10.1007-s12022-013-9285-4

  • Upload
    jeka222

  • View
    215

  • Download
    0

Embed Size (px)

Citation preview

  • 8/10/2019 10.1007-s12022-013-9285-4

    1/18

    Endocrine Pathology of the Ovary

    In Tribute to Robert E Scully, MD

    Esther Oliva &Robert H. Young

    Published online: 14 January 2014# Springer Science+Business Media New York 2014

    Keywords Ovary. Function . Endocrine. Pathology

    It is a pleasure and honor to be asked to contribute to this

    anniversary issue of the journal. Although tinged with sad-

    ness, it is fortuitous we were asked to write on the endocrine

    pathology of the ovary. This is the very same title as the

    wonderful book on the ovary written by our mentor, Dr.

    Robert E. Scully, with a gynecologist Dr. J.M Morris, in the

    mid 1950s and published in 1958 [1]. We began to work on

    this issue on Dr. Scullys birthday, August 31, the first since

    his death in late October 2012, and the day, and time since

    working on this essay, has produced much reflection. We

    dedicate these pages to Dr. Scully knowing he would be happy

    we are focusing on an area of ovarian pathology of greatinterest to him.

    As is usually the case, it is hard, if not impossible, to

    improve on Dr. Scullys approach; accordingly, we follow

    the outline of his book except that space constraints will not

    allow for a coverage of anatomic and embryological aspects

    or indeed all aspects of the topic overall. However, Dr.

    Scully devotes a chapter to this topic in his fascicle [2].

    We highlight areas we find of greatest interest, expand to a

    degree on aspects such as immunohistochemistry, and update

    the literature review.

    We discuss the following categories in turn: non-neoplastic

    lesions, sex cord-stromal tumors, other neoplasms with

    endocrine manifestations, the fascinating phenomenon of

    ovarian tumors with functioning stroma (Table1), a concept

    Dr. Morris and Dr. Scully had introduced in a review article ayear before their book was published [3], and finally, and

    briefly, paraendocrine disorders. Dr. Scullys interest in endo-

    crine manifestations of ovarian lesions was shown in many

    ways, such as making a reference to it in his seminal paper on

    gonadoblastoma in 1953 [4] and considering it in even more

    detail in his 1970 magnum opus (Fig.1)[5], and co-authoring

    a major contribution on metastatic tumors to the ovary with

    functioning stroma in 1961 [6]. He continued to explore

    endocrine function by non-neoplastic and neoplastic lesions

    over the years [715] and wrote one of the last comprehensive

    reviews on functioning stroma in 1987 [16]. Dr. Scully em-

    phasized traditional pathology, but his curious mind was al-ways interested in new techniques in a balanced way. Indeed,

    as soon as immunohistochemistry became available, he ex-

    plored its use and was one of the first to write about it

    regarding ovarian tumors in both peer-reviewed articles

    [1720] and in a review[21].

    Non-neoplastic Lesions

    This category (Table2) includes processes in which the ovary

    is not grossly abnormal or, if so, is to a limited degree as well

    as those that are typically associated with a mass and may be

    misconstrued as neoplasms, at least in some instances. The

    first group includes stromal hyperplasia/hyperthecosis (Fig. 2)

    and hilus cell hyperplasia. Pure stromal hyperplasia is rare as

    in most cases careful scrutiny shows at a least a minor com-

    ponent of lutein cells placing the process in the category of

    stromal hyperthecosis. Before the advent of immunohisto-

    chemistry, Dr. Scully reported oxidative enzyme activity by

    histochemistry in both luteinized and non-luteinized stromal

    cells in about 60 % of normal ovaries leading to the

    E. Oliva :R. H. Young

    James Homer Wright Pathology Laboratories, Department of

    Pathology, Massachusetts General Hospital, Harvard Medical

    School, Boston, MA, USA

    E. Oliva (*)

    Department of Pathology, Massachusetts General Hospital,

    55 Fruit Street, Warren 219, Boston, MA 02114, USA

    e-mail: [email protected]

    Endocr Pathol (2014) 25:102119

    DOI 10.1007/s12022-013-9285-4

  • 8/10/2019 10.1007-s12022-013-9285-4

    2/18

    introduction of the term enzymatically active stromal cells

    [15]. It was surmised that this was evidence of a spectrum

    from normal to histochemically abnormal to both histologi-

    cally and histochemically abnormal cells. These findings par-

    allel those observed today with inhibin or calretinin

    (Fig. 2c, d) [19, 22]. This spectrum may explain the occasional

    case in which there is some but usually not striking evidence

    of hormonal production (most often in the form of endometrialhyperplasia) in postmenopausal patients whose only ovarian

    pathology is stromal hyperplasia. These cells also express

    calretinin [23, 24]. When lutein cells are present, especially

    in significant numbers (stromal hyperthecosis) (Fig.2a), the

    frequency of endocrine manifestations, most often estrogenic

    but occasionally androgenic, increases [2527]. In some in-

    stances, the combined proliferation of stroma and lutein cells

    results in a grossly evident fibroma-like mass, but one that

    rarely exceeds 7 cm and in contrast to most fibromas is

    typically bilateral. When lutein cells are exuberant and form

    nodular aggregates up to 0.5 cm, the descriptive designation

    nodular hyperthecosismay be used (Fig.2b). A greater size

    by convention would be considered a so-called stromal

    luteoma, although this term is falling out of favor, as in the

    new World Health Organization (WHO) classification, they

    are considered a small steroid cell tumor without further

    subcategorization (see below) [28].

    Hilus cell hyperplasia, if strictly defined, is a much less

    common phenomenon and typically androgenic [29]. Small

    aggregates of hilar (Leydig) cells are common and hilus cell

    hyperplasia should only be diagnosed when one or more

    confluent nodules are present. As with the spectrum of stromal

    hyperthecosis/stromal luteoma, a cutoff of 0.5 cm is a reason-able arbitrary criterion for the distinction between hilus cell

    hyperplasia versus hilar cell tumor. It should be noted that as

    with hilar cell tumors, hilus cell hyperplasia may exhibit

    degenerative-type atypia. Rarely, these lesions may be associ-

    ated with androgenic manifestations [30]. These hyperplastic

    hilar cells are typically inhibin and calretinin positive [19,23,

    31] and they also express relaxin-like factor also known as

    Leydig cell insulin-like factor which also shows weak to

    moderate staining in theca and granulosa cell tumors [32].

    Non-neoplastic lesions that are grossly visible include

    massive edema and fibromatosis, both rare. They are some-

    times associated with menstrual irregularities presumptivelydue to estrogen production and more strikingly have been

    associated with androgenic manifestations, both explained

    by the presence of lutein cells in the background. Some have

    occurred during pregnancy; however, there is no specific

    relation to it [33].

    Non-neoplastic lesions that are cystic include polycystic

    ovarian disease (PCOD). It has been historically considered an

    important disorder in the realm of endocrine pathology but is

    now considered primarily a clinical diagnosis, and ovaries are

    rarely sent for pathologic examination. Furthermore, the his-

    tologic findings in isolation are not diagnostic. Of interest, the

    granulosa cells lining follicles in PCOD are negative for-

    inhibin and positive for-subunits. In contrast, the hyperplas-

    tic theca cells exhibit distinct positivity for all inhibin subunits

    [34]. They also produce excess activin or insufficient

    Table 1 Outlinecategories of functioning ovarian lesions

    1. Non-neoplastic lesions

    2. Thecoma

    3. Other stromal lesions

    4. Granulosa cell tumors

    5. Sertoli and SertoliLeydig cell tumors

    6. Sex cord tumor with annular tubules

    7. Miscellaneous other neoplasms with endocrine function

    8. Ovarian tumors with functioning stroma

    Fig. 1 Gonadoblastoma (right).

    Note large aggregate of lutein

    cells which may account for

    endocrine manifestations beneath

    typical nests containing sex cord

    and germ cells and the common

    calcification of this entity. Atleft

    are portions of the title pages of

    Dr. Scullys original description

    and his later study of 74 cases

    Endocr Pathol (2014) 25:102119 103

  • 8/10/2019 10.1007-s12022-013-9285-4

    3/18

    follistatin which may contribute to theca cell hyperplasia [35].

    Much more common are follicle cysts, a frequent cause of

    ovarian enlargement and symptomatology, the latter including

    endocrine manifestations. In the reproductive age group, the

    not infrequent menstrual irregularities are likely due to estro-

    gen production but it is in the premenarchal years that a more

    dramatic presentation, isosexual pseudoprecocity, may be

    seen [36,37]. Immunostaining of the cyst fluid with inhibin

    may be of value in confirming the presence of granulosa cells,

    thus establishing this diagnosis [38]. Theca and granulosa

    cells of follicle cysts are also typically positive for calretinin

    [23,24]. Occasionally, follicle cysts, often multiple and bilat-

    eral, are a component of the McCuneAlbright syndrome,

    characterized by the triad of polyostotic fibrous dysplasia,

    caf-au-lait skin pigmentation, and precocious puberty [39]

    due to post-zygotic activating mutations of arginine 201 in the

    guaninenucleotide-binding protein (G protein) -subunit

    [40]. Ovarian cyst formation and regression in these patients

    is often described as a sign of ovarian follicle hyperactivation;

    however, there is heterogeneity of the clinical manifestations

    [41]. A variant of follicle cyst, so-called large follicle cyst of

    pregnancy and the puerperium is, enigmatically, not associat-

    ed with endocrine function. Although the pathogenesis of

    these cysts is unknown, high levels of human chorionic go-

    nadotropin stimulation probably play an important role in their

    development [4244]. A characteristic feature of these cysts

    that also differs from conventional follicle cyst is the presence

    focally of bizarre nuclei in 10 to 50 % of the lining cells [ 42].Two important non-neoplastic lesions that may be func-

    tioning are pregnancy luteoma (Fig. 3) and hyperreactio

    luteinalis (Fig. 4), both of which may be associated with

    androgenic manifestations in about 25 and 15 %, respectively,

    but only the former is associated with virilization of female

    offspring, seen in 60 to 70 % of the cases [4550]. These two

    lesions differ dramatically grossly, as pregnancy luteoma is

    composed of multiple solid nodules (50 %) and shows a

    brown, reddish cut surface, whereas hyperreactio luteinalis is

    composed of multiple thin-walled cysts and it is almost in-

    variably bilateral in contrast to pregnancy luteoma (bilateral

    only in up to 40 %) [51,52]. The morphology of pregnancyluteoma has recently been reviewed in detail [53]. The cysts of

    hyperreactio are in isolation similar to typical follicle cysts,

    differing only in their number and additionally stromal edema

    and luteinization are common.

    Sex Cord-Stromal Tumors

    Thecoma

    This is one of the two ovarian tumors classically associated

    with estrogen production. The frequency of estrogenic

    Fig. 2 Stromal hyperthecosis

    (a). Steroid-type cells with

    eosinophilic cytoplasm are

    scattered within the ovarian

    stroma. A large nodule of such

    cells, but not forming a gross

    mass, is descriptively considered

    nodular hyperthecosis(b). In

    some cases, cortical stroma

    without evident lutein cells shows

    immunoreactivity for inhibin (c)

    and luxuriant staining for inhibin,or calretinin (d), is typical of

    overt hyperthecosis

    Table 2 Non-neoplastic lesions potentially associated with function

    1. Stromal hyperplasia

    2. Stromal hyperthecosis

    3. Hilus cell hyperplasia

    4. Polycystic ovarian disease

    5. Massive edema

    6. Fibromatosis

    7. Follicle cyst

    8. Hyperreactio luteinalis

    9. Pregnancy luteoma

    104 Endocr Pathol (2014) 25:102119

  • 8/10/2019 10.1007-s12022-013-9285-4

    4/18

    manifestations is hard to ascertain with certainty, but in one

    series as many as 21 % of patients had endometrial carcinoma,

    presumptively due to estrogen production [54]. These tumors

    are less common than granulosa cell tumors and differ from

    them from the clinical, gross, and microscopic viewpoints.

    They occur about 10 years later (63 versus 53 years) and areless often associated with pelvic symptomatology due to their

    smaller size (average 7 cm). They are typically solid, lobulat-

    ed, and yellow to white. Microscopic examination shows

    sheets and nodules of pale graycells with ill-defined cyto-

    plasmic borders (Fig.5) [55]. In our opinion, the lipid rich

    quality of the cells has been often overemphasized in the

    literature. Another well-known feature, hyaline plaques, are

    indeed common but may be seen in other tumors including

    fibromas, microcystic stromal tumors [56, 57], and even

    endometrioid stromal sarcoma [57, 58]. Thecomas are typi-

    cally positive for inhibin [19], a 32-kDa heterodimeric glyco-

    protein hormone composed of an- and a-subunits that innormal conditions is secreted by ovarian granulosa cells.

    Inhibin is also produced by testicular Sertoli cells, and extra-

    gonadal expression has been demonstrated in the placenta,

    pituitary gland, and adrenal gland. Inhibin has autocrine and

    paracrine effects in addition to its role in suppressing follicle-

    stimulating hormone secretion by the pituitary gland. Thus,

    inhibin acts as a modulator of folliculogenesis [9]. These

    tumors are also typically positive for calretinin, a more sensi-

    tive but less specific marker than inhibin in the diagnosis of

    sex cord-stromal tumors in general [59]. As calretinin is a

    calcium-binding protein, and these proteins as well as calciumions are involved in endocrine secretion by theca interna cells

    and corpus luteum in the normal ovary, calretinin positivity in

    theca interna cells, and some luteinized granulosa cells of the

    corpus luteum may suggest its expression is related to steroid

    secretion [60,61]. Although melan-A has not been reported in

    ovarian thecomas, Zhang and colleagues have reported

    fibrothecomas of the testis to be positive for this marker

    [62]. The FOXL2 gene encodes a transcription factor that is

    required for granulosa cell function and ovarian follicle de-

    velopment and it is typically expressed in adult and juvenile

    granulosa cell tumors. This is a sensitive marker of sex cord-

    stromal tumors and it can also be positive in thecomas [ 63].FOXL-2 mis-sense mutations have been reported in 20 % of

    thecomas [64]. However, it has to be noted that some tumors

    diagnosed as thecomas and having this mutation have been

    reclassified as granulosa cell tumors, highlighting the

    Fig. 4 Hyperreactio luteinalis. Part of four cysts separated by ovarian

    stroma are seen. Individually, the cysts are identical to follicle cysts of the

    non-pregnant ovary

    Fig. 5 Thecoma. Typical pale cytoplasm which is less lipid-rich than

    often stated in the literature. Calcification, focally seen here, may be

    striking, particularly in tumors of younger patients

    Fig. 6 Sclerosing stromal tumor. This example from a pregnant patient

    shows lutein cells in greater number and with a more robust appearance

    than is typical, likely due to the HCG stimulation of pregnancy. Note a

    striking ectatic vessel, a typical feature of this neoplasm, although it may

    be seen in other stromal tumors

    Fig. 3 Pregnancy luteoma. Follicle-like spaces containing colloid-like

    material are relatively common in this entity

    Endocr Pathol (2014) 25:102119 105

  • 8/10/2019 10.1007-s12022-013-9285-4

    5/18

    difficulty of the differential diagnosis of thecoma versus gran-

    ulosa cell tumor in some cases. Of note, and although unre-

    lated to endocrine manifestations, consistent numerical chro-

    mosomal aberrations have been described in 21 of 29 ovarian

    tumors in the thecomafibroma group, trisomy or tetrasomy

    12 being most common [65].

    Other Stromal Tumors

    Although fibromas are conventionally considered non-

    functioning, and usually are, they may be associated with

    endocrine manifestations if they contain lutein cells. Such

    tumors, until recently, were placed in the luteinized thecoma

    category [66,67], but the upcoming WHO classification ig-

    nores the lutein cells with regard to nomenclature, although

    they should be mentioned in a note if they help explain

    hormone function [28, 68]. A similar comment pertains to

    cases in which lutein-like cells in stromal neoplasms contain

    crystals of Reinke, enabling them to be designated as Leydig

    cells. These rare neoplasms have been reported as stromal

    Leydig cell tumor [69], but the current WHO classification

    does not include this tumor as a specific entity [28]. Some

    fibromas containing lutein cells have been associated with

    androgenic manifestations [67,68]. Finally, even when lutein

    cells are not seen, some fibromas may express inhibin as well

    as calretinin, indicative of limited endocrine activity [19,24,

    59]. These tumors although positive for FOXL-2 lack the

    FOXL-2 mutation, and this finding may be a helpful diagnos-

    tic adjunct in the differential diagnosis with diffuse type adult

    granulosa cell tumor [70]. They also express SF-1 (adrenal 4-

    binding protein), a nuclear transcription factor that regulates

    genes that are involved in steroidogenesis, development of the

    gonads and adrenal glands, sexual differentiation, reproduc-

    tion, and metabolism. Of interest, this gene is thought to

    regulate the inhibin gene, thus is expressed in cells that are

    also inhibin positive, but this marker has been reported to be

    more sensitive than inhibin in the diagnosis of sex cord-

    stromal tumors and it is more frequently positive in this

    category of tumors than inhibin [71].

    Sclerosing stromal tumor is a morphologically distinctive

    neoplasm with interesting clinical and pathological features

    but it is only briefly mentioned here as it is rarely functioning

    [72]. At first glance, this is surprising as a definitional feature

    of the tumor is a component of lutein cells but they usually

    have a degenerative appearance presumably explaining the

    lack of function. When robust lutein cells with abundant

    Fig. 7 Luteinized thecoma of

    type associated with sclerosing

    peritonitis. In some cases, the

    ovaries are not enlarged but have a

    striking cerebriform contour,

    something also seen

    microscopically (a). Lutein cells

    are seen in this lesion (b). Another

    common feature is stromal edema

    sometimes imparting amicrocysticappearance (c). A

    representative example of the

    sclerosing peritonitis of this entity

    is seen in d

    Fig. 8 Adult granulosa cell tumor. One of many gross appearances is a

    solid mass which may be characterized by multiple discrete yellow

    nodules separated by firm white areas representative of the common

    background stroma of this neoplasm

    106 Endocr Pathol (2014) 25:102119

  • 8/10/2019 10.1007-s12022-013-9285-4

    6/18

    eosinophilic cytoplasm (in contrast to the more common pale

    vacuolated appearance) are present in abundance, as seen forexample during pregnancy (Fig.6), hormone production may

    be striking including even virilization [7375]. These tumors,

    although usually not clinically functioning, express inhibin

    and calretinin [19,24,59]. They are also FOXL-2 positive but

    lack FOXL -2 mutations [63]. The so-called luteinized

    thecoma associated with sclerosing peritonitis (Fig. 7), al-

    though it has lutein cells, rarely is associated with estrogenic

    or androgenic manifestations. However, the lutein cells are

    typically inhibin positive in contrast to the spindle cells which

    show negative or only rarely show focal positive expression of

    this marker [76]. These tumors are also positive for FOXL2

    and SF-1 [70]. Parenthetically, although sclerosing peritonitis

    is typically associated with this distinctive stromal neoplasm,

    it has been described with one granulosa cell tumor [77]. As

    with sclerosing stromal tumor, the rarity of function is likely

    related to the weak nature of the luteinization (Fig.7b). Other

    rare entities in the stromal tumor category such as microcystic

    stromal tumor and signet ring cell stromal tumor have not

    been reported to be endocrinologically active, although the

    possibility exists [57,78]. However, the former often showspositivity for CD10, vimentin, and WT-1 and also has -

    catenin mutations [79] but is negative for inhibin [57].

    Granulosa Cell Tumors

    For approximately the last two decades, granulosa cell tumors

    have been subdivided into two categories following Dr.

    Scullys appreciation circa the late 1960s that in young fe-

    males these tumors often had distinctive microscopic features,

    leading him to designate them juvenile granulosa cell tumor

    [80]. This, of necessity, resulted in the need for a companion

    name for the well-known tumors that peak in the perimeno-

    pausal age group and the term adult granulosa cell tumor

    was introduced. It should be emphasized, however, that these

    designations are terms of convenience to capture a constella-

    tion of findings, and there is overlap between the two tumor

    types, some neoplasms having microscopic features of both

    subtypes. Furthermore, it must be noted that the classic adult

    Fig. 9 Adult granulosa cell

    tumor with bizarre nuclei.

    Characteristic features are seen

    (a), but a significant component

    of this tumor is characterized by

    bizarre nuclear atypia (b), a

    finding not of adverse prognostic

    significance

    Fig. 10 Adult granulosa cell

    tumor. A tumor that has a

    background resembling a cellular

    fibroma is punctuated by

    aggregates of cells with an

    epithelial-like arrangement (a)

    and their granulosa cell nature is

    highlighted by strong staining for

    inhibin (b). A reticulin stain from

    another case (c) shows large areas

    devoid of reticulin, a finding that

    helps substantiate granulosa cell

    differentiation in a tumor with a

    prominent stroma

    Endocr Pathol (2014) 25:102119 107

  • 8/10/2019 10.1007-s12022-013-9285-4

    7/18

    granulosa cell tumor may occur in children and young women[81] and conversely but less often the juvenile form can occur

    in older women [80].

    Adult granulosa cell tumors (AGCT) are often associated

    with estrogenic manifestations with a frequency that varies but

    can broadly be considered seen in half to two thirds of the

    patients. They include postmenopausal bleeding or menstrual

    irregularities in younger women. In children, they, like the

    juvenile form, may cause isosexual pseudoprecocity. If the

    endometrium is evaluated pathologically, it may show endo-

    metrial hyperplasia or even low-grade endometrioid adeno-

    carcinoma. Rarely, AGCT may be androgenic, a finding for

    unknown reasons disproportionally seen with cystic neo-plasms. Sometimes, estrogenic and androgenic manifestations

    can coexist. Cystic AGCTs are also notable grossly as they

    may have smooth cyst linings and suggest other more

    common cystic tumors. The gross spectrum of the AGCT iswide, ranging from cystic, to solid and cystic, to solid (Fig. 8.).

    They may be unilocular or multilocular and not uncommonly

    large, 20 cm or more [82]. Inhibin, mentioned earlier as an

    immunohistochemical adjunct in the identification of lutein or

    endocrine active ovarian stromal cells, can also be used as a

    serum marker to monitor disease course [83].

    Only a few comments will be made on the microscopic

    appearance of the AGCT which is remarkably varied (Figs.9,

    10, and11). The most common is a diffuse growth of round,

    oval, or, less often, spindle-shaped cells that almost always is

    associated with minor foci of obvious epithelial patterns, most

    often in the form of delicate cords. In these tumors, the stromais usually minimal to absent. Tumors in which spindled cells

    dominate have been referred to as sarcomatoid but that

    designation is discouraged as it may cause confusion from

    Fig. 11 Adult granulosa cell

    tumor. This neoplasm, in part, had

    a very typical pattern of regular

    anastomosing cords (a), but in

    other areas had a peculiar clear

    cell morphology (b), potentially

    leading to a broad differential

    diagnosis in the absence of typical

    granulosa cell foci

    Fig. 12 Juvenile granulosa cell

    tumor. Marked nuclear

    pleomorphism (a) is more

    common in this tumor than in the

    adult neoplasm. Note the helpful

    finding of focal follicular

    differentiation. Some juvenile

    granulosa cell tumors grow only

    in the form of large nodularaggregates (b)

    108 Endocr Pathol (2014) 25:102119

  • 8/10/2019 10.1007-s12022-013-9285-4

    8/18

    the management viewpoint. A second common appearance

    has obvious epithelial arrangements, including insular andtrabecular as well as anastomosing cords of granulosa cells,

    on a background of conspicuous but usually minor

    fibrothecomatous stroma. Although follicular patterns are of-

    ten emphasized, they are absent in the majority of tumors and

    are conspicuous in only a minority. The microfollicular pattern

    is characterized by small, generally regular, follicles (Call

    Exner bodies) that may contain eosinophilic material with

    nuclear debris, hyalinized basement membrane-like material,

    or, rarely, basophilic secretion. In our experience, CallExner

    bodies are overall uncommon. The macrofollicular pattern is

    even less common and is composed of large, relatively uni-

    form follicles typically containing eosinophilic secretions.

    Other patterns such as watered-silk(parallel, thin, winding

    cords) and gyriform (a zigzag arrangement of cords) are at

    least in aggregate usually more striking than follicular pat-

    terns. A pseudopapillary pattern has been recently described

    [56]. On gross examination, there is often a friable appearance

    that may suggest a surface epithelial tumor. Under the micro-

    scope, papillae are lined by several layers of typical granu-

    losa cells that often become detached from the surface are

    seen. The resultant appearance may make a papillary tran-

    sitional cell carcinoma, in particular, an initial consider-

    ation in differential diagnosis.

    A third low-power appearance of AGCT may closely re-

    semble either a cellular fibroma or a thecoma due to a prom-

    inent stroma but, when a minority (but >10 %) component of

    the tumor is composed of granulosa cells, the tumor is con-

    sidered a granulosa cell tumor. Sometimes, granulosa cell

    elements in these tumors are best seen at the periphery. Cystic

    AGCTs have cysts that are usually lined by many layers of

    granulosa cells which may show focal follicle formation and

    granulosa cells may be present in the cyst walls [82]. Denu-

    dation of the cyst lining may occasionally be significant and

    cause confusion with other cystic lesions of the ovary. Al-

    though granulosa cells usually have scant cytoplasm, it maybe abundant and eosinophilic, resulting in a luteinized appear-

    ance [84, 85], a feature as noted below more typical of

    juvenile neoplasms. Nuclei of AGCT are typically pale and

    round, oval, or angular and are often haphazardly oriented.

    Nuclear grooves are common but may be relatively inconspic-

    uous especially in tumors with a diffuse pattern or which are

    luteinized; nucleoli are occasionally moderately prominent,

    particularly in the latter. Significant pleomorphism is usually

    absent, but approximately 2 % of AGCTs contain cells with

    large, bizarre, hyperchromatic nuclei (Fig.9b)[86] that have

    no adverse impact on prognosis. The mitotic rate is usually2

    per 10 high-power fields, but higher rates do not exclude this

    diagnosis. A thecomatous or fibromatous stromal compo-

    nent is usually present and may, as noted above, pre-

    dominate and may be richly vascular. Exceptionally, he-

    patocytes and Leydig cells are seen but these findings

    have not been associated with endocrine manifestations

    [87, 88].

    Fig. 13 Juvenile granulosa cell

    tumor, cystic forms. One

    neoplasm (a) shows a cyst with a

    thick lining of neoplastic

    granulosa cells underlain by theca

    cells whereas another cyst has a

    much less conspicuous

    component of lining cells. In

    cystic juvenile granulosa cell

    tumors, as in the adultcounterpart, degenerative changes

    may result in a striking

    pseudopapillary appearance (b)

    Fig. 14 Sertoli cell tumor. This neoplasm has an alveolar arrangement

    with some fibrous septa and occasionally, as in the testis, this can result in

    confusion with germinoma, particularly if lymphocytes are present

    Endocr Pathol (2014) 25:102119 109

  • 8/10/2019 10.1007-s12022-013-9285-4

    9/18

    We restrict comments on the differential diagnosis of

    AGCT to one important tumor, small cell carcinoma of the

    hypercalcemic type, as this tumor, before being characterized,

    was likely misdiagnosed as AGCT because both tumors may

    have follicles and cells with scant cytoplasm. However, there

    are other morphologic features that differ between these two

    tumors including lack of grooves, high mitotic rate, extensive

    necrosis, and extension outside the ovary in approximatelytwo

    third of small cell carcinomas of the hypercalcemic type [89].Furthermore, inhibin is a useful marker in the diagnosis of

    AGCT and is negative in small cell carcinoma of the hyper-

    calcemic type [19,90]. However, of note, some AGCTs may

    be inhibin negative. A recurrent somatic point mutation

    (402CG) in FOXL -2 has been described in almost all

    AGCTs [91], and these tumors as mentioned earlier are

    FOXL-2 positive by immunohistochemistry [63]. They also

    express inhibin, calretinin, WT-1 and SF1 and not infrequently

    CD99 [24,59,71,92,93].

    Although, a known entity now for several decades, the

    juvenile granulosa cell tumor (JGCT) merits some emphasis

    here as it is often functioning and issues in differential diag-

    nosis remain common. The JGCT occurs in the first two

    decades in about 80 % of cases and most of the remainder

    prior to 30 years. The young age distribution results in many

    patients being pre-menarchal and estrogen produced by the

    tumor typically results in a dramatic clinical presentation with

    isosexual pseudoprecocity [80]. Rarely, androgenic manifes-

    tations may occur, an association with cystic tumors again

    being evident [82]. Like AGCT, the tumors are typically

    unilateral and stage I at diagnosis. The only clinical difference

    apparent to date is lack of late recurrences often seen inpatients with AGCT, and a tendency of the rare malignant

    JGCT to recur early [80].

    The spectrum of gross appearances is similar to that

    of the AGCT, and their microscopic spectrum is almost

    as varied (Figs. 12 and 13). Certain specific microscopic

    differences set this neoplasm apart. The first is the more

    immature mitotically active (including atypical forms)

    nature of the cells which typically lack nuclear grooves

    and in up to 15 % of cases are strikingly pleomorphic

    (Fig. 12a) [80]. As more experience accumulated other

    differences became apparent, specifically, abundant cy-

    toplasm, generally eosinophilic, and an irregular follic-ular architecture, follicles being variable in both size

    and shape. Other differences include a nodular architec-

    ture (Fig. 12b), in which some of the nodules occasion-

    ally may show marked sclerosis as well as a basophilic

    background. The variability in the aforementioned fea-

    tures results in a varied differential diagnosis, including

    yolk sac tumor, the coma (uncommon

  • 8/10/2019 10.1007-s12022-013-9285-4

    10/18

  • 8/10/2019 10.1007-s12022-013-9285-4

    11/18

    tumors are subclassified into well, intermediate, and poorly

    differentiated depending largely on the degree of tubular

    (Sertoli) differentiation. Although in general the Sertoli tu-

    bules have a characteristic morphology, they may show some

    variation that may be confusing, particularly when

    endometrioid-like [109]. There are some clinical and gross

    differences related to morphologic variants, perhaps the most

    important being SLCT with a retiform appearance [103] oc-

    curring on average a decade earlier. As these tumors are less

    often androgenic than the other subtypes, and have an appear-

    ance that is often unfamiliar to the pathologist, they are par-

    ticularly apt to be misdiagnosed. These tumors often exhibit

    polypoid grape-like excrescences or have a soft spongy

    consistency [110]. They are typically composed microscopi-

    cally of slit-like (retiform) branching tubules into which pro-

    trude cellular papillae or edematous fronds that may mimic to

    a striking degree a serous papillary tumor. Another category

    likely to cause confusion is SLCT with heterologous elements

    which include most commonly intestinal-type epithelium

    (Fig.15a, b) that may or not be associated with carcinoid

    and less often rhabdomyosarcoma, chondrosarcoma, or

    both. Heterologous mesenchymal elements tend to be seen

    in poorly differentiated tumors while endodermal differentia-

    tion occurs most frequently in SLCT of intermediate differen-

    tiation [103,110117].

    Given the young age of many patients with SLCTs, and for

    that matter some with either form of GCT, it is obvious that an

    occasional tumor will be discovered in a patient who is preg-

    nant. In this situation, the tumors may be particularly difficult

    to diagnose due to changes in their morphology apparently

    due to pregnancy. Most challenging is the tendency to show

    abundant intercellular edema as well as increased number of

    Leydig cells or lutein cells as the case may be [118]. The

    edema effaces the typical architecture of these tumors and

    additionally imparts a loose appearance reminiscent in some

    cases of a reticular pattern of a yolk sac tumor. Further

    confusion may be caused when, as is occasional, the SLCT

    is associated with elevation of serum -fetoprotein levels

    [116, 119125]. However, elevations are rarely as high as

    seen with yolk sac tumor and judicious sampling and aware-

    ness of this pitfall should enable correct diagnosis. As with

    JGCT, the morphologic diversity of SLCT results in a broad

    differential diagnosis ranging from endometrioid carcinoma to

    yolk sac tumor to malignant mixed mesodermal tumor (when

    mesenchymal heterologous elements present) to teratoma

    (when endodermal heterologous elements present).

    Inhibin may be used to confirm the diagnosis of Sertoli cell

    tumor or SLCT, being particularly helpful in the former in our

    experience. However, it is important to note that staining for

    inhibin is always less extensive and lighter in Sertoli cells

    when compared to Leydig cells. It is also important to remem-

    ber that Sertoli cells can be positive for keratins and rarely for

    EMA, thus a panel of antibodies that include inhibin and

    EMA, is helpful. Other markers that can be used include

    Table 4 Miscellaneous other neoplasms with endocrine function, i.e.,

    non sex-cord-stromal tumors with endocrine activity unrelated to stromal

    luteinization

    1. Steroid cell tumors (includes Leydig cell tumor) (usually androgenic)

    2. Struma ovarii (thyroid hyperfunction)

    3. Carcinoid tumors (carcinoid syndrome )

    4. Mucinous tumors (ZollingerEllison syndrome)

    5. Steroid cell tumorspituitary adenoma in dermoid(Cushings syndrome)

    6. Pituitary adenoma in dermoid (hyperprolactinemia)

    Table 5 Categories of ovarian tumors with functioning stroma

    1. Tumors with syncytiotrophoblast cells

    2. During pregnancy

    3. Idiopathic

    Primary and metastatic mucinous tumors

    Miscellaneous other tumors

    Rete cystadenoma and monodermal teratoma

    (luteinization usually peripheral)

    Fig. 18 Dysgerminoma with syncytiotrophoblast giant cells. Although

    not seen in this illustration, the giant cells frequently induce luteinization

    of the stroma which may lead to endocrine manifestations

    Fig. 19 Mucinous cystadenoma with stromal luteinization. Mucinous

    tumors are associated with this phenomenon more often than other

    primary ovarian neoplasms

    112 Endocr Pathol (2014) 25:102119

  • 8/10/2019 10.1007-s12022-013-9285-4

    12/18

    calretinin (positivity not related to hormonal function), WT-1,

    and SF1. The latter seems to be a very specific marker of sex

    cord differentiation [19,24,31,59,71,9499]. Approximate-

    ly 50 % of SertoliLeydig cell tumors have been recently

    reported to express FOXL2, and only rarely are associatedwithFOXL2 mutations [63,126].

    Sex Cord Tumor with Annular Tubules

    This is an unusual tumor (Fig. 16a) that may be associated

    with PeutzJeghers syndrome [127] in which instance it is

    typically bilateral, multiple, small, and non-functioning.

    When unassociated with the syndrome, they are typically

    unilateral and grossly visible, and 40 % are associated with

    some evidence of estrogen production [128] and occasional

    tumors have produced progesterone [129]. The latter is a rare

    manifestation of ovarian tumors overall and sex cord tumor

    with annular tubules (SCTAT) is the neoplasm most common-

    ly associated with it [130]. The non-PeutzJeghers associated

    tumors may be grossly cystic, and this may also be noted at the

    microscopic level (Fig.16b).

    Miscellaneous Other Neoplasms with Endocrine Function

    Steroid Cell Tumor

    This category has traditionally included Leydig cell tumor,

    stromal luteoma, and steroid cell tumor, not otherwise speci-

    fied (NOS). As noted earlier, the upcoming WHO classifica-

    tion no longer recognizes stromal luteoma as a separate entity

    [28]. Nonetheless, it is of note that in a series on stromal

    luteomas, the tumors were most often associated with estro-

    genic manifestations in postmenopausal patients [131] where-

    as other steroid cell tumors, when functioning, are more often

    androgenic, often occurring in younger patients.

    Leydig cell tumors are typically androgenic although usu-

    ally not to the degree present in SLCTs and not with such adramatic rapid onset. They usually occur in postmenopausal

    patients (mean age 63 years) and they are typically small

    (Fig. 17a) and located in the hilus. Although technically

    crystals of Reinke should ideally be seen, the diagnosis can

    be made in their absence when a variety of other morphologic

    features are seen including clustering of tumor cells and

    fibrinoid necrosis of vessel walls (Fig.17b)[132].

    Steroid cell tumors, NOS, occur in younger patients on

    average (mean age 43 years), compared to Leydig cell tumors

    and are also often androgenic, sometimes causing virilization

    [131]. These tumors like Leydig cell tumors are almost always

    unilateral but are typically larger. In contrast to Leydig cell

    tumors which usually have uniform eosinophilic cytoplasm,

    steroid cell tumors, NOS, usually have, at least in part, pale

    and vacuolated (lipid-rich) cytoplasm. It is also in this group

    Fig. 20 Krukenberg tumor. The tubular pattern and many intervening

    luteinized stromal cells may cause confusion with SertoliLeydig cell

    tumor

    Fig. 21 Ovarian hemangioma.

    Numerous small vessels are

    evident but their close apposition

    and additional content of

    numerous luteinized stromal cells

    may result in diagnostic difficulty

    (a). A CD31 stain highlights

    the vascular nature of the

    neoplasm (b)

    Endocr Pathol (2014) 25:102119 113

  • 8/10/2019 10.1007-s12022-013-9285-4

    13/18

    that malignant features may be seen and rare tumors have been

    associated with Cushings syndrome.

    These tumors in general are inhibin, calretinin, SF1, and

    CD99 positive, and frequently express Melan A and androgen

    receptor, and rarely may express AE 1/3 immunopositivity

    [19,59,71,133136]. FOXL2 has only been evaluated in a

    one study, and was not expressed in Leydig cell tumors or

    stromal luteomas, but was expressed in a single steroid celltumor NOS [63].

    Miscellaneous Other Neoplasms with Endocrine Function

    These are listed in Table 4 and are not considered here because

    of their overall rarity and lack of new information since

    reviewed by Dr. Scully; the reader is referred to that compre-

    hensive coverage and other reports [2,137].

    Ovarian Tumors with Functioning Stroma

    This is a much more common phenomenon than the rare,

    albeit interesting ones listed in Table 4 and merits more

    comment. It is also a phenomenon Dr. Scully first highlighted

    and furthermore can cause clinical confusion in as much as

    endocrine symptoms, either androgenic or estrogenic, due to

    stromal luteinization in diverse tumors, may suggest the pres-

    ence of an endocrine type tumor (sex cord-stromal or steroid

    cell tumor) when the responsible neoplasm is in fact in a

    different category. Tumors in this category can be considered

    in three groups (Table 5). In the first two, it is thought thatstromal luteinization is secondary to HCG stimulation, either

    due to syncytiotrophoblast cells, most often within a germ cell

    tumor (Fig.18) but occasionally with other neoplasms includ-

    ing carcinomas [138] or pregnancy. The greater number is

    however in the idiopathic group and the mechanism is obscure

    but HCG may again play a role. In a study of 100 ovarian

    tumors, HCG positivity was more frequently found in those

    with luteinized cells than in those with an inactive stroma [139].

    However, HCH-like substances as well as other factors may

    play a role in stromal activation. Among primary epithelial

    tumors, there is a particular association with mucinous tumors

    (Fig.19) and interestingly this association with mucinous epi-

    thelium also exists with metastatic tumors, as colorectal carci-

    nomas in the ovary and Krukenberg tumors (Fig. 20) frequently

    show luteinization [140]. Rarely, unusual primary ovarian tu-

    mors may be associated with luteinization, and this enhances

    diagnostic difficulty (Fig. 21). In the majority of cases, the

    lutein cells are randomly seen in the stroma but in a subset they

    are distributed peripherally. For unknown reasons, a dispropor-

    tionate number of these tumors have been monodermal terato-

    mas, particularly struma ovarii (Fig. 22). In the one series

    reported, 42 % of patients had androgenic manifestations, while

    29 % had evidence of estrogenic and 4 % progestational

    manifestations [10]. The second most common cause is related

    to rete cysts [141]. In some of these cases, the hormone-

    producing cells had crystals of Reinke, indicating a Leydig cell

    nature and not surprisingly were more often associated with

    androgenic manifestations than typically lutein cells [10].

    Paraendocrine Disorders

    In the final section of this review, we briefly note a few of the

    uncommon but fascinating tumors of different types that may

    be associated with hormone production of various types, the

    reason in most being unknown.

    Fig. 22 Struma ovarii. This neoplasm is associated with a thick band of

    lutein cells at its periphery

    Fig. 23 Small cell carcinoma of hypercalcemic type. Typical small cells

    with scant cytoplasm surround a follicle, the latter a helpful diagnostic

    feature in many cases

    Table 6 Paraendocrine disorders

    1. Hypercalcemia

    2. ACTH production and Cushings

    syndrome

    3. hCG production

    4. Hypoglycemia

    5. Renin production

    114 Endocr Pathol (2014) 25:102119

  • 8/10/2019 10.1007-s12022-013-9285-4

    14/18

    We first consider hypercalcemia because among the tumors

    responsible for it is the intriguing small cell carcinoma of the

    hypercalcemic type (Fig. 23). Indeed, it is comfortably the

    ovarian tumor that exhibits this phenomenon most often [142]

    and furthermore represents one of Dr. Scullys most astute

    original observations. He gradually became aware of the

    entity when he accrued cases of undifferentiated carcinoma

    in young women, itself uncommon, and was further struck bythe fact the carcinomas had a small cell morphology contrast-

    ing with the typical large cell morphology of undifferentiated

    carcinoma in general. When the first 11 cases all had hyper-

    calcemia clearly related to the neoplasm, he knew he had

    come across a distinctive tumor. In retrospect, designating this

    tumor small cell carcinoma is not ideal as the name may cause

    confusion with the better known small cell carcinoma of

    pulmonary type, an unrelated neoplasm. Additionally, as ex-

    perience with the small cell carcinoma of the hypercalcemic

    type has expanded, many tumors have been encountered in

    which the cells are large with abundant eosinophilic cyto-

    plasm [89]. This is the ovarian tumor most often associatedwith paraendocrine hypercalcemia, especially in young wom-

    en. The underlying mechanism of the hypercalcemia in these

    tumors is not well understood. Elevated serum calcium levels

    are typically associated with normal parathormone (PTH)

    serum levels and phosphate levels and occur in the absence

    of bone metastases. In general, tumors are PTH negative

    except in the series reported by Aguirre and colleagues where

    one patient with elevated serum levels had scattered PTH-

    positive cells in the tumor and in the series reported by Abeler

    and colleagues where two patients had elevated PTH serum

    levels and three of five tumors had PTH-positive cells [143,

    144]. Parathormone-related peptide (PTHrp) is a hormone

    closely related to PTH with autocrine and paracrine functions,

    which binds to PTH receptors in bone and kidney. The gene

    that encodes PTHrp is on the short arm of chromosome 12, in

    a position homologous to the location of the PTH gene on

    chromosome 11. As this substance has been isolated from

    malignant tumors associated with hypercalcemia, it has been

    postulated as a source of the hypercalcemia in these cases;

    however, no genetic alterations have been reported on the

    short arm of chromosome 11 [145]. Several authors found

    PTHrp staining in tumor cells in 8 O-SCCHTs [146148].

    However, the presence or absence of PTHrp staining in tu-

    mors does not correlate with the patients serum calcium levels

    [148]. The highly malignant nature of the tumor, apparent

    from the outset, sadly remains with little or no progress from

    the therapeutic point of view. The hypercalcemia itself only

    rarely causes clinical manifestations. The only other ovarian

    tumor associated with hypercalcemia with any frequency is

    clear cell carcinoma, the reason again being unknown.

    ACTH production has been seen with assorted ovarian

    tumors rarely, no symptoms often resulting. Dramatic cases

    of Cushings syndrome have been seen, mostly with steroid

    cell tumors, but also with a few pituitary adenomas within a

    dermoid cyst [2,149] and one carcinoid tumor [150]. Other

    paraendocrine manifestations that have been described are

    hypoglycemia and renin production (Table 6). The former

    has been associated with diverse lesions whereas renin pro-

    duction is most typically seen with Sertoli cell tumors [151].

    Concluding Remarks

    Although we may be biased due to our own interests, the

    already numerous fascinating aspects of ovarian tumors relat-

    ed to their morphology are only enhanced by the additional

    pre sen ce in a mea sur able number of cas es of var iou s

    endocrine/paraendocrine abnormalities which can make indi-

    vidual cases as, or more, clinically interesting than they are

    pathologically. This brief review presents an update on this

    topic. Our knowledge of the area is largely based on what was

    taught us over the years by our mentor Dr. Robert E Scullywhose teachings remain with us everyday and whose contri-

    butions to ovarian pathology are unlikely to be surpassed.

    References

    1. Morris JM, Scully RE: Endocrine Pathology of the Ovary, St. Louis:

    C. V. Mosby Co., 1958.

    2. Scully RE, Young RH, Clement PB: Tumors of the Ovary,

    Maldeveloped Gonads, Fallopian Tube, and Broad Ligament.

    Washington: Armed Forces Institute of Pathology, 1996.

    3. Scully RE, Morris JM. Functioning ovarian tumors. In: Meigs JV,

    Surgis S, eds. Progress in Gynecology. New York: Grune and

    Stratton, 1957; 20-34.

    4. Scully RE. Gonadoblastoma; a gonadal tumor related to the

    dysgerminoma (seminoma) and capable of sex-hormone produc-

    tion. Cancer 6: 455-463, 1953.

    5. Scully RE. Gonadoblastoma. A review of 74 cases. Cancer 25:

    1340-1356, 1970.

    6. Scully RE, Richardson GS. Luteinization of the stroma of metastatic

    cancer involving the ovary and its endocrine significance. Cancer

    14: 827-840, 1961.

    7. Benedict PH, Cohen RB, Cope O, Scully RE. Ovarian and adrenal

    morphology in cases of hirsutism or virilism and SteinLeventhal

    syndrome. Fertil Steril 13: 380-395, 1962.8. Boss JH, Scully RE, Wegner KH, Cohen RB. Structural variations

    in the adult ovary. Clinical significance. Obstet Gynecol 25: 747-

    764, 1965.

    9. Herrington JB, Scully RE. Endocrine aspects of germ cell tumors.

    In: Damjanov I, Knowles DM, Solter D, eds. The Human

    Teratomas. Clifton, NJ: Humana, 1983; 215-229.

    10. Rutgers JL, Scully RE. Functioning ovarian tumors with peripheral

    steroid cell proliferation:a report of twenty-four cases. Int J Gynecol

    Pathol 5: 319-337, 1986.

    11. Scully RE. Androgenic lesions of the ovary. In: Grady HG, Smith

    OE (eds) The Ovary. Baltimore: Williams & Wilkins Co., 1962;

    143-174.

    Endocr Pathol (2014) 25:102119 115

  • 8/10/2019 10.1007-s12022-013-9285-4

    15/18

    12. Scully RE. Germ cell tumors of the ovary and fallopian tube. In:

    Meigs JV, Surgis S, eds. Progress in Gynecology. New York: Grune

    & Stratton, 1963; 335-347.

    13. Scully RE. Ovarian tumors with endocrine manifestations. In:

    DeGroot LJ, ed. Metabolic Basis of Endocrinology. Philadelphia:

    W. B. Saunders Co., 1989; 1994-2008.

    14. Scully RE. Stromal Luteoma of the Ovary. A Distinctive Type of

    Lipoid-Cell Tumor. Cancer 17: 769-778, 1964.

    15. Scully RE, Cohen RB. Oxidative-Enzyme Activity in Normal and

    Pathologic Human Ovaries. Obstet Gynecol 24: 667-681, 1964.16. Scully RE. Ovarian tumors with functioning stroma. In: Fox H,

    Wells M, eds. Haines and Taylors Gynaecological and Obstetrical

    Pathology. 3rd ed. Edinburgh: Churchill Livingstone, 1987; 724-

    736.

    17. Aguirre P, Scully RE, Wolfe HJ, DeLellis RA. Argyrophil cells in

    Brenner tumors: histochemical and immunohistochemical analysis.

    Int J Gynecol Pathol 5: 223-234, 1986.

    18. Aguirre P, Thor AD, Scully RE. Ovarian endometrioid carcinomas

    resembling sex cord-stromal tumors. An immunohistochemical

    study. Int J Gynecol Pathol 8: 364-373, 1989.

    19. Kommoss F, Oliva E, Bhan AK, Young RH, Scully RE. Inhibin

    expression in ovarian tumors and tumor-like lesions: an immuno-

    histochemical study. Mod Pathol 11: 656-664, 1998.

    20. Scully RE, Aguirre P, DeLellis RA. Argyrophilia, serotonin, and

    peptide hormones in the female genital tract and its tumors. Int J

    Gynecol Pathol 3: 51-70, 1984.

    21. Scully RE. Immunohistochemistry of ovarian tumors. In: Russo J,

    Russo I, eds. Immunocytochemistry in Tumor Diagnosis. Boston:

    Martinus Nijhoff, 1985; 293-320.

    22. Stewart CJ, Jeffers MD, Kennedy A. Diagnostic value of inhibin

    immunoreactivity in ovarian gonadal stromal tumours and their

    histological mimics. Histopathology 31: 67-74, 1997.

    23. Lugli A, Forster Y, Haas P, et al. . Calretinin expression in human

    normal and neoplastic tissues: a tissue microarray analysis on 5233

    tissue samples. Hum Pathol 34: 994-1000, 2003.

    24. McCluggage WG, Maxwell P. Immunohistochemical staining for

    calretinin is useful in the diagnosis of ovarian sex cord-stromal

    tumours. Histopathology 38: 403-408, 2001.

    25. Krug E, Berga SL. Postmenopausal hyperthecosis: functional dys-

    regulation of androgenesis in climacteric ovary. Obstet Gynecol 99:

    893-897, 2002.

    26. Sasano H, Fukunaga M, Rojas M, Silverberg SG. Hyperthecosis of

    the ovary. Clinicopathologic study of 19 cases with immunohisto-

    chemical analysis of steroidogenic enzymes. Int J Gynecol Pathol 8:

    311-320, 1989.

    27. Vollaard ES, van Beek AP, Verburg FA, Roos A, Land JA.

    Gonadotropin-releasing hormone agonist treatment in postmeno-

    pausal women with hyperandrogenism of ovarian origin. J Clin

    Endocrinol Metab 96: 1197-1201, 2011.

    28. Kurman RJ, Carcangiu ML, Herrington CS, Young RH: WHO

    Classification of Tumours of Female Reproductive Organs. Lyon:

    International Agency for Research on Cancer, 2013 (in press).

    29. Meldrum DR, Frumar AM, Shamonki IM, Benirschke K, Judd HL.

    Ovarian and adrenal steroidogenesis in a virilized patient withgonadotropin-resistant ovaries and hilus cell hyperplasia. Obstet

    Gynecol 56: 216-221, 1980.

    30. Hofland M, Cosyns S, De Sutter P, Bourgain C, Velkeniers

    B. Leydig cell hyperplasia and Leydig cell tumour in post-

    menopausal women: report of two cases. Gynecol Endocrinol

    29: 213-215, 2013.

    31. Cao QJ, Jones JG, Li M. Expression of calretinin in human ovary,

    testis, and ovarian sex cord-stromal tumors. Int J Gynecol Pathol 20:

    346-352, 2001.

    32. Bamberger AM, Ivell R, Balvers M, et al. . Relaxin-like factor

    (RLF): a new specific marker for Leydig cells in the ovary. Int J

    Gynecol Pathol 18: 163-168, 1999.

    33. Young RH, Scully RE. Fibromatosis and massive edema of the

    ovary, possibly related entities: a report of 14 cases of fibromatosis

    and 11 cases of massive edema. Int J Gynecol Pathol 3: 153-178,

    1984.

    34. Yamoto M, Minami S, Nakano R. Immunohistochemical localiza-

    tion of inhibin subunits in polycystic ovary. J Clin Endocrinol

    Metab 77: 859-862, 1993.

    35. Duleba AJ, Pehlivan T, Carbone R, Spaczynski RZ. Activin stimu-

    lates proliferation of rat ovarian thecalinterstitial cells. Biol Reprod

    65: 704-709, 2001.36. Kosloske AM, Goldthorn JF, Kaufman E, Hayek A. Treatment of

    precocious pseudopuberty associated with follicular cysts of the

    ovary. Am J Dis Child 138: 147-149, 1984.

    37. Scully RE, Mark EJ, McNeely WF, McNeely BU. Case Records of

    the Massachusetts General Hospital, Case 47-1989. N Engl J Med

    321: 1463-1471, 1989.

    38. McCluggage WG, Patterson A, White J, Anderson NH.

    Immunocytochemical staining of ovarian cyst aspirates with mono-

    clonal antibody against inhibin. Cytopathology 9: 336-342, 1998.

    39. Scully RE, McNeely BU. Case Records of the Massachusetts

    General Hospital, Case 4-1975. N Engl J Med 202: 199-203, 1975.

    40. Lumbroso S, Paris F, Sultan C. Activating Gsalpha mutations:

    analysis of 113 patients with signs of McCuneAlbright syn-

    dromea European Collaborative Study. J Clin Endocrinol Metab

    89: 2107-2113, 2004.

    41. de Sanctis C, Lala R, MatarazzoP,Andreo M, de Sanctis L. Pubertal

    development in patients with McCuneAlbright syndrome or

    pseudohypoparathyroidism. J Pediatr Endocrinol Metab 16 Suppl

    2: 293-296, 2003.

    42. Clement PB, Scully RE. Large solitary luteinized follicle cyst of

    pregnancy and puerperium: A clinicopathological analysis of eight

    cases. Am J Surg Pathol 4: 431-438, 1980.

    43. Fang YM, Gomes J, Lysikiewicz A, Maulik D. Massive luteinized

    follicular cyst of pregnancy. Obstet Gynecol 105: 1218-1221, 2005.

    44. Wang XY, Vinta MK,Myers S, FanF. Solitary luteinized follicle cyst

    of pregnancy and puerperium. Pathol Res Pract 202: 471-473, 2006.

    45. Angioni S, Portoghese E, Milano F, Melis GB, Fulghesu AM.

    Hirsutism and hyperandrogenism associated with hyperreactio

    luteinalis in a singleton pregnancy: a case report. Gynecol

    Endocrinol 23: 248-251, 2007.

    46. Page SM, OBrien J. Hyperreactio luteinalis with clitoromegaly in a

    twin pregnancy. Rev Obstet Gynecol 2: 3-4, 2009.

    47. Masarie K, Katz V, Balderston K. Pregnancy luteomas: clinical

    presentations and management strategies. Obstet Gynecol Surv

    65: 575-582, 2010.

    48. Mazza V, Di Monte I, Ceccarelli PL, et al. . Prenatal diagnosis of

    female pseudohermaphroditism associated with bilateral luteoma of

    pregnancy: case report. Hum Reprod 17: 821-824, 2002.

    49. Spitzer RF, Wherrett D, Chitayat D, et al. . Maternal luteoma of

    pregnancy presenting with virilization of the female infant. J Obstet

    Gynaecol Can 29: 835-840, 2007.

    50. Wang YC, Su HY, Liu JY, Chang FW, Chen CH. Maternal and

    female fetal virilization caused by pregnancy luteomas. Fertil Steril

    84: 509, 2005.51. Clement PB. Tumor-like lesions of the ovary associated with preg-

    nancy. Int J Gynecol Pathol 12: 108-115, 1993.

    52. Norris HJ, Taylor HB. Nodular theca-lutein hyperplasia of pregnan-

    cy (so-called pregnancy luteoma). A clinical and pathologic study

    of 15 cases. Am J Clin Pathol 47: 557-566, 1967.

    53. Burandt E, Young RH. Pregnancy Luteoma: A study of 20 cases on

    the occasion of the 50th anniversary of its description by Dr.

    William H. Sternberg, with an emphasis on the common presence

    of follicle-like spaces and their diagnostic implications. Am J Surg

    Pathol, 2013 (in press).

    54. Bjorkholm E, Silfversward C. Theca-cell tumors. Clinical features

    and prognosis. Acta Radiol Oncol 19: 241-244, 1980.

    116 Endocr Pathol (2014) 25:102119

  • 8/10/2019 10.1007-s12022-013-9285-4

    16/18

    55. Roth LM, Czernobilsky B. Perspectives on pure ovarian stromal

    neoplasms and tumor-like proliferations of the ovarian stroma. Am J

    Surg Pathol 35: e15-33, 2011.

    56. Irving JA, Young RH. Granulosa cell tumors of the ovary with a

    pseudopapillary pattern: a study of 14 cases of an unusual morpho-

    logic variant emphasizing their distinction from transitional cell

    neoplasms and other papillary ovarian tumors. Am J Surg Pathol

    32: 581-586, 2008.

    57. Irving JA, Young RH. Microcystic stromal tumor of the ovary:

    report of 16 cases of a hitherto uncharacterized distinctive ovarianneoplasm. Am J Surg Pathol 33: 367-375, 2009.

    58. Oliva E, Egger JF, Young RH. Primary endometrioid stromal sarco-

    ma of the ovary: A clinicopathological study of 27 cases with

    morphologic and behavioral features similar to those of uterine

    low-grade endometrial stromal sarcoma. Am J Surg Pathol, 2013

    (in press).

    59. Movahedi-LankaraniS, Kurman RJ. Calretinin, a more sensitive but

    less specific marker than alpha-inhibin for ovarian sex cord-stromal

    neoplasms: an immunohistochemical study of 215 cases. Am J Surg

    Pathol 26: 1477-1483, 2002.

    60. Johnson AL, Tilly JL, Levorse JM. Possible role for arachidonic

    acid in the control of steroidogenesis in hen theca. Biol Reprod 44:

    338-344, 1991.

    61. Kawasaki H, Kretsinger RH. Calcium-binding proteins. 1: EF-

    hands. Protein Profile 1: 343-517, 1994.

    62. Zhang M, Kao CS, Ulbright TM, Epstein JI. Testicular

    fibrothecoma: a morphologic and immunohistochemical study of

    16 cases. Am J Surg Pathol 37: 1208-1214, 2013.

    63. Al-Agha OM, Huwait HF, Chow C, et al.. FOXL2 is a sensitive and

    specific marker for sex cord-stromal tumors of the ovary. Am J Surg

    Pathol 35: 484-494, 2011.

    64. Shah SP, Kobel M, Senz J, et al. Mutation of FOXL2 in granulosa-

    cell tumors of the ovary. N Engl J Med 360: 2719-2729, 2009.

    65. Micci F, Haugom L, Abeler VM, TropeCG, Danielsen HE, Heim S.

    Consistent numerical chromosome aberrations in thecofibromas of

    the ovary. Virchows Arch 452: 269-276, 2008.

    66. Roth LM, Sternberg WH. Partly luteinized theca cell tumor of the

    ovary. Cancer 51: 1697-1704, 1983.

    67. Zhang J, Young RH, Arseneau J, Scully RE. Ovarian stromal

    tumors containing lutein or Leydig cells (luteinized thecomas and

    stromal Leydig cell tumors)a clinicopathological analysis of fifty

    cases. Int J Gynecol Pathol 1: 270-285, 1982.

    68. Scully RE, Galdabini JJ, McNeely BU. Case Records of the

    Massachusetts General Hospital, Case 10-1980. N Engl J Med

    302: 621-626, 1980.

    69. Sternberg WH, Roth LM. Ovarian stromal tumors containing Leydig

    cells. I. StromalLeydig cell tumor and non-neoplastic transforma-

    tion of ovarian stroma to Leydig cells. Cancer 32: 940-951, 1973.

    70. McCluggage WG, Singh N, Kommoss S, Huntsman DG, Gilks CB.

    Ovarian Cellular Fibromas Lack FOXL2 Mutations: A Useful

    Diagnostic Adjunct in the Distinction From Diffuse Adult

    Granulosa Cell Tumor. Am J Surg Pathol 37: 1450-1455, 2013.

    71. Zhao C, Vinh TN, McManus K, Dabbs D, Barner R, Vang R.

    Identification of the most sensitive and robust immunohistochemi-cal markers in different categories of ovarian sex cord-stromal

    tumors. Am J Surg Pathol 33: 354-366, 2009.

    72. Chalvardjian A, Scully RE. Sclerosing stromal tumors of the ovary.

    Cancer 31: 664-670, 1973.

    73. Cashell AW, Cohen ML. Masculinizing sclerosing stromal tumor of

    the ovary during pregnancy. Gynecol Oncol 43: 281-285, 1991.

    74. Duska LR, Flynn C, Goodman A. Masculinizing sclerosing stromal

    cell tumor in pregnancy: report of a case and review of the literature.

    Eur J Gynaecol Oncol 19: 441-443, 1998.

    75. Quinn MA, Oster AO, Fortune D, Hudson B. Sclerosing stromal

    tumour of the ovary case report with endocrine studies. Br J Obstet

    Gynaecol 88: 555-558, 1981.

    76. Staats PN, McCluggage WG, Clement PB, Young RH. Luteinized

    thecomas (thecomatosis) of the type typically associated with scle-

    rosing peritonitis: a clinical, histopathologic, and immunohistochem-

    ical analysis of 27 cases. Am J Surg Pathol 32: 1273-1290, 2008.

    77. Walker J, Moss EL, Ganesan R, Hirschowitz L. Sclerosing perito-

    nitis associated with a luteinized adult granulosa cell tumor. Int J

    Gynecol Pathol 31: 141-144, 2012.

    78. Vang R, Bague S, Tavassoli FA, Prat J. Signet-ring stromaltumor of

    the ovary: clinicopathologic analysis and comparison with

    Krukenberg tumor. Int J Gynecol Pathol 23: 45-51, 2004.79. Maeda D, Shibahara J, Sakuma T, et al. Beta-catenin (CTNNB1)

    S33C mutation in ovarian microcystic stromal tumors. Am J Surg

    Pathol 35: 1429-1440, 2011.

    80. Young RH, Dickersin GR, Scully RE. Juvenile granulosa cell tumor

    of the ovary. A clinicopathological analysis of 125 cases. Am J Surg

    Pathol 8: 575-596, 1984.

    81. ZaloudekC, Norris HJ. Granulosa tumors of the ovary in children: a

    clinical and pathologic study of 32 cases. Am J Surg Pathol 6: 503-

    512, 1982.

    82. Nakashima N, Young RH, Scully RE. Androgenic granulosa cell

    tumors of the ovary. A clinicopathologic analysis of 17 cases and

    review of the literature. Arch Pathol Lab Med 108: 786-791, 1984.

    83. Geerts I, Vergote I, Neven P, Billen J. The role of inhibins B and

    antimullerianhormonefor diagnosis and follow-up of granulosa cell

    tumors. Int J Gynecol Cancer 19: 847-855, 2009.

    84. Ganesan R, Hirschowitz L, Baltrusaityte I, McCluggage WG.

    Luteinized adult granulosa cell tumora series of 9 cases: revisiting

    a rare variant of adult granulosa cell tumor. Int J Gynecol Pathol 30:

    452-459, 2011.

    85. Young RH, Oliva E, Scully RE. Luteinized adult granulosa cell

    tumors of the ovary: a report of four cases. Int J Gynecol Pathol 13:

    302-310, 1994.

    86. Young RH, Scully RE. Ovarian sex cord-stromal tumors with

    bizarre nuclei: a clinicopathologic analysis of 17 cases. Int J

    Gynecol Pathol 1: 325-335, 1983.

    87. Ahmed E, Young RH, Scully RE. Adult granulosa cell tumor of the

    ovary with foci of hepatic cell differentiation: a report of four cases

    and comparison with two cases of granulosa cell tumor with Leydig

    cells. Am J Surg Pathol 23: 1089-1093, 1999.

    88. Nogales FF, Concha A, Plata C, Ruiz-Avila I. Granulosa cell tumor

    of the ovary with diffuse true hepatic differentiation simulating

    stromal luteinization. Am J Surg Pathol 17: 85-90, 1993.

    89. Young RH, Oliva E, Scully RE. Small cell carcinoma of the ovary,

    hypercalcemic type. A clinicopathological analysis of 150 cases.

    Am J Surg Pathol 18: 1102-1116, 1994.

    90. McCluggage WG, Oliva E, Connolly LE, McBride HA, Young RH.

    An immunohistochemical analysis of ovarian small cell carcinoma

    of hypercalcemic type. Int J Gynecol Pathol 23: 330-336, 2004.

    91. Kobel M, Gilks CB, Huntsman DG. Adult-type granulosa cell

    tumors and FOXL2 mutation. Cancer Res 69: 9160-9162, 2009.

    92. Kommoss S, Anglesio MS, Mackenzie R, et al. FOXL2 molecular

    testing in ovarian neoplasms: diagnostic approach and procedural

    guidelines. Mod Pathol 26: 860-867, 2013.

    93. Rabban JT, Zaloudek CJ. A practical approach to immunohisto-chemical diagnosis of ovarian germ cell tumours and sex cord-

    stromal tumours. Histopathology 62: 71-88, 2013.

    94. Cathro HP, Stoler MH. The utility of calretinin, inhibin, and WT1

    immunohistochemical staining in the differential diagnosis of ovar-

    ian tumors. Hum Pathol 36: 195-201, 2005.

    95. Costa MJ, Ames PF, Walls J, Roth LM. Inhibin immunohistochem-

    istry applied to ovarian neoplasms: a novel, effective, diagnostic

    tool. Hum Pathol 28: 1247-1254, 1997.

    96. Deavers MT, Malpica A, Liu J, Broaddus R, Silva EG. Ovarian

    sex cord-stromal tumors: an immunohistochemical study includ-

    ing a comparison of calretinin and inhibin. Mod Pathol 16: 584-

    590, 2003.

    Endocr Pathol (2014) 25:102119 117

  • 8/10/2019 10.1007-s12022-013-9285-4

    17/18

    97. Matias-Guiu X, Pons C, Prat J. Mullerian inhibiting substance,

    alpha-inhibin, and CD99 expression in sex cord-stromal tumors

    and endometrioid ovarian carcinomas resembling sex cord-stromal

    tumors. Hum Pathol 29: 840-845, 1998.

    98. Riopel MA, Perlman EJ, Seidman JD, Kurman RJ, Sherman ME.

    Inhibin and epithelial membrane antigen immunohistochemistry

    assist in the diagnosis of sex cord-stromal tumors and provide clues

    to the histogenesis of hypercalcemic small cell carcinomas. Int J

    Gynecol Pathol 17: 46-53, 1998.

    99. Shah VI, Freites ON, Maxwell P, McCluggage WG. Inhibin is morespecific than calretinin as an immunohistochemical marker for

    differentiating sarcomatoid granulosa cell tumour of the ovary from

    other spindle cell neoplasms. J Clin Pathol 56: 221-224, 2003.

    100. Oliva E, Alvarez T, Young RH. Sertoli cell tumors of the ovary: a

    clinicopathologic and immunohistochemical study of 54 cases. Am

    J Surg Pathol 29: 143-156, 2005.

    101. Tavassoli FA, Norris HJ. Sertoli tumors of the ovary. A clinicopath-

    ologic study of 28 cases with ultrastructural observations. Cancer

    46: 2281-2297, 1980.

    102. Tracy SL, Askin FB, Reddick RL, Jackson B, Kurman RJ.

    Progesterone secreting Sertoli cell tumor of the ovary. Gynecol

    Oncol 22: 85-96, 1985.

    103. Young RH, Scully RE. Ovarian SertoliLeydig cell tumors. A

    clinicopathological analysis of 207 cases. Am J Surg Pathol 9:

    543-569, 1985.

    104. Gui T, Cao D, Shen K, et al. A clinicopathological analysis of 40

    cases of ovarian SertoliLeydig cell tumors. Gynecol Oncol 127:

    384-389, 2012.

    105. Hansen TP, Sorensen B. SertoliLeydig cell tumourof the ovarya

    rare cause of virilization after menopause. APMIS 101: 663-666,

    1993.

    106. Larsen WG, Felmar EA, Wallace ME, Frieder R. SertoliLeydig

    cell tumor of the ovary: a rare cause of amenorrhea. Obstet Gynecol

    79: 831-833, 1992.

    107. Ohashi M, Hasegawa Y, Haji M, Igarashi M, Nawata H. Production

    of immunoreactive inhibin by a virilizing ovarian tumour (Sertoli

    Leydig tumour). Clin Endocrinol (Oxf) 33: 613-618, 1990.

    108. Mango D, Scirpa P, Liberati M, Menini E, Fabiano A, Mancuso S.

    Ovarian and peripheral steroid hormonesin a case of SertoliLeydig

    cell tumor. J Endocrinol Invest 11: 521-525, 1988.

    109. McCluggage WG, Young RH. Ovarian SertoliLeydig cell tumors

    with pseudoendometrioid tubules (pseudoendometrioid Sertoli

    Leydig cell tumors). Am J Surg Pathol 31: 592-597, 2007.

    110. Mooney EE, Nogales FF, Bergeron C, Tavassoli FA. Retiform

    SertoliLeydig cell tumours: clinical, morphological and immuno-

    histochemical findings. Histopathology 41: 110-117, 2002.

    111. Ching B, Klink A, Wang L. Pathologic quiz case: a 22-year-old

    woman with a large right adnexal mass. Poorly differentiated

    SertoliLeydig cell tumor of the right ovary with retiform differen-

    tiation and heterologous elements (mucinous components). Arch

    Pathol Lab Med 128: e93-95, 2004.

    112. Young RH, Prat J, Scully RE. Ovarian Sertoli-Leydig cell tumors

    with heterologous elements. I. Gastrointestinal epithelium and car-

    cinoid: A clinicopathologic analysis of thirty-six cases. Cancer 50:2448-2456, 1982.

    113. Mooney EE, Nogales FF, Tavassoli FA. Hepatocytic differentiation

    in retiform SertoliLeydig cell tumors: distinguishing a heterolo-

    gous element from Leydig cells. Hum Pathol 30: 611-617, 1999.

    114. Prat J, Young RH, Scully RE. Ovarian SertoliLeydig cell tumors

    with heterologous elements. II. Cartilage and skeletal muscle: a clin-

    icopathologic analysis of twelve cases. Cancer 50: 2465-2475, 1982.

    115. Roth LM, Anderson MC, Govan AD, Langley FA, Gowing NF,

    Woodcock AS. SertoliLeydig cell tumors: a clinicopathologic

    study of 34 cases. Cancer 48: 187-197, 1981.

    116. Young RH, Perez-Atayde AR, Scully RE. Ovarian SertoliLeydig

    cell tumor with retiform and heterologous components. Report of a

    case with hepatocytic differentiation and elevated serum alpha-

    fetoprotein. Am J Surg Pathol 8: 709-718, 1984.

    117. Zaloudek C, Norris HJ. SertoliLeydig tumors of the ovary. A

    clinicopathologic study of 64 intermediate and poorly differentiated

    neoplasms. Am J Surg Pathol 8: 405-418, 1984.

    118. Young RH, Dudley AG, Scully RE. Granulosa cell, SertoliLeydig

    cell, and unclassified sex cord-stromal tumors associated with preg-

    nancy: a clinicopathological analysis of thirty-six cases. Gynecol

    Oncol 18: 181-205, 1984.

    119. Chadha S, Honnebier WJ, Schaberg A. Raised serum alpha-fetoprotein in SertoliLeydig cell tumor (androblastoma) of ovary:

    report of two cases. Int J Gynecol Pathol 6: 82-88, 1987.

    120. Farley JH, Taylor RR, Bosscher JR. Late presentation of an alpha-

    fetoprotein secreting isolated large upper abdominal retroperitoneal

    SertoliLeydig cell tumor recurrence. Gynecol Oncol 56: 319-322,

    1995.

    121. Gagnon S, Tetu B, Silva EG, McCaughey WT. Frequency of alpha-

    fetoprotein production by SertoliLeydig cell tumors of the ovary: an

    immunohistochemical study of eight cases.Mod Pathol 2: 63-67, 1989.

    122. Hammad A, Jasnosz KM, Olson PR. Expression of alpha-

    fetoprotein by ovarian SertoliLeydig cell tumors. Case report

    and review of the literature. Arch Pathol Lab Med 119: 1075-

    1079, 1995.

    123. Singh ZN, Singh MK, Chopra P. SertoliLeydig cell tumor with

    malignant heterologous elements and raised alpha-fetoprotein: a

    case report. J Obstet Gynaecol Res 22: 595-598, 1996.

    124. Tetu B, Ordonez NG, Silva EG. SertoliLeydig cell tumor of the

    ovary with alpha-fetoprotein production. Arch Pathol Lab Med 110:

    65-68, 1986.

    125. Watanabe T, Yamada H, Morimura Y, Abe M, Motoyama T, Sato A.

    Ovarian SertoliLeydig cell tumor with heterologous gastrointesti-

    nal epithelium as a source of alpha-fetoprotein: a case report. J

    Obstet Gynaecol Res 34: 418-421, 2008.

    126. Maillet D, Goulvent T, Rimokh R, et al. Impact of a second opinion

    using expression and molecular analysis of FOXL2 for sex cord-

    stromal tumors. A study of the GINECO group & the TMRO

    network. Gynecol Oncol, 2013. doi:10.1016/j.ygyno.2013.10.013

    127. Banno K, Kisu I, Yanokura M, et al. Hereditary gynecological

    tumors associated with PeutzJeghers syndrome (Review). Oncol

    Lett 6: 1184-1188, 2013.

    128. Ishikawa H, Kiyokawa T, Takatani T, Wen WG, Shozu M.

    Giant multilocular sex cord tumor with annular tubules asso-

    ciated with precocious puberty. Am J Obstet Gynecol 206:

    e14-16, 2012.

    129. Shen K, Wu PC, Lang JH, Huang RL, Tang MT, Lian LJ. Ovarian

    sex cord tumor with annular tubules: a report of six cases. Gynecol

    Oncol 48: 180-184, 1993.

    130. Young RH, Welch WR, Dickersin GR, Scully RE. Ovarian sex cord

    tumor with annular tubules: review of 74 cases including 27 with

    PeutzJeghers syndrome and four with adenoma malignum of the

    cervix. Cancer 50: 1384-1402, 1982.

    131. Hayes MC, Scully RE. Stromal luteoma of the ovary: a clinicopath-

    ologicalanalysis of 25 cases.Int J Gynecol Pathol 6: 313-321, 1987.

    132. Paraskevas M, Scully RE. Hilus cell tumor of the ovary. A clinico-pathological analysis of 12 Reinke crystal-positive and nine crystal-

    negative cases. Int J Gynecol Pathol 8: 299-310, 1989.

    133. Busam KJ, Iversen K, Coplan KA, et al. Immunoreactivity for

    A103, an antibody to melan-A (Mart-1), in adrenocortical and other

    steroid tumors. Am J Surg Pathol 22: 57-63, 1998.

    134. Deavers MT, Malpica A, Ordonez NG, Silva EG. Ovarian steroid

    cell tumors: an immunohistochemical study including a compar-

    ison of calretinin with inhibin. Int J Gynecol Pathol 22: 162-167,

    2003.

    135. Jones MW, Harri R, Dabbs DJ, Carter GJ. Immunohistochemical

    profile of steroid cell tumor of the ovary: a study of 14 cases and a

    review of the literature. Int J Gynecol Pathol 29: 315-320, 2010.

    118 Endocr Pathol (2014) 25:102119

    http://dx.doi.org/10.1016/j.ygyno.2013.10.013http://dx.doi.org/10.1016/j.ygyno.2013.10.013
  • 8/10/2019 10.1007-s12022-013-9285-4

    18/18

    136. Rishi M, Howard LN, Bratthauer GL, Tavassoli FA. Use of

    monoclonal antibody against human inhibin as a marker for

    sex cord-stromal tumors of the ovary. Am J Surg Pathol 21:

    583-589, 1997.

    137. Palmer PE, Bogojavlensky S, Bhan AK, Scully RE. Prolactinoma in

    wall of ovarian dermoid cyst with hyperprolactinemia. Obstet

    Gynecol 75: 540-543, 1990.

    138. Oliva E, Andrada E, Pezzica E, Prat J. Ovarian carcinomas

    with choriocarcinomatous differentiation. Cancer 72: 2441-

    2446, 1993.139. Matias-Guiu X, Prat J. Ovarian tumors with functioning stroma. An

    immunohistochemical study of 100 cases with human chorionic

    gonadotropin monoclonal and polyclonal antibodies. Cancer 65:

    2001-2005, 1990.

    140. Kiyokawa T, Young RH, Scully RE. Krukenberg tumors of the

    ovary: a clinicopathologic analysis of 120 cases with emphasis on

    their variable pathologic manifestations. Am J Surg Pathol 30: 277-

    299, 2006.

    141. Rutgers JL, Scully RE. Cysts (cystadenomas) and tumors of the rete

    ovarii. Int J Gynecol Pathol 7: 330-342, 1988.

    142. Clement PB, Young RH, Scully RE. Clinical syndromes associated

    with tumors of the female genital tract. Semin Diagn Pathol 8: 204-

    233, 1991.

    143. Abeler V, Kjorstad KE, Nesland JM. Small cell carcinoma of the

    ovary. A report of six cases. Int J Gynecol Pathol 7: 315-329, 1988.

    144. Aguirre P, Thor AD, Scully RE. Ovarian small cell carcinoma.

    Histogenetic considerations based on immunohistochemical and

    other findings. Am J Clin Pathol 92: 140-149, 1989.

    145. Walt H, Hornung R, Fink D, et al. Hypercalcemic-type of small cell

    carcinoma of the ovary: characterization of a new tumor line.

    Anticancer Res 21: 3253-3259, 2001.

    146. Burton PB, Knight DE, Quirke P, Smith R, Moniz C. Parathyroid

    hormone related peptide in ovarian carcinoma. J Clin Pathol 43:

    784, 1990.

    147. Chen L, Dinh TA, Haque A. Small cell carcinoma of the ovary withhypercalcemia and ectopic parathyroid hormone production. Arch

    Pathol Lab Med 129: 531-533, 2005.

    148. Matias-Guiu X, Prat J, Young RH, et al. Human parathyroid

    hormone-related protein in ovarian small cell carcinoma. An immu-

    nohistochemical study. Cancer 73: 1878-1881, 1994.

    149. Watson J, Taylor M, Pampiglione J, Rasbridge S, Armitage M. An

    exception to the rule: ectopic ACTH production from functional

    neuroendocrine tissue in an ovarian dermoid cyst. J Endocrinol

    Invest 24: 802-805, 2001.

    150. Schlaghecke R, Kreuzpaintner G, Burrig KF, Juli E, Kley HK.

    Cushings syndrome due to ACTH-production of an ovarian carci-

    noid. Klin Wochenschr 67: 640-644, 1989.

    151. Korzets A, Nouriel H, Steiner Z, et al. Resistant hypertension

    associated with a renin-producing ovarian Sertoli cell tumor. Am J

    Clin Pathol 85: 242-247, 1986.

    Endocr Pathol (2014) 25:102119 119