Journal of Hematology and Oncology Research

Journal of Hematology and Oncology Research

Journal of Hematology and Oncology Research

Current Issue Volume No: 1 Issue No: 2

In Brief Open Access Available online freely Peer Reviewed Citation

Identification of Novel Biomarker for Human Uterine Leiomyosarcoma

1Dept. Of Immunology And Infectious Disease, Shinshu University, School Of Medicine, Matsumoto, Nagano 390-8621, Japan

2SIGMA-Aldrich Collaboration Laboratory

3Promoting Business Using Advanced Technology, Japan Science And Technology Agency (JST), Chiyoda, Tokyo 102-8666, Japan

4Horiuchi Ladies Clinic, Matsumoto, Nagano 390-0821, Japan

5The Cancer System Laboratory, Research Center For Advanced Science And Technology, The University Of Tokyo, Meguro, Tokyo 153-9804 Japan

6Department Of Obstetrics And Gynecology, Osaka City University Graduate School Of Medicine, Osaka, Osaka 545-8585 Japan

7Department Of Obstetrics And Gynecology, Tohoku University Graduate School Of Medicine, Sendai, Miyagi 980-8574 Japan

8Pathology Division, National Cancer Center Research Institute, Chuoku, Tokyo 104-0045, Japan

9The International Human Epigenome Consortium (IHEC) And CREST, Japan Science And Technology Agency (JST), Chiyoda, Tokyo 102-8666, Japan

10Sigma-Aldrich Israel Ltd., Rehovot 76100, Israel

11Picower Institution And Department Of Biology, Massachusetts Institute Of Technology, Cambridge, MA 02139-4307 USA

12Department Of Obstetrics And Gynecology, Kyoto University Graduate School Of Medicine, Kyoto, Kyoto 606-8507, Japan

Abstract

Sarcomas are neoplastic malignancies that typically arise in tissues of mesenchymal origin. The identification of novel molecular mechanisms leading to sarcoma formation and the establishment of new therapies has been hampered by several critical factors. Human uterine leiomyosarcoma (Ut-LMS) develops more frequently in the muscle tissue layer of the uterine body than in the uterine cervix. Although the development of gynecologic tumors is often correlated with the secretion of female hormones; that of human Ut-LMS does not and its risk factors remain unknown. Importantly, a diagnostic biomarker that can distinguish malignant Ut-LMS from benign tumor uterine leiomyoma (LMA) has yet to be established. Therefore the risk factor(s) associated with human Ut-LMS to establish a diagnosis and novel therapeutic method. Proteasome b-ring subunit LMP2/b1i-deficient mice spontaneously develop Ut-LMS, with a disease prevalence of ~40% by 14 months of age. We shown that LMP2/b1i expression was absent in human Ut-LMS, but present in other human uterine mesenchymal tumors including uterine LMA. Therefore, defective-LMP2/b1i expression may be one of the risk factors for human Ut-LMS. LMP2/b1i is a potential diagnostic biomarker for human Ut-LMS, and may be a targeted-molecule for a new therapeutic approach.

Author Contributions
Received 22 Dec 2013; Accepted 29 Mar 2014; Published 02 Jun 2014;

Academic Editor: Krzysztof Roszkowski, Department of Radiotherapy, F. Lukaszczyk Oncology Center, Poland.

Checked for plagiarism: Yes

Review by: Single-blind

Copyright ©  2014 Takuma Hayashi, et al.

License
Creative Commons License     This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Competing interests

The authors have declared that no competing interests exist.

Citation:

Takuma Hayashi, Akiko Horiuchi, Hiroyuki Aburatani, Osamu Ishiko, Nobuo Yaegashi et al. (2014) Identification of Novel Biomarker for Human Uterine Leiomyosarcoma. Journal of Hematology and Oncology Research - 1(2):8-13. https://doi.org/10.14302/issn.2372-6601.jhor-13-379

Download as RIS, BibTeX, Text (Include abstract )

DOI 10.14302/issn.2372-6601.jhor-13-379

Introduction :

Background

Sarcomas are a rare form of malignant tumor with less than 15,000 new cases diagnosed each year in United States. Though rare, sarcomas are highly debilitating malignancies as they are often associated with significant morbidity and mortality. Sarcomas are biologically very heterogeneous as evidenced by the fact that these malignant tumors arise from a plethora of different tissues and cell types. They are classically defined by their tissue of origin and additionally stratified by their histopathology or patient’s age at diagnosis. While most tumors of the uterine body are adenocarcinomas, tumors of uterine cervix are classified into squamous tumors and adenocarcinomas. Uterine mesenchymal tumors that develop in the myometrium have been traditionally divided into benign uterine usual LMA, cellular LMA and malignant Ut-LMS based on cytological atypia, mitotic activity, and other criteria. Ut-LMS is relatively rare, having an estimated annual incidence of 0.64 per 100,000 women.1 Ut-LMS accounts for 2% ~ 5% of tumors of the uterine body and develops more frequently in the muscle layer of the uterine body than in the uterine cervix.As Surgical intervention is virtually the only means of treatment, because Ut-LMS is resistant to chemotherapy and radiotherapy.2, 3 The prognosis for Ut-LMS is poor, and the five-year survival rate is approximately 35%. However, the development of an efficient adjuvant therapy is expected to improve the prognosis for human Ut-LMS. Uterine LMA may occur in 70% ~ 80% of women by the age of 50 years.4 Difficulties have been reported in distinguishing Ut-LMS from other uterine mesenchymal tumors including uterine LMA, and a diagnosis generally requires surgery and cytoscopy. Diagnostic categories for uterine mesenchymal tumors and morphological criteria are used to assign cases. The non-standard subtypes of uterine mesenchymal tumors such as the epithelioid and myxoid types are classified in a different manner using these features; therefore a diagnostic method that can identify non-standard smooth muscle differentiation needs to be established.5, 6

High estrogen levels have been shown to significantly influence the development of tumors in the uterine body.7 Although the molecular mechanisms underlying the transformation of uterine LMA and Ut-LMS develop remain unknown. The tumors that have been initiated and grown in the myometrium increase in size due to the influence of the female hormone, estrogen, and generate more tumors. However, no correlation has been reported between the development of Ut-LMS and hormonal conditions, and no obvious risk factors have been identified. Although cases accompanied by hypocalcaemia or eosinophilia have been reported, neither clinical abnormality is an initial risk factor for human Ut-LMS. The identification of a risk factor associated with the development of human Ut-LMS would significantly contribute to the development of preventive and therapeutic treatments.

Cytoplasmic proteins are mostly degraded by a protease complex, which has many substrates consisting of twenty-eight 20 to 30 kDa subunits, referred to as the 20S proteasome.8, 9 The proteasomal degradation is essential for many cellular processes, including the cell cycle, regulation of gene expression, and immunological function.10 Interferon (IFN)-g treatment induces the expression of large numbers of responsive genes, the b-ring subunits of proteasome, i.e., low-molecular mass polypeptide (LMP)2/b1i, LMP7/b5i, and LMP10/multicatalytic endopeptidase complex-like (MECL)-1/b2i.11 A molecular approach to studying the correlation of IFN-g with tumor cell growth has drawn attention. Homozygous mice deficient in LMP2/b1i show tissue- and substrate-dependent abnormalities in the biological functions of the proteasome.12 Ut-LMS was reported in female LMP2/b1i-deficientmice at 6 months or older, with its incidence at 14 months being approximately 40%.13 Histopathological studies of LMP2/b1i-lacking uterine tumors have revealed the characteristic abnormalities of Ut-LMS.13 These tumors consist of uniform elongated myometrium cells arranged into bundles. The nuclei of tumor cells vary in size and shape, and mitosis is frequent. In contrast, the myometrium cells of its parental mice, C57BL/6 mice were shown to be normal in appearance. Whereas relatively few ki-67-positive cells, the proliferating cells of solid tumors, have been reported in the basal cell layer of a normal myometrium, the expression of ki-67/MIB1 was marked in most basal cells in LMP2/b1i-deficient mice.13 Marked body weight loss has been reported in LMP2/b1i-deficientmice that develop Ut-LMS, and these mice die by 14 months of age. The LMP2/b1i-deficientmice also exhibit skeletal muscle metastasis from Ut-LMS. Therefore, these research findings suggest that LMP2/b1i-deficient mice with Ut-LMS die as a result of the tumor mass and metastasis.

The non-standard subtypes of uterine mesenchymal tumors such as the epithelioid and myxoid types are classified in a different manner using these features; therefore, a diagnostic method that can the identify non-standard smooth muscle differentiation needs to be established.5, 6 Pathological studies have been performed to demonstrate the validity and reliability of LMP2/b1i as a diagnostic biomarker when combined with other candidate molecules, such as cyclin E and calponin h1, which reportedly function as anti-oncogenic factors in human Ut-LMS. Pathological examinations revealed that the ability to induce the expression of LMP2/b1i and calponin h1 was markedly lower in human Ut-LMS tissues than in uterine LMA or a normal myometrium located in the same section, and markedly expression of cyclin E in human Ut-LMS tissues only.14, 15, 16, 17 Histological findings for the skeletal muscle and rectum lesions were consistent with metastatic Ut-LMS.14, 15 Western blotting and RT-PCR experiments revealed that LMP2/b1i was expressed in a normal myometrium, but not in human Ut-LMS, and both findings strongly supported the pathological results.14, 15, 17, 18 Although we has previously demonstrated that abnormal expression of the ovarian steroid receptors, Tp53, ki-67 and mutations in Tp53 were frequently associated with Ut-LMS, defective LMP2/b1i expression appears to be more characteristic of human Ut-LMS than these factors.15, 16

A female hormonal imbalance is often a risk factor for development of tumors in the case of gynecological cancers.7 As in the case of uterine LMA, however, a correlation between the development of Ut-LMS, the female hormone, and hormone receptors has yet to be elucidated. Recent study reported the expression of Lmp2/b1i mRNA and protein in luminal and glandular epitheliua, placenta villi, trophoblastic shells, and arterial endothelial cells.19, 20, 21 These findings implicate LMP2/b1i in the invasion of placental villi, degradation of the extracellular matrix, immune tolerance, glandular secretion, and angiogenesis, but no more information for sarcomagenesis. Further studies should help to elucidate the molecular mechanism of human Ut-LMS tumorigenesis involved biological significance of LMP2/b1i; we are currently investigating the reliability and characteristics of LMP2/b1i as a diagnostic indicator with several clinical research facilities. Histopathological characteristics of human uterine mesenchymal tumors including mitotically active leiomyoma, bizarre leiomyoma, lipoleiomyoma, undifferentiated endometrial sarcoma, epithelioid variant leiomyosarcoma, myxoid variant leiomyosarcoma, smooth muscle tumors of uncertain malignant potential (STUMP), leiomyomatoid angiomatous neuroendocrin tumor (LANT) have been summarized already22, 23, 24, 25 (Table 1, Table 2). Clarifying correlations between these factors and the development of human Ut-LMS and the identifying specific risk factors may lead to the development of new clinical treatments for the disease.

Table 1. Differential protein expression in Uteriene tumours. SMA, smooth muscle actin; MSA, muscle specific actin; ER/PR, estrogen receptor/ progesterone receptor; Endoglin, CD105/TGFb receptor (stem cell marker); EGFR, epidermal growth factor receptor; LMP2, low-molecular mass polypeptide; CD56, neural cell adhesion molecule (N-CAM); WT-1, wilms tumor 1; NOS, not otherwise specified; MF, magnification factor; HPF, high power field; Foc., focal ; STUMP, smooth muscle tumors of uncertain malignant potential. Protein expression*, estimated-protein expressions by immunoblot analysis, immunohistochemistry (IHC) and/or RT-PCR (quantitative-PCR), +/-, partial expression; +, expression; ++, medium expression; +++, high expression; -, no evidence of expression; ER/PR(ref.16), LMP2(ref.14,15), cyclin E(ref.16,28), calponin h1(ref.17,26,27), Ki-67(ref.16,29). STUMP#(ref.29,30). Cyclin E, LMP2, calponin h1 are potential bio-marker for human uterine mesenchymal tumours. LANT##, leiomyomatoid angiomatous neuroendocrin tumor (LANT) is desvribed as a dimorphic neurosecretory tumor with a leiomyomatous vascular component (ref.22,23).
Tumor type protein expression*
Cytokeratin Desmin MSA SMA Vimentin ER/PR Endoglin EGFR Cyclin B1 Cycline E LMP2 Calponin h/ Ki-67
Endometrial stromal tumors
Endometrial stromal nodule + - + + + +++ + * + - ++ ++ -
Endometrial stromal sarcoma +/- - + +/- + +++ + + + - +/- ++ +/-
Undifferentiated endometrial sarcoma +/- Foc. * - - - + + + + +/- + +
Smooth muscle tumors
Leiomyoma, NOS foc. + + + * +++ +/- +/- + - ++ ++ +/-
Mitotically active leiomyoma * + + + * +++ +/- +/- + - ++ + +/-
Cellular leiomyoma * + + + * +++ +/- +/- + - ++ + +/-
Hemorrhagic cellular leiomyoma * + + + * +++ +/- +/- + - ++ + +/-
Epithelioid leiomyoma * + + + * +++ +/- +/- * - ++ ++ +/-
Myxoid leiomyoma * * * * * * +/- +/- * + +/- + +
Atypical leiomyoma - + + + * +++ + +/- + + +/- +/- +
Lipoleiomyoma * + + + * +++ * +/- * + +/- * +/-
STUMP# * + + + * * * +/- * +/- +/- * +/-
* + + + * * * +/- * +/- +/- * +/-
* + + + * * * +/- * +/- +/- * +/-
* + + + * * * +/- * +/- +/- * +/-
Leiomyosarcoma + + * +/- - - + +/- ++ +++ - - ++
Leiomyosarcoma, epithelioid variant + + * +/- - - + +/- ++ +++ - - ++
Leiomyosarcoma, myxoid variant + + * +/- - - + +/- ++ ++ - - ++
leiomyomatoid tumor
LANT## - - - + + * * + * ++ - - +/-

Table 2. LMP2 expression levels in human myometrium, uterine leiomyoma (Ordinaly leiomyoma, Cellular leiomyoma, Tumor of uncertain malignant potential), bizarre leiomyoma, uterine leiomyosarcoma.
  Age n LMP2 expression
- -/+1 focal+2 +++3
Normal 32~83 59       57
Leiomyoma (Ordinaly Ieiomyoma) (Cellular leiomyoma) (Tumor d uncertain malignant potential) 33~83 53 (32) (9) (12)       53
Bizarre Leiomyoma 44,49,55 3       3
Leiomyosarcoma 32~83 59 51 2 4 2

-/+1 : partially positive (5% to 10% of cells stained)
focal+ : focal-positive (focal or sporadic staining with less than 5% of cells stained)
+++ : diffuse-positive (homogeneous distribution with more than 90% of cells stained)
- : negative (no stained cells)

Acknowledgements

This study was supported in part by grants from the Ministry of Education, Culture, Science and Technology, and The Foundation of Osaka Cancer Research, and The foundation for the Promotion of Cancer Research, The Kanzawa Medical Research Foundation and The Takeda Foundation for Medical Science.

References

  1. 1.Zaloudek C, M R Hendrickson. (2002) Mesenchymal tumors of the uterus, in Kurman RJ.(ed):. Blaustein`s Pathology of the Female Genital Tract (ed 5) , New York, Springer-Verlag 5, 561-578.
  1. 2.T I Wu, T C Chang, Hsueh S, K H Hsu, H et al. (2006) Prognostic factors and impact of adjuvant chemotherapy for uterine leiomyosarcoma. , Gynecol Oncol 100, 166-172.
  1. 3.Leitao M M, Soslow R A, Nonaka D, Olshen A B, Aghajanian C et al. (2004) Tissue microarray immunohisto chemical expression of estrogen, progesterone, and androgen receptors in uterine leiomyomata and leiomyosarcoma. , Cancer 101, 1455-1462.
  1. 4.. http://cancer.gov/cancertopics/pdq/treatment/uterinesarcoma/HealthProfessional
  1. 5.R J Kurma. (2001) . Pathology of the Female Genital Tract, 4th ed , New York, Springer-Verlag 4, 499.
  1. 6.Diagnostic Criteria for LMS, Adapted from 2003, WHO Guidelines:(2003) World Health Organization Classification of Tumours:. Pathology and Genetics, Pathology and Genetics of Tumours of the Breast and Female Genital Organs.IARC Press,France .
  1. 7.J F Lin, B M Slomovitz. (2008) Uterine sarcoma. , Curr. Oncol. Rep 10, 512-518.
  1. 8.Peters J M, Franke W W, Kleinschmidt J A. (1994) Distinct 19S and 20S subcomplexes of the 26S proteasome and their distribution in the nucleus and the cytoplasm. , J. Biol. Chem 269, 7709-7718.
  1. 9.Lodish H, Berk A, Matsudaira P, Kaiser C A, Krieger M et al. (2004) . 3".Mol Cell Biol (5th ed.).New York:W.H. Freeman and CO 5, 66-72.
  1. 10.I M Konstantinova, A S Tsimokha, A G Mittenberg. (2008) Role of proteasomes in cellular regulation. , Intl. Rev. Cell Mol. Biol 267, 59-124.
  1. 11.Wang J, Maldonado M A. (2006) The Ubiquitin-Proteasome System and Its Role in Inflammatory and Autoimmune Diseases. Cell Mol. , Immunol 3, 255-261.
  1. 12.L Van Kaer, P G Ashton-Rickardt, Eichelberger M, Gaczynska M, Nagashima K et al. (1994) Altered peptidase and viral-specifi c T cell response in LMP2 mutant mice. , Immunity 1, 533-541.
  1. 13.Hayashi T, D L Faustman. (2002) Development of spontaneous uterine tumors in low molecular mass polypeptide-2 knockout mice. , Cancer Res 62, 24-27.
  1. 14.Hayashi T, Kobayashi Y, Kohsaka S, Sano K. (2006) Mutation in the ATP-binding region of JAK1, identified in human uterine leiomyosarcomas, results in defective interferon-gamma inducibility of TAP1 and LMP2. , Oncogene 25, 4016-4026.
  1. 15.Hayashi T, Horiuchi A, Sano K, Hiraoka N, Kasai M et al. (2011) Potential role of LMP2 as tumor-suppressor defines new targets for uterine leiomyosarcoma therapy. Sci. Rep.2011, 1,180| DOI: 10.1038/srep00180 .
  1. 16.Y L Zhai, Kobayashi Y, Mori A, Orii A, Nikaido T et al. (2004) Expression of steroid receptors, Ki-67, and p53 in uterine leiomyosarcomas. , Intl J Gynecol Pathol 18, 20-28.
  1. 17.Hayashi T, Horiuchi A, Sano K, Hiraoka N, Kasai M et al. (2012) Potential role of LMP2 as an anti-oncogenic factor in human uterine leiomyosarcoma: morphological significance of calponin h1. , FEBS Letter 586, 1824-1831.
  1. 18.Hayashi T, Horiuchi A, Konishi I. (2013) Tumor growth arrest: involvement of the mutation in the cataritic region of JAK1. , Europian Journal of Research in Medical Science 1, 8-21.
  1. 19.H X Wang, H M Wang, Q L Li, P L Judoson. (2004) Expression of proteasome subunits low molecular mass polypeptide (LMP) 2 and LMP7 in the endometrium and placenta of rhesus monkey (Macaca mulatta) during early pregnancy. , Biol. Reprod 71, 1317-1324.
  1. 20.H X Wang, H M Wang, H Y Lin, Yang Q, Zhang H et al. (2006) Proteasome subunit LMP2 is required for matrix metalloproteinase-2 and -9 expression and activities in human invasive extravillous trophoblast cell line. , J. Cell Physiol 206, 616-623.
  1. 21.Fu J J, Lin P, X Y Lv, X J Yan, H X Wang et al. (2009) Low molecular mass polypeptide-2 in human trophoblast: over-expression in hydatidiform moles and possible role in trophoblast cell invasion. , Placenta 30, 305-312.
  1. 22.Sakashita N, Yamada M, Nakagawa T, Yamasaki H, M A Takeya. (2008) leiomyomatoid angiomatous neuroendocrine tumor of the myometrium: case study with ultrastructural analysis. , Hum. Pathol 39, 788-792.
  1. 23.Vajtai I, Sahli R, Kappeler A, E R Christ, R W Seiler. (2006) Leiomyomatoid angiomatous neuroendocrine tumor (LANT) of the pituitary: a distinctive biphasic neoplasm with primitive secretory phenotype and smooth muscle-rich stroma. , Acta Neuropathol 111, 278-283.
  1. 24.Avritscher R, R B Iyer1, Ro J, Whitman G. (2001) . , Lipoleiomyoma of the Uterus. Am. J. Radiol 177, 856.
  1. 25.Takeuchi M, Matsuzaki K, Harada M. (2013) Preliminary observations and clinical value of lipid peak in high-grade uterine sarcomas using in vivo proton MR spectroscopy. , Eur. Radiol 23, 2358-2363.
  1. 26.Horiuchi A, Nikaido T, Ito K, Zhai Y, Orii A et al. (1998) Reduced expression of calponin h1 in leiomyosarcoma of the uterus. , Lab. Invest 78, 839-846.
  1. 27.Horiuchi A, Nikaido T, Taniguchi S, Fujii S. (1999) Possible role of calponin h1 as a tumor suppressor in human uterine leiomyosarcoma. , J. Natl. Cancer Inst 91, 790-796.
  1. 28.Zhai Y L, Nikaido T, Shiozawa T, Orii A, Fujii S. (1999) Expression of cyclins and cyclin-dependent kinases in smooth muscle tumors of the uterus. , Intl. J. Cancer 84, 224-250.
  1. 29.Ip P P, Cheung A N, Clement P B. (2009) Uterine smooth muscle tumors of uncertain malignant potential (STUMP): a clinicopathologic analysis of 16 cases. , Am. J. Surg. Pathol 33, 992-1005.
  1. 30.Ip P P, K Y Tse, K F Tam. (2010) Uterine smooth muscle tumors other than the ordinary leiomyomas and leiomyosarcomas: a review of selected variants with emphasis on recent advances and unusual morphology that may cause concern for malignancy. , Adv. Anat. Pathol 17, 91-112.