) an aggressive clinical behaviour was observed in 88% of cases. In conclusion, cathepsin K, CD68 (PG-M1), CK7, CA9, and PAX8 is a useful panel for the diagnosis. Larger tumour size, the presence of necrosis and MET immunohistochemical expression correlate with aggressive behaviour in Xp11 renal cell carcinomas, especially in combination. VEGF, MET, cathepsin K but not AXL may be potential predictive markers for targeted therapy in MiT family translocation renal cell carcinomas.
Collapse
|
43
|
Ooms AH, Vujanić GM, D’Hooghe E, Collini P, L’Herminé-Coulomb A, Vokuhl C, Graf N, van den Heuvel-Eibrink MM, de Krijger RR. Renal Tumors of Childhood-A Histopathologic Pattern-Based Diagnostic Approach. Cancers (Basel) 2020; 12:cancers12030729. [PMID: 32204536 PMCID: PMC7140051 DOI: 10.3390/cancers12030729] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/04/2020] [Accepted: 03/07/2020] [Indexed: 12/15/2022] Open
Abstract
Renal tumors comprise approximately 7% of all malignant pediatric tumors. This is a highly heterogeneous group of tumors, each with its own therapeutic management, outcome, and association with germline predispositions. Histopathology is the key in establishing the correct diagnosis, and therefore pathologists with expertise in pediatric oncology are needed for dealing with these rare tumors. While each tumor shows different histologic features, they do have considerable overlap in cell type and histologic pattern, making the diagnosis difficult to establish, if based on routine histology alone. To this end, ancillary techniques, such as immunohistochemistry and molecular analysis, can be of great importance for the correct diagnosis, resulting in appropriate treatment. To use ancillary techniques cost-effectively, we propose a pattern-based approach and provide recommendations to aid in deciding which panel of antibodies, supplemented by molecular characterization of a subset of genes, are required.
Collapse
Affiliation(s)
- Ariadne H.A.G. Ooms
- Princess Máxima Center for pediatric oncology, 3584 CS Utrecht, The Netherlands (M.M.v.d.H.-E.)
- Pathan B.V., 3045 PM Rotterdam, The Netherlands
| | | | - Ellen D’Hooghe
- Department of Pathology, Oslo University Hospital, Rikshospitalet, 0372 Oslo, Norway;
| | - Paola Collini
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Aurore L’Herminé-Coulomb
- Sorbonne Université, Department of Pathology, Hôpital Armand Trousseau, Hopitaux Universitaires Est Parisien, 75012 Paris, France;
| | - Christian Vokuhl
- Section of Pediatric Pathology, Department of Pathology, University Hospital Bonn, 53127 Bonn, Germany;
| | - Norbert Graf
- Department of Pediatric Oncology & Hematology, Saarland University, D-66421 Homburg, Germany;
| | | | - Ronald R. de Krijger
- Princess Máxima Center for pediatric oncology, 3584 CS Utrecht, The Netherlands (M.M.v.d.H.-E.)
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Correspondence: ; Tel.: +31-088-9727272
| |
Collapse
|
44
|
Gomez M, Whiting K, Naous R. Alveolar soft part sarcoma presenting as a uterine polyp: A case report. SAGE Open Med Case Rep 2020; 8:2050313X20910598. [PMID: 32180983 PMCID: PMC7057413 DOI: 10.1177/2050313x20910598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 01/30/2020] [Indexed: 11/15/2022] Open
Abstract
Alveolar soft part sarcoma is a rare malignant soft tissue tumor, mainly localized in the extremities and occurring principally in adolescents and young adults. Alveolar soft part sarcoma are uncommon in the female genital tract. We here report a case of alveolar soft part sarcoma in a 20-year-old nullipara, presenting with vaginal bleeding and profound anemia requiring blood transfusions. Ultrasonographic examination revealed a polyp in the lower uterine segment. Surgical resection of the polyp was performed, and pathological evaluation showed typical histological, immunohistochemical, and molecular features consistent with alveolar soft part sarcoma. Patient underwent for total hysterectomy. Currently, she follows up with her surgeon and has no new complains. Given the infrequency of alveolar soft part sarcoma, this case report raises the awareness of alveolar soft part sarcoma as one of the entities to consider when confronted with a uterine polyp in a young patient.
Collapse
Affiliation(s)
- Mariangela Gomez
- Department of Pathology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Kerry Whiting
- Department of Pathology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Rana Naous
- Department of Pathology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
45
|
Herrscher H, Boilève A, Lindner V, Barthélémy P, Hutt É, Pierard L, Kurtz JE, Rioux-Leclercq N, Lang H, Malouf GG. [MiT family translocation renal cell carcinomas: Natural history, molecular features and multidisciplinary management]. Bull Cancer 2020; 107:272-280. [PMID: 32044098 DOI: 10.1016/j.bulcan.2019.11.010] [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: 06/24/2019] [Revised: 11/10/2019] [Accepted: 11/16/2019] [Indexed: 10/25/2022]
Abstract
MiT family translocation renal cell carcinomas (tRCC) represent a rare subtype of renal cell carcinomas. These tumors have been introduced for the first time in the World Health Classification (WHO) classification of kidney cancers in 2004. tRCC are characterized by reccurent translocations involving members of the MiT family transcription factors, mainly TFE3 and TFEB. The estimated incidence of these tumors is ∼1-5 % among all renal cell carcinomas, with female prodominance. tRCC were initially described in children, and the spectrum has been expanded over time to encompass adolescents and adults. TFE3- and TFEB-rearranged RCC harbor characteristic clinicopathological and immunohistochemical features and fluorescent hybridization in situ is considered the gold standard for their diagnosis, although it has some limitations especially when the partners are located in the vicinity of TFE3. Nephron-sparing surgery is an efficient treatment of localized cases when achievable. In metastatic setting, targeted agents and immunotherapy showed modest efficacy, with response rates and median overall survival inferior to those observed in clear-cell renal cell carcinomas. Management of tRCC necessite a multidisciplinary team and accrual in clinical trials have to be encouraged when possible. Novel biological insights are urgently awaited to better understand the mechanisms associated with kidney oncogenesis in this setting, and ultimately help to identify therapeutic targets.
Collapse
Affiliation(s)
- Hugo Herrscher
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Alice Boilève
- Institut Gustave-Roussy, département de médecine, 67200 Villejuif, France
| | - Véronique Lindner
- Hôpitaux universitaires de Strasbourg, département de pathologie, 67200 Strasbourg, France
| | - Philippe Barthélémy
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Émilie Hutt
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Laure Pierard
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Jean-Emmanuel Kurtz
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Nathalie Rioux-Leclercq
- Université de Rennes, service d'anatomie et cytologie pathologiques, Centre Hospitalier Universitaire de Rennes, IRSET, 67200 Rennes, France
| | - Hervé Lang
- Hôpitaux universitaires de Strasbourg, service d'urologie, 35033 Strasbourg, France
| | - Gabriel G Malouf
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France.
| |
Collapse
|
46
|
Michalova K, Tretiakova M, Pivovarcikova K, Alaghehbandan R, Perez Montiel D, Ulamec M, Osunkoya A, Trpkov K, Yuan G, Grossmann P, Sperga M, Ferak I, Rogala J, Mareckova J, Pitra T, Kolar J, Michal M, Hes O. Expanding the morphologic spectrum of chromophobe renal cell carcinoma: A study of 8 cases with papillary architecture. Ann Diagn Pathol 2019; 44:151448. [PMID: 31918172 DOI: 10.1016/j.anndiagpath.2019.151448] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/08/2019] [Indexed: 02/06/2023]
Abstract
Although typically arranged in solid alveolar fashion, chromophobe renal cell carcinoma (RCC) may also show several other architectural growth patterns. We include in this series 8 chromophobe RCC cases with prominent papillary growth, a pattern very rarely reported or only mentioned as a feature of chromophobe RCC, which is lacking wider recognition The differential diagnosis of such cases significantly varies from the typical chromophobe RCC with its usual morphology, particularly its distinction from papillary RCC and other relevant and clinically important entities. Of 972 chromophobe RCCs in our files, we identified 8 chromophobe RCCs with papillary growth. We performed immunohistochemistry and array Comparative Genomic Hybridisation (aCGH) to investigate for possible chromosomal aberrations. Patients were 3 males and 5 females with age ranging from 30 to 84 years (mean 57.5, median 60 years). Tumor size was variable and ranged from 2 to 14 cm (mean 7.5, median 6.6 cm). Follow-up was available for 7 of 8 patients, ranging from 1 to 61 months (mean 20.1, median 12 months). Six patients were alive with no signs of aggressive behavior, and one died of the disease. Histologically, all cases were composed of dual cell population consisting of variable proportions of leaf-like cells with pale cytoplasm and eosinophilic cells. The extent of papillary component ranged from 15 to 100% of the tumor volume (mean 51%, median 50%). Sarcomatoid differentiation was identified only in the case with fatal outcome. Immunohistochemically, all tumors were positive for CK7, CD117 and Hale's Colloidal Iron. PAX8 was positive in 5 of 8 cases, TFE3 was focally positive 3 of 8 tumors, and Cathepsin K was focally positive in 2 of 8 tumors. All cases were negative for vimentin, AMACR and HMB45. Fumarate hydratase staining was retained in all tested cases. The proliferative activity was low (up to 1% in 7, up to 5% in one case). Three cases were successfully analyzed by aCGH and all showed a variable copy number variation profile with multiple chromosomal gains and losses. CONCLUSIONS: Chromophobe RCC demonstrating papillary architecture is an exceptionally rare carcinoma. The diagnosis can be challenging, although the cytologic features are consistent with the classic chromophobe RCC. Given the prognostic and therapeutic implications of accurately diagnosis other RCCs with papillary architecture (i.e., Xp11.2 translocation RCC, FH-deficient RCC), it is crucial to differentiate these cases from chromophobe RCC with papillary architecture. Based on this limited series, the presence of papillary architecture does not appear to have negative prognostic impact. However, its wider recognition may allow in depth studies on additional examples of this rare morphologic variant.
Collapse
Affiliation(s)
- Kvetoslava Michalova
- Department of Pathology, Charles University, Medical Faculty and Charles University Hospital Plzen, Czech Republic
| | - Maria Tretiakova
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Kristyna Pivovarcikova
- Department of Pathology, Charles University, Medical Faculty and Charles University Hospital Plzen, Czech Republic
| | - Reza Alaghehbandan
- Department of Pathology, Faculty of Medicine, University of British Columbia, Royal Columbian Hospital, Vancouver, BC, Canada
| | - Delia Perez Montiel
- Department of Pathology, Institute Nacional de Cancerologia, Mexico City, Mexico
| | - Monika Ulamec
- Ljudevit Jurak Pathology Department, University Clinical Hospital "Sestre milosrdnice", Pathology Department, School of Dental Medicine, University of Zagreb, Croatia
| | | | - Kiril Trpkov
- Department of Pathology and Laboratory Medicine, Calgary Laboratory Services and University of Calgary, Calgary, AB, Canada
| | - Gao Yuan
- Department of Pathology and Laboratory Medicine, Calgary Laboratory Services and University of Calgary, Calgary, AB, Canada
| | - Petr Grossmann
- Department of Pathology, Charles University, Medical Faculty and Charles University Hospital Plzen, Czech Republic
| | - Maris Sperga
- Department of Pathology, University of Split, Croatia
| | - Ivan Ferak
- Department of Pathology, AGEL, Novy Jicin, Czech Republic
| | - Joanna Rogala
- Department of Pathology, Charles University, Medical Faculty and Charles University Hospital Plzen, Czech Republic
| | - Jana Mareckova
- Department of Pathology, Charles University, Medical Faculty and Charles University Hospital Plzen, Czech Republic
| | - Tomas Pitra
- Department of Urology, Charles University, Medical Faculty and Charles University Hospital Plzen, Czech Republic
| | - Jiri Kolar
- Department of Urology, Charles University, Medical Faculty and Charles University Hospital Plzen, Czech Republic
| | - Michal Michal
- Department of Pathology, Charles University, Medical Faculty and Charles University Hospital Plzen, Czech Republic
| | - Ondrej Hes
- Department of Pathology, Charles University, Medical Faculty and Charles University Hospital Plzen, Czech Republic.
| |
Collapse
|
47
|
Ye H, Qin S, Li N, Lin M, Xu Y, Li X. A Rare Partner of TFE3 in the Xp11 Translocation Renal Cell Carcinoma: Clinicopathological Analyses and Detection of MED15-TFE3 Fusion. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5974089. [PMID: 31828108 PMCID: PMC6881588 DOI: 10.1155/2019/5974089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/29/2019] [Accepted: 10/16/2019] [Indexed: 01/09/2023]
Abstract
Xp11 translocation renal cell carcinoma (RCC), a member of the microphthalmia-associated transcription factor (MiTF) family, is a rare renal tumor characterized by different translocations involving the TFE3 gene. Here, we reported a case of Xp11 translocation RCC with a rare MED15-TFE3 gene fusion by RNA sequencing. Morphologically, the tumor cells were arranged in a solid and small nest pattern. The cytoplasm was voluminous, flocculent eosinophilic, and vacuolated. The nuclei were round or polygon with fine granular chromatin, and the nucleoli were unconspicuous. Psammoma bodies were observed in mesenchyma. Immunohistochemically, the tumor cells were diffuse moderately or strongly positive for CD10, P504S, vimentin, PAX8, RCC, AE1/AE3, and SDHB and focally positive for CK7 and CA IX while negative for cathepsin K, HMB45, Melan-A, Ksp-cadherin, and CD117. The Ki67 proliferation index was approximately 3%. However, TFE3 labeling showed an uncertainly weak nuclear staining and been considered negative. Fluorescence in situ hybridization (FISH) demonstrated a positive result that splits signals with a distance of > 2 signal diameters. Subsequently, RNA sequencing confirmed a fusion of MED15 gene exon 11 on chromosome 22 with TFE3 gene exon 6 in the tumor. The patient was alive with no evidence of recurrence. Our report contributes to the understanding on MED15-TFE3 RCC.
Collapse
MESH Headings
- Adult
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics
- Biomarkers, Tumor/genetics
- Carcinoma, Renal Cell/diagnostic imaging
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Chromosomes, Human, X/genetics
- Female
- Humans
- In Situ Hybridization, Fluorescence
- Male
- Mediator Complex/genetics
- Middle Aged
- Neoplasm Proteins/genetics
- Neoplasm Recurrence, Local/diagnostic imaging
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/pathology
- Oncogene Proteins, Fusion/genetics
- Sequence Analysis, RNA
- Tomography, X-Ray Computed
- Translocation, Genetic/genetics
Collapse
Affiliation(s)
- Hong Ye
- Department of Pathology, Tai'an Central Hospital, Tai'an 271000, China
| | - Shuming Qin
- Department of Pathology, Tai'an Central Hospital, Tai'an 271000, China
| | - Ning Li
- Department of Pathology, Tai'an Central Hospital, Tai'an 271000, China
| | - Min Lin
- Department of Pathology, Tai'an Central Hospital, Tai'an 271000, China
| | - Yanmei Xu
- Department of Pathology, Shanxian Central Hospital, Heze 274300, China
| | - Xiaomei Li
- Department of Pathology, Tai'an Central Hospital, Tai'an 271000, China
| |
Collapse
|
48
|
Caliò A, Segala D, Munari E, Brunelli M, Martignoni G. MiT Family Translocation Renal Cell Carcinoma: from the Early Descriptions to the Current Knowledge. Cancers (Basel) 2019; 11:E1110. [PMID: 31382581 PMCID: PMC6721505 DOI: 10.3390/cancers11081110] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 12/27/2022] Open
Abstract
The new category of MiT family translocation renal cell carcinoma has been included into the World Health Organization (WHO) classification in 2016. The MiT family translocation renal cell carcinoma comprises Xp11 translocation renal cell carcinoma harboring TFE3 gene fusions and t(6;11) renal cell carcinoma harboring TFEB gene fusion. At the beginning, they were recognized in childhood; nevertheless, it has been demonstrated that these neoplasms can occur in adults as well. In the nineties, among Xp11 renal cell carcinoma, ASPL, PRCC, and SFPQ (PSF) were the first genes recognized as partners in TFE3 rearrangement. Recently, many other genes have been identified, and a wide spectrum of morphologies has been described. For this reason, the diagnosis may be challenging based on the histology, and the differential diagnosis includes the most common renal cell neoplasms and pure epithelioid PEComa/epithelioid angiomyolipoma of the kidney. During the last decades, many efforts have been made to identify immunohistochemical markers to reach the right diagnosis. To date, staining for PAX8, cathepsin K, and melanogenesis markers are the most useful identifiers. However, the diagnosis requires the demonstration of the chromosomal rearrangement, and fluorescent in situ hybridization (FISH) is considered the gold standard. The outcome of Xp11 translocation renal cell carcinoma is highly variable, with some patients surviving decades with indolent disease and others dying rapidly of progressive disease. Despite most instances of t(6;11) renal cell carcinoma having an indolent clinical course, a few published cases demonstrate aggressive behavior. Recently, renal cell carcinomas with TFEB amplification have been described in connection with t(6;11) renal cell carcinoma. Those tumors appear to be associated with a more aggressive clinical course. For the aggressive cases of MiT family translocation carcinoma, the optimal therapy remains to be determined; however, new target therapies seem to be promising, and the search for predictive markers is mandatory.
Collapse
Affiliation(s)
- Anna Caliò
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona 37134, Italy
| | - Diego Segala
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda 37019, Italy
| | - Enrico Munari
- Department of Pathology, Sacro Cuore Hospital, Negrar 37024, Italy
| | - Matteo Brunelli
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona 37134, Italy
| | - Guido Martignoni
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona 37134, Italy.
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda 37019, Italy.
| |
Collapse
|
49
|
Fukuda H, Kato I, Furuya M, Tanaka R, Takagi T, Kondo T, Nagashima Y. A novel partner of TFE3 in the Xp11 translocation renal cell carcinoma: clinicopathological analyses and detection of EWSR1-TFE3 fusion. Virchows Arch 2018; 474:389-393. [DOI: 10.1007/s00428-018-2509-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/13/2018] [Accepted: 12/10/2018] [Indexed: 12/27/2022]
|
50
|
Damayanti NP, Budka JA, Khella HWZ, Ferris MW, Ku SY, Kauffman E, Wood AC, Ahmed K, Chintala VN, Adelaiye-Ogala R, Elbanna M, Orillion A, Chintala S, Kao C, Linehan WM, Yousef GM, Hollenhorst PC, Pili R. Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma. Clin Cancer Res 2018; 24:5977-5989. [PMID: 30061365 PMCID: PMC6279468 DOI: 10.1158/1078-0432.ccr-18-0269] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/19/2018] [Accepted: 07/23/2018] [Indexed: 01/05/2023]
Abstract
PURPOSE Translocation renal cell carcinoma (tRCC) represents a rare subtype of kidney cancer associated with various TFE3, TFEB, or MITF gene fusions that are not responsive to standard treatments for RCC. Therefore, the identification of new therapeutic targets represents an unmet need for this disease. EXPERIMENTAL DESIGN We have established and characterized a tRCC patient-derived xenograft, RP-R07, as a novel preclinical model for drug development by using next-generation sequencing and bioinformatics analysis. We then assessed the therapeutic potential of inhibiting the identified pathway using in vitro and in vivo models. RESULTS The presence of a SFPQ-TFE3 fusion [t(X;1) (p11.2; p34)] with chromosomal break-points was identified by RNA-seq and validated by RT-PCR. TFE3 chromatin immunoprecipitation followed by deep sequencing analysis indicated a strong enrichment for the PI3K/AKT/mTOR pathway. Consistently, miRNA microarray analysis also identified PI3K/AKT/mTOR as a highly enriched pathway in RP-R07. Upregulation of PI3/AKT/mTOR pathway in additional TFE3-tRCC models was confirmed by significantly higher expression of phospho-S6 (P < 0.0001) and phospho-4EBP1 (P < 0.0001) in established tRCC cell lines compared with clear cell RCC cells. Simultaneous vertical targeting of both PI3K/AKT and mTOR axis provided a greater antiproliferative effect both in vitro (P < 0.0001) and in vivo (P < 0.01) compared with single-node inhibition. Knockdown of TFE3 in RP-R07 resulted in decreased expression of IRS-1 and inhibited cell proliferation. CONCLUSIONS These results identify TFE3/IRS-1/PI3K/AKT/mTOR as a potential dysregulated pathway in TFE3-tRCC, and suggest a therapeutic potential of vertical inhibition of this axis by using a dual PI3K/mTOR inhibitor for patients with TFE3-tRCC.
Collapse
Affiliation(s)
- Nur P Damayanti
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Justin A Budka
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Heba W Z Khella
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science at the Li KaShing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
| | - Mary W Ferris
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Sheng Yu Ku
- Department of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, New York
| | - Eric Kauffman
- Department of Urology and Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York
| | - Anthony C Wood
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Khunsha Ahmed
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Venkata Nithinsai Chintala
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Remi Adelaiye-Ogala
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - May Elbanna
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Ashley Orillion
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Sreenivasulu Chintala
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Chinghai Kao
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | | | - George M Yousef
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science at the Li KaShing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
| | - Peter C Hollenhorst
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Roberto Pili
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana.
| |
Collapse
|