1
|
de Lima-Souza RA, Altemani A, Michal M, Mariano FV, Leivo I, Skálová A. Expanding the Molecular Spectrum of Carcinoma Ex Pleomorphic Adenoma: An Analysis of 84 Cases With a Novel HMGA2::LINC02389 Fusion. Am J Surg Pathol 2024:00000478-990000000-00418. [PMID: 39324957 DOI: 10.1097/pas.0000000000002307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Carcinoma ex pleomorphic adenoma (CXPA) is an aggressive epithelial and/or myoepithelial neoplasm that arises in association with a pleomorphic adenoma (PA). Its etiopathogenesis remains poorly understood, but it is believed that the development of this tumor is due to the accumulation of genetic, protein, metabolic, and epigenetic alterations in a PA. A retrospective review of the Salivary Gland Tumor Registry in Pilsen yielded 84 CXPA, namely 25/84 salivary duct carcinoma (SDC), 15/84 myoepithelial carcinoma (MC), 1/84 epithelial-myoepithelial carcinoma (EMC), and 1/84 adenoid cystic carcinoma (AdCC). All 84 CXPA cases were analyzed by next-generation sequencing (NGS) and/or fluorescence in situ hybridization (FISH). Forty-three tumors originally diagnosed as CXPA (43/84, 51.2%) showed some molecular alteration. Fusion transcripts were identified in 12/16 (75%) CXPA, including LIFR::PLAG1, CTNNB1::PLAG1, FGFR1::PLAG1, and a novel fusion, HMGA2::LINC02389. Most of the fusions were confirmed by FISH using PLAG1 (6/11) and HMGA2 (1/1) gene break probes. Split signals indicating gene break were identified by FISH for PLAG1 (12/17), HMGA2 (3/4), EWSR1 (7/22), and MYB (2/7). Concerning pathogenic mutations, only CXPA with epithelial differentiation (SDC) presented these alterations, including HRAS mutation (2/4), TP53 (1/4), PTEN (1/4), and ATK1 (1/4). In addition, amplifications in ERBB2 (17/35), MDM2 (1/4), and EWSR1 (1/7) were detected. A novel finding was the discovery of an HMGA2::LINC02389 fusion in 1 patient with EMC ex-PA. The present results indicate that molecular profiling of CXPA with myoepithelial differentiation (MC) tends to reveal chromosomal fusion events, whereas CXPA with epithelial differentiation (SDC) tends to have a higher frequency of pathogenic mutations and gene amplifications.
Collapse
Affiliation(s)
- Reydson Alcides de Lima-Souza
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba
- Department of Pathology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Albina Altemani
- Department of Pathology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Michal Michal
- Bioptic Laboratory Ltd
- Department of Pathology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Fernanda Viviane Mariano
- Department of Pathology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Ilmo Leivo
- Department of Pathology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Alena Skálová
- Bioptic Laboratory Ltd
- Department of Pathology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
2
|
Carillo AM, De Luca C, Pisapia P, Vigliar E, Ikenberg K, Freiberger SN, Troncone G, Rupp NJ, Bellevicine C. Molecular testing in salivary gland cytopathology: A practical overview in conjunction with the Milan system. Cytopathology 2024; 35:330-343. [PMID: 38308401 DOI: 10.1111/cyt.13363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/04/2024]
Abstract
Recently, significant advances in the molecular characterization of salivary gland neoplasms have facilitated the classification and diagnosis of specific diagnostic entities. In the highly challenging diagnostic scenario of salivary malignancies, molecular testing is increasingly being adopted in routine practice to refine the cytological diagnosis of salivary lesions. Here, we reviewed the most recent evidence in the field of salivary glands molecular cytopathology.
Collapse
Affiliation(s)
- Anna Maria Carillo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Caterina De Luca
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Elena Vigliar
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Kristian Ikenberg
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Sandra N Freiberger
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Claudio Bellevicine
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
3
|
Viehweger F, Hoop J, Tinger LM, Bernreuther C, Büscheck F, Clauditz TS, Hinsch A, Jacobsen F, Luebke AM, Steurer S, Hube-Magg C, Kluth M, Marx AH, Krech T, Lebok P, Fraune C, Burandt E, Sauter G, Simon R, Minner S. Frequency of Androgen Receptor Positivity in Tumors: A Study Evaluating More Than 18,000 Tumors. Biomedicines 2024; 12:957. [PMID: 38790919 PMCID: PMC11117763 DOI: 10.3390/biomedicines12050957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Androgen receptor (AR) is a transcription factor expressed in various normal tissues and is a therapeutic target for prostate and possibly other cancers. A TMA containing 18,234 samples from 141 different tumor types/subtypes and 608 samples of 76 different normal tissue types was analyzed by immunohistochemistry. AR positivity was found in 116 tumor types including 66 tumor types (46.8%) with ≥1 strongly positive tumor. Moderate/strong AR positivity was detected in testicular sex cord-stromal tumors (93.3-100%) and neoplasms of the prostate (79.3-98.7%), breast (25.0-75.5%), other gynecological tumors (0.9-100%), kidney (5.0-44.1%), and urinary bladder (5.4-24.2%). Low AR staining was associated with advanced tumor stage (pTa versus pT2-4; p < 0.0001) in urothelial carcinoma; advanced pT (p < 0.0001), high tumor grade (p < 0.0001), nodal metastasis (p < 0.0001), and reduced survival (p = 0.0024) in invasive breast carcinoma; high pT (p < 0.0001) and grade (p < 0.0001) in clear cell renal cell carcinoma (RCC); and high pT (p = 0.0055) as well as high grade (p < 0.05) in papillary RCC. AR staining was unrelated to histopathological/clinical features in 157 endometrial carcinomas and in 221 ovarian carcinomas. Our data suggest a limited role of AR immunohistochemistry for tumor distinction and a prognostic role in breast and clear cell RCC and highlight tumor entities that might benefit from AR-targeted therapy.
Collapse
Affiliation(s)
- Florian Viehweger
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Jennifer Hoop
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Lisa-Marie Tinger
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Franziska Büscheck
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Till S. Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
- Pathologie-Hamburg, Labor Lademannbogen Medizinisches Versorgungszentrum (MVZ) GmbH, 22339 Hamburg, Germany
| | - Andreas M. Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Andreas H. Marx
- Department of Pathology, Academic Hospital Fuerth, 90766 Fuerth, Germany;
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
- Institute of Pathology, Clinical Center Osnabrueck, 49076 Osnabrueck, Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
- Institute of Pathology, Clinical Center Osnabrueck, 49076 Osnabrueck, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
- Institute of Pathology, Clinical Center Osnabrueck, 49076 Osnabrueck, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (F.V.); (J.H.); (C.B.); (F.B.); (T.S.C.); (A.H.); (F.J.); (A.M.L.); (S.S.); (C.H.-M.); (M.K.); (T.K.); (P.L.); (C.F.); (E.B.); (G.S.); (S.M.)
| |
Collapse
|
4
|
Perri F, Fusco R, Sabbatino F, Fasano M, Ottaiano A, Cascella M, Marciano ML, Pontone M, Salzano G, Maiello ME, Montano M, Calogero E, D'Aniello R, Maiolino P, Ciardiello F, Zotta A, Alfieri S, Ionna F. Translational Insights in the Landscape of Salivary Gland Cancers: Ready for a New Era? Cancers (Basel) 2024; 16:970. [PMID: 38473330 DOI: 10.3390/cancers16050970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/15/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Salivary gland carcinomas (SGCs) are rare neoplasms, representing less than 10% of all head and neck tumors, but they are extremely heterogeneous from the histological point of view, their clinical behavior, and their genetics. The guidelines regarding their treatment include surgery in most cases, which can also play an important role in oligometastatic disease. Where surgery cannot be used, systemic therapy comes into play. Systemic therapy for many years has been represented by polychemotherapy, but recently, with the affirmation of translational research, it can also count on targeted therapy, at least in some subtypes of SGCs. Interestingly, in some SGC histotypes, predominant mutations have been identified, which in some cases behave as "driver mutations", namely mutations capable of governing the carcinogenesis process. Targeting these driver mutations may be an effective therapeutic strategy. Nonetheless, it is not always possible to have drugs suitable for targeting driver mutations-and targeting driver mutations is not always accompanied by a clinical benefit. In this review, we will analyze the main mutations predominant in the various histotypes of SGCs.
Collapse
Affiliation(s)
- Francesco Perri
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Roberta Fusco
- Medical Oncology Devision, IGEA S.p.A., 80013 Naples, Italy
| | - Francesco Sabbatino
- Medical Oncology Department, Università degli Studi di Salerno, Via Giovanni Paolo II, 132, 84084 Salerno, Italy
| | - Morena Fasano
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80128 Naples, Italy
| | - Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Marco Cascella
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Maria Luisa Marciano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Monica Pontone
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Giovanni Salzano
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Maria Elena Maiello
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Massimo Montano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Ester Calogero
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Roberta D'Aniello
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Piera Maiolino
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80128 Naples, Italy
| | - Alessia Zotta
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80128 Naples, Italy
| | - Salvatore Alfieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Franco Ionna
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy
| |
Collapse
|
5
|
McAfee JL, Hoda RS, Hoyle C, McCoy L, Sprague C, Reddy CA, Koyfman SA, Geiger JL, Komforti MK, Griffith CC. ERBB2 Amplification and HER2 Expression in Salivary Duct Carcinoma: Evaluation of Scoring Guidelines and Potential for Expanded Anti-HER2 Therapy. Mod Pathol 2023; 36:100273. [PMID: 37423585 DOI: 10.1016/j.modpat.2023.100273] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/09/2023] [Accepted: 06/30/2023] [Indexed: 07/11/2023]
Abstract
Salivary duct carcinoma (SDC) is aggressive with limited therapeutic options. A subset of SDC display human epidermal growth factor receptor 2 (HER2) protein overexpression by immunohistochemistry, and some show ERBB2 gene amplification. Guidelines for HER2 scoring are not firmly established. Recent advances in breast carcinoma have established a role for anti-HER2 therapies in lesions with low HER2 expression lacking ERBB2 amplification. Delineating HER2 staining patterns in SDC is critical for evaluating anti-HER2 treatments. In total, 53 cases of SDC resected at our institution between 2004 and 2020 were identified. Androgen receptor (AR) and HER2 immunohistochemistry and ERBB2 fluorescence in situ hybridization were performed in all cases. AR expression was scored for percentage positive cells and categorized as positive (>10% of cells), low positive (1%-10%), or negative (<1%). HER2 staining levels and patterns were recorded, scored using 2018 ASCO/CAP guidelines, and categorized into HER2-positive (3+ or 2+ with ERBB2 amplification), HER2-low (1+ or 2+ without ERBB2 amplification), HER2-very low (faint staining in <10% of cells), or HER2-absent types. Clinical parameters and vital status were recorded. Median age was 70 years, with a male predominance. ERBB2-amplified tumors (11/53; 20.8%) presented at lower pT stages (pTis/pT1/pT2; P = .005, Fisher exact test) and more frequently had perineural invasion (P = .007, Fisher exact test) compared with ERBB2 nonamplified tumors; no other pathologic features differed significantly by gene amplification status. In addition, 2+ HER2 staining by 2018 ASCO/CAP criteria was most common (26/53; 49%); only 4 cases (8%) were HER2-absent status; 3+ HER2 staining was found in 9 tumors, and all were ERBB2 amplified. Six patients with HER2-expressing tumors received trastuzumab therapy, including 2 with ERBB2-amplified tumors. Overall survival and recurrence-free survival did not differ significantly based on ERBB2 status. This work suggests that 2018 ASCO/CAP guidelines for HER2 evaluation in breast carcinoma could be applied to SDC. Our findings also show broad overexpression of HER2 in SDC raising the possibility that more patients may benefit from anti-HER2-directed therapies.
Collapse
Affiliation(s)
- John L McAfee
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Raza S Hoda
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Carrie Hoyle
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Lauren McCoy
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Cathy Sprague
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | | | | | | | - Miglena K Komforti
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, Florida
| | | |
Collapse
|
6
|
Rieke DT, Schröder S, Schafhausen P, Blanc E, Zuljan E, von der Emde B, Beule D, Keller U, Keilholz U, Klinghammer K. Targeted treatment in a case series of AR+, HRAS/PIK3CA co-mutated salivary duct carcinoma. Front Oncol 2023; 13:1107134. [PMID: 37427101 PMCID: PMC10325704 DOI: 10.3389/fonc.2023.1107134] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 05/26/2023] [Indexed: 07/11/2023] Open
Abstract
Background and purpose A subgroup of salivary duct carcinoma (SDC) harbor overexpression of the androgen receptor (AR), and co-occurring mutations in the HRAS- and PIK3CA-genes. The impact of genomic complexity on targeted treatment strategies in advanced cancer is unknown. Materials and methods We analyzed molecular and clinical data from an institutional molecular tumor board (MTB) to identify AR+, HRAS/PIK3CA co-mutated SDC. Follow-up was performed within the MTB registrational study or retrospective chart review after approval by the local ethics committee. Response was assessed by the investigator. A systematic literature search was performed in MEDLINE to identify additional clinically annotated cases. Results 4 patients with AR+ HRAS/PIK3CA co-mutated SDC and clinical follow-up data were identified from the MTB. An additional 9 patients with clinical follow-up were identified from the literature. In addition to AR overexpression and HRAS and PIK3CA-alterations, PD-L1 expression and Tumor Mutational Burden > 10 Mutations per Megabase were identified as additional potentially targetable alterations. Among evaluable patients, androgen deprivation therapy (ADT) was initiated in 7 patients (1 Partial Response (PR), 2 Stable Disease (SD), 3 Progressive Disease (PD), 2 not evaluable), tipifarnib was initiated in 6 patients (1 PR, 4 SD, 1 PD). One patient each was treated with immune checkpoint inhibition (Mixed Response) and combination therapies of tipifarnib and ADT (SD) and alpelisib and ADT (PR). Conclusion Available data further support comprehensive molecular profiling of SDC. Combination therapies, PI3K-inhibitors and immune therapy warrant further investigation, ideally in clinical trials. Future research should consider this rare subgroup of SDC.
Collapse
Affiliation(s)
- Damian T. Rieke
- Department of Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Comprehensive Cancer Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Schröder
- Department of Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philippe Schafhausen
- Department of Oncology, Hematology, and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eric Blanc
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Core Unit Bioinformatics (CUBI), Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Erika Zuljan
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Core Unit Bioinformatics (CUBI), Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Benjamin von der Emde
- Department of Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Core Unit Bioinformatics (CUBI), Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Ulrich Keilholz
- Comprehensive Cancer Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Konrad Klinghammer
- Department of Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
7
|
Rooper LM, Agaimy A, Assaad A, Bal M, Eugene H, Gagan J, Nonogaki H, Palsgrove DN, Shah A, Stelow E, Stoehr R, Thompson LDR, Weinreb I, Bishop JA. Recurrent IDH2 Mutations in Salivary Gland Striated Duct Adenoma Define an Expanded Histologic Spectrum Distinct From Canalicular Adenoma. Am J Surg Pathol 2023; 47:333-343. [PMID: 36510691 DOI: 10.1097/pas.0000000000002004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Striated duct adenoma (SDA) is a rare salivary gland neoplasm defined by histologic similarity to normal striated ducts. However, doubt persists about whether SDA represents a genuine entity distinct from canalicular adenoma and if a malignant counterpart exists. This study aims to evaluate the molecular underpinnings of SDA to clarify its pathogenesis and classification. We identified 10 SDA and 2 tumors called low-grade adenocarcinoma not otherwise specified that were retrospectively recognized to resemble SDA. All cases showed recurrent histologic features including (1) discrete monophasic tubules, (2) tall columnar eosinophilic cells, (3) monotonous oval nuclei, and (4) scant fibrous stroma, and most were positive for S100 protein (91%), SOX10 (80%), and CK7 (80%). Although 1 case was previously called adenocarcinoma based on interdigitation with normal acini, this pattern was also seen in some SDA, and likely does not indicate malignancy; the significance of growth surrounding nerve in 1 other case is less clear. Targeted sequencing identified IDH2 R172X mutations in all 8 cases with sufficient tissue, with positivity for IDH1/2 mutation-specific immunohistochemistry in 9 cases stained. In contrast, 5 canalicular adenomas lacked IDH2 mutations or other oncogenic alterations. Overall, IDH2 R172X mutations are a defining feature of SDA that, in combination with its recognizable pathologic profile, confirm it is a unique entity separate from canalicular adenoma. IDH1/2 mutation-specific immunohistochemistry may provide a convenient tool to facilitate diagnosis. Both morphology and IDH2 mutations raise parallels between SDA and breast tall cell carcinoma with reverse polarity.
Collapse
Affiliation(s)
- Lisa M Rooper
- Department of Pathology
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Abbas Agaimy
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital, Erlangen, Germany
| | - Adel Assaad
- Department of Pathology, Virginia Mason Hospital and Seattle Medical Center, Seattle, WA
| | - Munita Bal
- Department of Pathology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | | | - Jeffrey Gagan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | | | - Doreen N Palsgrove
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Akeesha Shah
- Department of Pathology, Cleveland Clinic Foundation, Cleveland, OH
| | - Edward Stelow
- Department of Pathology, University of Virginia, Charlottesville, VA
| | - Robert Stoehr
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital, Erlangen, Germany
| | | | - Ilan Weinreb
- Department of Laboratory Medicine and Pathobiology, University of Toronto
- Department of Pathology, University Health Network, Toronto, ON
| | - Justin A Bishop
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
8
|
[Salivary gland tumors-an overview : Advances in molecular characterization: Part II]. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:70-77. [PMID: 36622399 PMCID: PMC9877055 DOI: 10.1007/s00292-022-01171-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/02/2022] [Indexed: 01/10/2023]
Abstract
The second part of the article is devoted to the molecular characteristics of epithelial-myoepithelial carcinoma, polymorphous adenocarcinoma, myoepithelial carcinoma, basal cell adenocarcinoma, and salivary duct carcinoma. In addition, the new entities mucinous adenocarcinoma, sclerosing microcystic adenocarcinoma, and microsecretory adenocarcinoma are summarized. The molecular genotype can also be very helpful in diagnosing most of these entities. In this regard, overexpression of the androgen receptor and/or human epidermal growth factor receptor 2 (HER2)/neu can support diagnosis in the appropriate histopathologic context and may serve as a potential target for therapy in salivary duct carcinoma.
Collapse
|
9
|
Identification of novel prognostic and predictive biomarkers in salivary duct carcinoma via comprehensive molecular profiling. NPJ Precis Oncol 2022; 6:82. [PMCID: PMC9636405 DOI: 10.1038/s41698-022-00324-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
AbstractMolecular targets and predictive biomarkers for prognosis in salivary duct carcinoma (SDC) have not been fully identified. We conducted comprehensive molecular profiling to discover novel biomarkers for SDC. A total of 67 SDC samples were examined with DNA sequencing of 464 genes and transcriptome analysis in combination with the clinicopathological characteristics of the individuals. Prognostic biomarkers associated with response to combined androgen blockade (CAB) treatment were explored using mRNA expression data from 27 cases. Oncogenic mutations in receptor tyrosine kinase (RTK) genes or genes in the MAPK pathway were identified in 55 cases (82.1%). Alterations in the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway were identified in 38 cases (56.7%). Interestingly, patient prognosis could be predicted using mRNA expression profiles, but not genetic mutation profiles. The risk score generated from the expression data of a four-gene set that includes the ADAMTS1, DSC1, RNF39, and IGLL5 genes was a significant prognostic marker for overall survival in the cohort (HR = 5.99, 95% confidence interval (CI) = 2.73–13.1, p = 7.8 × 10−6). Another risk score constructed from the expression of CD3E and LDB3 was a strong prognostic marker for progression-free survival for CAB treatment (p = 0.03). Mutations in RTK genes, MAPK pathway genes, and PI3K/AKT pathway genes likely represent key mutations in SDC tumorigenesis. The gene expression profiles identified in this study may be useful for stratifying patients who are good candidates for CAB treatment and may benefit from additional systemic therapies.
Collapse
|
10
|
Kawakita D, Nagao T, Takahashi H, Kano S, Honma Y, Hirai H, Saigusa N, Akazawa K, Tani K, Ojiri H, Tsukahara K, Ozawa H, Okami K, Kondo T, Togashi T, Fushimi C, Shimura T, Shimizu A, Okamoto I, Okada T, Imanishi Y, Watanabe Y, Otsuka K, Sakai A, Ebisumoto K, Sato Y, Yamazaki K, Ueki Y, Hanazawa T, Saito Y, Ando M, Matsuki T, Nakaguro M, Sato Y, Urano M, Utsumi Y, Kohsaka S, Saotome T, Tada Y. Survival benefit of HER2-targeted or androgen deprivation therapy in salivary duct carcinoma. Ther Adv Med Oncol 2022; 14:17588359221119538. [PMID: 36090801 PMCID: PMC9459484 DOI: 10.1177/17588359221119538] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022] Open
Abstract
Background The efficacy and safety of human epidermal growth factor receptor 2 (HER2)-targeted therapy and androgen deprivation therapy (ADT) for locally advanced or recurrent or metastatic (LA/RM) salivary duct carcinoma (SDC) have been reported in prospective studies. However, the survival benefit of these therapies to conventional therapy remains controversial, and whether HER2-targeted therapy or ADT should be chosen in HER2- and androgen receptor (AR)-positive SDC patients remains unknown. Methods Overall, 323 LA/RM SDC patients treated at seven institutions between August 1992 and June 2020 were retrospectively enrolled. The primary aim was to analyze the effect of HER2-targeted therapy and ADT on overall survival from the diagnosis of LA/RM disease to death from any cause (OS1). The secondary indicators included the overall response rate (ORR), clinical benefit rate (CBR), overall survival from therapy initiation for LA/RM disease (OS2), progression-free survival (PFS), time to second progression (PFS2), duration of response (DoR), and duration of clinical benefit (DoCB) of HER2-targeted therapy or ADT as first-line therapy for HER2-positive/AR-positive SDC. Results Patients treated with HER2-targeted therapy or ADT had longer OS1 than those treated without these therapies (Median OS1: historical control, 21.6 months; HER2-targeted therapy, 50.6 months; ADT, 32.8 months; HER2-targeted therapy followed by ADT, 42.4 months; and ADT followed by HER2-targeted therapy, 45.2 months, p < 0.001). Among HER2-positive/AR-positive SDC patients, although HER2-targeted therapy had better ORR, CBR, and PFS than those of ADT as first-line therapy, we found no significant differences between HER2-targeted therapy and ADT regarding OS2, PFS2, DoR, and DoCB. Conclusion Patients treated with HER2-targeted therapy and ADT showed longer survival in LA/RM SDC. HER2-targeted therapy can be recommended prior to ADT for HER2-positive/AR-positive SDC. It is warranted to establish a biomarker that could predict the efficacy of clinical benefit or better response in ADT.
Collapse
Affiliation(s)
- Daisuke Kawakita
- Department of Otorhinolaryngology, Head and
Neck Surgery, Nagoya City University Graduate School of Medical Sciences,
Nagoya, Japan
| | - Toshitaka Nagao
- Department of Anatomic Pathology, Tokyo Medical
University, Shinjuku-ku, Tokyo, Japan
| | - Hideaki Takahashi
- Department of Otorhinolaryngology, Head and
Neck Surgery, Yokohama City University, School of Medicine, Yokohama,
Japan
| | - Satoshi Kano
- Department of Otolaryngology – Head and Neck
Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido
University, Sapporo, Japan
| | - Yoshitaka Honma
- Department of Head and Neck, Esophageal Medical
Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Hideaki Hirai
- Department of Anatomic Pathology, Tokyo Medical
University, Shinjuku-ku, Tokyo, Japan
| | - Natsuki Saigusa
- Department of Anatomic Pathology, Tokyo Medical
University, Shinjuku-ku, Tokyo, Japan
| | - Kohei Akazawa
- Department of Medical Informatics, Niigata
University Medical and Dental Hospital, Chuo-ku, Niigata, Japan
| | - Kaori Tani
- Department of Medical Informatics, Niigata
University Medical and Dental Hospital, Chuo-ku, Niigata, Japan
| | - Hiroya Ojiri
- Department of Radiology, The Jikei University
School of Medicine, Minato-ku, Tokyo, Japan
| | - Kiyoaki Tsukahara
- Department of Otorhinolaryngology, Head and
Neck Surgery, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Hiroyuki Ozawa
- Department of Otorhinolaryngology, Head and
Neck Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo,
Japan
| | - Kenji Okami
- Department of Otolaryngology, Head and Neck
Surgery, School of Medicine, Tokai University, Isehara, Japan
| | - Takahito Kondo
- Department of Otorhinolaryngology, Head and
Neck Surgery, Tokyo Medical University Hachioji Medical Center, Hachioji,
Japan
| | - Takafumi Togashi
- Department of Head and Neck Surgery, Niigata
Cancer Center Hospital, Niigata, Japan
| | - Chihiro Fushimi
- Department of Head and Neck Oncology and
Surgery, International University of Health and Welfare, Mita Hospital,
Minato-ku, Tokyo, Japan
| | - Tomotaka Shimura
- Department of Otolaryngology, Showa University
Fujigaoka Hospital, Aoba-ku, Yokohama, Japan
| | - Akira Shimizu
- Department of Otorhinolaryngology, Head and
Neck Surgery, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Isaku Okamoto
- Department of Otorhinolaryngology, Head and
Neck Surgery, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Takuro Okada
- Department of Otorhinolaryngology, Head and
Neck Surgery, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Yorihisa Imanishi
- Department of Otorhinolaryngology, Head and
Neck Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo,
Japan
| | - Yoshihiro Watanabe
- Department of Otorhinolaryngology, Head and
Neck Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo,
Japan
| | - Kuninori Otsuka
- Department of Otorhinolaryngology, Head and
Neck Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo,
Japan
| | - Akihiro Sakai
- Department of Otolaryngology, Head and Neck
Surgery, School of Medicine, Tokai University, Isehara, Japan
| | - Koji Ebisumoto
- Department of Otolaryngology, Head and Neck
Surgery, School of Medicine, Tokai University, Isehara, Japan
| | - Yuichiro Sato
- Department of Head and Neck Surgery, Niigata
Cancer Center Hospital, Niigata, Japan
| | - Keisuke Yamazaki
- Department of Otolaryngology Head and Neck
Surgery, Niigata University Graduate School of Medical and Dental Sciences,
Chuo-ku, Niigata, Japan
| | - Yushi Ueki
- Department of Otolaryngology Head and Neck
Surgery, Niigata University Graduate School of Medical and Dental Sciences,
Chuo-ku, Niigata, Japan
| | - Toyoyuki Hanazawa
- Department of Otorhinolaryngology/Head &
Neck Surgery, Chiba University Graduate School of Medicine, Chuo-ku, Chiba,
Japan
| | - Yuki Saito
- Department of Otolaryngology – Head and Neck
Surgery, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo,
Japan
| | - Mizuo Ando
- Department of Otorhinolaryngology/Head &
Neck Surgery, Okayama University Graduate School of Medicine, Kita-ku,
Okayama, Japan
| | - Takashi Matsuki
- Department of Otorhinolaryngology, Head and
Neck Surgery, Kitasato University School of Medicine, Minami-ku, Sagamihara,
Japan
| | - Masato Nakaguro
- Department of Pathology and Laboratory
Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Yukiko Sato
- Division of Pathology, Cancer Institute
Hospital, Japanese Foundation for Cancer Research, Koto-ku, Tokyo,
Japan
| | - Makoto Urano
- Department of Diagnostic Pathology, Bantane
Hospital, Fujita Health University, School of Medicine, Nakagawa-ku, Nagoya,
Japan
| | - Yoshitaka Utsumi
- Department of Anatomic Pathology, Tokyo
Medical University, Shinjuku-ku, Tokyo, Japan
| | - Shinji Kohsaka
- Division of Cellular Signaling, National
Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Takashi Saotome
- Division of Medical Oncology, Matsudo City
Hospital, Matsudo, Japan
| | - Yuichiro Tada
- Department of Head and Neck Oncology and
Surgery, International University of Health and Welfare, Mita Hospital,
1-4-3 Mita, Minato-ku, Tokyo 108-8329, Japan
| |
Collapse
|
11
|
Targeted molecular profiling of salivary duct carcinoma with rhabdoid features highlights parallels to other apocrine and discohesive neoplasms: which phenotype should drive classification? Head Neck Pathol 2022; 16:1063-1072. [PMID: 35794510 PMCID: PMC9729655 DOI: 10.1007/s12105-022-01464-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/17/2022] [Accepted: 05/22/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Salivary duct carcinoma with rhabdoid features (SDC-RF) is a recently-described salivary gland tumor that bears striking histologic similarity to lobular carcinoma of the breast. While this tumor has an apocrine phenotype that supports classification as a variant of SDC, it infrequently arises in association with conventional SDC. Furthermore, discohesive architecture can be seen in non-apocrine salivary carcinomas, raising the possibility that discohesive growth should define a separate entity. In this study, we aimed to perform comprehensive molecular profiling of SDC-RF to better understand its pathogenesis and classification. METHODS We documented the clinicopathologic features of 9 cases of SDC-RF and performed immunostains including AR, GCDFP, and e-cadherin on all cases. We also performed targeted next generation sequencing (NGS) panels on 7 cases that had sufficient tissue available. RESULTS The SDC-RF represented 8 men and 1 woman with a median age of 67 years (range 63-83 years) and included 6 parotid, 2 buccal, and 1 submandibular primary. All tumors were uniformly composed of discohesive cells with abundant eosinophilic cytoplasm; signet-ring cell features were seen in 2 cases. All tumors were also positive for AR (100%) and GCDFP (100%), and 7 tumors (78%) displayed lost or abnormal e-cadherin. NGS highlighted concomitant PIK3CA and HRAS mutations in 4 tumors, with additional cases harboring TP53, PTEN, and AKT1 mutations. Furthermore, CDH1 alterations were seen in 6 cases, including a novel CDH1::CORO7 fusion. Among 5 patients with follow-up available, 3 (60%) developed local recurrence and widespread distant metastasis and died of disease at a median 20 months (range 10-48 months). CONCLUSIONS Overall, our findings confirm frequent CDH1 mutations and e-cadherin inactivation in SDC-RF, similar to discohesive tumors from other sites. We also highlight an apocrine molecular profile similar to conventional SDC. However, occasional AKT1 mutation and signet-ring features suggest SDC-RF may also be related to mucinous adenocarcinoma. As more salivary tumors with discohesive growth are identified, it may become clearer whether SDC-RF should remain in the SDC family or be recognized as a separate entity.
Collapse
|
12
|
Wakely PE. Salivary duct carcinoma: A report of 70 FNA cases and review of the literature. Cancer Cytopathol 2022; 130:595-608. [PMID: 35255198 DOI: 10.1002/cncy.22568] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/22/2022] [Accepted: 02/15/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Although usually recognized as malignant, fine-needle aspiration (FNA) biopsy of salivary duct carcinoma (SDC) has been confused with other primary salivary gland (SG) neoplasms. This article undertook an analysis of a large collection of SDC FNA cases to assess diagnostic accuracy, specificity, and cytopathology. METHODS Cytopathology files were searched for SDC with histopathologic validation. FNA biopsy smears were performed using standard techniques. RESULTS Seventy cases from 56 patients (M:F, 1.9:1; age range, 26-92 years; mean age, 65 years) met inclusion criteria. All had tissue confirmation of SDC. FNA sites included: parotid gland (42, 60% cases), neck (10), submandibular gland (7), pre-/post-auricular area (5), face/cheek (3), mediastinal lymph nodes (2), and clavicle (1). Aspirates were from primary (52, 74%), metastatic (12, 17%), and locally recurrent (6, 9%) neoplasms. FNA diagnoses included: SDC (19, 27%), favor/suspicious for SDC (7, 10%), high-grade carcinoma (11), adenocarcinoma (9), carcinoma (6), malignant (6), SG neoplasm (5), atypia (3), SDC versus another malignancy (2), and pleomorphic adenoma (2). Large polygonal cells in groups and single forms showed cribriforming, variable necrosis, pseudopapillae, and oncocytic change. Androgen receptor staining was positive in all cases. CONCLUSIONS FNA biopsy is accurate and reliable in classifying SDC as a malignant neoplasm, but much less so for identification as a specific tumor type. Using the Milan system, 86% of aspirates were classified as either malignant or suspicious for malignancy. A recurring pitfall includes sampling error in cases of SDC ex pleomorphic adenoma.
Collapse
Affiliation(s)
- Paul E Wakely
- Department of Pathology, The Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| |
Collapse
|
13
|
Witte HM, Gebauer N, Steinestel K. Mutational and immunologic Landscape in malignant Salivary Gland Tumors harbor the potential for novel therapeutic strategies. Crit Rev Oncol Hematol 2022; 170:103592. [PMID: 35026433 DOI: 10.1016/j.critrevonc.2022.103592] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 11/24/2021] [Accepted: 01/06/2022] [Indexed: 12/18/2022] Open
Abstract
Salivary gland carcinomas (SGC) are rare (3-6 % of all head and neck cancers) and show biological heterogeneity depending on the respective histological subtype. While complete surgical resection is the standard treatment for localized disease, chemotherapy or radiation therapy are frequently insufficient for the treatment of unresectable or metastasized SGC. Therefore, new therapeutic approaches such as molecularly targeted therapy or the application of immune checkpoint inhibition enhance the treatment repertoire. Accordingly, comprehensive analyses of the genomic landscape and the tumor-microenvironment (TME) are of crucial importance in order to optimize and individualize SGC treatment. This manuscript combines the current scientific knowledge of the composition of the mutational landscape and the TME in SGCs harboring the potential for novel (immune-) targeted therapeutic strategies.
Collapse
Affiliation(s)
- Hanno M Witte
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Luebeck, 23538, Luebeck, Germany; Department of Hematology and Oncology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany; Institute of Pathology and Molecular Pathology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany.
| | - Niklas Gebauer
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Luebeck, 23538, Luebeck, Germany
| | - Konrad Steinestel
- Institute of Pathology and Molecular Pathology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany
| |
Collapse
|