Next Article in Journal
Screening for Prognostic microRNAs Associated with Treatment Failure in Diffuse Large B Cell Lymphoma
Next Article in Special Issue
Spinal Solitary Fibrous Tumors: An Original Multicenter Series and Systematic Review of Presentation, Management, and Prognosis
Previous Article in Journal
Current Surgical Management Strategies for Colorectal Cancer Liver Metastases
Previous Article in Special Issue
Clinical, Histological, and Molecular Features of Solitary Fibrous Tumor of Bone: A Single Institution Retrospective Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Therapeutic Options for Solitary Fibrous Tumor: Antiangiogenic Therapy and Beyond

1
Department of Medical Oncology, Centre Léon Bérard, 28 Rue Laennec, 69008 Lyon, France
2
Department of Pathology, Centre Jean Perrin, 58 Rue Montalembert, 63011 Clermont-Ferrand, France
3
Faculty of Medicine, Université Claude Bernard Lyon 1, 69003 Lyon, France
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(4), 1064; https://doi.org/10.3390/cancers14041064
Submission received: 30 December 2021 / Revised: 15 February 2022 / Accepted: 16 February 2022 / Published: 20 February 2022
(This article belongs to the Special Issue Solitary Fibrous Tumor)

Abstract

:

Simple Summary

In the latest WHO classification, solitary fibrous tumors (SFTs) are now subdivided into benign SFT (intermediate category (locally aggressive)), SFT NOS (intermediate category (rarely metastasizing)), and malignant SFT. Thanks to recent progress in molecular characterization, the identification of the NAB2–STAT6 fusion oncogene has emerged as a specific cytogenetic hallmark for SFT. Despite these recent advances in classification and understanding of the molecular pathophysiology of SFT, there are no consensus clinical guidelines regarding systemic treatment. Several new therapeutic options are of interest in this subtype of sarcoma considered as refractory to classical chemotherapy. In case of advanced disease, antiangiogenic therapy might be viewed as the best therapeutic option.

Abstract

SFT is an ultrarare mesenchymal ubiquitous tumor, with an incidence rate <1 case/million people/year. The fifth WHO classification published in April 2020 subdivided SFT into three categories: benign (locally aggressive), NOS (rarely metastasizing), and malignant. Recurrence can occur in up to 10–40% of localized SFTs, and several risk stratification models have been proposed to predict the individual risk of metastatic relapse. The Demicco model is the most widely used and is based on age at presentation, tumor size, and mitotic count. Total en bloc resection is the standard treatment of patients with a localized SFT; in case of advanced disease, the clinical efficacy of conventional chemotherapy remains poor. In this review, we discuss new insights into the biology and the treatment of patients with SFT. NAB2–STAT6 oncogenic fusion, which is the pathognomonic hallmark of SFT, is supposedly involved in the overexpression of vascular endothelial growth factor (VEGF). These specific biological features encouraged the successful assessment of antiangiogenic drugs. Overall, antiangiogenic therapies showed a significant activity toward SFT in the advanced/metastatic setting. Nevertheless, these promising results warrant additional investigation to be validated, including randomized phase III trials and biological translational analysis, to understand and predict mechanisms of efficacy and resistance. While the therapeutic potential of immunotherapy remains elusive, the use of antiangiogenics as first-line treatment should be considered.

1. Introduction

The fifth edition of the World Health Organization (WHO) classification of tumors of soft tissue and bone was published in April 2020, with more than 150 histological subtypes. The classification has been updated with the identification of new distinct molecular subtypes, thanks to the easier access to molecular tools, and these improvements have led to a better understanding of the tumorigenesis of many sarcomas. The classification of conjunctive tumors is crucial, not only for diagnosis and prognostication but also for correct management of patients.
In the fibroblastic and myofibroblastic tumor category, the recognized entity of solitary fibrous tumor (SFT) is described. SFT is a ubiquitous fibroblastic mesenchymal neoplasm originally reported by Klemperer et al. in 1931 [1], with locally invasive properties and a predilection for body cavity sites and serosal membranes; it has only recently been reported in the pleura and/or lung. SFT is known as the “great simulator” of soft-tissue neoplasm due to its many differential diagnoses [2]. The diagnosis of SFT is established by the conjunction of clinical, pathological, immunohistochemical, and molecular features. Identification of the NAB2 (NGFI-A-binding protein 2)–STAT6 (signal transduction and activator of transcription 6) fusion oncogene has emerged as a specific cytogenetic hallmark for SFT. Several risk stratification systems have been proposed for SFT to predict which tumors may harbor an aggressive behavior [2,3,4,5]. In the latest WHO classification, SFTs are now subdivided into benign SFT (intermediate category (locally aggressive)), SFT NOS (intermediate category (rarely metastasizing)), and malignant SFT [6].
In this review, we discuss recent progresses in the molecular characterization and therapeutics of SFT and future prospects of tailored therapeutic approaches.

2. Description of SFT

2.1. Epidemiology

SFT is an ultrarare mesenchymal tumor that represents 3.7% of all soft-tissue sarcomas (STSs) and mesenchymal tumors [7] with a reported incidence rate <1 case/million people/year [8,9]. SFT usually affects adult patients with a peak incidence in the fifth and sixth decades. Pleural SFTs are observed in older patients (median age at diagnosis 56 to 60) compared with intra-abdominal or meningeal SFTs that occur in younger patients (fourth decade) [10].

2.2. Clinical and Radiological Presentation

The thorax (pleura, mediastinum, lung parenchyma) is the most common site of initial presentation (30%), but SFT can occur at any extra-thoracic anatomical site [11]. The most frequent extra-pleural location is represented by the head and neck, including meninges (27%), followed by the intra-abdominal region (peritoneum, retroperitoneum, and pelvis) (20%), trunk (10%), and extremities (8%) [12]. SFTs localized in the retroperitoneum, peritoneum, or mediastinum tend to have a more aggressive course compared to other anatomical sites [5,13,14]. SFTs are often slow-growing tumors, with delayed symptom onset. Patients may present with nonspecific pulmonary symptoms such as dyspnea or cough, but often remain asymptomatic despite important tumor volume. Compression of adjacent anatomical structures can be observed for voluminous masses but remains unusual as the median tumor size at diagnosis ranges between 50 and 80 mm [15]. Additionally, few patients (<10%) develop paraneoplastic syndromes which can guide the diagnosis. Hypertrophic osteoarthropathy (Pierre Marie–Bamberger syndrome) is a nonspecific condition rarely associated with pleural SFT. Classical symptoms include distal digital clubbing, periostitis, and synovial effusions, supposedly linked to the paraneoplastic overexpression of vascular endothelial growth factor (VEGF). Fewer than 5% of SFT patients can also present with a refractory hypoglycemic syndrome due to the overproduction of insulin-like growth factor-2 (IGF-2) by large peritoneal or pleural SFTs, called Doege–Potter syndrome [16,17,18].
Consequently, SFTs are frequently diagnosed on incidental radiological findings [19]. Time to diagnosis is classically longer in pleural SFTs compared to extra-pleural SFTs. The radiographic features of SFTs are nonspecific, and computed tomography (CT) usually shows a well-circumscribed isodense mass to skeletal muscle with contrast enhancement in highly vascularized tumors (65%) [15]. SFTs are often characterized by the presence of low-signal-intensity foci on T1- and T2-weighted magnetic resonance imaging (MRI), corresponding to the collagen content. 18F-FDG PET/CT may have a limited role in diagnosing SFT in suspected patients, although the presence of multiple high-grade lesions associated with 18F-FDG PET hypermetabolism should raise suspicion of a malignant SFT.

2.3. Biopathology

2.3.1. Classification

SFTs belong to the group of fibroblastic and myofibroblastic tumors in the WHO classification [6]. The fifth edition published in April 2020 subdivided SFTs into three categories: benign (locally aggressive), NOS (rarely metastasizing), and malignant [20]. Meningeal SFT, previously known as hemangiopericytoma (HPC), is a rare form of extra-pleural SFT that derives from smooth muscle pericytes surrounding the intraparenchymal microvasculature [21]. Historically, SFTs were considered low-grade tumors contrary to HPCs that displayed aggressive patterns. Despite an apparent distinct clinical behavior, SFTs and HPCs were merged under one disease umbrella in the fourth WHO classification [22]. The fourth revised edition of the WHO Classification of Tumors of CNS published in 2016 introduced the hybrid “SFT/HPC” class, subdivided into three histological grades depending on mitotic count (grade 3 defined by at least five mitoses/10 HPFs) [23]. Most recently, the term of “hemangiopericytoma” was removed from the 2021 WHO Classification of Tumors of CNS to conform with soft-tissue pathology nomenclature [24]. In other words, updated classifications now consider SFTs and HPCs as the same entity at two opposite ends of the same histologic spectrum rather than the strict “benign or malignant” dichotomy that was used for decades (Figure 1).

2.3.2. Prognosis and Risk Stratification

Localized SFTs offer good prognosis after complete surgery. However, recurrence can occur in up to 10–25% of SFTs by 10 years [5,25,26,27]. The risk of metastatic recurrence at 5 years can even rise up to 40% in high-risk patients [13,28,29,30,31,32]. Preferential metastatic sites include lungs, liver, and bone.
SFT recurrence is more frequent in the case of incomplete resection (R1/R2) [25,28], tumor seeding within serosal membranes (pleura, peritoneum), or meningeal or distant hematogenous spread. Small retrospective series tend to show that patients with extra-pleural SFTs have a higher risk of local and metastatic recurrence compared to pleural SFTs [33,34,35]. Meningeal SFTs have a dismal prognosis with frequent local recurrence due to meningeal seeding and early bone metastatic recurrence. Notably, late relapse beyond 10 years and up to 20 years after initial presentation is common, which justifies a long-term follow-up [13,28,29,30,31,32].
Importantly, the development of multivariate risk stratification models has resulted in improved prognostication, such that the traditional benign/malignant distinction is now avoided. Among those models that integrate several clinicopathologic variables to predict the individual risk of metastatic recurrence, the Demicco model (“D-score” or “MDACC score”) is the most widely used in clinical practice and is applicable to SFTs of all extra-meningeal sites [36]. It is based on age at presentation, tumor size, and mitotic count to classify SFTs with a low, moderate, or high risk of developing a metastatic recurrence [37] (Table 1). Another version also includes tumor necrosis. Importantly, dedifferentiation, which corresponds to the abrupt transition from a conventional low-grade SFT component into a high-grade sarcoma and occurs in <1% of primary or recurrent SFTs, remains unpredictable by this model [38].
Ghanim et al. used blood-derived biomarkers to predict event-free-survival (EFS) in intrathoracic SFTs [39]. Elevated preoperative fibrinogen was an independent prognostic marker of poor outcome after curative surgery and associated with malignant SFT. This finding suggests a potential role of innate immune system overactivation in SFT prognosis.
The presence of TP53 and/or TERT mutations seems to be correlated with malignant SFTs associated with an aggressive biological behavior.

2.3.3. Tumorigenesis, Pathology, and Molecular Alterations

The macroscopic appearance of SFT is a well-defined, lobulated, firm mass surrounded by a serosal pseudo-capsule. The wide histological spectrum of SFT ranges from morphologically paucicellular to highly cellular tumors. SFTs are composed of atypical spindle cells with fusiform nuclei arranged haphazardly with a suggestive “patternless pattern”, surrounded by a dense stromal collagen with thin collagen bands (Figure 2A) and admixed with a characteristic branching staghorn (hemangiopericytoma-like) vasculature. Hypocellular phenotypes have a low mitotic count, and nuclear pleomorphism or necrosis is classically absent. These histopathologic patterns are not specific to SFTs and can also be observed in other mesenchymal tumors [40,41].
In practice, the detection of the NAB2–STAT6 fusion gene (detailed below) with cytogenetic methods such as fluorescent in situ hybridization (FISH) is impossible due to the small size of the inverted sequence and the proximity of the NAB2 and STAT6 loci. For routine diagnosis, STAT6 is a robust immunohistochemical surrogate marker of all NAB2–STAT6 fusion transcripts. A strong nuclear expression of the C-terminal part of STAT6 has good diagnostic performance with excellent sensitivity (98%) and specificity (>85%) [42,43,44,45,46] (Figure 2B). PCR-based detection of STAT6 is less sensitive for diagnosis due to the diversity of possible breakpoints in fusion transcripts. However, STAT6 is inconsistently expressed and might be absent in some SFT cases. STAT6 is rarely overexpressed in the nucleus and cytoplasm of well-differentiated (WD-LS) and dedifferentiated liposarcoma (DD-LPS) cells [47]. If that differential diagnosis is discussed, a complementary analysis of MDM2 and CDK4 expression in IHC can be useful as they are overexpressed in WD-LS/DD-LPS but not in SFT. Other standard IHC markers can be used in combination (CD34, Bcl2, CD99, vimentin, desmin, S100 protein, epithelial markers) with good sensitivity but a low specificity. Lastly, GRIA2 and ALDH1 are under investigation [48,49].
The WHO classification updates were justified by the discovery of a shared cytogenetic signature across all anatomical sites in 2013 by three different research groups: the NAB2–STAT6 fusion oncogene [41,50,51,52]. The NAB2–STAT6 fusion gene is detected in nearly all SFT cases, which suggests a driver role in tumor development. NAB2 and STAT6 are adjacent genes transcribed in opposite directions at the locus 12q13 [41,51,52].
NAB2 is a transcriptional repressor that binds to the inhibitory domain of EGR-1 (early growth response 1) through its N-terminal binding domain (NAB2 conserved domain 1—NCD1) to have transcriptional control over EGR-1 target genes, including IGF-2 or FGFR1. EGR-1 is a downstream gene of the MAPK/ERK pathway [53]. EGR-1 enhances the transcription of cell-cycle regulatory proteins such as cyclin D1 that promotes tumor cell proliferation, which in turns activates the MAPK/ERK signaling pathway and auto-activates the expression of EGR-1 through a positive feedback loop. Additionally, the transcription of EGR-1 is upregulated by various growth factors such as IGF-1 and its receptor IGFR-1. EGR-1 also plays a role in the systemic dissemination of tumor cells. Indeed, epithelial–mesenchymal transition (EMT) was reported to be triggered by the EGR-1-induced upregulation of Slug and Snail via the ERK1/2 and PI3K/Akt pathways in ovarian cancer cells [54]. Proangiogenic growth factors such as basic fibroblast growth factor (bFGF) and VEGF-A are EGR-1 target genes that contribute to tumor angiogenesis. EGR-1 can directly activate the transcription of these factors [55] or be stimulated via the ERK1/2 pathway after hypoxia-inducible factor (HIF)-1α overexpression in response to hypoxia in the tumoral microenvironment.
STAT6 (signal transducer and activator of transcription 6) is a transcription factor usually involved in allergic and immune signaling pathways with a role in tumorigenesis [56,57]. The Src homology 2 domain (SH2) is crucial to bind to IL-4R or IL-13R and trigger STAT6 phosphorylation by Janus (JAK) and TyK2 kinases [58]. In its dimerized form, STAT6 is activated, whereby it can enter the nucleus and bind to DNA promoters through its DNA-binding domain (DBD1).
In SFT, the recurrent intrachromosomal inversion in the long arm of chromosome 12 leads to the replacement of at least one of the three repressor domains of NAB2 (NCD1, NCD2, CID) by the transactivation domain (TAD) of STAT6. NAB2 and STAT6 are fused in a common direction of transcription, which results in the transcription of a chimeric NAB2–STAT6 fusion protein from the NAB2 promoter [50,52]. Then, the fusion transcript translocates to the nucleus and constitutively activates EGR-1-responsive genes [41] (Figure 3). Intriguingly, NAB2 and EGR-1 are mutual targets to each other, which creates a positive feedback loop and strengthens the abnormal accumulation of the NAB2–STAT6 fusion transcript in the nucleus of SFT cells. This specificity is a hallmark of SFT used for differential diagnosis with other tumors.
Barthelmess and colleagues first suggested that the NAB2 exon4–STAT6 exon2 (N4S2) and the NAB2 exon6–STAT6 exon16/17 (N6S16/17) fusion variants might be associated with distinct clinical features. N4S2 was mostly found in older patients with less aggressive SFTs and deep extra-thoracic lesions. On the other hand, N6S16/17 was more frequent in younger patients with aggressive phenotypes and clinical behavior usually found in meningeal SFTs [50]. Since then, other studies failed to demonstrate a clear impact of fusion variants on prognosis, probably due to a short follow-up that did not take into account late recurrences [59,60,61,62,63]. In a recent retrospective cohort with long-term follow-up, Georgiesh et al. investigated the clinicopathological and prognostic impact of the STAT6-Full (intact STAT6 domains) and STAT6-TAD (contains only the STAT6 TAD domain) variants by RNA sequencing [64]. A total of 39 patients with localized extra-meningeal SFTs were enrolled. Patients with STAT6-TAD tumors had a worse prognosis, with a higher mitotic count and a 10 year recurrence-free survival rate of 25% (vs. 78% for STAT6-Full patients). These promising results need further confirmation in prospective trials to conclude on their prognostic value.

3. Therapeutic Options for SFT

Patients with SFTs should be managed within sarcoma reference centers, by a dedicated multidisciplinary team with a pathologist, radiologist, surgical oncologist, radiation oncologist, and medical oncologist who are familiar with the nuances of this disease [65,66,67,68]. Each case has to be discussed in a specialized multidisciplinary tumor board (MTB) to determine the best individualized therapeutic strategy.
For response assessment, the use of both Response Evaluation Criteria in Solid Tumors (RECIST) and the new Choi criteria (i.e., a 10% decrease in tumor size or a more than 15% decrease in tumor density) might be of interest to evaluate therapeutic response to antiangiogenic agents [69,70,71]. Choi criteria were originally developed to predict the response of advanced gastrointestinal stromal tumor (GIST) to imatinib. Due to the hypervascularized nature of SFT, recent studies used both Choi and RECIST criteria to assess radiological response. The retrospective studies detailed below reported few responses with RECIST criteria but 46% to 79% response with Choi criteria [72,73,74,75,76]. However, the predictive value of Choi criteria in SFTs should be interpreted cautiously. There are no data on the use of Choi criteria in patients with advanced SFTs treated with chemotherapy.

3.1. Localized Disease

3.1.1. Surgery

Complete en bloc surgical resection with negative margins (R0) is the gold-standard treatment for localized disease [77]. The 10 year overall survival (OS) in patients with SFTs resected with negative margins ranges from 54% to 89% [78,79,80]. In pleural SFTs, the modalities of resection include wedge resection, lobectomy, or pneumonectomy, associated with chest wall or diaphragm resection if necessary [77,81,82]. In their retrospective series, Lahon et al. resected localized pleural SFTs in 157 patients. Despite R0 margins, 15 patients (10%) recurred with a median time to recurrence of 29 months (10 local and five metastatic recurrences) [81]. Re-resection of local recurrence could achieve local control in 9/10 patients. The 5 year and 10 year OS rates were 86% and 77%, respectively. Similarly, Lococo et al. reported disease recurrence in 15/50 patients (six local and nine metastatic recurrences) with localized pleural SFTs resected with negative margins, yielding 5 year and 10 year OS rates of 81% and 67%, respectively [83]. Complete surgical resection is also the primary approach in extra-pleural tumors, with procedures paralleling other sarcoma surgeries from similar anatomical sites.

3.1.2. Radiation Therapy

Several strategies can be discussed in the postoperative setting, depending on surgical margins and risk stratification models. Despite limited available prospective data, multiple observational studies report improved local control in high-risk SFT patients treated with adjuvant radiotherapy (RT). Nevertheless, there is no clear demonstration of an OS benefit of adjuvant RT [13,14,26,33,34,84,85,86,87,88]. In 2020, Haas et al. investigated the role of perioperative RT in localized extra-meningeal SFT [14]. In this cohort, 428/549 patients (78%) received surgery alone and 121 (22%) were treated with surgery and RT. Overall, 48% of patients (n = 58) received surgery followed by adjuvant RT, with a lower risk of local progression (p = 0.012), and a 96% local control rate at 5 years. However, this association did not translate into an OS benefit (p = 0.325). Preoperative RT was assessed in the STRASS phase III randomized trial, which included intra-abdominal sarcomas of different histotypes, but SFT cases were underrepresented, leading to inconclusive results in that population [88,89]. An international retrospective observational study performed across seven specialized sarcoma centers aimed to better define the benefit of definitive RT in this disease [90]. Forty patients with locally advanced or metastatic SFT (nine pleural, 16 soft-tissue, 10 meningeal, three head and neck, and two other SFTs) were treated with definitive RT, receiving approximately 60 Gy, with an objective response rate of 67%. At 5 years, the local control (LC) rate was 81.3%, and the OS rate was 87.5%. In case of palliative RT (39 Gy in conventional fractionation), the overall response rate (ORR) was 38%, and the 5 year LC and OS rates were 62.5% and 54.2% respectively. Therefore, radiation doses can range from 39 Gy (in conventional fractions) for palliative care to 60 Gy to achieve durable control.
In summary, observation is recommended in patients with negative margins (R0) without high-risk histologic features. In the case of intermediate- to high-risk SFT with positive margins (R1/R2), re-resection should be discussed for fit patients if complete resection can be achieved with minimal morbidity. If the patient is unfit for further resection or R0 surgery cannot be technically achieved (due to anatomical site), then adjuvant RT is a reasonable option. Surgeons may adopt a conservative attitude in selected cases to preserve organ functions when postoperative RT can be delivered, given the favorable long-term outcome. Adjuvant RT should systematically be considered for high-risk SFTs, such as malignant SFTs of the central nervous system (CNS), considering the very high risk of local recurrence. Neoadjuvant RT can be an option in selected cases to improve tumor resectability or when wound complications are predicted to be manageable.

3.1.3. (Neo)Adjuvant Chemotherapy

In patients with localized, resectable SFTs, there is no evidence supporting the use of systemic therapies in the (neo)adjuvant setting [18]. A review of the literature shows only limited data on this subject and consists mostly of a few case reports [91,92,93]. Even if the relevance of adjuvant chemotherapy (CT) in resected SFTs is still unknown due to a lack of data, it is important to note that SFTs marginally benefit from traditional sarcoma CT with low response rates (Table 2). Importantly, adjuvant CT should never be intended to rescue inadequate surgery. Nevertheless, for some patients with high-risk SFTs and/or large malignant tumors, the use of neoadjuvant CT should be discussed at a specialized MTB. Importantly, eligible patients should be managed within clinical studies. Therefore, in the case of locally advanced tumors, if R0 surgery is not feasible apart from mutilating surgery, neoadjuvant CT is an option. On the basis of the response rate data (Table 2), an anthracycline-based regimen (plus ifosfamide or dacarbazine) can be considered as the regimen of choice. Importantly, early tumor response assessment is required to avoid delaying surgery in the case of nonresponding disease.

3.2. Advanced and Metastatic Disease

3.2.1. Surgery, Ablations, or RT

Metachronous (disease-free interval >1 year), resectable lung metastases without extrapulmonary disease may be managed with surgery as standard treatment applied to sarcoma, if complete excision of all lesions is feasible. Surgery, ablations, or RT of extrapulmonary metastases may also be an option in highly selected cases [65,66,67].

3.2.2. Chemotherapy

In cases of synchronous and/or unresectable lung metastases and in cases of extrapulmonary metastatic disease, patients are candidates for systemic treatment, even though a standard medical approach is currently not established. Published data on the response of SFTs to conventional chemotherapy are limited, and results gathering small case series, retrospective studies, and predictive preclinical models show conflicting results. Table 2 summarizes the data available on the efficacy of systemic agents in SFT.
For anthracycline-based therapies, ORR ranges between 0% and 20%, and stable disease (SD) is achieved in 26–65% of cases with a median PFS (mPFS) and median OS (mOS) of 4–5.2 months and 11.5–14.6 months, respectively [72,74,94,95,99]. Ifosfamide monotherapy as first-line treatment was also assessed with a 10% ORR and 26% SD. Dacarbazine monotherapy showed a 37.5% ORR and an mPFS of 7 months [96]. Despite a 7.9–9.1% ORR, trabectedin is an important alternative in subsequent lines, as disease stabilization can be achieved in 42–73% of cases with an mPFS of 3.7–11.6 months and an mOS of 14.3–22.3 months [97,98,110]. Consequently, the use of trabectedin is often associated with a meaningful clinical benefit in patients with advanced SFTs. Subsequent lines of chemotherapy consisted of cytotoxic agents (in monotherapy or in combination) used routinely for STS treatment, but showed no objective response.
Common and frequent chemotherapy-induced adverse events such as high-grade mucositis, nausea and vomiting, or hematologic toxicities were reported with all the regimens. Additionally, patients could experience congestive heart failure (associated or not with a reduction in cardiac ejection fraction) with anthracycline, ifosfamide-induced encephalopathy or hepatotoxicity, and rhabdomyolysis with trabectedin. The toxicity profiles were similar to those classically observed in STS.
Overall, the consistent response rates observed across retrospective series tend to confirm the limited efficacy of conventional chemotherapy in advanced SFT. The STRADA randomized phase II study (NCT03023124) will prospectively compare trabectedin (1.3–1.5 mg/m2) to doxorubicin (75 mg/m2) plus dacarbazine (400 mg/m2/day, days 1, 2) in 50 patients with advanced SFTs. The Italian phase II single-arm ERASING trial (NCT03840772) will also evaluate the activity of eribulin in that population. Interestingly, the Choi response rate is one of the secondary outcomes of the ERASING and STRADA trials, which will provide data in these populations.
In summary, even if there is no formal gold standard, patients with metastatic SFTs are treated routinely in the same way as other STSs, using anthracycline as first-line therapy, while ifosfamide, dacarbazine, and trabectedin are options for second line and beyond. Best supportive care alone is an alternative for unfit patients, regarding the toxicities of those drugs. Otherwise, there is no demonstration that multiagent CT improves patient survival, and single-agent chemotherapy remains the standard.

3.2.3. Antiangiogenic Therapies and Other Targeted Therapies

SFTs are highly vascularized tumors with high expression rates of proteins involved in angiogenic pathways such as platelet-derived growth factor receptor (PDGFR) and vascular endothelial growth factor receptor (VEGFR) [111]. Angiogenesis is an important feature involved in tumor growth and metastatic diffusion of SFT. Accordingly, the inhibition of angiogenesis pathways was suspected to be a key therapeutic target to inhibit tumor cell proliferation. Considering the angiogenic properties of SFTs, several case reports and clinical trials investigated various inhibitors of angiogenesis with a promising activity in SFTs.
Prolonged disease control up to 30 months has been reported with antiangiogenic agents (sunitinib, sorafenib, pazopanib, temozolomide–bevacizumab) in case reports, retrospective studies, and phase I and phase II trials [76,95,101,102,103,104,105,106]. ORR varied from 0% to 79% with an mPFS ranging between 4.7 months and 9.7 months using Choi criteria. Antiangiogenic agents were positioned in subsequent lines after chemotherapy resistance.
A prospective single-arm phase II trial confirmed the efficacy of pazopanib in two cohorts of typical (n = 34) and malignant/dedifferentiated (DD) SFTs (n = 36) with 58% and 51% partial response (PR), respectively, according to Choi criteria. [108,109]. Interestingly, pazopanib was associated with a very high clinical benefit rate in the typical SFT cohort (97%). The outcomes of PFS were also more favorable in the typical SFT cohort than in the malignant/DD SFT cohort, with an mPFS of 9.8 month using Choi criteria and 12.1 months based on RECIST criteria in the typical SFT cohort, versus 5.6 months for the malignant/DD SFT group. Notably, the inclusion of patients with DD-SFT was stopped due to cases of hyper-progression under pazopanib in a planned interim analysis. The toxicity profiles were similar between the two cohorts and consistent with those reported in previous clinical trials.
Importantly, pazopanib harbors a multifaceted cardiovascular toxicity profile including cardiomyopathy, QTc-interval prolongation, and hypertension. Therefore, a cardiologic workup should always be proposed in patients with a preexisting cardiovascular condition. Even in the absence of comorbidity, it should be considered in patients previously treated with anthracycline-based CT.
In summary, according to the aforementioned results, pazopanib can be a treatment option in the first-line setting for typical SFT.

3.2.4. IGF-1 Inhibitors

IGF-1 is overexpressed in SFT, and treatment regimens using figitumumab, a fully human IgG2 anti-IGF-1 (IGF-1R) monoclonal antibody, demonstrated tumor responses in a few patients with advanced SFTs [106,112]. In a phase I trial assessing the combination of figitumumab and the mTOR inhibitor everolimus, the only PR (among 18 evaluable patients) was observed in a patient with SFT [112]. Unfortunately, Pfizer ceased the development of the drug in 2011 and has stopped its manufacture.

3.2.5. Immunotherapy

Immunotherapy is another promising approach for SFT. Available data on the SFT immune microenvironment mainly come from retrospective studies.
Tazzari et al. first reported an immunosuppressed environment at the tumor site marked by the absence of a granulocytic MDSC (gMDSC) infiltrate [113]. Samples treated with sunitinib malate were enriched in activated CD8+ and CD4+ cells, which suggests that antiangiogenic therapies might modulate the T-cell immune infiltrate in SFTs. A translational study on 16 intracranial SFT/HPC specimens (13 HPCs, three SFTs) evaluated the correlation of PD-1, PD-L1, and tumor-infiltrating lymphocyte (TIL) expression with prognosis [114]. PD-L1 was expressed in all tumors. As an individual biomarker, a diffuse or intense PD-L1 staining was associated with a shorter time to treatment failure (TTF). Diffuse PD-L1 staining coupled with the absence of CD8 expression was significantly associated with a shorter TTF (p = 0.005). Consequently, the conjunction of diffuse PD-L1 IHC staining and the absence of CD8 expression may predict the early occurrence of extracranial metastases in SFTs [114]. Dancsok et al. systematically evaluated the expression of immune checkpoint biomarkers and TILs in a variety of sarcoma subtypes [115]. Among the 16 SFT cases included in the study, PD-1 and PD-L1 were infrequently expressed with sparse TILs. On the basis of the results in the entire cohort, the authors suggested that T-cell immune infiltrate might be less frequent in translocation-associated sarcomas, such as SFTs. More recently, Berghoff et al. investigated the inflammatory tumor microenvironment in 74 specimens of meningeal tumors including 12 cases (16.2%) of HPC and seven cases (9.5%) of meningeal SFT [116]. TILs were present in all SFT cases and 11/12 cases (91.7%) of HPC. PD-L1 was only expressed in 1/12 (8.3%) cases of HPC.
Several cases of partial response to anti-PD1 or -PDL1 therapies have been reported in the recent literature. Boothe et al. published the case of a patient with advanced malignant pleural SFT who experienced a long-lasting near-complete response after 31 cycles of pembrolizumab. PD-L1 was positive in IHC (5%), and a mutation in exon 9 of MLH1 (E234Q) was detected after next-generation sequencing (NGS) [117]. Lastly, in a phase II trial of pembrolizumab in sarcomas, one case of SFT was the only PR reported in the study [118]. Given the suspected role of the immune system in these neoplasms, a phase III trial is currently ongoing to compare nivolumab + ipililumab to pazopanib in adults with advanced rare STSs including SFTs (NCT04741438).

4. Conclusions

Sarcomas represent a highly heterogeneous group of tumors, both in clinical and in genomic settings; thus, they should be treated separately. Furthermore, recent clinical trials exploring unselected sarcoma histotype populations [119] failed to improve patient outcome. Therefore, each subtype should still be treated separately.
SFTs are poorly sensitive to conventional chemotherapy. Nevertheless, the pathognomonic NAB2–STAT6 oncogenic fusion that induces IGF-1 overexpression and angiogenesis in the tumor microenvironment might help considering SFT as a targetable sarcoma. Those biological insights recently translated into clinical management, especially IGF-1 inhibitor (figitumumab) and antiangiogenic drugs (including pazopanib, sunitinib and sorafenib). The results presented in this review suggest that antiangiogenic therapies such as pazopanib could be of interest for first-line treatment, while data on the efficacy of immunotherapy remain scarce, and more results are needed.
Future approaches in advanced SFT treatments should focus on international collaborations to develop large, randomized phase III trials to assess the efficacy of antiangiogenics and/or immune checkpoint inhibitors (ICI) (PD-1 inhibitors and PD-L1 inhibitors) compared to conventional chemotherapy. Additionally, a randomized phase III trial comparing antiangiogenic therapy alone versus antiangiogenic therapy plus anti-PD-1/PD-L1, as well as ICI in combination, would be of interest. For localized disease, could (neo)adjuvant therapy based on antiangiogenics and/or ICI decrease recurrence rate in high-risk patients? Nevertheless, an international approach to research on this rare disease is unavoidable, and additional biological ancillary studies are highly recommended.
In conclusion, SFT is a rare STS subtype for which standard chemotherapy has been reported to have limited efficacy. Overall, our review underlines the modest activity of standard chemotherapy in SFT but confirms that antiangiogenic agents have interesting activity and might be considered as the best therapeutic option in the advanced setting. However, the prognosis remains poor, and the inclusion of patients in clinical trials is highly recommended.

Author Contributions

Conceptualization, A.d.B., I.R.-C. and M.B.; methodology, M.B.; software, A.d.B.; validation, A.D., J.-Y.B., I.R.-C. and M.B.; formal analysis, A.d.B. and M.B.; investigation, A.d.B. and M.B.; resources, F.M.; data curation, A.d.B.; writing—original draft preparation, A.d.B. and M.B.; writing—review and editing, A.d.B., A.D., F.M., J.-Y.B., I.R.-C. and M.B.; visualization, A.D., J.-Y.B., I.R.-C. and M.B.; supervision, M.B.; project administration, M.B.; funding acquisition, M.B. All authors read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

J.-Y. Blay reports grants and personal fees from PharmaMar and AstraZeneca outside the submitted work. I. Ray-Coquard reports personal fees from Roche, PharmaMar, AstraZeneca, Clovis, GlaxoSmithKline, Bristol Myers Squibb, Agenus, Mersana, ImmunoGen, MSD, EISAI, and Novartis outside the submitted work. The other authors declare no conflict of interest.

References

  1. Klemperer, P.; Rabin, C.B. Primary Neoplasms of the pleura. A report of five cases. Am. J. Ind. Med. 1992, 22, 4–31. [Google Scholar] [CrossRef] [PubMed]
  2. Demicco, E.G.; Wagner, M.J.; Maki, R.G.; Gupta, V.; Iofin, I.; Lazar, A.; Wang, W.-L. Risk assessment in solitary fibrous tumors: Validation and refinement of a risk stratification model. Mod. Pathol. 2017, 30, 1433–1442. [Google Scholar] [CrossRef] [PubMed]
  3. Diebold, M.; Soltermann, A.; Hottinger, S.; Haile, S.; Bubendorf, L.; Komminoth, P.; Jochum, W.; Grobholz, R.; Theegarten, D.; Berezowska, S.; et al. Prognostic value of MIB-1 proliferation index in solitary fibrous tumors of the pleura implemented in a new score—Amulticenter study. Respir. Res. 2017, 18, 210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Pasquali, S.; Gronchi, A.; Strauss, D.; Bonvalot, S.; Jeys, L.; Stacchiotti, S.; Hayes, A.; Honore, C.; Collini, P.; Renne, S.; et al. Resectable extra-pleural and extra-meningeal solitary fibrous tumours: A multi-centre prognostic study. Eur. J. Surg. Oncol. (EJSO) 2016, 42, 1064–1070. [Google Scholar] [CrossRef] [PubMed]
  5. Salas, S.; Resseguier, N.; Blay, J.Y.; Le Cesne, A.; Italiano, A.; Chevreau, C.; Rosset, P.; Isambert, N.; Soulie, P.; Cupissol, D.; et al. Prediction of local and metastatic recurrence in solitary fibrous tumor: Construction of a risk calculator in a multicenter cohort from the French Sarcoma Group (FSG) database. Ann. Oncol. 2017, 28, 1779–1787. [Google Scholar] [CrossRef] [PubMed]
  6. WHO. Classification of Tumours Soft Tissue and Bone Tumours, 5th ed.; IARC Press: Lyon, France, 2020. [Google Scholar]
  7. de Pinieux, G.; Karanian, M.; Le Loarer, F.; Le Guellec, S.; Chabaud, S.; Terrier, P.; Bouvier, C.; Batistella, M.; Neuville, A.; Robin, Y.-M.; et al. Nationwide incidence of sarcomas and connective tissue tumors of intermediate malignancy over four years using an expert pathology review network. PLoS ONE 2021, 16, e0246958. [Google Scholar] [CrossRef]
  8. Kinslow, C.J.; Bruce, S.S.; Rae, A.I.; Sheth, S.A.; McKhann, G.M.; Sisti, M.B.; Bruce, J.N.; Sonabend, A.M.; Wang, T.J.C. Solitary-fibrous tumor/hemangiopericytoma of the central nervous system: A population-based study. J. Neuro-Oncol. 2018, 138, 173–182. [Google Scholar] [CrossRef]
  9. Kinslow, C.J.; Wang, T.J.C. Incidence of extrameningeal solitary fibrous tumors. Cancer 2020, 126, 4067. [Google Scholar] [CrossRef]
  10. Kim, J.M.; Choi, Y.-L.; Kim, Y.J.; Park, H.K. Comparison and evaluation of risk factors for meningeal, pleural, and extrapleural solitary fibrous tumors: A clinicopathological study of 92 cases confirmed by STAT6 immunohistochemical staining. Pathol.-Res. Pract. 2017, 213, 619–625. [Google Scholar] [CrossRef]
  11. Goodlad, J.R.; Fletcher, C.D.M. Solitary fibrous tumour arising at unusual sites: Analysis of a series. Histopathology 1991, 19, 515–522. [Google Scholar] [CrossRef]
  12. Ronchi, A.; Cozzolino, I.; Zito Marino, F.; Accardo, M.; Montella, M.; Panarese, I.; Roccuzzo, G.; Toni, G.; Franco, R.; De Chiara, A. Extrapleural solitary fibrous tumor: A distinct entity from pleural solitary fibrous tumor. An update on clinical, molecular and diagnostic features. Ann. Diagn. Pathol. 2018, 34, 142–150. [Google Scholar] [CrossRef] [PubMed]
  13. Gholami, S.; Cassidy, M.R.; Kirane, A.; Kuk, D.; Zanchelli, B.; Antonescu, C.R.; Singer, S.; Brennan, M. Size and Location are the Most Important Risk Factors for Malignant Behavior in Resected Solitary Fibrous Tumors. Ann. Surg. Oncol. 2017, 24, 3865–3871. [Google Scholar] [CrossRef] [PubMed]
  14. Haas, R.L.; Walraven, I.; Lecointe-Artzner, E.; Van Houdt, W.J.; Strauss, D.; Schrage, Y.; Hayes, A.J.; Raut, C.P.; Fairweather, M.; Baldini, E.H.; et al. Extrameningeal solitary fibrous tumors—Surgery alone or surgery plus perioperative radiotherapy: A retrospective study from the global solitary fibrous tumor initiative in collaboration with the Sarcoma Patients EuroNet. Cancer 2020, 126, 3002–3012. [Google Scholar] [CrossRef] [PubMed]
  15. Wignall, O.J.; Moskovic, E.C.; Thway, K.; Thomas, J.M. Solitary Fibrous Tumors of the Soft Tissues: Review of the Imaging and Clinical Features With Histopathologic Correlation. Am. J. Roentgenol. 2010, 195, W55–W62. [Google Scholar] [CrossRef]
  16. Zafar, H.; Takimoto, C.H.; Weiss, G. Doege-Potter syndrome. Med. Oncol. 2003, 20, 403–407. [Google Scholar] [CrossRef]
  17. Steigen, S.E.; Schaeffer, D.F.; West, R.B.; Nielsen, T.O. Expression of insulin-like growth factor 2 in mesenchymal neoplasms. Mod. Pathol. 2009, 22, 914–921. [Google Scholar] [CrossRef] [Green Version]
  18. Le Roith, D. Tumor-Induced Hypoglycemia. N. Engl. J. Med. 1999, 341, 757–758. [Google Scholar] [CrossRef]
  19. Davanzo, B.; Emerson, R.E.; Lisy, M.; Koniaris, L.G.; Kays, J.K. Solitary fibrous tumor. Transl. Gastroenterol. Hepatol. 2018, 3. [Google Scholar] [CrossRef] [Green Version]
  20. Kallen, M.E.; Hornick, J.L. The 2020 WHO Classification: What’s New in Soft Tissue Tumor Pathology? Am. J. Surg. Pathol. 2021, 45, e1–e23. [Google Scholar] [CrossRef]
  21. Stout, A.P.; Murray, M.R. Hemangiopericytoma a Vascular Tumor Featuring Zimmermann’s Pericytes. Ann. Surg. 1942, 116, 26–33. [Google Scholar] [CrossRef]
  22. Fletcher, C.; Bridge, J.; Hogendoorn, P.; Mertens, F. World Health Organization Classification of Tumours of Soft Tissue and Bone, 4th ed.; IARC Press: Lyon, France, 2013. [Google Scholar]
  23. Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimlin, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef] [Green Version]
  24. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro-Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef]
  25. England, D.M.; Hochholzer, L.; McCarthy, M.J. Localized Benign and Malignant Fibrous Tumors of the Pleura: A Clinicopathologic Review of 223 Cases. Am. J. Surg. Pathol. 1989, 13, 640–658. [Google Scholar] [CrossRef]
  26. de Perrot, M.; Fischer, S.; Bründler, M.-A.; Sekine, Y.; Keshavjee, S. Solitary fibrous tumors of the pleura. Ann. Thorac. Surg. 2002, 74, 285–293. [Google Scholar] [CrossRef]
  27. Schirosi, L.; Lantuejoul, S.; Cavazza, A.; Murer, B.; Brichon, P.Y.; Migaldi, M.; Sartori, G.; Sgambato, A.; Rossi, G. Pleuro-pulmonary Solitary Fibrous Tumors: A Clinicopathologic, Immunohistochemical, and Molecular Study of 88 Cases Confirming the Prognostic Value of de Perrot Staging System and p53 Expression, and Evaluating the Role of c-kit, BRAF, PDGFRs (α/β), c-met, and EGFR. Am. J. Surg. Pathol. 2008, 32, 1627–1642. [Google Scholar]
  28. Baldi, G.G.; Stacchiotti, S.; Mauro, V.; Tos, A.P.D.; Gronchi, A.; Pastorino, U.; Duranti, L.; Provenzano, S.; Marrari, A.; Libertini, M.; et al. Solitary fibrous tumor of all sites: Outcome of late recurrences in 14 patients. Clin. Sarcoma Res. 2013, 3, 4. [Google Scholar] [CrossRef] [Green Version]
  29. Georgiesh, T.; Boye, K.; Bjerkehagen, B. A novel risk score to predict early and late recurrence in solitary fibrous tumour. Histopathology 2020, 77, 123–132. [Google Scholar] [CrossRef]
  30. Mohamed, H.; Mandal, A.K. Natural History of Multifocal Solitary Fibrous Tumors of the Pleura: A 25-Year Follow-up Report. J. Natl. Med. Assoc. 2004, 96, 659–662. [Google Scholar]
  31. Park, C.K.; Lee, D.H.; Park, J.Y.; Park, S.H.; Kwon, K.Y. Multiple Recurrent Malignant Solitary Fibrous Tumors: Long-Term Follow-Up of 24 Years. Ann. Thorac. Surg. 2011, 91, 1285–1288. [Google Scholar] [CrossRef] [Green Version]
  32. Okike, N.; Bernatz, P.E.; Woolner, L.B. Localized mesothelioma of the pleura. J. Thorac. Cardiovasc. Surg. 1978, 75, 363–372. [Google Scholar] [CrossRef]
  33. van Houdt, W.J.; Westerveld, C.M.A.; Vrijenhoek, J.E.P.; van Gorp, J.; van Coevorden, F.; Verhoef, C.; van Dalen, T. Prognosis of Solitary Fibrous Tumors: A Multicenter Study. Ann. Surg. Oncol. 2013, 20, 4090–4095. [Google Scholar] [CrossRef]
  34. Wilky, B.A.; Montgomery, E.A.; Guzzetta, A.A.; Ahuja, N.; Meyer, C.F. Extrathoracic Location and “Borderline” Histology are Associated with Recurrence of Solitary Fibrous Tumors After Surgical Resection. Ann. Surg. Oncol. 2013, 20, 4080–4089. [Google Scholar] [CrossRef] [Green Version]
  35. Cranshaw, I.; Gikas, P.; Fisher, C.; Thway, K.; Thomas, J.; Hayes, A. Clinical outcomes of extra-thoracic solitary fibrous tumours. Eur. J. Surg. Oncol. (EJSO) 2009, 35, 994–998. [Google Scholar] [CrossRef] [Green Version]
  36. Reisenauer, J.S.; Mneimneh, W.; Jenkins, S.; Mansfield, A.S.; Aubry, M.C.; Fritchie, K.J.; Allen, M.S.; Blackmon, S.H.; Cassivi, S.D.; Nichols, F.C.; et al. Comparison of Risk Stratification Models to Predict Recurrence and Survival in Pleuropulmonary Solitary Fibrous Tumor. J. Thorac. Oncol. 2018, 13, 1349–1362. [Google Scholar] [CrossRef] [Green Version]
  37. Demicco, E.G.; Park, M.S.; Araujo, D.M.; Fox, P.S.; Bassett, R.L.; Pollock, R.; Lazar, A.; Wang, W.-L. Solitary fibrous tumor: A clinicopathological study of 110 cases and proposed risk assessment model. Mod. Pathol. 2012, 25, 1298–1306. [Google Scholar] [CrossRef] [Green Version]
  38. Mosquera, J.-M.; Fletcher, C.D.M. Expanding the Spectrum of Malignant Progression in Solitary Fibrous Tumors: A Study of 8 Cases With a Discrete Anaplastic Component—Is This Dedifferentiated SFT? Am. J. Surg. Pathol. 2009, 33, 1314–1321. [Google Scholar] [CrossRef]
  39. Ghanim, B.; Hess, S.; Bertoglio, P.; Celik, A.; Bas, A.; Oberndorfer, F.; Melfi, F.; Mussi, A.; Klepetko, W.; Pirker, C.; et al. Intrathoracic solitary fibrous tumor—An international multicenter study on clinical outcome and novel circulating biomarkers. Sci. Rep. 2017, 7, 12557. [Google Scholar] [CrossRef] [Green Version]
  40. Enzinger, F.M.; Smith, B.H. Hemangiopericytoma: An analysis of 106 cases. Hum. Pathol. 1976, 7, 61–82. [Google Scholar] [CrossRef]
  41. Robinson, D.R.; Wu, Y.-M.; Kalyana-Sundaram, S.; Cao, X.; Lonigro, R.J.; Sung, Y.-S.; Chen, C.-L.; Zhang, L.; Wang, R.; Su, F.; et al. Identification of recurrent NAB2-STAT6 gene fusions in solitary fibrous tumor by integrative sequencing. Nat. Genet. 2013, 45, 180–185. [Google Scholar] [CrossRef] [Green Version]
  42. Demicco, E.G.; Harms, P.; Patel, R.; Smith, S.C.; Ingram, D.; Torres, K.E.; Carskadon, S.L.; Camelo-Piragua, S.; McHugh, J.B.; Siddiqui, J.; et al. Extensive Survey of STAT6 Expression in a Large Series of Mesenchymal Tumors. Am. J. Clin. Pathol. 2015, 143, 672–682. [Google Scholar] [CrossRef] [Green Version]
  43. Doyle, L.A.; Vivero, M.; Fletcher, C.D.M.; Mertens, F.; Hornick, J. Nuclear expression of STAT6 distinguishes solitary fibrous tumor from histologic mimics. Mod. Pathol. 2013, 27, 390–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Koelsche, C.; Schweizer, L.; Renner, M.; Warth, A.; Jones, D.T.W.; Sahm, F.; Reuss, D.E.; Capper, D.; Knösel, T.; Schulz, B.; et al. Nuclear relocation of STAT6 reliably predictsNAB2-STAT6fusion for the diagnosis of solitary fibrous tumour. Histopathology 2014, 65, 613–622. [Google Scholar] [CrossRef] [PubMed]
  45. Schweizer, L.; Koelsche, C.; Sahm, F.; Piro, R.M.; Capper, D.; Reuss, D.E.; Pusch, S.; Habel, A.; Meyer, J.; Göck, T.; et al. Meningeal hemangiopericytoma and solitary fibrous tumors carry the NAB2-STAT6 fusion and can be diagnosed by nuclear expression of STAT6 protein. Acta Neuropathol. 2013, 125, 651–658. [Google Scholar] [CrossRef]
  46. Yoshida, A.; Tsuta, K.; Ohno, M.; Yoshida, M.; Narita, Y.; Kawai, A.; Asamura, H.; Kushima, R. STAT6 Immunohistochemistry Is Helpful in the Diagnosis of Solitary Fibrous Tumors. Am. J. Surg. Pathol. 2014, 38, 552–559. [Google Scholar] [CrossRef]
  47. Doyle, L.A.; Tao, D.; Mariño-Enríquez, A. STAT6 is amplified in a subset of dedifferentiated liposarcoma. Mod. Pathol. 2014, 27, 1231–1237. [Google Scholar] [CrossRef] [Green Version]
  48. Bouvier, C.; Bertucci, F.; Métellus, P.; Finetti, P.; De Paula, A.M.; Forest, F.; Mokhtari, K.; Miquel, C.; Birnbaum, D.; Vasiljevic, A.; et al. ALDH1 is an immunohistochemical diagnostic marker for solitary fibrous tumours and haemangiopericytomas of the meninges emerging from gene profiling study. Acta Neuropathol. Commun. 2013, 1, 10. [Google Scholar] [CrossRef]
  49. Vivero, M.; Doyle, L.A.; Fletcher, C.D.M.; Mertens, F.; Hornick, J.L. GRIA2 is a novel diagnostic marker for solitary fibrous tumour identified through gene expression profiling. Histopathology 2014, 65, 71–80. [Google Scholar] [CrossRef]
  50. Barthelmeß, S.; Geddert, H.; Boltze, C.; Moskalev, E.A.; Bieg, M.; Sirbu, H.; Brors, B.; Wiemann, S.; Hartmann, A.; Agaimy, A.; et al. Solitary Fibrous Tumors/Hemangiopericytomas with Different Variants of the NAB2-STAT6 Gene Fusion Are Characterized by Specific Histomorphology and Distinct Clinicopathological Features. Am. J. Pathol. 2014, 184, 1209–1218. [Google Scholar] [CrossRef]
  51. Chmielecki, J.; Crago, A.; Rosenberg, M.; O’Connor, R.; Walker, S.R.; Ambrogio, L.; Auclair, D.; McKenna, A.; Heinrich, M.; Frank, D.A.; et al. Whole-exome sequencing identifies a recurrent NAB2-STAT6 fusion in solitary fibrous tumors. Nat. Genet. 2013, 45, 131–132. [Google Scholar] [CrossRef] [Green Version]
  52. Mohajeri, A.; Tayebwa, J.; Collin, A.; Nilsson, J.; Magnusson, L.; von Steyern, F.V.; Brosjö, O.; Domanski, H.A.; Larsson, O.; Sciot, R.; et al. Comprehensive genetic analysis identifies a pathognomonic NAB2/STAT6 fusion gene, nonrandom secondary genomic imbalances, and a characteristic gene expression profile in solitary fibrous tumor. Genes Chromosomes Cancer 2013, 52, 873–886. [Google Scholar] [CrossRef]
  53. Schwachtgen, J.L.; Houston, P.; Campbell, C.; Sukhatme, V.; Braddock, M. Fluid shear stress activation of egr-1 transcription in cultured human endothelial and epithelial cells is mediated via the extracellular signal-related kinase 1/2 mitogen-activated protein kinase pathway. J. Clin. Invest. 1998, 101, 2540–2549. [Google Scholar] [CrossRef] [Green Version]
  54. Cheng, J.-C.; Chang, H.-M.; Leung, P.C.K. Egr-1 mediates epidermal growth factor-induced downregulation of E-cadherin expression via Slug in human ovarian cancer cells. Oncogene 2012, 32, 1041–1049. [Google Scholar] [CrossRef] [Green Version]
  55. Ji, R.-C. Hypoxia and lymphangiogenesis in tumor microenvironment and metastasis. Cancer Lett. 2014, 346, 6–16. [Google Scholar] [CrossRef]
  56. Binnemars-Postma, K.; Bansal, R.; Storm, G.; Prakash, J. Targeting the Stat6 pathway in tumor-associated macrophages reduces tumor growth and metastatic niche formation in breast cancer. FASEB J. 2018, 32, 969–978. [Google Scholar] [CrossRef] [Green Version]
  57. Shahmarvand, N.; Nagy, A.; Shahryari, J.; Ohgami, R.S. Mutations in the signal transducer and activator of transcription family of genes in cancer. Cancer Sci. 2018, 109, 926–933. [Google Scholar] [CrossRef] [Green Version]
  58. Li, J.; Rodriguez, J.P.; Niu, F.; Pu, M.; Wang, J.; Hung, L.-W.; Shao, Q.; Zhu, Y.; Ding, W.; Liu, Y.; et al. Structural basis for DNA recognition by STAT6. Proc. Natl. Acad. Sci. USA 2016, 113, 13015–13020. [Google Scholar] [CrossRef] [Green Version]
  59. Akaike, K.; Kurisaki-Arakawa, A.; Hara, K.; Suehara, Y.; Takagi, T.; Mitani, K.; Kaneko, K.; Yao, T.; Saito, T. Distinct clinicopathological features of NAB2-STAT6 fusion gene variants in solitary fibrous tumor with emphasis on the acquisition of highly malignant potential. Hum. Pathol. 2014, 46, 347–356. [Google Scholar] [CrossRef]
  60. Yuzawa, S.; Nishihara, H.; Wang, L.; Tsuda, M.; Kimura, T.; Tanino, M.; Tanaka, S. Analysis of NAB2-STAT6 Gene Fusion in 17 Cases of Meningeal Solitary Fibrous Tumor/Hemangiopericytoma: Review of the Literature. Am. J. Surg. Pathol. 2016, 40, 1031–1040. [Google Scholar] [CrossRef]
  61. Chuang, I.-C.; Liao, K.-C.; Huang, H.-Y.; Kao, Y.-C.; Li, C.-F.; Huang, S.-C.; Tsai, J.-W.; Chen, K.-C.; Lan, J.; Lin, P.-C. NAB2-STAT6 gene fusion and STAT6 immunoexpression in extrathoracic solitary fibrous tumors: The association between fusion variants and locations: Extrathoracic solitary fibrous tumor. Pathol. Int. 2016, 66, 288–296. [Google Scholar] [CrossRef]
  62. Huang, S.; Li, C.; Kao, Y.; Chuang, I.; Tai, H.; Tsai, J.; Yu, S.; Huang, H.; Lan, J.; Yen, S.; et al. The clinicopathological significance of NAB 2- STAT 6 gene fusions in 52 cases of intrathoracic solitary fibrous tumors. Cancer Med. 2015, 5, 159–168. [Google Scholar] [CrossRef] [Green Version]
  63. Park, Y.-S.; Kim, H.-S.; Kim, J.-H.; Choi, S.-H.; Kim, D.-S.; Ryoo, Z.Y.; Kim, J.-Y.; Lee, S. NAB2-STAT6 fusion protein mediates cell proliferation and oncogenic progression via EGR-1 regulation. Biochem. Biophys. Res. Commun. 2020, 526, 287–292. [Google Scholar] [CrossRef]
  64. Georgiesh, T.; Namløs, H.M.; Sharma, N.; Lorenz, S.; Myklebost, O.; Bjerkehagen, B.; Meza-Zepeda, L.A.; Boye, K. Clinical and molecular implications of NAB2-STAT6 fusion variants in solitary fibrous tumour. Pathology 2021, 53, 713–719. [Google Scholar] [CrossRef]
  65. Casali, P.G.; Abecassis, N.; Bauer, S.; Biagini, R.; Bielack, S.; Bonvalot, S.; Boukovinas, I.; Bovee, J.V.M.G.; Brodowicz, T.; Broto, J.M.; et al. Soft tissue and visceral sarcomas: ESMO–EURACAN Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2018, 29, iv51–iv67. [Google Scholar] [CrossRef]
  66. Casali, P.G.; Bielack, S.; Abecassis, N.; Aro, H.T.; Bauer, S.; Biagini, R.; Bonvalot, S.; Boukovinas, I.; Bovee, J.V.M.G.; Brennan, B.; et al. Bone sarcomas: ESMO–PaedCan–EURACAN Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2018, 29, iv79–iv95. [Google Scholar] [CrossRef]
  67. Casali, P.G.; Blay, J.Y.; Abecassis, N.; Bajpai, J.; Bauer, S.; Biagini, R.; Bielack, S.; Bonvalot, S.; Boukovinas, I.; Bovee, J.V.M.G.; et al. Gastrointestinal stromal tumours: ESMO–EURACAN–GENTURIS Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2022, 33, 20–33. [Google Scholar] [CrossRef]
  68. Blay, J.-Y.; Honoré, C.; Stoeckle, E.; Meeus, P.; Jafari, M.; Gouin, F.; Anract, P.; Ferron, G.; Rochwerger, A.; Ropars, M.; et al. Surgery in reference centers improves survival of sarcoma patients: A nationwide study. Ann. Oncol. 2019, 30, 1143–1153. [Google Scholar] [CrossRef]
  69. Benjamin, R.S.; Choi, H.; Macapinlac, H.A.; Burgess, M.A.; Patel, S.R.; Chen, L.L.; Podoloff, D.A.; Charnsangavej, C. We Should Desist Using RECIST, at Least in GIST. JCO 2007, 25, 1760–1764. [Google Scholar] [CrossRef]
  70. Choi, H.; Charnsangavej, C.; Faria, S.C.; Macapinlac, H.A.; Burgess, M.A.; Patel, S.R.; Chen, L.L.; Podoloff, D.A.; Benjamin, R.S. Correlation of Computed Tomography and Positron Emission Tomography in Patients With Metastatic Gastrointestinal Stromal Tumor Treated at a Single Institution With Imatinib Mesylate: Proposal of New Computed Tomography Response Criteria. J. Clin. Oncol. 2007, 25, 1753–1759. [Google Scholar] [CrossRef]
  71. Stacchiotti, S.; Verderio, P.; Messina, A.; Morosi, C.; Collini, P.; Llombart-Bosch, A.; Martin, J.; Comandone, A.; Cruz, J.; Ferraro, A.; et al. Tumor response assessment by modified Choi criteria in localized high-risk soft tissue sarcoma treated with chemotherapy. Cancer 2012, 118, 5857–5866. [Google Scholar] [CrossRef]
  72. Park, M.S.; Ravi, V.; Conley, A.; Patel, S.R.; Trent, J.C.; Lev, D.C.; Lazar, A.J.; Wang, W.-L.; Benjamin, R.S.; Araujo, D.M. The role of chemotherapy in advanced solitary fibrous tumors: A retrospective analysis. Clin. Sarcoma Res. 2013, 3, 7. [Google Scholar] [CrossRef] [Green Version]
  73. Park, M.S.; Patel, S.R.; Ludwig, J.A.; Trent, J.C.; Conrad, C.A.; Lazar, A.J.; Wang, W.-L.; Boonsirikamchai, P.; Choi, H.; Wang, X.; et al. Activity of temozolomide and bevacizumab in the treatment of locally advanced, recurrent, and metastatic hemangiopericytoma and malignant solitary fibrous tumor: Temozolomide/Bevacizumab Therapy in HPC/SFT. Cancer 2011, 117, 4939–4947. [Google Scholar] [CrossRef]
  74. Stacchiotti, S.; Libertini, M.; Negri, T.; Palassini, E.; Gronchi, A.; Fatigoni, S.; Poletti, P.; Vincenzi, B.; Tos, A.D.; Mariani, L.; et al. Response to chemotherapy of solitary fibrous tumour: A retrospective study. Eur. J. Cancer 2013, 49, 2376–2383. [Google Scholar] [CrossRef]
  75. Stacchiotti, S.; Negri, T.; Libertini, M.; Palassini, E.; Marrari, A.; De Troia, B.; Gronchi, A.; Tos, A.D.; Morosi, C.; Messina, A.; et al. Sunitinib malate in solitary fibrous tumor (SFT). Ann. Oncol. 2012, 23, 3171–3179. [Google Scholar] [CrossRef]
  76. Maruzzo, M.; Martin-Liberal, J.; Messiou, C.; Miah, A.; Thway, K.; Alvarado, R.; Judson, I.; Benson, C. Pazopanib as first line treatment for solitary fibrous tumours: The Royal Marsden Hospital experience. Clin. Sarcoma Res. 2015, 5, 5. [Google Scholar] [CrossRef] [Green Version]
  77. Cardillo, G.; Lococo, F.; Carleo, F.; Martelli, M. Solitary fibrous tumors of the pleura. Curr. Opin. Pulm. Med. 2012, 18, 339–346. [Google Scholar] [CrossRef]
  78. Espat, N.J.; Lewis, J.J.; Leung, D.; Woodruff, J.M.; Antonescu, C.R.; Shia, J.; Brennan, M.F. Conventional hemangiopericytoma: Modern analysis of outcome. Cancer 2002, 95, 1746–1751. [Google Scholar] [CrossRef]
  79. Robinson, L.A. Solitary Fibrous Tumor of the Pleura. Cancer Control. 2006, 13, 6. [Google Scholar] [CrossRef]
  80. Spitz, F.R.; Bouvet, M.; Pisters, P.W.T.; Pollock, R.E.; Feig, B.W. Hemangiopericytoma: A 20-year single-institution experience. Ann. Surg. Oncol. 1998, 5, 350–355. [Google Scholar] [CrossRef]
  81. Lahon, B.; Mercier, O.; Fadel, E.; Ghigna, M.R.; Petkova, B.; Mussot, S.; Fabre, D.; Le Chevalier, T.; Dartevelle, P. Solitary Fibrous Tumor of the Pleura: Outcomes of 157 Complete Resections in a Single Center. Ann. Thorac. Surg. 2012, 94, 394–400. [Google Scholar] [CrossRef]
  82. Nomori, H. Contacting metastasis of a fibrous tumor of the pleura. Eur. J. Cardio-Thoracic Surg. 1997, 12, 928–930. [Google Scholar] [CrossRef] [Green Version]
  83. Lococo, F.; Cesario, A.; Cardillo, G.; Filosso, P.L.; Galetta, D.; Carbone, L.; Oliaro, A.; Spaggiari, L.; Cusumano, G.; Margaritora, S.; et al. Malignant Solitary Fibrous Tumors of the Pleura: Retrospective Review of a Multicenter Series. J. Thorac. Oncol. 2012, 7, 1698–1706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Bowe, S.N.; Wakely, P.E., Jr.; Ozer, E. Head and neck solitary fibrous tumors: Diagnostic and therapeutic challenges. Laryngoscope 2012, 122, 1748–1755. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, X.; Qian, J.; Bi, Y.; Ping, B.; Zhang, R. Malignant transformation of orbital solitary fibrous tumor. Int. Ophthalmol. 2013, 33, 299–303. [Google Scholar] [CrossRef] [PubMed]
  86. Yang, X.; Zheng, J.; Ye, W.; Wang, Y.; Zhu, H.; Wang, L.; Zhang, Z. Malignant solitary fibrous tumors of the head and neck: A clinicopathological study of nine consecutive patients. Oral Oncol. 2009, 45, 678–682. [Google Scholar] [CrossRef]
  87. Wushou, A.; Jiang, Y.-Z.; Liu, Y.-R.; Shao, Z.-M. The demographic features, clinicopathologic characteristics, treatment outcome and disease-specific prognostic factors of solitary fibrous tumor: A population-based analysis. Oncotarget 2015, 6, 41875–41883. [Google Scholar] [CrossRef] [Green Version]
  88. Bishop, A.J.; Zagars, G.K.; Demicco, E.G.; Wang, W.-L.; Feig, B.W.; Guadagnolo, B.A. Soft Tissue Solitary Fibrous Tumor: Combined Surgery and Radiation Therapy Results in Excellent Local Control. Am. J. Clin. Oncol. 2018, 41, 81–85. [Google Scholar] [CrossRef]
  89. Bonvalot, S.; Gronchi, A.; Le Péchoux, C.; Swallow, C.J.; Strauss, D.; Meeus, P.; van Coevorden, F.; Stoldt, S.; Stoeckle, E.; Rutkowski, P.; et al. Preoperative radiotherapy plus surgery versus surgery alone for patients with primary retroperitoneal sarcoma (EORTC-62092: STRASS): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2020, 21, 1366–1377. [Google Scholar] [CrossRef]
  90. Haas, R.L.; Walraven, I.; Lecointe-Artzner, E.; Scholten, A.N.; van Houdt, W.; Griffin, A.M.; Ferguson, P.C.; Miah, A.B.; Zaidi, S.; DeLaney, T.F.; et al. Radiation Therapy as Sole Management for Solitary Fibrous Tumors (SFT): A Retrospective Study From the Global SFT Initiative in Collaboration With the Sarcoma Patients EuroNet. Int. J. Radiat. Oncol. 2018, 101, 1226–1233. [Google Scholar] [CrossRef]
  91. Oike, N.; Kawashima, H.; Ogose, A.; Hotta, T.; Hirano, T.; Ariizumi, T.; Yamagishi, T.; Umezu, H.; Inagawa, S.; Endo, N. A malignant solitary fibrous tumour arising from the first lumbar vertebra and mimicking an osteosarcoma: A case report. World J. Surg. Oncol. 2017, 15, 100. [Google Scholar] [CrossRef]
  92. De Boer, J.; Jager, P.L.; Wiggers, T.; Nieboer, P.; Wymenga, A.N.M.; Pras, E.; Hoogenberg, K.; Sleijfer, D.T.; Suurmeijer, A.J.; Van Der Graaf, W.T. The therapeutic challenge of a nonresectable solitary fibrous tumor in a hypoglycemic patient. Int. J. Clin. Oncol. 2006, 11, 478–481. [Google Scholar] [CrossRef]
  93. Han, G.; Zhang, Z.; Shen, X.; Wang, K.; Zhao, Y.; He, J.; Gao, Y.; Shan, X.; Xin, G.; Li, C.; et al. Doege-Potter syndrome: A review of the literature including a new case report. Medicine 2017, 96, e7417. [Google Scholar] [CrossRef] [PubMed]
  94. Constantinidou, A.; Jones, R.L.; Olmos, D.; Thway, K.; Fisher, C.; Al-Muderis, O.; Judson, I. Conventional anthracycline-based chemotherapy has limited efficacy in solitary fibrous tumour. Acta Oncol. 2011, 51, 550–554. [Google Scholar] [CrossRef] [PubMed]
  95. Levard, A.; Derbel, O.; Méeus, P.; Ranchère, D.; Ray-Coquard, I.; Blay, J.-Y.; Cassier, P.A. Outcome of patients with advanced solitary fibrous tumors: The Centre Léon Bérard experience. BMC Cancer 2013, 13, 109. [Google Scholar] [CrossRef]
  96. Stacchiotti, S.; Tortoreto, M.; Bozzi, F.; Tamborini, E.; Morosi, C.; Messina, A.; Libertini, M.; Palassini, E.; Cominetti, D.; Negri, T.; et al. Dacarbazine in Solitary Fibrous Tumor: A Case Series Analysis and Preclinical Evidence vis-à-vis Temozolomide and Antiangiogenics. Clin. Cancer Res. 2013, 19, 5192–5201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Khalifa, J.; Ouali, M.; Chaltiel, L.; Le Guellec, S.; Le Cesne, A.; Blay, J.-Y.; Cousin, P.; Chaigneau, L.; Bompas, E.; Piperno-Neumann, S.; et al. Efficacy of trabectedin in malignant solitary fibrous tumors: A retrospective analysis from the French Sarcoma Group. BMC Cancer 2015, 15, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Le Cesne, A.; Ray-Coquard, I.; Duffaud, F.; Chevreau, C.; Penel, N.; Nguyen, B.B.; Piperno-Neumann, S.; Delcambre, C.; Rios, M.; Chaigneau, L.; et al. Trabectedin in patients with advanced soft tissue sarcoma: A retrospective national analysis of the French Sarcoma Group. Eur. J. Cancer 2015, 51, 742–750. [Google Scholar] [CrossRef] [PubMed]
  99. Schöffski, P.; Timmermans, I.; Hompes, D.; Stas, M.; Sinnaeve, F.; De Leyn, P.; Coosemans, W.; Van Raemdonck, D.; Hauben, E.; Sciot, R.; et al. Clinical Presentation, Natural History, and Therapeutic Approach in Patients with Solitary Fibrous Tumor: A Retrospective Analysis. Sarcoma 2020, 2020, 1–9. [Google Scholar] [CrossRef] [Green Version]
  100. Outani, H.; Kobayashi, E.; Wasa, J.; Saito, M.; Takenaka, S.; Hayakawa, K.; Endo, M.; Takeuchi, A.; Kobayashi, H.; Kito, M.; et al. Clinical Outcomes of Patients with Metastatic Solitary Fibrous Tumors: A Japanese Musculoskeletal Oncology Group (JMOG) Multiinstitutional Study. Ann. Surg. Oncol. 2020, 28, 3893–3901. [Google Scholar] [CrossRef]
  101. Mulamalla, K.; Truskinovsky, A.M.; Dudek, A.Z. Rare case of hemangiopericytoma responds to sunitinib. Transl. Res. 2008, 151, 129–133. [Google Scholar] [CrossRef]
  102. De Pas, T.; Toffalorio, F.; Colombo, P.; Trifirò, G.; Pelosi, G.; Della Vigna, P.; Manzotti, M.; Agostini, M.; De Braud, F.G.M. Brief Report: Activity of Imatinib in a Patient with Platelet-Derived-Growth-Factor Receptor Positive Malignant Solitary Fibrous Tumor of the Pleura. J. Thorac. Oncol. 2008, 3, 938–941. [Google Scholar] [CrossRef] [Green Version]
  103. George, S.; Merriam, P.; Maki, R.G.; Van den Abbeele, A.D.; Yap, J.T.; Akhurst, T.; Harmon, D.C.; Bhuchar, G.; O’Mara, M.M.; D’Adamo, D.R.; et al. Multicenter Phase II Trial of Sunitinib in the Treatment of Nongastrointestinal Stromal Tumor Sarcomas. JCO 2009, 27, 3154–3160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Domont, J.; Massard, C.; Lassau, N.; Armand, J.-P.; Le Cesne, A.; Soria, J.-C. Hemangiopericytoma and antiangiogenic therapy: Clinical benefit of antiangiogenic therapy (sorafenib and sunitinib) in relapsed Malignant Haemangioperyctoma /Solitary Fibrous Tumour. Investig. New Drugs 2010, 28, 199–202. [Google Scholar] [CrossRef] [PubMed]
  105. Valentin, T.; Fournier, C.; Penel, N.; Bompas, E.; Chaigneau, L.; Isambert, N.; Chevreau, C. Sorafenib in patients with progressive malignant solitary fibrous tumors: A subgroup analysis from a phase II study of the French Sarcoma Group (GSF/GETO). Investig. New Drugs 2013, 31, 1626–1627. [Google Scholar] [CrossRef] [PubMed]
  106. Stacchiotti, S.; Negri, T.; Palassini, E.; Conca, E.; Gronchi, A.; Morosi, C.; Messina, A.; Pastorino, U.; Pierotti, M.A.; Casali, P.G.; et al. Sunitinib Malate and Figitumumab in Solitary Fibrous Tumor: Patterns and Molecular Bases of Tumor Response. Mol. Cancer Ther. 2010, 9, 1286–1297. [Google Scholar] [CrossRef] [Green Version]
  107. Stacchiotti, S.; Tortoreto, M.; Baldi, G.; Grignani, G.; Toss, A.; Badalamenti, G.; Cominetti, D.; Morosi, C.; Tos, A.D.; Festinese, F.; et al. Preclinical and clinical evidence of activity of pazopanib in solitary fibrous tumour. Eur. J. Cancer 2014, 50, 3021–3028. [Google Scholar] [CrossRef]
  108. Martin-Broto, J.; Stacchiotti, S.; Lopez-Pousa, A.; Redondo, A.; Bernabeu, D.; de Alava, E.; Casali, P.G.; Italiano, A.; Gutierrez, A.; Moura, D.S.; et al. Pazopanib for treatment of advanced malignant and dedifferentiated solitary fibrous tumour: A multicentre, single-arm, phase 2 trial. Lancet Oncol. 2019, 20, 134–144. [Google Scholar] [CrossRef]
  109. Martin-Broto, J.; Cruz, J.; Penel, N.; Le Cesne, A.; Hindi, N.; Luna, P.; Moura, D.S.; Bernabeu, D.; de Alava, E.; Lopez-Guerrero, J.A.; et al. Pazopanib for treatment of typical solitary fibrous tumours: A multicentre, single-arm, phase 2 trial. Lancet Oncol. 2020, 21, 456–466. [Google Scholar] [CrossRef]
  110. Kobayashi, H.; Iwata, S.; Wakamatsu, T.; Hayakawa, K.; Yonemoto, T.; Wasa, J.; Oka, H.; Ueda, T.; Tanaka, S. Efficacy and safety of trabectedin for patients with unresectable and relapsed soft-tissue sarcoma in Japan: A Japanese Musculoskeletal Oncology Group study. Cancer 2019, 126, 1253–1263. [Google Scholar] [CrossRef]
  111. Hatva, E. Vascular Growth Factors and Receptors in Capillary Hemangioblastomas and Hemang iopericytomas. Am. J. Pathol. 1996, 148, 13. [Google Scholar]
  112. Quek, R.; Wang, Q.; Morgan, J.A.; Shapiro, G.I.; Butrynski, J.E.; Ramaiya, N.; Huftalen, T.; Jederlinic, N.; Manola, J.; Wagner, A.J.; et al. Combination mTOR and IGF-1R Inhibition: Phase I Trial of Everolimus and Figitumumab in Patients with Advanced Sarcomas and Other Solid Tumors. Clin. Cancer Res. 2010, 17, 871–879. [Google Scholar] [CrossRef] [Green Version]
  113. Tazzari, M.; Negri, T.; Rini, F.; Vergani, B.; Huber, V.; Villa, A.; Dagrada, P.; Colombo, C.; Fiore, M.; Gronchi, A.; et al. Adaptive immune contexture at the tumour site and downmodulation of circulating myeloid-derived suppressor cells in the response of solitary fibrous tumour patients to anti-angiogenic therapy. Br. J. Cancer 2014, 111, 1350–1362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Kamamoto, D.; Ohara, K.; Kitamura, Y.; Yoshida, K.; Kawakami, Y.; Sasaki, H. Association between programmed cell death ligand-1 expression and extracranial metastasis in intracranial solitary fibrous tumor/hemangiopericytoma. J. Neuro-Oncol. 2018, 139, 251–259. [Google Scholar] [CrossRef] [PubMed]
  115. Dancsok, A.R.; Setsu, N.; Gao, D.; Blay, J.-Y.; Thomas, D.; Maki, R.G.; Nielsen, T.O.; Demicco, E.G. Expression of lymphocyte immunoregulatory biomarkers in bone and soft-tissue sarcomas. Mod. Pathol. 2019, 32, 1772–1785. [Google Scholar] [CrossRef] [PubMed]
  116. Berghoff, A.S.; Kresl, P.; Rajky, O.; Widhalm, G.; Ricken, G.; Hainfellner, J.A.; Marosi, C.; Birner, P.; Preusser, M. Analysis of the inflammatory tumor microenvironment in meningeal neoplasms. Clin. Neuropathol. 2020, 39, 256–262. [Google Scholar] [CrossRef]
  117. Boothe, J.T.; Budd, G.T.; Smolkin, M.B.; Ma, P.C. Durable Near-Complete Response to Anti-PD-1 Checkpoint Immunotherapy in a Refractory Malignant Solitary Fibrous Tumor of the Pleura. Case Rep. Oncol. 2017, 10, 998–1005. [Google Scholar] [CrossRef] [Green Version]
  118. Toulmonde, M.; Penel, N.; Adam, J.; Chevreau, C.; Blay, J.-Y.; Le Cesne, A.; Bompas, E.; Piperno-Neumann, S.; Cousin, S.; Grellety, T.; et al. Use of PD-1 Targeting, Macrophage Infiltration, and IDO Pathway Activation in Sarcomas: A Phase 2 Clinical Trial. JAMA Oncol. 2018, 4, 93. [Google Scholar] [CrossRef]
  119. Tap, W.D.; Wagner, A.J.; Schöffski, P.; Martin-Broto, J.; Krarup-Hansen, A.; Ganjoo, K.N.; Yen, C.-C.; Abdul Razak, A.R.; Spira, A.; Kawai, A.; et al. Effect of Doxorubicin Plus Olaratumab vs Doxorubicin Plus Placebo on Survival in Patients With Advanced Soft Tissue Sarcomas: The ANNOUNCE Randomized Clinical Trial. JAMA 2020, 323, 1266. [Google Scholar] [CrossRef]
Figure 1. Evolution of the WHO classification of SFTs over time. A crucial update for SFT classification is the development of risk stratification models that resulted in improved prognostication over the traditional benign/malignant distinction (please see the section below).
Figure 1. Evolution of the WHO classification of SFTs over time. A crucial update for SFT classification is the development of risk stratification models that resulted in improved prognostication over the traditional benign/malignant distinction (please see the section below).
Cancers 14 01064 g001
Figure 2. (A) Morphological appearance of SFT composed of spindle cells with a patternless architecture and a dense hyalinized collagenous stroma. Magnification: ×40; staining: hematoxylin, eosin, and Safran (HES). (B) Diffuse and strong STAT6 nuclear staining in SFT tumor cells. Magnification: ×40; staining: STAT6 antibody.
Figure 2. (A) Morphological appearance of SFT composed of spindle cells with a patternless architecture and a dense hyalinized collagenous stroma. Magnification: ×40; staining: hematoxylin, eosin, and Safran (HES). (B) Diffuse and strong STAT6 nuclear staining in SFT tumor cells. Magnification: ×40; staining: STAT6 antibody.
Cancers 14 01064 g002
Figure 3. Suspected role of the NAB2–STAT6 fusion transcript in SFT tumorigenesis.
Figure 3. Suspected role of the NAB2–STAT6 fusion transcript in SFT tumorigenesis.
Cancers 14 01064 g003
Table 1. Risk stratification model proposed by Demicco et al. [2].
Table 1. Risk stratification model proposed by Demicco et al. [2].
Risk FactorCut-OffPoints Assigned
3-Variable Model4-Variable Model
Patient age (years)<5500
>5511
Mitoses/mm2000
0.5–1.511
≥222
Tumor size (cm)0–4.900
5–9.911
10–14.922
≥1533
Tumor necrosis<10%N/A0
≥10%N/A1
RiskLow0–2 points0–3 points
Intermediate3–4 points4–5 points
High5–6 points6–7 points
Table 2. Summary of available data on systemic agents in SFT.
Table 2. Summary of available data on systemic agents in SFT.
AuthorsDesignnDrug (n)Response/Duration/RRmPFS (Months)mOS (Months)
Constantinidou et al. 2012 [94]R24Anthracycline-based (75 mg/m2) (n = 17)
Non-anthracycline-based (n = 7)
1 PR, 40% SD, 50% PD
5 SD, 2 PD
4.2
(95% CI: 0–10.1)
14.6 (95% CI: 9.3–19.9)
Levard et al. 2013 [95]R23Doxorubicin alone (n = 9)
Pegylated liposomal doxorubicin (n = 1)
Doxorubicin + ifosfamide (n = 8)
Doxorubicin + palifosfamide (n = 1).
Vinorelbine (n = 1)
Paclitaxel (n = 1)
Carboplatin and paclitaxel (n = 1)
Brostallicin (n = 1)
2 PR (8.7%) in doxorubicin-based, 13 SD (59%), 8 PD (35%). 9 pts (39%) progression-free at 6 months5.2
(95% CI: 3.2–7.1)
33.5 (95% CI: 14.2–52.8)
(whole cohort, n = 30)
Stacchiotti et al. 2013 [74]R31Anthracycline monotherapy (n = 8), Anthracycline + ifosfamide (n = 23)
Ifosfamide (n = 19)
RECIST: 6 PR (20%), 8 SD (27%), 16 PD (53%). 20% progression-free at 6 months.
2 PR (10%), 5 SD (26%), 12 PD (63%)
4
(range 2–15)
11.5 (range 3–50)
Stacchiotti et al. 2013 [96]R8Dacarbazine monotherapy (n = 8), (1200 mg/m2)RECIST: 3 PR, 4SD, 1PD7
(range 2–12)
-
Park et al. 2013 [72]R21Doxorubicin-based (n = 15)
Gemcitabine-based (n = 5)
Paclitaxel (n = 5)
0 PR, 16 SD (89%), 2 PD (11%). 5 (28%) progression-free >6 months 4.6 (95% CI: 4.0–5.3)10.3 years (95% CI: 5.7– 14.9 years)
Khalifa et al. 2015 [97]R11Trabectedin (1.5 mg/m2)RECIST: 1 PR (9.1%), 8 SD (72.7%)11.6
(95 % CI = 2.0; 15.2)
22.3
(95 % CI = 9.1; NR)
Le Cesne et al. 2015 [98]R13Trabectedin 1.5 mg/m2-7.6 (95% CI: 1.6–13.7)14.3 (95% CI: 0.8–27.8)
Schöffski et al. 2020 [99]R26Doxorubicin (n = 15) (57.7%)
Doxorubicin + ifosfamide (n = 3) (11.5%)
Doxorubicin/olaratumab (n = 2) (7.7%)
Doxorubicin + evofosfamide (n = 1)
Doxorubicin + ifosfamide + cisplatin (n = 1)
2nd line: ifosfamide (n = 5), pazopanib (n = 5)
Doxorubicin-based: 2 PR (13.3%), 4 SD (26.7%)
Ifosfamide: no response
Pazopanib: 3 PR (60%)
34.1 (95% CI: 1.0–157.1)56.0 (95% CI: 0.3–258.3)
Outani et al. 2020 [100]R31Anthracycline-based (n = 11)
Eribulin mesylate (n = 4)
Gemcitabine + docetaxel (n = 10)
Ifosfamide-based (n = 7)
Pazopanib (n = 22)
Trabectedin (n = 6)
Other (n = 13)
Multidrug regimen (n = 21)
--55
(95% CI: 40–86)
Kobayashi et al. 2020R140Trabectedin (1.2 mg/m2) 11 PR (7.9%), 54 SD (41.9%) with 25/54 SD >6 months3.7 (95% CI: 2.8–5.7)16.4 (95% CI: 11.5–21.2)
Mulamalla et al. 2008 [101]CR1SunitinibSD for 6 months--
De Pas et al. 2008 [102]CR1ImatinibPR for 21 months with major clinical benefit --
George et al. 2009 [103]R48Sunitinib (37.5 mg)1 PR, 11 SD + PR (22%) at 16 weeks, 7 (14%) at 24 weeks. 1.8-
Domont et al. 2009 [104]P2Dacarbazine (1000 mg/m2) + sorafenib (400 mg)
Sunitinib (50 mg)
PR, PD at 1.5 year
SD > 6 months
--
Park et al. 2011 [73]R14Temozolomide (150 mg/m2)–bevacizumab (5 mg/kg)Choi: 11 PR (79%), 2 SD9.7 (95% CI: 7.31–not estimable)-
Valentin et al. 2013 [105]P5Sorafenib (800 mg)No objective response, 2/5 SD (9 months)-19.7
Levard et al. 2013 [95]R10Pazopanib (800 mg) (n = 6)
Sunitinib (37.5 mg) (n = 4)
No objective response, 5
SD, 5 PD
4 pts (40%) progression-free >6 months: at 8.0 and 14.0 (pazopanib), and 29.5 and 29.9 months (sunitinib).
5.1 (95% CI: 0.7–9.6).-
Stacchiotti et al. 2010 [106]R11Sunitinib (37.5 mg)6 PR, 1 SD
(Choi)Response >6 months in 5 pts
--
Stacchiotti et al. 2014 [107]R6Pazopanib (800 mg)No RECIST response
1 PR, 2 SD (Choi)
3 (range 1–15)-
Maruzzo et al. 2015 [76]P13Pazopanib 1st line (800 mg)5 PR (46%), 4 SD (36%) (Choi) 4.7 (95% CI: 4.8–7.4)13.3 (95% CI: 3.9–22.6)
Martin-Broto et al. 2019 [108]P36Pazopanib (800 mg)Choi: 18 PR (51%), 9 SD (26%)
RECIST: 2 PR (6%), 21 SD (60%)
5.6 (95% CI: 4.51–6.62) NR
Martin-Broto et al. 2020 [109]P31Pazopanib (800 mg)18 PR (58%), 12 SD (39%), 1 PD (3%)12.1 (range 2.6–21.7)49.8
(range 8.2–91.3)
(R = retrospective study; P = prospective study; CR = case report; pts = patients; CI = confidence interval; NR = not reached).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

de Bernardi, A.; Dufresne, A.; Mishellany, F.; Blay, J.-Y.; Ray-Coquard, I.; Brahmi, M. Novel Therapeutic Options for Solitary Fibrous Tumor: Antiangiogenic Therapy and Beyond. Cancers 2022, 14, 1064. https://doi.org/10.3390/cancers14041064

AMA Style

de Bernardi A, Dufresne A, Mishellany F, Blay J-Y, Ray-Coquard I, Brahmi M. Novel Therapeutic Options for Solitary Fibrous Tumor: Antiangiogenic Therapy and Beyond. Cancers. 2022; 14(4):1064. https://doi.org/10.3390/cancers14041064

Chicago/Turabian Style

de Bernardi, Axel, Armelle Dufresne, Florence Mishellany, Jean-Yves Blay, Isabelle Ray-Coquard, and Mehdi Brahmi. 2022. "Novel Therapeutic Options for Solitary Fibrous Tumor: Antiangiogenic Therapy and Beyond" Cancers 14, no. 4: 1064. https://doi.org/10.3390/cancers14041064

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