Inviting an author to review:
Find an author and click ‘Invite to review selected article’ near their name.
Search for authorsSearch for similar articles
2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Primary intrathoracic liposarcomas: A clinicopathologic and molecular study of 43 cases in one of the largest medical centers of China

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction

          Primary intrathoracic liposarcoma is extremely rare, and most published series lack genetic analyses. The aim of our study is to better understand the clinicopathologic and genetic features of these rare lesions.

          Materials and methods

          Forty-three primary intrathoracic liposarcomas were identified and most cases were analyzed by systematic genetic studies, including fluorescence in situ hybridization (FISH), whole-exome sequencing (WES), and Sanger sequencing.

          Results

          This series included 27 males and 16 females (ratios, 1.68:1) aged 24-73 years (median, 53 years). Tumors mainly occurred in the mediastinum (n=23, 53.5%), followed by pleural cavity (n=16, 37.2%) and lung (n=4, 9.3%). The study included 21 well-differentiated liposarcomas (WDLs), 19 dedifferentiated liposarcomas (DDLs), 2 myxoid pleomorphic liposarcomas (MPLs) and 1 pleomorphic liposarcoma (PL), without identification of myxoid liposarcoma. FISH analysis identified MDM2 amplification in 17 of 18 WDLs (94.4%) and all DDLs (16/16, 100.0%). The MDM2-nonamplified WDL was CDK4-nonamplified but FRS2-amplified. WES and Sanger sequencing found somatic TP53 mutation in the 2 MPLs. Follow-up information was available for 33 of 38 cases (86.8%). Thirteen patients (39.4%) showed no evidence of disease, 10 patients (30.3%) were alive with disease, and 8 patients (24.2%) died of disease. Fourteen cases developed recurrence and 1 with metastasis.

          Conclusions

          WDL/DDL was the overwhelming subtype in this location, followed by MPL and PL. Analysis of the FRS2 gene, in combination with MDM2 and other genes of 12q13-15, may more precisely characterize WDL/DDLs. MPL is the most fatal subtype of this site. Further studies are needed to explore the role of TP53 in the pathogenesis of MPL.

          Related collections

          Most cited references67

          • Record: found
          • Abstract: found
          • Article: not found

          Identification of recurrent NAB2-STAT6 gene fusions in solitary fibrous tumor by integrative sequencing.

          A 44-year old woman with recurrent solitary fibrous tumor (SFT)/hemangiopericytoma was enrolled in a clinical sequencing program including whole-exome and transcriptome sequencing. A gene fusion of the transcriptional repressor NAB2 with the transcriptional activator STAT6 was detected. Transcriptome sequencing of 27 additional SFTs identified the presence of a NAB2-STAT6 gene fusion in all tumors. Using RT-PCR and sequencing, we detected this fusion in all 51 SFTs, indicating high levels of recurrence. Expression of NAB2-STAT6 fusion proteins was confirmed in SFT, and the predicted fusion products harbor the early growth response (EGR)-binding domain of NAB2 fused to the activation domain of STAT6. Overexpression of the NAB2-STAT6 gene fusion induced proliferation in cultured cells and activated the expression of EGR-responsive genes. These studies establish NAB2-STAT6 as the defining driver mutation of SFT and provide an example of how neoplasia can be initiated by converting a transcriptional repressor of mitogenic pathways into a transcriptional activator.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Subtype-specific genomic alterations define new targets for soft tissue sarcoma therapy

            Introductory Paragraph Soft tissue sarcomas, which encompass approximately 10,700 diagnoses and 3800 deaths per year in the US1, exhibit remarkable histologic diversity, with more than 50 recognized subtypes2. However, knowledge of their genomic alterations is limited. We describe an integrative analysis of DNA sequence, copy number, and mRNA expression in 207 samples encompassing seven major subtypes. Frequently mutated genes included TP53 (17% of pleomorphic liposarcomas), NF1 (10.5% of myxofibrosarcomas and 8% of pleomorphic liposarcomas), and PIK3CA (18% of myxoid/round-cell liposarcomas). PIK3CA mutations in myxoid/round-cell liposarcomas were associated with AKT activation and poor clinical outcomes. In myxofibrosarcomas and pleomorphic liposarcomas, we found both point mutations and genomic deletions affecting the tumor suppressor NF1. Finally, we found that shRNA-based knockdown of several genes amplified in dedifferentiated liposarcoma, including CDK4 and YEATS4, decreased cell proliferation. Our study yields a detailed map of molecular alterations across diverse sarcoma subtypes and provides potential subtype-specific targets for therapy. Current knowledge of the key genomic aberrations in soft tissue sarcoma is limited to the most recurrent alterations or translocations. Subtypes with simple, near-diploid karyotypes bear few chromosomal rearrangements but have pathognomonic alterations: translocations in myxoid/round-cell liposarcoma (MRC) [t(12;16)(q13;p11), t(12;22)(q13;q12)] and synovial sarcomas (SS) [t(X;18)(p11;q11)]; activating mutations in KIT or PDGFRA in gastrointestinal stromal tumors (GIST)3,4. The discovery of the latter mutations led to the clinical deployment of imatinib for the treatment of GIST5, providing a model for genotype-directed therapies in molecularly defined sarcoma subtypes. Conversely, sarcomas with complex karyotypes, including dedifferentiated and pleomorphic liposarcoma, leiomyosarcoma, and myxofibrosarcoma, have no known characteristic mutations or fusion genes, although abnormalities are frequently observed in the Rb, p53, and specific growth-factor signaling pathways6. Recent large-scale analyses7–10 have established a standard for cancer genome studies, but soft tissue sarcomas have not yet been a focus of this type of effort. Given the urgent need for new treatments for the ~4000 patients who die each year in the US of soft tissue sarcoma1, we sought to identify novel genomic alterations that could serve as therapeutic targets. Here, we describe complementary genome and functional genetic analyses of seven subtypes of high-grade soft tissue sarcoma (Table 1 and Supplementary Table 1) to discover subtype-specific events. Several of our findings, detailed below, could have nearly immediate therapeutic implications. To study the genomic alterations in sarcomas, we initially analyzed 47 tumor/normal DNA pairs encompassing six soft tissue sarcoma subtypes by sequencing 722 protein-coding and microRNA genes, followed by verifying discovered mutations with mass spectrometry-based genotyping (see Methods, Supplementary Figure 1A, and Supplementary Table 2). The results revealed 28 somatic non-synonymous coding point mutations and 9 somatic insertions/deletions (indels) involving 21 genes in total (Table 2 and Supplementary Figure 1B). No mutations were detected in microRNAs genes. We extended the analysis to an additional 160 tumors, where we genotyped each of the mutations found above and re-sequenced exons of NF1 and ERBB4 in pleomorphic liposarcoma and myxofibrosarcoma, PIK3CA and KIT in myxoid/round cell liposarcoma, and CDH1 in dedifferentiated liposarcoma; this revealed nine additional mutations (Table 2 and Supplementary Table 3). KIT was frequently mutated in GISTs and unexpectedly, in one myxoid/round cell liposarcoma sample (Supplementary Note). The next most frequently mutated genes observed within specific sarcoma subtypes were PIK3CA, in 18% of myxoid/round cell liposarcomas, TP53 in 17% of pleomorphic liposarcomas (interestingly, the only subtype in which mutations of this gene were found), and NF1 in 10.5% of myxofibrosarcomas and 8% of pleomorphic liposarcomas (Table 2 and Figure 1). Additional genes, including protein and lipid kinases, as well as known or candidate tumor suppressor genes, were found mutated in just one sample for each sarcoma subtype (Table 2, Figure 1, and Supplementary Note). Further studies will be needed to establish the functional impact of these mutations in sarcoma. Below, we focus on three major specific genomic findings with therapeutic implications: point mutation and deletion of NF1 in a subset of soft tissue sarcomas, point mutation of PIK3CA in myxoid/round cell liposarcoma, and the complex pattern of amplification of chromosome 12q in dedifferentiated liposarcoma. Integrated analysis of DNA copy number, expression, and mutation data uncovered diverse alterations of the Neurofibromatosis type 1 gene (NF1) in several sarcoma subtypes. While germline and somatic inactivation of NF1 is associated with malignant peripheral nerve sheath tumors11 and GISTs in Neurofibromatosis type 1 patients12, no somatic NF1 alterations have been reported in other sarcomas. We detected six point mutations and twelve genomic deletions encompassing the NF1 locus, occurring in both myxofibrosarcoma and pleomorphic liposarcoma (Table 2 and Figure 1, 2A–B; copy number analysis discussed further below). Two of the mutations, R304* and Q369*, were previously reported as germline mutations in patients with Neurofibromatosis type 113,14, while the other four mutations (three missense and one nonsense) have not been previously reported. In some tumors, biallelic inactivation was evident, with heterozygous point mutations accompanied by deletion of the wild-type allele and correspondingly reduced gene expression compared to normal adipose tissue15 in most cases (Figure 2B). Together, these data indicate a diverse pattern of NF1 aberrations in myxofibrosarcomas and pleomorphic liposarcomas. These results complement recent reports of NF1 alterations in lung cancers and glioblastomas7,8. PIK3CA, encoding the catalytic subunit of phosphatidylinositol 3-kinase (PI3K), had one of the highest somatic mutation frequencies among the genes in this analysis (Table 2). Nucleotide substitutions in PIK3CA were initially detected in 4 of 21 myxoid/round-cell liposarcomas (MRCs). We measured the frequency of point mutations in PIK3CA in this subtype by genotyping an independent cohort of 50 MRCs16 for 13 common sites of PIK3CA mutation, including those discovered in our initial sequencing; mutations were detected in 9 additional patients (in total, 13 of 71). The mutations were clustered in two domains, the helical domain (E542K and E545K) and the kinase domain (H1047L and H1047R) (Table 2); both these domains are also mutated in epithelial tumors17. MRC patients whose tumors harbored mutations in PIK3CA had a shorter duration of disease–specific survival than did those with wildtype PIK3CA (p=0.036, log-rank test). Similar to observations in breast cancers18, patients with helical-domain PIK3CA mutations had worse outcomes than those with kinase-domain mutations (Figure 3A). However, this difference was not statistically significant given the small number of cases in our study. As both helical- and kinase-domain PIK3CA mutants are believed to activate Akt, although through different mechanisms19–21, we assessed Akt activation in MRC tumors harboring wildtype and mutated PIK3CA. Of note, only E545K helical-domain mutations were associated with increased Akt phosphorylation relative to wildtype, both at serine-473 and threonine-308 (TORC2 and PDK1 phosphorylation sites, respectively), and with increased phosphorylation of Akt substrates PRAS40 and S6 kinase (Figure 3B). Surprisingly, tumors with H1047R kinase-domain mutations did not have similar increases in Akt phosphorylation or activation (Figure 3B). However, H1047R-mutant tumors exhibited variably higher levels of PTEN, a negative regulator of PI3K activity, which may partly explain lower Akt activity. In addition, we detected a single MRC tumor with homozygous PTEN deletion and high Akt phosphorylation levels (data not shown). Further studies are needed to determine the relationship between activated PI3K signaling (resulting from PIK3CA mutations) and the pathognomonic t(12;16)(q13;p11) translocation in this subtype. In addition to sequencing, we characterized the spectrum of genomic aberrations in soft tissue sarcoma with 250K single nucleotide polymorphism (SNP) arrays for somatic copy number alterations (SCNAs: n=207; Figure 1 and Supplementary Figure 2A) and loss-of-heterozygosity (LOH) (n=200; Supplementary Figure 2B) and with oligonucleotide gene expression arrays (n=149) (see Methods). The patterns of statistically significant SCNAs22,23 (Figure 1) revealed substantial differences between subtypes with simple and complex karyotypes (Figure 1). Myxoid/round-cell liposarcoma, synovial sarcoma, and GIST had relatively normal karyotypes compared to dedifferentiated and pleomorphic liposarcoma, leiomyosarcoma, and myxofibrosarcoma. In addition, only the four complex subtypes harbored significant copy-neutral LOH (Supplementary Figure 2B and Supplementary Table 4). These types exhibit varied levels of complexity: both dedifferentiated liposarcoma and leiomyosarcoma are less complex than pleomorphic liposarcoma and myxofibrosarcoma (Figure 1). The latter two subtypes were strikingly similar (Figure 1 and Supplementary Figure 2A), indicating they might appropriately be considered a single entity in a molecular classification, as previously suggested24. Our copy number profiling revealed both focal and broad regions of recurrent amplification (Supplementary Table 5). The alteration with the highest prevalence in any subtype was chromosome 12q amplification in dedifferentiated liposarcoma (~90%; Figure 1 and Figure 4A). As amplification is a common mechanism of oncogenic activation, we designed an RNA interference (RNAi) screen to help identify genes in amplified regions that are necessary for cancer cell proliferation in this subtype. We performed knockdown with short hairpin RNAs (shRNA) on 385 genes (Supplementary Table 2) in three dedifferentiated liposarcoma cell lines (LPS141, DDLS8817, and FU-DDLS-1) with copy number profiles similar to those observed in primary tumors of this subtype. A total of 2,007 shRNA lentiviruses, a median of five per gene, were tested for their effects on cell proliferation after 5 days (see Methods). Using a statistical method, RSA (see Methods, Supplementary Note, and ref. 25), we identified 99 genes whose knockdown significantly decreased cell growth in at least one cell line (nominal p 10 days) inhibited proliferation when we assayed two of the three cell lines we screened (see Methods, Figure 4B). Furthermore, pharmacological inhibition of CDK4 in dedifferentiated liposarcoma cells with PD0332991, a selective CDK4/CDK6 inhibitor currently in clinical trials28, induced G1 arrest in the same two cell lines (Figure 4C). For MDM2, another oncogene found in focal 12q amplifications, knockdown did not significantly impair proliferation in our arrayed screen in any of the three cell lines tested. Nevertheless, proliferation was impaired by subsequent knockdown lasting more than a week when we assayed two of those three cell lines (Figure 4D). Interestingly, another gene whose knockdown reduced proliferation of cells in which it was amplified was YEATS4 (GAS41), encoding a putative transcription factor that represses the p53 tumor suppressor network during normal cell proliferation29. YEATS4, frequently co-amplified with MDM2 (Figure 4A), was transcriptionally upregulated both in tumors relative to normal adipose tissue and in tumors with amplification compared to those copy-neutral for the locus (Supplementary Figure 3). Repeat shRNA experiments confirmed the effect of YEATS4 knockdown seen in the arrayed screen (Figure 4E), consistent with the hypothesis that YEATS4 and MDM2 amplification may cooperatively repress the p53 network in dedifferentiated liposarcoma, as recently suggested30. This finding may have consequences for Nutlin-based antagonism of the p53-MDM2 interaction15,31 in dedifferentiated liposarcomas. Our findings lend additional support for YEATS4 serving as a likely key amplified gene in cancer, as recently suggested through a weight-of-evidence classification scheme proposed for identifying such amplified cancer genes32. This dataset provides the most comprehensive database of sarcoma genome alterations to date, revealing genes and signaling pathways not previously associated with this group of diseases. The study results are available as a community resource that might further the biological understanding of sarcomas and, eventually, shed light on additional strategies to improve patient care. Some of our findings already have potential therapeutic implications. For instance, the PIK3CA mutations found in MRC constitute the first report of such mutations in a mesenchymal cancer. These mutations identify a subset of tumors that might respond to treatment with PI3K inhibitors currently in clinical trials33. Our results also provide further rationale for use of CDK4 inhibitors in dedifferentiated liposarcoma and suggest the use of mTOR inhibitors in NF1-deficient sarcomas, since loss of NF1 function appears to cause mTOR pathway activation34. Finally, these data lend support for the clinical evaluation of agents targeting the p53/MDM2 interaction in dedifferentiated liposarcoma. This work argues for the therapeutic importance of genomic alterations in sarcoma and encourages us to pursue next-generation sequencing strategies that will continue to define the landscape of genomic aberrations in these deadly diseases. Methods Methods and any associated references are available in the online version of the paper at http://www.nature.com/naturegenetics/. Supplementary Material 1 2 3 4 5 6 01
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Molecular characterization of inflammatory myofibroblastic tumors with frequent ALK and ROS1 gene fusions and rare novel RET rearrangement.

              Approximately 50% of conventional inflammatory myofibroblastic tumors (IMTs) harbor ALK gene rearrangement and overexpress ALK. Recently, gene fusions involving other kinases have been implicated in the pathogenesis of IMT, including ROS1 and in 1 patient PDGFRB. However, it remains uncertain whether the emerging genotypes correlate with clinicopathologic characteristics of IMT. In this study, we expand the molecular investigation of IMT in a large cohort of different clinical presentations and analyze for potential genotype-phenotype associations. Criteria for inclusion in the study were typical morphology and tissue availability for molecular studies. The lack of ALK immunoreactivity was not an excluding factor. As overlapping gene fusions involving actionable kinases are emerging in both IMT and lung cancer, we set out to evaluate abnormalities in ALK, ROS1, PDGFRB, NTRK1, and RET by fluorescence in situ hybridization. In addition, next-generation paired-end RNA sequencing and FusionSeq algorithm was applied in 4 cases, which identified EML4-ALK fusions in 2 cases. Of the 62 IMTs (25 children and 37 adults), 35 (56%) showed ALK gene rearrangement. Of note, EML4-ALK inversion was noted in 7 (20%) cases, seen mainly in the lung and soft tissue of young children including 2 lesions from newborns. There were 6 (10%) ROS1-rearranged IMTs, all except 1 presenting in children, mainly in the lung and intra-abdominally and showed a distinctive fascicular growth of spindle cells with long cell processes, often positive for ROS1 immunohistochemistry. Two of the cases showed TFG-ROS1 fusions. Interestingly, 1 adult IMT revealed a RET gene rearrangement, a previously unreported finding. Our results show that 42/62 (68%) IMTs are characterized by kinase fusions, offering a rationale for targeted therapeutic strategies. Interestingly, 90% of fusion-negative IMTs were seen in adults, whereas >90% of pediatric IMT showed gene rearrangements. EML4-ALK inversion and ROS1 fusions emerge as common fusion abnormalities in IMT, closely recapitulating the pattern seen in lung cancer.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Oncol
                Front Oncol
                Front. Oncol.
                Frontiers in Oncology
                Frontiers Media S.A.
                2234-943X
                17 August 2022
                2022
                : 12
                : 949962
                Affiliations
                [1] 1 Department of Pathology, West China Hospital, Sichuan University , Chengdu, China
                [2] 2 Department of Pathology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to School of Medicine, University of Electronic Science and Technology of China , Chengdu, China
                Author notes

                Edited by: Ming Zhao, Zhejiang Provincial People’s Hospital, China

                Reviewed by: Joon Hyuk Choi, Yeungnam University, South Korea; Anjia Han, The First Affiliated Hospital of Sun Yat-sen University, China

                *Correspondence: Hongying Zhang, hy_zhang@ 123456scu.edu.cn

                †These authors have contributed equally to this work

                This article was submitted to Surgical Oncology, a section of the journal Frontiers in Oncology

                Article
                10.3389/fonc.2022.949962
                9432863
                36059611
                9a3ac1cb-8776-463f-89bc-8c6445a05701
                Copyright © 2022 Xie, Jing, Zhao, Peng, Chen, Lan, Peng, He, Chen, Zhang and Zhang

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 22 May 2022
                : 27 July 2022
                Page count
                Figures: 6, Tables: 3, Equations: 0, References: 67, Pages: 19, Words: 8504
                Funding
                Funded by: National Natural Science Foundation of China , doi 10.13039/501100001809;
                Categories
                Oncology
                Original Research

                Oncology & Radiotherapy
                liposarcoma,thorax,molecular analysis,well-differentiated liposarcoma,dedifferentiated liposarcoma,myxoid pleomorphic liposarcoma

                Comments

                Comment on this article