1
|
Li Z, Zhong Y, Ye D, Yang J, Chen L. Revealing NAPSA's role in ccRCC: Insights from single-cell RNA sequencing. Gene 2025; 959:149478. [PMID: 40194687 DOI: 10.1016/j.gene.2025.149478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is aggressive and heterogeneous, resulting in poor prognosis due to frequent metastasis. Napsin A, an aspartic proteinase encoded by the NAPSA gene, is involved in protein processing and is expressed in the kidney and lung, but its function is not well understood. Studying ccRCC's molecular characteristics, including Napsin A, is vital for enhancing diagnostics and treatment. METHODS Single-cell RNA sequencing data from the GEO database (GSE210042) were analyzed, including seven tumor and two normal samples. The Seurat package was used for data preprocessing, clustering, and visualization. Differential expression and enrichment analyses were conducted between tumor and normal cells, and cell-to-cell communication was assessed between NAPSA + and NAPSA- cells. The correlation between NAPSA expression and EMT score was analyzed using TCGA-KIRC data. In vitro experiments involved transfecting OS-RC-2 and Caki-1 ccRCC cell lines with siRNA targeting NAPSA. Effect on the cellular EMT process induced by TGF-β1 was assessed by immunofluorescence staining. RESULTS NAPSA was primarily expressed in podocytes and ccRCC epithelial cells, with significantly reduced levels in tumor tissues associated with poor prognosis. NAPSA downregulation may influence various biological pathways and enhance communication with tumor-associated macrophages and mast cells. Silencing NAPSA increased TGF-β1-induced epithelial-mesenchymal transition (EMT). CONCLUSION The study highlights NAPSA's expression characteristics and potential role in ccRCC, suggesting it may serve as a biomarker. Further research is needed to elucidate NAPSA's mechanisms and explore its applications in precision medicine.
Collapse
Affiliation(s)
- Zhichao Li
- Doctoral Candidate in School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750001, Ningxia, China; Shanghai Jiahui International Hospital, Shanghai, China
| | - Yuanjie Zhong
- Graduate Student in Ningxia Medical University, Yinchuan 750001, Ningxia, China
| | - Dan Ye
- Ningxia Medical University, Yinchuan 750001 Ningxia, China
| | - Jincheng Yang
- Department of Urology, Yinchuan First People's Hospital, Deputy Chief Physician, Yinchuan 750001, Ningxia, China
| | - Linbao Chen
- Yinchuan Women and Children Healthcare Hospital, Yinchuan 750001, Ningxia, China; Yinchuan First People's Hospital, Yinchuan 750001, Ningxia, China.
| |
Collapse
|
2
|
Larose C, Leduc C, Chevrier M. Napsin-A Immunohistochemistry in the Diagnosis of Pulmonary Alveolar Proteinosis. Arch Pathol Lab Med 2025; 149:568-572. [PMID: 39370148 DOI: 10.5858/arpa.2024-0136-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 10/08/2024]
Abstract
CONTEXT.— The diagnosis of pulmonary alveolar proteinosis (PAP) relies on a limited set of stains, namely hematoxylin-eosin and periodic acid-Schiff-diastase (PAS-D), demonstrating abundant alveolar material representing mostly surfactant. As cells harboring surfactant also express Napsin-A (pneumocytes and macrophages), we hypothesized that it would also be expressed within alveoli in PAP. OBJECTIVE.— To evaluate the sensitivity and specificity of Napsin-A in the diagnosis of PAP. DESIGN.— A 12-year retrospective case control study was designed to identify cases of PAP and potential histologic mimics (intra-alveolar fibrin, pulmonary edema, diffuse alveolar damage, and alveolar mucinosis). PAS-D staining and Napsin-A immunohistochemistry were performed. Distribution and intensity were evaluated by using a semiquantitative 3-point scale. Positivity was defined as 2+ intensity score, regardless of distribution. RESULTS.— Eleven cases of PAP and 46 control cases were identified. Napsin-A showed positivity in all PAP cases and 3 of 12 cases of edema. Among positive cases, all those with a 2+ distribution were PAP cases, with heterogeneous (1+) staining in all cases of edema. PAS-D showed positivity in all cases of PAP and most controls, except cases of edema. Sensitivity and specificity of Napsin-A for PAP were 100% and 94%, respectively, and of PAS-D for PAP, 100% and 21%, respectively. Double positivity for Napsin-A and PAS-D was 100% specific and sensitive for PAP. CONCLUSIONS.— This study is the first to demonstrate that Napsin-A is highly specific for the diagnosis of PAP, more so than PAS-D. It also shows that the combined positivity of Napsin-A and PAS-D is 100% specific and sensitive for PAP.
Collapse
Affiliation(s)
- Catherine Larose
- From the Department of Pathology, University of Montreal Health Centre, Montréal, Québec, Canada (Larose, Leduc)
| | - Charles Leduc
- From the Department of Pathology, University of Montreal Health Centre, Montréal, Québec, Canada (Larose, Leduc)
| | - Martin Chevrier
- the Department of Pathology, University of Sherbrooke Health Centre, Sherbrooke, Québec, Canada (Chevrier)
| |
Collapse
|
3
|
Maynard DM, Gochuico BR, Pri Chen H, Bleck CKE, Zerfas PM, Introne WJ, Gahl WA, Malicdan MCV. Insights into the renal pathophysiology in Hermansky-Pudlak syndrome-1 from urinary extracellular vesicle proteomics and a new mouse model. FEBS Lett 2025; 599:1055-1074. [PMID: 39739361 PMCID: PMC11995682 DOI: 10.1002/1873-3468.15088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/07/2024] [Accepted: 12/01/2024] [Indexed: 01/02/2025]
Abstract
Hermansky-Pudlak syndrome type 1 (HPS-1) is a rare, autosomal recessive disorder caused by defects in the biogenesis of lysosome-related organelles complex-3 (BLOC-3). Impaired kidney function is among its clinical manifestations. To investigate HPS-1 renal involvement, we employed 1D-gel-LC-MS/MS and compared the protein composition of urinary extracellular vesicles (uEVs) from HPS-1 patients to normal control individuals. We identified 1029 proteins, 149 of which were altered in HPS-1 uEVs. Ingenuity Pathway Analysis revealed disruptions in mitochondrial function and the LXR/RXR pathway that regulates lipid metabolism, which is supported by our novel Hps1 knockout mouse. Serum concentration of the LXR/RXR pathway protein ApoA1 in our patient cohort was positively correlated with kidney function (with the estimated glomerular filtration rate or eGFR). uEVs can be used to study epithelial cell protein trafficking in HPS-1 and may provide outcome measures for HPS-1 therapeutic interventions.
Collapse
Affiliation(s)
- Dawn M. Maynard
- Section on Human Biochemical Genetics, Medical Genetics Branch, NHGRINational Institutes of HealthBethesdaMDUSA
| | - Bernadette R. Gochuico
- Section on Human Biochemical Genetics, Medical Genetics Branch, NHGRINational Institutes of HealthBethesdaMDUSA
| | - Hadass Pri Chen
- Section on Human Biochemical Genetics, Medical Genetics Branch, NHGRINational Institutes of HealthBethesdaMDUSA
| | | | - Patricia M. Zerfas
- Office of Research Services, Office of the DirectorNational Institutes of HealthBethesdaMDUSA
| | - Wendy J. Introne
- Section on Human Biochemical Genetics, Medical Genetics Branch, NHGRINational Institutes of HealthBethesdaMDUSA
| | - William A. Gahl
- Section on Human Biochemical Genetics, Medical Genetics Branch, NHGRINational Institutes of HealthBethesdaMDUSA
| | - May C. V. Malicdan
- Section on Human Biochemical Genetics, Medical Genetics Branch, NHGRINational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
4
|
Tang L, Li N, Yang Z, Lin Y, Gao G, Lin Q, Wang Y. Targeted Nanoclusters for Intratracheal Delivery in Intraoperative Imaging of Lung Adenocarcinoma. Int J Nanomedicine 2025; 20:3575-3594. [PMID: 40125441 PMCID: PMC11930283 DOI: 10.2147/ijn.s509009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/06/2025] [Indexed: 03/25/2025] Open
Abstract
Background Computed tomography (CT) is widely used all over the world, and the detection rate of early lung adenocarcinoma is increasing. Minimally invasive thoracic surgery (MITS) has emerged as the preferred surgical approach for lung adenocarcinoma, but identifying small lung adenocarcinomas is difficult. Therefore, there is a need for a non-invasive, convenient and efficient way to localize lung adenocarcinomas. Materials and Methods A targeted gold nanocluster for intraoperative fluorescence imaging by intratracheal delivery has been designed. Au-GSH-anti Napsin A nanoclusters (NapA-AuNCs) were synthesized, and their physicochemical properties and optical properties were characterized. Target effect, cytotoxicity and fluorescence time curve of NapA-AuNCs, were tested in vivo and in vitro, and intratracheal delivery was also carried. Results NapA-AuNCs targeting lung adenocarcinoma with red fluorescence and good mucus penetration were synthesized, which had the targeting property of A549 and lung adenocarcinoma tissue, and also had very low toxicity to normal lung epithelial cells and organs. Intracheal delivery involves faster imaging of lung adenocarcinoma and less accumulation of other organs than intravenous administration. Conclusion NapA-AuNCs targeting lung adenocarcinoma were successfully conjugated, and intratracheal delivery is a safe, effective for lung adenocarcinoma intraoperative localization.
Collapse
Affiliation(s)
- Lu Tang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Ning Li
- Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Zhe Yang
- Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yanatai, Shandong, People’s Republic of China
| | - Yangliu Lin
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Ge Gao
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Yue Wang
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
5
|
Zhang J, Zang X, Jiao P, Wu J, Meng W, Zhao L, Lv Z. Alterations of Ceramides, Acylcarnitines, GlyceroLPLs, and Amines in NSCLC Tissues. J Proteome Res 2024; 23:4343-4358. [PMID: 39317643 DOI: 10.1021/acs.jproteome.4c00344] [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: 09/26/2024]
Abstract
Abnormal lipid metabolism plays an important role in cancer development. In this study, nontargeted lipidomic study on 230 tissue specimens from 79 nonsmall cell lung cancer (NSCLC) patients was conducted using ultraperformance liquid chromatography-high-resolution mass spectrometry (UPLC-HRMS). Downregulation of sphingosine and medium-long-chain ceramides and short-medium-chain acylcarnitine, upregulation of long-chain acylcarnitine C20:0, and enhanced histamine methylation were revealed in NSCLC tissues. Compared with paired noncancerous tissues, adenocarcinoma (AC) tissues had significantly decreased levels of sphingosine, medium-long-chain ceramides (Cer d18:1/12:0 and Cer d16:1/14:0, Cer d18:0/16:0, Cer d18:1/16:0, Cer d18:2/16:0, Cer d18:2/18:0), short-medium-chain (C2-C16) acylcarnitines, LPC 20:0 and LPC 22:1, and significantly increased levels of the long-chain acylcarnitine C20:0, LPC 16:0, LPC P-16:0, LPC 20:1, LPC 20:2, glyceroPC, LPE 16:0, and LPE 18:2. In squamous cell carcinoma (SCC) tissues, sphingosine, Cer d18:2/16:0 and Cer d18:2/18:0, and short-medium-chain acylcarnitines had significantly lower levels, while long-chain acylcarnitines (C20:0, and C22:0 or C22:0 M), LPC 20:1, LPC 20:2, and N1,N12-diacetylspermine had significantly higher levels compared to controls. In AC and SCC tissues, the levels of LPG 18:0, LPG 18:1, and LPS 18:1 were significantly decreased, while the levels of ceramide-1-phosphate (C1P) d18:0/3:0 or LPE P-16:0, N1-acetylspermidine, and 1-methylhistamine were significantly increased than controls. Furthermore, an orthogonal partial least-squares-discriminant analysis (OPLS-DA) model based on a 4-lipid panel was established, showing good discrimination ability between cancerous and noncancerous tissues.
Collapse
Affiliation(s)
- Jie Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P. R. China
| | - Xiaoling Zang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P. R. China
- Qingdao Marine Science and Technology Center, Qingdao, Shandong 266235, P. R. China
| | - Peng Jiao
- Department of Thoracic Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P. R. China
| | - Jiangyu Wu
- Department of Thoracic Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P. R. China
| | - Wei Meng
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P. R. China
| | - Lizhen Zhao
- College of Physics, Qingdao University, Qingdao, Shandong 266071, P. R. China
| | - Zhihua Lv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P. R. China
- Qingdao Marine Science and Technology Center, Qingdao, Shandong 266235, P. R. China
| |
Collapse
|
6
|
Rerkpichaisuth V, Lau RP, Meyerson C, Fishbein GA. The utility of the lineage specific immunohistochemical stains SATB2, CDX2, and villin, and the mucin glycoproteins MUC2, MUC5AC, and MUC6 to distinguish pulmonary invasive mucinous adenocarcinoma from metastatic colorectal carcinoma. Hum Pathol 2024; 151:105627. [PMID: 39029534 DOI: 10.1016/j.humpath.2024.105627] [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: 04/25/2024] [Revised: 07/03/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
CONTEXT The lungs are a common site of tumor metastasis. While morphology and immunophenotype can help differentiate primary from metastatic tumors, distinguishing pulmonary invasive mucinous adenocarcinoma (PIMA) from metastatic colorectal adenocarcinoma (CRC) may occasionally be challenging due to overlapping morphological and immunohistochemical features. Lineage-specific markers such as CDX2, TTF-1, and napsin A are helpful with pulmonary non-mucinous adenocarcinoma (PNMA), however they are non-specific and insensitive when applied to PIMA. SATB2 is a newer marker that distinguishes CRC from upper gastrointestinal and pancreaticobiliary tumors; its utility in distinguishing CRC from PIMA has not been fully elucidated. OBJECTIVE To evaluate the performance of lineage-specific and mucin glycoprotein immunostains in distinguishing PIMA and CRC. DESIGN We stained tissue microarrays comprising 34 PNMA, 31 PIMA, and 32 CRC with CK7, CK20, SATB2, CDX2, villin, TTF-1, napsin A, and gel-forming mucins MUC2, MUC5AC, and MUC6. RESULTS PIMA showed significant (>50% of cells) expression of SATB2 (6%), CDX2 (6%), villin (74%), TTF-1 (13%), and napsin A (23%). However, significant CK7 expression was seen in nearly all PIMA (30/31) and none of the metastatic CRC. CONCLUSION Our results suggest that CK7 remains one of the most useful markers for distinguishing primary PIMA from metastatic CRC. Expression of the mucin glycoproteins MUC5AC and MUC6 and lack of expression of MUC2 favored a diagnosis of PIMA, but expression of these markers was too heterogeneous to be of clinical utility. To our knowledge this is the only study comparing the immunohistochemical profile of PIMA and metastatic CRC in lung metastasectomy specimens.
Collapse
Affiliation(s)
- Vilasinee Rerkpichaisuth
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ryan P Lau
- Department of Pathology, Kaiser Permanente, Los Angeles Medical Center, Los Angeles, CA, USA
| | - Cherise Meyerson
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Gregory A Fishbein
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Knott EP, Kim EY, Kim EQ, Freire R, Medina JA, Wang Y, Chen CB, Wu C, Wangpaichitr M, Conejo-Garcia JR, Lim DC. Orthotopic Models Using New, Murine Lung Adenocarcinoma Cell Lines Simulate Human Non-Small Cell Lung Cancer Treated with Immunotherapy. Cells 2024; 13:1120. [PMID: 38994972 PMCID: PMC11240577 DOI: 10.3390/cells13131120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
Understanding tumor-host immune interactions and the mechanisms of lung cancer response to immunotherapy is crucial. Current preclinical models used to study this often fall short of capturing the complexities of human lung cancer and lead to inconclusive results. To bridge the gap, we introduce two new murine monoclonal lung cancer cell lines for use in immunocompetent orthotopic models. We demonstrate how our cell lines exhibit immunohistochemical protein expression (TTF-1, NapA, PD-L1) and common driver mutations (KRAS, p53, and p110α) seen in human lung adenocarcinoma patients, and how our orthotopic models respond to combination immunotherapy in vivo in a way that closely mirrors current clinical outcomes. These new lung adenocarcinoma cell lines provide an invaluable, clinically relevant platform for investigating the intricate dynamics between tumor and the immune system, and thus potentially contributes to a deeper understanding of immunotherapeutic approaches to lung cancer treatment.
Collapse
Affiliation(s)
- Eric P. Knott
- Research Services, Miami VA Healthcare System, Miami, FL 33125, USA; (E.P.K.); (E.Y.K.); (E.Q.K.); (C.W.); (M.W.)
- Division of Pulmonary & Critical Care Medicine, Miami VA Healthcare System, Miami, FL 33125, USA
| | - Emily Y. Kim
- Research Services, Miami VA Healthcare System, Miami, FL 33125, USA; (E.P.K.); (E.Y.K.); (E.Q.K.); (C.W.); (M.W.)
- South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125, USA
| | - Edison Q. Kim
- Research Services, Miami VA Healthcare System, Miami, FL 33125, USA; (E.P.K.); (E.Y.K.); (E.Q.K.); (C.W.); (M.W.)
| | - Rochelle Freire
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Justin A. Medina
- Department of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Yujie Wang
- Department of Industrial and Systems Engineering, University of Miami, Coral Gables, FL 33146, USA; (Y.W.); (C.-B.C.)
| | - Cheng-Bang Chen
- Department of Industrial and Systems Engineering, University of Miami, Coral Gables, FL 33146, USA; (Y.W.); (C.-B.C.)
| | - Chunjing Wu
- Research Services, Miami VA Healthcare System, Miami, FL 33125, USA; (E.P.K.); (E.Y.K.); (E.Q.K.); (C.W.); (M.W.)
| | - Medhi Wangpaichitr
- Research Services, Miami VA Healthcare System, Miami, FL 33125, USA; (E.P.K.); (E.Y.K.); (E.Q.K.); (C.W.); (M.W.)
- South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125, USA
- Department of Surgery, Cardiothoracic Surgery, University of Miami, Miami, FL 33136, USA
| | - Jose R. Conejo-Garcia
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA;
| | - Diane C. Lim
- Research Services, Miami VA Healthcare System, Miami, FL 33125, USA; (E.P.K.); (E.Y.K.); (E.Q.K.); (C.W.); (M.W.)
- Division of Pulmonary & Critical Care Medicine, Miami VA Healthcare System, Miami, FL 33125, USA
- Division of Pulmonary/Critical Care/Sleep, University of Miami, Miami, FL 33136, USA
- Division of Sleep Medicine, Miami VA Healthcare System, Miami, FL 33125, USA
| |
Collapse
|
8
|
Simbolo M, Centonze G, Gkountakos A, Monti V, Maisonneuve P, Golovco S, Sabella G, Del Gobbo A, Gobbo S, Ferrero S, Fabbri A, Pardo C, Garzone G, Prinzi N, Pusceddu S, Testi A, Rolli L, Mangogna A, Bercich L, Benvenuti MR, Bria E, Pilotto S, Berruti A, Pastorino U, Capella C, Infante M, Milella M, Scarpa A, Milione M. Characterization of two transcriptomic subtypes of marker-null large cell carcinoma of the lung suggests different origin and potential new therapeutic perspectives. Virchows Arch 2024; 484:777-788. [PMID: 38168015 PMCID: PMC11106141 DOI: 10.1007/s00428-023-03721-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/13/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024]
Abstract
Pulmonary large cell carcinoma (LCC) is an undifferentiated neoplasm lacking morphological, histochemical, and immunohistochemical features of small cell lung cancer, adenocarcinoma (ADC), or squamous cell carcinoma (SCC). The available molecular information on this rare disease is limited. This study aimed to provide an integrated molecular overview of 16 cases evaluating the mutational asset of 409 genes and the transcriptomic profiles of 20,815 genes. Our data showed that TP53 was the most frequently inactivated gene (15/16; 93.7%) followed by RB1 (5/16; 31.3%) and KEAP1 (4/16; 25%), while CRKL and MYB genes were each amplified in 4/16 (25%) cases and MYC in 3/16 (18.8%) cases; transcriptomic analysis identified two molecular subtypes including a Pure-LCC and an adenocarcinoma like-LCC (ADLike-LCC) characterized by different activated pathways and cell of origin. In the Pure-LCC group, POU2F3 and FOXI1 were distinctive overexpressed markers. A tuft cell-like profile and the enrichment of a replication stress signature, particularly involving ATR, was related to this profile. Differently, the ADLike-LCC were characterized by an alveolar-cell transcriptomic profile and association with AIM2 inflammasome complex signature. In conclusion, our study split the histological marker-null LCC into two different transcriptomic entities, with POU2F3, FOXI1, and AIM2 genes as differential expression markers that might be probed by immunohistochemistry for the differential diagnosis between Pure-LCC and ADLike-LCC. Finally, the identification of several signatures linked to replication stress in Pure-LCC and inflammasome complex in ADLike-LCC could be useful for designing new potential therapeutic approaches for these subtypes.
Collapse
Affiliation(s)
- Michele Simbolo
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Giovanni Centonze
- 1st Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Anastasios Gkountakos
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Valentina Monti
- 1st Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Patrick Maisonneuve
- Division of Epidemiology and Biostatistics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Stela Golovco
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Giovanna Sabella
- 1st Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Alessandro Del Gobbo
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Gobbo
- Department of Traslational Medicine, University of Ferrara, Ferrara, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessandra Fabbri
- 2nd Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Carlotta Pardo
- 1st Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Giovanna Garzone
- 1st Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Natalie Prinzi
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sara Pusceddu
- Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale Dei Tumori, Milan, Italy
| | - Adele Testi
- 2nd Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Luigi Rolli
- Thoracic Surgery Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, IRCCS Burlo Garofalo, Trieste, Italy
| | - Luisa Bercich
- Department of Pathology, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Mauro Roberto Benvenuti
- Thoracic Surgery Unit, Department of Medical and Surgical Specialties Radiological Sciences and Public Health, Medical Oncology, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Emilio Bria
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Sara Pilotto
- Section of Oncology, Department of Medicine, University of Verona, Verona, Italy
| | - Alfredo Berruti
- Medical Oncology Unit, ASST Spedali Civili of Brescia, Department of Medical and Surgical Specialties, Radiological Science and Public Health, University of Brescia, Brescia, Italy
| | - Ugo Pastorino
- Thoracic Surgery Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Carlo Capella
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | | | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona, Verona, Italy
| | - Aldo Scarpa
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy.
- ARC-NET Research Centre for Applied Research On Cancer, University and Hospital Trust of Verona, Piazzale Scuro, 10, 37134, Verona (VR), Italy.
| | - Massimo Milione
- 1st Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| |
Collapse
|
9
|
Ostmeyer J, Park JY, von Itzstein MS, Hsiehchen D, Fattah F, Gwin M, Catalan R, Khan S, Raj P, Wakeland EK, Xie Y, Gerber DE. T-cell tolerant fraction as a predictor of immune-related adverse events. J Immunother Cancer 2023; 11:e006437. [PMID: 37580069 PMCID: PMC10432621 DOI: 10.1136/jitc-2022-006437] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI) therapies may cause unpredictable and potentially severe autoimmune toxicities termed immune-related adverse events (irAEs). Because T cells mediate ICI effects, T cell profiling may provide insight into the risk of irAEs. Here we evaluate a novel metric-the T-cell tolerant fraction-as a predictor of future irAEs. METHODS We examined T-cell receptor beta (TRB) locus sequencing from baseline pretreatment samples from an institutional registry and previously published studies. For each patient, we used TRB sequences to calculate the T-cell tolerant fraction, which was then assessed as a predictor of future irAEs (classified as Common Terminology Criteria for Adverse Event grade 0-1 vs grade ≥2). We then compared the tolerant fraction to TRB clonality and diversity. Finally, the tolerant fraction was assessed on (1) T cells enriched against napsin A, a potential autoantigen of irAEs; (2) thymic versus peripheral blood T cells; and (3) TRBs specific for various infections and autoimmune diseases. RESULTS A total of 77 patients with cancer (22 from an institutional registry and 55 from published studies) receiving ICI therapy (43 CTLA4, 19 PD1/PDL1, 15 combination CTLA4+PD1/PDL1) were included in the study. The tolerant fraction was significantly lower in cases with clinically significant irAEs (p<0.001) and had an area under the receiver operating curve (AUC) of 0.79. The tolerant fraction was lower for each ICI treatment category, reaching statistical significance for CTLA4 (p<0.001) and demonstrating non-significant trends for PD1/PDL1 (p=0.21) and combination ICI (p=0.18). The tolerant fraction for T cells enriched against napsin A was lower than other samples. The tolerant fraction was also lower in thymic versus peripheral blood samples, and lower in some (multiple sclerosis) but not other (type 1 diabetes) autoimmune diseases. In our study cohort, TRB clonality had an AUC of 0.62, and TRB diversity had an AUC of 0.60 for predicting irAEs. CONCLUSIONS Among patients receiving ICI, the baseline T-cell tolerant fraction may serve as a predictor of clinically significant irAEs.
Collapse
Affiliation(s)
- Jared Ostmeyer
- Peter O'Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jason Y Park
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mitchell S von Itzstein
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - David Hsiehchen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Harold C Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Farjana Fattah
- Harold C Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mary Gwin
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rodrigo Catalan
- Harold C Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Shaheen Khan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Prithvi Raj
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Edward K Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yang Xie
- Peter O'Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Harold C Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - David E Gerber
- Peter O'Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Harold C Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
10
|
Tourigny DS, Zucker M, Kim M, Russo P, Coleman J, Lee CH, Carlo MI, Chen YB, Hakimi AA, Kotecha RR, Reznik E. Molecular Characterization of the Tumor Microenvironment in Renal Medullary Carcinoma. Front Oncol 2022; 12:910147. [PMID: 35837094 PMCID: PMC9275834 DOI: 10.3389/fonc.2022.910147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Renal medullary carcinoma (RMC) is a highly aggressive disease associated with sickle hemoglobinopathies and universal loss of the tumor suppressor gene SMARCB1. RMC has a relatively low rate of incidence compared with other renal cell carcinomas (RCCs) that has hitherto made molecular profiling difficult. To probe this rare disease in detail we performed an in-depth characterization of the RMC tumor microenvironment using a combination of genomic, metabolic and single-cell RNA-sequencing experiments on tissue from a representative untreated RMC patient, complemented by retrospective analyses of archival tissue and existing published data. Our study of the tumor identifies a heterogenous population of malignant cell states originating from the thick ascending limb of the Loop of Henle within the renal medulla. Transformed RMC cells displayed the hallmarks of increased resistance to cell death by ferroptosis and proteotoxic stress driven by MYC-induced proliferative signals. Specifically, genomic characterization of RMC tumors provides substantiating evidence for the recently proposed dependence of SMARCB1-difficient cancers on proteostasis modulated by an intact CDKN2A-p53 pathway. We also provide evidence that increased cystine-mTORC-GPX4 signaling plays a role in protecting transformed RMC cells against ferroptosis. We further propose that RMC has an immune landscape comparable to that of untreated RCCs, including heterogenous expression of the immune ligand CD70 within a sub-population of tumor cells. The latter could provide an immune-modulatory role that serves as a viable candidate for therapeutic targeting.
Collapse
Affiliation(s)
- David S. Tourigny
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, United States
- School of Mathematics, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: David S. Tourigny, ; A. Ari Hakimi, ; Ritesh R. Kotecha, ; Ed Reznik,
| | - Mark Zucker
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Minsoo Kim
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Paul Russo
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jonathan Coleman
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Chung-Han Lee
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Maria I. Carlo
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - A. Ari Hakimi
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- *Correspondence: David S. Tourigny, ; A. Ari Hakimi, ; Ritesh R. Kotecha, ; Ed Reznik,
| | - Ritesh R. Kotecha
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- *Correspondence: David S. Tourigny, ; A. Ari Hakimi, ; Ritesh R. Kotecha, ; Ed Reznik,
| | - Ed Reznik
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- *Correspondence: David S. Tourigny, ; A. Ari Hakimi, ; Ritesh R. Kotecha, ; Ed Reznik,
| |
Collapse
|
11
|
Weidemann S, Böhle JL, Contreras H, Luebke AM, Kluth M, Büscheck F, Hube-Magg C, Höflmayer D, Möller K, Fraune C, Bernreuther C, Rink M, Simon R, Menz A, Hinsch A, Lebok P, Clauditz T, Sauter G, Uhlig R, Wilczak W, Steurer S, Burandt E, Krech R, Dum D, Krech T, Marx A, Minner S. Napsin A Expression in Human Tumors and Normal Tissues. Pathol Oncol Res 2021; 27:613099. [PMID: 34257582 PMCID: PMC8262149 DOI: 10.3389/pore.2021.613099] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 02/26/2021] [Indexed: 11/25/2022]
Abstract
Background: Novel aspartic proteinase of the pepsin family A (Napsin A, TAO1/TAO2) is a functional aspartic proteinase which is involved in the maturation of prosurfactant protein B in type II pneumocytes and the lysosomal protein catabolism in renal cells. Napsin A is highly expressed in adenocarcinomas of the lung and is thus commonly used to affirm this diagnosis. However, studies have shown that other tumors can also express Napsin A. Methods: To comprehensively determine Napsin A expression in normal and tumor tissue, 11,957 samples from 115 different tumor types and subtypes as well as 500 samples of 76 different normal tissue types were evaluable by immunohistochemistry on tissue microarrays. Results: Napsin A expression was present in 16 different tumor types. Adenocarcinoma of the lung (85.6%), clear cell adenocarcinoma of the ovary (71.7%), clear cell adenocarcinoma of the endometrium (42.8%), papillary renal cell carcinoma (40.2%), clear cell (tubulo) papillary renal cell carcinoma (16.7%), endometrial serous carcinoma (9.3%), papillary thyroid carcinoma (9.3%) and clear cell renal cell carcinoma (8.2%) were among the tumors with the highest prevalence of Napsin A positivity. In papillary and clear cell renal cell carcinoma, reduced Napsin A expression was linked to adverse clinic-pathological features (p ≤ 0.03). Conclusion: This methodical approach enabled us to identify a ranking order of tumors according to their relative prevalence of Napsin A expression. The data also show that loss of Napsin A is linked to tumor dedifferentiation in renal cell carcinomas.
Collapse
Affiliation(s)
- Sören Weidemann
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Lukas Böhle
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hendrina Contreras
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas M Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Büscheck
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Doris Höflmayer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Möller
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Rink
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Menz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ria Uhlig
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Waldemar Wilczak
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rainer Krech
- Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck, Germany
| | - David Dum
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck, Germany
| | - Andreas Marx
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Pathology, Academic Hospital Fuerth, Fuerth, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
12
|
Barber J, Sikakana P, Sadler C, Baud D, Valentin JP, Roberts R. A target safety assessment of the potential toxicological risks of targeting plasmepsin IX/X for the treatment of malaria. Toxicol Res (Camb) 2021; 10:203-213. [PMID: 33884171 DOI: 10.1093/toxres/tfaa106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/28/2022] Open
Abstract
The aspartic proteases plasmepsin IX/X are important antimalarial drug targets due to their specificity to the malaria parasite and their vital role as mediators of disease progression. Focusing on parasite-specific targets where no human homologue exists reduces the possibility of on-target drug toxicity. However, there is a risk of toxicity driven by inadequate selectivity for plasmepsins IX/X in Plasmodium over related mammalian aspartic proteases. Of these, CatD/E may be of most toxicological relevance as CatD is a ubiquitous lysosomal enzyme present in most cell types and CatE is found in the gut and in erythrocytes, the clinically significant site of malarial infection. Based on mammalian aspartic protease physiology and adverse drug reactions (ADRs) to FDA-approved human immunodeficiency virus (HIV) aspartic protease inhibitors, we predicted several potential toxicities including β-cell and congenital abnormalities, hypotension, hypopigmentation, hyperlipidaemia, increased infection risk and respiratory, renal, gastrointestinal, dermatological, and other epithelial tissue toxicities. These ADRs to the HIV treatments are likely to be a result of host aspartic protease inhibition due a lack of specificity for the HIV protease; plasmepsins are much more closely related to human CatD than to HIV proteinase. Plasmepsin IX/X inhibition presents an opportunity to specifically target Plasmodium as an effective antimalarial treatment, providing adequate selectivity can be obtained. Potential plasmepsin IX/X inhibitors should be assayed for inhibitory activity against the main human aspartic proteases and particularly CatD/E. An investigative rodent study conducted early in drug discovery would serve as an initial risk assessment of the potential hazards identified.
Collapse
Affiliation(s)
- Jane Barber
- ApconiX, Alderley Park, Alderley Edge, SK10 4TG, UK
| | | | | | - Delphine Baud
- Medicines for Malaria Venture, 20 Route de Pré-Bois, Geneva 1215, Switzerland
| | - Jean-Pierre Valentin
- UCB Biopharma SRL, Building R9, Chemin du Foriest, 1420 Braine-l'Alleud, Belgium
| | - Ruth Roberts
- ApconiX, Alderley Park, Alderley Edge, SK10 4TG, UK
| |
Collapse
|
13
|
Grassystatin-derived peptides selectively inhibit cathepsin E and have low affinity to cathepsin D. Biochem Biophys Res Commun 2020; 527:238-241. [PMID: 32446374 DOI: 10.1016/j.bbrc.2020.04.070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 04/14/2020] [Indexed: 11/22/2022]
Abstract
Aspartic proteases are important biomarkers of human disease and interesting targets for modulation of immune response via MHC class II antigen processing inhibition. The lack of inhibitors with sufficient selectivity hampers precise analysis of the role of cathepsin E and napsin A in samples containing the ubiquitous and highly abundant homolog cathepsin D. Grassystatins from marine cyanobacteria show promising selectivity for cathepsin E but contain several ester bonds that make their synthesis cumbersome and thus limit availability of the inhibitors. Herewith, we present grassystatin-derived cathepsin E inhibitors with greatly facilitated synthesis but retained selectivity profile. We demonstrate their affinity and selectivity with both enzyme kinetic assays and streptavidin-based pull-down from cells and mouse organs. Our findings suggest that grassystatin-like inhibitors are useful tools for targeted inhibition of cathepsin E and thus provide a novel approach for cancer and immunology research.
Collapse
|
14
|
Nili F, Tavakoli M, Izadi Mood N, Saffar H, Sarmadi S. Napsin-A Expression, a Reliable Immunohistochemical Marker for Diagnosis of Ovarian and Endometrial Clear Cell Carcinomas. IRANIAN JOURNAL OF PATHOLOGY 2020; 15:81-85. [PMID: 32215023 PMCID: PMC7081762 DOI: 10.30699/ijp.2020.106598.2222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 02/23/2020] [Indexed: 11/10/2022]
Abstract
Background & Objective: Clear cell carcinomas (CCC) differ from other types of ovarian and endometrial carcinomas in biology, behavior and response to chemotherapy. Histopathologic diagnosis may be challenging in some situations which necessitates immunohistochemistary (IHC) assessment. In this study we investigated the diagnostic utility of Napsin-A in diagnosis of ovarian and endometrial CCCs. Methods: Ovarian and endometrial CCC samples from 2013 to 2018 in 3 general and women’s hospital in Tehran were re-evaluated by 2 expert pathologists. Forty-two samples were included as case and 42 non-clear cell carcinomas (Non-CCC) of ovary and endometrium were selected as control group. Based on IHC study tumors with sum intensity and percentage score ≥2 (at least 1+ staining in more than 1% of tumor cells) were considered positive. Results: The prevalence of endometrial and ovarian CCC in the case group were 15 and 27 respectively. The tumors in the control group included 22 cases of endometrioid, 2 high grade papillary serous carcinoma (HGSC) of endometrium, 6 endometrioid and 12 HGSC of ovary. Napsin-A positivity was observed in 35 (83%) of CCCs while 7 (17%) samples including 3 out of 15 endometrial and 4 out of 27 ovarian CCCs were Napsin-A negative. No positive reaction was seen in control group. The overall accuracy, specifity and sensitivity of Napsin-A for diagnosis of ovarian and endometrial CCCs were 83%, 100% and 83%, respectively. Sensitivity for ovarian and endometrial CCCs were 85% and 80%, orderly. Conclusion: Napsin-A is an accurate and reliable marker for distinction of CCCs from non-CCCs in ovary and endometrium. A panel of antibodies may yield the highest diagnostic accuracy.
Collapse
Affiliation(s)
- Fatemeh Nili
- Department of Pathology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mansoureh Tavakoli
- Department of Pathology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Narges Izadi Mood
- Department of Pathology, Mohebb-e-Yas Women Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Hana Saffar
- Department of Pathology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheila Sarmadi
- Department of Pathology, Mohebb-e-Yas Women Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Maurer A, Kalbacher H. Pepstatin pull-down at high pH is a powerful tool for detection and analysis of napsin A. Biochem Biophys Res Commun 2019; 515:145-148. [PMID: 31130231 DOI: 10.1016/j.bbrc.2019.05.094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 05/13/2019] [Indexed: 10/26/2022]
Abstract
Napsin A is an intracellular aspartic protease and biomarker of various malignancies like lung adenocarcinoma and ovarian clear cell carcinoma, but its detection is usually limited to immunohistochemical techniques gaining excellent information on its distribution but missing information about posttranslational modifications (e.g. maturation state) of the protein. We present a protocol for specific enrichment of napsin A from clinical or biological specimens, that facilitates detailed analysis of the protein. By using the exceptionally broad pH range under which napsin A binds to its inhibitor pepstatin A we achieve highly selective binding of napsin A while other aspartic proteases have negligible affinity. Using this method we demonstrate that lung napsin A in many mammals is a heterogeneous enzyme with a characteristic ladder-like appearance in SDS-PAGE that might be caused by proteolytically processed N- and/or C-termini, in contrast to the more homogeneous form found in kidneys and primary lung adenocarcinoma.
Collapse
Affiliation(s)
- Andreas Maurer
- Interfaculty Institute of Biochemistry, Eberhard Karls University Tübingen, Germany.
| | - Hubert Kalbacher
- Interfaculty Institute of Biochemistry, Eberhard Karls University Tübingen, Germany
| |
Collapse
|
16
|
Napsin A, Hepatocyte Nuclear Factor-1-Beta (HNF-1β), Estrogen and Progesterone Receptors Expression in Arias-Stella Reaction. Am J Surg Pathol 2019; 43:325-333. [DOI: 10.1097/pas.0000000000001212] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
17
|
Yatabe Y, Dacic S, Borczuk AC, Warth A, Russell PA, Lantuejoul S, Beasley MB, Thunnissen E, Pelosi G, Rekhtman N, Bubendorf L, Mino-Kenudson M, Yoshida A, Geisinger KR, Noguchi M, Chirieac LR, Bolting J, Chung JH, Chou TY, Chen G, Poleri C, Lopez-Rios F, Papotti M, Sholl LM, Roden AC, Travis WD, Hirsch FR, Kerr KM, Tsao MS, Nicholson AG, Wistuba I, Moreira AL. Best Practices Recommendations for Diagnostic Immunohistochemistry in Lung Cancer. J Thorac Oncol 2019; 14:377-407. [PMID: 30572031 PMCID: PMC6422775 DOI: 10.1016/j.jtho.2018.12.005] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 01/04/2023]
Abstract
Since the 2015 WHO classification was introduced into clinical practice, immunohistochemistry (IHC) has figured prominently in lung cancer diagnosis. In addition to distinction of small cell versus non-small cell carcinoma, patients' treatment of choice is directly linked to histologic subtypes of non-small cell carcinoma, which pertains to IHC results, particularly for poorly differentiated tumors. The use of IHC has improved diagnostic accuracy in the classification of lung carcinoma, but the interpretation of IHC results remains challenging in some instances. Also, pathologists must be aware of many interpretation pitfalls, and the use of IHC should be efficient to spare the tissue for molecular testing. The International Association for the Study of Lung Cancer Pathology Committee received questions on practical application and interpretation of IHC in lung cancer diagnosis. After discussions in several International Association for the Study of Lung Cancer Pathology Committee meetings, the issues and caveats were summarized in terms of 11 key questions covering common and important diagnostic situations in a daily clinical practice with some relevant challenging queries. The questions cover topics such as the best IHC markers for distinguishing NSCLC subtypes, differences in thyroid transcription factor 1 clones, and the utility of IHC in diagnosing uncommon subtypes of lung cancer and distinguishing primary from metastatic tumors. This article provides answers and explanations for the key questions about the use of IHC in diagnosis of lung carcinoma, representing viewpoints of experts in thoracic pathology that should assist the community in the appropriate use of IHC in diagnostic pathology.
Collapse
Affiliation(s)
- Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan.
| | - Sanja Dacic
- Department of Pathology University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alain C Borczuk
- Department of Pathology, Weill Cornell Medicine, New York, New York
| | - Arne Warth
- Institute of Pathology, Cytopathology, and Molecular Pathology MVZ UEGP Giessen, Wetzlar, Limburg, Germany
| | - Prudence A Russell
- Anatomical Pathology Department, St. Vincent's Hospital and the University of Melbourne, Fitzroy, Victoria, Australia
| | - Sylvie Lantuejoul
- Department of Biopathology, Centre Léon Bérard, Grenoble Alpes University, Lyon, France
| | - Mary Beth Beasley
- Department of Pathology, Mount Sinai Medical Center, New York, New York
| | - Erik Thunnissen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan and IRCCS MultiMedica, Milan, Italy
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Akihiko Yoshida
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Kim R Geisinger
- Department of Pathology, The University of Mississippi Medical Center, Jackson, Mississippi
| | - Masayuki Noguchi
- Department of Pathology, Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba, Japan
| | - Lucian R Chirieac
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Johan Bolting
- Department of Immunology Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jin-Haeng Chung
- Department of Pathology and Respiratory Center, Seoul National University Bundang Hospital, Seongnam city, Gyeonggi- do, Republic of Korea
| | - Teh-Ying Chou
- Division of Molecular Pathology, Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Republic of China
| | - Gang Chen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Claudia Poleri
- Office of Pathology Consultants, Buenos Aires, Argentina
| | - Fernando Lopez-Rios
- Laboratorio de Dianas Terapeuticas, Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Mauro Papotti
- Department of Oncology, University of Turin, Turin, Italy
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Minnesota
| | - William D Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fred R Hirsch
- University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Keith M Kerr
- Department of Pathology, Aberdeen Royal Infirmary, Aberdeen University Medical School, Aberdeen, Scotland, United Kingdom
| | - Ming-Sound Tsao
- Department of Pathology, University Health Network/Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Andrew G Nicholson
- Department of Histopathology, Royal Brompton and Harefield National Health Service Foundation Trust and National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, M. D. Anderson Cancer Center, Houston, Texas
| | - Andre L Moreira
- Department of Pathology, New York University Langone Health, New York, New York
| |
Collapse
|
18
|
Abstract
The differential diagnosis of hepatic mass lesions is broad and arriving at the right diagnosis can be challenging, especially on needle biopsies. The differential diagnosis of liver tumors in children is different from adults and is beyond the scope of this review. In adults, the approach varies depending on the age, gender, and presence of background liver disease. The lesions can be divided broadly into primary and metastatic (secondary), and the primary lesions can be further divided into those of hepatocellular origin and nonhepatocellular origin. The first category consists of benign and malignant lesions arising from hepatocytes, while the second category includes biliary, mesenchymal, hematopoietic, and vascular tumors. Discussion of nonepithelial neoplasms is beyond the scope of this review. The hepatocytic lesions comprise dysplastic nodules, focal nodular hyperplasia, hepatic adenoma, and hepatocellular carcinoma, and the differential diagnosis can be challenging requiring clinicopathological correlation and application of immunohistochemical (IHC) markers. Liver is a common site for metastasis, sometimes presenting with an unknown primary site, and proper workup is the key to arriving at the correct diagnosis. The correct diagnosis in this setting requires a systematic approach with attention to histologic features, imaging findings, clinical presentation, and judicious use of IHC markers. The list of antibodies that can be used for this purpose keeps on growing continually. It is important for pathologists to be up to date with the sensitivity and specificity of these markers and their diagnostic role and clinical implications. The purpose of this review is to outline the differential diagnosis of hepatic masses in adults and discuss an algorithmic approach to make a right diagnosis.
Collapse
Affiliation(s)
- Monika Vyas
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Dhanpat Jain
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
19
|
Al-Maghrabi JA, Butt NS, Anfinan N, Sait K, Sait H, Marzouki A, Khabaz MN. Infrequent Immunohistochemical Expression of Napsin A in Endometrial Carcinomas. Appl Immunohistochem Mol Morphol 2017; 25:632-638. [DOI: 10.1097/pai.0000000000000350] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
20
|
Viola D, Giani C, Mazzeo S, Ugolini C, Ciampi R, Molinaro E, Agate L, Borrelli N, Chella A, Fontanini G, Basolo F, Elisei R. KIF5B/RET Rearrangement in a Carcinoma of the Thyroid Gland: A Case Report of a Fatal Disease. J Clin Endocrinol Metab 2017; 102:3091-3096. [PMID: 28911147 DOI: 10.1210/jc.2017-00304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 07/10/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND The diffuse sclerosing variant of papillary thyroid cancer (DSV-PTC) is a rare variant of papillary thyroid cancer (PTC) with different clinicopathological features compared with conventional PTC. CASE An advanced DSV-PTC was diagnosed in a 39-year-old man. The radioiodine posttherapeutic whole-body-scan showed only an uptake in the central neck, whereas the computerized tomography showed multiple latero-cervical and mediastinum lymph node metastases, a single and spiculated lung lesion and multiple bilateral cerebellum metastases. The patient died after 6 months from the initial diagnosis. The histological revision of the thyroid tumor confirmed the diagnosis of DSV-PTC, and its molecular analysis revealed a KIF5B/RET rearrangement that, until now, was described only in a minority of lung adenocarcinoma. Other 18 cases of DSV-PTC were then studied for the presence of KIF5B/RET rearrangement, but all of them were negative. CONCLUSIONS This was a case of DSV-PTC positive for KIF5B/RET rearrangement, but considering that this alteration has been described only in lung adenocarcinoma and that the clinical course was more typical of lung carcinoma, we cannot completely rule out the possibility that this was a metastatic lesion from a lung tumor mimicking a DSV-PTC. As an alternative, we can also hypothesize that this was a case of fusion of two tumoral tissues deriving from a DSV-PTC and a metastasis of a KIF5B/RET positive lung adenocarcinoma. The question of whether the molecular findings, particularly when specifically reported only in some subtypes of human tumors, can overcome the morphological diagnosis is a matter of discussion.
Collapse
Affiliation(s)
- David Viola
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56124 Pisa, Italy
| | - Carlotta Giani
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56124 Pisa, Italy
| | - Salvatore Mazzeo
- Diagnostic and Interventional Radiology, Department of Translational Research and New Technologies in Medicine and Surgery, University Hospital of Pisa, 56124 Pisa, Italy
| | - Clara Ugolini
- Pathology Unit, Department of Surgical, Medical, Molecular Pathology and Critical Area, University Hospital of Pisa, 56124 Pisa, Italy
| | - Raffaele Ciampi
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56124 Pisa, Italy
| | - Eleonora Molinaro
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56124 Pisa, Italy
| | - Laura Agate
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56124 Pisa, Italy
| | - Nicla Borrelli
- Pathology Unit, Department of Surgical, Medical, Molecular Pathology and Critical Area, University Hospital of Pisa, 56124 Pisa, Italy
| | - Antonio Chella
- Unit of Pneumology, University Hospital of Pisa, 56124 Pisa, Italy
| | - Gabriella Fontanini
- Pathology Unit, Department of Surgical, Medical, Molecular Pathology and Critical Area, University Hospital of Pisa, 56124 Pisa, Italy
| | - Fulvio Basolo
- Pathology Unit, Department of Surgical, Medical, Molecular Pathology and Critical Area, University Hospital of Pisa, 56124 Pisa, Italy
| | - Rossella Elisei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56124 Pisa, Italy
| |
Collapse
|
21
|
Woo JS, Reddy OL, Koo M, Xiong Y, Li F, Xu H. Application of Immunohistochemistry in the Diagnosis of Pulmonary and Pleural Neoplasms. Arch Pathol Lab Med 2017. [PMID: 28644685 DOI: 10.5858/arpa.2016-0550-ra] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - A vast majority of neoplasms arising from lung or pleura are initially diagnosed based on the histologic evaluation of small transbronchial, endobronchial, or needle core biopsies. Although most diagnoses can be determined by morphology alone, immunohistochemistry can be a valuable diagnostic tool in the workup of problematic cases. OBJECTIVE - To provide a practical approach in the interpretation and immunohistochemical selection of lung/pleura-based neoplasms obtained from small biopsy samples. DATA SOURCES - A literature review of previously published articles and the personal experience of the authors were used in this review article. CONCLUSION - Immunohistochemistry is a useful diagnostic tool in the workup of small biopsies from the lung and pleura sampled by small biopsy techniques.
Collapse
|
22
|
Genova SN, Belovezhdov VT, Bichev SN, Danev VH. Comparative Characteristics of Napsin A, TTF 1 and EGFR Mutation Expression in Mucinous Lung Cell Carcinomas. Folia Med (Plovdiv) 2017; 59:174-182. [PMID: 28704186 DOI: 10.1515/folmed-2017-0020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 12/13/2016] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Invasive mucinous lung adenocarcinomas are rare and account for 2%-10% of all lung adenocarcinoma cases. It is believed that Napsin A exhibits a weaker expression in mucinous adenocarcinomas compared with TTF1, but such correlation is still poorly researched. AIM The aim of the study was to determine the frequency of mucinous to nonmucinous adenocarcinomas and compare specificity and sensitivity of monoclonal Napsin A with TTF1 in mucinous adenocarcinomas and define the frequency of EGFR mutations. MATERIALS AND METHODS Eighty-four resected lung carcinomas were prospectively evaluated. All biopsies were analysed with p63, TTF1, monoclonal Napsin A, CK7, CK20 and CDX2 and were studied with real-time PCR technology. RESULTS In resected material we detected 49/84 (58.3%) adenocarcinomas and selected 21 mucinous adenocarcinomas out of 46 non-mucinous adenocarcinomas (45.6%). The most common pattern of mucinous adenocarcinomas is papillary - 24% and colloidal - 24%, followed by acinar - 19.2% and lepidic - 19.2%. mNapsin A was positive in 18/21 (85.7%) mucinous adenocarcinomas v/s 17/21 TTF1 positive (80.9%). EGFR mutations were detected in 3/21 cases with mucinous adenocarcinomas (14.3%): mucinous papillary, mucinous acinar and "salivary gland-like". CONCLUSION Our study demonstrates a high proportion of primary mucinous lung adenocarcinomas to primary non-mucinous adenocarcinomas. Sensitivity and specificity of mNapsin A and TTF1 did not show significant difference in pulmonary mucinous and non-mucinous adenocarcinomas, as mNapsin A gave greater sensitivity to mucinous adenocarcinomas. Our results indicate the same mutation frequency of EGFR in mucinous adenocarcinomas as mutation frequency detected in non-mucinous adenocarcinomas in the Bulgarian region.
Collapse
Affiliation(s)
- Sylvia N Genova
- Department of General and Clinical Pathology and Forensic Medicine, Faculty of Medicine, Medical University of Plovdiv,
Plovdiv, Bulgaria
| | - Veselin T Belovezhdov
- Department of General and Clinical Pathology and Forensic Medicine, Faculty of Medicine, Medical University of Plovdiv,
Plovdiv, Bulgaria
| | - Stoyan N Bichev
- National Genetics Laboratory, University Hospital of Obstetrics and Gynecology, SBALAG “Maichin dom”, Sofi a, Bulgaria
| | - Vladimir H Danev
- Department of General and Clinical Pathology and Forensic Medicine, Faculty of Medicine, Medical University of Plovdiv,
Plovdiv, Bulgaria
| |
Collapse
|
23
|
Role of minimal panel immunostaining in accurate diagnosis of lung cancer using small biopsies. EGYPTIAN JOURNAL OF CHEST DISEASES AND TUBERCULOSIS 2017. [DOI: 10.1016/j.ejcdt.2016.10.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
24
|
Peat TJ, Edmondson EF, Miller MA, DuSold DM, Ramos-Vara JA. Pax8, Napsin A, and CD10 as Immunohistochemical Markers of Canine Renal Cell Carcinoma. Vet Pathol 2017; 54:588-594. [PMID: 28346124 DOI: 10.1177/0300985817698211] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Pax8, napsin A, and CD10 are useful immunohistochemical markers of human renal cell carcinoma (RCC); however, their diagnostic utility in canine RCC is unclear. Forty formalin-fixed paraffin-embedded renal cell carcinomas from dogs (15 papillary, 12 solid, and 13 tubular) and 10 metastases were evaluated for expression of Pax8, napsin A, and CD10. Thirty-nine (98%), 24 (60%), and 19 (50%) tumors expressed Pax8 (nuclear labeling), napsin A (cytoplasmic labeling), and CD10 (cytoplasmic and membranous labeling), respectively. Pax8 was expressed in 92% of solid, 100% of papillary, and 100% of tubular tumors. Napsin A was expressed in 58% of solid, 60% of papillary, and 62% of tubular RCC. CD10 was expressed in 33% of solid, 47% of papillary, and 62% of tubular RCC. Pax8 was expressed in 80% of the metastatic tumors, napsin A in 60%, and CD10 in 50%. Additionally, Pax8 immunoreactivity was stronger overall than that of napsin A or CD10. In summary, Pax8 is a more sensitive marker than napsin A or CD10 for primary and metastatic canine RCC; its nuclear and more intense reactivity also makes it easier to interpret. Tubular and papillary RCCs were more likely than solid RCC to express all 3 markers. These findings highlight the utility of Pax8 as an immunohistochemical marker in diagnosing all major subtypes of canine primary and metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Tyler J. Peat
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Elijah F. Edmondson
- NCI, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Margaret A. Miller
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Dee M. DuSold
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - José A. Ramos-Vara
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
25
|
Bir F, Çeliker D, Evyapan BF, Yaren A, Edirne T. New immunohistochemical markers in the differential diagnosisof nonsmall cell lung carcinoma. Turk J Med Sci 2016; 46:1854-1861. [PMID: 28081338 DOI: 10.3906/sag-1501-68] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/27/2016] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND/AIM The aim of this study was to investigate Napsin-A, NTRK-1, NTRK-2, Desmoglein-3, and Desmocollin-3 in the differential diagnosis and prognosis of nonsmall cell lung cancer. MATERIALS AND METHODS The expression of Napsin-A, NTRK-1, NTRK-2, and Desmoglein-3 was examined by immunohistochemistry in 50 squamous cell carcinomas and 50 adenocarcinomas. Desmocollin-3 was investigated in 29 squamous cell carcinoma and 29 adenocarcinoma cases. Associations between expression profiles of Napsin-A, NTRK-1, NTRK-2, Desmoglein-3, and Desmocollin-3 in lung cancers and clinicopathological variables were analyzed. RESULTS Napsin-A staining was statistically significant in detecting adenocarcinomas versus squamous cell carcinomas. The sensitivity of Napsin-A for adenocarcinomas was 96% and the specificity was 100%. NTRK-2 and Desmocollin-3 staining were statistically significant in detecting squamous cell carcinomas versus adenocarcinomas. Desmoglein-3, Napsin-A, and NTRK-2 had no effect on survival. Disease-free survival time was significantly shorter in cases that were moderately positive with NTRK-1. CONCLUSION Our data suggest that Napsin-A, NTRK-2, and Desmocollin-3 are useful markers in the differentiation of nonsmall cell lung cancer.
Collapse
Affiliation(s)
- Ferda Bir
- Department of Pathology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Duygu Çeliker
- Department of Pathology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Binnaz Fatma Evyapan
- Department of Chest Diseases, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Arzu Yaren
- Department of Medical Oncology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Tamer Edirne
- Department of Family Medicine, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| |
Collapse
|
26
|
Ramos-Vara J, Frank C, DuSold D, Miller M. Immunohistochemical Detection of Pax8 and Napsin A in Canine Thyroid Tumours: Comparison with Thyroglobulin, Calcitonin and Thyroid Transcription Factor 1. J Comp Pathol 2016; 155:286-298. [DOI: 10.1016/j.jcpa.2016.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 07/06/2016] [Accepted: 07/18/2016] [Indexed: 12/21/2022]
|
27
|
Shi J, Liu H, Ma XJ, Chen Z, He MX, Luo Y, Lin F. Ribonucleic Acid In Situ Hybridization Is a More Sensitive Method Than Immunohistochemistry in Detection of Thyroid Transcription Factor 1 and Napsin A Expression in Lung Adenocarcinomas. Arch Pathol Lab Med 2016; 140:332-40. [PMID: 27028392 DOI: 10.5858/arpa.2014-0644-oa] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT TTF-1 and napsin A immunomarkers have a crucial role in differentiating lung adenocarcinoma from lung squamous cell carcinoma and in identifying a primary lung adenocarcinoma when working on a tumor of unknown origin. OBJECTIVES To investigate the diagnostic sensitivity of ribonucleic acid in situ hybridization (RNAscope) in the detection of expression of these biomarkers in lung adenocarcinomas and to compare RNAscope to immunohistochemical techniques. DESIGN Both RNAscope and the immunohistochemical assays for TTF-1 and napsin A were performed on tissue microarray sections containing 80 lung adenocarcinomas and 80 lung squamous cell carcinomas. The RNAscope assay for both TTF-1 and napsin A was also performed on 220 adenocarcinomas from various organs. RESULTS The RNAscope assay for TTF-1 gave positive results in 92.5% (74 of 80) of the lung adenocarcinomas; in contrast, immunohistochemistry gave positive results in 82.5% (66 of 80) of those cases. The RNAscope assay for napsin A gave positive results in 90% (72 of 80) of lung adenocarcinomas; immunohistochemistry results were positive in 77.5% (62 of 80) of those cases. Napsin A expression was not seen in lung squamous cell carcinomas by either method. In contrast, TTF-1 expression was seen in 3.8% (3 of 80) (1(+)) and 10% (8 of 80) (1(+)) of the squamous cell carcinomas by immunochemistry and the RNAscope, respectively. All nonpulmonary adenocarcinoma results were negative for TTF-1 by the RNAscope assay. CONCLUSIONS Preliminary data suggest that RNAscope is superior to immunohistochemistry in detecting TTF-1 and napsin A expression in primary lung adenocarcinomas. Therefore, performing an RNAscope assay may be considered for both TTF-1(-) and napsin A(-) cases with a clinical suspicion of lung adenocarcinoma. The TTF-1 results should be interpreted with caution because a small percentage of squamous cell carcinomas can be focally positive by either assay.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fan Lin
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania (Drs Shi, Liu, Chen, and Lin); and the Department of Research and Development, Advanced Cell Diagnostics, Inc, Hayward, California (Drs Ma, He, and Luo)
| |
Collapse
|
28
|
Immunohistochemical Comparison of Ovarian and Uterine Endometrioid Carcinoma, Endometrioid Carcinoma With Clear Cell Change, and Clear Cell Carcinoma. Am J Surg Pathol 2015; 39:1061-9. [PMID: 25871622 DOI: 10.1097/pas.0000000000000436] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Accurate distinction of clear cell carcinoma (CCC) from endometrioid carcinoma (EC) has important clinical implications, but, not infrequently, EC demonstrates clear cell change (EC-CC), mimicking CCC. We examined whether a panel of immunomarkers can help distinguish between these tumors. Sixty-four CCCs (40 ovarian and 24 uterine), 34 ECs (21 ovarian and 13 uterine), and 34 EC-CCs (6 ovarian and 28 uterine) were stained for HNF1β, BAF250a, Napsin A, ER, and PR. Intensity and extent of immunoreactivity was assessed. Fifty-seven of 64 (89%) CCCs, 14/34 (41%) EC-CCs, and 16/34 (47%) ECs expressed HNF1β, and 56/64 (88%) CCCs, 4/34 (12%) EC-CCs, and 1/34 (3%) ECs stained for Napsin A. Most CCCs demonstrated at least moderate and diffuse staining for both markers, whereas only focal and weak expression was identified in most EC-CC/EC. Compared to HNF1β, Napsin A showed increased specificity (93.0% vs. 55.9%, P<0.0001) and similar sensitivity (87.5% vs. 89.1%) in distinguishing CCC from EC-CC/EC. Thirteen of 64 (20%) CCCs, 6/34 (18%) EC-CCs, and 2/34 (6%) ECs showed loss of BAF250a. ER was expressed by 10/64 (16%) CCCs, 30/34 (88%) EC-CCs, and 33/34 (97%) ECs, whereas PR positivity was identified in 9/64 (14%) CCCs, 26/34 (77%) EC-CCs, and 33/34 (97%) ECs. The majority of EC and EC-CC demonstrated diffuse staining for ER/PR, whereas most CCCs showed very focal positivity. There is a statistically significant difference in HNF1β, Napsin A, ER, and PR immunoexpression between CCC and EC/EC-CC, with Napsin A being a more specific marker for CCC than HNF1β. Overall, the immunoprofile of EC-CC is more comparable to that of EC than CCC. The use of a panel of immunostains can help distinguish EC-CC from CCC.
Collapse
|
29
|
Qu Y, Zhao D, Mu J, Che N, Zhang C, Liu Z, Su D, Zhou L, Zhang H, Wei L. Prognostic analysis of primary mucin-producing adenocarcinoma of the lung: a comprehensive retrospective study. Tumour Biol 2015; 37:887-96. [PMID: 26254613 DOI: 10.1007/s13277-015-3869-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/30/2015] [Indexed: 01/15/2023] Open
Abstract
Although primary mucin-producing adenocarcinoma of the lung is uncommon, each subtype has distinct clinical, pathological, molecular, and prognostic characteristics. This study aimed to determine correlations between clinical and pathological features and genetic phenotypes with the prognosis. We immunohistochemically examined the protein levels of thyroid transcription factor 1 (TTF-1), Napsin A, and anaplastic lymphoma kinase (ALK) and genetically examined epidermal growth factor receptor (EGFR) and KRAS mutations in these mucin-producing tumors. A total of 75 cases of mucin-producing adenocarcinoma of the lung were examined. ALK protein positivity was 33.3 % (25/75), and primarily occurred in solid predominant with mucin production subtype (SA). KRAS mutations occurred in 22.7 % (17/75) of patients, predominantly in invasive mucinous adenocarcinoma (IMA). Positive TTF-1 and Napsin A expression was more common in SA, while they were both negative in IMA. The 1-, 3-, and 5-year progression-free survival rates of mucin-producing lung adenocarcinoma were 85, 64, and 38 %, respectively; the overall survival rates were 90, 67, and 50 %, respectively. Larger tumors, advanced stage, and lymph node metastasis were associated with poor prognosis. Mucinous minimally invasive adenocarcinoma (m-MIA) had the best prognosis, followed by IMA, SA, and acinar or papillary predominant adenocarcinoma with mucin production (A/P). KRAS mutations were an independent positive prognostic factor for postoperative progress.
Collapse
Affiliation(s)
- Yang Qu
- Department of Pathology, The General Hospital of People's Liberation Army, 28# Fuxing Road, Haidian District, Beijing, 100853, China
- Department of pathology, Beijing Chest Hospital, Capital Medical University, 97# Machang, Tongzhou District, Beijing, 101149, China
| | - Dan Zhao
- Department of pathology, Beijing Chest Hospital, Capital Medical University, 97# Machang, Tongzhou District, Beijing, 101149, China
| | - Jing Mu
- Department of pathology, Beijing Chest Hospital, Capital Medical University, 97# Machang, Tongzhou District, Beijing, 101149, China
| | - Nanying Che
- Department of pathology, Beijing Chest Hospital, Capital Medical University, 97# Machang, Tongzhou District, Beijing, 101149, China
| | - Chen Zhang
- Department of pathology, Beijing Chest Hospital, Capital Medical University, 97# Machang, Tongzhou District, Beijing, 101149, China
| | - Zichen Liu
- Department of pathology, Beijing Chest Hospital, Capital Medical University, 97# Machang, Tongzhou District, Beijing, 101149, China
| | - Dan Su
- Department of pathology, Beijing Chest Hospital, Capital Medical University, 97# Machang, Tongzhou District, Beijing, 101149, China
| | - Lijuan Zhou
- Department of pathology, Beijing Chest Hospital, Capital Medical University, 97# Machang, Tongzhou District, Beijing, 101149, China
| | - Haiqing Zhang
- Department of pathology, Beijing Chest Hospital, Capital Medical University, 97# Machang, Tongzhou District, Beijing, 101149, China.
| | - Lixin Wei
- Department of Pathology, The General Hospital of People's Liberation Army, 28# Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
30
|
Ma Y, Fan M, Dai L, Kang X, Liu Y, Sun Y, Yan W, Liang Z, Xiong H, Chen K. The expression of TTF-1 and Napsin A in early-stage lung adenocarcinoma correlates with the results of surgical treatment. Tumour Biol 2015; 36:8085-92. [PMID: 25982999 DOI: 10.1007/s13277-015-3478-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 04/20/2015] [Indexed: 01/15/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for 80 % of lung cancers, and lung adenocarcinoma (ADC) is one of the main types of NSCLC. Although there are several studies on the relationship between lung ADC immunohistochemical diagnostic markers (thyroid transcription factor 1 (TTF-1) and Napsin A) and survival, some aspects of those studies could be improved. We examined the significance of the commonly used lung ADC diagnostic markers, including TTF-1, Napsin A, and CK7, in the prognosis of early-stage lung ADC. One hundred and nineteen cases of early-stage lung ADC (N0) were selected from the prospective database of lung cancer (Jan 2000 to Dec 2009). The expression levels of TTF-1, Napsin A, and CK7 in inventoried specimens were analyzed using tissue microarray (TMA) and immunohistochemical (IHC) analysis, and the effect of the expression level of each marker on patients' survival was examined. The diagnostic sensitivity and specificity of each marker for lung ADC were as follows: TTF-1, 87.0 and 90.1 %; Napsin A, 72.2 and 90.4 %; and CK7, 94.6 and 76.0 %, respectively. Patients with high expression levels of TTF-1 and Napsin A, and high co-expression levels of TTF-1/Napsin A had better survival rates than those with low levels of expression (P < 0.05). The expression levels of CK7 were not related to patients' survival. Multivariate analysis showed that the expression levels of Napsin A and TTF-1/Napsin A are independent prognostic factors for survival. The IHC detection of TTF-1 and Napsin A in specimens should be routinely performed in postoperative early-stage lung ADC patients. Its significance lies not only in the differential diagnosis, but also in determining the prognosis.
Collapse
Affiliation(s)
- Yunfan Ma
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital & Institute, 52 Fucheng Rd. Haidian District, Beijing, China, 100142
| | - Mengying Fan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital & Institute, 52 Fucheng Rd. Haidian District, Beijing, China, 100142
| | - Liang Dai
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital & Institute, 52 Fucheng Rd. Haidian District, Beijing, China, 100142
| | - Xiaozheng Kang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital & Institute, 52 Fucheng Rd. Haidian District, Beijing, China, 100142
| | - Yiqiang Liu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yu Sun
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Wanpu Yan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital & Institute, 52 Fucheng Rd. Haidian District, Beijing, China, 100142
| | - Zhen Liang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital & Institute, 52 Fucheng Rd. Haidian District, Beijing, China, 100142
| | - Hongchao Xiong
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital & Institute, 52 Fucheng Rd. Haidian District, Beijing, China, 100142
| | - Keneng Chen
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital & Institute, 52 Fucheng Rd. Haidian District, Beijing, China, 100142.
| |
Collapse
|
31
|
Lin F, Liu H. Immunohistochemistry in undifferentiated neoplasm/tumor of uncertain origin. Arch Pathol Lab Med 2015; 138:1583-610. [PMID: 25427040 DOI: 10.5858/arpa.2014-0061-ra] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT Immunohistochemistry has become an indispensable ancillary study in the identification and classification of undifferentiated neoplasms/tumors of uncertain origin. The diagnostic accuracy has significantly improved because of the continuous discoveries of tissue-specific biomarkers and the development of effective immunohistochemical panels. OBJECTIVES To identify and classify undifferentiated neoplasms/tumors of uncertain origin by immunohistochemistry. DATA SOURCES Literature review and authors' research data and personal practice experience were used. CONCLUSIONS To better guide therapeutic decisions and predict prognostic outcomes, it is crucial to differentiate the specific lineage of an undifferentiated neoplasm. Application of appropriate immunohistochemical panels enables the accurate classification of most undifferentiated neoplasms. Knowing the utilities and pitfalls of each tissue-specific biomarker is essential for avoiding potential diagnostic errors because an absolutely tissue-specific biomarker is exceptionally rare. We review frequently used tissue-specific biomarkers, provide effective panels, and recommend diagnostic algorithms as a standard approach to undifferentiated neoplasms.
Collapse
Affiliation(s)
- Fan Lin
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania
| | | |
Collapse
|
32
|
Rekhtman N, Kazi S. Nonspecific Reactivity of Polyclonal Napsin A Antibody in Mucinous Adenocarcinomas of Various Sites: A Word of Caution. Arch Pathol Lab Med 2014; 139:434-6. [DOI: 10.5858/arpa.2014-0452-le] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering, Cancer Center, New York, NY 10065
| | - Sofia Kazi
- Department of Pathology, Memorial Sloan Kettering, Cancer Center, New York, NY 10065
| |
Collapse
|
33
|
In search of the ideal immunopanel to distinguish metastatic mammary carcinoma from primary lung carcinoma: a tissue microarray study of 207 cases. Appl Immunohistochem Mol Morphol 2014; 22:266-74. [PMID: 23958551 DOI: 10.1097/pai.0b013e318297cc0b] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Distinguishing metastatic carcinoma of breast origin (MCBO) to lung from primary lung carcinomas (PLC) is a diagnostic quandary with clinical ramifications. Immunostains CK7, CK20, ER, PR, and Mammaglobin as well as pertinent negative stains are utilized but prove insufficient. We set out to identify stains either alone or as a group that would better discern between these 2 entities. DESIGN Tissue microarrays of 109 MCBO to lung and 102 PLC were stained with CK7, CK20, ER, PR, AR, Mammaglobin, Napsin A, GATA-3, and TTF-1. An H-score was calculated for each case and stain. RESULTS The highest area under the receiver-operating characteristic curves for each stain was seen with GATA-3 (0.817), Napsin A (0.817), and TTF-1 (0.854). When all possible combinations were analyzed, GATA-3 and TTF-1 proved to correctly classify with the highest accuracy (0.934). Combinations of GATA-3 and Napsin A (0.920) and GATA-3, Napsin A, and TTF-1 (0.933) were not significantly different from GATA-3 and TTF-1. The odds ratios for each stain and combination of stains showed that those for GATA-3 and TTF-1 were divergent, signifying that cases with higher H-scores for GATA-3 and TTF-1 were more likely to be classified as MCBO and PLC, respectively. CONCLUSIONS GATA-3 and TTF-1 can correctly classify a case as either MCBO or PLC in 93.4% of cases. Although highly specific and sensitive for PLC, Napsin A in lieu of TTF-1 or as an additional stain did not improve classification accuracy.
Collapse
|
34
|
Lin F, Chen Z. Standardization of Diagnostic Immunohistochemistry: Literature Review and Geisinger Experience. Arch Pathol Lab Med 2014; 138:1564-77. [DOI: 10.5858/arpa.2014-0074-ra] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context
Immunohistochemistry has become an indispensable ancillary technique in anatomic pathology laboratories. Standardization of every step in preanalytic, analytic, and postanalytic phases is crucial to achieve reproducible and reliable immunohistochemistry test results.
Objective
To standardize immunohistochemistry tests from preanalytic, analytic, to postanalytic phases.
Data Sources
Literature review and Geisinger (Geisinger Medical Center, Danville, Pennsylvania) experience.
Conclusions
This review article delineates some critical points in preanalytic, analytic, and postanalytic phases; reiterates some important questions, which may or may not have a consensus at this time; and updates the newly proposed guidelines on antibody validation from the College of American Pathologists Pathology and Laboratory Quality Center. Additionally, the article intends to share Geisinger's experience with (1) testing/optimizing a new antibody and troubleshooting; (2) interpreting and reporting immunohistochemistry assay results; (3) improving and implementing a total immunohistochemistry quality management program; and (4) developing best practices in immunohistochemistry.
Collapse
Affiliation(s)
- Fan Lin
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania
| | - Zongming Chen
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania
| |
Collapse
|
35
|
Falchook GS, Ordóñez NG, Bastida CC, Stephens PJ, Miller VA, Gaido L, Jackson T, Karp DD. Effect of the RET Inhibitor Vandetanib in a Patient With RET Fusion-Positive Metastatic Non-Small-Cell Lung Cancer. J Clin Oncol 2014; 34:e141-4. [PMID: 25366691 DOI: 10.1200/jco.2013.50.5016] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
| | | | | | | | | | - Lindsay Gaido
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tiffiny Jackson
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Daniel D Karp
- The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
36
|
Heymann JJ, Hoda RS, Scognamiglio T. Polyclonal Napsin A Expression: A Potential Diagnostic Pitfall in Distinguishing Primary From Metastatic Mucinous Tumors in the Lung. Arch Pathol Lab Med 2014; 138:1067-71. [DOI: 10.5858/arpa.2013-0403-oa] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context.—Napsin A is a useful marker for distinguishing primary from metastatic lung tumors. Mucinous lung tumors may be difficult to distinguish from metastatic mucinous tumors.
Objectives.—To evaluate napsin A expression in lung and extrapulmonary mucinous tumors on both histology and cytology specimens and to determine napsin A's utility in differentiating primary from metastatic mucinous tumors.
Design.—Napsin A immunohistochemistry was performed using a rabbit polyclonal antibody on formalin-fixed, paraffin-embedded surgical and fine-needle aspiration biopsy–derived, paraffin-embedded cell block specimens. Positive expression was defined as coarse, granular, cytoplasmic staining in 10% or more of tumor cells.
Results.—Sixteen of 32 mucinous lung tumors (50%) and 16 of 33 extrapulmonary mucinous tumors (48%), including 15 of 18 of gastrointestinal origin (83%), expressed napsin A. Positivity was concordant between surgical and cell block specimens in 5 of 9 cases (56%). In 3 of 4 discordant cases, napsin A expression was detected on the surgical specimen but not the cell block. The cell block material in these cases was paucicellular.
Conclusions.—Napsin A shows decreased sensitivity and specificity for mucinous lung tumors and is unlikely to be reliable as a sole immunohistochemical marker of lung origin for such tumors (52% specificity in this study). The high frequency of napsin A expression in gastrointestinal mucinous tumors makes it particularly unreliable in distinguishing metastatic gastrointestinal from primary lung mucinous tumors. However, napsin A expression analysis may facilitate distinguishing mucinous tumors of pulmonary from those of nongastrointestinal origin. Interpretation of napsin A staining may be problematic in mucinous tumor specimens of low cellularity such as cell blocks.
Collapse
Affiliation(s)
- Jonas J. Heymann
- From the Department of Pathology & Laboratory Medicine, NewYork-Presbyterian Hospital–Weill Cornell Medical College, New York, New York
| | - Rana S. Hoda
- From the Department of Pathology & Laboratory Medicine, NewYork-Presbyterian Hospital–Weill Cornell Medical College, New York, New York
| | - Theresa Scognamiglio
- From the Department of Pathology & Laboratory Medicine, NewYork-Presbyterian Hospital–Weill Cornell Medical College, New York, New York
| |
Collapse
|
37
|
Ordóñez NG, Sahin AA. Diagnostic utility of immunohistochemistry in distinguishing between epithelioid pleural mesotheliomas and breast carcinomas: a comparative study. Hum Pathol 2014; 45:1529-40. [PMID: 24816068 DOI: 10.1016/j.humpath.2014.03.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/06/2014] [Accepted: 03/12/2014] [Indexed: 02/03/2023]
Abstract
Epithelioid mesotheliomas and breast carcinomas can present a variety of morphologic patterns. Because of this, breast carcinomas that metastasize to the pleura and lung may be confused with mesotheliomas. The aim of the present study is to compare the immunohistochemical markers currently available for the diagnosis of these 2 malignancies and to determine the best panel of markers that can be used to assist in discriminating between them. Sixty epithelioid mesotheliomas and 80 breast carcinomas (40 triple negative and 40 estrogen receptor positive) were investigated for expression of the positive mesothelioma markers calretinin, keratin 5/6, mesothelin, podoplanin, thrombomodulin, and WT1; the positive carcinoma marker claudin 4; and the breast-associated markers gross cystic disease fluid protein 15 (GCDFP-15), mammaglobin, and GATA3. All of the epithelioid mesotheliomas reacted for calretinin and keratin 5/6, 93% for WT1; 88% for podoplanin; 77% for thrombomodulin; 23% for GATA3; and 0% for claudin 4, GCDFP-15, and mammaglobin, respectively. Of the triple-negative breast carcinomas, 100% expressed claudin 4; 5%, keratin 5/6; 30%, GATA3; 18%, mammaglobin; 15%, GCDFP-15; 56%, mesothelin; 38%, calretinin; 18%, thrombomodulin; 5%, WT1; and 3%, podoplanin. Among the estrogen receptor-positive breast carcinomas, 100% were claudin 4 and GATA3 positive; 70% expressed GCDFP-15; 63%, mammaglobin; 13%, calretinin; 13%, thrombomodulin; 8%, WT1; 5%, keratin 5/6; 3%, mesothelin; and 0%, podoplanin. It is concluded that podoplanin and WT1 are the best positive mesothelioma markers for differentiating epithelioid mesotheliomas from breast carcinomas. An accurate differential diagnosis can be reached with the use of these two markers in combination with the breast-associated markers GCDFP-15, mammaglobin, and GATA3.
Collapse
Affiliation(s)
- Nelson G Ordóñez
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Aysegul A Sahin
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
38
|
Kim MY, Go H, Koh J, Lee K, Min HS, Kim MA, Jeon YK, Lee HS, Moon KC, Park SY, Kim WH, Chung DH. Napsin A is a useful marker for metastatic adenocarcinomas of pulmonary origin. Histopathology 2014; 65:195-206. [DOI: 10.1111/his.12383] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 01/27/2014] [Indexed: 11/29/2022]
Affiliation(s)
- Moon-Young Kim
- Department of Pathology; Seoul National University Hospital; Seoul South Korea
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| | - Heounjeong Go
- Department of Pathology; Seoul National University Hospital; Seoul South Korea
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
- Department of Pathology; Asan Medical Center; University of Ulsan College of Medicine; Seoul South Korea
| | - Jaemoon Koh
- Department of Pathology; Seoul National University Hospital; Seoul South Korea
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| | - Kyoungbun Lee
- Department of Pathology; Seoul National University Hospital; Seoul South Korea
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| | - Hye-Sook Min
- Department of Pathology; Seoul National University Hospital; Seoul South Korea
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| | - Min-A Kim
- Department of Pathology; Seoul National University Hospital; Seoul South Korea
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| | - Yoon Kyung Jeon
- Department of Pathology; Seoul National University Hospital; Seoul South Korea
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| | - Hye-Seung Lee
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
- Department of Pathology; Seoul National University Bundang Hospital; Seongnam Gyeonggi-do South Korea
| | - Kyung-Chul Moon
- Department of Pathology; Seoul National University Hospital; Seoul South Korea
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| | - So Yeon Park
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
- Department of Pathology; Seoul National University Bundang Hospital; Seongnam Gyeonggi-do South Korea
| | - Woo-Ho Kim
- Department of Pathology; Seoul National University Hospital; Seoul South Korea
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| | - Doo Hyun Chung
- Department of Pathology; Seoul National University Hospital; Seoul South Korea
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| |
Collapse
|
39
|
Rossi G, Cavazza A, Righi L, Sartori G, Bisagni A, Longo L, Pelosi G, Papotti M. Napsin-A, TTF-1, EGFR, and ALK Status Determination in Lung Primary and Metastatic Mucin-Producing Adenocarcinomas. Int J Surg Pathol 2014; 22:401-7. [PMID: 24651909 DOI: 10.1177/1066896914527609] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Pulmonary mucin-producing adenocarcinomas may be indistinguishable on conventional histology from a metastasis, as thyroid transcription factor-1 (TTF-1) expression often is lacking and KRAS mutations are widely present even in extrapulmonary sites. Few data have been reported on the diagnostic role of napsin-A and epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) gene alterations in this challenging differential diagnosis. Seventy-seven surgically resected cases, including 53 primary and 24 metastatic tumors from different sites, were evaluated for napsin-A, TTF-1, and ALK by immunohistochemistry and for EGFR mutations by direct sequencing. Overall, napsin-A expression in primary lung mucin-producing adenocarcinomas was 36% (8% mucinous, 17% colloid, 87.5% solid, and 100% signet ring cell) and TTF-1 expression reached an overall figure of 42% (12.5% mucinous, 33% colloid, 87.5% solid, and 100% signet ring cell). Metastatic mucinous adenocarcinomas did not react with napsin-A or with TTF-1. All primary and metastatic tumors lacked EGFR mutations, while a single case of signet ring cell lung adenocarcinoma showed ALK expression and rearrangement at fluorescent in situ hybridization analysis. Napsin-A has a lower sensitivity compared with TTF-1 in primary mucin-producing adenocarcinomas of the lung. However, both antibodies have an absolute specificity, being always negative in metastatic mucinous adenocarcinomas. EGFR mutations and ALK translocation or expression are exceedingly rare in mucin-producing adenocarcinomas of the lung, resulting unnecessary as diagnostic tool in this setting.
Collapse
Affiliation(s)
- Giulio Rossi
- Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Alberto Cavazza
- Azienda Arcispedale S. Maria Nuova/IRCCS, Reggio Emilia, Italy
| | - Luisella Righi
- University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Giuliana Sartori
- Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | | | | | - Giuseppe Pelosi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy University of Milan, Milan, Italy
| | - Mauro Papotti
- University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| |
Collapse
|
40
|
Diagnostic and Prognostic Role of Immunohistochemical Expression of Napsin-A Aspartic Peptidase in Clear Cell and Papillary Renal Cell Carcinoma. Appl Immunohistochem Mol Morphol 2014; 22:206-12. [DOI: 10.1097/pai.0b013e31828ef24e] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
|
42
|
Terra SBSP, Aubry MC, Yi ES, Boland JM. Immunohistochemical study of 36 cases of pulmonary sarcomatoid carcinoma--sensitivity of TTF-1 is superior to napsin. Hum Pathol 2013; 45:294-302. [PMID: 24331839 DOI: 10.1016/j.humpath.2013.09.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 09/06/2013] [Accepted: 09/11/2013] [Indexed: 10/25/2022]
Abstract
Immunohistochemistry is often used to distinguish pulmonary sarcomatoid carcinoma from morphologic mimics. Napsin-A is a pulmonary adenocarcinoma marker, but literature on expression in sarcomatoid carcinoma is limited. Thirty-six cases of sarcomatoid carcinoma were stained for napsin, TTF-1, Oscar, CAM5.2, AE1/AE3, desmin, SMA, S-100, CK5/6, calretinin, D2-40, and WT1. Patients were 24 men and 12 women (mean, 70 years; range, 46-93). There were 27 pleomorphic carcinomas, 5 spindle cell carcinomas, 3 carcinosarcomas, and 1 giant cell carcinoma. Cases were positive for at least 1 keratin: AE1/3 was positive in all 36 cases; Oscar, in 34 cases (94%); and CAM5.2, in 32 cases (89%, weaker/more focal). Napsin was positive in 14 cases (39%): 8 diffuse, 3 focal, and 3 rare cells. TTF-1 was positive in 22 cases (61%): 15 diffuse, 3 focal, and 4 rare cells. No cases were napsin positive and negative for TTF-1. Variable staining for mesothelial markers was observed, including positivity for calretinin (12 cases, 33%), WT1 (6 cases, 17%), D2-40 (5 cases, 14%), and CK5/6 (9 cases, 25%). Mesenchymal markers were also sometimes positive (usually focal), including S-100 (4 cases, 11%), desmin (4 cases, 11%), and SMA (7 cases, 19%, 1 diffuse). In conclusion, TTF-1 is more sensitive than napsin for detection of sarcomatoid carcinoma, and no cases were positive for napsin but negative for TTF-1. CAM5.2 is less sensitive than AE1/AE3 and Oscar. Use of a thoughtful immunohistochemical panel is important in the evaluation of sarcomatoid carcinoma because mesothelial and mesenchymal markers can be expressed.
Collapse
Affiliation(s)
- Simone B S P Terra
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Marie Christine Aubry
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Eunhee S Yi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jennifer M Boland
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
43
|
Skirnisdottir I, Bjersand K, Åkerud H, Seidal T. Napsin A as a marker of clear cell ovarian carcinoma. BMC Cancer 2013; 13:524. [PMID: 24191930 PMCID: PMC4228360 DOI: 10.1186/1471-2407-13-524] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 10/30/2013] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Clear cell carcinomas are aggressive tumors with a distinct biologic behaviour. In a genome-wide screening for genes involved in chemo-resistance, NAPA was over-expressed in cisplatin-resistant cells. The NAPA (protein) Napsin A was described to promote resistance to cisplatin by degradation of the tumor suppressor p53. METHODS Totally 131 patients were included in this study all in FIGO-stages I-II; 16 were clear cell tumors which were compared with 40 Type I tumors and 75 type II tumors according to the markers Napsin A, p21, p53 and p27 and some clinical features. For detection of the markers tissue microarrays and immunohistochemistry were used. RESULTS Positivity for Napsin A was detected in 12 (80%) out of the 15 clear cell tumors available for analysis compared with 3 (4%) out of the Type I and II tumors in one group (p<0.001). Differences in p21 status, p53 status, and p21+p53- status were striking when clear cell tumors were compared with Type I, Type II, and Type I and II tumors in one group, respectively. The p21+p53-status was associated to positive staining of Napsin A (p=0.0015) and clear cell morphology (p=0.0003). In two separate multivariate logistic regression analyses with Napsin A as endpoint both clear cell carcinoma with OR=153 (95% C.I. 21-1107); (p<001) and p21+p53- status with OR=5.36 (95% C.I. 1.6-17.5); (p=0.005) were independent predictive factors. ROC curves showed that AUC for Napsin A alone was 0.882, for p21+p53- it was 0.720 and for p21+p53-Napsin A+AUC was 0.795. Patients with clear cell tumors had lower (p=0.013) BMI than Type I patients and were younger (p=0.046) at diagnosis than Type II patients. Clear cell tumors had a higher frequency (p=0.039) of capsule rupture at surgery than Type I and II tumors. CONCLUSIONS Positivity of Napsin A in an epithelial ovarian tumor might strengthen the morphological diagnosis of clear cell ovarian carcinoma in the process of differential diagnosis between clear cell ovarian tumors and other histological subtypes.
Collapse
Affiliation(s)
| | - Kathrine Bjersand
- Department of Women’s and Children’s Health, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Helena Åkerud
- Department of Women’s and Children’s Health, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Tomas Seidal
- Department of Pathology, Halmstad Medical Center Hospital, Halmstad, Sweden
| |
Collapse
|
44
|
Abstract
GATA3 is a member of a group of zinc-finger transcription factors that is involved in cell development and differentiation. Recent studies have shown that, among tumors, GATA3 is commonly expressed in both urothelial tumors and breast epithelial neoplasms. With the exception of salivary gland and parathyroid tumors, GATA3 has been reported to be either absent or only rarely expressed in other epithelial tumors. Owing to its restricted expression in urothelial and breast carcinomas, GATA3 has proved to be a useful immunohistochemical marker for assisting in distinguishing these 2 groups of neoplasms from other malignancies with which they may be confused.
Collapse
|
45
|
Matsukuma S, Kono T, Takeo H, Hamakawa Y, Sato K. Tumor-to-tumor metastasis from lung cancer: a clinicopathological postmortem study. Virchows Arch 2013; 463:525-34. [PMID: 23913165 DOI: 10.1007/s00428-013-1455-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 06/17/2013] [Accepted: 07/09/2013] [Indexed: 12/12/2022]
Abstract
This study examined 47 cases of lung cancer concomitant with other tumors and found eight cases (17 %) with nine foci of tumor-to-tumor metastasis, defined as metastasis of lung cancer into another tumor. Donor lung cancers were four adenocarcinomas, two squamous cell carcinomas, and two small cell carcinomas. Tumor-to-tumor metastasis was found in five of six renal cell carcinomas (83 %), one of eight thyroid papillary carcinomas (13 %), one of three adrenocortical adenomas (33 %), one of three pancreatic endocrine microadenomas (33 %), and another lung cancer (one of six cases of multiple lung cancers, 17 %). The higher recipient incidence in renal cell carcinoma was statistically significant compared with prostatic carcinoma (0/16, P < 0.001), colorectal carcinoma (0/7, P = 0.005), and gastric carcinoma (0/5, P = 0.015). Generalized metastases were found in 88 % of the tumor-to-tumor metastasis cases. The total clinical course of patients with tumor-to-tumor metastasis was shorter than that of the patients without tumor-to-tumor metastasis (mean, 5.4 versus 18.8 months; P = 0.046). Tumor-to-tumor metastasis sometimes mimicked undifferentiated recipient tumor cells. Immunostains for thyroid transcription factor 1 (TTF-1), Napsin A, cytokeratin 7 (CK7), and CK5/6 were useful to confirm tumor-to-tumor metastasis. However, TTF-1-, Napsin A-, and/or CK7-negative lung adenocarcinoma components metastasized to renal cell carcinoma in three cases, and recipient renal cell carcinomas were focally Napsin A+ (two cases) or CK7+ (two cases). Tumor-to-tumor metastasis can occur as a result of metastasis from lung cancer with more aggressive behavior. Tumor-to-tumor metastasis should be carefully distinguished from undifferentiated recipient tumor cells.
Collapse
Affiliation(s)
- Susumu Matsukuma
- Department of Pathology, Japan Self-Defense Forces Central Hospital, 1-2-24 Ikejiri, Setagaya-ku, 154-0001, Tokyo, Japan,
| | | | | | | | | |
Collapse
|
46
|
Ordóñez NG. Value of PAX8, PAX2, napsin A, carbonic anhydrase IX, and claudin-4 immunostaining in distinguishing pleural epithelioid mesothelioma from metastatic renal cell carcinoma. Mod Pathol 2013; 26:1132-43. [PMID: 23503645 DOI: 10.1038/modpathol.2013.34] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Revised: 12/12/2012] [Accepted: 12/28/2012] [Indexed: 01/05/2023]
Abstract
Both mesotheliomas and renal cell carcinomas can present a wide variety of cytomorphologic features and histologic patterns. Because of this, renal cell carcinomas metastatic to the pleura and lung can be confused with mesotheliomas. Recently, a variety of positive carcinoma markers, including kidney-associated markers, have become available. The aim of this study is to investigate the value of some of these markers, specifically PAX8, PAX2, napsin A, carbonic anhydrase IX, and claudin-4, for assisting in distinguishing pleural epithelioid mesotheliomas from metastatic renal cell carcinomas. To do so, a total of 40 pleural epithelioid mesotheliomas and 55 renal cell carcinomas (33 clear cell, 10 papillary, and 12 chromophobe) were investigated. In all, 91% of the renal cell carcinomas expressed claudin-4, 89% PAX8, 60% PAX2, 71% carbonic anhydrase IX, and 29% napsin A. All of the mesotheliomas were positive for carbonic anhydrase IX and were negative for all of the other markers. On the basis of these results, it is concluded that claudin-4 and PAX8 have a higher sensitivity and specificity for assisting in discriminating between pleural epithelioid mesotheliomas and renal cell carcinomas when compared with all of the other positive carcinoma markers that are, at present, recommended to be included in the immunohistochemical panels used in this differential diagnosis. Even though PAX2 and napsin A are highly specific, because of their low sensitivity, they have only a limited value. Carbonic anhydrase IX is not useful.
Collapse
Affiliation(s)
- Nelson G Ordóñez
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
47
|
Abstract
Signet-ring cell mesothelioma is uncommon and only two case reports have been published on this mesothelioma variant, both of which were initially misdiagnosed as signet-ring cell carcinoma. Herein are reported 23 signet-ring cell mesotheliomas that were investigated by immunohistochemistry, 12 of which were also studied by electron microscopy. Twenty-one of the cases originated in the pleura and two in the peritoneum. For comparison purposes and in order to determine the value of these techniques in the differential diagnosis of these tumors, seven cases of signet-ring cell lung adenocarcinoma were also studied. All signet-ring cell mesotheliomas were positive for calretinin, keratin 5/6, keratin 7, and mesothelin, 93% for podoplanin, and 91% for WT1; whereas, none reacted for MOC-31, CEA, TAG-72, CD15, TTF-1, napsin A, or CDX2. Among signet-ring cell lung adenocarcinomas, 100% were positive for keratin 7, CEA, and napsin A, 86% each for TTF-1 and TAG-72, 71% for CD15, and 14% for mesothelin, while all were negative for calretinin, keratin 5/6, WT1, podoplanin, and CDX2. After analyzing the results, it is concluded that the panels of markers used in the differential diagnosis of this mesothelioma variant should include those markers that are usually expressed in mesotheliomas (eg, calretinin, keratin 5/6, WT1, and podoplanin), broad-spectrum carcinoma markers that are frequently expressed in adenocarcinomas regardless of their site of origin (eg, MOC-31 and CEA), and organ-associated markers (eg, TTF-1 and napsin A for lung), which allow the site of origin of a metastatic adenocarcinoma to be established. Electron microscopy can be very useful as it permits the identification of characteristic ultrastructural mesothelioma and adenocarcinoma markers, and it also allows a better understanding of the morphologic features seen on routine light microscopy. Pathologists should be aware of this mesothelioma subtype as it can potentially be confused with other tumors that exhibit signet-ring features.
Collapse
|
48
|
Wu J, Chu PG, Jiang Z, Lau SK. Napsin A expression in primary mucin-producing adenocarcinomas of the lung: an immunohistochemical study. Am J Clin Pathol 2013; 139:160-6. [PMID: 23355200 DOI: 10.1309/ajcp62wjuamszcom] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Immunohistochemical expression of napsin A in primary pulmonary mucinous tumors is not well established. Napsin A immunoreactivity was evaluated in 43 mucin-producing adenocarcinomas of the lung consisting of 18 tumors formerly classified as mucinous bronchioloalveolar carcinoma, 15 colloid adenocarcinomas, 5 solid predominant adenocarcinomas with mucin production, and 5 adenocarcinomas with signet ring cell features, as well as in 25 extrapulmonary mucinous adenocarcinomas of different anatomic sites. Immunohistochemical expression of thyroid transcription factor 1 (TTF-1) was also compared. Thirty-three percent of mucinous lung tumors exhibited positive immunoreactivity for napsin A, whereas 42% expressed TTF-1. All 25 extrapulmonary mucinous adenocarcinomas lacked expression of napsin A and TTF-1. Mucin-producing neoplasms of the lung infrequently express napsin A, suggesting that immunohistochemical assessment of napsin A may have limited diagnostic usefulness for distinguishing primary and metastatic mucinous adenocarcinomas involving the lung.
Collapse
Affiliation(s)
- Jeffrey Wu
- Department of Pathology, City of Hope National Medical Center, Duarte, CA
| | - Peiguo G. Chu
- Department of Pathology, City of Hope National Medical Center, Duarte, CA
| | - Zhong Jiang
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA
| | - Sean K. Lau
- Department of Pathology, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
49
|
Mukhopadhyay S, Katzenstein ALA. Comparison of monoclonal napsin A, polyclonal napsin A, and TTF-1 for determining lung origin in metastatic adenocarcinomas. Am J Clin Pathol 2012; 138:703-11. [PMID: 23086771 DOI: 10.1309/ajcpkvbxti9o3tem] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Thyroid transcription factor 1 (TTF-1) is currently the best immunohistochemical marker for carcinomas of lung origin. Our aim was to compare napsin A to TTF-1 for identifying pulmonary origin in metastatic adenocarcinoma and its mimics. One hundred fifty-five metastatic carcinomas (55 pulmonary, 100 nonpulmonary) were stained with monoclonal napsin A and TTF-1, and most also with polyclonal napsin A. The sensitivity of monoclonal napsin A, polyclonal napsin A, and TTF-1 for metastatic adenocarcinomas of pulmonary origin was 76%, 81%, and 82%, respectively. Two lung carcinomas were diffusely positive for monoclonal napsin A, but negative or equivocal for TTF-1. TTF-1 stained 9 of 100 nonpulmonary carcinomas (all thyroid), monoclonal napsin A stained 12 of 100 (4 sites), and polyclonal napsin A stained 27 of 91 (8 sites). Napsin A is expressed in a wider variety of metastatic nonpulmonary carcinomas than TTF-1, and the monoclonal antibody is more specific. Napsin A is a useful adjunct to TTF-1, because occasional lung adenocarcinomas are TTF-1 negative but napsin A positive.
Collapse
|
50
|
Ordóñez NG. Application of immunohistochemistry in the diagnosis of epithelioid mesothelioma: a review and update. Hum Pathol 2012; 44:1-19. [PMID: 22963903 DOI: 10.1016/j.humpath.2012.05.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 05/14/2012] [Accepted: 05/16/2012] [Indexed: 01/12/2023]
Abstract
A large number of immunohistochemical markers that can assist in the differential diagnosis of epithelioid mesotheliomas are currently available. Because these markers are expressed differently in the various types of carcinomas that can metastasize to the serosal membranes and can potentially be confused with epithelioid mesothelioma, their selection for inclusion in a diagnostic panel largely depends on the differential diagnosis, as well as on which ones work the best in a given laboratory. Traditionally, the panels used in the differential diagnosis of epithelioid mesothelioma have consisted of a combination of positive mesothelioma markers and broad-spectrum carcinoma markers. At present, a wide variety of organ-associated carcinoma markers such as thyroid transcription factor-1 and napsin A for the lung, PAX 8 and PAX 2 for the kidney, and Müllerian-derived tumors; gross cystic disease fluid protein-15 and mammaglobin for the breast; and CDX2 for intestinal differentiation are available, which can assist in establishing the site of origin of an adenocarcinoma when included in a diagnostic panel. This article provides updated information on the composition of the panels of markers recommended in the various differential diagnoses.
Collapse
Affiliation(s)
- Nelson G Ordóñez
- The University of Texas MD Anderson Cancer Center, Department of Pathology, Houston, TX 77030, USA.
| |
Collapse
|