1
|
Turashvili G, Hanley K. Malignant Brenner Tumor of the Ovary: A Critical Reappraisal. Int J Gynecol Pathol 2025; 44:182-192. [PMID: 39778112 DOI: 10.1097/pgp.0000000000001060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Malignant Brenner tumors (MBTs) are rare epithelial tumors of the ovary, most likely arising from benign and borderline Brenner tumors. MBTs may be misdiagnosed as other primary carcinomas or nonepithelial tumors of the ovary as well as metastatic carcinomas. Accurate diagnosis usually requires clinical-radiologic correlation, extensive sampling, and immunohistochemical studies. Treatment is not standardized and may include surgery with or without chemotherapy. More than half of MBTs are diagnosed at stage I, with 47.7% and at least 20% recurrence and mortality rates, respectively. Awareness of key diagnostic features and pitfalls is essential to differentiate MBT from its mimics and ensure optimal clinical management. This comprehensive review includes classification, etiopathogenesis, historical overview, epidemiology, clinical features, treatment, prognosis, gross pathology, key morphologic features, ancillary testing, and differential diagnostic considerations for ovarian MBTs.
Collapse
Affiliation(s)
- Gulisa Turashvili
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Krisztina Hanley
- Department of Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, Georgia
| |
Collapse
|
2
|
Xu J, Weisman PS. Immunohistochemistry as an adjunct for challenging histological patterns of borderline Brenner tumors: An illustrative study of 4 cases. Ann Diagn Pathol 2024; 72:152324. [PMID: 38733672 DOI: 10.1016/j.anndiagpath.2024.152324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024]
Abstract
Borderline Brenner tumors (BBT) have a range of morphology that shows considerable overlap with that of malignant Brenner tumors (MBT). In particular, two histological patterns of BBT can be particularly challenging: 1) BBT with intraepithelial carcinoma (BBT-IEC) and 2) BBT with a small nested pattern (BBT-SNP). BBT-IEC is characterized by a tumor with the low-power non-infiltrative silhouette of a conventional BBT, but with increased cytological atypia and mitotic activity similar to that of MBT. Conversely, BBT-SNP is characterized by a complex proliferation of small tumor nests that closely resemble the infiltrative growth pattern of MBT, but without the obligate cytologic atypia and mitotic activity of MBT. We suggest that the combination of p16, p53 and Ki-67 may be helpful in distinguishing these 2 patterns of BBT from both conventional BBT and from MBT. While both conventional BBT and BBT-IEC show a null pattern of p16 expression, our case of BBT-IEC showed aberrant p53 overexpression, albeit with a maturation pattern similar to that described for TP53 mutant mucinous ovarian carcinoma and differentiated vulvar intraepithelial neoplasia (dVIN). Similarly, while BBT-SNP shows an infiltrative-like growth pattern similar to that of MBT, our case also showed a wild-type pattern of p53 expression and a Ki-67 proliferative index similar to areas with conventional BBT histology. In conclusion, in our small case series, we show that the use of immunohistochemistry for p53 and Ki-67 may help to distinguish challenging patterns of BBT from MBT. Further studies are needed to validate this finding in a larger case cohort.
Collapse
Affiliation(s)
- Jin Xu
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States of America
| | - Paul S Weisman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States of America.
| |
Collapse
|
3
|
Somasegar S, Kertowidjojo E, Chui MH, Ramalingam P, Liu Y, Aviki E. Other rare ovarian cancers: Transitional cell carcinoma, malignant Brenner tumor, endometrioid carcinoma, mesothelioma, squamous cell carcinoma, sarcoma. DIAGNOSIS AND TREATMENT OF RARE GYNECOLOGIC CANCERS 2023:121-141. [DOI: 10.1016/b978-0-323-82938-0.00008-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
4
|
Kohlruss M, Krenauer M, Grosser B, Pfarr N, Jesinghaus M, Slotta-Huspenina J, Novotny A, Hapfelmeier A, Schmidt T, Steiger K, Gaida MM, Reiche M, Bauer L, Ott K, Weichert W, Keller G. Diverse 'just-right' levels of chromosomal instability and their clinical implications in neoadjuvant treated gastric cancer. Br J Cancer 2021; 125:1621-1631. [PMID: 34671125 PMCID: PMC8651679 DOI: 10.1038/s41416-021-01587-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The Cancer Genome Atlas (TCGA) consortium described EBV positivity(+), high microsatellite instability (MSI-H), genomic stability (GS) and chromosomal instability (CIN) as molecular subtypes in gastric carcinomas (GC). We investigated the predictive and prognostic value of these subtypes with emphasis on CIN in the context of neoadjuvant chemotherapy (CTx) in GC. METHODS TCGA subgroups were determined for 612 resected adenocarcinomas of the stomach and gastro-oesophageal junction (291 without, 321 with CTx) and 143 biopsies before CTx. EBV and MSI-H were analysed by standard assays. CIN was detected by multiplex PCRs analysing 22 microsatellite markers. Besides the TCGA classification, CIN was divided into four CIN-subgroups: low, moderate, substantial, high. Mutation profiling was performed for 52 tumours by next-generation sequencing. RESULTS EBV(+) (HR, 0.48; 95% CI, 0.23-1.02), MSI-H (HR, 0.56; 95% CI, 0.35-0.89) and GS (HR, 0.72; 95% CI, 0.45-1.13) were associated with increased survival compared to CIN in the resected tumours. Considering the extended CIN-classification, CIN-substantial was a negative prognostic factor in uni- and multivariable analysis in resected tumours with CTx (each p < 0.05). In biopsies before CTx, CIN-high predicted tumour regression (p = 0.026), but was not prognostically relevant. CONCLUSION A refined CIN classification reveals tumours with different biological characteristics and potential clinical implications.
Collapse
Affiliation(s)
- Meike Kohlruss
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Marie Krenauer
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Bianca Grosser
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany ,grid.419801.50000 0000 9312 0220Institute of Pathology and Molecular Diagnostics, University Hospital Augsburg, Augsburg, Germany
| | - Nicole Pfarr
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Moritz Jesinghaus
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany ,grid.411067.50000 0000 8584 9230Institute of Pathology, University Hospital Marburg, Marburg, Germany
| | - Julia Slotta-Huspenina
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Novotny
- grid.6936.a0000000123222966Department of Surgery, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Hapfelmeier
- grid.6936.a0000000123222966Institute of Medical Informatics, Statistics and Epidemiology, Technical University of Munich, Munich, Germany ,grid.6936.a0000000123222966Institute of General Practice and Health Services Research, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas Schmidt
- grid.7700.00000 0001 2190 4373Department of Surgery, University of Heidelberg, Heidelberg, Germany ,grid.411097.a0000 0000 8852 305XDepartment of Surgery, Universitätsklinikum Köln, Köln, Germany
| | - Katja Steiger
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany ,German Cancer Consortium (DKTK), Partner Site Munich, Institute of Pathology, Munich, Germany
| | - Matthias M. Gaida
- grid.7700.00000 0001 2190 4373Institute of Pathology, University of Heidelberg, Heidelberg, Germany ,grid.410607.4Institute of Pathology, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
| | - Magdalena Reiche
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Lukas Bauer
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Katja Ott
- grid.477776.20000 0004 0394 5800Department of Surgery, RoMed Klinikum Rosenheim, Rosenheim, Germany
| | - Wilko Weichert
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany ,German Cancer Consortium (DKTK), Partner Site Munich, Institute of Pathology, Munich, Germany
| | - Gisela Keller
- grid.6936.a0000000123222966Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
5
|
MDM2 amplification in malignant Brenner tumors may play a role in progression to malignancy and aid in separation from urothelial and other ovarian carcinomas. Hum Pathol 2021; 117:42-50. [PMID: 34391746 DOI: 10.1016/j.humpath.2021.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 11/20/2022]
Abstract
Malignant Brenner tumor (MBT) is diagnosed in the setting of invasive high-grade carcinoma with urothelial-like morphology and the presence of an adjacent benign Brenner tumor (BBT) or borderline Brenner tumor (BLBT). MDM2 amplification was recently detected by next-generation sequencing on a small number of MBTs, potentially significant for future targeted therapy. Experience is limited, however, and evaluation of widely available MDM2 immunohistochemistry (IHC) has not been performed to determine clinical utility. After confirming all diagnoses morphologically and immunohistochemically, we performed MDM2 IHC on 4 MBTs, 3 BLBTs, 26 BBTs, 142 high-grade serous carcinomas (HGSC), 6 ovarian endometrioid carcinomas (OEC) with urothelial-like morphology, and 49 high-grade urothelial carcinomas (HGUC). MDM2 IHC was considered positive with diffuse (>25%) nuclear reactivity; in cases of patchy staining (10-25% nuclear reactivity), MDM2 was considered equivocal. Positive staining in <10% of cells was considered negative. In cases with positive or equivocal staining, MDM2 amplification was evaluated by fluorescence in-situ hybridization (FISH). Three MBTs (75%) showed diffuse nuclear reactivity for MDM2 by IHC, a finding corroborated by amplification of MDM2 in all three cases. One MBT and 2 BLBTs showed equivocal MDM2 IHC, but all three were negative for MDM2 amplification. The final BLBT, as well as all BBTs, HGSC, OEC, and HGUC, were negative for MDM2. In conclusion, our limited cohort confirms MDM2 amplification in MBT and suggests that MDM2 IHC may have an influence in rare diagnostically challenging cases.
Collapse
|
6
|
Whole Exome Sequencing of Biliary Tubulopapillary Neoplasms Reveals Common Mutations in Chromatin Remodeling Genes. Cancers (Basel) 2021; 13:cancers13112742. [PMID: 34205964 PMCID: PMC8198366 DOI: 10.3390/cancers13112742] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Intraductal tubulopapillary neoplasms (ITPN) have recently been described as rare precursor lesions of pancreatic ductal adenocarcinoma and cholangiocarcinoma. Despite a high number of associated invasive adenocarcinomas at the time of diagnosis, patients with ITPN tend to have a much better clinical outcome than those with classical pancreato-biliary adenocarcinoma. Furthermore, rare molecular studies of ITPN show an unexpected lack of hotspot mutations in common driver genes of pancreato-biliary adenocarcinoma, including KRAS. This article reports the first large comprehensive and comparative molecular study of pancreato-biliary ITPN. In the absence of KRAS mutations, we found a high genetic heterogeneity with enrichment in core signaling pathways, including putative actionable genomic targets in one-third of the cases. Whereas, pancreatic ITPN demonstrates a highly distinct genetic profile, differing from classical pancreatic carcinogenesis, biliary ITPN and classical cholangiocarcinoma share common alterations in key genes of the chromatin remodeling pathway, and therefore, appear more closely related than pancreatic ITPN and classical pancreatic ductal adenocarcinoma PDAC. Abstract The molecular carcinogenesis of intraductal tubulopapillary neoplasms (ITPN), recently described as rare neoplasms in the pancreato-biliary tract with a favorable prognosis despite a high incidence of associated pancreato-biliary adenocarcinoma, is still poorly understood. To identify driver genes, chromosomal gains and losses, mutational signatures, key signaling pathways, and potential therapeutic targets, the molecular profile of 11 biliary and 6 pancreatic ITPNs, associated with invasive adenocarcinoma in 14/17 cases, are studied by whole exome sequencing (WES). The WES of 17 ITPNs reveals common copy number variants (CNVs) broadly distributed across the genome, with recurrent chromosomal deletions primarily in 1p36 and 9p21 affecting the tumor suppressors CHD5 and CDKN2A, respectively, and gains in 1q affecting the prominent oncogene AKT3. The identified somatic nucleotide variants (SNVs) involve few core signaling pathways despite high genetic heterogeneity with diverse mutational spectra: Chromatin remodeling, the cell cycle, and DNA damage/repair. An OncoKB search identifies putative actionable genomic targets in 35% of the cases (6/17), including recurrent missense mutations of the FGFR2 gene in biliary ITPNs (2/11, 18%). Our results show that somatic SNV in classical cancer genes, typically associated with pancreato-biliary carcinogenesis, were absent (KRAS, IDH1/2, GNAS, and others) to rare (TP53 and SMAD4, 6%, respectively) in ITPNs. Mutational signature pattern analysis reveals a predominance of an age-related pattern. Our findings highlight that biliary ITPN and classical cholangiocarcinoma display commonalities, in particular mutations in genes of the chromatin remodeling pathway, and appear, therefore, more closely related than pancreatic ITPN and classical pancreatic ductal adenocarcinoma.
Collapse
|
7
|
Recurrent urothelial carcinoma-like FGFR3 genomic alterations in malignant Brenner tumors of the ovary. Mod Pathol 2021; 34:983-993. [PMID: 33077920 DOI: 10.1038/s41379-020-00699-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/24/2020] [Indexed: 12/27/2022]
Abstract
Malignant Brenner tumor is a rare primary ovarian carcinoma subtype that may present diagnostic and therapeutic conundrums. Here, we characterize the genomics of 11 malignant Brenner tumors, which represented 0.1% of 14,153 clinically advanced ovarian carcinomas submitted for genomic profiling during the course of clinical care. At the time of molecular profiling, there was no evidence of a primary urothelial carcinoma of the urinary tract in any case. Cases with transitional-like morphologic features in the setting of variant ovarian serous or endometrioid carcinoma morphology were excluded from the final cohort. Malignant Brenner tumors exhibited CDKN2A/2B loss and oncogenic FGFR1/3 genomic alterations in 55% of cases, respectively; including recurrent FGFR3 S249C or FGFR3-TACC3 fusion in 45% of cases. FGFR3-mutated cases had an associated benign or borderline Brenner tumor pre-cursor components, further confirming the diagnosis and the ovarian site of origin. Malignant Brenner tumors were microsatellite stable, had low tumor mutational burden and exhibited no evidence of homologous recombination deficiency. PIK3CA mutations were enriched with FGFR3 alterations, while FGFR3 wild-type cases featured MDM2 amplification or TP53 mutations. The FGFR3 S249C short variant mutation was absent in 14,142 non-Brenner, ovarian carcinomas subtypes. In contrast to malignant Brenner tumors, FGFR1/2/3 alterations were present in ~5% of non-Brenner, ovarian serous, clear cell and endometrioid carcinoma subtypes, most often as FGFR1 amplification in serous carcinoma or FGFR2 short variant alterations in clear cell or endometrioid carcinomas, respectively. Finally, malignant Brenner tumors had overall distinct genomic signatures compared to FGFR-mutated ovarian serous, endometrioid, and clear cell carcinoma subtypes. This study provides insights into the molecular pathogenesis of malignant Brenner tumors, contrasts the extent of FGFR1/2/3 alterations in ovarian serous, clear cell and endometrioid carcinomas and emphasizes the potential value of novel and FDA-approved, anti-FGFR inhibitors, such as erdafitinib and pemigatinib, in refractory, FGFR3-mutated malignant Brenner tumors.
Collapse
|
8
|
Miree O, Srivastava SK, Dasgupta S, Singh S, Rocconi R, Singh AP. Current and Futuristic Roadmap of Ovarian Cancer Management: An Overview. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1330:1-19. [PMID: 34339027 DOI: 10.1007/978-3-030-73359-9_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy among women worldwide. In most cases, it is diagnosed late at an advanced stage and does not respond well to existing therapies leading to its poor prognosis. In addition, other factors including epidemiological, complex histological diversity, multiple molecular alterations, and overlapping signaling pathways are also important contributors to poor disease outcome. Efforts have continued to develop a deeper understanding of the molecular pathogenesis and altered signaling nodes that provide hope for better clinical management through the development of novel approaches for early diagnosis, disease subtyping, prognosis, and therapy. In this chapter, we provide a detailed overview of OC and its histological subtypes and discuss prevalent molecular aberrations and active signaling pathways that drive OC progression. We also summarize various diagnostic and prognostic markers and therapeutic approaches currently being employed and discuss emerging findings that hold the potential to change the future course of OC management.
Collapse
Affiliation(s)
- Orlandric Miree
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA.,Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Sanjeev Kumar Srivastava
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA.,Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Santanu Dasgupta
- Department of Medicine, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - Seema Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA.,Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Rodney Rocconi
- Division of Gynecologic Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Ajay Pratap Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, AL, USA. .,Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA. .,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA.
| |
Collapse
|
9
|
Dantes Z, Yen HY, Pfarr N, Winter C, Steiger K, Muckenhuber A, Hennig A, Lange S, Engleitner T, Öllinger R, Maresch R, Orben F, Heid I, Kaissis G, Shi K, Topping G, Stögbauer F, Wirth M, Peschke K, Papargyriou A, Rezaee-Oghazi M, Feldmann K, Schäfer AP, Ranjan R, Lubeseder-Martellato C, Stange DE, Welsch T, Martignoni M, Ceyhan GO, Friess H, Herner A, Liotta L, Treiber M, von Figura G, Abdelhafez M, Klare P, Schlag C, Algül H, Siveke J, Braren R, Weirich G, Weichert W, Saur D, Rad R, Schmid RM, Schneider G, Reichert M. Implementing cell-free DNA of pancreatic cancer patient-derived organoids for personalized oncology. JCI Insight 2020; 5:137809. [PMID: 32614802 DOI: 10.1172/jci.insight.137809] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/24/2020] [Indexed: 01/05/2023] Open
Abstract
One of the major challenges in using pancreatic cancer patient-derived organoids (PDOs) in precision oncology is the time from biopsy to functional characterization. This is particularly true for endoscopic ultrasound-guided fine-needle aspiration biopsies, typically resulting in specimens with limited tumor cell yield. Here, we tested conditioned media of individual PDOs for cell-free DNA to detect driver mutations already early on during the expansion process to accelerate the genetic characterization of PDOs as well as subsequent functional testing. Importantly, genetic alterations detected in the PDO supernatant, collected as early as 72 hours after biopsy, recapitulate the mutational profile of the primary tumor, indicating suitability of this approach to subject PDOs to drug testing in a reduced time frame. In addition, we demonstrated that this workflow was practicable, even in patients for whom the amount of tumor material was not sufficient for molecular characterization by established means. Together, our findings demonstrate that generating PDOs from very limited biopsy material permits molecular profiling and drug testing. With our approach, this can be achieved in a rapid and feasible fashion with broad implications in clinical practice.
Collapse
Affiliation(s)
- Zahra Dantes
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Hsi-Yu Yen
- Institute of Pathology.,Comparative Experimental Pathology, and
| | | | - Christof Winter
- Institute of Clinical Chemistry and Pathobiochemistry, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), partner site Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katja Steiger
- Institute of Pathology.,Comparative Experimental Pathology, and
| | | | - Alexander Hennig
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technical University of Dresden, Dresden, Germany
| | - Sebastian Lange
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Thomas Engleitner
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Rupert Öllinger
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Roman Maresch
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Felix Orben
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | | | | | - Kuangyu Shi
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
| | - Geoffrey Topping
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
| | | | - Matthias Wirth
- Medical Department, Division of Hematology and Oncology at Campus Benjamin Franklin, Charité, Berlin, Germany
| | - Katja Peschke
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | | | | | - Karin Feldmann
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Arlett Pg Schäfer
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Raphela Ranjan
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | | | - Daniel E Stange
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technical University of Dresden, Dresden, Germany.,DKTK, partner site Dresden, Germany
| | - Thilo Welsch
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technical University of Dresden, Dresden, Germany.,DKTK, partner site Dresden, Germany
| | - Marc Martignoni
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Güralp O Ceyhan
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Alexander Herner
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Lucia Liotta
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Matthias Treiber
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Guido von Figura
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | | | - Peter Klare
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Christoph Schlag
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Hana Algül
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Jens Siveke
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, Essen, Germany.,Division of Solid Tumor Translational Oncology, DKTK, partner site Essen, Germany
| | - Rickmer Braren
- German Cancer Consortium (DKTK), partner site Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technical University of Dresden, Dresden, Germany
| | | | - Wilko Weichert
- Institute of Pathology.,German Cancer Consortium (DKTK), partner site Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dieter Saur
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar.,German Cancer Consortium (DKTK), partner site Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Roland Rad
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar.,German Cancer Consortium (DKTK), partner site Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Roland M Schmid
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar
| | - Günter Schneider
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar.,German Cancer Consortium (DKTK), partner site Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maximilian Reichert
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar.,German Cancer Consortium (DKTK), partner site Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
10
|
Impact of Tumor Localization and Molecular Subtypes on the Prognostic and Predictive Significance of p53 Expression in Gastric Cancer. Cancers (Basel) 2020; 12:cancers12061689. [PMID: 32630478 PMCID: PMC7352381 DOI: 10.3390/cancers12061689] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/10/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022] Open
Abstract
We investigated the prognostic and predictive impact of p53 expression for gastric cancer (GC) patients treated without or with preoperative chemotherapy (CTx) and its relationship with specific molecular GC subtypes. Specimens from 694 GC patients (562 surgical resection specimens without or after CTx, 132 biopsies before CTx) were analyzed by p53 immunohistochemistry. High (H) and low (L) microsatellite instability (MSI) and Epstein–Barr virus positivity were determined previously. Our results show that aberrant p53 expression was a negative prognostic factor in uni- and multivariable analysis in the resection specimens cohort (each p < 0.01). Subgroup analysis showed the strongest prognostic effect for patients with distally located tumors or no CTx treatment. In the biopsy cohort before CTx, p53 did not predict response or survival. p53 expression was significantly different among the molecular subtypes in surgical resection and bioptic specimens with strong association of altered p53 with MSI-L. Patients with MSI-H and aberrant p53 showed the worst survival in the biopsy cohort. In conclusion, the prognostic impact of p53 in GC differs according to tumor localization and CTx. Altered p53 is characteristic for MSI-L, and the p53 status in biopsies before CTx delineates MSI-H subtypes with inverse prognostic impact.
Collapse
|
11
|
Wang L, Allison D, Shukla PS. Amplification of MDM2 and Loss of p16 Expression: Do They Have a Role in Malignant Transformation of Ovarian Brenner Tumor? Am J Clin Pathol 2020; 154:133-141. [PMID: 32322877 DOI: 10.1093/ajcp/aqaa033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES To review the significance of MDM2 and cyclin D1 expression and loss of p16 expression in malignant and borderline Brenner tumors (BTs) of the ovary. METHODS We describe 2 new cases of ovarian BT, 1 malignant and 1 borderline. We studied MDM2, p16, and cyclin D1 expression by immunohistochemistry in the benign, borderline, and malignant components of these 2 cases and in 5 additional cases of benign BT. We also reviewed and summarized the literature on the clinical, immunohistochemical and molecular characteristics of borderline and malignant BTs (BdBTs and MBTs). RESULTS Nuclear expression of MDM2 was seen only in the MBT. Loss of p16 expression was seen in both BdBT and MBT. Cyclin D1 expression was in proportion to the degree of malignancy. Amplification of MDM2, loss of CDKN2A (p16-encoding gene), and amplification of CCND1 (cyclin D1-encoding gene) were confirmed by commercial next-generation sequencing in the case of MBT. CONCLUSIONS We are the first to report immunohistochemical expression of MDM2 in an MBT. Amplification of MDM2 and loss of p16 expression may have a role in malignant transformation of BT.
Collapse
Affiliation(s)
- Lucy Wang
- Department of Pathology, NYU Langone Medical Center, New York, NY
| | - Douglas Allison
- Department of Pathology, NYU Langone Medical Center, New York, NY
| | | |
Collapse
|
12
|
Moret S, Just PA. [Histoseminary: Epithelial ovarian tumors - Case n o 6]. Ann Pathol 2020; 40:214-219. [PMID: 32331810 DOI: 10.1016/j.annpat.2020.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 02/18/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Sandrine Moret
- Service de pathologie, AP-HP, centre - université de Paris, hôpital Cochin, 27, rue du Faubourg-Saint-Jacques, 75014 Paris, France
| | - Pierre-Alexandre Just
- Service de pathologie, AP-HP, centre - université de Paris, hôpital Cochin, 27, rue du Faubourg-Saint-Jacques, 75014 Paris, France.
| |
Collapse
|
13
|
Mogler C, Koschny R, Heilig CE, Frohling S, Schirmacher P, Weichert W, Pfarr N. Molecular characterization of hepatic epithelioid hemangioendothelioma reveals alterations in various genes involved in DNA repair, epigenetic regulation, signaling pathways, and cell cycle control. Genes Chromosomes Cancer 2019; 59:106-110. [PMID: 31390105 DOI: 10.1002/gcc.22795] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/31/2022] Open
Abstract
Epithelioid hemangioendotheliomas (EHE) of the liver are rare, low-malignant vascular tumors whose molecular pathogenesis is incompletely understood. The diagnosis of EHE is challenging, and the course of the disease can be highly variable. Therapeutic options for EHE are limited, including resection of primary and metastatic tumors, organ transplantation and rather ineffective systemic approaches. Driver mutations have been reported (fusion transcripts of either YAP-TFE3 or WWTR1-CAMTA1) but comprehensive molecular profiling has not been performed. Our aim was to molecularly characterize hepatic EHE to identify new molecular targets. Eight primary hepatic EHE were analyzed by next-generation sequencing using a 409-gene panel. The majority of primary hepatic EHE revealed a low number of mutations. Genes that were mutated primarily are involved in DNA repair, epigenetic regulation, signaling pathways and cell cycle control, indicating that EHE present with mutations in various functions. Although only detecting a low mutation rate, a comparison with comprehensive databases (target db V3) revealed mutations in five genes with putative therapeutical options. Therefore, our findings help to shed light on the molecular background of EHE and might pave the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Carolin Mogler
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Ronald Koschny
- Internal Medicine, Department of Gastroenterology, University Hospital Heidelberg, Heidelberg, Germany
| | - Christoph E Heilig
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and DKFZ, Heidelberg, Germany
| | - Stefan Frohling
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and DKFZ, Heidelberg, Germany.,DKFZ-Heidelberg Center for Personalized Oncology (HIPO), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | | | - Wilko Weichert
- Institute of Pathology, Technical University Munich, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Nicole Pfarr
- Institute of Pathology, Technical University Munich, Munich, Germany
| |
Collapse
|
14
|
Several genotypes, one phenotype: PIK3CA/AKT1 mutation-negative hidradenoma papilliferum show genetic lesions in other components of the signalling network. Pathology 2019; 51:362-368. [PMID: 31010589 DOI: 10.1016/j.pathol.2019.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/06/2018] [Accepted: 01/23/2019] [Indexed: 12/24/2022]
Abstract
About 60-70% of hidradenoma papilliferum (HP), a benign tumour of the anogenital region, were recently described to harbour mutations in major driver genes of the PI3K/AKT/MAPK-signalling pathways. However, the underlying genetic defects of the non-mutant cases are still unknown. Using a 409 gene panel, we employed targeted next generation sequencing to investigate the mutational landscape in a cohort of seven PI3K/AKT-negative cases and five cases with known hotspot mutations in either PIK3CA or AKT1. In total, we identified 29 mutations in 22 of 409 genes. The four cases with PIK3CA hotspot mutations carried no or only few additional mutations. The AKT1 hotspot mutated case harboured additional mutations in four genes (SYNE1, ADAMTS20, EP400 and CASC5). At least two of these genes are involved in or contribute to the PI3K/AKT-pathway. In the seven non-hotspot mutated cases we observed 18 mutations. Each case carried at least one mutation in a gene contributing to or involved in PI3K/AKT-signalling. Affected genes were PIK3CA (n=1, non-hotspot mutation), PIK3R1 (n=3), SYNE1, AR, IL6ST, PDGFRB, KMT2C, AR, BTK, DST, KAT6A, BRD3, RNF213, USP9X, ADGRB3, MAGI1, and IL7R (each gene mutated once). The identified PIK3CA and PIK3R1 mutations lead to constitutive activated PI3K/AKT-signalling. In conclusion, we demonstrate the genetic basis of HP in all cases. Our data suggest that tumourigenic alterations in the PI3K/AKT-pathway are indispensable in HP and establish a homogenous morphomolecular entity with a functionally converging and selecting tumourigenic mechanism.
Collapse
|
15
|
Konukiewitz B, Jesinghaus M, Steiger K, Schlitter AM, Kasajima A, Sipos B, Zamboni G, Weichert W, Pfarr N, Klöppel G. Pancreatic neuroendocrine carcinomas reveal a closer relationship to ductal adenocarcinomas than to neuroendocrine tumors G3. Hum Pathol 2018; 77:70-79. [PMID: 29596894 DOI: 10.1016/j.humpath.2018.03.018] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 12/29/2022]
Abstract
Pancreatic neuroendocrine carcinoma is a rare aggressive tumor commonly harboring TP53 and RB1 alterations and lacking neuroendocrine-related genetic changes such as mutations in MEN1 and ATRX/DAXX. Little is known about its genetic profile with regard to that of pancreatic ductal adenocarcinoma. We therefore conducted a detailed genetic study in 12 pancreatic neuroendocrine carcinomas of large cell (n = 9) and small cell type (n = 3) using massive parallel sequencing applying a 409-gene panel on an Ion Torrent system. The genetic data were compared with known data of pancreatic ductal adenocarcinoma and correlated with exocrine lineage marker expression. A similar analysis was performed in 11 pancreatic neuroendocrine tumors G3. Neuroendocrine carcinomas harbored 63 somatic mutations in 45 different genes, affecting most commonly TP53 (8/12 cases), KRAS (5/12 cases), and RB1 (loss of expression with or without deletion in 4/12 cases). Five carcinomas had both TP53 and KRAS mutations. Neuroendocrine tumors G3 only shared singular mutations in 5 different genes with neuroendocrine carcinomas, including TP53, CDKN2A, ARID1A, LRP1B, and APC, affecting 5 different cases. Most KRAS-positive neuroendocrine carcinomas also expressed MUC1 (4/5) and carcinoembryonic antigen (3/5) as markers of ductal differentiation. Our data indicate that almost half of the pancreatic neuroendocrine carcinomas are genetically and phenotypically related to pancreatic ductal adenocarcinoma, and might therefore respond to chemotherapies targeting the latter carcinomas.
Collapse
Affiliation(s)
- Björn Konukiewitz
- Institute of Pathology, Technical University of Munich, 81675 Munich, Germany.
| | - Moritz Jesinghaus
- Institute of Pathology, Technical University of Munich, 81675 Munich, Germany.
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich, 81675 Munich, Germany.
| | | | - Atsuko Kasajima
- Institute of Pathology, Technical University of Munich, 81675 Munich, Germany.
| | - Bence Sipos
- Institute of Pathology, University Hospital of Tuebingen, 72076 Tuebingen, Germany.
| | - Giuseppe Zamboni
- Institute of Pathology, Sacro Cuore Don Calabria Hospital, 37024 Negrar, Verona, Italy.
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, 81675 Munich, Germany.
| | - Nicole Pfarr
- Institute of Pathology, Technical University of Munich, 81675 Munich, Germany.
| | - Günter Klöppel
- Institute of Pathology, Technical University of Munich, 81675 Munich, Germany.
| |
Collapse
|
16
|
Jesinghaus M, Konukiewitz B, Foersch S, Stenzinger A, Steiger K, Muckenhuber A, Groß C, Mollenhauer M, Roth W, Detlefsen S, Weichert W, Klöppel G, Pfarr N, Schlitter AM. Appendiceal goblet cell carcinoids and adenocarcinomas ex-goblet cell carcinoid are genetically distinct from primary colorectal-type adenocarcinoma of the appendix. Mod Pathol 2018; 31:829-839. [PMID: 29327707 DOI: 10.1038/modpathol.2017.184] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/24/2017] [Accepted: 11/08/2017] [Indexed: 12/23/2022]
Abstract
The appendix gives rise to goblet cell carcinoids, which represent special carcinomas with distinct biological and histological features. Their genetic background and molecular relationship to colorectal adenocarcinoma is largely unknown. We therefore performed a next-generation sequencing analysis of 25 appendiceal carcinomas including 11 goblet cell carcinoids, 7 adenocarcinomas ex-goblet cell carcinoid, and 7 primary colorectal-type adenocarcinomas, using a modified Colorectal Cancer specific Panel comprising 32 genes linked to colorectal and neuroendocrine tumorigenesis. The mutational profiles of these neoplasms were compared with those of conventional adenocarcinomas, mixed adenoneuroendocrine carcinomas, and neuroendocrine carcinomas of the colorectum. In addition, a large-scale pan-cancer sequencing panel covering 409 genes was applied to selected cases of goblet cell carcinoid/adenocarcinoma ex-goblet cell carcinoid (n=2, respectively). Mutations in colorectal cancer-related genes (eg, TP53, KRAS, APC) were rare to absent in both, goblet cell carcinoids and adenocarcinomas ex-goblet cell carcinoid, but frequent in primary colorectal-type adenocarcinomas of the appendix. Additional large-scale sequencing of selected goblet cell carcinoids and adenocarcinomas ex-goblet cell carcinoid revealed mutations in Wnt-signaling-associated genes (USP9X, NOTCH1, CTNNA1, CTNNB1, TRRAP). These data suggest that appendiceal goblet cell carcinoids and adenocarcinomas ex-goblet cell carcinoid constitute a morphomolecular entity, histologically and genetically distinct from appendiceal colorectal-type adenocarcinomas and its colorectal counterparts. Altered Wnt-signaling associated genes, apart from APC, may act as potential drivers of these neoplasms. The absence of KRAS/NRAS mutations might render some of these tumors eligible for anti-EGFR directed therapy regimens.
Collapse
Affiliation(s)
- Moritz Jesinghaus
- Institute of Pathology, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Björn Konukiewitz
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | | | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Alexander Muckenhuber
- Institute of Pathology, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Claudia Groß
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | | | - Wilfried Roth
- Institute of Pathology, University Hospital Mainz, Mainz, Germany
| | - Sönke Detlefsen
- Department of Clinical Pathology, University Hospital Odense, Odense, Denmark
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Günter Klöppel
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Nicole Pfarr
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Anna Melissa Schlitter
- Institute of Pathology, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
17
|
Ovarian Cancers: Genetic Abnormalities, Tumor Heterogeneity and Progression, Clonal Evolution and Cancer Stem Cells. MEDICINES 2018; 5:medicines5010016. [PMID: 29389895 PMCID: PMC5874581 DOI: 10.3390/medicines5010016] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 02/07/2023]
Abstract
Four main histological subtypes of ovarian cancer exist: serous (the most frequent), endometrioid, mucinous and clear cell; in each subtype, low and high grade. The large majority of ovarian cancers are diagnosed as high-grade serous ovarian cancers (HGS-OvCas). TP53 is the most frequently mutated gene in HGS-OvCas; about 50% of these tumors displayed defective homologous recombination due to germline and somatic BRCA mutations, epigenetic inactivation of BRCA and abnormalities of DNA repair genes; somatic copy number alterations are frequent in these tumors and some of them are associated with prognosis; defective NOTCH, RAS/MEK, PI3K and FOXM1 pathway signaling is frequent. Other histological subtypes were characterized by a different mutational spectrum: LGS-OvCas have increased frequency of BRAF and RAS mutations; mucinous cancers have mutation in ARID1A, PIK3CA, PTEN, CTNNB1 and RAS. Intensive research was focused to characterize ovarian cancer stem cells, based on positivity for some markers, including CD133, CD44, CD117, CD24, EpCAM, LY6A, ALDH1. Ovarian cancer cells have an intrinsic plasticity, thus explaining that in a single tumor more than one cell subpopulation, may exhibit tumor-initiating capacity. The improvements in our understanding of the molecular and cellular basis of ovarian cancers should lead to more efficacious treatments.
Collapse
|