ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Pathogenetic factors in soft tissue and bone sarcomas

Pathogenetic factors in soft tissue and bone sarcomas
Literature review current through: Jan 2024.
This topic last updated: Jun 26, 2023.

INTRODUCTION — Sarcomas are malignant tumors arising from skeletal and extraskeletal connective tissues, including the peripheral nervous system. Approximately 76 percent arise in soft tissue, the remainder in bone.

There is no clearly defined etiology in most cases of soft tissue sarcoma, but a number of associated or predisposing factors have been identified [1]. These include a genetic predisposition, gene mutations, radiation therapy (RT), chemotherapy, chemical carcinogens, chronic irritation, and lymphedema. In addition, an association between viral infection and sarcoma has been shown for human herpesvirus 8 (HHV-8) in Kaposi sarcoma, and for Epstein-Barr virus (EBV) and smooth muscle tumors in immunocompromised patients.

GENETIC PREDISPOSITION — Some patients with bone and soft tissue sarcomas, particularly children, have a genetic predisposition to cancer [1-5]. In some cases, individuals are from families with a defined inherited predisposing condition, such as Li-Fraumeni syndrome (LFS) or retinoblastoma, but many cases do not fit recognized inherited cancer syndromes. One analysis, in which 1162 patients with sarcoma, unselected for family history, underwent targeted exon sequencing of 72 genes selected for their association with cancer risk, concluded that approximately one-half of the patients had putatively pathogenic, monogenic, and polygenic variation in known and novel cancer genes [6]. In a pooled analysis of all sarcoma probands, 240 carried multiple variants, suggesting a polygenic contribution to sarcoma risk. Only 155 (17 percent) of the 911 families with informative pedigrees fit recognizable cancer syndromes.

The major genetic syndromes are briefly outlined below.

Li-Fraumeni syndrome — Mutations in TP53 are the most common germline mutations that predispose to pediatric sarcomas, including osteosarcoma, undifferentiated pleomorphic sarcoma, rhabdomyosarcoma, leiomyosarcoma, and liposarcoma [5]. As many as 7 percent of children with soft tissue sarcomas may have LFS [7]. In a series of 151 children with soft tissue sarcomas, for example, five of the families (3.3 percent) manifested the classic LFS familial cancer syndrome, 10 (6.6 percent) had features consistent with the syndrome, and 16 (10.5 percent) had one parent with a possible hereditary cancer syndrome or with cancer before the age of 60 [4].

The LFS is inherited as an autosomal dominant trait, and a germline mutation in the TP53 tumor suppressor gene is found in most affected families [8-10]. LFS is primarily characterized by soft tissue and bone sarcomas and breast cancer; other features include brain tumors, leukemia, and adrenocortical carcinoma occurring before the age of 45. The cumulative case risk among affected individuals is 100 percent by age 70 [11]. Sarcomas account for 25 to 33 percent of tumors in affected individuals, and they arise at a younger age than those unassociated with LFS [11,12]. In one series of individuals from the International Agency for Research on Cancer (IARC) database, 96 percent of the sarcomas arising in individuals with LFS arose before the age of 50, compared with 38 percent before 50 years of age in the general population [12]. (See 'TP53 gene' below and "Li-Fraumeni syndrome".)

Familial adenomatous polyposis and Gardner syndrome — There is a high frequency of intra-abdominal desmoid fibromatosis (also called desmoid tumors) among patients with familial adenomatous polyposis (FAP); this combination is also known as Gardner syndrome (see "Desmoid tumors: Epidemiology, molecular pathogenesis, clinical presentation, diagnosis, and local therapy") [13]. This disorder is characterized by mutations in the APC (adenomatous polyposis coli) gene. (See "Gardner syndrome".)

Retinoblastoma — Sarcomas of soft tissue and bone, particularly osteosarcoma, develop later in life in surviving patients with retinoblastoma (RB), particularly of the familial or bilateral type, in which individuals inherit a mutant copy of the RB gene [14-16]. The gene itself appears to predispose to second cancers of bone and soft tissue, with radiation therapy (RT) shortening the latent period and increasing the risk [16,17].

These relationships were illustrated in a longitudinal study of 1601 patients with RB (963 with the inherited type) [15,17,18]. The cumulative frequency of a second cancer at 50 years after diagnosis was significantly different based on whether the patient had the genetic form of RB (bilateral tumors or unilateral disease with a positive family history) versus nongenetic form of RB (ie, unilateral disease in the absence of a positive family history, due to a somatic mutation; 36 versus 6 percent), and whether the second tumor was associated with RT or not (38 versus 21 percent, respectively) [15]. More than 60 percent of the cancers were sarcomas.

A later publication provided a detailed description of 69 secondary soft tissue sarcomas among 68 children with hereditary RB [17]. Almost all of the sarcomas (n = 66) developed in children who had received orbital RT, but 18 did not arise within the irradiated field. The predominant histology was leiomyosarcoma (33 percent), followed by fibrosarcoma, undifferentiated pleomorphic sarcoma (UPS), soft tissue tumors/sarcomas not otherwise specified, rhabdomyosarcoma, and liposarcoma (19, 17, 15, 12, and 4 percent of cases, respectively).

There were some major differences between histologic types. Leiomyosarcomas were diagnosed more often outside of the RT field than within it (14 of 23), suggesting that genetic predisposition may have played a more important role than RT. Leiomyosarcoma was also the only histology for which risk was significantly higher among patients receiving chemotherapy plus RT versus RT alone. Furthermore, these were also late developing tumors; 18 were diagnosed more than 30 years after diagnosis (ie, at the same age as individuals in the general population).

In contrast, seven of eight secondary rhabdomyosarcomas occurred within the RT field, and seven developed within 20 years of treatment, during a normal growth period.

These data underscore the need for long-term surveillance for second cancers in RB survivors, particularly for those with the bilateral or inherited form of the disease. (See "Retinoblastoma: Treatment and outcome", section on 'Long-term follow-up'.)

Neurofibromatosis — Some of the multiple benign neurofibromas of von Recklinghausen disease (type 1 neurofibromatosis, NF1) can undergo malignant change to malignant peripheral nerve sheath tumors (MPNSTs (image 1)). The karyotypic changes seen in MPNSTs are typically complex [19,20].

NF1 is associated with a mutation in the NF1 gene; it has been proposed that malignant degeneration reflects the two-hit hypothesis in which one allele is constitutionally inactivated in the germline while the other allele undergoes somatic inactivation (the second hit). (See "Neurofibromatosis type 1 (NF1): Pathogenesis, clinical features, and diagnosis".)

Among nonneurogenic sarcomas, rhabdomyosarcomas also tend to arise more often in NF1 patients than in the general population. In one series, the risk of developing a malignant peripheral nerve sheath tumor among patients with NF1 was 4.6 percent, compared with 0.001 percent among the general population [21]. (See "Rhabdomyosarcoma in childhood and adolescence: Epidemiology, pathology, and molecular pathogenesis", section on 'Inherited syndromes'.)

Other — Rothmund-Thomson syndrome (poikiloderma congenitale) is an autosomal recessive condition characterized by a distinctive skin finding (atrophy, telangiectasias, pigmentation), sparse hair, cataracts, small stature, skeletal anomalies, and a significantly increased risk for osteosarcoma. (See "Osteosarcoma: Epidemiology, pathology, clinical presentation, and diagnosis", section on 'Genetic conditions'.)

GENETICS AND MOLECULAR PATHOGENESIS — The genetics of sarcomas segregate into two major types: those with specific genetic alterations and usually simple karyotypes, including fusion genes due to reciprocal translocations (eg, PAX3-FOXO1A in alveolar rhabdomyosarcomas (table 1)) or specific point mutations (eg, KIT mutations in gastrointestinal stromal tumors [GIST]), and those with nonspecific genetic alterations and complex, unbalanced karyotypes, reflected by numerous genetic losses and gains (eg, osteosarcoma, undifferentiated pleomorphic sarcoma [UPS], angiosarcoma, leiomyosarcoma).

Fusion gene-related sarcomas account for approximately one-third of all sarcomas [22]. In many cases, the aberrant protein product of the fused gene acts as an abnormal transcriptional regulator, thus providing the molecular basis for oncogenesis. Just as specific mutations in KIT impact the diagnosis and response to therapy in GIST, for many fusion gene-related sarcomas, molecular testing to identify specific translocations that are unique to certain subtypes of sarcoma can be used for diagnostic refinement, prognostic assessment, or both [23]. (See 'Chromosomal translocations' below and "Epidemiology, pathology, and molecular genetics of Ewing sarcoma", section on 'Molecular diagnostics' and "Dermatofibrosarcoma protuberans: Epidemiology, pathogenesis, clinical presentation, diagnosis, and staging", section on 'Molecular diagnostics' and "Clinical presentation, diagnosis, and prognosis of gastrointestinal stromal tumors", section on 'Pathogenesis'.)

In contrast, the molecular pathogenesis of sarcomas with unbalanced karyotypic complexity has not been definitively proven, but inactivation of the p53 pathway appears to be a main differentiating feature between these tumors and those with simple genetic alterations [24]. Among the mechanisms for p53 pathway inactivation are TP53 point mutations, homozygous deletion in CDKN2A, and MDM2 amplification. (See 'Somatic gene mutations' below.)

Somatic gene mutations — There is a high incidence of acquired (somatic) gene alterations in soft tissue and bone sarcomas. In one study of tumor biopsies of soft tissue sarcomas, for example, the concurrent application of standard cytogenetics and DNA ploidy by flow cytometry identified an abnormal cell population by one or both methods in 84 percent of the tumors [25]. This discussion will focus on those mutations that are seen in soft tissue sarcomas. The degree to which mutations affect prognosis in soft tissue sarcomas, and the molecular pathogenesis of bone sarcomas, are discussed elsewhere. (See "Osteosarcoma: Epidemiology, pathology, clinical presentation, and diagnosis", section on 'Risk factors' and "Epidemiology, pathology, and molecular genetics of Ewing sarcoma", section on 'Molecular genetics' and "Clinical presentation, histopathology, diagnostic evaluation, and staging of soft tissue sarcoma", section on 'Introduction'.)

Somatic mutations in specific genes vary across the histologic subtypes of soft tissue sarcoma. One study performed DNA sequencing of 722 protein-coding genes that have been implicated in cancer in 200 samples of soft tissue sarcomas encompassing seven distinct histologic subtypes [26]. Frequently mutated genes included the TP53 tumor suppressor gene (17 percent of pleomorphic liposarcomas), type 1 NF1 (10.5 percent of myxofibrosarcomas and 8 percent of pleomorphic liposarcomas), and PIK3CA (18 percent of myxoid liposarcomas). The retinoblastoma (RB1) gene was mutated in 4 percent of pleomorphic liposarcomas. DNA amplifications of chromosome 12q were observed in 90 percent of patients with dedifferentiated liposarcomas. Genes amplified in the 12q13-15 amplicon in well differentiated and dedifferentiated liposarcomas include MDM2, HMGA2, YEATS4, CDK4, and SAS [27].

Whole-genome sequencing has been performed on childhood rhabdomyosarcomas. One study analyzed 16 rhabdomyosarcoma tumors from 13 patients and compared the results with matched normal tissue [28]. Another study analyzed 44 rhabdomyosarcoma tumors and matched normal leukocyte DNA [29]. These studies provide a comprehensive genomic analysis that reveals the landscape of gene mutations in rhabdomyosarcoma. These studies show that rhabdomyosarcomas fall into two broad categories. The first group includes alveolar rhabdomyosarcomas that contain a PAX3 or PAX7 translocation without other recurrent cancer mutations. The second group is embryonal rhabdomyosarcomas, which are characterized by mutations in the receptor tyrosine kinase, RAS, PIK3CA axis. Interestingly, mutations in RAS or NF1, which negatively regulates Ras, were significantly associated with more aggressive tumors (ie, intermediate and high-risk rhabdomyosarcoma). Therefore, the RAS is a critical driver mutation for embryonal rhabdomyosarcoma and a potential therapeutic target [30].

Mutations in the KIT protooncogene, which are associated with GISTs, are discussed elsewhere. (See "Rhabdomyosarcoma in childhood and adolescence: Epidemiology, pathology, and molecular pathogenesis" and "Clinical presentation, diagnosis, and prognosis of gastrointestinal stromal tumors".)

NF1 gene — The NF1 gene encodes the neurofibromin protein that functions as a Ras-GTPase to suppress the Ras pathway [31,32]. Loss of NF1 expression, which occurs in malignant peripheral nerve sheath tumors (MPNSTs) in patients with neurofibromatosis type I and in a variety of other sporadic soft tissue sarcomas [26], activates Ras signaling. Most MPNSTs also have loss-of-function mutations in subunits of the histone-modifying PRC2 complex (SUZ12 or EED), which amplify Ras-driven transcription [33,34].

PIKC3A gene — The PI3KCA gene encodes for the catalytic subunit of phosphatidylinositol 3-kinase (PI3K), which phosphorylates lipids to create second messengers that regulate cell proliferation, survival, and motility. Mutations of PIK3CA in myxoid liposarcomas are associated with a shorter duration of disease-specific survival [26].

TP53 gene — The p53 protein is a transcriptional activator that plays a key role in the integration of signals inducing cell division, arrest of DNA synthesis following DNA damage, and programmed cell death (apoptosis). DNA damage results in increased levels of p53 protein, which induces cell cycle arrest at the G1/S interface, thereby permitting the cell to repair genomic damage or to initiate apoptosis [35-37]. The TP53 gene also functions as a tumor suppressor gene, and alterations are the most commonly detected mutations in a diverse group of malignancies. The wild-type p53 in normal tissue has a short half-life and is not detectable by immunohistochemical methods; in comparison, mutations of the gene result in a stabilized p53 protein, which accumulates in the cell and often becomes detectable by immunohistochemistry.

As noted above, germline mutations in the TP53 gene are present in most families with the Li-Fraumeni syndrome [8-10]. (See "Li-Fraumeni syndrome".)

Germline mutations in this gene also may occur in other patients with soft tissue sarcoma, particularly those with other cancers that are not considered indicative of the Li-Fraumeni syndrome [38-40]. In one study of patients with sarcoma, for example, germline TP53 mutations were relatively common (5 of 15) in those with multiple primary cancers or a positive family history of cancer but rare (3 of 185) in those without such a history [38]. Germline mutations in TP53 also may predispose to sarcoma formation in NF1 [41].

Somatic mutations in the TP53 gene are the most frequently detected molecular alteration in sporadic soft tissue sarcomas. These mutations have been detected in a variety of soft tissue sarcomas including UPS, leiomyosarcoma, liposarcoma, and rhabdomyosarcoma [42-51].

TP53 gene mutations are present in approximately one-third of tumors [42,44,45]. In one series of 127 bone and soft tissue sarcomas, 42 had somatic alterations in the TP53 gene: one-half were gross rearrangements and one-half were subtle missense or nonsense mutations [42]. Nearly all of the rearrangements and nonsense mutations showed no immunostaining for p53 (the "null pattern"), in contrast to the strong and diffuse pattern observed with pathogenic missense mutations (indicative of increased protein half-life) [43]. Furthermore, there was staining (usually weak) in 29 percent of the cases without detectable alterations in the TP53 gene.

Somatic loss of the TP53 gene appears to be quite variable in soft tissue sarcomas. Some tumors show a loss of both alleles or a loss of one allele with the remaining allele being either normal or with a point mutation [47]. In a case report, a synovial sarcoma metastasis was composed exclusively of cells bearing a distinct TP53 mutation. This same mutation was found as a small clone within the primary tumor, indicating that the metastasis resulted from expansion of a mutant TP53 clone [52].

Animal models are consistent with a pathogenetic role for TP53 defects in sarcoma development, as suggested by the observations:

Mice deficient for p53 expression develop a variety of neoplasms including bone and soft tissue sarcomas [53].

Irradiated transgenic mice harboring mutant TP53 show higher frequencies of sarcomas [54].

Transduction of wild type TP53 genes into soft tissue sarcomas bearing mutated p53 genes restores enhanced cell cycle control and suppresses sarcoma growth [55].

Restoring p53 function in primary sarcomas in mice lacking p53 expression causes sarcoma regression [56].

ATRX gene — Alpha-thalassemia/mental retardation syndrome X-linked (ATRX) is one of the most commonly mutated genes after TP53 in several types of sarcoma, including leiomyosarcoma [57] and osteosarcoma [58]. Loss of expression of ATRX frequently occurs in a number of complex karyotype sarcomas, including angiosarcoma, UPS, and dedifferentiated liposarcoma, which correlates with tumors utilizing the alternative lengthening of telomeres (ALT) as a mechanism to maintain telomeres [59]. ATRX binds to death-associated protein 6 (DAXX) to remodel chromatin. Cells lacking ATRX may be sensitive to inhibitors of the ATR kinase [60].

Retinoblastoma gene — Deletions or mutations of the tumor suppressor RB1 gene are critical in the pathogenesis of retinoblastoma and a variety of solid tumors. Even before the RB1 gene had been identified, it was recognized that some sporadic sarcomas had deletions on chromosome 13 similar to those observed in some patients with retinoblastoma. Alterations in the RB1 gene are common in soft tissue sarcoma, occurring in up to 70 percent of tumors in some series [61-63]. It has been proposed that RB1 alterations are primary events in human sarcomas and may be involved in tumorigenesis or the early phases of tumor progression [62]. The RB1 gene is critical for proper entry into DNA replication and transition through the cell cycle, and mutations are believed to perturb normal cell cycle function. (See "Retinoblastoma: Clinical presentation, evaluation, and diagnosis", section on 'Pathogenesis'.)

MDM2 gene — The MDM2 (murine double minute 2 homolog) gene, located at 12q15, is overexpressed in a variety of human tumors including soft tissue sarcomas [64-67]. Its gene product localizes predominantly to the nucleus, where it acts as an inhibitor of the TP53 tumor suppressor gene product. The MDM2 product functions by concealing the activation domain of the p53 protein, thereby inhibiting TP53 transcriptional activity [68,69].

In a series of 24 soft tissue sarcomas, an alteration in p53 was found in eight tumors and MDM2 amplification in another eight [70]. No tumor contained alterations in both genes, which is consistent with the hypothesis that TP53 and MDM2 genetic alterations are alternative mechanisms for inactivating the same regulatory pathway for suppressing cell growth. In another study, 22 of 211 soft tissue sarcomas showed increased immunoreactivity to both MDM2 and p53 [71]. However, the overexpression of p53 and MDM2 proteins in the nuclei of these cells did not always correlate well with gene amplification at the MDM2 locus or mutation at the TP53 gene.

CDK4 gene — The CDK4 gene, which encodes a cyclin dependent kinase, occurs in an adjacent amplicon at 12q14. Amplification of CDK4 has been found in a variety of sarcomas including well-differentiated and dedifferentiated liposarcomas [27,72,73]. The frequent association with MDM2 has suggested a synergistic effect in opposing p53 function [73]. A role for CDK4 amplification in the pathogenesis of soft tissue sarcomas is suggested by the fact that knockdown of CDK4 in liposarcoma cell lines or treatment of these cells with a CDK4/6 inhibitor inhibits proliferation [27].

Detection of MDM2 and CDK4 overexpression by immunohistochemical staining may be helpful in diagnosing well-differentiated and dedifferentiated liposarcomas [74].

YEATS4 gene — The YEATS4 gene, which is adjacent to MDM2 at 12q15, is amplified along with MDM2 in well-differentiated and dedifferentiated liposarcomas [27]. YEATS4 functions as a transcription factor and negatively regulates p53 function [75]. Therefore, overexpressed YEATS4 may act in concert with amplified MDM2 to block p53 function.

HMGA2 gene — The HMGA2 (high mobility group-AT hook 2) gene is not only amplified with MDM2 in liposarcomas, but also frequently undergoes intragenic or close extragenic rearrangements [27]. The product of the HMGA2 gene is a high-mobility group protein that can accumulate on the chromatin of senescent cells to form senescent associated heterochromatic foci [76]. The ability of HMGA2 to promote senescence (permanent cell cycle arrest) is cancelled by overexpression of MDM2 and CDK4. Overexpression of HMGA2 has been shown to promote anchorage independent growth, a feature of oncogenic transformation [77].

SAS gene — A gene originally called SAS (sarcoma amplified sequence), designated tetraspanin 31 (TSPAN31), is also located at 12q14 and often coamplified with CDK4 in some soft tissue sarcomas [27,78]. SAS encodes a new member of the transmembrane 4 superfamily of proteins that may function in signal transduction and growth control [79]. One study of 12q13-14 amplification in 98 sarcomas found, among others, SAS amplification in 10, MDM2 in nine (eight of which had amplification of SAS), and the DDIT3 gene (formerly known as CHOP or GADD153) in four; it was suggested that the target of these amplifications may be an unidentified gene [80]. (See 'Myxoid liposarcomas' below.)

Chromosomal translocations — As noted above, a number of soft tissue sarcomas display recurrent nonrandom chromosomal translocations, which serve as definitive diagnostic criteria for the tumors in which they occur (table 1) [23].

Furthermore, these chromosomal abnormalities have been characterized at the molecular level, and many of the chimeric genes have been identified as being important to the biology of these tumors, acting as abnormal transcription factors that deregulate the transcription of multiple downstream genes and pathways [24]. Thus, the identification of these molecular signatures has provided clues as to the molecular alterations that are fundamental for the development and often maintenance of sarcoma. Because these translocations generate fusion proteins that are specific to the sarcoma cell, they are attractive targets for sarcoma therapy. A Surveillance, Epidemiology, and End Results (SEER) database study noted an increased subsequent malignant neoplasm (SMN) risk experienced by survivors of sarcoma and demonstrated higher SMN rates in survivors of fusion-negative sarcomas compared with those with a history of fusion-associated sarcomas.

Ewing sarcoma — Approximately 85 to 90 percent of Ewing sarcomas (EWS) display a reciprocal exchange t(11;22)(q24;q12) [81-83]. In this translocation, the EWSR1 gene from chromosome 22q12 is covalently linked to the ETS family member, FLI1 [84]. A less common translocation t(21;22)(q22;q12) has also been identified and links EWSR1 to a different ETS family member, ERG [85]. In one study, t(11;22)(q24;q12) fusion transcripts were identified in 83 of 87 of EWS [83]. In this series, 6 of 15 tumors classified as undifferentiated (ie, not EWS) showed fusion transcripts indicating that they might have to be reclassified as EWS.

Undifferentiated round cell sarcoma that lack an EWSR1-FLI1 or EWSR1-ERG translocation may harbor other translocations that do not include the EWSR1 gene. As an example, a t(4;19)(q35;q13) or t(10;19)(q26;q13) translocation results in fusion of the transcriptional repressor CIC with DUX4, a double homeobox transcription factor [86]. These tumors, which are classified as CIC-rearranged sarcomas [87], characteristically express ETV family transcription factors such as ETV4, along with WT1 [88,89]. CIC-rearranged sarcomas can also result from a t(X;19)(q13;q13.3) translocation that leads to the fusion of CIC and the transcription factor FOXO4 [90].

Furthermore, an intrachromosomal X-chromosome fusion of the BCL6 corepressor (BCOR) gene and CCNB3, which encodes the testis-specific cyclin B3, has been found in a distinct undifferentiated bone and soft tissue tumor (table 1) [91-95].

It is thought that the chimeric proteins that result from these translocations act as transcriptional regulators. (See "Epidemiology, pathology, and molecular genetics of Ewing sarcoma".)

Desmoplastic small round cell tumor — The rare desmoplastic small round cell tumor contains a characteristic translocation t(11;22) EWSR1-WT1, in which the breakpoints are between the Ewing sarcoma breakpoint region 1 (EWSR1) and a second gene on chromosome 11, WT1. This specific translocation differentiates these tumors from Ewing sarcoma and other small round blue cell tumors.

Myxoid liposarcomas — Myxoid liposarcoma (including high-grade examples formerly known as "round cell" liposarcoma) usually harbors the reciprocal translocation t(12;16)(q13;p11) [96,97]. In this translocation, the DDIT3 (DNA damage inducible transcript 3) gene (formerly known as CHOP) is inserted adjacent to an FUS (fused in sarcoma; also known as TLS, translocated in liposarcoma). The fusion gene FUS-DDIT3 is found in 95 percent of myxoid liposarcomas; around 5 percent of tumors harbor an alternate EWSR1-DDIT3 fusion [96-99]. These fusions fail to induce G1/S arrest, which is one of the functions of the nononcogenic form of DDIT3 [100]. Identification of the fusion gene has been used as a diagnostic aid for this type of liposarcoma; immunohistochemistry using an antibody directed against DDIT3 has been developed as a diagnostic tool [98,101,102].

Synovial sarcoma — Synovial sarcomas is characterized by the translocation t(X;18)(p11.2;q11.2) [103]. The breakpoint of this translocation fuses the SS18 (synovial sarcoma translocation, chromosome 18, previously called SYT) gene from chromosome 18 to one of three homologous genes, SSX1, SSX2, and SSX4, on the X chromosome [104-106]. An SS18-SSX fusion-specific antibody has been developed for immunohistochemistry; this antibody is highly sensitive and specific for synovial sarcoma [107].

The SS18 gene encodes for a protein subunit of the mSWI/SNF (BAF) chromatin remodeling complex. The SS18-SSX fusion gene product competes with the endogenous SS18 protein, forming an altered complex lacking the tumor suppressor BAF47 (hSNF5). The altered protein complex binds the SOX2 locus and reverses polycomb-mediated repression, resulting in activation of the transcription factor SOX2, which promotes self-renewal and a stem cell phenotype [108].

SS18-SSX1 is associated with biphasic tumors (glandular epithelial differentiation on a background of spindle tumor cells), while SS18-SSX2 is associated with monophasic tumors that lack glandular epithelial differentiation [109].

Alveolar rhabdomyosarcoma — Alveolar rhabdomyosarcomas (ARMS) have a translocation t(2;13)(q35;q14) or less often t(1;13)(p36;q14); the corresponding chimeric genes are termed PAX3-FOXO1A and PAX7-FOXO1A (formerly known as FKHR) [110,111].

These translocations are associated with overexpression of a fusion product, which is thought to be involved in the pathogenesis of ARMS, although the specific mechanisms are incompletely understood [111-113].

Compared with PAX3-FOXO1A tumors, PAX7-FOXO1A tumors are more often extremity located and localized [114]. Molecular determination of minimal residual disease in ARMS is possible, but the clinical significance of this finding is uncertain [115].

Clear cell sarcoma — Clear cell sarcoma is a translocation-associated soft tissue sarcoma with melanocytic differentiation; this tumor type was previously also known as "melanoma of the soft parts." Although these tumors stain immunohistochemically for melanoma markers, the genetics is entirely different from melanoma.

Cytogenetically, most clear cell sarcomas exhibit a translocation at t(12;22)(q13-14;q12), which is not seen in melanoma [116,117]. This results in the fusion of the EWSR1 with ATF1, a gene encoding a member of the cyclic AMP-responsive element binding protein (CREB) family of transcription factors. A variant translocation (EWSR1-CREB1) is commonly found in clear cell sarcoma-like tumors of the gastrointestinal tract (also known as malignant gastrointestinal neuroectodermal tumor [GNET]); these tumors lack melanocytic differentiation [118]. (See "Uncommon sarcoma subtypes", section on 'Clear cell sarcoma'.)

Alveolar soft part sarcoma — Alveolar soft part sarcomas are highly vascular, rare malignant soft tissue tumors that tend to arise on the extremities of adolescents and young adults, with a female predilection [119]. A characteristic nonreciprocal translocation (derivative or der(17)t(X;17)(p11;q25)) is found in nearly all cases [120,121]. The translocation involves the ASPSCR1 (previously called ASPL) gene on chromosome 17 and the TFE3 gene on the X chromosome. The resulting ASPSCR1-TFE3 fusion gene is postulated to be involved in the pathogenesis of this tumor [122], possibly by upregulating angiogenesis-related genes [123]. It is hypothesized that the female predominance is due to their possession of an extra X chromosome, and the fusion gene not being subject to X-inactivation [119]. (See "Uncommon sarcoma subtypes", section on 'Alveolar soft part sarcoma'.)

Extraskeletal myxoid chondrosarcoma — Extraskeletal myxoid chondrosarcoma (EMC) is a tumor of uncertain histogenesis (despite its name suggesting cartilaginous differentiation) that is characterized by abundant myxoid matrix and often presents in the lower extremities [124,125]. (See "Uncommon sarcoma subtypes", section on 'Extraskeletal myxoid chondrosarcoma'.)

The majority are characterized by a unique reciprocal translocation t(9;22) that results in the fusion of the EWSR1 gene on chromosome 22 with a novel gene at 9q22 called NR4A3 (previously known as NOR1, CHN, or TEC) [126-129]. The biological function of the NR4A3 nuclear receptor is not yet fully elucidated. The fusion protein appears to function as a transcriptional activator and has been shown to induce cellular transformation [130-133].

A small proportion have a different translocation, either t(9;17), which results in a TAF15-NR4A3 fusion gene [134,135], or t(9;15), which results in an TCF12-NR4A3 fusion gene [136].

Dermatofibrosarcoma protuberans — DFSP is characterized by a specific nonreciprocal translocation t(17;22)(q22;q13), often within ring chromosomes. (See "Dermatofibrosarcoma protuberans: Epidemiology, pathogenesis, clinical presentation, diagnosis, and staging".)

Solitary fibrous tumor — Solitary fibrous tumors (SFTs) of all sites are characterized by a recurrent inversion of the long arm of chromosome 12 (12q13). This inversion results in a fusion of two genes, NAB2 (NGFIA-binding protein 2) and STAT6 (signal transducer and activator of transcription 6). The fusion of NAB2 and STAT6 creates a chimeric transcription factor in which the NAB2 repressor domain is substituted by a carboxy-terminal STAT6 transactivation domain or near-full-length STAT6. The NAB2-STAT6 chimeric transcription factor constitutively localizes to the nucleus, where it is thought to serve as a driver of tumorigenesis by constitutively activating NAB2 target genes [137]. The NAB2-STAT6 fusion gene is a distinct molecular feature of SFTs, present in up to 100 percent of cases, which has not been detected in other tumors. (See "Solitary fibrous tumor", section on 'Molecular pathogenesis and molecular diagnostics'.)

Epithelioid hemangioendothelioma — Epithelioid hemangioendothelioma (EHE) is a malignant vascular neoplasm that is characterized by recurrent translocations involving chromosome regions 1p36.3 and 3q25; the resulting translocation between the WWTR1 (WW domain-containing transcription regulator 1; 3q25) and CAMTA1 (calmodulin-binding transcription activator 1; 1p36) genes results in the formation of a disease-defining WWTR1-CAMTA1 fusion gene [138,139]. The fusion gene is under the transcriptional control of the WWTR1 promoter and encodes a putative chimeric transcription factor that leads to overexpression of both genes. The fusion gene appears to be present in approximately 90 percent of EHEs tested, but not in other vascular neoplasms. Immunohistochemical staining for CAMTA1 protein expression is useful for distinguishing EHE from other epithelioid mesenchymal neoplasms, which are negative for expression of CAMTA1 [140].

A small subset of EHE have a different translocation, which results in a gene rearrangement between Yes-associated protein 1 (YAP1) and transcription factor for the immunoglobulin heavy chain enhancer 3 (TFE3) [141]. (See "Uncommon sarcoma subtypes", section on 'Epithelioid hemangioendothelioma'.)

Chromosomal instability — Although approximately one-third of all soft tissue sarcomas have a defining translocation as described above, the remaining sarcomas often have a complex karyotype. A positive correlation exists between the number of genomic alterations and the histologic grade of soft tissue sarcomas [142]. Because high-grade soft tissue sarcomas have worse outcome, genomic complexity is correlated with more aggressive sarcomas. (See "Clinical presentation, histopathology, diagnostic evaluation, and staging of soft tissue sarcoma", section on 'Histologic grade'.)

Molecular genetic testing for tumor classification — Immunohistochemistry, cytogenetics, and next-generation sequencing are important tools that aid in sarcoma classification. As an example, molecular assays for specific fusion genes can provide a genetic approach to the differential diagnosis of soft tissue sarcomas [143].

Gene expression profiling (GEP) by means of DNA microarrays has been applied to soft tissue sarcomas [144]. In one report, transcriptional profiling for 5520 different known genes was used to separate 41 soft tissue sarcomas into five distinct groups [145]:

GIST highly expressed a cluster of 125 genes, which separated them from other sarcomas.

Synovial sarcomas expressed a specific cluster of 104 genes, among them, molecules involved in retinoic acid pathways, and the epidermal growth factor receptor.

Neural tumors (eg, malignant peripheral nerve sheath tumors).

One-half of all leiomyosarcomas, in which 24 specific genes were highly expressed.

A broad group, containing all of the liposarcomas, UPS, and the remainder of the leiomyosarcomas, in which molecular profiles were not predicted by histologic features or immunohistochemistry.

The clinical utility of such a classification system has been realized only for the GISTs, in which tyrosine kinase-activating KIT mutations predict responsiveness to targeted therapy. (See "Clinical presentation, diagnosis, and prognosis of gastrointestinal stromal tumors".)

However, the potential for future benefit of improved classification of soft tissue tumors includes better prediction of natural history and patient-tailored therapy based on the specific targets identified by microarray analysis [146]. For example, a prognostic gene expression signature termed the complexity index in sarcomas (CINSARC) has been developed that includes 67 genes related to mitosis and chromosome management [142]. In multivariate analysis, CINSARC predicted metastasis outcome in a training set and in a validation set of 127 independent soft tissue sarcomas. CINSARC was validated as a predictor of metastasis in patients with synovial sarcoma [147]. Interestingly, chromosomal complexity was more prevalent in adult patients with synovial sarcoma who had worse outcome than pediatric patients with synovial sarcoma. The CINSARC signature had already been shown to be a predictor of clinical outcomes in soft tissue sarcomas, GIST, and other cancers [142].

RADIATION THERAPY AND CHEMOTHERAPY

Epidemiology — Radiation therapy (RT) is recognized as a cause of sarcoma of bone and soft tissue; sarcomas were one of the first solid cancers to be linked to ionizing radiation exposure. The most frequent histopathologic type of radiation-induced soft tissue sarcoma arising in soft tissues is undifferentiated pleomorphic sarcoma (UPS). The most common type of bone sarcoma is osteosarcoma. Among females treated with RT for breast cancer, the most common secondary sarcoma is cutaneous angiosarcoma. (See "Radiation-associated sarcomas", section on 'Histologic distribution' and "Breast sarcoma: Epidemiology, risk factors, clinical presentation, diagnosis, and staging".)

The frequency increases with the RT dose and with the post-RT observation period, and it decreases with age. Sarcoma is primarily a complication of high-dose therapy; it is rarely seen after low doses (<40 Gy). However, the actuarial frequency at 15 to 20 years is small, approximately 0.5 percent for RT of normal bone and soft tissue in the adult treated with RT alone to full doses and without chemotherapy. (See "Radiation-associated sarcomas", section on 'Radiation dose and age of exposure'.)

Childhood cancer survivors — The frequency of secondary sarcomas is even higher following RT in survivors of childhood cancer. The risk is highest in children who receive both RT and chemotherapy (particularly anthracyclines and alkylating agents) and in those who are treated for a primary sarcoma or retinoblastoma. (See "Radiation-associated sarcomas", section on 'Childhood RT' and "Radiation-associated sarcomas", section on 'Chemotherapy agents'.)

In general, secondary sarcomas have a poorer prognosis than do de novo sarcomas. However, these patients warrant aggressive treatment, as secondary sarcomas are potentially curable. This subject is discussed in detail separately. (See "Radiation-associated sarcomas" and "Breast sarcoma: Epidemiology, risk factors, clinical presentation, diagnosis, and staging" and "Breast sarcoma: Treatment" and "Head and neck sarcomas", section on 'Risk factors'.)

INDUSTRIAL CHEMICALS — There are a number of inherent problems in occupational epidemiology with small numbers of patients in any given series and the difficulty in isolating a single agent [148]. For these reasons, few associations can be considered established and causal [149]. As examples, there is a clear association between vinyl chloride or arsenic exposure and hepatic angiosarcoma [150,151] and a probable association between phenoxy herbicides and soft tissue sarcoma [152-154]. The last risk may be greater with exposure to phenoxy herbicides contaminated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or higher chlorinated dioxins [155,156]. A role for dioxin per se is controversial. A population-based case control study, however, found no increased risk for soft tissue sarcoma among Vietnam veterans, including those exposed to Agent Orange, which contains dioxin [157].

High intensity chlorophenol exposure in jobs involving wood preservation, machinists and the use of cutting fluids may increase the risk of soft tissue sarcoma, independent of phenoxy herbicides [153,158]. However, some studies have not confirmed this association [152].

CHRONIC EDEMA, CHRONIC IRRITATION, AND TRAUMA — Sarcomas of soft tissue (primarily angiosarcomas) may be observed following massive and quite protracted edema. This has classically been seen in the postmastectomy lymphedematous arm (Stewart-Treves syndrome) [159,160]. It has also been described with chronic lymphedema due to filarial infection [161].

Chronic irritation secondary to foreign bodies also may be a factor in the induction of sarcomas. Trauma may also be a factor in the development of soft tissue sarcomas including desmoid tumors. In an occasional patient, there is a history of major trauma to the affected site many months prior to the appearance of local symptoms of tumor. The usual history is of a traumatic incident occurring shortly prior to the awareness of the mass. Clinicians often explain this sequence of events that the trauma merely brought the patient's attention to the presence of the mass. However, injury has been shown to promote sarcoma development in animal models [162,163]. Therefore, additional study is needed in patients to determine whether injury promotes sarcomagenesis.

VIRAL INFECTION — Viral infections are rarely associated with the development of sarcomas:

Human herpesvirus 8 (HHV-8) has been implicated in the pathogenesis of Kaposi sarcoma. (See "AIDS-related Kaposi sarcoma: Clinical manifestations and diagnosis" and "Classic Kaposi sarcoma: Epidemiology, risk factors, pathology, and molecular pathogenesis", section on 'Anti-HHV-8 antibodies and viremia'.)

Epstein-Barr virus (EBV)-associated smooth muscle tumors have been described in patients with HIV/AIDS and in other immunocompromised hosts (eg, solid organ transplant, NK cell deficiency) [164-167]. (See "NK cell deficiency syndromes: Clinical manifestations and diagnosis".)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Soft tissue sarcoma".)

SUMMARY

There is no clearly defined etiology in most cases of soft tissue sarcoma, but a number of associated or predisposing factors have been identified. These include familial predisposition (Li-Fraumeni syndrome, familial adenomatous polyposis [FAP], retinoblastoma, and neurofibromatosis), gene mutations, radiation therapy (RT), chemotherapy, chemical carcinogens, chronic irritation, and lymphedema. In addition, viruses, including human herpesvirus 8 (HHV-8), have been implicated in the pathogenesis of Kaposi sarcoma. (See "Classic Kaposi sarcoma: Epidemiology, risk factors, pathology, and molecular pathogenesis" and "AIDS-related Kaposi sarcoma: Clinical manifestations and diagnosis".)

There is a high incidence of acquired (somatic) gene alterations in soft tissue and bone sarcomas. Somatic mutations in specific genes vary across the histologic subtypes of soft tissue sarcoma. (See 'Somatic gene mutations' above.)

A number of soft tissue sarcomas display recurrent nonrandom chromosomal translocations, which serve as definitive diagnostic criteria for the tumors in which they occur (table 1). Furthermore, the identification of these molecular signatures has provided clues as to the molecular alterations that are fundamental for the development and often maintenance of sarcoma. (See 'Chromosomal translocations' above.)

RT is recognized as a cause of sarcomas of soft tissue and bone. The frequency increases with the RT dose and with the post-RT observation period, and it decreases with age. The risk of a radiation-induced sarcoma is particularly high in survivors of childhood cancer, and it is highest in children who receive both RT and chemotherapy (particularly anthracyclines and alkylating agents) and in those who are treated for a primary sarcoma. (See 'Radiation therapy and chemotherapy' above.)

Chronic edema has been linked to soft tissue sarcomas, particularly angiosarcoma in the setting of an edematous extremity. (See 'Chronic edema, chronic irritation, and trauma' above.)

Viral infections are associated with the development of specific sarcomas (Kaposi sarcoma and Epstein-Barr virus [EBV]-associated smooth muscle tumors). (See 'Viral infection' above.)

Sarcomas of soft tissue (primarily angiosarcomas) may be observed following massive and protracted edema. This has classically been seen in the postmastectomy lymphedematous arm and is termed Stewart-Treves syndrome. (See 'Chronic edema, chronic irritation, and trauma' above.)

ACKNOWLEDGMENTS — The UpToDate editorial staff acknowledges Thomas DeLaney, MD, and David G Kirsch, MD, PhD, who contributed to earlier versions of this topic review.

  1. Zahm SH, Fraumeni JF Jr. The epidemiology of soft tissue sarcoma. Semin Oncol 1997; 24:504.
  2. Li FP. Cancer families: human models of susceptibility to neoplasia--the Richard and Hinda Rosenthal Foundation Award lecture. Cancer Res 1988; 48:5381.
  3. Li FP, Fraumeni JF Jr, Mulvihill JJ, et al. A cancer family syndrome in twenty-four kindreds. Cancer Res 1988; 48:5358.
  4. Hartley AL, Birch JM, Blair V, et al. Patterns of cancer in the families of children with soft tissue sarcoma. Cancer 1993; 72:923.
  5. Zhang J, Walsh MF, Wu G, et al. Germline Mutations in Predisposition Genes in Pediatric Cancer. N Engl J Med 2015; 373:2336.
  6. Ballinger ML, Goode DL, Ray-Coquard I, et al. Monogenic and polygenic determinants of sarcoma risk: an international genetic study. Lancet Oncol 2016; 17:1261.
  7. Carnevale A, Lieberman E, Cárdenas R. Li-Fraumeni syndrome in pediatric patients with soft tissue sarcoma or osteosarcoma. Arch Med Res 1997; 28:383.
  8. Evans SC, Lozano G. The Li-Fraumeni syndrome: an inherited susceptibility to cancer. Mol Med Today 1997; 3:390.
  9. Varley JM, McGown G, Thorncroft M, et al. Germ-line mutations of TP53 in Li-Fraumeni families: an extended study of 39 families. Cancer Res 1997; 57:3245.
  10. Malkin D. p53 and the Li-Fraumeni syndrome. Cancer Genet Cytogenet 1993; 66:83.
  11. Mai PL, Best AF, Peters JA, et al. Risks of first and subsequent cancers among TP53 mutation carriers in the National Cancer Institute Li-Fraumeni syndrome cohort. Cancer 2016; 122:3673.
  12. Ognjanovic S, Olivier M, Bergemann TL, Hainaut P. Sarcomas in TP53 germline mutation carriers: a review of the IARC TP53 database. Cancer 2012; 118:1387.
  13. Hizawa K, Iida M, Mibu R, et al. Desmoid tumors in familial adenomatous polyposis/Gardner's syndrome. J Clin Gastroenterol 1997; 25:334.
  14. DerKinderen DJ, Koten JW, Nagelkerke NJ, et al. Non-ocular cancer in patients with hereditary retinoblastoma and their relatives. Int J Cancer 1988; 41:499.
  15. Kleinerman RA, Tucker MA, Tarone RE, et al. Risk of new cancers after radiotherapy in long-term survivors of retinoblastoma: an extended follow-up. J Clin Oncol 2005; 23:2272.
  16. Yu CL, Tucker MA, Abramson DH, et al. Cause-specific mortality in long-term survivors of retinoblastoma. J Natl Cancer Inst 2009; 101:581.
  17. Kleinerman RA, Tucker MA, Abramson DH, et al. Risk of soft tissue sarcomas by individual subtype in survivors of hereditary retinoblastoma. J Natl Cancer Inst 2007; 99:24.
  18. Wong FL, Boice JD Jr, Abramson DH, et al. Cancer incidence after retinoblastoma. Radiation dose and sarcoma risk. JAMA 1997; 278:1262.
  19. Brekke HR, Ribeiro FR, Kolberg M, et al. Genomic changes in chromosomes 10, 16, and X in malignant peripheral nerve sheath tumors identify a high-risk patient group. J Clin Oncol 2010; 28:1573.
  20. Mertens F, Rydholm A, Bauer HF, et al. Cytogenetic findings in malignant peripheral nerve sheath tumors. Int J Cancer 1995; 61:793.
  21. Ducatman BS, Scheithauer BW, Piepgras DG, et al. Malignant peripheral nerve sheath tumors. A clinicopathologic study of 120 cases. Cancer 1986; 57:2006.
  22. Borden EC, Baker LH, Bell RS, et al. Soft tissue sarcomas of adults: state of the translational science. Clin Cancer Res 2003; 9:1941.
  23. Italiano A, Di Mauro I, Rapp J, et al. Clinical effect of molecular methods in sarcoma diagnosis (GENSARC): a prospective, multicentre, observational study. Lancet Oncol 2016; 17:532.
  24. Ladanyi M, Bridge JA. Contribution of molecular genetic data to the classification of sarcomas. Hum Pathol 2000; 31:532.
  25. Mohamed AN, Zalupski MM, Ryan JR, et al. Cytogenetic aberrations and DNA ploidy in soft tissue sarcoma. A Southwest Oncology Group Study. Cancer Genet Cytogenet 1997; 99:45.
  26. Barretina J, Taylor BS, Banerji S, et al. Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy. Nat Genet 2010; 42:715.
  27. Italiano A, Bianchini L, Keslair F, et al. HMGA2 is the partner of MDM2 in well-differentiated and dedifferentiated liposarcomas whereas CDK4 belongs to a distinct inconsistent amplicon. Int J Cancer 2008; 122:2233.
  28. Chen X, Stewart E, Shelat AA, et al. Targeting oxidative stress in embryonal rhabdomyosarcoma. Cancer Cell 2013; 24:710.
  29. Shern JF, Chen L, Chmielecki J, et al. Comprehensive genomic analysis of rhabdomyosarcoma reveals a landscape of alterations affecting a common genetic axis in fusion-positive and fusion-negative tumors. Cancer Discov 2014; 4:216.
  30. Zhang M, Linardic CM, Kirsch DG. RAS and ROS in rhabdomyosarcoma. Cancer Cell 2013; 24:689.
  31. Martin GA, Viskochil D, Bollag G, et al. The GAP-related domain of the neurofibromatosis type 1 gene product interacts with ras p21. Cell 1990; 63:843.
  32. Xu GF, Lin B, Tanaka K, et al. The catalytic domain of the neurofibromatosis type 1 gene product stimulates ras GTPase and complements ira mutants of S. cerevisiae. Cell 1990; 63:835.
  33. Lee W, Teckie S, Wiesner T, et al. PRC2 is recurrently inactivated through EED or SUZ12 loss in malignant peripheral nerve sheath tumors. Nat Genet 2014; 46:1227.
  34. De Raedt T, Beert E, Pasmant E, et al. PRC2 loss amplifies Ras-driven transcription and confers sensitivity to BRD4-based therapies. Nature 2014; 514:247.
  35. el-Deiry WS, Tokino T, Velculescu VE, et al. WAF1, a potential mediator of p53 tumor suppression. Cell 1993; 75:817.
  36. Lane DP. Cancer. p53, guardian of the genome. Nature 1992; 358:15.
  37. Levine AJ, Perry ME, Chang A, et al. The 1993 Walter Hubert Lecture: the role of the p53 tumour-suppressor gene in tumorigenesis. Br J Cancer 1994; 69:409.
  38. Toguchida J, Yamaguchi T, Dayton SH, et al. Prevalence and spectrum of germline mutations of the p53 gene among patients with sarcoma. N Engl J Med 1992; 326:1301.
  39. Malkin D, Jolly KW, Barbier N, et al. Germline mutations of the p53 tumor-suppressor gene in children and young adults with second malignant neoplasms. N Engl J Med 1992; 326:1309.
  40. McIntyre JF, Smith-Sorensen B, Friend SH, et al. Germline mutations of the p53 tumor suppressor gene in children with osteosarcoma. J Clin Oncol 1994; 12:925.
  41. Vogel KS, Klesse LJ, Velasco-Miguel S, et al. Mouse tumor model for neurofibromatosis type 1. Science 1999; 286:2176.
  42. Toguchida J, Yamaguchi T, Ritchie B, et al. Mutation spectrum of the p53 gene in bone and soft tissue sarcomas. Cancer Res 1992; 52:6194.
  43. Wadayama B, Toguchida J, Yamaguchi T, et al. p53 expression and its relationship to DNA alterations in bone and soft tissue sarcomas. Br J Cancer 1993; 68:1134.
  44. Porter PL, Gown AM, Kramp SG, Coltrera MD. Widespread p53 overexpression in human malignant tumors. An immunohistochemical study using methacarn-fixed, embedded tissue. Am J Pathol 1992; 140:145.
  45. Soini Y, Vähäkangas K, Nuorva K, et al. p53 immunohistochemistry in malignant fibrous histiocytomas and other mesenchymal tumours. J Pathol 1992; 168:29.
  46. Stratton MR, Moss S, Warren W, et al. Mutation of the p53 gene in human soft tissue sarcomas: association with abnormalities of the RB1 gene. Oncogene 1990; 5:1297.
  47. Mulligan LM, Matlashewski GJ, Scrable HJ, Cavenee WK. Mechanisms of p53 loss in human sarcomas. Proc Natl Acad Sci U S A 1990; 87:5863.
  48. Patterson H, Gill S, Fisher C, et al. Abnormalities of the p53 MDM2 and DCC genes in human leiomyosarcomas. Br J Cancer 1994; 69:1052.
  49. Andreassen A, Oyjord T, Hovig E, et al. p53 abnormalities in different subtypes of human sarcomas. Cancer Res 1993; 53:468.
  50. Blom R, Guerrieri C, Stâl O, et al. Leiomyosarcoma of the uterus: A clinicopathologic, DNA flow cytometric, p53, and mdm-2 analysis of 49 cases. Gynecol Oncol 1998; 68:54.
  51. Simms WW, Ordóñez NG, Johnston D, et al. p53 expression in dedifferentiated chondrosarcoma. Cancer 1995; 76:223.
  52. Pollock RE, Lang A, Luo J, et al. Soft tissue sarcoma metastasis from clonal expansion of p53 mutated tumor cells. Oncogene 1996; 12:2035.
  53. Donehower LA, Harvey M, Slagle BL, et al. Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature 1992; 356:215.
  54. Lee JM, Abrahamson JL, Kandel R, et al. Susceptibility to radiation-carcinogenesis and accumulation of chromosomal breakage in p53 deficient mice. Oncogene 1994; 9:3731.
  55. Pollock R, Lang A, Ge T, et al. Wild-type p53 and a p53 temperature-sensitive mutant suppress human soft tissue sarcoma by enhancing cell cycle control. Clin Cancer Res 1998; 4:1985.
  56. Ventura A, Kirsch DG, McLaughlin ME, et al. Restoration of p53 function leads to tumour regression in vivo. Nature 2007; 445:661.
  57. Yang CY, Liau JY, Huang WJ, et al. Targeted next-generation sequencing of cancer genes identified frequent TP53 and ATRX mutations in leiomyosarcoma. Am J Transl Res 2015; 7:2072.
  58. Chen X, Bahrami A, Pappo A, et al. Recurrent somatic structural variations contribute to tumorigenesis in pediatric osteosarcoma. Cell Rep 2014; 7:104.
  59. Liau JY, Lee JC, Tsai JH, et al. Comprehensive screening of alternative lengthening of telomeres phenotype and loss of ATRX expression in sarcomas. Mod Pathol 2015; 28:1545.
  60. Flynn RL, Cox KE, Jeitany M, et al. Alternative lengthening of telomeres renders cancer cells hypersensitive to ATR inhibitors. Science 2015; 347:273.
  61. Cance WG, Brennan MF, Dudas ME, et al. Altered expression of the retinoblastoma gene product in human sarcomas. N Engl J Med 1990; 323:1457.
  62. Karpeh MS, Brennan MF, Cance WG, et al. Altered patterns of retinoblastoma gene product expression in adult soft-tissue sarcomas. Br J Cancer 1995; 72:986.
  63. Wunder JS, Czitrom AA, Kandel R, Andrulis IL. Analysis of alterations in the retinoblastoma gene and tumor grade in bone and soft-tissue sarcomas. J Natl Cancer Inst 1991; 83:194.
  64. Oliner JD, Kinzler KW, Meltzer PS, et al. Amplification of a gene encoding a p53-associated protein in human sarcomas. Nature 1992; 358:80.
  65. Flørenes VA, Maelandsmo GM, Forus A, et al. MDM2 gene amplification and transcript levels in human sarcomas: relationship to TP53 gene status. J Natl Cancer Inst 1994; 86:1297.
  66. Khatib ZA, Matsushime H, Valentine M, et al. Coamplification of the CDK4 gene with MDM2 and GLI in human sarcomas. Cancer Res 1993; 53:5535.
  67. Nilbert M, Rydholm A, Willén H, et al. MDM2 gene amplification correlates with ring chromosome in soft tissue tumors. Genes Chromosomes Cancer 1994; 9:261.
  68. Momand J, Zambetti GP, Olson DC, et al. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell 1992; 69:1237.
  69. Oliner JD, Pietenpol JA, Thiagalingam S, et al. Oncoprotein MDM2 conceals the activation domain of tumour suppressor p53. Nature 1993; 362:857.
  70. Leach FS, Tokino T, Meltzer P, et al. p53 Mutation and MDM2 amplification in human soft tissue sarcomas. Cancer Res 1993; 53:2231.
  71. Cordon-Cardo C, Latres E, Drobnjak M, et al. Molecular abnormalities of mdm2 and p53 genes in adult soft tissue sarcomas. Cancer Res 1994; 54:794.
  72. Kanoe H, Nakayama T, Murakami H, et al. Amplification of the CDK4 gene in sarcomas: tumor specificity and relationship with the RB gene mutation. Anticancer Res 1998; 18:2317.
  73. Pilotti S, Della Torre G, Lavarino C, et al. Molecular abnormalities in liposarcoma: role of MDM2 and CDK4-containing amplicons at 12q13-22. J Pathol 1998; 185:188.
  74. Aleixo PB, Hartmann AA, Menezes IC, et al. Can MDM2 and CDK4 make the diagnosis of well differentiated/dedifferentiated liposarcoma? An immunohistochemical study on 129 soft tissue tumours. J Clin Pathol 2009; 62:1127.
  75. Llanos S, Efeyan A, Monsech J, et al. A high-throughput loss-of-function screening identifies novel p53 regulators. Cell Cycle 2006; 5:1880.
  76. Narita M, Narita M, Krizhanovsky V, et al. A novel role for high-mobility group a proteins in cellular senescence and heterochromatin formation. Cell 2006; 126:503.
  77. Mayr C, Hemann MT, Bartel DP. Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation. Science 2007; 315:1576.
  78. Smith SH, Weiss SW, Jankowski SA, et al. SAS amplification in soft tissue sarcomas. Cancer Res 1992; 52:3746.
  79. Jankowski SA, Mitchell DS, Smith SH, et al. SAS, a gene amplified in human sarcomas, encodes a new member of the transmembrane 4 superfamily of proteins. Oncogene 1994; 9:1205.
  80. Forus A, Flørenes VA, Maelandsmo GM, et al. Mapping of amplification units in the q13-14 region of chromosome 12 in human sarcomas: some amplica do not include MDM2. Cell Growth Differ 1993; 4:1065.
  81. Ladanyl M, Heinemann FS, Huvos AG, et al. Neural differentiation in small round cell tumors of bone and soft tissue with the translocation t(11;22)(q24;q12): an immunohistochemical study of 11 cases. Hum Pathol 1990; 21:1245.
  82. Turc-Carel C, Aurias A, Mugneret F, et al. Chromosomes in Ewing's sarcoma. I. An evaluation of 85 cases of remarkable consistency of t(11;22)(q24;q12). Cancer Genet Cytogenet 1988; 32:229.
  83. Delattre O, Zucman J, Melot T, et al. The Ewing family of tumors--a subgroup of small-round-cell tumors defined by specific chimeric transcripts. N Engl J Med 1994; 331:294.
  84. Zucman J, Melot T, Desmaze C, et al. Combinatorial generation of variable fusion proteins in the Ewing family of tumours. EMBO J 1993; 12:4481.
  85. Sorensen PH, Lessnick SL, Lopez-Terrada D, et al. A second Ewing's sarcoma translocation, t(21;22), fuses the EWS gene to another ETS-family transcription factor, ERG. Nat Genet 1994; 6:146.
  86. Kawamura-Saito M, Yamazaki Y, Kaneko K, et al. Fusion between CIC and DUX4 up-regulates PEA3 family genes in Ewing-like sarcomas with t(4;19)(q35;q13) translocation. Hum Mol Genet 2006; 15:2125.
  87. Antonescu CR, Owosho AA, Zhang L, et al. Sarcomas With CIC-rearrangements Are a Distinct Pathologic Entity With Aggressive Outcome: A Clinicopathologic and Molecular Study of 115 Cases. Am J Surg Pathol 2017; 41:941.
  88. Hung YP, Fletcher CD, Hornick JL. Evaluation of ETV4 and WT1 expression in CIC-rearranged sarcomas and histologic mimics. Mod Pathol 2016; 29:1324.
  89. Le Guellec S, Velasco V, Pérot G, et al. ETV4 is a useful marker for the diagnosis of CIC-rearranged undifferentiated round-cell sarcomas: a study of 127 cases including mimicking lesions. Mod Pathol 2016; 29:1523.
  90. Sugita S, Arai Y, Tonooka A, et al. A novel CIC-FOXO4 gene fusion in undifferentiated small round cell sarcoma: a genetically distinct variant of Ewing-like sarcoma. Am J Surg Pathol 2014; 38:1571.
  91. Pierron G, Tirode F, Lucchesi C, et al. A new subtype of bone sarcoma defined by BCOR-CCNB3 gene fusion. Nat Genet 2012; 44:461.
  92. Puls F, Niblett A, Marland G, et al. BCOR-CCNB3 (Ewing-like) sarcoma: a clinicopathologic analysis of 10 cases, in comparison with conventional Ewing sarcoma. Am J Surg Pathol 2014; 38:1307.
  93. Boyar RM. Regulation of gonadotropin secretion in man. Med Clin North Am 1978; 62:367.
  94. Cohen-Gogo S, Cellier C, Coindre JM, et al. Ewing-like sarcomas with BCOR-CCNB3 fusion transcript: a clinical, radiological and pathological retrospective study from the Société Française des Cancers de L'Enfant. Pediatr Blood Cancer 2014; 61:2191.
  95. Machado I, Navarro L, Pellin A, et al. Defining Ewing and Ewing-like small round cell tumors (SRCT): The need for molecular techniques in their categorization and differential diagnosis. A study of 200 cases. Ann Diagn Pathol 2016; 22:25.
  96. Crozat A, Aman P, Mandahl N, Ron D. Fusion of CHOP to a novel RNA-binding protein in human myxoid liposarcoma. Nature 1993; 363:640.
  97. Rabbitts TH, Forster A, Larson R, Nathan P. Fusion of the dominant negative transcription regulator CHOP with a novel gene FUS by translocation t(12;16) in malignant liposarcoma. Nat Genet 1993; 4:175.
  98. Hisaoka M, Tsuji S, Morimitsu Y, et al. Detection of TLS/FUS-CHOP fusion transcripts in myxoid and round cell liposarcomas by nested reverse transcription-polymerase chain reaction using archival paraffin-embedded tissues. Diagn Mol Pathol 1998; 7:96.
  99. Aman P, Ron D, Mandahl N, et al. Rearrangement of the transcription factor gene CHOP in myxoid liposarcomas with t(12;16)(q13;p11). Genes Chromosomes Cancer 1992; 5:278.
  100. Barone MV, Crozat A, Tabaee A, et al. CHOP (GADD153) and its oncogenic variant, TLS-CHOP, have opposing effects on the induction of G1/S arrest. Genes Dev 1994; 8:453.
  101. Scapa JV, Cloutier JM, Raghavan SS, et al. DDIT3 Immunohistochemistry Is a Useful Tool for the Diagnosis of Myxoid Liposarcoma. Am J Surg Pathol 2021; 45:230.
  102. Baranov E, Black MA, Fletcher CDM, et al. Nuclear expression of DDIT3 distinguishes high-grade myxoid liposarcoma from other round cell sarcomas. Mod Pathol 2021; 34:1367.
  103. dos Santos NR, de Bruijn DR, van Kessel AG. Molecular mechanisms underlying human synovial sarcoma development. Genes Chromosomes Cancer 2001; 30:1.
  104. Clark J, Rocques PJ, Crew AJ, et al. Identification of novel genes, SYT and SSX, involved in the t(X;18)(p11.2;q11.2) translocation found in human synovial sarcoma. Nat Genet 1994; 7:502.
  105. Kawai A, Woodruff J, Healey JH, et al. SYT-SSX gene fusion as a determinant of morphology and prognosis in synovial sarcoma. N Engl J Med 1998; 338:153.
  106. Agus V, Tamborini E, Mezzelani A, et al. Re: A novel fusion gene, SYT-SSX4, in synovial sarcoma. J Natl Cancer Inst 2001; 93:1347.
  107. Baranov E, McBride MJ, Bellizzi AM, et al. A Novel SS18-SSX Fusion-specific Antibody for the Diagnosis of Synovial Sarcoma. Am J Surg Pathol 2020; 44:922.
  108. Kadoch C, Crabtree GR. Reversible disruption of mSWI/SNF (BAF) complexes by the SS18-SSX oncogenic fusion in synovial sarcoma. Cell 2013; 153:71.
  109. Paulino AC. Synovial sarcoma prognostic factors and patterns of failure. Am J Clin Oncol 2004; 27:122.
  110. Barr FG, Nauta LE, Hollows JC. Structural analysis of PAX3 genomic rearrangements in alveolar rhabdomyosarcoma. Cancer Genet Cytogenet 1998; 102:32.
  111. Davis RJ, Barr FG. Fusion genes resulting from alternative chromosomal translocations are overexpressed by gene-specific mechanisms in alveolar rhabdomyosarcoma. Proc Natl Acad Sci U S A 1997; 94:8047.
  112. Ginsberg JP, Davis RJ, Bennicelli JL, et al. Up-regulation of MET but not neural cell adhesion molecule expression by the PAX3-FKHR fusion protein in alveolar rhabdomyosarcoma. Cancer Res 1998; 58:3542.
  113. Linardic CM, Naini S, Herndon JE 2nd, et al. The PAX3-FKHR fusion gene of rhabdomyosarcoma cooperates with loss of p16INK4A to promote bypass of cellular senescence. Cancer Res 2007; 67:6691.
  114. Kelly KM, Womer RB, Sorensen PH, et al. Common and variant gene fusions predict distinct clinical phenotypes in rhabdomyosarcoma. J Clin Oncol 1997; 15:1831.
  115. Kelly KM, Womer RB, Barr FG. Minimal disease detection in patients with alveolar rhabdomyosarcoma using a reverse transcriptase-polymerase chain reaction method. Cancer 1996; 78:1320.
  116. Zucman J, Delattre O, Desmaze C, et al. EWS and ATF-1 gene fusion induced by t(12;22) translocation in malignant melanoma of soft parts. Nat Genet 1993; 4:341.
  117. Coindre JM, Hostein I, Terrier P, et al. Diagnosis of clear cell sarcoma by real-time reverse transcriptase-polymerase chain reaction analysis of paraffin embedded tissues: clinicopathologic and molecular analysis of 44 patients from the French sarcoma group. Cancer 2006; 107:1055.
  118. Antonescu CR, Nafa K, Segal NH, et al. EWS-CREB1: a recurrent variant fusion in clear cell sarcoma--association with gastrointestinal location and absence of melanocytic differentiation. Clin Cancer Res 2006; 12:5356.
  119. Bu X, Bernstein L. A proposed explanation for female predominance in alveolar soft part sarcoma. Noninactivation of X; autosome translocation fusion gene? Cancer 2005; 103:1245.
  120. Heimann P, Devalck C, Debusscher C, et al. Alveolar soft-part sarcoma: further evidence by FISH for the involvement of chromosome band 17q25. Genes Chromosomes Cancer 1998; 23:194.
  121. Joyama S, Ueda T, Shimizu K, et al. Chromosome rearrangement at 17q25 and xp11.2 in alveolar soft-part sarcoma: A case report and review of the literature. Cancer 1999; 86:1246.
  122. Ladanyi M, Lui MY, Antonescu CR, et al. The der(17)t(X;17)(p11;q25) of human alveolar soft part sarcoma fuses the TFE3 transcription factor gene to ASPL, a novel gene at 17q25. Oncogene 2001; 20:48.
  123. Lazar AJ, Das P, Tuvin D, et al. Angiogenesis-promoting gene patterns in alveolar soft part sarcoma. Clin Cancer Res 2007; 13:7314.
  124. Drilon AD, Popat S, Bhuchar G, et al. Extraskeletal myxoid chondrosarcoma: a retrospective review from 2 referral centers emphasizing long-term outcomes with surgery and chemotherapy. Cancer 2008; 113:3364.
  125. Lucas DR, Stenman G. Extraskeletal myxoid chondrosarcoma. In: WHO classification of tumours of soft tissue and bone, 4th, Fletcher CDM, Bridge JA, Hogendoorn PCW, Mertens F (Eds), IARC, Lyon 2013. p.223.
  126. Brody RI, Ueda T, Hamelin A, et al. Molecular analysis of the fusion of EWS to an orphan nuclear receptor gene in extraskeletal myxoid chondrosarcoma. Am J Pathol 1997; 150:1049.
  127. Sandberg AA. Genetics of chondrosarcoma and related tumors. Curr Opin Oncol 2004; 16:342.
  128. Hirabayashi Y, Ishida T, Yoshida MA, et al. Translocation (9;22)(q22;q12). A recurrent chromosome abnormality in extraskeletal myxoid chondrosarcoma. Cancer Genet Cytogenet 1995; 81:33.
  129. Stenman G, Andersson H, Mandahl N, et al. Translocation t(9;22)(q22;q12) is a primary cytogenetic abnormality in extraskeletal myxoid chondrosarcoma. Int J Cancer 1995; 62:398.
  130. Labelle Y, Bussières J, Courjal F, Goldring MB. The EWS/TEC fusion protein encoded by the t(9;22) chromosomal translocation in human chondrosarcomas is a highly potent transcriptional activator. Oncogene 1999; 18:3303.
  131. Filion C, Motoi T, Olshen AB, et al. The EWSR1/NR4A3 fusion protein of extraskeletal myxoid chondrosarcoma activates the PPARG nuclear receptor gene. J Pathol 2009; 217:83.
  132. Hisaoka M, Hashimoto H. Extraskeletal myxoid chondrosarcoma: updated clinicopathological and molecular genetic characteristics. Pathol Int 2005; 55:453.
  133. Filion C, Labelle Y. The oncogenic fusion protein EWS/NOR-1 induces transformation of CFK2 chondrogenic cells. Exp Cell Res 2004; 297:585.
  134. Panagopoulos I, Mertens F, Isaksson M, et al. Molecular genetic characterization of the EWS/CHN and RBP56/CHN fusion genes in extraskeletal myxoid chondrosarcoma. Genes Chromosomes Cancer 2002; 35:340.
  135. Attwooll C, Tariq M, Harris M, et al. Identification of a novel fusion gene involving hTAFII68 and CHN from a t(9;17)(q22;q11.2) translocation in an extraskeletal myxoid chondrosarcoma. Oncogene 1999; 18:7599.
  136. Sjögren H, Wedell B, Meis-Kindblom JM, et al. Fusion of the NH2-terminal domain of the basic helix-loop-helix protein TCF12 to TEC in extraskeletal myxoid chondrosarcoma with translocation t(9;15)(q22;q21). Cancer Res 2000; 60:6832.
  137. Robinson DR, Wu YM, Kalyana-Sundaram S, et al. Identification of recurrent NAB2-STAT6 gene fusions in solitary fibrous tumor by integrative sequencing. Nat Genet 2013; 45:180.
  138. Tanas MR, Sboner A, Oliveira AM, et al. Identification of a disease-defining gene fusion in epithelioid hemangioendothelioma. Sci Transl Med 2011; 3:98ra82.
  139. Patel NR, Salim AA, Sayeed H, et al. Molecular characterization of epithelioid haemangioendotheliomas identifies novel WWTR1-CAMTA1 fusion variants. Histopathology 2015; 67:699.
  140. Doyle LA, Fletcher CD, Hornick JL. Nuclear Expression of CAMTA1 Distinguishes Epithelioid Hemangioendothelioma From Histologic Mimics. Am J Surg Pathol 2016; 40:94.
  141. Antonescu CR, Le Loarer F, Mosquera JM, et al. Novel YAP1-TFE3 fusion defines a distinct subset of epithelioid hemangioendothelioma. Genes Chromosomes Cancer 2013; 52:775.
  142. Chibon F, Lagarde P, Salas S, et al. Validated prediction of clinical outcome in sarcomas and multiple types of cancer on the basis of a gene expression signature related to genome complexity. Nat Med 2010; 16:781.
  143. Barr FG, Chatten J, D'Cruz CM, et al. Molecular assays for chromosomal translocations in the diagnosis of pediatric soft tissue sarcomas. JAMA 1995; 273:553.
  144. Golub TR, Slonim DK, Tamayo P, et al. Molecular classification of cancer: class discovery and class prediction by gene expression monitoring. Science 1999; 286:531.
  145. Nielsen TO, West RB, Linn SC, et al. Molecular characterisation of soft tissue tumours: a gene expression study. Lancet 2002; 359:1301.
  146. Hernando E, Charytonowicz E, Dudas ME, et al. The AKT-mTOR pathway plays a critical role in the development of leiomyosarcomas. Nat Med 2007; 13:748.
  147. Lagarde P, Przybyl J, Brulard C, et al. Chromosome instability accounts for reverse metastatic outcomes of pediatric and adult synovial sarcomas. J Clin Oncol 2013; 31:608.
  148. Sathiakumar N, Delzell E. A review of epidemiologic studies of triazine herbicides and cancer. Crit Rev Toxicol 1997; 27:599.
  149. Dich J, Zahm SH, Hanberg A, Adami HO. Pesticides and cancer. Cancer Causes Control 1997; 8:420.
  150. Lee FI, Smith PM, Bennett B, Williams DM. Occupationally related angiosarcoma of the liver in the United Kingdom 1972-1994. Gut 1996; 39:312.
  151. Lander JJ, Stanley RJ, Sumner HW, et al. Angiosarcoma of the liver associated with Fowler's solution (potassium arsenite). Gastroenterology 1975; 68:1582.
  152. Hardell L, Eriksson M. The association between soft tissue sarcomas and exposure to phenoxyacetic acids. A new case-referent study. Cancer 1988; 62:652.
  153. Wingren G, Fredrikson M, Brage HN, et al. Soft tissue sarcoma and occupational exposures. Cancer 1990; 66:806.
  154. Vineis P, Faggiano F, Tedeschi M, Ciccone G. Incidence rates of lymphomas and soft-tissue sarcomas and environmental measurements of phenoxy herbicides. J Natl Cancer Inst 1991; 83:362.
  155. Kogevinas M, Becher H, Benn T, et al. Cancer mortality in workers exposed to phenoxy herbicides, chlorophenols, and dioxins. An expanded and updated international cohort study. Am J Epidemiol 1997; 145:1061.
  156. Fingerhut MA, Halperin WE, Marlow DA, et al. Cancer mortality in workers exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin. N Engl J Med 1991; 324:212.
  157. The association of selected cancers with service in the US military in Vietnam. II. Soft-tissue and other sarcomas. The Selected Cancers Cooperative Study Group. Arch Intern Med 1990; 150:2485.
  158. Hoppin JA, Tolbert PE, Herrick RF, et al. Occupational chlorophenol exposure and soft tissue sarcoma risk among men aged 30-60 years. Am J Epidemiol 1998; 148:693.
  159. Stewart FW, Treves N. Classics in oncology: lymphangiosarcoma in postmastectomy lymphedema: a report of six cases in elephantiasis chirurgica. CA Cancer J Clin 1981; 31:284.
  160. Tomita K, Yokogawa A, Oda Y, Terahata S. Lymphangiosarcoma in postmastectomy lymphedema (Stewart-Treves syndrome): ultrastructural and immunohistologic characteristics. J Surg Oncol 1988; 38:275.
  161. Muller R, Hajdu SI, Brennan MF. Lymphangiosarcoma associated with chronic filarial lymphedema. Cancer 1987; 59:179.
  162. Sieweke MH, Thompson NL, Sporn MB, Bissell MJ. Mediation of wound-related Rous sarcoma virus tumorigenesis by TGF-beta. Science 1990; 248:1656.
  163. Martins-Green M, Boudreau N, Bissell MJ. Inflammation is responsible for the development of wound-induced tumors in chickens infected with Rous sarcoma virus. Cancer Res 1994; 54:4334.
  164. Purgina B, Rao UN, Miettinen M, Pantanowitz L. AIDS-Related EBV-Associated Smooth Muscle Tumors: A Review of 64 Published Cases. Patholog Res Int 2011; 2011:561548.
  165. Cheuk W, Li PC, Chan JK. Epstein-Barr virus-associated smooth muscle tumour: a distinctive mesenchymal tumour of immunocompromised individuals. Pathology 2002; 34:245.
  166. Lee ES, Locker J, Nalesnik M, et al. The association of Epstein-Barr virus with smooth-muscle tumors occurring after organ transplantation. N Engl J Med 1995; 332:19.
  167. McClain KL, Leach CT, Jenson HB, et al. Association of Epstein-Barr virus with leiomyosarcomas in young people with AIDS. N Engl J Med 1995; 332:12.
Topic 7737 Version 46.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟