Abstract
Genomic and functional study of existing and emerging sarcoma targets, such as fusion proteins, chromosomal aberrations, reduced tumor suppressor activity, and oncogenic drivers, is broadening our understanding of sarcomagenesis. Among these mechanisms, the tumor suppressor p53 (TP53) plays significant roles in the suppression of bone and soft tissue sarcoma progression. Although mutations in TP53 were thought to be relatively low in sarcomas, modern techniques including whole-genome sequencing have recently illuminated unappreciated alterations in TP53 in osteosarcoma. In addition, oncogenic gain-of-function activities of missense mutant p53 (mutp53) have been reported in sarcomas. Moreover, new targeting strategies for TP53 have been discovered: restoration of wild-type p53 (wtp53) activity through inhibition of TP53 negative regulators, reactivation of the wtp53 activity from mutp53, depletion of mutp53, and targeting of vulnerabilities in cells with TP53 deletions or mutations. These discoveries enable development of novel therapeutic strategies for therapy-resistant sarcomas. We have outlined nine bone and soft tissue sarcomas for which TP53 plays a crucial tumor suppressive role. These include osteosarcoma, Ewing sarcoma, chondrosarcoma, rhabdomyosarcoma (RMS), leiomyosarcoma (LMS), synovial sarcoma, liposarcoma (LPS), angiosarcoma, and undifferentiated pleomorphic sarcoma (UPS).
Keywords: TP53, mutation, MDM2, bone, sarcoma, targeted therapy
1. Introduction
Bone and soft tissue sarcomas are tumors of mesenchymal origin. Sarcomas are exceptionally rare tumors, comprising less than 1% of all human cancers. Most sarcomas arise from abnormal differentiation processes of mesenchymal stem cells (MSCs) and their derived cell lineages (Eid & Garcia, 2015, Lye, Nordin, Vidyadaran, & Thilakavathy, 2016). Sarcomas commonly occur in children and young adults (Ognjanovic, Olivier, Bergemann, & Hainaut, 2012). Sarcoma prognosis remains poor, especially in cases of distant metastasis (Meyer & Seetharam, 2019). Although advanced chemotherapy and surgery have improved patients’ prognosis, treatment options for most sarcomas have remained stagnant for the past 30–40 years (Meyer & Seetharam, 2019, Scheidt, et al., 2019). Therefore, understanding the molecular mechanisms driving sarcoma malignancy would significantly accelerate development of novel therapeutic strategies.
Advances in molecular biology have enabled precise and detailed discovery in the genome and expression profiles of tumor tissues, as compared with those of non-tumor tissues. Several unappreciated mutations and chromosomal abnormalities are found in osteosarcoma, including genes associated with IGF signaling, alterations in the PI3K/mTOR pathway, and somatic structural variations in TP53 (Behjati, et al., 2017, X. Chen, et al., 2014, Perry, et al., 2014, Sayles, et al., 2019). Genome sequencing of Ewing sarcoma reveals mutations related to DNA repair machineries (Brohl, et al., 2017). Indeed, Anderson et al. (Anderson, et al., 2018) show that complex chromosome rearrangements at the early stage of Ewing sarcoma development may contribute to EWS-ETS gene fusions and other DNA rearrangements. Additionally, multi-platform molecular landscape analysis of 206 adult soft tissue sarcomas, including leiomyosarcoma (LMS), de-differentiated liposarcoma (LPS), undifferentiated pleomorphic sarcoma (UPS), and synovial sarcoma, has recently revealed previously unappreciated sarcoma-type-specific changes in copy number variation, methylation pattern, RNA expression profile, and protein expression, which could have an impact on sarcoma diagnosis and therapy (Cancer Genome Atlas Research Network. Electronic address & Cancer Genome Atlas Research, 2017). These findings have further confirmed the significance of mutations and genetic abnormalities in the TP53 gene on bone and soft tissue sarcoma progression. Mutations in TP53 are detected in about half of all tumors, remaining the most frequently mutated gene in human cancers (Hung & Anderson, 1997, Taubert, Meye, & Wurl, 1998, Zhou, Hao, & Lu, 2018). Previously, mutations in TP53 were thought to occur at a relatively low frequency in sarcomas (Toguchida, et al., 1992). This is mainly because mutations in TP53 were identified by sequencing only exonic regions in the DNA binding domain or by performing immunohistochemistry (IHC) to detect positive staining in p53-mutated tumors due to the long half-life of the mutant protein. However, recent whole-genome sequencing analyses have revealed more frequent alterations in TP53, including structural alterations in TP53 intron 1. Thus, the significance of TP53 in sarcoma could have been underestimated, which drives us to revisit the roles of TP53 in sarcomagenesis. Since the variety of alterations to TP53 and TP53 upstream regulators are detected in sarcomas, we will also discuss several distinct TP53 targeting strategies in this review article (Table 1).
Table 1.
Strategy targeting TP53
TP53 status | Targeting Strategy |
---|---|
Wild-type TP53 (reduced activity) |
Restoration of wild-type TP53 activity MDM2 antagonists (e.g., Nutlin-3a, RG7112, RG7388), MDM4 inhibitors, MDM2/4 dual inhibitors, RITA |
TP53-null |
Target vulnerabilities in cells lacking wild-type TP53 activity Inhibitors for wee1 kinase, Chk1, and polo-like kinase 1 |
Mutant TP53 |
Target vulnerabilities in cells lacking wild-type TP53 activity Inhibitors for wee1 kinase, Chk1, and polo-like kinase 1 Reactivation of wild-type TP53 activity from mutant TP53 e.g. PRIMA-1, PRIMA-1Met, CP-31398, STIMA-1, Stictic acid Mutant TP53 depletion Statins, HSP90 inhibitors |
TP53 is a transcription factor that is stabilized following genotoxic stress and induces transcription of genes associated with cell cycle arrest, apoptosis, and metabolism, thereby functioning as a tumor suppressor (D. Lane & Levine, 2010, Ranjan & Iwakuma, 2016). Typically, tumor suppressors have loss-of-function (LOF) mutations or deletions in cancers; however, the majority of TP53 mutations are missense mutations in the DNA binding domain, such that mutant TP53 (mutp53) not only loses the tumor suppressive function but also gains oncogenic functions (GOF: gain of function) independent of wild-type TP53 (wtp53) (Parrales & Iwakuma, 2015). The most frequently mutated codons are R175, R248, and R273. There are roughly two types of TP53 mutations. The first is a DNA contact type in which mutations occur at amino acids that directly interact with DNA (e.g., K120, R248, R273, R280). The second is a conformational or misfolded type, in which mutations do not occur in DNA-contacting amino acids but instead alter the three-dimensional structure of TP53, thereby losing the DNA binding activity (Adhikari & Iwakuma, 2009, D. Lane & Levine, 2010). Increasing evidence suggests that not all TP53 mutants have identical GOF activity. However, overall, the majority of TP53 mutants lose tumor suppressive function, at least partially, and some mutants show distinct oncogenic activities to promote malignant progression (M. P. Kim & Lozano, 2018, Mantovani, Collavin, & Del Sal, 2019, Parrales & Iwakuma, 2015). These mutants’ oncogenic properties are seen throughout a variety of cancers including bone and soft tissue sarcomas.
Mutp53’s oncogenic properties are also demonstrated in Li-Fraumeni syndrome (LFS), an autosomal dominant familial cancer-prone syndrome. The link between germline mutations of TP53 and LFS was confirmed by molecular testing in 1990 (Malkin, et al., 1990). Over 70% of patients with LFS inherently harbor a mutation in the TP53 gene in their germlines and frequently develop various types of cancer at early ages, including osteosarcoma, rhabdomyosarcoma (RMS), brain tumors, breast cancer, leukemias, and adrenocortical carcinoma (Correa, 2016, Malkin, et al., 1990). Osteosarcoma is the most common sarcoma in patients with LFS, though rates are only slightly higher for osteosarcoma than brain tumors and RMS (Correa, 2016, Guha & Malkin, 2017). Even a low-penetrance TP53 R337H mutation, found in a clustered population in southeast Brazil, is associated with an increased frequency of adrenocortical tumors, choroid plexus carcinoma, and osteosarcoma (Seidinger, et al., 2011). Additionally, Mirabello et al. (Mirabello, Yeager, et al., 2015) report that young-onset osteosarcoma has a higher frequency of LFS-associated TP53 mutations. Moreover, they show that the presence of a rare TP53 variant leading to an exonic splice site change, rs1800372 (p.R213R), is associated with metastasis at diagnosis of osteosarcoma (Mirabello, Yeager, et al., 2015).
In this review article, we revisit the significance of TP53 in bone and soft tissue sarcoma with updated literatures by focusing on osteosarcoma, Ewing sarcoma, chondrosarcoma, RMS, LMS, synovial sarcoma, LPS, angiosarcoma, and UPS. For each sarcoma we explore the roles of TP53 in tumor development, malignancy, and the prognosis of these sarcomas, as well as potential and optimal therapeutic strategies targeting TP53 for each sarcoma (Table 2).
Table 2.
TP53 mutation rate in sarcomas and suitable targeting strategies
Sarcoma | Overall TP53 mutation rate | Notable TP53/MDM2 alterations | Suitable TP53 targeting strategies |
---|---|---|---|
Osteosarcoma | ~80% | 1) TP53 intron 1 rearrangements, 2) MDM2/MDM4 gene amplification | 1) MDM2/MDM4 antagonists to restore wild-type TP53 activity, 2) Targeting vulnerability imposed by TP53 deletion/mutations |
Ewing sarcoma | ~10% | 1) Inhibition of wild type TP53 by EWS-FLI fusion protein | 1) Restoring wild-type TP53 activity |
Chondrosarcoma | ~20% | 1) MDM2 gene amplification, 2) Alterations in the TP53 pathway in >50% of cases | 1) Restoring wild-type TP53 activity by MDM2 antagonists, 2) Targeting vulnerabilities imposed byTP53 deletion/mutations |
Rhabdomyosarcoma | < 15% | 1) Reduced wild-type TP53 activity and expression by PAX-FHKR and PAX proteins, 2) TP53 deletions/mutations in E2 subtype of ERMS | 1) Restoring wild-type TP53 activity, 2) Targeting vulnerabilities imposed by TP53 deletion/mutations, reactivating wild-type TP53 deletions/mutations in activity from mutant TP53, or depleting mutant TP53 (ERMS) |
Leiomyosarcoma | ~50% | 1) TP53 missense mutations | 1) Targeting vulnerabilities imposed by TP53 mutations, reactivating wild-type TP53 activity from mutant TP53, or depleting mutant TP53 |
Synovial sarcoma | ~5% | 1) Inhibition of wild-type TP53 by SS18-SSX and MDM2/MDM4 gene amplification | 1) Restoring wild-type TP53 activity by MDM2/MDM4 antagonists |
Liposarcoma | 10–20% | 1) MDM2 gene amplification, 2) TP53 missense mutations in pleomorphic LPS | 1) Restoring wild-type TP53 activity by MDM2 antagonists, 2) Targeting vulnerabilities imposed by TP53 deletion/mutations, reactivating wild-type TP53 activity from mutant TP53, or depleting mutant TP53 (Pleomorphic LPS) |
Angiosarcoma | < 50% | 1) TP53 missense mutations, 2) MDM2 upregulation | 1) Targeting vulnerabilities imposed by TP53 deletion/mutations, reactivating wild-type TP53 activity from mutant TP53, or depleting mutant TP53, 2) Restoring wild-type TP53 activity by MDM2 antagonists |
Undifferentiated pleomorphic sarcoma | ~30% | 1) Retaining one wild-type TP53 allele in > 85% or p14ARF deletion/silencing, 2) TP53 missense mutations | 1) Restoring wild-type TP53 activity by MDM2 antagonists, 2) Targeting vulnerabilities imposed by TP53 deletion/mutations, reactivating wild-type TP53 activity from mutant TP53, or depleting mutant TP53 |
2. Osteosarcoma
Osteosarcoma is the most common primary cancer of the bone and arises during the process of osteoblastic differentiation from MSCs (Mortus, Zhang, & Hughes, 2014, Tang, Song, Luo, Haydon, & He, 2008). Osteosarcoma is commonly detected near the epiphyseal plate of long bones, most commonly in children. A combination of surgery and chemotherapy, including high-dose methotrexate, cisplatin, and doxorubicin, is the main therapeutic strategy for osteosarcoma. Although this treatment has improved 5-year survival rate of patients to 70% in the past three decades, some patients develop resistance to chemotherapy, and the long-term survival of patients with relapse remains below 20% (Harrison, Geller, Gill, Lewis, & Gorlick, 2018, Y. Zhang, J. Yang, et al., 2018). To overcome this poor prognosis, it is crucial to learn about the genetics and molecular pathogenesis of this disease.
Osteosarcomas frequently show complex karyotypes and genomic instability including gene amplifications (MYC, CCNE, Rad21, VEGFQ, AURKB, CDK4), deletions (TP53, RB1, PTEN), somatic nucleotide variants (SNVs) or short indels (TP53, ATRX, RBs, PRKDC), and structural variants (SVs: TP53, LRP1B, RB1, FHIT) (Martin, Squire, & Zielenska, 2012, Sayles, et al., 2019). While many other sarcomas are often characterized by chromosomal translocations (including Ewing sarcoma, RMS, synovial sarcoma, and LPS), until now no such representative translocation has been identified in osteosarcoma (Martin, et al., 2012). Mutation of TP53 is well-correlated with genomic and chromosomal instability in human high-grade osteosarcoma (Al-Romaih, et al., 2003, Zuffa, et al., 2008). One early immunohistochemistry (IHC) study shows that 27 out of 46 (58.7%) osteosarcoma specimens exhibit TP53 overexpression, a common indicator of TP53 missense mutations (W. Guo, Wang, & Feng, 1996), while other studies show fewer TP53 mutations in osteosarcoma (Mendoza, Konishi, Dernell, Withrow, & Miller, 1998, Miller, et al., 1996, Mirabello, Koster, et al., 2015). Intriguingly, Masuda et al. (Masuda, Miller, Koeffler, Battifora, & Cline, 1987) first reported intron 1 rearrangements in the TP53 gene in 3 out of 6 human osteosarcoma samples in 1987. Indeed, a recent whole-genome sequencing of 34 osteosarcoma samples has discovered similar mutations in the TP53 gene. Nine (26%) have TP53 point mutations, and 19 (55%) have structural variations in the TP53 gene, the majority of which are translocations with breakpoints in the first intron (X. Chen, et al., 2014). This finding is supported by the report by Ribi et al. (Ribi, et al., 2015), where 16% of sporadic osteosarcomas show intron 1 rearrangements, while no other tumors have such TP53 rearrangements. The intron 1 rearrangements are also detected in a four-generation LFS family (Ribi, et al., 2015). The most recent study by Sayles et al. (Sayles, et al., 2019) demonstrates that TP53 alterations including structural variation (SV) and somatic nucleotide variants (SNVs) are detected in 74% of human osteosarcoma. Since rearrangements in intron 1 cannot be detected by exon sequencing and IHC, alterations in TP53 have likely been underestimated. Biologically, loss of TP53 activity is shown to promote osteogenic differentiation of bone marrow stromal cells, as well as osteosarcoma development from MSCs (He, et al., 2015, Velletri, et al., 2016). Thus, TP53 has strong impact on preventing the malignant transformation of MSCs.
Genetic deletion or mutations of TP53 in mice with the C57BL/6 background often results in osteosarcoma development (Lang, et al., 2004). Also, mice expressing gain-of-function (GOF) TP53R172H (R172H is equivalent to human R175H) spontaneously develop metastatic tumors, including osteosarcoma at a high frequency (~30% in the C57BL/6 background) (Lang, et al., 2004, Olive, et al., 2004). This metastatic GOF activity by mutp53 can be mediated by upregulation of the ONZIN-CXCL5-MAPK axis or by binding of mutp53 with TP63, TP73, and ETS2 (Do, et al., 2012, Lang, et al., 2004, Pourebrahim, et al., 2017, Xiong, et al., 2014, Y. Zhang, Q. Hu, et al., 2018). Moreover, combined deletion of TP53 and RB1 in mouse osteoblasts results in development of metastatic osteosarcoma at a high frequency (Walkley, et al., 2008). Thus, deletions and mutations in TP53 significantly contribute to osteosarcoma progression in mouse models, supporting findings in humans.
TP53 has been implicated in chemotherapy sensitivity, mainly through induction of apoptosis and senescence as well as inhibition of autophagy (J. Fan & Bertino, 1999, Hu, et al., 2017, Z. Wang & Sun, 2010). Previous studies have shown that mutations in TP53 are associated with chemoresistance or poor event-free survival in human osteosarcoma (Goto, et al., 1998, Tsuchiya, et al., 2000). Loss of heterozygosity (LOH) of TP53 in osteosarcomas is associated with chemoresistance (Goto, et al., 1998). The work by Asada et al. (Asada, Tsuchiya, & Tomita, 1999) supports this, showing that deletion of TP53 is found in an osteosarcoma cell line with acquired resistance to cisplatin. However, multiple meta-analyses suggest that while TP53 mutations could serve as an effective prognostic marker only for 2- or 3-year overall survival of patients with osteosarcoma, TP53 status does not appear to be correlated with development of metastases and chemotherapy response in patients with osteosarcoma (Z. Chen, Guo, Zhang, & Guo, 2016, Fu, et al., 2013, Gokgoz, et al., 2001, Pakos, Kyzas, & Ioannidis, 2004, Wunder, et al., 2005, Yao, et al., 2014). These clinical studies do not examine the structural alterations in TP53 introns. Considering frequent observations of the TP53 intron 1 rearrangement, it may be important to re-evaluate the clinical significance of TP53 alterations in malignancy, response to chemotherapy, and metastasis of human osteosarcoma and other types of cancer.
Alterations to regulators of TP53 can also contribute to development of osteosarcoma (Morrow & Khanna, 2015). One crucial regulator of TP53 stability and degradation is the protein MDM2 (murine double minute 2). MDM2, which acts as the major E3 ubiquitin ligase for TP53, is overexpressed in ~30% of human cancers. MDM2 copy number is amplified more than 3-fold in 14.7% of high grade osteosarcoma (Overholtzer, et al., 2003). Ito et al. (Ito, et al., 2011) show that 35% of osteosarcoma cases have over 3-fold MDM2 amplification. However, mutations in TP53, but not copy number of MDM2, are correlated with overall genomic instability in high-grade human osteosarcoma (Overholtzer, et al., 2003). Although a multivariate analysis shows that the MDM2 polymorphism T309G which increases MDM2 expression levels via an extra SP1 binding site in the MDM2 promoter is associated with an increased risk of developing high-grade osteosarcomas in female patients (Toffoli, et al., 2009), there is no conclusive literature showing correlation of MDM2 overexpression with survival and prognosis of human osteosarcoma.
Another major inhibitor of TP53 is MDM4 (Haupt, Hernandez, Vijayakumaran, Keam, & Haupt, 2019). Although it is a homolog of MDM2, MDM4 does not have ubiquitin ligase activity like MDM2. However, MDM4 still binds with TP53 and inhibits TP53 activity. Also, MDM4 hetero-dimerizes with MDM2 through the C-terminal RING finger domain to induce TP53 degradation as a cofactor of MDM2 (Haupt, et al., 2019). Similar to MDM2, 35% of osteosarcoma cases show MDM4 amplification over 3-fold (Duhamel, et al., 2012, Ito, et al., 2011). In addition, multiple studies have highlighted the expression of MDM4 splice variants in different tumor types (Bardot & Toledo, 2017). One MDM4 splice variant, MDM4-S, which skips exon 6 and prematurely terminates in exon 7, encodes a truncated MDM4 protein carrying only the N-terminal TP53-binding domain along with 13 novel amino acids (Bardot & Toledo, 2017, Pant, et al., 2017). Overexpression of MDM4-S has been linked to poor prognosis in osteosarcoma, soft tissue sarcoma, and other cancers (Dewaele, et al., 2016, Lenos, et al., 2012, Lenos & Jochemsen, 2011). Thus, MDM4-S overexpression can serve as an effective biomarker for TP53 pathway attenuation in cancers (Lenos, et al., 2012).
Pagetic osteosarcoma and TP53
Paget’s disease of bone (PDB), which occurs in approximately 3% of adults over 50 years old, is a metabolic bone disease characterized by increased bone resorption and subsequent excessive disorganized bone formation (Ouslander & Beck, 1982). PDB occurs rarely in Asian and African regions, while individuals in Europe, North America, and Australia show a high prevalence rate of PDB (Gennari, Rendina, Falchetti, & Merlotti, 2019, Gruener & Camacho, 2014). One of the genes implicated in PDB is Sqstm1/p62, which plays roles in NF-κB activation as a scaffolding/adaptor protein and selective macroautophagy as an autophagosome cargo protein (Duran, et al., 2008, L. Fan, Yin, Zhang, & Hu, 2018, McManus & Roux, 2012). Patients with PDB have a high incidence of osteosarcoma, called Pagetic osteosarcoma. The prevalence of Pagetic osteosarcoma is estimated to be about 1%, which is several hundred- to thousand-fold higher than the prevalence of osteosarcoma in the general population depending on the age (Greditzer, McLeod, Unni, & Beabout, 1983, Hansen, Nellissery, & Bhatia, 1999, Mirabello, Troisi, & Savage, 2009). Pagetic osteosarcoma accounts for 50% of osteosarcoma cases in patients over 60 years of age (Gennari, et al., 2019, Yochum, 1984). Although MYC gene amplification and TP53 mutations are detected in Pagetic osteosarcomas and loss of TP53 activity is reported to reduce Sqstm1/p62 levels, the direct role of TP53 in Pagetic osteosarcoma development remains unclear (Goiran, et al., 2018, Reddy, 2004, Ueda, Healey, Huvos, & Ladanyi, 1997).
TP53-targeted therapy in osteosarcoma
Because TP53 alterations have been underestimated in osteosarcoma, most early studies in osteosarcoma focus on restoring wtp53 activity by inhibiting a TP53 negative regulator, MDM2 (Table 1) (Hientz, Mohr, Bhakta-Guha, & Efferth, 2017). The first report for an MDM2 inhibitor is Nutlin-3a (Vassilev, et al., 2004). Nutlin-3a binds MDM2 in the TP53-binding pocket and activates TP53 without causing DNA damage in cancer cells, leading to cell cycle arrest, apoptosis, and growth inhibition in osteosarcoma xenograft mouse models (Tovar, et al., 2006, Vassilev, et al., 2004, B. Wang, Fang, Zhao, Xiang, & Wang, 2012). Nutlin-3a shows synergy with other chemotherapy drugs and CDK inhibitors, and oridonin, a diterpenoid extracted from medicinal herbs (Cheok, Dey, & Lane, 2007, X. H. Wang, Zhang, Bao, & Liu, 2017). Unfortunately, clinical trials for Nutlin-3a in various tumor types have yet to show much success due to significant side-effects including bone marrow suppression (Ray-Coquard, et al., 2012). Several other compounds that inhibit MDM2-TP53 binding have been identified, including RG7112 (Tovar, et al., 2013), SAR405838 (S. Wang, et al., 2014), APG-115 (Yi, et al., 2018), AMG 232 (D. Sun, et al., 2014), and MK-8242 (Wagner, et al., 2017). Some of these MDM2 inhibitors are currently in clinical trials (D. P. Lane, Brown, Verma, & Cheok, 2011, Morrow & Khanna, 2015, Tovar, et al., 2013, Wagner, et al., 2017, S. Wang, et al., 2014). Besides MDM2 inhibitors, investigators have discovered MDM4 inhibitors (e.g., CTX1, K-181, SJ-172550), as well as MDM2/MDM4 dual inhibitors (e.g., DIMP53–1, pDI analogs) (Bista, et al., 2012, Karan, et al., 2016, Philippe, et al., 2016, Soares, et al., 2017, Uesato, et al., 2016). The effects of these inhibitors on osteosarcoma need to be tested in the future.
When tumors carry missense mutp53, restoration of wtp53 activity from mutp53 can be an efficient strategy for anti-cancer therapy (Table 1) (Parrales & Iwakuma, 2015). PRIMA-1 is the first small molecule that is shown to restore sequence-specific DNA binding and transcriptional activities of TP53 with tumor suppressive effects in mutp53-expressing cells (V. J. Bykov, Issaeva, et al., 2005). Since then, many compounds have been identified, and details of these compounds are summarized elsewhere (Binayke, Mishra, Suman, Das, & Chander, 2019, V. J. N. Bykov, Eriksson, Bianchi, & Wiman, 2018, Parrales & Iwakuma, 2015). Of these, APR-246, a PRIMA-1 analog, shows synergy in inhibiting tumor cell growth with camptothecin, a quinoline alkaloid, in the Saos2 osteosarcoma cell line exogenously expressing TP53 R273H (Saos2-TP53R273H) (V. J. Bykov, Zache, et al., 2005). STIMA-1, a small molecule compound with structural similarities to CP-31398 that stabilizes the active conformation of TP53 (Wischhusen, Naumann, Ohgaki, Rastinejad, & Weller, 2003), is also shown to stimulate DNA binding of mutp53, induce expression of TP53 target genes, and trigger apoptosis in H1299-TP53R175H lung adenocarcinoma and Saos2-TP53R273H osteosarcoma cells (Zache, et al., 2008). Additionally, stictic acid, which was identified though computational methods for a transiently open binding pocket in the TP53 core domain, is shown to upregulate p21 and PUMA in Saos2 cells expressing TP53 R175H and G245S (Wassman, et al., 2013).
Other than these compounds to increase wtp53 activity, drugs that can deplete mutp53 can also be used to inhibit tumor growth of mutp53-expressing osteosarcoma. This is based on the observations that cancer cells are addicted to oncogenes, including mutp53, and that depletion of mutp53 may lead to reactivation of proteins or pathways suppressed by mutp53 (Iyer, et al., 2016, Parrales & Iwakuma, 2015). HSP90 inhibitors and cholesterol-lowering drugs (statins) are agents have been shown to induce degradation of mutp53 mediated by MDM2 and/or CHIP ubiquitin ligases and inhibit tumor progression including osteosarcoma (Alexandrova, et al., 2015, Parrales, et al., 2016).
Another way to target TP53 mutations is to treat cells with a compound that inhibits proteins or pathways unique and essential for survival and proliferation (vulnerabilities) in TP53-null or mutated cells with minimal impact on wtp53-expressing cells (Table 1) (Lu, et al., 2016, Tongyang, et al., 2015). Such a compound may inhibit function of proteins that show a synthetically lethal interaction with TP53 mutations, including CHK1, ATM/CHK2, Plk1, Wee1, and MK2 (Chung, et al., 2018, Gurpinar & Vousden, 2015, Harada, et al., 2011, Jiang, et al., 2009, Morandell, et al., 2013, Origanti, Cai, Munir, White, & Piwnica-Worms, 2013, Tongyang, et al., 2015, X. Wang & Simon, 2013, Weidle, Maisel, & Eick, 2011). Since ~80% of osteosarcoma cases have TP53 alterations, targeting vulnerable pathways and proteins in cells lacking wtp53 activity would be a reasonable treatment strategy. Intriguingly, Yu et al. (Yu, et al., 2015) identify HDAC (histone deacetylase) inhibitors through a 54 FDA-approved agent screen as agents having anti-growth activity and synergistic effects with proteasome inhibitors in five TP53-inactive pediatric osteosarcoma cell lines, although the underlying mechanism remains unclear.
3. Ewing sarcoma
Ewing sarcoma accounts for approximately 1% of childhood cancers and is the second most common bone cancer in children, although it can also arise in soft tissues (Grunewald, et al., 2018). The femur is the most frequently affected bone. The origin of Ewing sarcoma remains controversial as both MSCs and neural crest stem cells (NCSCs) have been implicated as the cell of origin (Lin, Wang, & Lozano, 2011, Todorova, 2014). Microscopically, the tumor consists of small round cells with regular round nuclei and a narrow rim of clear or faintly eosinophilic cytoplasm (Burchill, 2003).
The current chemotherapy protocols for Ewing sarcoma include combinations of doxorubicin, cyclophosphamide, vincristine, actinomycin-D, ifosfamide, and etoposide (Ozaki, 2015). These treatments have improved the overall 5-year survival of localized Ewing sarcoma to 70%. However, overall survival of metastatic Ewing sarcoma remains stagnant at ~30% (Balamuth & Womer, 2010, Kridis, et al., 2017).
This malignancy is characterized by translocation between a gene of the RNA-binding TET family (e.g., EWSR1, FUS) and a gene of the ETS-transcription family (e.g., FLI1, ERG, ETV1, ETV4, and FEV). In the majority of cases (~85%), the EWS-FLI1 fusion resulting from a t(11;22) translocation is detected (Renzi, Anderson, Light, & Gupta, 2018). However, it remains unclear why this fusion occurs and how exactly the fusion protein contributes to Ewing sarcoma development. A recent study by Anderson et al. (Anderson, et al., 2018) using whole-genome sequence analyses of 124 Ewing sarcomas indicates that chromoplexy, a sudden burst of loop-like genomic rearrangements involving multiple chromosomes and genes, rather than reciprocal translocation, is the potential cause of the EWSR1–ETS fusion in 42% of cases. The loop-like genomic rearrangements and fusions are also detected in other sarcomas including chondromyxoid fibroma and synovial sarcoma (Anderson, et al., 2018).
Molecular and genetic profiles of Ewing sarcoma have been assessed in a genome-wide association study of 733 Ewing sarcoma cases and 1,346 unaffected individuals of European ancestry by Crompton et al. (Crompton, et al., 2014). They reveal new susceptibility loci at 6p25.1, 20p11.22, and 20p11.23, in addition to previously reported loci at 1p36.22, 10q21.3, and 15q15.1. They identify candidate genes at 6p25.1 (RREB1) and 20p11.23 (KIZ) (Crompton, et al., 2014). Brohl et al. (Brohl, et al., 2014) also show that Ewing sarcoma has frequent mutations in the cohesin complex subunit STAG2 (21.5%), homozygous deletion of CDKN2A (13.8%) and mutations of TP53 (6.2%), as well as an increased prevalence of the BRCA2 K3326X polymorphism (7.3%). Other genome sequencing studies also identify mutations in cancer-related genes including KDR, STK11, MLH1, KRAS, and PTPN11, as well as in DNA double-strand break repair, in Ewing sarcoma tissues (Brohl, et al., 2017, Zhang, et al., 2016).
With regard to TP53 mutations, multiple articles have found that genetic alterations in the TP53 gene are detected only in ~10% of Ewing sarcomas (Komuro, et al., 1993, Lerman, et al., 2015, Neilsen, Pishas, Callen, & Thomas, 2011, Radig, et al., 1998). The two most frequently detected TP53 mutations in Ewing sarcoma are the C176F and R273X (Sand, Szuhai, & Hogendoorn, 2015). Although mutation frequency of TP53 is low in Ewing sarcoma, TP53 still plays a crucial role in inhibiting Ewing sarcoma progression. Li et al (Y. Li, et al., 2010) report that EWS-FLI1 binds to and inhibits TP53’s transcriptional activity, which may explain why TP53 mutations are detected only in ~10% of Ewing sarcoma. To support this finding, silencing of EWS-FLI1 reactivates NOTCH signaling and subsequently activates TP53 to induce cell cycle arrest in Ewing sarcoma cells (Ban, et al., 2008). Additionally, EWS-FLI1 significantly inhibits p300-mediated acetylation of TP53 at Lys-382 (Y. Li, et al., 2012). These results strongly suggest the inhibitory role of EWS-FLI1 in TP53 activity as well as the vital tumor inhibitory role of TP53 in Ewing sarcoma progression. However, a study by Lessenick et al. (Lessnick, Dacwag, & Golub, 2002) shows that EWS-FLI1 induces a TP53-dependent growth arrest in primary human fibroblasts, which is rescued by wtp53 inhibition. The observed discrepancy may be due to difference in the experimental settings including cancer cells vs non-transformed cells.
Several clinical studies also support the important role of TP53 in inhibiting Ewing sarcoma progression. An IHC study by de Alava et al. (de Alava, et al., 2000) shows that positive TP53 staining in Ewing’s sarcoma is a poor prognostic factor. This finding is supported by studies by Huang et al. (H. Y. Huang, et al., 2005) and Abdu et al. (Abudu, et al., 1999), demonstrating significant correlation of TP53 mutations with poor overall survival and response to chemotherapy. On the other hand, a study by Lerman et al. (Lerman, et al., 2015) fails to observe an association between TP53 mutations and event-free survival of patients. The discrepancy among these studies may be caused by the methodology used to determine TP53 mutations. Whole-genome sequencing, rather than IHC and exome sequencing, would be necessary to clarify the significance of TP53 mutations in Ewing sarcoma.
Similar to osteosarcoma, MDM2 overexpression is not high (~10%) (Kovar, et al., 1993). Ladanyi et al. (Ladanyi, et al., 1995) report that only three out of 30 Ewing sarcoma specimens have MDM2 gene amplification, all of which have metastasis at diagnosis, while only one out of 15 specimens without MDM2 amplification has metastasis, suggesting correlation between MDM2 amplification and metastasis. Thus, the TP53 pathway may play a crucial role in the suppression of Ewing sarcoma progression.
Given the infrequent rate of mutations of TP53 in Ewing sarcoma, restoring TP53 activity by inhibiting upstream inhibitors of TP53 is a rational therapeutic strategy (Neilsen, et al., 2011, Stolte, et al., 2018). Through a genome-scale CRISPR-Cas9 screening, Stolte et al. (Stolte, et al., 2018) identify MDM2, MDM4, USP7, and PPM1D as druggable targets for Ewing sarcoma. They also demonstrate that ATSP-7041, a stapled peptide inhibitor of MDM2 and MDM4, shows anti-tumor efficacy in vitro and in mouse models. Additionally, P5091, a USP7 inhibitor, and GSK2830371, a Wip1/PPM1D inhibitor, decrease viability of Ewing sarcoma cells, in a TP53-dependent manner (Stolte, et al., 2018). Pishas et al. (Pishas, et al., 2011) show that Nutlin-3a induces cell death, mainly apoptosis, in Ewing sarcoma cells expressing wtp53, and Nutlin-3a’s effect is synergistic with the chemotherapeutic agents: vincristine, actinomycin D, doxorubicin, and etoposide. Similarly, Sonnemann et al. (Sonnemann, et al., 2011) observe that nutlin-3a induces apoptosis and senescence with increased TP53 level and mRNA expression of TP53 downstream target genes in wtp53-expressing Ewing sarcoma cells (Sonnemann, et al., 2011). Also, YK-4–279, a small molecule inhibitor of EWSR1–ETS’s transcriptional activity through inhibition of its interaction with RNA helicase A, is shown to reduce Ewing sarcoma development regardless of TP53 status. The effects of YK-4–279 are additive with Nutlin-3a, in vitro and in a xenograft zebrafish model of human Ewing sarcoma cell lines (van der Ent, et al., 2014). Another way to restore wtp53 activity in Ewing sarcoma is to inhibit CD99. CD99 is expressed in most cases of Ewing sarcoma and is required to maintain malignancy (Pasello, Manara, & Scotlandi, 2018, Ventura, et al., 2016). Guerzoni et al. (Guerzoni, et al., 2015) find that dAbd C7, a novel human monospecific bivalent single-chain fragment variable diabody against CD99, induces cell death in Ewing sarcoma cells, through MDM2 degradation and TP53 reactivation; the anti-CD99 dAbd C7 shows additive effects with doxorubicin to suppress Ewing sarcoma malignancy.
When Ewing sarcoma expresses mutp53, APR-246 can be used to induce apoptosis by reactivating wtp53 activity in tumors. APR-246 is currently in early-phase clinical trials for Ewing sarcomas (Aryee, et al., 2013). Intriguingly, curcumin may radiosensitize TP53-mutated Ewing sarcoma cells by upregulating p21 and Bax and downregulating BCL-XL and MCL1, although the mechanism behind reactivation of wtp53 from mutp53 remains unclear (Veeraraghavan, Natarajan, Herman, & Aravindan, 2010). Another small molecule TP53 reactivator, RITA/NSC652287, is shown to induce apoptosis and effectively reduce tumor growth of Ewing sarcoma cell lines; however, this effect of RITA is independent of the TP53 status. As an additional mechanism of action of RITA in Ewing sarcoma cells, RITA induces degradation of IGF-1R, a regulator of anchorage-independent growth, in a manner dependent on MDM2 (Di Conza, et al., 2012). More studies are required for evaluating efficacy of targeting the TP53 pathway for Ewing sarcoma therapy.
4. Chondrosarcoma
Chondrosarcoma is a heterogeneous group of bone malignancies with cartilage-forming tumor cells, and it is the second most common form of primary malignant bone tumor (Chow, 2018, M. J. Kim, Cho, Ayala, & Ro, 2011). Chondrosarcoma generally grows slowly and rarely metastasizes, and the prognosis of surgically resected chondrosarcoma is good with a ~50% 5-year survival rate (M. J. Kim, et al., 2011). However, chondrosarcoma is generally chemo- and radiotherapy resistant, likely due to a low percentage of dividing cells and poor tumor vascularization. Because surgical resection is the primary treatment for chondrosarcoma, more efficient treatment options for metastatic and recurrent disease are required (Gelderblom, et al., 2008).
Genetically, chondrosarcoma can be subtyped to chromosomal translocation-positive and translocation-negative types. Extraskeletal myxoid chondrosarcoma, a slow-growing, low-grade sarcoma with high frequency of metastases and local recurrence, has a t(9;22)(q22;q12) translocation, creating the EWSR1-NR4A3, RBP56-NR4A3, and TCF12-NR4A3 fusion genes, while mesenchymal chondrosarcoma, a rare, fast-growing, high-grade sarcoma, has a HEY1-NCOA2 fusion gene resulting from intrachromosomal rearrangement of 8q21.13 and 8q13.3 or a IRF2BP2-CDX1 fusion gene resulting from t(1;5)(q42;q32) (Chow, 2018, Nazeri, Gouran Savadkoohi, Majidzadeh, & Esmaeili, 2018, Nyquist, et al., 2012, Panagopoulos, et al., 2002, Sandberg, 2004, Stenman, Andersson, Mandahl, Meis-Kindblom, & Kindblom, 1995, L. Wang, et al., 2012). On the other hand, de-differentiated chondrosarcoma, a high grade, non-chondroid sarcoma associated with low-grade cartilaginous lesions, have structural aberrations in chromosomes 1 and 9, as well as numerical aberrations (trisomy and tetrasomy) in chromosomes 7 and 19, without having distinct chromosomal translocations (Bridge, Bhatia, Anderson, & Neff, 1993, Dijkhuizen, et al., 1994, Hameed, et al., 2009, Olsson, Paulsson, Bovee, & Nord, 2011, Sakamoto, 2014). Also, clear cell chondrosarcoma, a rare, slow-growing low-grade sarcoma with low metastatic potential, does not have chromosomal translocations.
A whole-genome sequencing analysis by Tarpey et al. (Tarpey, et al., 2013) reveals that 37% of chondrosarcoma cases have COL2A1γ gene deletion and rearrangements, while mutations in IDH½ are found in 59% of cases, similar to the previous findings by Amary et al. (Amary, et al., 2011) and Totoki et al. (Totoki, et al., 2014). Additionally, they detect mutations in TP53 (20%), the RB pathway (33%), and Hedgehog signaling (18%) (Tarpey, et al., 2013). Mutations in TP53 and RB1 are the most common changes involved in the later stage of chondrosarcoma (Oshiro, et al., 1998, Samuel, Costa, & Lindskog, 2014). Also, deletion and silencing of p16, an upstream regulator of RB1, are implicated in de-differentiated chondrosarcoma (Asp, et al., 2000, Sakamoto, 2014). In conventional and de-differentiated chondrosarcoma, alterations in the TP53 gene are observed in 20–50% of cases, while alterations in the RB pathway are detected in 30–96% of cases, depending on the methods of detection (exome sequencing or IHC) (Blasenbreu, et al., 1998, Schrage, et al., 2009, Tarpey, et al., 2013). A higher incidence of TP53 mutation is found in atypical chondrosarcomas, de-differentiated types, and mesenchymal types, while overexpression (indicating missense mutations) or alteration in TP53 is correlated with high histologic grade, presence of metastasis or local recurrence, and reduced overall survival (Dobashi, et al., 1993, Oshiro, et al., 1998, Simms, Ordonez, Johnston, Ayala, & Czerniak, 1995, Wadayama, Toguchida, Yamaguchi, Sasaki, & Yamamuro, 1993). Moreover, overexpression of MDM2 by IHC is found in 33% of high-grade chondrosarcoma, which is positively correlated with histological grades (Schrage, et al., 2009). Thus, the TP53 pathway is altered in over half of high-grade chondrosarcomas. Furthermore, a mouse model overexpressing Gli2, a protein that plays a role in regulating growth plate chondrocyte differentiation, in combination with TP53 heterozygosity develops chondrosarcoma. This is due to additive effects of overexpression of Gli2 and TP53 deficiency on inhibition of apoptosis mediated through IGFBP3 downregulation in chondrocytes (Ho, et al., 2009, M. J. Kim, et al., 2011).
Chemotherapy is generally not effective in chondrosarcoma with the exception of mesenchymal chondrosarcoma (Dai, Ma, He, & Jha, 2011). As mentioned above, signaling pathways of IGF, Hedgehog, RB, and TP53, as well as these interactions, are implicated in chondrosarcoma progression, and these pathways can be potential therapeutic targets (Bovee, Hogendoorn, Wunder, & Alman, 2010, Jamil, Howie, & Salter, 2010). Intriguingly, van Oosterwijk et al. (van Oosterwijk, et al., 2013) identify that the Src pathway is involved in chondrosarcoma chemoresistance. To support their finding, dasatinib, a dual BCR/ABL and Src family tyrosine kinase inhibitor, is shown to be effective in a phase 2 clinical study for multiple sarcomas, including chondrosarcoma, and significantly sensitizes chondrosarcoma cell lines harboring TP53 mutations to doxorubicin, (Schuetze, et al., 2017). Appropriate TP53-tageting agents may be chosen to enhance efficacy of current treatment regimens for chondrosarcoma, depending on TP53 status (Polychronidou, et al., 2017).
5. Rhabdomyosarcoma (RMS)
Rhabdomyosarcoma (RMS) is a malignant tumor most commonly found in the head & neck, trunk, genitourinary tract, or limbs, and accounts for about half of all childhood soft tissue sarcomas (Egas-Bejar & Huh, 2014). RMS is generally thought to derive from the myogenic progenitors of striated muscle or MSCs, with the specific cell of origin (e.g. satellite cells, myoblasts) determining RMS subtype and therapy (Abraham, et al., 2014, X. Sun, et al., 2015). The main varieties of RMS are alveolar RMS (ARMS), embryonal RMS (ERMS) which has two histopathologic variants: spindle cell and botryoid (Esiashvili, Prabhu, Kahn, & Paulino, 2013), and pleomorphic RMS (PRMS) (Ruiz-Mesa, Goldberg, Coronado Munoz, Dumont, & Trent, 2015). The ERMS and ARMS subtypes are most common in children and adolescents, while adults typically develop PRMS (Egas-Bejar & Huh, 2014, Ruiz-Mesa, et al., 2015). Remission rates for RMS with modern therapies are now >90% in children without metastasis (Mazzoleni, et al., 2005); however, the 5-year survival rate for metastatic RMS remains less than 30%, and adult RMS also shows poor prognosis (Egas-Bejar & Huh, 2014).
RMS can also be subtyped to chromosome translocation-positive (ARMS) and -negative (ERMS, PRMS) types. The majority of ARMS cases (~85%) express PAX-FKHR fusion proteins resulting from translocation of a PAX family member (i.e. PAX3, PAX7) located on chromosome 1 or 2 to the FKHR gene locus on chromosome 13 (Stegmaier, et al., 2011). The PAX-FKHR fusion protein in ARMS maintains myogenic lineage but inhibits terminal myogenic differentiation through induction of MyoD and inhibition of cell cycle arrest (Graf Finckenstein, Shahbazian, Davicioni, Ren, & Anderson, 2007). Normal myogenic cells also express the PAX/FKHR mRNA, as well as other chimeric fusion mRNAs, during normal myogenesis, suggesting that a portion of this population of cells may become transformed during this process, producing ARMS tumors (Xie, et al., 2016). ERMS tumors frequently display chromosomal abnormalities including aneuploidy and polyploidy, and are characterized by loss of heterozygosity on the short arm of chromosome 11 (Goldstein, Meller, Issakov, & Orr-Urtreger, 2006). Although the t(1 or 2;13) chromosome translocation is rare in ERMS, both PAX3 and PAX7 are frequently overexpressed in ERMS (Goldstein, et al., 2006). PRMS also do not normally have the PAX-FKHR fusion and can have a highly complex karyotype, sometimes with abnormalities in every chromosome (Goldstein, et al., 2006). Similar to ERMS, PRMS tumors overexpress both PAX3 and FOXO1 (FKHR) (Goldstein, et al., 2006).
Neither ARMS nor ERMS harbors many somatic mutations or genetic alterations (Shern, et al., 2014), but tumors lacking the PAX-FKHR fusion protein tend to have a higher burden of somatic mutations in the TP53 and RB1 pathways (S. J. Xia, Pressey, & Barr, 2002). Interestingly, ERMS is proposed to originate at an earlier stage of myogenic development than ARMS (Stewart, et al., 2018). Using whole-genome and transcriptomic sequencing, Seki et al. (Seki, et al., 2015), further subdivide RMS to into four ARMS/ERMS subtypes based on somatic mutations: A1, A2, E1, and E2. While A1 and A2 show expression of PAX-FKHR and cell cycle regulators, the E1 and E2 subtypes contain mutations in the FGFR4/RAS/AKT pathways, as well as PTEN mutations (Seki, et al., 2015). E1 and E2 also have greater chromosomal and epigenetic aberrations evidenced by changes in allelic imbalance and gene copy number (Seki, et al., 2015).
Mutations in TP53 in RMS range from 0.02% to 15% (Ognjanovic, Martel, et al., 2012, Seki, et al., 2015, Taylor, et al., 2000), while amplification of MDM2 is detected at less than 10% of RMS (Ognjanovic, Martel, et al., 2012, Seki, et al., 2015, Taylor, et al., 2000). Because ARMS rarely shows inactivation or mutations of TP53, other mechanisms may be responsible for reduced wtp53 activity. One such mechanism may be the overexpression of PAX proteins or the PAX-FHKR oncoprotein, since these proteins repress wtp53 transcriptional activity in RMS (Stuart, Haffner, Oren, & Gruss, 1995). PAX3 specifically downregulates TP53 protein levels, but not transcription (Pani, Horal, & Loeken, 2002). Interestingly, PAX5 can directly inhibit TP53 mRNA expression by binding to the TP53 promoter, a trait shared by PAX2 and PAX8 (Stuart, et al., 1995).
On the other hand, ERMS tissues and their derived cell lines have a relatively high frequency of mutations or aberrations in the TP53/MDM2 axis (Felix, et al., 1992). Specifically, Seki et al. (Seki, et al., 2015) reveal that the E2 subtype of ERMS has a high rate (>45%) of TP53 mutation or copy number loss of TP53 (Seki, et al., 2015). Also, metastases of ERMS have high levels of TP53 expression (Leuschner, et al., 2003), and the presence of TP53 mutations significantly reduces survival of patients of the E1/E2 subtypes (Seki, et al., 2015). Intriguingly, in a kRASG12D-induced zebrafish ERMS model, deletion of TP53 enhances invasion and metastasis (Ignatius, et al., 2018), while expression of a dominant-negative mutp53 in mouse MSCs expressing PAX-FKHR fusion protein is sufficient to induce ARMS tumors in mice (Ren, et al., 2008). Thus, although clinical relevance of TP53 appears not to be robust, TP53 and its upstream regulators may contribute to malignant progression of RMS, in specific subtypes or cellular contexts. Additionally, DNp73, the N-terminal deleted dominant-negative form of a TP53 family member, is frequently overexpressed in RMS (>80%) and is also shown to inhibit myogenic differentiation and contribute to transformation of mouse myoblast cells and RMS progression in vivo (Cam, et al., 2006).
As mentioned above, prognosis of recurrent or metastatic RMS remains poor, and new treatment strategies should be developed (Shern, et al., 2014). CP-31398, a small molecule drug which enhances wtp53 activity and restores wtp53 function from mutp53, is shown to induce ROS-dependent cell cycle arrest and apoptosis in wtp53-carrying A204 and mutp53-expressing ERMS cell lines to reduce their tumor growth in xenograft mouse models (Xu, et al., 2010). Given that ARMS and PRMS rarely have TP53 mutations, compounds that restore wtp53 activity would also be efficient. On the other hand, for ERMS which often carries TP53 mutations, reactivating wtp53 activity from mutp53 may be an effective way to treat patients.
6. Leiomyosarcoma (LMS)
Leiomyosarcoma (LMS) is a soft tissue sarcoma arising from the smooth muscle cells lining small blood vessels (Danielson, et al., 2010, Rubio, et al., 2010). LMS most frequently occurs in the uterus (ULMS), but also arises in the abdomen, retroperitoneum, and large blood vessels (non-uterine LMS) (Beck, et al., 2010, Bleeker, Quevedo, & Folpe, 2012, X. Guo, et al., 2015). Although ULMS is considered distinct from non-uterine LMS, these two types are genetically more similar to one another than other sarcomas (Abeshouse, et al., 2017, Miettinen & Fetsch, 2006). Individuals with LFS or the condition called hereditary leiomyomatosis and renal cell carcinoma (HLRCC) have a higher risk of developing LMS (Farid & Ngeow, 2016). Another risk factor for ULMS is tamoxifen exposure for unrelated conditions, such as breast cancer (Bleeker, et al., 2012). The prognosis for ULMS is worse than for other LMS, with a recurrence rate between 53%−71%, and the five-year overall survival for these patients is 15%−25% (D’Angelo & Prat, 2010). On the other hand, 39% of non-uterine LMS patients experience recurrence either locally or distantly, with retroperitoneal LMS cases having a higher recurrence rate of 51% (Gladdy, et al., 2013). The five-year overall survival for non-uterine LMS is 64% (Mankin, et al., 2004). The first line chemotherapy for LMS is presently a combination of gemcitabine and docetaxel (Momtahen, Curtin, & Mittal, 2016, Seddon, et al., 2017); however, a recent study suggests that doxorubicin may have better outcomes in combination with gemcitabine than docetaxel (Seddon, et al., 2017).
Unlike other sarcomas, LMS does not have a key fusion protein driving its tumorigenesis, although a small group of ULMS (<2.5%) is found to express an ALK fusion protein (KANK2-ALK, ACTG2-ALK) (Davis, et al., 2019). Rather, the general molecular alterations in LMS affect activities of the tumor suppressors TP53, PTEN, and/or RB1 with deletion of PTEN and RB1 and mutations of TP53 (Chudasama, et al., 2018, Grossmann, Layfield, & Randall, 2012, Gunderson, et al., 2016, Yang, et al., 2009). Cytogenetic studies of LMS suggest that loss of tumor suppressors can be an initiating event in LMS, while oncogene activation may occur at a later stage of malignant progression, because low-grade tumors contain more DNA copy number losses, while high-grade tumors have more copy number gains (Grossmann, et al., 2012). Interestingly, loss of TP53, but not RB1, is sufficient to transform MSCs and induce LMS formation in mice (Rubio, et al., 2010). Besides these tumor suppressors, LMS often has mutations in ATRX (Alpha thalassemia/mental retardation syndrome X-linked) with a mutation rate of ~30% in ULMS (Gunderson, et al., 2016, Mäkinen, et al., 2016). ATRX is a SWI/SNF chromatin remodeling protein which acts as a tumor suppressor (Kadoch & Crabtree, 2015). ULMS also harbors mutations in MED12 (mediator complex subunit 12), a coactivator involved in the interaction of transcription factors with RNA polymerase II, at a ~20% frequency, but this does not occur in non-uterine LMS (Mäkinen, et al., 2016, Mäkinen, Kämpjärvi, Frizzell, Bützow, & Vahteristo, 2017). MED12 mutation is also common in the benign tumor leiomyoma (Ravegnini, et al., 2012), suggesting that ULMS harboring MED12 mutations may represent the small population of tumors known to originate from leiomyoma (Mäkinen, et al., 2017). Intriguingly, ULMS tumors frequently have one or more hallmarks of “BRCAness”, enabling the use of a PARP inhibitor for ULMS treatment (Chudasama, et al., 2018, Seligson, et al., 2018).
TP53 is mutated in ~50% of all LMS and ~35% of ULMS (Abeshouse, et al., 2017, Gao, Seebacher, Hornicek, Guo, & Duan, 2018, O’neill, McBride, Connolly, & McCluggage, 2007). LMS frequently shows gene amplifications near the gene locus of MDM2, 12q15 (Ragazzini, et al., 2004); however, MDM2 amplification is rare in LMS (Miura, et al., 2012, Miyajima, et al., 2001, Yang, et al., 2009). Importantly, there is a correlation between abnormalities in TP53 and an advanced clinicopathological stage or poor prognosis in LMS (Konomoto, Fukuda, Hayashi, Kumazawa, & Tsuneyoshi, 1998, Patterson, et al., 1994), suggesting potential value for TP53-tageting therapies. Indeed, a mutp53 reactivator, PRIMA-1, is shown to reduce viability of IB134 ULMS and IB138 soft tissue LMS cell lines harboring TP53 mutations, while it is less effective in the wtp53-expressing IB139 LMS cell line (Grellety, et al., 2015). Also, Gendicine, a recombinant adenoviral vector expressing wtp53, is clinically used in China and shows a 66.7% remission rate and a 91.7% responsive rate in combination with chemotherapy in ULMS (Y. Xia, Du, Wang, & Li, 2018, W.-W. Zhang, et al., 2018). These results support the idea that restoring wtp53 activity in tumors may be beneficial for LMS patients.
7. Synovial Sarcoma
Synovial sarcoma comprises up to 10% of all soft tissue sarcomas and is the second most common soft tissue sarcoma in patients younger than 30 years old, although the majority (70%) of patients are older than 30 years old (Mark D. Murphey, et al., 2006, Stacchiotti & Van Tine, 2017). The prognosis of synovial sarcoma is worse with increased age (Jang, et al., 2007, Stacchiotti & Van Tine, 2017). Tumors frequently occur in the lower half of the body, particularly near the knee joint, but can occur in the head and neck areas of the upper body (Gopalakrishnan, et al., 2017, Mark D. Murphey, et al., 2006). MSCs are thought to be the cell of origin for synovial sarcoma (Garcia, et al., 2012, Norifumi Naka, et al., 2010). Tumors have the appearance of synovial tissue, but are unrelated to the synovium, and they are comprised of two main cell types: epithelial cells and mesenchymal spindle cells (El Beaino, Araujo, Lazar, & Lin, 2017, Mark D. Murphey, et al., 2006). The presence of one or both of these cell types as well as the degree of cellular differentiation defines the sarcoma’s subtype: monophasic (mesenchymal), biphasic (epithelial and mesenchymal), and small cell (poorly differentiated) (El Beaino, et al., 2017). Importantly, over half (50–70%) of synovial sarcomas metastasize, which makes overall survival for synovial sarcoma less than 40% (Krieg, et al., 2011).
A recent whole-genome sequencing analysis has revealed that 95% of synovial sarcomas express the fusion oncoprotein SS18 (synovial sarcoma translocation, chromosome 18)-SSX, making it a defining characteristic of synovial sarcoma (Vlenterie, et al., 2015). The t(X:18)(p11.2;q11.2) chromosomal translocation creates an in-frame fusion of SS18 to SSX1, SSX2, or SSX4, leading to generation of a SS18-SSX protein (Clark, et al., 1994, de Leeuw, Balemans, Weghuis, & van Kessel, 1995, Skytting, et al., 1999). Although the SS18-SSX lacks a DNA binding domain, it interacts with other chromatin regulators, including components of the SWI/SNF complex (e.g., hBRM, BRG1), to act as an oncoprotein that drives synovial sarcoma progression (Clark, et al., 1994, de Leeuw, et al., 1995, Skytting, et al., 1999). To support this, SS18-SSX proteins have been found to bind with a non-canonical polycomb repressive complex 1 (PRC1.1) and co-associate with SWI/SNF and KDM2B complexes to aberrantly regulate the expression of developmentally regulated transcription factors and mesenchymal differentiation genes (Banito, et al., 2018). Additionally, SS18-SSX plays roles in activation of the WNT/β-catenin and PI3K/AKT pathways and stabilization of MDM2, leading to inhibition of TP53 activity (Arcy, Maruwge, Ryan, & Brodin, 2008, Bozzi, et al., 2008).
Synovial sarcomas do not frequently display mutations in the TP53 gene (D’Arcy, Ryan, & Brodin, 2009). Despite its low rate of mutations (~6%), TP53 alteration may be an effective prognostic indicator because tumors with missense mutations in the TP53 gene show significantly reduced 5-year survival when compared to non-mutated tumors (Schhneider-stock, et al., 1999). Also, over 3-fold gene amplifications of MDM2 and MDM4 are observed in 33% and 44% of synovial sarcoma tissues, respectively, which has an inverse correlation with TP53 mutations (Ito, et al., 2011). This is supported by the finding of that SS18-SSX1 induces TP53 degradation by MDM2 (Arcy, et al., 2008). Thus, TP53 activity appears to be lowered in synovial sarcoma through MDM2/MDM4.
Presently, synovial sarcomas are mainly treated with doxorubicin, ifosfamide, and pazopanib, a multi-targeted tyrosine kinase inhibitor, in addition to surgical resection (In, Hu, & Tseng, 2017). These therapies are mainly effective to non-metastatic synovial sarcoma but are limited in effectiveness in tumors with metastases. Hence, the overall 10-year survival rates are 50–60% (Lewis, et al., 2000). Obviously, new therapeutic strategies must be developed for metastatic synovial sarcomas. Since TP53 mutation is uncommon in synovial sarcoma, restoration of wtp53 activity in tumors by MDM2/MDM4 inhibitors may be a rational strategy for this type of sarcoma. Indeed, Nutlin-3a and MI-219, two small molecule inhibitors of MDM2, are shown to inhibit the growth of synovial sarcoma cells in vitro (Banito, et al., 2018, D’Arcy, et al., 2009, Wade, Li, & Wahl, 2013).
8. Liposarcoma (LPS)
Liposarcoma (LPS) is the most common adult soft tissue sarcoma and represents ~25% of all soft tissue sarcomas. It is a tumor derived from the mesenchymal adipogenic lineage, and most commonly arises in the limbs, retroperitoneum, and the paratesticular areas of the body (Brill, et al., 2010, Bui, et al., 2011, Peng, et al., 2011). LPS is often confused for a benign tumor (L. G. Dodd, 2012), posing a threat for patient care. There are four key subtypes of LPS which have been identified based on cytogenetic features: myxoid/round cell LPS, pleomorphic LPS, well-differentiated LPS, and de-differentiated LPS (Bui, et al., 2011, Genadry, Pietrobono, Rota, & Linardic, 2018). The subtypes can be distinguished by the presence of the FUS-CHOP fusion oncoprotein (myxoid/round cell LPS), MDM2 overexpression (well-differentiated, and de-differentiated LPS), and chromosomal abnormalities (pleomorphic LPS) (L. G. Dodd, 2012). LPS has about a 20% rate of recurrence overall, with de-differentiated LPS showing a higher local recurrence rate of 37.6% (Vos, et al., 2018). Distant metastasis occurs in ~20% of all LPS, while in the pleomorphic subtype metastasis occurs in ~45% of tumors (Vos, et al., 2018). Although LPS has a relatively good prognosis overall with a five-year overall survival rate of ~80%, de-differentiated and pleomorphic LPS show significantly worse prognosis with overall 5-year survival rates at ~40% (Ng, Scharschmidt, Mayerson, & Fisher, 2013, Vos, et al., 2018). Pleomorphic LPS also tends to have highly variable karyotypes (Mandahl, et al., 2017).
A chromosomal translocation t(12;16)(q13;p11) results in the creation of a fusion oncoprotein, FUS-CHOP (also referred to as TLS-CHOP), which is present in myxoid/round cell LPS but not in other types (Rodriguez, et al., 2011, Tornin, et al., 2018). In rare cases, t(12;22)(q13;q12) translocation creates the EWS-CHOP fusion protein (Rodriguez, et al., 2011). FUS is an RNA-binding protein involved in regulation of gene expression, genomic integrity, and mRNA/microRNA processing, while CHOP is an ER-stress response transcription factor (Nishitoh, 2011). Overexpression of FUS-CHOP in the mouse genome under the human EF1α promoter is shown to induce LPS in 100% of mice (Pérez-Losada, et al., 2000). LPS tumors developed in FUS-CHOP transgenic mice express high levels of the adipocyte regulatory protein PPARγ (Pérez-Losada, et al., 2000). FUS-CHOP–overexpressing primary mesenchymal progenitor cells (MPC) also form tumors resembling human myxoid LPS (Riggi, et al., 2006). Transcription profile analysis of these tumors and non-xenografted MPCs expressing FUS-CHOP reveals repression of CTGF, PERP, and TFPI, as well as induction of growth factors (PDGFA, HGF), cytokines (IL6), growth factor receptors (MET), cell cycle regulators (CDK4, MDM2), proteolytic enzymes (MMP-11, CTSD, PLAT), and adipocyte differentiation-associated factors, (ADFP, FASN, HMGCR, RGS2) (Riggi, et al., 2006). Moreover, the FUS-CHOP fusion protein enhances invasiveness of LPS cells by activating the SRC/FAK/RHO/ROCK pathway, and enhances metastatic potential of LPS cells by transcriptionally upregulating MMP2 proteases (Patil, et al., 2014, Tornin, et al., 2018).
Intriguingly, supernumerary ring chromosomes and giant rod marker chromosomes are observed in myxoid/round cell, well-differentiated, and de-differentiated LPS (Laroche-Clary, et al., 2017, Macchia, et al., 2018, Mandahl, et al., 2017, Pedeutour, et al., 1999). These supernumerary chromosomes frequently amplify the CDK4, HMGA2, and MDM2 genes from chromosome 12q (Mandahl, et al., 2017, Pedeutour, et al., 1999, Szymanska, et al., 1996). Also, well-differentiated and de-differentiated LPSs carry neochromosomes due to amplifications and rearrangements of chromosome 12q encoding oncogenes (MDM2, CDK4, YEATS2) and adipocytic differentiation factors (HMGA2, CPM) (Beird, et al., 2018). A recent study with exome and RNA sequencing analyses using 17 patients with both well-differentiated and de-differentiated LPSs reveals that the two subtypes share only 8.3% of their somatic mutations between matched tumors, suggesting the possibility that these tumors arise from a common origin and diverge at an early stage of tumor development (Beird, et al., 2018). Moreover, de-differentiated tumors show greater genomic instability attributed to an early clonal divergence from well-differentiated LPS tumors (Beird, et al., 2018).
An earlier study by Barretina et al. (Barretina, et al., 2010) identify mutations in TP53 and NF1, as well as PIK3CA mutations in myxoid/round-cell LPSs. To support their finding, SNP arrays, whole exome sequencing, and targeted exome sequencing by Kanojia et al. (Kanojia, et al., 2015) identify carboxypeptidase M (CPM), which cleaves C-terminal arginine or lysine residues from polypeptides, as an oncogene which is involved in the EGFR pathway and is recurrently amplified in well-differentiated and de-differentiated LPS. They also report FGFR1 amplification at chromosome 8p11.23, classical CDK4, HMGA2, and MDM2 amplification at the chromosome 12q13–15 region, and deletion of TP53BP1 at chromosome 15q15 in de-differentiated LPS. Moreover, their functional analyses reveal the tumor suppressive role of neurofibromin 1 (NF1), which negatively regulates RAS through hydrolysis of RAS-GTP, in LPS, irrespective of subtype (Kanojia, et al., 2015).
Mutations in TP53 are not usually high in LPS (10–20%), except for pleomorphic LPS in which ~60% of cases show TP53 mutations. This is likely because the MDM2 gene is amplified in the majority of well-differentiated and de-differentiated LPSs, leading to functional inactivation of TP53 without mutations (Abeshouse, et al., 2017, Barretina, et al., 2010, Ghadimi, et al., 2011, Kawai, et al., 1994, Nakayama, et al., 1995, Taubert, et al., 1998). However, mutations in TP53 are associated with proliferation, tumor aggressiveness, reduced patient survival, and advanced disease in LPS (Antonescu, et al., 2001, Kawai, et al., 1994, Schneider-Stock, et al., 1999). Moreover, subcutaneous transplantation of mouse adipose-derived mesenchymal stem/stromal cells (ASCs) expressing FUS-CHOP results in formation of LPS-like tumors only when cells are null for TP53, although this is not observed in human ASCs (Rodriguez, et al., 2011). Since TP53-null mouse ASCs develop LMS, these data may indicate that FUS-CHOP predisposes TP53-null mouse ASCs to adipogenic differentiation (Rodriguez, et al., 2011). To support this finding, Charytonowicz et al. (Charytonowicz, et al., 2012) generate a mouse model expressing FUS-CHOP specifically in mesoderm tissues. Although the FUS-CHOP transgenic mice are not tumor-prone, mice show development of sarcoma resembling myxoid/round cell LPS upon deletion of the TP53 allele(s). Clinically, TP53 mutations are detected in 15–30% of cases of human myxoid/round cell LPS and are correlated with unfavorable outcomes (Antonescu, et al., 2001, Dei Tos, et al., 1997, Schneider-Stock, et al., 1999). Thus, FUS-CHOP and TP53 loss may have cooperative effects on myxoid round cell LPS progression.
Chemotherapeutic drugs for primary and metastatic LPS include doxorubicin and ifosfamide in the first-line setting, and other drugs including docetaxel and gemcitabine are also used (Kollár & Benson, 2014). However, due to high frequency of MDM2 amplification and low frequency of TP53 mutations in LPS, especially in well-differentiated and de-differentiated LPS, use of MDM2 antagonists to restore TP53 activity (e.g., Nutlin-3a, RG7112, SAR405838) can be a major therapeutic strategy (Ray-Coquard, et al., 2012). Nutlin-3a has been extensively studied in multiple cancers, including in MDM2-amplified LPS cells, to restore wtp53 activity and induce apoptosis (Muller, et al., 2007). A Nutlin-3a analog, RG7112, has been clinically tested for the treatment of patients with well-differentiated or de-differentiated LPS (Obrador-Hevia, et al., 2015, Ray-Coquard, et al., 2012). Although RG7112 stabilizes disease progression in many cases, it has significant adverse effects including neutropenia and thrombocytopenia, in as many as 40% of patients (Ray-Coquard, et al., 2012). SAR405838 is also shown to inhibit de-differentiated LPS progression in in vitro and in vivo mouse models; it induces stabilization of TP53, upregulation of TP53 targets, and induction of apoptosis in de-differentiated LPS, which has enabled this drug to enter early-phase clinical trials for multiple malignancies (Bill, et al., 2016). As stated above, TP53 mutation status is correlated with recurrence, metastasis, and advanced stage of LPS (Antonescu, et al., 2001, Kawai, et al., 1994, McGovern, Zhou, & Jones, 2017, Schneider-Stock, et al., 1999). Hence, mutp53-targeted therapies may be efficacious for these aggressive LPS, which should be tested clinically in the future.
9. Angiosarcoma
Angiosarcomas are malignant endothelial-cell tumors originating from vascular or lymphatic tissues (Young, Brown, Reed, Hughes, & Woll, 2010). Angiosarcomas can occur spontaneously or secondarily to ionizing radiation or chronic lymphedema; they are subdivided into cutaneous, lymphoedema-associated, radiation-induced, primary-breast, and soft-tissue types (Behjati, et al., 2014, Young, et al., 2010). Surgical resection followed by high-dose radiotherapy, is the standard of care. However, recurrence is frequent, and most patients ultimately develop metastases, leading to poor prognosis (Florou & Wilky, 2018). Tyrosine kinase inhibitors and vascular-targeted therapies can be efficacious, but some patients still develop drug resistance (Florou & Wilky, 2018, Spiker & Ramsey, 2018, Young, et al., 2010). Better understanding of the molecular mechanisms of angiosarcoma malignancy is essential for discovery of new treatments and improvement of current therapeutic regimens.
Analyses of whole-genome, exome, and targeted sequencing with primary and secondary angiosarcomas have provided useful information in determining the potential causes of tumorigenesis. Behjati et al. (Behjati, et al., 2014) identify recurrent mutations of angiogenesis-related genes, PTPRB (receptor-type tyrosine-protein phosphatase β) and PLCG1 (phospholipase C, γ1) in angiosarcomas. The PTPRB gene has predominantly truncating mutations in 26% of tumors, with an activating R707Q missense mutation in 9% of cases (Behjati, et al., 2014). Also, Murali et al. (Murali, et al., 2015) report that more than 50% of angiosarcomas carry some genetic alterations impacting the MAPK pathway. These include mutations in KRAS, HRAS, NRAS, BRAF, MAPK1, and NF1, as well as amplifications in MAPK1/CRKL, CRAF, or BRAF. Moreover, the most frequently detected genetic aberrations are mutations in TP53 (35%) and losses of CDKN2A (26%). MYC amplifications are detected in the majority (88%) of radiation-induced angiosarcomas, which is mutually exclusive of alterations in TP53 and CDKN2A. Additionally, FLT4 amplifications and mutations or rearrangements of PTPRB, PLCG1, CIC, FLT4, and KDR genes are detected in angiosarcomas (S. C. Huang, et al., 2016). Thus, whole-genome sequencing analyses further confirm the molecular heterogeneity of angiosarcomas.
The involvement of TP53 in angiosarcomas is also shown by Naka et al (N. Naka, et al., 1997), where frequency of TP53 mutations is dependent on the site of tumors with an overall occurrence of ~50%. In addition to TP53 mutations, upregulation of MDM2 is reported in two-thirds of angiosarcoma cases (Zietz, et al., 1998). Intriguingly, increased VEGF levels are observed in ~80% of cases, which is correlated with increased protein levels of TP53 and MDM2 (Zietz, et al., 1998). The involvement of TP53 in angiosarcoma is also shown by mouse studies showing occurrence of angiosarcoma in TP53 knockout mice (Landuzzi, et al., 2014, Lang, et al., 2004). Specifically, over 65% of alymphocytic TP53 knockout mice (Rag2−/−;II2rg−/−;TP53−/−, referred to as RGKO-TP53−/−) spontaneously develop hemangiosarcoma (Landuzzi, et al., 2014). Furthermore, Li et al. (Q. Li, et al., 2014) observe that ~34% of mice carrying hypomorphic TP53 alleles with overexpression of MDM2 (MDM2Tg TP53Neo/Neo) spontaneously develop angiosarcoma. Moreover, cre-mediated restoration of wtp53 expression in angiosarcoma developed in MDM2Tg TP53Neo/Neo mice results in inhibition of tumor growth in a syngeneic transplant model, suggesting that TP53 restoration is a potential therapeutic strategy for angiosarcomas (Q. Li, et al., 2014). Considering the high frequency of TP53 mutations in angiosarcoma, TP53-targeted therapies may be an option for anti-angiosarcoma treatment. Meanwhile, continuous efforts to understand the genetic heterogeneity of angiosarcomas and the identification of molecular targets for each subtype of angiosarcoma are necessary to determine the best therapy for each subtype (Florou & Wilky, 2018). (Mehren & Joensuu, 2018)
10. Undifferentiated Pleomorphic Sarcoma (UPS)
Undifferentiated pleomorphic sarcoma (UPS) is an aggressive bone and soft tissue sarcoma, originally called malignant fibrous histiocytoma and later reclassified as UPS in 2002 by the World Health Organization (WHO) (Matushansky, et al., 2009, M. D. Murphey, 2007). UPS is categorized to four subtypes based on the histology: storiform-pleomorphic, myxoid, giant cell, and inflammatory types. The storiform-pleomorphic type is the most common (~70%), followed by the myxoid variant (~20%), while giant cell and inflammatory types are rare. Of these subtypes, the myxoid variant is the least aggressive and has the best prognosis. Most patients are between 50 and 70 years old, and the most common tumor location is the lower extremities (Dei Tos, 2006, M. D. Murphey, 2007).
Due to clinical and genetic heterogeneity, few studies have analyzed the molecular profiles of UPS (R. D. Dodd, 2016). Conventional comparative genomic hybridization (CGH) and gene expression profiling analyses reveal similarity between UPS and LMS including loss of 13q14–21 as the most common deletion and alterations of the RB and TP53 pathways (Carneiro, et al., 2009). Using Ptch1, TP53 and/or RB1 conditional knockout mouse models, Rubin et al. (Rubin, et al., 2011) show that ERMS and UPS are a continuum of myodifferentiation, with satellite cells giving rise to UPS. Also, loss of RB1, but not TP53, promotes undifferentiated phenotypes to mimic UPS (Rubin, et al., 2011). However, recent multi-platform molecular landscape analyses using human soft tissue sarcomas identify molecular similarity between UPS and myxofibrosarcoma and suggest the involvement of the Hippo pathway in UPS pathogenesis (Cancer Genome Atlas Research Network. Electronic address & Cancer Genome Atlas Research, 2017). Moreover, some previously diagnosed peripheral UPSs, which do not have characteristics of well-differentiated LPS but have MDM2 gene amplifications, later turned out to be de-differentiated LPS (Le Guellec, et al., 2014). Overall, due to complex molecular heterogeneity and difficulty of diagnosis, the genetic and molecular profiling of UPS remains unclear. Nonetheless, Serrano et al. (Serrano, et al., 2016) recently show that the RAS/MAPK and PI3K/mTOR pathways are activated in the majority of cases of UPS, while activation of the RAS/MAPK pathway is correlated with an increased risk of disease recurrence and impaired overall survival (R. D. Dodd, 2016).
Regarding the involvement of TP53 in UPS, a study using a large series of 143 soft tissue sarcomas identifies genomic deletion of the TP53 locus in 18.4% of tumors and TP53 mutations in 32% (Perot, et al., 2010). Intriguingly, most UPS (87.2%) retain one wild-type TP53 allele. Furthermore, most tumors which do not have TP53 alterations show deletion or silencing of the p14ARF gene, a negative regulator of MDM2 (Perot, et al., 2010). These results suggest that the p14ARF-MDM2-p53 pathway plays crucial roles in UPS pathogenesis. Hence, restoring wtp53 activity may be one option for treating UPS.
11. Conclusions and future perspectives
In this review we have summarized the role of TP53 in bone and soft tissue sarcomas with emphasis on TP53 and mutations/deletions in TP53 as therapeutic targets. Alterations to TP53 or other genes in the TP53 pathway often occur in sarcomas. Notably, over 70% of osteosarcoma has structural variants or mutations in the TP53 gene, Ewing sarcoma is rarely mutated for TP53 due to EWS-FLI1’s inhibitory effect on TP53, non-uterine LMS has a high rate (~50%) of TP53 mutation, and well- and de-differentiated LPSs are defined by amplification of MDM2. Evolving new technologies, including next generation sequencing, would enable identification of novel therapeutic targets, some of which may directly or indirectly alter TP53 activity. While current TP53-targeted therapies have some drawbacks in a clinical setting, including bone marrow suppression and other side effects, improved TP53-targeted therapies to restore wtp53 activity, reactivate wtp53 activity from mutp53, deplete mutp53, and target vulnerabilities in TP53-mutated/deleted cells may prove effective in therapy-resistant bone and soft tissue sarcomas in the near future.
Acknowledgments
ET and AC summarized and organized literatures, while ET, AC, and TI wrote the manuscript. This manuscript is supported by NIH R01 CA174735 (TI), R01 CA214916 (TI), and Braden’s Hope (TI) grants in U.S.A. These funding agencies had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Abbreviations
- TP53
tumor protein p53
- MDM2
mouse double minute 2
- MDM4
mouse double minute 4
- IHC
immunohistochemistry
- RMS
rhabdomyosarcoma
- ERMS
embryonic rhabdomyosarcoma
- ARMS
alveolar rhabdomyosarcoma
- PRMS
pleomorphic rhabdomyosarcoma
- LPS
liposarcoma
- LMS
leiomyosarcoma
- ULMS
uterine leiomyosarcoma
- ASCs
adipose-derived mesenchymal stem/stromal cells
- LFS
Li-Fraumeni Syndrome
- LOF
loss of function
- GOF
gain of function
- PDB
Paget’s disease of bone
- MSC
mesenchymal stem cell
Footnotes
Conflicts of interest statement
The authors declare no conflict of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Abeshouse A, Adebamowo C, Adebamowo SN, Akbani R, Akeredolu T, Ally A, Anderson ML, Anur P, Appelbaum EL, Armenia J, Auman JT, Bailey MH, Baker L, Balasundaram M, Balu S, Barthel FP, Bartlett J, Baylin SB, Behera M, Belyaev D, Bennett J, Benz C, Beroukhim R, Birrer M, Bocklage T, Bodenheimer T, Boice L, Bootwalla MS, Bowen J, Bowlby R, Boyd J, Brohl AS, Brooks D, Byers L, Carlsen R, Castro P, Chen H-W, Cherniack AD, Chibon F, Chin L, Cho J, Chuah E, Chudamani S, Cibulskis C, Cooper LAD, Cope L, Cordes MG, Crain D, Curley E, Danilova L, Dao F, Davis IJ, Davis LE, Defreitas T, Delman K, Demchok JA, Demetri GD, Demicco EG, Dhalla N, Diao L, Ding L, DiSaia P, Dottino P, Doyle LA, Drill E, Dubina M, Eschbacher J, Fedosenko K, Felau I, Ferguson ML, Frazer S, Fronick CC, Fulidou V, Fulton LA, Fulton RS, Gabriel SB, Gao J, Gao Q, Gardner J, Gastier-Foster JM, Gay CM, Gehlenborg N, Gerken M, Getz G, Godwin AK, Godwin EM, Gordienko E, Grilley-Olson JE, Gutman DA, Gutmann DH, Hayes DN, Hegde AM, Heiman DI, Heins Z, Helsel C, Hepperla AJ, Higgins K, Hoadley KA, Hobensack S, Holt RA, Hoon DB, Hornick JL, Hoyle AP, Hu X, Huang M, Hutter CM, Iacocca M, Ingram DR, Ittmann M, Iype L, Jefferys SR, Jones KB, Jones CD, Jones SJM, Kalir T, Karlan BY, Karseladze A, Kasaian K, Kim J, Kundra R, Kuo H, Ladanyi M, Lai PH, Laird PW, Larsson E, Lawrence MS, Lazar AJ, Lee S, Lee D, Lehmann K-V, Leraas KM, Lester J, Levine DA, Li I, Lichtenberg TM, Lin P, Liu J, Liu W, Liu EM, Lolla L, Lu Y, Ma Y, Madan R, Maglinte DT, Magliocco A, Maki RG, Mallery D, Manikhas G, Mardis ER, Mariamidze A, Marra MA, Martignetti JA, Martinez C, Mayo M, McLellan MD, Meier S, Meng S, Meyerson M, Mieczkowski PA, Miller CA, Mills GB, Moore RA, Morris S, Mose LE, Mozgovoy E, Mungall AJ, Mungall K, Nalisnik M, Naresh R, Newton Y, Noble MS, Novak JE, Ochoa A, Olvera N, Owonikoko TK, Paklina O, Parfitt J, Parker JS, Pastore A, Paulauskis J, Penny R, Pereira E, Perou CM, Perou AH, Pihl T, Pollock RE, Potapova O, Radenbaugh AJ, Ramalingam SS, Ramirez NC, Rathmell WK, Raut CP, Riedel RF, Reilly C, Reynolds SM, Roach J, Robertson AG, Roszik J, Rubin BP, Sadeghi S, Saksena G, Salner A, Sanchez-Vega F, Sander C, Schein JE, Schmidt HK, Schultz N, Schumacher SE, Sekhon H, Senbabaoglu Y, Setdikova G, Shelton C, Shelton T, Shen R, Shi Y, Shih J, Shmulevich I, Sica GL, Simons JV, Singer S, Sipahimalani P, Skelly T, Socci N, Sofia HJ, Soloway MG, Spellman P, Sun Q, Swanson P, Tam A, Tan D, Tarnuzzer R, Thiessen N, Thompson E, Thorne LB, Tong P, Torres KE, van de Rijn M, Van Den Berg DJ, Van Tine BA, Veluvolu U, Verhaak R, Voet D, Voronina O, Wan Y, Wang Z, Wang J, Weinstein JN, Weisenberger DJ, Wilkerson MD, Wilson RK, Wise L, Wong T, Wong W, Wrangle J, Wu Y, Wyczalkowski M, Yang L, Yau C, Yellapantula V, Zenklusen JC, Zhang J, Zhang H, Zhang H, & Zmuda E (2017). Comprehensive and Integrated Genomic Characterization of Adult Soft Tissue Sarcomas. Cell, 171, 950–965.e928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abraham J, Nuñez-Álvarez Y, Hettmer S, Carrió E, Chen H-IH, Nishijo K, Huang ET, Prajapati SI, Walker RL, Davis S, Rebeles J, Wiebush H, McCleish AT, Hampton ST, Bjornson CRR, Brack AS, Wagers AJ, Rando TA, Capecchi MR, Marini FC, Ehler BR, Zarzabal LA, Goros MW, Michalek JE, Meltzer PS, Langenau DM, LeGallo RD, Mansoor A, Chen Y, Suelves M, Rubin BP, & Keller C (2014). Lineage of origin in rhabdomyosarcoma informs pharmacological response. Genes & Development, 28, 1578–1591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abudu A, Mangham DC, Reynolds GM, Pynsent PB, Tillman RM, Carter SR, & Grimer RJ (1999). Overexpression of p53 protein in primary Ewing’s sarcoma of bone: relationship to tumour stage, response and prognosis. Br J Cancer, 79, 1185–1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adhikari AS, & Iwakuma T (2009). Mutant p53 gain of oncogenic function: in vivo evidence, mechanism of action and its clinical implications. Fukuoka Igaku Zasshi, 100, 217–228. [PubMed] [Google Scholar]
- Al-Romaih K, Bayani J, Vorobyova J, Karaskova J, Park PC, Zielenska M, & Squire JA (2003). Chromosomal instability in osteosarcoma and its association with centrosome abnormalities. Cancer Genet Cytogenet, 144, 91–99. [DOI] [PubMed] [Google Scholar]
- Alexandrova EM, Yallowitz AR, Li D, Xu S, Schulz R, Proia DA, Lozano G, Dobbelstein M, & Moll UM (2015). Improving survival by exploiting tumour dependence on stabilized mutant p53 for treatment. Nature, 523, 352–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amary MF, Bacsi K, Maggiani F, Damato S, Halai D, Berisha F, Pollock R, O’Donnell P, Grigoriadis A, Diss T, Eskandarpour M, Presneau N, Hogendoorn PC, Futreal A, Tirabosco R, & Flanagan AM (2011). IDH1 and IDH2 mutations are frequent events in central chondrosarcoma and central and periosteal chondromas but not in other mesenchymal tumours. J Pathol, 224, 334–343. [DOI] [PubMed] [Google Scholar]
- Anderson ND, de Borja R, Young MD, Fuligni F, Rosic A, Roberts ND, Hajjar S, Layeghifard M, Novokmet A, Kowalski PE, Anaka M, Davidson S, Zarrei M, Id Said B, Schreiner LC, Marchand R, Sitter J, Gokgoz N, Brunga L, Graham GT, Fullam A, Pillay N, Toretsky JA, Yoshida A, Shibata T, Metzler M, Somers GR, Scherer SW, Flanagan AM, Campbell PJ, Schiffman JD, Shago M, Alexandrov LB, Wunder JS, Andrulis IL, Malkin D, Behjati S, & Shlien A (2018). Rearrangement bursts generate canonical gene fusions in bone and soft tissue tumors. Science, 361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Antonescu CR, Tschernyavsky SJ, Decuseara R, Leung DH, Woodruff JM, Brennan MF, Bridge JA, Neff JR, Goldblum JR, & Ladanyi M (2001). Prognostic impact of P53 status, TLS-CHOP fusion transcript structure, and histological grade in myxoid liposarcoma: a molecular and clinicopathologic study of 82 cases. Clin Cancer Res, 7, 3977–3987. [PubMed] [Google Scholar]
- Arcy P, Maruwge W, Ryan BA, & Brodin B (2008). The Oncoprotein SS18-SSX1 Promotes p53 Ubiquitination and Degradation by Enhancing HDM2 Stability. Molecular Cancer Research, 6, 127. [DOI] [PubMed] [Google Scholar]
- Aryee DN, Niedan S, Ban J, Schwentner R, Muehlbacher K, Kauer M, Kofler R, & Kovar H (2013). Variability in functional p53 reactivation by PRIMA-1(Met)/APR-246 in Ewing sarcoma. Br J Cancer, 109, 2696–2704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Asada N, Tsuchiya H, & Tomita K (1999). De novo deletions of p53 gene and wild-type p53 correlate with acquired cisplatin-resistance in human osteosarcoma OST cell line. Anticancer Res, 19, 5131–5137. [PubMed] [Google Scholar]
- Asp J, Sangiorgi L, Inerot SE, Lindahl A, Molendini L, Benassi MS, & Picci P (2000). Changes of the p16 gene but not the p53 gene in human chondrosarcoma tissues. Int J Cancer, 85, 782–786. [DOI] [PubMed] [Google Scholar]
- Balamuth NJ, & Womer RB (2010). Ewing’s sarcoma. Lancet Oncol, 11, 184–192. [DOI] [PubMed] [Google Scholar]
- Ban J, Bennani-Baiti IM, Kauer M, Schaefer KL, Poremba C, Jug G, Schwentner R, Smrzka O, Muehlbacher K, Aryee DN, & Kovar H (2008). EWS-FLI1 suppresses NOTCH-activated p53 in Ewing’s sarcoma. Cancer Res, 68, 7100–7109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banito A, Li X, Laporte AN, Roe J-S, Sanchez-Vega F, Huang C-H, Dancsok AR, Hatzi K, Chen C-C, Tschaharganeh DF, Chandwani R, Tasdemir N, Jones KB, Capecchi MR, Vakoc CR, Schultz N, Ladanyi M, Nielsen TO, & Lowe SW (2018). The SS18-SSX Oncoprotein Hijacks KDM2B-PRC1.1 to Drive Synovial Sarcoma. Cancer Cell, 33, 527–541.e528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bardot B, & Toledo F (2017). Targeting MDM4 Splicing in Cancers. Genes (Basel), 8. [Google Scholar]
- Barretina J, Taylor BS, Banerji S, Ramos AH, Lagos-Quintana M, DeCarolis PL, Shah K, Socci ND, Weir BA, Ho A, Chiang DY, Reva B, Mermel CH, Getz G, Antipin Y, Beroukhim R, Major JE, Hatton C, Nicoletti R, Hanna M, Sharpe T, Fennell TJ, Cibulskis K, Onofrio RC, Saito T, Shukla N, Lau C, Nelander S, Silver SJ, Sougnez C, Viale A, Winckler W, Maki RG, Garraway LA, Lash A, Greulich H, Root DE, Sellers WR, Schwartz GK, Antonescu CR, Lander ES, Varmus HE, Ladanyi M, Sander C, Meyerson M, & Singer S (2010). Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy. Nature Genetics, 42, 715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beck AH, Lee CH, Witten DM, Gleason BC, Edris B, Espinosa I, Zhu S, Li R, Montgomery KD, Marinelli RJ, Tibshirani R, Hastie T, Jablons DM, Rubin BP, Fletcher CD, West RB, & van de Rijn M (2010). Discovery of molecular subtypes in leiomyosarcoma through integrative molecular profiling. Oncogene, 29, 845–854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Behjati S, Tarpey PS, Haase K, Ye H, Young MD, Alexandrov LB, Farndon SJ, Collord G, Wedge DC, Martincorena I, Cooke SL, Davies H, Mifsud W, Lidgren M, Martin S, Latimer C, Maddison M, Butler AP, Teague JW, Pillay N, Shlien A, McDermott U, Futreal PA, Baumhoer D, Zaikova O, Bjerkehagen B, Myklebost O, Amary MF, Tirabosco R, Van Loo P, Stratton MR, Flanagan AM, & Campbell PJ (2017). Recurrent mutation of IGF signalling genes and distinct patterns of genomic rearrangement in osteosarcoma. Nat Commun, 8, 15936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Behjati S, Tarpey PS, Sheldon H, Martincorena I, Van Loo P, Gundem G, Wedge DC, Ramakrishna M, Cooke SL, Pillay N, Vollan HKM, Papaemmanuil E, Koss H, Bunney TD, Hardy C, Joseph OR, Martin S, Mudie L, Butler A, Teague JW, Patil M, Steers G, Cao Y, Gumbs C, Ingram D, Lazar AJ, Little L, Mahadeshwar H, Protopopov A, Al Sannaa GA, Seth S, Song X, Tang J, Zhang J, Ravi V, Torres KE, Khatri B, Halai D, Roxanis I, Baumhoer D, Tirabosco R, Amary MF, Boshoff C, McDermott U, Katan M, Stratton MR, Futreal PA, Flanagan AM, Harris A, & Campbell PJ (2014). Recurrent PTPRB and PLCG1 mutations in angiosarcoma. Nat Genet, 46, 376–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beird HC, Wu C-C, Ingram DR, Wang W-L, Alimohamed A, Gumbs C, Little L, Song X, Feig BW, & Roland CL (2018). Genomic profiling of dedifferentiated liposarcoma compared to matched well-differentiated liposarcoma reveals higher genomic complexity and a common origin. Molecular Case Studies, 4, a002386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bill KLJ, Garnett J, Meaux I, Ma X, Creighton CJ, Bolshakov S, Barriere C, Debussche L, Lazar AJ, Prudner BC, Casadei L, Braggio D, Lopez G, Zewdu A, Bid H, Lev D, & Pollock RE (2016). SAR405838: A Novel and Potent Inhibitor of the MDM2:p53 Axis for the Treatment of Dedifferentiated Liposarcoma. Clinical Cancer Research, 22, 1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Binayke A, Mishra S, Suman P, Das S, & Chander H (2019). Awakening the “guardian of genome”: reactivation of mutant p53. Cancer Chemother Pharmacol, 83, 1–15. [DOI] [PubMed] [Google Scholar]
- Bista M, Smithson D, Pecak A, Salinas G, Pustelny K, Min J, Pirog A, Finch K, Zdzalik M, Waddell B, Wladyka B, Kedracka-Krok S, Dyer MA, Dubin G, & Guy RK (2012). On the mechanism of action of SJ-172550 in inhibiting the interaction of MDM4 and p53. PLoS One, 7, e37518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blasenbreu S, Baretton GB, Bender C, Haas CJ, Diebold J, & Lohrs U (1998). [TP53 gene aberrations in chondromatous neoplasms: correlation with immunohistochemical p53 accumulation and MDM2 expression]. Verh Dtsch Ges Pathol, 82, 284–289. [PubMed] [Google Scholar]
- Bleeker JS, Quevedo JF, & Folpe AL (2012). “Malignant” perivascular epithelioid cell neoplasm: risk stratification and treatment strategies. Sarcoma, 2012, 541626–541626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bovee JV, Hogendoorn PC, Wunder JS, & Alman BA (2010). Cartilage tumours and bone development: molecular pathology and possible therapeutic targets. Nat Rev Cancer, 10, 481–488. [DOI] [PubMed] [Google Scholar]
- Bozzi F, Ferrari A, Negri T, Conca E, Luca DR, Losa M, Casieri P, Orsenigo M, Lampis A, Meazza C, Casanova M, Pierotti MA, Tamborini E, & Pilotti S (2008). Molecular characterization of synovial sarcoma in children and adolescents: evidence of akt activation. Translational oncology, 1, 95–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bridge JA, Bhatia PS, Anderson JR, & Neff JR (1993). Biologic and clinical significance of cytogenetic and molecular cytogenetic abnormalities in benign and malignant cartilaginous lesions. Cancer Genet Cytogenet, 69, 79–90. [DOI] [PubMed] [Google Scholar]
- Brill E, Gobble R, Angeles C, Lagos-Quintana M, Crago A, Laxa B, Decarolis P, Zhang L, Antonescu C, Socci ND, Taylor BS, Sander C, Koff A, & Singer S (2010). ZIC1 overexpression is oncogenic in liposarcoma. Cancer research, 70, 6891–6901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brohl AS, Patidar R, Turner CE, Wen X, Song YK, Wei JS, Calzone KA, & Khan J (2017). Frequent inactivating germline mutations in DNA repair genes in patients with Ewing sarcoma. Genet Med, 19, 955–958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brohl AS, Solomon DA, Chang W, Wang J, Song Y, Sindiri S, Patidar R, Hurd L, Chen L, Shern JF, Liao H, Wen X, Gerard J, Kim JS, Lopez Guerrero JA, Machado I, Wai DH, Picci P, Triche T, Horvai AE, Miettinen M, Wei JS, Catchpool D, Llombart-Bosch A, Waldman T, & Khan J (2014). The genomic landscape of the Ewing Sarcoma family of tumors reveals recurrent STAG2 mutation. PLoS Genet, 10, e1004475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bui B, Lortal B, Toulmonde M, Italiano A, Antonescu CR, Singer S, Coindre J-M, Schwartz GK, Cioffi A, Maki RG, Le Cesne A, Patrikidou A, Penel N, Isambert N, Bompas E, Duffaud F, & Blay J-Y (2011). Advanced well-differentiated/dedifferentiated liposarcomas: role of chemotherapy and survival. Annals of Oncology, 23, 1601–1607. [DOI] [PubMed] [Google Scholar]
- Burchill SA (2003). Ewing’s sarcoma: diagnostic, prognostic, and therapeutic implications of molecular abnormalities. J Clin Pathol, 56, 96–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bykov VJ, Issaeva N, Zache N, Shilov A, Hultcrantz M, Bergman J, Selivanova G, & Wiman KG (2005). Reactivation of mutant p53 and induction of apoptosis in human tumor cells by maleimide analogs. J Biol Chem, 280, 30384–30391. [DOI] [PubMed] [Google Scholar]
- Bykov VJ, Zache N, Stridh H, Westman J, Bergman J, Selivanova G, & Wiman KG (2005). PRIMA-1(MET) synergizes with cisplatin to induce tumor cell apoptosis. Oncogene, 24, 3484–3491. [DOI] [PubMed] [Google Scholar]
- Bykov VJN, Eriksson SE, Bianchi J, & Wiman KG (2018). Targeting mutant p53 for efficient cancer therapy. Nat Rev Cancer, 18, 89–102. [DOI] [PubMed] [Google Scholar]
- Cam H, Griesmann H, Beitzinger M, Hofmann L, Beinoraviciute-Kellner R, Sauer M, Hüttinger-Kirchhof N, Oswald C, Friedl P, Gattenlöhner S, Burek C, Rosenwald A, & Stiewe T (2006). p53 family members in myogenic differentiation and rhabdomyosarcoma development. Cancer Cell, 10, 281–293. [DOI] [PubMed] [Google Scholar]
- Cancer Genome Atlas Research Network. Electronic address, e. d. s. c., & Cancer Genome Atlas Research, N. (2017). Comprehensive and Integrated Genomic Characterization of Adult Soft Tissue Sarcomas. Cell, 171, 950–965 e928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carneiro A, Francis P, Bendahl PO, Fernebro J, Akerman M, Fletcher C, Rydholm A, Borg A, & Nilbert M (2009). Indistinguishable genomic profiles and shared prognostic markers in undifferentiated pleomorphic sarcoma and leiomyosarcoma: different sides of a single coin? Lab Invest, 89, 668–675. [DOI] [PubMed] [Google Scholar]
- Charytonowicz E, Terry M, Coakley K, Telis L, Remotti F, Cordon-Cardo C, Taub RN, & Matushansky I (2012). PPARγ agonists enhance ET-743–induced adipogenic differentiation in a transgenic mouse model of myxoid round cell liposarcoma. The Journal of Clinical Investigation, 122, 886–898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen X, Bahrami A, Pappo A, Easton J, Dalton J, Hedlund E, Ellison D, Shurtleff S, Wu G, Wei L, Parker M, Rusch M, Nagahawatte P, Wu J, Mao S, Boggs K, Mulder H, Yergeau D, Lu C, Ding L, Edmonson M, Qu C, Wang J, Li Y, Navid F, Daw NC, Mardis ER, Wilson RK, Downing JR, Zhang J, Dyer MA, & St. Jude Children’s Research Hospital-Washington University Pediatric Cancer Genome, P. (2014). Recurrent somatic structural variations contribute to tumorigenesis in pediatric osteosarcoma. Cell Rep, 7, 104–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Z, Guo J, Zhang K, & Guo Y (2016). TP53 Mutations and Survival in Osteosarcoma Patients: A Meta-Analysis of Published Data. Dis Markers, 2016, 4639575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheok CF, Dey A, & Lane DP (2007). Cyclin-dependent kinase inhibitors sensitize tumor cells to nutlin-induced apoptosis: a potent drug combination. Mol Cancer Res, 5, 1133–1145. [DOI] [PubMed] [Google Scholar]
- Chow WA (2018). Chondrosarcoma: biology, genetics, and epigenetics. F1000Res, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chudasama P, Mughal SS, Sanders MA, Hubschmann D, Chung I, Deeg KI, Wong SH, Rabe S, Hlevnjak M, Zapatka M, Ernst A, Kleinheinz K, Schlesner M, Sieverling L, Klink B, Schrock E, Hoogenboezem RM, Kasper B, Heilig CE, Egerer G, Wolf S, von Kalle C, Eils R, Stenzinger A, Weichert W, Glimm H, Groschel S, Kopp HG, Omlor G, Lehner B, Bauer S, Schimmack S, Ulrich A, Mechtersheimer G, Rippe K, Brors B, Hutter B, Renner M, Hohenberger P, Scholl C, & Frohling S (2018). Integrative genomic and transcriptomic analysis of leiomyosarcoma. Nat Commun, 9, 144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chung SW, Kim GC, Kweon S, Lee H, Choi JU, Mahmud F, Chang HW, Kim JW, Son WC, Kim SY, & Byun Y (2018). Metronomic oral doxorubicin in combination of Chk1 inhibitor MK-8776 for p53-deficient breast cancer treatment. Biomaterials, 182, 35–43. [DOI] [PubMed] [Google Scholar]
- Clark J, Rocques PJ, Crew AJ, Gill S, Shipley J, Chan AML, Gusterson BA, & Cooper CS (1994). Identification of novel genes, SYT and SSX, involved in the t(X;18)(p11.2;q11.2) translocation found in human synovial sarcoma. Nature Genetics, 7, 502. [DOI] [PubMed] [Google Scholar]
- Correa H (2016). Li-Fraumeni Syndrome. J Pediatr Genet, 5, 84–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crompton BD, Stewart C, Taylor-Weiner A, Alexe G, Kurek KC, Calicchio ML, Kiezun A, Carter SL, Shukla SA, Mehta SS, Thorner AR, de Torres C, Lavarino C, Sunol M, McKenna A, Sivachenko A, Cibulskis K, Lawrence MS, Stojanov P, Rosenberg M, Ambrogio L, Auclair D, Seepo S, Blumenstiel B, DeFelice M, Imaz-Rosshandler I, Schwarz-Cruz YCA, Rivera MN, Rodriguez-Galindo C, Fleming MD, Golub TR, Getz G, Mora J, & Stegmaier K (2014). The genomic landscape of pediatric Ewing sarcoma. Cancer Discov, 4, 1326–1341. [DOI] [PubMed] [Google Scholar]
- D’Angelo E, & Prat J (2010). Uterine sarcomas: A review. Gynecologic Oncology, 116, 131–139. [DOI] [PubMed] [Google Scholar]
- D’Arcy P, Ryan BA, & Brodin B (2009). Reactivation of p53 function in synovial sarcoma cells by inhibition of p53–HDM2 interaction. Cancer Letters, 275, 285–292. [DOI] [PubMed] [Google Scholar]
- Dai X, Ma W, He X, & Jha RK (2011). Review of therapeutic strategies for osteosarcoma, chondrosarcoma, and Ewing’s sarcoma. Med Sci Monit, 17, RA177–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Danielson LS, Menendez S, Attolini CS-O, Guijarro MV, Bisogna M, Wei J, Socci ND, Levine DA, Michor F, & Hernando E (2010). A Differentiation-Based MicroRNA Signature Identifies Leiomyosarcoma as a Mesenchymal Stem Cell-Related Malignancy. The American Journal of Pathology, 177, 908–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis LE, Nusser KD, Przybyl J, Pittsenbarger J, Hofmann NE, Varma S, Vennam S, Debiec-Rychter M, van de Rijn M, & Davare MA (2019). Discovery and characterization of recurrent, targetable ALK fusions in leiomyosarcoma. Molecular Cancer Research, 17, 676–685. [DOI] [PubMed] [Google Scholar]
- de Alava E, Antonescu CR, Panizo A, Leung D, Meyers PA, Huvos AG, Pardo-Mindan FJ, Healey JH, & Ladanyi M (2000). Prognostic impact of P53 status in Ewing sarcoma. Cancer, 89, 783–792. [PubMed] [Google Scholar]
- de Leeuw B, Balemans M, Weghuis DO, & van Kessel AG (1995). Identification of two alternative fusion genes, SYT-SSX1 and SYT-SSX2, in t(X; 18)(p11.2;q11.2)-positive synoviaol sarcomas. Human Molecular Genetics, 4, 1097–1099. [DOI] [PubMed] [Google Scholar]
- Dei Tos AP (2006). Classification of pleomorphic sarcomas: where are we now? Histopathology, 48, 51–62. [DOI] [PubMed] [Google Scholar]
- Dei Tos AP, Piccinin S, Doglioni C, Vukosavljevic T, Mentzel T, Boiocchi M, & Fletcher CD (1997). Molecular aberrations of the G1-S checkpoint in myxoid and round cell liposarcoma. Am J Pathol, 151, 1531–1539. [PMC free article] [PubMed] [Google Scholar]
- Dewaele M, Tabaglio T, Willekens K, Bezzi M, Teo SX, Low DH, Koh CM, Rambow F, Fiers M, Rogiers A, Radaelli E, Al-Haddawi M, Tan SY, Hermans E, Amant F, Yan H, Lakshmanan M, Koumar RC, Lim ST, Derheimer FA, Campbell RM, Bonday Z, Tergaonkar V, Shackleton M, Blattner C, Marine JC, & Guccione E (2016). Antisense oligonucleotide-mediated MDM4 exon 6 skipping impairs tumor growth. J Clin Invest, 126, 68–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Conza G, Buttarelli M, Monti O, Pellegrino M, Mancini F, Pontecorvi A, Scotlandi K, & Moretti F (2012). IGF-1R/MDM2 relationship confers enhanced sensitivity to RITA in Ewing sarcoma cells. Mol Cancer Ther, 11, 1247–1256. [DOI] [PubMed] [Google Scholar]
- Dijkhuizen T, van den Berg E, Molenaar WM, Oosterhuis JW, Dam A, Wiersema J, Schraffordt Koops H, & de Jong B (1994). Cytogenetics as a tool in the histologic subclassification of chondrosarcomas. Cancer Genet Cytogenet, 76, 100–105. [DOI] [PubMed] [Google Scholar]
- Do PM, Varanasi L, Fan S, Li C, Kubacka I, Newman V, Chauhan K, Daniels SR, Boccetta M, Garrett MR, Li R, & Martinez LA (2012). Mutant p53 cooperates with ETS2 to promote etoposide resistance. Genes Dev, 26, 830–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dobashi Y, Sugimura H, Sato A, Hirabayashi T, Kanda H, Kitagawa T, Kawaguchi N, Imamura T, & Machinami R (1993). Possible association of p53 overexpression and mutation with high-grade chondrosarcoma. Diagn Mol Pathol, 2, 257–263. [PubMed] [Google Scholar]
- Dodd LG (2012). Update on liposarcoma: a review for cytopathologists. Diagnostic cytopathology, 40, 1122–1131. [DOI] [PubMed] [Google Scholar]
- Dodd RD (2016). Emerging targets in sarcoma: Rising to the challenge of RAS signaling in undifferentiated pleomorphic sarcoma. Cancer, 122, 17–19. [DOI] [PubMed] [Google Scholar]
- Duhamel LA, Ye H, Halai D, Idowu BD, Presneau N, Tirabosco R, & Flanagan AM (2012). Frequency of Mouse Double Minute 2 (MDM2) and Mouse Double Minute 4 (MDM4) amplification in parosteal and conventional osteosarcoma subtypes. Histopathology, 60, 357–359. [DOI] [PubMed] [Google Scholar]
- Duran A, Linares JF, Galvez AS, Wikenheiser K, Flores JM, Diaz-Meco MT, & Moscat J (2008). The signaling adaptor p62 is an important NF-kappaB mediator in tumorigenesis. Cancer Cell, 13, 343–354. [DOI] [PubMed] [Google Scholar]
- Egas-Bejar D, & Huh WW (2014). Rhabdomyosarcoma in adolescent and young adult patients: current perspectives. Adolescent health, medicine and therapeutics, 5, 115–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eid JE, & Garcia CB (2015). Reprogramming of mesenchymal stem cells by oncogenes. Semin Cancer Biol, 32, 18–31. [DOI] [PubMed] [Google Scholar]
- El Beaino M, Araujo DM, Lazar AJ, & Lin PP (2017). Synovial Sarcoma: Advances in Diagnosis and Treatment Identification of New Biologic Targets to Improve Multimodal Therapy. Annals of Surgical Oncology, 24, 2145–2154. [DOI] [PubMed] [Google Scholar]
- Esiashvili N, Prabhu R, Kahn S, & Paulino AC (2013). Current strategies and challenges in treatment of childhood rhabdomyosarcoma. Journal of Radiation Oncology, 2, 159–168. [Google Scholar]
- Fan J, & Bertino JR (1999). Modulation of cisplatinum cytotoxicity by p53: effect of p53-mediated apoptosis and DNA repair. Mol Pharmacol, 56, 966–972. [DOI] [PubMed] [Google Scholar]
- Fan L, Yin S, Zhang E, & Hu H (2018). Role of p62 in the regulation of cell death induction. Apoptosis, 23, 187–193. [DOI] [PubMed] [Google Scholar]
- Farid M, & Ngeow J (2016). Sarcomas associated with genetic cancer predisposition syndromes: a review. The oncologist, 21, 1002–1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Felix CA, Kappel CC, Mitsudomi T, Nau MM, Tsokos M, Crouch GD, Nisen PD, Winick NJ, & Helman LJ (1992). Frequency and diversity of p53 mutations in childhood rhabdomyosarcoma. Cancer Research, 52, 2243–2247. [PubMed] [Google Scholar]
- Florou V, & Wilky BA (2018). Current and Future Directions for Angiosarcoma Therapy. Curr Treat Options Oncol, 19, 14. [DOI] [PubMed] [Google Scholar]
- Fu HL, Shao L, Wang Q, Jia T, Li M, & Yang DP (2013). A systematic review of p53 as a biomarker of survival in patients with osteosarcoma. Tumour Biol, 34, 3817–3821. [DOI] [PubMed] [Google Scholar]
- Gao P, Seebacher NA, Hornicek F, Guo Z, & Duan Z (2018). Advances in sarcoma gene mutations and therapeutic targets. Cancer Treatment Reviews, 62, 98–109. [DOI] [PubMed] [Google Scholar]
- Garcia CB, Shaffer CM, Alfaro MP, Smith AL, Sun J, Zhao Z, Young PP, VanSaun MN, & Eid JE (2012). Reprogramming of mesenchymal stem cells by the synovial sarcoma-associated oncogene SYT-SSX2. Oncogene, 31, 2323–2334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gelderblom H, Hogendoorn PC, Dijkstra SD, van Rijswijk CS, Krol AD, Taminiau AH, & Bovee JV (2008). The clinical approach towards chondrosarcoma. Oncologist, 13, 320–329. [DOI] [PubMed] [Google Scholar]
- Genadry KC, Pietrobono S, Rota R, & Linardic CM (2018). Soft Tissue Sarcoma Cancer Stem Cells: An Overview. Frontiers in oncology, 8, 475–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gennari L, Rendina D, Falchetti A, & Merlotti D (2019). Paget’s Disease of Bone. Calcif Tissue Int. [DOI] [PubMed] [Google Scholar]
- Ghadimi MP, Liu P, Peng T, Bolshakov S, Young ED, Torres KE, Colombo C, Hoffman A, Broccoli D, Hornick JL, Lazar AJ, Pisters P, Pollock RE, & Lev D (2011). Pleomorphic liposarcoma. Cancer, 117, 5359–5369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gladdy RA, Qin L-X, Moraco N, Agaram NP, Brennan MF, & Singer S (2013). Predictors of Survival and Recurrence in Primary Leiomyosarcoma. Annals of Surgical Oncology, 20, 1851–1857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goiran T, Duplan E, Rouland L, El Manaa W, Lauritzen I, Dunys J, You H, Checler F, & Alves da Costa C (2018). Nuclear p53-mediated repression of autophagy involves PINK1 transcriptional down-regulation. Cell Death Differ, 25, 873–884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gokgoz N, Wunder JS, Mousses S, Eskandarian S, Bell RS, & Andrulis IL (2001). Comparison of p53 mutations in patients with localized osteosarcoma and metastatic osteosarcoma. Cancer, 92, 2181–2189. [DOI] [PubMed] [Google Scholar]
- Goldstein M, Meller I, Issakov J, & Orr-Urtreger A (2006). Novel Genes Implicated in Embryonal, Alveolar, and Pleomorphic Rhabdomyosarcoma: A Cytogenetic and Molecular Analysis of Primary Tumors. Neoplasia, 8, 332–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gopalakrishnan V, Amini B, Wagner MJ, Nowell EN, Lazar AJ, Lin PP, Benjamin RS, & Araujo DM (2017). Synovial Sarcoma of the Head and Neck: A Single Institution Review. Sarcoma, 2017, 2016752–2016752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goto A, Kanda H, Ishikawa Y, Matsumoto S, Kawaguchi N, Machinami R, Kato Y, & Kitagawa T (1998). Association of loss of heterozygosity at the p53 locus with chemoresistance in osteosarcomas. Jpn J Cancer Res, 89, 539–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Graf Finckenstein F, Shahbazian V, Davicioni E, Ren YX, & Anderson MJ (2007). PAX-FKHR function as pangenes by simultaneously inducing and inhibiting myogenesis. Oncogene, 27, 2004. [DOI] [PubMed] [Google Scholar]
- Greditzer HG 3rd, McLeod RA, Unni KK, & Beabout JW (1983). Bone sarcomas in Paget disease. Radiology, 146, 327–333. [DOI] [PubMed] [Google Scholar]
- Grellety T, Laroche-Clary A, Chaire V, Lagarde P, Chibon F, Neuville A, & Italiano A (2015). PRIMA-1(MET) induces death in soft-tissue sarcomas cell independent of p53. BMC cancer, 15, 684–684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grossmann AH, Layfield LJ, & Randall RL (2012). Classification, molecular characterization, and the significance of pten alteration in leiomyosarcoma. Sarcoma, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gruener G, & Camacho P (2014). Paget’s disease of bone. Handb Clin Neurol, 119, 529–540. [DOI] [PubMed] [Google Scholar]
- Grunewald TGP, Cidre-Aranaz F, Surdez D, Tomazou EM, de Alava E, Kovar H, Sorensen PH, Delattre O, & Dirksen U (2018). Ewing sarcoma. Nat Rev Dis Primers, 4, 5. [DOI] [PubMed] [Google Scholar]
- Guerzoni C, Fiori V, Terracciano M, Manara MC, Moricoli D, Pasello M, Sciandra M, Nicoletti G, Gellini M, Dominici S, Chiodoni C, Fornasari PM, Lollini PL, Colombo MP, Picci P, Cianfriglia M, Magnani M, & Scotlandi K (2015). CD99 triggering in Ewing sarcoma delivers a lethal signal through p53 pathway reactivation and cooperates with doxorubicin. Clin Cancer Res, 21, 146–156. [DOI] [PubMed] [Google Scholar]
- Guha T, & Malkin D (2017). Inherited TP53 Mutations and the Li-Fraumeni Syndrome. Cold Spring Harb Perspect Med, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gunderson C, McMahon C, Elvin JA, Ross JS, Moore KN, Gay LM, Ali S, & Ramkissoon S (2016). Comprehensive genomic profiling of uterine leiomyosarcomas identifies opportunities for personalized therapies. Annals of Oncology, 27. [Google Scholar]
- Guo W, Wang X, & Feng C (1996). P53 gene abnormalities in osteosarcoma. Chin Med J (Engl), 109, 752–755. [PubMed] [Google Scholar]
- Guo X, Jo VY, Mills AM, Zhu SX, Lee C-H, Espinosa I, Nucci MR, Varma S, Forgó E, Hastie T, Anderson S, Ganjoo K, Beck AH, West RB, Fletcher CD, & van de Rijn M (2015). Clinically Relevant Molecular Subtypes in Leiomyosarcoma. Clinical cancer research : an official journal of the American Association for Cancer Research, 21, 3501–3511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gurpinar E, & Vousden KH (2015). Hitting cancers’ weak spots: vulnerabilities imposed by p53 mutation. Trends Cell Biol, 25, 486–495. [DOI] [PubMed] [Google Scholar]
- Hameed M, Ulger C, Yasar D, Limaye N, Kurvathi R, Streck D, Benevenia J, Patterson F, Dermody JJ, & Toruner GA (2009). Genome profiling of chondrosarcoma using oligonucleotide array-based comparative genomic hybridization. Cancer Genet Cytogenet, 192, 56–59. [DOI] [PubMed] [Google Scholar]
- Hansen MF, Nellissery MJ, & Bhatia P (1999). Common mechanisms of osteosarcoma and Paget’s disease. J Bone Miner Res, 14 Suppl 2, 39–44. [DOI] [PubMed] [Google Scholar]
- Harada N, Watanabe Y, Yoshimura Y, Sakumoto H, Makishima F, Tsuchiya M, Nakanishi K, Nakanishi M, & Aoki Y (2011). Identification of a checkpoint modulator with synthetic lethality to p53 mutants. Anticancer Drugs, 22, 986–994. [DOI] [PubMed] [Google Scholar]
- Harrison DJ, Geller DS, Gill JD, Lewis VO, & Gorlick R (2018). Current and future therapeutic approaches for osteosarcoma. Expert Rev Anticancer Ther, 18, 39–50. [DOI] [PubMed] [Google Scholar]
- Haupt S, Hernandez OM, Vijayakumaran R, Keam S, & Haupt Y (2019). The long and the short of it: the MDM4 tail so far. J Mol Cell Biol. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He Y, de Castro LF, Shin MH, Dubois W, Yang HH, Jiang S, Mishra PJ, Ren L, Gou H, Lal A, Khanna C, Merlino G, Lee M, Robey PG, & Huang J (2015). p53 loss increases the osteogenic differentiation of bone marrow stromal cells. Stem Cells, 33, 1304–1319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hientz K, Mohr A, Bhakta-Guha D, & Efferth T (2017). The role of p53 in cancer drug resistance and targeted chemotherapy. Oncotarget, 8, 8921–8946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ho L, Stojanovski A, Whetstone H, Wei QX, Mau E, Wunder JS, & Alman B (2009). Gli2 and p53 cooperate to regulate IGFBP-3- mediated chondrocyte apoptosis in the progression from benign to malignant cartilage tumors. Cancer Cell, 16, 126–136. [DOI] [PubMed] [Google Scholar]
- Hu F, Guo XL, Zhang SS, Zhao QD, Li R, Xu Q, & Wei LX (2017). Suppression of p53 potentiates chemosensitivity in nutrient-deprived cholangiocarcinoma cells via inhibition of autophagy. Oncol Lett, 14, 1959–1966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang HY, Illei PB, Zhao Z, Mazumdar M, Huvos AG, Healey JH, Wexler LH, Gorlick R, Meyers P, & Ladanyi M (2005). Ewing sarcomas with p53 mutation or p16/p14ARF homozygous deletion: a highly lethal subset associated with poor chemoresponse. J Clin Oncol, 23, 548–558. [DOI] [PubMed] [Google Scholar]
- Huang SC, Zhang L, Sung YS, Chen CL, Kao YC, Agaram NP, Singer S, Tap WD, D’Angelo S, & Antonescu CR (2016). 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, 40, 645–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hung J, & Anderson R (1997). p53: functions, mutations and sarcomas. Acta Orthop Scand Suppl, 273, 68–73. [DOI] [PubMed] [Google Scholar]
- Ignatius MS, Hayes MN, Moore FE, Tang Q, Garcia SP, Blackburn PR, Baxi K, Wang L, Jin A, Ramakrishnan A, Reeder S, Chen Y, Nielsen GP, Chen EY, Hasserjian RP, Tirode F, Ekker SC, & Langenau DM (2018). tp53 deficiency causes a wide tumor spectrum and increases embryonal rhabdomyosarcoma metastasis in zebrafish. eLife, 7, e37202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- In GK, Hu JS, & Tseng WW (2017). Treatment of advanced, metastatic soft tissue sarcoma: latest evidence and clinical considerations. Therapeutic advances in medical oncology, 9, 533–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ito M, Barys L, O’Reilly T, Young S, Gorbatcheva B, Monahan J, Zumstein-Mecker S, Choong PF, Dickinson I, Crowe P, Hemmings C, Desai J, Thomas DM, & Lisztwan J (2011). Comprehensive mapping of p53 pathway alterations reveals an apparent role for both SNP309 and MDM2 amplification in sarcomagenesis. Clin Cancer Res, 17, 416–426. [DOI] [PubMed] [Google Scholar]
- Iyer SV, Parrales A, Begani P, Narkar A, Adhikari AS, Martinez LA, & Iwakuma T (2016). Allele-specific silencing of mutant p53 attenuates dominant-negative and gain-of-function activities. Oncotarget, 7, 5401–5415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jamil N, Howie S, & Salter DM (2010). Therapeutic molecular targets in human chondrosarcoma. Int J Exp Pathol, 91, 387–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jang KS, Min KW, Jang SH, Paik SS, Tae K, Jang SJ, & Park MH (2007). Primary synovial sarcoma of the thyroid gland. Journal of Korean medical science, 22 Suppl, S154–S158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang H, Reinhardt HC, Bartkova J, Tommiska J, Blomqvist C, Nevanlinna H, Bartek J, Yaffe MB, & Hemann MT (2009). The combined status of ATM and p53 link tumor development with therapeutic response. Genes Dev, 23, 1895–1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kadoch C, & Crabtree GR (2015). Mammalian SWI/SNF chromatin remodeling complexes and cancer: Mechanistic insights gained from human genomics. Science advances, 1, e1500447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kanojia D, Nagata Y, Garg M, Lee DH, Sato A, Yoshida K, Sato Y, Sanada M, Mayakonda A, Bartenhagen C, Klein H-U, Doan NB, Said JW, Mohith S, Gunasekar S, Shiraishi Y, Chiba K, Tanaka H, Miyano S, Myklebost O, Yang H, Dugas M, Meza-Zepeda LA, Silberman AW, Forscher C, Tyner JW, Ogawa S, & Koeffler HP (2015). Genomic landscape of liposarcoma. Oncotarget, 6, 42429–42444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karan G, Wang H, Chakrabarti A, Karan S, Liu Z, Xia Z, Gundluru M, Moreton S, Saunthararajah Y, Jackson MW, Agarwal MK, & Wald DN (2016). Identification of a Small Molecule That Overcomes HdmX-Mediated Suppression of p53. Mol Cancer Ther, 15, 574–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kawai A, Noguchi M, Beppu Y, Yokoyama R, Mukai K, Hirohashi S, Inoue H, & Fukuma H (1994). Nuclear immunoreaction of p53 protein in soft tissue sarcomas. A possible prognostic factor. Cancer, 73, 2499–2505. [DOI] [PubMed] [Google Scholar]
- Kim MJ, Cho KJ, Ayala AG, & Ro JY (2011). Chondrosarcoma: with updates on molecular genetics. Sarcoma, 2011, 405437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim MP, & Lozano G (2018). Mutant p53 partners in crime. Cell Death Differ, 25, 161–168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kollár A, & Benson C (2014). Current management options for liposarcoma and challenges for the future. Expert Review of Anticancer Therapy, 14, 297–306. [DOI] [PubMed] [Google Scholar]
- Komuro H, Hayashi Y, Kawamura M, Hayashi K, Kaneko Y, Kamoshita S, Hanada R, Yamamoto K, Hongo T, Yamada M, & et al. (1993). Mutations of the p53 gene are involved in Ewing’s sarcomas but not in neuroblastomas. Cancer Res, 53, 5284–5288. [PubMed] [Google Scholar]
- Konomoto T, Fukuda T, Hayashi K, Kumazawa J, & Tsuneyoshi M (1998). Leiomyosarcoma in soft tissue: examination of p53 status and cell proliferating factors in different locations. Hum Pathol, 29, 74–81. [DOI] [PubMed] [Google Scholar]
- Kovar H, Auinger A, Jug G, Aryee D, Zoubek A, Salzer-Kuntschik M, & Gadner H (1993). Narrow spectrum of infrequent p53 mutations and absence of MDM2 amplification in Ewing tumours. Oncogene, 8, 2683–2690. [PubMed] [Google Scholar]
- Kridis WB, Toumi N, Chaari H, Khanfir A, Ayadi K, Keskes H, Boudawara T, Daoud J, & Frikha M (2017). A Review of Ewing Sarcoma Treatment: Is it Still a Subject of Debate? Rev Recent Clin Trials, 12, 19–23. [DOI] [PubMed] [Google Scholar]
- Krieg AH, Hefti F, Speth BM, Jundt G, Guillou L, Exner UG, von Hochstetter AR, Cserhati MD, Fuchs B, Mouhsine E, Kaelin A, Klenke FM, & Siebenrock KA (2011). Synovial sarcomas usually metastasize after >5 years: a multicenter retrospective analysis with minimum follow-up of 10 years for survivors. Annals of Oncology, 22, 458–467. [DOI] [PubMed] [Google Scholar]
- Ladanyi M, Lewis R, Jhanwar SC, Gerald W, Huvos AG, & Healey JH (1995). MDM2 and CDK4 gene amplification in Ewing’s sarcoma. J Pathol, 175, 211–217. [DOI] [PubMed] [Google Scholar]
- Landuzzi L, Ianzano ML, Nicoletti G, Palladini A, Grosso V, Ranieri D, Dall’Ora M, Raschi E, Laranga R, Gambarotti M, Picci P, De Giovanni C, Nanni P, & Lollini PL (2014). Genetic prevention of lymphoma in p53 knockout mice allows the early development of p53-related sarcomas. Oncotarget, 5, 11924–11938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lane D, & Levine A (2010). p53 Research: the past thirty years and the next thirty years. Cold Spring Harb Perspect Biol, 2, a000893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lane DP, Brown CJ, Verma C, & Cheok CF (2011). New insights into p53 based therapy. Discov Med, 12, 107–117. [PubMed] [Google Scholar]
- Lang GA, Iwakuma T, Suh YA, Liu G, Rao VA, Parant JM, Valentin-Vega YA, Terzian T, Caldwell LC, Strong LC, El-Naggar AK, & Lozano G (2004). Gain of function of a p53 hot spot mutation in a mouse model of Li-Fraumeni syndrome. Cell, 119, 861–872. [DOI] [PubMed] [Google Scholar]
- Laroche-Clary A, Chaire V, Algeo M-P, Derieppe M-A, Loarer FL, & Italiano A (2017). Combined targeting of MDM2 and CDK4 is synergistic in dedifferentiated liposarcomas. Journal of hematology & oncology, 10, 123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le Guellec S, Chibon F, Ouali M, Perot G, Decouvelaere AV, Robin YM, Larousserie F, Terrier P, Coindre JM, & Neuville A (2014). Are peripheral purely undifferentiated pleomorphic sarcomas with MDM2 amplification dedifferentiated liposarcomas? Am J Surg Pathol, 38, 293–304. [DOI] [PubMed] [Google Scholar]
- Lenos K, Grawenda AM, Lodder K, Kuijjer ML, Teunisse AF, Repapi E, Grochola LF, Bartel F, Hogendoorn PC, Wuerl P, Taubert H, Cleton-Jansen AM, Bond GL, & Jochemsen AG (2012). Alternate splicing of the p53 inhibitor HDMX offers a superior prognostic biomarker than p53 mutation in human cancer. Cancer Res, 72, 4074–4084. [DOI] [PubMed] [Google Scholar]
- Lenos K, & Jochemsen AG (2011). Functions of MDMX in the modulation of the p53-response. J Biomed Biotechnol, 2011, 876173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lerman DM, Monument MJ, McIlvaine E, Liu XQ, Huang D, Monovich L, Beeler N, Gorlick RG, Marina NM, Womer RB, Bridge JA, Krailo MD, Randall RL, Lessnick SL, & Children’s Oncology Group Ewing Sarcoma Biology, C. (2015). Tumoral TP53 and/or CDKN2A alterations are not reliable prognostic biomarkers in patients with localized Ewing sarcoma: a report from the Children’s Oncology Group. Pediatr Blood Cancer, 62, 759–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lessnick SL, Dacwag CS, & Golub TR (2002). The Ewing’s sarcoma oncoprotein EWS/FLI induces a p53-dependent growth arrest in primary human fibroblasts. Cancer Cell, 1, 393–401. [DOI] [PubMed] [Google Scholar]
- Leuschner I, Langhans I, Schmitz R, Harms D, Mattke A, & Treuner J (2003). p53 and mdm-2 expression in Rhabdomyosarcoma of childhood and adolescence: clinicopathologic study by the Kiel Pediatric Tumor Registry and the German Cooperative Soft Tissue Sarcoma Study. Pediatric and Developmental Pathology, 6, 128–136. [DOI] [PubMed] [Google Scholar]
- Lewis JJ, Antonescu CR, Leung DHY, Blumberg D, Healey JH, Woodruff JM, & Brennan MF (2000). Synovial Sarcoma: A Multivariate Analysis of Prognostic Factors in 112 Patients With Primary Localized Tumors of the Extremity. Journal of Clinical Oncology, 18, 2087–2094. [DOI] [PubMed] [Google Scholar]
- Li Q, Zhang Y, El-Naggar AK, Xiong S, Yang P, Jackson JG, Chau G, & Lozano G (2014). Therapeutic efficacy of p53 restoration in Mdm2-overexpressing tumors. Mol Cancer Res, 12, 901–911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Li X, Fan G, Fukushi J, Matsumoto Y, Iwamoto Y, & Zhu Y (2012). Impairment of p53 acetylation by EWS-Fli1 chimeric protein in Ewing family tumors. Cancer Lett, 320, 14–22. [DOI] [PubMed] [Google Scholar]
- Li Y, Tanaka K, Fan X, Nakatani F, Li X, Nakamura T, Takasaki M, Yamamoto S, & Iwamoto Y (2010). Inhibition of the transcriptional function of p53 by EWS-Fli1 chimeric protein in Ewing Family Tumors. Cancer Lett, 294, 57–65. [DOI] [PubMed] [Google Scholar]
- Lin PP, Wang Y, & Lozano G (2011). Mesenchymal Stem Cells and the Origin of Ewing’s Sarcoma. Sarcoma, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu S, Cai C, Yan G, Zhou Z, Wan Y, Chen V, Chen L, Cooper GF, Obeid LM, Hannun YA, Lee AV, & Lu X (2016). Signal-Oriented Pathway Analyses Reveal a Signaling Complex as a Synthetic Lethal Target for p53 Mutations. Cancer Res, 76, 6785–6794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lye KL, Nordin N, Vidyadaran S, & Thilakavathy K (2016). Mesenchymal stem cells: From stem cells to sarcomas. Cell Biol Int, 40, 610–618. [DOI] [PubMed] [Google Scholar]
- Macchia G, Severgnini M, Purgato S, Tolomeo D, Casciaro H, Cifola I, L’abbate A, Loverro A, Palumbo O, & Carella M (2018). The hidden genomic and transcriptomic plasticity of giant marker chromosomes in cancer. Genetics, 208, 951–961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mäkinen N, Aavikko M, Heikkinen T, Taipale M, Taipale J, Koivisto-Korander R, Bützow R, & Vahteristo P (2016). Exome sequencing of uterine leiomyosarcomas identifies frequent mutations in TP53, ATRX, and MED12. PLoS genetics, 12, e1005850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mäkinen N, Kämpjärvi K, Frizzell N, Bützow R, & Vahteristo P (2017). Characterization of MED12, HMGA2, and FH alterations reveals molecular variability in uterine smooth muscle tumors. Molecular Cancer, 16, 101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malkin D, Li FP, Strong LC, Fraumeni JF Jr., Nelson CE, Kim DH, Kassel J, Gryka MA, Bischoff FZ, Tainsky MA, & et al. (1990). Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science, 250, 1233–1238. [DOI] [PubMed] [Google Scholar]
- Mandahl N, Magnusson L, Nilsson J, Viklund B, Arbajian E, von Steyern FV, Isaksson A, & Mertens F (2017). Scattered genomic amplification in dedifferentiated liposarcoma. Molecular cytogenetics, 10, 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mankin HJ, Casas-Ganem J, Kim J-I, Gebhardt MC, Hornicek FJ, & Zeegen EN (2004). Leiomyosarcoma of somatic soft tissues. Clinical orthopaedics and related research, 421, 225–231. [DOI] [PubMed] [Google Scholar]
- Mantovani F, Collavin L, & Del Sal G (2019). Mutant p53 as a guardian of the cancer cell. Cell Death Differ, 26, 199–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martin JW, Squire JA, & Zielenska M (2012). The genetics of osteosarcoma. Sarcoma, 2012, 627254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Masuda H, Miller C, Koeffler HP, Battifora H, & Cline MJ (1987). Rearrangement of the p53 gene in human osteogenic sarcomas. Proc Natl Acad Sci U S A, 84, 7716–7719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matushansky I, Charytonowicz E, Mills J, Siddiqi S, Hricik T, & Cordon-Cardo C (2009). MFH classification: differentiating undifferentiated pleomorphic sarcoma in the 21st Century. Expert Rev Anticancer Ther, 9, 1135–1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mazzoleni S, Bisogno G, Garaventa A, Cecchetto G, Ferrari A, Sotti G, Donfrancesco A, Madon E, Casula L, Carli M, & Associazione Italiana di Ematologia e Oncologia Pediatrica Soft Tissue Sarcoma, C. (2005). Outcomes and prognostic factors after recurrence in children and adolescents with nonmetastatic rhabdomyosarcoma. Cancer, 104, 183–190. [DOI] [PubMed] [Google Scholar]
- McGovern Y, Zhou CD, & Jones RL (2017). Systemic Therapy in Metastatic or Unresectable Well-Differentiated/Dedifferentiated Liposarcoma. Frontiers in oncology, 7, 292–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McManus S, & Roux S (2012). The adaptor protein p62/SQSTM1 in osteoclast signaling pathways. J Mol Signal, 7, 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mehren M. v., & Joensuu H (2018). Gastrointestinal Stromal Tumors. Journal of Clinical Oncology, 36, 136–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mendoza S, Konishi T, Dernell WS, Withrow SJ, & Miller CW (1998). Status of the p53, Rb and MDM2 genes in canine osteosarcoma. Anticancer Res, 18, 4449–4453. [PubMed] [Google Scholar]
- Meyer M, & Seetharam M (2019). First-Line Therapy for Metastatic Soft Tissue Sarcoma. Curr Treat Options Oncol, 20, 6. [DOI] [PubMed] [Google Scholar]
- Miettinen M, & Fetsch JF (2006). Evaluation of biological potential of smooth muscle tumours. Histopathology, 48, 97–105. [DOI] [PubMed] [Google Scholar]
- Miller CW, Aslo A, Won A, Tan M, Lampkin B, & Koeffler HP (1996). Alterations of the p53, Rb and MDM2 genes in osteosarcoma. J Cancer Res Clin Oncol, 122, 559–565. [DOI] [PubMed] [Google Scholar]
- Mirabello L, Koster R, Moriarity BS, Spector LG, Meltzer PS, Gary J, Machiela MJ, Pankratz N, Panagiotou OA, Largaespada D, Wang Z, Gastier-Foster JM, Gorlick R, Khanna C, de Toledo SR, Petrilli AS, Patino-Garcia A, Sierrasesumaga L, Lecanda F, Andrulis IL, Wunder JS, Gokgoz N, Serra M, Hattinger C, Picci P, Scotlandi K, Flanagan AM, Tirabosco R, Amary MF, Halai D, Ballinger ML, Thomas DM, Davis S, Barkauskas DA, Marina N, Helman L, Otto GM, Becklin KL, Wolf NK, Weg MT, Tucker M, Wacholder S, Fraumeni JF Jr., Caporaso NE, Boland JF, Hicks BD, Vogt A, Burdett L, Yeager M, Hoover RN, Chanock SJ, & Savage SA (2015). A Genome-Wide Scan Identifies Variants in NFIB Associated with Metastasis in Patients with Osteosarcoma. Cancer Discov, 5, 920–931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mirabello L, Troisi RJ, & Savage SA (2009). Osteosarcoma incidence and survival rates from 1973 to 2004: data from the Surveillance, Epidemiology, and End Results Program. Cancer, 115, 1531–1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mirabello L, Yeager M, Mai PL, Gastier-Foster JM, Gorlick R, Khanna C, Patino-Garcia A, Sierrasesumaga L, Lecanda F, Andrulis IL, Wunder JS, Gokgoz N, Barkauskas DA, Zhang X, Vogt A, Jones K, Boland JF, Chanock SJ, & Savage SA (2015). Germline TP53 variants and susceptibility to osteosarcoma. J Natl Cancer Inst, 107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miura Y, Keira Y, Ogino J, Nakanishi K, Noguchi H, Inoue T, & Hasegawa T (2012). Detection of specific genetic abnormalities by fluorescence in situ hybridization in soft tissue tumors. Pathol Int, 62, 16–27. [DOI] [PubMed] [Google Scholar]
- Miyajima K, Tamiya S, Oda Y, Adachi T, Konomoto T, Toyoshiba H, Masuda K, & Tsuneyoshi M (2001). Relative quantitation of p53 and MDM2 gene expression in leiomyosarcoma; real-time semi-quantitative reverse transcription-polymerase chain reaction. Cancer Lett, 164, 177–188. [DOI] [PubMed] [Google Scholar]
- Momtahen S, Curtin J, & Mittal K (2016). Current Chemotherapy and Potential New Targets in Uterine Leiomyosarcoma. Journal of clinical medicine research, 8, 181–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morandell S, Reinhardt HC, Cannell IG, Kim JS, Ruf DM, Mitra T, Couvillon AD, Jacks T, & Yaffe MB (2013). A reversible gene-targeting strategy identifies synthetic lethal interactions between MK2 and p53 in the DNA damage response in vivo. Cell Rep, 5, 868–877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morrow JJ, & Khanna C (2015). Osteosarcoma Genetics and Epigenetics: Emerging Biology and Candidate Therapies. Crit Rev Oncog, 20, 173–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mortus JR, Zhang Y, & Hughes DP (2014). Developmental pathways hijacked by osteosarcoma. Adv Exp Med Biol, 804, 93–118. [DOI] [PubMed] [Google Scholar]
- Muller CR, Paulsen EB, Noordhuis P, Pedeutour F, Saeter G, & Myklebost O (2007). Potential for treatment of liposarcomas with the MDM2 antagonist Nutlin-3A. Int J Cancer, 121, 199–205. [DOI] [PubMed] [Google Scholar]
- Murali R, Chandramohan R, Moller I, Scholz SL, Berger M, Huberman K, Viale A, Pirun M, Socci ND, Bouvier N, Bauer S, Artl M, Schilling B, Schimming T, Sucker A, Schwindenhammer B, Grabellus F, Speicher MR, Schaller J, Hillen U, Schadendorf D, Mentzel T, Cheng DT, Wiesner T, & Griewank KG (2015). Targeted massively parallel sequencing of angiosarcomas reveals frequent activation of the mitogen activated protein kinase pathway. Oncotarget, 6, 36041–36052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murphey MD (2007). World Health Organization classification of bone and soft tissue tumors: modifications and implications for radiologists. Semin Musculoskelet Radiol, 11, 201–214. [DOI] [PubMed] [Google Scholar]
- Murphey MD, Gibson MS, Jennings BT, Crespo-Rodríguez AM, Fanburg-Smith J, & Gajewski DA (2006). Imaging of Synovial Sarcoma with Radiologic-Pathologic Correlation. RadioGraphics, 26, 1543–1565. [DOI] [PubMed] [Google Scholar]
- Naka N, Takenaka S, Araki N, Miwa T, Hashimoto N, Yoshioka K, Joyama S, Hamada K. i., Tsukamoto Y, Tomita Y, Ueda T, Yoshikawa H, & Itoh K (2010). Synovial Sarcoma Is a Stem Cell Malignancy. STEM CELLS, 28, 1119–1131. [DOI] [PubMed] [Google Scholar]
- Naka N, Tomita Y, Nakanishi H, Araki N, Hongyo T, Ochi T, & Aozasa K (1997). Mutations of p53 tumor-suppressor gene in angiosarcoma. Int J Cancer, 71, 952–955. [DOI] [PubMed] [Google Scholar]
- Nakayama T, Toguchida J, Wadayama B, Kanoe H, Kotoura Y, & Sasaki MS (1995). MDM2 gene amplification in bone and soft-tissue tumors: association with tumor progression in differentiated adipose-tissue tumors. Int J Cancer, 64, 342–346. [DOI] [PubMed] [Google Scholar]
- Nazeri E, Gouran Savadkoohi M, Majidzadeh AK, & Esmaeili R (2018). Chondrosarcoma: An overview of clinical behavior, molecular mechanisms mediated drug resistance and potential therapeutic targets. Crit Rev Oncol Hematol, 131, 102–109. [DOI] [PubMed] [Google Scholar]
- Neilsen PM, Pishas KI, Callen DF, & Thomas DM (2011). Targeting the p53 Pathway in Ewing Sarcoma. Sarcoma, 2011, 746939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ng VY, Scharschmidt TJ, Mayerson JL, & Fisher JL (2013). Incidence and survival in sarcoma in the United States: a focus on musculoskeletal lesions. Anticancer Res, 33, 2597–2604. [PubMed] [Google Scholar]
- Nishitoh H (2011). CHOP is a multifunctional transcription factor in the ER stress response. The Journal of Biochemistry, 151, 217–219. [DOI] [PubMed] [Google Scholar]
- Nyquist KB, Panagopoulos I, Thorsen J, Haugom L, Gorunova L, Bjerkehagen B, Fossa A, Guriby M, Nome T, Lothe RA, Skotheim RI, Heim S, & Micci F (2012). Whole-transcriptome sequencing identifies novel IRF2BP2-CDX1 fusion gene brought about by translocation t(1;5)(q42;q32) in mesenchymal chondrosarcoma. PLoS One, 7, e49705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’neill C, McBride H, Connolly L, & McCluggage W (2007). Uterine leiomyosarcomas are characterized by high p16, p53 and MIB1 expression in comparison with usual leiomyomas, leiomyoma variants and smooth muscle tumours of uncertain malignant potential. Histopathology, 50, 851–858. [DOI] [PubMed] [Google Scholar]
- Obrador-Hevia A, Martinez-Font E, Felipe-Abrio I, Calabuig-Farinas S, Serra-Sitjar M, Lopez-Guerrero JA, Ramos R, Alemany R, & Martin-Broto J (2015). RG7112, a small-molecule inhibitor of MDM2, enhances trabectedin response in soft tissue sarcomas. Cancer Invest, 33, 440–450. [DOI] [PubMed] [Google Scholar]
- Ognjanovic S, Martel G, Manivel C, Olivier M, Langer E, & Hainaut P (2012). Low Prevalence of TP53 Mutations and MDM2 Amplifications in Pediatric Rhabdomyosarcoma. Sarcoma, 2012, 492086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ognjanovic S, Olivier M, Bergemann TL, & Hainaut P (2012). Sarcomas in TP53 germline mutation carriers: a review of the IARC TP53 database. Cancer, 118, 1387–1396. [DOI] [PubMed] [Google Scholar]
- Olive KP, Tuveson DA, Ruhe ZC, Yin B, Willis NA, Bronson RT, Crowley D, & Jacks T (2004). Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome. Cell, 119, 847–860. [DOI] [PubMed] [Google Scholar]
- Olsson L, Paulsson K, Bovee JV, & Nord KH (2011). Clonal evolution through loss of chromosomes and subsequent polyploidization in chondrosarcoma. PLoS One, 6, e24977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Origanti S, Cai SR, Munir AZ, White LS, & Piwnica-Worms H (2013). Synthetic lethality of Chk1 inhibition combined with p53 and/or p21 loss during a DNA damage response in normal and tumor cells. Oncogene, 32, 577–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oshiro Y, Chaturvedi V, Hayden D, Nazeer T, Johnson M, Johnston DA, Ordonez NG, Ayala AG, & Czerniak B (1998). Altered p53 is associated with aggressive behavior of chondrosarcoma: a long term follow-up study. Cancer, 83, 2324–2334. [DOI] [PubMed] [Google Scholar]
- Ouslander JG, & Beck JC (1982). Paget’s disease of bone. J Am Geriatr Soc, 30, 410–414. [DOI] [PubMed] [Google Scholar]
- Overholtzer M, Rao PH, Favis R, Lu XY, Elowitz MB, Barany F, Ladanyi M, Gorlick R, & Levine AJ (2003). The presence of p53 mutations in human osteosarcomas correlates with high levels of genomic instability. Proc Natl Acad Sci U S A, 100, 11547–11552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ozaki T (2015). Diagnosis and treatment of Ewing sarcoma of the bone: a review article. J Orthop Sci, 20, 250–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pakos EE, Kyzas PA, & Ioannidis JP (2004). Prognostic significance of TP53 tumor suppressor gene expression and mutations in human osteosarcoma: a meta-analysis. Clin Cancer Res, 10, 6208–6214. [DOI] [PubMed] [Google Scholar]
- Panagopoulos I, Mertens F, Isaksson M, Domanski HA, Brosjo O, Heim S, Bjerkehagen B, Sciot R, Dal Cin P, Fletcher JA, Fletcher CD, & Mandahl N (2002). Molecular genetic characterization of the EWS/CHN and RBP56/CHN fusion genes in extraskeletal myxoid chondrosarcoma. Genes Chromosomes Cancer, 35, 340–352. [DOI] [PubMed] [Google Scholar]
- Pani L, Horal M, & Loeken MR (2002). Rescue of neural tube defects in Pax-3-deficient embryos by p53 loss of function: implications for Pax-3- dependent development and tumorigenesis. Genes & development, 16, 676–680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pant V, Larsson CA, Aryal N, Xiong S, You MJ, Quintas-Cardama A, & Lozano G (2017). Tumorigenesis promotes Mdm4-S overexpression. Oncotarget, 8, 25837–25847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parrales A, & Iwakuma T (2015). Targeting Oncogenic Mutant p53 for Cancer Therapy. Front Oncol, 5, 288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parrales A, Ranjan A, Iyer SV, Padhye S, Weir SJ, Roy A, & Iwakuma T (2016). DNAJA1 controls the fate of misfolded mutant p53 through the mevalonate pathway. Nat Cell Biol, 18, 1233–1243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pasello M, Manara MC, & Scotlandi K (2018). CD99 at the crossroads of physiology and pathology. J Cell Commun Signal, 12, 55–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patil N, Ahmed Kabeer Rasheed S, Abba M, Hendrik Leupold J, Schwarzbach M, & Allgayer H (2014). A mechanistic study on the metastasis inducing function of FUS-CHOP fusion protein in liposarcoma. International Journal of Cancer, 134, 2808–2819. [DOI] [PubMed] [Google Scholar]
- Patterson H, Gill S, Fisher C, Law MG, Jayatilake H, Fletcher CD, Thomas M, Grimer R, Gusterson BA, & Cooper CS (1994). Abnormalities of the p53 MDM2 and DCC genes in human leiomyosarcomas. Br J Cancer, 69, 1052–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pedeutour F, Forus A, Coindre JM, Berner JM, Nicolo G, Michiels JF, Terrier P, Ranchere‐Vince D, Collin F, & Myklebost O (1999). Structure of the supernumerary ring and giant rod chromosomes in adipose tissue tumors. Genes, Chromosomes and Cancer, 24, 30–41. [PubMed] [Google Scholar]
- Peng T, Zhang P, Liu J, Nguyen T, Bolshakov S, Belousov R, Young ED, Wang X, Brewer K, Lopez-Terrada DH, Oliveira AM, Lazar AJ, & Lev D (2011). An experimental model for the study of well-differentiated and dedifferentiated liposarcoma; deregulation of targetable tyrosine kinase receptors. Lab Invest, 91, 392–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pérez-Losada J, Pintado B, Gutiérrez-Adán A, Flores T, Bañares-González B, Calabia del Campo J, Martín-Martín JF, Battaner E, & Sánchez-García I (2000). The chimeric FUS/TLS-CHOP fusion protein specifically induces liposarcomas in transgenic mice. Oncogene, 19, 2413. [DOI] [PubMed] [Google Scholar]
- Perot G, Chibon F, Montero A, Lagarde P, de The H, Terrier P, Guillou L, Ranchere D, Coindre JM, & Aurias A (2010). Constant p53 pathway inactivation in a large series of soft tissue sarcomas with complex genetics. Am J Pathol, 177, 2080–2090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perry JA, Kiezun A, Tonzi P, Van Allen EM, Carter SL, Baca SC, Cowley GS, Bhatt AS, Rheinbay E, Pedamallu CS, Helman E, Taylor-Weiner A, McKenna A, DeLuca DS, Lawrence MS, Ambrogio L, Sougnez C, Sivachenko A, Walensky LD, Wagle N, Mora J, de Torres C, Lavarino C, Dos Santos Aguiar S, Yunes JA, Brandalise SR, Mercado-Celis GE, Melendez-Zajgla J, Cardenas-Cardos R, Velasco-Hidalgo L, Roberts CW, Garraway LA, Rodriguez-Galindo C, Gabriel SB, Lander ES, Golub TR, Orkin SH, Getz G, & Janeway KA (2014). Complementary genomic approaches highlight the PI3K/mTOR pathway as a common vulnerability in osteosarcoma. Proc Natl Acad Sci U S A, 111, E5564–5573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Philippe G, Huang YH, Cheneval O, Lawrence N, Zhang Z, Fairlie DP, Craik DJ, de Araujo AD, & Henriques ST (2016). Development of cell-penetrating peptide-based drug leads to inhibit MDMX:p53 and MDM2:p53 interactions. Biopolymers, 106, 853–863. [DOI] [PubMed] [Google Scholar]
- Pishas KI, Al-Ejeh F, Zinonos I, Kumar R, Evdokiou A, Brown MP, Callen DF, & Neilsen PM (2011). Nutlin-3a is a potential therapeutic for ewing sarcoma. Clin Cancer Res, 17, 494–504. [DOI] [PubMed] [Google Scholar]
- Polychronidou G, Karavasilis V, Pollack SM, Huang PH, Lee A, & Jones RL (2017). Novel therapeutic approaches in chondrosarcoma. Future Oncol, 13, 637–648. [DOI] [PubMed] [Google Scholar]
- Pourebrahim R, Zhang Y, Liu B, Gao R, Xiong S, Lin PP, McArthur MJ, Ostrowski MC, & Lozano G (2017). Integrative genome analysis of somatic p53 mutant osteosarcomas identifies Ets2-dependent regulation of small nucleolar RNAs by mutant p53 protein. Genes Dev, 31, 1847–1857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Radig K, Schneider-Stock R, Rose I, Mittler U, Oda Y, & Roessner A (1998). p53 and ras mutations in Ewing’s sarcoma. Pathol Res Pract, 194, 157–162. [DOI] [PubMed] [Google Scholar]
- Ragazzini P, Gamberi G, Pazzaglia L, Serra M, Magagnoli G, Ponticelli F, Ferrari C, Ghinelli C, Alberghini M, & Bertoni F (2004). Amplification of CDK4, MDM2, SAS and GLI genes in leiomyosarcoma, alveolar and embryonal rhabdomyosarcoma. Histology and histopathology, 19, 401–412. [DOI] [PubMed] [Google Scholar]
- Ranjan A, & Iwakuma T (2016). Non-Canonical Cell Death Induced by p53. Int J Mol Sci, 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ravegnini G, Mariño-Enriquez A, Slater J, Eilers G, Wang Y, Zhu M, Nucci MR, George S, Angelini S, Raut CP, & Fletcher JA (2012). MED12 mutations in leiomyosarcoma and extrauterine leiomyoma. Modern Pathology, 26, 743. [DOI] [PubMed] [Google Scholar]
- Ray-Coquard I, Blay J-Y, Italiano A, Le Cesne A, Penel N, Zhi J, Heil F, Rueger R, Graves B, Ding M, Geho D, Middleton SA, Vassilev LT, Nichols GL, & Bui BN (2012). Effect of the MDM2 antagonist RG7112 on the P53 pathway in patients with MDM2-amplified, well-differentiated or dedifferentiated liposarcoma: an exploratory proof-of-mechanism study. The Lancet Oncology, 13, 1133–1140. [DOI] [PubMed] [Google Scholar]
- Reddy SV (2004). Etiology of Paget’s disease and osteoclast abnormalities. J Cell Biochem, 93, 688–696. [DOI] [PubMed] [Google Scholar]
- Ren Y-X, Finckenstein FG, Abdueva DA, Shahbazian V, Chung B, Weinberg KI, Triche TJ, Shimada H, & Anderson MJ (2008). Mouse mesenchymal stem cells expressing PAX-FKHR form alveolar rhabdomyosarcomas by cooperating with secondary mutations. Cancer research, 68, 6587–6597. [DOI] [PubMed] [Google Scholar]
- Renzi S, Anderson ND, Light N, & Gupta A (2018). Ewing-like sarcoma: An emerging family of round cell sarcomas. J Cell Physiol. [DOI] [PubMed] [Google Scholar]
- Ribi S, Baumhoer D, Lee K, Edison, Teo AS, Madan B, Zhang K, Kohlmann WK, Yao F, Lee WH, Hoi Q, Cai S, Woo XY, Tan P, Jundt G, Smida J, Nathrath M, Sung WK, Schiffman JD, Virshup DM, & Hillmer AM (2015). TP53 intron 1 hotspot rearrangements are specific to sporadic osteosarcoma and can cause Li-Fraumeni syndrome. Oncotarget, 6, 7727–7740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Riggi N, Cironi L, Provero P, Suva ML, Stehle JC, Baumer K, Guillou L, & Stamenkovic I (2006). Expression of the FUS-CHOP fusion protein in primary mesenchymal progenitor cells gives rise to a model of myxoid liposarcoma. Cancer Res, 66, 7016–7023. [DOI] [PubMed] [Google Scholar]
- Rodriguez R, Rubio R, Gutierrez‐Aranda I, Melen GJ, Elosua C, García‐Castro J, & Menendez P (2011). FUS‐CHOP fusion protein expression coupled to p53 deficiency induces liposarcoma in mouse but not in human adipose‐derived mesenchymal stem/stromal cells. Stem Cells, 29, 179–192. [DOI] [PubMed] [Google Scholar]
- Rubin BP, Nishijo K, Chen HI, Yi X, Schuetze DP, Pal R, Prajapati SI, Abraham J, Arenkiel BR, Chen QR, Davis S, McCleish AT, Capecchi MR, Michalek JE, Zarzabal LA, Khan J, Yu Z, Parham DM, Barr FG, Meltzer PS, Chen Y, & Keller C (2011). Evidence for an unanticipated relationship between undifferentiated pleomorphic sarcoma and embryonal rhabdomyosarcoma. Cancer Cell, 19, 177–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rubio R, García-Castro J, Gutiérrez-Aranda I, Paramio J, Santos M, Catalina P, Leone PE, Menendez P, & Rodríguez R (2010). Deficiency in p53 but not retinoblastoma induces the transformation of mesenchymal stem cells in vitro and initiates leiomyosarcoma in vivo. Cancer research, 70, 4185–4194. [DOI] [PubMed] [Google Scholar]
- Ruiz-Mesa C, Goldberg JM, Coronado Munoz AJ, Dumont SN, & Trent JC (2015). Rhabdomyosarcoma in Adults: New Perspectives on Therapy. Current Treatment Options in Oncology, 16, 27. [DOI] [PubMed] [Google Scholar]
- Sakamoto A (2014). The molecular pathogenesis of dedifferentiated chondrosarcoma. Indian J Orthop, 48, 262–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samuel AM, Costa J, & Lindskog DM (2014). Genetic alterations in chondrosarcomas - keys to targeted therapies? Cell Oncol (Dordr), 37, 95–105. [DOI] [PubMed] [Google Scholar]
- Sand LG, Szuhai K, & Hogendoorn PC (2015). Sequencing Overview of Ewing Sarcoma: A Journey across Genomic, Epigenomic and Transcriptomic Landscapes. Int J Mol Sci, 16, 16176–16215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sandberg AA (2004). Genetics of chondrosarcoma and related tumors. Curr Opin Oncol, 16, 342–354. [DOI] [PubMed] [Google Scholar]
- Sayles LC, Breese MR, Koehne AL, Leung SG, Lee AG, Liu HY, Spillinger A, Shah AT, Tanasa B, Straessler K, Hazard FK, Spunt SL, Marina N, Kim GE, Cho SJ, Avedian RS, Mohler DG, Kim MO, DuBois SG, Hawkins DS, & Sweet-Cordero EA (2019). Genome-Informed Targeted Therapy for Osteosarcoma. Cancer Discov, 9, 46–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scheidt S, Jacobs C, Koob S, Welle K, Walter S, Jakob J, Kehrer M, & Schmolders J (2019). [Soft Tissue Sarcoma - a Current Review of the Diagnostic and Treatment Strategies]. Z Orthop Unfall. [DOI] [PubMed] [Google Scholar]
- Schhneider-stock R, Onnasch D, Haeckel C, Mellin W, S Franke D, & Roessner A (1999). Prognostic significance of p53 gene mutations and p53 protein expression in synovial sarcomas (Vol. 435). [DOI] [PubMed] [Google Scholar]
- Schneider-Stock R, Ziegeler A, Haeckel C, Franke D-S, Rys J, & Roessner A (1999). Prognostic relevance of p53 alterations and Mib-1 proliferation index in subgroups of primary liposarcomas. Clinical cancer research, 5, 2830–2835. [PubMed] [Google Scholar]
- Schrage YM, Lam S, Jochemsen AG, Cleton-Jansen AM, Taminiau AH, Hogendoorn PC, & Bovee JV (2009). Central chondrosarcoma progression is associated with pRb pathway alterations: CDK4 down-regulation and p16 overexpression inhibit cell growth in vitro. J Cell Mol Med, 13, 2843–2852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schuetze SM, Bolejack V, Choy E, Ganjoo KN, Staddon AP, Chow WA, Tawbi HA, Samuels BL, Patel SR, von Mehren M, D’Amato G, Leu KM, Loeb DM, Forscher CA, Milhem MM, Rushing DA, Lucas DR, Chugh R, Reinke DK, & Baker LH (2017). Phase 2 study of dasatinib in patients with alveolar soft part sarcoma, chondrosarcoma, chordoma, epithelioid sarcoma, or solitary fibrous tumor. Cancer, 123, 90–97. [DOI] [PubMed] [Google Scholar]
- Seddon B, Strauss SJ, Whelan J, Leahy M, Woll PJ, Cowie F, Rothermundt C, Wood Z, Benson C, Ali N, Marples M, Veal GJ, Jamieson D, Küver K, Tirabosco R, Forsyth S, Nash S, Dehbi H-M, & Beare S (2017). Gemcitabine and docetaxel versus doxorubicin as first-line treatment in previously untreated advanced unresectable or metastatic soft-tissue sarcomas (GeDDiS): a randomised controlled phase 3 trial. The Lancet Oncology, 18, 1397–1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seidinger AL, Mastellaro MJ, Paschoal Fortes F, Godoy Assumpcao J, Aparecida Cardinalli I, Aparecida Ganazza M, Correa Ribeiro R, Brandalise SR, Dos Santos Aguiar S, & Yunes JA (2011). Association of the highly prevalent TP53 R337H mutation with pediatric choroid plexus carcinoma and osteosarcoma in southeast Brazil. Cancer, 117, 2228–2235. [DOI] [PubMed] [Google Scholar]
- Seki M, Nishimura R, Yoshida K, Shimamura T, Shiraishi Y, Sato Y, Kato M, Chiba K, Tanaka H, Hoshino N, Nagae G, Shiozawa Y, Okuno Y, Hosoi H, Tanaka Y, Okita H, Miyachi M, Souzaki R, Taguchi T, Koh K, Hanada R, Kato K, Nomura Y, Akiyama M, Oka A, Igarashi T, Miyano S, Aburatani H, Hayashi Y, Ogawa S, & Takita J (2015). Integrated genetic and epigenetic analysis defines novel molecular subgroups in rhabdomyosarcoma. Nature Communications, 6, 7557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seligson ND, Kautto EA, Passen EN, Stets C, Toland AE, Millis SZ, Meyer CF, Hays JL, & Chen JL (2018). BRCA½ Functional Loss Defines a Targetable Subset in Leiomyosarcoma. Oncologist. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Serrano C, Romagosa C, Hernandez-Losa J, Simonetti S, Valverde C, Moline T, Somoza R, Perez M, Velez R, Verges R, Dominguez R, Carles J, & Ramon YCS (2016). RAS/MAPK pathway hyperactivation determines poor prognosis in undifferentiated pleomorphic sarcomas. Cancer, 122, 99–107. [DOI] [PubMed] [Google Scholar]
- Shern JF, Chen L, Chmielecki J, Wei JS, Patidar R, Rosenberg M, Ambrogio L, Auclair D, Wang J, Song YK, Tolman C, Hurd L, Liao H, Zhang S, Bogen D, Brohl AS, Sindiri S, Catchpoole D, Badgett T, Getz G, Mora J, Anderson JR, Skapek SX, Barr FG, Meyerson M, Hawkins DS, & Khan J (2014). Comprehensive Genomic Analysis of Rhabdomyosarcoma Reveals a Landscape of Alterations Affecting a Common Genetic Axis in Fusion-Positive and Fusion-Negative Tumors. Cancer Discovery, 4, 216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simms WW, Ordonez NG, Johnston D, Ayala AG, & Czerniak B (1995). p53 expression in dedifferentiated chondrosarcoma. Cancer, 76, 223–227. [DOI] [PubMed] [Google Scholar]
- Skytting B, Nilsson G, Brodin B, Xie Y, Lundeberg J, Uhlén M, & Larsson O (1999). A Novel Fusion Gene, SYT-SSX4, in Synovial Sarcoma. JNCI: Journal of the National Cancer Institute, 91, 974–975. [DOI] [PubMed] [Google Scholar]
- Soares J, Espadinha M, Raimundo L, Ramos H, Gomes AS, Gomes S, Loureiro JB, Inga A, Reis F, Gomes C, Santos MMM, & Saraiva L (2017). DIMP53–1: a novel small-molecule dual inhibitor of p53-MDM2/X interactions with multifunctional p53-dependent anticancer properties. Mol Oncol, 11, 612–627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sonnemann J, Palani CD, Wittig S, Becker S, Eichhorn F, Voigt A, & Beck JF (2011). Anticancer effects of the p53 activator nutlin-3 in Ewing’s sarcoma cells. Eur J Cancer, 47, 1432–1441. [DOI] [PubMed] [Google Scholar]
- Spiker AM, & Ramsey ML (2018). Cancer, Angiosarcoma. In StatPearls; Treasure Island (FL). [Google Scholar]
- Stacchiotti S, & Van Tine BA (2017). Synovial Sarcoma: Current Concepts and Future Perspectives. Journal of Clinical Oncology, 36, 180–187. [DOI] [PubMed] [Google Scholar]
- Stegmaier S, Poremba C, Schaefer K-L, Leuschner I, Kazanowska B, Békássy AN, Bielack SS, Klingebiel T, & Koscielniak E (2011). Prognostic value of PAX–FKHR fusion status in alveolar rhabdomyosarcoma: A report from the cooperative soft tissue sarcoma study group (CWS). Pediatric Blood & Cancer, 57, 406–414. [DOI] [PubMed] [Google Scholar]
- Stenman G, Andersson H, Mandahl N, Meis-Kindblom JM, & Kindblom LG (1995). Translocation t(9;22)(q22;q12) is a primary cytogenetic abnormality in extraskeletal myxoid chondrosarcoma. Int J Cancer, 62, 398–402. [DOI] [PubMed] [Google Scholar]
- Stewart E, McEvoy J, Wang H, Chen X, Honnell V, Ocarz M, Gordon B, Dapper J, Blankenship K, & Yang Y (2018). Identification of therapeutic targets in rhabdomyosarcoma through integrated genomic, epigenomic, and proteomic analyses. Cancer cell, 34, 411–426. e419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stolte B, Iniguez AB, Dharia NV, Robichaud AL, Conway AS, Morgan AM, Alexe G, Schauer NJ, Liu X, Bird GH, Tsherniak A, Vazquez F, Buhrlage SJ, Walensky LD, & Stegmaier K (2018). Genome-scale CRISPR-Cas9 screen identifies druggable dependencies in TP53 wild-type Ewing sarcoma. J Exp Med, 215, 2137–2155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stuart ET, Haffner R, Oren M, & Gruss P (1995). Loss of p53 function through PAX‐mediated transcriptional repression. The EMBO journal, 14, 5638–5645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun D, Li Z, Rew Y, Gribble M, Bartberger MD, Beck HP, Canon J, Chen A, Chen X, Chow D, Deignan J, Duquette J, Eksterowicz J, Fisher B, Fox BM, Fu J, Gonzalez AZ, Gonzalez-Lopez De Turiso F, Houze JB, Huang X, Jiang M, Jin L, Kayser F, Liu JJ, Lo MC, Long AM, Lucas B, McGee LR, McIntosh J, Mihalic J, Oliner JD, Osgood T, Peterson ML, Roveto P, Saiki AY, Shaffer P, Toteva M, Wang Y, Wang YC, Wortman S, Yakowec P, Yan X, Ye Q, Yu D, Yu M, Zhao X, Zhou J, Zhu J, Olson SH, & Medina JC (2014). Discovery of AMG 232, a potent, selective, and orally bioavailable MDM2-p53 inhibitor in clinical development. J Med Chem, 57, 1454–1472. [DOI] [PubMed] [Google Scholar]
- Sun X, Guo W, Shen JK, Mankin HJ, Hornicek FJ, & Duan Z (2015). Rhabdomyosarcoma: Advances in Molecular and Cellular Biology. Sarcoma, 2015, 232010–232010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szymanska J, Tarkkanen M, Wiklund T, Virolainen M, Blomqvist C, Asko-Seljavaara S, Tukiainen E, Elomaa I, & Knuutila S (1996). Gains and losses of DNA sequences in liposarcomas evaluated by comparative genomic hybridization. Genes, Chromosomes and Cancer, 15, 89–94. [DOI] [PubMed] [Google Scholar]
- Tang N, Song WX, Luo J, Haydon RC, & He TC (2008). Osteosarcoma development and stem cell differentiation. Clin Orthop Relat Res, 466, 2114–2130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tarpey PS, Behjati S, Cooke SL, Van Loo P, Wedge DC, Pillay N, Marshall J, O’Meara S, Davies H, Nik-Zainal S, Beare D, Butler A, Gamble J, Hardy C, Hinton J, Jia MM, Jayakumar A, Jones D, Latimer C, Maddison M, Martin S, McLaren S, Menzies A, Mudie L, Raine K, Teague JW, Tubio JM, Halai D, Tirabosco R, Amary F, Campbell PJ, Stratton MR, Flanagan AM, & Futreal PA (2013). Frequent mutation of the major cartilage collagen gene COL2A1 in chondrosarcoma. Nat Genet, 45, 923–926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taubert H, Meye A, & Wurl P (1998). Soft tissue sarcomas and p53 mutations. Mol Med, 4, 365–372. [PMC free article] [PubMed] [Google Scholar]
- Taylor AC, Shu L, Danks MK, Poquette CA, Shetty S, Thayer MJ, Houghton PJ, & Harris LC (2000). P53 mutation and MDM2 amplification frequency in pediatric rhabdomyosarcoma tumors and cell lines. Medical and Pediatric Oncology: The Official Journal of SIOP—International Society of Pediatric Oncology (Societé Internationale d’Oncologie Pédiatrique, 35, 96–103. [DOI] [PubMed] [Google Scholar]
- Todorova R (2014). Ewing’s sarcoma cancer stem cell targeted therapy. Curr Stem Cell Res Ther, 9, 46–62. [DOI] [PubMed] [Google Scholar]
- Toffoli G, Biason P, Russo A, De Mattia E, Cecchin E, Hattinger CM, Pasello M, Alberghini M, Ferrari C, Scotlandi K, Picci P, & Serra M (2009). Effect of TP53 Arg72Pro and MDM2 SNP309 polymorphisms on the risk of high-grade osteosarcoma development and survival. Clin Cancer Res, 15, 3550–3556. [DOI] [PubMed] [Google Scholar]
- Toguchida J, Yamaguchi T, Ritchie B, Beauchamp RL, Dayton SH, Herrera GE, Yamamuro T, Kotoura Y, Sasaki MS, Little JB, & et al. (1992). Mutation spectrum of the p53 gene in bone and soft tissue sarcomas. Cancer Res, 52, 6194–6199. [PubMed] [Google Scholar]
- Tongyang L, Haiqiang G, Meiyan Z, Yingze H, Shuting J, Ying L, & Jihong Z (2015). [Synthetic lethal genes to mutant p53]. Yi Chuan, 37, 321–326. [DOI] [PubMed] [Google Scholar]
- Tornin J, Hermida-Prado F, Padda RS, Gonzalez MV, Alvarez-Fernandez C, Rey V, Martinez-Cruzado L, Estupiñan O, Menendez ST, & Fernandez-Nevado L (2018). FUS-CHOP promotes invasion in myxoid liposarcoma through a SRC/FAK/RHO/ROCK-dependent pathway. Neoplasia, 20, 44–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Totoki Y, Yoshida A, Hosoda F, Nakamura H, Hama N, Ogura K, Yoshida A, Fujiwara T, Arai Y, Toguchida J, Tsuda H, Miyano S, Kawai A, & Shibata T (2014). Unique mutation portraits and frequent COL2A1 gene alteration in chondrosarcoma. Genome Res, 24, 1411–1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tovar C, Graves B, Packman K, Filipovic Z, Higgins B, Xia M, Tardell C, Garrido R, Lee E, Kolinsky K, To KH, Linn M, Podlaski F, Wovkulich P, Vu B, & Vassilev LT (2013). MDM2 small-molecule antagonist RG7112 activates p53 signaling and regresses human tumors in preclinical cancer models. Cancer Res. [DOI] [PubMed] [Google Scholar]
- Tovar C, Rosinski J, Filipovic Z, Higgins B, Kolinsky K, Hilton H, Zhao X, Vu BT, Qing W, Packman K, Myklebost O, Heimbrook DC, & Vassilev LT (2006). Small-molecule MDM2 antagonists reveal aberrant p53 signaling in cancer: implications for therapy. Proc Natl Acad Sci U S A, 103, 1888–1893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsuchiya T, Sekine K, Hinohara S, Namiki T, Nobori T, & Kaneko Y (2000). Analysis of the p16INK4, p14ARF, p15, TP53, and MDM2 genes and their prognostic implications in osteosarcoma and Ewing sarcoma. Cancer Genet Cytogenet, 120, 91–98. [DOI] [PubMed] [Google Scholar]
- Ueda T, Healey JH, Huvos AG, & Ladanyi M (1997). Amplification of the MYC Gene in Osteosarcoma Secondary to Paget’s Disease of Bone. Sarcoma, 1, 131–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uesato S, Matsuura Y, Matsue S, Sumiyoshi T, Hirata Y, Takemoto S, Kawaratani Y, Yamai Y, Ishida K, Sasaki T, & Enari M (2016). Discovery of new low-molecular-weight p53-Mdmx disruptors and their anti-cancer activities. Bioorg Med Chem, 24, 1919–1926. [DOI] [PubMed] [Google Scholar]
- van der Ent W, Jochemsen AG, Teunisse AF, Krens SF, Szuhai K, Spaink HP, Hogendoorn PC, & Snaar-Jagalska BE (2014). Ewing sarcoma inhibition by disruption of EWSR1-FLI1 transcriptional activity and reactivation of p53. J Pathol, 233, 415–424. [DOI] [PubMed] [Google Scholar]
- van Oosterwijk JG, van Ruler MA, Briaire-de Bruijn IH, Herpers B, Gelderblom H, van de Water B, & Bovee JV (2013). Src kinases in chondrosarcoma chemoresistance and migration: dasatinib sensitises to doxorubicin in TP53 mutant cells. Br J Cancer, 109, 1214–1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z, Kong N, Kammlott U, Lukacs C, Klein C, Fotouhi N, & Liu EA (2004). In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science, 303, 844–848. [DOI] [PubMed] [Google Scholar]
- Veeraraghavan J, Natarajan M, Herman TS, & Aravindan N (2010). Curcumin-altered p53-response genes regulate radiosensitivity in p53-mutant Ewing’s sarcoma cells. Anticancer Res, 30, 4007–4015. [PubMed] [Google Scholar]
- Velletri T, Xie N, Wang Y, Huang Y, Yang Q, Chen X, Chen Q, Shou P, Gan Y, Cao G, Melino G, & Shi Y (2016). P53 functional abnormality in mesenchymal stem cells promotes osteosarcoma development. Cell Death Dis, 7, e2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ventura S, Aryee DN, Felicetti F, De Feo A, Mancarella C, Manara MC, Picci P, Colombo MP, Kovar H, Care A, & Scotlandi K (2016). CD99 regulates neural differentiation of Ewing sarcoma cells through miR-34a-Notch-mediated control of NF-kappaB signaling. Oncogene, 35, 3944–3954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vlenterie M, Hillebrandt-Roeffen MHS, Flucke UE, Groenen PJTA, Tops BBJ, Kamping EJ, Pfundt R, de Bruijn DRH, Geurts van Kessel AHM, van Krieken HJHJM, van der Graaf WTA, & Versleijen-Jonkers YMH (2015). Next generation sequencing in synovial sarcoma reveals novel gene mutations. Oncotarget, 6, 34680–34690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vos M, Koseła-Paterczyk H, Rutkowski P, van Leenders GJLH, Normantowicz M, Lecyk A, Sleijfer S, Verhoef C, & Grünhagen DJ (2018). Differences in recurrence and survival of extremity liposarcoma subtypes. European Journal of Surgical Oncology, 44, 1391–1397. [DOI] [PubMed] [Google Scholar]
- Wadayama B, Toguchida J, Yamaguchi T, Sasaki MS, & Yamamuro T (1993). p53 expression and its relationship to DNA alterations in bone and soft tissue sarcomas. Br J Cancer, 68, 1134–1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wade M, Li Y-C, & Wahl GM (2013). MDM2, MDMX and p53 in oncogenesis and cancer therapy. Nature Reviews Cancer, 13, 83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wagner AJ, Banerji U, Mahipal A, Somaiah N, Hirsch H, Fancourt C, Johnson-Levonas AO, Lam R, Meister AK, Russo G, Knox CD, Rose S, & Hong DS (2017). Phase I Trial of the Human Double Minute 2 Inhibitor MK-8242 in Patients With Advanced Solid Tumors. J Clin Oncol, 35, 1304–1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walkley CR, Qudsi R, Sankaran VG, Perry JA, Gostissa M, Roth SI, Rodda SJ, Snay E, Dunning P, Fahey FH, Alt FW, McMahon AP, & Orkin SH (2008). Conditional mouse osteosarcoma, dependent on p53 loss and potentiated by loss of Rb, mimics the human disease. Genes Dev, 22, 1662–1676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang B, Fang L, Zhao H, Xiang T, & Wang D (2012). MDM2 inhibitor Nutlin-3a suppresses proliferation and promotes apoptosis in osteosarcoma cells. Acta Biochim Biophys Sin (Shanghai), 44, 685–691. [DOI] [PubMed] [Google Scholar]
- Wang L, Motoi T, Khanin R, Olshen A, Mertens F, Bridge J, Dal Cin P, Antonescu CR, Singer S, Hameed M, Bovee JV, Hogendoorn PC, Socci N, & Ladanyi M (2012). Identification of a novel, recurrent HEY1-NCOA2 fusion in mesenchymal chondrosarcoma based on a genome-wide screen of exon-level expression data. Genes Chromosomes Cancer, 51, 127–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang S, Sun W, Zhao Y, McEachern D, Meaux I, Barriere C, Stuckey JA, Meagher JL, Bai L, Liu L, Hoffman-Luca CG, Lu J, Shangary S, Yu S, Bernard D, Aguilar A, Dos-Santos O, Besret L, Guerif S, Pannier P, Gorge-Bernat D, & Debussche L (2014). SAR405838: an optimized inhibitor of MDM2-p53 interaction that induces complete and durable tumor regression. Cancer Res, 74, 5855–5865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang X, & Simon R (2013). Identification of potential synthetic lethal genes to p53 using a computational biology approach. BMC Med Genomics, 6, 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang XH, Zhang SF, Bao JT, & Liu FY (2017). Oridonin synergizes with Nutlin-3 in osteosarcoma cells by modulating the levels of multiple Bcl-2 family proteins. Tumour Biol, 39, 1010428317701638. [DOI] [PubMed] [Google Scholar]
- Wang Z, & Sun Y (2010). Targeting p53 for Novel Anticancer Therapy. Transl Oncol, 3, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wassman CD, Baronio R, Demir O, Wallentine BD, Chen CK, Hall LV, Salehi F, Lin DW, Chung BP, Hatfield GW, Richard Chamberlin A, Luecke H, Lathrop RH, Kaiser P, & Amaro RE (2013). Computational identification of a transiently open L1/S3 pocket for reactivation of mutant p53. Nat Commun, 4, 1407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weidle UH, Maisel D, & Eick D (2011). Synthetic lethality-based targets for discovery of new cancer therapeutics. Cancer Genomics Proteomics, 8, 159–171. [PubMed] [Google Scholar]
- Wischhusen J, Naumann U, Ohgaki H, Rastinejad F, & Weller M (2003). CP-31398, a novel p53-stabilizing agent, induces p53-dependent and p53-independent glioma cell death. Oncogene, 22, 8233–8245. [DOI] [PubMed] [Google Scholar]
- Wunder JS, Gokgoz N, Parkes R, Bull SB, Eskandarian S, Davis AM, Beauchamp CP, Conrad EU, Grimer RJ, Healey JH, Malkin D, Mangham DC, Rock MJ, Bell RS, & Andrulis IL (2005). TP53 mutations and outcome in osteosarcoma: a prospective, multicenter study. J Clin Oncol, 23, 1483–1490. [DOI] [PubMed] [Google Scholar]
- Xia SJ, Pressey JG, & Barr FG (2002). Molecular pathogenesis of rhabdomyosarcoma. Cancer biology & therapy, 1, 97–104. [DOI] [PubMed] [Google Scholar]
- Xia Y, Du Z, Wang X, & Li X (2018). Treatment of uterine sarcoma with rAd-p53 (gendicine) followed by chemotherapy: clinical study of TP53 gene therapy. Human gene therapy, 29, 242–250. [DOI] [PubMed] [Google Scholar]
- Xie Z, Babiceanu M, Kumar S, Jia Y, Qin F, Barr FG, & Li H (2016). Fusion transcriptome profiling provides insights into alveolar rhabdomyosarcoma. Proceedings of the National Academy of Sciences of the United States of America, 113, 13126–13131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiong S, Tu H, Kollareddy M, Pant V, Li Q, Zhang Y, Jackson JG, Suh YA, Elizondo-Fraire AC, Yang P, Chau G, Tashakori M, Wasylishen AR, Ju Z, Solomon H, Rotter V, Liu B, El-Naggar AK, Donehower LA, Martinez LA, & Lozano G (2014). Pla2g16 phospholipase mediates gain-of-function activities of mutant p53. Proc Natl Acad Sci U S A, 111, 11145–11150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu J, Timares L, Heilpern C, Weng Z, Li C, Xu H, Pressey JG, Elmets CA, Kopelovich L, & Athar M (2010). Targeting wild-type and mutant p53 with small molecule CP-31398 blocks the growth of rhabdomyosarcoma by inducing reactive oxygen species-dependent apoptosis. Cancer Res, 70, 6566–6576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang J, Du X, Chen K, Ylipää A, Lazar AJF, Trent J, Lev D, Pollock R, Hao X, Hunt K, & Zhang W (2009). Genetic aberrations in soft tissue leiomyosarcoma. Cancer letters, 275, 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao D, Cai GH, Chen J, Ling R, Wu SX, & Li YP (2014). Prognostic value of p53 alterations in human osteosarcoma: a meta analysis. Int J Clin Exp Pathol, 7, 6725–6733. [PMC free article] [PubMed] [Google Scholar]
- Yi H, Yan X, Luo Q, Yuan L, Li B, Pan W, Zhang L, Chen H, Wang J, Zhang Y, Zhai Y, Qiu MZ, & Yang DJ (2018). A novel small molecule inhibitor of MDM2-p53 (APG-115) enhances radiosensitivity of gastric adenocarcinoma. J Exp Clin Cancer Res, 37, 97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yochum TR (1984). Paget’s sarcoma of bone. Radiologe, 24, 428–433. [PubMed] [Google Scholar]
- Young RJ, Brown NJ, Reed MW, Hughes D, & Woll PJ (2010). Angiosarcoma. Lancet Oncol, 11, 983–991. [DOI] [PubMed] [Google Scholar]
- Yu D, Kahen E, Cubitt CL, McGuire J, Kreahling J, Lee J, Altiok S, Lynch CC, Sullivan DM, & Reed DR (2015). Identification of Synergistic, Clinically Achievable, Combination Therapies for Osteosarcoma. Sci Rep, 5, 16991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zache N, Lambert JM, Rokaeus N, Shen J, Hainaut P, Bergman J, Wiman KG, & Bykov VJ (2008). Mutant p53 targeting by the low molecular weight compound STIMA-1. Mol Oncol, 2, 70–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang N, Liu H, Yue G, Zhang Y, You J, & Wang H (2016). Molecular Heterogeneity of Ewing Sarcoma as Detected by Ion Torrent Sequencing. PLoS One, 11, e0153546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang W-W, Li L, Li D, Liu J, Li X, Li W, Xu X, Zhang MJ, Chandler LA, & Lin H (2018). The first approved gene therapy product for cancer Ad-p53 (Gendicine): 12 years in the clinic. Human gene therapy, 29, 160–179. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Hu Q, Li G, Li L, Liang S, Zhang Y, Liu J, Fan Z, Li L, Zhou B, Ruan Y, Yang X, Chen S, Mu T, Wang G, & Xiong S (2018). ONZIN Upregulation by Mutant p53 Contributes to Osteosarcoma Metastasis Through the CXCL5-MAPK Signaling Pathway. Cell Physiol Biochem, 48, 1099–1111. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Yang J, Zhao N, Wang C, Kamar S, Zhou Y, He Z, Yang J, Sun B, Shi X, Han L, & Yang Z (2018). Progress in the chemotherapeutic treatment of osteosarcoma. Oncol Lett, 16, 6228–6237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou X, Hao Q, & Lu H (2018). Mutant p53 in Cancer Therapy - the Barrier or the Path. J Mol Cell Biol. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zietz C, Rossle M, Haas C, Sendelhofert A, Hirschmann A, Sturzl M, & Lohrs U (1998). MDM-2 oncoprotein overexpression, p53 gene mutation, and VEGF up-regulation in angiosarcomas. Am J Pathol, 153, 1425–1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zuffa E, Mancini M, Brusa G, Pagnotta E, Hattinger CM, Serra M, Remondini D, Castellani G, Corrado P, Barbieri E, & Santucci MA (2008). P53 oncosuppressor influences selection of genomic imbalances in response to ionizing radiations in human osteosarcoma cell line SAOS-2. Int J Radiat Biol, 84, 591–601. [DOI] [PubMed] [Google Scholar]