Next Article in Journal
Peritoneal Cancer Index Dominates Prognosis After CRS–HIPEC for Colorectal Peritoneal Metastases: A Consecutive Single-Centre Cohort with 3-Year Follow-Up
Previous Article in Journal
Delay in Diagnosis and Treatment of Breast Cancer: Regional Disparities
Previous Article in Special Issue
Integration of an OS-Based Machine Learning Score (AS Score) and Immunoscore as Ancillary Tools for Predicting Immunotherapy Response in Sarcomas
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic Heterogeneity of Undifferentiated Pleomorphic Sarcoma: Is There Potential for Targeted Therapy?

by
Ekaterina A. Lesovaya
1,2,3,
Timur I. Fetisov
1,
Beniamin Yu. Bokhyan
1,
Maria A. Senchenko
1,
Dmitry V. Rogozhin
1,
Varvara P. Maksimova
1,
Anna N. Demko
2,
Gennady A. Belitsky
1,
Marianna G. Yakubovskaya
1,3,*,† and
Kirill I. Kirsanov
1,3,†
1
Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow 115478, Russia
2
Oncology Department, Ryazan State Medical University Named After Academician I.P. Pavlov, Ministry of Health of Russia, Ryazan 390026, Russia
3
Institute of Medicine, RUDN University, Moscow 117198, Russia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2025, 17(22), 3613; https://doi.org/10.3390/cancers17223613
Submission received: 9 September 2025 / Revised: 27 October 2025 / Accepted: 6 November 2025 / Published: 10 November 2025
(This article belongs to the Special Issue Genomics and Transcriptomics in Sarcoma)

Simple Summary

Despite advances in multitargeted anticancer drugs and personalized treatment approaches, the management of soft tissue sarcoma remains challenging. Among the most difficult subtypes to diagnose and treat with targeted therapies is UPS, owing to its high degree of genetic heterogeneity. This review summarizes recent findings on the genetics and epigenetics of this sarcoma subtype and discusses the potential application of various targeted therapies.

Abstract

Undifferentiated pleomorphic sarcoma (UPS) is the most morphologically and genetically heterogeneous form of soft tissue sarcoma. UPS tumors can exhibit a wide range of genetic abnormalities, including activating and inactivating mutations, gene amplifications, chromosomal translocations, and copy number variations. Owing to this extensive genetic heterogeneity, no UPS-specific therapeutic targets have yet been validated, complicating diagnosis, prognosis, and the selection of targeted treatment strategies. Currently, immune checkpoint inhibitors (targeting PD-1, PD-L1, and CTLA-4) are the only validated targeted therapy for UPS, reflecting the frequent mutational events that activate immune response pathways. Because molecular genetic profiling alone provides limited prognostic value for chemoresistance in UPS, the development of experimental ex vivo and in vitro testing approaches may help to identify and exclude potentially ineffective targeted therapies.

1. Introduction

Undifferentiated pleomorphic sarcoma (UPS), formerly known as malignant fibrous histiocytoma, is a heterogeneous group of pleomorphic sarcomas that lack a definable line of differentiation and specific molecular features and thus remain a diagnosis of exclusion [1]. This subtype of soft tissue sarcoma (STS) is an aggressive malignancy characterized by a high metastatic potential and limited responsiveness to current therapeutic modalities. Therefore, the identification of specific diagnostic and prognostic markers, along with the development of novel targeted therapeutic strategies, is urgently needed.
UPS typically occurs in late adulthood, most commonly between 50 and 70 years of age, with a higher prevalence among white males [1]. The most frequent sites of UPS development are the lower extremities—particularly the thighs—followed by the upper extremities and the retroperitoneum [2,3,4,5]. Clinically, UPS usually presents as a painless, slowly enlarging mass. Microscopically, it exhibits considerable morphological variability and is generally composed of highly pleomorphic cells. Spindle cell, round cell, and epithelioid variants have been described, and multiple histological components may coexist in the same tumor. The neoplastic cells often form fascicles or storiform structures; however, in some cases, no distinct growth pattern can be recognized. Additional histopathological features include high mitotic activity, areas of necrosis, and variable degrees of chronic inflammation [6].
Immunohistochemical (IHC) findings in UPS are generally limited and non-specific. The primary role of immunohistochemistry in UPS diagnosis is to exclude other, more specific sarcoma subtypes. Typically, UPS lacks expression of distinct lineage-specific markers but shows diffuse positivity for vimentin and variable or focal expression of CD68, smooth muscle actin (SMA), and CD34, all of which are considered low-specificity markers [7]. Over the past decades, numerous studies have identified various genetic and epigenetic changes associated with UPS; however, further comprehensive analyses are required to validate these molecular abnormalities as potential diagnostic markers and to refine criteria guiding therapeutic decision-making. In particular, several transcriptomic analyses of large STS cohorts have failed to clearly distinguish UPS from other STS subtypes, such as leiomyosarcoma (LMS). Therefore, a comprehensive analysis of recent molecular genetic studies, case reports, and findings from emerging optimized therapeutic approaches is essential for identifying molecular and genetic characteristics of UPS that may hold prognostic and therapeutic significance.

2. Molecular Genetic Heterogeneity of UPS

Owing to the absence of distinctive morphological or molecular features, UPS remains a diagnosis of exclusion in clinical practice. Specifically, myxofibrosarcoma (MFS) has been reclassified as a separate entity from UPS based on its fibroblastic differentiation and characteristic myxoid stroma; however, from a genetic standpoint, UPS and MFS are still considered part of the same sarcoma group. According to the current WHO Classification of Soft Tissue and Bone Tumors, UPS lacks specific gene transcripts and is characterized by a highly unstable genome [1,8].
Genetic aberrations in UPS—including chromosomal translocations, mutations, and deletions—are numerous but non-specific and may vary considerably among individual cases. Therefore, the significance of data derived from single case reports should not be overlooked. The karyotype of UPS cells is typically complex, with multiple chromosomes affected simultaneously. Reported chromosomal translocations include t(1;2), t(1;3), t(1;7), t(1;10), t(1;17), t(2;3), t(5;10), t(5;11), t(5;17), t(6;8), t(6;10), t(7;10), t(9;10), t(10;11), t(10;12), t(11;17), and t(15;21) [8,9,10]. Recently, isolated cases of UPS harboring NTRK rearrangements—including TMTC–NTRK3, DCTN1–NTRK1, and SARM1–NTRK1—have been reported in both adult and pediatric patients [11,12,13,14,15]. In a retrospective study, the TRIO::TERT fusion gene was identified in patients with UPS [16]. This finding was further supported by evidence from additional studies involving several non-translocation-related STS, such as MFS, pleomorphic rhabdomyosarcoma, and dedifferentiated liposarcoma, including UPS. Multiple TRIO fusions involving different partners and TERT exons were described in these studies, including TRIO(ex33)–TERT(ex2), TRIO(ex33)–TERT(ex3), TRIO(ex34)–CDH18(ex2)–TERT(ex2), TRIO(ex34)–CDH18(ex2)–TERT(ex3), TRIO(ex33)LINC01504(intron2), TRIO(ex33)LINC01504(intron3), and TRIO(ex33)LINC01504(exon4) [17]. In another study, a similar fusion—lacking MDM2 amplification—was reported in a case of spindle cell liposarcoma [18]. Because TRIO fusions have been detected in tumors of various histological types, it has been proposed that these changes do not represent primary oncogenic drivers but instead contribute to tumor progression as secondary oncogenic events [17].
An EML4ALK gene rearrangement associated with brain metastasis was reported in a case of primary malignant fibrous histiocytoma of the lung in a 59-year-old male patient [19]. ALK rearrangements, particularly the EML4ALK fusion, are well documented in non-small cell lung cancer [20,21]. Therefore, this tumor may be related to a sarcomatoid carcinoma of the lung.
Integration of multiple omics datasets has advanced the genetic profiling of UPS. In a study analyzing 19 UPS tumors, including 2 paired recurrent and re-recurrent samples, 66 fusion genes were identified, of which 10 were novel. Specifically, retinoblastoma (RB1) fusions—such as RB1-RNASEH2B, RB1-FGF14-AS1, and E2F6-FKBP4—were observed in two tumor samples and correlated with increased Rb/E2F signaling activation [22]. Additional targeted fusions included pseudogene-related fusions (CIC-DUX4L8 and EIF2AK4-ANXA2P2), as well as PDGFRA-MACROD2 and NCOR1-MAP2K1. Other rare fusion genes reported in primary UPS included CLTC-VMP1, FARP1-STK24, and PRDM10 fusions with MED12 and CITED2 partners [14,21,22]. Since the release of the 2020 WHO Classification, tumors harboring CIC rearrangements are recognized as a distinct entity—CIC-rearranged sarcoma [1]. However, the biological significance of most other identified fusion transcripts remains largely unknown.
Importantly, positive regulatory domain (PRDM) proteins play critical roles in cell proliferation, differentiation, and malignant transformation. PRDM10-containing fusion genes have been identified in low-grade UPS and are present in approximately 5% of cases, representing a clinically important UPS subset [23,24,25]. Additionally, in secondary radiation-induced UPS following primary breast cancer, a novel COL3A1-GULP1 fusion (COL3A1: exon23–GULP1: exon5) was detected [26].
Both loss-of-function and gain-of-function mutations, as well as gene amplifications, are common genetic abnormalities in UPS; however, their clinical correlations remain under investigation. Several growth factors have been shown to be overexpressed in UPS and may serve as potential therapeutic targets. For example, a tumor subgroup exhibiting increased IGF2 and FGFR3 expression could warrant more aggressive treatment strategies. Despite these findings, no validated prognostic markers currently exist for UPS [27]. Amplifications and activating mutations of PDGFRA, PDGFRB, and EGFR have been reported in cardiac and rare intracranial UPS, providing a rationale for exploring therapies targeting PDGF receptors and EGFR [28,29]. Additionally, leucine-rich repeat-containing protein 15 (LRRC15), a target of TGF-β, is frequently overexpressed in UPS cells. LRRC15 mediates cell–cell and cell–matrix interactions and has emerged as a promising anticancer target owing to its high expression in mesenchymal-derived tumors, including UPS [30,31]. Increased expression of WWTR1 and YAP1 has also been observed in 50–60% of sarcomas, predominantly in UPS and dedifferentiated liposarcoma [32].
Several amplifications and activating mutations in genes commonly associated with other STS subtypes have been identified in UPS. Specifically, mutations and amplifications in KIT, KRAS, PDGFRA, PDGFRB, PIK3CA, AKT, AXL, MMP13, and WNT7B have been reported [10,29,33,34,35,36,37,38,39]. Amplification of VGLL3, which encodes a mechanosensitive transcriptional regulator, has also been observed in UPS [40]. Comprehensive genomic profiling has revealed six recurrent genomic alterations across UPS tumors of different anatomical sites: TP53 R248W, ATR I2435V, GNAS P423H, MKI67 A1493T, PDCD11 Q838H, and SF3B1 A263V. In one case of orbital UPS, additional alterations in NOTCH1, PCLO, MYST1, and NPM1 were detected [41].
Bioinformatic analysis of TCGA data and the gene expression profile GSE21050, combined with immunohistochemical evaluation of a tissue microarray (TMA) and in vitro validation, revealed that high expression of adenosine monophosphate deaminase 2 (AMPD2)—a key enzyme in purine metabolism—is associated with poorer patient outcomes across independent cohorts, potentially by promoting UPS cell proliferation [42]. In another study, overexpression of serine/threonine-protein kinase 13 (Plk1) and the DNA replication inhibitor geminin was identified as biomarkers of poor prognosis and provided novel insights into UPS biology [43]. Additionally, in 24 UPS samples, strong negative correlations were observed between the multidrug resistance genes ABCB1 and ABCG2, while a positive correlation between MVP expression and favorable response to doxorubicin/gemcitabine therapy was reported [44].
In addition, UPS frequently harbors inactivating mutations and deletions in tumor suppressor genes, including CDKN2A (mutations and deletions), TP53, CSF2RB, RB1, PTEN, ATM, and ATRX, but lacks a single defining driver mutation [10,39,45,46,47,48]. Notably, loss of function in RB1 and TP53 leads to activation of SKP2 and increased cell proliferation, suggesting that SKP2 inhibition may represent a potential therapeutic strategy [49].
In a study of 60 UPS cases using cDNA microarray analysis, a 300-gene signature (11% FDR) was identified. The most upregulated gene clusters comprised cathepsins and regulators of protein degradation, inflammation, cell motility, and proliferation [50]. Interestingly, increased expression of the cytoskeletal component COL6A3 and biglycan (BGN) was associated with a favorable prognosis in studies of 46 and 38 UPS patients, respectively, despite the potential oncogenic role of the COL6A3-GULP1 fusion [26,51]. Furthermore, dysregulation of alternative splicing events has been implicated in UPS pathogenesis and progression. Specifically, exon skipping in the EWSR1 gene correlates with poor prognosis, although this finding requires further validation [52].
Copy number variations (CNVs) in UPS have been reported as follows: gains at 1p36.33–p31.3, 1q21.2–q24.3, 4p16.3, 5p15.33–p13.1, 7p22.3, 7p15.2–7p11.2, 7q32.1–q32.2, 9q34.3, 14q11.2, 14q32.33, 16p13.3, 17q12, 17q21.33, 17q23.3, 19p13.3, 19q13.11–q13.2, 19q13.42, 20q11.21–q13.33, 21q22.3, and X; losses at 1q32.1, 2p25.3, 2q36.1–q37.3, 8p23.3, 9p24.2–9p22.3, 9p21.3–p21.1, 10q21.1–q23.2, 11q22.3, 13q12.11–q31.1, 13q33.3, 16q11.2, and 16q23.1; and amplifications at 1p36, 1p32, 1q21–q23, 1q32, 3q26, 4q, 5p, 6q23, 7q, 8p23.1, 8q21.2–q22, 8q24, 9q31–q34, 10q26, 11q, 12p, 12q13–q15, 17q12, and 20q. Among these, deletion of 13q, particularly 13q14–21, was the most common and has recently been identified as the most frequent copy-number alteration in UPS [8,53].
Near-haploidization, characterized by the loss of one copy of chromosomes, is relatively rare in most tumors but may represent a specific feature of UPS. However, clinical validation of this phenomenon remains limited because only a single study has been reported in the literature. Whole-genome and transcriptome sequencing of two UPS samples revealed chromosomal rearrangements in the form of copy number variants, specifically affecting SMC1A, a gene encoding a component of the cohesin complex and a key regulator of the S-phase [54].

3. Epigenetic Alterations in UPS

UPS is not defined by specific epigenetic changes; however, subsets of epigenetic regulators and microRNAs (miRNAs) have been reported. Genome-wide studies indicate that UPS exhibits aberrant DNA methylation patterns. In particular, DNA methyltransferase DNMT3B is overexpressed in UPS and is associated with poor prognosis. Patient samples also show substantially elevated methylation of histones H3K4me3 and H3K9me3 compared with normal muscle tissue. However, current DNMT inhibitors, including 5-aza-2ʹ-deoxycytidine and nanaomycin A, are ineffective in UPS owing to unfavorable safety profiles [55,56].
MiRNA profiling has been investigated in several UPS studies. In one study of 10 high-grade UPS samples, miRNA microarray analysis identified differentially expressed miRNAs, along with their target genes, including miR-199b-5p, miR-320a, miR-199a-3p, miR-126, and miR-22, targeting IMP3, ROR2, MDM2, CDK4, and UPA. In a subsequent series of 27 UPS samples, these findings were validated using quantitative polymerase chain reaction (PCR) [57]. Another study identified a distinct subset of differentially expressed miRNAs, predominantly with tumor suppressor functions, including miR-451, miR-1260, miR-1274a, miR-34a, miR-152, miR-199b-5p, and miR-320a [58]. Interestingly, in UPS samples with mutated BRAF V600E and KRAS G12D and increased MAPK signaling, higher levels of mature miRNAs were detected. In vitro and in vivo experiments showed that mutations in Dicer, a key enzyme in miRNA biogenesis, cooperated with oncogenic KRAS and BRAF mutations to promote tumor progression in vivo [59].
In addition, long noncoding RNAs (lncRNAs) act as tissue-specific regulators of gene expression, and their upregulation can promote carcinogenesis, particularly sarcomagenesis. Data on the role of lncRNAs in UPS are limited; however, an in vivo study demonstrated that upregulation of Nuclear Enriched Abundant Transcript 1 (NEAT1) may facilitate UPS metastasis to the lungs [60].
To date, no epigenetic therapy has been proven effective, nor have any of the described epigenetic markers demonstrated prognostic value in UPS. Because epigenetic regulation directly influences chromatin organization, it can contribute to genomic instability and tumor progression, or conversely, enhance sensitivity to standard therapies as a result of drug-induced epigenetic modifications. Further studies in this field are therefore warranted.

4. Changes in UPS Signaling and Associated Therapeutic Approaches

UPS remains poorly understood both clinically and molecularly, largely owing to its intrinsic phenotypic and cytogenetic complexity. As described above, UPS may harbor multiple genetic and epigenetic abnormalities, yet no specific prognostic or predictive biomarkers have been definitively established. The complex genetics of UPS limit the effectiveness of standard chemotherapy, leaving surgical resection and adjuvant radiotherapy as the primary treatment options. Therefore, identifying more effective therapies for UPS patients is urgently needed. Although numerous studies have investigated aberrant signaling in UPS cells, only a few of the most common signaling alterations have been characterized. The therapeutic targets with potential clinical validation and the corresponding possible therapies are summarized below and shown in Figure 1.
Figure 1. Potential prognostic/therapeutic targets in UPS.
Figure 1. Potential prognostic/therapeutic targets in UPS.
Cancers 17 03613 g001
Aberrations in the RB and TP53 genes result in decreased activation of their corresponding signaling pathways. For example, in most tumors without TP53 mutations, deletion or silencing of the p14ARF gene, a negative regulator of MDM2, has been observed, highlighting the potential role of the p14ARF–MDM2–TP53 axis in UPS pathogenesis. Restoring TP53 activity may represent a potential therapeutic approach for UPS. Alterations in RB/TP53 have been shown to shift cancer cell survival toward the oncogenic protein SKP2. TMA data revealed a correlation between loss of RB and TP53 expression and positive SKP2 expression. Inhibition of SKP2 using the neddylation-activating enzyme (NAE) inhibitor pevonedistat suppressed proliferative activity in both patient-derived UPS cells and murine models, providing a rationale for novel systemic therapies [59]. TP53 has been shown to negatively regulate aurora kinases, and loss of p53 results in increased aurora kinase levels, which could serve as a target for therapy. In particular, ENMD-2076, an aurora-A kinase inhibitor with anti-angiogenic properties, demonstrated activity in a phase II study in patients with STS, including three patients with UPS [61,62].
The RAS/mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) pathways are frequently activated in UPS, contributing to tumor progression and poor prognosis [63]. Components of these signaling pathways represent potential therapeutic targets. In preclinical studies, the MEK inhibitor PD325901 slowed tumor growth in vivo [64]. Several clinical trials have evaluated RAS/MAPK inhibitors in large cohorts of patients with STS; however, no responses to sorafenib were observed in UPS [65]. The MEK inhibitor selumetinib induced a partial response in 1 of 2 UPS patients [66]. Similarly, in a phase II study of the mTOR inhibitor temsirolimus, none of the eight UPS patients achieved a response [67]. Notably, the mTOR inhibitor ridaforolimus elicited a partial response in one UPS patient [68]. It is important to note that in vitro, HRAS G12V-driven UPS cells rapidly developed resistance to treatment with either a single MEK or ERK inhibitor; however, combination therapy effectively overcame this resistance [69].
Immunohistochemical analyses of UPS samples frequently reveal phosphorylated AKT (p-AKT), p-mTOR, p-S6RP, p-4EBP, as well as HGF, c-Met, and MEK/ERK, supporting the involvement of PI3K/AKT/mTOR and c-Met signaling in UPS pathogenesis. In preclinical studies, HSP90 overexpression was associated with elevated levels of p-AKT, p-mTOR, and p-S6RP, suggesting its potential as both a poor prognostic marker and a therapeutic target [70]. Notably, the selective HSP90 inhibitor SNX-2112 demonstrated antitumor activity in vitro in UPS cells by inducing apoptosis and autophagy, inhibiting mTOR phosphorylation, and suppressing MAPK signaling [71,72].
Another signaling pathway with oncogenic potential in UPS is the Janus kinase (JAK)–signal transducer and activator of transcription (STAT) pathway, which regulates gene transcription and cell proliferation. In a study of 79 UPS patient samples, phosphorylated STAT3 and its negative regulator SOCS3 were detected in 59.7% and 55.8% of samples, respectively, correlating with favorable and unfavorable prognosis. These findings suggest that STAT3 may serve both as a prognostic marker and a therapeutic target in UPS [73].
Additionally, loss of ATRX expression occurs in 20–30% of UPS cases and confers selective sensitivity to Wnt pathway activation. The Wnt signaling inhibitor tegavivint reduces UPS cell viability, representing a potential therapeutic approach for ATRX-deficient UPS [48].
Aberrant activation of Hedgehog and Notch signaling also contributes to UPS proliferation, involving multiple effectors, including NOTCH3, JAG1, GLI1, PTCH1, HHIP, HES1, HEY1, and HEY2. Deregulation of the Hedgehog-linked tumor suppressor Hippo pathway may further promote tumor progression [74,75,76]. Notably, YAP1, a transcriptional regulator and central effector of the Hippo pathway, is aberrantly stabilized in UPS owing to epigenetic silencing of its inhibitor Angiomotin (AMOT) and Hippo kinase copy number loss. In vivo experiments demonstrated that YAP1–TGFβ crosstalk enhanced cell proliferation and motility [40,74,75,77]. Treatment with epigenetic modulators vorinostat and JQ1 restored AMOT expression and Hippo pathway signaling, highlighting a potential therapeutic strategy warranting further investigation [78]. The TGF-β–linked membrane protein LRRC15 is expressed in several STS, including UPS, highlighting the urgent need for effective therapies. The antibody–drug conjugate ABBV-085, developed to target LRRC15, demonstrated an overall response rate of 20% in a clinical trial of ABBV-085 monotherapy in patients with osteosarcoma or UPS [31,79]. In addition, YAP1 contributes to the activation of NF-κB and VEGF signaling, and its inhibition is related to the modulation of the unfolded protein response (UPR), suggesting these pathways as therapeutically relevant targets [77,80,81]. Overexpression of proteins in NF-κB, VEGF, and UPR pathways may also serve as biomarkers of therapeutic sensitivity. Notably, higher levels of CREB3L1 correlate with increased doxorubicin sensitivity in in vivo studies [82].
A distinct therapeutic approach for UPS is immunotherapy owing to its status as a highly mutated STS subtype, which elicits a pronounced immune response. UPS tumors often exhibit elevated PD-L1 and PD-1 expression, increased T-cell infiltration (positive for CD3, CD8, CD127/IL7 receptor, CD99, CD68, CD10, and negative for TIGIT), and abundant tumor-associated macrophages (positive for CD163, ionized calcium-binding adaptor molecule 1 (Iba1), MSR1, CD204, and SIRPα), alongside a high tumor mutational burden (TMB-H) compared with other STS subtypes [38,83,84,85,86,87,88,89,90,91,92]. Anti-PD-1/PD-L1 drugs have been introduced into UPS therapy [89,93,94], and several clinical trials are ongoing, including: Pembrolizumab (PD-1) + Gemcitabine (pyrimidine antimetabolite) [87,95,96]; Toripalimab (PD-1) + Anlotinib (tyrosine kinase inhibitor, TKI) [90,97]; Carilizumab (PD-1) + Apatinib (TKI) [98]; Envafolimab (PD-L1) + Ipilimumab (CTLA-4) [99]; Nivolumab (PD-1) + Ipilimumab (CTLA-4) [100]; Pembrolizumab (PD-1) + Cyclophosphamide (alkylating agent) [101]; Pembrolizumab (PD-1) monotherapy [101]; checkpoint blockade inhibitors combined with radiation therapy [100,102,103,104]; Nivolumab (PD-L1) + Bempegaldesleukin (CD122-preferential IL-2 pathway agonist) [105,106]; Nivolumab (PD-L1) + Nab-sirolimus (mTOR inhibitor) [107,108]; Pembrolizumab (PD-1) + Eribulin (non-taxane microtubule inhibitor) [109,110]; and Pembrolizumab (PD-1) + Doxorubicin (topoisomerase II inhibitor) [111].
The SARC028 clinical trial demonstrated the efficacy of anti-PD-1 therapy in UPS [89,94]. This study also revealed a functional association between PD-L1 and CKLF-like MARVEL transmembrane domain-containing 6 (CMTM6), identifying CMTM6 as a novel regulator of PD-L1 expression and a prognostic marker associated with poor outcomes. Therapeutic strategies could potentially be guided by CMTM6 expression [112]. Additionally, the collagen-modifying enzyme procollagen-lysine,2-oxoglutarate 5-dioxygenase 2 (PLOD2), which is overexpressed in many tumors relative to normal tissues, has been shown to promote immune evasion in UPS, contributing to tumor metastasis and CD8+ T cell dysfunction. An in vivo study demonstrated that inhibition of PLOD2 reduced tumor growth and enhanced the efficacy of anti-PD-1 therapy, highlighting PLOD2 as a potential novel therapeutic target in UPS immunotherapy [113]. Several additional potential targets are currently being investigated in preclinical studies. The glutaminase inhibitor CB-839 showed therapeutic efficacy in a murine UPS model [114]. The epigenetic modifier NEO2734, which targets BET/EP300, demonstrated antitumor activity both in vitro and in vivo in murine UPS models characterized by activation of MYC-target pathways [115]. Furthermore, cotreatment of UPS xenografts in immunodeficient mice with a dual PI3K/mTOR inhibitor and an anti-IGF1R kinase inhibitor reduced tumor growth in vivo, while also decreasing UPS cell migration and invasion in vitro [116]. Using a comparative oncology approach, DNMT3B was identified as a potential therapeutic target; however, currently available anti-methylation drugs have not yet demonstrated effective clinical activity in UPS [56,117]. Another epigenetic modifier, the HDAC inhibitor LBH589, exhibited anticancer effects both in vitro and in vivo through downregulation of FOS-like antigen 1 (FOSL1) [118]. Additional potential targets include neurotensin receptor 1 (NTSR1) and human tumor endothelin 1 (TEM1), with the NTSR1 inhibitor SR48692 showing antitumor activity in vitro [119,120,121]. The ATM inhibitor AZD0156, in combination with the ATR inhibitor AZD6738, abolished UPS growth in vitro and in vivo [122]. Moreover, the FGFR inhibitor JNJ-42756493 demonstrated anticancer activity in cultured cells and patient-derived xenograft models of UPS, encompassing tumors with diverse phenotypes, prognoses, and molecular features [123].
UPSs are not commonly characterized by the expression of cancer-testis antigens (CTAs), which could potentially be targeted by T-cell receptor (TCR) gene therapy. Notably, a case was reported in which a patient with UPS received NY-ESO-1 TCR-transgenic T cells combined with dendritic cell vaccination and anti-PD-1 therapy, resulting in a durable antitumor response [124]. Similarly, MAGE-A3 TCR-engineered T cells, recognizing epitopes in MAGE-A3, could represent another potential immunotherapeutic approach for UPS [125].
Additional clinical trials investigating UPS therapy are summarized in Table 1.

5. Conclusions

UPS represents an STS subtype with highly heterogeneous genetics, creating considerable challenges in developing individualized treatment strategies. Numerous genetic and epigenetic aberrations in UPS have been investigated, but only a few may contribute to the development of targeted therapies for UPS. The accumulation of multiple genetic abnormalities may also result in hyperactivation of the immune system, making UPS a potential candidate for immunotherapy with checkpoint inhibitors, including anti-PD-1, anti-PD-L1, and anti-CTLA-4. Excluding ineffective therapies could improve treatment selection and patient outcomes. Developing experimental approaches for ex vivo and in vitro testing may help identify and eliminate therapies unlikely to benefit individual UPS patients.

Author Contributions

Conceptualization, E.A.L., K.I.K., G.A.B., B.Y.B., M.A.S., D.V.R. and M.G.Y.; analysis of the literature and table preparation, T.I.F., V.P.M., M.A.S., D.V.R. and A.N.D.; writing—original draft preparation, E.A.L., T.I.F. and V.P.M.; writing—review and editing, E.A.L., K.I.K., G.A.B., B.Y.B., M.A.S., D.V.R. and M.G.Y.; supervision, M.G.Y.; project administration, E.A.L. and K.I.K.; funding acquisition, M.G.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Russian Science Foundation, grant number 23-65-00003.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data for this study were collected from online resources only.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ABCB1ATP Binding Cassette Subfamily B Member 1
ABCG2ATP Binding Cassette Subfamily G Member 2 (JR Blood Group)
AKTAKT Serine/Threonine Kinase
ALKAnaplastic Lymphoma Kinase
AMOTAngiomotin
AMPD2Adenosine Monophosphate Deaminase 2
ANXA2P2Annexin A2 Pseudogene 2
ASPSAlveolar Soft Part Sarcoma
ATMAtaxia Telangiectasia Mutated
ATRATR Serine/Threonine Kinase
ATRXATRX Chromatin Remodeler
AXLAXL Receptor Tyrosine Kinase
BCORBCL6 Corepressor
BET/EP300BET and CBP/EP300 bromodomain
BGNBiglycan
BRAFB-Raf Proto-Oncogene, Serine/Threonine Kinase
CCNBCyclin B
CDCluster Of Differentiation
CDHCadherin
CDK4Cyclin Dependent Kinase 4
CDKN2ACyclin Dependent Kinase Inhibitor 2A
CICCapicua Transcriptional Repressor
CITED2Cbp/P300 Interacting Transactivator with Glu/Asp Rich Carboxy-Terminal Domain 2
CKLFChemokine Like Factor
CLTCClathrin Heavy Chain
CMTM6CKLF Like MARVEL Transmembrane Domain Containing 6
COL3A1Collagen Type III Alpha 1 Chain
CSF2RBColony Stimulating Factor 2 Receptor Subunit Beta
CTLA4Cytotoxic T-Lymphocyte Antigen-4
DCDendritic Cells
DCTN1Dynactin-1
DDLPSDedifferentiated Liposarcoma
DNMT3BDNA Methyltransferase 3 Beta
DUX4L8Double Homeobox 4 Like 8
E2F6E2F Transcription Factor 6
4EBPEukaryotic Translation Initiation Factor 4E Binding Protein 1
EGFREpidermal Growth Factor Receptor
EIF2AK4Eukaryotic Translation Initiation Factor 2 Alpha Kinase 4
EML4Echinoderm Microtubule-Associated Protein-Like 4 
ETV6ETS Variant Transcription Factor 6
EWSR1Ewing Sarcoma RNA Binding Protein 1
FARP1FERM, ARH/Rhogef and Pleckstrin Domain Protein 1
FDRFalse Discovery Rate
FGFFibroblast Growth Factor
FGFRFibroblast Growth Factor Receptor
FKBP4FK506-Binding Protein 4
FOSL1FOS Like 1, AP-1 Transcription Factor Subunit
GLI1GLI Family Zinc Finger 1
GNASGuanine Nucleotide Binding Protein (G Protein), Alpha Stimulating Activity Polypeptide
GULP1GULP PTB Domain Containing Engulfment Adaptor 1
HES1Hes Family BHLH Transcription Factor 1
HEYHes Related Family BHLH Transcription Factor with YRPW Motif 
HGFHepatocyte Growth Factor
HHIPHedgehog Interacting Protein
HSP90Heat Shock Protein 90
Iba1Ionized Calcium-Binding Adapter Molecule 1
IGF1RInsulin Like Growth Factor 1 Receptor
IL-7Interleukin-7
IMP3IMP U3 Small Nucleolar Ribonucleoprotein 3
JAG1Jagged Canonical Notch Ligand 1
JAKJanus Kinase
KITKIT Proto-Oncogene, Receptor Tyrosine Kinase
KRASKRAS Proto-Oncogene, Gtpase
LINCLinkers Of Nucleoskeleton and Cytoskeleton
LMNALamin A/C
lncRNALong Non-Coding RNA
LPSLiposarcoma
LRRC15Leucine Rich Repeat Containing 15
MACROD2Mono-ADP Ribosylhydrolase 2
MAGE-A3Melanoma-Associated Antigen 3, Family Member A3
MAPKMitogen-Activated Protein Kinase
MDM2Mouse Double Minute 2
MED12Mediator Complex Subunit
MEK (MAP2K1)Mitogen-Activated Protein Kinase Kinase 1
MFHMalignant Fibrous Histiocytoma
MFSMyxofibrosarcoma
MKI67Marker of Proliferation Ki-67
MMPMatrix Metallopeptidase
MPNSTMalignant Peripheral Nerve Sheath Tumor
MRIMagnetic Resonance Imaging
MSR1Macrophage Scavenger Receptor 1 
mTORMechanistic Target of Rapamycin Kinase
MVPMajor Vault Protein
MYST1MYST Histone Acetyltransferase 1
NAENEDD8 Activating Enzyme
NCOR1Nuclear Receptor Corepressor 1
NEAT1Nuclear Enriched Abundant Transcript 1
NF-kBNuclear Factor Kappa B
NOTCHNotch Receptor
NPM1Nucleophosmin 1
NTRKNeurotrophic Tropomyosin Receptor Kinase
NTSR1Neurotensin Receptor 1
NY-ESO- 1New York esophageal squamous cell carcinoma 1
PCLOPiccolo Presynaptic Cytomatrix Protein
PCRPolymerase Chain Reaction
PD1Programmed Cell Death Protein 1
PDCD11Programmed Cell Death 11
PDGFRPlatelet Derived Growth Factor Receptor
PD-L1Programmed Cell Death Protein Ligand 1
PI3KPhosphatidylinositol-4,5-Bisphosphate 3-Kinase
PIK3CAPhosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha
PLK1Polo Like Kinase 1
PLOD2Procollagen-Lysine,2-Oxoglutarate 5-Dioxygenase 2 
PRDMPR/SET Domain
PTCH1Patched 1
PTENPhosphatase and Tensin Homolog
RB1Retinoblastoma
RNASEH2BRibonuclease H2, Subunit B
ROR2Receptor Tyrosine Kinase Like Orphan Receptor 2
S6RPRibosomal Protein S6
SARMSterile Alpha and TIR Motif Containing
SF3B1Splicing Factor 3b Subunit 1
SIRPαSignal Regulatory Protein Alpha
SKP2S-Phase Kinase Associated Protein 2
SMC1AStructural Maintenance of Chromosomes 1A
SOCS3Suppressor of Cytokine Signaling 3
SSSynovial Sarcoma
STATSignal Transducer and Activator of Transcription
STK24Serine/Threonine Kinase 24
STSSoft Tissue Sarcoma
TCGAThe Cancer Genome Atlas
TEM-1Tumor endothelin 1
TGFβTransforming Growth Factor Beta
TIGITT Cell Immunoreceptor with Ig and ITIM Domains
TKITyrosine Kinase Inhibitor
TMATissue Microarray
TMBTumor Mutational Burden
TMTCTransmembrane And Tetratricopeptide Repeat Containing
TP53Tumor Protein P53
TRIOTrio Rho Guanine Nucleotide Exchange Factor
VEGFVascular Endothelial Growth Factor
VGLL3Vestigial Like Family Member 3 
VMP1Vacuole Membrane Protein 1
WNT7BWingless-Type MMTV Integration Site Family, Member 7B
WWTR1 (TAZ)WW Domain Containing Transcription Regulator 1
UPAPlasminogen Activator, Urokinase
UPSUndifferentiated Pleomorphic Sarcoma
UPRUnfolded Protein Response
YAP1Yes1 Associated Transcriptional Regulator

References

  1. Sbaraglia, M.; Bellan, E.; Dei Tos, A.P. The 2020 WHO Classification of Soft Tissue Tumours: News and Perspectives. Pathologica 2020, 113, 70–84. [Google Scholar] [CrossRef]
  2. Goldblum, J.R. An Approach to Pleomorphic Sarcomas: Can We Subclassify, and Does It Matter? Mod. Pathol. 2014, 27, S39–S46. [Google Scholar] [CrossRef]
  3. Carter, C.S.; Patel, R.M. Cutaneous Soft Tissue Tumors: Diagnostically Disorienting Epithelioid Tumors That Are Not Epithelial, and Other Perplexing Mesenchymal Lesions. Mod. Pathol. 2020, 33, 66–82. [Google Scholar] [CrossRef] [PubMed]
  4. Vaitiekiene, A.; Vaitiekus, D.; Urbonaite, L.; Jankauskas, A.; Portacenko, J.; Lapinskas, T.; Benetis, R.; Siudikas, A.; Veikutiene, A.; Poskiene, L.; et al. Multidisciplinary Approach to Rare Primary Cardiac Sarcoma: A Case Report and Review. BMC Cancer 2019, 19, 529. [Google Scholar] [CrossRef] [PubMed]
  5. Chen, S.X.; Eichenfield, D.Z.; Orme, C.; Hinds, B. Pleomorphic Dermal Sarcoma in a Man with HIV: Report with next-Generation Sequencing Analysis and Review of the Atypical Fibroxanthoma/Pleomorphic Dermal Sarcoma Spectrum. Dermatol. Online J. 2019, 25, 13030. [Google Scholar] [CrossRef]
  6. Jibbe, A.; Worley, B.; Miller, C.H.; Alam, M. Surgical Excision Margins for Fibrohistiocytic Tumors, Including Atypical Fibroxanthoma and Undifferentiated Pleomorphic Sarcoma: A Probability Model Based on a Systematic Review. J. Am. Acad. Dermatol. 2022, 87, 833–840. [Google Scholar] [CrossRef]
  7. Thway, K.; Fisher, C. Undifferentiated and dedifferentiated soft tissue neoplasms: Immunohistochemical surrogates for differential diagnosis. Semin. Diagn. Pathol. 2021, 38, 170–186. [Google Scholar] [CrossRef]
  8. Sun, H.; Liu, J.; Hu, F.; Xu, M.; Leng, A.; Jiang, F.; Chen, K. Current Research and Management of Undifferentiated Pleomorphic Sarcoma/Myofibrosarcoma. Front. Genet. 2023, 14, 1109491. [Google Scholar] [CrossRef]
  9. Mairal, A.; Chibon, F.; Rousselet, A.; Couturier, J.; Terrier, P.; Aurias, A. Establishment of a Human Malignant Fibrous Histiocytoma Cell Line, COMA. Cancer Genet. Cytogenet. 2000, 121, 117–123. [Google Scholar] [CrossRef]
  10. Stefano, S.; Giovanni, S. The PTEN Tumor Suppressor Gene in Soft Tissue Sarcoma. Cancers 2019, 11, 1169. [Google Scholar] [CrossRef]
  11. Bai, C.; Zhang, L.; Wang, Y.; You, X.; Ju, Y.; Sun, T.; Fan, Z. A Novel TMTC2-NTRK3 Fusion in Undifferentiated High-Grade Pleomorphic Sarcoma. J. Cancer Res. Clin. Oncol. 2022, 148, 2933–2937. [Google Scholar] [CrossRef] [PubMed]
  12. Goh, X.N.; Seng, M.S.-F.; Loh, A.H.P.; Gupta, A.; Chang, K.T.E.; Iyer, P. Larotrectinib Followed by Selitrectinib in a Novel DCTN1–NTRK1 Fusion Undifferentiated Pleomorphic Sarcoma. J. Oncol. Pharm. Pract. 2021, 27, 485–489. [Google Scholar] [CrossRef] [PubMed]
  13. Zhou, N.; Schäfer, R.; Li, T.; Fang, M.; Liu, L. A Primary Undifferentiated Pleomorphic Sarcoma of the Lumbosacral Region Harboring a LMNA-NTRK1 Gene Fusion with Durable Clinical Response to Crizotinib: A Case Report. BMC Cancer 2018, 18, 842. [Google Scholar] [CrossRef] [PubMed]
  14. Ali, N.M.; Niada, S.; Brini, A.T.; Morris, M.R.; Kurusamy, S.; Alholle, A.; Huen, D.; Antonescu, C.R.; Tirode, F.; Sumathi, V.; et al. Genomic and Transcriptomic Characterization of Undifferentiated Pleomorphic Sarcoma of Bone. J. Pathol. 2019, 247, 166–176. [Google Scholar] [CrossRef]
  15. Bou-Maroun, L.M.; Hoff, L.; Joshi, A.; Bloom, D.A.; Heider, A.; Geiger, J.D.; Wu, Y.; Robinson, D.; Mody, R.; Rao, R.J. Undifferentiated Pleomorphic Sarcoma of the Pancreas with Novel SARM1-NTRK 1 Gene Fusion and Associated Pancreatitis, Panniculitis, and Polyarthritis Syndrome. Pediatr. Blood Cancer 2024, 71, e30819. [Google Scholar] [CrossRef]
  16. Goffinet, S.; Di Mauro, I.; Doyen, J.; Boyer, J.; Birtwisle-Peyrottes, I.; Keslair, F.; Pedeutour, F.; Dadone-Montaudie, B. Vanished MDM2 Amplification in Multiple Recurrences of an Irradiated Poorly Differentiated Sarcoma with Amplified TRIO::TERT Fusion Gene. Genes. Chromosomes Cancer 2023, 62, 342–352. [Google Scholar] [CrossRef]
  17. Delespaul, L.; Lesluyes, T.; Pérot, G.; Brulard, C.; Lartigue, L.; Baud, J.; Lagarde, P.; Le Guellec, S.; Neuville, A.; Terrier, P.; et al. Recurrent TRIO Fusion in Nontranslocation–Related Sarcomas. Clin. Cancer Res. 2017, 23, 857–867. [Google Scholar] [CrossRef]
  18. Suster, D.I.; Deshpande, V.; Chebib, I.; Taylor, M.S.; Mullen, J.; Bredella, M.A.; Nielsen, G.P. Spindle Cell Liposarcoma with a TRIO-TERT Fusion Transcript. Virchows Arch. 2019, 475, 391–394. [Google Scholar] [CrossRef]
  19. Zhang, S.; Liao, X.; Chen, J. EML4-ALK Rearrangement in Primary Malignant Fibrous Histiocytoma of the Lung Treated with Alectinib: A Case Report. Front. Oncol. 2022, 12, 978327. [Google Scholar] [CrossRef]
  20. Schneider, J.L.; Lin, J.J.; Shaw, A.T. ALK-positive lung cancer: A moving target. Nat. Cancer 2023, 4, 330–343. [Google Scholar] [CrossRef]
  21. Elshatlawy, M.; Sampson, J.; Clarke, K.; Bayliss, R. EML4-ALK biology and drug resistance in non-small cell lung cancer: A new phase of discoveries. Mol. Oncol. 2023, 17, 950–963. [Google Scholar] [CrossRef] [PubMed]
  22. Zheng, B.; Zhang, S.; Cai, W.; Wang, J.; Wang, T.; Tang, N.; Shi, Y.; Luo, X.; Yan, W. Identification of Novel Fusion Transcripts in Undifferentiated Pleomorphic Sarcomas by Transcriptome Sequencing. Cancer Genom.-Proteom. 2019, 16, 399–408. [Google Scholar] [CrossRef] [PubMed]
  23. Hofvander, J.; Tayebwa, J.; Nilsson, J.; Magnusson, L.; Brosjö, O.; Larsson, O.; Vult von Steyern, F.; Mandahl, N.; Fletcher, C.D.M.; Mertens, F. Recurrent PRDM10 Gene Fusions in Undifferentiated Pleomorphic Sarcoma. Clin. Cancer Res. 2015, 21, 864–869. [Google Scholar] [CrossRef] [PubMed]
  24. Hofvander, J.; Puls, F.; Pillay, N.; Steele, C.D.; Flanagan, A.M.; Magnusson, L.; Nilsson, J.; Mertens, F. Undifferentiated Pleomorphic Sarcomas with PRDM10 Fusions Have a Distinct Gene Expression Profile. J. Pathol. 2019, 249, 425–434. [Google Scholar] [CrossRef]
  25. Mistik, O.; Sayar, H. Immunohistochemical Positive Regulatory Domain Member 10 Expression in Soft Tissue Sarcomas. Pol. J. Pathol. 2022, 73, 223–232. [Google Scholar] [CrossRef]
  26. Lei, T.; Shen, Z.; Shen, M.; Du, L.; Shi, Y.; Peng, Y.; Zhou, Z.; Da, W.; Chen, X.; Li, Q. Clinicopathological and Genetic Characterization of Radiotherapy-Induced Undifferentiated Pleomorphic Sarcoma Following Breast Cancer: A Case Series of Three Tumors and Comprehensive Literature Review. Diagn. Pathol. 2024, 19, 110. [Google Scholar] [CrossRef]
  27. Rüping, K.; Altendorf-Hofmann, A.; Chen, Y.; Kampmann, E.; Gibis, S.; Lindner, L.; Katenkamp, D.; Petersen, I.; Knösel, T. High IGF2 and FGFR3 Are Associated with Tumour Progression in Undifferentiated Pleomorphic Sarcomas, but EGFR and FGFR3 Mutations Are a Rare Event. J. Cancer Res. Clin. Oncol. 2014, 140, 1315–1322. [Google Scholar] [CrossRef]
  28. Urbini, M.; Astolfi, A.; Indio, V.; Nannini, M.; Pizzi, C.; Paolisso, P.; Tarantino, G.; Pantaleo, M.A.; Saponara, M. Genetic Aberrations and Molecular Biology of Cardiac Sarcoma. Ther. Adv. Med. Oncol. 2020, 12, 1758835920918492. [Google Scholar] [CrossRef]
  29. Hong, C.S.; Partovi, E.; Clune, J.; Huttner, A.; Park, H.S.; Omay, S.B. Genomic Characterization of Radiation-Induced Intracranial Undifferentiated Pleomorphic Sarcoma. Case Rep. Genet. 2021, 2021, 1–5. [Google Scholar] [CrossRef]
  30. Ray, U.; Pathoulas, C.L.; Thirusangu, P.; Purcell, J.W.; Kannan, N.; Shridhar, V. Exploiting LRRC15 as a Novel Therapeutic Target in Cancer. Cancer Res. 2022, 82, 1675–1681. [Google Scholar] [CrossRef]
  31. Demetri, G.D.; Luke, J.J.; Hollebecque, A.; Powderly, J.D.; Spira, A.I.; Subbiah, V.; Naumovski, L.; Chen, C.; Fang, H.; Lai, D.W.; et al. First-in-Human Phase I Study of ABBV-085, an Antibody–Drug Conjugate Targeting LRRC15, in Sarcomas and Other Advanced Solid Tumors. Clin. Cancer Res. 2021, 27, 3556–3566. [Google Scholar] [CrossRef]
  32. Fullenkamp, C.A.; Hall, S.L.; Jaber, O.I.; Pakalniskis, B.L.; Savage, E.C.; Savage, J.M.; Ofori-Amanfo, G.K.; Lambertz, A.M.; Ivins, S.D.; Stipp, C.S.; et al. TAZ and YAP Are Frequently Activated Oncoproteins in Sarcomas. Oncotarget 2016, 7, 30094–30108. [Google Scholar] [CrossRef] [PubMed]
  33. Roland, C.L.; May, C.D.; Watson, K.L.; Al Sannaa, G.A.; Dineen, S.P.; Feig, R.; Landers, S.; Ingram, D.R.; Wang, W.-L.; Guadagnolo, B.A.; et al. Analysis of Clinical and Molecular Factors Impacting Oncologic Outcomes in Undifferentiated Pleomorphic Sarcoma. Ann. Surg. Oncol. 2016, 23, 2220–2228. [Google Scholar] [CrossRef] [PubMed]
  34. Williams, E.A.; Vegas, I.; El-Senduny, F.F.; Zhang, J.; Mata, D.A.; Hiemenz, M.C.; Hughes, S.R.; Sa, B.C.; Kraft, G.P.; Gorbatov, N.; et al. Pan-Cancer Genomic Analysis of AXL Mutations Reveals a Novel, Recurrent, Functionally Activating AXL W451C Alteration Specific to Myxofibrosarcoma. Am. J. Surg. Pathol. 2024, 48, 699–707. [Google Scholar] [CrossRef] [PubMed]
  35. Matsuoka, H.; Yoshida, K.I.; Nakai, S.; Suzuki, R.; Imura, Y.; Takami, H.; Watanabe, M.; Wakamatsu, T.; Tamiya, H.; Outani, H.; et al. Successful pazopanib treatment of undifferentiated pleomorphic sarcoma with coamplification of PDGFRA, VEGFR2 and KIT: A case report. Mol. Clin. Oncol. 2024, 21, 69. [Google Scholar] [CrossRef]
  36. Li, B.; Li, L.; Li, X.; Wang, Y.; Xie, Y.; Liu, C.; Li, F. Undifferentiated Pleomorphic Sarcoma with Co-Existence of KRAS/PIK3CA Mutations. Int. J. Clin. Exp. Pathol. 2015, 8, 8563–8567. [Google Scholar]
  37. Vanni, S.; Fausti, V.; Fonzi, E.; Liverani, C.; Miserocchi, G.; Spadazzi, C.; Cocchi, C.; Calabrese, C.; Gurrieri, L.; Riva, N.; et al. Unveiling the Genomic Basis of Chemosensitivity in Sarcomas of the Extremities: An Integrated Approach for an Unmet Clinical Need. Int. J. Mol. Sci. 2023, 24, 6926. [Google Scholar] [CrossRef]
  38. Higuchi, M.; Yamada, H.; Machino, K.; Oshibe, I.; Soeta, N.; Saito, T.; Uramoto, H.; Yamada, S.; Hojo, H.; Suzuki, H. Successful Multidisciplinary Treatment for Aggressive Primary Pulmonary Undifferentiated Pleomorphic Sarcoma. Case Rep. Oncol. 2020, 13, 385–391. [Google Scholar] [CrossRef]
  39. Suzuki, H.; Fukuda, M.; Shirono, T.; Kondo, R.; Tanaka, T.; Niizeki, T.; Akiba, J.; Koga, H.; Kawaguchi, T. A Rare Case of Primary Hepatic Undifferentiated Pleomorphic Sarcoma: Exploring Cancer-Related Gene Mutations. Intern. Med. 2025, 64, 1653–1658. [Google Scholar] [CrossRef]
  40. Abeshouse, A.; Adebamowo, C.; Adebamowo, S.N.; Akbani, R.; Akeredolu, T.; Ally, A.; Anderson, M.L.; Anur, P.; Appelbaum, E.L.; Armenia, J.; et al. Comprehensive and Integrated Genomic Characterization of Adult Soft Tissue Sarcomas. Cell 2017, 171, 950–965.e28. [Google Scholar] [CrossRef]
  41. Ramsey, J.K.; Chen, J.L.; Schoenfield, L.; Cho, R.I. Undifferentiated Pleomorphic Sarcoma Metastatic to the Orbit. Ophthalmic Plast. Reconstr. Surg. 2018, 34, e193–e195. [Google Scholar] [CrossRef] [PubMed]
  42. Orth, M.F.; Gerke, J.S.; Knösel, T.; Altendorf-Hofmann, A.; Musa, J.; Alba-Rubio, R.; Stein, S.; Hölting, T.L.B.; Cidre-Aranaz, F.; Romero-Pérez, L.; et al. Functional Genomics Identifies AMPD2 as a New Prognostic Marker for Undifferentiated Pleomorphic Sarcoma. Int. J. Cancer 2019, 144, 859–867. [Google Scholar] [CrossRef] [PubMed]
  43. Cullen, M.M.; Lazarides, A.L.; Pittman, P.D.; Flamant, E.M.; Stoeber, K.L.; Stoeber, K.; Visguass, J.D.; Brigman, B.E.; Riedel, R.F.; Cardona, D.M.; et al. Cell-Cycle Phase Progression Analysis Identifies Three Unique Phenotypes in Soft Tissue Sarcoma. BMC Cancer 2024, 24, 1288. [Google Scholar] [CrossRef] [PubMed]
  44. Moiseeva, N.I.; Laletina, L.A.; Fetisov, T.I.; Makhmudova, L.F.; Manikaylo, A.E.; Fomina, L.Y.; Burov, D.A.; Lesovaya, E.A.; Bokhyan, B.Y.; Zinovieva, V.Y.; et al. Analysis of Multiple Drug Resistance Mechanism in Different Types of Soft Tissue Sarcomas: Assessment of the Expression of ABC-Transporters, MVP, YB-1, and Analysis of Their Correlation with Chemosensitivity of Cancer Cells. Int. J. Mol. Sci. 2022, 23, 3183. [Google Scholar] [CrossRef]
  45. Lewin, J.; Garg, S.; Lau, B.Y.; Dickson, B.C.; Traub, F.; Gokgoz, N.; Griffin, A.M.; Ferguson, P.C.; Andrulis, I.L.; Sim, H.; et al. Identifying Actionable Variants Using next Generation Sequencing in Patients with a Historical Diagnosis of Undifferentiated Pleomorphic Sarcoma. Int. J. Cancer 2018, 142, 57–65. [Google Scholar] [CrossRef]
  46. Pan, M.; Zhou, M.Y.; Jiang, C.; Zhang, Z.; Bui, N.Q.; Bien, J.; Siy, A.; Achacoso, N.; Solorzano, A.V.; Tse, P.; et al. Sex-Dependent Prognosis of Patients with Advanced Soft Tissue Sarcoma. Clin. Cancer Res. 2024, 30, 413–419. [Google Scholar] [CrossRef]
  47. Kirilin, E.M.; Fetisov, T.I.; Moiseeva, N.I.; Lesovaya, E.A.; Laletina, L.A.; Makhmudova, L.F.; Manikaylo, A.E.; Fomina, L.Y.; Burov, D.A.; Bokhyan, B.Y.; et al. Soft Tissue Sarcoma Study: Association of Genetic Alterations in the Apoptosis Pathways with Chemoresistance to Doxorubicin. Cancers 2022, 14, 1796. [Google Scholar] [CrossRef]
  48. Fang, Y.; Barrows, D.; Dabas, Y.; Carroll, T.S.; Singer, S.; Tap, W.D.; Nacev, B.A. ATRX Guards against Aberrant Differentiation in Mesenchymal Progenitor Cells. Nucleic Acids Res. 2024, 52, 4950–4968. [Google Scholar] [CrossRef]
  49. Li, G.Z.; Okada, T.; Kim, Y.-M.; Agaram, N.P.; Sanchez-Vega, F.; Shen, Y.; Tsubokawa, N.; Rios, J.; Martin, A.S.; Dickson, M.A.; et al. Rb and P53-Deficient Myxofibrosarcoma and Undifferentiated Pleomorphic Sarcoma Require Skp2 for Survival. Cancer Res. 2020, 80, 2461–2471. [Google Scholar] [CrossRef]
  50. Francis, P.; Namløs, H.; Müller, C.; Edén, P.; Fernebro, J.; Berner, J.-M.; Bjerkehagen, B.; Åkerman, M.; Bendahl, P.-O.; Isinger, A.; et al. Diagnostic and Prognostic Gene Expression Signatures in 177 Soft Tissue Sarcomas: Hypoxia-Induced Transcription Profile Signifies Metastatic Potential. BMC Genom. 2007, 8, 73. [Google Scholar] [CrossRef]
  51. Klein, J.C.; Wang, L.; Strand, D.; Lastufka, C.; Hosler, G.A.; Hon, G.C. Single-Cell and Spatial Transcriptomics Identify COL6A3 as a Prognostic Biomarker in Undifferentiated Pleomorphic Sarcoma. Mol. Cancer 2024, 23, 257. [Google Scholar] [CrossRef] [PubMed]
  52. Yang, X.; Huang, W.; He, R.; Ma, J.; Lin, P.; Xie, Z.; Ma, F.; Chen, G. Determining the Prognostic Significance of Alternative Splicing Events in Soft Tissue Sarcoma Using Data from The Cancer Genome Atlas. J. Transl. Med. 2019, 17, 283. [Google Scholar] [CrossRef] [PubMed]
  53. Carneiro, A.; Francis, P.; Bendahl, P.-O.; Fernebro, J.; Åkerman, M.; Fletcher, C.; Rydholm, A.; Borg, Å.; Nilbert, M. Indistinguishable Genomic Profiles and Shared Prognostic Markers in Undifferentiated Pleomorphic Sarcoma and Leiomyosarcoma: Different Sides of a Single Coin? Lab. Investig. 2009, 89, 668–675. [Google Scholar] [CrossRef] [PubMed]
  54. Ibstedt, S.; Piccinelli, P.; Sydow, S.; Köster, J.; Mertens, F. Structural Variants in the SMC1A Gene Associated with Near-Haploidy in Undifferentiated Pleomorphic Sarcomas. Genes Chromosomes Cancer 2024, 63, e23255. [Google Scholar] [CrossRef]
  55. Aoki, Y.; Yamamoto, J.; Tome, Y.; Hamada, K.; Masaki, N.; Inubushi, S.; Tashiro, Y.; Bouvet, M.; Endo, I.; Nishida, K.; et al. Over-Methylation of Histone H3 Lysines Is a Common Molecular Change Among the Three Major Types of Soft-Tissue Sarcoma in Patient-Derived Xenograft (PDX) Mouse Models. Cancer Genom.-Proteom. 2021, 18, 715–721. [Google Scholar] [CrossRef]
  56. Fuller, A.M.; DeVine, A.; Murazzi, I.; Mason, N.J.; Weber, K.; Eisinger-Mathason, T.S.K. Comparative Oncology Reveals DNMT3B as a Molecular Vulnerability in Undifferentiated Pleomorphic Sarcoma. Cell. Oncol. 2022, 45, 1277–1295. [Google Scholar] [CrossRef]
  57. Guled, M.; Pazzaglia, L.; Borze, I.; Mosakhani, N.; Novello, C.; Benassi, M.S.; Knuutila, S. Differentiating Soft Tissue Leiomyosarcoma and Undifferentiated Pleomorphic Sarcoma: A MiRNA Analysis. Genes Chromosomes Cancer 2014, 53, 693–702. [Google Scholar] [CrossRef]
  58. Zoroddu, S.; Lucariello, A.; De Luca, A.; Bagella, L. Dysregulation of MiRNAs in Soft Tissue Sarcomas. Cells 2024, 13, 1853. [Google Scholar] [CrossRef]
  59. Mito, J.K.; Min, H.D.; Ma, Y.; Carter, J.E.; Brigman, B.E.; Dodd, L.; Dankort, D.; McMahon, M.; Kirsch, D.G. Oncogene-dependent Control of miRNA Biogenesis and Metastatic Progression in a Model of Undifferentiated Pleomorphic Sarcoma. J. Pathol. 2013, 229, 132–140. [Google Scholar] [CrossRef]
  60. Huang, J.; Sachdeva, M.; Xu, E.; Robinson, T.J.; Luo, L.; Ma, Y.; Williams, N.T.; Lopez, O.; Cervia, L.D.; Yuan, F.; et al. The Long Noncoding RNA NEAT1 Promotes Sarcoma Metastasis by Regulating RNA Splicing Pathways. Mol. Cancer Res. 2020, 18, 1534–1544. [Google Scholar] [CrossRef]
  61. Thoenen, E.; Curl, A.; Iwakuma, T. TP53 in Bone and Soft Tissue Sarcomas. Pharmacol. Ther. 2019, 202, 149–164. [Google Scholar] [CrossRef] [PubMed]
  62. Veitch, Z.; Zer, A.; Loong, H.; Salah, S.; Masood, M.; Gupta, A.; Bradbury, P.A.; Hogg, D.; Wong, A.; Kandel, R.; et al. A Phase II Study of ENMD-2076 in Advanced Soft Tissue Sarcoma (STS). Sci. Rep. 2019, 9, 7390. [Google Scholar] [CrossRef] [PubMed]
  63. Serrano, C.; Romagosa, C.; Hernández-Losa, J.; Simonetti, S.; Valverde, C.; Moliné, T.; Somoza, R.; Pérez, M.; Vélez, R.; Vergés, R.; et al. RAS/MAPK Pathway Hyperactivation Determines Poor Prognosis in Undifferentiated Pleomorphic Sarcomas. Cancer 2016, 122, 99–107. [Google Scholar] [CrossRef] [PubMed]
  64. Dodd, R.D.; Mito, J.K.; Eward, W.C.; Chitalia, R.; Sachdeva, M.; Ma, Y.; Barretina, J.; Dodd, L.; Kirsch, D.G. NF1 Deletion Generates Multiple Subtypes of Soft-Tissue Sarcoma That Respond to MEK Inhibition. Mol. Cancer Ther. 2013, 12, 1906–1917. [Google Scholar] [CrossRef]
  65. Maki, R.G.; D’Adamo, D.R.; Keohan, M.L.; Saulle, M.; Schuetze, S.M.; Undevia, S.D.; Livingston, M.B.; Cooney, M.M.; Hensley, M.L.; Mita, M.M.; et al. Phase II Study of Sorafenib in Patients with Metastatic or Recurrent Sarcomas. J. Clin. Oncol. 2009, 27, 3133–3140. [Google Scholar] [CrossRef]
  66. Eroglu, Z.; Tawbi, H.A.; Hu, J.; Guan, M.; Frankel, P.H.; Ruel, N.H.; Wilczynski, S.; Christensen, S.; Gandara, D.R.; Chow, W.A. A Randomised Phase II Trial of Selumetinib vs. Selumetinib plus Temsirolimus for Soft-Tissue Sarcomas. Br. J. Cancer 2015, 112, 1644–1651. [Google Scholar] [CrossRef]
  67. Okuno, S.; Bailey, H.; Mahoney, M.R.; Adkins, D.; Maples, W.; Fitch, T.; Ettinger, D.; Erlichman, C.; Sarkaria, J.N. A Phase 2 Study of Temsirolimus (CCI-779) in Patients with Soft Tissue Sarcomas. Cancer 2011, 117, 3468–3475. [Google Scholar] [CrossRef]
  68. Chawla, S.P.; Blay, J.; Ray-Coquard, I.L.; Le Cesne, A.; Staddon, A.P.; Milhem, M.M.; Penel, N.; Riedel, R.F.; Bui Nguyen, B.; Cranmer, L.D.; et al. Results of the Phase III, Placebo-Controlled Trial (SUCCEED) Evaluating the MTOR Inhibitor Ridaforolimus (R) as Maintenance Therapy in Advanced Sarcoma Patients (Pts) Following Clinical Benefit from Prior Standard Cytotoxic Chemotherapy (CT). J. Clin. Oncol. 2011, 29, 10005. [Google Scholar] [CrossRef]
  69. Catalano, A.; Adlesic, M.; Kaltenbacher, T.; Klar, R.F.U.; Albers, J.; Seidel, P.; Brandt, L.P.; Hejhal, T.; Busenhart, P.; Röhner, N.; et al. Sensitivity and Resistance of Oncogenic RAS-Driven Tumors to Dual MEK and ERK Inhibition. Cancers 2021, 13, 1852. [Google Scholar] [CrossRef]
  70. Bekki, H.; Kohashi, K.; Maekawa, A.; Yamada, Y.; Yamamoto, H.; Harimaya, K.; Hakozaki, M.; Nabeshima, K.; Iwamoto, Y.; Oda, Y. Elevated Expression of HSP90 and the Antitumor Effect of an HSP90 Inhibitor via Inactivation of the Akt/MTOR Pathway in Undifferentiated Pleomorphic Sarcoma. BMC Cancer 2015, 15, 804. [Google Scholar] [CrossRef]
  71. Fukuoka, N.; Nakamura, O.; Yamagami, Y.; Nishimura, H.; Ishibashi, Y.; Yamamoto, T. SNX-2112 Induces Apoptosis and Autophagy of Nara-H Cells. Anticancer. Res. 2018, 38, 5177–5181. [Google Scholar] [CrossRef]
  72. Lahat, G.; Zhang, P.; Zhu, Q.-S.; Torres, K.; Ghadimi, M.; Smith, K.D.; Wang, W.-L.; Lazar, A.J.; Lev, D. The Expression of C-Met Pathway Components in Unclassified Pleomorphic Sarcoma/Malignant Fibrous Histiocytoma (UPS/MFH): A Tissue Microarray Study. Histopathology 2011, 59, 556–561. [Google Scholar] [CrossRef]
  73. Bekki, H.; Kohashi, K.; Yamada, Y.; Iura, K.; Ishii, T.; Maekawa, A.; Otsuka, H.; Yamamoto, H.; Hakozaki, M.; Nabeshima, K.; et al. Phosphorylation of STAT3 in Undifferentiated Pleomorphic Sarcoma Is Correlated with a Favorable Prognosis. Pathobiology 2017, 84, 161–169. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, C.Y.Y.; Wei, Q.; Han, I.; Sato, S.; Azarnier, R.G.-; Whetstone, H.; Poon, R.; Hu, J.; Zheng, F.; Zhang, P.; et al. Hedgehog and Notch Signaling Regulate Self-Renewal of Undifferentiated Pleomorphic Sarcomas. Cancer Res. 2012, 72, 1013–1022. [Google Scholar] [CrossRef] [PubMed]
  75. Amm, H.M.; DeVilliers, P.; Srivastava, A.R.; Diniz, M.G.; Siegal, G.P.; MacDougall, M. Mandibular Undifferentiated Pleomorphic Sarcoma: Molecular Analysis of a Primary Cell Population. Clin. Exp. Dent. Res. 2020, 6, 495–505. [Google Scholar] [CrossRef] [PubMed]
  76. Ye, S.; Liu, Y.; Fuller, A.M.; Katti, R.; Ciotti, G.E.; Chor, S.; Alam, M.Z.; Devalaraja, S.; Lorent, K.; Weber, K.; et al. TGFβ and Hippo Pathways Cooperate to Enhance Sarcomagenesis and Metastasis through the Hyaluronan-Mediated Motility Receptor (HMMR). Mol. Cancer Res. 2020, 18, 560–573. [Google Scholar] [CrossRef]
  77. Rivera-Reyes, A.; Ye, S.; Marino, G.E.; Egolf, S.; Ciotti, G.E.; Chor, S.; Liu, Y.; Posimo, J.M.; Park, P.M.C.; Pak, K.; et al. YAP1 Enhances NF-ΚB-Dependent and Independent Effects on Clock-Mediated Unfolded Protein Responses and Autophagy in Sarcoma. Cell Death Dis. 2018, 9, 1108. [Google Scholar] [CrossRef]
  78. Ye, S.; Lawlor, M.A.; Rivera-Reyes, A.; Egolf, S.; Chor, S.; Pak, K.; Ciotti, G.E.; Lee, A.C.; Marino, G.E.; Shah, J.; et al. YAP1-Mediated Suppression of USP31 Enhances NFκB Activity to Promote Sarcomagenesis. Cancer Res. 2018, 78, 2705–2720. [Google Scholar] [CrossRef]
  79. Available online: https://Clinicaltrials.Gov/Study/NCT02565758 (accessed on 11 August 2025).
  80. Washimi, K.; Kasajima, R.; Shimizu, E.; Sato, S.; Okubo, Y.; Yoshioka, E.; Narimatsu, H.; Hiruma, T.; Katayama, K.; Yamaguchi, R.; et al. Histological Markers, Sickle-Shaped Blood Vessels, Myxoid Area, and Infiltrating Growth Pattern Help Stratify the Prognosis of Patients with Myxofibrosarcoma/Undifferentiated Sarcoma. Sci. Rep. 2023, 13, 6744. [Google Scholar] [CrossRef]
  81. Lv, X.; Liu, J.; Islam, K.; Ruan, J.; He, C.; Chen, P.; Huang, C.; Wang, H.; Dhar, A.; Moness, M.; et al. Hyperactivated YAP1 Is Essential for Sustainable Progression of Renal Clear Cell Carcinoma. Oncogene 2025, 44, 2142–2157. [Google Scholar] [CrossRef]
  82. Denard, B.; Pavia-Jimenez, A.; Chen, W.; Williams, N.S.; Naina, H.; Collins, R.; Brugarolas, J.; Ye, J. Identification of CREB3L1 as a Biomarker Predicting Doxorubicin Treatment Outcome. PLoS ONE 2015, 10, e0129233. [Google Scholar] [CrossRef]
  83. Pollack, S.M.; He, Q.; Yearley, J.H.; Emerson, R.; Vignali, M.; Zhang, Y.; Redman, M.W.; Baker, K.K.; Cooper, S.; Donahue, B.; et al. T-cell Infiltration and Clonality Correlate with Programmed Cell Death Protein 1 and Programmed Death-ligand 1 Expression in Patients with Soft Tissue Sarcomas. Cancer 2017, 123, 3291–3304. [Google Scholar] [CrossRef]
  84. Wunder, J.S.; Lee, M.J.; Nam, J.; Lau, B.Y.; Dickson, B.C.; Pinnaduwage, D.; Bull, S.B.; Ferguson, P.C.; Seto, A.; Gokgoz, N.; et al. Osteosarcoma and Soft-Tissue Sarcomas with an Immune Infiltrate Express PD-L1: Relation to Clinical Outcome and Th1 Pathway Activation. Oncoimmunology 2020, 9, 1737385. [Google Scholar] [CrossRef] [PubMed]
  85. Budczies, J.; Mechtersheimer, G.; Denkert, C.; Klauschen, F.; Mughal, S.S.; Chudasama, P.; Bockmayr, M.; Jöhrens, K.; Endris, V.; Lier, A.; et al. PD-L1 (CD274) Copy Number Gain, Expression, and Immune Cell Infiltration as Candidate Predictors for Response to Immune Checkpoint Inhibitors in Soft-Tissue Sarcoma. Oncoimmunology 2017, 6, e1279777. [Google Scholar] [CrossRef] [PubMed]
  86. Munuswamy, N.; Sundar, M.; Krishnan, K.; Chandran, M.; R K, K. Undifferentiated Pleomorphic Sarcoma: A Case Report. Cureus 2024, 16, e73422. [Google Scholar] [CrossRef] [PubMed]
  87. Zając, A.E.; Czarnecka, A.M.; Rutkowski, P. The Role of Macrophages in Sarcoma Tumor Microenvironment and Treatment. Cancers 2023, 15, 5294. [Google Scholar] [CrossRef]
  88. Ishihara, S.; Iwasaki, T.; Kohashi, K.; Kawaguchi, K.; Toda, Y.; Fujiwara, T.; Setsu, N.; Endo, M.; Matsumoto, Y.; Nakashima, Y.; et al. Clinical Significance of Signal Regulatory Protein Alpha and T Cell Immunoreceptor with Immunoglobulin and Immunoreceptor Tyrosine-Based Inhibition Motif Domain Expression in Undifferentiated Pleomorphic Sarcoma. J. Cancer Res. Clin. Oncol. 2023, 149, 2425–2436. [Google Scholar] [CrossRef]
  89. Tang, F.; Tie, Y.; Wei, Y.-Q.; Tu, C.-Q.; Wei, X.-W. Targeted and Immuno-Based Therapies in Sarcoma: Mechanisms and Advances in Clinical Trials. Biochim. Biophys. Acta (BBA)-Rev. Cancer 2021, 1876, 188606. [Google Scholar] [CrossRef]
  90. Yuan, L.-L.; Chen, Z.; Qin, J.; Qin, C.-J.; Bian, J.; Dong, R.-F.; Yuan, T.-B.; Xu, Y.-T.; Kong, L.-Y.; Xia, Y.-Z. Single-Cell Sequencing Reveals the Landscape of the Tumor Microenvironment in a Skeletal Undifferentiated Pleomorphic Sarcoma Patient. Front. Immunol. 2022, 13, 1019870. [Google Scholar] [CrossRef]
  91. Cheung, L.S.; Chen, L.; Oke, T.F.; Schaffer, T.B.; Boudadi, K.; Ngo, J.T.; Gross, J.M.; Kemberling, H.; Diaz, L.A.; Lipson, E.; et al. Anti-PD-1 Elicits Regression of Undifferentiated Pleomorphic Sarcomas with UV-Mutation Signatures. J. Immunother. Cancer 2021, 9, e002345. [Google Scholar] [CrossRef]
  92. Li, Y.; Liu, Y.; Qu, Y.; Chen, X.; Qu, X.; Ye, Y.; Du, X.; Cheng, Y.; Xu, M.; Zhang, H. Case Report: Two Cases of Soft-Tissue Sarcomas: High TMB as a Potential Predictive Biomarker for Anlotinib Combined with Toripalimab Therapy. Front. Immunol. 2022, 13, 832593. [Google Scholar] [CrossRef] [PubMed]
  93. Italiano, A.; Bellera, C.; D’Angelo, S. PD1/PD-L1 Targeting in Advanced Soft-Tissue Sarcomas: A Pooled Analysis of Phase II Trials. J. Hematol. Oncol. 2020, 13, 55. [Google Scholar] [CrossRef] [PubMed]
  94. Burgess, M.A.; Bolejack, V.; Schuetze, S.; Van Tine, B.A.; Attia, S.; Riedel, R.F.; Hu, J.S.; Davis, L.E.; Okuno, S.H.; Priebat, D.A.; et al. Clinical Activity of Pembrolizumab (P) in Undifferentiated Pleomorphic Sarcoma (UPS) and Dedifferentiated/Pleomorphic Liposarcoma (LPS): Final Results of SARC028 Expansion Cohorts. J. Clin. Oncol. 2019, 37, 11015. [Google Scholar] [CrossRef]
  95. Available online: https://Clinicaltrials.Gov/Study/NCT03123276 (accessed on 11 August 2025).
  96. Available online: https://Clinicaltrials.Gov/Study/NCT02406781 (accessed on 11 August 2025).
  97. Available online: https://Clinicaltrials.Gov/Study/NCT03946943 (accessed on 11 August 2025).
  98. Available online: https://Clinicaltrials.Gov/Study/NCT04447274 (accessed on 11 August 2025).
  99. Available online: https://Clinicaltrials.Gov/Study/NCT04480502 (accessed on 11 August 2025).
  100. Available online: https://Clinicaltrials.Gov/Study/NCT03307616 (accessed on 11 August 2025).
  101. Available online: https://Clinicaltrials.Gov/Study/NCT02301039 (accessed on 11 August 2025).
  102. Available online: https://Clinicaltrials.Gov/Study/NCT03116529 (accessed on 11 August 2025).
  103. Available online: https://Clinicaltrials.Gov/Study/NCT03092323 (accessed on 11 August 2025).
  104. Guram, K.; Nunez, M.; Einck, J.; Mell, L.K.; Cohen, E.; Sanders, P.D.; Miyauchi, S.; Weihe, E.; Kurzrock, R.; Boles, S.; et al. Radiation Therapy Combined with Checkpoint Blockade Immunotherapy for Metastatic Undifferentiated Pleomorphic Sarcoma of the Maxillary Sinus with a Complete Response. Front. Oncol. 2018, 8, 435. [Google Scholar] [CrossRef]
  105. D’Angelo, S.P.; Richards, A.L.; Conley, A.P.; Woo, H.J.; Dickson, M.A.; Gounder, M.; Kelly, C.; Keohan, M.L.; Movva, S.; Thornton, K.; et al. Pilot Study of Bempegaldesleukin in Combination with Nivolumab in Patients with Metastatic Sarcoma. Nat. Commun. 2022, 13, 3477. [Google Scholar] [CrossRef]
  106. Available online: https://Clinicaltrials.Gov/Study/NCT03282344 (accessed on 11 August 2025).
  107. Gordon, E.M.; Angel, N.L.; Omelchenko, N.; Chua-Alcala, V.S.; Moradkhani, A.; Quon, D.; Wong, S. A Phase I/II Investigation of Safety and Efficacy of Nivolumab and Nab -Sirolimus in Patients with a Variety of Tumors with Genetic Mutations in the MTOR Pathway. Anticancer. Res. 2023, 43, 1993–2002. [Google Scholar] [CrossRef]
  108. Available online: https://Clinicaltrials.Gov/Study/NCT05103358 (accessed on 11 August 2025).
  109. Haddox, C.L.; Nathenson, M.J.; Mazzola, E.; Lin, J.-R.; Baginska, J.; Nau, A.; Weirather, J.L.; Choy, E.; Marino-Enriquez, A.; Morgan, J.A.; et al. Phase II Study of Eribulin plus Pembrolizumab in Metastatic Soft-Tissue Sarcomas: Clinical Outcomes and Biological Correlates. Clin. Cancer Res. 2024, 30, 1281–1292. [Google Scholar] [CrossRef]
  110. Available online: https://Clinicaltrials.Gov/Study/NCT03899805 (accessed on 11 August 2025).
  111. Pollack, S.M.; Redman, M.W.; Baker, K.K.; Wagner, M.J.; Schroeder, B.A.; Loggers, E.T.; Trieselmann, K.; Copeland, V.C.; Zhang, S.; Black, G.; et al. Assessment of Doxorubicin and Pembrolizumab in Patients with Advanced Anthracycline-Naive Sarcoma. JAMA Oncol. 2020, 6, 1778. [Google Scholar] [CrossRef]
  112. Ishihara, S.; Iwasaki, T.; Kohashi, K.; Yamada, Y.; Toda, Y.; Ito, Y.; Susuki, Y.; Kawaguchi, K.; Takamatsu, D.; Kawatoko, S.; et al. The Association between the Expression of PD-L1 and CMTM6 in Undifferentiated Pleomorphic Sarcoma. J. Cancer Res. Clin. Oncol. 2021, 147, 2003–2011. [Google Scholar] [CrossRef]
  113. Pan, H.; Liu, Y.; Fuller, A.M.; Williams, E.F.; Fraietta, J.A.; Eisinger, T.S.K. Collagen Modification Remodels the Sarcoma Tumor Microenvironment and Promotes Resistance to Immune Checkpoint Inhibition. bioRxiv 2024. [Google Scholar] [CrossRef]
  114. Lee, P.; Malik, D.; Perkons, N.; Huangyang, P.; Khare, S.; Rhoades, S.; Gong, Y.-Y.; Burrows, M.; Finan, J.M.; Nissim, I.; et al. Targeting Glutamine Metabolism Slows Soft Tissue Sarcoma Growth. Nat. Commun. 2020, 11, 498. [Google Scholar] [CrossRef] [PubMed]
  115. Verbeke, S.; Bourdon, A.; Lafon, M.; Chaire, V.; Frederic, B.; Naït Eldjoudi, A.; Derieppe, M.-A.; Giles, F.; Italiano, A. Dual Inhibition of BET and EP300 Has Antitumor Activity in Undifferentiated Pleomorphic Sarcomas and Synergizes with Ferroptosis Induction. Transl. Oncol. 2025, 52, 102236. [Google Scholar] [CrossRef] [PubMed]
  116. May, C.D.; Landers, S.M.; Bolshakov, S.; Ma, X.; Ingram, D.R.; Kivlin, C.M.; Watson, K.L.; Sannaa, G.A.A.; Bhalla, A.D.; Wang, W.-L.; et al. Co-Targeting PI3K, MTOR, and IGF1R with Small Molecule Inhibitors for Treating Undifferentiated Pleomorphic Sarcoma. Cancer Biol. Ther. 2017, 18, 816–826. [Google Scholar] [CrossRef] [PubMed]
  117. Lyko, F. The DNA Methyltransferase Family: A Versatile Toolkit for Epigenetic Regulation. Nat. Rev. Genet. 2018, 19, 81–92. [Google Scholar] [CrossRef]
  118. Saitoh, Y.; Bureta, C.; Sasaki, H.; Nagano, S.; Maeda, S.; Furukawa, T.; Taniguchi, N.; Setoguchi, T. The Histone Deacetylase Inhibitor LBH589 Inhibits Undifferentiated Pleomorphic Sarcoma Growth via Downregulation of FOS-like Antigen 1. Mol. Carcinog. 2019, 58, 234–246. [Google Scholar] [CrossRef]
  119. Diaz, L.A.; Coughlin, C.M.; Weil, S.C.; Fishel, J.; Gounder, M.M.; Lawrence, S.; Azad, N.; O’Shannessy, D.J.; Grasso, L.; Wustner, J.; et al. A First-in-Human Phase I Study of MORAb-004, a Monoclonal Antibody to Endosialin in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2015, 21, 1281–1288. [Google Scholar] [CrossRef]
  120. Tokumoto, H.; Setoguchi, T.; Saitoh, Y.; Sasaki, H.; Nagano, S.; Maeda, S.; Tanimoto, A.; Taniguchi, N. Neurotensin Receptor 1 Is a New Therapeutic Target for Human Undifferentiated Pleomorphic Sarcoma Growth. Mol. Carcinog. 2019, 58, 2230–2240. [Google Scholar] [CrossRef]
  121. Yu, K.; Wang, L.; Bu, F.; Zhang, J.; Hai, Y.; Hu, R.; Lu, J.; Shi, X. Retroperitoneal Undifferentiated Pleomorphic Sarcoma with Total Nephrectomy: A Case Report and Literature Review. Front. Surg. 2023, 10, 1166764. [Google Scholar] [CrossRef]
  122. Spalato-Ceruso, M.; Laroche-Clary, A.; Perret, R.; Valverde, Y.; Chaire, V.; Derieppe, M.-A.; Velasco, V.; Bourdon, A.; Italiano, A. Genome-Wide CRISPR/Cas9 Library Screening Identified ATM Signaling Network Genes as Critical Drivers for Resistance to ATR Inhibition in Soft-Tissue Sarcomas: Synthetic Lethality and Therapeutic Implications. Exp. Hematol. Oncol. 2023, 12, 51. [Google Scholar] [CrossRef]
  123. Toulmonde, M.; Lucchesi, C.; Verbeke, S.; Crombe, A.; Adam, J.; Geneste, D.; Chaire, V.; Laroche-Clary, A.; Perret, R.; Bertucci, F.; et al. High Throughput Profiling of Undifferentiated Pleomorphic Sarcomas Identifies Two Main Subgroups with Distinct Immune Profile, Clinical Outcome and Sensitivity to Targeted Therapies. EBioMedicine 2020, 62, 103131. [Google Scholar] [CrossRef]
  124. Frankiw, L.; Singh, A.; Peters, C.; Comin-Anduix, B.; Berent-Maoz, B.; Macabali, M.; Shammaie, K.; Quiros, C.; Kaplan-Lefko, P.; Baselga Carretero, I.; et al. Immunotherapy Resistance Driven by Loss of NY-ESO-1 Expression in Response to Transgenic Adoptive Cellular Therapy with PD-1 Blockade. J. Immunother. Cancer 2023, 11, e006930. [Google Scholar] [CrossRef]
  125. Conley, A.P.; Wang, W.-L.; Livingston, J.A.; Ravi, V.; Tsai, J.-W.; Ali, A.; Ingram, D.R.; Lowery, C.D.; Roland, C.L.; Somaiah, N.; et al. MAGE-A3 Is a Clinically Relevant Target in Undifferentiated Pleomorphic Sarcoma/Myxofibrosarcoma. Cancers 2019, 11, 677. [Google Scholar] [CrossRef]
Table 1. Clinical trials on UPS treatment.
Table 1. Clinical trials on UPS treatment.
Type of SarcomaTherapyTrial Registration Number
Unresectable or metastatic UPSAnlotinib Hydrochloride + ToripalimabNCT03946943
UPSEnvafolimab/Envafolimab + IpilimumabNCT04480502
UPSMecbotamab vedotin/Mecbotamab vedotin + NivolumabNCT03425279
Liposarcoma (LPS), leiomyosarcoma, UPSEribulin + PembrolizumabNCT03899805
Leiomyosarcoma, UPSPembrolizumab + GemcitabineNCT03123276
Recurrent or resectable UPS, dedifferentiated liposarcoma (DDLPS)Nivolumab/Nivolumab + Ipilimumab and radiation therapyNCT03307616
Advanced angiosarcoma, UPSPropranolol + PembrolizumabNCT05961761
UPS, alveolar soft part sarcoma (ASPS)Pembrolizumab + Melphalan + DactinomycinNCT04332874
Advanced UPSRecombinant anti-PD-1 humanized monoclonal antibody (609A)NCT05193214
Unresectable UPS and ASPSCarilizumab + ApatinibNCT04447274
Leiomyosarcoma, UPS, DDLPSCSF1R Inhibitor (DCC-3014) + AvelumabNCT04242238 
Ewing sarcoma, osteosarcoma, UPSPembrolizumab + CabozantinibNCT05182164
Metastatic or unresectable UPSdoxorubicin and pembrolizumabNCT06422806
Advanced STSMAGE-12 Peptide VaccineNCT00020267
Advanced or metastatic STSBrostallicin (PNU-166196A)/DoxorubicinNCT00410462
Leiomyosarcoma, synovial sarcoma (SS), osteosarcoma, malignant peripheral nerve sheath tumor (MPNST), neurofibrosarcoma, desmoplastic small round cell tumor fibrosarcoma, ASPS, UPS, hemangiopericytoma, chondrosarcoma, epithelioid sarcoma, malignant mesenchymoma Doxorubicin hydrochloride + TrabectedinNCT01189253
Leiomyosarcoma, LPS, SS, MPNST, neurofibrosarcoma, ASPS, UPS, hemangiopericytoma, epithelioid sarcoma malignant mesenchymoma Caelyx (pegylated liposomal doxorubin hydrochloride) + Ifosfamide NCT00030784
Leiomyosarcoma, LPS, SS, fibrosarcoma, UPS, hemangiopericytoma SoblidotinNCT00064220
Leiomyosarcoma, LPS, SS, osteosarcoma, Ewing sarcoma, MPNST, neurofibrosarcoma, UPS, chondrosarcoma Torisel + liposomal DoxorubicinNCT00949325
UPSXmAb®23104NCT03752398
Multiple STS subtypes including adult UPSGemcitabine + PazopanibNCT01532687
Non small cell lung cancer, head and neck squamous cell carcinoma, pancreatic adenocarcinoma, colorectal cancer, UPS, solitary fibrous tumors, DDLPSOKN4395/OKN4395 + PembrolizumabNCT06789172
High grade sarcoma, metastatic leiomyosarcoma, metastatic MPNST, metastatic SS, metastatic UPS, unresectable sarcoma, recurrent leiomyosarcoma, recurrent MPNST, recurrent SS, recurrent UPSSapanisertib (MLN0128 [TAK-228])NCT02601209
Metastatic UPS and other multiple STS subtypesNivolumab/Nivolumab + IpilimumabNCT02500797
Recurrent adult STS, recurrent leiomyosarcoma, recurrent LPS, recurrent MPNST, recurrent UPSMLN8237 (alisertib)NCT01653028
Osteosarcoma, Ewing sarcoma, MFH, synovial fibrosarcoma, leiomyosarcomaReolysis (oncolytic virus)NCT00503295
Advanced solid tumors, UPS, squamous cell carcinoma of the head and neck, carcinoma of the breastABBV-085NCT02565758
Multiple STS subtypes including adult UPSEpirubicin + Ifosfamide + NivolumabNCT03277924
Multiple STS subtypes including adult UPSBO-112/BO-112 + NivolumabNCT04420975
Multiple STS subtypes including adult UPSTalimogene laherparepvec (T-VEC) + RadiotherapyNCT02923778
Fibrosarcoma, leiomyosarcoma, LPS, myosarcoma, histiocytic sarcoma, SS, lymphangiosarcoma, MPNST, UPS, DDLPS, pleomorphic rhabdomyosarcoma, malignant triton tumorRadiotherapy + Sequential Doxorubicin and IfosfamideNCT03651375
UPS, SS, myxoid LPS and DDLPSSintilimab + Doxorubicin + IfosfamideNCT04356872
Extraskeletal myxoid chondrosarcoma, leiomyosarcoma, LPS, UPSIpilimumab + Nivolumab/Cabozantinib + Nivolumab + Ipilimumab NCT05836571
Metastatic DDLPS, metastatic leiomyosarcoma, metastatic SS, metastatic UPS, DDLPS, unresectable leiomyosarcoma, unresectable SS, unresectable UPSPeposertib + liposomal DoxorubicinNCT05711615
Leiomyosarcoma, MPNST, UPSGemcitabine + Docetaxel + PazopanibNCT01418001
Leiomyosarcoma, LPS, SS, angiosarcoma, UPS, epithelioid sarcoma, MPNST, fibrosarcoma, pleomorphic rhabdomyosarcoma, endometrial stromal sarcoma, desmoplastic small round cell tumorDoxorubicin + IfosfamideNCT06277154
Metastatic leiomyosarcoma, metastatic SS, metastatic UPS, advanced myxoid LPS, advanced STS, metastatic myxoid LPS, metastatic round cell LPS, metastatic STS, refractory leiomyosarcoma, refractory myxoid LPS, refractory round cell LPS, refractory STS, refractory SS, refractory UPS advanced leiomyosarcoma, advanced SS, advanced UPS, metastatic chondrosarcomaItacitinib NCT03670069
LPS, leiomyosarcoma, UPSSunitinibNCT00400569
UPS, leiomyosarcoma, LPS, SS, angiosarcomaOlaratumab (Lartruvo) + DoxorubicinNCT02451943
NCT02584309
Multiple STS subtypes including adult UPSRibociclib + DoxorubicinNCT03009201
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lesovaya, E.A.; Fetisov, T.I.; Bokhyan, B.Y.; Senchenko, M.A.; Rogozhin, D.V.; Maksimova, V.P.; Demko, A.N.; Belitsky, G.A.; Yakubovskaya, M.G.; Kirsanov, K.I. Genetic Heterogeneity of Undifferentiated Pleomorphic Sarcoma: Is There Potential for Targeted Therapy? Cancers 2025, 17, 3613. https://doi.org/10.3390/cancers17223613

AMA Style

Lesovaya EA, Fetisov TI, Bokhyan BY, Senchenko MA, Rogozhin DV, Maksimova VP, Demko AN, Belitsky GA, Yakubovskaya MG, Kirsanov KI. Genetic Heterogeneity of Undifferentiated Pleomorphic Sarcoma: Is There Potential for Targeted Therapy? Cancers. 2025; 17(22):3613. https://doi.org/10.3390/cancers17223613

Chicago/Turabian Style

Lesovaya, Ekaterina A., Timur I. Fetisov, Beniamin Yu. Bokhyan, Maria A. Senchenko, Dmitry V. Rogozhin, Varvara P. Maksimova, Anna N. Demko, Gennady A. Belitsky, Marianna G. Yakubovskaya, and Kirill I. Kirsanov. 2025. "Genetic Heterogeneity of Undifferentiated Pleomorphic Sarcoma: Is There Potential for Targeted Therapy?" Cancers 17, no. 22: 3613. https://doi.org/10.3390/cancers17223613

APA Style

Lesovaya, E. A., Fetisov, T. I., Bokhyan, B. Y., Senchenko, M. A., Rogozhin, D. V., Maksimova, V. P., Demko, A. N., Belitsky, G. A., Yakubovskaya, M. G., & Kirsanov, K. I. (2025). Genetic Heterogeneity of Undifferentiated Pleomorphic Sarcoma: Is There Potential for Targeted Therapy? Cancers, 17(22), 3613. https://doi.org/10.3390/cancers17223613

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

Article Metrics

Back to TopTop