<<

www.oncotarget.com Oncotarget, 2018, Vol. 9, (No. 28), pp: 19753-19766

Research Paper Mouse genetic background influences whether HrasG12V expression plus Cdkn2a knockdown causes angiosarcoma or undifferentiated pleomorphic

Laura P. Brandt1,2, Joachim Albers1, Tomas Hejhal1, Svende Pfundstein1,3, Ana Filipa Gonçalves1, Antonella Catalano1,6, Peter J. Wild4 and Ian J. Frew1,2,5,6 1Institute of Physiology, University of Zurich, Zurich, Switzerland 2Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland 3Zurich Integrative Rodent Physiology, University of Zurich, Zurich, Switzerland 4Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland 5BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany 6Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany Correspondence to: Ian J. Frew, email: [email protected] Keywords: angiosarcoma; undifferentiated pleomorphic sarcoma; MuLE lentivirus; H-Ras; mouse model Received: March 22, 2017 Accepted: February 28, 2018 Published: April 13, 2018 Copyright: Brandt et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

ABSTRACT

Soft tissue are rare mesenchymal tumours accounting for 1% of adult malignancies and are fatal in approximately one third of patients. Two of the most aggressive and lethal forms of soft tissue sarcomas are angiosarcomas and undifferentiated pleomorphic sarcomas (UPS). To examine sarcoma-relevant molecular pathways, we employed a lentiviral gene regulatory system to attempt to generate in vivo models that reflect common molecular alterations of human angiosarcoma and UPS. Mice were intraveneously injected with MuLE lentiviruses expressing combinations of shRNA against Cdkn2a, Trp53, Tsc2 and Pten with or without expression of HrasG12V, PIK3CAH1047R or Myc. The systemic injection of an ecotropic lentivirus expressing oncogenic HrasG12V together with the knockdown of Cdkn2a or Trp53 was sufficient to initiate angiosarcoma and/or UPS development, providing a flexible system to generate autochthonous mouse models of these diseases. Unexpectedly, different mouse strains developed different types of sarcoma in response to identical genetic drivers, implicating genetic background as a contributor to the genesis and spectrum of sarcomas.

INTRODUCTION profiles, including TP53 mutation, CDKN2A deletion and MDM2 amplification [2–4]. Soft tissue sarcomas are rare mesenchymal Undifferentiated pleomorphic sarcomas (UPS), malignancies that account for approximately 1% of all previously referred to as malignant fibrous histiocytomas . The WHO has defined over 100 different soft tissue (MFH), account for approximately 5% of adult soft tissue sarcoma subtypes named after the tissue that they most sarcomas and represent one of the most common types of closely resemble [1]. Based on molecular characteristics, high-grade soft tissue sarcoma. Standard treatment options soft tissue sarcomas can be divided in two broad categories: are surgical resection, radiotherapy, and chemotherapy, which sarcomas with simple karyotypes, such as chromosomal in many cases are not curative, highlighting the necessity to translocations, and sarcomas with more complex genetic develop novel targeted treatments. It is not clear whether www.oncotarget.com 19753 Oncotarget UPS represents a group of de-differentiated sarcomas that driver oncogenes and tumour suppressor genes to the share a common morphology but which originated from formation of angiosarcoma and UPS. Different mouse different cell types or if all UPS tumours arise from an as- strains were injected intraveneously with ecotropic MuLE yet-unidentified common cell of origin [5]. The genetic lentiviruses expressing combinations of shRNA against alterations responsible for the development of UPS are Cdkn2a, Trp53, Tsc2 and Pten with or without expression also incompletely understood. TP53 alterations have been of HrasG12V, PIK3CAH1047R or Myc. Tumour development identified in 17% of human UPS [2] and CDKN2A loss was monitored by in vivo imaging. We successfully seems to be an alternative to TP53 deletion [3]. HRAS and generated new models of angiosarcoma and of UPS based KRAS mutations have been identified in up to 50% of human on oncogenic HrasG12V expression in combination with UPS tumours [6–8]. Mouse studies have confirmed that knockdown of Cdkn2a or Trp53. Unexpectedly, different the cooperation of oncogenic Kras and Trp53 or Cdkn2a mouse strains developed different types of sarcoma in deficiency resulted in the development of undifferentiated response to identical genetic drivers. pleomorphic sarcomas in different tissues [9–12]. Another clinically aggressive subtype of high- RESULTS grade soft tissue sarcoma is angiosarcoma. These tumours represent rare malignancies of endothelial Expression of oncogenic HrasG12V plus differentiation that account for approximately 1% of all knockdown of Cdkn2a causes angiosarcoma soft tissue sarcomas. Angiosarcomas show a wide anatomic development in SCID/beige mice distribution and arise spontaneously or secondarily to radiation, toxic chemicals (e.g. vinyl chloride) or chronic To functionally test the contributions of different lymphoedema (Stewart-Treves syndrome). Treatment candidate driver oncogenes and tumour suppressor genes options are limited and the prognosis is poor [13]. to the formation of angiosarcoma, we generated a panel Genetic mutations and amplifications of VEGF, MDM2, of lentiviral vectors based on the MuLE system [10] TP53, CDKN2A, KRAS and MYC have been described in (Supplementary Figure 1A), to induce genetic alterations angiosarcoma patients [14–17]. MYC gene amplifications that reflect some of the most commonly found alterations are commonly found in radiation-induced angiosarcomas of human angiosarcomas. We first utilised these ecotropic [18]. A recent publication reported that the majority of MuLE lentiviruses expressing combinations of shRNA or genetic alterations were found in the p53 and MAPK shRNA-miR30 against Cdkn2a, Trp53, Tsc2 and Pten with pathways. TP53 was mutated in 35% of the lesions and or without expression of oncogenic HrasG12V, oncogenic CDKN2A lost in 26%. 53% of angiosarcomas displayed PIK3CAH1047R or Myc vectors to attempt to generate MAPK pathway activation, and harboured genetic panels of genetically-engineered angiosarcoma cell lines activating mutations in KRAS, HRAS, NRAS, BRAF, by infecting a disease-relevant cell type, namely primary MAPK1 or inactivating mutations in NF1 and PTPRB1 murine endothelial cells from the spleen (pMSECs). [19, 20]. Several in vivo mouse studies showed the Western blotting and real time PCR assays of puromycin- involvement of loss of function of the p53 tumour selected cultured cells infected with these vectors verified suppressor in angiosarcoma development [21–23]. In that they effectively induced the desired changes in gene addition, the in vivo deletion of Cdkn2a in mice lead to expression (Supplementary Figure 1B–1E). Consistent the development of lesions which recapitulate human with an oncogenic activity of these genetic changes, all cell angiosarcoma, however, only 30% of the mice displayed lines, with the exception of HrasG12V expression alone and angiosarcomas within 100 days [24]. Furthermore, HrasG12V expression plus shTrp53, exhibited increased rates alterations in the PI3K/AKT/mTOR pathway have of proliferation (Supplementary Figure 2A). The absence been identified in a small percentage of patients [19, of increased proliferation induced by oncogenic HrasG12V 25, 26] and deletion of Tsc1, a tumour suppressor that or oncogenic HrasG12V plus Trp53 knockdown is likely to negatively regulates the pathway, induced the formation of be mediated by the upregulation of p16INK4A protein in mice [27]. Another report showed that expression observed in pMSEC cells infected with these the in vivo deletion of Notch1 resulted in the development vectors (Supplementary Figure 1B) as removal of this of hepatic angiosarcomas with a penetrance of 86% at 50 putative proliferative barrier by knockdown of Cdkn2a weeks after gene deletion [28], although genetic alterations increased cellular proliferation (Supplementary Figure 2A). in the Notch pathway have not been reported in human To further investigate potential transformed angiosarcomas. Although these studies have been helpful cellular behaviour we cultured cells of all genotypes in uncovering aspects of sarcomagenesis, there is limited on low-attachment plates, however none of the genetic understanding of the interactions between cooperating combinations allowed the proliferation of cells as spheres genetic alterations. or masses (data not shown), indicating that they do not In this study we employed a mouse genetic approach have anchorage-independent proliferation capacity. We using the MuLE lentiviral gene regulatory system [10] to next asked whether these cellular systems might represent functionally test the contributions of different candidate experimentally tractable allograft tumour models by www.oncotarget.com 19754 Oncotarget injecting wild type pMSECs or shCdkn2a plus HrasG12V cells. Tumour cells exhibited high levels of expression pMSECs subcutaneously into SCID/beige immunodeficient of H-RAS in comparison to surrounding normal tissue, mice. Within one month of injection, both the wild type and indicating that the MuLE virus is functional in vivo shCdkn2a plus HrasG12V cells formed blood-filled lesions (Supplementary Figure 3B). Positive immunoreactivity (Supplementary Figure 2B) that were lined with CD31- to antibodies against the endothelial cell marker proteins positive endothelial cells with atypical nuclei growing either CD31 and von Willebrand Factor (vWF) confirmed the as single layers or in papillary projections (Supplementary endothelial differentiation of the tumour cells (Figure 2A). Figure 2C). The injected cells apparently have the capacity The lesions showed variable staining for VIMENTIN, to co-opt or integrate into local blood vessels, resulting in DESMIN and SMOOTH MUSCLE ACTIN (SMA), large blood-filled vascular structures. Based on the fact ranging from an absence of staining to some tumours that this phenotype also arose following injection of wild showing strong positivity (Figure 2A). None of the tumours type pMSECs in the absence of oncogene activation or exhibited nuclear staining for the skeletal muscle markers tumour suppressor inactivation, we conclude that this ex MYOD1 and MYOGENIN (Figure 2A). Supplementary vivo engineered cellular system does not represent a good Figure 4 shows positive and negative control stainings allograft model system for angiosarcoma. for the different antibodies employed throughout this We next sought to assess the tumour forming study. Tumours that arose in Trp53 knockdown plus capacity of the same genetic changes that were tested in oncogenic HrasG12V injected mice displayed an identical the experiments above directly in vivo. 4–6-week-old histological appearance and immunohistochemical staining SCID/beige mice were intravenously injected via the tail profile to tumours in Cdkn2a knockdown plus oncogenic vein with concentrated ecotropic MuLE vectors that carried HrasG12V injected mice (Figure 2B). In summary, these an expression element for firefly luciferase (Figure 1A) histological and molecular features are consistent with in order to label infected cells and to trace potential a diagnosis of angiosarcoma. Indeed, analyses of three tumour development in vivo over time. The intravenous human angiosarcomas revealed that they have a simlar injection of an ecotropic lentivirus expressing oncogenic histological appearance and exhibit a similar pattern of HrasG12V together with knockdown of Cdkn2a (n = 21 of immunoreactivity to the mouse tumours (Figure 2C). We 32 injected mice) or Trp53 (n = 2 of 4 injected mice) in conclude that we have developed a rapid autochthonous SCID/beige mice induced increases in luciferase signals mouse model of angiosarcoma that is trackable via live over 4–8 weeks (Figure 1B and Supplementary Figure 3A). animal imaging and that reflects the frequent genetic These signals were widely distributed in different organs alterations that arise in human angiosarcoma tumours. throughout the body. One of three mice injected with a vector expressing only oncogenic HrasG12V developed Different immunocompetent mouse strains signals in the brain approximately 6 months after injection. display different types of soft tissue sarcomas in None of the other viruses was sufficient to cause any large response to expression of oncogenic HrasG12V plus increases in luciferase signal within 6 months of injection, loss of Cdkn2a function demonstrating that these combinations of genetic alterations are not oncogenic in this setting. To more accurately model the complexities of Dissections of mice revealed that the increased tumour development in humans it would be desirable to luciferase signals corresponded to the presence of bloody- have tumour models that arise in immunocompetent mice. appearing tumours in different organs (Figure 1C and 1D We therefore investigated whether similar tumours arose and Supplementary Figure 3). From 32 mice injected with in response to intravenous injection of shCdkn2a plus shCdkn2a plus HrasG12V MuLE vectors, 21 mice developed HrasG12V MuLE vectors in the Fox Chase CB17, 129/Sv a total of 24 tumours in various tissues including testicle and C57BL/6 mouse strains. (n = 9, 38%), brain (n = 7, 30%), spleen (n = 2, 8%), uterus Fox Chase CB17 mice carry the immunoglobulin (n = 2, 8%), ovary (n = 1, 4%), lung (n = 1, 4%), colon heavy chain allele from C57BL/Ka mice on a BALB/c (n = 1, 4%) and eye (n = 1, 4%). Histological analysis background. They serve as an ideal control for SCID/beige of these tumours revealed poorly demarcated malignant mice as they represent the identical genetic background neoplasms with hemorrhage and irregular, anastomosing but have a normal immune system. Within 4 weeks, 7 of vascular channels. Endothelial lining showed 8 (88%) Fox Chase CB17 mice injected intraveneously multilayering and intraluminal tufting with nuclear with shCdkn2a plus HrasG12V MuLE vectors developed atypia, hyperchromasia, enlargement and irregularity angiosarcomas with comparable growth kinetics and (Supplementary Figure 3A, arrowheads). Mitotic activity histology to those that arose in SCID/beige mice was variable and back-to-back vascular channels appeared (Supplementary Figure 5A). The anatomic distribution of sieve-like. Atypical cells were either spindled, epitheloid the tumours in Fox Chase CB17 mice was similar to the or mixed. There was more solid growth in more poorly tumour distribution seen in SCID/beige mice and included differentiated areas. Intraluminal erythrocytes were a brain (n = 4, 50%), testicle (n = 3, 38%) and spleen (n = 1, common feature as well as large areas filled with red blood 12%) (Supplementary Figure 5B). The tumours presented www.oncotarget.com 19755 Oncotarget as bloody lesions and stained positively for CD31 and affect tumour formation and provide a new autochthonous vWF by immunohistochemistry. Like the tumours in angiosarcoma model in an immunocompetent background. SCID/beige mice, these tumours also showed variable To investigate sarcoma formation in mouse staining for VIMENTIN, DESMIN and SMA as well as backgrounds that are more commonly utilised for an absence of staining for MYOD1 and MYOGENIN biomedical research we next employed 129/Sv mice. (Supplementary Figure 5C). These experiments Intraveneous injections of shCdkn2a plus HrasG12V MuLE demonstrate that a competent immune system does not viruses in 129/Sv mice (n = 8) caused a strong luciferase signal increase and the development of multiple tumours

Figure 1: HrasG12V expression plus knockdown of Cdkn2a or Trp53 causes angiosarcomas in SCID/beige mice. (A) Schematic of MuLE vectors simultaneously expressing combinations of shRNAs against Cdkn2a, Trp53, Pten and Tsc2 with or without expression of HrasG12V, Myc or PIK3CAH1047R and a Luciferase reporter. (B) Bioluminescence imaging 4, 11, 20, 28 and 36 days after the injection of MuLE lentiviruses expressing shRNA against Cdkn2a together with HrasG12V into the tail veins of 4-6-week-old SCID/beige mice. Numbers of mice that developed tumours and total number of injected mice are shown for each construct. (C) Bioluminescence imaging and (D) photographs showing examples of tumour-bearing organs from shCdkn2a plus HrasG12V and shTrp53 plus HrasG12V injected animals. Scale bar: 10 mm. www.oncotarget.com 19756 Oncotarget with 100% penetrance within 4 weeks (Figure 3A and 3B, (n = 4) additionally developed subcutaneous tumours (n = 5) Supplementary Figure 6A). Bloody-appearing tumours that were located in the head and neck region and close to were observed in testicles of 75% of male mice (n = 3) the subcutis of the vulva. These tumours were solid and and in uteri (n = 2) and ovaries (n = 2) of 50% of female white in appearance (Figure 3C, last column). Neither mice. 25% of mice carried lesions in the spleen (n = 2), the sub-cutaneous location, nor the gross morphological and 13% in lung (n = 1) and brain (n = 1), similar to appearance, were ever seen in tumours in SCID/beige or results in SCID/beige mice. However, all female mice Fox Chase CB17 mice. While all of the bloody-appearing

Figure 2: Histological and molecular characterisation of angiosarcomas. H&E and immunohistochemical stainings using the indicated antibodies of tumours derived from the systemic injection of (A) shCdkn2a plus HrasG12V and (B) shTrp53 plus HrasG12V viruses. (C) Stainings of a human angiosarcoma tumour. Similar staining patterns were observed in two additional cases from different patients. Low magnification scale bars: 1000 µm and high magnification scale bars: 100 µm. www.oncotarget.com 19757 Oncotarget Figure 3: HrasG12V expression plus knockdown of Cdkn2a causes angiosarcomas and undifferentiated pleomorphic sarcomas in 129Sv mice. (A) Bioluminescence imaging 5, 13, 20, 26 and 33 days after the injection of MuLE lentiviruses expressing shRNA against Cdkn2a together with HrasG12V into the tail veins of 4-6-week-old 129/Sv mice. (B) Bioluminescence imaging showing examples of tumour-bearing organs.. (C) H&E and immunohistochemical stainings using the indicated antibodies. Low magnification scale bars: 1000 µm and high magnification scale bars: 100 µm. www.oncotarget.com 19758 Oncotarget tumours exhibited an identical histological appearance control stainings of all of these antibodies are shown in and pattern of immunoreactivity similar to tumours that Supplementary Figure 4. arose in SCID/beige mice (Figure 3C, Supplementary Given the absence of morphologic features of Figure 6B), classifying them as angiosarcomas, the any cellular lineage, the absence of clear evidence for subcutaneous tumours exhibited a completely different positivity of tumour cells for a panel of lineage markers histology and immunohistochemical staining profile. and absence of strong and diffuse pan-CYTOKERATIN These tumours contained cells with rhabdoid features; staining we therefore favour the diagnosis of these i.e., large polygonal cells with gigantic bizarre nuclei tumours as high-grade UPS Not Otherwise Specified in (Figure 4A; arrowheads), abundant, deeply eosinophilic keeping with WHO diagnostic guidelines. However, since cytoplasm in a tadpole- or racquet shape and growing the differential diagnosis of UPS versus sarcomatoid in a storiform pattern. Nuclei displayed high mitotic carcinoma is necessarily a diagnosis of exclusion, it indices, irregular nuclear membranes, and eosinophilic remains formally possible that these tumours might cytoplasmic inclusions. These tumors showed necrotic represent an unknown type of undifferentiated sarcomatoid regions, acute inflammatory responses and were highly carcinoma arising from a cell type that we have not been invasive, infiltrating surrounding tissues including able to identify. As already known from human tumour muscle and fat (Figure 4A, arrows). The absence specimens, the differential diagnosis of these lesions of apparent features of any definable cell lineage is remains a continuous matter of debate. Importantly, these suggestive of a diagnosis of undifferentiated pleomorphic analyses show that 129/Sv mice develop two different sarcoma (UPS). Indeed, while these tumours showed high types of tumours, angiosarcomas and UPS, in response to levels of H-RAS expression in comparison to adjacent the same oncogenic stimulus. normal tissue (Figure 4C, 4D) and were immunoreactive To further investigate the effect of genetic for the common mesenchymal marker VIMENTIN, the background on tumour formation, we injected C57BL/6 tumour cells did not stain for lineage markers including mice with shCdkn2a plus HrasG12V MuLE lentiviruses. CD31, vWF, DESMIN, SMA, MYOD1 or MYOGENIN Within 4–8 weeks of injection both male and female (Figure 3C, last column). Given the subcutaneous C57BL/6 mice showed large increases in luciferase signal location of these tumours, we further investigated and developed subcutaneous lesions with 92% penetrance whether the tumours might potentially represent (12 of 13 injected mice) (Figure 5A and 5B). Tumours sarcomatoid variants of malignancies derived from a (n = 12) that developed in C57BL/6 mice were solid skin cell. Tumours in 129/Sv mice stained negatively and white in appearance like those seen in female 129/ for the melanoma marker protein PMEL (the HMB45 Sv mice. They were subcutaneous and located either in antigen) (Figure 4E). Melanomas, as well as other types the head and neck area, lower leg or at the junction of of neural lineage-derived tumours are typically diffusely the tail and spine. These tumours exhbited an identical positive for S100 and PMEL. The majority of tumour histological appearance to the UPS tumours that arose cells in tumours in 129/Sv mice were negative for S100 in 129/Sv mice (Figure 4B). These tumours similarly but some scattered cells within the tumour displayed showed an absence of staining for all of the markers that positive immunoreactivity for S100 (Figure 4F). Since were used to characterise the tumours in 129Sv mice, macrophages also stain positively for S100, it is likely except VIMENTIN (Figure 5C). Based on these results that this staining is due to the presence of inflammatory we conclude that shCdkn2a plus HrasG12V MuLE viruses cells. Tumour cells in human sarcomatoid squamous cell solely cause high-grade UPS in C57BL/6 mice. carcinomas typically exhibit nuclear immunoreactivity for p63 [29]. While normal skin showed strong nuclear DISCUSSION p63 immunoreactivity as an internal positive control, only a small number of scattered cells in tumours in Major hurdles in studying sarcoma pathologies are 129/Sv mice displayed weak cytoplasmic staining the relative rarities of the human diseases and the absence for p63 (Figure 4G), arguing against a diagnosis of for many sarcoma subtypes of good pre-clinical models. sarcomatoid squamous cell carcinoma. Tumours were Here, we used the MuLE lentiviral gene regulatory system also completely negative for the epithelial marker [10] to investigate the molecular genetics underlying the EpCAM (Figure 4H), but scattered cells within the 129/ pathogenesis of two types of soft tissue sarcomas, namely Sv subcutaneous tumours (Figure 4I), as well as rare angiosarcoma and UPS. The MuLE system allows the cells in angiosarcomas in SCID/beige mice (Figure 4J), direct introduction of multiple genetic alterations in reacted with a pan-CYTOKERATIN antibody, another somatic cells in vivo by lentiviral injection. Bypassing epithelial marker. However, since human sarcomas, germline transgenic approaches has benefits in terms of including UPS and angiosarcoma, can contain cells that time and costs and offers flexibility in terms of the strains are immunoreactive for pan-CYTOKERATIN caution of mice that can be used for the experiments, allowing should be taken in interpreting this staining as providing comparisons between different genetic backgrounds. strong evidence of epithelial origin [30]. Positive Guided by the genetics of human angiosarcomas and UPS www.oncotarget.com 19759 Oncotarget Figure 4: High grade, locally invasive undifferentiated pleomorphic sarcomas form in 129/Sv and C57BL/6 mice. (A, B) High magnification images of H&E stained tumour sections derived from the intraveneous injection of shCdkn2a plus H-RasG12V viruses in (A) 129/Sv mice and (B) C57BL/6 mice. Arrowheads indicate atypical nuclei and arrows indicate entrapped muscle fibres. Images depict tumour invasion into the surrounding muscle or fat tissue. (C, D) Immunohistochemical staining using an anti-H-RAS antibody showing high H-RAS expression in a subcutaneous tumour (C) but absence of staining in adjacent normal tissue (D). (E–I) Representative immunohistochemical staining patterns for the indicated antibodies in subcutaneous tumours. In (G) the margin between normal skin (p63 nuclear positive) and the tumour is shown by a dotted line. (J) Occasional cells in angiosarcoma tumours from SCID/ beige mice stain positively for pan-CYTOKERATIN. Scale bars: 100 µm. www.oncotarget.com 19760 Oncotarget Figure 5: HrasG12V expression plus knockdown of Cdkn2a causes undifferentiated pleomorphic sarcomas in C57BL/6 mice. (A) Bioluminescence imaging 8, 15, 30 and 36 days after the injection of MuLE lentiviruses expressing shRNA against Cdkn2a together with HrasG12V into the tail veins of 4-6-week-old C57Bl/6 mice. (B) Bioluminescence imaging showing examples of a subcutaneous tumour prior to (left) and after (right) resection. (C) H&E and immunohistochemical stainings using the indicated antibodies. Scale bars: 100 µm. www.oncotarget.com 19761 Oncotarget tumours, we functionally tested the contributions of the viruses into skeletal muscle in SCID-beige mice caused candidate sarcoma tumour suppressors Cdkn2a, Trp53, UPS [10] but intravenous injection caused angiosarcoma, Tsc2 and Pten and the candidate oncogenes HrasG12V, whereas in C57/BL6 mice both modes of injection caused PIK3CAH1047R or Myc. We discovered that the systemic UPS formation [10]. Our data demonstrate that the same injection of ecotropic lentiviruses expressing oncogenic combination of genetic drivers can cause different types of HrasG12V together with the knockdown of Cdkn2a or tumours based not only on the site of viral delivery, likely Trp53 was sufficient to initiate angiosarcoma formation due to infection of different cell types, but also based on in multiple organs in SCID/beige mice. shCdkn2a plus the genetic background of the mouse strain. HrasG12V MuLE viruses also induced angiosarcoma Why does intraveneous, lentiviral-mediated delivery formation in Fox Chase CB17 and 129/Sv mice, but of the same genetic alterations cause different tumours in surprisingly additionally caused UPS development in 129/ different strains of mice? One possibility relates to genetic Sv mice and only UPS development in C57/BL6 mice. modifiers such as allelic variants, sequence differences, These observations are consistent with the fact that RAS- epigenetic modifications and gene expression levels that MAPK and p53 pathway alterations are frequently found could potentially influence tumour phenotypes [36]. There in high-grade soft tissue sarcomas, such as angiosarcomas is precedent for the fact that identical genetic alterations can and UPS [2, 3, 6, 7, 19]. result in different phenotypes in different mouse strains. Our experimental approach provides new models of One example is that the incidence of mammary tumours at least some of the genetic subsets of human angiosarcomas varies among strains heterozygous for Trp53, with C57BL/6 and UPS tumours. It should also be noted that while mice being resistant and BALB/c mice being susceptible histological and immunohistochemical analyses revealed [37, 38]. Indeed, BALB/c mice carry an allelic variant numerous similarities between mouse and human tumours, of Cdkn2a that leads to compromised p16 activity, likely there are differences in terms of the anatomical sites where causing an increased susceptibility to develop certain the mouse model tumours arise and the most frequent sites tumours [39, 40]. In the context of the results of the present where naturally occurring human counterpart tumours study, it might be possible in future long-term and large- arise. Mouse angiosarcomas in our model arose most scale studies to utilise multi-generational interbreeding frequently in the brain, genital organs and spleen. Human strategies between C57BL/6 and Fox Chase CB17 strains, angiosarcomas are most frequently found in the skin (60%), coupled with genomic analyses, to narrow down loci that deep soft tissue (25%), breast (8%) and more rarely in liver contribute to the type of sarcoma that develops in response and spleen [1]. There are also examples of cases of human to oncogenic HrasG12V expression and Cdkn2a knockdown. angiosarcomas arising in the testes [31], uterus and ovaries By extension, our observations highlight the fact that also [32], brain [33], lungs [34] and colon [35], which are all in humans the genetic background of every individual may sites at which tumours were observed in our mouse models. potentially influence the outcome of oncogenic mutations Since endothelial cells are present in all tissues in the body, in terms of what type of sarcoma develops. In this context, it appears that angiosarcomas have the potential to arise in it is noteworthy that a study of UPS tumours in Korean any organ in humans. In our mouse model, the anatomical patients identified a high frequency of oncogenic KRAS distribution of tumours is likely to be influenced by the and HRAS mutations [6], but a study by the same authors method of intravascular injection, which will presumably of UPS tumours in American patients revealed that these influence the likelihood of viral infection at different sites tumours lacked KRAS and HRAS mutations [7], arguing in the animal. In contrast, UPS tumours in our models that different human genetic backgrounds (or environmental were always localised subcutaneously. In humans UPS can factors) may select for different oncogenic mutations during arise anywhere in the body but most frequently occurs in the course of sarcoma development. To further investigate lower extremeties and sometimes in the retroperitoneum, this issue, we analysed the mutation spectrum of a series of head and neck and breast. These tumours typically arise in 48 human UPS tumours from the TCGA provisional dataset deep subfascial tissue, but about 10% of UPS are primarily using cBioPortal software (http://www.cbioportal.org/ subcutaneous [1], similarly to the tumours that arise in our study?id=sarc_tcga#summary) (Supplementary Figure 7). mouse models. The ethnicity of this patient cohort is 42 white, 4 black Genetic cooperation between RAS pathway or African American, 2 hispanic or latino, 2 unknown but activation and loss of Cdkn2a tumour suppressor function no reported Asian patients. Interestingly, this set of UPS has been shown in other mouse models of sarcomas. tumours does not display activating point mutations in Intramuscular injection of Adeno-Cre in the leg of loxP- any of the RAS-family genes in keeping with the previous STOP-loxP-KrasG12D/+;Cdkn2afl/fl mice caused UPS study of American patients [7]. However, the majority of development [9, 11] and we have previously shown these tumours exhibit loss-of-function point mutations or that intramuscular injection of the same shCdkn2a plus copy number deletions of the TP53 and CDKN2A tumour HrasG12V or shTrp53 plus HrasG12V MuLE lentiviruses that suppressor genes and all but one of these tumours exhibit were used in this study caused the developement of high- multiple copy number gains or amplifications of genes grade UPS [10]. It is noteworthy that injection of these involved in the RAS-RAF-MEK-MAPK signalling cascade, www.oncotarget.com 19762 Oncotarget as well as copy number losses of the NF1 tumour suppressor proposed as the cell of origin of human UPS [42–45]. It gene that negatively regulates signalling by this cascade. has been shown that the overexpression of oncogenic Kras These results give rise to a hypothesis that could be tested in cultured MSCs isolated from the bone marrow of Trp53 in future studies, namely that chromosomal copy number knockout C57BL/6 mice caused transformation of these alterations rather than point mutations account for activation cells in vitro [46]. We have shown that ecotropic MuLE of the RAS-signalling pathway in combination with loss lentiviruses can infect a variety of cultured primary cells, of the CDKN2A-TP53 tumour suppressor pathways to including mouse embryonic stem cells [10] and although drive UPS tumour formation in non-Asian individuals. In bone marrow represents the main source of MSCs, MSCs summary, we believe that our functional tumour modelling also exist in low numbers in peripheral blood [47]. It is studies, combined with the above-described genetic theoretically possible that infected MSCs in the peripheral analyses, further emphasise the need to consider individual blood could give rise to UPS in 129/Sv and C57BL/6 mice. tumours at the molecular level rather than at the level of In summary, these new experimental models will histo-pathological appearance when thinking about the facilitate future pre-clinical studies for establishing development and clinical application of new molecularly new therapeutic interventions for these aggressive targeted sarcoma therapies. malignancies. While it is somewhat surprising that the A second possible explanation for the different other tested candidate tumour suppressors and oncogenes tumour types that arise in the different mouse stains relates were not sufficient to cause tumour formation, given their to cellular tropism. It remains possible that MuLE viruses frequent mutational alteration in human angiosarcomas might infect different spectra of cells in different mouse and UPS, the flexible nature of the MuLE system should strains, potentially due to different expression levels of also allow the testing of other candidate oncogenes, the mouse cationic amino acid transporter 1 (mCAT1) modifier genes or tumour suppressor genes that will likely protein that serves as the virus receptor. However, to continue to emerge from ongoing genomic studies of these our knowledge, nothing is known about the expression rare tumours. patterns of this protein in different strains of mice and in our experience identifying infected cells in vivo is only MATERIALS AND METHODS possible using genetic reporter mouse lines [10]. However, the identification of infection of a particular cell type is Mice at best a hint that it might be able to be transformed and act as the cell of origin of a tumour. Consistent with the SCID/beige mutant mice (C.B-17/CrHsd- idea that angiosarcoma and UPS tumours might arise due PrkdcScidLystbg-J) and C57BL/6JRccHsd mice were obtained to the infection of different cell types it is noteworthy that from Envigo. 129S2/SvPasOrlRj mice were obtained from all of the UPS tumours that arose in 129/Sv and C57BL/6 Janvier Labs and Fox Chase CB17™ mice (C.BKa-lghb/ mice were subcutaneous, whereas angiosargomas in 129/ lcrCrl) were obtained from Charles River Laboratories. All Sv, Fox Chase CB17 and SCID/beige mice were found in mouse experiments were approved by the Veterinary Office several different organs. These differences in anatomical of the Canton of Zurich under the licence 137/2013. distribution are suggestive of different cells of origin of these tumours. Proof of cell of orgin of a tumour Generation of MuLE vectors requires a lineage tracing experiment. Given the fact that angiosarcomas express markers of endothelial cells and The majority of the MuLE Entry and Destination are intimately connected to the normal vascular network vectors used in this study were previously described of the mouse (blood-filled lesions) it appears likely that [10] and were recombined using MultiSite Gateway LR the cell of origin of these tumours are vascular endothelial 2-fragment recombinations to generate the final viral cells. Future experiments could involve utilising lineage constructs. New MuLE Entry vectors carrying 7SK tracing experiments using a Cre-driver such as Tie2- promotor-driven expression of shRNA against Tsc2 (Sigma, CreERT2 [41] to genetically label endothelial cells prior to TRCN0000306244) and CMV promotor-driven expression injection of shCdkn2a plus HrasG12V MuLE lentiviruses. of hemagglutinin (HA) tagged phosphoinositide 3-kinase A potential caveat of these studies may however be that H1047R (HA-PIK3CA H1047R, Addgene 12524, deposited the tumour type that will arise will also be dictated by by Dr. Jean Zhao) were generated. Ecotropic lentiviral the (likely mixed) genetic background of the strain of the vectors were produced by using calcium phosphate- lineage tracing mouse. The cell of origin of UPS remains mediated transfection of HEK293T cells and the viral unknown and it is not clear whether UPS represent a group preparation was concentrated as described previously [10]. of de-differentiated sarcomas which share a common morphology but originated from different cell types or if all Culture, infection and assays of pMSECs UPS tumours arise from a common cell of origin [5]. One interesting hypothesis that could be experimentally tested C57BL/6 mouse primary spleen endothelial cells is that adult mesenchymal stem cells (MSCs) have been (pMSECs, C57-6057, Cell Biologics) were cultured according www.oncotarget.com 19763 Oncotarget to the instructions of the supplier in complete mouse endothelial Sigma-Aldrich), anti-H-RAS (1:1000, sc-520, Santa cell medium (M1168, Cell Biologics) in a humidified 5% Cruz Biotechnology Inc.), anti-MYC (1:1000, AB32072,

(v/v) CO2 incubator at 37° C. 50% confluent pMSECs were Epitomics (Abcam)), anti-p16 (1:1000, sc-1207, Santa transduced with lentiviral vectors in the presence of 4 μg/ml Cruz Biotechnology Inc.), anti-p19 (1:1000, sc-32748, polybrene (Hexadimethrine bromide, 107689, Sigma-Aldrich). Santa Cruz Biotechnology Inc.), anti-p53 (1:500, NCL- Puromycin (ANT-PR-1, InvivoGen) was added to cultures 48 p53-CM5p, Novocastra), anti-PTEN (1:1000, sc-7974, h after infection and drug selection continued until all control Santa Cruz Biotechnology Inc.), anti-TSC2 (1:1000, cells were dead. pMSECs were seeded in triplicates at density 3990s, Cell signaling) and anti-VINCULIN (1:5000, of 2,000 cells per well in 96-well plates and analysed after 1, 3, ab129002, Abcam). 5 and 7 days using the SRB assay [48]. For allograft assays, 1 × 106 pMSECs were injected subcutaneously into SCID/beige Real-time PCR mice. Total RNA was isolated from cells using In vivo tumour formation assays NucleoSpin RNA kit (Macherey-Nagel). cDNA systhesis was performed with random hexamer primers and Ready- Concentrated ecotropic lentiviruses were injected To-Go You-Prime First-Strand Beads (GE Healthcare). (3 × 105 infectious viral particles in a volume of 10 ml/kg) Real-time PCR analysis was performed with SYBR using a 30G insulin syringe into the lateral tail vein of Green JumpStart Taq Ready Mix (Sigma-Aldrich) 4–6-week old mice. Noninvasive in vivo bioluminescence and the following primer pairs: S12 (5′-GAAGCTGC imaging was performed using the IVIS Spectrum (Perkin CAAAGCCTTAGA-3′, 5′-AACTGCAACCAACCACC Elmer) together with the Living Image software (version TTC-3′) and PIK3CA (5′- CCACGACCATCATCAGGT 4.4). Mice were anaesthetized by using 2.5% isoflurane. GAA-3′, 5′- CCTCACGGAGGCATTCTAAAGT-3′). During imaging, the isoflurane levels were reduced to 1.5%. All fluorescence measurements were performed in Author contributions epi-fluorescence mode. For bioluminescence imaging, mice were injected subcutaneously with 150 mg/kg D-luciferin LPB, JA, TH, SP, AFG and AC performed (Caliper, no. 122796) and imaged 15 min after injection. experiments, LPB, JA and IJF analysed data, PJW performed histopathological analyses and LPB and IJF Immunohistochemistry wrote the manuscript with input from all authors.

Tumour-bearing organs were resected, fixed in ACKNOWLEDGMENTS 10% formalin, paraffin-embedded and cut in 5 µm thick sections. Immunohistochemical analysis was performed as We are grateful to Fabiola Prutek, Susanne described [49]. The antibodies used in this study were anti- Dettwiler, Peter Schraml, Christiane Mittmann, Marcel CD31 (1:200, ab28364, Abcam), anti-DESMIN (1:100, Glönkler, and Andre Fitsche for excellent technical D1033, Sigma-Aldrich), anti-EpCAM (1:500, ab71916, assistance. Silvia Behnke (Sophistolab, Muttenz) is Abcam), anti-H-RAS (1:100, GTX116041, GeneTex), thanked for her help with antibody validation. anti-MYOD1 (1:100, M3512, Dako), anti-MYOGENIN (1:500, M3559, Dako), anti-pan-CYTOKERATIN (1:100, CONFLICTS OF INTEREST BMA Biomedicals AG, T1302), anti-PMEL(HMB45) (1:100, Abcam, ab137078), anti-p63 (1:500, ab124762, The authors declare that they have no conflicts of Abcam), anti-alpha-SMA (1:5000, ab5694, Abcam), interest related to this manuscript. anti-S100 (1:2000, S100 DAKO A/S), anti-VIMENTIN (1:500, D21H3, Cell Signaling) and anti-von Willebrand Factor (vWF) (1:1000, F3520, Sigma-Aldrich). FUNDING This research was supported by funding to IJF from Western blotting the European Research Council (260316). This work was Cells were lysed in RIPA buffer (50 mM Tris- funded in part by a University of Zurich grant (Stiftung für HCl pH 8.0, 150 mM sodium chloride, 1% (v/v) NP- Wissenschaftliche Forschung) to PJW. 40, 0.5% (w/v) sodium deoxycholate, 0.1% (w/v) SDS, 5mM sodium fluoride, 1 mM sodium orthovanadate, 1 REFERENCES mM PMSF, 1:100 Protease Inhibitor Cocktail (Sigma- Aldrich)) for protein extraction. Proteins were run on 1. Fletcher C. WHO classification of tumours of soft tissue 8–12% acrylamide gels, transferred to nitrocellulose and bone. Fourth Edition. 2013. membranes and visualized by immunoblotting with the 2. Barretina J, Taylor BS, Banerji S, Ramos AH, Lagos- following antibodies anti-ß-ACTIN (1:2000, A2228, Quintana M, Decarolis PL, Shah K, Socci ND, Weir BA, www.oncotarget.com 19764 Oncotarget Ho A, Chiang DY, Reva B, Mermel CH, et al. Subtype- 15. Manner J, Radlwimmer B, Hohenberger P, Mössinger K, specific genomic alterations define new targets for soft- Küffer S, Sauer C, Belharazem D, Zettl A, Coindre JM, tissue sarcoma therapy. Nat Genet. 2010; 42:715–21. Hallermann C, Hartmann JT, Katenkamp D, Katenkamp K, 3. Pérot G, Chibon F, Montero A, Lagarde P, de Thé H, et al. MYC High Level Gene Amplification Is a Distinctive Terrier P, Guillou L, Ranchère D, Coindre JM, Aurias A. Feature of Angiosarcomas after Irradiation or Chronic Constant p53 Pathway Inactivation in a Large Series of Soft Lymphedema. The American Journal of Pathology. 2010; Tissue Sarcomas with Complex Genetics. The American 176:34–9. Journal of Pathology. 2010; 177:2080–90. 16. Weihrauch M, Bader M, Lehnert G, Koch B, Wittekind 4. Borden EC, Baker LH, Bell RS, Bramwell V, Demetri GD, C, Wrbitzky R, Tannapfel A. Mutation analysis of K-ras-2 Eisenberg BL, Fletcher CD, Fletcher JA, Ladanyi M, in liver angiosarcoma and adjacent nonneoplastic liver Meltzer P, O'Sullivan B, Parkinson DR, Pisters PW, et al. tissue from patients occupationally exposed to vinyl Soft tissue sarcomas of adults: state of the translational chloride. Environmental and Molecular Mutagenesis. 2002; 40:36–40. science. Clin Res. 2003; 9:1941–56. 17. Hayashi T, Koike K, Kumasaka T, Saito T, Mitani K, Terao Y, 5. Matushansky I, Charytonowicz E, Mills J, Siddiqi S, Hricik T, Ogishima D, Yao T, Takeda S, Takahashi K, Seyama K. Uterine Cordon-Cardo C. MFH classification: differentiating angiosarcoma associated with in undifferentiated pleomorphic sarcoma in the 21st Century. a patient with tuberous sclerosis complex: an autopsy case Expert Rev Anticancer Ther. 2009; 9:1135–44. report with immunohistochemical and genetic analysis. Human 6. Yoo J, Robinson RA, Lee JY. H-ras and K-ras gene mutations Pathology. 2012; 43:1777–84. in primary human soft tissue sarcoma: concomitant mutations 18. Guo T, Zhang L, Chang NE, Singer S, Maki RG, of the ras genes. Mod Pathol. 1999; 12:775–80. Antonescu CR. Consistent MYC and FLT4 gene 7. Yoo J, Robinson RA. H-ras and K-ras mutations in soft tissue amplification in radiation-induced angiosarcoma but not in sarcoma. Cancer. 1999; 86:58–63. other radiation-associated atypical vascular lesions. Genes 8. Bohle RM, Brettreich S, Repp R, Borkhardt A, Kosmehl H, Chromosomes Cancer. 2011; 50:25–33. Altmannsberger HM. Single somatic ras gene point 19. Murali R, Chandramohan R, Möller I, Scholz SL, Berger M, mutation in soft tissue malignant fibrous histiocytomas. The Huberman K, Viale A, Pirun M, Socci ND, Bouvier N, American Journal of Pathology. 1996; 148:731–8. Bauer S, Artl M, Schilling B, et al. Targeted massively 9. Kirsch DG, Dinulescu DM, Miller JB, Grimm J, parallel sequencing of angiosarcomas reveals frequent Santiago PM, Young NP, Nielsen GP, Quade BJ, Chaber CJ, activation of the mitogen activated protein kinase pathway. Schultz CP, Takeuchi O, Bronson RT, Crowley D, et al. A Oncotarget. 2015; 6:36041–52. https://doi.org/10.18632/ spatially and temporally restricted mouse model of soft oncotarget.5936. tissue sarcoma. Nature Medicine. 2007; 13:992–7. 20. Behjati S, Tarpey PS, Sheldon H, Martincorena I, Van Loo P, Gundem G, Wedge DC, Ramakrishna M, Cooke SL, 10. Albers J, Danzer C, Rechsteiner M, Lehmann H, Brandt LP, Pillay N, Vollan HKM, Papaemmanuil E, Koss H, et al. Hejhal T, Catalano A, Busenhart P, Gonçalves AF, Brandt S, Recurrent PTPRB and PLCG1 mutations in angiosarcoma. Bode PK, Bode-Lesniewska B, Wild PJ, et al. A versatile Nat Genet. 2014; 46:376–9. modular vector system for rapid combinatorial mammalian genetics. J Clin Invest. 2015; 125:1603–19. 21. Landuzzi L, Ianzano ML, Nicoletti G, Palladini A, Grosso V, Ranieri D, Dall’Ora M, Raschi E, Laranga R, 11. Hettmer S, Liu J, Miller CM, Lindsay MC, Sparks CA, Gambarotti M, Picci P, De Giovanni C, Nanni P, et al. Guertin DA, Bronson RT, Langenau DM, Wagers AJ. Genetic prevention of lymphoma in p53 knockout mice Sarcomas induced in discrete subsets of prospectively allows the early development of p53-related sarcomas. isolated skeletal muscle cells. Proc Natl Acad Sci U S A. Oncotarget. 2014; 5:11924–38. https://doi.org/10.18632/ 2011; 108:20002–7. oncotarget.2650. 12. Blum JM, Añó L, Li Z, Van Mater D, Bennett BD, 22. Ghahremani MF, Radaelli E, Haigh K, Bartunkova S, Sachdeva M, Lagutina I, Zhang M, Mito JK, Dodd LG, Haenebalcke L, Marine JC, Goossens S, Haigh JJ. Loss of Cardona DM, Dodd RD, Williams N, et al. Distinct and autocrine endothelial-derived VEGF significantly reduces Overlapping Sarcoma Subtypes Initiated from Muscle Stem development in conditional p53-deficient and Progenitor Cells. Cell Reports. 2013; 5:933–40. mice. Cell Cycle. 2014; 13:1501–7. 13. Young RJ, Brown NJ, Reed MW, Hughes D, Woll PJ. 23. Papa A, Cordon-Cardo C, Bernardi R, Pandolfi PP. Angiosarcoma. Lancet Oncol. 2010; 11:983–91. Compound In Vivo Inactivation of Pml and p53 14. Zietz C, Rössle M, Haas C, Sendelhofert A, Hirschmann A, Uncovers a Functional Interaction in Angiosarcoma Stürzl M, Löhrs U. MDM-2 Oncoprotein Overexpression, Suppression. Genes Cancer. 2012; 3:599–603. https://doi. p53 Gene Mutation, and VEGF Up-Regulation in org/10.1177/1947601912473604. Angiosarcomas. The American Journal of Pathology. 1998; 24. Yang J, Kantrow S, Sai J, Hawkins OE, Boothby M, 153:1425–33. Ayers GD, Young ED, Demicco EG, Lazar AJ, Lev D, www.oncotarget.com 19765 Oncotarget Richmond A. Ikk4a/Arf Inactivation with Activation of the 37. Medina D. Mammary tumorigenesis in chemical NF-κB/IL-6 Pathway Is Sufficient to Drive the Development carcinogen-treated mice. I. Incidence in BALB-c and and Growth of Angiosarcoma. Cancer Res. 2012; C57BL mice. J Natl Cancer Inst. 1974; 53:213–21. 72:4682–95. 38. Ullrich RL, Bowles ND, Satterfield LC, Davis CM. Strain- 25. Italiano A, Chen CL, Thomas R, Breen M, Bonnet F, dependent susceptibility to radiation-induced mammary Sevenet N, Longy M, Maki RG, Coindre JM, Antonescu CR. cancer is a result of differences in epithelial cell sensitivity Alterations of the p53 and PIK3CA/AKT/mTOR pathways in to transformation. Radiat Res. 1996; 146:353–5. angiosarcomas: a pattern distinct from other sarcomas with 39. Zhang S, Qian X, Redman C, Bliskovski V, Ramsay ES, complex genomics. Cancer. 2012; 118:5878–87. Lowy DR, Mock BA. p16INK4a gene promoter variation 26. Tate G, Suzuki T, Mitsuya T. Mutation of the PTEN gene and differential binding of a repressor, the ras-responsive in a human hepatic angiosarcoma. Cancer Genetics and zinc-finger transcription factor, RREB. Oncogene. 2003; Cytogenetics. 2007; 178:160–2. 22:2285–95. 40. Blackburn AC, Brown JS, Naber SP, Otis CN, Wood JT, 27. Leech JD, Lammers SH, Goldman S, Auricchio N, Jerry DJ. BALB/c alleles for Prkdc and Cdkn2a interact to Bronson RT, Kwiatkowski DJ, Sahin M. A Vascular Model modify tumor susceptibility in Trp53+/– mice. Cancer Res. of Tsc1 Deficiency Accelerates Renal Tumor Formation 2003; 63:2364–8. with Accompanying Hemangiosarcomas. Mol Cancer Res. 2015; 13:548–5. 41. Forde A, Constien R, Gröne HJ, Hämmerling G, Arnold B. Temporal Cre-mediated recombination exclusively in 28. Dill MT, Rothweiler S, Djonov V, Hlushchuk R, Tornillo L, endothelial cells using Tie2 regulatory elements. Genesis. Terracciano L, Meili Butz S, Radtke F, Heim MH, Semela D. 2002; 33:191–7. Disruption of Notch1 Induces Vascular Remodeling, 42. Rodriguez R, Rubio R, Menendez P. Modeling Intussusceptive Angiogenesis, and Angiosarcomas in Livers sarcomagenesis using multipotent mesenchymal stem cells. of Mice. Gastroenterology. 2012; 142:967–977.e2. Cell Research. 2011; 22:62–77. 29. Hall JM, Saenger JS, Fadare O. Diagnostic utility of P63 and 43. Mohseny AB, Hogendoorn PCW. Concise review: CD10 in distinguishing cutaneous spindle cell/sarcomatoid mesenchymal tumors: when stem cells go mad. Stem Cells. squamous cell carcinomas and atypical fibroxanthomas. Int J 2011; 29:397–403. Clin Exp Pathol. 2008; 1:524–30. 44. Rubio R, García-Castro J, Gutiérrez-Aranda I, Paramio J, 30. Goldblum JR. An approach to pleomorphic sarcomas: can Santos M, Catalina P, Leone PE, Menendez P, Rodriguez R. we subclassify, and does it matter? Mod Pathol. 2014; Deficiency in p53 but not Retinoblastoma Induces the 27:39–46. Transformation of Mesenchymal Stem Cells In vitro and 31. Armah HB, Rao UN, Parwani AV. Primary angiosarcoma of Initiates Leiomyosarcoma In vivo. Cancer Res. 2010; the testis: report of a rare entity and review of the literature. 70:4185–94. Diagn Pathol. 2007; 2:23. 45. Tolar J, Nauta AJ, Osborn MJ, Panoskaltsis Mortari A, 32. Kruse AJ, Sep S, Slangen BF, Vandevijver NM, Van Gorp McElmurry RT, Bell S, Xia L, Zhou N, Riddle M, T, Kruitwagen RF, Van de Vijver KK. Angiosarcomas of Schroeder TM, Westendorf JJ, McIvor RS, Hogendoorn primary gynecologic origin: a clinicopathologic review PCW, et al. Sarcoma derived from cultured mesenchymal and quantitative analysis of survival. Int J Gynecol Cancer. stem cells. Stem Cells. 2007; 25:371–9. 2014; 24:4–12. 46. Guarnerio J, Riccardi L, Taulli R, Maeda T, Wang G, Hobbs RM, Song MS, Sportoletti P, Bernardi R, Bronson RT, 33. Hackney JR, Palmer CA, Riley KO, Cure JK, Fathallah- Castillo-Martin M, Cordon-Cardo C, Lunardi A, et al. A Shaykh HM, Nabors LB. Primary central nervous system genetic platform to model sarcomagenesis from primary adult angiosarcoma: two case reports. J Med Case Rep. 2012; mesenchymal stem cells. Cancer Discov. 2015; 5:396–409. 6:251. 47. He Q, Wan C, Li G. Concise review: multipotent 34. Ren Y, Zhu M, Liu Y, Diao X, Zhang Y. Primary pulmonary mesenchymal stromal cells in blood. Stem Cells. 2007; angiosarcoma: Three case reports and literature review. 25:69–77. Thorac Cancer. 2016; 7:607–13. 48. Vichai V, Kirtikara K. Sulforhodamine B colorimetric 35. Al Beteddini OS, Brenez D, Firket C, Algaba R, Tabech A. assay for cytotoxicity screening. Nature Protocols. 2006; Colonic angiosarcoma: A case report and review of 1:1112–6. literature. Int J Surg Case Rep. 2013; 4:208–11. 49. Thoma CR, Frew IJ, Hoerner CR, Montani M, Moch H, 36. Sigmund CD. Viewpoint: are studies in genetically altered Krek W. pVHL and GSK3β are components of a primary mice out of control? Arterioscler Thromb Vasc Biol. 2000; cilium-maintenance signalling network. Nat Cell Biol. 20:1425–9. 2007; 9:588–95.

www.oncotarget.com 19766 Oncotarget