Skip to main content

Advertisement

Log in

Etiology, Pathogenesis, and Malignant Potential of Uterine Leiomyoma – A Review

  • Management of Uterine Fibroid (I Meinhold-Heerlein, Section Editor)
  • Published:
Current Obstetrics and Gynecology Reports Aims and scope Submit manuscript

Abstract

In uterine smooth muscle tumors (SMT), benign leiomyoma (LM) consist of smooth muscle cells, side population cells, and fibroblasts acting together within a network regulating cell proliferation and production of extracellular matrix molecules. Chromosomal alterations, activation of the WNT/ß-catenin pathway, and overexpression of genes of the HMG group are important pathogenetic steps. The features of malignancy are tumor cell necrosis (TCN), cytological atypia, and mitotic activity. However, hormones and hypoxic stress induce reactive mitotic activity. Therefore, the mitotic index by itself is not an independent predictor of malignancy. SMT with mitotic activity up to 15 mitoses/10 HPF in the absence of atypia and TCN are designated mitotically active LM. These tumors are mostly benign. SMT with cytological atypia and a mitotic count below 10 figures /10 HPF without TCN are classified as atypical LM. Tumors with questionable TCN or without TCN and atypia but with a mitotic activity of > 15 figures/10 HPF belong to the SMT of “uncertain malignant potential”. In the presence of definite TCN, LMS are diagnosed irrespective of mitotic count. In the absence of TCN, a mitotic activity of ≥ 10 figures /10 HPF and severe atypia argue for LMS. Uterine LMS are not graded. Whether uterine LMS arise from LM or de novo is still debated, although molecular studies suggest that both are formed through distinct molecular pathways.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Cramer SF, Patel A. The frequency of uterine leiomyomas. Am J Clin Pathol. 1990;94:435–8.

    CAS  PubMed  Google Scholar 

  2. Laughlin SK, Schroeder JC, Baird DD. New directions in the epidemiology of uterine fibroids. Semin Reprod Med. 2010;28:204–17.

    Article  CAS  PubMed  Google Scholar 

  3. Ryan GL, Syrop CH, Van Voorhis BJ. Role, epidemiology, and natural history of benign uterine mass lesions. Clin Obstet Gynecol. 2005;48:312–24.

    Article  PubMed  Google Scholar 

  4. Schwartz SM. Epidemiology of uterine leiomyomata. Clin Obstet Gynecol. 2001;44:316–26.

    Article  CAS  PubMed  Google Scholar 

  5. Parker WH. Etiology, symptomatology, and diagnosis of uterine myomas. Fertil Steril. 2007;87:725–36.

    Article  PubMed  Google Scholar 

  6. Wallach EE, Vlahos NF. Uterine myomas: an overview of development, clinical features, and management. Obstet Gynecol. 2004;104:393–406.

    Article  PubMed  Google Scholar 

  7. Andersen J. Growth factors and cytokines in uterine leiomyomas. Semin Reprod Endocrinol. 1996;14:269–82.

    Article  CAS  PubMed  Google Scholar 

  8. Rein MS, Barbieri RL, Friedman AJ. Progesterone: a critical role in the pathogenesis of uterine myomas. Am J Obstet Gynecol. 1995;172:14–8.

    Article  CAS  PubMed  Google Scholar 

  9. Fields KR, Neinstein LS. Uterine myomas in adolescents: case reports and a review of the literature. J Pediatr Adolesc Gynecol. 1996;9:195–8.

    Article  CAS  PubMed  Google Scholar 

  10. Ip PP et al. 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. 2010;17:91–112.

    Article  PubMed  Google Scholar 

  11. Baird DD, Dunson DB. Why is parity protective for uterine fibroids? Epidemiology. 2003;14:247–50.

    PubMed  Google Scholar 

  12. Wise LA, Palmer JR, Harlow BL, et al. Reproductive factors, hormonal contraception, and risk of uterine leiomyomata in African-American women: a prospective study. Am J Epidemiol. 2004;59:113–23.

    Article  Google Scholar 

  13. Catherino WH, Eltoukhi HM, Al-Hendy A. Racial and ethnic differences in the pathogenesis and clinical manifestations of uterine leiomyoma. Semin Reprod Med. 2013;31:370–9.

    Article  PubMed  Google Scholar 

  14. Hodge JC, Park PJ, et al. Identifying the molecular signature oft he interstitial deletion 7q subgroup of uterine leiomyoma using a paired analysis. Genes Chromosom Cancer. 2009;48:865–85.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  15. Cleynen I, van de Ven WJM. The HMGA proteins: a myriad of functions. Int J Oncol. 2008;32:289–305.

    CAS  PubMed  Google Scholar 

  16. Segars JH et al. Proceedings from the third national institutes of health international congress on advances in uterine leiomyoma research: comprehensive review, conference summary and future recommendations. Hum Reprod Update. 2013;20:309–33.

    Article  Google Scholar 

  17. Velegaleti GV, Tonk VS, et al. Fusion of HMGA2 to COG5 in uterine leiomyoma. Cancer Genet Cytogenet. 2010;202:11–6.

    Article  Google Scholar 

  18. Makinen N, Mehine M, et al. MED12, the mediator complex subunit 12 gene, is mutated in high frequency in uterine leiomyoma. Science. 2011;334:252–5.

    Article  PubMed  Google Scholar 

  19. Makinen N, Vahteristo P, Bützow R, Sjöberg J, Aaltonen LA. Exomic landscape of MED12 mutation-negative and -positive uterine leiomyomas. Int J Cancer. 2014;134:1008–12.

    PubMed  Google Scholar 

  20. Kim S, Xu X, Hecht A, Boyer TG. Mediator is a transducer of Wnt/beta-catenin signaling. J Biol Chem. 2006;281:14066–75.

    Article  CAS  PubMed  Google Scholar 

  21. Catherino W, Salama A, Potlog-Nahari C, Leppert P, Tsibris J, Segars J. Gene expression studies in leiomyomata: new directions for research. Semin Reprod Med. 2004;22:83–90.

    Article  CAS  PubMed  Google Scholar 

  22. Markowski DN et al. MED12 mutations in uterine fibroids—their relationship to cytogenetic subgroups. Int J Cancer. 2012;131:1528–36.

    CAS  PubMed  Google Scholar 

  23. Ishikawa H, Reierstad S, et al. High aromatase expression in uterine leiomyoma tissues of African-American woman. J Clin Endocrinol Metab. 2009;94:1752–6.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  24. Orthman EE, Al-Hendy A. Molecular genetics and racial disparities of uterine leiomyomas. Best Pract Res Clin Obstet Gynecol. 2008;22:589–601.

    Article  Google Scholar 

  25. Yin P, Lin Z, et al. Progesteron receptor regulates Bcl-2 gene expression through direct binding to ist promotor region in leiomyoma cells. J Clin Endocinol Metab. 2007;92:4459–66.

    Article  CAS  Google Scholar 

  26. Yin P, Roqueiro D, et al. Genome-wide progesteron receptor binding: cell type-specific and shared mechanismsin T47D breast cancer cells and primary leiomyoma cells. PLoS One. 2012;7:e29021.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  27. Qiang W, Liu Z, Serna VA, Druschitz SA, Liu Y, Espona-Fiedler M, et al. Down-Regulation of miR-29b is Essential for Pathogenesis of Uterine Leiomyoma. Endocrinology. 2014;155:663–9.

    Article  PubMed  Google Scholar 

  28. Luo N, Guan Q, Zheng L, Qu X, Dai H, Cheng Z. Estrogen-mediated activation of fibroblasts and its effects on the fibroid cell proliferation. Transl Res. 2014;163:232–41.

  29. Mas A et al. Identification and characterization of the human leiomyoma side population as putative tumor-initiating cells. Fertil Steril. 2012;98:741–51.

    Article  PubMed  Google Scholar 

  30. Cervelló I et al. Reconstruction of endometrium from human endometrial side population cell lines. PLoS ONE. 2011;6(6):e21221.

    Article  PubMed Central  PubMed  Google Scholar 

  31. Ono M et al. Side population in human uterine myometrium displays phenotypic and functional characteristics of myometrial stem cells. Proc Natl Acad Sci U S A. 2007;104:18700–5.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  32. Ono M et al. Role of stem cells in human uterine leiomyoma growth. PLoS ONE. 2012;7(5):e36935.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  33. Smalley MJ, Clarke RB. The mammary gland “side population”: a putative stem/progenitor cell marker? J Mammary Gland Biol Neoplasia. 2005;10:37–47.

    Article  PubMed  Google Scholar 

  34. Gottardi CJ, Königshoff M. Considerations for targeting β-catenin signaling in fibrosis. Am J Respir Crit Care Med. 2013;187:566–8.

    CAS  PubMed  Google Scholar 

  35. Lam AP, Gottardi CJ. β-catenin signaling: a novel mediator of fibrosis and potential therapeutic target. Curr Opin Rheumatol. 2011;23:562–7.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  36. Tanwar PS et al. Constitutive activation of Beta-catenin in uterine stroma and smooth muscle leads to the development of mesenchymal tumors in mice. Biol Reprod. 2009;81:545–52.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  37. Schwab KE, Chan RW, et al. Putative stem cell activity of human endometrial epithelial and stromal cells during the menstrual cycle. Fertil Steril. 2005;84 Suppl 2:1124–30.

    Article  CAS  PubMed  Google Scholar 

  38. Catherino WH et al. Reduced dermatopontin expression is a molecular linkbetween uterine leiomyomas and keloids. Genes Chromosom Cancer. 2004;40:204–17.

    Article  CAS  PubMed  Google Scholar 

  39. Leppert PC, Catherino WH, Segars JH. A new hypothesis about the origin of uterine fibroids based on gene expression profiling with microarrays. Am J Obstet Gynecol. 2006;195:415–20.

    Article  CAS  PubMed  Google Scholar 

  40. Tal R, Segars JH. The role of angiogenic factors in fibroid pathogenesis: potential implications for future therapy. Hum Reprod Update. 2014;20:194–216.

    Article  CAS  PubMed  Google Scholar 

  41. Cesen-Cummings K et al. Lessons from pregnancc and parturition: uterine leiomyomas result from discordant differentiation and dedifferentiation responses in smooth muscle cells. Med Hypotheses. 2000;55:485–90.

    Article  CAS  PubMed  Google Scholar 

  42. Koivisto-Korander R et al. Incidence of uterine leiomyosarcoma and endometrial stromal sarcoma in Nordic countries: results from NORDCAN and NOCCA databases. Maturitas. 2012;72:56–60.

    Article  CAS  PubMed  Google Scholar 

  43. Lin J, Song X, Liu C. Pelvic intravascular leiomyomatosis associated with benign pulmonary metastasizing leiomyoma: clinicopathologic, clonality, and copy number variance analysis. Int J Gynecol Pathol. 2014;33:140–5.

    Article  PubMed  Google Scholar 

  44. Chiang S, Oliva E. Recent developments in uterine mesenchymal neoplasms. Histopathology. 2013;62:124–37.

    Article  PubMed  Google Scholar 

  45. Zaloudek CJ, Hendrickson MR, Soslow RA. Mesenchymal tumors of the uterus. In: Kurman RJ, Ellenson LH, Ronnett BM, editors. Blaustein’s pathology of the female genital tract. 6th ed. New York: Springer; 2011. p. 453–527.

    Chapter  Google Scholar 

  46. Roth LM et al. Recurrent cotyledonoid dissecting leiomyoma of the uterus. Int J Gynecol Pathol. 2013;32:215–20.

    Article  PubMed  Google Scholar 

  47. Giuntoli RL et al. Diagnostic criteria for uterine smooth muscle tumors: leiomyoma variants associated with malignant behavior. J Reprod Med. 2007;52:1001–10.

    PubMed  Google Scholar 

  48. Ly A et al. Atypical leiomyomas of the uterus: a clinicopathologic study of 51 cases. Am J Surg Pathol. 2013;37:643–9.

    Article  PubMed  Google Scholar 

  49. Veras E, Zivanovic O, Jacks L, Chiappetta D, Hensley M, Soslow R. “Low-grade leiomyosarcoma” and late-recurring smooth muscle tumors of the uterus: a heterogenous collection of frequently misdiagnosed tumors associated with an overall favorable prognosis relative to conventional uterine leiomyosarcomas. Am J Surg Pathol. 2011;35:1626–37.

    Article  PubMed  Google Scholar 

  50. Oduyebo T et al. The value of re-exploration in patinets with inadvertently morcellated uterine sarcoma. Gynecol Oncol. 2014;132:360–5.

    Article  PubMed  Google Scholar 

  51. Seidman MA et al. Peritoneal dissemination complicating morcellation of uterine mesenchymal neoplasms. PLoS ONE. 2012;7:e50058.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  52. Czarkowski K, Chetty N, Berkes E, Hackethal A. Role and Risks of Morcellation Associated with Laparoscopic Management of Myomas. Curr Obstet Gynecol Rep. doi:10.1007/s13669-014-0084-9

  53. Wang WL et al. Histopathologic prognostic factors in stage I leiomyosarcoma of the uterus: a detailed analysis of 27 cases. Am J Surg Pathol. 2011;35:522–9.

    Article  PubMed  Google Scholar 

Download references

Compliance with Ethics Guidelines

Conflict of Interest

Steffen Hauptmann and Günter Köhler declare that they have no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Steffen Hauptmann.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hauptmann, S., Köhler, G. Etiology, Pathogenesis, and Malignant Potential of Uterine Leiomyoma – A Review. Curr Obstet Gynecol Rep 3, 186–190 (2014). https://doi.org/10.1007/s13669-014-0091-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13669-014-0091-x

Keywords

Navigation