pathophysiology of pituitary adenomas

download pathophysiology of pituitary adenomas

of 17

Transcript of pathophysiology of pituitary adenomas

  • 8/10/2019 pathophysiology of pituitary adenomas

    1/17

    1

    The pathophysiology of pituitary adenomas

    Dorota Dworakowska, MD, PhD,Doctor a,b, Ashley B. Grossman, MD, PhD,Professor of Neuroendocrinology a,*

    a Centre for Endocrinology, Barts and the London School of Medicine, EC1M 6BQ, UKb Department of Endocrinology and Internal Medicine, Medical University of Gdansk, 80-211, 7 Debinki Str., Poland

    Keywords:

    pathophysiology

    pituitary adenoma

    molecular alteration

    The pathogenesis of tumour formation in the anterior pituitary has

    been intensively studied, but the causative mechanisms involved

    in pituitary cell transformation and tumourigenesis remain elusive.

    Most pituitary tumours are sporadic, but some arise as a compo-

    nent of genetic syndromes such as the McCuneAlbright

    syndrome, multiple endocrine neoplasia type 1, Carney complex

    and, the most recently described, a MEN1-like phenotype (MEN4)

    and pituitary adenoma predisposition syndromes. Some specificgenes have been identified that predispose to pituitary neoplasia

    (GNAS, MEN1, PRKAR1A, CDKN1B and AIP), but these are rarely

    involved in the pathogenesis of sporadic tumours. Mutations of

    tumour suppressor genes or oncogenes, as seen in more common

    cancers, do not seem to play an important role in the great

    majority of pituitary adenomas. The pituitary tumour transforming

    gene (PTTG; securin) was the first transforming gene found to be

    highly expressed in pituitary tumour cells, and seems to play an

    important role in the process of oncogenesis. Many tumour

    suppressor genes, especially those involved in the regulation of the

    cell cycle, are under-expressed, most often by epigenetic modu-

    lation usually promoter hypermethylation but the regulator ofthese co-ordinated series of methylations is also unclear. Cell sig-

    nalling abnormalities have been identified in pituitary tumours,

    but their genetic basis is unknown. Both Raf/MEK/ERK and PI3K/

    Akt/mTOR pathways are over-expressed and/or over-activated in

    pituitary tumours: these pathways share a common root, including

    initial activation related to the tyrosine kinase receptor, and we

    * Corresponding author. Centre for Endocrinology, St Bartholomews Hospital, West Smithfield, London EC1A 7BE, UK. Tel.:

    44 20 76018343; Fax: 44 20 76018505.

    E-mail address: [email protected](A.B. Grossman).

    Contents lists available atScienceDirect

    Best Practice & Research Clinical

    Endocrinology & Metabolismj o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / lo c a t e / b e e m

    1521-690X/$ see front matter 2009 Elsevier Ltd. All rights reserved.

    doi:10.1016/j.beem.2009.05.004

    Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541

    mailto:[email protected]://www.sciencedirect.com/science/journal/1521690Xhttp://www.elsevier.com/locate/beemhttp://www.elsevier.com/locate/beemhttp://www.sciencedirect.com/science/journal/1521690Xmailto:[email protected]
  • 8/10/2019 pathophysiology of pituitary adenomas

    2/17

    speculate that a change to these receptors or their relationship to

    membrane matrix-related proteins may be an early event in

    pituitary tumourigenesis.

    2009 Elsevier Ltd. All rights reserved.

    Pituitary adenomas are the third most common intracranial tumours in surgical practice,

    accounting for approximately 1025% of intracranial neoplasms.1 Pituitary tumours are frequently seen

    in autopsy specimens (14%).2 Radiological series suggest that pituitary incidentalomas may be present

    in one in six people. Furthermore, recent epidemiological data suggest that clinically apparent pituitary

    adenomas have a prevalence of approximately one in 1000 in the general population.3

    Although only very rarely malignant, pituitary tumours may cause significant morbidity in affected

    patients. First, given the critical location of the gland, large tumours may lead to mass effects, and,

    second, proliferation of hormone-secreting pituitary cells leads to endocrine syndromes. Approxi-

    mately 50% of newly diagnosed pituitary adenomas are prolactinomas (PRL-omas). Endocrinologically

    inactive adenomas non-functioning pituitary adenomas (NFPAs) represent about 30%, somatotrophadenomas (GH-omas) 1520%, corticotroph adenomas (ACTH-omas) 510% and thyrotroph adenomas

    (TSH-omas) less than 1%.4,5 True gonadotroph-secreting pituitary adenomas resulting in clinical

    syndromes are extremely rare, but it seems likely that the great majority, if not all, NFPAs are in fact

    very-low-grade FSH/LH-omas. Pituitary carcinomas comprise around 0.2% of pituitary adenomas1,

    distinct from thew1% of pituitary mass lesions, which are metastatic tumours from non-pituitary sites.

    The initiating molecularaberrations in the great majority of sporadicpituitarytumours remain elusive.6

    The majority of pituitary adenomas are sporadic, although some arise as a component of familial

    syndromes. Due to clonal analysis (which allows one to make the important distinction between a poly-

    clonal proliferation in response to a stimulatory factor versus a monoclonal expansion of a genetically

    aberrant cell), it has been demonstrated that almost all hormone-secreting as well as non-functioning

    pituitary adenomas are unicellular in origin, arising from a monoclonal expansion of a genetically mutatedcell.68 Pituitary adenomas have attracted considerable attention because they can act as a model system

    for more aggressive cancers. The aim of this review is to summarise recent advances in pituitary tumour

    pathophysiology rather than to provide exhaustive information about all molecular abnormalities seen in

    these tumours, to which several recent reviews have been dedicated.5,914

    Genetic/familial syndromes

    Pituitary adenomas may be a manifestation of an underlying germ line syndrome; such disorders

    include the McCuneAlbright syndrome (MAS), multiple endocrine neoplasia type 1 (MEN1), Carney

    complex (CNC) and, most recently, the MEN1-like phenotype and pituitary adenoma predisposition

    (PAP) syndromes.11,13

    The genes for GNAS (at 20q13), menin (at 11q13) and protein kinase A regulatorysubunit-1-a (PRKAR1A, at 17q24) have been associated with MAS, MEN1 and CNC, respectively. The

    cyclin-dependent kinase inhibitor 1B (CDKN1B, which codes for p27, at 12p13) and aryl hydrocarbon

    receptor (AHR)-interacting protein (AIP, also at 11q13, but distinct from menin)15,16 are associated with

    an MEN1-like phenotype and PAP, respectively. CDKN1B was identified from a rat phenotype referred

    to as MENX, but still only accounts for a fraction of human MEN1-like syndromes, which are MEN1-

    gene negative.17 Indeed, the syndrome, now designated MEN4 by OMIM, seems less often than

    originally thought to include pituitary adenomas. AIP is mainly mutated in the subset of families with

    familial isolated pituitary adenomas, which have familial acromegaly and prolactinomas, isolated

    familial somatotropinomas (IFSs)18, but even in this syndrome the majority of patients do not show

    a mutation in AIP, and other genes are almost certainly involved.19

    The McCuneAlbright syndrome (MAS) is caused by mosaicism for a mutation in the GNASoncogene. The GNAS1 mutation is the only mutation that has been identified in a significant proportion

    of sporadic pituitary tumours, occurring in approximately 3040% of GH-omas. However, in spite of

    extensive work, there is little direct evidence that the gsp mutation plays an essential primary onco-

    genic role in such tumours, or that it alters tumour growth or recurrence rate.20,21

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541526

  • 8/10/2019 pathophysiology of pituitary adenomas

    3/17

    Multiple endocrine neoplasia type 1 (MEN1) is familial disorder with a mutation in the MEN1gene

    located on chromosome 11q13, inherited as an autosomal-dominant trait.22 Even though allelic

    deletions on 11q13 are often observed in sporadic tumours23, somatic MEN1 mutations or loss of

    expression24 are very rare in sporadic pituitary tumours25, and the evidence to date suggests that

    mutation or loss of this gene plays only a limited role in sporadic tumours.

    Up to 1025% of patients with MEN1 have normal MEN1 genes26,27, implicating other predispositiongenes in this phenotype. A germ line nonsense mutation in the human cyclin-dependent kinase

    inhibitor 1B (CDKN1B, also known as p27 and KIP1) gene was recently identified in a family with an

    MEN1-like condition15; however, the occurrence of this mutation seems to be very rare in such

    families.17 No mutations in the p27 gene have been found in sporadic pituitary tumours.28 As discussed

    below, p27 negatively regulates cell cycle progression by inhibiting cyclin and cyclin-dependent kinase

    complexes, with p27 protein showing under-expression in most human pituitary tumours. 2931 More

    recent studies indicate that p27 is an important down-stream signal in the menin signalling pathway

    and a possible target of oncogenic RET in endocrine cells.32 It is also a direct transcriptional target of

    AHR, the partner of AIP, which is known to affect cell proliferation.33

    Carney complex (CNC) is an inherited neoplasia syndrome characterised by spotty skin pigmen-

    tation, myxomas, endocrine tumours and Schwannomas. Some 40% of CNCs are associated withinactivating mutations in the gene encoding for the protein kinase A (PKA) type 1A regulatory (R1a)

    subunit (PRKAR1A).34 PRKAR1A, like menin, acts as a tumour suppressor gene in affected tissues; loss

    of its normal allele at chromosomal region 17q2224 is present in pituitary tumours associated with

    CNC35, but it is normally expressed and unmutated in almost all sporadic somatotroph tumours. 36,37

    Familial isolated pituitary adenoma (FIPA) is a rare syndrome that embraces IFS as well as the PAP

    syndrome. The most frequently described forms of FIPA are familial somatotropinomas or prolacti-

    nomas; however, pituitary adenomas of all types can occur in a familial setting. FIPA differs from MEN1

    in terms of a lower proportion of prolactinomas and more frequent somatotropinomas. Patients with

    FIPA are significantly younger at diagnosis38 and have significantly larger pituitary adenomas than

    matched sporadic pituitary adenoma counterparts.12,39 PAP constitutes the group of patients with FIPA,

    especially those with IFS, who show germ line mutations in AIP. A linkage in patients with IFS tochromosome 11q13.111q13.3 was first established by Gadelha et al.40, and it is now known that many

    such patients show a mutation in AIP.

    In a recent series from our group, about 66% of patients with AIP mutations showed pituitary

    adenomas.41 Mutations to the AIP gene also resulted in unchanged cell proliferation when inserted into

    GH3 pituitary cell lines, HEK293 cell lines and TIG3 cell lines (compared with markedly reduced cell

    proliferation in cell lines engineered to over-express AIP).41 AIP thus appears to be a tumour suppressor

    gene, and mutations in this gene are involved at least in some patients with IFS. On the other hand,

    approximately 85% of the FIPA cohort and 50% of those with familial somatotropinomas were negative

    for AIP mutations.39 Furthermore, AIP can be ruled out as a candidate gene for involvement in the great

    majority of sporadic pituitary adenomas, as it is extremely rarely mutated and, if anything, is over-

    expressed (possibly as a compensatory phenomenon) in all tumour types.41

    Others tumour suppressor genes, oncogenes, growth factors and cell cycle regulators

    Molecular studies on pituitary tumours have shown that alterations in the best-known tumour

    suppressor genes in other neoplasms (e.g.,P53 andRB) or common oncogenes (e.g., theRas-family) are

    only rarely involved in the development of these tumours.4245 Although point mutations of the Ras

    Practice points and research agenda

    Current studies have greatly increased our knowledge of the genetic basis of MAS, MEN1, CNC

    and familial pituitary adenoma syndromes, but unfortunately they have done little to elucidate

    the causes of sporadic pituitary tumours.

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541 527

  • 8/10/2019 pathophysiology of pituitary adenomas

    4/17

    oncogene43, loss of heterozygosity (LOH) near the RB locus on chromosome 1346,47 and LOH on

    chromosome 1144,45 have all been implicated in some pituitary tumours, these are clearly in the

    minority. For instance, a mutation of the H-Ras gene (codon 12, Gly to Val) was found in recurrent,

    highly invasive prolactinomas.43 A recent study showed the presence of P53 gene mutations in

    pituitary carcinomas but confirmed their absence in pituitary adenomas (in carcinomas, a P53gene

    mutation was additionally related to p53 protein over-expression in tumour cells, which may bediagnostically helpful).48

    Several other genetic alterations have been implicated in pituitary tumourigenesis: increased

    expression of pituitary tumour transforming gene (PTTG), alternative transcription initiation of pitu-

    itary-tumour-derived fibroblast growth factor receptor 4 (Pdt-FGFR4) or promoter methylation of

    tumour suppressor genes (e.g., RB). Growth arrest and DNA damage-inducible protein gamma

    (GADD45G), maternally expressed protein 3A (MEG3A), the zinc-finger protein pleiomorphic adenoma

    gene-like 1 (ZAC), death-associated protein kinase (DAP kinase), pituitary tumour apoptosis gene

    (PTAG) and p27 (Kip 1) gene have all been found to be abnormally expressed in pituitary adenomas and

    thus may play a possible role in pituitary tumourigenesis.14,4952

    The novel PTTG was identified and cloned in rat GH4 pituitary tumour cells.53 PTTG over-expression

    occurs in a wide variety of endocrine and non-endocrine tumours, including those of the pituitary,thyroid, ovary, breast, prostate, lung, esophagus, colon and the central nervous system.54 By acting as

    a securin protein and inhibiting premature sister chromatid separation, PTTG plays an important role in

    mitosis. Cohesin binds sister chromatids during metaphase, and is subsequently degraded by separin to

    allow sister chromatid separation at anaphase (Fig. 1).55 It has been shown that PTTG blocks sister

    chromatid separation during metaphase by binding to separin and preventing cohesin degradation.56

    PTTG induces cell transformation when over-expressed in vitro in NIH 3T3 fibroblasts, and tumour

    formation in nude mice. It stimulates basic fibroblast growth factor (bFGF), which is a potent mitogenic

    and angiogenic factor,in vitro49, and interacts with theP53tumour suppressor gene andc-mycproto-

    oncogene.54 In addition, phosphoinositol-3-kinase (PI3K) and MAPK both physically interact with

    Fig. 1. Molecular mechanism of chromosome segregation. At the metaphaseanaphase transition, APC/CCdc20 ubiquitinates securin

    (PTTG). Degradation of securin activates separase. Separase then cleaves the Scc1 subunit of cohesin, allowing chromosome

    segregation. In response to sister-chromatids not being properly attached to the mitotic spindle, the spindle checkpoint promotes

    the assembly of checkpoint protein complexes that inhibit the activity of APC/C, leading to the stabilisation of securin, preservation

    of sister-chromatid cohesion, and a delay in the onset of anaphase. 63

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541528

  • 8/10/2019 pathophysiology of pituitary adenomas

    5/17

    PTTG, thus suggesting that PTTG phosphorylation occurs through these two cascades.57,58 Interaction

    of PTTG with Sp1, an important transcription factor involved in the regulation of several genes

    associated with cellular growth and differentiation, is well documented.54 PTTG and Sp1 were found to

    cooperatively mediate G1/S-phase transition59, and recently it was shown that PTTG binds to the p21

    promoter region through Sp1, thereby suppressing p21activity.60 Conversely, knock-out of PTTG causes

    decreased cellular proliferation and premature cellular senescence.61 Thus, PTTG acts at the centre ofa network of processes that control cell proliferation and division, and both up- and down-regulation

    are inimical to regulated cell growth. However, specific mutation of PTTG has not been shown in

    sporadic pituitary adenomas.

    Changes in PTTG expression may relate to pituitary tumour angiogenesis as well as to tumour

    invasiveness and aggressiveness.49,54,62 PTTG expression was found to be correlated with Ki-67

    expression in pituitary adenomas.62 In addition, in pituitary adenomas, a significant positive correla-

    tion was found between VEGF and PTTG mRNA expression, as well as between PTTG and KDR (kinase

    insert domain receptor, one of the family of VEGF receptors) mRNA 63,64:in situhybridisation revealed

    PTTG expression in NFPAs and in GH-secreting adenomas but not in normal pituitary tissue.49

    Immunohistochemistry showed lack of PTTG expression in normal pituitary tissue and a very high rate

    of PTTG positivity in different types of pituitary tumours (approximately 90% of tumours showed PTTGexpression, but with considerable variability).62 More sensitive techniques based on reverse

    transcriptase polymer chain reaction (RT-PCR) showed that PTTG expression was present in normal

    pituitaries, but at a much lower level than in pituitary adenomas. PTTG was mostly over-expressed in

    adenomas with a >50% increase in NFPA, GH-, PRL- and ACTH-omas. In some tumours, more than

    a 10-fold increase in PTTG expression was observed, suggesting that PTTG is indeed involved in some

    manner in pituitary tumourigenesis.49 It has not yet been clarified whether PTTG functions as

    a cytoplasmic or a nuclear protein. In pituitaryadenomas, some studies showed that PTTG might be

    predominantly expressed in the cytoplasm64,65, but another showed PTTG to be expressed predomi-

    nantly in the nucleus.62 PTTG was found to be a secretory protein in human pituitary adenomas and in

    mouse pituitary tumour cell lines, and may exert autocrine and/or paracrine functions.66 In hormone-

    secreting tumours, over-expression of PTTG correlated with tumour invasiveness, since higher PTTGexpression was observed in tumours that had invaded the sphenoid bone (stages III and IV; 95%

    confidence interval (CI): 3.1189.715) compared with tumours that were confined to the pituitary fossa

    (stages I and II; 95% CI: 1.6813.051).49

    Fibroblast growth factors (FGFs) and fibroblast growth factor receptors (FGFRs) are known to be

    important for a variety of biological processes, including mitogenesis, differentiation, development,

    angiogenesis and tumourigenesis.67 The FGFs mediate their biological effects by binding to high-

    affinity tyrosine kinase receptors, the FGFRs.68 FGF-2 (also known as basic or bFGF) is over-expressed

    by pituitary tumour cells with higher levels in more aggressive tumours.69 The role of the pituitary

    tumour-derived FGFR-4 isoform (ptd-FGFR4), a constitutively phosphorylated protein with trans-

    forming properties in vitro and in vivo70, has been recently investigated in sporadic pituitary adenomas.

    Cytoplasmic expression of ptd-FGFR4 was found in around 60% of cases (containing GH-, ACTH-, FSH/LH- and NFPAs) but was relatively rare in PRL-omas and absent in normal adenohypophysial tissue.

    Expression of ptd-FGFR4 was stronger in macroadenomas in comparison to microadenomas, and

    correlated with cell proliferation assessed by Ki-67.71 Up-stream regulators, particularly transcription

    factors of theicarusgroup, controlling this aberrant splice variant are under study, and, if confirmed in

    other laboratories, could signify a novel pathway regulating proliferation.

    The cell cycle

    Cell proliferation is dependent on a highly organised and temporally linked series of kinases, which

    rise and fall with each cell division. In the cell cycle the cyclins are phosphorylated (by cyclin-

    dependent kinases or CDKs) and then degraded in sequence, leading to DNA synthesis, chromosomalseparation and mitosis. We have already emphasised the role of PTTG, or securin, in this process. In

    addition, so important is the regulation of this process and its exquisite co-ordination, that a series of

    CDK inhibitors (CDKIs) are also involved. Most importantly, the step from cyclin D/CDKs 4/6 to cyclin E/

    CDK2 is precisely tuned by a series of CDKIs known as the Ink4 family (p15, p16, p18, for CDK 4 and 6)

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541 529

  • 8/10/2019 pathophysiology of pituitary adenomas

    6/17

    and the kip/cip family (p21, p27, p57 for CDK2). A cell passing through this checkpoint phosphorylates

    Rb, leading to dissociation from E2F and induction of pro-proliferative capacity (Fig. 2).72 It then

    becomes irreversibly committed to cell division; thus, disruption at this level is of fundamental

    importance. It is of considerable interest that transgenic knockouts of many of these CDKIs will usually

    greatly increase the probability of pituitary tumour formation. In rodent transformed pituitary cell

    lines, p16 was shown to be under-expressed by promoter hypermethylation in the mouse AtT20 lineand p27 under-expressed by a similar process in the rat GH3 line. In the human, the vast majority of

    adenomas display alterations of the RB1 pathway (90%); specifically, promoter hypermethylation of the

    p15(INK4b), p16(INK4a) and RB1 genes was detected in 3236%, 5971% and 2935% of cases,

    respectively.73,74 Yoshino et al. additionally demonstrated promoter hypermethylation of p14 (ARF),

    p21(Waf1/Cip1) and p73 genes in 6%, 3% and 12% of adenomas, respectively, whereas the promoter of

    p27(Kip1) was not hypermethylated.74 However, our group and that of Ricardo Lloyd reported under-

    expression of p27 at the protein level in all types of pituitary adenomas, especially corticotroph

    tumours.29,30 Thus, dysregulation of the cell cycle appears to be a hallmark of pituitary tumour

    formation, occurring both by CDKI promoter methylation as well as by enhanced degradation.

    Inappropriate methylation of CpG islands of other key cell cycle control and growth-regulatory

    genes has been demonstrated: such genes include the growth arrest and DNA damage-inducibleprotein gamma (GADD45G)75, and death-associated protein kinase (DAP kinase).76 GADD45G nega-

    tively regulates cell growth and is significantly under-expressed in GH-secreting and PRL-secreting

    pituitary tumours.50 The DAP kinase gene encodes for a calmodulin-dependent serine/threonine

    kinase, which positively mediates programmed cell death: loss of DAP kinase expression was

    demonstrated in highly metastatic cells, whilst re-expression of the protein resulted in delayed tumour

    Fig. 2. Cell cycle progression. In early G1, cyclin D activates CDK4/6 which partially phosphorylates Rb protein. After progression

    beyond the restriction point, CDK2 in complex with cyclin E further phosphorylates pRb rendering it inactive and thus allowing

    transcription of cyclin A. The accumulating cyclin A captures CDK2 from cyclin E and promotes progression through S phase. At the

    transition between G2-M phase, cyclin B complexes with and activates CDK1 to further activate metaphase promoting complex

    (MPC) required for entering M phase. CDK inhibitors oppose CDK activation: in early G1, the INK4 family bind to CDK4/6 and prevent

    cyclin D activation of this kinase. At G1-S transition, WAF/KIP family members sequester CDK2 away from cyclin E/A when they are

    in stoichiometric amounts with CDK2. When CDK2 exceeds p27 levels, the former phosphorylates the latter and triggers its

    degradation. Resetting of pRb occurs in M-phase by dephosphorylation, probably by protein-phosphatase type 1 and this correlates

    with the time of cyclin A degradation.80

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541530

  • 8/10/2019 pathophysiology of pituitary adenomas

    7/17

    growth and a decreased incidence of metastasis. Loss of DAP kinase expression (associated with either

    CpG island methylation or homozygous deletion) preferentially segregates with pituitary tumours that

    show an invasive phenotype.76

    A novel differentially methylated chromosome 22 CpG island-associated gene (C22orf3), also

    known as pituitary tumour apoptosis gene (PTAG), has been recently identified. 52 The majority of

    pituitary adenomas (80%) failed to express this gene in comparison to normal pituitary, and in thisgroup approximately 20% showed methylation of the CpG islands of the PTAG promoter.52 Where the

    promoter was methylated, it was invariably associated with loss of transcript expression.52 In a model

    pituitary tumour cell line, AtT20, expression of PTAG per sehad no discernible effects on proliferation,

    cell cycle profile or viability. However, enforced expression was associated with a significantly

    increased sensitivity to the apoptotic effects induced by the bromocriptine challenge.77

    Other genes with suggested role in pituitary tumourigenesis on the basis of their promoter

    methylation include maternally expressed protein 3A (MEG3A)78 and the ZAC.79 MEG3 is highly

    expressed in the normal human pituitary, but is not detected in human NFPAs. Two functionally

    important 50- flanking regions, immediately preceding and approximately 1.62.1 kb up-stream of the

    first exon, respectively, were found to be hypermethylated in tumours without MEG3 expression.78 The

    ZAC gene encodes a novel zinc-finger protein that concomitantly induces apoptosis and cell cyclearrest; it localises to chromosome 6q24q25, a well-known hot spot related to cancer. ZAC is highly

    expressed in the anterior pituitary gland, and its ablation by antisense targeting promotes pituitary cell

    proliferation. Strong down-regulation of ZAC mRNA and protein expression was reported in NFPAs 79,

    although this was not associated with gene mutations.

    We have reviewed the evidence for disruption of the cell cycle in pituitary adenomas, usually by

    promoter hypermethylation of CDKIs. However, in the case of p27, this is due to a decrease in p27

    protein levels,while p27 mRNA levels are normal or even increased, and somatic mutations have not

    been reported.11,30 Knockout mice lacking p27 have been shown to develop multi-organ hyperplasia

    and intermediate lobe pituitary tumours secreting ACTH. The expression of p27 is reduced in pituitary

    adenomas (including ACTH-, PRL-, GH-, LH/FSH- and TSH-adenomas), compared with the normal cells,

    and completely lost in metastatic pituitary carcinomas.30

    Importantly, p27 expression is inverselyrelated to Ki-67.80 Phosphorylation of p27 is an essential step for its degradation, and this has been

    shown to be higher in pituitary adenomas than in the normal pituitary, leading to the cytoplasmic

    sequestration and reduced nuclear expression of p27.80 Decreased levels of p27 particularly lead to

    increased levels of cyclin E, and this has been reported in corticotroph adenomas. 80,81

    RNA microarrays and proteomics of pituitary adenomas

    Microarray technology allows for the expression profile of many thousands of genes to be quantified

    at the same time and has resulted in novel discoveries regarding the tumour biology of

    pituitary adenomas. A study by Ruebel at al. underlined how major are the differences between

    Practice points and research agenda

    Recent studies have highlighted multiple molecular alterations in pituitary adenomas, espe-

    cially involving the cell cycle. Many of the changes seem to reflect epigenetic mechanisms

    associated with gene silencing, particularly of cell cycle inhibitors. PTTG is a gene that is

    importantly over-expressed, which may lead to enhanced proliferation and chromosomal de-

    stabilisation. The isolation of novel genes in pituitary adenomas (e.g., PTTG, Pdt-FGFR4, GADD45G,

    MEG3A, ZAC, DAP kinase and PTAG) offers a significant advantage with respect to our under-

    standing of tumourigenesis in pituitary tumours. The reversal of apparent gene silencing may

    eventually lead to tumour cell sensitisation to chemo- and radiotherapeutic treatment strategies.

    A summary of the reported changes in expression in pituitary tumours is shown in Table 1.

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541 531

  • 8/10/2019 pathophysiology of pituitary adenomas

    8/17

    Table 1

    A summary of gene inactivation studies due to promoter hypermethylation in pituitary adenomas.

    Gene Remarks Ref.

    p15(INK4b)

    p16(INK4a)RB1

    91% of the adenomas displayed alterations of the RB1 pathway.

    Promoter hypermethylation of the p15(INK4b), p16(INK4a), and RB1 genes wasdetected in 15 (35.7%), 30 (71.4%), and 12 (28.6%) of the adenomas, respectively.

    Promoter hypermethylation of the p15(INK4b) gene coincided with a p16(INK4a)

    alteration and/or RB1 methylation, whereas p16(INK4a) and RB1 methylations tended

    to be mutually exclusive.

    73

    RB1, p14(ARF)

    p15(INK4b)p16(INK4a)

    p21(Waf1/Cip1)

    p27(Kip1) p73

    Promoter hypermethylation of the RB1, p14(ARF), p15(INK4b), p16(INK4a), p21(Waf1/

    Cip1), p27(Kip1), and p73 genes was detected in 12 (35%), 2 (6%), 11 (32%), 20 (59%), 1

    (3%), 0 (0%), and 4 (12%) of the adenomas, respectively. In total, 88% (30 of 34) of the

    adenomas displayed methylation of at least one of such cell cycle regulatory genes,

    especially methylation of the member genes of the RB1 pathway (29 of 34; 85%).

    Promoter hypermethylation of p15(INK4b) coincided with RB1 and/or p16(INK4a)

    methylation, whereas RB1 and p16(INK4a) methylations tended to be mutually exclu-

    sive (p 0.005).

    Promoter hypermethylations of p14(ARF), p21(Waf1/Cip1), and p73 (not belonging tothe member genes of the RB1 pathway) were coincident with RB1 and/or p16(INK4a)

    methylation except in one p73 methylated case.

    74

    RB1 Methylation of the CpG island within the RB1 promoter region was detected in 6 of 10

    tumors that failed to express pRb.

    18 of 20 tumors and all six histologically normal postmortem pituitaries that expressed

    pRb were unmethylated.

    131

    GADD45G 67% of pituitary adenomas (22/33) do not express GADD45G as determined by RT-PCR

    analysis.

    Loss of expression was not associated with either LOH or mutations within the coding

    region of this gene.

    Methylation of the GADD45G genes CpG island, was found in 19 of 33 adenomas (58%)

    and was significantly associated with tumours in which GADD45G transcript was notexpressed (18 of 22; 82%; P 0.002).

    75

    DAP kinase 34% (11/32) of tumours had undetectable DAP kinase expression, by Western blot and/

    or RT-PCR analysis.

    Of 11 tumours that failed to express DAP kinase, five (45%) showedde novomethylation

    of the CpG island contained within the promoter region, while four (36%) had evidence

    of homozygous deletion of this region.

    Loss of DAP kinase expression was significantly (P

  • 8/10/2019 pathophysiology of pituitary adenomas

    9/17

    pituitary adenomas and carcinomas, indicating 4298 genes that were differentially expressed among

    the adenomas compared to the carcinoma, with 2057 genes over-expressed and 2241 genes under-

    expressed in the adenomas.82 In particular, they showed that the b-galactoside binding protein

    galectin-3, the human achaetescute homologue-1 ASCL1 (hASH-1) gene and ID2 (which plays an

    important role in cell development and tumourigenesis), were under-expressed in adenomas

    compared to the carcinomas.82

    A pioneering complementary DNA microarray study indicated that 128 of 7075 genes examined

    were differentially expressed in pituitary adenomas compared to normal pituitaries. Further RT-real

    time quantitative PCR (RT-qPCR) examination established that at least three genes involved in

    carcinogenesis in other tissues were also aberrantly regulated in the major types of pituitary

    tumours, including the folate receptor gene (over-expressed in NFPAs and under-expressed in PRL-

    and GH-adenomas), the ornithine decarboxylase gene (over-expressed in GH-omas adenomas and

    under-expressed in ACTH-omas), and the C-mer proto-oncogene tyrosine kinase gene (over-

    expressed in ACTH-omas and under-expressed in PRL-omas).83 The most recent microarray analysis

    performed by the Evans group focussed on the molecular pathogenesis of human prolactinomas

    (based on gene expression profiling, RT-qPCR, and proteomic analyses) and identified 726 unique

    genes that were statistically significantly different between prolactinomas and normal glands,whereas proteomic analysis identified four differentially up-regulated and 19 down-regulated

    proteins.84

    b-Catenin is a major component of the Wingless (Wnt) signalling pathway and an important factor

    in pituitary development. Its activation regulates the functioning of the somatotroph, mammotroph,

    thyrotroph and gonadotroph pituitary axes, and it regulates Pit-1 expression/transcription. Recent

    studies have explored the possible involvement ofb-catenin signaling in pituitary tumour develop-

    ment.9 The data are sometimes discordant and contradictory, although there is indeed evidence that

    there are alterations in Wnt/b-catenin signalling in these benign neoplasms, leading to aberrant

    activation and transcription of specific target genes.9 In one study based on microarray analysis and

    proteomics, several changes in the expression of genes involved in pituitary development were found

    in NFPAs, including up-regulation of Pitx2, Pitx1, SFRP1 (secreted frizzled related protein 1) and TLE2(transducin-like enhancer of split 2, which not only represses targets ofb-catenin, but also interacts

    with Hes1 during neuronal development).85 The same group has recently shown that several

    components of the Notch pathway were altered in prolactinomas, and there was an increased

    expression of the Pit-1 transcription factor, and the survival factor BAG1, but decreased E-cadherin and

    N-cadherin expression in comparison to normal pituitaries.84 An Australian group has shown by

    microarray profiling and real-time quantitative RT-PCR that the mRNA expression of a series of

    inhibitors of the Wnt pathway (WIF1, SFRP2, frizzled B SFRP3 (FZDB), SFRP4) were all down-regu-

    lated in pituitary tumours compared to normal pituitaries.86

    A recent microarray study performed by our group showed that the expression of some genes was

    altered in particular subtypes of pituitary adenomas. For instance, lysosomal-associated protein

    transmembrane-4-b (LAPTM4B), a novel gene up-regulated in hepatocellular carcinoma, was signifi-cantly over-expressed in ACTH-omas and NFPAs. Bcl-2-associated athanogene (BAG1), an anti-

    apoptotic protein found at high levels in a number of human cancers, was significantly over-expressed

    in GH- and PRL-omas as well as in NFPAs. Furthermore, the cyclin-dependent kinase inhibitor p18,

    which was shown on murine gene deletion to induce pituitary ACTH cell hyperplasia and adenomas,

    was significantly under-expressed in ACTH-secreting adenomas.87 However, it should be noted that

    quantitative variations in gene expression between normal pituitary and human pituitary adenoma

    biopsy samples require careful interpretation, as the expression profiles of highly uniform cell pop-

    ulations in human pituitary adenomas are necessarily different from the composite derived from the

    complex admixture of different cell types that constitute the normal pituitary.5

    There are even fewer studies on protein arrays in pituitary tumours. One study from our group

    detected 316 different proteins present in pituitary tissue, of which 116 had not previously beendescribed in the human pituitary. In protein array analysis, heat-shock protein 110 (HSP110) and B2

    bradykinin receptor were significantly over-expressed in all adenoma subtypes, while C-terminal src

    kinase (CSK) and annexin II were significantly under-expressed in all adenoma subtypes. The immu-

    nohistochemical analysis confirmed the over-expression of HSP110 and B2 bradykinin receptor and

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541 533

  • 8/10/2019 pathophysiology of pituitary adenomas

    10/17

    under-expression of CSK and annexin II in pituitary adenoma cells when compared to their corre-

    sponding normal pituitary cells, but results were more variable using standard Western blotting. 88 In

    NFPAs, a proteomics and transcriptomics study demonstrated secretagogin down-regulation at both

    protein and mRNA levels.89

    A new phosphoproteomics study showed 50 phosphorylation sites characterised in 26 proteins90,

    and post-translational modifications of proteins from the human pituitary gland play an important rolein the regulation of different pituitary functions. The proteome database of the human pituitary can be

    accessed at the website http://www.utmem.edu/proteomics.

    Pituitary-specific signalling pathways

    Since deregulation of normal feedback processes seems to occur in GH- and ACTH- secreting

    tumours, abnormalities in pituitary-specific signalling pathways have been studied.

    In Cushings disease, there are several possible sites of derangement of corticotroph signalling.

    The ACTH receptor (ACTH-R), the second member of the melanocortin (MC-2) receptor family that

    includes five seven-transmembrane G protein-coupled receptors, has been shown to be predomi-

    nantly expressed in the adrenal cortex. It has been postulated that ACTH may regulate its own

    secretion through ultra-short-loop feedback within the pituitary. In the study performed by ourgroup, we identified ACTH-R mRNA expression in normal pituitaries, but not in 16/22 ACTH-

    adenomas. In addition, we did not find any ACTH-R gene mutation in a subgroup analysis of eight of

    these tumours. Interestingly, diagnostic preoperative plasma ACTH levels were significantly lower in

    the ACTH-R positive ACTH-omas, compared with those who were ACTH-R negative. In the absence of

    any gene defects, we concluded that the absence of ACTH-R expression in Cushings disease is

    probably a consequence of the tumoural process rather than a cause, but may aid in tumour

    progression.91

    The ACTH-secreting tumours are at least partially glucocorticoid resistant, but no mutations in the

    glucocorticoid receptor (GR) were found in any of the tumours. A majority of the ACTH-omas tested

    (16 out of 19) showed variable but increased overall GR expression, and thus these findings could

    not account for the dexamethasone resistance characteristic of these tumours.92

    Over-expression ofCRH-R has been shown in corticotroph tumours in comparison to normal pituitaries; interestingly, in

    ACTH-secreting bronchial carcinoids, CRH-R signals were detected in only four of the six tumours.93

    The CRH-R does not appear to be constitutively activated by mutation.94

    On the other hand, vasopressin is an important regulator of hypothalamicpituitaryadrenal axis

    activation, primarily acting through the V3 receptor (V3R). Many patients with ACTH-secreting

    pituitary adenomas, but not normal individuals, respond to desmopressin, a relatively V2-specific

    vasopressin agonist, with increased ACTH and cortisol levels. No mutation of the V3R gene was found in

    12 ACTH-omas; the V2R gene was expressed in the majority of the samples tested, while V3R was

    expressed in all of these tumours.95

    Derangement has been demonstrated in ACTH-omas in the two isoenzymes 11b-hydroxysteroid

    dehydrogenase (11b-HSD) 1 and 2, which convert cortisone to cortisol, and cortisol to cortisone,respectively. These enzymes may play an important pre-receptor role in regulating corticosteroid

    hormone action. The 11b-HSD2 is up-regulated and 11b-HSD1 is down-regulated in some pituitary

    tumours, resulting in increased cortisone and decreased cortisol production. Decreased cortisol in

    the region of the GR could diminish functional feedback and allow selective secretion of

    Practice points and research agenda

    The techniques of modern genetics, genomics and molecular biological approaches may be

    expected to reveal new mechanisms of endocrine tumourigenesis aiming at development of

    better diagnostic, prognostic and therapeutic tools. However, current results have provided much

    information but a less impressive increase in understanding.

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541534

    http://www.utmem.edu/proteomicshttp://www.utmem.edu/proteomics
  • 8/10/2019 pathophysiology of pituitary adenomas

    11/17

    corticotrophs essentially re-setting glucocorticoid feedback control.96 However, these changes may

    be a general feature of all pituitary adenomas so their functional significance in corticotroph

    tumours remains unclear.97

    An additional molecular basis for glucocorticoid resistance in pituitary adenomas is the observation

    that approximately 50% of tumours are deficient in the expression of Brg1 (the ATPase subunit of the

    Swi/Snf complex) or histone deacetylase-2 (HDAC2).98 These two proteins (Brg1 and HDAC2) areinvolved in chromatin re-modelling and may also participate in the tumourigenic process, as Brg1 is

    a tumour suppressor.99 The GR-dependent repression of the pro-opiomelanocortin (POMC) gene,

    which is at the centre of the hypothalamicpituitaryadrenal axis that controls glucocorticoid

    synthesis, involves orphan nuclear receptors related to NGFI-B: this repression of POMC requires Brg1

    to stabilise interactions between GR and NGFI-B. Brg1 also stabilises the interaction between GR and

    HDAC2, which results in de-acetylation of the POMC promoter region and thus repression. Loss of

    either HDAC2 or Brg1, therefore, produces glucocorticoid resistance as seen in Cushings disease.98

    In acromegaly, there is less information available than for Cushings disease. However, GHRH receptor

    variants were found to be common in GH-producing pituitary adenomas, but constitutively activating

    mutations, as a mechanism for GH-omas, appear to be rare.100,101 The type 1 insulin-like growth factor

    (IGF1)/GH negative feedback loop is one of the several factors that regulate GH secretion. It has beenpostulated that this is a result of changes at the GH-receptor (GH-R) level. However, it seems that

    decreased feedback inhibition of GH because of somatic mutations of the coding region of the GH-R is

    unlikely to be a common factor in the pathogenesis of GH-omas.102 Nevertheless, the decreased

    expression of the GH-R and of IGF-R in 18 somatotroph tumours (both at the mRNA and protein level)

    may, at least in part, help explain the continuous secretion of GH from the tumour despite the high

    circulating levels of IGF-I and GH.102 Recently, a selective somatic histidine-to-leucine substitution in

    codon 49 of the extracellular domain of the GH receptor has been described in a morphologic subtype of

    human GH-producingpituitary tumoursthatis characterised by the presenceof cytoskeletal aggresomes.

    ThisGH-R mutation significantlyimpairs glycosylation-mediatedreceptor processing,maturation, ligand

    binding and signalling, and provides evidence for impaired hormone auto-feedback in the pathogenesis

    of these pituitary tumours. It might explain the lack of responsiveness to somatostatin analogue therapyof this tumour type, in contrast to the exquisite sensitivity of tumours that lack aggresomes, and this has

    therapeutic implications for the safety of GH antagonism as a therapeutic modality in acromegaly.103 By

    contrast, no mutations of the IGF-1 receptor were found in another study.104

    A relationship between somatostatin sensitivity and tumour pathogenesis has been suggested in

    some GH-omas. However, no mutations affecting the sst2A protein were found in any of 15 analysed

    tumours, suggesting that these mutations are unlikely to be involved in the pathogenesis of acro-

    megaly.105 Molecular analysis of genomic DNA from pituitary tumour and peripheral blood obtained

    from an acromegalic resistant to octreotide confirmed a lack of mutation in sst2, but showed one

    polymorphism (Pro109Ser) and one germ line mutation (Arg240Trp) in sst5.106

    In addition, it is well documented that cytokines of the gp130 family, such as interleukin-6, which

    use gp130 as a common signalling protein, stimulate not only the proliferation but also the hormonesecretion of pituitary cells. Experiments in vivo have shown that the over-expression of the gp130

    receptor resulted in abnormal pituitary growth.107 Bone morphogenetic protein-4 (BMP-4), a member

    of the TGF-b family, promotes the development of prolactinomas but prevents Cushings disease

    progression107; mRNA differential display showed that the BMP inhibitor noggin is down-regulated in

    prolactinomas from dopamine D2-receptor-deficient mice, whereas BMP-4 is over-expressed in

    prolactinomas taken from dopamine D2-receptor-deficient female mice.108 Based on Western blot

    analysis, BMP-4 was found to be over-expressed in other prolactinoma models, including oestradiol-

    induced rat prolactinomas and human prolactinomas, compared with normal tissue and other

    pituitary adenoma types. In addition, BMP-4 stimulates, and noggin blocks, cell proliferation and the

    expression of c-myc in human prolactinomas.108

    On the other hand, ghrelin, originally isolated from the stomach, is an endogenous ligand of the GHsecretagogue receptor (GHS-R). Several groups, including our own, have demonstrated the presence of

    both ghrelin and GHS-R mRNA in the normal human hypothalamus, as well as normal and

    adenomatous human pituitary. ACTH-omas showed significantly less expression of ghrelin mRNA,

    whereas GHS-R mRNA levels were similar to those in normal pituitary tissue. Gonadotroph tumours

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541 535

  • 8/10/2019 pathophysiology of pituitary adenomas

    12/17

    showed a particularly low level of expression of GHS-R mRNA. It seems that the presence of ghrelin

    mRNA and peptide in the pituitary implies that the locally synthesised hormone may have an auto-

    crine/paracrine modulatory effect on pituitary hormone release.109 In addition, in a rat pituitary

    somatotroph cell line, ghrelin was shown to exert a proliferative effect through the mitogen-activated

    protein kinase pathway.110

    Abnormal cell signalling pathways

    Abnormalities in cell signalling pathways are frequently seen in pituitary adenomas. Clarifying the

    role of the particular component involved in regulation of these pathways may represent potential

    selective targets for anti-tumour therapy, and identify the locus or loci of the initiating abnormalities.

    The serinethreonine kinases, including key mediators of tumourigenesis such as Raf, mitogen-

    activated protein kinase (MAPK) cascades and Akt/protein kinase B, appear to be of particular

    interest.111

    The PI3K/Akt/mTOR pathway is altered in many tumours, now including pituitary adenomas.

    Phosphatidylinositol 3-kinase (PI3K) is activated as a result of the ligand-dependent activation of

    tyrosine kinase receptors, G-protein-coupled receptors or integrins. Receptor-independent activationof PI3K can also occur in, for instance, cells expressing a constitutively active Ras protein.112,113 The

    best-characterised phosphorylation target of PI3K is Akt (also known as protein kinase B), resulting in

    the phosphorylation of a host of other proteins that affect cell growth, cell cycle entry and cell survival.

    Akt phosphorylation activates a serinethreonine kinase mTOR (mammalian target of rapamycin),

    which activates 40S ribosomal protein S6 kinase (p70S6K)114 and inactivates 4E-binding protein (4E-

    BP1). It has been shown that the tuberous sclerosis complex (TSC), which mediates between PI3K/Akt

    and mTOR, inhibits mTOR. Mitogenic stimuli activating Akt can directly phosphorylate TSC2 (tuberin),

    causing destabilisation of TSC2 and inhibiting the formation of the TSC1/2 complex, leading to an

    increase of mTOR activity through rheb (Ras homolog enriched in brain).115 4E-BP1 inhibits the initi-

    ation of translation of mRNA for many factors, including c-myc and cyclin D1, through its association

    with eIF-4E116,117

    , and thus loss of the binding protein in response to mTOR activation will lead toenhanced proliferation. Akt is over-expressed (at both mRNA and protein levels) as well as over-

    activated (through phosphorylation) in all pituitary tumours, especially NFPAs.118 This up-regulation of

    Akt will increase the phosphorylation of p27, preventing its nuclear import and causing at least some of

    the changes in the cell cycle discussed above. Therefore, changes in the cell cycle occurring in pituitary

    adenomas may be secondary to activation of the Akt pathway.118 It is also salient that constitutive

    activation of the TSC1/2 complex, as occurs in tuberous sclerosis, appears to be associated with a risk of

    neuroendocrine tumourigenesis, especially insulinomas and Cushings disease.119

    The Raf/MEK/ERK pathway is a hierarchical cascade originating at the cell membrane with receptors

    for mitogens or growth factors, which recruit, via adapter proteins and exchange factors, the small

    guanosine triphosphatase (GTPase) Ras.111 Activated Ras in turn activates Raf (MAPKKK), which is

    a serinethreonine kinase. Raf activates mitogen-activated protein kinase kinase (MAPKK), also knownas MEK; MEK, in turn, phosphorylates and activates mitogen-activated protein kinase (MAPK, or ERK1

    and ERK2), which translocates to the nucleus and transactivates transcription factors, changing gene

    expression to promote growth and mitosis.120 This MAPK pathway activation causes phosphorylation

    and activation of ribosomal S6 kinase and transcription factors such as c-myc, Elk1, c-Fos and cyclin

    D1121,122, similar to the Akt/mTOR cascade and resulting in the activation of genes associated with

    proliferation123 and leading to cell transformation.124 Over-activation of Akt may also lead, via changes

    in the kinase GSK3b, to activation of the b-catenin pathway, which has been implicated in pituitary

    tumourigenesis.9

    The most sensitive Raf, B-Raf, is frequently mutated at the V600E position in melanomas and

    papillary thyroid cancer leading to constitutive activity, but it is not similarly mutated in sporadic

    pituitary adenomas; however, it is over-expressed in pituitary adenomas, particularly NFPAs.

    125

    Bcl-associated athanogene (BAG1), which is over-expressed in somatotroph adenomas and non-functioning

    NFPAs, may also increase B-Raf activity.87 We have recent preliminary data indicating that MEK1/2 as

    well as its down-stream regulator ERK1/2 is also over-phosphorylated, and hence over-activated in all

    types of pituitaryadenomas, compared to normal pituitary.126 Over-activation of ERK, an end-product of

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541536

  • 8/10/2019 pathophysiology of pituitary adenomas

    13/17

    the MAPK pathway, was also associated with increased cyclin D1126, as previously reported.81,127 Future

    studies should be focussed on assessing the role of consecutive ERK1/2 down-stream regulators and

    possible correlations between B-Raf/MEK/ERK and PI3K/Akt/mTOR pathways, especially as these two

    pathways have been shown recently to play an important role not only in malignant transformation but

    also in drug resistance.128 Furthermore, activation of at least one type of growth factor receptor, EGFR,

    increased proliferation in a cell line model.129 This in turn raises the possibility that tyrosine kinasereceptor antagonists, such as gefitinib, may play a role in the treatment of intractable tumours.

    Finally, a very recent study has shown by differential display that a sumoylation factor, now referred

    to as RSUME, is over-expressed in pituitary tumours. This agent sumoylates and thereby stabilises HIF-

    1a in response to hypoxia, and thereby feeds into an important angiogenic pathway implicated, in

    particular, in pheochromocytomas and paragangliomas.130 Study of the interplay between this

    pathway and the preceding signalling pathways will be of immense interest in unraveling pituitary

    oncogenesis.

    In summary, a number of genetic factors, including hormones, growth factors and cell cycle regu-

    lators, are believed to act together with different epigenetic events in the process of pituitary tumourformation. Most pituitary tumours are sporadic, but some arise as a component of hereditary

    syndromes. Our understanding of these genetic conditions has expanded rapidly due to the identifi-

    cation of new predisposing genes, including MEN1, MEN4, PRKAR1A, CDKN1B and AIP, but there is little

    information that the mutations occurring in genetic syndromes are common in sporadic tumours.

    While significant progress has been made in characterising the molecular basis of pituitary

    tumourigenesis, the current evidence does not point towards any of these changes being the primary

    event responsible for sporadic tumours. Mutations in recognised tumour suppressor genes and

    oncogenes do not seem to play an important role in the great majority of pituitary adenomas. However,

    methylation-mediated or -associated gene silencing, in particular of tumour suppressor genes, has

    been reported by numerous investigators. Several genes (e.g., PTTG, Pdt-FGFR4, GADD45G, MEG3A,

    ZAC, DAP kinase, PTAG and p27) have been implicated to be involved in pituitary tumourigenesis. PTTG(securin) seems to be related to pituitary adenoma invasiveness and aggressiveness. Cell signalling

    abnormalities have been identified in pituitary tumours, but their genetic basis is unknown. Both the

    PI3K/Akt/mTOR and the Raf/MEK/ERK pathways are over-expressed and/or over-active in many pitu-

    itary tumours, which results in the inhibition of cell cycle inhibitors, as may b-catenin and HIF path-

    ways. These pathways may share a common root at multiple stages, including initial pathway

    activation related to the tyrosine kinase receptor as well as interactions with down-stream regulators,

    including c-myc or cyclin D1. For a significant proportion of pituitary tumours, the molecular defect is

    not yet elucidated, and more genes for susceptibility are likely to be identified. Modern genetics,

    genomics and molecular biology approaches are expected to reveal new mechanisms of endocrine

    tumourigenesis aiming at the development of better diagnostic, prognostic and therapeutic tools.

    Acknowledgements

    Dorota Dworakowska thanks the Polish Science Foundation for a post-doctoral fellowship to Barts

    and the London School of Medicine, London, England.

    Practice points and research agenda

    Pituitary tumours are characterised by over-activity of both the Akt pathway and the MAPK

    pathways, and, while this is a finding in many tumour types, it suggests that cell cycle changes

    may be secondary to cell signalling abnormalities. There are also changes in the b-catenin

    pathway, which is involved in cell contact inhibition through the cadherins. We speculate that

    changes at the level of the cell surface growth factor receptors and their modulation by integrins

    and cadherins may hold the key to pituitary pathogenesis.

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541 537

  • 8/10/2019 pathophysiology of pituitary adenomas

    14/17

    References

    1. Scheithauer BW, Gaffey TA, Lloyd RV et al. Pathobiology of pituitary adenomas and carcinomas. Neurosurgery 2006; 59:341353 [discussion -53].

    2. Ezzat S, Asa SL, Couldwell WT et al. The prevalence of pituitary adenomas: a systematic review. Cancer 2004; 101:613619.

    3. Daly AF, Burlacu MC, Livadariu E et al. The epidemiology and management of pituitary incidentalomas.Hormone Research2007; 6 8(Suppl. 5): 195198.

    4. Lindholm J, Juul S, Jorgensen JO et al. Incidence and late prognosis of Cushings syndrome: a population-based study.TheJournal of Clinical Endocrinology and Metabolism2001; 86: 117123.

    5. Levy A. Molecular and trophic mechanisms of tumorigenesis.Endocrinology and Metabolism Clinics of North America2008;37:2350. vii.

    6. Alexander JM, Biller BM, Bikkal H et al. Clinically nonfunctioning pituitary tumors are monoclonal in origin.The Journal ofClinical Investigation 1990; 86: 336340.

    7. Herman V, Fagin J, Gonsky R et al. Clonal origin of pituitary adenomas.The Journal of Clinical Endocrinology and Metabolism1990;71: 14271433.

    8. Melmed S. Pituitary neoplasia.Endocrinology and Metabolism Clinics of North America 1994; 23: 8192.9. Gueorguiev M & Grossman AB. Pituitary gland and beta-catenin signaling: from ontogeny to oncogenesis.Pituitary2008.

    10. Levy A & Lightman S. Molecular defects in the pathogenesis of pituitary tumours.Frontiers in Neuroendocrinology 2003;24:94127.

    11. Boikos SA & Stratakis CA. Molecular genetics of the cAMP-dependent protein kinase pathway and of sporadic pituitarytumorigenesis.Human Molecular Genetics 2007; 16(Spec No 1): R80R87.

    12. Beckers A & Daly AF. The clinical, pathological, and genetic features of familial isolated pituitary adenomas. EuropeanJournal of Endocrinology2007; 157:371382.

    *13. Karhu A & Aaltonen LA. Susceptibility to pituitary neoplasia related to MEN-1, CDKN1B and AIP mutations: an update.Human Molecular Genetics 2007; 16(Spec No 1): R73R79.

    *14. Horvath A & Stratakis CA. Clinical and molecular genetics of acromegaly: MEN1, Carney complex, McCune-Albright syndrome,familial acromegaly and genetic defects in sporadic tumors. Reviews in Endocrine & Metabolic Disorders 2008;9:111.

    15. Pellegata NS, Quintanilla-Martinez L, Siggelkow H et al. Germ-line mutations in p27Kip1 cause a multiple endocrineneoplasia syndrome in rats and humans. Proceedings of the National Academy of Sciences of the United States of America2006; 103: 1555815563.

    16. Vierimaa O, Georgitsi M, Lehtonen R et al. Pituitary adenoma predisposition caused by germline mutations in the AIPgene.Science 2006; 312: 12281230.

    17. Igreja S, Chahal HS, Akker SA et al. Assessment of p27 (cyclin-dependent kinase inhibitor 1B) and AIP (aryl hydrocarbonreceptor-interacting protein) genes in MEN1 syndrome patients without any detectable MEN1 gene mutations. Clinical

    Endocrinology2008.18. Luccio-Camelo DC, Une KN, Ferreira RE et al. A meiotic recombination in a new isolated familial somatotropinomakindred.European Journal of Endocrinology 2004; 150: 643648.

    19. Daly AF, Jaffrain-Rea ML, Ciccarelli A et al. Clinical characterization of familial isolated pituitary adenomas.The Journal ofClinical Endocrinology and Metabolism 2006; 91: 33163323.

    20. Spada A, Arosio M, Bochicchio D et al. Clinical, biochemical, and morphological correlates in patients bearing growthhormone-secreting pituitary tumors with or without constitutively active adenylyl cyclase. The Journal of ClinicalEndocrinology and Metabolism 1990; 71: 14211426.

    21. Lania A, Mantovani G & Spada A. Genetics of pituitary tumors: focus on G-protein mutations. Experimental Biology andMedicine (Maywood, N.J.) 2003; 228:10041017.

    22. Burgess JR, Greenaway TM & Shepherd JJ. Expression of the MEN-1 gene in a large kindred with multiple endocrineneoplasia type 1. Journal of Internal Medicine 1998; 243: 465470.

    23. Farrell WE, Simpson DJ, Bicknell J et al. Sequence analysis and transcript expression of the MEN1 gene in sporadicpituitary tumours. British Journal of Cancer1999; 80: 4450.

    24. Satta MA, Korbonits M, Jacobs RA et al. Expression of menin gene mRNA in pituitary tumours. European Journal of

    Endocrinology1999; 140: 358361.25. Poncin J, Stevenaert A & Beckers A. Somatic MEN1 gene mutation does not contribute significantly to sporadic pituitary

    tumorigenesis.European Journal of Endocrinology 1999; 140:573576.26. Dahia PL, Aguiar RC, Honegger J et al. Mutation and expression analysis of the p27/kip1 gene in corticotrophin-secreting

    tumours. Oncogene 1998; 16: 6976.27. Hai N, Aoki N, Shimatsu A et al.Clinical featuresof multipleendocrine neoplasiatype1 (MEN1)phenocopywithout germline

    MEN1 gene mutations: analysis of 20 Japanese sporadic cases with MEN1. Clinical Endocrinology2000;52:509518.28. Georgitsi M, Raitila A, Karhu A et al. Germline CDKN1B/p27Kip1 mutation in multiple endocrine neoplasia.The Journal of

    Clinical Endocrinology and Metabolism 2007; 92: 33213325.29. Lloyd RV, Jin L, Qian X et al. Aberrant p27kip1 expression in endocrine and other tumors. The American Journal of

    Pathology1997; 150:401407.30. Lidhar K, Korbonits M, Jordan S et al. Low expression of the cell cycle inhibitor p27Kip1 in normal corticotroph cells,

    corticotroph tumors, and malignant pituitary tumors. The Journal of Clinical Endocrinology and Metabolism 1999; 84:38233830.

    31. Milne TA, Hughes CM, Lloyd R et al. Menin and MLL cooperatively regulate expression of cyclin-dependent kinaseinhibitors.Proceedings of the National Academy of Sciences of the United States of America2005; 102: 749754.

    32. Drosten M, Hilken G, Bockmann M et al. Role of MEN2A-derived RET in maintenance and proliferation of medullarythyroid carcinoma. Journal of the National Cancer Institute 2004; 96: 12311239.

    33. Kolluri SK, Weiss C, Koff A et al. p27(Kip1) induction and inhibition of proliferation by the intracellular Ah receptor indeveloping thymus and hepatoma cells. Genes & Development 1999; 13: 17421753.

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541538

  • 8/10/2019 pathophysiology of pituitary adenomas

    15/17

    34. Kirschner LS, Carney JA, Pack SD et al. Mutations of the gene encoding the protein kinase A type I-alpha regulatorysubunit in patients with the Carney complex. Nature Genetics 2000; 26: 8992.

    35. Bossis I, Voutetakis A, Matyakhina L et al. A pleiomorphic GH pituitary adenoma from a Carney complex patient displaysuniversal allelic loss at the protein kinase A regulatory subunit 1A (PRKARIA) locus.Journal of Medical Genetics 2004; 41: 596600.

    36. Sandrini F, Kirschner LS, Bei T et al. PRKAR1A, one of the Carney complex genes, and its locus (17q2224) are rarely

    altered in pituitary tumours outside the Carney complex. Journal of Medical Genetics 2002; 39: e78.37. Kaltsas GA, Kola B, Borboli N et al. Sequence analysis of the PRKAR1A gene in sporadic somatotroph and other pituitarytumours.Clinical Endocrinology 2002; 57:443448.

    38. Georgitsi M, Raitila A, Karhu A et al. Molecular diagnosis of pituitary adenoma predisposition caused by aryl hydrocarbonreceptor-interacting protein gene mutations. Proceedings of the National Academy of Sciences of the United States of

    America2007; 104: 41014105.39. Daly AF, Vanbellinghen JF, Khoo SK et al. Aryl hydrocarbon receptor-interacting protein gene mutations in familial iso-

    lated pituitary adenomas: analysis in 73 families. The Journal of Clinical Endocrinology and Metabolism 2007; 92:18911896.

    40. Gadelha MR, Une KN, Rohde K et al. Isolated familial somatotropinomas: establishment of linkage to chromosome 11q13.111q13.3 and evidence for a potential second locus at chromosome 2p1612. The Journal of Clinical Endocrinology andMetabolism2000; 85: 707714.

    41. Leontiou CA, Gueorguiev M, van der Spuy J et al. The role of the aryl hydrocarbon receptor-interacting protein gene infamilial and sporadic pituitary adenomas. The Journal of Clinical Endocrinology and Metabolism 2008; 93: 23902401.

    *42. Asa SL & Ezzat S. The pathogenesis of pituitary tumours. Nature Reviews. Cancer2002; 2: 836849.

    43. Karga HJ, Alexander JM, Hedley-Whyte ET et al. Ras mutations in human pituitary tumors. The Journal of Clinical Endo-crinology and Metabolism 1992; 74:914919.

    44. Herman V, Drazin NZ, Gonsky R et al. Molecular screening of pituitary adenomas for gene mutations and rearrangements.The Journal of Clinical Endocrinology and Metabolism 1993; 77: 5055.

    45. Boggild MD, Jenkinson S, Pistorello M et al. Molecular genetic studies of sporadic pituitary tumors.The Journal of ClinicalEndocrinology and Metabolism 1994; 78:387392.

    46. Cryns VL, Alexander JM, Klibanski A et al. The retinoblastoma gene in human pituitary tumors. The Journal of ClinicalEndocrinology and Metabolism 1993; 77:644646.

    47. Pei L, Melmed S, Scheithauer B et al. Frequent loss of heterozygosity at the retinoblastoma susceptibility gene (RB) locusin aggressive pituitary tumors: evidence for a chromosome 13 tumor suppressor gene other than RB. Cancer Research1995;55: 16131616.

    48. Tanizaki Y, Jin L, Scheithauer BW et al. P53 gene mutations in pituitary carcinomas. Endocrine Pathology 2007; 18: 217222.49. Zhang X, Horwitz GA, Heaney AP et al. Pituitary tumor transforming gene (PTTG) expression in pituitary adenomas.

    The Journal of Clinical Endocrinology and Metabolism 1999; 84: 761767.50. Zhang X, Sun H, Danila DC et al. Loss of expression of GADD45 gamma, a growth inhibitory gene, in human pituitary

    adenomas: implications for tumorigenesis. The Journal of Clinical Endocrinology and Metabolism 2002; 87: 12621267.51. Zhang X, Zhou Y, Mehta KR et al. A pituitary-derived MEG3 isoform functions as a growth suppressor in tumor cells. The

    Journalof Clinical Endocrinology and Metabolism 2003; 88: 51195126.52. Bahar A, Simpson DJ, Cutty SJ et al. Isolation and characterization of a novel pituitary tumor apoptosis gene.Molecular

    Endocrinology2004; 18: 18271839.53. Pei L & Melmed S. Isolation and characterization of a pituitary tumor-transforming gene (PTTG).Molecular Endocrinology

    1997; 11:433441.*54. Salehi F, Kovacs K, Scheithauer BW et al. Pituitary tumor-transforming gene in endocrine and other neoplasms: a review

    and update. Endocrine-related Cancer2008; 15: 721743.55. Bharadwaj R & Yu H. The spindle checkpoint, aneuploidy, and cancer.Oncogene 2004; 23: 20162027.56. Zou H, McGarry TJ, Bernal T et al. Identification of a vertebrate sister-chromatid separation inhibitor involved in trans-

    formation and tumorigenesis. Science 1999; 285: 418422.57. Wang Z & Melmed S. Pituitary tumor transforming gene (PTTG) transforming and transactivation activity.The Journal of

    Biological Chemistry 2000; 275: 74597461.

    58. Chamaon K, Kirches E, Kanakis D et al. Regulation of the pituitary tumor transforming gene by insulin-like-growth factor-I and insulin differs between malignant and non-neoplastic astrocytes. Biochemical and Biophysical Research Communi-cations 2005; 331: 8692.

    59. Tong Y, Tan Y, Zhou C et al. Pituitary tumor transforming gene interacts with Sp1 to modulate G1/S cell phase transition.Oncogene2007; 26: 55965605.

    *60. Chesnokova V, Zonis S, Rubinek T et al. Senescence mediates pituitary hypoplasia and restrains pituitary tumor growth.Cancer Research2007; 67:1056410572.

    61. Chesnokova V, Kovacs K, Castro AV et al. Pituitary hypoplasia in Pttg/ mice is protective for Rb/ pituitary tumor-igenesis.Molecular Endocrinology 2005; 19: 23712379.

    62. Filippella M, Galland F, Kujas M et al. Pituitary tumour transforming gene (PTTG) expression correlates with theproliferative activity and recurrence status of pituitary adenomas: a clinical and immunohistochemical study. ClinicalEndocrinology2006; 65: 536543.

    63. McCabe CJ, Boelaert K, Tannahill LA et al. Vascular endothelial growth factor, its receptor KDR/Flk-1, and pituitary tumortransforming gene in pituitary tumors. The Journal of Clinical Endocrinology and Metabolism 2002; 87:42384244.

    64. Minematsu T, Suzuki M, Sanno N et al. PTTG overexpression is correlated with angiogenesis in human pituitary

    adenomas.Endocrine Pathology 2006; 17: 143153.65. Saez C, Japon MA, Ramos-Morales F et al. hpttg is over-expressed in pituitary adenomas and other primary epithelial

    neoplasias.Oncogene 1999; 18: 54735476.66. Minematsu T, Egashira N, Kajiya H et al. PTTG is a secretory protein in human pituitary adenomas and in mouse pituitary

    tumor cell lines. Endocrine Pathology 2007; 18:815.

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541 539

  • 8/10/2019 pathophysiology of pituitary adenomas

    16/17

    67. Powers CJ, McLeskey SW & Wellstein A. Fibroblast growth factors, their receptors and signaling.Endocrine-related Cancer2000;7: 165197.

    68. Givol D & Yayon A. Complexity of FGF receptors: genetic basis for structural diversity and functional specificity.The FASEBJournal1992; 6: 33623369.

    69. Ezzat S, Smyth HS, Ramyar L et al. Heterogenous in vivo and in vitro expression of basic fibroblast growth factor byhuman pituitary adenomas. The Journal of Clinical Endocrinology and Metabolism 1995; 80: 878884.

    70. Ezzat S, Zheng L, Zhu XF et al. Targeted expression of a human pituitary tumor-derived isoform of FGF receptor-4recapitulates pituitary tumorigenesis. The Journal of Clinical Investigation 2002; 109:6978.71. Qian ZR, Sano T, Asa SL et al. Cytoplasmic expression of fibroblast growth factor receptor-4 in human pituitary adenomas:

    relation to tumor type, size, proliferation, and invasiveness.The Journal of Clinical Endocrinology and Metabolism2004;89:19041911.

    72. Musat M, Vax VV, Borboli N et al. Cell cycle dysregulation in pituitary oncogenesis.Frontiers of Hormone Research2004;32:3462.

    73. Ogino A, Yoshino A, Katayama Y et al. The p15(INK4b)/p16(INK4a)/RB1 pathway is frequently deregulated in humanpituitary adenomas. Journal of Neuropathology and Experimental Neurology 2005; 64: 398403.

    74. Yoshino A, Katayama Y, Ogino A et al. Promoter hypermethylation profile of cell cycle regulator genes in pituitaryadenomas.Journal of Neuro-oncology 2007; 83: 153162.

    75. Bahar A, Bicknell JE, Simpson DJ et al. Loss of expression of the growth inhibitory gene GADD45gamma, in humanpituitary adenomas, is associated with CpG island methylation. Oncogene 2004; 23: 936944.

    76. Simpson DJ, Clayton RN & Farrell WE. Preferential loss of Death Associated Protein kinase expression in invasive pituitarytumours is associated with either CpG island methylation or homozygous deletion. Oncogene 2002; 21: 12171224.

    *77. Farrell WE. A novel apoptosis gene identified in the pituitary gland.Neuroendocrinology2006; 84: 217221.78. Zhao J, Dahle D, Zhou Y et al. Hypermethylation of the promoter region is associated with the loss of MEG3 gene

    expression in human pituitary tumors. The Journal of Clinical Endocrinology and Metabolism 2005; 90: 21792186.79. Pagotto U, Arzberger T, Theodoropoulou M et al. The expression of the antiproliferative gene ZAC is lost or highly reduced

    in nonfunctioning pituitary adenomas. Cancer Research2000; 60: 67946799.*80. Korbonits M, Chahal HS, Kaltsas G et al. Expression of phosphorylated p27(Kip1) protein and Jun activation domain-

    binding protein 1 in human pituitary tumors. The Journal of Clinical Endocrinology and Metabolism 2002;87:26352643.81. Jordan S, Lidhar K, Korbonits M et al. Cyclin D and cyclin E expression in normal and adenomatous pituitary. European

    Journal of Endocrinology2000; 143: R1R6.82. Ruebel KH, Leontovich AA, Jin L et al. Patterns of gene expression in pituitary carcinomas and adenomas analyzed by

    high-density oligonucleotide arrays, reverse transcriptase-quantitative PCR, and protein expression. Endocrine2006;29:435444.

    83. Evans CO, Young AN, Brown MR et al. Novel patterns of gene expression in pituitary adenomas identified by comple-mentary deoxyribonucleic acid microarrays and quantitative reverse transcription-polymerase chain reaction.The Journalof Clinical Endocrinology and Metabolism 2001; 86: 30973107.

    84. Evans CO, Moreno CS, Zhan X et al. Molecular pathogenesis of human prolactinomas identified by gene expressionprofiling, RT-qPCR, and proteomic analyses. Pituitary2008;11: 231245.

    *85. Moreno CS, Evans CO, Zhan X et al. Novel molecular signaling and classification of human clinically nonfunctionalpituitary adenomas identified by gene expression profiling and proteomic analyses. Cancer Research 2005; 65:1021410222.

    86. Elston MS, Gill AJ, Conaglen JV et al. Wnt pathway inhibitors are strongly down-regulated in pituitary tumors.Endo-crinology2008; 149: 12351242.

    87. Morris DG, Musat M, Czirjak S et al. Differential gene expression in pituitary adenomas by oligonucleotide array analysis.European Journal of Endocrinology 2005; 153: 143151.

    88. Ribeiro-Oliveira A, Franchi G, Kola B et al. Protein western array analysis in human pituitary tumours: insights andlimitations.Endocrine-related Cancer2008.

    89. Zhan X, Evans CO, Oyesiku NM et al. Proteomics and transcriptomics analyses of secretagogin down-regulation in humannon-functional pituitary adenomas. Pituitary 2003; 6: 189202.

    90. Beranova-Giorgianni S, Zhao Y, Desiderio DM et al. Phosphoproteomic analysis of the human pituitary.Pituitary2006;9:

    109120.91. Morris DG, Kola B, Borboli N et al. Identification of adrenocorticotropin receptor messenger ribonucleic acid in the humanpituitary and its loss of expression in pituitary adenomas.The Journal of Clinical Endocrinology and Metabolism 2003;88:60806087.

    92. Dahia PL, Honegger J, Reincke M et al. Expression of glucocorticoid receptor gene isoforms in corticotropin-secretingtumors. The Journal of Clinical Endocrinology and Metabolism 1997; 82: 10881093.

    93. de Keyzer Y, Rene P, Beldjord C et al. Overexpression of vasopressin (V3) and corticotrophin-releasing hormone receptorgenes in corticotroph tumours. Clinical Endocrinology 1998; 49: 475482.

    94. Dieterich KD, Gundelfinger ED, Ludecke DK et al. Mutation and expression analysis of corticotropin-releasing factor 1receptor in adrenocorticotropin-secreting pituitary adenomas.The Journal of Clinical Endocrinology and Metabolism 1998;83: 33273331.

    95. Dahia PL, Ahmed-Shuaib A, Jacobs RA et al. Vasopressin receptor expression and mutation analysis in corticotropin-secreting tumors. The Journal of Clinical Endocrinology and Metabolism 1996; 81: 17681771.

    96. Korbonits M, Bujalska I, Shimojo M et al. Expression of 11 beta-hydroxysteroid dehydrogenase isoenzymes in the humanpituitary: induction of the type 2 enzyme in corticotropinomas and other pituitary tumors. The Journal of Clinical

    Endocrinology and Metabolism 2001; 86: 27282733.97. Rabbitt EH, Ayuk J, Boelaert K et al. Abnormal expression of 11 beta-hydroxysteroid dehydrogenase type 2 in human

    pituitary adenomas: a prereceptor determinant of pituitary cell proliferation. Oncogene 2003; 22: 16631667.98. Bilodeau S, Vallette-Kasic S, Gauthier Y et al. Role of Brg1 and HDAC2 in GR trans-repression of the pituitary POMC gene

    and misexpression in Cushing disease. Genes & Development2006; 20: 28712886.

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541540

  • 8/10/2019 pathophysiology of pituitary adenomas

    17/17

    99. Drouin J, Bilodeau S & Vallette S. Of old and new diseases: genetics of pituitary ACTH excess (Cushing) and deficiency.Clinical Genetics 2007; 72: 175182.

    100. Lee EJ, Kotlar TJ, Ciric I et al. Absence of constitutively activating mutations in the GHRH receptor in GH-producingpituitary tumors. The Journal of Clinical Endocrinology and Metabolism 2001; 86: 39893995.

    101. Salvatori R, Thakker RV, Lopes MB et al. Absence of mutations in the growth hormone (GH)-releasing hormone receptorgene in GH-secreting pituitary adenomas. Clinical Endocrinology 2001; 54: 301307.

    102. Kola B, Korbonits M, Diaz-Cano S et al. Reduced expression of the growth hormone and type 1 insulin-like growth factorreceptors in human somatotroph tumours and an analysis of possible mutations of the growth hormone receptor. ClinicalEndocrinology2003; 59: 328338.

    *103. Asa SL, Digiovanni R, Jiang J et al. A growth hormone receptor mutation impairs growth hormone autofeedback signalingin pituitary tumors. Cancer Research2007; 67:75057511.

    104. Greenman Y, Prager D & Melmed S. The IGF-I receptor sub-membrane domain is intact in GH-secreting pituitarytumours.Clinical Endocrinology 1995; 42: 169172.

    105. Petersenn S, Heyens M, Ludecke DK et al. Absence of somatostatin receptor type 2 A mutations and gip oncogene inpituitary somatotroph adenomas. Clinical Endocrinology 2000; 52: 3542.

    106. Ballare E, Persani L, Lania AG et al. Mutation of somatostatin receptor type 5 in an acromegalic patient resistant tosomatostatin analog treatment. The Journal of Clinical Endocrinology and Metabolism 2001; 86: 38093814.

    107. Giacomini D, Acuna M, Gerez J et al. Pituitary action of cytokines: focus on BMP-4 and gp130 family.Neuroendocrinology2007; 85: 94100.

    108. Paez-Pereda M, Giacomini D, Refojo D et al. Involvement of bone morphogenetic protein 4 (BMP-4) in pituitary pro-lactinoma pathogenesis through a Smad/estrogen receptor crosstalk. Proceedings of the National Academy of Sciences of

    the United States of America 2003; 100:10341039.109. Korbonits M, Kojima M, Kangawa K et al. Presence of ghrelin in normal and adenomatous human pituitary.Endocrine

    2001;14: 101104.110. Nanzer AM, Khalaf S, Mozid AM et al. Ghrelin exerts a proliferative effect on a rat pituitary somatotroph cell line via the

    mitogen-activated protein kinase pathway. European Journal of Endocrinology 2004; 151: 233240.111. Adjei AA & Hidalgo M. Intracellular signal transduction pathway proteins as targets for cancer therapy.Journal of Clinical

    Oncology2005; 23: 53865403.112. Rodriguez-Viciana P, Warne PH, Khwaja A et al. Role of phosphoinositide 3-OH kinase in cell transformation and control

    of the actin cytoskeleton by Ras. Cell 1997; 89: 457467.113. Kauffmann-Zeh A, Rodriguez-Viciana P, Ulrich E et al. Suppression of c-Myc-induced apoptosis by Ras signalling through

    PI(3)K and PKB. Nature 1997; 385: 544548.114. Volarevic S & Thomas G. Role of S6 phosphorylation and S6 kinase in cell growth.Progress in Nucleic Acid Research and

    Molecular Biology 2001; 65: 101127.115. Inoki K, Li Y, Zhu T et al. TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling.Nature Cell Biology

    2002; 4: 648657.116. Sonenberg N & Gingras AC. The mRNA 50 cap-binding protein eIF4E and control of cell growth.Current Opinion in Cell

    Biology1998; 10: 268275.117. Rosenwald IB, Kaspar R, Rousseau D et al. Eukaryotic translation initiation factor 4E regulates expression of cyclin D1 at

    transcriptional and post-transcriptional levels. The Journal of Biological Chemistry 1995; 270: 2117621180.*118. Musat M, Korbonits M, Kola B et al. Enhanced protein kinase B/Akt signalling in pituitary tumours.Endocrine-related

    Cancer2005; 12: 423433.119. Dworakowska D & Grossman A. Are neuroendocrine tumours a feature of tuberous sclerosis?Endocrine-related Cancer

    2009; 16:4558.120. Haystead TA, Dent P, Wu J et al. Ordered phosphorylation of p42mapk by MAP kinase kinase. FEBS Letters 1992; 306: 1722.121. Terada Y, Nakashima O, Inoshita S et al. Mitogen-activated protein kinase cascade and transcription factors: the opposite

    role of MKK3/6-p38K and MKK1-MAPK. Nephrology, Dialysis, Transplantation 1999; 14(Suppl. 1):4547.122. Terada Y, Inoshita S, Nakashima O et al. Regulation of cyclin D1 expression and cell cycle progression by mitogen-acti-

    vated protein kinase cascade. Kidney International 1999; 56: 12581261.123. Guan KL. The mitogen activated protein kinase signal transduction pathway: from the cell surface to the nucleus.Cellular

    Signalling1994; 6: 581589.124. Joneson T & Bar-Sagi D. Ras effectors and their role in mitogenesis and oncogenesis.Journal of Molecular Medicine 1997;75:587593.

    125. Ewing I, Pedder-Smith S, Franchi G et al. A mutation and expression analysis of the oncogene BRAF in pituitary adenomas.Clinical Endocrinology 2007; 66: 348352.

    126. Dworakowska D, Teng M, Grozinsky-Glasberg S, et al. Extracellular signal-regulated kinase (ERK 1/2) overactivation inpituitary adenomas. The Endocrine Societys 90th Annual Meeting 2008, San Francisco, USA; 2008: [P3514].

    127. Hibberts NA, Simpson DJ, Bicknell JE et al. Analysis of cyclin D1 (CCND1) allelic imbalance and overexpression in sporadichuman pituitary tumors. Clinical Cancer Research 1999; 5: 21332139.

    128. McCubrey JA, Steelman LS, Abrams SL et al. Roles of the RAF/MEK/ERK and PI3K/PTEN/AKT pathways in malignanttransformation and drug resistance. Advances in Enzyme Regulation 2006; 46: 249279.

    129. Vlotides G, Siegel E, Donangelo I et al. Rat prolactinoma cell growth regulation by epidermal growth factor receptorligands.Cancer Research 2008; 68: 63776386.

    130. Carbia-Nagashima A, Gerez J, Perez-Castro C et al. RSUME, a small RWD-containing protein, enhances SUMO conjugationand stabilizes HIF-1alpha during hypoxia. Cell 2007; 131: 309323.

    131. Simpson DJ, Hibberts NA, McNicol AM et al. Loss of pRb expression in pituitary adenomas is associated with methylationof the RB1 CpG island. Cancer Research 2000; 60: 12111216.

    D. Dworakowska, A.B. Grossman / Best Practice & Research Clinical Endocrinology & Metabolism 23 (2009) 525541 541