Skip to main content
  • Minireview
  • Published:

The new molecular biology of granulosa cell tumors of the ovary

Abstract

Granulosa cell tumors (GCTs) of the ovary belong to the group of ovarian sex-cord stromal tumors and represent 5 to 10% of ovarian malignancies. GCTs exhibit several morphological, biochemical and hormonal features of normal proliferating pre-ovulatory granulosa cells, such as estrogen biosynthesis. Prognostic factors of this condition are lacking, and alternative treatment options to preserve future fertility are needed. Several groups have shown that two genetic factors implicated in GCTs are of particular interest. The gsp oncogene is a constitutive activating mutation of the prognosis of the tumor. FOXL2 is a transcription factor gene involved in ovarian development and function, whose expression is reduced and which is mutated in the majority of GCTs. FOXL2 appears to play a major role in cell cycle regulation. These recent findings open new pathophysiological insights into GCT development as well as revisitation of granulosa cell and ovarian function.

Natural history of granulosa cell tumors of the ovary

Granulosa cell tumors (GCTs) of the ovary are relatively uncommon neoplasms, representing approximately 5 to 10% of all ovarian malignancies [1, 2]; the others are germ cell tumors (teratomas and yolk sac tumors, 60 to 70%) and epithelial adenomas (10 to 20%) [3]. GCTs of the ovary belong to the group of ovarian sex-cord stromal tumors. Other tumors in this class are thecoma-fibromas, Sertoli cell tumors, sex cord tumors with annular tubules, and gynandroblastomas. The majority of patients with GCTs are adults, but 5% are pre- or peri-pubertal [4]. Consequently, two histological types of GCTs can be distinguished on a histopathological basis: juvenile and adult. GCTs exhibit several morphological, biochemical and hormonal features of normal proliferating pre-ovulatory granulosa cells, including both estrogen and inhibin biosynthesis [5]. This tumoral hyperestrogenism induces precocious puberty in children (premature breast development, vaginal bleeding) and advanced growth and bone maturation. The prognosis of juvenile GCTs is excellent overall: tumor recurrence and metastasis are rare and usually occur early [6]. On the other hand, adult GCTs are low-grade indolent malignant neoplasms that display a significant propensity for recurrence and metastasis, and they may cause menorrhagia or inter-menstrual bleeding. The cornerstone of treatment remains surgery. The disease in young patients is often confined to one ovary and thus, in order to preserve fertility, a unilateral salpingo-oophorectomysubunit andis is preferable.correlated In the case of post-menopausal women, a total abdominal hysterectomy and bilateral salpingo-oophorectomy should be performed [7].

Challenges in GCT management

Nevertheless, management of patients with ovarian GCTs needs to be improved. First, the recurrent or metastatic tumor may manifest many years after removal of the primary neoplasm, with intervals of 10 or even 20 years being not uncommon, especially in the adult disease [8, 9]. Clinical follow-up of these patients is thus critical, but prognostic factors are lacking and assessment of the recurrence risk remains imprecise. For instance, in International Federation of Obstetricians and Gynecologists (FIGO) stage I disease (that is, intra-ovarian tumor with no extension, the most frequent stage at diagnosis according to FIGO), no association between outcome and the clinical signs or tumor histology (mitotic, nuclear atypia, ploidy, Ki67 expression) has been undisputedly demonstrated [10, 11]. Furthermore, in more advanced disease, aggressive debulking surgery and postoperative chemotherapy may be required [8]. Alternative treatment options and the use of molecular markers are thus necessary, especially for young women whose fertility should be preserved.

FOXL2 and GCTs of the ovary

FOXL2 is a recent candidate gene in the pathophysiology of GCTs and could prove useful to evaluate their prognosis. Indeed, FOXL2 is a winged helix/forkhead transcription factor gene involved in ovarian development and function [12, 13]. FOXL2 ovarian expression in mammals starts before the morphological differentiation of the gonad is recognizable, and persists until adulthood, mainly in granulosa cells [14, 15]. FOXL2 is thought to be a key factor in the early development and maintenance of the vertebrate ovary [12, 15]. Its expression in granulosa cells is suppressed, or at least lowered, in the majority of juvenile GCTs, particularly in those with the most aggressive pattern of progression [16]. Similar expression studies have yet to be performed in adult GCTs.

Interestingly, Shah et al. [17] recently studied four adult GCTs using whole-transcriptome paired-end RNA sequencing. After removing the previously described germline genetic variants and non-specific mutations present in non-GCTs, the authors identified the somatic missense point mutation c.402C→G (p.C134W) in all four specimens. Subsequently, they used direct sequencing, restriction fragment length polymorphism analyses (RFLPs), and a real-time PCR-based allelic discrimination assay to genotype FOXL2 in a large collection of tumors. The p. C134W mutation was found in 97% of 89 additional adult GCTs studied, and was detected in only one case out of eight juvenile GCTs. The mutation was also found in 21% of thecomas, that is, 3 out of 14, in the study of Shah et al. Further studies are needed to determine the link between this mutation and GCTs, and whether this mutation alone is sufficient to induce tumor progression.

Other genetic factors implicated in ovarian GCTs

Genetic defects in other signaling pathways have been investigated in GCTs. Although Peutz-Jeghers syndrome is associated with an increased risk for GCTs, neither allele loss at the disease locus (19p13.3) nor mutations in the LKB1 gene are associated with sporadic GCTs [18, 19]. Similarly, despite the association of epithelial ovarian cancer with the familial breast cancer genes BRCA1 and BRCA2, sporadic mutations have not been reported for GCTs.

Other interesting candidate pathways are those involving the mitogen-activated protein kinases [20] and growth factors, because they are known to induce increased oncogene functioning or a loss-of-anti-oncogene function in several solid tumors. However, King et al. [21] failed to identify any prognostic value for the oncogenes c-myc, p21-ras, c-erb B2, and p53 in a group of 40 GCTs. Similarly, the Wilms' tumor suppressor gene (WT1) and TP53 genes, which play a role in follicular development, have been tested [22]. WT1 is expressed in GCT, but neither mutations nor loss of heterozygosity have been identified [23]. TP53 mutations are frequent in numerous cancers, especially in epithelial tumors of the ovary, but there is neither hyperexpression [24] nor mutation [25] of this gene in GCT.

The role of the follicle-stimulating hormone (FSH) signaling pathway is also strongly suspected in GCTs for two reasons: firstly, normal granulosa cells are under the control of FSH and, secondly, the gene expression profile of GCTs is consistent with a constitutive activation of FSH signaling [26]. However, there is no evidence of activating mutations in the gene encoding the FSH receptor [2729]. FSH signaling involves the coupling of heterotrimeric G proteins to activate intracellular second messenger systems, mainly the cAMP-dependent protein kinase (PKA).

Conflicting data have been reported regarding the presence of a constitutive activating mutation of the Gsα subunit in GCTs, the so-called gsp oncogene. It has rarely been described in adult GCTs [30, 31], but our group has identified it frequently in the juvenile type, where mutations in hot spot position 201 (p.R201C or p.R201H) were found in 30% of patients [32]. The precise role of this mutation in the transformation of ovarian cells into malignant cells remains unclear, but it has been demonstrated in other tissues that the rate of cell proliferation and invasiveness can be influenced by the constitutive activation of Gαs [33]. The oncological stages are indeed significantly different according to the gsp oncogene status. Patients with a cantly more advanced tumors, and almost 80% of gsp-positive patients present with an extra-ovarian extension (stage Ic) or have a recurrence [32].

Potential diagnostic and therapeutic applications

The clinical applications of these findings may be promising in the near future, for several reasons. Firstly, identification of a recurrent mutation in FOXL2 may be used as another diagnostic tool for adult GCTs, in addition to the classical pathological and immunohistochemical features. It may be particularly useful to explore granulosa tumor participation in heterogeneous ovarian non-germinal tumors with components of different origins. Also, the low frequency of the FOXL2 mutation in juvenile GCTs compared with adult cases, as reported by Shah and collaborators [17], supports the distinction made between the juvenile type and the adult type based on clinical presentation, pathological features and natural progression [1]. Clearly, differentiating the two types of tumor is clinically relevant, since relapses of adult GCTs are more frequent and may occur later than juvenile GCTs. The mutational or expressional status of FOXL2 could thus be of importance in adapting the length of postoperative follow-up. Another genetic abnormality that could be critical in the follow-up of these patients is the gsp oncogene status, especially for juvenile GCTs. A constitutively activated Gsα is indeed involved in the natural history of the tumor and is a prognostic factor of its extension. Finally, the finding of both extinction [16] and mutation [17] of FOXL2 in GCTs highlights the role of this gene in the regulation of cell proliferation. These findings increase the body of evidence implicating the FOX family genes, whether as oncogenes or anti-oncogenes, in malignant processes such as rhabdomyosarcomas [34], secondary acute myeloblastic leukemia [35], and laryngeal, lung, breast [3638] and pancreatic cancers [39]. FOXL2 may act as a transcriptional regulator and a coordinator of SMAD3 downstream targets [40, 41] that, with SMAD2 and the TGF-β superfamily ligands, regulate granulosa cell proliferation [42]. These recently identified steps could be the targets of the next generation of therapies.

Abbreviations

FIGO:

International Federation of Obstetricians and Gynaecologists

FOXL2:

forkhead transcription factor FOXL2

FSH:

follicle-stimulating hormone

GCT:

granulosa cell tumor

PKA:

cAMP-dependent protein kinase

RFLP:

restriction fragment length polymorphism

WT1:

Wilms' tumor suppressor gene.

References

  1. Schumer ST, Cannistra SA: Granulosa cell tumor of the ovary. J Clin Oncol. 2003, 21: 1180-1189. 10.1200/JCO.2003.10.019

    Article  PubMed  Google Scholar 

  2. Colombo N, Parma G, Zanagnolo V, Insinga A: Management of ovarian stromal cell tumors. J Clin Oncol. 2007, 25: 2944-2951. 10.1200/JCO.2007.11.1005

    Article  CAS  PubMed  Google Scholar 

  3. Merras-Salmio L, Vettenranta K, Mottonen M, Heikinheimo M: Ovarian granulosa cell tumors in childhood. Pediatr Hematol Oncol. 2002, 19: 145-156. 10.1080/088800102753541297

    Article  PubMed  Google Scholar 

  4. Young RH, Dickersin GR, Scully RE: Juvenile granulosa cell tumor of the ovary. A clinicopathological analysis of 125 cases. Am J Surg Pathol. 1984, 8: 575-596.

    Article  CAS  PubMed  Google Scholar 

  5. Fuller PJ, Chu S, Fikret S, Burger HG: Molecular pathogenesis of granulosa cell tumours. Mol Cell Endocrinol. 2002, 191: 89-96. 10.1016/S0303-7207(02)00059-X

    Article  CAS  PubMed  Google Scholar 

  6. Kalfa N, Patte C, Orbach D, Lecointre C, Pienkowski C, Philippe F, Thibault E, Plantaz D, Brauner R, Rubie H, Guedj AM, Ecochard A, Paris F, Jeandel C, Baldet P, Sultan C: A nationwide study of granulosa cell tumors in pre- and postpubertal girls: missed diagnosis of endocrine manifestations worsens prognosis. J Pediatr Endocrinol Metab. 2005, 18: 25-31.

    Article  CAS  PubMed  Google Scholar 

  7. Evans AT, Gaffey TA, Malkasian GD, Annegers JF: Clinicopathologic review of 118 granulosa and 82 theca cell tumors. Obstet Gynecol. 1980, 55: 231-238.

    PubMed  Google Scholar 

  8. Pectasides D, Papaxoinis G, Fountzilas G, Aravantinos G, Pectasides E, Mouratidou D, Economopoulos T, Andreadis C: Adult granulosa cell tumors of the ovary: a clinicopatho-logical study of 34 patients by the Hellenic Cooperative Oncology Group (HeCOG). Anticancer Res. 2008, 28 (2B): 1421-1427.

    CAS  PubMed  Google Scholar 

  9. Hasiakos D, Papakonstantinou K, Karvouni E, Fotiou S: Recurrence of granulosa cell tumor 25 years after initial diagnosis. Report of a case and review of the literature. Eur J Gynaecol Oncol. 2008, 29: 86-88.

    CAS  PubMed  Google Scholar 

  10. Leuverink EM, Brennan BA, Crook ML, Doherty DA, Hammond IG, Ruba S, Stewart CJ: Prognostic value of mitotic counts and Ki-67 immunoreactivity in adult-type granulosa cell tumour of the ovary. J Clin Pathol. 2008, 61: 914-919. 10.1136/jcp.2008.056093

    Article  CAS  PubMed  Google Scholar 

  11. Kalfa N, Sultan C: Juvenile ovarian granulosa cell tumor: a benign or malignant condition?. Gynecol Endoc. 2009, 25: 299-10.1080/09513590802630153.

    Article  Google Scholar 

  12. Schmidt D, Ovitt CE, Anlag K, Fehsenfeld S, Gredsted L, Treier AC, Treier M: The murine winged-helix transcription factor Foxl2 is required for granulosa cell differentiation and ovary maintenance. Development. 2004, 131: 933-942. 10.1242/dev.00969

    Article  CAS  PubMed  Google Scholar 

  13. Uda M, Ottolenghi C, Crisponi L, Garcia JE, Deiana M, Kimber W, Forabosco A, Cao A, Schlessinger D, Pilia G: Foxl2 disruption causes mouse ovarian failure by pervasive blockage of follicle development. Hum Mol Genet. 2004, 13: 1171-1181. 10.1093/hmg/ddh124

    Article  CAS  PubMed  Google Scholar 

  14. Cocquet J, Pailhoux E, Jaubert F, Servel N, Xia X, Pannetier M, De Baere E, Messiaen L, Cotinot C, Fellous M, Veitia RA: Evolution and expression of FOXL2. J Med Genet. 2002, 39: 916-921. 10.1136/jmg.39.12.916

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  15. Cocquet J, De Baere E, Gareil M, Pannetier M, Xia X, Fellous M, Veitia RA: Structure, evolution and expression of the FOXL2 transcription unit. Cytogenet Genome Res. 2003, 101: 206-211. 10.1159/000074338

    Article  CAS  PubMed  Google Scholar 

  16. Kalfa N, Philibert P, Patte C, Ecochard A, Duvillard P, Baldet P, Jaubert F, Fellous M, Sultan C: Extinction of FOXL2 expression in aggressive ovarian granulosa cell tumors in children. Fertil Steril. 2007, 87: 896-901. 10.1016/j.fertnstert.2006.11.016

    Article  CAS  PubMed  Google Scholar 

  17. Shah SP, Köbel M, Senz J, Morin RD, Clarke BA, Wiegand KC, Leung G, Zayed A, Mehl E, Kalloger SE, Sun M, Giuliany R, Yorida E, Jones S, Varhol R, Swenerton KD, Miller D, Clement PB, Crane C, Madore J, Provencher D, Leung P, DeFazio A, Khattra J, Turashvili G, Zhao Y, Zeng T, Glover JN, Vanderhyden B, Zhao C, et al.: Mutation of FOXL2 in granu-losa-cell tumors of the ovary. N Engl J Med. 2009, 360: 2719-2729. 10.1056/NEJMoa0902542

    Article  CAS  PubMed  Google Scholar 

  18. Wang ZJ, Churchman M, Campbell IG, Xu WH, Yan ZY, McCluggage WG, Foulkes WD, Tomlinson IP: Allele loss and mutation screen at the Peutz-Jeghers (LKB1) locus (19p13.3) in sporadic ovarian tumours. Br J Cancer. 1999, 80: 70-72. 10.1038/sj.bjc.6690323

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  19. Kato N, Romero M, Catasus L, Prat J: The STK11/LKB1 Peutz-Jegher gene is not involved in the pathogenesis of sporadic sex cord-stromal tumors, although loss of heter-ozygosity at 19p13.3 indicates other gene alteration in these tumors. Hum Pathol. 2004, 35: 1101-1104. 10.1016/j.humpath.2004.05.011

    Article  CAS  PubMed  Google Scholar 

  20. Kim K, Lindstrom MJ, Gould MN: Regions of H- and K-ras that provide organ specificity/potency in mammary cancer induction. Cancer Res. 2002, 62: 1241-1245.

    CAS  PubMed  Google Scholar 

  21. King LA, Okagaki T, Gallup DG, Twiggs LB, Messing MJ, Carson LF: Mitotic count, nuclear atypia, and immunohisto-chemical determination of Ki-67, c-myc, p21-ras, c-erbB2, and p53 expression in granulosa cell tumors of the ovary: mitotic count and Ki-67 are indicators of poor prognosis. Gynecol Oncol. 1996, 61: 227-232. 10.1006/gyno.1996.0130

    Article  CAS  PubMed  Google Scholar 

  22. Makrigiannakis A, Amin K, Coukos G, Tilly JL, Coutifaris C: Regulated expression and potential roles of p53 and Wilms' tumor suppressor gene (WT1) during follicular development in the human ovary. J Clin Endocrinol Metab. 2000, 85: 449-459. 10.1210/jc.85.1.449

    CAS  PubMed  Google Scholar 

  23. Coppes MJ, Ye Y, Rackley R, Zhao XL, Liefers GJ, Casey G, Williams BR: Analysis of WT1 in granulosa cell and other sex cord-stromal tumors. Cancer Res. 1993, 53: 2712-2714.

    CAS  PubMed  Google Scholar 

  24. Liu FS, Ho ES, Lai CR, Chen JT, Shih RT, Yang CH, Tsao CM: Overexpression of p53 is not a feature of ovarian granu-losa cell tumors. Gynecol Oncol. 1996, 61: 50-53. 10.1006/gyno.1996.0095

    Article  CAS  PubMed  Google Scholar 

  25. Kappes S, Milde-Langosch K, Kressin P, Passlack B, Dockhorn-Dworniczak B, Rohlke P, Loning T: p53 mutations in ovarian tumors, detected by temperature-gradient gel electrophoresis, direct sequencing and immunohistochemis-try. Int J Cancer. 1995, 64: 52-59. 10.1002/ijc.2910640111

    Article  CAS  PubMed  Google Scholar 

  26. Chu S, Rushdi S, Zumpe ET, Mamers P, Healy DL, Jobling T, Burger HG, Fuller PJ: FSH-regulated gene expression profiles in ovarian tumours and normal ovaries. Mol Hum Reprod. 2002, 8: 426-433. 10.1093/molehr/8.5.426

    Article  CAS  PubMed  Google Scholar 

  27. Fuller PJ, Verity K, Shen Y, Mamers P, Jobling T, Burger HG: No evidence of a role for mutations or polymorphisms of the follicle-stimulating hormone receptor in ovarian granu-losa cell tumors. J Clin Endocrinol Metab. 1998, 83: 274-279. 10.1210/jc.83.1.274

    CAS  PubMed  Google Scholar 

  28. Kotlar T, Young RH, Albanese C, Crowley WF, Scully RE, Jameson JL: Absence of mutations in the FSH receptor in ovarian granulosa cell tumors. J Clin Endocrinol Metab. 1998, 83: 3001- 10.1210/jc.83.8.3001

    Article  CAS  PubMed  Google Scholar 

  29. Bas F, Pescovitz OH, Steinmetz R: No activating mutations of FSH receptor in four children with ovarian juvenile gran-ulosa cell tumors and the association of these tumors with central precocious puberty. J Pediatr Adolesc Gynecol. 2009, 22: 173-179. 10.1016/j.jpag.2008.10.003

    Article  PubMed  Google Scholar 

  30. Fragoso MC, Latronico AC, Carvalho FM, Zerbini MC, Marcondes JA, Araujo LM, Lando VS, Frazzatto ET, Mendonca BB, Villares SM: Activating mutation of the stimulatory G protein (gsp) as a putative cause of ovarian and testicular human stromal Leydig cell tumors. J Clin Endocrinol Metab. 1998, 83: 2074-2078. 10.1210/jc.83.6.2074

    CAS  PubMed  Google Scholar 

  31. Ligtenberg MJ, Siers M, Themmen AP, Hanselaar TG, Willemsen W, Brunner HG: Analysis of mutations in genes of the follicle-stimulating hormone receptor signaling pathway in ovarian granulosa cell tumors. J Clin Endocrinol Metab. 1999, 84: 2233-2234. 10.1210/jc.84.6.2233

    CAS  PubMed  Google Scholar 

  32. Kalfa N, Ecochard A, Patte C, Duvillard P, Audran F, Pienkowski C, Thibaud E, Brauner R, Lecointre C, Plantaz D, Guedj AM, Paris F, Baldet P, Lumbroso S, Sultan C: Activating mutations of the stimulatory g protein in juvenile ovarian granulosa cell tumors: a new prognostic factor?. J Clin Endocrinol Metab. 2006, 91: 1842-1847. 10.1210/jc.2005-2710

    Article  CAS  PubMed  Google Scholar 

  33. Chien J, Wong E, Nikes E, Noble MJ, Pantazis CG, Shah GV: Constitutive activation of stimulatory guanine nucleotide binding protein (G(S)alphaQL)-mediated signaling increases invasiveness and tumorigenicity of PC-3M prostate cancer cells. Oncogene. 1999, 18: 3376-3382. 10.1038/sj.onc.1202690

    Article  CAS  PubMed  Google Scholar 

  34. Barr FG: Gene fusions involving PAX and FOX family members in alveolar rhabdomyosarcoma. Oncogene. 2001, 20: 5736-5746. 10.1038/sj.onc.1204599

    Article  CAS  PubMed  Google Scholar 

  35. Hillion J, Le Coniat M, Jonveaux P, Berger R, Bernard OA: AF6q21, a novel partner of the MLL gene in t(6;11) (q21;q23), defines a forkhead transcriptional factor subfamily. Blood. 1997, 90: 3714-3719.

    CAS  PubMed  Google Scholar 

  36. Mani H, Shilo K, Galvin JR, Stocker JT, Franks TJ: Spectrum of precursor and invasive neoplastic lesions in type 1 congenital pulmonary airway malformation: case report and review of the literature. Histopathology. 2007, 51: 561-565. 10.1111/j.1365-2559.2007.02806.x

    Article  CAS  PubMed  Google Scholar 

  37. Mani N, Lowe D, Pope L, El-Daly H, Pfleiderer A: An unusual case of laryngeal spindle cell carcinoma metastasising to the orbit and heart. J Laryngol Otol. 2007, 121: e19- 10.1017/S0022215107009498

    Article  CAS  PubMed  Google Scholar 

  38. Mani SA, Yang J, Brooks M, Schwaninger G, Zhou A, Miura N, Kutok JL, Hartwell K, Richardson AL, Weinberg RA: Mesenchyme Forkhead 1 (FOXC2) plays a key role in metastasis and is associated with aggressive basal-like breast cancers. Proc Natl Acad Sci USA. 2007, 104: 10069-10074. 10.1073/pnas.0703900104

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  39. Nakamura T, Furukawa Y, Nakagawa H, Tsunoda T, Ohigashi H, Murata K, Ishikawa O, Ohgaki K, Kashimura N, Miyamoto M, Hirano S, Kondo S, Katoh H, Nakamura Y, Katagiri T: Genome-wide cDNA microarray analysis of gene expression profiles in pancreatic cancers using populations of tumor cells and normal ductal epithelial cells selected for purity by laser microdissection. Oncogene. 2004, 23: 2385-2400. 10.1038/sj.onc.1207392

    Article  CAS  PubMed  Google Scholar 

  40. Blount AL, Schmidt K, Justice NJ, Vale WW, Fischer WH, Bilezikjian LM: FoxL2 and Smad3 coordinately regulate fol-listatin gene transcription. J Biol Chem. 2009, 284: 7631-7645. 10.1074/jbc.M806676200

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  41. Ellsworth BS, Burns AT, Escudero KW, Duval DL, Nelson SE, Clay CM: The gonadotropin releasing hormone (GnRH) receptor activating sequence (GRAS) is a composite regulatory element that interacts with multiple classes of transcription factors including Smads, AP-1 and a forkhead DNA binding protein. Mol Cell Endocrinol. 2003, 206: 93-111. 10.1016/S0303-7207(03)00235-1

    Article  CAS  PubMed  Google Scholar 

  42. Dragovic RA, Ritter LJ, Schulz SJ, Amato F, Thompson JG, Armstrong DT, Gilchrist RB: Oocyte-secreted factor activation of SMAD 2/3 signaling enables initiation of mouse cumulus cell expansion. Biol Reprod. 2007, 76: 848-857. 10.1095/biolreprod.106.057471

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to acknowledge Dr and Pr F Paris, Unité d'endocrinologie pédiatrique, Hôpital Aranud-de-Villeneuve, CHU Montpellier, France; P Philibert and A Ecochard, Service d'Hormonologie, Hôpital Lapeyronie, CHU Montpellier, France; C Patte, Département d'Oncologie, Institut Gustave Roussy, Villejuif, France; E Thibaud, Service d'Endocrinologie Pédiatrique, Hôpital Necker, Paris, France; C Pienkowski, Service d'Endocrinologie Pédiatrique, Hôpital des Enfants, CHU Toulouse, France; M Fellous, Génétique Humaine, Inserm 709, Université Paris 7, Hôpital Cochin, Paris, France; and F Jaubert, Service d'anatompathologie, Hôpital Necker, Paris, France, for their contribution in this work. This work was supported by a national grant from the Association de Recherche contre le Cancer (ARC: JR/MLD/MDV-P05/5).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Charles Sultan.

Additional information

Competing interests

The authors declare that they have no competing interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kalfa, N., Veitia, R.A., Benayoun, B.A. et al. The new molecular biology of granulosa cell tumors of the ovary. Genome Med 1, 81 (2009). https://doi.org/10.1186/gm81

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/gm81

Keywords