Background: Myxofibrosarcoma (MFS) is among the most highly complex sarcoma types. Molecular cytogenetic studies have identified a high level of genomic complexity. Summary: This review provides an update of the current research related to MFS, with particular emphasis on emerging mechanisms of tumorigenesis and their potential therapeutic impact. Many novel possible molecular markers have been identified, not only for prognostication in MFS, but also to serve as possible therapeutic targets, and thereby improve clinical outcomes. However, the molecular pathogenesis of MFS remains incompletely understood. Key Messages: Patients suffering from advanced MFS might benefit from expanded molecular evaluation in order to detect specific expression profiles and identify drug-able targets. Moreover, immunotherapy represents an intriguingly perspective due to the presence of “T-cell inflamed” tumor microenvironment.

Myxofibrosarcoma (MFS) is currently regarded as a distinct fibroblastic/myofibroblastic tumor type defined by cellular pleomorphism, a curvilinear vascular pattern, and a myxoid stromal component [1].

Genomic instability is a hallmark of all human tumors [2, 3] and can be related to gene mutations, gene copy number alterations, and structural chromosomal abnormalities such as translocations, telomere dysfunction, and whole-chromosome aneuploidy that later results in chromosome instability [3].

Different studies have shown that MFS is among the most highly complex sarcoma types [4, 5]. Molecular cytogenetic studies have identified a high level of genomic complexity with a recurrent amplification of the chromosome 5p and 7q regions, the biological significance of which is unknown [6, 7].

Clonal heterogeneity may be evident within single samples but can also be observed between different tumor regions within the same primary site (so-called “regional heterogeneity”) or even between primary and metastatic sites [8]. This has recently been highlighted also in MFS by Lohberger et al. [9].

Many authors attempted to identify novel molecular markers, not only for prognostication in MFS, but also to serve as possible therapeutic targets, and thereby improve clinical outcomes. However, the molecular pathogenesis of MFS remains incompletely understood.

This review provides an update of the current research related to MFS, with particular emphasis on emerging mechanisms of tumorigenesis and their potential therapeutic impact.

Integrin-α10 is overexpressed in primary and metastatic melanoma cells and is associated with melanoma cell migration [10]. In contrast, in other solid tumors, the downregulation of integrin-α10 is associated with loss of phosphorylated retinoblastoma protein (RB) and with disease progression [11]. These results suggest that the role of integrin-α10 in human cancer is context dependent. The role of integrin-α10 in sarcomagenesis remains unknown.

Okada et al. [12] analyzed the gene expression profiles of 64 primary high-grade MFS and found that expression of ITGA10, which encodes integrin-10, was amplified on chromosome 5p and overexpressed in about 50% of MFS. In addition, 42% MFS possessed coamplification of TRIO and RICTOR and had worse outcomes.

Heitzer et al. [13], in a series of 25 MFS, observed that while 44% of G3 tumors showed a coamplification of TRIO and RICTOR, the same phenomenon was observed in only one G1 tumor (10%). However, overrepresentation of RICTOR alone was found in 40% of G1 tumors indicating that TRIO amplification is significantly more abundant in G3 tumors and therefore a late event.

Okada et al. [12] also carried on functional studies in patient-derived MFS cell lines. Knockdown of integrin-10 specifically inhibited growth and decreased RAC and AKT activation in MFS cells but not in normal mesenchymal cells, which suggests that integrin-10 signals through RAC and AKT in a tumor-specific manner. The authors hypothesized that integrin-10 signals through TRIO for the control of RAC and through RICTOR for the control of AKT. Also, cells overexpressing integrin-α10 exhibited greater migration and invasion than the control cells.

Hu et al. [14] evaluated 61 MFS and undifferentiated pleomorphic sarcoma (UPS). They found that increased ITGA10 and decreased PPP2R2B expression had independent prognostic value. PP2R2B encodes the regulatory B subunit of protein phosphatase 2A. It directly interacts with PDK1 and suppresses its activation. Therefore, both ITGA10 and PPP2R2B might act as upstream regulators of AKT. Somatic mutation was not common in these genes, but DNA methylation showed a profound effect on their expression.

Activation of the AKT/mTORC2 pathway was correlated with histological grade and tumor progression by Takahashi et al. [15] in a series including 68 primary MFS.

On MFS cell lines, Okada et al. [12] tested two drugs (NSC27366 and its newly developed derivative EHop-016) which inhibit RAC activation by TRIO [16, 17]. They used selective kinase inhibitor (INK128-MLN0128) to inhibit mTORC2 [18]. Alternatively, the pathway could be targeted with PAK inhibitors, such as IPA-3. Because integrin-α10 exerts its tumor-specific growth signal through both RAC and mTORC2, they hypothesized that inhibitors of RAC and mTORC2 may have cooperative effects in MFS cells.

Integrin-α10 itself is an attractive therapeutic target. Even though no direct inhibitor is currently available, it could plausibly be targeted using chimeric antigen receptor T-cell immunotherapy [19] or agents that interfere with integrin-α10 ligand binding, such as a ligand-mimetic peptide or a monoclonal antibody against its I domain [20].

Integrin-mediated signaling is also known to cross-talk with mesenchymal epithelial transition factor (MET) [21, 22].

MET encodes a transmembrane receptor tyrosine kinase, which constitutes the only known high-affinity receptor of hepatocyte growth factor (HGF). Through combination with HGF, MET could activate the RAS-MAPK or PI3K-AKT signaling pathway to promote cell motility and proliferation [22]. Besides mitogenic and antiapoptotic activities common to many growth factor receptors, enhanced MET activation can stimulate cell-cell detachment, migration, invasiveness, and angiogenesis [23-26].

Lee et al. [21] first investigated the role of MET in 86 primary localized MFS. Approximately two thirds of MFS displayed MET overexpression at immunohistochemistry (HIC), which correlated with adverse clinicopathological factors (tumor size and mitotic rate) and was independently predictive of shorter survival.

The authors quantified MET transcripts by real-time reverse transcription polymerase chain reaction (RT-PCR) for 16 laser-microdissected tumors and 2 MFS cell lines. Nine (56%) specimens showed apparently upregulated MET transcript, suggesting their frequent upregulation in MFS. Lee et al. [21] found wild-type MET oncogene in both cell lines. The authors suggested that, as proven in a variety of cancers, MET protein expression might be upregulated by several small noncoding micro-RNAs [27-29].

More recently, Ma et al. [30] confirmed the relationship between MET and MFS. They used HIC and fluorescence in situ hybridization to detect the MET expression and gene status in 30 MFS. The authors observed MET overexpression in 14 cases (46.7%), with a correlation with FNLCC grade and mitotic rate. They also found a polysomy of chromosome 7 in 11 of these cases, thus suggesting that chromosome 7 polysomy, rather than MET amplification, led to the overexpression of MET protein. Patients with MET overexpression or chromosome 7 polysomy also had a high risk of local recurrence and metastasis.

Ogura et al. [31] observed also that recurrent mutational targets included genes encoding components of the receptor tyrosine kinase-RAS-PI3K cancer pathway in 31% of the cases.

The findings of the studies [21, 30] strengthen the possible causative function of MET in conferring an aggressive phenotype, implying the potentiality of HGF/MET as an attractive target of therapeutics in MFS [32-34]. Nowadays, there are two approved drugs tested in other cancers, crizotinib [35] and cabozantinib [36].

Regarding the NF1 gene, Ogura et al. [31] found 4 homozygous deletions and 9 somatic mutations in 116 MFS.

These data suggest that a distinctive pattern of NF1 aberrations may play a role in MFS tumorigenesis, similarly to other cancers [37]. Loss-of-function mutations in NF1 gene were also reported by Barretina et al. [38] in 5 out of 35 MFS. Heitzer et al. [13] were unable to inform about the NF1 mutation status since they used a hotspot panel for mutation analysis which did not cover NF1. However, loss of NF1 was observed in 2 out of 25 patients.

The use of MEK inhibitors may be a potential therapeutic option in NF1-deficient MFS, as recent studies revealed that tumors harboring NF1 inactivation (inactivating/deleterious NF1 mutations) exhibited activation of the MAPK/ERK pathway and hence are potential targets for MEK inhibitors [39].

The tyrosine kinase receptor MET and its ligand HGF, splice variants of CD44 and ezrin cooperate [40].

CD44 are ubiquitously expressed on all cell types, where they act as receptors for hyaluronic acid. They play various roles and are involved in cellular differentiation, cellular migration, and cell-cell contact [41]. Abnormal expression of CD44 may promote cell invasion and was correlated with more aggressive behavior in various cancers, including soft tissue sarcomas (STS) [42-44].

Matuschek et al. [45] analyzed with PCR 4 variants of CD44 (CD44, CD44s, CD44v6, and CD44v8) in 34 MFS. They found a significant difference in tumor-related survival only for CD44s and CD44v6, with increased CD44s and decreased CD44v6 expression associated with a better prognosis. CD44v6 was shown to be strictly required for MET activation by HGF in rat and human carcinoma cells, in established cell lines as well as in primary keratinocytes [40]. This is consistent with prostate cancer, in which CD44s is a tumor suppressor but certain CD44 variants are oncogenes and promote growth [28, 29, 46].

More recently, Tsuchie et al. [47] evaluated the HIC expression of CD44s in 44 MFS. The overall expression rate of CD44s in all patients was relatively high. The authors found that high expression of CD44s was associated with local recurrence but did not affect survival.

Ezrin functions as a linker between the plasma membrane and cortical actin cytoskeleton [48]. Increased ezrin levels have been reported to be associated with a high metastatic propensity in different cancers, including rhabdomyosarcoma and osteosarcoma [49-51]. Its underlying mechanistic basis may be ascribed to increased activated ezrin to modulate pleiotropic cellular phenotypes related to cancerous states, such as substrate adhesion, cell survival, cell migration, and formation of cell-cell junctions [50, 51].

Huang et al. [52] evaluated the expression of ezrin in 78 primary localized MFS with HIC. Ezrin overexpression was present in approximately half of the tumors (49%), and it was significantly associated with important variables related to tumor aggressiveness, including necrosis and FNLCC grading. Additionally, ezrin overexpression was an independent poor prognosticator for survival.

They also measured ezrin mRNA expression levels in 2 MFS cell lines, through real-time RT-PCR and Western blot test [53, 54]. The ezrin mRNA expression level of MFS cell lines was apparently lower than that of normal fibroblasts. However, active ezrin (phosphorylated form at the residue of Thr567) was only detectable in MFS cells, but not in fibroblasts.

α-Methylacyl coenzyme A racemase (AMACR) is a peroxisomal and mitochondrial enzyme encoded on chromosome 5p13.3, which acts as a gatekeeper for the β-oxidation of dietary branched-chain fatty acids and bile acid synthesis [55]. It was identified as a protein which drives tumor growth in prostate cancer and other neoplasias, because most malignancies increase the need for fatty acids as an energy source [55-57].

The role of AMACR in MFS has recently been investigated by Li et al. [58] in a series of 105 primary MFS and in 2 cell lines. AMACR amplification was found in 21% MFS by fluorescence in situ hybridization and was strongly correlated with HIC overexpression. AMACR overexpression was correlated with FNLCC grade and worse survival. However, approximately 40% of AMACR-overexpressing MFS lacked gene amplification. Thus, involvement of alternative regulatory mechanisms was likely in a subset of MFS.

The authors also found that in MFS cell lines and xenografts, AMACR overexpression could increase cyclin D1 expression at the mRNA and protein levels, whereas the mechanisms underlying this regulatory link remain to be elucidated.

Finally, they tested ebselen oxide [59]. Inducing pro-teasome-mediated AMACR degradation and apoptosis, ebselen oxide demonstrates selective cytotoxicity in AMACR-expressing MFS cell lines and dose-dependent inhibition of derived xenografts, suggesting that AMACR is a potential therapeutic target in MFS.

Melanoma-associated antigen 3 (MAGE-A3) protein was found to be overexpressed in multiple cancers [60].

Conley et al. [61] explored expression of MAGE-A3 among a diverse number of sarcomas and sarcoma cell lines [62, 63].

MAGE-A3 was overexpressed in 41% of MFS and UPS, significantly higher than in other STS histotypes. High expression was more likely to be seen in recurrences than in primary tumors and was associated with an adverse survival.

Immunotherapies targeting MAGE-A3 have shown both positive and negative results in the treatment of various cancers [64].

Ogura et al. [31] analyzed a total of 106 MFS. They found frequent alterations in genes related to p53 signaling (51%), along with those associated with the cell cycle checkpoint (43%), including RB1, CDKN2A/CDKN2B, CCND1, and CDK6. Alterations of any of the cell cycle regulators were associated with poorer overall survival. The authors [31] also identified a novel BRAF fusion gene (SLC37A-BRAF) which could be targeted with anti-BRAF therapies [65].

Heitzer et al. [13] evaluated MFS samples from 25 patients. Somatic mutations were identified in only 11 (44%) patients. Grade 3 tumors showed a higher amount of somatic copy number alterations than grade 1. All these 11 patients showed at least one somatic TP53 mutation. TP53 mutations are relatively common in sarcomas with nonspecific genetic aberrations and complex karyotypes compared to sarcomas with reciprocal specific translocations [66]. Moreover, they identified focal amplification/deletion in a variety of known cancer driver genes such as CDKN2A, CCND1, CCNE1, EGFR, EPHA3, EPHB1, FGFR1, JUN, NF1, RB1, or RET, in particular in G3 tumors. In addition, G3 tumor areas of the same tumor showed novel emerged focal amplifications compared to the G1 areas. Many of these, such as BRAF, EGFR, FGFR, KIT, or RET, are indeed actionable targets and are actively being used for precision medicine in different tumor entities.

Ogura et al. [31] also identified novel recurrently mutated genes such as GNAS (9%), ATRX (9%), KRAS (7%), and JAK1 (4%). The presence of GNAS mutations was significantly associated with local recurrence-free survival [31].

Amplification of JAK1 could be a therapeutic target in MFS because aberrant activation of the JAK/STAT pathway has been shown to be a promising target in various cancer types [67, 68].

S-phase kinase-associated protein 2 (SKP2) is a negative regulator of p27kip1 cell-cycle inhibitor. Its overexpression was associated with metastatic propensity in common carcinomas, such as prostatic and esophageal cancers [69, 70].

The role of SKP2 in MFS was evaluated in two consecutive series [71, 72] which included a total of 82 cases. SKP2 gene amplification was detected in 38% of cases. Its amplification strongly correlated with SKP2 overexpression in HIC and with higher FNLCC grades. However, 14% of such tumors and SKP2-expressing cell lines lacked gene amplification. Both SKP2 protein overexpression and gene amplification were highly predictive of worse outcomes.

In addition, pharmacological assays were evaluated both on cell lines and in xenograft models for the therapeutic relevance of bortezomib [73, 74]. The authors showed that bortezomib treatment in SKP2-overexpressing MFS could achieve cytotoxicity in vitro. They also found a remarkably decreased mRNA expression, rather than increased protein degradation, in both MFS cell lines treated. On MFS in vivo studies of MFS xenografts, they observed significant tumor regression with lowered SKP2 labeling, increased p27Kip1 expression, and increased apoptosis [75].

CD109 is a cell surface glycoprotein that is expressed on endothelial cells, activated T lymphocytes, platelets, and a subpopulation of bone marrow CD34 cells. CD109 is a transforming growth factor β (TGF-β) coreceptor that regulates TGF-β receptor endocytosis and degrading, thus inhibiting TGF-β signaling [76]. The roles of TGF-β in remodeling the tumor microenvironment, by suppressing T-cell differentiation and activity, inducing fibrosis and angiogenesis, have been extensively characterized [77]. TGF-β also appears to have a role in chemotherapy resistance, as reported in several tumors including breast cancer and squamous cell carcinoma [78-80].

CD109 has been found to be overexpressed in various cancers, including STS [81, 82]. Emori et al. [83] evaluated 37 MFS with HIC and found an overexpression of the CD109 protein in 10% of patients, which was significantly associated with decreased survival.

De Vita et al. [84] analyze 3 patient-derived high-grade MFS cell cultures. HIC analysis showed that tumor cells were strongly positive for CD109. Conversely, CD109 was weakly expressed in normal tissue. Also, higher levels of CD109 were observed with gene expression analysis in all primary MFS cultures with respect to normal tissue.

Over the years, numerous preclinical and clinical studies have sought to leverage these insights and use inhibitors of TGF-β synthesis, receptor binding, or signal transduction in order to inhibit cancer progression [85].

Programmed death-1 protein (PD-1) is normally expressed on the surface of activated T cells and suppresses unwanted or excessive immune responses, including autoimmune reactions. Its ligand PD-L1 can be expressed by various cells, including macrophages and tumor cells. The PD-1/PD-L1 interaction is a major pathway used by tumors to suppress immune control. Several studies have assessed the expression of PD-L1 in sarcomas [86].

T-cell infiltration and PD-L1 expression were found to be higher in sarcomas with complex genomics and particularly in UPS than in other STS [87, 88]. Among STS subtypes, UPS/MFS has the highest median macrophage infiltration and the infiltration of immature dendritic cells was positively correlated with survival. Ogura et al. [31] observed that the average fraction of infiltrated CD8+ T cells was significantly higher in the better prognostic cluster in MFS. Hu et al. [14] reported that a high level of CD4+ T-cell infiltration was associated with a significantly better relapse-free survival in UPS and MFS. They also found a moderate negative correlation between the 2-gene signature (increased ITGA10 and decreased PPP2R2B) and CD4+ T-cell infiltration. Conley et al. [61] also found that HLA-A and HLA-DPB1 mRNA expression was significantly higher in UPS/MFS compared to other STS subtypes. Also, the lymphocyte infiltrate in UPS/MFS was significantly higher compared to other histotypes. These findings suggest that UPS and MFS may have immunologically mutated protein targets and thus might respond to immune checkpoint therapy [89].

The results of successful immunotherapy trials using an anti-PD-1 antibody and combination anti-PD-1/anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) showed that a significant number of UPS experienced a reduction in tumor size. A phase II study of single-agent pembrolizumab (anti-PD-1 antibody) in multiple sarcoma types showed an objective response rate of 40% in UPS [90, 91]. A separate trial involving nivolumab alone and in combination with ipilimumab demonstrated responses in UPS [92].

Studies in melanoma, non-small-cell lung cancer, and colorectal cancer have identified the presence of a “T-cell inflamed” tumor microenvironment, high tumor mutation burden, microsatellite instability, and PD-L1 expression as biomarkers of response to PD-1/PD-L1 blockade in some patients [93]. A pair of recent papers [94, 95] identified TGF-β signaling in the tumor microenvironment as a determinant of tumor T-cell exclusion and poor response to PD-1/PD-L1 blockade.

In murine models, Mariathasan et al. [94] demonstrated that combined treatment with antibodies targeting PD-L1 and TGF-β induces CD8+ T-cell infiltration and tumor regression, with complete response rates of 70%, while monotherapy with either agent alone is ineffective. Beyond TGF-β, therapies targeting other oncogenic signaling pathways, including MAPK and AKT-PI3K-mTOR, have also shown promising results in combination with immune checkpoint blockade, at least partly due to their effect on the tumor microenvironment [96].

Although the outcomes for MFS have not changed, substantial advances in the understanding of the natural history and pathogenesis of this sarcoma have been made. Many possible drug-able targets have been identified. However, MFS harbor a very heterogeneous karyotype with very different clones observed in each patient, but also in distinctive areas of the same tumor. Thus, patients suffering from advanced MFS might benefit from expanded molecular evaluation in order to detect specific expression profiles and identify drug-able targets.

Moreover, promising results have been found with immunotherapy. Future research that combines treatment modalities, including immunotherapy, are warranted.

No ethic approval was needed for reviews.

All the authors have no conflict of interests to declare.

Conceived the paper: A.S., D.M.D., M.D.P. Collected the data: G.B., P.S., M.D.P. Wrote the paper: A.S., G.B., M.D.P., D.M.D. Manuscript final editing: A.S., P.S., M.D.P.

1.
Fletcher
CD
,
Gustafson
P
,
Rydholm
A
,
Willén
H
,
Akerman
M
.
Clinicopathologic re-evaluation of 100 malignant fibrous histiocytomas: prognostic relevance of subclassification
.
J Clin Oncol
.
2001
Jun
;
19
(
12
):
3045
50
.
[PubMed]
0732-183X
2.
Gordon
DJ
,
Resio
B
,
Pellman
D
.
Causes and consequences of aneuploidy in cancer
.
Nat Rev Genet
.
2012
Jan
;
13
(
3
):
189
203
.
[PubMed]
1471-0056
3.
Hanahan
D
,
Weinberg
RA
.
Hallmarks of cancer: the next generation
.
Cell
.
2011
Mar
;
144
(
5
):
646
74
.
[PubMed]
0092-8674
4.
Gibault
L
,
Pérot
G
,
Chibon
F
,
Bonnin
S
,
Lagarde
P
,
Terrier
P
, et al
New insights in sarcoma oncogenesis: a comprehensive analysis of a large series of 160 soft tissue sarcomas with complex genomics
.
J Pathol
.
2011
Jan
;
223
(
1
):
64
71
.
[PubMed]
0022-3417
5.
Willems
SM
,
Debiec-Rychter
M
,
Szuhai
K
,
Hogendoorn
PC
,
Sciot
R
.
Local recurrence of myxofibrosarcoma is associated with increase in tumour grade and cytogenetic aberrations, suggesting a multistep tumour progression model
.
Mod Pathol
.
2006
Mar
;
19
(
3
):
407
16
.
[PubMed]
0893-3952
6.
Idbaih
A
,
Coindre
JM
,
Derré
J
,
Mariani
O
,
Terrier
P
,
Ranchère
D
, et al
Myxoid malignant fibrous histiocytoma and pleomorphic liposarcoma share very similar genomic imbalances
.
Lab Invest
.
2005
Feb
;
85
(
2
):
176
81
.
[PubMed]
0023-6837
7.
Ohguri
T
,
Hisaoka
M
,
Kawauchi
S
,
Sasaki
K
,
Aoki
T
,
Kanemitsu
S
, et al
Cytogenetic analysis of myxoid liposarcoma and myxofibrosarcoma by array-based comparative genomic hybridisation
.
J Clin Pathol
.
2006
Sep
;
59
(
9
):
978
83
.
[PubMed]
0021-9746
8.
Gerlinger
M
,
Rowan
AJ
,
Horswell
S
,
Math
M
,
Larkin
J
,
Endesfelder
D
, et al
Intratumor heterogeneity and branched evolution revealed by multiregion sequencing
.
N Engl J Med
.
2012
Mar
;
366
(
10
):
883
92
.
[PubMed]
0028-4793
9.
Lohberger
B
,
Stuendl
N
,
Leithner
A
,
Rinner
B
,
Sauer
S
,
Kashofer
K
, et al
Establishment of a novel cellular model for myxofibrosarcoma heterogeneity
.
Sci Rep
.
2017
Mar
;
7
(
1
):
44700
.
[PubMed]
2045-2322
10.
Wenke
AK
,
Kjellman
C
,
Lundgren-Akerlund
E
,
Uhlmann
C
,
Haass
NK
,
Herlyn
M
, et al
Expression of integrin alpha10 is induced in malignant melanoma
.
Cell Oncol
.
2007
;
29
(
5
):
373
86
.
[PubMed]
1570-5870
11.
Engel
BE
,
Welsh
E
,
Emmons
MF
,
Santiago-Cardona
PG
,
Cress
WD
.
Expression of integrin alpha 10 is transcriptionally activated by pRb in mouse osteoblasts and is downregulated in multiple solid tumors
.
Cell Death Dis
.
2013
Nov
;
4
(
11
):
e938
.
[PubMed]
2041-4889
12.
Okada
T
,
Lee
AY
,
Qin
LX
,
Agaram
N
,
Mimae
T
,
Shen
Y
, et al
Integrin-α10 Dependency Identifies RAC and RICTOR as Therapeutic Targets in High-Grade Myxofibrosarcoma
.
Cancer Discov
.
2016
Oct
;
6
(
10
):
1148
65
.
[PubMed]
2159-8274
13.
Heitzer
E
,
Sunitsch
S
,
Gilg
MM
,
Lohberger
B
,
Rinner
B
,
Kashofer
K
, et al
Expanded molecular profiling of myxofibrosarcoma reveals potentially actionable targets
.
Mod Pathol
.
2017
Dec
;
30
(
12
):
1698
709
.
[PubMed]
0893-3952
14.
Hu
Q
,
Zhou
S
,
Hu
X
,
Zhang
H
,
Huang
S
,
Wang
Y
.
Systematic screening identifies a 2-gene signature as a high-potential prognostic marker of undifferentiated pleomorphic sarcoma/myxofibrosarcoma
.
J Cell Mol Med
.
2020
Jan
;
24
(
1
):
1010
21
.
[PubMed]
1582-1838
15.
Takahashi
Y
,
Kohashi
K
,
Yamada
Y
,
Endo
M
,
Setsu
N
,
Ishii
T
, et al
Activation of the Akt/mammalian target of rapamycin pathway in myxofibrosarcomas
.
Hum Pathol
.
2014
May
;
45
(
5
):
984
93
.
[PubMed]
0046-8177
16.
Montalvo-Ortiz
BL
,
Castillo-Pichardo
L
,
Hernández
E
,
Humphries-Bickley
T
,
De la Mota-Peynado
A
,
Cubano
LA
, et al
Characterization of EHop-016, novel small molecule inhibitor of Rac GTPase
.
J Biol Chem
.
2012
Apr
;
287
(
16
):
13228
38
.
[PubMed]
0021-9258
17.
Gao
Y
,
Dickerson
JB
,
Guo
F
,
Zheng
J
,
Zheng
Y
.
Rational design and characterization of a Rac GTPase-specific small molecule inhibitor
.
Proc Natl Acad Sci USA
.
2004
May
;
101
(
20
):
7618
23
.
[PubMed]
0027-8424
18.
Slotkin
EK
,
Patwardhan
PP
,
Vasudeva
SD
,
de Stanchina
E
,
Tap
WD
,
Schwartz
GK
.
MLN0128, an ATP-competitive mTOR kinase inhibitor with potent in vitro and in vivo antitumor activity, as potential therapy for bone and soft-tissue sarcoma
.
Mol Cancer Ther
.
2015
Feb
;
14
(
2
):
395
406
.
[PubMed]
1535-7163
19.
Fu
X
,
Rivera
A
,
Tao
L
,
Zhang
X
.
Genetically modified T cells targeting neovasculature efficiently destroy tumor blood vessels, shrink established solid tumors and increase nanoparticle delivery
.
Int J Cancer
.
2013
Nov
;
133
(
10
):
2483
92
.
[PubMed]
0020-7136
20.
Brennan
M
,
Cox
D
.
The therapeutic potential of I-domain integrins
.
Adv Exp Med Biol
.
2014
;
819
:
157
78
.
[PubMed]
0065-2598
21.
Lee
JC
,
Li
CF
,
Fang
FM
,
Wang
JW
,
Jeng
YM
,
Yu
SC
, et al
Prognostic implication of MET overexpression in myxofibrosarcomas: an integrative array comparative genomic hybridization, real-time quantitative PCR, immunoblotting, and immunohistochemical analysis
.
Mod Pathol
.
2010
Oct
;
23
(
10
):
1379
92
.
[PubMed]
0893-3952
22.
Weidner
KM
,
Sachs
M
,
Birchmeier
W
.
The Met receptor tyrosine kinase transduces motility, proliferation, and morphogenic signals of scatter factor/hepatocyte growth factor in epithelial cells
.
J Cell Biol
.
1993
Apr
;
121
(
1
):
145
54
.
[PubMed]
0021-9525
23.
Koochekpour
S
,
Jeffers
M
,
Rulong
S
,
Taylor
G
,
Klineberg
E
,
Hudson
EA
, et al
Met and hepatocyte growth factor/scatter factor expression in human gliomas
.
Cancer Res
.
1997
Dec
;
57
(
23
):
5391
8
.
[PubMed]
0008-5472
24.
Eder
JP
,
Vande Woude
GF
,
Boerner
SA
,
LoRusso
PM
.
Novel therapeutic inhibitors of the c-Met signaling pathway in cancer
.
Clin Cancer Res
.
2009
Apr
;
15
(
7
):
2207
14
.
[PubMed]
1078-0432
25.
Longati
P
,
Bardelli
A
,
Ponzetto
C
,
Naldini
L
,
Comoglio
PM
.
Tyrosines1234-1235 are critical for activation of the tyrosine kinase encoded by the MET proto-oncogene (HGF receptor)
.
Oncogene
.
1994
Jan
;
9
(
1
):
49
57
.
[PubMed]
0950-9232
26.
Jankowski
K
,
Kucia
M
,
Wysoczynski
M
,
Reca
R
,
Zhao
D
,
Trzyna
E
, et al
Both hepatocyte growth factor (HGF) and stromal-derived factor-1 regulate the metastatic behavior of human rhabdomyosarcoma cells, but only HGF enhances their resistance to radiochemotherapy
.
Cancer Res
.
2003
Nov
;
63
(
22
):
7926
35
.
[PubMed]
0008-5472
27.
Migliore
C
,
Petrelli
A
,
Ghiso
E
,
Corso
S
,
Capparuccia
L
,
Eramo
A
, et al
MicroRNAs impair MET-mediated invasive growth
.
Cancer Res
.
2008
Dec
;
68
(
24
):
10128
36
.
[PubMed]
0008-5472
28.
Duan
Z
,
Choy
E
,
Nielsen
GP
,
Rosenberg
A
,
Iafrate
J
,
Yang
C
, et al
Differential expression of microRNA (miRNA) in chordoma reveals a role for miRNA-1 in Met expression
.
J Orthop Res
.
2010
Jun
;
28
(
6
):
746
52
.
[PubMed]
1554-527X
29.
Kim
S
,
Lee
UJ
,
Kim
MN
,
Lee
EJ
,
Kim
JY
,
Lee
MY
, et al
MicroRNA miR-199a* regulates the MET proto-oncogene and the downstream extracellular signal-regulated kinase 2 (ERK2)
.
J Biol Chem
.
2008
Jun
;
283
(
26
):
18158
66
.
[PubMed]
0021-9258
30.
Ma
S
,
Fan
L
,
Liu
Y
,
Wang
Y
,
Yu
K
,
Wang
L
, et al
MET-overexpressing myxofibrosarcoma frequently exhibit polysomy of chromosome 7 but not MET amplification, especially in high-grade cases: clinical and pathological review of 30 myxofibrosarcoma cases
.
Diagn Pathol
.
2018
Aug
;
13
(
1
):
56
.
[PubMed]
1746-1596
31.
Ogura
K
,
Hosoda
F
,
Arai
Y
,
Nakamura
H
,
Hama
N
,
Totoki
Y
, et al
Integrated genetic and epigenetic analysis of myxofibrosarcoma
.
Nat Commun
.
2018
Jul
;
9
(
1
):
2765
.
[PubMed]
2041-1723
32.
Lv
PC
,
Wang
ZC
,
Zhu
HL
.
Recent Advances in the Design and Synthesis of c-Met Inhibitors as Anticancer Agents (2014-Present)
.
Curr Med Chem
.
2017
;
24
(
1
):
57
64
.
[PubMed]
0929-8673
33.
Kim
NA
,
Hong
S
,
Kim
KH
,
Choi
DH
,
Kim
JS
,
Park
KE
, et al
New Preclinical Development of a c-Met Inhibitor and Its Combined Anti-Tumor Effect in c-Met-Amplified NSCLC
.
Pharmaceutics
.
2020
Feb
;
12
(
2
):
E121
.
[PubMed]
1999-4923
34.
Comoglio
PM
,
Giordano
S
,
Trusolino
L
.
Drug development of MET inhibitors: targeting oncogene addiction and expedience
.
Nat Rev Drug Discov
.
2008
Jun
;
7
(
6
):
504
16
.
[PubMed]
1474-1776
35.
Cui
JJ
,
Tran-Dubé
M
,
Shen
H
,
Nambu
M
,
Kung
PP
,
Pairish
M
, et al
Structure based drug design of crizotinib (PF-02341066), a potent and selective dual inhibitor of mesenchymal-epithelial transition factor (c-MET) kinase and anaplastic lymphoma kinase (ALK)
.
J Med Chem
.
2011
Sep
;
54
(
18
):
6342
63
.
[PubMed]
0022-2623
36.
Song
EK
,
Tai
WM
,
Messersmith
WA
,
Bagby
S
,
Purkey
A
,
Quackenbush
KS
, et al
Potent antitumor activity of cabozantinib, a c-MET and VEGFR2 inhibitor, in a colorectal cancer patient-derived tumor explant model
.
Int J Cancer
.
2015
Apr
;
136
(
8
):
1967
75
.
[PubMed]
0020-7136
37.
Ding
L
,
Getz
G
,
Wheeler
DA
,
Mardis
ER
,
McLellan
MD
,
Cibulskis
K
, et al
Somatic mutations affect key pathways in lung adenocarcinoma
.
Nature
.
2008
Oct
;
455
(
7216
):
1069
75
.
[PubMed]
0028-0836
38.
Barretina
J
,
Taylor
BS
,
Banerji
S
,
Ramos
AH
,
Lagos-Quintana
M
,
Decarolis
PL
, et al
Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy
.
Nat Genet
.
2010
Aug
;
42
(
8
):
715
21
.
[PubMed]
1061-4036
39.
Arcila
ME
,
Drilon
A
,
Sylvester
BE
,
Lovly
CM
,
Borsu
L
,
Reva
B
, et al
MAP2K1 (MEK1) Mutations Define a Distinct Subset of Lung Adenocarcinoma Associated with Smoking
.
Clin Cancer Res
.
2015
Apr
;
21
(
8
):
1935
43
.
[PubMed]
1078-0432
40.
Orian-Rousseau
V
,
Chen
L
,
Sleeman
JP
,
Herrlich
P
,
Ponta
H
.
CD44 is required for two consecutive steps in HGF/c-Met signaling
.
Genes Dev
.
2002
Dec
;
16
(
23
):
3074
86
.
[PubMed]
0890-9369
41.
Li
Y
,
Heldin
P
.
Hyaluronan production increases the malignant properties of mesothelioma cells
.
Br J Cancer
.
2001
Aug
;
85
(
4
):
600
7
.
[PubMed]
0007-0920
42.
Washimi
O
,
Ueda
R
,
Ariyoshi
Y
,
Suyama
M
,
Seki
T
,
Takahashi
T
, et al
Expression of CD44 variant isoforms in normal and neoplastic cells of the lung
.
Jpn J Cancer Res
.
1994
Nov
;
85
(
11
):
1112
6
.
[PubMed]
0910-5050
43.
Henderson
T
,
Chen
M
,
Darrow
MA
,
Li
CS
,
Chiu
CL
,
Monjazeb
AM
, et al
Alterations in cancer stem-cell marker CD44 expression predict oncologic outcome in soft-tissue sarcomas
.
J Surg Res
.
2018
Mar
;
223
:
207
14
.
[PubMed]
0022-4804
44.
Peiper
M
,
Sato
T
,
Zurakowski
D
,
Eisenberger
C
,
Heinecke
A
,
Hosch
S
, et al
CD44s expression is associated with improved survival in soft tissue sarcoma
.
Anticancer Res
.
2004
Mar-Apr
;
24
2C
:
1053
6
.
[PubMed]
0250-7005
45.
Matuschek
C
,
Lehnhardt
M
,
Gerber
PA
,
Poremba
C
,
Hamilton
J
,
Lammering
G
, et al
Increased CD44s and decreased CD44v6 RNA expression are associated with better survival in myxofibrosarcoma patients: a pilot study
.
Eur J Med Res
.
2014
Feb
;
19
(
1
):
6
.
[PubMed]
0949-2321
46.
Yang
K
,
Tang
Y
,
Habermehl
GK
,
Iczkowski
KA
.
Stable alterations of CD44 isoform expression in prostate cancer cells decrease invasion and growth and alter ligand binding and chemosensitivity
.
BMC Cancer
.
2010
Jan
;
10
(
1
):
16
.
[PubMed]
1471-2407
47.
Tsuchie
H
,
Emori
M
,
Miyakoshi
N
,
Nagasawa
H
,
Okada
K
,
Nanjyo
H
, et al
Prognostic Impact of CD44 Expression in Patients With Myxofibrosarcoma
.
In Vivo
.
2019
Nov-Dec
;
33
(
6
):
2095
102
.
[PubMed]
0258-851X
48.
McClatchey
AI
,
Fehon
RG
.
Merlin and the ERM proteins—regulators of receptor distribution and signaling at the cell cortex
.
Trends Cell Biol
.
2009
May
;
19
(
5
):
198
206
.
[PubMed]
0962-8924
49.
Bruce
B
,
Khanna
G
,
Ren
L
,
Landberg
G
,
Jirström
K
,
Powell
C
, et al
Expression of the cytoskeleton linker protein ezrin in human cancers
.
Clin Exp Metastasis
.
2007
;
24
(
2
):
69
78
.
[PubMed]
0262-0898
50.
Yu
Y
,
Khan
J
,
Khanna
C
,
Helman
L
,
Meltzer
PS
,
Merlino
G
.
Expression profiling identifies the cytoskeletal organizer ezrin and the developmental homeoprotein Six-1 as key metastatic regulators
.
Nat Med
.
2004
Feb
;
10
(
2
):
175
81
.
[PubMed]
1078-8956
51.
Khanna
C
,
Wan
X
,
Bose
S
,
Cassaday
R
,
Olomu
O
,
Mendoza
A
, et al
The membrane-cytoskeleton linker ezrin is necessary for osteosarcoma metastasis
.
Nat Med
.
2004
Feb
;
10
(
2
):
182
6
.
[PubMed]
1078-8956
52.
Huang
HY
,
Li
CF
,
Fang
FM
,
Tsai
JW
,
Li
SH
,
Lee
YT
, et al
Prognostic implication of ezrin overexpression in myxofibrosarcomas
.
Ann Surg Oncol
.
2010
Dec
;
17
(
12
):
3212
9
.
[PubMed]
1068-9265
53.
Krause
AK
,
Hinrichs
SH
,
Orndal
C
,
DeBoer
J
,
Neff
JR
,
Bridge
JA
.
Characterization of a human myxoid malignant fibrous histiocytoma cell line, OH931
.
Cancer Genet Cytogenet
.
1997
Apr
;
94
(
2
):
138
43
.
[PubMed]
0165-4608
54.
Kawashima
H
,
Ogose
A
,
Gu
W
,
Nishio
J
,
Kudo
N
,
Kondo
N
, et al
Establishment and characterization of a novel myxofibrosarcoma cell line
.
Cancer Genet Cytogenet
.
2005
Aug
;
161
(
1
):
28
35
.
[PubMed]
0165-4608
55.
Zhou
M
,
Chinnaiyan
AM
,
Kleer
CG
,
Lucas
PC
,
Rubin
MA
.
Alpha-Methylacyl-CoA racemase: a novel tumor marker over-expressed in several human cancers and their precursor lesions
.
Am J Surg Pathol
.
2002
Jul
;
26
(
7
):
926
31
.
[PubMed]
0147-5185
56.
Langner
C
,
Rupar
G
,
Leibl
S
,
Hutterer
G
,
Chromecki
T
,
Hoefler
G
, et al
Alpha-methylacyl-CoA racemase (AMACR/P504S) protein expression in urothelial carcinoma of the upper urinary tract correlates with tumour progression
.
Virchows Arch
.
2006
Mar
;
448
(
3
):
325
30
.
[PubMed]
0945-6317
57.
Wu
LC
,
Chen
LT
,
Tsai
YJ
,
Lin
CM
,
Lin
CY
,
Tian
YF
, et al
Alpha-methylacyl coenzyme A racemase overexpression in gallbladder carcinoma confers an independent prognostic indicator
.
J Clin Pathol
.
2012
Apr
;
65
(
4
):
309
14
.
[PubMed]
0021-9746
58.
Li
CF
,
Fang
FM
,
Lan
J
,
Wang
JW
,
Kung
HJ
,
Chen
LT
, et al
AMACR amplification in myxofibrosarcomas: a mechanism of overexpression that promotes cell proliferation with therapeutic relevance
.
Clin Cancer Res
.
2014
Dec
;
20
(
23
):
6141
52
.
[PubMed]
1078-0432
59.
Wilson
BA
,
Wang
H
,
Nacev
BA
,
Mease
RC
,
Liu
JO
,
Pomper
MG
, et al
High-throughput screen identifies novel inhibitors of cancer biomarker α-methylacyl coenzyme A racemase (AMACR/P504S)
.
Mol Cancer Ther
.
2011
May
;
10
(
5
):
825
38
.
[PubMed]
1535-7163
60.
Lee
SY
,
Obata
Y
,
Yoshida
M
,
Stockert
E
,
Williamson
B
,
Jungbluth
AA
, et al
Immunomic analysis of human sarcoma
.
Proc Natl Acad Sci USA
.
2003
Mar
;
100
(
5
):
2651
6
.
[PubMed]
0027-8424
61.
Conley
AP
,
Wang
WL
,
Livingston
JA
,
Ravi
V
,
Tsai
JW
,
Ali
A
, et al
MAGE-A3 is a Clinically Relevant Target in Undifferentiated Pleomorphic Sarcoma/Myxofibrosarcoma
.
Cancers (Basel)
.
2019
May
;
11
(
5
):
E677
.
[PubMed]
2072-6694
62.
Groisberg
R
,
Roszik
J
,
Conley
A
,
Patel
SR
,
Subbiah
V
.
The Role of Next-Generation Sequencing in Sarcomas: Evolution From Light Microscope to Molecular Microscope
.
Curr Oncol Rep
.
2017
Oct
;
19
(
12
):
78
.
[PubMed]
1523-3790
63.
Barretina
J
,
Caponigro
G
,
Stransky
N
,
Venkatesan
K
,
Margolin
AA
,
Kim
S
, et al
The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity
.
Nature
.
2012
Mar
;
483
(
7391
):
603
7
.
[PubMed]
0028-0836
64.
Lu
YC
,
Parker
LL
,
Lu
T
,
Zheng
Z
,
Toomey
MA
,
White
DE
, et al
Treatment of Patients With Metastatic Cancer Using a Major Histocompatibility Complex Class II-Restricted T-Cell Receptor Targeting the Cancer Germline Antigen MAGE-A3
.
J Clin Oncol
.
2017
Oct
;
35
(
29
):
3322
9
.
[PubMed]
0732-183X
65.
Ross
JS
,
Wang
K
,
Chmielecki
J
,
Gay
L
,
Johnson
A
,
Chudnovsky
J
, et al
The distribution of BRAF gene fusions in solid tumors and response to targeted therapy
.
Int J Cancer
.
2016
Feb
;
138
(
4
):
881
90
.
[PubMed]
0020-7136
66.
Borden
EC
,
Baker
LH
,
Bell
RS
,
Bramwell
V
,
Demetri
GD
,
Eisenberg
BL
, et al
Soft tissue sarcomas of adults: state of the translational science
.
Clin Cancer Res
.
2003
Jun
;
9
(
6
):
1941
56
.
[PubMed]
1078-0432
67.
Balko
JM
,
Schwarz
LJ
,
Luo
N
,
Estrada
MV
,
Giltnane
JM
,
Dávila-González
D
, et al
Triple-negative breast cancers with amplification of JAK2 at the 9p24 locus demonstrate JAK2-specific dependence
.
Sci Transl Med
.
2016
Apr
;
8
(
334
):
334ra53
.
[PubMed]
1946-6234
68.
Xiang
Z
,
Zhao
Y
,
Mitaksov
V
,
Fremont
DH
,
Kasai
Y
,
Molitoris
A
, et al
Identification of somatic JAK1 mutations in patients with acute myeloid leukemia
.
Blood
.
2008
May
;
111
(
9
):
4809
12
.
[PubMed]
0006-4971
69.
Chan
CH
,
Lee
SW
,
Li
CF
,
Wang
J
,
Yang
WL
,
Wu
CY
, et al
Deciphering the transcriptional complex critical for RhoA gene expression and cancer metastasis
.
Nat Cell Biol
.
2010
May
;
12
(
5
):
457
67
.
[PubMed]
1465-7392
70.
Wang
XC
,
Wu
YP
,
Ye
B
,
Lin
DC
,
Feng
YB
,
Zhang
ZQ
, et al
Suppression of anoikis by SKP2 amplification and overexpression promotes metastasis of esophageal squamous cell carcinoma
.
Mol Cancer Res
.
2009
Jan
;
7
(
1
):
12
22
.
[PubMed]
1541-7786
71.
Li
CF
,
Wang
JM
,
Kang
HY
,
Huang
CK
,
Wang
JW
,
Fang
FM
, et al
Characterization of gene amplification-driven SKP2 overexpression in myxofibrosarcoma: potential implications in tumor progression and therapeutics
.
Clin Cancer Res
.
2012
Mar
;
18
(
6
):
1598
610
.
[PubMed]
1078-0432
72.
Huang
HY
,
Kang
HY
,
Li
CF
,
Eng
HL
,
Chou
SC
,
Lin
CN
, et al
Skp2 overexpression is highly representative of intrinsic biological aggressiveness and independently associated with poor prognosis in primary localized myxofibrosarcomas
.
Clin Cancer Res
.
2006
Jan
;
12
(
2
):
487
98
.
[PubMed]
1078-0432
73.
Uddin
S
,
Ahmed
M
,
Hussain
AR
,
Jehan
Z
,
Al-Dayel
F
,
Munkarah
A
, et al
Bortezomib-mediated expression of p27Kip1 through S-phase kinase protein 2 degradation in epithelial ovarian cancer
.
Lab Invest
.
2009
Oct
;
89
(
10
):
1115
27
.
[PubMed]
0023-6837
74.
Wang
Y
,
Rishi
AK
,
Puliyappadamba
VT
,
Sharma
S
,
Yang
H
,
Tarca
A
, et al
Targeted proteasome inhibition by Velcade induces apoptosis in human mesothelioma and breast cancer cell lines
.
Cancer Chemother Pharmacol
.
2010
Aug
;
66
(
3
):
455
66
.
[PubMed]
0344-5704
75.
Wright
JJ
.
Combination therapy of bortezomib with novel targeted agents: an emerging treatment strategy
.
Clin Cancer Res
.
2010
Aug
;
16
(
16
):
4094
104
.
[PubMed]
1078-0432
76.
Bizet
AA
,
Liu
K
,
Tran-Khanh
N
,
Saksena
A
,
Vorstenbosch
J
,
Finnson
KW
, et al
The TGF-β co-receptor, CD109, promotes internalization and degradation of TGF-β receptors
.
Biochim Biophys Acta
.
2011
May
;
1813
(
5
):
742
53
.
[PubMed]
0006-3002
77.
Pickup
M
,
Novitskiy
S
,
Moses
HL
.
The roles of TGFβ in the tumour microenvironment
.
Nat Rev Cancer
.
2013
Nov
;
13
(
11
):
788
99
.
[PubMed]
1474-175X
78.
Oshimori
N
,
Oristian
D
,
Fuchs
E
.
TGF-β promotes heterogeneity and drug resistance in squamous cell carcinoma
.
Cell
.
2015
Feb
;
160
(
5
):
963
76
.
[PubMed]
0092-8674
79.
Bhola
NE
,
Balko
JM
,
Dugger
TC
,
Kuba
MG
,
Sánchez
V
,
Sanders
M
, et al
TGF-β inhibition enhances chemotherapy action against triple-negative breast cancer
.
J Clin Invest
.
2013
Mar
;
123
(
3
):
1348
58
.
[PubMed]
0021-9738
80.
Yegodayev
KM
,
Novoplansky
O
,
Golden
A
,
Prasad
M
,
Levin
L
,
Jagadeeshan
S
, et al
TGF-Beta-Activated Cancer-Associated Fibroblasts Limit Cetuximab Efficacy in Preclinical Models of Head and Neck Cancer
.
Cancers (Basel)
.
2020
Feb
;
12
(
2
):
E339
.
[PubMed]
2072-6694
81.
Emori
M
,
Tsukahara
T
,
Murase
M
,
Kano
M
,
Murata
K
,
Takahashi
A
, et al
High expression of CD109 antigen regulates the phenotype of cancer stem-like cells/cancer-initiating cells in the novel epithelioid sarcoma cell line ESX and is related to poor prognosis of soft tissue sarcoma
.
PLoS One
.
2013
Dec
;
8
(
12
):
e84187
.
[PubMed]
1932-6203
82.
Hasegawa
M
,
Moritani
S
,
Murakumo
Y
,
Sato
T
,
Hagiwara
S
,
Suzuki
C
, et al
CD109 expression in basal-like breast carcinoma
.
Pathol Int
.
2008
May
;
58
(
5
):
288
94
.
[PubMed]
1320-5463
83.
Emori
M
,
Tsukahara
T
,
Murata
K
,
Sugita
S
,
Sonoda
T
,
Kaya
M
, et al
Prognostic impact of CD109 expression in myxofibrosarcoma
.
J Surg Oncol
.
2015
Jun
;
111
(
8
):
975
9
.
[PubMed]
0022-4790
84.
De Vita
A
,
Recine
F
,
Mercatali
L
,
Miserocchi
G
,
Liverani
C
,
Spadazzi
C
, et al
Myxofibrosarcoma primary cultures: molecular and pharmacological profile
.
Ther Adv Med Oncol
.
2017
Dec
;
9
(
12
):
755
67
.
[PubMed]
1758-8340
85.
Neuzillet
C
,
Tijeras-Raballand
A
,
Cohen
R
,
Cros
J
,
Faivre
S
,
Raymond
E
, et al
Targeting the TGFβ pathway for cancer therapy
.
Pharmacol Ther
.
2015
Mar
;
147
:
22
31
.
[PubMed]
0163-7258
86.
Penn
I
.
Sarcomas in organ allograft recipients
.
Transplantation
.
1995
Dec
;
60
(
12
):
1485
91
.
[PubMed]
0041-1337
87.
Bertucci
F
,
Finetti
P
,
Perrot
D
,
Leroux
A
,
Collin
F
,
Le Cesne
A
, et al
PDL1 expression is a poor-prognosis factor in soft-tissue sarcomas
.
OncoImmunology
.
2017
Feb
;
6
(
3
):
e1278100
.
[PubMed]
2162-4011
88.
Pollack
SM
,
He
Q
,
Yearley
JH
,
Emerson
R
,
Vignali
M
,
Zhang
Y
, 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
Sep
;
123
(
17
):
3291
304
.
[PubMed]
0008-543X
89.
Keung
EZ
,
Lazar
AJ
,
Torres
KE
,
Wang
WL
,
Cormier
JN
,
Ashleigh Guadagnolo
B
, et al
Phase II study of neoadjuvant checkpoint blockade in patients with surgically resectable undifferentiated pleomorphic sarcoma and dedifferentiated liposarcoma
.
BMC Cancer
.
2018
Sep
;
18
(
1
):
913
.
[PubMed]
1471-2407
90.
Kelly
CM
,
Antonescu
CR
,
Bowler
T
,
Munhoz
R
,
Chi
P
,
Dickson
MA
, et al
Objective Response Rate Among Patients With Locally Advanced or Metastatic Sarcoma Treated With Talimogene Laherparepvec in Combination With Pembrolizumab: A Phase 2 Clinical Trial
.
JAMA Oncol
.
2020
Jan
;
6
(
3
):
402
.
[PubMed]
2374-2437
91.
Tawbi
HA
,
Burgess
M
,
Bolejack
V
,
Van Tine
BA
,
Schuetze
SM
,
Hu
J
, et al
Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial
.
Lancet Oncol
.
2017
Nov
;
18
(
11
):
1493
501
.
[PubMed]
1470-2045
92.
D’Angelo
SP
,
Mahoney
MR
,
Van Tine
BA
,
Atkins
J
,
Milhem
MM
,
Jahagirdar
BN
, et al
Nivolumab with or without ipilimumab treatment for metastatic sarcoma (Alliance A091401): two open-label, non-comparative, randomised, phase 2 trials
.
Lancet Oncol
.
2018
Mar
;
19
(
3
):
416
26
.
[PubMed]
1470-2045
93.
Gibney
GT
,
Weiner
LM
,
Atkins
MB
.
Predictive biomarkers for checkpoint inhibitor-based immunotherapy
.
Lancet Oncol
.
2016
Dec
;
17
(
12
):
e542
51
.
[PubMed]
1470-2045
94.
Mariathasan
S
,
Turley
SJ
,
Nickles
D
,
Castiglioni
A
,
Yuen
K
,
Wang
Y
, et al
TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells
.
Nature
.
2018
Feb
;
554
(
7693
):
544
8
.
[PubMed]
0028-0836
95.
Tauriello
DV
,
Palomo-Ponce
S
,
Stork
D
,
Berenguer-Llergo
A
,
Badia-Ramentol
J
,
Iglesias
M
, et al
TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis
.
Nature
.
2018
Feb
;
554
(
7693
):
538
43
.
[PubMed]
0028-0836
96.
Ott
PA
,
Hodi
FS
,
Kaufman
HL
,
Wigginton
JM
,
Wolchok
JD
.
Combination immunotherapy: a road map
.
J Immunother Cancer
.
2017
Feb
;
5
(
1
):
16
.
[PubMed]
2051-1426
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.