17 results on '"SMARCB1 Protein deficiency"'
Search Results
2. Molecular and Treatment Characteristics of SMARCB1 or SMARCA4-Deficient Undifferentiated Tumor: Retrospective Case Series.
- Author
-
Kang HG, Koh J, Kim TM, Han DH, Won TB, Kim DW, Kim DY, and Keam B
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Biomarkers, Tumor genetics, Nuclear Proteins genetics, Nuclear Proteins deficiency, Prognosis, Retrospective Studies, Thoracic Neoplasms genetics, Thoracic Neoplasms therapy, Thoracic Neoplasms pathology, DNA Helicases genetics, SMARCB1 Protein genetics, SMARCB1 Protein deficiency, Transcription Factors genetics
- Abstract
SMARCB1 or SMARCA4-deficient sinonasal carcinoma or thoracic undifferentiated tumor has aggressive nature with a poor prognosis. Patients with this disease were diagnosed by immunohistochemistry or next-generation sequencing. Those who were able to receive a surgery tended to be cured, while the others treated with chemotherapy, radiation therapy, or immune checkpoint inhibitor were often insensitive to these therapies. However, one having CD274 (PD-L1) amplification showed the response to immune checkpoint inhibitor and a good prognosis. We believed that this report could provide promising information for determining the optimal treatment option.
- Published
- 2024
- Full Text
- View/download PDF
3. SMARCB1/INI1-deficient undifferentiated tumour of the thorax: a case report and review of the literature.
- Author
-
Zagni M, Marando A, Negrelli M, Lauricella C, Motta V, Paglino G, Veronese S, Valtorta E, Bonoldi E, and Pelosi G
- Subjects
- Humans, Nuclear Proteins genetics, Nuclear Proteins deficiency, Male, Female, Biomarkers, Tumor analysis, Biomarkers, Tumor genetics, Middle Aged, Lung Neoplasms pathology, Lung Neoplasms genetics, Lung Neoplasms diagnosis, SMARCB1 Protein deficiency, SMARCB1 Protein genetics, Transcription Factors genetics, Transcription Factors deficiency, Thoracic Neoplasms pathology, Thoracic Neoplasms genetics, DNA Helicases deficiency, DNA Helicases genetics
- Abstract
The 5th WHO classification of thoracic tumours includes thoracic SMARCA4-deficient undifferentiated tumour (SMARCA4-UT) among the "other epithelial tumours of the lung" chapter. Herein, we present a case of undifferentiated thoracic neoplasm with retention of SMARCA4 expression, lack of NUT fusion protein and loss of SMARCB1/INI1 expression. After presenting the clinical and pathological features of the tumour, we carried out a review of the literature on the same topic. Albeit very rare, we believe this entity should be included in the heterogeneous group of undifferentiated neoplasms of the thorax., (Copyright © 2024 Società Italiana di Anatomia Patologica e Citopatologia Diagnostica, Divisione Italiana della International Academy of Pathology.)
- Published
- 2024
- Full Text
- View/download PDF
4. Targeting DCAF5 suppresses SMARCB1-mutant cancer by stabilizing SWI/SNF.
- Author
-
Radko-Juettner S, Yue H, Myers JA, Carter RD, Robertson AN, Mittal P, Zhu Z, Hansen BS, Donovan KA, Hunkeler M, Rosikiewicz W, Wu Z, McReynolds MG, Roy Burman SS, Schmoker AM, Mageed N, Brown SA, Mobley RJ, Partridge JF, Stewart EA, Pruett-Miller SM, Nabet B, Peng J, Gray NS, Fischer ES, and Roberts CWM
- Subjects
- Animals, Female, Humans, Male, Mice, Cell Line, Tumor, CRISPR-Cas Systems, Gene Editing, Tumor Suppressor Proteins deficiency, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Proteolysis, Ubiquitin metabolism, Mutation, Neoplasms genetics, Neoplasms metabolism, SMARCB1 Protein deficiency, SMARCB1 Protein genetics, SMARCB1 Protein metabolism, Multiprotein Complexes chemistry, Multiprotein Complexes metabolism
- Abstract
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 Project
1-3 . 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., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
5. SMARCB1 deletion in atypical teratoid rhabdoid tumors results in human endogenous retrovirus K (HML-2) expression.
- Author
-
Doucet-O'Hare TT, DiSanza BL, DeMarino C, Atkinson AL, Rosenblum JS, Henderson LJ, Johnson KR, Kowalak J, Garcia-Montojo M, Allen SJ, Orr BA, Santi M, Wang T, Fathi S, Lee MH, Sampson K, Li W, Zhuang Z, and Nath A
- Subjects
- 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, Cell Transformation, Neoplastic genetics, Endogenous Retroviruses genetics, Rhabdoid Tumor etiology, Rhabdoid Tumor pathology, SMARCB1 Protein deficiency, Sequence Deletion, Virus Activation genetics
- Abstract
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.
- Published
- 2021
- Full Text
- View/download PDF
6. SMARCB1/INI1 Deficient Sino-Nasal Carcinoma: Extending the Histomorphological Features.
- Author
-
Ayyanar P, Mishra P, Preetam C, and Adhya AK
- Subjects
- Adult, Biomarkers, Tumor analysis, Biomarkers, Tumor metabolism, Female, Humans, Male, Middle Aged, Retrospective Studies, Carcinoma pathology, Maxillary Sinus Neoplasms pathology, SMARCB1 Protein deficiency
- Abstract
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.
- Published
- 2021
- Full Text
- View/download PDF
7. Genomic and Immunologic Characterization of INI1-Deficient Pediatric Cancers.
- Author
-
Forrest SJ, Al-Ibraheemi A, Doan D, Ward A, Clinton CM, Putra J, Pinches RS, Kadoch C, Chi SN, DuBois SG, Leavey PJ, LeBoeuf NR, Mullen E, Collins N, Church AJ, and Janeway KA
- Subjects
- 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, B7-H1 Antigen metabolism, Biomarkers, Tumor genetics, Immune Checkpoint Inhibitors therapeutic use, Mutation, Neoplasms pathology, SMARCB1 Protein deficiency
- Abstract
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., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
8. SMARCB1 (INI-1)-Deficient Adenocarcinoma of the Sinonasal Tract: A Potentially Under-Recognized form of Sinonasal Adenocarcinoma with Occasional Yolk Sac Tumor-Like Features.
- Author
-
Shah AA, Jain D, Ababneh E, Agaimy A, Hoschar AP, Griffith CC, Magliocca KR, Wenig BM, Rooper LM, and Bishop JA
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Tumor analysis, Endodermal Sinus Tumor pathology, Female, Humans, Male, Middle Aged, Paranasal Sinuses pathology, Young Adult, Adenocarcinoma pathology, Paranasal Sinus Neoplasms pathology, SMARCB1 Protein deficiency
- Abstract
The classification of sinonasal adenocarcinoma (SNAC) is complex. The high-grade, non-intestinal SNAC group is particularly heterogeneous, with tumors showing widely variable morphology. SMARCB1 (INI-1)-deficient sinonasal carcinoma is a newly described, aggressive tumor that usually resembles sinonasal undifferentiated carcinoma (SNUC) or non-keratinizing squamous cell carcinoma; however, glandular differentiation has been rarely reported and this feature may be under-recognized. We present a dedicated series of 12 SMARCB1-deficient SNACs. All tumors had an oncocytoid/plasmacytoid cytomorphology with variable degrees of glandular differentiation consisting of tubules and cribriform structures with foci of intracellular or intraluminal mucin. Three of 12 tumors exhibited foci of yolk sac tumor-like histologic features. The tumors were uniformly high-grade, with nuclear pleomorphism, elevated mitotic rates and frequent necrosis. By immunohistochemistry, all tumors were entirely SMARCB1-deficient, and 10 of 12 were CK7-positive. Occasional expression of CDX2 (4 of 12), CK20 (3 of 12), and p40 (3 of 10) was seen. Expression of yolk sac markers was variably present in tumors that harbored yolk sac-like areas but also tumors that did not: glypican-3 (10 of 11), SALL4 (6 of 11), HepPar-1 (4 of 11), PLAP (1 of 10), and AFP (1 of 11). SMARCB1-deficient sinonasal carcinoma, particularly the oncocytoid/plasmacytoid form, can demonstrate variable degrees of glandular differentiation. This unexpected morphology combined with variable immunohistochemical results may lead to misdiagnoses of high-grade intestinal or non-intestinal SNAC, myoepithelial carcinoma, or even yolk sac tumor or metastatic hepatocellular carcinoma.
- Published
- 2020
- Full Text
- View/download PDF
9. SMARC-B1 deficient sinonasal carcinoma metastasis to the brain with next generation sequencing data: a case report of perineural invasion progressing to leptomeningeal invasion.
- Author
-
Gomez-Acevedo H, Patterson JD, Sardar S, Gokden M, Das BC, Ussery DW, and Rodriguez A
- Subjects
- Adult, Biomarkers, Tumor, Carcinoma diagnostic imaging, Disease Progression, Female, High-Throughput Nucleotide Sequencing, Humans, Immunohistochemistry, Meningeal Carcinomatosis diagnosis, Meningeal Carcinomatosis secondary, Neoplasm Metastasis, Neoplasm Staging, Paranasal Sinus Neoplasms diagnostic imaging, Polymorphism, Single Nucleotide, Tomography, X-Ray Computed, Carcinoma etiology, Carcinoma pathology, Paranasal Sinus Neoplasms etiology, Paranasal Sinus Neoplasms pathology, SMARCB1 Protein deficiency
- Abstract
Background: SMARCB1-deficient sinonasal carcinoma (SDSC) is an aggressive subtype of head and neck cancers that has a poor prognosis despite multimodal therapy. We present a unique case with next generation sequencing data of a patient who had SDSC with perineural invasion to the trigeminal nerve that progressed to a brain metastasis and eventually leptomeningeal spread., Case Presentation: A 42 year old female presented with facial pain and had resection of a tumor along the V2 division of the trigeminal nerve on the right. She underwent adjuvant stereotactic radiation. She developed further neurological symptoms and imaging demonstrated the tumor had infiltrated into the cavernous sinus as well as intradurally. She had surgical resection for removal of her brain metastasis and decompression of the cavernous sinus. Following her second surgery, she had adjuvant radiation and chemotherapy. Several months later she had quadriparesis and imaging was consistent with leptomeningeal spread. She underwent palliative radiation and ultimately transitioned quickly to comfort care and expired. Overall survival from time of diagnosis was 13 months. Next generation sequencing was carried out on her primary tumor and brain metastasis. The brain metastatic tissue had an increased tumor mutational burden in comparison to the primary., Conclusions: This is the first report of SDSC with perineural invasion progressing to leptomeningeal carcinomatosis. Continued next generation sequencing of the primary and metastatic tissue by clinicians is encouraged toprovide further insights into metastatic progression of rare solid tumors.
- Published
- 2019
- Full Text
- View/download PDF
10. Immunologic Correlates of the Abscopal Effect in a SMARCB1/INI1-negative Poorly Differentiated Chordoma after EZH2 Inhibition and Radiotherapy.
- Author
-
Gounder MM, Zhu G, Roshal L, Lis E, Daigle SR, Blakemore SJ, Michaud NR, Hameed M, and Hollmann TJ
- Subjects
- Adult, Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Biopsy, Cell Line, Tumor, Chordoma metabolism, Chordoma pathology, Chordoma therapy, Combined Modality Therapy, Disease Models, Animal, Exons, Female, Humans, Immunohistochemistry, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Lymphocytes, Tumor-Infiltrating pathology, Mice, Mutation, Neoplasm Grading, Neoplasm Staging, Radiotherapy, SMARCB1 Protein metabolism, Tomography, X-Ray Computed, Treatment Outcome, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Xenograft Model Antitumor Assays, Chordoma etiology, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Immunomodulation, SMARCB1 Protein deficiency
- Abstract
Purpose: We sought to determine the mechanism of an exceptional response in a patient diagnosed with a SMARCB1/INI1-negative chordoma treated with tazemetostat, an EZH2 inhibitor, and followed by radiotherapy. Patient and Methods: In an attempt to investigate the mechanism behind this apparent abscopal effect, we interrogated tumor tissues obtained over the clinical course. We utilized next-generation sequencing, standard IHC, and employed a novel methodology of multiplex immunofluorescence analysis., Results: We report an exceptional and durable response (2+ years) in a patient with SMARCB1-deleted, metastatic, poorly differentiated chordoma, a lethal disease with an overall survival of 6 months. The patient was treated for 4 weeks with tazemetostat, an EZH2 inhibitor, in a phase II clinical trial. At the time of progression she underwent radiation to the primary site and unexpectedly had a complete response at distant metastatic sites. We evaluated baseline and on-treatment tumor biopsies and demonstrate that tazemetostat resulted in pharmacodynamic inhibition of EZH2 as seen by decrease in histone trimethylation at H3K27. Tazemetostat resulted in a significant increase in intratumoral and stromal infiltration by proliferative (high Ki-67), CD8
+ T cells, FoxP3+ regulatory T cells, and immune cells expressing checkpoint regulators PD-1 and LAG-3. These changes were pronounced in the stroma., Conclusions: These observations are the first demonstration in patient samples confirming that EZH2 inhibition can promote a sustained antitumor response that ultimately leads to T-cell exhaustion and checkpoint activation. This suggests that targeted alteration of the epigenetic landscape may sensitize some tumors to checkpoint inhibitors., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
11. p53 Is a Master Regulator of Proteostasis in SMARCB1-Deficient Malignant Rhabdoid Tumors.
- Author
-
Carugo A, Minelli R, Sapio L, Soeung M, Carbone F, Robinson FS, Tepper J, Chen Z, Lovisa S, Svelto M, Amin S, Srinivasan S, Del Poggetto E, Loponte S, Puca F, Dey P, Malouf GG, Su X, Li L, Lopez-Terrada D, Rakheja D, Lazar AJ, Netto GJ, Rao P, Sgambato A, Maitra A, Tripathi DN, Walker CL, Karam JA, Heffernan TP, Viale A, Roberts CWM, Msaouel P, Tannir NM, Draetta GF, and Genovese G
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p16 genetics, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Female, Gene Expression Regulation, Neoplastic, Humans, Male, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Knockout, Proteasome Inhibitors pharmacology, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, Rhabdoid Tumor drug therapy, Rhabdoid Tumor genetics, Rhabdoid Tumor pathology, SMARCB1 Protein genetics, Signal Transduction, Tumor Cells, Cultured, Tumor Suppressor Protein p53 deficiency, Tumor Suppressor Protein p53 genetics, Unfolded Protein Response, Autophagy drug effects, Endoplasmic Reticulum Stress drug effects, Proteostasis drug effects, Rhabdoid Tumor metabolism, SMARCB1 Protein deficiency, Tumor Suppressor Protein p53 metabolism
- Abstract
Alterations in chromatin remodeling genes have been increasingly implicated in human oncogenesis. Specifically, the biallelic inactivation of the SWI/SNF subunit SMARCB1 results in the emergence of extremely aggressive pediatric malignancies. Here, we developed embryonic mosaic mouse models of malignant rhabdoid tumors (MRTs) that faithfully recapitulate the clinical-pathological features of the human disease. We demonstrated that SMARCB1-deficient malignancies exhibit dramatic activation of the unfolded protein response (UPR) and ER stress response via a genetically intact MYC-p19
ARF -p53 axis. As a consequence, these tumors display an exquisite sensitivity to agents inducing proteotoxic stress and inhibition of the autophagic machinery. In conclusion, our findings provide a rationale for drug repositioning trials investigating combinations of agents targeting the UPR and autophagy in SMARCB1-deficient MRTs., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
12. Clinical Benefit to an Aurora A Kinase Inhibitor in a Patient with Metastatic Integrase Interactor 1-Deficient Carcinoma.
- Author
-
Karantanos T, Rooper L, Kang Y, Lin CT, Wenga P, Sagorsky S, Lauring J, and Kang H
- Subjects
- Adult, Carcinoma pathology, Female, Humans, Neoplasm Metastasis, Aurora Kinase A antagonists & inhibitors, Carcinoma drug therapy, SMARCB1 Protein deficiency
- Abstract
Integrase interactor 1 (INI-1)-deficient carcinoma is a rare cancer characterized by the loss of the SWItch/Sucrose Non-Fermentable-related matrix-associated actin-dependent regulator of chromatin subfamily B member 1 gene ( SMARCB1 ) and tends to follow an aggressive clinical course. There is no currently available standard therapy option, although a few promising treatment strategies, including enhancer of zeste homolog 2 (EZH2) inhibition, are under active investigation. This report describes a 30-year-old woman with INI-1-deficient carcinoma who progressed on combination chemotherapy and an EZH2 inhibitor. Next-generation-sequencing-based targeted cancer-related gene assay confirmed SMARCB1 loss and revealed other mutations in breast cancer 1 gene and checkpoint kinase 2 gene, which may have impacted her clinical course. After discussion at the molecular tumor board, she was offered alisertib, an aurora A kinase inhibitor, on a single-patient expanded-use program and achieved prolonged disease stabilization. Aurora A kinase inhibition may have an important role in the management of patients with INI-1-deficient tumors, warranting further evaluation in clinical studies. KEY POINTS: Loss of the SWItch/Sucrose Non-Fermentable-related matrix-associated actin-dependent regulator of chromatin subfamily B member 1 gene ( SMARCB1 ), which encodes integrase interactor 1 (INI-1), is associated with various mesenchymal malignancies, but a few carcinomas with rhabdoid features have been recently described as a distinct entity.INI-1-deficient carcinoma can be very aggressive, and there is no known treatment option available.There are encouraging preliminary data with an enhancer of zeste homolog 2 inhibitor, tazematostat, in INI-1-deficient malignancies, including INI-1-deficient carcinomas.Loss of INI-1 can activate aurora A kinase (AurkA), and inhibition of AurkA by alisertib could be a viable option and warrants further investigation in this cancer.Clinical genomic profiling can confirm diagnosis of molecularly defined malignancy and provide insights on therapeutic options., Competing Interests: Disclosures of potential conflicts of interest may be found at the end of this article., (© AlphaMed Press 2018.)
- Published
- 2019
- Full Text
- View/download PDF
13. SMARCB1 Deficiency Integrates Epigenetic Signals to Oncogenic Gene Expression Program Maintenance in Human Acute Myeloid Leukemia.
- Author
-
Chatterjee SS, Biswas M, Boila LD, Banerjee D, and Sengupta A
- Subjects
- Epigenesis, Genetic, Gene Expression Regulation, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, SMARCB1 Protein genetics, SMARCB1 Protein metabolism, Signal Transduction, Transfection, Leukemia, Myeloid, Acute metabolism, SMARCB1 Protein deficiency
- Abstract
SWI/SNF is an evolutionarily conserved multi-subunit chromatin remodeling complex that regulates epigenetic architecture and cellular identity. Although SWI/SNF genes are altered in approximately 25% of human malignancies, evidences showing their involvement in tumor cell-autonomous chromatin regulation and transcriptional plasticity are limiting. This study demonstrates that human primary acute myeloid leukemia (AML) cells exhibit near complete loss of SMARCB1 (BAF47 or SNF5/INI1) and SMARCD2 (BAF60B) associated with nucleation of SWI/SNF
Δ SMARCC1 (BAF155), an intact core component of SWI/SNFΔ , colocalized with H3K27Ac to target oncogenic loci in primary AML cells. Interestingly, gene ontology (GO) term and pathway analysis suggested that SMARCC1 occupancy was enriched on genes regulating Rac GTPase activation, cell trafficking, and AML-associated transcriptional dysregulation. Transcriptome profiling revealed that expression of these genes is upregulated in primary AML blasts, and loss-of-function studies confirmed transcriptional regulation of Rac GTPase guanine nucleotide exchange factors ( GEF ) by SMARCB1. Mechanistically, loss of SMARCB1 increased recruitment of SWI/SNFΔ and associated histone acetyltransferases (HAT) to target loci, thereby promoting H3K27Ac and gene expression. Together, SMARCB1 deficiency induced GEFs for Rac GTPase activation and augmented AML cell migration and survival. Collectively, these findings highlight tumor suppressor role of SMARCB1 and illustrate SWI/SNFΔ function in maintaining an oncogenic gene expression program in AML. Implications: Loss of SMARCB1 in AML associates with SWI/SNFΔ nucleation, which in turn promotes Rac GTPase GEF expression, Rac activation, migration, and survival of AML cells, highlighting SWI/SNFΔ downstream signaling as important molecular regulator in AML. Mol Cancer Res; 16(5); 791-804. ©2018 AACR ., (©2018 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
14. Cribriform neuroepithelial tumor: molecular characterization of a SMARCB1-deficient non-rhabdoid tumor with favorable long-term outcome.
- Author
-
Johann PD, Hovestadt V, Thomas C, Jeibmann A, Heß K, Bens S, Oyen F, Hawkins C, Pierson CR, Aldape K, Kim SP, Widing E, Sumerauer D, Hauser P, van Landeghem F, Ryzhova M, Korshunov A, Capper D, Jones DTW, Pfister SM, Schneppenheim R, Siebert R, Paulus W, Frühwald MC, Kool M, and Hasselblatt M
- Subjects
- Child, Child, Preschool, DNA Methylation genetics, Female, Humans, Infant, Kaplan-Meier Estimate, Male, Neoplasms, Neuroepithelial pathology, Rhabdoid Tumor pathology, Statistics, Nonparametric, Brain Neoplasms genetics, Mutation genetics, Neoplasms, Neuroepithelial genetics, Rhabdoid Tumor genetics, SMARCB1 Protein deficiency, SMARCB1 Protein genetics
- Abstract
Rhabdoid phenotype and loss of SMARCB1 expression in a brain tumor are characteristic features of atypical teratoid/rhabdoid tumors (ATRT). Rare non-rhabdoid brain tumors showing cribriform growth pattern and SMARCB1 loss have been designated cribriform neuroepithelial tumor (CRINET). Small case series suggest that CRINETs may have a relatively favorable prognosis. However, the long-term outcome is unclear and it remains uncertain whether CRINET represents a distinct entity or a variant of ATRT. Therefore, 10 CRINETs were clinically and molecularly characterized and compared with 10 ATRTs of each of three recently described molecular subgroups (i.e. ATRT-TYR, ATRT-SHH and ATRT-MYC) using Illumina Infinium HumanMethylation450 arrays, FISH, MLPA, and sequencing. Furthermore, outcome was compared to a larger cohort of 27 children with ATRT-TYR. Median age of the 6 boys and 4 girls harboring a CRINET was 20 months. On histopathological examination, all CRINETs demonstrated a cribriform growth pattern and distinct tyrosinase staining. On unsupervised cluster analysis of methylation data, all CRINETs examined exclusively clustered within the ATRT-TYR molecular subgroup. As ATRT-TYR, CRINETs mainly showed large heterozygous 22q deletions (9/10) and SMARCB1 mutations of the other allele. In two patients, SMARCB1 mutations were also present in the germline. Estimated mean overall survival in patients with CRINETs was 125 months (95% confidence interval 100-151 months) as compared to only 53 (33-74) months in patients with ATRTs of the ATRT-TYR subgroup (Log-Rank P < 0.05). In conclusion, CRINET represents a SMARCB1-deficient non-rhabdoid tumor, which shares molecular similarities with the ATRT-TYR subgroup but has distinct histopathological features and favorable long-term outcome., (© 2016 International Society of Neuropathology.)
- Published
- 2017
- Full Text
- View/download PDF
15. Embryonic signature distinguishes pediatric and adult rhabdoid tumors from other SMARCB1-deficient cancers.
- Author
-
Richer W, Masliah-Planchon J, Clement N, Jimenez I, Maillot L, Gentien D, Albaud B, Chemlali W, Galant C, Larousserie F, Boudou-Rouquette P, Leruste A, Chauvin C, Han ZY, Coindre JM, Varlet P, Freneaux P, Ranchère-Vince D, Delattre O, and Bourdeaut F
- Subjects
- Adolescent, Adult, Age of Onset, Child, Child, Preschool, DNA Methylation, Diagnosis, Differential, Gene Regulatory Networks, Humans, Infant, Male, Young Adult, Gene Expression Profiling methods, Rhabdoid Tumor diagnosis, Rhabdoid Tumor genetics, SMARCB1 Protein deficiency, Exome Sequencing methods
- Abstract
Extra-cranial rhabdoid tumors (RT) are highly aggressive malignancies of infancy, characterized by undifferentiated histological features and loss of SMARCB1 expression. The diagnosis is all the more challenging that other poorly differentiated cancers lose SMARCB1 expression, such as epithelioid sarcomas (ES), renal medullary carcinomas (RMC) or undifferentiated chordomas (UC). Moreover, late cases occurring in adults are now increasingly reported, raising the question of differential diagnoses and emphasizing nosological issues. To address this issue, we have analyzed the expression profiles of a training set of 32 SMARCB1-deficient tumors (SDT), with ascertained diagnosis of RT (n = 16, all < 5 years of age), ES (n = 8, all > 10 years of age), UC (n = 3) and RMC (n = 5). As compared with other SDT, RT are characterized by an embryonic signature, and up-regulation of key-actors of de novo DNA methylation processes. Using this signature, we then analysed the expression profiling of 37 SDT to infer the appropriate diagnosis. Thirteen adult onset tumors showed strong similarity with pediatric RT, in spite of older age; by exome sequencing, these tumors also showed genomic features indistinguishable from pediatric RT. In contrary, 8 tumors were reclassified within carcinoma, ES or UC categories, while the remaining could not be related to any of those entities. Our results demonstrate that embryonic signature is shared by all RT, whatever the age at diagnosis; they also illustrate that many adult-onset SDT of ambiguous histological diagnosis are clearly different from RT. Finally, our study paves the way for the routine use of expression-based signatures to give accurate diagnosis of SDT.
- Published
- 2017
- Full Text
- View/download PDF
16. Oncogenic roles of SMARCB1/INI1 and its deficient tumors.
- Author
-
Kohashi K and Oda Y
- Subjects
- Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Carcinogenesis genetics, Cell Proliferation genetics, Disease Progression, Humans, Oncogenes genetics, Rhabdoid Tumor diagnosis, Rhabdoid Tumor metabolism, SMARCB1 Protein deficiency, SMARCB1 Protein metabolism, Gene Expression Regulation, Neoplastic, Rhabdoid Tumor genetics, SMARCB1 Protein genetics, Signal Transduction genetics
- Abstract
SMARCB1/INI1 is one of the core subunit proteins of the ATP-dependent SWI/SNF chromatin remodeling complex, and is identified as a potent and bona fide tumor suppressor. Interactions have been demonstrated between SMARCB1/INI1 and key proteins in various pathways related to tumor proliferation and progression: the p16-RB pathway, WNT signaling pathway, sonic hedgehog signaling pathway and Polycomb pathway. Initially, no detectable SMARCB1/INI1 protein expression was found in malignant rhabdoid tumor cells, whereas all other kinds of tumor cells and non-tumorous tissue showed SMARCB1/INI1 protein expression. Therefore, immunohistochemical testing for the SMARCB1/INI1 antibody has been considered useful in confirming the histologic diagnosis of malignant rhabdoid tumors. However, recently, aberrant expression of SMARCB1/INI1 has been found in various tumors such as epithelioid sarcomas, schwannomatosis, synovial sarcomas, and so on. In addition, it has been reported that aberrant expression can be classified into three patterns: complete loss, mosaic expression and reduced expression. Although the various pathways related to mechanisms of tumorigenesis and tumor proliferation are complexly intertwined, the clarification of these mechanisms may contribute to therapeutic strategies in SMARCB1/INI1-deficient tumors. In terms of pathological classifications, SMARCB1/INI1-deficient tumors may be re-classified by genetic backgrounds., (© 2017 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.)
- Published
- 2017
- Full Text
- View/download PDF
17. Synthetic vulnerabilities of mesenchymal subpopulations in pancreatic cancer.
- Author
-
Genovese G, Carugo A, Tepper J, Robinson FS, Li L, Svelto M, Nezi L, Corti D, Minelli R, Pettazzoni P, Gutschner T, Wu CC, Seth S, Akdemir KC, Leo E, Amin S, Molin MD, Ying H, Kwong LN, Colla S, Takahashi K, Ghosh P, Giuliani V, Muller F, Dey P, Jiang S, Garvey J, Liu CG, Zhang J, Heffernan TP, Toniatti C, Fleming JB, Goggins MG, Wood LD, Sgambato A, Agaimy A, Maitra A, Roberts CW, Wang H, Viale A, DePinho RA, Draetta GF, and Chin L
- Subjects
- Animals, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Deoxycytidine analogs & derivatives, Deoxycytidine pharmacology, Deoxycytidine therapeutic use, Endoplasmic Reticulum Stress genetics, Female, Genes, myc, Genes, ras, Humans, MAP Kinase Kinase 4 metabolism, MAP Kinase Signaling System, Male, Mesoderm metabolism, Mice, Mosaicism, Oncogene Protein p55(v-myc) metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms metabolism, Proteolysis, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, SMARCB1 Protein deficiency, SMARCB1 Protein metabolism, Transcriptome genetics, Gemcitabine, Mesoderm pathology, Pancreatic Neoplasms pathology
- Abstract
Malignant neoplasms evolve in response to changes in oncogenic signalling. Cancer cell plasticity in response to evolutionary pressures is fundamental to tumour progression and the development of therapeutic resistance. Here we determine the molecular and cellular mechanisms of cancer cell plasticity in a conditional oncogenic Kras mouse model of pancreatic ductal adenocarcinoma (PDAC), a malignancy that displays considerable phenotypic diversity and morphological heterogeneity. In this model, stochastic extinction of oncogenic Kras signalling and emergence of Kras-independent escaper populations (cells that acquire oncogenic properties) are associated with de-differentiation and aggressive biological behaviour. Transcriptomic and functional analyses of Kras-independent escapers reveal the presence of Smarcb1-Myc-network-driven mesenchymal reprogramming and independence from MAPK signalling. A somatic mosaic model of PDAC, which allows time-restricted perturbation of cell fate, shows that depletion of Smarcb1 activates the Myc network, driving an anabolic switch that increases protein metabolism and adaptive activation of endoplasmic-reticulum-stress-induced survival pathways. Increased protein turnover renders mesenchymal sub-populations highly susceptible to pharmacological and genetic perturbation of the cellular proteostatic machinery and the IRE1-α-MKK4 arm of the endoplasmic-reticulum-stress-response pathway. Specifically, combination regimens that impair the unfolded protein responses block the emergence of aggressive mesenchymal subpopulations in mouse and patient-derived PDAC models. These molecular and biological insights inform a potential therapeutic strategy for targeting aggressive mesenchymal features of PDAC.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.