Cancer Letters

Cancer Letters

Volume 275, Issue 1, 8 March 2009, Pages 1-8
Cancer Letters

Mini-review
Genetic aberrations in soft tissue leiomyosarcoma

https://doi.org/10.1016/j.canlet.2008.06.013Get rights and content

Abstract

Leiomyosarcoma is a malignant mesenchymal tumor composed of cells showing smooth muscle differentiation. This tumor usually occurs in middle-aged or older adults, and forms a significant percentage of retroperitoneal, vascular, extremity, and uterine sarcomas. Leiomyosarcomas are most often associated with complex karyotypes with numerous chromosomal gains and losses. Some of these cytogenetic and molecular genetic aberrations correlate with histopathologic features and clinical outcomes. Identification of genetic alterations with specific identification of oncogenes and tumor suppressor genes may lead to additional insights into the tumorigenesis of leiomyosarcoma and the opportunity to confer the benefits of targeted therapy.

Introduction

Bone and soft tissue sarcomas are uncommon neoplasms representing no more than 1% of malignant tumors [1]. Some of these tumors, such as synovial sarcoma, Ewing’s sarcoma, and osteosarcoma, occur most often in adolescents or in young adults; other sarcomas, however, such as leiomyosarcoma or well-differentiated liposarcoma, are more frequent in older individuals. On the basis of histology alone, there are more than 50 distinct types of sarcoma [2]. From the molecular point of view, these neoplasms can be bifurcated into two major groups: (a) sarcomas showing specific, recurrent genetic alterations, and relatively simple karyotypes (such as the EWSR1-FLI1 gene fusion in Ewing’s sarcoma) and (b) sarcomas showing multiple and often variable gene alterations and very complex karyotypes, such as leiomyosarcoma and osteosarcoma [3].

Leiomyosarcoma is a malignant tumor composed of cells showing distinct features of the smooth muscle lineage. It usually occurs in middle-aged or older adults. These tumors arise most commonly in five distinct anatomic sites: (1) retroperitoneum; (2) deep extremity; (3) uterus; (4) blood vessels; and (5) superficial dermis. When confined to the dermis, this latter group has an indolent clinical course and rarely metastasizes. Leiomyosarcoma constitutes a significant percentage of retroperitoneal and pelvic sarcomas. It is comparatively less common at other sites, accounting for at most 10–15% of extremity sarcomas. The deep leiomyosarcomas frequently arise in association with the smooth muscle wall of a vessel or a tubular digestive organ. Uterine leiomyosarcomas arise in the context of the myometrium. Deep leiomyosarcoma is a deadly cancer with significant mortality associated with pulmonary metastases. Risk for local recurrence, metastasis and tumor-specific mortality correlates with the three-tiered histologic grade (low, intermediate and high) assigned under the French sarcoma grading system guidelines [4], though these criteria are not clearly applicable to the uterine and dermal categories.

Leiomyosarcoma is often present as an enlarging mass. Imaging studies demonstrate a nonspecific soft tissue mass, but are helpful in delineating the relationship to adjacent structures, particularly in the retroperitoneum. The typical histologic pattern of leiomyosarcoma is of intersecting, sharply marginated fascicles of spindle cells with abundant eosinophilic cytoplasmic and elongated (cigar-shaped) nuclei. The great majority of leiomyosarcomas are reactive for SMA, desmin, and h-caldesmon on immunohistochemistry, though none of these markers are absolutely specific for smooth muscle differentiation.

Until recent years gastrointestinal stromal tumors (GISTs) were not clearly delineated from leiomyosarcoma. In contrast to GISTs, where >90% of cases are positive for c-kit protein in immunohistochemisty analysis, leiomyosarcoma only rarely expresses c-kit and even then, only at low levels [5], [6]. The signature mutation and overexpression of KIT and PDGFRA genes provide a target for selective therapy with the kinase inhibitor imatinib mesylate (Gleevec) [7]. In contrast, therapeutically relevant targets in leiomyosarcoma have yet to be discerned. Currently, leiomyosarcoma is not amenable or unlikely to be controlled by surgery alone and is treated by conventional cytotoxic chemotherapy at many centers, yet only 50% of patients respond with less than 10% long-term survival [6]. Clearly, efforts are needed to reveal recurrent genetic and molecular changes in leiomyosarcoma that may lead to improved therapeutic interventions.

Section snippets

Cytogenetic aberrations

The existing published data show that the cytogenetic and molecular genetic changes in leiomyosarcoma are complex [8]. Wang et al. [9] in 2001 examined karyotypes of about 100 leiomyosarcomas and found that most karyotypes were complex and there were no consistent, recurrent aberrations demonstrated at the chromosomal level. DNA copy number changes in 29 leiomyosarcomas were further investigated by comparative genomic hybridization (CGH). The most frequent losses were detected in 10q (20 of 29)

Molecular aberrations

Little is known about the underlying molecular determinants driving soft tissue leiomyosarcoma inception, proliferation, and metastasis. Although 17p, which harbors p53 tumor suppressor gene has been found to be amplified rather than deleted in leiomyosarcoma [9], frequent deletion of 19p, where ARF is located, suggests inactivation of p53 tumor suppressor gene in leiomyosarcoma via a different mechanism. Amplification of the COPS3 gene located in the 17p11-12 region was proposed to target p53

Organ-specific gene activities

There appear to be organ-related phenotypes among leiomyosarcomas. The uterine leiomyosarcoma have different clinical and genetic features from the other leiomyosarcomas. Cho et al. showed that uterine leiomyosarcoma had specific gains and losses using the methods of genome-wide array-based comparative genomic hybridization (array CGH) and fluorescence in situ hybridization (FISH). The regions of high-level gain were 7q36.3, 7q33-q35, 12q13-q15, and 12q23.3, while the regions of homozygous loss

Gene expression profiling studies

The focus of this review is on genetic changes in leiomyosarcoma at gene copy number changes and related molecular alterations. However, it should be noted that gene expression profiling studies using expression microarrays have also been studied by several groups. Nielsen et al. [44] showed that gene expression profiles can be used to classify different types of soft tissue tumors and improve on the histology-based classification method. Pathway analysis of the gene expression profiles of

Summary

Soft tissue leiomyosarcomas show multiple gene alterations and very complex karyotypes, including numerous gains and losses. Some of the cytogenetic and molecular genetic aberrations are correlated with the clinical pathologic features and with prognosis in small pilot studies with limited clinical information. Further exploration of aberrations of the oncogenes and the tumor suppressor genes in soft tissue leiomyosarcomas in a large cohort of patient samples using high resolution genomic

Conflict of interest

There is no conflict of interest associated with this manuscript.

Acknowledgments

We thank Kathryn Hale in the Department of Scientific Publications for her editorial assistance. The work was partially supported by NIH Grants RO1 CA098570 (WZ) and K23CA109060 (JCT) and the UTMDACC Physician Scientist Program (AJL, JCT).

References (45)

  • J. Hu et al.

    Loss of DNA copy number of 10q is associated with aggressive behavior of leiomyosarcomas: a comparative genomic hybridization study

    Cancer Genet. Cytogenet.

    (2005)
  • M.L. Larramendy et al.

    Gene copy number profiling of soft-tissue leiomyosarcomas by array-comparative genomic hybridization

    Cancer Genet. Cytogenet.

    (2006)
  • K. Kawaguchi et al.

    Death-associated protein kinase (DAP kinase) alteration in soft tissue leiomyosarcoma: promoter methylation or homozygous deletion is associated with a loss of DAP kinase expression

    Hum. Pathol.

    (2004)
  • S. Shimada et al.

    The value of MDM2 and CDK4 amplification levels using real-time polymerase chain reaction for the differential diagnosis of liposarcomas and their histologic mimickers

    Hum. Pathol.

    (2006)
  • K. Kawaguchi et al.

    Genetic and epigenetic alterations of the PTEN gene in soft tissue sarcomas

    Hum. Pathol.

    (2005)
  • Y.L. Cho et al.

    Array comparative genomic hybridization analysis of uterine leiomyosarcoma

    Gynecol. Oncol.

    (2005)
  • L. Wang et al.

    The proto-oncogene c-kit is expressed in leiomyosarcomas of the uterus

    Gynecol. Oncol.

    (2003)
  • T.O. Nielsen et al.

    Molecular characterisation of soft tissue tumours: a gene expression study

    Lancet

    (2002)
  • A. Jemal et al.

    Cancer statistics 2008

    CA Cancer J. Clin.

    (2008)
  • C.D.M. Fletcher et al.

    Soft tissue tumours: epidemiology, clinical features, histopathological typing and grading

  • E. de Alava

    Molecular pathology in sarcomas

    Clin. Transl. Oncol.

    (2007)
  • J.M. Coindre, P. Terrier, L. Guillou, V. Le Doussal, F. Collin, D. Ranchère, X. Sastre, M.O. Vilain, F. Bonichon,...
  • Cited by (87)

    • The biology and treatment of leiomyosarcomas

      2023, Critical Reviews in Oncology/Hematology
    • Molecular pathogenesis of soft tissue and bone tumors

      2023, Diagnostic Molecular Pathology: A Guide to Applied Molecular Testing, Second Edition
    • A genomic survey of sarcomas on sun-exposed skin reveals distinctive candidate drivers and potentially targetable mutations

      2020, Human Pathology
      Citation Excerpt :

      Although our targeted approach does not provide genome-wide copy number data, we hypothesize that chromosomal numerical alterations (including CDKN2A deletion) might overall play a more significant role relative to mutations in PDS as compared with AFX. Previous studies have demonstrated that LMSs have complex karyotypes [18,46]. Extracutaneous LMS cases harbor frequent copy number alterations that predominate over mutations [47].

    • TP53 in bone and soft tissue sarcomas

      2019, Pharmacology and Therapeutics
      Citation Excerpt :

      Unlike other sarcomas, LMS does not have a key fusion protein driving its tumorigenesis, although a small group of ULMS (<2.5%) is found to express an ALK fusion protein (KANK2-ALK, ACTG2-ALK) (Davis et al., 2019). Rather, the general molecular alterations in LMS affect activities of the tumor suppressors TP53, PTEN, and/or RB1 with deletion of PTEN and RB1 and mutations of TP53 (Chudasama et al., 2018; Grossmann, Layfield, & Randall, 2012; Gunderson et al., 2016; Yang et al., 2009). Cytogenetic studies of LMS suggest that loss of tumor suppressors can be an initiating event in LMS, while oncogene activation may occur at a later stage of malignant progression, because low-grade tumors contain more DNA copy number losses, while high-grade tumors have more copy number gains (Grossmann et al., 2012).

    View all citing articles on Scopus
    View full text