Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 48
1.
Nature ; 628(8007): 442-449, 2024 Apr.
Article En | MEDLINE | ID: mdl-38538798

Whereas oncogenes can potentially be inhibited with small molecules, the loss of tumour suppressors is more common and is problematic because the tumour-suppressor proteins are no longer present to be targeted. Notable examples include SMARCB1-mutant cancers, which are highly lethal malignancies driven by the inactivation of a subunit of SWI/SNF (also known as BAF) chromatin-remodelling complexes. Here, to generate mechanistic insights into the consequences of SMARCB1 mutation and to identify vulnerabilities, we contributed 14 SMARCB1-mutant cell lines to a near genome-wide CRISPR screen as part of the Cancer Dependency Map Project1-3. We report that the little-studied gene DDB1-CUL4-associated factor 5 (DCAF5) is required for the survival of SMARCB1-mutant cancers. We show that DCAF5 has a quality-control function for SWI/SNF complexes and promotes the degradation of incompletely assembled SWI/SNF complexes in the absence of SMARCB1. After depletion of DCAF5, SMARCB1-deficient SWI/SNF complexes reaccumulate, bind to target loci and restore SWI/SNF-mediated gene expression to levels that are sufficient to reverse the cancer state, including in vivo. Consequently, cancer results not from the loss of SMARCB1 function per se, but rather from DCAF5-mediated degradation of SWI/SNF complexes. These data indicate that therapeutic targeting of ubiquitin-mediated quality-control factors may effectively reverse the malignant state of some cancers driven by disruption of tumour suppressor complexes.


Multiprotein Complexes , Mutation , Neoplasms , SMARCB1 Protein , Animals , Female , Humans , Male , Mice , Cell Line, Tumor , CRISPR-Cas Systems , Gene Editing , Neoplasms/genetics , Neoplasms/metabolism , SMARCB1 Protein/deficiency , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Proteolysis , Ubiquitin/metabolism
2.
Am J Surg Pathol ; 46(2): 169-178, 2022 02 01.
Article En | MEDLINE | ID: mdl-34265804

So-called primary yolk sac tumors of the vulva are very rare and often have an aggressive disease course. Their molecular features have not been previously characterized. There is also a well-documented group of SMARCB1 (INI-1)-deficient vulvar neoplasms, which includes proximal-type epithelioid sarcoma and myoepithelial carcinoma. Until now, "vulvar yolk sac tumors" and SMARCB1-deficient neoplasms were considered unrelated diseases. After reviewing an index case of a vulvar yolk sac tumor with loss of SMARCB1 by immunohistochemistry, we retrospectively identified 2 additional cases diagnosed as vulvar yolk sac tumors. Patient ages were 34, 32, and 25 years old, and 2 tumors were associated with a pregnancy. All 3 cases showed morphology typical of a yolk sac tumor, and by immunohistochemistry all were positive for SALL4, glypican-3, keratins, and lacked CD34 positivity. All tumors also demonstrated loss of SMARCB1 in tumor cells. Targeted molecular profiling was performed in 2 cases and identified 2 copy deletion of SMARCB1, without genomic alterations typically seen in gonadal yolk sac tumors. In the third case, isochromosome 12p was not identified by fluorescence in situ hybridization. All 3 patients had either local recurrences or distant metastases, and 2 died of disease. One patient had progressive disease while receiving the enhancer of zeste homolog 2 inhibitor tazemetostat. Overall, these findings suggest that vulvar tumors with pure yolk sac-like morphology may represent morphologic variants of SMARCB1-deficient tumors and not veritable germ cell neoplasia. This potential reclassification may have both prognostic and treatment implications and warrants study of additional extragonadal yolk sac tumors.


Biomarkers, Tumor/deficiency , Endodermal Sinus Tumor/chemistry , SMARCB1 Protein/deficiency , Vulvar Neoplasms/chemistry , Adult , Biomarkers, Tumor/genetics , Chromosome Deletion , Chromosomes, Human, Pair 12 , Disease Progression , Endodermal Sinus Tumor/genetics , Endodermal Sinus Tumor/secondary , Endodermal Sinus Tumor/surgery , Female , Gene Deletion , Humans , Neoplasm Recurrence, Local , Retrospective Studies , SMARCB1 Protein/genetics , Treatment Outcome , Vulvar Neoplasms/genetics , Vulvar Neoplasms/pathology , Vulvar Neoplasms/surgery
3.
Virchows Arch ; 479(6): 1209-1219, 2021 Dec.
Article En | MEDLINE | ID: mdl-34350470

Mimickers of neuroendocrine neoplasms (NEN) include a number of important pitfall tumors. Here, we describe our experience with mesenchymal mimics of NENs to illustrate their spectrum and draw the attention particularly to a group of mesenchymal/non-epithelial neoplasms (MN) that combine epithelioid histology with neuroendocrine (NE-) features and peculiar genetic abnormalities. In a consultation series of 4498 cases collected between 2009 and 2021, 2099 neoplasms expressing synaptophysin and/or chromograninA were reviewed and analyzed. A total of 364 (18%) were diagnosed as non-NENs, while the remaining tumors were NEN. The group of mesenchymal/non-epithelial neoplasms with NE-features (MN-NE) included 31/364 (8%) cases. These mostly malignant neoplasms showed an epithelioid morphology. While all but one tumor expressed synaptophysin, mostly patchy, only 10/29 (34%) co-expressed chromograninA. A total of 13/31 (42%) of the MN-NE showed EWSR1-related gene fusions (6 Ewing sarcomas, 5 clear cell sarcomas, and 1 desmoplastic small round cell tumor, 1 neoplasm with FUS-CREM gene fusion) and 7 (23%) were SWI/SNF (SMARCB1 or SMARCA4)-deficient neoplasms. The remaining MN-NE included synovial sarcoma, sclerosing epithelioid mesenchymal neoplasm, melanoma, alveolar soft part sarcoma, solitary fibrous tumor, and chordoma. A total of 27/31 MN-NE were from the last 8 years, and 6 of them were located in the pancreas. Eleven MN-NE were initially diagnosed as neuroendocrine carcinomas (NECs). MN-NE with epithelioid features play an increasing role as mimickers of NECs. They mostly belong to tumors with gene fusions involving the EWSR1 gene, or with SWI/SNF complex deficiency. Synaptophysin expression is mostly patchy and chromograninA expression is infrequent in MN-NE of this series and data extracted from literature.


Biomarkers, Tumor/deficiency , Biomarkers, Tumor/genetics , Carcinoma, Neuroendocrine/genetics , DNA Helicases/deficiency , Gene Fusion , Neoplasms, Connective Tissue/genetics , Nuclear Proteins/deficiency , RNA-Binding Protein EWS/genetics , SMARCB1 Protein/deficiency , Transcription Factors/deficiency , Carcinoma, Neuroendocrine/chemistry , Carcinoma, Neuroendocrine/pathology , Chromogranin A/analysis , Cyclic AMP Response Element Modulator/genetics , Decision Support Techniques , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Neoplasms, Connective Tissue/chemistry , Neoplasms, Connective Tissue/pathology , Predictive Value of Tests , RNA-Binding Protein FUS/genetics , Synaptophysin/analysis
4.
Sci Rep ; 11(1): 12893, 2021 06 18.
Article En | MEDLINE | ID: mdl-34145313

Atypical Teratoid Rhabdoid Tumor (AT/RT) is a rare pediatric central nervous system cancer often characterized by deletion or mutation of SMARCB1, a tumor suppressor gene. In this study, we found that SMARCB1 regulates Human Endogenous Retrovirus K (HERV-K, subtype HML-2) expression. HML-2 is a repetitive element scattered throughout the human genome, encoding several intact viral proteins that have been associated with stem cell maintenance and tumorigenesis. We found HML-2 env expression in both the intracellular and extracellular compartments in all AT/RT cell lines (n = 4) and in 95% of AT/RT patient tissues (n = 37) evaluated. SMARCB1 knock-down in neural stem cells (NSCs) led to an upregulation of HML-2 transcription. We found that SMARCB1 binds adjacent to the HML-2 promoter, repressing its transcription via chromatin immunoprecipitation; restoration of SMARCB1 expression in AT/RT cell lines significantly downregulated HML-2 expression. Further, targeted downregulation of HML-2 transcription via CRISPR-dCas9 coupled with suppressor proteins led to cellular dispersion, decreased proliferation, and cell death in vitro. HML-2 knock-down with shRNA, siRNA, and CRISPR-dCas9 significantly decreased Ras expression as measured by qRT-PCR, suggesting that HML-2 modulates MAPK/ERK signaling in AT/RT cells. Overexpression of NRAS was sufficient to restore cellular proliferation, and MYC, a transcription factor downstream of NRAS, was bound to the HERV-K LTR significantly more in the absence of SMARCB1 expression in AT/RT cells. We show a mechanism by which these undifferentiated tumors remain pluripotent, and we demonstrate that their formation is aided by aberrant HML-2 activation, which is dependent on SMARCB1 and its interaction with MYC.


Cell Transformation, Neoplastic/genetics , Endogenous Retroviruses/genetics , Rhabdoid Tumor/etiology , Rhabdoid Tumor/pathology , SMARCB1 Protein/deficiency , Sequence Deletion , Virus Activation/genetics , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation , Cell-Derived Microparticles/metabolism , Disease Susceptibility , GTP Phosphohydrolases/metabolism , Gene Expression Regulation , Humans , Membrane Proteins/metabolism , Repetitive Sequences, Nucleic Acid , Signal Transduction
5.
Histopathology ; 79(4): 629-641, 2021 Oct.
Article En | MEDLINE | ID: mdl-33932047

AIMS: Complete loss of SMARCB1/INI1 in soft-tissue tumours such as malignant rhabdoid tumour, epithelioid sarcoma, myoepithelial tumour of soft tissue and extraskeletal myxoid chondrosarcoma is often associated with high-grade malignancy and poor prognosis. The diagnosis is sometimes challenging, owing to histological similarities, so careful differential diagnosis is required. Therefore, soft-tissue tumours with complete SMARCB1/INI1 loss could potentially include an unknown entity. METHODS AND RESULTS: We analysed 160 cases of SMARCB1/INI1-deficient soft-tissue tumour, and found 14 cases that were not classifiable into already existing categories and had common clinical and histological features. These involved two male and 12 female patients, ranging in age from 20 years to 61 years. The tumours were located in the the puboinguinal region (n = 13) and pelvic cavity (n = 1). Histologically, the tumours showed relatively uniform epithelioid to spindle-shaped cells with myxoid stroma. All tumours showed immunoreactivity for brachyury, epithelial membrane antigen, and progesterone receptor, and 12 of 14 cases did so for oestrogen receptor. Variable positive staining for α-smooth muscle actin, S100 and glial fibrillary acidic protein (GFAP) was seen. NR4A3 and EWSR1 gene rearrangements were not detected in 13 and 11 examined cases, respectively. Clinical follow-up data for the 14 patients showed that 13 were alive without disease and one had been lost to follow-up; four patients developed local recurrence and/or metastases. CONCLUSION: The designation 'myxoepithelioid tumour with choroid features' (METC) was proposed as a tumour with intermediate malignancy controllable with appropriate treatment, including the entity of myoepithelioma-like tumour of the vulvar region. METC represents a novel and independent subset that is histologically, biologically and clinically distinct from already existing SMARCB1/INI1-deficient soft-tissue tumours.


SMARCB1 Protein/deficiency , Soft Tissue Neoplasms/diagnosis , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/pathology , Adult , Diagnosis, Differential , Female , Humans , Immunohistochemistry , Male , Middle Aged , SMARCB1 Protein/genetics , Young Adult
6.
J Pathol ; 255(1): 1-15, 2021 09.
Article En | MEDLINE | ID: mdl-33999421

Extracranial rhabdoid tumours (ECRTs) are an aggressive malignancy of infancy and early childhood. The vast majority of cases demonstrate inactivation of SMARCB1 (ECRTSMARCB1 ) on a background of a remarkably stable genome, a low mutational burden, and no other recurrent mutations. Rarely, ECRTs can harbour the alternative inactivation of SMARCA4 (ECRTSMARCA4 ) instead of SMARCB1. However, very few ECRTSMARCA4 cases have been published to date, and a systematic characterization of ECRTSMARCA4 is missing from the literature. In this study, we report the clinical, pathological, and genomic features of additional cases of ECRTSMARCA4 and show that they are comparable to those of ECRTSMARCB1. We also assess whether ECRTSMARCB1 , ECRTSMARCA4 , and small cell carcinomas of the ovary, hypercalcaemic type (SCCOHT) represent distinct or overlapping entities at a molecular level. Using DNA methylation and transcriptomics-based tumour classification approaches, we demonstrate that ECRTSMARCA4 display molecular features intermediate between SCCOHT and ECRTSMARCB1 ; however, ECRTSMARCA4 appear to be more closely related to SCCOHT by DNA methylation. Conversely, both transcriptomics and DNA methylation show a larger gap between SCCOHT and ECRTSMARCB1 , potentially supporting their continuous separate classification. Lastly, we show that ECRTSMARCA4 display concomitant lack of SMARCA4 (BRG1) and SMARCA2 (BRM) expression at the protein level, similar to what is seen in SCCOHT. Overall, these results expand our knowledge on this rare tumour type and explore the similarities and differences among entities from the 'rhabdoid tumour' spectrum. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


DNA Helicases/deficiency , Nuclear Proteins/deficiency , Rhabdoid Tumor/genetics , Rhabdoid Tumor/pathology , Transcription Factors/deficiency , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/pathology , Child, Preschool , DNA Helicases/genetics , Female , Humans , Infant , Male , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , SMARCB1 Protein/deficiency , SMARCB1 Protein/genetics , Transcription Factors/genetics
7.
Hum Pathol ; 112: 1-8, 2021 06.
Article En | MEDLINE | ID: mdl-33741347

Loss of SMARCB1 protein expression has recently been identified in a variety of tumor types such as poorly differentiated chordoma (PCh) and malignant rhabdoid tumor (MRT) including atypical teratoid/rhabdoid tumor (AT/RT). PCh is characterized by poorly differentiated epithelioid tumor cells, sheet arrangement, and coexpression of nonepithelial and epithelial markers. Rhabdoid cells are sometimes present. Therefore, the differentiation of these tumors is often difficult. Brachyury is a transcription factor within the T-box family typically expressed in notochord tissue and chordomas. Some studies have reported high specificity and sensitivity of brachyury expression in chordomas. In the present study, we analyzed immunohistochemical brachyury expression in SMARCB1-deficient tumors and discuss important clinicopathological and diagnostic points, especially in cases of intracranial SMARCB1-deficient tumors with brachyury expression. Brachyury and cytokeratin immunoexpression status was examined in 42 formalin-fixed paraffin-embedded SMARCB1-deficient tumor specimens (PCh, 6 cases; extra-central nervous system [CNS] MRT, 26 cases; AT/RT, 10 cases) and 25 cases of conventional chordoma (CCh). All cases of PCh and CCh showed diffuse immunopositivities for cytokeratin 8, pan-cytokeratin, and brachyury. Brachyury immunoexpression was present in 2 extra-CNS MRT (8%) and 5 AT/RT (50%) cases, but immunopositivity was focal not diffuse. Indeed, in almost all cases of AT/RT (cytokeratin 8, 7/10 cases; pan-cytokeratin, 7/10 cases) and extra-CNS MRT (cytokeratin 8, 23/26 cases; pan-cytokeratin, 25/26 cases), fewer than 50% of cells showed immunoreactivity. Although the histological and clinical features of PCh resemble those of AT/RT, semiquantitative evaluations of the degree of brachyury and cytokeratin immunoexpressivity may help to distinguish PCh from AT/RT.


Biomarkers, Tumor/metabolism , Brain Neoplasms/diagnosis , Chordoma/diagnosis , Fetal Proteins/biosynthesis , Rhabdoid Tumor/diagnosis , T-Box Domain Proteins/biosynthesis , Teratoma/diagnosis , Adult , Aged , Aged, 80 and over , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Child , Child, Preschool , Chordoma/metabolism , Chordoma/pathology , Diagnosis, Differential , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Rhabdoid Tumor/metabolism , Rhabdoid Tumor/pathology , SMARCB1 Protein/deficiency , Teratoma/metabolism , Teratoma/pathology
8.
J Cutan Pathol ; 48(8): 1051-1060, 2021 Aug.
Article En | MEDLINE | ID: mdl-33625734

BACKGROUND: SMARCB1-deficient malignancies can arise in various sites. We describe a novel primary SMARCB1-deficient carcinoma of skin (SDCS) and characterize SMARCB1 mutations in non-melanoma skin cancers (NMSC). METHODS: Cases underwent immunophenotyping and targeted exome sequencing (MSK-IMPACT) assay interrogating somatic mutations in 468 cancer-related genes. The MSK-IMPACT database from 2014 to 2020 encompassing 55, 000 cases was searched for NMSC with SMARCB1 mutations. RESULTS: SDCS arose on the scalp of an 18-year-old woman showing homozygous SMARCB1 deletion with a LATS2 G963E variant. Another case arose on the temple of a 76-year-old man harboring a SMARCB1 W206* mutation associated with loss of heterozygosity (LOH), 59 concurrent mutations, and a UV mutation signature (UV-MS). Both tumors exhibited INI1 loss, positive CK5/6, p40, p63, and claudin-4 with negative CD34. Of 378 NMSC cases, including 370 carcinomas, 7 SMARCB1-mutated tumors were identified: 3 squamous cell, 3 Merkel cell, and one basal cell carcinoma. Six showed UV-MS. Five INI1-interrogated cases retained protein expression suggesting they were SMARCB1-proficient. CONCLUSIONS: SDCS can be clinically aggressive, harbor SMARCB1 homozygous deletions or truncating SMARCB1 mutations associated with LOH, and can occur with or without UV-MS. Overall, SMARCB1 mutations in NMSC are rare with most being of undetermined significance and associated with retained INI1 and UV-MS.


Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , SMARCB1 Protein/deficiency , Skin Neoplasms/pathology , Adolescent , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/therapy , Fatal Outcome , Female , Homozygote , Humans , Immunohistochemistry/methods , Immunophenotyping/methods , Immunotherapy/methods , Loss of Heterozygosity/genetics , Male , Middle Aged , Mutation/genetics , Protein Serine-Threonine Kinases/genetics , Proton Therapy/methods , Scalp/pathology , Skin Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Exome Sequencing/methods
9.
Am J Clin Pathol ; 156(2): 229-245, 2021 07 06.
Article En | MEDLINE | ID: mdl-33608696

OBJECTIVES: The SWI/SNF complex core subunit SMARCB1 is inactivated in a variety of neoplasms that share characteristic "rhabdoid" cytomorphology. The aim of this study was to evaluate SMARCB1-deficient soft tissue neoplasms on cytology to identify diagnostic clues. METHODS: Eleven SMARCB1-deficient tumors, including six epithelioid sarcomas, three malignant rhabdoid tumors, one epithelioid malignant peripheral nerve sheath tumor (MPNST), and one poorly differentiated chordoma with fine-needle aspiration (FNA), serous effusion, or touch prep (TP) from two institutions, were included. Targeted next-generation sequencing (NGS) was performed in two cases. RESULTS: Evaluation of FNA (n = 4), effusion (n = 4), and TP (n = 3) in nine adult and two pediatric patients demonstrated cellular samples (n = 11), epithelioid cells with rhabdoid morphology (n = 9), eccentrically located nuclei with prominent nucleoli (n = 7), and cytoplasmic bodies (n = 4); two patients were diagnosed on FNA with cell block. Immunohistochemistry (IHC) demonstrated SMARCB1 loss in all cases and keratin and/or EMA expression in all but the epithelioid MPNST; NGS identified SMARCB1 inactivation in both cases. CONCLUSIONS: SMARCB1-deficient soft tissue neoplasms comprise a variety of tumors with epithelioid morphology and frequent expression of keratin and/or EMA. Recognition of characteristic rhabdoid morphology on cytology can prompt IHC and/or NGS testing for SMARCB1 deficiency and help establish the diagnosis.


SMARCB1 Protein/deficiency , Soft Tissue Neoplasms/pathology , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Male , Middle Aged , Molecular Biology , SMARCB1 Protein/genetics , Soft Tissue Neoplasms/genetics , Young Adult
10.
Histopathology ; 79(2): 160-167, 2021 Aug.
Article En | MEDLINE | ID: mdl-33432679

AIMS: Genomic inactivation of ARID1B in ARID1A-inactivated tumour and genomic inactivation of SMARCB1 represent two recurrent mechanisms, core SWItch/sucrose non-fermentable (SWI/SNF) complex inactivation, that are associated with de-differentiation in endometrial carcinoma. Approximately one-third of dedifferentiated/undifferentiated endometrial carcinomas (DDEC/UEC) show loss of ARID1B expression with a minor subset showing loss of SMARCB1 expression, but little is known regarding the specificity of ARID1B or SMARCB1 loss in gynaecological tract tumours in general. The aim of this study was to examine the frequency of ARID1B and SMARCB1 loss by immunohistochemistry in a series of gynaecological tract epithelial/mesenchymal neoplasms. METHODS AND RESULTS: We evaluated 1849 tumours that included 748 endometrial carcinomas, 101 uterine carcinosarcomas/adenosarcomas, 64 uterine sarcomas, 221 cervical carcinomas and 715 ovarian carcinomas/borderline tumours by tissue microarrays (TMA). We observed ARID1B loss in 35 of 86 (41%) and SMARCB1 loss in three of 86 (3%) DDEC/UEC, but not in any other uterine tumour types examined. ARID1B-deficient DDEC/UEC also showed concurrent loss of ARID1A expression. All SMARCB1-deficient tumours showed loss of MLH1 and PMS2, while 29 of 35 ARID1B-deficient tumours showed loss of MLH1 and PMS2 or loss of MSH6. All ovarian carcinomas/borderline tumours and cervical carcinomas showed intact expression of ARID1B and SMARCB1. CONCLUSION: Our findings indicate that the loss of expression of ARID1B or SMARCB1 by immunohistochemistry is highly specific for undifferentiated carcinoma among tumours of the upper gynaecological tract and cervix, and therefore can be used to identify these highly aggressive malignant tumours.


Carcinoma/diagnosis , Cell Dedifferentiation , DNA-Binding Proteins/deficiency , Genital Neoplasms, Female/diagnosis , SMARCB1 Protein/deficiency , Transcription Factors/deficiency , Biomarkers, Tumor/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Cell Dedifferentiation/genetics , Cohort Studies , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/metabolism , Genital Neoplasms, Female/pathology , Humans , Immunohistochemistry , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Sarcoma/diagnosis , Sarcoma/metabolism , Sarcoma/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
11.
Genes Chromosomes Cancer ; 60(3): 190-209, 2021 03.
Article En | MEDLINE | ID: mdl-33252159

Our knowledge regarding the role of genes encoding the chromatin remodeling switch/sucrose non-fermenting (SWI/SNF) complex in the initiation and progression of gynecologic malignancies continues to evolve. This review focuses on gynecologic tumors in which the sole or primary genetic alteration is in SMARCA4 or SMARCB1, two members of the SWI/SNF chromatin remodeling complex. In this review, we present a brief overview of the classical example of such tumors, ovarian small cell carcinoma of hypercalcemic type, and then a detailed review and update of SMARCB1-deficient and SMARCA4-deficient tumors of the uterus and vulva.


Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/deficiency , Nuclear Proteins/deficiency , Ovarian Neoplasms/metabolism , SMARCB1 Protein/deficiency , Transcription Factors/deficiency , Transcription Factors/metabolism , Uterine Neoplasms/metabolism , Vulvar Neoplasms/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , Female , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovarian Neoplasms/genetics , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Sarcoma/genetics , Sarcoma/metabolism , Transcription Factors/genetics , Uterine Neoplasms/genetics , Vulvar Neoplasms/genetics
12.
Head Neck Pathol ; 15(2): 555-565, 2021 Jun.
Article En | MEDLINE | ID: mdl-33141418

SMARCB1/INI1 deficient sinonasal carcinoma is a variant of sinonasal undifferentiated carcinoma (SNUC). There is a paucity of literature describing the histomorphological features of this relatively new entity. Herein we describe the histomorphological features of three such cases and review the literature. We retrospectively reviewed the cases of SNUC diagnosed in our institute in the last 6 years. Immunohistochemistry for INI1, NUT & p16 were performed on these cases. Three cases showed loss of INI1. The histomorphology and clinicopathological features of these cases were studied and compared with non INI1 deficient SNUC. A total of 9 cases of SNUC were identified. Three of these cases showed loss of INI1. These three cases had presented with large sinonasal mass and with intracranial extension. Histopathology of these cases showed a diffuse infiltrative pattern, nest, and islands of predominantly basaloid cells with focal rhabdoid morphology. Additional features like small cell carcinoma like pattern, pseudoalveolar and pseudoglandular patterns, clear vacuoles and pseudopapillary appearance were also noted. We conclude that in presence of a mixed pattern of cells with a predominance of basaloid morphology, the possibility of SMARCB1/INI1 deficient sinonasal carcinoma must be strongly suspected and immunohistochemistry for INI1 must be performed.


Carcinoma/pathology , Maxillary Sinus Neoplasms/pathology , SMARCB1 Protein/deficiency , Adult , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Female , Humans , Male , Middle Aged , Retrospective Studies
14.
Genes Chromosomes Cancer ; 60(1): 43-48, 2021 01.
Article En | MEDLINE | ID: mdl-32920865

Evolution of poorly differentiated chordoma from conventional chordoma has not been previously reported. We encountered a case of a poorly differentiated chordoma with evidence of whole-genome doubling arising from a SMARCB1-deficient conventional chordoma. The tumor presented as a destructive sacral mass in a 43-year-old man and was comprised of a highly cellular poorly differentiated chordoma with small, morphologically distinct nodules of conventional chordoma accounting for <5% of the total tumor volume. Immunohistochemistry (IHC) revealed both components were strongly reactive for brachyury and lacked normal staining for INI1. Single nucleotide polymorphism (SNP) array analysis identified multiple genomic imbalances in the conventional component, including deletions of 1p, 3p, and 22q (involving SMARCB1) and loss of chromosomes 5 and 15, while the poorly differentiated component exhibited the same aberrations at a more profound level with additional loss of chromosome 4, low level focal deletion of 17p (involving TP53), and tetraploidy. Homozygous deletion of SMARCB1 was present in both components. Fluorescence in situ hybridization (FISH) analysis confirmed the relevant deletions in both components as well as genome doubling in the poorly differentiated tumor. This case suggests that SMARCB1 loss is an early event in rare conventional chordomas that could potentially evolve into poorly differentiated chordoma through additional genomic aberrations such as genome doubling. Further studies with additional patients will be needed to determine if genome doubling is a consistent pathway for evolution of poorly differentiated chordoma.


Chordoma/genetics , Chromosome Deletion , SMARCB1 Protein/genetics , Spinal Neoplasms/genetics , Tetraploidy , Adult , Chordoma/pathology , Fetal Proteins/genetics , Fetal Proteins/metabolism , Humans , Male , SMARCB1 Protein/deficiency , Sacrum/pathology , Spinal Neoplasms/pathology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
15.
Am J Surg Pathol ; 44(12): 1712-1724, 2020 12.
Article En | MEDLINE | ID: mdl-32910019

Switch/sucrose nonfermenting complex subunits, such as BRG1, INI1, and ARID1B, are inactivated in a subset of endometrial undifferentiated carcinoma and dedifferentiated carcinoma (DC). Limited information is currently available on their prevalence in other subtypes or the nosological status of endometrial carcinoma with their deficiencies. This study immunohistochemically examined the expression status of BRG1, INI1, and ARID1B using 570 archived cases of endometrial carcinoma and carcinosarcoma resected at a single institution. We identified 1 BRG1-deficient undifferentiated carcinoma, 8 BRG1/INI1/ARID1B-deficient DC, and 3 BRG1-deficient clear-cell carcinomas. None of the cases of endometrioid and serous carcinomas or carcinosarcoma showed deficiencies of these subunits. We then compared 8 BRG1/INI1/ARID1B-deficient DC with 6 BRG1/INI1/ARID1B-intact DC and 28 carcinosarcomas, the latter of which was often confused with DC. Histologically, BRG1/INI1/ARID1B-intact and BRG1/INI1/ARID1B-deficient DC shared a monotonous solid appearance with rhabdoid and epithelioid cells and a myxoid stroma; however, abrupt keratinization and cell spindling was absent in BRG1/INI1/ARID1B-deficient tumors. The median overall survival of patients with BRG1/INI1/ARID1B-deficient DC was 3.8 months, which was worse than those with BRG1/INI1/ARID1B-intact DC (P=0.008) and with carcinosarcoma (P=0.004). BRG1/INI1/ARID1B-deficient DC may be a separate entity with an aggressive behavior to be distinguished from BRG1/INI1/ARID1B-intact DC and carcinosarcoma. Regarding clear-cell carcinoma (n=12), BRG1 deficiency appeared to be mutually exclusive with abnormal ARID1A, BRM, and p53 expression. Further studies are needed to clarify whether BRG1 deficiency plays a role in the pathogenesis of clear-cell carcinoma.


Biomarkers, Tumor/deficiency , Carcinoma/chemistry , Carcinosarcoma/chemistry , DNA Helicases/deficiency , DNA-Binding Proteins/deficiency , Endometrial Neoplasms/chemistry , Immunohistochemistry , Nuclear Proteins/deficiency , SMARCB1 Protein/deficiency , Transcription Factors/deficiency , Adult , Aged , Aged, 80 and over , Carcinoma/pathology , Carcinoma/surgery , Carcinoma, Endometrioid/chemistry , Carcinoma, Endometrioid/pathology , Carcinoma, Endometrioid/surgery , Carcinosarcoma/pathology , Carcinosarcoma/surgery , Cell Dedifferentiation , Endometrial Neoplasms/pathology , Endometrial Neoplasms/surgery , Female , Humans , Middle Aged
16.
Hum Pathol ; 104: 105-116, 2020 10.
Article En | MEDLINE | ID: mdl-32818509

SMARCB1-deficient sinonasal carcinoma (SNC) is an aggressive malignancy characterized by INI1 loss mostly owing to homozygous SMARCB1 deletion. With the exception of a few reported cases, these tumors have not been thoroughly studied by massive parallel sequencing (MPS). A retrospective cohort of 22 SMARCB1-deficient SNCs were studied by light microscopy, immunohistochemistry, fluorescence in situ hybridization (n = 9), targeted exome MPS (n = 12), and Fraction and Allele-Specific Copy Number Estimates from Tumor Sequencing (FACETS) (n = 10), a bioinformatics pipeline for copy number/zygosity assessment. SMARCB1-deficient SNC was found in 13 (59%) men and 9 (41%) women. Most common growth patterns were the basaloid pattern (59%), occurring mostly in men (77%), and plasmacytoid/eosinophilic/rhabdoid pattern (23%), arising mostly in women (80%). The former group was significantly younger (median age = 46 years, range = 24-54, vs 79 years, range = 66-95, p < 0.0001). Clear cell, pseudoglandular, glandular, spindle cell, and sarcomatoid features were variably present. SMARCB1-deficient SNC expressed cytokeratin (100%), p63 (72%), neuroendocrine markers (52%), CDX-2 (44%), S-100 (25%), CEA (4/4 cases), Hepatocyte (2/2 cases), and aberrant nuclear ß-catenin (1/1 case). SMARCB1 showed homozygous deletion (68%), hemizygous deletion (16%), or truncating mutations associated with copy neutral loss of heterozygosity (11%). Coexisting genetic alterations were 22q loss including loss of NF2 and CHEK2 (50%), chromosome 7 gain (25%), and TP53 V157F, CDKN2A W110∗, and CTNNB1 S45F mutations. At 2 years and 5 years, the disease-specific survival and disease-free survival were 70% and 35% and 13% and 0%, respectively. SMARCB1-deficient SNCs are phenotypically and genetically diverse, and these distinctions warrant further investigation for their biological and clinical significance.


Biomarkers, Tumor/genetics , Genetic Heterogeneity , Nose Neoplasms/genetics , Paranasal Sinus Neoplasms/genetics , SMARCB1 Protein/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/deficiency , Disease-Free Survival , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Mutation , Neoplasm Staging , Nose Neoplasms/chemistry , Nose Neoplasms/pathology , Nose Neoplasms/therapy , Paranasal Sinus Neoplasms/chemistry , Paranasal Sinus Neoplasms/pathology , Paranasal Sinus Neoplasms/therapy , Phenotype , Retrospective Studies , SMARCB1 Protein/deficiency , Time Factors , Young Adult
17.
J Am Soc Cytopathol ; 9(6): 494-501, 2020.
Article En | MEDLINE | ID: mdl-32839151

INTRODUCTION: SMARCB1 encodes for a component of the SWI/SNF complex and is widely implicated in carcinogenesis. In the head and neck, SMARCB1-deficient carcinomas typically arise in the sinonasal tract but can be found at other sites. EZH2 inhibitors have emerged as potential targeted therapy against SWI/SNF-deficient tumors. We sought to characterize the cytomorphology of head and neck carcinomas with SMARCB1 deficiencies to identify potential candidates for targeted therapy. MATERIALS AND METHODS: Head and neck carcinomas with SMARCB1 mutations were retrospectively identified and confirmed to be SMARCB1-deficient by both molecular (fluorescent in-situ hybridization or next generation sequencing) and immunohistochemical means. Cases with positive cytology were reviewed and their cytologic features cataloged. RESULTS: A total of 19 specimens from 13 patients were reviewed, including 8 specimens from 7 sinonasal carcinomas, 4 specimens from 3 thyroid carcinomas, 3 specimens from 2 skin carcinomas, and 4 specimens from 1 carcinoma of unknown primary origin. High-grade features were common, including mitoses (11 of 19) necrosis (13 of 19) and multinucleation (16 of 19). Tumors showed either dense cytoplasm with distinct cell borders (10 of 19) or delicate cytoplasm with indistinct cell borders (9 of 19). Most tumors showed no distinct epithelial differentiation (12 of 19), while some (7 of 19) showed glandular or signet ring features. A minor cohort demonstrated rhabdoid cells (4 of 19). CONCLUSIONS: Head and neck carcinomas with SMARCB1 deficiencies have a wide array of morphologies and tend to demonstrate high-grade features. Only a minor cohort demonstrate rhabdoid-type cells. Evaluation of SMARCB1 deficiency for potential targeted therapy should not be limited to tumors with rhabdoid morphology.


Carcinoma/metabolism , Carcinoma/pathology , Paranasal Sinus Neoplasms/metabolism , Paranasal Sinus Neoplasms/pathology , SMARCB1 Protein/deficiency , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Adult , Aged , Biomarkers, Tumor/deficiency , Biomarkers, Tumor/genetics , Carcinoma/genetics , Cell Nucleolus/pathology , Female , Humans , Immunohistochemistry/methods , Male , Middle Aged , Mitosis , Mutation , Necrosis/pathology , Paranasal Sinus Neoplasms/genetics , Retrospective Studies , SMARCB1 Protein/genetics , Skin Neoplasms/genetics , Thyroid Neoplasms/genetics
18.
Clin Cancer Res ; 26(12): 2882-2890, 2020 06 15.
Article En | MEDLINE | ID: mdl-32122923

PURPOSE: Several aggressive pediatric cancers harbor alterations in SMARCB1, including rhabdoid tumors, epithelioid sarcoma, and chordoma. As tumor profiling has become more routine in clinical care, we investigated the relationship between SMARCB1 genetic variants identified by next-generation sequencing (NGS) and INI1 protein expression. Therapeutic approaches for INI1-deficient tumors are limited. Early reports suggest a potential role for immune checkpoint inhibition in these patients. Thus, we also investigated PD-L1 and CD8 expression in INI1-negative pediatric brain and solid tumors. EXPERIMENTAL DESIGN: We performed immunohistochemistry (IHC) for INI1 and immune markers (PD-L1, CD8, and CD163) and NGS on tumor samples from 43 pediatric patients who had tumors with INI1 loss on previous IHC or SMARCB1 genomic alterations on prior somatic sequencing. RESULTS: SMARCB1 two-copy deletions and inactivating mutations on NGS were associated with loss of INI1 protein expression. Single-copy deletion of SMARCB1 was not predictive of INI1 loss in tumor histologies not known to be INI1-deficient. In the 27 cases with INI1 loss and successful tumor sequencing, 24 (89%) had a SMARCB1 alteration detected. In addition, 47% (14/30) of the patients with INI1-negative tumors had a tumor specimen that was PD-L1 positive and 60% (18/30) had positive or rare CD8 staining. We report on 3 patients with INI1-negative tumors with evidence of disease control on immune checkpoint inhibitors. CONCLUSIONS: A significant proportion of the INI1-negative tumors express PD-L1, and PD-L1 positivity was associated with extracranial tumor site. These results suggest that clinical trials of immune checkpoint inhibitors are warranted in INI1-negative pediatric cancers.


B7-H1 Antigen/metabolism , Biomarkers, Tumor/genetics , Immune Checkpoint Inhibitors/therapeutic use , Mutation , Neoplasms/pathology , SMARCB1 Protein/deficiency , Adolescent , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , Child , Child, Preschool , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Prognosis , SMARCB1 Protein/genetics
19.
World Neurosurg ; 136: 305-310, 2020 Apr.
Article En | MEDLINE | ID: mdl-32001406

INTRODUCTION: To describe the current state of literature involving SMARCB1/INI-1 deficient sinonasal carcinoma (SDSC) and examine a case at our institution. METHODS: A systematic search was performed using the Population, Intervention, Comparator, Outcome, and Study Design approach. Search criteria included all occurrences in the title or abstract of the terms: "integrase interactor 1 deficient," "INI1 deficient," or "SMARCB1 deficient" and "sinonasal carcinoma." The main outcomes were disease-free survival, all-cause mortality, rates of recurrence, or metastases. RESULTS: Systematic search yielded 13 studies for final review. All studies were either case series or case reports with 82 cases of SDSC published since 2014. Age on presentation ranged from 19 to 75 years, with the majority of patients being male. Surgical resection was the primary modality of treatment with adjuvant radiation or chemoradiation therapy. Overall, the prognosis was poor, with most tumors presenting at advanced stages with an overall median (range) survival of 22 (12-44) months with an average (standard deviation) of 45.3% (33.1%) of patients dying of the disease. An average (standard deviation) of 38.2% (34.0%) of patients had no evidence of disease at follow-up. Studies comparing sinonasal undifferentiated carcinoma to SDSC reported worse prognosis for SDSC and increased risk for locoregional recurrence in the latter cohort. CONCLUSIONS: SDSC represents a highly aggressive tumor presenting at advanced stage with propensity of metastasis. More research is necessary to determine the optimal treatment modality and management.


Carcinoma/genetics , Maxillary Sinus Neoplasms/genetics , Nose Neoplasms/genetics , SMARCB1 Protein/genetics , Adult , Aged , Carcinoma/surgery , Female , Headache/etiology , Humans , Male , Maxillary Sinus Neoplasms/surgery , Middle Aged , Neurosurgical Procedures , Nose Neoplasms/surgery , SMARCB1 Protein/deficiency , Young Adult
20.
Cancer Biomark ; 27(3): 399-406, 2020.
Article En | MEDLINE | ID: mdl-32083567

SMARCB1 is a tumor suppressor gene, which is part of SWI/SNF complex involved in transcriptional regulation. Recently, loss of SMARCB1 expression has been reported in gastrointestinal carcinomas. Our purpose was to evaluate the incidence and prognostic value of SMARCB1 loss in colon carcinoma (CC). Patients with stage III CC (n= 1695), and a second cohort of 23 patients with poorly differentiated CC were analyzed. Immunohistochemistry for SMARCB1 was performed on tissue microarrays, and cases with loss of expression were controlled on whole sections. Loss of SMARCB1 was compared with the clinico-pathological and molecular characteristics, and the prognostic value was evaluated. Loss of SMARCB1 was identified in 12 of 1695 (0.7%) patients with stage III CC. Whole section controls showed a complete loss in only one of these cases, corresponding to a medullary carcinoma. SMARCB1 loss was not associated with histological grade, tumor size nor survival. In the cohort of poorly differentiated CC, we detected 2/23 (8.7%) cases with loss of SMARCB1; one was rhabdoid while the other had medullary and mucinous histology. These 2 cases were deficient for MisMatched Repair (dMMR) and mutated for BRAF. SMARCB1 loss is rare in stage III CC, but appears more frequent in poorly differentiated CC.


Colonic Neoplasms/metabolism , SMARCB1 Protein/deficiency , Adult , Aged , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , SMARCB1 Protein/biosynthesis , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Young Adult
...