Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Human Reproduction Update, Vol.13, No.3 pp. 265–273, 2007 doi:10.

1093/humupd/dml060
Advance Access publication January 9, 2007

The Anti-Mullerian hormone and ovarian cancer

Antonio La Marca 1 and Annibale Volpe


Mother – Infant Department, Institute of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Modena, Italy
1
To whom correspondence should be addressed at: Mother – Infant Department, Institute of Obstetrics and Gynecology, University of
Modena and Reggio Emilia, Policlinico di Modena, Largo del Pozzo, 41100 Modena, Italy. Tel: 39 0 594 224 379; Fax: 39 0 594 224 394;

Downloaded from https://academic.oup.com/humupd/article/13/3/265/2457878 by guest on 14 August 2021


E-mail: antlamarca@libero.it

The Anti-Mullerian hormone (AMH), which is produced by fetal Sertoli cells, is responsible for regression of Mullerian
ducts, the anlagen for uterus and Fallopian tubes, during male sex differentiation. Ovarian granulosa cells also secrete
AMH from late in fetal life. The patterns of expression of AMH and its type II receptor in the post-natal ovary indicate
that AMH may play an important role in ovarian folliculogenesis. Recent advances in the physiological role of AMH
has stimulated interest in the significance of AMH as a diagnostic marker and therapeutic agent for ovarian cancer.
Currently, AMH has been shown to be a circulating marker specifically for granulosa cell tumour (GCT). Its diagnostic
performance seems to be very good, with a sensitivity ranging between 76 and 93%. In patients treated for GCT, AMH
may be used post-operatively as marker for the efficacy of surgery and for disease recurrence. Based on the physiologi-
cal inhibitory role of AMH in the Mullerian ducts, it has been proposed that AMH may inhibit epithelial ovarian cancer
cell both in vitro and in vivo. These observations will be the basis for future research aiming to investigate the possible
clinical role of AMH as neo-adjuvant, or most probably adjuvant, therapy for ovarian cancer.

Key words: AMH/cancer therapy/diagnostic markers/epithelial ovarian cancer/granulosa cell tumour

Introduction
Ovarian cancer comprises a heterogeneous group of tumours of of cases the effects are estrogenic in nature, though rarely andro-
three major types: epithelial, germ cell and sex cord stromal. genic effects have been documented (Nakashima et al., 1984).
Epithelial tumours arise from the coelomic mesothelium, which In the prepubertal female, these effects may manifest as preco-
is capable of differentiating into both benign and malignant cious isosexual development, whereas in premenopausal patients,
tumours. Epithelial malignancies represent about 82% of all menstrual irregularities are seen (Biscotti and Hart, 1989). In older
ovarian malignancies. The predominant cell types are: serous, women, the most frequent symptom is post-menopausal bleeding.
mucinous and endometrioid. As a consequence of this hormonal secretion, an association
The malignant germ cell tumours are believed to arise from between GCT with both endometrial hyperplasia and adenocarci-
primitive germ cells in the ovary. These tumours represent about noma has been well documented (Salerno, 1962).
5% of all ovarian malignancies. They include dysgerminoma, In GCT the routes of dissemination are not completely clear.
embryonal carcinoma, endodermal sinus tumour, choriocarcinoma These tumours grow locally and then spread to the adjacent intra-
and malignant teratomas. peritoneal organs and when systemic disease occurs it does so late
The sex cord stromal neoplasms are derived from mesenchymal in the course of the disease. The majority of GCT present as stage I
stem cells in the ovarian cortex and represent about 10% of all and the 5-year survival rate is 90%.
ovarian tumours. They include granulosa – theca cell tumours, The initial form of therapy for most patients should be a surgical
granulosa cell tumours (GCTs) and Sertoli –Leydig cell tumours. procedure so that a histological diagnosis and appropriate staging
GCT constitutes 3% of all ovarian tumours. Two distinct types may be obtained. In patients with disease localized to one ovary
of GCTs have been described on the basis of clinical presentation and those wishing to preserve fertility, conservative therapy seems
and histological characteristics: the juvenile and the adult form. to be indicated. In those with more advanced disease or in post-
The more common adult form generally presents in pre- or post- menopausal women, Total abdominal hysterectomy and bilateral
menopausal women with a median age of 50 years during presen- salpingo-oophorectomy (TAH/BSO) would be an appropriate
tation (Pankratz et al., 1978; Bjorkholm and Silfversward, 1981). initial treatment. Chemotherapy should be considered in those
A majority (two-thirds) of these GCT patients have endocrine patients with advanced, recurrent or metastatic disease. Although
manifestations as a direct consequence of hormone secretion by response rates to chemotherapy are high, the impact on both disease-
the tumour (Bjorkholm and Silfversward, 1981). In the majority free states and overall long-term survival is currently unknown.

# The Author 2007. Published by Oxford University Press on behalf of the European Society of Human Reproduction and Embryology. All rights reserved. For
Permissions, please email: journals.permissions@oxfordjournals.org 265
A. La Marca and A. Volpe

The GCTs have the potential to secrete estrogen and inhibin. In the female, AMH expression begins post-natally and acts in
Hence, these products are used as markers for GCT. modulating follicular growth (Durlinger et al., 2001; Ikeda
In several studies, elevated serum levels of estradiol have been et al., 2002) and preventing recruitment of non-dominant follicles
found in some patients with known disease. The levels generally (Durlinger et al., 1999; Josso et al., 1998).
fluctuate with response to or failure of treatment or with the Granulosa cells of primary follicles show homogeneous AMH
bulk of the disease. Unfortunately, the cases that have been expression, whereas in larger follicles, AMH is mainly produced
described are few and the marker itself lacks sensitivity. Although in cells near the oocyte and in a few cells surrounding the
it has been shown that the granulosa cells produce estradiol, they antrum. AMH continues to be expressed in the growing follicles
do so only if testosterone is presented to them. Testosterone is in the ovary until they have reached the size and differentiation
produced by the adjacent theca cells, which are present in some state at which they are to be selected for dominance by the
ovarian tumours but not in extraovarian tumours (Lappohn action of pituitary FSH.
et al., 1989). In mouse this occurs at the early antral stage in small growing
A second marker, inhibin, has also been reported in patients follicles (Durlinger et al., 2002), whereas in human it is in antral

Downloaded from https://academic.oup.com/humupd/article/13/3/265/2457878 by guest on 14 August 2021


with GCT. Inhibin is a dimeric glycoprotein secreted by the follicles of size 4– 6 mm (Weenen et al., 2004). Thus, AMH is
gonads to regulate pituitary FSH. It exists as a dimer of two expressed in follicles that have undergone recruitment from the
subunits (a and bA or bB) to form inhibin A (abA) or B (abB) primordial follicle pool and have not been selected for dominance.
(Bilezijkian et al., 2004). AMH is not expressed in atretic follicles and theca cells (Rey
Studies investigating the specific assays for inhibin A and B et al., 2000) (Figure 1).
have shown that almost all GCT have elevated inhibin B levels AMH employs a heteromeric receptor system consisting of a
and the majority also have elevated inhibin A levels (Robertson single membrane spanning serine threonine kinase receptors of
et al., 1999a). Concentrations of immunoreactive inhibin and the types I and II, respectively. The type II receptor imparts ligand-
specific dimeric inhibin in GCT are by far the highest seen in binding specificity and the type I receptor mediates downstream
any form of ovarian malignancy or in other disease. signalling when activated by the type II receptor (Figure 2). The
However, as a diagnostic test, neither inhibin A nor B are as AMH type II receptor (AMH-RII) is localized to the mesenchyme
effective as a total a subunit as measured by the inhibin radio- around the Müllerian duct in the urogenital ridge of both the male
immunoassay (RIA) (Robertson et al., 1999a). Unfortunately, and female rat and mouse. It is interesting to note that loss of func-
the inhibin RIA is limited as a diagnostic test due to its lack of tion mutations in the AMH-RII as well as the AMH ligand itself, are
practicability (5-day assay). A two-site immunofluorometric causes of persistent Müllerian Duct Syndrome in humans (Imbeaud
assay (aC IFMA) was subsequently developed (Robertson et al., et al., 1994). In the ovary of rats, the AMH-RII is expressed both in
1999b). However, the aC IFMA is not appropriate for wide-spread granulosa and theca cells (Ingraham et al., 2000).
diagnostic use as it has proved difficult to maintain the assay
reliability between different batches of antisera. In conclusion
Role of AMH in ovarian physiology
there is controversy surrounding the use of different antisera and
methodologies, which have resulted in conflicting results. AMH is considered as a negative regulator of the early stages of follicu-
More than 10 years ago, an alteration in anti-Mullerian hormone lar development (Themmen 2005). Earlier studies have shown that
(AMH) [also called mullerian inhibiting substance) secretion was AMH inhibits the first meiotic division of diplotene oocytes in imma-
described in a patient affected by GCT (Gustafson et al., 1992). ture rats (Ueno et al., 1989), but this has not been confirmed by others
After this first observation, several studies investigating the (Tsafriri et al., 1988). In addition, AMH blocks the proliferation of
dynamics of serum AMH in patients with ovarian malignancies human granulosa–luteal cells in vitro (Kim et al., 1992) and the con-
have been published. centration of AMH in follicular fluid is inversely proportional to the
In the present article, the main findings and possible clinical mitotic indices of granulosa cells in vivo (Seifer et al., 1993). Thus,
implications of these studies are explored. AMH appears to have an autocrine role in maturation of normal fol-
licles (Figure 3).
Based on this observation it has been hypothesized that AMH
AMH and AMH receptors
could be one of the factors involved in determining the responsive-
AMH is a member of the transforming growth factor (TGF) super- ness of ovarian follicles to FSH during cyclic recruitment
family. AMH is strongly expressed in Sertoli cells from testicular (Skinner, 2005). It has been recently shown that oocytes
differentiation up to puberty and to a much lesser degree in granu- up-regulate AMH expression in granulosa cells in a fashion that
losa cells from birth up to menopause (Munsterberg and is dependent upon the developmental stage of the oocyte
Lovell-Badge, 1991; Lee and Donahoe, 1993; Josso et al., 1998; (Salmon et al., 2004). This observation leads to the hypothesis
Durlinger et al., 1999; Durlinger et al., 2001; Ikeda et al., 2002). that oocytes in the pool of growing follicles could control the
AMH seems to act only in the reproductive organs (Lee and pool of primordial follicles by modulating the expression of the
Donahoe, 1993). The most striking effect of AMH is its capacity inhibiting factor AMH.
to induce regression of the Müllerian ducts, the anlage of the
female internal reproductive organs. In the absence of AMH,
Circulating AMH in women
Müllerian ducts of both sexes develop into the uterus, Fallopian
tubes and the upper part of the vagina (Munsterberg and In males, AMH levels after birth are low, but rapidly rise to peak
Lovell-Badge, 1991). During fetal life, only the male testis values by late infancy, then slowly decrease to the adult range at
expresses the hormone (Munsterberg and Lovell-Badge, 1991). puberty (Josso et al., 1990; Lee et al., 1996). In women, AMH

266
AMH and ovarian cancer

Downloaded from https://academic.oup.com/humupd/article/13/3/265/2457878 by guest on 14 August 2021


Figure 1. AMH immunostaining in normal human adult ovary. (A) Primordial follicles are negative (arrowheads), whereas a weak positive reaction can already be
observed in the cytoplasm of a few cells in a primary follicle (arrow). (B) All granulosa cells of pre-antral follicles are AMH-positive. (C) AMH-positive and AMH-
negative cells are intermingled in the cumulus oophorus of a pre-ovulatory follicle. (D) Only a few granulosa cells surrounding the antrum (ANT) are AMH-positive.
Thecal cells (TC) are not immunoreactive (original magnifications—A and B: 200; C: 100; and D: 300) (Reprinted from “Anti-Müllerian hormone is a specific
marker of sertoli- and granulosa-cell origin in conadal tuitors” by Rey R. et al. (2000) Human Pathology, 31:1202-1208, with permission from Elsevier).

serum levels are almost undetectable at birth (Rajpert-De Meyts time in young normovulatory women (de Vet et al., 2002), and
et al., 1999), with a subtle increase noted after puberty (Hudson to correlate with age, FSH and the number of antral follicles. In
et al., 1990). a recent study, a group of women was studied twice and the inter-
Serum AMH levels have been measured at different time-points val between the two visits ranged from 1.1 to 7.3 years. A
during the menstrual cycle, suggesting minimal fluctuation reduction in mean AMH levels of about 38% was observed,
(La Marca et al., 2006a). Minimal fluctuations in serum AMH whereas the number of antral follicles and the levels of FSH and
levels may be consistent with continuous non-cyclic growth of inhibin B did not change (de Vet et al., 2002). The decrease in
small follicles. AMH with advancing age may occur before changes in the
Hence, AMH is relatively convenient to determine, especially
as it seems to exhibit a relatively stable expression during the men-
strual cycle, making it an attractive determinant of ovarian activity
(Gruijters et al., 2003).
Recent studies indicate the AMH as a novel measure of ovarian
reserve. Serum AMH levels have been shown to decrease over

Figure 3. Role of AMH in human folliculogenesis. In women, AMH


expression can first be observed in granulosa cells of primary follicles, and
expression is strongest in pre-antral and small antral follicles (4 mm). AMH
expression disappears in follicles of increasing size and is almost lost in follicles
larger than 8–10 mm, where only very weak staining remains, restricted to the
granulosa cells of the cumulus. This expression pattern suggests that AMH may
Figure 2. AMH receptor binding. Upon ligand binding to the type II receptor, play a role in initial recruitment and in the selection of the dominant follicle:
the type I receptor is recruited into the receptor complex and becomes phos- AMH seems to be a negative regulator of the early stages of follicular develop-
phorylated by the type II receptor. Activation of the type I receptor results in ment. AMH prevents recruitment of non-dominant follicles and reduces the
the phosphorylation of the downstream Smad proteins. Phosphorylated responsiveness of ovarian follicles to FSH during cyclic recruitment by
receptor-specific Smads translocate to the nucleus where they regulate gene Stephen et al. (2002) Highly purified Müllerian inhibiting substance inhibits
expression. human ovarian cancer in vitro. Clin Cancer Res. 8, 2640-2646.

267
A. La Marca and A. Volpe

currently known ageing-related variables and this indicates that induces gonadal tumourigenesis in transgenic mice (Peschon
serum AMH levels may be the best marker for ovarian ageing et al., 1992; Dutertre et al., 1997). They also showed that granu-
and menopausal transition. losa cell tumours of less than 100 mg (10-fold the size of
the normal ovary) were positive for both AMH and AMH-RII
mRNA and proteins. Moreover, expression was not found in
AMH and AMH receptor in ovarian cancer larger tumours (Dutertre et al., 2001). They also showed that
Granulosa cell tumour AMHR-II was expressed on the cell surface and was able to
bind to its ligand AMH. They concluded that GCT of transgenic
In the first description of increased AMH in serum from a patient mice produce AMH and secrete it into the blood when the
with GCT (Gustafson et al., 1992), the AMH gene expression in tumour is relatively small. In advanced stages, the GCT invades
neoplasm specimens was also investigated. RNA was extracted organs and develops metastases and loses both AMH and
and analysed by northern blotting showing that a 2.0 kb AMH AMH-RII expression. Considering the inhibitory effect of AMH
RNA transcript was present. on GCT growth (Kim et al., 1992), this phenomenon could indi-

Downloaded from https://academic.oup.com/humupd/article/13/3/265/2457878 by guest on 14 August 2021


In a small series of ovarian tumours, AMH expression was cate a counter-selection to favour further tumour progression.
detected in all of the GC tumours but not in the seven epithelial
carcinomas (Matias-Guiu et al., 1998). Using paraffin-fixed sec-
Epithelial ovarian cancer
tions and antibody directed to AMH, authors demonstrated that
AMH immunoreactivity was always present in the GC tumours More recently, the AMH-RII expression has been found in human
but was not as strong or diffuse as alpha-inhibin. The finding of epithelial ovarian cancer (Masiakos et al., 1999). Human epithelial
AMH expression in both juvenile- and adult-type GCT of either ovarian cancer cells and ascites cells isolated from patients with
ovarian or metastatic localization has been recently confirmed ovarian papillary serous cystoadenocancer were processed and
(Rey et al., 2000) (Figure 4). analysed by RT – PCR for the detection of AMH-RII mRNA.
Dutertre et al. (2001) developed a murine transgenic model of AMH-RII transcripts were detected in cell lines from 10 of the
GCT of the ovary. They showed that driving the expression of 15 patients. In 89% of the cases, the presence of AMH-RII
the simian virus 40 (SV40) oncogene by the AMH promoter mRNA was predictive of the binding to AMH. In the solid
tumours from four patients AMH-RII immunostaining was also
demonstrated.

AMH as circulating tumour marker for GCT


Gustafson et al. (1992) have demonstrated abnormalities of AMH
secretion in relation to ovarian malignancy in stored sera from 18
patients. Elevated serum concentrations of AMH was present in
four women with GCT. Serum AMH levels were normal in six
women who had undergone resection of GCT 2– 45 months
before and who were disease-free. AMH was undetectable in
serum from women with metastatic epithelial ovarian cancer and
in three women with previously resected ovarian carcinoma. Simi-
larly, AMH was undetectable in serum from one woman with
Fallopian tube carcinoma and from a woman with mesonephric
adenocancer. Two women with bilateral gonadoblastoma or
bilateral dysgerminoma AMH did not exhibit increased serum
AMH levels.
Hence, based on these findings it seems that AMH is increased
only in the serum from women with GCT.
One patient with GCT was followed in the clinical course for
about 6 years and the results of hormonal evaluation demonstrated
that serum AMH levels decreased after each of the subsequent
major tumour resections. Furthermore, elevations of AMH pre-
ceded or accompanied recurrences of disease.
A successive study confirmed that AMH is a serum marker
limited to GCT. Rey et al., 1996 showed normal AMH levels in
93.3% of patients with non-gynaecologic cancer or gynaecologic
cancer other than GCT. In eight of nine patients with progressive
GCT, AMH levels were above the normal range, and in one patient
with clinical remission of the disease, AMH was below the detec-
Figure 4. AMH-immunostaining of primary GCT. (A) Juvenile-type GCT of
ovarian localization. (B) Adult-type GCT of ovarian localization. In all tion limit.
cases, staining is in the cytoplasm of positive cells (original magnifications— In their longitudinal study of 11 patients with recurrent GCT
A: 300; B: 500) (for permission see fig. 1). (Rey et al., 1996), AMH showed a good correlation with evolution

268
AMH and ovarian cancer

Table I. Sensitivity of AMH measurement as marker of GCT

Reference N Type of AMH assay Sensitivity (%) Specificity (%) PPV (%) NPV (%)

Gustafson (1992) 4 Traditional 100


Rey (1996) 9 Traditional 88.8
Lane (1999) 8 (JGCT) Traditional 75
9 (AGCT) 78
Long (2000) 31 New ultrasensitive 93 96 93 96

JGCT, Juvenile granulosa-cell tumor.


AGCT, Adult granulosa-cell tumor.

of the disease during follow-up in nine cases. Serum AMH fell to and one-quarter reaching complete pathological response.
undetectable values after successful treatment or rose early in One-half of those patients with complete pathological response

Downloaded from https://academic.oup.com/humupd/article/13/3/265/2457878 by guest on 14 August 2021


cases of recurrence of the disease. These findings indicate that will relapse. It is expected that 70 – 80% of all patients with
AMH can be used to evaluate the efficacy of the treatment. Inter- advanced ovarian cancer will need second-line therapy (Kalil
estingly, comparing AMH and alpha-inhibin, it has been shown and McGuire, 2002). The object of second-line therapy is to
that serum alpha-inhibin fluctuated between normal and high reduce symptoms, minimize toxicity and prolong the progression-
values more frequently than did AMH. AMH has also been free interval, with palliative intent.
measured intraoperatively in cystic fluid from two children with AMH may also be applicable as a chemotherapeutic agent for
juvenile GCT, showing elevated concentrations in comparison epithelial ovarian cancer. AMH induces regression of the Muller-
with circulating ones, thus indicating the local production (Rey ian ducts, which arise from a common embryological precursor to
et al., 1996). the surface epithelium of the ovary, from which epithelial ovarian
Globally, AMH serum levels seem to be elevated in 76% (Lane tumours originate (La Marca et al., 2006b). The inhibitory effect
et al., 1999) to 93% (Long et al., 2000) of GCT patients (Table I). of the hormone is believed to be consequent to its binding to the
The mean AMH level was 190.3 ng ml21 (range 2 – 1124 ng ml21) AMH-RII. Indeed, expression of AMH-RII has been demonstrated
in patients with GCT in one study (Lane et al., 1999). In patients in at the transcriptional and translational level in a series of human
whom serial measurements were made following initial tumour ovarian cancer cell lines (Masiakos et al., 1999).
resection, the length of time that an elevation in AMH levels Masiakos et al. (1999) demonstrated that the growth of five of
preceded clinical detection of a recurrence was 3 months (Lane six epithelial ovarian carcinoma cell lines expressing AMH-RII
et al., 1999). was inhibited by exogenous recombinant AMH. Moreover, 56%
An ultrasensitive ELISA assay for AMH was developed some of primary ascites cells seem to bind AMH (Masiakos et al.,
years ago (Long et al., 2000). The new ultrasensitive ELISA 1999). Using the colony inhibition assay, it has been demonstrated
assay is a sandwich-type assay where sensitivity is as low as that AMH binding to its receptor is followed by inhibition of the
0.1 ng ml21 (in comparison with 2 ng ml21 of the traditional cell growth (Chin et al., 1991; Masiakos et al., 1999; Ha et al.,
assay used in the studies published before the year 2000). Using 2000; Stephen et al., 2001).
the new assay, Long et al. (2000) found that sensitivity, specificity, The growth inhibition correlates with a block in cell cycle
positive predictive value (PPV) and negative predictive value progression and is characterized by apoptosis. These events are
(NPV) for the presence of GCT were 93, 96, 93, 96%, respectively. mediated by induction of the cyclin-dependent kinase inhibitor
The relevance of using an ultrasensitive assay is demonstrated p16, which results in the up-regulation of the E2F family of
by the fact that in 6 of 31 patients the new and the traditional transcription factors that are regulated by members of the pocket
assays gave discordant results. Indeed in these patients, AMH protein family (Ha et al., 2000).
was undetectable using the traditional assay but readily detectable In a recent study human ovarian cancer cells were implanted in
with the ultrasensitive AMH ELISA (Long et al., 2000). immunosuppressed mice and treated with recombinant AMH
(Stephen et al., 2002). Two to three weeks of AMH treatment
was followed by a significant reduction in the graft size ratio
AMH as a chemotherapeutic agent for epithelial
indicating that AMH delivered parenterally may cause inhibition
ovarian cancer
of human cancer cell lines in vivo (Figure 5).
About two-thirds of the new cases of epithelial ovarian cancer
diagnosed every year are at an advanced stage (Greenlee et al.,
Conclusions
2001). Once the tumour has spread beyond the confines of the
ovary, surgery alone is unlikely to provide adequate therapy. Ovarian cancer is one of the most frequent gynaecological malig-
After aggressive debulking surgery, primary chemotherapy with nancies. Three major types are recognized: epithelial, stromal and
curative intent with platinum and taxanes is the standard of care. germ cell. Epithelial ovarian tumours are the most common human
Carboplatin and cisplatin are equally effective, but carboplatin ovarian carcinomas accounting for 82% of all the human ovarian
has less toxicity and is the most common platinum compound cancers. The majority of these cancers present at III or IV stage,
used as first line in combination with paclitaxel. hence the 5-year mortality rate for the disease is about 70% (Can-
In patients with advanced epithelial ovarian disease, the overall nistra, 1993).
response to primary platinum-based therapy is 70 – 80%, with GCT constitutes 3% of all ovarian tumours. GCT is divided in to
one-half of these patients achieving complete clinical response the adult and the juvenile form. Of women with adult GCT, 90%

269
A. La Marca and A. Volpe

Downloaded from https://academic.oup.com/humupd/article/13/3/265/2457878 by guest on 14 August 2021


Figure 5. Effects of AMH on ovarian cancer. OVCAR 8 is a human ovarian cancer cell line, which expresses the AMH-RII. OVCAR 8 tumours were implanted
under the renal capsules of nude mice after 3 weeks. (A) Control tumour shows neovascularization and minimal necrosis or inflammation. (magnification: 40). (B)
AMH-treated tumour (10-mg day213 weeks delivered i.p.) is about one-third the size of the control tumour and has minimal necrosis or inflammation
(magnification:  40) (Reprinted from “Highly purified Müllerian inhibiting substance inhibits human ovarian cancer in vivo” by Stephen et al. Clinical Cancer
Research 2002, 8: 2640– 2646, with permission from American Association for Cancer Research).

present with stage I and the survival rate at 5 years remains very for GCT. Its diagnostic performance seems to be very good,
high (Bjorkholm and Silfverswaard, 1981). The recurrence for with a sensitivity ranging between 76 and 93%. AMH seems to
adult GCT is late occurring: 4 – 6 years after the first treatment. be superior to alpha-inhibin and estradiol in the early diagnosis
In contrast, a large subset experience recurrence of juvenile and in the follow-up of GCT.
GCT occurs within one year, with a mean of 6 months (Calaminus It is largely recognized that AMH and alpha-inhibin
et al., 1997). exhibit a higher degree of sensitivity than estradiol in pro-
In recent years, the advances in the physiological role of AMH gressive GCT. Indeed, estradiol production by GCT is
has stimulated interest in the significance of AMH as diagnostic widely variable (Lappohn et al., 1989) while AMH seems to
marker and therapeutic agent for ovarian cancer. At this be a marker of comparable value to alpha-inhibin. However,
moment, AMH has only been shown to be a circulating marker AMH is elevated only in sex cord stromal tumours, while

270
AMH and ovarian cancer

inhibin may be elevated in different types of cancer (Healy (vii) Recurrences in juvenile GCTs occur within a year of
et al., 1993). initial diagnosis at a mean of 5 – 6 months. Hence, in
Doubts remain on the possible role of AMH in the pathogenesis juvenile GCT, AMH serum levels should be determined
of GCT. Indeed, AMH is considered a powerful inhibitor of every month following the surgery.
granulosa cell proliferation. Hence, the high serum AMH levels At this moment, data on the role of AMH measurement in moni-
found in patients with GCT, are in contrast with the progressive toring the response to chemotherapy is lacking.
characteristic of the disease. One possible explanation is that the These recommendations are based on a limited number of
binding of AMH-RII by its ligand in tumour cells is not followed studies. Hence, they should be validated in clinical trials.
by the inhibitory signal. Otherwise from animal studies it has been Several authors have also hypothesized a role for recombinant
hypothesized that the inhibitory signal is functional when the AMH in the treatment of epithelial ovarian cancer (Stephen
tumours are limited (Dutertre et al., 2001). Successively, when et al., 2001, 2002). Based on the physiological inhibiting role of
the tumours become progressive and reach a large dimension, AMH on the mullerian ducts, researchers have demonstrated
they lose AMH and AMH-RII expression. This could be a that AMH inhibits epithelial ovarian cancer cell in vitro (Fuller

Downloaded from https://academic.oup.com/humupd/article/13/3/265/2457878 by guest on 14 August 2021


counter selection to favour further tumour progression (Dutertre et al., 1982, 1985; Stephen et al., 2002; Masiakos et al., 1999).
et al., 2001). In a small series, it has been shown that 56% of ovarian cancer
In patients treated for GCT, AMH may be followed post- ascite cells express the receptor for AMH, and when tested in
operatively as a marker for efficacy of surgery and disease recur- solid tumours from patients with epithelial ovarian cancer, in
rence. Following surgery, AMH decreases to normal values within each case AMH-RII was expressed (Masiakos et al., 1999). The
days to weeks. The AMH half-life is estimated to be 48 h (Vigier administration of recombinant AMH in immunosuppressed mice
et al., 1983). Hence, reduced levels of AMH should be detected with human epithelial ovarian cancer implants has been followed
72 h after successful surgery. Indeed, bilateral ovariectomy per- by reduced dimensions of the graft (Stephen et al., 2002).
formed for benign disease is followed by undetectable AMH This of course will be the basis for future research aiming to
levels 3 – 5 days after surgery (La Marca et al., 2005). investigate the possible clinical role of AMH as neo-adjuvant or
In the past, anti-AMH antibodies were not commercially avail- most probably adjuvant therapy for ovarian cancer.
able. Only few laboratories in the world performed the assay and
this has precluded the diffusion of AMH as a marker for GCT. The
References
recent availability of ultrasensitive commercial ELISA will permit
its clinical application (Rey et al., 1999; Long et al., 2000). This Al-Qahtani A, Muttukrishna S, Appasamy M, Johns J, Cranfield M, Visser JA,
Themmen AP and Groome NP (2005) Development of a sensitive enzyme
ELISA, which is valid both for males and females, is a sandwich- immunoassay for anti-Müllerian hormone and the evaluation of potential
type assay where sensitivity is as low as 0.1 ng ml21 (in compari- clinical applications in males and females. Clin Endocrinol 63,267–273.
son with 2 ng ml21 for the traditional assay used in the studies Bilezikjian LM, Blount AL, Leal AM, Donaldson CJ, Fischer WH and Vale
WW (2004) Autocrine/paracrine regulation of pituitary function by
published before 2000). More recently, a new double-antibody activin, inhibin and follistatin. Mol Cell Endocrinol. 225,29–36.
ELISA for AMH has been developed, with a detection limit of Biscotti CV and Hart WR (1989) Juvenile granulosa cell tumors of the ovary.
,0.078 ng ml21 (Al-Qahtani et al., 2005). Arch Pathol Lab Med 113,40– 46.
The specificity of the commercially available ultrasensitive Bjorkholm E and Silfversward C (1981) Prognostic factors in granulosa-cell
tumors. Gynecol Oncol 11,261–274.
assay was tested by assaying samples containing known concen- Calaminus G, Wessalowski R, Harms D and Gobel U (1997) Juvenile granulosa
trations of most of the AMH-related members of a large family cell tumors of the ovary in children and adolescents: results from 33 patients
of growth and differentiation factors: TGF-b, inhibin A and B, registered in a prospective cooperative study. Gynecol Oncol 65,447–452.
inhibin pro-aC subunit and activin A. No cross reactivity was Cannistra SA (1993) Cancer of the ovary. N Engl J Med 329,1550– 1559.
Chin TW, Parry RL and Donahoe PK (1991) Human mullerian inhibiting
observed in any of the cases (Long et al., 2000). substance inhibits tumor growth in vitro and in vivo. Cancer Res
Based on the published studies it is possible to elaborate the fol- 51,2101–2106.
lowing recommendations for the use of AMH as a marker of GCT: de Vet A, Loven JS, de Jong FH, Themmen AP and Fauser BC (2002)
Anti-Mullerian hormone serum levels: a putative marker for ovarian
aging. Fertil Steril 77,357–362.
(i) High serum AMH levels are found in 76 – 93% of patients Donahoe PK, Fuller AF Jr, Scully RE, Guy SR and Budzik GP (1981)
with GCT. Mullerian inhibiting substance inhibits growth of a human ovarian
(ii) Serum AMH levels should normalize within few days fol- cancer in nude mice. Ann Surg,194,472– 480.
Durlinger AL, Kramer P, Karels B, de Jong FH, Uilenbroek JT, Grootegoed JA
lowing surgery. and Themmen AP (1999) Control of primordial follicle recruitment by
(iii) Persistent AMH levels are indicative of residual disease. anti-Mullerian hormone in the mouse ovary. Endocrinology 140,5789–
(iv) When bilateral ovariectomy is performed AMH must be 5796.
Durlinger AL, Gruijters MJ, Kramer P, Karels B, Kumar TR, Matzuk MM,
undetectable in serum. Even very low levels may be sug- Rose UM, de Jong FH, Uilenbroek JT and Grootegoed JA et al. (2001)
gestive of residual disease. Anti-Mullerian hormone attenuates the effects of FSH on follicle
(v) A post-operative rise in serum AMH levels may precede development in the mouse ovary. Endocrinology, 142,4891–4899.
the clinical recurrence. Hence, the clinical examination Durlinger AL, Visser JA and Themmen AP (2002) Regulation of ovarian
function: the role of anti-Mullerian hormone. Reproduction 124,601–609.
and imaging should be anticipated. Dutertre M, Rey R, Porteu A, Josso N and Picard JY (1997) A mouse Sertoli
(vi) AMH levels should be obtained every 6 months for at least cell line expressing anti-Mullerian hormone and its type II receptor.
5 years after the surgery. However, it should not be Mol Cell Endocrinol 136,57–65.
Dutertre M, Gouedard L, Xavier F, Long WQ, di Clemente N, Picard JY
ignored that a late recurrence (up to 30 years after initial
and Rey R (2001) Ovarian granulosa cell tumors express a functional
treatment) has been reported in up to 33% of patients membrane receptor for anti-Mullerian hormone in transgenic mice.
(Stenwig et al., 1979). Endocrinology 142,4040– 4046.

271
A. La Marca and A. Volpe

Fuller AF, Guy S, Budzik GP and Donahoe PK (1982) Mullerian inhibiting Long WQ, Ranchin V, Pautier P, Belville C, Denizot P, Cailla H, Lhomme C,
substance inhibits colony growth of a human ovarian carcinoma cell Picard JY, Bidart JM and Rey R (2000) Detection of minimal levels of
line. J Clin Endocrinol Metab 54,1051 –1055. serum anti-Mullerian hormone during follow-up of patients with ovarian
Fuller AF Jr, Krane IM, Budzik GP and Donahoe PK (1985) Mullerian granulosa cell tumor by means of a highly sensitive enzyme-linked
inhibiting substance reduction of colony growth of human gynecologic immunosorbent assay. J Clin Endocrinol Metab 85,540–544.
cancers in a stem cell assay. Gynecol Oncol. 22,135– 148. Masiakos PT, MacLaughlin DT, Maheswaran S, Teixeira J, Fuller AF Jr,
Greenlee RT, Hill-Harmon MB, Murray T and Thun M (2001) Cancer Shah PC, Kehas DJ, Kenneally MK, Dombkowski DM and Ha TU
statistics. Cancer J Clin 51,144–154. et al. (1999) Human ovarian cancer, cell lines, and primary ascites
Gruijters MJ, Visser JA, Durlinger AL and Themmen AP (2003) cells express the human Mullerian inhibiting substance (MIS) type II
Anti-Mullerian hormone and its role in ovarian function. Mol Cell receptor, bind, and are responsive to MIS. Clin Cancer Res 5,3488–
Endocrinol 211,85–90. 3499.
Gustafson ML, Lee MM, Scully RE, Moncure AC, Hirakawa T, Goodman A, Matias-Guiu X, Pons C and Prat J (1998) Mullerian inhibiting substance,
Muntz HG, Donahoe PK, MacLaughlin DT and Fuller AF (1992) alpha-inhibin, and CD99 expression in sex cord-stromal tumors and
Mullerian inhibiting substance as a marker for ovarian sex-cord tumor. endometrioid ovarian carcinomas resembling sex cord-stromal tumors.
N Engl J Med 326,466 –471. Hum Pathol 29,840– 845.
Ha TU, Segev DL, Barbie D, Masiakos PT, Tran TT, Dombkowski D, Munsterberg A and Lovell-Badge R (1991) Expression of the mouse

Downloaded from https://academic.oup.com/humupd/article/13/3/265/2457878 by guest on 14 August 2021


Glander M, Clarke TR, Lorenzo HK and Donahoe PK et al. (2000) anti-mullerian hormone gene suggests a role in both male and female
Mullerian inhibiting substance inhibits ovarian cell growth through an sexual differentiation. Development 113,613–624.
Rb-independent mechanism. J Biol Chem 275,37101– 37109. Nakashima N, Young RH and Scully RE (1984) Androgenic granulosa cell
Healy DL, Burger HG, Mamers P, Jobling T, Bangah M Quinn M, Grant P, tumors of the ovary. A clinicopathologic analysis of 17 cases and
Day AJ, Rome R and Campbell JJ (1993) Elevated serum inhibin review of the literature. Arch Pathol Lab Med 108,786–791.
concentrations in postmenopausal women with ovarian tumors. N Pankratz E, Boyes DA, White GW, Galliford BW, Fairey RN and Benedet JL
Engl J Med 329,1539–1542. (1978) Granulosa cell tumors. A clinical review of 61 cases. Obstet
Hudson PL, Dougas I, Donahoe PK, Cate RL, Epstein J, Pepinsky RB Gynecol 52,718–723.
and MacLaughlin DT (1990) An immunoassay to detect human Peschon JJ, Behringer RR, Cate RL, Harwood KA, Idzerda RL, Brinster RL
mullerian inhibiting substance in males and females during normal and Palmiter RD (1992) Directed expression of an oncogene to Sertoli
development. J Clin Endocrinol Metab 70,16– 22. cells in transgenic mice using mullerian inhibiting substance regulatory
Ikeda Y, Nagai A, Ikeda MA and Hayashi S (2002) Increased expression of sequences. Mol Endocrinol 6,1403–1411.
Mullerian-inhibiting substance correlates with inhibition of follicular Rajpert-De Meyts E, Jorgensen N, Graem N, Muller J, Cate RL
growth in the developing ovary of rats treated with E2 benzoate. and Skakkebaek NE (1999) Expression of anti-Mullerian hormone
Endocrinology 143,304– 312. during normal and pathological gonadal development: association with
Imbeaud S, Carre-Eusebe D, Rey R, Belville C, Josso N and Picard JY (1994) differentiation of Sertoli and granulosa cells. J Clin Endocrinol Metab
Molecular genetics of the persistent mullerian duct syndrome: a study of 84,3836– 3844.
19 families. Hum Mol Genet 3,125–131. Rey RA, Lhomme C, Marcillac I, Lahlou N, Duvillard P, Josso N and Bidart
Ingraham HA, Hirokawa Y, Roberts LM, Mellon SH, McGee E, Nachtigal MW JM (1996) Anti-mullerian hormone as a serum marker of granulosa cell
and Visser JA (2000) Autocrine and paracrine Mullerian inhibiting tumorsof the ovary: comparative study with serum alpha-inhibin and
substance hormone signaling in reproduction. Recent Prog Horm Res estradiol. Am J Obstet Gynecol 174,958–965.
55,53–67. Rey RA, Belville C, Nihoul-Fekete C, Michel-Calemard L, Forest M.G, Lahlou
Josso N, Legeai L, Forest MG, Chaussain JL and Brauner R (1990) An enzyme N, Jaubert F, Mowszowicz I, David M and Saka N et al. (1999) Evaluation of
linked immunoassay for anti-mullerian hormone: a new tool for the gonadal function in 107 intersex patients by means of serum antimüllerian
evaluation of testicular function in infants and children. J Clin hormone measurement. J Clin Endocrinol Metab 84,627–631.
Endocrinol Metab 70,23– 27. Rey R, Sabourin JC, Venara M, Long WQ, Jaubert F, Zeller WP, Duvillard P,
Josso N, Racine C, di Clemente N, Rey R and Xavier F (1998) The role of Chemes H and Bidart JM (2000) Anti-Mullerian hormone is a specific
anti-Mullerian hormone in gonadal development. Mol Cell Endocrinol marker of sertoli- and granulosa-cell origin in gonadal tumors. Hum
145,3–7. Pathol 31,1202 –1208.
Kalil NG and McGuire WP (2002) Chemotherapy for advanced epithelial Robertson DM, Cahir N, Burger HG, Mamers P and Groome N (1999a) Inhibin
ovarian carcinoma. Best Pract Res Clin Obstet Gynaecol 16,553–571. forms in serum from postmenopausal women with ovarian cancers. Clin
Kim JH, Seibel MM, MacLaughlin DT, Donahoe PK, Ransil BJ, Hametz PA Endocrinol (Oxf) 50,381–386.
and Richards CJ (1992) The inhibitory effects of Müllerian-inhibiting Robertson DM, Cahir N, Burger HG, Mamers P, McCloud PI, Pettersson K
substance on epidermal growth factor induced proliferation and and McGuckin M (1999b) Combined inhibin and CA125 assays in the
progesterone production of human granulosa-luteal cells. J Clin detection of ovarian cancer. Clin Chem. 45,651–658.
Endocrinol Metab 75,911–917. Salerno LJ (1962) Feminizing mesenchymomas of the ovary. An analysis of 28
LaMarca A, De Leo V, Giulini S, Orvieto R, Malmusi S, Giannella L and Volpe granulosa-theca cell tumors and their relationship to coexistent carcinoma.
A (2005) Anti-Mullerian hormone in premenopausal women and after Am J Obstet Gynecol 84,731–738.
spontaneous or surgically induced menopause. J Soc Gynecol Invest Salmon NA, Handyside AH and Joyce IM (2004) Oocyte regulation of
12,545– 548. anti-Mullerian hormone expression in granulosa cells during ovarian
La Marca A and Volpe A (2006b) Anti-Mullerian hormone (AMH) in female follicle development in mice. Dev Biol 266,201– 208.
reproduction: is measurement of circulating AMH a useful tool? Clin Seifer DB, MacLaughlin DT, Penzias AS, Behrman HR, Asmundson L,
Endocrinol (Oxf) 64,603– 610. Donahoe PK, Haning RVJ and Flynn SD (1993) Gonadotropin-releasing
La Marca A, Stabile G, Artenisio AC and Volpe A (2006a) Serum hormone agonist-induced differences in granulosa cell cycle kinetics are
anti-Mullerian hormone throughout the human menstrual cycle. Hum associated with alterations in follicular fluid müllerian-inhibiting
Reprod 21,3103–3107. substance and androgen content. J Clin Endocrinol Metab 76,711–714.
Lane AH Jr, Lee MM, Fuller AF, Kehas DJ, Donahoe PK and MacLaughlin DT Skinner MK (2005) Regulation of primordial follicle assembly and
(1999) Diagnostic utility of Mullerian inhibiting substance determination development. Hum Reprod Update 11,461– 471.
in patients with primary and recurrent granulosa cell tumors. Gynecol Stenwig JT, Hazekamp JT and Beecham JB (1979) Granulosa cell tumors of the
Oncol 21,3103– 3107. ovary. A clinicopathological study of 118 cases with long-term follow-up.
Lappohn RE, Burger HG, Bouma J, Bangah M, Krans M and de Bruijn HW Gynecol Oncol 7,136– 152.
(1989) Inhibin as a marker for granulosa-cell tumors. N Engl J Med Stephen AE, Masiakos PT, Segev DL, Vacanti JP, Donahoe PK
321,790–793. and MacLaughlin DT (2001) Tissue-engineered cells producing
Lee MM and Donahoe PK (1993) Mullerian inhibiting substance: a gonadal complex recombinant proteins inhibit ovarian cancer in vivo. Proc Natl
hormone with multiple functions. Endocr Rev 14,152–164. Acad Sci USA 98,3214–3219.
Lee MM, Donahoe PK, Hasegawa T, Silverman B, Crist GB, Best S, Stephen AE, Pearsall LA, Christian BP, Donahoe PK, Vacanti JP
Hasegawa Y, Noto RA, Schoenfeld D and MacLaughlin DT (1996) and MacLaughlin DT (2002) Highly purified mullerian inhibiting
Mullerian inhibiting substance in humans: normal levels from infancy substance inhibits human ovarian cancer in vivo. Clin Cancer Res.
to adulthood. J Clin Endocrinol Metab 81,571– 576. 8,2640–2646.

272
AMH and ovarian cancer

Themmen AP (2005) Anti-Mullerian hormone: its role in follicular growth Vigier B, Tran D, du Mesnil du Buisson F, Heyman Y and Josso N (1983) Use
initiation and survival and as an ovarian reserve marker. J Natl Cancer of monoclonal antibody techniques to study the ontogeny of bovine
Inst Monogr 34,18–21. anti-Mullerian hormone. J Reprod Fertil 69,207–214.
Tsafriri A, Picard JY and Josso N (1988) Immunopurified anti-Müllerian Weenen C, Laven JS, Von Bergh AR, Cranfield M, Groome NP, Visser JA,
hormone does not inhibit spontaneous resumption of meiosis in vitro of Kramer P, Fauser BC and Themmen AP (2004) Anti-Mullerian
rat oocytes. Biol Reprod 38,481– 485. hormone expression pattern in the human ovary: potential implications
Ueno S, Kuroda T, Maclaughlin DT, Ragin RC, Manganaro TF and for initial and cyclic follicle recruitment. Mol Hum Reprod 10,77– 83.
Donahoe PK (1989) Mullerian inhibiting substance in the adult rat
ovary during various stages of the estrous cycle. Endocrinology Submitted on July 10, 2006; resubmitted on November 3, 2006; accepted on
125,1060– 1066. November 29, 2006

Downloaded from https://academic.oup.com/humupd/article/13/3/265/2457878 by guest on 14 August 2021

273

You might also like