1
|
Matsas A, Stefanoudakis D, Troupis T, Kontzoglou K, Eleftheriades M, Christopoulos P, Panoskaltsis T, Stamoula E, Iliopoulos DC. Tumor Markers and Their Diagnostic Significance in Ovarian Cancer. Life (Basel) 2023; 13:1689. [PMID: 37629546 PMCID: PMC10455076 DOI: 10.3390/life13081689] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Ovarian cancer (OC) is characterized by silent progression and late-stage diagnosis. It is critical to detect and accurately diagnose the disease early to improve survival rates. Tumor markers have emerged as valuable tools in the diagnosis and management of OC, offering non-invasive and cost-effective options for screening, monitoring, and prognosis. PURPOSE This paper explores the diagnostic importance of various tumor markers including CA-125, CA15-3, CA 19-9, HE4,hCG, inhibin, AFP, and LDH, and their impact on disease monitoring and treatment response assessment. METHODS Article searches were performed on PubMed, Scopus, and Google Scholar. Keywords used for the searching process were "Ovarian cancer", "Cancer biomarkers", "Early detection", "Cancer diagnosis", "CA-125","CA 15-3","CA 19-9", "HE4","hCG", "inhibin", "AFP", "LDH", and others. RESULTS HE4, when combined with CA-125, shows improved sensitivity and specificity, particularly in early-stage detection. Additionally, hCG holds promise as a prognostic marker, aiding treatment response prediction and outcome assessment. Novel markers like microRNAs, DNA methylation patterns, and circulating tumor cells offer potential for enhanced diagnostic accuracy and personalized management. Integrating these markers into a comprehensive panel may improve sensitivity and specificity in ovarian cancer diagnosis. However, careful interpretation of tumor marker results is necessary, considering factors such as age, menopausal status, and comorbidities. Further research is needed to validate and refine diagnostic algorithms, optimizing the clinical significance of tumor markers in ovarian cancer management. In conclusion, tumor markers such as CA-125, CA15-3, CA 19-9, HE4, and hCG provide valuable insights into ovarian cancer diagnosis, monitoring, and prognosis, with the potential to enhance early detection.
Collapse
Affiliation(s)
- Alkis Matsas
- Laboratory of Experimental Surgery and Surgical Research ‘N.S. Christeas’, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitrios Stefanoudakis
- Second Department of Obstetrics and Gynecology, Medical School, “Aretaieion” University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Theodore Troupis
- Department of Anatomy, Faculty of Health Sciences, Medical School, National and Kapodistrian University of Athens, MikrasAsias Str. 75, 11627 Athens, Greece
| | - Konstantinos Kontzoglou
- Laboratory of Experimental Surgery and Surgical Research ‘N.S. Christeas’, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Makarios Eleftheriades
- Second Department of Obstetrics and Gynecology, Medical School, “Aretaieion” University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Panagiotis Christopoulos
- Second Department of Obstetrics and Gynecology, Medical School, “Aretaieion” University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Theodoros Panoskaltsis
- Second Department of Obstetrics and Gynecology, Medical School, “Aretaieion” University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eleni Stamoula
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, University Campus Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Dimitrios C. Iliopoulos
- Laboratory of Experimental Surgery and Surgical Research ‘N.S. Christeas’, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
2
|
Li J, Bao R, Peng S, Zhang C. The molecular mechanism of ovarian granulosa cell tumors. J Ovarian Res 2018; 11:13. [PMID: 29409506 PMCID: PMC5802052 DOI: 10.1186/s13048-018-0384-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/30/2018] [Indexed: 12/12/2022] Open
Abstract
Over these years, more and more sex cord-stromal tumors have been reported. Granulosa cell tumor (GCT) is a rare tumor in ovaries, accounts for 2% to 5% of ovarian cancers. The main different feature of GCTs from other ovarian cancers is that GCTs can lead to abnormally secreted hormones (estrogen, inhibin and Müllerian inhibiting substance). The GCT is divided into two categories according to the age of patients, namely AGCT (adult granulosa cell tumor) and JGCT (Juvenile granulosa cell tumor). AGCT patients accounts for 95%. Although the pathogenesis is not clear, FOXL2 (Forkhead box L2) mutation was considered as the most critical factor in AGCT development. The current treatment is dominated by surgery. Target therapy remains in the adjuvant therapy stage, such as hormone therapy. During these years, other pathogenic factors were also explored, such as PI3K/AKT (phosphatidylinositol-3-kinase; serine/threonine kinase), TGF-β (Transforming growth factor beta) signaling pathway, Notch signaling pathway, GATA4 and VEGF (vascular endothelial growth factor). These factors and signaling pathway play important roles in GCT cell proliferation, apoptosis, or angiogenesis. The purpose of this review is to summarize the possible pathogenic factors and signaling pathways, which may shed lights on developing potential therapeutic targets for GCT.
Collapse
Affiliation(s)
- Jiaheng Li
- Joint programme of Nanchang University and Queen Mary University of London, Nanchang, China
| | - Riqiang Bao
- Joint programme of Nanchang University and Queen Mary University of London, Nanchang, China
| | - Shiwei Peng
- Department of Gynecology and Obstetrics, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Chunping Zhang
- Department of Cell Biology, School of Medicine, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.
| |
Collapse
|
3
|
Ray-Coquard I, Brown J, Harter P, Provencher DM, Fong PC, Maenpaa J, Ledermann JA, Emons G, Rigaud DB, Glasspool RM, Mezzanzanica D, Colombo N. Gynecologic Cancer InterGroup (GCIG) consensus review for ovarian sex cord stromal tumors. Int J Gynecol Cancer 2014; 24:S42-7. [PMID: 25341579 DOI: 10.1097/igc.0000000000000249] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Sex cord stromal tumors (SCST) are rare cancers of the ovarian area in adults. They constitute a heterogeneous group of tumors that develop from the sex cords and the ovarian stroma. These tumors are detected typically at an early stage, and they may recur as late as 30 years after the initial treatment. Because 70% of the patients present with stage I tumors, surgery represents the most important therapeutic arm. There are no data to support any kind of postoperative adjuvant treatment for patients with stage IA or IB SCSTs, given the indolent nature of these neoplasms and the overall good prognosis. The long natural history of the disease may lead to repeated surgical procedure should a relapse occurs. Platinum-based chemotherapy is currently used for patients with advanced stage SCSTs or recurrent disease, with an overall response rate of 63% to 80%. The indolent nature of SCSTs with the tendency for late recurrence requires long-term follow-up.
Collapse
Affiliation(s)
- Isabelle Ray-Coquard
- *Centre Léon Bérard, Lyon, France (GINECO); †MD Anderson Cancer Center, Houston, TX (GOG); ‡Kliniken-Essen-Mitte, Essen, Germany (AGO); §CHUM Hôpital Notre Dame, Montréal, Québec, Canada (NCIC-CTG); ∥FMHS, University of Auckland, Auckland, New Zealand (ANZGOG); ¶Tampere University Hospital, Tampere, Finland (NSGO); #UCL Cancer Institute, London, United Kingdom (NCRI MRC); **University Medecine, Göttingen, Germany (AGO); ††Institut de Cancérologie de l'Ouest René Gauducheau, Saint Herblain, France (GINECO); ‡‡Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom (SGCTG); §§Fondazione IRCCS Instituto Nazionale dei Tumori, Milan, Italy (MITO); and ∥∥University of Milan-Bicocca, Milan, Italy (MaNGO)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Makanji Y, Zhu J, Mishra R, Holmquist C, Wong WPS, Schwartz NB, Mayo KE, Woodruff TK. Inhibin at 90: from discovery to clinical application, a historical review. Endocr Rev 2014; 35:747-94. [PMID: 25051334 PMCID: PMC4167436 DOI: 10.1210/er.2014-1003] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
When it was initially discovered in 1923, inhibin was characterized as a hypophysiotropic hormone that acts on pituitary cells to regulate pituitary hormone secretion. Ninety years later, what we know about inhibin stretches far beyond its well-established capacity to inhibit activin signaling and suppress pituitary FSH production. Inhibin is one of the major reproductive hormones involved in the regulation of folliculogenesis and steroidogenesis. Although the physiological role of inhibin as an activin antagonist in other organ systems is not as well defined as it is in the pituitary-gonadal axis, inhibin also modulates biological processes in other organs through paracrine, autocrine, and/or endocrine mechanisms. Inhibin and components of its signaling pathway are expressed in many organs. Diagnostically, inhibin is used for prenatal screening of Down syndrome as part of the quadruple test and as a biochemical marker in the assessment of ovarian reserve. In this review, we provide a comprehensive summary of our current understanding of the biological role of inhibin, its relationship with activin, its signaling mechanisms, and its potential value as a diagnostic marker for reproductive function and pregnancy-associated conditions.
Collapse
Affiliation(s)
- Yogeshwar Makanji
- Department of Obstetrics and Gynecology (Y.M., J.Z., C.H., W.P.S.W., T.K.W.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60610; Center for Molecular Innovation and Drug Discovery (R.M., C.H.), Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208; and Department of Molecular Biosciences (N.B.S., K.E.M., T.K.W.), Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208
| | | | | | | | | | | | | | | |
Collapse
|
5
|
WANG XINGXING, GUI LU, ZHANG YOUYUAN, ZHANG JIHONG, SHI JIMIN, XU GUOXIONG. Cystatin B is a progression marker of human epithelial ovarian tumors mediated by the TGF-β signaling pathway. Int J Oncol 2014; 44:1099-1106. [PMID: 24452274 PMCID: PMC3977810 DOI: 10.3892/ijo.2014.2261] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 12/24/2013] [Indexed: 01/23/2023] Open
Abstract
Advanced ovarian cancer is a devastating disease. Gaining biomarkers of early detection during ovarian tumorigenesis may lead to earlier diagnosis and better therapeutic strategies. Cystatin B (CSTB) functions as an inhibitor to suppress intracellular cysteine proteases and has been implicated in several types of cancers. The present study explored the expression of CSTB in human ovarian tumors, to investigate CSTB expression associated with clinicopathological features, and to examine the effect of transforming growth factor-β (TGF-β), which plays a key role in ovarian tumorigenesis, on CSTB expression in ovarian cancer cells. The ovarian tissue samples from 33 patients were retrieved. The expression of CSTB in ovarian tissue was examined by immunohistochemistry. We found that CSTB was over-expressed in human ovarian surface epithelial tumors, including serous, mucinous and clear cell tumors. The immunoreactive staining of CSTB was strong in borderline and malignant tumors, weak in benign tumors, and negative in normal tissue counterparts, but was not correlated with the clinicopathological features of patients with ovarian tumors, such as age, histological types, tumor size, lymph node metastasis and clinical stages. The CSTB at mRNA and protein levels in two types of epithelial ovarian cancer cells, OVCAR-3 and SK-OV-3, was decreased after TGF-β1 treatment detected by quantitative PCR and western blot analysis, respectively. The inhibitory effect of TGF-β1 on CSTB expression was abolished in the presence of SB-431542, a TGF-β type I receptor kinase inhibitor. Our data suggest that CSTB is tumor tissue-specific and overexpressed in ovarian borderline and malignant tumors. The increased CSTB expression in ovarian tissue represents tumor progression and is dysregulated by the TGF-β signaling pathway. CSTB may become a novel diagnostic intracellular biomarker for the early detection of ovarian cancer.
Collapse
Affiliation(s)
- XINGXING WANG
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508
- Department of Pathology, Jinshan Hospital, Fudan University, Shanghai 201508
| | - LU GUI
- Department of Pathology, Jinshan Hospital, Fudan University, Shanghai 201508
| | - YOUYUAN ZHANG
- Department of Pathology, Jinshan Hospital, Fudan University, Shanghai 201508
| | - JIHONG ZHANG
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508
| | - JIMIN SHI
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508
| | - GUOXIONG XU
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032,
P.R. China
| |
Collapse
|
6
|
Ulcova-Gallova Z, Babcova K, Micanova Z, Bibkova K, Rumpik D. Hyperstimulation syndrome: the levels of inhibin A and B in sera and follicular fluids. Gynecol Endocrinol 2014; 30:298-301. [PMID: 24455972 DOI: 10.3109/09513590.2013.875999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Ovarial hyperstimulation syndrome (OHSS) represents a serious problem encountered during in vitro fertilization (IVF). We examined 10 patients with OHSS and 50 women also hormonally stimulated in the process of IVF who had no complications. In all women, we evaluated the number of obtained oocytes and the level of inhibins A and B in sera and follicular fluid collected at the time of ovarial puncture, the day embryo transfer and on the day of positivity for hCG. The level of inhibin B in both fluids was significantly higher (t = 0.0403) in women with high quality of oocytes. The higher level of inhibin A was detected in patients with OHSS at the time of oocyte collection and on the day of embryo transfer. Inhibin B was elevated only at the time of oocyte collection. The levels of inhibin A and B were identical in follicular fluids collected from both ovaries. We observed no statistically significant differences between the levels of inhibin A and B in follicular fluids of women in the absence of OHSS. Evaluation of serum levels of inhibin A and B at the time of oocyte collection may contribute to the prognosis and prevention of OHSS.
Collapse
|
7
|
Drummond AE, Fuller PJ. Activin and inhibin, estrogens and NFκB, play roles in ovarian tumourigenesis is there crosstalk? Mol Cell Endocrinol 2012; 359:85-91. [PMID: 21839804 DOI: 10.1016/j.mce.2011.07.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Revised: 07/14/2011] [Accepted: 07/14/2011] [Indexed: 01/24/2023]
Abstract
Ovarian cancer may be the most frequently lethal gynaecological malignancy but the heterogeneous nature of the disease and the advanced stage at which it is usually diagnosed, have contributed to the paucity of information relating to its aetiology and pathogenesis. Members of the TGF-β superfamily, estrogen and NFκB have all been implicated in the development and progression of cancers from a wide range of tissues. In the ovary, TGF-β superfamily members and estrogen play key roles in maintaining normal function. To date, little is known about the capacity of NFκB to influence normal ovarian function except that it is ubiquitously expressed. In this review we will highlight the roles that inhibin/activin, estrogen and NFκB, have been attributed within carcinogenesis and examine the potential for crosstalk between these pathways in ovarian cancer pathogenesis.
Collapse
Affiliation(s)
- Ann E Drummond
- Prince Henry's Institute of Medical Research, PO Box 5152, Clayton, Victoria 3168, Australia.
| | | |
Collapse
|
8
|
van Liempt SWJD, van Rheenen-Flach LE, van Waesberghe JHTM, Bleeker MCG, Piek JMJ, Lambalk CB. Solely inhibin B producing ovarian tumour as a cause of secondary amenorrhoea with hot flushes: case report and review of literature. Hum Reprod 2012; 27:1144-8. [DOI: 10.1093/humrep/der475] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
9
|
Inhibin/activin expression in human and rodent liver: subunits α and βB as new players in human hepatocellular carcinoma? Br J Cancer 2011; 104:1303-12. [PMID: 21407220 PMCID: PMC3078591 DOI: 10.1038/bjc.2011.53] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background: Activins and inhibins belong to the TGFβ-superfamily, which controls cell proliferation and differentiation in many organs. Activin A, the dimer of inhibin βA subunit, acts strongly anti-proliferative in hepatocytes. Little is known on the other activin/inhibin subunits in human liver and hepatocellular carcinoma (HCC). Methods: We studied the expression of the complete inhibin family α, βA, βB, βC, βE in normal liver, tumour-adjacent and HCC tissue, 12 additional organs and rodent liver. A total of 16 HCC and 10 disease-free livers were analysed. Expression of inhibin subunits was determined by qRT–PCR, normalised to RNA input and by geNorm algorithm, and confirmed by immunohistochemistry. Results: Remarkably, βA expression was not decreased in HCC. Similarly, βC and βE exhibited no major changes. In contrast, inhibin α, barely detectable in normal liver, was strongly increased in tumour-adjacent liver and dramatically enhanced in HCC. βB was strongly enhanced in some HCC. At variance with human liver, rodent liver showed higher inhibin α and βC expression, but βA was somewhat, and βB dramatically lower. Conclusions: Upregulation of inhibin α – and possibly of βB – may shield HCC cells from anti-proliferative effects of activin A. Dramatic variations between humans and rodents may reflect different functions of some inhibins/activins.
Collapse
|
10
|
Abstract
Inhibin A and B, dimeric glycoproteins comprising an α- and β((A/B))-subunit, negatively regulate follicle stimulating hormone (FSH) synthesis by the pituitary. The expression of α- and β-subunits within Sertoli cells of the testis and granulosa cells of the ovary is controlled by a range of transcription factors, including CREB, SP-1, Smads, and GATA factors. The inhibin α- and β-subunits are synthesized as precursor molecules consisting of an N-terminal propeptide and a C-terminal mature domain. Recently, we showed that hydrophobic residues within the propeptides of the α- and β-subunits interact noncovalently with their mature domains, maintaining the molecules in a conformation competent for dimerization. Dimeric precursors are cleaved by proprotein convertases and mature inhibins are secreted from the cell noncovalently associated with their propeptides. Propeptides may increase the half-life of inhibin A and B in circulation, but they are readily displaced in the presence of the high-affinity receptors, betaglycan, and ActRII.
Collapse
|
11
|
Les tumeurs rares de l’ovaire : stratégies thérapeutiques en 2010, Observatoire francophone des tumeurs rares de l’ovaire et émergence des centres de références. Bull Cancer 2010; 97:123-35. [DOI: 10.1684/bdc.2010.1017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
12
|
Datta M, Winter-Roach BA, Fitzgerald C. Secondary amenorrhoea associated with an isolated elevated serum leuteinising hormone: An unusual presentation of a granulosa cell tumour. J OBSTET GYNAECOL 2009; 26:830-2. [PMID: 17130057 DOI: 10.1080/01443610600994858] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- M Datta
- Department of Obstetrics and Gynaecology, St Mary's Hospital, Manchester, UK.
| | | | | |
Collapse
|
13
|
Bittinger S, Alexiadis M, Fuller PJ. Expression status and mutational analysis of the PTEN and P13K subunit genes in ovarian granulosa cell tumors. Int J Gynecol Cancer 2009; 19:339-42. [PMID: 19407556 DOI: 10.1111/igc.0b013e3181a1cdfd] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Granulosa cell tumors (GCT) are a unique subset of ovarian tumors which have a molecular phenotype resembling that of follicle stimulating hormone (FSH)-stimulated pre-ovulatory granulosa cells. FSH acts via its receptor to stimulate signaling pathways including the phosphatidylinositol 3-kinase (PI3K)-AKT pathway. Activation of this pathway occurs in solid tumors, including ovarian epithelial tumors, through mutation of the PI3K subunit genes or inactivation of the tumor suppressor, PTEN. Activation of this pathway would be predicted to be tumorigenic in granulosa cells.Expression of the 2 PI3K subunit genes, PIK3CA, which encodes the catalytic subunit, and PIK3R1, which encodes the regulatory subunit, together with the PTEN gene was determined in a panel of GCT, 2 human GCT-derived cell lines, COV434 and KGN, and normal ovary. Direct sequence analysis was used to screen for mutations. Expression of all 3genes was observed in the GCT without evidence of overexpression for the PI3K subunit genes or loss of expression for PTEN. Sequence analysis of amplicons spanning exons 9and 20, in which greater than 75% of mutations occur in the PIK3CA gene did not identify any missense mutations. Similarly, the previously reported deletions in exons 12 and 13 of the PIK3R1 were not found in the GCT. Three amplicons spanning the entire coding sequence of the PTEN gene were sequenced; neither deletions nor mutations were identified.These findings suggest that activation of PI3K signaling through PI3K/PTEN mutation or altered expression, in contrast to many other types of solid tumor, is not associated with GCT.
Collapse
Affiliation(s)
- Sophie Bittinger
- Prince Henry's Institute of Medical Research, Victoria, Australia
| | | | | |
Collapse
|
14
|
Agha-Hosseini M, Aleyaseen A, Safdarian L, Kashani L. Secondary Amenorrhea with Low Serum Luteinizing Hormone and Follicle-stimulating Hormone Caused by an Inhibin A- and Inhibin B-producing Granulosa Cell Tumor. Taiwan J Obstet Gynecol 2009; 48:72-5. [DOI: 10.1016/s1028-4559(09)60040-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
15
|
Kalfa N, Philibert P, Patte C, Thibaud E, Pienkowski C, Ecochard A, Boizet-Bonhoure B, Fellous M, Sultan C. [Juvenile granulosa-cell tumor: clinical and molecular expression]. ACTA ACUST UNITED AC 2008; 37:33-44. [PMID: 19119048 DOI: 10.1016/j.gyobfe.2008.06.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Accepted: 06/23/2008] [Indexed: 01/09/2023]
Abstract
Ovarian sex cord-stromal tumors are rare tumors that originate from the nongerminal cells of ovary. Two decades ago, the identification of juvenile granulosa-cell tumors (GCT), as a specific entity inside this group, allowed a better treatment of these tumors in children. However, little data have been reported on the natural course of the disease and reliable prognostic factors have not been yet defined. We here review the clinical and genetics aspects of granulosa tumors, based on a series of 40 children. This national collaborative study involved the French Society of Children Cancer and eight clinical departments of pediatric endocrinology. We found that early diagnosis of a tumor, revealed by clinical signs of hyperoestrogeny, is an important prognostic factor. The pathophysiology of these tumors is still debatable and several cellular- and molecular-abnormal signals could be implicated in their development. The role of growth factors and oncogenes through the signaling pathway of MAP kinase is still discussed. According to our data, FSH signaling-transduction pathway, such as a constitutionally activated Galphas, could also be implicated in the induction of granulosa cell proliferation and seems to modulate the invasiveness of the tumor. Last, we have described a low-expression pattern or an extinction of an ovarian-determination gene, FOXL2, which is related to a worse prognosis of this tumor.
Collapse
Affiliation(s)
- N Kalfa
- Service d'hormonologie, hôpital Lapeyronie, CHU de Montpellier, Montpellier, France
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
OBJECTIVE Pancreatic mucinous cystic tumor (MCT(P)) and ovarian mucinous cystic tumor (MCT(O)) show common features. However, there are few studies showing a comparison of both types of tumor. We immunohistochemically studied both types of tumor to clarify their characteristics. METHODS Eight patients with MCT(P) and 21 patients with MCT(O) were examined. The tumors were immunohistochemically examined using antibodies against female sex hormone receptors (estrogen receptor, progesterone receptor, and alpha-inhibin), pancreatobiliary tissue markers (carbohydrate antigen 19-9 and DUPAN2) and cell cycle regulators (p27kip1 and phosphorylated retinoblastoma). Samples from 7 female patients with invasive pancreatic ductal carcinoma (DC), 8 female patients with normal pancreatic tissue, and 10 patients with normal ovarian tissue were also examined. RESULTS In the tumor epithelial cells, the expressions of DUPAN2 and p27/kip1 were similar between MCT(P) (38% and 88%, respectively) and MCT(O) (14% and 76%, respectively), but significantly different between both tumors and DC (100% and 0%). In the stromal cells, the expressions of estrogen receptor, progesterone receptor, alpha-inhibin, and p27/kip1 were similar between MCT(P) (63%, 75%, 50%, and 63%, respectively) and MCT(O) (57%, 71%, 81%, and 57%, respectively), but significantly different between both tumors and DC (0%, 0%, 0%, and 0%, respectively). CONCLUSIONS MCT(P) and MCT(O) have several immunohistochemical similarities and are significantly different from DC. The female sex hormone system may play a major role in the development of both MCT(P) and MCT(O). A frequent p27/kip1 expression level was associated with nonaggressive progression of both tumors.
Collapse
|
17
|
Ray-Coquard I, Guastalla JP, Treilleux I, Weber B, Guardiola E, Lotz JP, Méeus P, Mignot L, Raudrant D, Tournigand C, Duvillard P, Pujade-Lorraine E. Sex cord-stromal tumours, rare events in oncology necessitating multidisciplinary approach and referral pathways. EJC Suppl 2007. [DOI: 10.1016/s1359-6349(07)70048-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
18
|
MEDAN MS, ARAI KY, WATANABE G, TAYA K. Inhibin: Regulation of reproductive function and practical use in females. Anim Sci J 2007. [DOI: 10.1111/j.1740-0929.2006.00399.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
19
|
Robertson DM, Oehler MK. Emerging role of inhibin as a biomarker for ovarian cancer. WOMENS HEALTH 2005; 1:51-7. [DOI: 10.2217/17455057.1.1.51] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
20
|
Kurihara S, Hirakawa T, Amada S, Ariyoshi K, Nakano H. Inhibin-producing ovarian granulosa cell tumor as a cause of secondary amenorrhea: Case report and review of the literature. J Obstet Gynaecol Res 2004; 30:439-43. [PMID: 15566459 DOI: 10.1111/j.1447-0756.2004.00231.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We report the case of 31-year-old patient with an inhibin B-secreting granulosa cell tumor of the left ovary who presented with secondary amenorrhea. Preoperative serum hormonal levels were as follows: follicle-stimulating hormone (FSH) 0.3 mIU/mL, luteinizing hormone (LH) 9.81 mIU/mL, estradiol 142.0 pg/mL and inhibin B 2429 pg/mL. Gonadotropin-releasing hormone (GnRH) test revealed no FSH response and a normal LH response. After removal of the tumor, the levels of FSH and inhibin B returned to within the normal range, and regular menses resumed 27 days postoperatively. In premenopausal women, secondary amenorrhea may be the initial manifestation of granulosa cell tumor. A low FSH level coupled with normal levels of E2 and LH, the inhibition of the FSH response to GnRH and an elevated inhibin level suggest the presence of an inhibin-secreting ovarian tumor and also rule out the possibility of isolated FSH deficiency.
Collapse
Affiliation(s)
- Shuichi Kurihara
- Department of Obstetrics and Gynecology, Kyushu University Hospital, Fukuoka, Japan
| | | | | | | | | |
Collapse
|
21
|
Abstract
The inhibins are produced and secreted by several ovarian cancers. Monitoring serum levels by immunoassay may be a useful diagnostic aid in the initial assessment of this disease and in monitoring its potential recurrence following surgery. The assays are applicable to women after menopause when the majority of ovarian cancers are detected, and when the normal ovarian production of inhibin is low to negligible. A new inhibin immunoassay (total inhibin ELISA) has been developed with the intention of widespread clinical application. The assay readily detects granulosa cell and mucinous tumours. CA125, a widely used ovarian cancer marker, detects the other main ovarian cancer types (serous, endometrioid, undifferentiated) with high sensitivity. The combination of the two tests detects the majority of ovarian cancers with high specificity (95%) and sensitivity (95%). Studies have been undertaken to assess its application to women in the perimenopausal stage and to younger women during normal reproductive life. These studies are providing a platform for the introduction of the test into clinical practice.
Collapse
Affiliation(s)
- David M Robertson
- Prince Henry's Institute of Medical Research, Monash Medical Centre, P.O. Box 5152, Clayton, Vic. 3168, Australia.
| | | | | | | | | | | |
Collapse
|
22
|
Mylonas I, Makovitzky J, Richter DU, Jeschke U, Briese V, Friese K. Expression of the inhibin-alpha subunit in normal, hyperplastic and malignant endometrial tissue: an immunohistochemical analysis. Gynecol Oncol 2004; 93:92-7. [PMID: 15047219 DOI: 10.1016/j.ygyno.2003.12.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2003] [Indexed: 10/26/2022]
Abstract
OBJECTIVES Determination of the frequency and tissue distribution of inhibin-alpha (INH-alpha) in normal, hyperplastic and malignant endometrium. MATERIALS AND METHODS Endometrial tissue was obtained from normal, hyperplastic (glandular-cystic hyperplasia), adenomatous hyperplasia (AH grade I to III) and endometroid adenocarcinoma and immunohistochemically characterized with INH-alpha antibody. RESULTS INH-alpha was expressed in normal, hyperplastic and malignant endometrium. Highest expression was observed during secretory phase and glandular-cystic hyperplasia compared to all groups. A continuous decline was noted from AH grade I to III with a significance between AH I and II. DISCUSSION A menstrual cycle associated expression of INH-alpha in glandular endometrial epithelium was observed. Since AH grade II and III can be considered as a precursor of endometrial cancer, INH-alpha could be a marker of cell transformation and an endometrial tumor-suppressor agent.
Collapse
Affiliation(s)
- Ioannis Mylonas
- Department of Obstetrics and Gynecology, University of Rostock, Rostock, Germany
| | | | | | | | | | | |
Collapse
|
23
|
Chan LF, Storr HL, Scheimberg I, Perry LA, Banerjee K, Miraki-Moud F, Camacho-Hübner C, Savage MO. Pseudo-precocious puberty caused by a juvenile granulosa cell tumour secreting androstenedione, inhibin and insulin-like growth factor-I. J Pediatr Endocrinol Metab 2004; 17:679-84. [PMID: 15198302 DOI: 10.1515/jpem.2004.17.4.679] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We report a female child who presented at age 3.92 years with a 2-year history of consonant pubertal development caused by a large right-sided ovarian juvenile granulosa cell tumour (JGCT). Although JGCTs causing pseudo-precocious puberty have been previously described, they remain rare and endocrine data are often incomplete. In this case the tumour was associated with raised serum oestradiol, androstenedione, inhibin and IGF-I. Histological changes were consistent with JGCT. Immunohistochemical studies revealed positive reactivity to MIC-2, inhibin, melan A, IGF-I and IGFBP-2.
Collapse
Affiliation(s)
- L F Chan
- Department of Paediatric Endocrinology, St Bartholomew's and The Royal London School of Medicine and Dentistry, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Peng C. The TGF-beta superfamily and its roles in the human ovary and placenta. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2004; 25:834-44. [PMID: 14532952 DOI: 10.1016/s1701-2163(16)30674-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The transforming growth factor-beta (TGF-beta) superfamily consists of a large group of growth and differentiation factors, such as TGF-betas, activins, inhibins, growth and differentiation factors (GDFs), and bone morphogenetic proteins (BMPs). These molecules act through specific receptor complexes that are composed of type I and type II serine/threonine receptor kinases. The receptor kinases subsequently activate Smad proteins, which then propagate the signals into the nucleus to regulate target gene expression. Several ligands in this family, such as TGF-betas, activins, inhibins, BMP-15, and GDF-9, play important roles in regulating human ovarian functions, including follicle development and maturation. Activin and TGF-beta are also involved in regulating placental development and functions. Abnormal expression or function of these ligands has been found in several pathological conditions. This review summarizes the role of the TGF-beta superfamily in human ovarian and placental regulation and function, and the potential clinical implications.
Collapse
Affiliation(s)
- Chun Peng
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
25
|
Borgoño CA, Fracchioli S, Yousef GM, Rigault de la Longrais IA, Luo LY, Soosaipillai A, Puopolo M, Grass L, Scorilas A, Diamandis EP, Katsaros D. Favorable prognostic value of tissue human kallikrein 11 (hK11) in patients with ovarian carcinoma. Int J Cancer 2003; 106:605-610. [PMID: 12845660 DOI: 10.1002/ijc.11296] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Human kallikrein 11 (hK11/trypsin-like serine protease/TLSP, encoded by the KLK11 gene) is a member of the kallikrein family of secreted serine proteases. Recently, we developed a highly sensitive and specific immunoassay for hK11 and found that this protease is expressed in the prostate, stomach and trachea as well as in amniotic fluid and milk of lactating women. Elevated serum hK11 levels were found in 60% of men with prostate cancer and 70% of women with ovarian cancer. Also, hK11 expression was found to be under the regulation of steroid hormones, particularly estrogens, at the level of KLK11 transcription. We hypothesized that hK11 may be implicated in endocrine-related malignancies and serve as a novel prostate and ovarian cancer serological marker. The aim of our study was to examine if hK11 expression in ovarian tumors bears any prognostic significance. The concentration of hK11 (ng per mg of total protein) in 104 ovarian tumor cytosolic extracts was quantified and correlated with clinicopathologic variables and outcome over a median follow-up period of 67 months. Outcome was defined as progression-free survival (PFS) and overall survival (OS). hK11 concentration in ovarian tumor cytosols ranged from 0-21 ng/mg of total protein, with a median of 0.54 ng/mg. An optimal cutoff value of 0.54 ng/mg was selected to categorize tumors as hK11-positive or -negative. hK11-positive tumors were more frequently associated with early stage (Stage I/II) disease, pre-/peri-menopausal status and patients who exhibited complete or partial response to chemotherapy (p < 0.05). Univariate analysis revealed that patients with hK11-positive tumors had a significantly decreased risk of relapse with a hazard ratio (HR) of 0.45 (p = 0.007) and death (HR of 0.34, p = 0.005). Cox multivariate analysis indicated that hK11 was an independent prognostic indicator of OS (HR of 0.41, p = 0.025). Kaplan-Meier survival curves further confirmed that women with hK11-positive tumors have longer PFS and OS (p = 0.005 and p = 0.003, respectively). Similarly, in the subgroup of patients with grade 1-2 tumors, hK11-positivity was associated with higher OS in both univariate and multivariate analysis (HR of 0.23 and 0.17, p < 0.05). Finally, in women with optimal debulking after surgery (<1 cm residual tumor), hK11 positivity was associated with a slower disease progression. These results indicate that hK11 is a novel, independent marker of favorable prognosis in patients with ovarian cancer.
Collapse
MESH Headings
- Adenocarcinoma, Clear Cell/metabolism
- Adenocarcinoma, Clear Cell/pathology
- Adenocarcinoma, Mucinous/metabolism
- Adenocarcinoma, Mucinous/pathology
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/metabolism
- Carcinoma, Endometrioid/metabolism
- Carcinoma, Endometrioid/pathology
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/pathology
- Disease Progression
- Disease-Free Survival
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Middle Aged
- Neoplasm Proteins/metabolism
- Neoplasm Staging
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Prognosis
- Serine Endopeptidases/metabolism
- Survival Rate
- Up-Regulation
Collapse
Affiliation(s)
- Carla A Borgoño
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Stefano Fracchioli
- Department of Gynecology, Gynecologic Oncology Unit, University of Turin, Turin, Italy
| | - George M Yousef
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Liu-Ying Luo
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Antoninus Soosaipillai
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Manuela Puopolo
- Department of Gynecology, Gynecologic Oncology Unit, University of Turin, Turin, Italy
| | - Linda Grass
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Andreas Scorilas
- National Center of Scientific Research "Demokritos", IPC, Athens, Greece
| | - Eleftherios P Diamandis
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Dionyssios Katsaros
- Department of Gynecology, Gynecologic Oncology Unit, University of Turin, Turin, Italy
| |
Collapse
|
26
|
Affiliation(s)
- Stephen Tong
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | | | | |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Granulosa cell tumours of the ovary are an uncommon ovarian sex-cord stromal tumour. These neoplasms provide a spectrum of clinical presentations that span from the first to the tenth decade. Surgery represents the primary therapy for early stage disease; however, management of women with advanced disease is less clear. Because of their relative rarity, evidence to support decision-making in the management of granulosa cell tumours is limited. The purpose of this review is to provide the clinician with an updated knowledge of the clinical and molecular aspects of granulosa cell tumours in order to guide therapy. RECENT FINDINGS The clinical stage, mitotic index and cellular atypia correlate most strongly with prognosis. However, these tumours may demonstrate heterogeneous genetic aberrations that can predict behaviour and response to therapy. Case series and reports suggest that postoperative combination chemotherapy is of most benefit in advanced disease. Serial measurements of serum inhibin may be helpful in the follow-up of these women, particularly in the post-menopausal group. SUMMARY The pathology and treatment of women with granulosa cell tumours of the ovary is complex. Such women should be managed in a multidisciplinary gynaecological oncology unit. A better understanding of the molecular pathology may assist treatment.
Collapse
Affiliation(s)
- Gavin C E Stuart
- Department of Oncology, University of Calgary, Tom Baker Cancer Centre, 1331-29th Street NW, Calgary, Alberta T2N 4N2, Canada.
| | | |
Collapse
|
28
|
Welt C, Sidis Y, Keutmann H, Schneyer A. Activins, inhibins, and follistatins: from endocrinology to signaling. A paradigm for the new millennium. Exp Biol Med (Maywood) 2002; 227:724-52. [PMID: 12324653 DOI: 10.1177/153537020222700905] [Citation(s) in RCA: 225] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
It has been 70 years since the name inhibin was used to describe a gonadal factor that negatively regulated pituitary hormone secretion. The majority of this period was required to achieve purification and definitive characterization of inhibin, an event closely followed by identification and characterization of activin and follistatin (FS). In contrast, the last 15-20 years saw a virtual explosion of information regarding the biochemistry, physiology, and biosynthesis of these proteins, as well as identification of activin receptors, and a unique mechanism for FS action-the nearly irreversible binding and neutralization of activin. Many of these discoveries have been previously summarized; therefore, this review will cover the period from the mid 1990s to present, with particular emphasis on emerging themes and recent advances. As the field has matured, recent efforts have focused more on human studies, so the endocrinology of inhibin, activin, and FS in the human is summarized first. Another area receiving significant recent attention is local actions of activin and its regulation by both FS and inhibin. Because activin and FS are produced in many tissues, we chose to focus on a few particular examples with the most extensive experimental support, the pituitary and the developing follicle, although nonreproductive actions of activin and FS are also discussed. At the cellular level, it now seems that activin acts largely as an autocrine and/or paracrine growth factor, similar to other members of the transforming growh factor beta superfamily. As we discuss in the next section, its actions are regulated extracellularly by both inhibin and FS. In the final section, intracellular mediators and modulators of activin signaling are reviewed in detail. Many of these are shared with other transforming growh factor beta superfamily members as well as unrelated molecules, and in a number of cases, their physiological relevance to activin signal propagation remains to be elucidated. Nevertheless, taken together, recent findings suggest that it may be more appropriate to consider a new paradigm for inhibin, activin, and FS in which activin signaling is regulated extracellularly by both inhibin and FS whereas a number of intracellular proteins act to modulate cellular responses to these activin signals. It is therefore the balance between activin and all of its modulators, rather than the actions of any one component, that determines the final biological outcome. As technology and model systems become more sophisticated in the next few years, it should become possible to test this concept directly to more clearly define the role of activin, inhibin, and FS in reproductive physiology.
Collapse
Affiliation(s)
- Corrine Welt
- Reproductive Endocrine Unit and Endocrine Unit, Massachusetts General Hospital, Boston 02114, USA
| | | | | | | |
Collapse
|
29
|
Welt CK. The physiology and pathophysiology of inhibin, activin and follistatin in female reproduction. Curr Opin Obstet Gynecol 2002; 14:317-23. [PMID: 12032389 DOI: 10.1097/00001703-200206000-00012] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In the last 2 years, major advances have been made in the understanding of inhibin physiology. Discovery of an inhibin receptor and binding protein has expanded our knowledge of the mechanism whereby inhibin antagonizes activin action. Controlled experimental studies have clarified the regulation and physiology of inhibin A and inhibin B, providing evidence for their use as markers of ovarian function. Clinical studies continue to uphold the use of inhibin as a marker for ovarian cancer, but have not generally supported its use over standard prognostic markers in assisted reproductive technologies. Finally, ongoing work suggests alterations in inhibin and follistatin that may be linked to the pathophysiology of polycystic ovary syndrome. Thus, the mechanism of inhibin action and its role in normal and abnormal ovarian function continues to emerge.
Collapse
Affiliation(s)
- Corrine K Welt
- Reproductive Endocrine Unit, Reproductive Endocrine Sciences Center and National Center for Infertility Research, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| |
Collapse
|