Abstract

Context

Craniopharyngiomas, which are categorized as adamantinomatous (ACPs) or papillary (PCPs), have traditionally been treated with surgery and/or radiotherapy, although when the tumors progress or recur, therapeutic possibilities are very limited. Following recent advances in their molecular pathogenesis, new medical therapeutic options have emerged.

Evidence Acquisition

The search strategy that we selected to identify the appropriate evidence involved the following medical subject headings (MeSH) terms: (“Craniopharyngioma” [MeSH] AND “Craniopharyngioma/drug therapy” [MeSH]) NOT (“review” [Publication Type] OR “review literature as topic” [MeSH Terms] OR “review” [All Fields]) AND (“2009/05/01” [PDat]: “2019/04/28” [PDat]).

Evidence Synthesis

Mutations of β-catenin causing Wnt activation with alterations of the MEK/ERK pathway are encountered in the great majority of patients with ACPs; specific alterations also stratify patients to a more aggressive behavior. In most PCPs there is primary activation of the Ras/Raf/MEK/ERK pathway secondary to BRAF-V600E mutations. BRAF inhibitors, such as dabrafenib or vemurafenib, either alone or in combination with the MEK inhibitors trametinib and cobimetinib, have been administered to patients with PCPs producing clinically useful and, in some cases, sustained responses. In contrast to PCPs, drugs targeting β-catenin and its downstream MAPK pathway in ACPs have so far only been used in in vitro studies, but there appear to be promising new targets clinically.

Conclusions

The identification of specific genetic alterations in patients with craniopharyngiomas has expanded the therapeutic options, providing evidence for a customized approach using newer molecular agents. More studies including a larger number of carefully selected patients are required to evaluate the response to currently available and evolving agents alone and in combination.

The title of this review may sound like an oxymoron, as until very recently the mainstays of treatment of craniopharyngiomas (CPs) have been surgery and radiotherapy (RT). However, during the last few years there have been major developments in our understanding of the molecular pathology of these tumors that could set the scene for specific targeted therapy.

CPs develop as malformations of embryonic remnants along the original pathway of the craniopharyngeal duct. They are characterized as mostly benign epithelial tumors according to the World Health Organization Classification of Tumors of the Central Nervous System, namely, World Health Organization grade I tumors (13). They are relatively rare, with an incidence of 0.5 to 2.5 per 106 cases per year (2, 47), and are divided into two distinct subtypes, adamantinomatous CPs (ACPs) and papillary CPs (PCPs), differing in both histological features and genetic alterations (8). ACP is more prevalent and displays a bimodal age distribution, with peaks between the ages 5 to 15 years and 45 to 60 years, but it can occur at any age, even during the neonatal period (3, 9, 10); PCP has been classically considered as an adult entity peaking between 40 and 55 years of age (2, 4, 7, 11), although pediatric cases have occasionally been reported (12).

Harvey Cushing not only suggested the term “craniopharyngioma” but also recognized the substantial challenges in their management (13). Indeed, their anatomic vicinity to vital structures (hypothalamus, optic pathways, brain) and their intrinsic local infiltrative tendency render their growth pattern unpredictable and complete safe surgical removal problematic (14). Furthermore, malignant transformation may also rarely be seen and, overall, the long-term morbidities and mortality of patients with CPs are disappointedly high, particularly compared with patients with treated pituitary neuroendocrine tumors (15, 16).

Current therapeutic options are relatively limited: surgery, either with the intention to treat by gross total resection (GTR) or with the intention to reduce their mass by subtotal/partial resection (STR) (17, 18), with or without postoperative RT (19), is the primary therapeutic modality (3). In a systematic review, STR with adjuvant RT, and GTR, showed similar rates of long-term tumor control (20). Currently, STR followed by RT is recommended, unless there is a clear margin between the tumor and surrounding vital structures, allowing complete safe tumor removal. Whenever feasible, the tumors are approached by the transsphenoidal route (17, 18). The intracystic administration of sclerosing substances, mainly bleomycin and interferon (IFN)-α, has been suggested in the past to attenuate fluid formation and diminish cyst size (2123). Bleomycin use is limited by its toxicity (21, 22), whereas IFN-α has shown some benefit with an acceptable safety profile (24); however, no prospective or randomized study has been performed, and there are isolated reports of relatively poor responses and major adverse effects following IFN-α instillation, limiting its use (21).

Recently, progress in molecular biology has unraveled novel potentially druggable molecular pathways in CPs that offer new promising therapeutic options. The use of such drugs is especially important to minimize the otherwise adverse effects associated with surgery and RT, particularly in recurrent tumors where there are few or no alternative options. The purpose of this mini-review is to present the available literature on clinical studies and case reports describing targeted treatments that are based on the recently identified molecular alterations in both ACPs and PCPs. The search strategy that we selected to identify the appropriate evidence involved the following MeSH terms: (“Craniopharyngioma” [MeSH] AND “Craniopharyngioma/drug therapy” [MeSH]) NOT (“review” [Publication Type] OR “review literature as topic” [MeSH Terms] OR “review” [All Fields]) AND (“2009/05/01” [PDat]: “2019/04/28” [PDat]) (Fig. 1).

The MeSH terms used in the search strategy: (“Craniopharyngioma” [MeSH] AND “Craniopharyngioma/drug therapy” [MeSH]) NOT (“review” [Publication Type] OR “review literature as topic” [MeSH Terms] OR “review” [All Fields]) AND (“2009/05/01” [PDat]: “2019/04/28” [PDat]).
Figure 1.

The MeSH terms used in the search strategy: (“Craniopharyngioma” [MeSH] AND “Craniopharyngioma/drug therapy” [MeSH]) NOT (“review” [Publication Type] OR “review literature as topic” [MeSH Terms] OR “review” [All Fields]) AND (“2009/05/01” [PDat]: “2019/04/28” [PDat]).

Molecular Biological Advances in ACPs and PCPs

It has recently been shown that each CP histological subtype (Figs. 2 and 3) is characterized by alterations in oncogenic molecular signaling pathways, harboring distinct mutational, transcriptomic, and epigenomic profiles as a result of different gene mutations, gene expression, and methylation patterns (2528). These findings have shed light on the identification of tumor-specific signaling pathway activation that may lead to CP-targeted therapies. An additional role of the different molecular profiles may be to differentiate pathologically indeterminate cases of CPs (28, 29).

Photomicrograph of a representative case of a PCP. Hematoxylin and eosin staining; original magnification, ×100 magnification. [Dr. A. Ramanathan and Prof. A. B. Grossman, unpublished data.]
Figure 2.

Photomicrograph of a representative case of a PCP. Hematoxylin and eosin staining; original magnification, ×100 magnification. [Dr. A. Ramanathan and Prof. A. B. Grossman, unpublished data.]

(A) Histology of ACP. Basal epithelium demonstrating the aberrant nuclear accumulation of β-catenin in nodular whorls (arrowheads) due to β-catenin mutation. Anti–β-catenin stain; original magnification, ×400. (B) Histology of PCP. Squamous epithelium showing membranous immunoreactivity of β-catenin, lacking clusters with aberrant nuclear accumulation. Original magnification, ×400. [Reproduced from Lithgow K, Pohl U, Karavitaki N. Craniopharyngiomas. In: Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2019 Mar 14.]
Figure 3.

(A) Histology of ACP. Basal epithelium demonstrating the aberrant nuclear accumulation of β-catenin in nodular whorls (arrowheads) due to β-catenin mutation. Anti–β-catenin stain; original magnification, ×400. (B) Histology of PCP. Squamous epithelium showing membranous immunoreactivity of β-catenin, lacking clusters with aberrant nuclear accumulation. Original magnification, ×400. [Reproduced from Lithgow K, Pohl U, Karavitaki N. Craniopharyngiomas. In: Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-2019 Mar 14.]

Sekine et al. (30) first described activating mutations of the β-catenin encoding gene CTNNB1 in ACPs as early as 2002. Whole-exome sequencing revealed mutations in CTNNB1 in most of a small group of ACPs (11/12, 92%) (25). Additionally, targeted genotyping revealed CTNNB1 mutations in 96% of ACPs (51/53) (25). Next-generation panel sequencing did not identify any mutations other than those in CTNNB1 (27). In 76% of the ACPs, a mutation in exon 3 of CTNNB1 was found, and there was a trend toward a worse event-free survival in cases mutated at Thr41 (27). When primary and recurrent ACP tumor samples from the same patient were tested, the same CTNNB1 mutation (S45P) was found without additional ones (28). No substantial large chromosomal aberrations have been found, although a fraction of ACPs showed recurrent focal gains of chromosomal material, whereas in other cases there was loss of chromosomal region Xq28 (27).

These CTNNB1 mutations are exclusively found in exon 3, which encodes the destruction/degradation complex of β-catenin: β-catenin is held in a multiprotein complex that can be phosphorylated at specific residues, and is then very quickly degraded. If such phosphorylation sites are mutated then there is aberrant nucleocytoplasmic accumulation of β-catenin (30) (Fig. 3A). This can also be demonstrated by immunocytochemistry (IHC), which is highly reliable in differentiating ACPs from other (para)sellar lesions (31, 32). β-Catenin is a dominant mediator of the classic Wnt molecular signaling pathway and is also part of the adherens complex (8). Normally, there are only low levels of β-catenin in the cytoplasm, as when phosphorylated it is rapidly degraded in a multiprotein destruction–regulator complex, facilitating its ubiquitination and degradation by the proteasome. Thus, absent Wnt ligands, such signaling is inactive, the default or “off” state (8, 3335) (Fig. 4A). Other proteins held in this regulatory complex include the scaffold proteins Axin and Axin2, the tumor suppressor protein adenomatosis polyposis coli (APC), and the phosphokinases glycogen synthase kinase-3β (GSK-3β), casein kinase 1α (CK1), protein phosphatase 2A (PP2A) or, alternatively, by IκB kinase α (IKK-α) (36). The kinases phosphorylate APC, which in turn recruits β-catenin to the complex (34, 36); in this way the complex is marked to be degraded via phosphorylation of serine (Ser33, Ser37, Ser45) or threonine (Thr41) residues (of exon 3 on the CTNNB1) (34, 3639). Additionally, recognition of such phosphorylation by multiple ubiquitin molecules, such as a component of E3 ubiquitin ligase, β-TrCP, allows ubiquitination of β-catenin and dissociation of α-catenin from β-catenin, resulting in loss of cadherin adhesion (34, 35, 3741). In an alternative scenario, exogenous Wnt signaling ligands “switch on” (i.e., activate) Wnt signaling (41, 42), The Wnt protein family comprises ∼20 different proteins that bind to the Frizzled family of receptors. When activated, the Wnt pathway (34) leads to an intracellular signaling cascade that includes dimerization of Frizzled with LRP5/6 (34, 43), which in turn binds to Axin, GSK-3β, and CK1. These latter mediators are then sequestered at the cell membrane by Dishevelled (Dvl) distant to the β-catenin destruction complex, diminishing phosphorylation of β-catenin and thus stabilizing it by rendering it refractory to degradation (3436, 4345). The accumulation of β-catenin allows for its translocation to the nucleus, possibly mediated by its Armadillo-like sequences, and then leads to the transcription of target genes, including CyclinD1, c-Myc, CD44, Survivin, vascular endothelial growth factor (VEFG), and fascin (26, 4649). These various factors acting alone or in combination stimulate cellular proliferation and other Wnt-regulated processes (50). Importantly, note that it is the nuclear accumulation of β-catenin that eventuates in Wnt-signaling target gene expression. It has been shown that the target genes Axin2, bone morphogenetic protein (BMP)4, and Fascin-1, stem cell markers (CD133 and CD44), and the cell cycle inhibitor p21Waf1/CIP1 are specifically concentrated in cells with nuclear β-catenin (8). One particular target of special interest is Fascin (48), which cross-links actin and is involved in filopodia formation, and thus mediates tumor cell migration and related tumoral invasion (36); β-catenin binds fascin by its armadillo-repeat sequence (as noted above, this is also involved in nuclear pore complex translocation of β-catenin) (51), leading to the relocation of β-catenin changing cell adhesion and decreasing β-catenin degradation (36). As a consequence, loss of either fascin or β-catenin expression has been shown to attenuate the ability of ACP tumor cells to migrate when cultured (36). The armadillo-repeat sequences are clearly significant in that they also bind to the TCP family transcription factors, Axin2, and APC (52). Furthermore, other growth factors pathways are also involved: β-catenin cell clusters demonstrate activation of epidermal growth factor (EGF) receptor (EGFR), or src-driven phosphorylation at the tyrosine of codon 654 (Y654) and sonic hedgehog (SHH) signaling pathways (8, 26, 28, 48, 5255). Such activation reduces the binding affinity of β-catenin to E-cadherin such that the adherens complex is destabilized (40, 56) and may additionally aid in tumor stem cell maintenance (5759). Additionally, Fyn, Fer, or c-Met promote phosphorylation of β-catenin at tyrosine residue 142, which also facilitates the loosened binding of α-catenin to β-catenin (Fig. 4A). RNA profiling (60) has confirmed that human ACP clusters express high levels of the fibroblast growth factor (FGF), BMP, and Wnt families (50); these are growth-related signaling mediators that have been shown to be involved in a variety of pathophysiological and embryological situations. These molecules may be useful in terms of predicting tumor recurrence: FGF-2 is only seen in recurrent ACPs, whereas platelet-derived growth factor (PDGF) receptor-α expression is significantly higher in recurrent ACP compared with nonrecurrent tumors. In contrast, VEGF and fibronectin are expressed in both ACP and PCP (61). BMP2 and BMP4 are downstream of the Wnt/β-catenin pathway and are present at high expression levels in ACP (52, 53, 62). Of particular importance from the therapeutic perspective, as discussed below, is that EGFR is a member of the human EGFR (HER) family, and its most significant signal transduction pathways are those involving the Ras-Raf MAPK (MAPK/ERK pathway) and the phosphatidylinositol 3-kinase (PI3K)/Akt pathways (8) (Fig. 4B).

(a) Schematic presentation of adherens junction complex (including β- and α-catenin and E-cadherin) and the molecular signaling pathways that may crosstalk in both ACPs or PCPs. In ACPs, β-catenin cluster cells show an activation of the Wnt, SHH, and EGFR pathway, which are reported to crosstalk with each other. When Wnt signaling is inactive, axin recruits and binds to GSK-3β, CK1α, and PP2A. APC is phosphorylated by these kinases, which recruit β-catenin to the complex and tag it for degradation through phosphorylation of the serine (S33, S37, S45) or threonine (T41) residues of exon 3 on the CTNNB1 gene. This phosphorylation pattern is recognized by β-TrCP, which ubiquitinates β-catenin, When Wnt signaling is active, by the activation of a Wnt ligand or when extracellular SHH binds to the transmembrane receptor PTCH1, the protein degradation complex is inhibited and β-catenin is accumulated first in the cytoplasm and then in the nucleus. There, β-catenin represents a transcription cofactor by interacting with transcription factors of the0020TCF family TCF/LEF1, initiating expression of the target genes. (b) In ACPs, paracrine growth-related signaling molecules such as SOX2. FGFs, EGF, and PDGF bind to their receptors, activating the Ras/Raf/MAPK/ERK1/2 pathway and the PI3K/Akt pathway. Similarly, in PCPs the BRAF-V600E serine/threonine kinase is a critical component of the Ras-Raf-Mek-ERK1/2 pathway that has been mutated. Red lines show the inhibitors that may be used as molecular-targeted therapy for CPs. BMP, bone morphogenetic protein; Dvl, Dishevelled; FGF, fibroblast growth factor; Fz, Frizzled; IKK-α, IκB kinase α; LEF1, lymphocyte enhancer factor 1; PI3K, phosphatidylinositol 3-kinase; PP2A, protein phosphatase 2A; PTCH1, patched 1; TCF, T-cell factor; VEGF-R, vascular endothelial growth factor receptor.
Figure 4.

(a) Schematic presentation of adherens junction complex (including β- and α-catenin and E-cadherin) and the molecular signaling pathways that may crosstalk in both ACPs or PCPs. In ACPs, β-catenin cluster cells show an activation of the Wnt, SHH, and EGFR pathway, which are reported to crosstalk with each other. When Wnt signaling is inactive, axin recruits and binds to GSK-3β, CK1α, and PP2A. APC is phosphorylated by these kinases, which recruit β-catenin to the complex and tag it for degradation through phosphorylation of the serine (S33, S37, S45) or threonine (T41) residues of exon 3 on the CTNNB1 gene. This phosphorylation pattern is recognized by β-TrCP, which ubiquitinates β-catenin, When Wnt signaling is active, by the activation of a Wnt ligand or when extracellular SHH binds to the transmembrane receptor PTCH1, the protein degradation complex is inhibited and β-catenin is accumulated first in the cytoplasm and then in the nucleus. There, β-catenin represents a transcription cofactor by interacting with transcription factors of the0020TCF family TCF/LEF1, initiating expression of the target genes. (b) In ACPs, paracrine growth-related signaling molecules such as SOX2. FGFs, EGF, and PDGF bind to their receptors, activating the Ras/Raf/MAPK/ERK1/2 pathway and the PI3K/Akt pathway. Similarly, in PCPs the BRAF-V600E serine/threonine kinase is a critical component of the Ras-Raf-Mek-ERK1/2 pathway that has been mutated. Red lines show the inhibitors that may be used as molecular-targeted therapy for CPs. BMP, bone morphogenetic protein; Dvl, Dishevelled; FGF, fibroblast growth factor; Fz, Frizzled; IKK-α, IκB kinase α; LEF1, lymphocyte enhancer factor 1; PI3K, phosphatidylinositol 3-kinase; PP2A, protein phosphatase 2A; PTCH1, patched 1; TCF, T-cell factor; VEGF-R, vascular endothelial growth factor receptor.

Of the pathways disrupted in human cancers, Ras-Raf-Mek-ERK1/2 is one of the most important (63, 64). Extracellular growth factors (FGFs, EGF, PDGF) bind to and activate receptor tyrosine kinases, causing a downstream phosphorylation cascade that eventually leads to transcription of target genes regulating cellular proliferation, differentiation, apoptosis, and senescence (63). Downstream activation of the MAPK/ERK pathway, as evidenced by phosphorylation of ERK1/2, was identified by IHC around the clusters with particularly prominent staining at the forefront, known as the leading edge, of the tumor (50). Phospholipase Cγ (PLCγ), the signal transducer and activator of transcription (STAT), and src/FAK cascades are also stimulated (8, 65, 66). SHH signaling pathway gene products, such as Gli2 and patched (Ptch)1, are overexpressed in ACP, and their cleaved active forms are especially highly expressed (8, 26, 28, 53). Upregulation of certain matrix metallopeptidases (MMPs) has also been reported in ACP transcriptome studies (23, 50). A variable degree of colocalization between Ki-67 and pERK1/2 expression has also been also shown (60). In ACP, whole-exome sequencing showed additional mutations in genes listed in the Cancer Gene Census (67), although many of these may be “passenger” mutations (25); there is also evidence for recurrent copy number changes in some tumors (27). Thus, mutation-produced loss of phosphorylation sites on β-catenin leads to a plethora of downstream changes initiating and augmenting tumor growth.

Considering PCPs, a BRAF-V600E mutation was documented (25) by targeted genotyping in 95% of PCPs (36/39), whereas whole-exome sequencing revealed recurrent mutations in BRAF-V600E in all PCPs (3/3) (25). In a parallel study, an analysis of known oncogenes revealed that BRAF-V600E mutations were seen in 81% (17/21) of PCPs by targeted Sanger sequencing and in 86% (18/21) of PCPs by IHC (68). Agreement between methods was seen in 95% (20/21) of cases; however, interpretation of anti–BRAF-V600E staining was challenging due to occasional nonspecific reactivity (68). It is well established that the gain-of-function mutation BRAF-V600E, a critical serine/threonine kinase in the Ras-Raf-Mek-ERK1/2 pathway, renders it a potent oncogene, leading to increased cell proliferation and survival, resulting in cell transformation and tumorigenesis (69, 70) (Fig. 4B). Such mutations render the BRAF kinase constitutively active. The MAPK pathway also plays a role in controlling stem-cell specification during development and perhaps during stem-cell homeostasis in the postnatal period (70), modulating the balance between the proliferation and differentiation of Sox2+ cells, implying that persistent proliferative capacity of Sox2+ cells may underlie the pathogenesis of PCP (70). Sox2+ progenitor cells appear to show tumor-inducing potential, driving tumor formation in a paracrine manner by inducing tumorigenic events in adjacent cells but without gains or losses of genetic material (53, 54, 71). In PCPs (25), whole-exome sequencing with a recently described novel technique—MuTectalso showed isolated mutations of cancer-related genes encoding chromatin remodeling factors (CHD5, CHD6) and cell adhesion molecules (CDH26, PTPRT), and one (KIAA1549) that is fused to BRAF in most cases of pilocytic astrocytoma (72, 73). However, it is presently unclear whether these mutations are pathogenetic or merely passenger mutations (25).

CTNNB1 and BRAF mutations were mutually exclusive and clonal in ACP and PCP, according to an original observation by Brastianos et al. (25), and there were no other recurrent mutations or genomic aberrations in either subtype (Fig. 3A and 3B). Thus, the specific oncogenic change appears to be Wnt activation in ACP, as well as activation of the Ras/Raf/MEK/ERK pathway by BRAF-V600E mutations in PCP (27). It was then suggested that one should classify CPs as ACP CTNNB1 mutated, ACP CTNNB1 wild type, PCP BRAF-V600E mutated, and PCP BRAF wild type (74). Comparison microarray analysis of 18 ACP and 10 PCP samples revealed significant upregulation of several direct targets of the Wnt/β-catenin signaling pathway in ACPs compared with PCPs, including LEF1 and AXIN, whereas components of the SHH such as GLI2, PTCH1, and SHH were also overexpressed specifically in ACPs in comparison with PCPs (8, 36, 48, 52, 71, 75, 76). Unsupervised consensus clustering of the gene expression values of the 5000 most variable genes resulted in two distinct and stable clusters that perfectly separated ACP and PCP samples. Gene expression data showed increased expression of microtubule-associated protein 2 (MAP2), tenascin C (TNC), and the stem cell marker CD133 (PROM1) in ACPs. CD44 and claudin-1 (CLDN1) were significantly downregulated in ACPs, but showed higher expression in PCPs (28, 52, 75). Moreover, reduction of claudin-1 protein levels point toward an invasive growth pattern in ACPs (75). Downregulation of miRNA miR-132 was identified as a marker of aggressiveness, playing a role in epithelial–mesenchymal transition, in a study of 754 miRNAs from childhood CPs (77). However, in contrast to the view of a complete molecular separation between these two tumor subtypes, Larkin et al. (68), in a small cohort of ACPs, reported a dual aberration of CTNNB1 mutated at Thr41 together with BRAF-V600E mutation in two tumors. This finding was validated by review of morphology and comparison with IHC findings. Sequencing in forward and reverse directions from two DNA samples extracted on different occasions, confirmed these findings. Whether this represents the rare occurrence of a collision tumor needs to be established in future larger studies (68). The expression of additional stem cell markers SOX9, KLF4, and Okt4 was also identified by IHC in both ACPs and PCPs (8, 78).

Other transcriptional molecular analyses of ACPs have revealed the expression of immune system gene expression, including IL-1β, IL-6, IL-8 (CXCL-8), IL-10, IL-18. and TNF-α. In particular, there is a characteristic inflammatory cytokine/chemokine reaction in both ACP cyst fluid and solid tumor components. Cytokines and chemokines with elevated concentrations in ACPs included IL-6, CXCL-1 (GRO), CXCL-8, and the immunosuppressive cytokine IL-10, and their relevant receptors are also expressed in ACPs, as were IL-10 as well as the immunosuppressive factor IDO-1 at both mRNA and protein levels (79). This interaction between tumor cells and chemokines is not dissimilar to that seen in some pituitary neuroendocrine tumors (80).

In summary, in the era of molecular-targeted treatments, the molecular pathways that characterize either ACPs or PCPs are under vigorous investigation. Currently, ACPs are characterized and identified by activating mutations of the β-catenin–encoding gene CTNNB1, whereas PCPs are characterized by the activating BRAF-V600E mutation. In both cases, there is upregulation of the MAPK pathway, which is critically involved in cell proliferation.

Targeting Molecular Aberrations in CPs

Currently, there are no approved targeted or cytotoxic therapies available for the systemic treatment of CPs. The development of molecular-targeted treatments in oncology has opened new horizons in the pharmacological treatment of tumors caused by genetic alterations. Recently, in cancers harboring BRAF-V600E alterations, V600E mutation–specific BRAF inhibitors, such as dabrafenib or vemurafenib (81, 82), have been shown to be effective antitumor agents. Additionally, the MEK inhibitors trametinib and cobimetinib have been administered in combination with BRAF inhibitors, because these compounds override any resistance to BRAF inhibition (81, 83). These data have led to the use of BRAF inhibitors in patients with aggressive PCPs (see Table 1 for summary). Aylwin et al. (83) described the beneficial effect of the off-label administration of vemurafenib (960 mg twice daily) in a 41-year-old female with a 16-year history of a recurrent PCP (carrying a BRAF-V600E mutation) with progressive visual failure following three transsphenoidal operations. MRI 2 weeks after starting treatment showed marked reduction in the size of the tumor with resolution of the surrounding edema, and 3 months later there was evidence of radiological improvement. The CP recurred 6 weeks later and vemuranefib was restarted with reduction of tumor size followed by stabilization for 7 months, but this was followed by further growth. In the same year, Brastianos et al. (84) reported on the use of dabrafenib (150 mg twice daily) in a 39-year-old male with a PCP harboring a BRAF-V600E mutation. This patient had undergone four craniotomies along with cyst decompression during a period of 11 months: as early as 4 days after treatment, the tumor showed a 23% size reduction, with the cyst volume having decreased by 32%. The beneficial effect was even more obvious 17 days after treatment, achieving a 52% reduction in tumor size and 70% decrease in the cyst volume. The MEK inhibitor trametinib (2 mg, orally, twice daily) was added to the therapeutic scheme on day 21 aiming to enhance the effect. Fourteen days after this combined therapy, there was an 85% reduction in tumor size and 81% decrease in the cyst volume. On day 38 of treatment, the patient underwent endoscopic transsphenoidal surgery: 2 weeks later the combined treatment was discontinued, and a week later RT was offered (50.4 Gy in 28 fractions). Seven months later, the patient has remained free of symptomatic recurrence. In the following year, Roque and Odia (29) described a 47-year-old female with acute visual loss and a 4-month history of amenorrhea, cold intolerance, and headache. A 2.7-cm cystic lesion was identified in the suprasellar area harboring a BRAF-V600E mutation (29). Subsequently, she underwent right frontal craniotomy, but the tumor regrew, causing bilateral visual impairment necessitating an Ommaya catheter placement into the cyst and then RT (54 Gy in 30 fractions). Seven months later, the tumor regrew and dabrafenib (150 mg twice daily) and trametinib (2 mg daily) were administered. After 2 months, a 52% reduction in tumor volume was documented, and after 5 months, the tumor size was reduced by 75%. The patient was continuing to improve on this treatment both radiologically and clinically 7 months posttreatment. Subsequently, Rostami et al. (85) reported a 65-year-old male with visual deficits who underwent a partial transsphenoidal resection of a PCP harboring a BRAF-V600E mutation. After rapid tumor growth, dabrafenib (150 mg twice daily) was initiated, followed by trametinib (2 mg daily) 3 weeks later for a total of 7 weeks. An 11% tumor reduction was seen at 4 weeks and 91% by the 8th week.

Table 1.

Case Reports and Current Clinical Trials of Patients With PCPs and BRAF-V600E Alterations Treated With V600E Mutation-Specific BRAF Inhibitors (Dabrafenib or Vemurafenib) With or Without Combined Treatment With MEK Inhibitors (Trametinib or Cobimetinib)

Case Series/ReportsCPPrevious TreatmentHistopathological And Molecular Genetic AnalysisTreatmentDuration Of TreatmentOutcome (Mean Of Measurement)Comments
Aylwin et al., 2015 (83)PCPTSS ×2, RTPyrosequencing analysis indicated the presence of BRAF mutation c.1799T>A (p.Val600Glu).Vemurafenib 960 mg bd3 moNear complete resolution (MRI)Recurrence 6 wk off treatment; restart of treatment and stabilization for 7 mo with a regrowth
Brastianos et al., 2015 (84)PCPCraniotomy ×3Uniform staining of the BRAFV600E protein (VE1 antibody) by IHC; V600E mutation confirmed by allele-specific genetic testing. Mutation detectable in peripheral blood during treatment. Absence of nuclear staining for β-catenin by IHC.Dabrafenib 150 mg bd + after 3 wk trametinib 2 mg od35 d (followed by other treatments)85% reduction in tumor volume and 81% reduction in tumor-associated
cyst (MRI)
Roque et al., 2016 (29)PCPCraniotomy, Ommaya catheter with percutaneous aspiration of cyst fluid, RT 54 Gy in 30 fractionsPositive staining for BRAFV600E by IHC; next-generation sequencing confirmed BRAFV600E mutation and FGFR3 splice site (437_445 + 3del12) with two variants of unknown significance (BRCA2-G1771D and FGFR4-S551F).Dabrafenib 150 mg orally bd and trametinib 2 mg orally od7 moAlmost disappearance of 2.6 × 2.3 × 3.2-cm tumor (MRI); reduced volume and intensity of FDG uptake on PET
Rostami et al., 2017 (85)PCPTSSWeak staining of mutated BRAFV600E (VE1 antibody) by IHC; BRAFV600E genotype confirmed by pyrosequencing mutational analysis.Dabrafenib 150 mg bd plus after 3 wk trametinib 2 mg od7 wk91% reduction tumor size (MRI)Combined BRAF and MEK-targeted therapy
Himes et al., 2019 (86)PCPCraniotomy, RT 36 Gy in 12 fractionsBRAF V600E mutation confirmed in specimens from original resection.Dabrafenib 150 mg bd (shortly) → od (several weeks) → 225 mg od1 y under treatment and follow-up for 1 y off treatment.Minimal residual tumor
Juratli et al., 2019 (87)PCPNo treatmentBRAF V600E confirmed by biopsyDabrafenib 150 mg bd plus trametinib 2 mg od6 mo>80% reduction of tumor size (MRI)Characterized as neoadjuvant therapy
NCT03224767 (88, 89)PCPCohort A: with/without surgeryIHC for BRAF V600E mutation (VE1) and β-catenin IHC (membranous, nonnuclear pattern)Vemurafenib and cobimetinibVemurafenib bd days 1–28 plus cobimetinibEvery 28 d for up to five courses in the absence of disease progression or unacceptable toxicity (MRI)Patients may then receive RT, surgery, or continued treatment
Cohort B: RT with/without other treatment (except for BRAF or MEK inhibitors)od on days 1–21
Case Series/ReportsCPPrevious TreatmentHistopathological And Molecular Genetic AnalysisTreatmentDuration Of TreatmentOutcome (Mean Of Measurement)Comments
Aylwin et al., 2015 (83)PCPTSS ×2, RTPyrosequencing analysis indicated the presence of BRAF mutation c.1799T>A (p.Val600Glu).Vemurafenib 960 mg bd3 moNear complete resolution (MRI)Recurrence 6 wk off treatment; restart of treatment and stabilization for 7 mo with a regrowth
Brastianos et al., 2015 (84)PCPCraniotomy ×3Uniform staining of the BRAFV600E protein (VE1 antibody) by IHC; V600E mutation confirmed by allele-specific genetic testing. Mutation detectable in peripheral blood during treatment. Absence of nuclear staining for β-catenin by IHC.Dabrafenib 150 mg bd + after 3 wk trametinib 2 mg od35 d (followed by other treatments)85% reduction in tumor volume and 81% reduction in tumor-associated
cyst (MRI)
Roque et al., 2016 (29)PCPCraniotomy, Ommaya catheter with percutaneous aspiration of cyst fluid, RT 54 Gy in 30 fractionsPositive staining for BRAFV600E by IHC; next-generation sequencing confirmed BRAFV600E mutation and FGFR3 splice site (437_445 + 3del12) with two variants of unknown significance (BRCA2-G1771D and FGFR4-S551F).Dabrafenib 150 mg orally bd and trametinib 2 mg orally od7 moAlmost disappearance of 2.6 × 2.3 × 3.2-cm tumor (MRI); reduced volume and intensity of FDG uptake on PET
Rostami et al., 2017 (85)PCPTSSWeak staining of mutated BRAFV600E (VE1 antibody) by IHC; BRAFV600E genotype confirmed by pyrosequencing mutational analysis.Dabrafenib 150 mg bd plus after 3 wk trametinib 2 mg od7 wk91% reduction tumor size (MRI)Combined BRAF and MEK-targeted therapy
Himes et al., 2019 (86)PCPCraniotomy, RT 36 Gy in 12 fractionsBRAF V600E mutation confirmed in specimens from original resection.Dabrafenib 150 mg bd (shortly) → od (several weeks) → 225 mg od1 y under treatment and follow-up for 1 y off treatment.Minimal residual tumor
Juratli et al., 2019 (87)PCPNo treatmentBRAF V600E confirmed by biopsyDabrafenib 150 mg bd plus trametinib 2 mg od6 mo>80% reduction of tumor size (MRI)Characterized as neoadjuvant therapy
NCT03224767 (88, 89)PCPCohort A: with/without surgeryIHC for BRAF V600E mutation (VE1) and β-catenin IHC (membranous, nonnuclear pattern)Vemurafenib and cobimetinibVemurafenib bd days 1–28 plus cobimetinibEvery 28 d for up to five courses in the absence of disease progression or unacceptable toxicity (MRI)Patients may then receive RT, surgery, or continued treatment
Cohort B: RT with/without other treatment (except for BRAF or MEK inhibitors)od on days 1–21

Abbreviations: bd, twice a day; FDG, 18fluoro-d-glucose; od, once a day; PET, positron emission tomography; TSS, transsphenoidal surgery.

Table 1.

Case Reports and Current Clinical Trials of Patients With PCPs and BRAF-V600E Alterations Treated With V600E Mutation-Specific BRAF Inhibitors (Dabrafenib or Vemurafenib) With or Without Combined Treatment With MEK Inhibitors (Trametinib or Cobimetinib)

Case Series/ReportsCPPrevious TreatmentHistopathological And Molecular Genetic AnalysisTreatmentDuration Of TreatmentOutcome (Mean Of Measurement)Comments
Aylwin et al., 2015 (83)PCPTSS ×2, RTPyrosequencing analysis indicated the presence of BRAF mutation c.1799T>A (p.Val600Glu).Vemurafenib 960 mg bd3 moNear complete resolution (MRI)Recurrence 6 wk off treatment; restart of treatment and stabilization for 7 mo with a regrowth
Brastianos et al., 2015 (84)PCPCraniotomy ×3Uniform staining of the BRAFV600E protein (VE1 antibody) by IHC; V600E mutation confirmed by allele-specific genetic testing. Mutation detectable in peripheral blood during treatment. Absence of nuclear staining for β-catenin by IHC.Dabrafenib 150 mg bd + after 3 wk trametinib 2 mg od35 d (followed by other treatments)85% reduction in tumor volume and 81% reduction in tumor-associated
cyst (MRI)
Roque et al., 2016 (29)PCPCraniotomy, Ommaya catheter with percutaneous aspiration of cyst fluid, RT 54 Gy in 30 fractionsPositive staining for BRAFV600E by IHC; next-generation sequencing confirmed BRAFV600E mutation and FGFR3 splice site (437_445 + 3del12) with two variants of unknown significance (BRCA2-G1771D and FGFR4-S551F).Dabrafenib 150 mg orally bd and trametinib 2 mg orally od7 moAlmost disappearance of 2.6 × 2.3 × 3.2-cm tumor (MRI); reduced volume and intensity of FDG uptake on PET
Rostami et al., 2017 (85)PCPTSSWeak staining of mutated BRAFV600E (VE1 antibody) by IHC; BRAFV600E genotype confirmed by pyrosequencing mutational analysis.Dabrafenib 150 mg bd plus after 3 wk trametinib 2 mg od7 wk91% reduction tumor size (MRI)Combined BRAF and MEK-targeted therapy
Himes et al., 2019 (86)PCPCraniotomy, RT 36 Gy in 12 fractionsBRAF V600E mutation confirmed in specimens from original resection.Dabrafenib 150 mg bd (shortly) → od (several weeks) → 225 mg od1 y under treatment and follow-up for 1 y off treatment.Minimal residual tumor
Juratli et al., 2019 (87)PCPNo treatmentBRAF V600E confirmed by biopsyDabrafenib 150 mg bd plus trametinib 2 mg od6 mo>80% reduction of tumor size (MRI)Characterized as neoadjuvant therapy
NCT03224767 (88, 89)PCPCohort A: with/without surgeryIHC for BRAF V600E mutation (VE1) and β-catenin IHC (membranous, nonnuclear pattern)Vemurafenib and cobimetinibVemurafenib bd days 1–28 plus cobimetinibEvery 28 d for up to five courses in the absence of disease progression or unacceptable toxicity (MRI)Patients may then receive RT, surgery, or continued treatment
Cohort B: RT with/without other treatment (except for BRAF or MEK inhibitors)od on days 1–21
Case Series/ReportsCPPrevious TreatmentHistopathological And Molecular Genetic AnalysisTreatmentDuration Of TreatmentOutcome (Mean Of Measurement)Comments
Aylwin et al., 2015 (83)PCPTSS ×2, RTPyrosequencing analysis indicated the presence of BRAF mutation c.1799T>A (p.Val600Glu).Vemurafenib 960 mg bd3 moNear complete resolution (MRI)Recurrence 6 wk off treatment; restart of treatment and stabilization for 7 mo with a regrowth
Brastianos et al., 2015 (84)PCPCraniotomy ×3Uniform staining of the BRAFV600E protein (VE1 antibody) by IHC; V600E mutation confirmed by allele-specific genetic testing. Mutation detectable in peripheral blood during treatment. Absence of nuclear staining for β-catenin by IHC.Dabrafenib 150 mg bd + after 3 wk trametinib 2 mg od35 d (followed by other treatments)85% reduction in tumor volume and 81% reduction in tumor-associated
cyst (MRI)
Roque et al., 2016 (29)PCPCraniotomy, Ommaya catheter with percutaneous aspiration of cyst fluid, RT 54 Gy in 30 fractionsPositive staining for BRAFV600E by IHC; next-generation sequencing confirmed BRAFV600E mutation and FGFR3 splice site (437_445 + 3del12) with two variants of unknown significance (BRCA2-G1771D and FGFR4-S551F).Dabrafenib 150 mg orally bd and trametinib 2 mg orally od7 moAlmost disappearance of 2.6 × 2.3 × 3.2-cm tumor (MRI); reduced volume and intensity of FDG uptake on PET
Rostami et al., 2017 (85)PCPTSSWeak staining of mutated BRAFV600E (VE1 antibody) by IHC; BRAFV600E genotype confirmed by pyrosequencing mutational analysis.Dabrafenib 150 mg bd plus after 3 wk trametinib 2 mg od7 wk91% reduction tumor size (MRI)Combined BRAF and MEK-targeted therapy
Himes et al., 2019 (86)PCPCraniotomy, RT 36 Gy in 12 fractionsBRAF V600E mutation confirmed in specimens from original resection.Dabrafenib 150 mg bd (shortly) → od (several weeks) → 225 mg od1 y under treatment and follow-up for 1 y off treatment.Minimal residual tumor
Juratli et al., 2019 (87)PCPNo treatmentBRAF V600E confirmed by biopsyDabrafenib 150 mg bd plus trametinib 2 mg od6 mo>80% reduction of tumor size (MRI)Characterized as neoadjuvant therapy
NCT03224767 (88, 89)PCPCohort A: with/without surgeryIHC for BRAF V600E mutation (VE1) and β-catenin IHC (membranous, nonnuclear pattern)Vemurafenib and cobimetinibVemurafenib bd days 1–28 plus cobimetinibEvery 28 d for up to five courses in the absence of disease progression or unacceptable toxicity (MRI)Patients may then receive RT, surgery, or continued treatment
Cohort B: RT with/without other treatment (except for BRAF or MEK inhibitors)od on days 1–21

Abbreviations: bd, twice a day; FDG, 18fluoro-d-glucose; od, once a day; PET, positron emission tomography; TSS, transsphenoidal surgery.

Very recently, Himes et al. (86) presented the longer-term effects of monotherapy with dabrafenib in a patient with a history of non-Hodgkin lymphoma and colon cancer who had a PCP with the BRAF-V600E mutation treated by cranial surgery and RT. Three years after the RT he developed tumor recurrence, and dabrafenib was initiated (150 mg twice daily, reduced to 150 mg daily due to joint pains, but then increased to 225 mg daily). Two months following treatment, an enlargement of the cystic component was documented in parallel with a reduction of the solid component. However, at 6 months posttreatment substantial reduction in the size of both components was identified, and by 9 months only minimal residual tumor was evident and this remained stable until 12 months; treatment was discontinued, and the tumor remained stable for further 12 months. A sixth case was recently reported at the European Congress of Endocrinology 2019 by Jurtali (87), who administered dabrafenib in a neoadjuvant setting to a 21-year-old male prior to surgery and found a >80% reduction of tumor size. All of these cases support the potential benefits of BRAF inhibitors, at least in achieving transient tumor responses, either as neoadjuvant treatment or following recurrence (Table 1). What is unclear from these single reports relates to the efficacy of different inhibitors, the mechanisms of tumor resistance, and the means to overcome such resistance. An interesting observation from one of these case reports was the documentation of a circulating BRAF-V600E mutation in the peripheral blood of the patient during treatment with the BRAF inhibitor (84). The authors stated that their finding of detectable BRAF-V600E in peripheral blood was unexpected, because there was not any previously reported case of circulating tumor cells or cell-free DNA in patients with intracranial benign tumors. However, if this is not a result of the surgical procedures or drug treatment, and the presence of BRAF-V600E can be detected in the blood before any treatment, then this assessment may be of value as a selection criterion of the patients who may benefit from BRAF-inhibitor neoadjuvant therapy (84). Following these observations, an ongoing cooperative group trial (Alliance A071601) is testing the combination of vemurafenib and cobimetinib in an open-label, phase 2 study in patients with BRAF-V600E–mutant CPs (88) to determine the frequency, durability, and extent of BRAF treatment responses in patients with PCPs. In this study, vemurafenib is administered twice daily on days 1 through 28, and cobimetinib is administered four times daily on days 1 through 21; treatment is repeated every 28 days to a maximum of five cycles as long as the disease shows a failure to progress and there is no unacceptable toxicity. Patients may then be treated with RT, surgery, or continue the combination of medical treatment. In another published series, a patient in whom GTR was achieved had a recurrence of a cystic tumor component after 2 years and has been referred to a phase 2 trial of combined BRAF/MEK inhibitors (NCT03224767) (88, 89).

Adverse effects of compounds currently used in the management of CPs

Data on the adverse effects profile of these treatments in patients with CPs are limited. The phenomenon of “pseudo-progression” with dabrafenib, defined as early initial cystic enlargement together with solid component reduction, followed by lesion shrinkage, has been observed, similar to that seen in gliomas after RT or chemotherapy (86). Additionally, transient fever has been described with BRAF inhibitors (29, 84, 85). Finally, the addition of the MEK inhibitor to BRAF inhibition may play a role in reducing the incidence of secondary squamous cell skin carcinomas, as previously shown in patients with melanoma (90).

Other potential treatments targeting CPs

Despite the identification of the deranged molecular pathways in ACP pathogenesis, drugs targeting β-catenin and its downstream MAPK pathway have been used only in in vitro studies. A transcriptome study used mRNA microarray gene expression analysis and unraveled several “druggable” pharmaceutical targets that were consistently overexpressed in a panel of 15 ACPs, compared with other brain tumors and normal brain tissue (26). Lymphocyte-specific protein tyrosine kinase (LCK), ephrin type-A2 (EPHA2), and SRC were identified as targets of the tyrosine kinase inhibitor dasatinib. AREG, EGFR, and ERBB-3 in the EGFR pathway may respond to inhibitors such as cetuximab, erlotinib, and lapatinib, and MMP9 and MMP12 could potentially respond to the oral MMP9/12 inhibitor AZD1236 (26, 50). Recurrence in ACPs has been associated with increased VEGF expression, suggesting a possible therapeutic role for the VEGF inhibitor bevacizumab (49, 61). Indeed, some of these targets can be shown to be expressed in the primary ACP tumor using Western blot analysis (26). Alternatively, although the SHH pathway may be implicated in the pathogenesis of ACPs, inhibition of this pathway may be tumorigenic (91). This relies on data showing that vismodegib, a well-established inhibitor of SHH, results in premature tumor formation and increased tumor cell proliferation in both genetically engineered and patient-derived xenograft mouse models (91). Inhibition of EGFR signaling by the tyrosine kinase inhibitor gefitinib reduced ACP cell migration in vitro by decreasing Fascin expression; this was attributed to the finding of nuclear colocalization of activated EGFR, β-catenin, and Fascin in ACP cell migration in vitro (48), implying crosstalk of these pathways in ACPs. Because annexin A2 expression, a Ca2+-regulated binding protein, correlates with gefitinib sensitivity in vitro, this may serve as a biomarker of a response to treatment (92). Small samples of human ACP tumors were cultured with and without trametinib, a specific MEK inhibitor; the inhibition of the MAPK/ERK pathway resulted in reduced proliferation along with a dose-dependent significant decrease in the Ki-67 proliferation index in the trametinib-treated tumors, suggesting a possible role in the management of not only PCPs but also ACPs (91). Notably, the well-established synergistic effects of combined targeted therapies in other neuroendocrine tumor models provide support for the potential use of combinatorial targeted therapy in CPs (93, 94).

Given the potential role of inflammation in the pathogenesis of CPs and previous studies on intratumor administration of IFN-α in patients with a CP (22), an open-label, phase 2 study is underway aiming to estimate the 1-year disease stabilization rate associated with the use of Peg–IFN-α2b in patients with progressive unresectable or recurrent CPs after surgery alone (without adjuvant RT). The study will also estimate the sustained objective response rate (partial and complete response) to Peg–IFN-α2b in patients with CPs that progress or recur following RT (95). Finally, noting the immune microenvironment in ACPs, IL-1R inhibitors, such as anakinra, may have a place in our therapeutic armamentarium (60).

Conclusions

The discovery of distinct oncogenic molecular signaling pathways in CPs has opened new avenues for the personalized treatment of patients with these challenging tumors. Τhe key pathogenetic event in ACPs appears to be Wnt activation along with alterations of the MEK/ERK pathway, whereas in PCPs there is primary activation of the Ras/Raf/MEK/ERK pathway by BRAF-V600E mutations. Current literature on PCPs includes single case reports, with both BRAF and MAPK inhibitors, in patients with V600E mutations. Their results are promising, but well-designed studies are needed that aim to provide robust data on treatment regimens and protocols, as well as on efficacy and safety. It also remains unclear whether single or combination therapies will be necessary, although in vitro data from other endocrine tumors have highlighted the use of multiple simultaneous combination treatments. The use of MAPK inhibitors for the more common ACPs also requires assessment in clinical trials, particularly in view of the risk of acquired drug resistance. The value of combination therapies aiming to target multiple molecular mediators implicated in the pathogenesis of ACP and possibly minimizing drug toxicities also requires further investigation.

Nevertheless, the recently expanding data on the pathogenesis of CPs offer substantial ground for the translation of molecular insights into practical therapies for primary, recurrent, or very aggressive tumors that will, hopefully, improve the prognosis of the patients. The eventual use of neoadjuvant medical treatment or even an entirely nonoperative treatment strategy for CPs should improve the quality of life of these patients, minimizing the neurologic and endocrinological sequelae caused either by the tumor extension or by the neurosurgical or radiotherapeutic complications. Combination therapy strategies could be also be effective in cases of ACPs to target the multiple molecular mediators that are implicated in ACP pathogenesis and to mimimize any adverse treatment effects.

Additional Information

Disclosure Summary: The authors have nothing to disclose.

Data Availability: All data generated or analyzed during this study are included in this published article or in the data repositories listed in References.

Abbreviations:

    Abbreviations:
     
  • ACP

    adamantinomatous craniopharyngioma

  •  
  • APC

    adenomatosis polyposis coli

  •  
  • BMP

    bone morphogenetic protein

  •  
  • CK1

    casein kinase 1α

  •  
  • CP

    craniopharyngioma

  •  
  • EGF

    epidermal growth factor

  •  
  • EGFR

    epidermal growth factor receptor

  •  
  • FGF

    fibroblast growth factor

  •  
  • GSK-3β

    glycogen synthase kinase-3β

  •  
  • GTR

    gross total resection

  •  
  • IFN

    interferon

  •  
  • IHC

    immunocytochemistry

  •  
  • MeSH

    medical subject headings

  •  
  • MMP

    matrix metallopeptidase

  •  
  • PCP

    papillary craniopharyngioma

  •  
  • PDGF

    platelet-derived growth factor

  •  
  • RT

    radiotherapy

  •  
  • SHH

    sonic hedgehog

  •  
  • STR

    subtotal/partial resection

  •  
  • VEGF

    vascular endothelial growth factor

References and Notes

1.

Karavitaki
N
,
Cudlip
S
,
Adams
CB
,
Wass
JA
.
Craniopharyngiomas
.
Endocr Rev
.
2006
;
27
(
4
):
371
397
.

2.

Louis
DN
,
Ohgaki
H
,
Wiestler
OD
,
Cavenee
WK.
WHO Classification of Tumours of the Central Nervous System
. Revised 4th ed.
Lyon, France
:
International Agency for Research on Cancer
;
2016
:
324
328
.

3.

Lithgow
K
,
Pohl
U
,
Karavitaki
N
.
Craniopharyngiomas. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, Korbonits M, McLachlan R, Morley JE, New M, Perreault L, Purnell J, Rebar R, Singer F, Trence DL, Vinik A, Wilson DP, eds. Endotext [Internet].
South Dartmouth, MA
:
MDText.com, Inc
. Available at: https://www.ncbi.nlm.nih.gov/books/NBK538819/. Accessed 6 October 2019
.

4.

Bunin
GR
,
Surawicz
TS
,
Witman
PA
,
Preston-Martin
S
,
Davis
F
,
Bruner
JM
.
The descriptive epidemiology of craniopharyngioma
.
J Neurosurg
.
1998
;
89
(
4
):
547
551
.

5.

Bülow
B
,
Attewell
R
,
Hagmar
L
,
Malmström
P
,
Nordström
CH
,
Erfurth
EM
.
Postoperative prognosis in craniopharyngioma with respect to cardiovascular mortality, survival, and tumor recurrence
.
J Clin Endocrinol Metab
.
1998
;
83
(
11
):
3897
3904
.

6.

Nielsen
EH
,
Feldt-Rasmussen
U
,
Poulsgaard
L
,
Kristensen
LO
,
Astrup
J
,
Jørgensen
JO
,
Bjerre
P
,
Andersen
M
,
Andersen
C
,
Jørgensen
J
,
Lindholm
J
,
Laurberg
P
.
Incidence of craniopharyngioma in Denmark (n = 189) and estimated world incidence of craniopharyngioma in children and adults
.
J Neurooncol
.
2011
;
104
(
3
):
755
763
.

7.

Olsson
DS
,
Andersson
E
,
Bryngelsson
IL
,
Nilsson
AG
,
Johannsson
G
.
Excess mortality and morbidity in patients with craniopharyngioma, especially in patients with childhood onset: a population-based study in Sweden
.
J Clin Endocrinol Metab
.
2015
;
100
(
2
):
467
474
.

8.

Hölsken
A
.
Pathogenesis of human ACP
. In:
Martinez-Barbera
JP
,
Andoniadou
CL
, eds.
Basic Research and Clinical Aspects of Adamantinomatous Craniopharyngioma
.
Cham, Switzerland
:
Springer International Publishing
;
2017
:
1
26
.

9.

Bailey
W
,
Freidenberg
GR
,
James
HE
,
Hesselink
JR
,
Jones
KL
.
Prenatal diagnosis of a craniopharyngioma using ultrasonography and magnetic resonance imaging
.
Prenat Diagn
.
1990
;
10
(
10
):
623
629
.

10.

Müller-Scholden
J
,
Lehrnbecher
T
,
Müller
HL
,
Bensch
J
,
Hengen
RH
,
Sörensen
N
,
Stockhausen
HB
.
Radical surgery in a neonate with craniopharyngioma. Report of a case
.
Pediatr Neurosurg
.
2000
;
33
(
5
):
265
269
.

11.

Nielsen
EH
,
Lindholm
J
,
Laurberg
P
.
Excess mortality in women with pituitary disease: a meta-analysis
.
Clin Endocrinol (Oxf)
.
2007
;
67
(
5
):
693
697
.

12.

Borrill
R
,
Cheesman
E
,
Stivaros
S
,
Kamaly-Asl
ID
,
Gnanalingham
K
,
Kilday
JP
.
Papillary craniopharyngioma in a 4-year-old girl with BRAF V600E mutation: a case report and review of the literature
.
Childs Nerv Syst
.
2019
;
35
(
1
):
169
173
.

13.

Cushing
H
.
Intracranial Tumors. Notes Upon Series of Two Thousand Verified Cases With Surgical Mortality Percentages Pertaining Thereto
.
Springfield, IL
:
Thomas
;
1932
.

14.

Vardon
AJ
,
Karavitaki
N
.
Clinical diagnosis of human ACP
. In:
Martinez-Barbera
JP
,
Andoniadou
CL
, eds.
Basic Research and Clinical Aspects of Adamantinomatous Craniopharyngioma
.
Cham, Switzerland
:
Springer International Publishing
;
2017
:
57
66
.

15.

Erfurth
EM
.
Obesity and metabolic disturbances in adamantinomatous craniopharyngioma patients
. In:
Martinez-Barbera
JP
,
Andoniadou
CL
, eds.
Basic Research and Clinical Aspects of Adamantinomatous Craniopharyngioma
.
Cham, Switzerland
:
Springer International Publishing
;
2017
:
85
99
.

16.

Asa
SL
,
Casar-Borota
O
,
Chanson
P
,
Delgrange
E
,
Earls
P
,
Ezzat
S
,
Grossman
A
,
Ikeda
H
,
Inoshita
N
,
Karavitaki
N
,
Korbonits
M
,
Laws
ER
Jr
,
Lopes
MB
,
Maartens
N
,
McCutcheon
IE
,
Mete
O
,
Nishioka
H
,
Raverot
G
,
Roncaroli
F
,
Saeger
W
,
Syro
LV
,
Vasiljevic
A
,
Villa
C
,
Wierinckx
A
,
Trouillas
J
;
attendees of 14th Meeting of the International Pituitary Pathology Club, Annecy, France, November 2016
.
From pituitary adenoma to pituitary neuroendocrine tumor (PitNET): an International Pituitary Pathology Club proposal
.
Endocr Relat Cancer
.
2017
;
24
(
4
):
C5
C8
.

17.

Aquilina
K
,
Buchfelder
M
.
Surgical treatment of human ACP
. In:
Martinez-Barbera
JP
,
Andoniadou
CL
, eds.
Basic Research and Clinical Aspects of Adamantinomatous Craniopharyngioma
.
Cham, Switzerland
:
Springer International Publishing
;
2017
:
137
158
.

18.

Müller
HL
.
Long-term management and clinical trials in adamantinomatous craniopharyngioma
. In:
Martinez-Barbera
JP
,
Andoniadou
CL
, eds.
Basic Research and Clinical Aspects of Adamantinomatous Craniopharyngioma
.
Cham, Switzerland
:
Springer International Publishing
;
2017
:
179
214
.

19.

Jyoti
B
,
Indelicato
DJ
,
Bradley
JA
,
Rotondo
RL
.
Radiology and radiotherapy of craniopharyngioma
. In:
Martinez-Barbera
JP
,
Andoniadou
CL
, eds.
Basic Research and Clinical Aspects of Adamantinomatous Craniopharyngioma
.
Cham, Switzerland
:
Springer International Publishing
;
2017
:
101
135
.

20.

Yang
I
,
Sughrue
ME
,
Rutkowski
MJ
,
Kaur
R
,
Ivan
ME
,
Aranda
D
,
Barani
IJ
,
Parsa
AT
.
Craniopharyngioma: a comparison of tumor control with various treatment strategies
.
Neurosurg Focus
.
2010
;
28
(
4
):
E5
.

21.

Kilday
JP
,
Bartels
U
.
Intracystic administration of interferon-alpha for reduction of cystic tumour burden
. In:
Martinez-Barbera
JP
,
Andoniadou
CL
, eds.
Basic Research and Clinical Aspects of Adamantinomatous Craniopharyngioma
.
Cham, Switzerland
:
Springer International Publishing
;
2017
:
159
177
.

22.

Mrowczynski
OD
,
Langan
ST
,
Rizk
EB
.
Craniopharyngiomas: a systematic review and evaluation of the current intratumoral treatment landscape
.
Clin Neurol Neurosurg
.
2018
;
166
:
124
130
.

23.

Pettorini
BL
,
Inzitari
R
,
Massimi
L
,
Tamburrini
G
,
Caldarelli
M
,
Fanali
C
,
Cabras
T
,
Messana
I
,
Castagnola
M
,
Di Rocco
C
.
The role of inflammation in the genesis of the cystic component of craniopharyngiomas
.
Childs Nerv Syst
.
2010
;
26
(
12
):
1779
1784
.

24.

Kilday
JP
,
Caldarelli
M
,
Massimi
L
,
Chen
RH
,
Lee
YY
,
Liang
ML
,
Parkes
J
,
Naiker
T
,
van Veelen
ML
,
Michiels
E
,
Mallucci
C
,
Pettorini
B
,
Meijer
L
,
Dorfer
C
,
Czech
T
,
Diezi
M
,
Schouten-van Meeteren
AY
,
Holm
S
,
Gustavsson
B
,
Benesch
M
,
Müller
HL
,
Hoffmann
A
,
Rutkowski
S
,
Flitsch
J
,
Escherich
G
,
Grotzer
M
,
Spoudeas
HA
,
Azquikina
K
,
Capra
M
,
Jiménez-Guerra
R
,
MacDonald
P
,
Johnston
DL
,
Dvir
R
,
Constantini
S
,
Kuo
MF
,
Yang
SH
,
Bartels
U
.
Intracystic interferon-alpha in pediatric craniopharyngioma patients: an international multicenter assessment on behalf of SIOPE and ISPN
.
Neuro-Oncol
.
2017
;
19
(
10
):
1398
1407
.

25.

Brastianos
PK
,
Taylor-Weiner
A
,
Manley
PE
,
Jones
RT
,
Dias-Santagata
D
,
Thorner
AR
,
Lawrence
MS
,
Rodriguez
FJ
,
Bernardo
LA
,
Schubert
L
,
Sunkavalli
A
,
Shillingford
N
,
Calicchio
ML
,
Lidov
HG
,
Taha
H
,
Martinez-Lage
M
,
Santi
M
,
Storm
PB
,
Lee
JY
,
Palmer
JN
,
Adappa
ND
,
Scott
RM
,
Dunn
IF
,
Laws
ER
Jr
,
Stewart
C
,
Ligon
KL
,
Hoang
MP
,
Van Hummelen
P
,
Hahn
WC
,
Louis
DN
,
Resnick
AC
,
Kieran
MW
,
Getz
G
,
Santagata
S
.
Exome sequencing identifies BRAF mutations in papillary craniopharyngiomas
.
Nat Genet
.
2014
;
46
(
2
):
161
165
.

26.

Gump
JM
,
Donson
AM
,
Birks
DK
,
Amani
VM
,
Rao
KK
,
Griesinger
AM
,
Kleinschmidt-DeMasters
BK
,
Johnston
JM
,
Anderson
RC
,
Rosenfeld
A
,
Handler
M
,
Gore
L
,
Foreman
N
,
Hankinson
TC
.
Identification of targets for rational pharmacological therapy in childhood craniopharyngioma
.
Acta Neuropathol Commun
.
2015
;
3
(
1
):
30
.

27.

Goschzik
T
,
Gessi
M
,
Dreschmann
V
,
Gebhardt
U
,
Wang
L
,
Yamaguchi
S
,
Wheeler
DA
,
Lauriola
L
,
Lau
CC
,
Müller
HL
,
Pietsch
T
.
Genomic alterations of adamantinomatous and papillary craniopharyngioma
.
J Neuropathol Exp Neurol
.
2017
;
76
(
2
):
126
134
.

28.

Hölsken
A
,
Sill
M
,
Merkle
J
,
Schweizer
L
,
Buchfelder
M
,
Flitsch
J
,
Fahlbusch
R
,
Metzler
M
,
Kool
M
,
Pfister
SM
,
von Deimling
A
,
Capper
D
,
Jones
DT
,
Buslei
R
.
Adamantinomatous and papillary craniopharyngiomas are characterized by distinct epigenomic as well as mutational and transcriptomic profiles
.
Acta Neuropathol Commun
.
2016
;
4
(
1
):
20
.

29.

Roque
A
,
Odia
Y
.
BRAF-V600E mutant papillary craniopharyngioma dramatically responds to combination BRAF and MEK inhibitors
.
CNS Oncol
.
2017
;
6
(
2
):
95
99
.

30.

Sekine
S
,
Shibata
T
,
Kokubu
A
,
Morishita
Y
,
Noguchi
M
,
Nakanishi
Y
,
Sakamoto
M
,
Hirohashi
S
.
Craniopharyngiomas of adamantinomatous type harbor β-catenin gene mutations
.
Am J Pathol
.
2002
;
161
(
6
):
1997
2001
.

31.

Hofmann
BM
,
Kreutzer
J
,
Saeger
W
,
Buchfelder
M
,
Blümcke
I
,
Fahlbusch
R
,
Buslei
R
.
Nuclear β-catenin accumulation as reliable marker for the differentiation between cystic craniopharyngiomas and rathke cleft cysts: a clinico-pathologic approach
.
Am J Surg Pathol
.
2006
;
30
(
12
):
1595
1603
.

32.

Martinez-Barbera
JP
.
Molecular and cellular pathogenesis of adamantinomatous craniopharyngioma
.
Neuropathol Appl Neurobiol
.
2015
;
41
(
6
):
721
732
.

33.

Kemler
R
.
From cadherins to catenins: cytoplasmic protein interactions and regulation of cell adhesion
.
Trends Genet
.
1993
;
9
(
9
):
317
321
.

34.

Larkin
SJ
,
Ansorge
O
.
Pathology and pathogenesis of craniopharyngiomas
.
Pituitary
.
2013
;
16
(
1
):
9
17
.

35.

Cani
CM
,
Matushita
H
,
Carvalho
LR
,
Soares
IC
,
Brito
LP
,
Almeida
MQ
,
Mendonça
BB
.
PROP1 and CTNNB1 expression in adamantinomatous craniopharyngiomas with or without β-catenin mutations
.
Clinics (São Paulo)
.
2011
;
66
(
11
):
1849
1854
.

36.

Hölsken
A
,
Buchfelder
M
,
Fahlbusch
R
,
Blümcke
I
,
Buslei
R
.
Tumour cell migration in adamantinomatous craniopharyngiomas is promoted by activated Wnt-signalling
.
Acta Neuropathol
.
2010
;
119
(
5
):
631
639
.

37.

Behrens
J
,
Jerchow
BA
,
Würtele
M
,
Grimm
J
,
Asbrand
C
,
Wirtz
R
,
Kühl
M
,
Wedlich
D
,
Birchmeier
W
.
Functional interaction of an axin homolog, conductin, with β-catenin, APC, and GSK3
β.
Science
.
1998
;
280
(
5363
):
596
599
.

38.

Rubinfeld
B
,
Albert
I
,
Porfiri
E
,
Fiol
C
,
Munemitsu
S
,
Polakis
P
.
Binding of GSK3β to the APC-β-catenin complex and regulation of complex assembly
.
Science
.
1996
;
272
(
5264
):
1023
1026
.

39.

Rubinfeld
B
,
Souza
B
,
Albert
I
,
Müller
O
,
Chamberlain
SH
,
Masiarz
FR
,
Munemitsu
S
,
Polakis
P
.
Association of the APC gene product with beta-catenin
.
Science
.
1993
;
262
(
5140
):
1731
1734
.

40.

Lilien
J
,
Balsamo
J
.
The regulation of cadherin-mediated adhesion by tyrosine phosphorylation/dephosphorylation of β-catenin
.
Curr Opin Cell Biol
.
2005
;
17
(
5
):
459
465
.

41.

Giles
RH
,
van Es
JH
,
Clevers
H
.
Caught up in a Wnt storm: Wnt signaling in cancer
.
Biochim Biophys Acta
.
2003
;
1653
(
1
):
1
24
.

42.

Hölsken
A
,
Stache
C
,
Schlaffer
SM
,
Flitsch
J
,
Fahlbusch
R
,
Buchfelder
M
,
Buslei
R
.
Adamantinomatous craniopharyngiomas express tumor stem cell markers in cells with activated Wnt signaling: further evidence for the existence of a tumor stem cell niche
?
Pituitary
.
2014
;
17
(
6
):
546
556
.

43.

Esheba
GE
,
Hassan
AA
.
Comparative immunohistochemical expression of β-catenin, EGFR, ErbB2, and p63 in adamantinomatous and papillary craniopharyngiomas
.
J Egypt Natl Canc Inst
.
2015
;
27
(
3
):
139
145
.

44.

Buslei
R
,
Nolde
M
,
Hofmann
B
,
Meissner
S
,
Eyupoglu
IY
,
Siebzehnrübl
F
,
Hahnen
E
,
Kreutzer
J
,
Fahlbusch
R
.
Common mutations of β-catenin in adamantinomatous craniopharyngiomas but not in other tumours originating from the sellar region
.
Acta Neuropathol
.
2005
;
109
(
6
):
589
597
.

45.

Andoniadou
CL
,
Martinez-Barbera
JP
.
Genetically modified mouse models of adamantinomatous craniopharyngioma
. In:
Martinez-Barbera
JP
,
Andoniadou
CL
, eds.
Basic Research and Clinical Aspects of Adamantinomatous Craniopharyngioma
.
Cham, Switzerland
:
Springer International Publishing
;
2017
:
41
56
.

46.

Chen
Z
,
He
X
,
Jia
M
,
Liu
Y
,
Qu
D
,
Wu
D
,
Wu
P
,
Ni
C
,
Zhang
Z
,
Ye
J
,
Xu
J
,
Huang
J
.
β-Catenin overexpression in the nucleus predicts progress disease and unfavourable survival in colorectal cancer: a meta-analysis
.
PLoS One
.
2013
;
8
(
5
):
e63854
.

47.

Weiss
V
,
Dueber
J
,
Wright
JP
,
Cates
J
,
Revetta
F
,
Parikh
AA
,
Merchant
NB
,
Shi
C
.
Immunohistochemical analysis of the Wnt/β-catenin signaling pathway in pancreatic neuroendocrine neoplasms
.
World J Gastrointest Oncol
.
2016
;
8
(
8
):
615
622
.

48.

Hölsken
A
,
Gebhardt
M
,
Buchfelder
M
,
Fahlbusch
R
,
Blümcke
I
,
Buslei
R
.
EGFR signaling regulates tumor cell migration in craniopharyngiomas
.
Clin Cancer Res
.
2011
;
17
(
13
):
4367
4377
.

49.

Gupta
S
,
Bi
WL
,
Giantini Larsen
A
,
Al-Abdulmohsen
S
,
Abedalthagafi
M
,
Dunn
IF
.
Craniopharyngioma: a roadmap for scientific translation
.
Neurosurg Focus
.
2018
;
44
(
6
):
E12
.

50.

Robinson
L
,
Santagata
S
,
Hankinson
TC
.
Transcriptomic and genomic analyses of human craniopharyngioma
. In:
Martinez-Barbera
JP
,
Andoniadou
CL
, eds.
Basic Research and Clinical Aspects of Adamantinomatous Craniopharyngioma
.
Cham, Switzerland
:
Springer International Publishing
;
2017
:
27
39
.

51.

Koike
M
,
Kose
S
,
Furuta
M
,
Taniguchi
N
,
Yokoya
F
,
Yoneda
Y
,
Imamoto
N
.
β-Catenin shows an overlapping sequence requirement but distinct molecular interactions for its bidirectional passage through nuclear pores
.
J Biol Chem
.
2004
;
279
(
32
):
34038
34047
.

52.

Hölsken
A
,
Kreutzer
J
,
Hofmann
BM
,
Hans
V
,
Oppel
F
,
Buchfelder
M
,
Fahlbusch
R
,
Blümcke
I
,
Buslei
R
.
Target gene activation of the Wnt signaling pathway in nuclear β-catenin accumulating cells of adamantinomatous craniopharyngiomas
.
Brain Pathol
.
2009
;
19
(
3
):
357
364
.

53.

Andoniadou
CL
,
Gaston-Massuet
C
,
Reddy
R
,
Schneider
RP
,
Blasco
MA
,
Le Tissier
P
,
Jacques
TS
,
Pevny
LH
,
Dattani
MT
,
Martinez-Barbera
JP
.
Identification of novel pathways involved in the pathogenesis of human adamantinomatous craniopharyngioma
.
Acta Neuropathol
.
2012
;
124
(
2
):
259
271
.

54.

Gaston-Massuet
C
,
Andoniadou
CL
,
Signore
M
,
Jayakody
SA
,
Charolidi
N
,
Kyeyune
R
,
Vernay
B
,
Jacques
TS
,
Taketo
MM
,
Le Tissier
P
,
Dattani
MT
,
Martinez-Barbera
JP
.
Increased Wingless (Wnt) signaling in pituitary progenitor/stem cells gives rise to pituitary tumors in mice and humans
.
Proc Natl Acad Sci USA
.
2011
;
108
(
28
):
11482
11487
.

55.

Sekine
S
,
Takata
T
,
Shibata
T
,
Mori
M
,
Morishita
Y
,
Noguchi
M
,
Uchida
T
,
Kanai
Y
,
Hirohashi
S
.
Expression of enamel proteins and LEF1 in adamantinomatous craniopharyngioma: evidence for its odontogenic epithelial differentiation
.
Histopathology
.
2004
;
45
(
6
):
573
579
.

56.

Roura
S
,
Miravet
S
,
Piedra
J
,
García de Herreros
A
,
Duñach
M
.
Regulation of E-cadherin/catenin association by tyrosine phosphorylation
.
J Biol Chem
.
1999
;
274
(
51
):
36734
36740
.

57.

Mimeault
M
,
Batra
SK
.
Frequent deregulations in the hedgehog signaling network and cross-talks with the epidermal growth factor receptor pathway involved in cancer progression and targeted therapies
.
Pharmacol Rev
.
2010
;
62
(
3
):
497
524
.

58.

Reya
T
,
Clevers
H
.
Wnt signalling in stem cells and cancer
.
Nature
.
2005
;
434
(
7035
):
843
850
.

59.

Reya
T
,
Morrison
SJ
,
Clarke
MF
,
Weissman
IL
.
Stem cells, cancer, and cancer stem cells
.
Nature
.
2001
;
414
(
6859
):
105
111
.

60.

Apps
JR
,
Carreno
G
,
Gonzalez-Meljem
JM
,
Haston
S
,
Guiho
R
,
Cooper
JE
,
Manshaei
S
,
Jani
N
,
Hölsken
A
,
Pettorini
B
,
Beynon
RJ
,
Simpson
DM
,
Fraser
HC
,
Hong
Y
,
Hallang
S
,
Stone
TJ
,
Virasami
A
,
Donson
AM
,
Jones
D
,
Aquilina
K
,
Spoudeas
H
,
Joshi
AR
,
Grundy
R
,
Storer
LC
,
Korbonits
M
,
Hilton
DA
,
Tossell
K
,
Thavaraj
S
,
Ungless
MA
,
Gil
J
,
Buslei
R
,
Hankinson
T
,
Hargrave
D
,
Goding
C
,
Andoniadou
CL
,
Brogan
P
,
Jacques
TS
,
Williams
HJ
,
Martinez-Barbera
JP
.
Tumour compartment transcriptomics demonstrates the activation of inflammatory and odontogenic programmes in human adamantinomatous craniopharyngioma and identifies the MAPK/ERK pathway as a novel therapeutic target
.
Acta Neuropathol
.
2018
;
135
(
5
):
757
777
.

61.

Sun
HI
,
Akgun
E
,
Bicer
A
,
Ozkan
A
,
Bozkurt
SU
,
Kurtkaya
O
,
Koc
DY
,
Pamir
MN
,
Kilic
T
.
Expression of angiogenic factors in craniopharyngiomas: implications for tumor recurrence
.
Neurosurgery
.
2010
;
66
(
4
):
744
750, discussion 750
.

62.

Baker
JC
,
Beddington
RS
,
Harland
RM
.
Wnt signaling in Xenopus embryos inhibits Bmp4 expression and activates neural development
.
Genes Dev
.
1999
;
13
(
23
):
3149
3159
.

63.

Dhillon
AS
,
Hagan
S
,
Rath
O
,
Kolch
W
.
MAP kinase signalling pathways in cancer
.
Oncogene
.
2007
;
26
(
22
):
3279
3290
.

64.

Zhang
W
,
Liu
HT
.
MAPK signal pathways in the regulation of cell proliferation in mammalian cells
.
Cell Res
.
2002
;
12
(
1
):
9
18
.

65.

Herbst
RS
.
Review of epidermal growth factor receptor biology
.
Int J Radiat Oncol Biol Phys
.
2004
;
59
(
2
Suppl
):
21
26
.

66.

Laurent-Puig
P
,
Lievre
A
,
Blons
H
.
Mutations and response to epidermal growth factor receptor inhibitors
.
Clin Cancer Res
.
2009
;
15
(
4
):
1133
1139
.

67.

Futreal
PA
,
Coin
L
,
Marshall
M
,
Down
T
,
Hubbard
T
,
Wooster
R
,
Rahman
N
,
Stratton
MR
.
A census of human cancer genes
.
Nat Rev Cancer
.
2004
;
4
(
3
):
177
183
.

68.

Larkin
SJ
,
Preda
V
,
Karavitaki
N
,
Grossman
A
,
Ansorge
O
.
BRAF V600E mutations are characteristic for papillary craniopharyngioma and may coexist with CTNNB1-mutated adamantinomatous craniopharyngioma
.
Acta Neuropathol
.
2014
;
127
(
6
):
927
929
.

69.

Davies
H
,
Bignell
GR
,
Cox
C
,
Stephens
P
,
Edkins
S
,
Clegg
S
,
Teague
J
,
Woffendin
H
,
Garnett
MJ
,
Bottomley
W
,
Davis
N
,
Dicks
E
,
Ewing
R
,
Floyd
Y
,
Gray
K
,
Hall
S
,
Hawes
R
,
Hughes
J
,
Kosmidou
V
,
Menzies
A
,
Mould
C
,
Parker
A
,
Stevens
C
,
Watt
S
,
Hooper
S
,
Wilson
R
,
Jayatilake
H
,
Gusterson
BA
,
Cooper
C
,
Shipley
J
,
Hargrave
D
,
Pritchard-Jones
K
,
Maitland
N
,
Chenevix-Trench
G
,
Riggins
GJ
,
Bigner
DD
,
Palmieri
G
,
Cossu
A
,
Flanagan
A
,
Nicholson
A
,
Ho
JW
,
Leung
SY
,
Yuen
ST
,
Weber
BL
,
Seigler
HF
,
Darrow
TL
,
Paterson
H
,
Marais
R
,
Marshall
CJ
,
Wooster
R
,
Stratton
MR
,
Futreal
PA
.
Mutations of the BRAF gene in human cancer
.
Nature
.
2002
;
417
(
6892
):
949
954
.

70.

Haston
S
,
Pozzi
S
,
Carreno
G
,
Manshaei
S
,
Panousopoulos
L
,
Gonzalez-Meljem
JM
,
Apps
JR
,
Virasami
A
,
Thavaraj
S
,
Gutteridge
A
,
Forshew
T
,
Marais
R
,
Brandner
S
,
Jacques
TS
,
Andoniadou
CL
,
Martinez-Barbera
JP
.
MAPK pathway control of stem cell proliferation and differentiation in the embryonic pituitary provides insights into the pathogenesis of papillary craniopharyngioma
.
Development
.
2017
;
144
(
12
):
2141
2152
.

71.

Larkin
S
,
Karavitaki
N
.
Recent advances in molecular pathology of craniopharyngioma
.
F1000 Res
.
2017
;
6
:
1202
.

72.

Jones
DT
,
Kocialkowski
S
,
Liu
L
,
Pearson
DM
,
Ichimura
K
,
Collins
VP
.
Oncogenic RAF1 rearrangement and a novel BRAF mutation as alternatives to KIAA1549:BRAF fusion in activating the MAPK pathway in pilocytic astrocytoma
.
Oncogene
.
2009
;
28
(
20
):
2119
2123
.

73.

Tian
Y
,
Rich
BE
,
Vena
N
,
Craig
JM
,
Macconaill
LE
,
Rajaram
V
,
Goldman
S
,
Taha
H
,
Mahmoud
M
,
Ozek
M
,
Sav
A
,
Longtine
JA
,
Lindeman
NI
,
Garraway
LA
,
Ligon
AH
,
Stiles
CD
,
Santagata
S
,
Chan
JA
,
Kieran
MW
,
Ligon
KL
.
Detection of KIAA1549-BRAF fusion transcripts in formalin-fixed paraffin-embedded pediatric low-grade gliomas
.
J Mol Diagn
.
2011
;
13
(
6
):
669
677
.

74.

Brastianos
PK
,
Santagata
S
.
Endocrine tumors: BRAF V600E mutations in papillary craniopharyngioma
.
Eur J Endocrinol
.
2016
;
174
(
4
):
R139
R144
.

75.

Stache
C
,
Hölsken
A
,
Fahlbusch
R
,
Flitsch
J
,
Schlaffer
SM
,
Buchfelder
M
,
Buslei
R
.
Tight junction protein claudin-1 is differentially expressed in craniopharyngioma subtypes and indicates invasive tumor growth
.
Neuro-oncol
.
2014
;
16
(
2
):
256
264
.

76.

Stache
C
,
Hölsken
A
,
Schlaffer
SM
,
Hess
A
,
Metzler
M
,
Frey
B
,
Fahlbusch
R
,
Flitsch
J
,
Buchfelder
M
,
Buslei
R
.
Insights into the infiltrative behavior of adamantinomatous craniopharyngioma in a new xenotransplant mouse model
.
Brain Pathol
.
2015
;
25
(
1
):
1
10
.

77.

Samis
J
,
Vanin
EF
,
Sredni
ST
,
de Bonaldo
MF
,
Costa
FF
,
Tomita
T
,
Habiby
R
,
Zimmerman
D
,
Soares
MB
.
Extensive miRNA expression analysis in craniopharyngiomas
.
Childs Nerv Syst
.
2016
;
32
(
9
):
1617
1624
.

78.

Garcia-Lavandeira
M
,
Saez
C
,
Diaz-Rodriguez
E
,
Perez-Romero
S
,
Senra
A
,
Dieguez
C
,
Japon
MA
,
Alvarez
CV
.
Craniopharyngiomas express embryonic stem cell markers (SOX2, OCT4, KLF4, and SOX9) as pituitary stem cells but do not coexpress RET/GFRA3 receptors
.
J Clin Endocrinol Metab
.
2012
;
97
(
1
):
E80
E87
.

79.

Donson
AM
,
Apps
J
,
Griesinger
AM
,
Amani
V
,
Witt
DA
,
Anderson
RC
,
Niazi
TN
,
Grant
G
,
Souweidane
M
,
Johnston
JM
,
Jackson
EM
,
Kleinschmidt-DeMasters
BK
,
Handler
MH
,
Tan
AC
,
Gore
L
,
Virasami
A
,
Gonzalez-Meljem
JM
,
Jacques
TS
,
Martinez-Barbera
JP
,
Foreman
NK
,
Hankinson
TC
;
Advancing Treatment for Pediatric Craniopharyngioma Consortium
.
Molecular analyses reveal inflammatory mediators in the solid component and cyst fluid of human adamantinomatous craniopharyngioma
.
J Neuropathol Exp Neurol.
2017
;
76
(
9
):
779
788
.

80.

Barry
S
,
Carlsen
E
,
Marques
P
,
Stiles
CE
,
Gadaleta
E
,
Berney
DM
,
Roncaroli
F
,
Chelala
C
,
Solomou
A
,
Herincs
M
,
Caimari
F
,
Grossman
AB
,
Crnogorac-Jurcevic
T
,
Haworth
O
,
Gaston-Massuet
C
,
Korbonits
M
.
Tumor microenvironment defines the invasive phenotype of AIP-mutation-positive pituitary tumors
.
Oncogene
.
2019
;
38
(
27
):
5381
5395
.

81.

Ascierto
PA
,
Minor
D
,
Ribas
A
,
Lebbe
C
,
O’Hagan
A
,
Arya
N
,
Guckert
M
,
Schadendorf
D
,
Kefford
RF
,
Grob
JJ
,
Hamid
O
,
Amaravadi
R
,
Simeone
E
,
Wilhelm
T
,
Kim
KB
,
Long
GV
,
Martin
AM
,
Mazumdar
J
,
Goodman
VL
,
Trefzer
U
.
Phase II trial (BREAK-2) of the BRAF inhibitor dabrafenib (GSK2118436) in patients with metastatic melanoma
.
J Clin Oncol
.
2013
;
31
(
26
):
3205
3211
.

82.

Tsoukalas
N
,
Tsapakidis
K
,
Alexandraki
KI
.
The role of palbociclib in thyroid carcinoma with BRAF mutation
.
Gland Surg
.
2018
;
7
(
Suppl 1
):
S82
S85
.

83.

Aylwin
SJ
,
Bodi
I
,
Beaney
R
.
Pronounced response of papillary craniopharyngioma to treatment with vemurafenib, a BRAF inhibitor
.
Pituitary
.
2016
;
19
(
5
):
544
546
.

84.

Brastianos
PK
,
Shankar
GM
,
Gill
CM
,
Taylor-Weiner
A
,
Nayyar
N
,
Panka
DJ
,
Sullivan
RJ
,
Frederick
DT
,
Abedalthagafi
M
,
Jones
PS
,
Dunn
IF
,
Nahed
BV
,
Romero
JM
,
Louis
DN
,
Getz
G
,
Cahill
DP
,
Santagata
S
,
Curry
WT
Jr
,
Barker
FG
II
.
Dramatic response of BRAF V600E mutant papillary craniopharyngioma to targeted therapy
.
J Natl Cancer Inst
.
2015
;
108
(
2
):
djv310
.

85.

Rostami
E
,
Witt Nyström
P
,
Libard
S
,
Wikström
J
,
Casar-Borota
O
,
Gudjonsson
O
.
Recurrent papillary craniopharyngioma with BRAFV600E mutation treated with neoadjuvant-targeted therapy
.
Acta Neurochir (Wien)
.
2017
;
159
(
11
):
2217
2221
.

86.

Himes
BT
,
Ruff
MW
,
Van Gompel
JJ
,
Park
SS
,
Galanis
E
,
Kaufmann
TJ
,
Uhm
JH
.
Recurrent papillary craniopharyngioma with BRAF V600E mutation treated with dabrafenib: case report
.
J Neurosurg
.
2018
;
130
(
4
):
1299
1303
.

87.

Juratli
TA
,
Jones
PS
,
Wang
N
,
Subramanian
M
,
Aylwin
SJB
,
Odia
Y
,
Rostami
E
,
Gudjonsson
O
,
Shaw
BL
,
Cahill
DP
,
Galanis
E
,
Barker
FG
2nd, Santagata S, Brastianos PK. Targeted treatment of papillary craniopharyngiomas harboring BRAF V600E mutations. Cancer. 2019;125(17):2910–2914
.

88.

Vemurafenib and cobimetinib in treating patients with BRAF V600E mutation positive craniopharyngioma. Phase II Trial of BRAF/MEK Inhibitors in Papillary Craniopharyngiomas. ClinicalTrials.gov No. NCT03224767 (registered 21 July 2017). Available at: https://clinicaltrials.gov/ct2/results?cond=&term=NCT03224767&cntry=&state=&city=&dist=. Accessed 6 October 2019
.

89.

La Corte
E
,
Younus
I
,
Pivari
F
,
Selimi
A
,
Ottenhausen
M
,
Forbes
JA
,
Pisapia
DJ
,
Dobri
GA
,
Anand
VK
,
Schwartz
TH
.
BRAF V600E mutant papillary craniopharyngiomas: a single-institutional case series
.
Pituitary
.
2018
;
21
(
6
):
571
583
.

90.

Flaherty
KT
,
Infante
JR
,
Daud
A
,
Gonzalez
R
,
Kefford
RF
,
Sosman
J
,
Hamid
O
,
Schuchter
L
,
Cebon
J
,
Ibrahim
N
,
Kudchadkar
R
,
Burris
HA
III
,
Falchook
G
,
Algazi
A
,
Lewis
K
,
Long
GV
,
Puzanov
I
,
Lebowitz
P
,
Singh
A
,
Little
S
,
Sun
P
,
Allred
A
,
Ouellet
D
,
Kim
KB
,
Patel
K
,
Weber
J
.
Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations
.
N Engl J Med
.
2012
;
367
(
18
):
1694
1703
.

91.

Carreno
G
,
Boult
JK
,
Apps
JR
,
Gonzalez-Meljem
JM
,
Haston
S
,
Guiho
R
,
Stache
C
,
Danielson
LS
,
Koers
A
,
Smith
LS
,
Virasami
A
,
Panousopoulos
L
,
Buchfelder
M
,
Jacques
TS
,
Chesler
L
,
Robinson
S
,
Martinez-Barbera
JP
.
SHH pathway inhibition is protumourigenic in adamantinomatous craniopharyngioma [published online ahead of print 1 January 2019]
.
Endocr Relat Cancer
. doi: 10.1530/ERC-18-0538.

92.

Wang
Y
,
Deng
J
,
Guo
G
,
Tong
A
,
Peng
X
,
Chen
H
,
Xu
J
,
Liu
Y
,
You
C
,
Zhou
L
.
Clinical and prognostic role of annexin A2 in adamantinomatous craniopharyngioma
.
J Neurooncol
.
2017
;
131
(
1
):
21
29
.

93.

Nölting
S
,
Garcia
E
,
Alusi
G
,
Giubellino
A
,
Pacak
K
,
Korbonits
M
,
Grossman
AB
.
Combined blockade of signalling pathways shows marked anti-tumour potential in phaeochromocytoma cell lines
.
J Mol Endocrinol
.
2012
;
49
(
2
):
79
96
.

94.

Giubellino
A
,
Bullova
P
,
Nölting
S
,
Turkova
H
,
Powers
JF
,
Liu
Q
,
Guichard
S
,
Tischler
AS
,
Grossman
AB
,
Pacak
K
.
Combined inhibition of mTORC1 and mTORC2 signaling pathways is a promising therapeutic option in inhibiting pheochromocytoma tumor growth: in vitro and in vivo studies in female athymic nude mice
.
Endocrinology
.
2013
;
154
(
2
):
646
655
.

95.

Peginterferon alfa-2b in treating younger patients with craniopharyngioma that is recurrent or cannot be removed by surgery. Phase II Trial of BRAF/MEK Inhibitors in Papillary Craniopharyngiomas. ClinicalTrials.gov no. NCT01964300 (registered 17 October 2013). Available at: https://clinicaltrials.gov/ct2/results?cond=&term=NCT01964300&cntry=&state=&city=&dist=. Accessed 6 October 2019.

Author notes

N.K. and A.B.G. are joint senior authors.