Endocr Pathol DOI 10.1007/s12022-014-9322-y

In Search of a Prognostic Classification of Endocrine Pituitary Tumors Jacqueline Trouillas

# Springer Science+Business Media New York 2014

Abstract Pituitary tumors, the most frequent intracranial tumors, are historically considered benign. However, various pieces of clinical evidence and recent advances in pathological and molecular data suggest the need to consider that these tumors are more than an endocrinological disease despite the low incidence of metastasis. We present here a historical and critical review of the classifications of pituitary tumors, including a new prognostic clinicopathological classification based on tumor size, immunohistological subtype (prolactin (PRL), growth hormone (GH), follicle-stimulating hormone and luteinizing hormone (FSH-LH), adrenocorticotropic hormone (ACTH), and thyroid-stimulating hormone (TSH)), and five grades, which take into account invasion and proliferation of the tumors. We also present a brief review of the main markers of tumor behavior. We believe that a better classification of these tumors and the identification of prognostic markers will help the clinician to choose the appropriate therapeutic management.

This paper is dedicated to my friend Professor Carlo Capella, one of the pioneers of neuroendocrinology and a member of the “International Pituitary Pathology Club” founded in 1980 by Jules Hardy, Françoise Robert (deceased 1995), Kalman Kovacs and me. J. Trouillas Neuro-oncology and Neuro-inflammation team, Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR5292, 69372 Lyon, France J. Trouillas Université de Lyon, Université Lyon 1, 69372 Lyon, France J. Trouillas (*) Laboratoire d’Histologie et Embryologie Moléculaires, Faculté de Médecine Lyon Est, Site Laennec, and Centre de Pathologie Est, Groupement Hospitalier Est, Hospices Civils de Lyon, Rue Guillaume Paradin, 69372 Lyon, Cedex 8, France e-mail: [email protected]

Keywords Classification . Pituitary tumor . Pituitary adenoma . Prognostic marker

Introduction Endocrine pituitary tumors are the most frequent intracranial neoplasm affecting 1/1,000 of the population [1, 2]. They arise from adenohypophyseal cells and are clinically classified into functioning (mainly growth hormone (GH) with acromegaly; prolactin (PRL) with amenorrhea-galactorrhea; adrenocorticotropic hormone (ACTH) with Cushing’s disease) and nonfunctioning (mainly follicle-stimulating hormone and luteinizing hormone (FSH-LH) tumors. Over the years, the increase of technology and knowledge has allowed the evolution of the pathological classification of pituitary endocrine tumors from a tinctorial classification (three types: acidophilic, basophilic, and chromophobic) to an immunohistochemical classification (five types) plus a dozen ultrastructural subtypes. These tumors were considered as benign. But, 30 to 45 % of them are invasive for the cavernous or the sphenoid sinus [3, 4], and a significant number are considered as aggressive based on recurrence during follow-up [5, 6]. Some of them are named “atypical” adenoma. This new subtype with uncertain malignancy has been identified in the World Health Organization (WHO) 2004 classification [7]. Some of these tumors develop metastasis during the follow-up and are considered as carcinomas [8–10]. While pituitary carcinoma with metastasis is rare, recent evidence suggests that the benign status of the pituitary tumors must be revised. The major hurdles in the management of pituitary tumors are the lack of prognostic markers and also of prognostic classification. I will present a historical and critical review of the pathological classifications of pituitary tumors to date and propose a new prognostic clinicopathological classification [11]. I will

Endocr Pathol

list some pathological, molecular, and genetic markers associated with tumor behavior (not tumorigenesis) which allow personalized management of patients with these tumors.

The Endocrine Pituitary Tumors’ Classifications: 1970–2010 From the Tinctorial Classification to the 2004 WHO Classification Until the 1980s, pituitary tumors were classified based on their tinctorial properties with hematoxylin-eosin-safran/hematoxylin-phloxine-safran (HES or HPS) and were correlated with clinical disease into eosinophilic or acidophilic with acromegaly, basophilic with Cushing’s disease, and chromophobic adenomas. Later, with the development of electron microscopy (EM) and immunohistochemistry (IHC), the tumors were classified based on the appearance of their organelles (granulations and mitochondria) and their hormonal secretion [12–15]. Taking into account the clinical signs and the ultrastructural and IHC data, many subtypes were described: the PRL adenoma or prolactinoma, the GH adenoma with both the sparsely and densely granulated ultrastructural subtypes, the gonadotroph adenoma with hypersecretion of FSH-LH (rev in the study of Snyder [16]) or with normal plasma values [17, 18], the null cell adenoma [19] or immunonegative adenoma [17], the thyroid-stimulating hormone (TSH) adenoma (rev in the study of Beck-Peccoz et al. and Bertholon-Gregoire et al. [20, 21], the “silent” corticotroph subtypes 1 and 2 [22], and the acidophilic stem cell adenomas [23]. In 1996, Kovacs et al. [24] proposed the WHO classification of adenohypophysial neoplasms using a five-tier scheme on the basis of functional, imaging/surgical, histological, immunohistochemical, and ultrastructural findings. While this detailed classification was very interesting, it is highly complex and can only be used by specialized pathologists. For 17 years (1970–1987), I systematically performed EM, but I am convinced that IHC is more powerful than EM and with the promise of molecular biology, I decided to freeze the pieces of tumors rather than keep fragments for EM. Nowadays, expensive and time-consuming EM is only rarely performed by some specialists. The more powerful IHC technique is now used on a routine basis, and tumor fragments are frozen for molecular analyses when possible. Tumors are currently classified into five main IHC types PRL, GH, ACTH, TSH, and FSH-LH which can be monohormonal or plurihormonal, with or without (silent) signs of hypersecretion (Table 1). Almost all the ultrastuctural subtypes were found to be morphological variants identifiable by IHC, and with the improvement of the IHC technique (automation and new antibodies), some disappeared. An

Table 1 Immunohistochemical classification of endocrine pituitary tumors Tumor types

Immunoprofiles

PRL tumor Densely granulateda Sparsely granulated GH tumor

PRL PRL, α-SU

Monohormonal Densely granulated Sparsely granulated Plurihormonal Mixed GH-PRL Mammosomatotroph ACTH tumor Densely granulated Sparsely granulated TSH tumor Monohormonala Plurihormonal FSH-LH tumor Immunonegative tumora “Silent tumors Mono/plurihormonal ACTH GH TSH

GH, (α-SU), Ck, (CgA) GH, (α-SU), Ck, (CgA) GH, PRL, (α-SU, β-TSH) GH, PRL, (α-SU, β-TSH) ACTH, β-endorphin, β-LPH, Ck, (CgA) ACTH, β-endorphin, β-LPH, Ck, (CgA) β-TSH, α-SU β-TSH, α-SU, GH, PRL β-FSH, β-LH, (α-SU), CgA (CgA)

ACTH, β-endorphin, β-LPH GH, PRL (β-TSH) β-TSH, α-SU, GH, PRL

PRL prolactin, SU sub unit, GH growth hormone, FSH-LH folliclestimulating hormone and luteinizing hormone, ACTH adrenocorticotropic hormone, TSH thyroid-stimulating hormone, LPH lipotropin, CgA chromogranin A, CK cytokeratin a

Rare subtype

example is the null cell adenoma [19], which corresponds to FSH-LH tumors with low intracellular hormonal content, undetected by some antibodies yet identified in culture or with a more sensitive IHC technique [25]. We prefer the term of nonimmunoreactive or immunonegative tumors which are now very rare (from 10 % in 1992 to 1 % in 2012 in Lyon’s pathological series). The oncocytoma [26, 27] is not an entity per se but rather a tumor, most often of FSH-LH type with numerous mitochondria. The ultrastructurally densely and sparsely granulated GH tumors are most easily identified using the IHC detection of low-molecular-weight keratins (CAM 5.2 or cytokeratin 8). The juxtanuclear dots, known as “fibrous bodies,” are characteristic of this latter subtype. The expression of cytokeratin is also helpful when identifying ACTH tumors which are usually strongly positive, especially those undergoing Crooke’s changes, although there can be weak staining in silent ACTH tumors which are also positive with galectin-3 [28]. The detection of chromogranin A, characteristic of gonadotroph tumors [29], is also very useful to

Endocr Pathol

establish pituitary origin, especially in the cases of negative immunostaining of the pituitary hormones. Some other subtypes must be considered because they may have a particular behavior. For example, silent corticotroph subtypes 1 and 2 [22] are clinical or cytological variants of ACTH tumors. As recently reported [30, 31], a number of patients with a silent ACTH tumor may present with clinical symptoms of Cushing’s disease at some stage during their follow-up; evidence also exists of a continuum from the macroadenoma with Cushing’s to the silent ACTH tumor [31]. We propose therefore renaming such silent tumors as “ACTH tumor without signs of Cushing’s” similar to the renaming of silent GH tumors [32] as GH adenoma without acromegaly [33]. Acidophilic stem cell adenomas [23] are either GH-PRL or PRL tumors with giant and dilated mitochondria. I believe that it is necessary to provide such precision to clarify the current terminology since some of these subtypes, e.g., the sparsely granulated GH subtype [34], the silent ACTH, silent GH-PRL, or subtype 3 adenomas, may be considered as poorly differentiated tumors with potentially aggressive behavior. Such subtypes must be taken into account for postoperative management, especially in cases of residual tumor. These clinically nonfunctioning tumors should not be confused with the silent FSH-LH tumors, which have a less aggressive course (rev in the study of Cooper and Melmed [30]). The WHO 2004 Classification and a New Prognostic Clinicopathological Classification In the decade after the millenium, it appeared that some tumors without metastasis behaved differently from benign tumors. MRI indicated that some of these tumors invaded the skull base or both cavernous sinuses, but crucially, because the pathologist had no proof of the invasion and metastasis, these tumors continued to be named adenoma and considered benign. However, in 2004, the WHO classification [7] classified all benign tumors as typical adenomas (ICD-0 8272/0) but identified atypical adenomas showing “borderline or uncertain behavior” (ICD-0 8272/1). Such tumors have atypical morphologic features suggestive of aggressive behavior such as invasive growth. Other features include an elevated mitotic index and a Ki-67 labeling index greater than 3 %, as well as extensive nuclear staining for p53 immunoreactivity. The term of pituitary carcinomas (ICD-0 8272/2) continues to be restricted to the pituitary tumor with systemic or cerebrospinal metastases [35, 36]. Although markers of proliferation, p53 detection, and invasion were all mentioned as criteria of the “atypical adenoma,” invasion has not been systematically considered, as illustrated by the two following papers. In Laws’ series of 121 consecutive patients [4], the frequency of atypical adenoma was 15 % compared to 2.5 % in the Saeger et al.’s series of 241 tumors from the German registry [37]. This discrepancy is explained by the differences in

criteria taken into account for the diagnosis: a Ki-67 > 3 %, p53 positivity, and increased mitotic activity without a cutoff value in the Laws’ series [4] and invasion associated with Ki67 > 3 % and p53 > 5 % in the Saeger’s series [37]. Thus, as advised by Wolfsberger and Knosp [38], “the definition of invasiveness is needed and should be included in this classification as in a previous classification” [39], and the proliferation must be evaluated by markers of the cell cycle with welldefined thresholds. Moreover, as some experts [40–42] have pointed out, this classification needs to be correlated to clinical evolution with postoperative results, progression, and recurrence. Thus, we initiated a multicentric collaborative study in 2006 to validate the clinical prognostic value of a clinicopathological classification which has been recently published [11]. This classification takes into account both tumor size and the five IHC types (PRL, GH, FSH-LH, ACTH, and TSH). We set up a grading system, such as that used for other endocrine tumors of the foregut, [43] based on invasion and proliferation status (Table 2). MRI is needed to evaluate the invasion since the histological proof of invasion is rare (9 % of invasive tumors in this series). Only invasion into the sphenoid sinus, confirmed by the infiltrated respiratory mucosae on histology, and unequivocal invasion of the cavernous sinus were considered [44]. Indeed, recent anatomical studies have shown that the medial wall of the cavernous sinus is composed of dura [45], and moreover, the intracavernous invasion can be assessed peroperatively by an endoscopic technique. Using multivariate statistical analysis and a ROC curve [11], we

Table 2 Clinicopathological classification of the endocrine pituitary tumors The classification is based on the three following characteristics: 1. Tumor size into micro (40 mm) by MRI 2. Tumor type into GH, PRL, ACTH, FSH-LH, and TSH by IHC 3. Tumor grade based on the following criteria: Invasion defined as histological and/or radiological (MRI) signs of cavernous or sphenoid sinus invasion Proliferation considered on the presence of at least two of the three criteria: Mitoses: n>2/10 HPF; Ki-67: ≥3 %; p53: positive (>10 strongly positive nuclei/10 HPF) The five grades are the following: Grade 1a: noninvasive tumor Grade 1b: noninvasive and proliferative tumor Grade 2a: invasive tumor Grade 2b: invasive and proliferative tumor Grade 3: metastatic tumor (cerebrospinal or systemic metastases) From Trouillas J et al. [11]

Endocr Pathol

confirmed that invasion is a major prognostic factor in predicting both the disease-free status following surgical removal of pituitary tumors [46] and recurrence/progression [47]. In our opinion, invasion must be added to the synoptic checklist for pituitary lesions recently published by a group of experts [48], especially considering that data from pituitary imaging and surgical findings are included in the patient database, which is now available to pathologists. Tumor proliferation is evaluated by the two most commonly used cell cycle markers in oncology (Ki-67 index, and mitotic count) as well as p53. Considering the controversial value of these markers, which will be discussed later, and the lack of methodological standards and validated cutoff for p53, we defined proliferation as the presence of at least two of these markers with cutoff values of 3 % for Ki-67 index with formol fixative and 1 % for Bouin-Hollande fixative and the number of mitoses n>2/10 high power fields (HPFs), as for endocrine pancreatic tumors [43] and the positivity for p53 if at least 10 nuclei/10 HPFs were strongly positive. With this grading, nearly half of the tumors were grade 1a (47.3 %), 10 % were grade 1b, 27.6 % were grade 2a, and 15.1 % were grade 2b. Eight tumors (2.0 %), four PRL, and four ACTH, classified as grade 1b (n=1), 2a (n=1), and 2b (n=6) at the time of initial surgery, were later classified as carcinomas (grade 3) based on the occurrence of metastases during the 8-year follow-up. The series of 410 patients in our case-control study is representative of pituitary tumor behavior with a similar percentage of invasive tumors (43 %) to that in other series [3] and a percentage of grade 2b (15 %) similar to the percentage of the atypical adenoma in the Laws’ surgical series [4]. However, prospective studies must now be performed to evaluate the exact percentage of each grade for each IHC type. By multivariate analysis, we confirmed the poor prognostic value of young age in predicting disease-free outcome and recurrence/progression status at 8 years (rev in the study of Roelfsema et al. [49]). However, while tumor size was not associated with the recurrence/progression status, it was with disease-free outcome [50]. Patient sex had no prognostic value, as suggested previously [49]. Our proposed classification displayed highly significant prognostic value for predicting postoperative disease-free outcome or recurrence/ progression status, across all tumors and for each type of tumor. At 8-year follow-up, the probability of a patient showing evidence of disease or tumor progression was 25and 12-fold higher, respectively, if they had an invasive and proliferative tumor (grade 2b) as compared to if they had a noninvasive and nonproliferative tumor (grade 1a). These results confirm those obtained in our preliminary study on 94 PRL tumors [51] that were classified into three groups, i.e., noninvasive, invasive, and aggressiveinvasive, corresponding respectively to the grades 1a, 2a, and 2b of the present classification.

Challenges of Pituitary Tumors As there are no obvious signs of malignancy, pituitary carcinoma is only defined when the tumor has metastasized. It is rare and accounts for about 0.2 % of pituitary tumors with only 132 cases reported in the literature from 1961 to 2009 [9, 10, 36]. Most pituitary carcinomas are functioning, secreting PRL (36 % cases) (rev in the study of Kars et al. [52]) or ACTH hormone (30 % cases) (rev in the study of Landman et al. [53]). Nonfunctioning tumors are less frequent (23 % cases), especially the FSH-LH carcinoma [54]. Previously, histological signs of malignancy were thought to be absent; however, based on the frequency of certain signs in previous pathological series of pituitary carcinomas [55–57], a potential malignancy could reasonably be suspected based on the association of the following pathological signs: invasion, neoangiogenesis, vascular invasion, abnormal mitoses, very high index of Ki-67> 10 %, and p53>5 % and genomic alteration (chromosome 11 for PRL tumors), which if combined might be considered as criteria of malignancy. However, these cutoff values were not validated, and some of the above signs were found to be absent in certain metastatic pituitary carcinomas [56, 57]. The observation that six out of the eight carcinomas of our series were grade 2b at the first surgery [11], together with the comparison of human PRL tumors with our animal model (SMtTW model) [58], led us to postulate that grade 2b or aggressive-invasive tumors are in fact malignant tumors without metastasis [59–61]. In oncology, the term “adenoma” signifies that a tumor is benign. I propose that the generic term should be “pituitary tumor” instead of “pituitary adenoma” because this tumor can be benign or malignant. Furthermore, we propose that, instead of atypical adenoma or “aggressive adenoma,” the term “tumor suspected of malignancy” (tumor grade 2b) be used. The characteristics of the tumors classified according to malignancy are summarized in Table 3 [55, 62].

Molecular Markers Associated with Tumor Behavior While the expression of several biological markers has been investigated by IHC and correlated to invasiveness and/or aggressive behavior, no single marker has been found to predict the tumor behavior, and until now, their detection is only performed at the research level. As some exhaustive reviews have been published on this subject [63–66], here, I will focus on biomarkers that appear to correlate with invasive and aggressive behavior of pituitary tumors.

Endocr Pathol Table 3 Classification of endocrine pituitary tumors according to malignancy

From Pernicone PJ et al. [55], Trouillas J et al. [62] HPF high power field, LI labeling index

Tumor characteristics

Benign tumor/adenoma

Tumor suspected of malignancy/grade 2b

Malignant tumor/carcinoma

Tumor size (MRI) Invasion Metastasis Mitoses (n/10 HPF) Ki-67 LI p53 positivity Neoangiogenesis Vascular invasion

< or >10 mm No No 0 or 2 >3 % Variable 0 0

>10 mm Yes Yes 2–10 (abnormal) >10 % or low Usually positive Variable Rare

Cell Cycle Markers Ki-67 Nuclear proteins, such as Ki-67, are also used to assess proliferation. Ki-67, which is expressed throughout the cell cycle, is now detected in routine by a monoclonal antibody (MIB-1). The Ki-67 labeling index (LI), defined as a percentage of positive nuclei, is a very easy reproducible and reliable method. Although the Ki-67 LI varied according to the fixative (it is lower in Bouin-Hollande than in formol fixative), its correlation with tumor growth and invasion is conflicting [46, 47, 67–72]. Ki-67 LI >3 % is considered as a good sign of proliferation, but it varies according to IHC type [47], and it is higher in the functioning tumors than in nonfunctioning ones [73], and so by itself, it does not predict the tumor behavior, and the use of other makers is recommended. Mitoses The presence and the number of mitoses are important predictive factors of a rapid growth. However, given the low proliferative rate of the majority of pituitary tumors, only scarce mitoses can be identified, more easily with Ki-67 immunostaining than with HPS staining. As for endocrine pancreatic tumors [43], I consider that a tumor is proliferative if the number of mitoses is higher than 2/10 HPFs. p53 and Other Cell Cycle Markers The expression of p53 gene product is an important marker in many cancers. But, p53 mutations have not been described in pituitary tumors, and the value of its detection is controversial. Indeed, its evaluation may differ from one laboratory to another and from one pathologist to another, and its correlation with invasion and aggressiveness/malignancy was found in some series [71] and not in others [74]. Since p53 has been found elevated in almost all pituitary carcinomas [71] and “an

extensive p53 positivity” is one of the criteria of the atypical adenoma [7], we decided to consider p53. In our classification, we considered its positivity and not its precise percentage. I propose to stipulate its percentage for future studies. Compared to their noninvasive counterparts, invasive pituitary tumors also express higher levels of pituitary tumor transforming gene (PTTG) which is a member of the securin family that regulates sister chromatid separation during mitosis [75]. Other cell cycle markers have also been studied (topoisomerase IIα, cyclins, cyclin-dependant kinases, etc.) but are not used routinely.

Growth Factors, Metalloproteinases, and Angiogenesis Of the fibroblast growth factors (FGFs) and their receptors (FGFRs), which regulate growth, differentiation, migration, and angiogenesis, FGF2 and FGF4 are expressed in the pituitary. Tumors show a loss of FGFR2, with a resultant upregulation of MAGEA3, and a truncated isoform of FGFR4, known as pituitary tumor-derived (ptd-FGFR4). The expression of the latter induces invasive growth of pituitary tumor cells in vivo with loss of membranous N-cadherin expression [76]. Moreover, ptd-FGFR4 interacts with a polysialated form of neuronal cell adhesion molecule (PSA-NCAM), which correlates with invasiveness [72, 77]. The matrix metalloproteinase (MMP) family of proteolytic enzymes cleaves extracellular matrix molecules and plays a role in invasiveness of many neoplasms. The expression of MMP9 is significantly higher in invasive pituitary tumors [78] and may be correlated with the activation of protein kinase C, which is also known to be involved in the invasion of pituitary tumors. Although the tumors are less vascular than the normal pituitary, a significantly increased microvessel density has been found in invasive PRL tumors than in the noninvasive ones. Moreover, the expression of endocan, a proteoglycan secreted by endothelial cells, has also been associated with size and progression of pituitary tumors [79].

Endocr Pathol

Transcription Factors

Table 4 Checklist for pathological diagnosis of pituitary endocrine tumors

Transcription factors involved in pituitary cell differentiation can also be examined and may be useful to confirm the IHC diagnosis, particularly of immunonegative or “silent tumors.” Pit-1 is expressed in GH, PRL, and TSH tumors [80], T-pit in ACTH tumors with and without Cushing’s disease [81], and SF-1 in FSH-LH tumors [82]. Moreover, the detection of somatostatin receptors type 2 (SSTR2) and 5 (SSTR5) could be helpful in GH, TSH, ACTH, and FSH-LH tumors to identify those with low SSTR2 expression, shown to represent a higher risk of resistance to octreotide/lanreotide, or those with high SSTR5R expression more likely to respond to pasireotide treatment. This detection is always negative in PRL tumors [83]. My pathological diagnosis of endocrine pituitary tumors uses this checklist, which is different from the recent published one [48], in that it takes into account the MRI and the peroperative data. It also includes freezing tumor samples for future molecular biology analyses (Table 4).

I - Preoperative pituitary MRI Tumor size: Micro, Macro, Giant tumor Invasion: No yes localization: cavernous sinus sphenoidal sinus other Peroperative invasion: yes no ND II - Histology 1) Description 2) Invasion ND No yes localization: Juxtatumoral pituitary: 3) Immunohistochemistry (Note the fixative and the clones of the tested antibodies)

Genetic Markers Recent progress in molecular and genetic techniques (transcriptome, comparative genetic hybridization, and epigenetic) allowed the identification of variations in gene expression (microRNA and miRNA), loss of heterozygosity (LOH), and gene mutation in pituitary tumors (rev in the study of Raverot et al. [66]). These techniques need quickly frozen tissue in sufficient quantity and a control of the frozen samples by the pathologist to avoid contamination by normal pituitary. Moreover, the molecular data must be correlated to precise clinical and pathological data. Using rigorous conditions, some of these markers were related to invasion and aggressiveness in gonadotroph tumors [75, 84] and PRL tumors [51, 59, 60]. Fifteen years ago, Bates et al. identified a significantly higher frequency of LOH in invasive tumors compared to noninvasive tumors [85], and this has been confirmed in other publications. We recently demonstrated that the 11p region was commonly deleted in PRL aggressive tumors (grade 2b) [61] and a loss of the 11q arm and a gain in the 1q arm in the three aggressive tumors that developed metastasis during follow-up and so became malignant. These results suggested that accumulation of new chromosomal alterations (11q loss and 1q gain) may transform an “aggressive” pituitary tumor into a pituitary carcinoma. Pituitary tumors are mostly sporadic, but some familial cases have been identified leading to the characterization of genetic forms of pituitary adenoma. The search for MEN1 or AIP mutation is necessary in patients with a familial history [86], or in young patients with a large macroadenoma [87, 88],

Antibodies to be tested: a) For the tumor typage Chromogranine A, βFSH, βLH, αSU → FSH or FSH-LH or LH or αSU tumor PRL, GH, CK → GH or GH-PRL or PRL tumor, silent GH or GH-PRL tumor ACTH, βend, CK → ACTH tumor, silent ACTH tumor -βTSH, CK → TSH tumor -SSTR 2-5 for GH, TSH, FSH-LH and ACTH tumors If all the antibodies negative: immunonegative tumor b) For evaluation of proliferation (count for 10 HPF) Index of Ki-67:(cutoff >3 %); Number of mitoses: (cutoff n > 2) Expression of p53: + or − (cutoff n > 10 positive nuclei); Index: c) Grading: taking into account invasion (MRI) and proliferation (two or three markers > cutoff) Grade 1a, Grade 1b, Grade 2a, Grade 2b, Grade 3 (if metastasis) III - Molecular biology Frozen samples of tumor after control by smears or frozen sections. Keep at −80° or in liquid nitrogen for microarrays, PCR, and CGH.

or in the case of unusual plurihormonality or double adenomas [89]. Recent advances in pituitary tumor classification now allow the early identification of pituitary tumors with high risk of recurrence, associated with a malignant potential. In such cases (grade 2b), after discussion by a designated team of neurosurgeon, endocrinologist, pathologist, and oncologist, an optimized therapeutic strategy should be proposed taking into account new therapeutic options [6] in addition to conventional therapies associating surgery and radiotherapy [90].

Conclusion The evidence against pituitary tumors being considered only as benign tumors inducing hormonal disease is mounting. Besides the rare carcinomas and the frequent noninvasive, nonproliferative benign pituitary tumors, a group of tumors

Endocr Pathol

representing almost 15 % of all pituitary tumors should be individualized. We propose to name such grade 2b tumors with high risk of recurrence as tumor suspected of malignancy. Indeed, clinical, pathological, and molecular evidence suggests that some of these tumors may develop metastasis during follow-up. Recent genomic studies identifying new molecular markers associated with tumor behavior may help to classify these tumors. Until markers of malignancy were discovered, comprehensive classification of pituitary tumors remains the best indicator of clinical behavior in patients with pituitary tumors, but their prognostic value has to be proved with multivariate statistic methods in prospective studies. Acknowledgments I thank G Sassolas who, in 1970, suggested that I study pituitary tumors, which, at this time, were not considered as the most frequent cause of acromegaly and of amenorrhea-galactorrhea. I am delighted that G. Raverot, Prof. of endocrinology, now the leader of the research on pituitary tumors in Lyon, along with E. Jouanneau, Prof. of neurosurgery, and Dr A. Vasiljevic, neuropathologist, all cooperate in the diagnosis and patient management of pituitary tumors. Many thanks to P Chevallier, A. Reynaud, and M.P. Guigard for expert technical assistance during 10 to 30 years and to my secretary P. Gerardi for typing our manuscripts. At least, I thank all the members of HYPOPRONOS who gave data of their patients: R. Assaker, C. Auger, A. Barlier, M. Bernier, F. Bonnet, J. F. Bonneville, F. Borson-Chazot, G. Brassier, T. Brue, S. CauletMaugendre, O. Chabre, F. Chapuis, P. Chanson, A. Cornelius, C. Cortet-Rudelli, J. F. Cottier, E. Dantony, B. Delemer, E. Delgrange, L. Di Tommaso, H. Dufour, S. Eimer, D. Figarella-Branger, P. François, S. Gaillard, F. Galland, J. J. Girard, M. Jan, J. Lachuer, V. Lapras, H. Loiseau, C. A. Maurage, F. Mougel, J. G. Passagia, M. Patey, A. Penfornis, J. Y. Poirier, G. Perrin, P. Roy, N. Sturm, A. Tabarin, G. Viennet, L. Villeneuve, and A. Wierinckx. Conflict of Interest The authors have nothing to disclose. This work was supported by grants from the Ministère de la Santé (Programme Hospitalier de Recherche Clinique National no. 27-43, HYPOPRONOS) and research contracts with the Institut National de la Santé et de la Recherche Médicale and the Ligue Contre le Cancer Rhône-Alpes.

References 1. Daly AF, Rixhon M, Adam C, Dempegioti A, Tichomirowa MA, Beckers A (2006) High prevalence of pituitary adenomas: a crosssectional study in the province of Liege, Belgium. J Clin Endocrinol Metab 91: 4769-4775 2. Fernandez A, Karavitaki N, Wass JA (2010) Prevalence of pituitary adenomas: a community-based, cross-sectional study in Banbury (Oxfordshire, UK). Clin Endocrinol (Oxf) 72: 377-382 3. Meij BP, Lopes MB, Ellegala DB, Alden TD, Laws ER, Jr. (2002) The long-term significance of microscopic dural invasion in 354 patients with pituitary adenomas treated with transsphenoidal surgery. J Neurosurg 96: 195-208 4. Zada G, Woodmansee WW, Ramkissoon S, Amadio J, Nose V, Laws ER, Jr. (2011) Atypical pituitary adenomas: incidence, clinical characteristics, and implications. J Neurosurg 114: 336-344 5. McCormack AI, Wass JA, Grossman AB (2011) Aggressive pituitary tumours: the role of temozolomide and the assessment of MGMT status. Eur J Clin Invest 41: 1133-1148

6. Raverot G, Castinetti F, Jouanneau E, Morange I, Figarella-Branger D, Dufour H, Trouillas J, Brue T (2012) Pituitary carcinomas and aggressive pituitary tumours: merits and pitfalls of temozolomide treatment. Clin Endocrinol (Oxf) 76: 769-775 7. Lloyd RV, Kovacs K, Young WF J, Farrell WE, Asa SL, Trouillas J, Kontogeorgos G, Sano T, Scheithauer BW, E. H: (2004) Pituitary tumours: introduction. In chaper 1: Tumours of the pituitary. DeLellis RA, Lloyd RV, Heitz PU, Eng C (ed) World Health Organization Classification of Tumours - Pathology and genetics of tumours of endocrine organs, IARC Press, Lyon, pp 10-13 8. Batisse M, Raverot G, Maqdasy S, Durando X, Sturm N, Montoriol PF, Kemeny JL, Chazal J, Trouillas J, Tauveron I (2013) Aggressive silent GH pituitary tumor resistant to multiple treatments, including temozolomide. Cancer Invest 31: 190-196 9. Dudziak K, Honegger J, Bornemann A, Horger M, Mussig K (2011) Pituitary carcinoma with malignant growth from first presentation and fulminant clinical course–case report and review of the literature. J Clin Endocrinol Metab 96: 2665-2669 10. Heaney AP (2011) Clinical review: pituitary carcinoma: difficult diagnosis and treatment. J Clin Endocrinol Metab 96: 3649-3660 11. Trouillas J, Roy P, Sturm N, Dantony E, Cortet-Rudelli C, Viennet G, Bonneville JF, Assaker R, Auger C, Brue T, Cornelius A, Dufour H, Jouanneau E, Francois P, Galland F, Mougel F, Chapuis F, Villeneuve L, Maurage CA, Figarella-Branger D, Raverot G (2013) A new prognostic clinicopathological classification of pituitary adenomas: a multicentric case-control study of 410 patients with 8 years postoperative follow-up. Acta Neuropathol 126: 123-135 12. Asa SL, Kovacs K (1983) Histological classification of pituitary disease. Clin Endocrinol Metab 12: 567-596 13. Trouillas J, Cure M, Lheritier M, Guichard Y, Girod C (1974) [Attempted at a cytofunctional classification of 19 pituitary adenomas using histological, immunocytochemical, biometric and ultrastructural data]. Rev Otoneuroophtalmol 46: 223-236 14. Trouillas J, Girod C (1996) Pathology of pituitary adenomas. In Landolt AM, Vance ML, Reilly PL (ed) Pituitary Adenomas, Churchill Livingstone, New-York, pp 27-46 15. Girod C, Mazucca M, Trouillas J, Tramu G, Lheritier M, Beauvillain JC, Claustat B, Dubois MP (1980) Light microscopy, fine structure and immunohistochemistry studies of 278 pituitary adenomas. In Derome PJ, Jedynak CP, Peillon F (ed) Pituitary adenomas, Asclepios, Paris, pp 3-18 16. Snyder PJ (1985) Gonadotroph cell adenomas of the pituitary. Endocr Rev 6: 552-563 17. Trouillas J, Girod C, Sassolas G, Claustrat B (1986) The human gonadotropic adenoma: pathologic diagnosis and hormonal correlations in 26 tumors. Semin Diagn Pathol 3: 42-57 18. Trouillas J, Girod C, Sassolas G, Claustrat B, Lheritier M, Dubois MP, Goutelle A (1981) Human pituitary gonadotropic adenoma; histological, immunocytochemical, and ultrastructural and hormonal studies in eight cases. J Pathol 135: 315-336 19. Kovacs K, Horvath E, Ryan N, Ezrin C (1980) Null cell adenoma of the human pituitary. Virchows Arch A Pathol Anat Histol 387: 165-174 20. Beck-Peccoz P, Brucker-Davis F, Persani L, Smallridge RC, Weintraub BD (1996) Thyrotropin-secreting pituitary tumors. Endocr Rev 17: 610-638 21. Bertholon-Gregoire M, Trouillas J, Guigard MP, Loras B, Tourniaire J (1999) Mono- and plurihormonal thyrotropic pituitary adenomas: pathological, hormonal and clinical studies in 12 patients. Eur J Endocrinol 140: 519-527 22. Horvath E, Kovacs K, Killinger DW, Smyth HS, Platts ME, Singer W (1980) Silent corticotropic adenomas of the human pituitary gland: a histologic, immunocytologic, and ultrastructural study. Am J Pathol 98: 617-638 23. Horvath E, Kovacs K, Singer W, Smyth HS, Killinger DW, Erzin C, Weiss MH (1981) Acidophil stem cell adenoma of the human pituitary: clinicopathologic analysis of 15 cases. Cancer 47: 761-771

Endocr Pathol 24. Kovacs K, Scheithauer BW, Horvath E, Lloyd RV (1996) The World Health Organization classification of adenohypophysial neoplasms. A proposed five-tier scheme. Cancer 78: 502-510 25. Trouillas J, Girod C, Loras B, Sassolas G, Claustrat B, Perrin G (1990) [Normal and tumorous human gonadotropic cells]. Ann Endocrinol (Paris) 51: 54-64 26. Trouillas J, Lheritier M, Cure M, Girod C, Guinet P, Tommasi M, Goutelle A, Allegre GE (1975) L’oncocytome hypophysaire est-il une entité ? Discussion anatomo-clinique à propos de 3 observations personnelles. Lyon Med. 234: 25-35 27. Kovacs K, Horvath E (1973) Pituitary "chromophobe" adenoma composed of oncocytes. A light and electron microscopic study. Arch Pathol 95: 235-239 28. Thodou E, Argyrakos T, Kontogeorgos G (2007) Galectin-3 as a marker distinguishing functioning from silent corticotroph adenomas. Hormones (Athens) 6: 227-232 29. Riva C, Leutner M, Capella C, Usellini L, la Rosa S, Brianza MR, Buffa R (1993) Different expression of chromogranin A and chromogranin B in various types of pituitary adenomas. Zentralbl Pathol 139: 165-170 30. Cooper O, Melmed S (2012) Subclinical hyperfunctioning pituitary adenomas: the silent tumors. Best Pract Res Clin Endocrinol Metab 26: 447-460 31. Raverot G, Wierinckx A, Jouanneau E, Auger C, Borson-Chazot F, Lachuer J, Pugeat M, Trouillas J (2010) Clinical, hormonal and molecular characterization of pituitary ACTH adenomas without (silent corticotroph adenomas) and with Cushing’s disease. Eur J Endocrinol 163: 35-43 32. Horvath E, Kovacs K, Smith H, Cusimano M, Singer W (2005) Silent adenoma subtype 3 of the pituitary-immunohistochemical and ultrastructural classification: a review of 29 cases. Ultrastruct Pathol 29: 511-524 33. Tourniaire J, Trouillas J, Chalendar D, Bonneton-Emptoz A, Goutelle A, Girod C (1985) Somatotropic adenoma manifested by galactorrhea without acromegaly. J. Clin. Endocrinol. Metab. 61: 451-453 34. Obari A, Sano T, Ohyama K, Kudo E, Qian ZR, Yoneda A, Rayhan N, Mustafizur Rahman M, S. Y (2008) Clinicopathological features of growth hormone-producing pituitary adenomas: difference among various types defined by cytokeratin distribution pattern including a transitional form. Endocr Pathol 19: 82-91 35. Scheithauer BW, Gaffey TA, Lloyd RV, Sebo TJ, Kovacs KT, Horvath E, Yapicier O, Young WF, Jr., Meyer FB, Kuroki T, Riehle DL, Laws ER, Jr. (2006) Pathobiology of pituitary adenomas and carcinomas. Neurosurgery 59: 341-353; discussion 341-353 36. Kaltsas GA, Nomikos P, Kontogeorgos G, Buchfelder M, Grossman AB (2005) Diagnosis and Management of Pituitary Carcinomas. J Clin Endocrinol Metab 90: 3089-3099 37. Saeger W, Ludecke DK, Buchfelder M, Fahlbusch R, Quabbe HJ, Petersenn S (2007) Pathohistological classification of pituitary tumors: 10 years of experience with the German Pituitary Tumor Registry. Eur J Endocrinol 156: 203-216 38. Wolfsberger S, Knosp E (2006) Comments on the WHO 2004 classification of pituitary tumors. Acta Neuropathol 111: 66-67 39. Solcia E, Klöppel G, Sobin L: (2000) Histological classification of adenohypophyseal tumours. Springer Heidelberg New-York pp 6-29 40. Figarella-Branger D, Trouillas J (2006) The new WHO classification of human pituitary tumors: comments. Acta Neuropathol 111: 71-72 41. Grossman AB (2006) The 2004 World Health Organization classification of pituitary tumors: is it clinically helpful? Acta Neuropathol (Berl) 111: 76-77 42. Laws ER, Jr., Lopes MB (2006) The new WHO classification of pituitary tumors: highlights and areas of controversy. Acta Neuropathol 111: 80-81 43. Rindi G, Kloppel G, Alhman H, Caplin M, Couvelard A, de Herder WW, Erikssson B, Falchetti A, Falconi M, Komminoth P, Korner M, Lopes JM, McNicol AM, Nilsson O, Perren A, Scarpa A, Scoazec JY,

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

55.

56.

57. 58.

59.

60.

Wiedenmann B (2006) TNM staging of foregut (neuro)endocrine tumors: a consensus proposal including a grading system. Virchows Arch 449: 395-401 Cottier JP, Destrieux C, Brunereau L, Bertrand P, Moreau L, Jan M, D. H (2000) Cavernous sinus invasion by pituitary adenoma: MR imaging. Radiology 215: 463-469 Francois P, Travers N, Lescanne E, Arbeille B, Jan M, Velut S (2010) The interperiosteo-dural concept applied to the perisellar compartment: a microanatomical and electron microscopic study. J Neurosurg 113: 1045-1052 Knosp E, Steiner E, Kitz K, Matula C (1993) Pituitary adenomas with invasion of the cavernous sinus space: a magnetic resonance imaging classification compared with surgical findings. Neurosurgery 33: 610-617; discussion 617-618 Righi A, Agati P, Sisto A, Frank G, Faustini-Fustini M, Agati R, Mazzatenta D, Farnedi A, Menetti F, Marucci G, Foschini MP (2012) A classification tree approach for pituitary adenomas. Hum Pathol 43: 1627-1637 Nose V, Ezzat S, Horvath E, Kovacs K, Laws ER, Lloyd R, Lopes MB, Asa SL (2011) Protocol for the examination of specimens from patients with primary pituitary tumors. Arch Pathol Lab Med 135: 640-646 Roelfsema F, Biermasz NR, Pereira AM (2012) Clinical factors involved in the recurrence of pituitary adenomas after surgical remission: a structured review and meta-analysis. Pituitary 15: 71-83 Losa M, Mortini P, Barzaghi R, Ribotto P, Terreni M, Marzoli S, Pieralli S, Giovanelli M (2008) Early results of surgery in patients with nonfunctioning pituitary adenoma and analysis of the rick of tumor recurrence. J Neurosurg 108: 525-532 Raverot G, Wierinckx A, Dantony E, Auger C, Chapas G, Villeneuve L, Brue T, Figarella-Branger D, Roy P, Jouanneau E, Jan M, Lachuer J, Trouillas J (2010) Prognostic factors in prolactin pituitary tumors: clinical, histological, and molecular data from a series of 94 patients with a long postoperative follow-up. J Clin Endocrinol Metab 95: 1708-1716 Kars M, Roelfsema F, Romijn J, Pereira A (2006) Malignant prolactinoma: case report and review of the literature. Eur J Endocrinol 155: 523-534 Landman RE, Horwith M, Peterson RE, Khandji AG, Wardlaw SL (2002) Long-term survival with ACTH-secreting carcinoma of the pituitary: a case report and review of the literature. J Clin Endocrinol Metab 87: 3084-3089 Beauchesne P, Trouillas J, Barral F, Brunon J (1995) Gonadotropic pituitary carcinoma: case report. Neurosurgery 37: 810-815; discussion 815-816 Pernicone PJ, Scheithauer BW, Sebo TJ, Kovacs KT, Horvath E, Young WF, Jr., Lloyd RV, Davis DH, Guthrie BL, Schoene WC (1997) Pituitary carcinoma: a clinicopathologic study of 15 cases. Cancer 79: 804-812 Scheithauer BW, Kurtkaya-Yapicier O, Kovacs KT, Young WF, Jr., Lloyd RV (2005) Pituitary carcinoma: a clinicopathological review. Neurosurgery 56: 1066-1074; discussion 1066-1074 Lubke D, Saeger W (1995) Carcinomas of the pituitary: definition and review of the literature. Gen Diagn Pathol 141: 81-92 Trouillas J, Chevallier P, Claustrat B, Hooghe-Peters E, Dubray C, Rousset B, Girod C (1994) Inhibitory effects of the dopamine agonists quinagolide (CV 205-502) and bromocriptine on prolactin secretion and growth of SMtTW pituitary tumors in the rat. Endocrinology 134: 401-410 Wierinckx A, Auger C, Devauchelle P, Reynaud A, Chevallier P, Jan M, Perrin G, Fevre-Montange M, Rey C, Figarella-Branger D, Raverot G, Belin MF, Lachuer J, Trouillas J (2007) A diagnostic marker set for invasion, proliferation, and aggressiveness of prolactin pituitary tumors. Endocr Relat Cancer 14: 887-900 Wierinckx A, Raverot G, Nazaret N, Jouanneau E, Auger C, Lachuer J, Trouillas J (2010) Proliferation markers of human pituitary tumors:

Endocr Pathol

61.

62.

63. 64.

65. 66.

67.

68.

69.

70.

71.

72.

73.

74.

75.

76. 77.

contribution of a genome-wide transcriptome approach. Mol Cell Endocrinol 326: 30-39 Wierinckx A, Roche M, Raverot G, Legras-Lachuer C, Croze S, Nazaret N, Rey C, Auger C, Jouanneau E, Chanson P, Trouillas J, Lachuer J (2011) Integrated genomic profiling identifies loss of chromosome 11p impacting transcriptomic activity in aggressive pituitary PRL tumors. Brain Pathol 21: 533-543 Trouillas J, Raverot G, Jouanneau E, Auger C: (2010) Carcinome hypophysaire. In Tumeurs malignes rares.Droz JP, Ray-Coquard I, Peix JL (ed) Springer, Paris, pp 441-445 Kontogeorgos G (2006) Predictive markers of pituitary adenoma behavior. Neuroendocrinology 83: 179-188 Gurlek A, Karavitaki N, Ansorge O, Wass JA (2007) What are the markers of aggressiveness in prolactinomas? Changes in cell biology, extracellular matrix components, angiogenesis and genetics. Eur J Endocrinol 156: 143-153 Mete O, Ezzat S, Asa SL (2012) Biomarkers of aggressive pituitary adenomas. J Mol Endocrinol 49: R69-78 Raverot G, Jouanneau E, Trouillas J (2014) Management of endocrine disease: clinicopathological classification and molecular markers of pituitary tumours for personalized therapeutic strategies. Eur J Endocrinol Mar 13 doi: 10.1530/EJE-13-1031 Amar AP, Hinton DR, Krieger MD, Weiss MH (1999) Invasive pituitary adenomas: significance of proliferation parameters. Pituitary 2: 117-122 Dubois S, Guyetant S, Menei P, Rodien P, Illouz F, Vielle B, Rohmer V (2007) Relevance of Ki-67 and prognostic factors for recurrence/ progression of gonadotropic adenomas after first surgery. Eur J Endocrinol 157: 141-147 Knosp E, Kitz K, Perneczky A (1989) Proliferation activity in pituitary adenomas: measurement by monoclonal antibody Ki-67. Neurosurgery 25: 927-930 Saeger W, B L, DK L (2008) Clinical tumor growth and comparison with proliferation markers in non-functioning (inactive) pituitary adenomas. Exp Clin Endocrinol Diabetes 116: 80-85 Thapar K, Kovacs K, Scheithauer BW, Stefaneanu L, Horvath E, Pernicone PJ, Murray D, Laws ER, Jr. (1996) Proliferative activity and invasiveness among pituitary adenomas and carcinomas: an analysis using the MIB-1 antibody. Neurosurgery 38: 99-106; discussion 106-107 Trouillas J, Daniel L, Guigard MP, Tong S, Gouvernet J, Jouanneau E, Jan M, Perrin G, Fischer G, Tabarin A, Rougon G, FigarellaBranger D (2003) Polysialylated neural cell adhesion molecules expressed in human pituitary tumors and related to extrasellar invasion. J Neurosurg 98: 1084-1093 Jaffrain-Rea ML, Di Stefano D, Minniti G, Esposito V, Bultrini A, Ferretti E, Santoro A, Faticanti Scucchi L, Gulino A, Cantore G (2002) A critical reappraisal of MIB-1 labelling index significance in a large series of pituitary tumours: secreting versus non-secreting adenomas. Endocr Relat Cancer 9: 103113 Gejman R, Swearingen B, Hedley-Whyte ET (2008) Role of Ki-67 proliferation index and p53 expression in predicting progression of pituitary adenomas. Hum Pathol 39: 758-766 Zhang X, Zhou Y, Mehta KR, Danila DC, Scolavino S, Johnson SR, Klibanski A (2003) A pituitary-derived MEG3 isoform functions as a growth suppressor in tumor cells. J Clin Endocrinol Metab 88: 51195126 Ezzat S (2004) Growth factors and their receptors in the genesis and treatment of thyroid cancer. Cancer Treat Res 122: 121-129 Daniel L, Trouillas J, Renaud W, Chevallier P, Gouvernet J, Rougon G, Figarella-Branger D (2000) Polysialylated-neural cell adhesion molecule expression in rat pituitary transplantable tumors

78.

79.

80.

81.

82.

83.

84.

85.

86.

87.

88.

89.

90.

(spontaneous mammotropic transplantable tumor in Wistar-Furth rats) is related to growth rate and malignancy. Cancer Res 60: 80-85 Kawamoto H, Kawamoto K, Mizoue T, Uozumi T, Arita K, Kurisu K (1996) Matrix metalloproteinase-9 secretion by human pituitary adenomas detected by cell immunoblot analysis. Acta Neurochir (Wien) 138: 1442-1448 Cornelius A, Cortet-Rudelli C, Assaker R, Kerdraon O, Gevaert MH, Prevot V, Lassalle P, Trouillas J, Delehedde M, Maurage CA (2012) Endothelial expression of endocan is strongly associated with tumor progression in pituitary adenoma. Brain Pathol 22: 757-764 Delhase M, Vergani P, Malur A, Velkeniers B, Teugels E, Trouillas J, Hooghe-Peters EL (1993) Pit-1/GHF-1 expression in pituitary adenomas: further analogy between human adenomas and rat SMtTW tumours. J Mol Endocrinol 11: 129-139 Vallette-Kasic S, Figarella-Branger D, Grino M, Pulichino AM, Dufour H, Grisoli F, Enjalbert A, Drouin J, Brue T (2003) Differential regulation of proopiomelanocortin and pituitaryrestricted transcription factor (TPIT), a new marker of normal and adenomatous human corticotrophs. J Clin Endocrinol Metab 88: 3050-3056 Asa SL, Bamberger AM, Cao B, Wong M, Parker KL, Ezzat S (1996) The transcription activator steroidogenic factor-1 is preferentially expressed in the human pituitary gonadotroph. J Clin Endocrinol Metab 81: 2165-2170 Chinezu L, Vasiljevic A, Jouanneau E, Francois P, Borda A, Trouillas J, Raverot G (2014) Expression of somatostatin receptors, SSTR2A and SSTR5, in 108 endocrine pituitary tumors using immunohistochemical detection with new specific monoclonal antibodies. Hum Pathol 45: 71-77 Galland F, Lacroix L, Saulnier P, Dessen P, Meduri G, Bernier M, Gaillard S, Guibourdenche J, Fournier T, Evain-Brion D, Bidart JM, Chanson P (2010) Differential gene expression profiles of invasive and non-invasive non-functioning pituitary adenomas based on microarray analysis. Endocr Relat Cancer 17: 361-371 Bates AS, Farrell WE, Bicknell EJ, McNicol AM, Talbot AJ, Broome JC, Perrett CW, Thakker RV, Clayton RN (1997) Allelic deletion in pituitary adenomas reflects aggressive biological activity and has potential value as a prognostic marker. J Clin Endocrinol Metab 82: 818-824 Beckers A, Aaltonen LA, Daly AF, Karhu A (2013) Familial isolated pituitary adenomas (FIPA) and the pituitary adenoma predisposition due to mutations in the aryl hydrocarbon receptor interacting protein (AIP) gene. Endocr Rev 34: 239-277 Cazabat L, Libe R, Perlemoine K, Rene-Corail F, Burnichon N, Gimenez-Roqueplo AP, Dupasquier-Fediaevsky L, Bertagna X, Clauser E, Chanson P, Bertherat J, Raffin-Sanson ML (2007) Germline inactivating mutations of the aryl hydrocarbon receptorinteracting protein gene in a large cohort of sporadic acromegaly: mutations are found in a subset of young patients with macroadenomas. Eur J Endocrinol 157: 1-8 Verges B, Boureille F, Goudet P, Murat A, Beckers A, Sassolas G, Cougard P, Chambe B, Montvernay C, Calender A (2002) Pituitary disease in MEN type 1 (MEN1): data from the France-Belgium MEN1 multicenter study. J Clin Endocrinol Metab 87: 457-465 Trouillas J, Labat-Moleur F, Sturm N, Kujas M, Heymann MF, Figarella-Branger D, Patey M, Mazucca M, Decullier E, Verges B, Chabre O, Calender A (2008) Pituitary tumors and hyperplasia in multiple endocrine neoplasia type 1 syndrome (MEN1): a casecontrol study in a series of 77 patients versus 2509 non-MEN1 patients. Am J Surg Pathol 32: 534-543 Zemmoura I, Wierinckx A, Vasiljevic A, Jan M, Trouillas J, Francois P (2013) Aggressive and malignant prolactin pituitary tumors: pathological diagnosis and patient management. Pituitary 16: 515-522

In search of a prognostic classification of endocrine pituitary tumors.

Pituitary tumors, the most frequent intracranial tumors, are historically considered benign. However, various pieces of clinical evidence and recent a...
259KB Sizes 0 Downloads 0 Views