Abstract
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 2014–2020 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.
Keywords: SMARCB1, INI1, BAF47, Squamous cell carcinoma, Epithelioid sarcoma
Introduction
The BAF complex represents the mammalian analogue to the yeast SWI/SNF chromatin remodeling complex. Dysregulation of components in this complex may be found in up to 20% of human cancers in the form of both driver and passenger mutations1. SMARCB1 (BAF47/INI1) is a subunit of the BAF complex and plays a role in tumor suppression via the p16-Rb, Wnt, and sonic hedgehog pathways among others. SMARCB1 loss and deficiency in its activity has been implicated in a growing number of malignancies including epithelioid sarcoma (ES), malignant rhabdoid tumors of the kidney, medullary renal carcinoma, and SMARCB1-deficient sinonasal carcinoma among others2, 3, 4, 5.
SMARCB1 loss is primarily associated with ES in malignant primary cutaneous tumors. Classic-type ES typically shows mildly atypical cells with abundant cytoplasm often arranged in a granuloma-like pattern with central necrosis. Proximal-type ES exhibits rhabdoid morphology with nuclear pleomorphism arranged in sheets or nodules. Immunohistochemically, ES often expresses CD34 as well as a variety of cytokeratins and BAF47 immunohistochemistry demonstrates loss of nuclear expression. The main differential diagnosis in the skin is squamous cell carcinoma (SCC) which shows retention of BAF47 expression and typically stains for p63, p40 and CK5/6. In contrast, only a small minority of ES can show labeling for CK5/6 in rare tumor cells, and p63 and p40 expression is consistently absent in ES4, 6, 7. Molecular studies would demonstrate genetic alterations in SMARCB1 in ES and a UV-mutation signature (UV-MS) in SCCs8. Epithelial neoplasms with alterations in SMARCB1 and INI1 protein loss arising primarily in skin have not been described.
Herein, we present two cases of a novel primary SMARCB1-deficient carcinoma of skin (SDCS) delineating their clinical, pathological and molecular features, and explore the type and frequency of SMARCB1 somatic mutations within a large clinical sequencing cohort of non-melanoma skin cancers (NMSC).
Methods
Cases
The study was approved by the Memorial Sloan Kettering Cancer Center institutional research board (IRB). Pathology slides and molecular results on the 2 SMARCB1-deficient cutaneous carcinomas were reviewed by all authors including two trained dermatopathologists (YH and TJH) and two molecular and head and neck pathologists (PC and SD). The clinical MSK-IMPACT molecular cohort (>55,000 cases) was screened for the presence of SMARCB1 genetic alterations in NMSC profiled from 2014–2020.
Immunohistochemistry (IHC)
IHC was performed using clinical protocols with primary antibodies listed in (Table 1).
Table 1:
Immunohistochemistry
Primary antibody | Type | Clone | Manufacturer |
---|---|---|---|
Cytokeratin | mouse monoclonal | Cam 5.2 | Becton-Dickinson, Franklin Lakes, NJ |
BAF-47 | mouse monoclonal | 25/BAF47 | BD Bioscience, San Jose, CA |
BRG-1 | mouse monoclonal | G-7 | Santa Cruz Biotechnology, Dallas, TX |
BRST2 | mouse monoclonal | D6 | Biolegend, San Diego, CA |
p40 | mouse monoclonal | BC28 | Biocare Medical, Concord, CA |
p63 | mouse monoclonal | VS38C | Agilent Dako, Santa Clara, CA |
Cytokeratin | mouse monoclonal | AE1/AE3 | Agilent Dako, Santa Clara, CA |
CD34 | mouse monoclonal | QBend-10 | Ventana Medical Systems, Oro Valley, AZ |
CK7 | mouse monoclonal | OV-TL-12/30 | Agilent Dako, Santa Clara, CA |
34betaE12 | mouse monoclonal | 34betaE12 | Ventana Medical Systems, Oro Valley, AZ |
CK 5/6 | mouse monoclonal | D5/16B4 | Ventana Medical Systems, Oro Valley, AZ |
TTF1 | mouse monoclonal | 8G7G3/1 | Ventana Medical Systems, Oro Valley, AZ |
PAX8 | rabbit polyclonal | 10336–1-AP | Proteintech Group, Rosemont, IL |
chromogranin | mouse monoclonal | LK2H10 | Ventana Medical Systems, Oro Valley, AZ |
ER | mouse monoclonal | 6F11 | Leica Biosystems, Buffalo Grove, IL |
PR | mouse monoclonal | 16 | Leica Biosystems, Buffalo Grove, IL |
NUT | rabbit monoclonal | C52B1 | Cell Signaling, Danvers, MA |
HMB45 | mouse monoclonal | HMB45 | Agilent Dako, Santa Clara, CA |
S100 | rabbit polyclonal | S100 | Agilent Dako, Santa Clara, CA |
Molecular profiling and copy number alterations (CNA) analysis
Formalin-fixed paraffin-embedded (FFPE) tissue was submitted for tumor DNA extraction and corresponding germline DNA was obtained from peripheral blood samples. All cases were clinically tested by a targeted hybridization capture-based massively parallel sequencing assay, Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT™), a clinically validated molecular assay interrogating somatic variants in 468 cancer-related genes as previously described9, 10. Sequence coverage of targeted regions in a tumor sample were compared relative to a standard diploid normal sample to identify CNA. CNA were expressed as the log2 transformed tumor/normal ratio and a cutoff of 2.0-fold change was required to consider a gene amplified or deleted9, 10. Copy number/zygosity assessment was assessed by Fraction and Allele-Specific Copy Number Estimates from Tumor Sequencing (FACETS) as previously described11 and was performed on all SMARCB1 alterations and on coexisting mutations if designated as likely or predicted oncogenic. The OncoKB annotation in cBioPortal was utilized to evaluate oncogenicity12.
Results
a). SMARCB1-deficient carcinoma of skin – two case reports
Case 1
Clinical course and pathologic findings
An 18-year-old female with no exposures and a prior history of left parietal alopecia presented with a subcutaneous nodule. The initial clinical impression was that of a benign cyst and she was observed for one year. She then returned with multiple nodules in the occipital region, scalp and left posterior neck skin and lymph nodes and underwent surgical resection (Table 2). Review of the pathology slides on her initial resection revealed an infiltrative, undifferentiated, non-keratinizing malignancy involving the dermis with no connection to the overlying epidermis noted in the provided biopsy material (Figure 1). The tumor cells were positive for AE1/AE3 and p63 and negative for ER, PR, BRST2, Melan A and TTF-1 by immunohistochemistry. She was diagnosed with a poorly differentiated carcinoma involving dermis, metastatic to lymph nodes and was further treated with radiation. No other primary tumors were found on imaging. After one year, she recurred with left neck and right axillary lymph node metastasis when she presented to our institution for further management. Her axillary lymph node metastasis was morphologically similar to the primary tumor and was positive for AE1/AE3, Cam5.2 and p40 and negative for SOX10. The concurrent cytologic preparations showed clusters and single polygonal tumor cells with indistinct nuclear borders and moderate to scant, delicate cytoplasm. Nuclei were irregular, with coarse to vesicular chromatin and prominent nucleoli. A molecular assay showed SMARCB1 deletion and prompted an additional IHC work-up of the tumor revealing nuclear loss of SMARCB1 protein, expression of claudin-4 and CK5/6, and lack of CD34 expression (Figure 1). The patient was treated with chemotherapy and immunotherapy but recurred within 7 months with cutaneous metastases on the chest showing an interstitial arrangement of the tumor cells morphologically similar to the primary scalp carcinoma. In addition, imaging work-up identified pulmonary nodules suspicious for metastases. The patient continued with the immunotherapy but succumbed to her disease 3 years and 10 months after her initial clinical presentation.
Table 2.
Clinical course of patients with SMARCB1-deficient poorly differentiated carcinoma of skin.
Case 1 | Year | 2015 | 2016 | 2017 | 2018 | |||
DX | Left parietal skin nodularity (clinically) | PDCA in dermis | Metastatic SDCS | SDCS in the dermis | ||||
Site | Left parietal skin | Left parietal, occipital, inferior and superior scalp, left posterior neck skin, left neck lymph nodes | Left neck lymph nodes, right axillary lymph node | Right chest skin; lung nodules (clinically consistent with metastasis) | ||||
RX | Observation | Surgical excision and RT | Chemo, IT | IT | ||||
Case 2 | Year | 2010 | 2012 | 2013 | 2014 | 2015 | 2016 | 2017 |
DX | BCC | BCC, SCC | SCC | SCC | Poorly differentiated SCC | Poorly differentiated malignant neoplasm (cytology) | SDCS | |
Site | Right temple | Nose, left neck, upper back (BCC), Left scalp (SCC) | Left scalp | Left scalp | Right temple, parotid, LVI, PNI, right neck lymph node metastasis with extranodal extension (stage pT2 pN2b pMx) | Right upper neck | Right submental soft tissue, right retroauricular area, right abdominal skin | |
RX | Surgical excision | Surgical excision | Surgical excision with skin graft | Surgical excision with reconstruction | Surgical resection, chemo, RT | RT, IT | RT, IT |
Abbreviations: DX, diagnosis; RX, treatment; PDCA, poorly differentiated carcinoma; SDCS, SMARCB1-deficient carcinoma of skin; RT, radiation therapy; IT, immunotherapy, BCC, basal cell carcinoma; SCC, squamous cell carcinoma; LVI, lymphovascular invasion; PNI, perineural invasion.
Figure 1.
SMARCB1-deficient carcinoma involving scalp of an 18-year-old female. The tumor cells infiltrate dermis in the form of nests and cords intersected by variably thick, dense collagenous stroma and is associated with lymphoid aggregates at the tumor periphery [A, H&E, scanning magnification]. The tumor cells surround adnexal structures and no connection to the overlying epidermis was identified [B, H&E, 200x magnification]. Perineural invasion is seen (arrow). The tumor cells are of intermediate size and show smooth cell borders. No prominent rhabdoid features are seen although in areas, they do have a subtle plasmacytoid appearance. The nuclear/cytoplasmic ratio is high and the nuclei are round with finely dispersed chromatin and one or multiple conspicuous nucleoli [C, H&E, 400x magnification]. A cytologic preparation reveals clusters of cells with indistinct nuclear borders and scant delicate cytoplasm, vesicular chromatin and prominent nucleoli [D, Papanicolaou stain, 600x magnification]. Immunohistochemistry revealed loss of nuclear INI1 expression [E, 400x magnification] and positive staining for claudin-4 [F, 400x magnification], p40 [G, 400x magnification], with patchy CK 5/6 expression throughout the tumor [H, 400x magnification].
A CNA plot of Case 1 shows homozygous deletion of SMARCB1 (green arrow). The Y-axis depicts copy number changes expressed as the log2 transformed tumor/normal ratio by their genomic positions indicated on the x-axis. The blue dots represent individual exons and red dots indicate a ≥2-fold tumor/normal ratio [I]. FACETS analysis shows deletion of both SMARCB1 alleles as indicated by the total integer copy number 0 (black line, y-axis). A red line indicates a minor allele. SMARCB1 genomic position on chromosome 22 is indicated by the vertical green line [H].
Abbreviations: FACETS, Fraction and Allele-Specific Copy Number Estimates from Tumor Sequencing; CNA, copy number alterations.
Molecular diagnosis
A targeted exome sequencing of the axillary lymph node metastasis revealed a SMARCB1 deletion and a single coexistent mutation in LATS2 of unknown functional significance (Table 3). FACETS analysis confirmed homozygous SMARCB1 deletion and an otherwise relatively quiet genome without many notable gains or losses (Figure 1).
Table 3.
Genetic alterations in SMARCB1-deficient carcinomas of skin.
Case | Functional significance | Gene | cDNA | AA | Mutation type | Zygosity/ploidy status | Variant allele frequency |
---|---|---|---|---|---|---|---|
18 F | Likely oncogenic | SMARCB1 | Whole gene deletion | N/A | Deep deletion | Homozygous deletion | N/A |
Unknown | LATS2 | c.2888G>A | p.G963E | Missense | 0.35 | ||
| |||||||
75 M | Likely or predicted oncogenic | SMARCB1 | c.617G>A | p.W206* | Nonsense | LOH | 0.44 |
KDM6A | c.3878+2T>G | p.X1293_splice | Splice site | LOH | 0.62 | ||
EP300 | c.4192C>T | p.L1398F | Missense | LOH | 0.39 | ||
TP53 | c.529_546delCCCCACCATGAGCGCTGC | p.P177_C182del | In frame deletion | LOH | 0.39 | ||
TP53 | c.867_868delinsTT | p.R290C | Missense | LOH | 0.38 | ||
FAT1 | c.9568G>T | p.E3190* | Nonsense | LOH | 0.38 | ||
KMT2D | c.3810_3811delAC | p.L1271Ifs*14 | Frameshift deletion | Diploid | 0.27 | ||
CIC | Inversion of exons 16–20 | N/A | Structural variant | N/A | N/A | ||
CDK12 | Intragenic deletion of exon 2 | N/A | Structural variant | N/A | N/A | ||
CDKN2A/p16INK4A | Whole gene deletion | N/A | Deep deletion | Homozygous deletion | N/A | ||
CDKN2A/p14ARF | Whole gene deletion | N/A | Deep deletion | Homozygous deletion | N/A | ||
CDKN2B | Whole gene deletion | N/A | Deep deletion | Homozygous deletion | N/A | ||
PTPRD | Whole gene deletion | N/A | Deep deletion | Homozygous deletion | N/A | ||
| |||||||
Unknown | KDM5C | c.1318T>C | p.Y440H | Missense | 0.57 | ||
EIF4A2 | c.307C>T | p.Q103* | Nonsense | 0.49 | |||
FGFR1 | c.2378C>T | p.S793F | Missense | 0.44 | |||
MST1R | c.4189C>T | p.P1397S | Missense | 0.40 | |||
INPP4B | c.1672G>A | p.G558R | Missense | 0.40 | |||
NOTCH1 | c.517C>T | p.P173S | Missense | 0.40 | |||
AGO2 | c.1276G>A | p.A426T | Missense | 0.37 | |||
U2AF1 | c.598C>T | p.R200W | Missense | 0.36 | |||
JAK2 | c.2780T>C | p.L927S | Missense | 0.36 | |||
MEF2B | c.980C>G | p.P327R | Missense | 0.32 | |||
IKBKE | c.571C>T | p.R191* | Nonsense | 0.31 | |||
ASXL2 | c.2060G>A | p.G687E | Missense | 0.31 | |||
RET | c.3095G>A | p.G1032D | Missense | 0.30 | |||
MDM4 | c.272C>T | p.S91F | Missense | 0.30 | |||
CIC | c.338C>T | p.P113L | Missense | 0.29 | |||
EPAS1 | c.1558G>A | p.D520N | Missense | 0.29 | |||
TERT | c.1247G>T | p.R416L | Missense | 0.29 | |||
MDC1 | c.5498C>T | p.S1833F | Missense | 0.29 | |||
NSD2 | c.2089C>T | p.L697F | Missense | 0.29 | |||
KIT | c.447_448delinsAA | p.G150R | Missense | 0.29 | |||
SF3B1 | c.104C>T | p.S35F | Missense | 0.29 | |||
ROS1 | c.2822C>T | p.S941F | Missense | 0.28 | |||
SDHA | c.589G>A | p.G197S | Missense | 0.28 | |||
ZFHX3 | c.4027G>A | p.D1343N | Missense | 0.28 | |||
POLD1 | c.1139_1140delinsTT | p.A380V | Missense | 0.28 | |||
KMT2D | c.4388_4389delinsTT | p.T1463I | Missense | 0.28 | |||
ASXL1 | c.3500C>T | p.P1167L | Missense | 0.28 | |||
FGFR2 | c.1268C>T | p.P423L | Missense | 0.28 | |||
ROS1 | c.1855C>T | p.P619S | Missense | 0.28 | |||
CENPA | c.263C>T | p.A88V | Missense | 0.27 | |||
MGA | c.775C>T | p.R259W | Missense | 0.27 | |||
NOTCH2 | c.3707T>C | p.L1236P | Missense | 0.27 | |||
ERBB4 | c.716C>T | p.S239L | Missense | 0.26 | |||
CBL | c.1282C>T | p.P428S | Missense | 0.26 | |||
ARID2 | c.1418C>T | p.S473F | Missense | 0.25 | |||
CDC73 | c.251C>T | p.P84L | Missense | 0.25 | |||
BLM | c.2186C>T | p.S729F | Missense | 0.25 | |||
PREX2 | c.1673C>T | p.P558L | Missense | 0.24 | |||
IL7R | c.801–1G>T | p.X267_splice | Splice site | 0.24 | |||
RPS6KA4 | c.2300G>A | p.G767D | Missense | 0.24 | |||
EPHB1 | c.2732C>T | p.T911M | Missense | 0.23 | |||
ARID5B | c.1759_1760delinsAT | p.E587I | Missense | 0.22 | |||
TGFBR1 | c.1183T>C | p.S395P | Missense | 0.18 | |||
PAK5 | c.1259_1260delinsTT | p.S420F | Missense | 0.12 | |||
PTCH1 | c.1322G>A | p.R441H | Missense | 0.10 | |||
MDC1 | Intragenic deletion of exons 10–12 | N/A | Structural variant | 0 | |||
TEK | Whole gene deletion | N/A | Deep deletion | N/A |
Abbreviations: AA, amino acid; LOH, loss of heterozygosity; N/A, not available
Case 2
Clinical course and pathology findings
A 76-year-old male with a prior history of prostatic adenocarcinoma presented with a 6-year history of multiple basal cell carcinomas (BCC) and SCC of the scalp, face and neck skin treated with surgical excisions, chemoradiation and immunotherapy. His clinical course and pathologic diagnoses are summarized in Table 2. He presented to our hospital in 2017 for further management. Pathology material before 2015 was not available for our review and comparison. Representative sections of the right temple and parotid resection specimen slides (2015) were reviewed. No other primary sites were suggested on imaging studies. The tumor involved the dermis and subcutis comprised of nests, cords and sheets of undifferentiated tumor cells within a collagenous and myxoid stroma. Connection to the overlying epidermis, keratinization, or definitive morphologic evidence of squamous differentiation was not seen in representatively submitted sections. However, in view of positive p63 and 34BE12 cytokeratin protein expression along with negative SOX10, p16, chromogranin, CK7 and CK20, the diagnostic interpretation was most consistent with poorly differentiated SCC of skin (Figure 2). At the same time, the patient underwent a submental soft tissue and retroauricular mass biopsy. On microscopic examination, the undifferentiated tumor cells were morphologically similar to the tumor of the right temple (2015). An extensive diagnostic work-up of a new biopsy revealed the tumor cells were positive for 34BE12, CK5/6, and p40, while negative for CD34, CK7, CK20, TTF-1, PAX8, NUT, S-100, chromogranin and HMB45. In addition, the tumor showed nuclear loss of SMARCB1 (INI1), retained nuclear SMARCA4 (BRG1) and positive labeling for claudin-4. A diagnosis of poorly differentiated carcinoma, SMARCB1-deficient was rendered. The tumor cells in the concurrent cytology specimen were similar, also showing multinucleation (Figure 2). Molecular testing results supported this interpretation (Table 3). Subsequently, he developed abdominal cutaneous metastases. In the view of molecular findings, he was considered for targeted therapy, but his performance status precluded eligibility for a clinical trial. He underwent proton therapy but expired from progression of his disease 61 months after his first skin cancer diagnosis and 24 months after his diagnosis of poorly differentiated carcinoma on the right temple.
Figure 2.
SMARCB1-deficient carcinoma involving the temple skin of a 75-year-old man. Infiltrative undifferentiated tumor cells in dermis are surrounded by collagenous and myxoid stroma [A, H&E, scanning magnification, inset 200x magnification]. Areas of necrosis were present, and apoptotic bodies and mitotic figures are notable. The tumor cells are variably sized, show high nuclear/cytoplasmic ratio, small to moderate amount of eosinophilic, focally very dense/pink cytoplasm [B, H&E, 200x magnification]. The undifferentiated tumor cells show no distinct rhabdoid features. Nuclei are round to oval with open or speckled chromatin, with one or multiple, focally prominent nucleoli [C, H&E, 400x magnification]. A cytologic preparation shows dishesive cells with moderate cytoplasm, prominent nucleoli and multinucleation [D, Diff-Quik stain, 400x magnification]. Immunohistochemistry also revealed loss of nuclear INI1 expression [E, 400x magnification] and positive staining for claudin-4 [F, 400x magnification], p40 [G, 400x magnification], and CK 5/6 [H, 400x magnification].
A CNA plot for Case 2 shows multiple gains and losses including homozygous CDKN2A deletion on chromosome 9p [red dots, I], which is confirmed by FACETS as the total copy number 0 [blue arrow, J]. LOH in SMARCB1 (green line) is indicated as the total copy number of 1 [J].
Abbreviations: FACETS, Fraction and Allele-Specific Copy Number Estimates from Tumor Sequencing; CNA, copy number alterations; LOH, loss of heterozygosity.
Molecular diagnosis
The submental biopsy was subjected to the targeted exome sequencing assay revealing 60 somatic genetic alterations including a truncating SMARCB1 W206* variant at a 44% variant allele frequency (VAF) (Table 3). FACETS analysis showed loss of heterozygosity (LOH) in SMARCB1 consistent with INI1 protein loss (Figure 2). Twelve additional likely or predicted oncogenic variants were detected including TP53 mutations associated with LOH and CDKN2A homozygous deletion. An abundance of G>A and C>T transitions were compatible with a UV mutation signature (UV-MS) (Table 3).
c). Molecular epidemiology of SMARCB1 genetic alterations in NMSC
Prompted by the identification of these two SDCS cases described above, the MSK-IMPACT database was interrogated for somatic SMARCB1 mutations in NMSC excluding ES of skin. A total of 378 patients NMSC, mostly with carcinomas (n=370, 98%) comprised of 406 specimens (239 primaries and 167 metastases) were genotyped by MSK-IMPACT. These included Merkel cell carcinoma (MCC, n=122), squamous cell carcinoma (n=119), extramammary Paget disease (n=65), basal cell carcinoma (n=48), skin adnexal carcinomas (n=40), dermatofibrosarcoma protuberans (n=8), aggressive digital papillary adenocarcinoma (n=2) and the two SDCS described herein. Seven additional cases with SMARCB1 genetic alterations including 3 SCC, 3 MCC and one BCC were identified. In addition to a single G>T transversion, six of 8 SMARCB1 genetic alterations were either C>T, G>A, CC>TT or GG>AA transitions and all but one case showed UV-MS. INI1 IHC performed on all cases with possible loss-of-function mutations with available material (n=5) was retained precluding the diagnosis of SMARCB1-deficient carcinoma in any of 7 additional cases (Table 4).
Table 4.
SMARCB1 mutations of undetermined significance in carcinomas of skin.
Age/sex | Diagnosis | cDNA | AA | Mutation type | Zygosity/ploidy status | Functional significance | INI1 IHC | # Mut | UV-S |
---|---|---|---|---|---|---|---|---|---|
71 M | SCC of scalp, BCC of forehead, arm and shoulder met to LN | AMP | N/A | AMP | AMP | Unknown | N/A | 38 | Yes |
53 M | MCC met to parotid | c.1102C>T | p.Q368* | Nonsense | Diploid | Likely oncogenic | Retained | 53 | Yes |
79 M | MCC met to chest wall | c.1130G>A | p.R377H | Missense | Diploid | Predicted oncogenic | N/A* | 41 | Yes |
83 M | SCC of chest and shoulder met to LN | c.1130G>A | p.R377H | Missense | Diploid | Predicted oncogenic | Retained | 7 | No |
63 M | SCC met to parotid | c.494C>T | p.P165L | Missense | Heterozygous gain | Unknown | Retained | 68 | Yes |
66 F | BCC met to LN | c.868_869delinsTT | p.P290L | Missense | LOH | Unknown | Retained | 48 | Yes |
68 M | MCC met to LN | c.378G>T | p.K126N | Missense | Diploid | Unknown | N/A | 46 | Yes |
c.1143_1144delinsAA | p.A382T | Missense | Diploid | Unknown | N/A |
Abbreviations: SCC, squamous cell carcinoma; BCC, basal cell carcinoma; MCC, Merkel cell carcinoma; LN, lymph node; met, metastasis; PD, poorly differentiated; AMP, amplification; AA, aminoacid; LOH, loss of heterozygosity; N/A, not available; # Mut, number of mutations in the sample; UV-S, UV signature.
not performed but presumed retained
Discussion
We describe 2 cases of SDCS and examined the frequency and the type of SMARCB1 mutations in a large cohort of NMSC, which is enriched for metastatic and advanced cases, predominantly carcinomas, profiled by massive parallel sequencing. We found that in NMSC, SMARCB1 mutations are rare and most are variants of unknown functional significance. Retained INI1 protein expression in most cases and the presence of UV-MS evidenced by C>T, G>A, CC>TT, or GG>AA transitions affecting SMARCB1 suggest these are UV-induced passenger mutations. However, true SMARCB1-deficient carcinomas can arise in skin and can harbor a SMARCB1 homozygous deletion or truncating SMARCB1 mutations associated with LOH and clinically aggressive behavior.
Both our cases were histopathologically undifferentiated, morphologically similar and involved the dermis without demonstrated connection to the overlying epidermis in representatively submitted specimens and/or limited biopsy material. Unlike other SMARCB1-deficient tumors, rhabdoid morphology was inconspicuous in both cases with tumor cells instead displaying a plasmacytoid appearance4. Despite having an immunophenotype commonly seen in SCC, such as expression of CK5/6, p40 and p63, absence of squamous differentiation features, such as keratinization, can aid in distinguishing these cases from typical cutaneous SCC. Moreover, SCC arising in the scalp of a very young woman (Case 1) with biologically aggressive behavior similar to that in SMARCB1-deficient sinonasal carcinoma13, 14 would be unusual. While her age, clinical presentation and the tumor genotype would be more in line with ES, positive p40 and claudin-4, and negative CD34 expression argue against this being ES and support epithelial rather than mesenchymal differentiation. Indeed, Schaefer et al. reported the use of claudin-4 expression to distinguish SWI/SNF complex-deficient undifferentiated carcinomas from sarcomas. They demonstrated no expression of claudin-4 in ES whereas 80% of carcinomas harboring SWI/SNF complex deficiency were positive15.
In Case 2, the patient’s age, tumor location, prior history of multiple recurrent SCC and BCC along with a finding of UV-MS16, 17 could be compatible with another SCC. A positive p40/p63, diffuse CK 5/6 expression and negative CD34 would further support squamous differentiation and reinforced an immunophenotype that was incongruous with ES. Nevertheless, while the clinical history and the immunophenotype in Case 2 could justify a diagnosis of poorly differentiated SCC, we suggest that such cases should be separated from a variety of cutaneous SCC and recognized as SDCS for the following reasons: First, a pathogenic SMARCB1 variant consistent with loss of INI1 represents an opportunity to apply novel targeted therapies. Second, the tumor morphology does not support a conventional cutaneous SCC. Third, this tumor appears to be clinically more aggressive than most SCC.
EZH2 inhibitors are novel therapeutic agents for SMARCB1-deficient malignancies. The SWI/SNF complex which includes SMARCB1 is a regulator of the Polycomb Repressive Complex 2 (PRC2) of which EZH2 is a member. Accordingly, loss of SMARCB1 results in deregulation of EZH2 and PRC2-mediated gene silencing that support retention of a stem cell phenotype and, in turn, oncogenesis18, 19. In SDCS, deficiency in SMARCB1 may render them sensitive to EZH2 inhibitors. Of note, our second patient had been considered for this therapy as part of a clinical trial after an initial course of chemotherapy but did not meet the qualifications of the study. The FDA has recently approved tazemetostat as the first EZH2 inhibitor for the treatment of advanced or metastatic ES20. With this approval, detection of SMARCB1 alterations in these carcinomas may now have theragnostic implications.
At the molecular level, the cases were distinct. Case 1 showed a homozygous SMARCB1 deletion and very few coexisting genetic alterations of unknown functional significance. Such molecular profile would be similar to those seen in most other SMARCB1 deficiency-driven malignancies5, 21, 22, 23 and emphasize the oncogenic role of SMARCB1 protein loss. In contrast, Case 2 harbored a truncating SMARCB1 W206* variant associated with LOH coexisting with multiple other, likely oncogenic variants involving mainly tumor suppressor genes. While the question of a putative driver in the latter case remains open, a relatively high SMARCB1 VAF suggests its presence in the majority of tumor cells arguing against this being a subclonal event present only in a minor subset of tumor cells. Furthermore, the SMARCB1 variant in Case 2 occurred in the context of UV-MS and was also G>A transition suggesting a UV-induced etiology.
Finally, the tumor phenotype and molecular alterations detected also lent credence to our conclusion that these tumors represent a novel category of malignant neoplasm that should be recognized and distinguished from its mimics. SMARCB1-deficient tumors have been described arising in skin of special sites including the eyelid but these involved the lacrimal gland24. Folpe et al. reviewed a series of malignant vulvar tumors exhibiting SMARCB1-loss classifying them into proximal-type ES, distal-type ES, and malignant myoepithelioma categories25. However, the cases in the latter studies have not been characterized by p40, p63 or CK5/6 and is difficult to make comparison with the SDCS cases in the current study.
In summary, we present two cases of a novel malignant neoplasm with epithelial phenotype carrying genetic alterations in SMARCB1 resulting in INI1 loss arising in the skin that exhibits an aggressive clinical course. An IHC work-up including INI1 and claudin-4 should be performed in any poorly differentiated/undifferentiated cutaneous malignancy exhibiting a squamous immunophenotype but lacking any morphologic evidence of squamous differentiation. An accurate, timely diagnosis of SDCS can appropriately direct disease management and be critical in qualifying these patients for novel targeted therapeutic agents.
Ethics statement:
This study was approved by the MSKCC institutional research review board to be compliant with ethical standards.
Acknowledgments
Research reported in this publication was supported by the Cancer Center Support Grant of the National Institutes of Health/National Cancer Institute under award number P30CA008748.
Footnotes
Declarations of interest: None.
Funding disclosures: None.
References:
- 1.Arnaud O, Le Loarer F, Tirode F. BAFfling pathologies: Alterations of BAF complexes in cancer. Cancer Lett 2018; 419(1):266–279. [DOI] [PubMed] [Google Scholar]
- 2.Kohashi K, Oda Y. Oncogenic roles of SMARCB1/INI1 and its deficient tumors. Cancer Sci 2017; 108(4):547–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Pawel BR. SMARCB1-deficient tumors of childhood: A practical guide. Pediatr Dev Pathol 2018. 21(1):6–28. [DOI] [PubMed] [Google Scholar]
- 4.Thway K, Jones RL, Noujaim J, Fisher C. Epithelioid sarcoma: Diagnostic features and genetics. Adv Anat Pathol 2016; 23(1):41–49. [DOI] [PubMed] [Google Scholar]
- 5.Forrest SJ, Al-Ibraheemi A, Doan D, et al. Genomic and immunologic characterization of INI-deficient pediatric cancers. Clin Cancer Res 2020; 26(12):2882–2890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Lin L, Skacel M, Sigel JE, et al. Epithelioid sarcoma: an immunohistochemical analysis evaluation the utility of cytokeratin 5/6 in distinguishing superficial epithelioid sarcoma from spindled squamous cell carcinoma. J Cutan Pathol 2003; 30(2):114–117. [DOI] [PubMed] [Google Scholar]
- 7.Agaimy A SWI/SNF complex-deficient soft tissue neoplasms. A pattern-based approach to diagnosis and differential diagnosis. Surg Pathol Clin 2019; 12(1):149–163. [DOI] [PubMed] [Google Scholar]
- 8.Shen Y, Kim AL, Du R, Liu L. Transcriptome analysis identifies the dysregulation of ultraviolet target genes in human cancers. PLoS One 2016; 11(9):e0163054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Cheng DT, Mitchell TN, Zehir A, et al. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): A hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J Mol Diagn 2015; 17(3):251–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zehir A, Benayed R, Shah RH, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med 2017; 23(6):703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Shen R, Seshan VE. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res 2016; 44(16):e131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chakravarty D, Gao J, Phillips SM, et al. OncoKB: A Precision Oncology Knowledge Base. JCO Precis Oncol 2017; 2017(1):1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Dogan S, Vasudevaraja V, Xu B, et al. DNA methylation-based classification of sinonasal undifferentiated carcinoma. Mod Pathol 2019; 32(10):1447–1459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Agaimy A, Hartmann A, Antonescu CR, et al. SMARCB1 (INI-1)-deficient Sinonasal Carcinoma: A Series of 39 Cases Expanding the Morphologic and Clinicopathologic Spectrum of a Recently Described Entity. Am J Surg Pathol 2017; 41(4):458–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Schaefer IM, Agaimy A, Fletcher CD, Hornick JL. Claudin-4 expression distinguishes SWI/SNF complex-deficient undifferentiated carcinomas from sarcomas. Mod Pathol 2017; 30(4):539–548. [DOI] [PubMed] [Google Scholar]
- 16.Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature 2013; 500(7463):415–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Pfeifer GP. Mechanisms of UV-induced mutations and skin cancer. Genome Instability & Disease 2020; 1(1):99–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Rothbart SB, Baylin SB. Epigenetic therapy for epithelioid sarcoma. Cell 2020; 181(2):211. [DOI] [PubMed] [Google Scholar]
- 19.Italiano A Targetiing epigenetics in sarcomas through EZH2 inhibition. J Hematol Oncol 2020; 13(1):33–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hoy SM. Tazemetostat: First approval. Drugs 2020; 80(5):513–521. [DOI] [PubMed] [Google Scholar]
- 21.Biegel JA, Fogelgren B, Zhou J-Y, et al. Mutations of the INI1 rhabdoid tumor suppressor gene in medulloblastomas and primitive neuroectodermal tumors of the central nervous system. Clin Cancer Res 2000; 6(7):2759–2763. [PubMed] [Google Scholar]
- 22.Modena P, Lualdi E, Facchinetti F, et al. SMARCB1/INI1 tumor suppressor gene is frequently inactivated in epithelioid sarcomas. Cancer Res 2005; 65(10):4012–4019. [DOI] [PubMed] [Google Scholar]
- 23.Cotzia P, Ptashkin RN, Gounder MM, et al. Genetic and Histologic Spectrum of SMARCB1-deficient Carcinomas of the Head and Neck Including Sinonasal Tract, Thyroid and Skin. Lab Investigation 2018; 98(suppl 1):474–475. [Google Scholar]
- 24.Srinivasan A, Tuluc M, Jordan AJ, Curry JM, Bilyk JR. INI/SMARCB1-deficient carcinoma (rhabdoid tumor) of the lacrimal gland. Ophthalmic Plast Reconstr Surg 2019; 35(2):e41–e43. [DOI] [PubMed] [Google Scholar]
- 25.Folpe AL, Schoolmeester JK, McCluggage WG, et al. SMARCB1-deficient vulvar neoplasms: A Clinicopathologic, immunohistochemical, and molecular genetic study of 14 cases. Am J Surg Pathol 2015; 39(6):836–849. [DOI] [PubMed] [Google Scholar]