Next Article in Journal
Tanshinone IIA Facilitates Efficient Cartilage Regeneration under Inflammatory Factors Caused Stress via Upregulating LncRNA NEAT1_2
Previous Article in Journal
Estimating the Prevalence and Characteristics of Patients Potentially Eligible for Lipoprotein(a)-Lowering Therapies in a Real-World Setting
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genomic Landscape Comparison of Cardiac versus Extra-Cardiac Angiosarcomas

1
Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy
2
Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
3
Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
4
Unit of Cardiology, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
5
Division of Pathology, Cardiovascular and Cardiac Transplant Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
6
Cardiac Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
7
IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
8
Interdepartmental Center Alma Mater Institute on Healthy Planet, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy
9
Division of Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(12), 3290; https://doi.org/10.3390/biomedicines11123290
Submission received: 15 March 2023 / Revised: 29 November 2023 / Accepted: 7 December 2023 / Published: 12 December 2023
(This article belongs to the Section Cancer Biology and Oncology)

Abstract

:
Angiosarcomas (ASs) are rare malignant vascular entities that can affect several regions in our body, including the heart. Cardiac ASs comprise 25–40% of cardiac sarcomas and can cause death within months of diagnosis. Thus, our aim was to identify potential differences and/or similarities between cardiac and extra-cardiac ASs to enhance targeted therapies and, consequently, patients’ prognosis. Whole-transcriptome analysis of three cardiac and eleven extra-cardiac non-cutaneous samples was performed to investigate differential gene expression and mutational events between the two groups. The gene signature of cardiac and extra-cardiac non-cutaneous ASs was also compared to that of cutaneous angiosarcomas (n = 9). H/N/K-RAS and TP53 alterations were more recurrent in extra-cardiac ASs, while POTE-gene family overexpression was peculiar to cardiac ASs. Additionally, in vitro functional analyses showed that POTEH upregulation conferred a growth advantage to recipient cells, partly supporting the cardiac AS aggressive phenotype and patients’ scarce survival rate. These features should be considered when investigating alternative treatments.

1. Introduction

Angiosarcomas (ASs) are malignant soft-tissue tumors characterized by an incidence of 1–2% among all sarcomas and poor prognosis [1,2,3]. This subtype arises from endothelial cells of blood or lymphatic vessels and can affect different areas, including the head, neck, breast, bone, and viscera [3]. The median age at diagnosis ranges between 52 and 67 years old, and the distribution between men and women is analogous [2,4].
Low-grade angiosarcomas present a solid component along with low-grade cytology and vessels with open lumina, while high-grade angiosarcomas display high mitotic rates and atypical cells [5]. Angiosarcomas can also be classified according to their onset. Primary ASs arise de novo, whereas secondary ASs develop following chronic lymphedema or exposure to radiotherapy or vinyl chloride [2,3,6]. Other potential risk factors include UV radiation, arteriovenous fistula, and a compromised immune system [6].
Visceral angiosarcomas comprise cardiac sarcomas, which represent 25–40% of cardiac sarcomas [7]. Cardiac ASs primarily arise in the right atrium and have a higher incidence in men with a 2/3:1 ratio [7,8,9]. Malignant cells can also invade the pericardium and cardiac chambers causing obstruction and, subsequently, heart failure [8]. Additionally, this subgroup is characterized by a high proliferation rate and late diagnosis, since symptoms are aspecific and onset is rare. Thus, patients are usually deceased within months of diagnosis [9]. Surgical resection alone remains the most effective option, especially when a tumor is strictly localized; however, a combination of surgery, chemotherapy, and radiation is crucial to prolong overall survival.
Considering the rarity and the heterogeneity in their possible site of origin, depicting a well-defined AS mutational profile has been challenging. Therefore, in the present study, we evaluated the molecular differences and similarities between cardiac and extra-cardiac angiosarcomas using next-generation sequencing.

2. Materials and Methods

From 2005 to 2017, 11 patients diagnosed with primary cardiac sarcomas were identified in our center. This case series comprised several rare subtypes including tree angiosarcomas (27%) [10,11], two of which were suitable for genomic analysis (L359 and L360) (SRA Accession Number: PRJNA896891).
Case L359: A 74-year-old male affected by a right atrial lesion of 7 cm was diagnosed with cardiac MRI and PET-FDG (SUVmax 43.7). He underwent surgery with explorative and diagnostic intent followed by biopsy. The histology was fused, high-grade angiosarcoma (Figure 1). He began one cycle of chemotherapy with doxorubicin and died of the disease after a few weeks.
Case L360: A 39-year-old male affected by a right atrial lesion of 6 cm was diagnosed with a CT scan, MRI, and PET-FDG (SUVmax 8). He underwent tumor resection, which was not completed due to vena cava infiltration. The histology was angiosarcoma (grade 3) with a high mitotic index (14/10 high-power field—HPF) (Figure 1). He began chemotherapy with taxol and died of the disease.
In addition to these two cases, to evaluate the molecular differences and similarities between cardiac and extra-cardiac angiosarcomas, AS samples from the Gene Expression Omnibus database (https://www.ncbi.nlm.nih.gov/geo/, accessed on 27 September 2022; accession number: GSE163359) were included in this study—one cardiac and twelve extra-cardiac non-cutaneous ASs, whose RNA had been extracted from fresh tissue (Table 1) [12].

2.1. Coding Transcriptome Sequencing

Formalin-fixed paraffin-embedded (FFPE) slides were manually macrodissected by a pathologist to achieve a tumor tissue enrichment of at least 70%. Total RNA was extracted using a RecoverAll Total Nucleic Acid Isolation Kit (Thermo Fisher Scientific, Waltham, MA, USA), and cDNA libraries were synthesized from 100 ng total RNA using a TruSeq RNA Exome kit (Illumina, San Diego, CA, USA) following manufacturer’s instructions. cDNA library synthesis included RNA fragmentation, adapter ligation, amplification, and probe hybridization to select coding exon sequences. Subsequently, libraries were sized with Agilent DNA 7500 chips on the Bioanalyzer 2100 (Agilent Technologies, Taipei, Taiwan) and quantified with a fluorometric assay (Quant-IT Picogreen assay; Life Technologies, Carlsbad, CA, USA). Paired-end libraries were sequenced at 80 bp on a NextSeq500 instrument (Illumina, San Diego, CA, USA).

2.2. Bioinformatic Analysis

First, quality control of all the FASTQ files was performed with FastQC [13], and the results across all samples were summarized using MultiQC [14]. Two samples (L410 and SRR13260960) were excluded from the analysis due to a high number of overrepresented sequences. Reads of the remaining samples were aligned and mapped on the reference human genome hg38 (No Alts, with decoys) followed by RNA quantification on BaseSpace, an Illumina web tool (http://euc1.sh.basespace.illumina.com/, accessed on 7 October 2022). Small variant calling combined with variant annotation with Nirvana was performed.
Regarding gene alterations, filters were applied to vcf output files to select only stop gain/splice donor/splice acceptor/splice region/frameshift indels/inframe deletion/inframe insertion/initiator codon/ATG loss/missense mutations predicted as deleterious or probably damaging with the SIFT predictor, with a GnomAD frequency <0.01, a QC metric quality >29, a total read depth >14, an alt allele depth >3, and a variant read frequency >0.15. Variants were validated using the Integrative Genomic Viewer (IGV—https://igv.org/, accessed on 10 December 2022). Lastly, only genes belonging to Tier 1 of the Cancer Gene Census list (https://cancer.sanger.ac.uk/census, accessed on 10 December 2022) were considered for further analysis. Sequencing data were analyzed using the DRAGEN RNA app (version 3.10.4). Our mutational events were also compared to the ones from the Angiosarcoma Project—Count Me In, with a focus on cutaneous ASs [15]. These additional data (48 samples from 36 patients) were retrieved from the cBioPortal for Cancer Genomics database (https://www.cbioportal.org, accessed on 1 December 2022), including one cardiac angiosarcoma and nine cutaneous ASs of the head, neck, face, and scalp (HNFS). Further analysis was performed using STAR to map paired reads on the reference human genome hg38 [16]; subsequently, duplicate removal, sorting, and indexing were applied using Samtools [17]. Gene expression was quantified and normalized as transcripts per million (TPM) from raw gene counts generated with the Python package HTseq-count [18]. TPM were then used to perform a principal component analysis (PCA) using the R package prcomp [19] and a differential gene expression (DGE) analysis applying the Mann–Whitney U test (p-value < 0.05), following batch correction with the sva package [20]. Batch correction was necessary since, as previously mentioned, our RNA samples had been extracted from paraffin-embedded tissue, while the GEO samples were derived from fresh tissue.
A gene set enrichment analysis (GSEA) was performed using the whole expression matrix and selecting the Hallmarks, Reactome, and KEGG gene sets [21]. Moreover, parameters were set as follows: “number of permutations” = “1000”, “permutation type” = “gene set”, “enrichment statistic” = “weighted”, “metric for ranking gene” = “Signal2Noise”, “min size: exclude smaller sets” = “15”, and “normalization mode” = “meandiv”.

2.3. Transfection of the POTEH Gene Transcript

To investigate the potential role of POTEH overexpression, a plasmid DNA (pcDNA3.1(+)-C-eGFP) containing the whole POTEH transcript under the control of a CMV promoter and upstream of an eGFP-tag sequence was synthesized (GenScript, Rijswijk, NL, USA). The expression plasmid was transformed and amplified in One Shot TOP10 Chemically Competent E. coli cells (Life Technologies, Carlsbad, CA, USA) and then purified with an E.Z.N.A. Endo-Free Plasmid DNA Maxi Kit (Omega Bio-tek, Norcross, GA, USA). HEK293 cells were obtained from CLS Cell Lines Service (Eppelheim, Germany) and cultured in Dulbecco’s Modified Eagle Medium high glucose (DMEM, Gibco) enriched with 10% fetal bovine serum (FBS), 1% L-glutamine, and 1% penicillin–streptomycin. Subsequently, the cells were seeded on 6-well plates (0.5 × 106 cells/well) in DMEM 10% FBS without antibiotics, and after 24 h, they were transiently transfected with POTEH plasmid or with empty pcDNA3.1 vector using Lipofectamine 2000 (Life Technologies, Carlsbad, CA, USA). Mock-transfected (treated only with Lipofectamine) and pcDNA3.1-transfected cells were used as controls. To assess transfection efficiency, GFP positivity was quantified using a FACSCanto flow cytometer (Beckton Dickinson, Franklin Lakes, NJ, USA). To measure the POTEH effect on cell viability, an MTT assay was performed 48 and 72 h after seeding transfected cells in complete medium. Total RNA was extracted 72 h after transfection using the RNeasy Mini Kit (Zymo Research, Irvine, CA, USA), and reverse transcribed with the iScript™ Reverse Transcription Supermix (Bio-Rad, Hercules, CA, USA) into cDNA to quantify POTEH overexpression.

2.4. Real-Time PCR Analysis

POTEH expression was evaluated in transfected HEK293 cells using quantitative RT-PCR on a Roche (LightCycler480, Roche Diagnostics) with a Luna Universal One-Step RT-qPCR kit (New England BioLabs, Ipswich, MA, USA). GAPDH and ATPS were used as housekeeping genes. GraphPad PRISM Software was used for statistical analysis. The p-value was estimated against pcDNA3.1 using an unpaired t-test (** p < 0.01; *** p < 0.001).

3. Results

3.1. Gene Expression Analysis

First and foremost, the expression profile of three cardiac versus eleven extra-cardiac non-cutaneous ASs was analyzed (Table 1). The unsupervised PCA did not show a distinct cluster of extra-cardiac angiosarcomas along any principal component; however, the three cardiac samples were located close to one another (Figure 2). This suggested the presence of, at least, some common features among cardiac cases and confirmed AS heterogeneity.
Additionally, a supervised analysis of cardiac versus extra-cardiac angiosarcomas detected 248 differentially expressed genes (p-value < 0.05). Interestingly, several POTE-family members, including POTEH, POTEG, and POTEM, were upregulated in cardiac ASs (Figure 3). These findings were further validated by the GSEA rank-ordered gene list, where the aforementioned POTE-related genes were identified as specific gene markers of cardiac ASs. Moreover, the epithelial–mesenchymal transition (EMT), myogenesis, gene downregulation in response to UV, and cardiac and striated muscle contraction pathways were upregulated in cardiac samples compared with extra-cardiac ASs (GSEA Hallmarks, Reactome, and KEGG databases) (Figure 4).

3.2. POTEH Overexpression

Due to the significance of POTE-family overexpression in cardiac ASs, we decided to investigate the role of POTE genes and selected POTEH among its family members due to its pronounced upregulation. We transfected HEK293 cells with an expression vector containing the full-length POTEH transcript and an eGFP tag. After 24 h, 49% of POTEH-transfected cells exhibited GFP positivity and RT-PCR effectively demonstrated a significant increase in POTEH mRNA levels in POTEH-transfected cells when compared with pcDNA3.1 controls. Regarding the phenotypic effect, POTEH overexpression significantly increased cell viability, with a 63% and 42% increase with respect to pcDNA3.1-transfected cells at 48 and 72 h, respectively (Figure 5).

3.3. Gene Alteration Analysis

First, comparing mutational signatures between cardiac and extra-cardiac non-cutaneous angiosarcomas led to the identification of the following events in ASs of the heart (Table 2). The RAD51B missense mutation (Lys243Arg) is classified as pathogenic on COSMIC and probably damaging with a score = 1 on PolyPhen. In contrast, it is considered likely benign and benign on ClinVar and Varsome, respectively. Additionally, PSIP1 presented a deletion involving four nucleotides (c.651_654del), which caused a frameshift. The KDR c.2267-1_2267 insertion shifting the reading frame is defined as likely pathogenic on Varsome; while the JAK2 point mutation (c.2840G>A) is probably damaging according to PolyPhen and pathogenic with a score = 0.96 on COSMIC.
Alterations concerning RNF213 and ATRX were present in both cardiac and extra-cardiac ASs (Figure 6) [15]. Regarding RNF213, two point mutations (Ile3318Val and Thr1705Lys) are considered benign on ClinVar and Varsome. According to PolyPhen, the Ile3318Val substitution is also benign, while the other two alterations (Thr1705Lys and Pro2274Leu) are probably damaging with a score of 0.98 and 0.99, respectively. In L359, ATRX presented an in-frame deletion (c.6792_6794del) involving exon 31, where a whole triplet encoding glutamic acid was depleted. The same gene presented a point mutation (c.4675A>T) in SRR13260959, causing stop-codon gain. ATRX nonsense mutations were also found in two cutaneous samples [15].
Including cutaneous AS, the most frequent events concerned TP53 (n = 11). TP53 missense mutations (Arg248Gln, Arg248Trp, Arg280Gly, Tyr220Cys, Pro250Leu, Ser241Phe, and Glu286Lys) are frequently associated with cancer development (Figure 6). For instance, on COSMIC, the Arg248Gln variant is present in 173/3610 mutated samples (4.8%), while Arg248Trp is in 152/3610 (4.2%). The c.920-1G>A splice acceptor variant and the Trp53*, Gln317*, and Gln165* nonsense mutations are classified as pathogenic on ClinVar.
Other remarkable alterations involved the following genes related to tumor onset. NRAS and HRAS point mutations (Gln61Leu) are considered deleterious on SIFT and possibly damaging with a score = 0.86 and 0.58 on PolyPhen, respectively. On COSMIC, Gln61Leu affecting NRAS is observed in 125/1183 (10.6%) mutated samples, while the same variant affecting HRAS is observed in 30/234 (12.8%).
Lastly, the TSC2 missense mutation (Ile885Val) is of uncertain significance according to ClinVar and Varsome, tolerated on SIFT, and possibly damaging with a score = 0.88 on PolyPhen. The KMT2C point mutation (c.5053G>T) is defined as deleterious on SIFT, probably damaging on PolyPhen, and pathogenic with a score = 0.99 on COSMIC, but also as benign on ClinVar. FANCE harbored a missense variant (c.1095A>C) classified as deleterious on SIFT and as possibly damaging on PolyPhen. Regarding the NBN alteration c.758C>T, there are conflicting interpretations: it is considered deleterious on SIFT, but benign and likely benign on PolyPhen and ClinVar, respectively.

4. Discussion

The PCA findings confirmed AS heterogeneity, which partly depends on the tumor site. Indeed, the results also highlighted common features between cardiac and extra-cardiac angiosarcomas.
Among AS-mutated genes, TP53 is well-known for its role as a tumor suppressor; however, the role of its alterations has remained unclear in cardiac angiosarcomas thus far. A study by Garcia and colleagues and the whole-exome sequencing analysis by Zhrebker’s team did not identify any TP53 mutations in cardiac ASs [22,23]. In support of this, TP53 seems to be frequently altered in extra-cardiac ASs, especially in UV-induced head and neck ASs [24,25,26]. In contrast, in a patient affected by cardiac AS, immunohistochemical analyses detected high levels of mutated p53 gene products in tumor cells [22]. Another work showed the presence of TP53 point mutations mostly in extra-cardiac ASs, but also in two cases of cardiac ASs [27]. These results were further supported by the Angiosarcoma Project [15]: TP53 was the most altered gene (29.2%), and mutational events occurred in one cardiac AS and in ten extra-cardiac ASs, five of which were cutaneous.
Combining these five cutaneous samples [15] with our findings, TP53 alterations were identified mainly in extra-cardiac cases (n = 9) and in one cardiac AS. They were missense and truncating variants, which are known for negatively affecting apoptosis, senescence, and DNA repair. Therefore, although our study only included four cardiac ASs, these findings might suggest that TP53 mutations are, in fact, more common in ASs located outside of the heart.
The second most mutated gene was RNF213, which was recently identified as an early-stage lung cancer biomarker [28]. RNF213 alterations might lead to carcinogenesis because of its involvement in angiogenesis, vasculogenesis, inflammation, and proliferation [29]. In our study and the Angiosarcoma Project [15], this gene was altered in two cardiac ASs and three extra-cardiac ASs. Thus, RNF213 involvement in AS onset does not seem to strongly correlate with tumor localization.
Alterations affecting the RAS pathway are also remarkable events in AS onset as they can alter cell division, which greatly contributes to tumor development. These signatures are far more common in extra-cardiac ASs [25,26]; indeed, 5/39 extra-cardiac AS cases presented H/K/N-RAS mutations [24]. NRAS and HRAS were exclusively mutated in three and two extra-cardiac cases, respectively [15], while KRAS point mutations were only detected in two cases of cardiac ASs [22]. Accordingly, our results showed deleterious NRAS and HRAS alterations in two extra-cardiac ASs. NRAS and HRAS are proto-oncogenes involved in cell division; thus, their mutations can lead to uncontrolled proliferation, a feature of cancer cells. Moreover, in the Angiosarcoma Project NRAS was mutated in two breast ASs and one cutaneous AS, while HRAS was mutated in two ASs of the breast [15].
Unsurprisingly, other genes associated with angiogenesis (KDR and PSIP1) were altered in our analysis. KDR, which encodes a vascular endothelial growth factor (VEGF) receptor, was found mutated in 7–10% of soft-tissue ASs, mainly in the breast [7,15,30]. This was not confirmed in our samples since both breast ASs lacked KDR alterations. Conversely, seven out of eight patients presenting KDR mutations were diagnosed with breast AS in the Angiosarcoma Project [15]. Furthermore, this gene was mutated in a few cases of cardiac ASs [23,31], as it occurred in one of our cardiac samples (SRR13260952). KDR and PLCG1, which also contributes to angiogenesis, are mutually exclusive [30]. In this regard, the SRR13260952 case presented solely KDR alterations. Mutations in TP53 and KDR are also mutually exclusive [15]; indeed, as previously mentioned, TP53 alterations only concerned extra-cardiac AS. Both exclusivities were further confirmed in the Angiosarcoma Project [15].
When altered, PSIP1 can enhance angiogenesis and prevent apoptosis in cancer cells [32]. Additionally, PSIP1 contributes to homology-directed repair (HDR) [33]. In our study, it is not the only altered gene involved in DNA repair: RAD51B participates in homologous recombination repair (HRR) of double-strand DNA breaks [34,35]; FANCE is a member of the Fanconi anemia complementation group (FANC) and plays a role in DNA cross-links repair [36]; NBN is an MRE11-RAD50-NBN (MRN) component involved in DNA recombination, telomere maintenance, cell division regulation and double-strand DNA break repair [35,36]; and lastly, KMT2C is a histone methyltransferase that marks sites for transcription and DNA repair [37]. Genes involved in DNA repair were altered in many extra-cardiac ASs and in one cardiac AS.
Alterations in ATRX, JAK2, and TSC2 are also noteworthy since these genes contribute to cell growth and proliferation. ATRX is involved in chromatin remodeling, transcription, and telomere maintenance (ALT pathway) [36,38]. In our analysis it was mutated in one cardiac (L359) and in one extra-cardiac case (SRR13260959), while it presented nonsense mutations in two cutaneous ASs. This suggests that ATRX alterations might not be subtype-specific biomarkers. JAK2 plays a major role in growth factor signaling and histone modifications [39,40], while TSC2 is a tumor suppressor that inhibits cell growth by downregulating the mTORC1 pathway [41]. Consequently, when altered, these genes might cause uncontrolled cell proliferation.
To better distinguish cardiac from extra-cardiac angiosarcomas, we conducted multiple gene expression analyses, whose results will now be explored.
The POTE-gene family comprises 14 homologous genes, including cancer-testis antigens (CTAs), located on several chromosomes. These genes are usually upregulated in prostate and epithelial ovarian cancer (EOC), including high-grade serous carcinoma (HGSC) [42,43,44] but also in breast and lung cancer [45]. Their overexpression in ovarian cancer is associated with more advanced stages and poor prognosis [44]. Therefore, POTE-related genes might be therapeutically targeted. To validate these observations, we focused on POTEH, the most upregulated POTE family member in cardiac ASs. This gene is located on chromosome 22 and is expressed in normal prostate, ovarian, and testis tissues and also prostate, ovarian (e.g., HGSC), and lung cancer [43,44,46]. In our research, we transfected mammalian cells with a plasmid containing its transcript sequence and, as a result, successfully transfected cells presented greater growth levels compared with pcDNA3.1-transfected samples. Since POTEH-overexpressing cells can be considered more proliferative and resistant over time, gene upregulation in cardiac angiosarcomas highlights a more aggressive phenotype and could at least partly explain patients’ scarce survival rate.
Furthermore, in cardiac sarcomas, we observed an overexpression of genes involved in the epithelial–mesenchymal transition, which is in line with their threatening histotype. In EMT, endothelial cells tend to detach from surrounding cells and, eventually, acquire the phenotype of mesenchymal stem cells, including the ability to migrate and colonize other tissues.
Two common events in angiosarcomas were not detected in our samples. POT1 is usually mutated in ASs, especially in those located in the head and neck [15,26,47,48]. As proof, it was mutated in three cutaneous cases from the Angiosarcoma Project [15]. Since it is involved in telomere maintenance and apoptosis, POT1 alterations can lead to longer telomeres and, subsequently, to cell immortality. Mutated POT1 can enhance ATR-dependent DNA damage signaling, which induces cell cycle arrest [48]. Subsequently, cell senescence can cause progenitor cell depletion and tissue stress. Therefore, cells tend to bypass apoptosis and acquire additional somatic mutations, which characterize tumor-aggressive phenotypes.
Another typical event in ASs is MYC amplification, especially in secondary angiosarcomas following breast cancer [3,49]. MYC is relevant to cancer development because of its involvement in oncogenic signaling. Unfortunately, the whole transcriptome analysis applied in this study did not allow us to verify this event.
Until now, the medical treatment of advanced or unresectable ASs has been independent of tumor localization and comprises the administration of anthracyclines and/or gemcitabine/Taxotere, as stated by consensus conference guidelines [50]. Although our molecular findings did not suggest site-specific variations in the chemotherapy regimen, emerging data regarding cutaneous AS immune sensitivity are intriguing [15,51,52]. Indeed, ipilimumab–nivolumab combined therapy has demonstrated a durable response in ASs, especially in cutaneous tumors of the scalp or face [51]. Additionally, cutaneous angiosarcomas of the head, neck, face, and scalp appeared to be characterized by high tumor mutational burden (TMB) and signatures corresponding to UV light exposure [15,52]. These features suggest that UV damages might greatly contribute to HNFS-AS development. Cutaneous samples from the Angiosarcoma Project presented a greater number of mutational events compared with cardiac and extra-cardiac non-cutaneous ASs, which can be correlated with abundant immune infiltrate [15]. Thus, this AS subtype might be more sensitive to checkpoint inhibitors.
Ultimately, mutational events were more homogeneous in HNFS angiosarcomas than in cardiac ASs, even though both areas are localized. Among the cutaneous samples, the proto-oncogene RET, FGFR2, and NOTCH2 involved in cell growth, proliferation, and apoptosis, and POT1 presented elevated mutational rates, highlighting their role as potential therapeutic targets. Consequently, these differences between cutaneous and non-cutaneous angiosarcomas, and also between cardiac and extra-cardiac ASs, should be carefully considered during treatment selection, although high TMB and UV signature do not ensure promising response to immunotherapy [52].

5. Conclusions

Our molecular results supported AS heterogeneity but also highlighted some distinct features between cardiac and extra-cardiac ASs. TP53 and H/K/NRAS mutations seemed to be predominant causative factors in extra-cardiac AS development. Conversely, POTE alterations were signatures of cardiac ASs. Since our in vitro results showed that POTEH upregulation enhances cell viability, these biomarkers might be relevant to define and, eventually, to predict AS onset in patients.
However, considering the limited sample size as a drawback in our study and more broadly in rare tumor studies, future attempts should be directed toward analyzing more samples to further confirm our findings and potentially identify new alterations. Whole-exome-sequencing analyses should also be performed to investigate gene amplifications and deletions, which might indicate other biomarkers.
From a therapeutic perspective, our results highlighted a higher mutational burden in cutaneous angiosarcomas compared with non-cutaneous ASs. Moreover, since POTE genes are cancer-testis antigens, their overexpression in cardiac ASs might suggest their amenability to immunotherapy. This might pave the way to explore alternative targets and treatments in non-cutaneous ASs, especially in angiosarcomas of the heart.

Author Contributions

Conceptualization, L.G., M.N., M.U., M.A.P. and A.A.; methodology, L.G., M.N., M.U., M.A.P. and A.A.; validation, L.G. and M.U.; formal analysis, L.G. and M.N.; investigation, C.P., F.A., A.F., D.P. and G.F.; resources, O.L., B.C. and C.B.; data curation, L.G., M.N. and M.U.; writing—original draft preparation, L.G., M.N. and A.A.; writing—review and editing, L.G., A.C., T.P., M.C.N., G.P., M.A.P. and A.A.; supervision, M.N., M.A.P. and A.A.; project administration, M.A.P. and A.A. All authors have read and agreed to the published version of this manuscript.

Funding

The work reported in this publication was supported by the Italian Ministry of Health, RC-2022-2773439.

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki and was approved by the Institutional Ethics Committee of Policlinico Sant’Orsola-Malpighi, Bologna, Italy (approval number: 95/2013/U/Tess; date: 8 October 2013).

Informed Consent Statement

Informed consent was obtained from all living subjects involved in the study.

Data Availability Statement

Publicly available datasets were analyzed in this study. Our data can be found at https://www.ncbi.nlm.nih.gov/sra/PRJNA896891, accessed on 27 September 2022. Data from the Gene Expression Omnibus database can be retrieved from https://www.ncbi.nlm.nih.gov/geo/, accessed on 27 September 2022—accession number: GSE163359, and data from the Angiosarcoma Project—Count me in can be found in the cBioPortal for Cancer Genomics database (https://www.cbioportal.org, accessed on 1 December 2022).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fayette, J.; Martin, E.; Piperno-Neumann, S.; le Cesne, A.; Robert, C.; Bonvalot, S.; Ranchère, D.; Pouillart, P.; Coindre, J.M.; Blay, J.Y. Angiosarcomas, a Heterogeneous Group of Sarcomas with Specific Behavior Depending on Primary Site: A Retrospective Study of 161 Cases. Ann. Oncol. 2007, 18, 2030–2036. [Google Scholar] [CrossRef] [PubMed]
  2. Weidema, M.E.; Versleijen-Jonkers, Y.M.H.; Flucke, U.E.; Desar, I.M.E.; van der Graaf, W.T.A. Targeting Angiosarcomas of the Soft Tissues: A Challenging Effort in a Heterogeneous and Rare Disease. Crit. Rev. Oncol. Hematol. 2019, 138, 120–131. [Google Scholar] [CrossRef] [PubMed]
  3. Chan, J.Y.; Lim, J.Q.; Yeong, J.; Ravi, V.; Guan, P.; Boot, A.; Tay, T.K.Y.; Selvarajan, S.; Md Nasir, N.D.; Loh, J.H.; et al. Multiomic Analysis and Immunoprofiling Reveal Distinct Subtypes of Human Angiosarcoma. J. Clin. Investig. 2020, 130, 5833–5846. [Google Scholar] [CrossRef] [PubMed]
  4. Young, R.J.; Robin, A.; Young, J.; Brown, N.J.; Reed, M.W.; Hughes, D.; Woll, P.J. Angiosarcoma. Lancet Oncol. 2010, 11, 983–991. [Google Scholar] [CrossRef] [PubMed]
  5. Gaballah, A.H.; Jensen, C.T.; Palmquist, S.; Pickhardt, P.J.; Duran, A.; Broering, G.; Elsayes, K.M. Angiosarcoma: Clinical and Imaging Features from Head to Toe. Br. J. Radiol. 2017, 90, 20170039. [Google Scholar] [CrossRef]
  6. Sturm, E.C.; Marasco, I.S.; Katz, S.C. Multidisciplinary Management of Angiosarcoma—A Review. J. Surg. Res. 2021, 257, 213–220. [Google Scholar] [CrossRef] [PubMed]
  7. Urbini, M.; Astolfi, A.; Indio, V.; Nannini, M.; Pizzi, C.; Paolisso, P.; Tarantino, G.; Pantaleo, M.A.; Saponara, M. Genetic Aberrations and Molecular Biology of Cardiac Sarcoma. Ther. Adv. Med. Oncol. 2020, 12, 1758835920918492. [Google Scholar] [CrossRef] [PubMed]
  8. Scicchitano, P.; Chiara Sergi, M.; Cameli, M.; Miglioranza, M.H.; Ciccone, M.M.; Gentile, M.; Porta, C.; Tucci, M. Primary Soft Tissue Sarcoma of the Heart: An Emerging Chapter in Cardio-Oncology. Biomedicines 2021, 9, 774. [Google Scholar] [CrossRef]
  9. Patel, S.D.; Peterson, A.; Bartczak, A.; Lee, S.; Chojnowski, S.; Gajewski, P.; Loukas, M. Primary Cardiac Angiosarcoma—A Review. Med. Sci. Monit. 2014, 20, 103–109. [Google Scholar] [CrossRef]
  10. Gozzellino, L.; Nannini, M.; Pizzi, C.; Leone, O.; Corti, B.; Indio, V.; Baldovini, C.; Paolisso, P.; Foà, A.; Pacini, D.; et al. Genomic Characterization of Rare Primary Cardiac Sarcoma Entities. Diagnostics 2023, 13, 214. [Google Scholar] [CrossRef]
  11. Saponara, M.; Indio, V.; Pizzi, C.; Serban, E.D.; Urbini, M.; Astolfi, A.; Paolisso, P.; Suarez, S.M.; Nannini, M.; Pacini, D.; et al. Successful Multidisciplinary Clinical Approach and Molecular Characterization by Whole Transcriptome Sequencing of a Cardiac Myxofibrosarcoma: A Case Report. World J. Clin. Cases 2019, 7, 3018–3026. [Google Scholar] [CrossRef] [PubMed]
  12. Kim, J.H.; Megquier, K.; Thomas, R.; Sarver, A.L.; Song, J.M.; Kim, Y.T.; Cheng, N.; Schulte, A.J.; Linden, M.A.; Murugan, P.; et al. Genomically Complex Human Angiosarcoma and Canine Hemangiosarcoma Establish Convergent Angiogenic Transcriptional Programs Driven by Novel Gene Fusions. Mol. Cancer Res. 2021, 19, 847–861. [Google Scholar] [CrossRef] [PubMed]
  13. Andrews, S. FastQC: A Quality Control Tool for High Throughput Sequence Data. 2010. Available online: http://www.bioinformatics.babraham.ac.uk/projects/fastqc (accessed on 7 October 2022).
  14. Ewels, P.; Magnusson, M.; Lundin, S.; Käller, M. MultiQC: Summarize Analysis Results for Multiple Tools and Samples in a Single Report. Bioinformatics 2016, 32, 3047–3048. [Google Scholar] [CrossRef] [PubMed]
  15. Painter, C.A.; Jain, E.; Tomson, B.N.; Dunphy, M.; Stoddard, R.E.; Thomas, B.S.; Damon, A.L.; Shah, S.; Kim, D.; Gómez Tejeda Zañudo, J.; et al. The Angiosarcoma Project: Enabling Genomic and Clinical Discoveries in a Rare Cancer through Patient-Partnered Research. Nat. Med. 2020, 26, 181–187. [Google Scholar] [CrossRef] [PubMed]
  16. Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast Universal RNA-Seq Aligner. Bioinformatics 2013, 29, 15–21. [Google Scholar] [CrossRef] [PubMed]
  17. Danecek, P.; Bonfield, J.K.; Liddle, J.; Marshall, J.; Ohan, V.; Pollard, M.O.; Whitwham, A.; Keane, T.; McCarthy, S.A.; Davies, R.M.; et al. Twelve Years of SAMtools and BCFtools. Gigascience 2021, 10, giab008. [Google Scholar] [CrossRef] [PubMed]
  18. Putri, G.H.; Anders, S.; Pyl, P.T.; Pimanda, J.E.; Zanini, F. Analysing High-Throughput Sequencing Data in Python with HTSeq 2.0. Bioinformatics 2022, 38, 2943–2945. [Google Scholar] [CrossRef]
  19. Sigg, C.D.; Buhmann, J.M. Expectation-Maximization for Sparse and Non-Negative PCA. In Proceedings of the Twenty-Fifth International Conference on Machine Learning, Helsinki, Finland, 5–9 June 2008. [Google Scholar] [CrossRef]
  20. Leek, J.T.; Johnson, W.E.; Parker, H.S.; Fertig, E.J.; Jaffe, A.E.; Zhang, Y.; Storey, J.D.; Torres, L.C. Package “sva”: Surrogate Variable Analysis; Bioconductor: Boston, MA, USA, 2022. [Google Scholar]
  21. Subramanian, A.; Tamayo, P.; Mootha, V.K.; Mukherjee, S.; Ebert, B.L.; Gillette, M.A.; Paulovich, A.; Pomeroy, S.L.; Golub, T.R.; Lander, E.S.; et al. Gene Set Enrichment Analysis: A Knowledge-Based Approach for Interpreting Genome-Wide Expression Profiles. Proc. Natl. Acad. Sci. USA 2005, 2, 15545–15550. [Google Scholar] [CrossRef]
  22. Garcia, J.M.; Gonzalez, R.; Silva, J.M.; Dominguez, G.; Vegazo, I.S.; Gamallo, C.; Provencio, M.; España, P.; Bonilla, F. Mutational Status of K-Ras and TP53 Genes in Primary Sarcomas of the Heart. Br. J. Cancer 2000, 82, 1183–1185. [Google Scholar] [CrossRef]
  23. Zhrebker, L.; Cherni, I.; Gross, L.M.; Hinshelwood, M.M.; Reese, M.; Aldrich, J.; Guileyardo, J.M.; Roberts, W.C.; Craig, D.; von Hoff, D.D.; et al. Case Report: Whole Exome Sequencing of Primary Cardiac Angiosarcoma Highlights Potential for Targeted Therapies. BMC Cancer 2017, 17, 17. [Google Scholar] [CrossRef]
  24. Behjati, S.; Tarpey, P.S.; Sheldon, H.; Martincorena, I.; van Loo, P.; Gundem, G.; Wedge, D.C.; Ramakrishna, M.; Cooke, S.L.; Pillay, N.; et al. Recurrent PTPRB and PLCG1 Mutations in Angiosarcoma. Nat. Genet. 2014, 46, 376–379. [Google Scholar] [CrossRef] [PubMed]
  25. Murali, R.; Chandramohan, R.; Möller, I.; Scholz, S.L.; Berger, M.; Huberman, K.; Viale, A.; Socci, N.D.; Bouvier, N.; Bauer, S.; et al. Targeted Massively Parallel Sequencing of Angiosarcomas Reveals Frequent Activation of the Mitogen Activated Protein Kinase Pathway. Oncotarget 2015, 6, 36041–36052. [Google Scholar] [CrossRef] [PubMed]
  26. Espejo-Freire, A.P.; Elliott, A.; Rosenberg, A.; Costa, P.A.; Barreto-Coelho, P.; Jonczak, E.; D’amato, G.; Subhawong, T.; Arshad, J.; Diaz-Perez, J.A.; et al. Genomic Landscape of Angiosarcoma: A Targeted and Immunotherapy Biomarker Analysis. Cancers 2021, 13, 4816. [Google Scholar] [CrossRef] [PubMed]
  27. Naka, N.; Tomita, Y.; Nakamshi, H.; Araki, N.; Hongyo, T.; Ochi, T.; Aozasa, K. Mutations of P53 Tumor-Suppressor Gene in Angiosarcoma. Int. J. Cancer 1997, 71, 952–955. [Google Scholar] [CrossRef]
  28. Jiang, N.; Zhou, J.; Zhang, W.; Li, P.; Liu, Y.; Shi, H.; Zhang, C.; Wang, Y.; Zhou, C.; Peng, C.; et al. RNF213 Gene Mutation in Circulating Tumor DNA Detected by Targeted Next-Generation Sequencing in the Assisted Discrimination of Early-Stage Lung Cancer from Pulmonary Nodules. Thorac. Cancer 2021, 12, 181–193. [Google Scholar] [CrossRef] [PubMed]
  29. Pollaci, G.; Gorla, G.; Potenza, A.; Carrozzini, T.; Canavero, I.; Bersano, A.; Gatti, L. Novel Multifaceted Roles for RNF213 Protein. Int. J. Mol. Sci. 2022, 23, 4492. [Google Scholar] [CrossRef] [PubMed]
  30. Huang, S.C.; Zhang, L.; Sung, Y.S.; Chen, C.L.; Kao, Y.C.; Agaram, N.P.; Singer, S.; Tap, W.D.; D’Angelo, S.; Antonescu, C.R. Recurrent CIC Gene Abnormalities in Angiosarcomas: A Molecular Study of 120 Cases with Concurrent Investigation of PLCG1, KDR, MYC, and FLT4 Gene Alterations. Am. J. Surg. Pathol. 2016, 40, 645–655. [Google Scholar] [CrossRef]
  31. Kunze, K.; Spieker, T.; Gamerdinger, U.; Nau, K.; Berger, J.; Dreyer, T.; Sindermann, J.R.; Hoffmeier, A.; Gattenlöhner, S.; Bräuninger, A. A Recurrent Activating PLCG1 Mutation in Cardiac Angiosarcomas Increases Apoptosis Resistance and Invasiveness of Endothelial Cells. Cancer Res. 2014, 74, 6173–6183. [Google Scholar] [CrossRef]
  32. Singh, D.K.; Gholamalamdari, O.; Jadaliha, M.; Li, X.L.; Lin, Y.C.; Zhang, Y.; Guang, S.; Hashemikhabir, S.; Tiwari, S.; Zhu, Y.J.; et al. PSIP1/P75 Promotes Tumorigenicity in Breast Cancer Cells by Promoting the Transcription of Cell Cycle Genes. Carcinogenesis 2017, 38, 966–975. [Google Scholar] [CrossRef]
  33. Liedtke, V.; Schröder, C.; Roggenbuck, D.; Weiss, R.; Stohwasser, R.; Schierack, P.; Rödiger, S.; Schenk, L. LEDGF/P75 Is Required for an Efficient Dna Damage Response. Int. J. Mol. Sci. 2021, 22, 5866. [Google Scholar] [CrossRef]
  34. Setton, J.; Selenica, P.; Mukherjee, S.; Shah, R.; Pecorari, I.; McMillan, B.; Pei, I.X.; Kemel, Y.; Ceyhan-Birsoy, O.; Sheehan, M.; et al. Germline RAD51B Variants Confer Susceptibility to Breast and Ovarian Cancers Deficient in Homologous Recombination. NPJ Breast Cancer 2021, 7, 135. [Google Scholar] [CrossRef] [PubMed]
  35. Westphalen, C.B.; Fine, A.D.; André, F.; Ganesan, S.; Heinemann, V.; Rouleau, E.; Turnbull, C.; Palacios, L.G.; Lopez, J.A.; Sokol, E.S.; et al. Pan-Cancer Analysis of Homologous Recombination Repair-Associated Gene Alterations and Genome-Wide Loss-of-Heterozygosity Score. Clin. Cancer Res. 2022, 28, 1412–1421. [Google Scholar] [CrossRef] [PubMed]
  36. Heeke, A.L.; Pishvaian, M.J.; Lynce, F.; Xiu, J.; Brody, J.R.; Chen, W.J.; Baker, T.M.; Marshall, J.L.; Isaacs, C. Prevalence of Homologous Recombination-Related Gene Mutations Across Multiple Cancer Types. JCO Precis. Oncol. 2018, 2, 1–13. [Google Scholar] [CrossRef] [PubMed]
  37. Chang, A.; Liu, L.; Ashby, J.M.; Wu, D.; Chen, Y.; O’Neill, S.S.; Huang, S.; Wang, J.; Wang, G.; Cheng, D.; et al. Recruitment of KMT2C/MLL3 to DNA Damage Sites Mediates DNA Damage Responses and Regulates PARP Inhibitor Sensitivity in Cancer. Cancer Res. 2021, 81, 3358–3373. [Google Scholar] [CrossRef] [PubMed]
  38. Valenzuela, M.; Amato, R.; Sgura, A.; Antoccia, A.; Berardinelli, F. The Multiple Facets of Atrx Protein. Cancers 2021, 13, 2211. [Google Scholar] [CrossRef] [PubMed]
  39. Perner, F.; Perner, C.; Ernst, T.; Heidel, F.H. Roles of JAK2 in Aging, Inflammation, Hematopoiesis and Malignant Transformation. Cells 2019, 8, 854. [Google Scholar] [CrossRef]
  40. Ernst, P.; Schnöder, T.M.; Huber, N.; Perner, F.; Jayavelu, A.K.; Eifert, T.; Hsu, C.J.; Tubío-Santamaría, N.; Crodel, C.C.; Ungelenk, M.; et al. Histone Demethylase KDM4C Is a Functional Dependency in JAK2-Mutated Neoplasms. Leukemia 2022, 36, 1843–1849. [Google Scholar] [CrossRef]
  41. Inoki, K.; Zhu, T.; Guan, K.-L. TSC2 Mediates Cellular Energy Response to Control Cell Growth and Survival. Cell 2003, 115, 577–590. [Google Scholar] [CrossRef]
  42. Bera, T.K.; Zimonjic, D.B.; Popescu, N.C.; Sathyanarayana, B.K.; Kumar, V.; Lee, B.; Pastan, I. POTE, a Highly Homologous Gene Family Located on Numerous Chromosomes and Expressed in Prostate, Ovary, Testis, Placenta, and Prostate Cancer. Proc. Natl. Acad. Sci. USA 2002, 99, 16975–16980. [Google Scholar] [CrossRef]
  43. Sharma, A.; Albahrani, M.; Zhang, W.; Kufel, C.N.; James, S.R.; Odunsi, K.; Klinkebiel, D.; Karpf, A.R. Epigenetic Activation of POTE Genes in Ovarian Cancer. Epigenetics 2019, 14, 185–197. [Google Scholar] [CrossRef]
  44. Barger, C.J.; Zhang, W.; Sharma, A.; Chee, L.; James, S.R.; Kufel, C.N.; Miller, A.; Meza, J.; Drapkin, R.; Odunsi, K.; et al. Expression of the POTE Gene Family in Human Ovarian Cancer. Sci. Rep. 2018, 8, 17136. [Google Scholar] [CrossRef] [PubMed]
  45. Maggiolini, F.A.M.; Mercuri, L.; Antonacci, F.; Anaclerio, F.; Calabrese, F.M.; Lorusso, N.; L’abbate, A.; Sorensen, M.; Giannuzzi, G.; Eichler, E.E.; et al. Evolutionary Dynamics of the POTE Gene Family in Human and Nonhuman Primates. Genes 2020, 11, 213. [Google Scholar] [CrossRef] [PubMed]
  46. Bera, T.K.; Fleur, A.S.; Lee, Y.; Kydd, A.; Hahn, Y.; Popescu, N.C.; Zimonjic, D.B.; Lee, B.; Pastan, I. POTE Paralogs Are Induced and Differentially Expressed in Many Cancers. Cancer Res. 2006, 66, 52–56. [Google Scholar] [CrossRef] [PubMed]
  47. Calvete, O.; Garcia-Pavia, P.; Domínguez, F.; Bougeard, G.; Kunze, K.; Braeuninger, A.; Teule, A.; Lasa, A.; Ramón, Y.; Cajal, T.; et al. The Wide Spectrum of POT1 Gene Variants Correlates with Multiple Cancer Types. Eur. J. Hum. Genet. 2017, 25, 1278–1281. [Google Scholar] [CrossRef] [PubMed]
  48. Calvete, O.; Garcia-Pavia, P.; Domınguez, F.; Mosteiro, L.; Perez-Cabornero, L.; Cantalapiedra, D.; Zorio, E.; Ramon, Y.; Cajal, T.; Crespo-Leiro, M.G.; et al. Pot1 and Damage Response Malfunction Trigger Acquisition of Somatic Activating Mutations in the Vegf Pathway in Cardiac Angiosarcomas. J. Am. Heart Assoc. 2019, 8, e012875. [Google Scholar] [CrossRef] [PubMed]
  49. Motaparthi, K.; Lauer, S.R.; Patel, R.M.; Vidal, C.I.; Linos, K. MYC Gene Amplification by Fluorescence in Situ Hybridization and MYC Protein Expression by Immunohistochemistry in the Diagnosis of Cutaneous Angiosarcoma: Systematic Review and Appropriate Use Criteria. J. Cutan. Pathol. 2021, 48, 578–586. [Google Scholar] [CrossRef] [PubMed]
  50. Gronchi, A.; Miah, A.B.; Dei Tos, A.P.; Abecassis, N.; Bajpai, J.; Bauer, S.; Biagini, R.; Bielack, S.; Blay, J.Y.; Bolle, S.; et al. Soft Tissue and Visceral Sarcomas: ESMO–EURACAN–GENTURIS Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann. Oncol. 2021, 32, 1348–1365. [Google Scholar] [CrossRef]
  51. Wagner, M.J.; Othus, M.; Patel, S.P.; Ryan, C.; Sangal, A.; Powers, B.; Budd, G.T.; Victor, A.I.; Hsueh, C.T.; Chugh, R.; et al. Multicenter Phase II Trial (SWOG S1609, Cohort 51) of Ipilimumab and Nivolumab in Metastatic or Unresectable Angiosarcoma: A Substudy of Dual Anti-CTLA-4 and Anti-PD-1 Blockade in Rare Tumors (DART). J. Immunother. Cancer 2021, 9, e002990. [Google Scholar] [CrossRef]
  52. Rosenbaum, E.; Antonescu, C.R.; Smith, S.; Bradic, M.; Kashani, D.; Richards, A.L.; Donoghue, M.; Kelly, C.M.; Nacev, B.; Chan, J.E.; et al. Clinical, Genomic, and Transcriptomic Correlates of Response to Immune Checkpoint Blockade-Based Therapy in a Cohort of Patients with Angiosarcoma Treated at a Single Center. J. Immunother. Cancer 2022, 10, e004149. [Google Scholar] [CrossRef]
Figure 1. Histological findings representing the L359 and L360 samples. (A) L359: the sheet-like proliferation of round to spindle cells associated with focal necrosis and collagen stroma (hematoxylin–eosin, 200×); (B) L360: epithelioid pattern with high cell proliferation prevalently made up of epithelioid cells with marked nuclear pleomorphism and intracytoplasmic lumina (hematoxylin–eosin, 200×); and (C) L360: immunohistochemistry of neoplastic cells diffusely positive for endothelial marker CD34 (200×).
Figure 1. Histological findings representing the L359 and L360 samples. (A) L359: the sheet-like proliferation of round to spindle cells associated with focal necrosis and collagen stroma (hematoxylin–eosin, 200×); (B) L360: epithelioid pattern with high cell proliferation prevalently made up of epithelioid cells with marked nuclear pleomorphism and intracytoplasmic lumina (hematoxylin–eosin, 200×); and (C) L360: immunohistochemistry of neoplastic cells diffusely positive for endothelial marker CD34 (200×).
Biomedicines 11 03290 g001
Figure 2. Unsupervised principal component analysis (PCA) on cardiac angiosarcomas (n = 3) versus extra-cardiac non-cutaneous angiosarcomas (n = 11), showing a close localization of cardiac ASs.
Figure 2. Unsupervised principal component analysis (PCA) on cardiac angiosarcomas (n = 3) versus extra-cardiac non-cutaneous angiosarcomas (n = 11), showing a close localization of cardiac ASs.
Biomedicines 11 03290 g002
Figure 3. Hierarchical clustering of the most overexpressed gene markers in cardiac (n = 3) versus extra-cardiac non-cutaneous (n = 11) angiosarcomas.
Figure 3. Hierarchical clustering of the most overexpressed gene markers in cardiac (n = 3) versus extra-cardiac non-cutaneous (n = 11) angiosarcomas.
Biomedicines 11 03290 g003
Figure 4. GSEA analysis of enriched pathways in cardiac versus extra-cardiac non-cutaneous angiosarcomas applying the following gene sets: Hallmarks, Reactome, and KEGG. ES: Enrichment Score; NES: Normalized Enrichment Scores; FDR q-val: False Discovery Rate q-value.
Figure 4. GSEA analysis of enriched pathways in cardiac versus extra-cardiac non-cutaneous angiosarcomas applying the following gene sets: Hallmarks, Reactome, and KEGG. ES: Enrichment Score; NES: Normalized Enrichment Scores; FDR q-val: False Discovery Rate q-value.
Biomedicines 11 03290 g004
Figure 5. (A) GFP expression levels in POTEH-transfected cells (red) versus mock- (green) and pcDNA3.1-transfected (blue) cells. (B) Higher POTEH mRNA expression levels in the POTEH-transfected cells compared with pcDNA3.1-transfected cells (RT-PCR). (C) MTT assay results showing cell viability in pcDNA3.1- and POTEH-transfected cells 48 and 72 h after seeding transfected cells in complete medium (** p < 0.01; *** p < 0.001).
Figure 5. (A) GFP expression levels in POTEH-transfected cells (red) versus mock- (green) and pcDNA3.1-transfected (blue) cells. (B) Higher POTEH mRNA expression levels in the POTEH-transfected cells compared with pcDNA3.1-transfected cells (RT-PCR). (C) MTT assay results showing cell viability in pcDNA3.1- and POTEH-transfected cells 48 and 72 h after seeding transfected cells in complete medium (** p < 0.01; *** p < 0.001).
Biomedicines 11 03290 g005
Figure 6. Mutational events classified according to their consequences and occurring in at least 3 samples among cardiac (n = 4), visceral (n = 11), and cutaneous (n = 9) angiosarcomas.
Figure 6. Mutational events classified according to their consequences and occurring in at least 3 samples among cardiac (n = 4), visceral (n = 11), and cutaneous (n = 9) angiosarcomas.
Biomedicines 11 03290 g006
Table 1. Sample features.
Table 1. Sample features.
SampleSexAgeHistotypeLocalization
L359male74angiosarcomaheart
L360male39angiosarcomaheart
SRR13260949female69angiosarcomaomentum
SRR13260950male45angiosarcomalung
SRR13260951female39angiosarcomaperihepatic tissue
SRR13260952male62angiosarcomaheart
SRR13260953female64angiosarcomabreast
SRR13260954unknownunknownangiosarcomaadrenal gland
SRR13260955female40angiosarcomaovary
SRR13260956female73angiosarcomaleft chest wall
SRR13260957male47angiosarcomalung
SRR13260958female65angiosarcomabreast
SRR13260959male85angiosarcomaliver
SRR13260961male73angiosarcomasoft tissue
Table 2. Mutational profile of cardiac (n = 3) and extra-cardiac non-cutaneous (n = 11) angiosarcomas.
Table 2. Mutational profile of cardiac (n = 3) and extra-cardiac non-cutaneous (n = 11) angiosarcomas.
SampleGeneChrcDNAProteinTypeCOSMICClinVarVarsomeGnom-
AD Freq.1
Alt
Allele Depth
Total Read Depth
L359RAD51B14c.728A>Gp.(Lys243Arg)missensepathogeniclik. Benign 2benign0.006791119
L359RNF21317c.9952A>Gp.(Ile3318Val)missenseNA 3benignbenign0.001981421
L359PSIP19c.651_654delp.(Ser217Argfs
Ter47)
frameshiftNA 3NA 3NA 3NA 34364
L359ATRX23c.6792_6794delp.(Glu2265del)inframeNA 3NA 3NA 3NA 3142168
SRR13260952KDR4c.2267-1_2267insTTTACATGTTp.(Gly756Valfs
Ter38)
frameshiftNA 3NA 3lik. Pathog. 4NA 3178213
SRR13260952JAK29c.2840G>Ap.(Arg947Gln)missensepathogenicNA 3uncertain0.0000194076
SRR13260950KMT2C7c.10763C>Tp.(Ser3588Leu)missensepathogenicbenignuncertain0.002811529
SRR13260950TSC216c.2653A>Gp.(Ile885Val)missenseNA 3uncertainuncertainNA 35696
SRR13260951TP5317c.920-1G>ANA 3splice
acceptor
NA 3pathogenicuncertainNA 33234
SRR13260953HRAS11c.182A>Tp.(Gln61Leu)missensepathogenicuncertainuncertainNA 3135332
SRR13260955TP5317c.743G>Ap.(Arg248Gln)missensepathogenicpathogenicpathogenic0.00001968120
SRR13260955RNF21317c.5114C>Ap.(Thr1705Lys)missenseNA 3benignbenign0.0045101131
SRR13260955NBN8c.758C>Tp.(Thr253Ile)missenseNA 3benignbenign0.00009271130
SRR13260956RNF21317c.6821C>Tp.(Pro2274Leu)missenseNA 3NA 3benign0.0000193463
SRR13260956NRAS1c.182A>Tp.(Gln61Leu)missensepathogenicpathogenicpathogenicNA 323126
SRR13260957FANCE6c.1095A>Cp.(Arg365Ser)missenseNA 3uncertainbenign0.0005262036
SRR13260959TP5317c.659A>Gp.(Tyr220Cys)missensepathogenicpathogenicpathogenic0.00000754143
SRR13260959ATRX23c.4675A>Tp.(Lys1559Ter)stop gainNA 3NA 3lik. Pathog. 4NA 32241
SRR13260961TP5317c.749C>Tp.(Pro250Leu)missensepathogenicuncertainpathogenicNA 32668
1 Freq. = frequency; 2 Lik. Benign = likely benign; 3 NA = not applicable; 4 Lik. Pathog. = likely pathogenic.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gozzellino, L.; Nannini, M.; Urbini, M.; Pizzi, C.; Leone, O.; Corti, B.; Baldovini, C.; Angeli, F.; Foà, A.; Pacini, D.; et al. Genomic Landscape Comparison of Cardiac versus Extra-Cardiac Angiosarcomas. Biomedicines 2023, 11, 3290. https://doi.org/10.3390/biomedicines11123290

AMA Style

Gozzellino L, Nannini M, Urbini M, Pizzi C, Leone O, Corti B, Baldovini C, Angeli F, Foà A, Pacini D, et al. Genomic Landscape Comparison of Cardiac versus Extra-Cardiac Angiosarcomas. Biomedicines. 2023; 11(12):3290. https://doi.org/10.3390/biomedicines11123290

Chicago/Turabian Style

Gozzellino, Livia, Margherita Nannini, Milena Urbini, Carmine Pizzi, Ornella Leone, Barbara Corti, Chiara Baldovini, Francesco Angeli, Alberto Foà, Davide Pacini, and et al. 2023. "Genomic Landscape Comparison of Cardiac versus Extra-Cardiac Angiosarcomas" Biomedicines 11, no. 12: 3290. https://doi.org/10.3390/biomedicines11123290

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop