Abstract

Adamantinomatous craniopharyngioma (ACP) is the most common benign tumor in the sellar region of children and originates from embryonic remnants. Owing to its unique location and frequent tight adhesion to and invasion of surrounding structures, the ACP poses significant challenges for neurosurgical treatment. Traditionally, the core treatment for ACPs has been surgical resection supplemented with radiotherapy in cases of residual or recurrent tumors. As a result, ACP classification has been based primarily on the tumor’s relationship with surrounding anatomical and histological structures, guiding the selection of surgical approaches and the prevention of complications. Moreover, efforts to explore pharmacological treatments for ACP have yielded varying results across different cases, creating confusion among researchers. This variability also suggests the possibility of different molecular subtypes within ACPs, despite being driven by a single-gene mutation. With advancements in molecular biological studies, such as ACP RNA sequencing, whole-exome sequencing, and methylation analysis, along with the discovery of interactions between different molecular pathways within ACP, researchers have been continuously exploring the molecular subtyping of ACP and predicting the efficacy of targeted therapies on the basis of these subtypes. This review focuses on summarizing and synthesizing the molecular mechanisms and potential subtypes of ACP, aiming to provide theoretical support for future research on the molecular subtyping of ACP.

Craniopharyngiomas are rare intracranial tumors originating from the craniopharyngeal duct at the base of the skull. It primarily occurs in children and adolescents, although adults can also be affected, with an incidence rate of approximately 0.5–2 cases per million person-years.1 Although craniopharyngiomas are histologically classified as benign epithelial tumors (World Health Organization [WHO] Grade I), their location, often near or adhered to critical neural and vascular structures, makes surgical treatment challenging, with high perioperative complication rates, high recurrence rates, and poor prognoses.2 As the pioneering neurosurgeon Professor Harvey Cushing once noted, craniopharyngioma represents one of the most formidable challenges faced by neurosurgeons.3 Previously, craniopharyngiomas were classified into adamantinomatous craniopharyngiomas (ACPs) and papillary craniopharyngiomas (PCPs) on the basis of their histological characteristics.4 However, owing to differences in their molecular biology and histopathology, these have now been recognized as distinct independent types of craniopharyngiomas.5 Among them, ACP is more common, accounting for approximately 90% of all craniopharyngioma cases. Therefore, an in-depth exploration of the detailed mechanisms of ACP is crucial for addressing craniopharyngiomas as a whole.6

Currently, there are 2 conventional treatment options for ACP: aggressive total resection or more conservative partial resection followed by radiotherapy.7 However, both options are associated with potential complications. Aggressive total resection may damage surrounding tissues, leading to cerebrovascular injury, cognitive decline, metabolic changes, and frequent panhypopituitarism.8–11 On the other hand, partial resection combined with radiotherapy may leave residual tumor tissue, increasing the risk of recurrence and repeat surgeries and increasing the risk of radiation-induced secondary malignancies.12–14 Consequently, there has been an ongoing exploration of anti-inflammatory treatments, immunotherapies, and other targeted therapies for ACP.15,16 Recent advances in the study of ACP molecular mechanisms have made significant progress, suggesting the potential for more effective targeted therapies that could reduce recurrence rates and treat related complications. Researchers have also explored the possibility of different molecular subtypes of ACP.17–20

This paper aims to systematically review the latest research advancements in ACP molecular mechanisms and the various molecular subtypes proposed on the basis of these advancements. This will further the development of ACP molecular research, aiding in the clinical prognosis assessment and precision treatment of patients.

Histopathological Features of ACP

Adamantinomatous craniopharyngioma is a nonneuroepithelial tumor located at the base of the third ventricle and the pia mater.21 It can occur at any age, with a peak incidence between the ages of 5–15 years and 45–60 years.7 In most cases, the tumor is tightly adhered to surrounding tissues, often infiltrating adjacent structures such as the pituitary gland, hypothalamus, and optic chiasm, making surgical separation challenging and the prognosis poor.22–25 Imaging of the ACP reveals different subtypes, including cystic, solid, or mixed cystic‒solid features (Figure 1A‒F). The cystic component may consist of one or more cysts filled with dark fluid, commonly referred to as “motor oil,” which is rich in lipids and inflammatory mediators.26,27 The solid part is primarily composed of epithelial tumor cells, which are highly heterogeneous and include rows of epithelium and stellate reticulum at the invasive front of the tumor. The epithelial cells form finger-like projections that often contain whorled cell structures23,28 (Figure 1G, H). The tumor is frequently surrounded by gliotic tissue, which consists mainly of astrocytes and immune cells,29 and this gliotic zone facilitates complete tumor resection. Additionally, ACPs often contain calcifications, which can be observed via computed tomography (CT), as well as wet keratin nodules (containing cells without visible nuclei), both of which are crucial for ACP diagnosis22 (Figure 1G). Notably, ACPs originate from precursor cells of the anterior pituitary gland, which are located outside the blood‒brain barrier, a histological feature that provides a natural advantage for ACP drug therapy.30

A series of medical imaging and histological staining images related to ACPs. (A) CT scan showing egg-shell calcification of the ACP, with arrows indicating the calcified region. (B) CT scan showing cluster-like calcification of the ACP, with an arrow marking the site. (C) MRI sagittal T1 image with contrast enhancement showing a completely cystic ACP lesion, with arrows indicating the cystic region. (D) MRI sagittal T1 image with contrast enhancement showing a mixed cystic and solid ACP lesion; an arrow points to the solid lesion in the upper left, and two arrows indicate the cystic lesion in the lower region. (E) MRI sagittal T1 image with contrast enhancement showing a solid ACP lesion, marked by an arrow. (F) MRI sagittal T1 image with contrast enhancement showing a mixed cystic and solid ACP lesion; an arrow points to the solid lesion on the right, while three arrows indicate the cystic lesion on the left. (G) HE staining of an ACP sample showing tumor papillary projections (marked by four arrows on the left), wet keratin (indicated by a circle), and calcification (marked by an arrow near the left side of the circle). (H) HE staining showing the tumor basal layer (indicated by three arrows on the left) and clusters of tumor cells (marked by a circle). (I) IHC staining showing β-catenin expression in the nuclei and cytoplasm of some tumor cells within tumor cell clusters (marked by three arrows on the left); the remaining tumor components predominantly show membranous expression, as indicated by arrows at the top, right, and bottom. (J) IHC staining showing β-catenin nuclear expression, indicated by an arrow.
Figure 1.

Imaging, HE, and IHC data related to ACPs. (A) CT scan showing egg-shell calcification of the ACP (arrows). (B) CT scan showing cluster-like calcification of the ACP (arrow). (C) MRI: sagittal T1 image with contrast enhancement showing a completely cystic lesion of the ACP (arrows). (D) MRI: sagittal T1 image with contrast enhancement showing a mixed cystic and solid lesion of the ACP. Solid lesion (upper left arrow), cystic lesion (two lower arrows). (E) MRI: sagittal T1 image with contrast enhancement showing a solid lesion of the ACP (arrow). (F) MRI: sagittal T1 image with contrast enhancement showing a mixed cystic and solid lesion of the ACP. Solid lesion (right arrow), cystic lesion (three left arrows). (G) HE staining: tumor papillary projection (four left arrows), wet keratin (circle), and calcification (arrow near the left side of the circle). (H) HE staining: tumor basal layer (arrows) and clusters of tumor cells (circles). (I) IHC: β-catenin expression in the nuclei and cytoplasm of some tumor cells within tumor cell clusters (three left arrows); the remaining tumor components show predominant membranous expression (top, right, and bottom arrows). (J) IHC: β-catenin nuclear expression (arrows).

Molecular Biology of ACP

On immunohistochemistry, ACPs typically exhibit abnormal accumulation of β-catenin (Figure 1I, J). Studies have shown that ACP is driven primarily by somatic mutations in exon 3 of the CTNNB1 gene, which encodes β-catenin. These mutations prevent the phosphorylation of β-catenin, increasing its resistance to degradation, thereby activating the Wnt/β-catenin signaling pathway, which induces the formation and development of ACPs28,31–33 (Figure 2A, B). The Wnt/β-catenin signaling pathway has been extensively studied in colorectal cancer, hepatocellular carcinoma, endometrial cancer, and medulloblastoma.34–36 These findings indicate that abnormal activation of this pathway can significantly promote tumorigenesis and progression, potentially disrupting cancer immunosurveillance, promoting immune evasion, and contributing to resistance to immunotherapies, including immune checkpoint inhibitors. In ACP studies, the reported incidence of CTNNB1 gene mutations varies significantly across different sources (16%~100%),37–40 and some researchers speculate that this variation may stem from differences in tumor site detection and the sensitivity of the techniques used. In ACPs, nuclear and cytoplasmic β-catenin accumulation is observed only in small clusters of cells, in areas with epithelial whorl-like structures, or a few cells near the invasive edge of the tumor. These regions influence ACP cell proliferation and differentiation through paracrine effects through the secretion of factors.22,41 No other somatic gene mutations associated with sporadic ACP have been identified, whereas somatic or germline mutations in the APC gene have been detected in some familial (nonsporadic) ACP patients, suggesting that familial ACP may involve a non-CTNNB1-dependent pathway.42

A schematic diagram illustrating the molecular mechanisms and targeted therapies related to ACP development. (A) Diagram showing common CTNNB1 mutation sites and associated nucleic acid mutations in ACPs. (B) Illustration of known intracellular signaling pathways involved in ACP tumor cells. (C) Schematic representation of matrix metalloproteinases (MMPs), cytokines (IL-6, CXCL1, IL-8, and IL-10), and the SASP-related paracrine pathway, highlighting their roles in the ACP tumor microenvironment. (D) Diagram summarizing targeted therapies and molecular subtype classification of ACP, showing potential therapeutic strategies based on molecular features.
Figure 2.

Schematic diagram of the molecular mechanisms and targeted therapies related to ACP development. (A) Common CTNNB1 mutation sites and nucleic acid mutations in ACPs. (B) Known intracellular signaling pathways in ACP tumor cells. (C) Schematic representation of MMPs; the cytokines IL-6, CXCL1, IL-8, and IL-10; and the role of the SASP-related paracrine pathway in the ACP microenvironment. (D) Schematic diagram of targeted therapies and molecular subtype classification of ACP. All panels were created with BioRender.com.

Recent research by Lu et al has shown that the TGF-β signaling pathway plays a significant role in promoting aggressive biological behavior and poor prognosis of ACP and is closely associated with recurrent ACP.43 The TGF-β pathway promotes epithelial‒mesenchymal transition (EMT) through various mechanisms, including autocrine and paracrine mechanisms, and coactivation with the WNT pathway, which is also common in other tumors.44–46 Additionally, through immunostaining and mouse model experiments, Apps et al reported that the MAPK/ERK pathway is highly active in certain areas of the ACP.27 Fibroblast growth factor (FGF), epidermal growth factor (EGF), platelet-derived growth factor (PDGF), and ERK1/2 colocalize with the proliferation marker Ki67 in the palisading epithelium around cluster structures and adjacent reactive tissues. In ACP treated with trametinib, pERK1/2 immunofluorescence is decreased, accompanied by dose-dependent apoptosis and significantly reduced proliferation,27 suggesting that the MAPK/ERK pathway could be a novel therapeutic target for ACP, providing preclinical evidence for the use of MEK inhibitors. In another concurrent study, researchers also reported that CD47—an immune checkpoint highly expressed in various tumor cells47–49—is overexpressed in ACPs, promoting ACP cell behavior through the activation of the MAPK/ERK pathway.50 Similarly, the sonic hedgehog (SHH) signaling pathway is also excessively activated in ACPs. Studies have shown that the mRNA expression of SHH, GLI1, and GLI3 is significantly greater in ACP samples than in normal pituitary tissues. These proteins belong to the SHH pathway, and analysis suggests that coactivation of the SHH and WNT pathways may play an important role in ACP pathogenesis.51–53 Moreover, the role of the epidermal growth factor receptor (EGFR) signaling pathway in ACP progression has garnered increasing attention.54 The EGFR pathway regulates cell migration by upregulating the expression of the low- and intermediate-molecular-weight keratins CK8 and CK18, a pattern of keratin expression also observed in ACP tumor cells.55 Hölsken reported that activation of the EGFR pathway significantly promotes ACP cell migration and invasion, a finding confirmed in a mouse ACP model experiment56,57 (Figure 2B).

In addition to the above signaling pathways, several other molecular mechanisms are involved in ACP development (Figure 2C), such as the role of matrix metalloproteinases (MMPs) in tumor invasion.18 Matrix metalloproteinases are highly expressed in both the stromal capillaries and epithelial components of ACPs, inducing the expression of the antiapoptotic protein B-cell leukemia/lymphoma 2 (Bcl-2) and participating in the regulation of tumor cell growth, thereby promoting ACP growth and recurrence through autocrine‒paracrine mechanisms.58,59 Immunomodulatory factors also play crucial roles in the tumor microenvironment.60 Donson et al reported high levels of cytokines and chemokines, especially IL-6, CXCL1, IL-8, IL-10, and their receptors, in ACP cyst fluid and tumor tissues.61 Grob et al reported significant shrinkage of ACP cysts upon treatment with monoclonal antibodies against VEGF and IL-6 receptor antagonists.62 In light of this evidence, Apps et al suggested that inflammation in ACPs is a key driver of ACP pathogenesis.2

The role of the senescence phenotype in ACP development is noteworthy, as recent studies have investigated how senescent cells induce tumorigenesis through paracrine pathways in ACPs.28,63–65 For a long time, cellular senescence has been widely regarded as a natural protective mechanism that limits cancer cell proliferation and tumor growth. However, this view is being increasingly challenged.66 Research suggests that the senescence-associated secretory phenotype (SASP) produced by senescent cells can trigger various protumorigenic cellular and molecular processes through paracrine pathways, leading to changes in the cellular microenvironment characterized by inflammation and immune response. This phenomenon has been confirmed in ACP studies.63,66 Single-cell and single-nucleus gene expression analyses further confirmed the presence of senescent cells with active SASP in ACPs, revealing their complex relationship with tumor progression.64

Historical Classification of Craniopharyngiomas

The classification of any disease is rooted in the evolving understanding of that disease, and craniopharyngiomas (CPs) are no exception. Since CP is considered a benign tumor (CNS WHO Grade I), it was historically believed that complete radical resection was the best treatment for craniopharyngiomas.67 However, radical resection increases the risk of intraoperative damage to the hypothalamus, significantly increasing surgical risk.68 This finding indicates that the extent of tumor adherence to the hypothalamus dictates the surgical treatment strategy for craniopharyngiomas. Consequently, classifying craniopharyngiomas on the basis of the degree of adhesion between the tumor and the hypothalamus has become natural. Currently, 3 main grading systems are mentioned in the literature to assess hypothalamic involvement or the adherence of the tumor to the hypothalamus: the Puget grading system,69 the Prieto grading system,70 and the Hori T grading system.71 It is indisputable that the adhesion and invasion of craniopharyngiomas to surrounding tissues significantly influence treatment decisions and patient prognosis.72 Therefore, a systematic and comprehensive preoperative analysis of the adhesion between the craniopharyngioma and surrounding structures, such as the hypothalamus, is crucial in determining the surgical approach.73

Once the surgical principles for craniopharyngiomas have been established, whether for aggressive or conservative treatment, selecting the correct and appropriate surgical approach becomes key to a successful operation. The goal of surgical resection of craniopharyngiomas is to remove as much of the tumor as possible while minimizing damage to the hypothalamus.74 To achieve this surgical goal, surgeons should choose the correct and appropriate surgical approach on the basis of the anatomical relationship between the tumor and surrounding neurovascular structures, as well as the surgeon’s personal experience. Over the past few decades, researchers have proposed a series of topographic classifications of craniopharyngiomas to facilitate precise assessment of the anatomical location of the tumor, thereby enabling the determination of the correct and appropriate surgical approach (Table 1).

Table 1.

The Topographic (Anatomical) Classification of CPs

AuthorYearClassification basisClassification system
Ciric IS et al751980Relationship of the tumor with the surrounding arachnoid and pia mater-Intraventricular type
-Subpial type
-Subarachnoid extrameningeal type
-Intraextraarachnoid type
-Intra-arachnoid suprasellar type
-Extra-arachnoid intrasellar type
Steno J761985Tumor location-Intrasellar type
-Suprasellar type
(a) Extraventricular subtype
(b) Intraextraventricular subtype
(c) Intraventricular subtype
Yaşargil MG et al771990Relationship of the tumor with the diaphragm sellae-Entirely intrasellar–subdiaphragmatic type
-Intrasellar and suprasellar–sellar diaphragm type
-Suprasellar para-chiasmatic–extraventricular type
-Intraextraventricular type
-Paraventricular type
-Intraventricular type
Hoffman HJ781994Relationship of the tumor with the optic chiasm-Intrasellar type
-Prechiasmatic type
-Postchiasmatic type
-Giant type
Samii M et al791997The levels of tumor vertical invasion (5 grades)-I: intrasellar type
-II: suprasellar cistern type
-III: inferior third ventricle type
-IV: superior third ventricle type
-V: lateral ventricle septum pellucidum type
Pascual JM et al802004Relationship of the tumor with the third ventricle-I: suprasellar tumor compressing the third ventricle (third ventricle wall intact)
-II: suprasellar tumor breaking through the floor of the third ventricle and invading inside
-III: tumor inside the third ventricle, invading the floor of the third ventricle
-IV: tumor inside the third ventricle, with the third ventricle floor intact
Wang KC et al812005Tumor origin and relationship with the diaphragma sellae-Origin below the diaphragma sellae, diaphragma sellae intact
-Origin below the diaphragma sellae, diaphragma sellae incomplete
-Origin above the diaphragma sellae
Kassam AB et al82
Tang B et al83
2008
2019
Relationship of the tumor with the pituitary stalk (endoscopic perspective)
Subclassification of Type II (endoscopic perspective)
-I: preinfundibular type
-II: trans-infundibular type
-II1: tumor confined to the pituitary stalk
-II2: (a) tumor extends upward into the third ventricle; (b) tumor extends downward into the sella
-II3: tumor extends both upward into the third ventricle and downward into the sella
-III: postinfundibular type
-IIIa: tumor extends upward into the third ventricle
-IIIb: tumor extends upward into the interpeduncular cistern
-IIIa + b: tumor extends in both directions
IV: completely intrathird ventricle type
Fatemi N et al842009Anatomical extension of the tumor-Postchiasmatic type
-Intrasellar-suprasellar type
-Cavernous sinus involvement type
-Far-lateral extension type
Yamada S et al852010Location of the tumor-Below the sellar diaphragm type
(a) Pure intrasellar type
(b) Suprasellar extension type
-Above the sellar diaphragm type
Qi S et al86,872011, 2021Growth pattern of the tumor around the arachnoid sleeve near the pituitary stalk-Q type: tumor originating from the area below the sellar diaphragm
-S type: tumor originating from the suprasellar pituitary stalk area
-T type: tumor originating from the infundibular tubercle region
Matsuo T et al882014Anatomical relationships of the tumor with the diaphragma sellae, pituitary stalk, and optic nerves-Relationship with the sellar diaphragm: below the sellar diaphragm type (complete, incomplete), above the sellar diaphragm type
-Relationship with the pituitary stalk: preinfundibular, lateral-infundibular, postinfundibular, and Infiltrating infundibulum type
-Relationship with optic nerve: prechiasmatic type, postchiasmatic type, and other (pure intrasellar type)
-Tumor extension: third ventricle, interpeduncular cistern, prepontine cistern, frontal base, and cavernous sinus
-Sphenoid sinus: intrasellar type, anterior sellar type, and external ear type
Morisako H et al892016Tumor origin-Intrasellar type
-Prechiasmatic type
-Postchiasmatic type
-Intrathird ventricle type
Jeswani S et al902016Relationship of the tumor with the pituitary stalk (endoscopic view)-I: tumor mainly anterior to the pituitary stalk
-II: tumor mainly within the pituitary stalk
-III: tumor mainly posterior to the pituitary stalk
Tang B et al912018Relationship of the tumor with the pituitary stalk-Central type
-Lateral type (based on lesion origin):
hypothalamic-pituitary stalk subtype
Suprasellar pituitary stalk subtype
Intrasellar pituitary stalk subtype
Prieto R et al92,932017, 2020Mammillary body angle (MBA)-Intrasellar-suprasellar type
-Suprasellar and third ventricle extraventricular type
-Suprasellar–intraventricular invasive type
-Infundibulum-tuber cinereum type
-Pure intrathird ventricle type
Almeida JP et al942020Tumor extension-A. Lower-middle type (intrasellar type)
-B. Upper-middle type (suprasellar type)
-C. Lateral type (lateral fissure type)
-D. Intraventricular type
-E. Posterior type (interpeduncular cistern and prepontine cistern type)
Lei C et al952021Location of the tumor (endoscopic perspective)-Intrasellar type
-Intrasellar-suprasellar type
-Suprasellar type
-Intrathird ventricle type
AuthorYearClassification basisClassification system
Ciric IS et al751980Relationship of the tumor with the surrounding arachnoid and pia mater-Intraventricular type
-Subpial type
-Subarachnoid extrameningeal type
-Intraextraarachnoid type
-Intra-arachnoid suprasellar type
-Extra-arachnoid intrasellar type
Steno J761985Tumor location-Intrasellar type
-Suprasellar type
(a) Extraventricular subtype
(b) Intraextraventricular subtype
(c) Intraventricular subtype
Yaşargil MG et al771990Relationship of the tumor with the diaphragm sellae-Entirely intrasellar–subdiaphragmatic type
-Intrasellar and suprasellar–sellar diaphragm type
-Suprasellar para-chiasmatic–extraventricular type
-Intraextraventricular type
-Paraventricular type
-Intraventricular type
Hoffman HJ781994Relationship of the tumor with the optic chiasm-Intrasellar type
-Prechiasmatic type
-Postchiasmatic type
-Giant type
Samii M et al791997The levels of tumor vertical invasion (5 grades)-I: intrasellar type
-II: suprasellar cistern type
-III: inferior third ventricle type
-IV: superior third ventricle type
-V: lateral ventricle septum pellucidum type
Pascual JM et al802004Relationship of the tumor with the third ventricle-I: suprasellar tumor compressing the third ventricle (third ventricle wall intact)
-II: suprasellar tumor breaking through the floor of the third ventricle and invading inside
-III: tumor inside the third ventricle, invading the floor of the third ventricle
-IV: tumor inside the third ventricle, with the third ventricle floor intact
Wang KC et al812005Tumor origin and relationship with the diaphragma sellae-Origin below the diaphragma sellae, diaphragma sellae intact
-Origin below the diaphragma sellae, diaphragma sellae incomplete
-Origin above the diaphragma sellae
Kassam AB et al82
Tang B et al83
2008
2019
Relationship of the tumor with the pituitary stalk (endoscopic perspective)
Subclassification of Type II (endoscopic perspective)
-I: preinfundibular type
-II: trans-infundibular type
-II1: tumor confined to the pituitary stalk
-II2: (a) tumor extends upward into the third ventricle; (b) tumor extends downward into the sella
-II3: tumor extends both upward into the third ventricle and downward into the sella
-III: postinfundibular type
-IIIa: tumor extends upward into the third ventricle
-IIIb: tumor extends upward into the interpeduncular cistern
-IIIa + b: tumor extends in both directions
IV: completely intrathird ventricle type
Fatemi N et al842009Anatomical extension of the tumor-Postchiasmatic type
-Intrasellar-suprasellar type
-Cavernous sinus involvement type
-Far-lateral extension type
Yamada S et al852010Location of the tumor-Below the sellar diaphragm type
(a) Pure intrasellar type
(b) Suprasellar extension type
-Above the sellar diaphragm type
Qi S et al86,872011, 2021Growth pattern of the tumor around the arachnoid sleeve near the pituitary stalk-Q type: tumor originating from the area below the sellar diaphragm
-S type: tumor originating from the suprasellar pituitary stalk area
-T type: tumor originating from the infundibular tubercle region
Matsuo T et al882014Anatomical relationships of the tumor with the diaphragma sellae, pituitary stalk, and optic nerves-Relationship with the sellar diaphragm: below the sellar diaphragm type (complete, incomplete), above the sellar diaphragm type
-Relationship with the pituitary stalk: preinfundibular, lateral-infundibular, postinfundibular, and Infiltrating infundibulum type
-Relationship with optic nerve: prechiasmatic type, postchiasmatic type, and other (pure intrasellar type)
-Tumor extension: third ventricle, interpeduncular cistern, prepontine cistern, frontal base, and cavernous sinus
-Sphenoid sinus: intrasellar type, anterior sellar type, and external ear type
Morisako H et al892016Tumor origin-Intrasellar type
-Prechiasmatic type
-Postchiasmatic type
-Intrathird ventricle type
Jeswani S et al902016Relationship of the tumor with the pituitary stalk (endoscopic view)-I: tumor mainly anterior to the pituitary stalk
-II: tumor mainly within the pituitary stalk
-III: tumor mainly posterior to the pituitary stalk
Tang B et al912018Relationship of the tumor with the pituitary stalk-Central type
-Lateral type (based on lesion origin):
hypothalamic-pituitary stalk subtype
Suprasellar pituitary stalk subtype
Intrasellar pituitary stalk subtype
Prieto R et al92,932017, 2020Mammillary body angle (MBA)-Intrasellar-suprasellar type
-Suprasellar and third ventricle extraventricular type
-Suprasellar–intraventricular invasive type
-Infundibulum-tuber cinereum type
-Pure intrathird ventricle type
Almeida JP et al942020Tumor extension-A. Lower-middle type (intrasellar type)
-B. Upper-middle type (suprasellar type)
-C. Lateral type (lateral fissure type)
-D. Intraventricular type
-E. Posterior type (interpeduncular cistern and prepontine cistern type)
Lei C et al952021Location of the tumor (endoscopic perspective)-Intrasellar type
-Intrasellar-suprasellar type
-Suprasellar type
-Intrathird ventricle type
Table 1.

The Topographic (Anatomical) Classification of CPs

AuthorYearClassification basisClassification system
Ciric IS et al751980Relationship of the tumor with the surrounding arachnoid and pia mater-Intraventricular type
-Subpial type
-Subarachnoid extrameningeal type
-Intraextraarachnoid type
-Intra-arachnoid suprasellar type
-Extra-arachnoid intrasellar type
Steno J761985Tumor location-Intrasellar type
-Suprasellar type
(a) Extraventricular subtype
(b) Intraextraventricular subtype
(c) Intraventricular subtype
Yaşargil MG et al771990Relationship of the tumor with the diaphragm sellae-Entirely intrasellar–subdiaphragmatic type
-Intrasellar and suprasellar–sellar diaphragm type
-Suprasellar para-chiasmatic–extraventricular type
-Intraextraventricular type
-Paraventricular type
-Intraventricular type
Hoffman HJ781994Relationship of the tumor with the optic chiasm-Intrasellar type
-Prechiasmatic type
-Postchiasmatic type
-Giant type
Samii M et al791997The levels of tumor vertical invasion (5 grades)-I: intrasellar type
-II: suprasellar cistern type
-III: inferior third ventricle type
-IV: superior third ventricle type
-V: lateral ventricle septum pellucidum type
Pascual JM et al802004Relationship of the tumor with the third ventricle-I: suprasellar tumor compressing the third ventricle (third ventricle wall intact)
-II: suprasellar tumor breaking through the floor of the third ventricle and invading inside
-III: tumor inside the third ventricle, invading the floor of the third ventricle
-IV: tumor inside the third ventricle, with the third ventricle floor intact
Wang KC et al812005Tumor origin and relationship with the diaphragma sellae-Origin below the diaphragma sellae, diaphragma sellae intact
-Origin below the diaphragma sellae, diaphragma sellae incomplete
-Origin above the diaphragma sellae
Kassam AB et al82
Tang B et al83
2008
2019
Relationship of the tumor with the pituitary stalk (endoscopic perspective)
Subclassification of Type II (endoscopic perspective)
-I: preinfundibular type
-II: trans-infundibular type
-II1: tumor confined to the pituitary stalk
-II2: (a) tumor extends upward into the third ventricle; (b) tumor extends downward into the sella
-II3: tumor extends both upward into the third ventricle and downward into the sella
-III: postinfundibular type
-IIIa: tumor extends upward into the third ventricle
-IIIb: tumor extends upward into the interpeduncular cistern
-IIIa + b: tumor extends in both directions
IV: completely intrathird ventricle type
Fatemi N et al842009Anatomical extension of the tumor-Postchiasmatic type
-Intrasellar-suprasellar type
-Cavernous sinus involvement type
-Far-lateral extension type
Yamada S et al852010Location of the tumor-Below the sellar diaphragm type
(a) Pure intrasellar type
(b) Suprasellar extension type
-Above the sellar diaphragm type
Qi S et al86,872011, 2021Growth pattern of the tumor around the arachnoid sleeve near the pituitary stalk-Q type: tumor originating from the area below the sellar diaphragm
-S type: tumor originating from the suprasellar pituitary stalk area
-T type: tumor originating from the infundibular tubercle region
Matsuo T et al882014Anatomical relationships of the tumor with the diaphragma sellae, pituitary stalk, and optic nerves-Relationship with the sellar diaphragm: below the sellar diaphragm type (complete, incomplete), above the sellar diaphragm type
-Relationship with the pituitary stalk: preinfundibular, lateral-infundibular, postinfundibular, and Infiltrating infundibulum type
-Relationship with optic nerve: prechiasmatic type, postchiasmatic type, and other (pure intrasellar type)
-Tumor extension: third ventricle, interpeduncular cistern, prepontine cistern, frontal base, and cavernous sinus
-Sphenoid sinus: intrasellar type, anterior sellar type, and external ear type
Morisako H et al892016Tumor origin-Intrasellar type
-Prechiasmatic type
-Postchiasmatic type
-Intrathird ventricle type
Jeswani S et al902016Relationship of the tumor with the pituitary stalk (endoscopic view)-I: tumor mainly anterior to the pituitary stalk
-II: tumor mainly within the pituitary stalk
-III: tumor mainly posterior to the pituitary stalk
Tang B et al912018Relationship of the tumor with the pituitary stalk-Central type
-Lateral type (based on lesion origin):
hypothalamic-pituitary stalk subtype
Suprasellar pituitary stalk subtype
Intrasellar pituitary stalk subtype
Prieto R et al92,932017, 2020Mammillary body angle (MBA)-Intrasellar-suprasellar type
-Suprasellar and third ventricle extraventricular type
-Suprasellar–intraventricular invasive type
-Infundibulum-tuber cinereum type
-Pure intrathird ventricle type
Almeida JP et al942020Tumor extension-A. Lower-middle type (intrasellar type)
-B. Upper-middle type (suprasellar type)
-C. Lateral type (lateral fissure type)
-D. Intraventricular type
-E. Posterior type (interpeduncular cistern and prepontine cistern type)
Lei C et al952021Location of the tumor (endoscopic perspective)-Intrasellar type
-Intrasellar-suprasellar type
-Suprasellar type
-Intrathird ventricle type
AuthorYearClassification basisClassification system
Ciric IS et al751980Relationship of the tumor with the surrounding arachnoid and pia mater-Intraventricular type
-Subpial type
-Subarachnoid extrameningeal type
-Intraextraarachnoid type
-Intra-arachnoid suprasellar type
-Extra-arachnoid intrasellar type
Steno J761985Tumor location-Intrasellar type
-Suprasellar type
(a) Extraventricular subtype
(b) Intraextraventricular subtype
(c) Intraventricular subtype
Yaşargil MG et al771990Relationship of the tumor with the diaphragm sellae-Entirely intrasellar–subdiaphragmatic type
-Intrasellar and suprasellar–sellar diaphragm type
-Suprasellar para-chiasmatic–extraventricular type
-Intraextraventricular type
-Paraventricular type
-Intraventricular type
Hoffman HJ781994Relationship of the tumor with the optic chiasm-Intrasellar type
-Prechiasmatic type
-Postchiasmatic type
-Giant type
Samii M et al791997The levels of tumor vertical invasion (5 grades)-I: intrasellar type
-II: suprasellar cistern type
-III: inferior third ventricle type
-IV: superior third ventricle type
-V: lateral ventricle septum pellucidum type
Pascual JM et al802004Relationship of the tumor with the third ventricle-I: suprasellar tumor compressing the third ventricle (third ventricle wall intact)
-II: suprasellar tumor breaking through the floor of the third ventricle and invading inside
-III: tumor inside the third ventricle, invading the floor of the third ventricle
-IV: tumor inside the third ventricle, with the third ventricle floor intact
Wang KC et al812005Tumor origin and relationship with the diaphragma sellae-Origin below the diaphragma sellae, diaphragma sellae intact
-Origin below the diaphragma sellae, diaphragma sellae incomplete
-Origin above the diaphragma sellae
Kassam AB et al82
Tang B et al83
2008
2019
Relationship of the tumor with the pituitary stalk (endoscopic perspective)
Subclassification of Type II (endoscopic perspective)
-I: preinfundibular type
-II: trans-infundibular type
-II1: tumor confined to the pituitary stalk
-II2: (a) tumor extends upward into the third ventricle; (b) tumor extends downward into the sella
-II3: tumor extends both upward into the third ventricle and downward into the sella
-III: postinfundibular type
-IIIa: tumor extends upward into the third ventricle
-IIIb: tumor extends upward into the interpeduncular cistern
-IIIa + b: tumor extends in both directions
IV: completely intrathird ventricle type
Fatemi N et al842009Anatomical extension of the tumor-Postchiasmatic type
-Intrasellar-suprasellar type
-Cavernous sinus involvement type
-Far-lateral extension type
Yamada S et al852010Location of the tumor-Below the sellar diaphragm type
(a) Pure intrasellar type
(b) Suprasellar extension type
-Above the sellar diaphragm type
Qi S et al86,872011, 2021Growth pattern of the tumor around the arachnoid sleeve near the pituitary stalk-Q type: tumor originating from the area below the sellar diaphragm
-S type: tumor originating from the suprasellar pituitary stalk area
-T type: tumor originating from the infundibular tubercle region
Matsuo T et al882014Anatomical relationships of the tumor with the diaphragma sellae, pituitary stalk, and optic nerves-Relationship with the sellar diaphragm: below the sellar diaphragm type (complete, incomplete), above the sellar diaphragm type
-Relationship with the pituitary stalk: preinfundibular, lateral-infundibular, postinfundibular, and Infiltrating infundibulum type
-Relationship with optic nerve: prechiasmatic type, postchiasmatic type, and other (pure intrasellar type)
-Tumor extension: third ventricle, interpeduncular cistern, prepontine cistern, frontal base, and cavernous sinus
-Sphenoid sinus: intrasellar type, anterior sellar type, and external ear type
Morisako H et al892016Tumor origin-Intrasellar type
-Prechiasmatic type
-Postchiasmatic type
-Intrathird ventricle type
Jeswani S et al902016Relationship of the tumor with the pituitary stalk (endoscopic view)-I: tumor mainly anterior to the pituitary stalk
-II: tumor mainly within the pituitary stalk
-III: tumor mainly posterior to the pituitary stalk
Tang B et al912018Relationship of the tumor with the pituitary stalk-Central type
-Lateral type (based on lesion origin):
hypothalamic-pituitary stalk subtype
Suprasellar pituitary stalk subtype
Intrasellar pituitary stalk subtype
Prieto R et al92,932017, 2020Mammillary body angle (MBA)-Intrasellar-suprasellar type
-Suprasellar and third ventricle extraventricular type
-Suprasellar–intraventricular invasive type
-Infundibulum-tuber cinereum type
-Pure intrathird ventricle type
Almeida JP et al942020Tumor extension-A. Lower-middle type (intrasellar type)
-B. Upper-middle type (suprasellar type)
-C. Lateral type (lateral fissure type)
-D. Intraventricular type
-E. Posterior type (interpeduncular cistern and prepontine cistern type)
Lei C et al952021Location of the tumor (endoscopic perspective)-Intrasellar type
-Intrasellar-suprasellar type
-Suprasellar type
-Intrathird ventricle type

Among all the anatomical classifications of craniopharyngiomas, the Yaşargil MG classification, Samii M classification, Morisako H classification, and Qi S classification are the most widely referenced. The Yaşargil MG classification is commonly used in clinical practice, primarily because of the widespread adoption of pterional surgical approaches and microsurgical techniques at the time. The Samii M and Morisako H classifications are, in fact, modifications of the Yaşargil MG classification. However, different surgical practices and the frequent need to adapt surgical approaches on the basis of the tumor’s location and morphological characteristics during surgery make it difficult to assess the strengths and weaknesses of each classification solely on the basis of surgical outcomes from different centers.

The QST classification, which is based on the tissue of origin, is particularly noteworthy for researchers. As individualized surgical plans become more common and the integration of endoscopic techniques with transcranial microsurgery advances,96–98 different anatomical classifications may increasingly focus on hypothalamic protection during resection and the decision of whether to pursue total tumor removal.99,100

Molecular Subtyping of ACP

As described in the previous discussion on molecular biology, the development of ACPs involves the interaction of multiple signaling pathways and molecular mechanisms, including Wnt/β-catenin, MAPK, SHH, and others, which play significant roles in the formation and progression of ACPs. In recent years, with the application of high-resolution technologies such as single-cell RNA sequencing, researchers have gradually revealed significant heterogeneity within ACP tumors.101 This internal heterogeneity also leads to considerable differences in the clinical presentations of patients with ACP.102 For example, the age of onset varies widely, affecting both children and adults; radiologically, ACP can present in various forms, including cystic, solid, or a combination of cystic and solid components.103 These diverse presentations contrast sharply with the traditional view of ACP as a tumor driven solely by single-gene mutations. More notably, different patients also exhibit significant variability in their responses to drug treatments.104 For example, some patients respond well to anti-inflammatory therapy and achieve disease control, whereas others may show no significant response to the same treatment.15 These findings further suggest that ACP might exist in different molecular subtypes, each with distinct mechanisms of onset, progression, and treatment response. Tumor classification has gradually shifted from histological to molecular subtyping, and new molecular subtyping is not only the basis for precision therapy but also crucial for elucidating tumor mechanisms, identifying prognostic factors, and developing personalized treatment strategies.105,106

Research on the molecular subtyping of ACP has evolved from the discovery of single-gene mutations to comprehensive multiomics analyses. Early studies focused primarily on mutations in the CTNNB1 gene (encoding β-catenin), revealing aberrant activation of the Wnt signaling pathway in ACP.107–109 Subsequently, specific epigenetic markers, such as DNA methylation and histone modifications, were identified, revealing different patterns among numerous ACP patients.110 With technological advancements, researchers have employed high-throughput sequencing techniques, including transcriptomics, proteomics, single-cell sequencing, and spatial transcriptomics, to perform comprehensive analyses of the ACP genome, discovering additional gene mutations and structural variations.4,101,111,112 Clinical studies indicate that the biological heterogeneity of ACP tumors results in significant variations in clinical presentation, pathological features, and prognosis. For example, ACPs with certain molecular characteristics may exhibit increased invasiveness and recurrence rates, whereas others demonstrate relatively benign behavior.39,113,114 These findings suggest the possibility of finer molecular subtypes within ACPs. Current research on the molecular subtypes of ACP has focused mainly on defining different tumor subtypes through gene mutation profiles, expression profiles, and epigenetic changes22,110,115 (Figure 2D).

In early studies on ACP, due to significant variations in the reported incidence of CTNNB1 gene mutations among different studies, classifying ACP into wild-type and mutant-type seemed to be a feasible approach.37–40 However, this classification currently lacks sufficient clinical data to support it.33,116 Moreover, the so-called wild-type ACP, IHC, and HE staining images have shown the presence of similar whorled cluster structures and nuclear accumulation of β-catenin in these samples.20 A recent study employed laser capture microdissection to isolate tumor cells and conducted deep sequencing of targeted amplified fragments. The results revealed that all ACP samples tested carried CTNNB1 mutations.40 The study by Zhang et al also confirmed that the detection of wild-type cases may be affected by the low proportion of tumor cells and the limited sensitivity of current sequencing technologies.117 Therefore, the existence of wild-type and mutant-type still requires further investigation.

Given the challenges of molecular genotyping, researchers have begun exploring other classification criteria. The cyst wall cells of craniopharyngiomas express programmed death-ligand 1 (PD-L1), while programmed death-1 (PD-1) is expressed in whorl-like cells with nuclear accumulation of β-catenin, suggesting that PD-1/PD-L1 immune checkpoint-targeted therapy for craniopharyngiomas might have potential clinical value.118 Nevertheless, in subsequent studies, we observed that the expression of PD-L1 in ACP tumor cells is challenging to quantify definitively, including determining the level of expression and how to classify it. It remains to be further determined which types of patients could benefit from this treatment.119–121 Yuan et al retrieved RNA data from 39 pediatric ACP patients from the public databases GSE60815 and GSE94349 and, through screening and analysis, classified 725 ACP-related immune genes into 2 types: immune resistance subtype and immunogenic subtype.115 These results indicate that these 2 types respond differently to immune checkpoint blockade (ICB) therapy. However, Yuan et al’s classification is merely a bioinformatics-based classification, lacking comparative analysis with clinical data such as imaging or histology, and it has not been validated in clinical practice. Moreover, this classification does not clarify the relationship between these 2 subtypes and the gene-driven pathways of ACP.118

Wu et al reported that ACPs are diverse at the genetic level, providing an important basis for the molecular subtyping of ACPs. They introduced the concept that ACP patients with high expression of RHOC and CD109 have different sensitivities to various drugs on the basis of sequencing databases.112 However, like previous studies, they did not obtain clinical research support. Many scholars pessimistically believe that it might not be possible to subtype ACPs on the basis of RNA omics.122 A recently published study proposed a new classification method that classifies certain tumor cells as the BRAF V600E mutation-like type on the basis of proteomics and phosphoproteomics theories.123 According to this new classification method, the proteomic changes in the MAPK/MEK pathway of these tumor cells resemble the previously observed BRAF V600E gene variation expression, suggesting that MEK pathway inhibitors might be effective for ACP treatment.27,124 Additionally, an epigenetic study by Gutiérrez et al identified 2 distinct ACP methylation clusters, with the ACP-2 cluster exhibiting significantly low methylation and being associated with CTNNB1 mutations, which are more aggressive.110 These findings suggest that DNA methylation features might be used for future ACP diagnosis and prognosis.

Recently, Professor Lin Zhixiong’s research team conducted whole-exome sequencing (WES) on 151 samples from 143 craniopharyngioma patients, along with RNA sequencing on 84 samples and DNA methylation analysis on 95 samples (the largest dataset globally).20 The results revealed completely distinct molecular subtypes of ACP at the RNA level, which was consistent with the findings of the methylation-based subtypes, indicating that ACP has 3 different molecular subtypes: the WNT type, ImA type, and ImB type. Among them, the WNT type can be overactivated through the Wnt/β-catenin pathway, whereas the ImA type and ImB type exhibit increased levels of immune infiltration, with the ImA type showing a significant gliosis reaction. These molecular characteristics of the 3 subtypes were confirmed through immunohistochemistry and proteomics analysis. Computed tomography and MRI observations indicate that the WNT type is predominantly solid, whereas the ImA type and ImB type are primarily cystic. Prognostic statistics show that patients with the WNT type almost never experience recurrence, and their survival time is significantly longer than that of patients with the ImB type. A multivariate Cox regression analysis suggested that this classification is the most significant prognostic predictor apart from the extent of tumor resection. According to the TIDE (tumor immune dysfunction and exclusion) model predictions, the ImA type is expected to benefit the most from immunosuppressive therapy among the 3 subtypes, whereas the WNT type shows an immune exclusion status. Single-sample gene set enrichment analysis (GSEA) revealed that only ImA type and ImB type samples were enriched for the interferon-α response pathway, which is consistent with a retrospective analysis of clinical trial results: interferon-α treatment responses were observed only in ACP cases with predominantly cystic components.125 These findings are highly important for the future precision diagnosis and treatment of ACP. However, the study did not clarify why tumors driven by single-gene mutations present different molecular subtypes. This may result from interactions between the molecular pathways driven by single-gene mutations and the tumor microenvironment.

Precision Therapy for ACP on the Basis of Molecular Characteristics

In recent years, there has been a gradual exploration of precision-targeted therapies for ACP126 (Figure 2D). Ros et al investigated the inflammatory environment of ACP and its therapeutic importance and reported that ACP displays numerous inflammatory and immune markers, suggesting the presence of numerous potential treatment targets.15 Nowadays, various related drugs are being tested for the treatment of ACP.127 Earlier, Jakacki et al studied the application of interferon-2α (IFN-2α) in ACP patients,128 followed by clinical studies evaluating the response of ACPs to locally and systemically administered pegylated IFN-α-2b.129 However, a recent phase II clinical trial indicated that pegylated IFN-α-2b had limited efficacy.130,131 Transcriptomic analyses have shown that both solid and cystic components of ACP exhibit inflammasome activation.61 In this context, Grob, Vos-Kerkhof, and others explored the use of the monoclonal antibody tocilizumab, which targets IL-6, for the treatment of ACP and reported good efficacy in cystic ACP patients.62,132 Currently, a phase II clinical trial (NCT05233397) is further evaluating the efficacy of tocilizumab (Actemra) for progressive or recurrent ACP.133 Additionally, De Rosa et al reported a case where ACP was treated with bevacizumab, resulting in a 66.1% reduction in tumor volume after 3 months, with partial remission achieved.134 In another case study by Patel et al, the use of MEK inhibitor binimetinib was shown to reduce the tumor volume and stabilize the condition of the patient.124 Related to this, a phase II clinical trial (NCT05286788) is also assessing the efficacy of binimetinib (Mektovi) in pediatric ACP patients.135 In addition to the aforementioned studies, another phase II trial (NCT05465174) is investigating the potential of the PD-1 inhibitor nivolumab in combination with the pan-RAF kinase inhibitor tovorafenib for the treatment of ACP in children and adolescents, which is also worth ongoing attention.136 While research on targeted therapies for ACP highlights the sustained efforts of scholars to develop effective interventions, current treatment options remain limited.126 Additionally, the efficacy of most treatments is limited, with relatively few cases and inconsistent outcomes. Given the heterogeneity of ACPs and the urgent need for personalized treatment strategies, further research is needed to enrich and validate targeted therapy options for ACPs.

Conclusion and Outlook

Adamantinomatous craniopharyngioma is a complex and challenging type of intracranial tumor. Although histologically classified as benign, its invasive growth and high recurrence rate make treatment and prognosis extremely difficult.137 Over the past few decades, significant progress has been made in understanding ACP, particularly in the field of molecular biology. Research has revealed the critical roles of several key signaling pathways, including the Wnt/β-catenin signaling pathway, the TGF-β signaling pathway, and the MAPK/ERK pathway, in the development and progression of ACP. Additionally, the senescence phenotype, immune modulators, and MMPs play significant roles in the tumor microenvironment. These discoveries have laid the groundwork for molecular subtyping and have provided potential targets for the development of new therapeutic strategies.

Currently, molecular subtyping is gradually emerging in ACP research. Through the analysis of mutation profiles, expression profiles, and epigenetic changes, researchers have identified multiple molecular subtypes of ACP. Recent studies have indicated that ACPs can be classified into the WNT type, ImA type, and ImB type. These subtypes differ significantly in their clinical presentation and prognosis, suggesting that future treatment strategies should fully consider the characteristics of these molecular subtypes to enable precise treatment, in line with the growing trend toward precision medicine in cancer research.138

Future research on the molecular biology of ACP should focus on understanding why tumors driven by single-gene mutations present different molecular subtypes. Previous studies have suggested a close relationship between inflammation and ACP development.41 This raises the following question: do the different molecular subtypes of ACP result from interactions between single-gene mutation-driven molecular pathways and the inflammatory microenvironment of the tumor? If this hypothesis is confirmed, future targeted therapies for ACP must inevitably involve a combination of both approaches.

Funding

The study was supported by the Sanbo Brain Hospital Management Group [SBJT-KY-2020-002 to Z.L.], the Capital Funds for Health Improvement and Research [2022-2-8013 to Z.L.], and the National Key R&D Program of China [2023YFC2411603 to Z.L.].

Conflict of interest statement. None declared.

Acknowledgments

Figure 2 was created with BioRender.com.

Author contributions

W.A., S.L., and Y.A. were responsible for the conception and design of the study. W.A. drafted and edited the manuscript. Z.L. reviewed and revised the manuscript. All the authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

This study was performed in accordance with the principles of the Declaration of Helsinki. Approval was granted by the Institutional Review Board/Ethics Committee of Sanbo Brain Hospital of Capital Medical University.

Consent for publication

All authors have read and approved the final manuscript.

Data availability

Not applicable.

References

1.

Zhu
 
L
,
Zhang
 
L
,
Hu
 
W
, et al.  
A multi-task two-path deep learning system for predicting the invasiveness of craniopharyngioma
.
Comput Methods Programs Biomed.
 
2022
;
216
:
106651
.

2.

Apps
 
JR
,
Muller
 
HL
,
Hankinson
 
TC
,
Yock
 
TI
,
Martinez-Barbera
 
JP.
 
Contemporary biological insights and clinical management of craniopharyngioma
.
Endocr Rev.
 
2023
;
44
(
3
):
518
538
.

3.

Pascual
 
JM
,
Prieto
 
R.
 
Harvey cushing and pituitary case number 3 (Mary D.): the origin of this most baffling problem in neurosurgery
.
Neurosurg Focus.
 
2016
;
41
(
1
):
E6
.

4.

Kim
 
JH
,
Kim
 
H
,
Dan
 
K
, et al.  
In-depth proteomic profiling captures subtype-specific features of craniopharyngiomas
.
Sci Rep.
 
2021
;
11
(
1
):
21206
.

5.

Louis
 
DN
,
Perry
 
A
,
Wesseling
 
P
, et al.  
The 2021 WHO classification of tumors of the central nervous system: a summary
.
Neuro Oncol
.
2021
;
23
(
8
):
1231
1251
.

6.

Jannelli
 
G
,
Calvanese
 
F
,
Paun
 
L
,
Raverot
 
G
,
Jouanneau
 
E.
 
Current advances in papillary craniopharyngioma: state-of-the-art therapies and overview of the literature
.
Brain Sci
.
2023
;
13
(
3
):
515
.

7.

Henderson
 
F
,
Schwartz
 
TH.
 
Update on management of craniopharyngiomas
.
J Neurooncol.
 
2022
;
156
(
1
):
97
108
.

8.

Baqai
 
MWS
,
Shah
 
Z
,
Malik
 
MJA
, et al.  
Quality of life of pediatric patients with craniopharyngioma: a retrospective series from a low-middle-income country with more than 4 years follow-up
.
Surg Neurol Int
.
2024
;
15
:
199
.

9.

Aldave
 
G
,
Okcu
 
MF
,
Chintagumpala
 
M
, et al.  
Comparison of neurocognitive and quality-of-life outcomes in pediatric craniopharyngioma patients treated with partial resection and radiotherapy versus gross-total resection only
.
J Neurosurg Pediatr
.
2023
;
31
(
5
):
453
462
.

10.

Chen
 
A
,
Ai
 
M
,
Sun
 
T.
 
Advances in the treatment of adamantinomatous craniopharyngioma: how to balance tumor control and quality of life in the current environment: a narrative review
.
Front Oncol.
 
2023
;
13
:
1326595
.

11.

Yousuf
 
OK
,
Salehani
 
A
,
Zimmerman
 
K
, et al.  
Does subtotal resection ameliorate hypothalamic morbidity in pediatric craniopharyngioma? A 30-year retrospective cohort study
.
J Neurosurg Pediatr
.
2023
;
32
(
5
):
569
575
.

12.

Sofela
 
AA
,
Hettige
 
S
,
Curran
 
O
,
Bassi
 
S.
 
Malignant transformation in craniopharyngiomas
.
Neurosurgery.
 
2014
;
75
(
3
):
306
14
.

13.

Fukuhara
 
N
,
Nishihara
 
T
,
Sato
 
K
, et al.  
Long-term outcomes of neuroendoscopic cyst partial resection combined with stereotactic radiotherapy for craniopharyngioma
.
Acta Neurochir (Wien).
 
2024
;
166
(
1
):
218
.

14.

Del Baldo
 
G
,
Vennarini
 
S
,
Cacchione
 
A
, et al.  
Multidisciplinary management of craniopharyngiomas in children: a single center experience
.
Diagnostics
.
2022
;
12
(
11
):
2745
.

15.

Whelan
 
R
,
Prince
 
E
,
Gilani
 
A
,
Hankinson
 
T.
 
The inflammatory milieu of adamantinomatous craniopharyngioma and its implications for treatment
.
J Clin Med
.
2020
;
9
(
2
):
519
.

16.

Reyes
 
M
,
Taghvaei
 
M
,
Yu
 
S
, et al.  
Targeted therapy in the management of modern craniopharyngiomas
.
Front Biosci
.
2022
;
27
(
4
):
136
.

17.

Apps
 
JR
,
Martinez-Barbera
 
JP.
 
Molecular pathology of adamantinomatous craniopharyngioma: review and opportunities for practice
.
Neurosurg Focus.
 
2016
;
41
(
6
):
E4
.

18.

Gupta
 
S
,
Bi
 
WL
,
Giantini Larsen
 
A
, et al.  
Craniopharyngioma: a roadmap for scientific translation
.
Neurosurg Focus.
 
2018
;
44
(
6
):
E12
.

19.

Gritsch
 
D
,
Santagata
 
S
,
Brastianos
 
PK.
 
Integrating systemic therapies into the multimodality therapy of patients with craniopharyngioma
.
Curr Treat Options Oncol.
 
2024
;
25
(
2
):
261
273
.

20.

Wang
 
X
,
Zhao
 
C
,
Lin
 
J
, et al.  
Multi-omics analysis of adamantinomatous craniopharyngiomas reveals distinct molecular subgroups with prognostic and treatment response significance
.
Chin Med J (Engl).
 
2024
;
137
(
7
):
859
870
.

21.

Qi
 
S
,
Liu
 
Y
,
Wang
 
C
, et al.  
Membrane structures between craniopharyngioma and the third ventricle floor based on the QST classification and its significance: a pathological study
.
J Neuropathol Exp Neurol.
 
2020
;
79
(
9
):
966
974
.

22.

Müller
 
HL
,
Merchant
 
TE
,
Warmuth-Metz
 
M
,
Martinez-Barbera
 
JP
,
Puget
 
S. Craniopharyngioma.
 
Nat Rev Dis Primers
.
2019
;
5
(
1
):
75
.

23.

Martinez-Barbera
 
JP
,
Andoniadou
 
CL.
 
Biological behaviour of craniopharyngiomas
.
Neuroendocrinology.
 
2020
;
110
(
9-10
):
797
804
.

24.

Zacharia
 
BE
,
Bruce
 
SS
,
Goldstein
 
H
, et al.  
Incidence, treatment and survival of patients with craniopharyngioma in the surveillance, epidemiology and end results program
.
Neuro Oncol
.
2012
;
14
(
8
):
1070
1078
.

25.

Müller
 
HL
,
Merchant
 
TE
,
Puget
 
S
,
Martinez-Barbera
 
JP.
 
New outlook on the diagnosis, treatment and follow-up of childhood-onset craniopharyngioma
.
Nat Rev Endocrinol.
 
2017
;
13
(
5
):
299
312
.

26.

Johnson
 
LN
,
Hepler
 
RS
,
Yee
 
RD
,
Frazee
 
JG
,
Simons
 
KB.
 
Magnetic resonance imaging of craniopharyngioma
.
Am J Ophthalmol.
 
1986
;
102
(
2
):
242
244
.

27.

Apps
 
JR
,
Carreno
 
G
,
Gonzalez-Meljem
 
JM
, et al.  
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
.

28.

Gonzalez-Meljem
 
JM
,
Martinez-Barbera
 
JP.
 
Adamantinomatous craniopharyngioma as a model to understand paracrine and senescence-induced tumourigenesis
.
Cell Mol Life Sci.
 
2021
;
78
(
10
):
4521
4544
.

29.

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

30.

Prince
 
EW
,
Hoffman
 
LM
,
Vijmasi
 
T
, et al.  
Adamantinomatous craniopharyngioma associated with a compromised blood–brain barrier: patient series
.
J Neurosurg Case Lessons.
 
2021
;
1
(
19
):
CASE2150
.

31.

Gaston-Massuet
 
C
,
Andoniadou
 
CL
,
Signore
 
M
, et al.  
Increased Wingless (Wnt) signaling in pituitary progenitor/stem cells gives rise to pituitary tumors in mice and humans
.
Proc Natl Acad Sci U S A.
 
2011
;
108
(
28
):
11482
11487
.

32.

Brastianos
 
PK
,
Taylor-Weiner
 
A
,
Manley
 
PE
, et al.  
Exome sequencing identifies BRAF mutations in papillary craniopharyngiomas
.
Nat Genet.
 
2014
;
46
(
2
):
161
165
.

33.

Hara
 
T
,
Akutsu
 
H
,
Takano
 
S
, et al.  
Clinical and biological significance of adamantinomatous craniopharyngioma with CTNNB1 mutation
.
J Neurosurg.
 
2019
;
131
(
1
):
217
226
.

34.

Du
 
L
,
Lee
 
JH
,
Jiang
 
H
, et al.  
β-Catenin induces transcriptional expression of PD-L1 to promote glioblastoma immune evasion
.
J Exp Med.
 
2020
;
217
(
11
):
e20191115
.

35.

Galluzzi
 
L
,
Spranger
 
S
,
Fuchs
 
E
,
López-Soto
 
A.
 
WNT signaling in cancer immunosurveillance
.
Trends Cell Biol.
 
2019
;
29
(
1
):
44
65
.

36.

Liu
 
J
,
Xiao
 
Q
,
Xiao
 
J
, et al.  
Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities
.
Signal Transduct Target Ther
.
2022
;
7
(
1
):
3
.

37.

Goschzik
 
T
,
Gessi
 
M
,
Dreschmann
 
V
, et al.  
Genomic alterations of adamantinomatous and papillary craniopharyngioma
.
J Neuropathol Exp Neurol.
 
2017
;
76
(
2
):
126
134
.

38.

Yoshimoto
 
K
,
Hatae
 
R
,
Suzuki
 
SO
, et al.  
High-resolution melting and immunohistochemical analysis efficiently detects mutually exclusive genetic alterations of adamantinomatous and papillary craniopharyngiomas
.
Neuropathol
.
2018
;
38
(
1
):
3
10
.

39.

Jucá
 
CEB
,
Colli
 
LM
,
Martins
 
CS
, et al.  
Impact of the canonical Wnt pathway activation on the pathogenesis and prognosis of adamantinomatous craniopharyngiomas
.
Horm Metab Res.
 
2018
;
50
(
7
):
575
581
.

40.

Apps
 
JR
,
Stache
 
C
,
Gonzalez-Meljem
 
JM
, et al.  
CTNNB1 mutations are clonal in adamantinomatous craniopharyngioma
.
Neuropathol Appl Neurobiol.
 
2020
;
46
(
5
):
510
514
.

41.

Campanini
 
ML
,
Almeida
 
JP
,
Martins
 
CS
,
de Castro
 
M.
 
The molecular pathogenesis of craniopharyngiomas
.
Arch Endocrinol Metab
.
2023
;
67
(
2
):
266
275
.

42.

Passos
 
J
,
Quidet
 
M
,
Brahimi
 
A
, et al.  
Familial adenomatous polyposis associated craniopharyngioma secondary to both germline and somatic mutations in the APC gene
.
Acta Neuropathol.
 
2020
;
140
(
6
):
967
969
.

43.

Jin
 
L
,
Cai
 
K
,
Wu
 
W
, et al.  
Correlations between the expression of molecules in the TGF-β signaling pathway and clinical factors in adamantinomatous craniopharyngiomas
.
Front Endocrinol (Lausanne)
.
2023
;
14
:
1167776
.

44.

Yeh
 
HW
,
Hsu
 
EC
,
Lee
 
SS
, et al.  
PSPC1 mediates TGF-β1 autocrine signalling and Smad2/3 target switching to promote EMT, stemness and metastasis
.
Nat Cell Biol.
 
2018
;
20
(
4
):
479
491
.

45.

Brabletz
 
T
,
Hlubek
 
F
,
Spaderna
 
S
, et al.  
Invasion and metastasis in colorectal cancer: epithelial-mesenchymal transition, mesenchymal-epithelial transition, stem cells and β-catenin
.
Cells Tissues Organs
.
2005
;
179
(
1-2
):
56
65
.

46.

Sato
 
R
,
Imamura
 
K
,
Semba
 
T
, et al.  
TGFβ signaling activated by cancer-associated fibroblasts determines the histological signature of lung adenocarcinoma
.
Cancer Res.
 
2021
;
81
(
18
):
4751
4765
.

47.

Lian
 
S
,
Xie
 
R
,
Ye
 
Y
, et al.  
Simultaneous blocking of CD47 and PD-L1 increases innate and adaptive cancer immune responses and cytokine release
.
EBioMed
.
2019
;
42
:
281
295
.

48.

Lu
 
Y
,
Liang
 
H
,
Yu
 
T
, et al.  
Isolation and characterization of living circulating tumor cells in patients by immunomagnetic negative enrichment coupled with flow cytometry
.
Cancer.
 
2015
;
121
(
17
):
3036
3045
.

49.

Anderson
 
KL
,
Snyder
 
KM
,
Ito
 
D
, et al.  
Evolutionarily conserved resistance to phagocytosis observed in melanoma cells is insensitive to upregulation of pro-phagocytic signals and to CD47 blockade
.
Melanoma Res.
 
2020
;
30
(
2
):
147
158
.

50.

Zhang
 
H
,
Wang
 
C
,
Fan
 
J
, et al.  
CD47 promotes the proliferation and migration of adamantinomatous craniopharyngioma cells by activating the MAPK/ERK pathway, and CD47 blockade facilitates microglia‐mediated phagocytosis
.
Neuropathology Appl Neurobio.
 
2022
;
48
(
4
):
e12795
.

51.

Musani
 
V
,
Gorry
 
P
,
Basta-Juzbasic
 
A
, et al.  
Mutation in exon 7 of PTCH deregulates SHH/PTCH/SMO signaling: possible linkage to WNT
.
Int J Mol Med.
 
2006
;
17
(
5
):
755
759
.

52.

Gomes
 
DC
,
Jamra
 
SA
,
Leal
 
LF
, et al.  
Sonic Hedgehog pathway is upregulated in adamantinomatous craniopharyngiomas
.
Eur J Endocrinol.
 
2015
;
172
(
5
):
603
608
.

53.

Zhao
 
J
,
Yang
 
Y
,
Pan
 
Y
, et al.  
Transcription factor GLI1 induces IL-6-mediated inflammatory response and facilitates the progression of adamantinomatous craniopharyngioma
.
ACS Chem Neurosci.
 
2023
;
14
(
18
):
3347
3356
.

54.

Halder
 
S
,
Basu
 
S
,
Lall
 
SP
, et al.  
Targeting the EGFR signaling pathway in cancer therapy: what’s new in 2023
?
Expert Opin Ther Targets.
 
2023
;
27
(
4-5
):
305
324
.

55.

Robinson
 
LC
,
Santagata
 
S
,
Hankinson
 
TC.
 
Potential evolution of neurosurgical treatment paradigms for craniopharyngioma based on genomic and transcriptomic characteristics
.
Neurosurg Focus.
 
2016
;
41
(
6
):
E3
.

56.

Hölsken
 
A
,
Gebhardt
 
M
,
Buchfelder
 
M
, et al.  
EGFR signaling regulates tumor cell migration in craniopharyngiomas
.
Clin Cancer Res
.
2011
;
17
(
13
):
4367
4377
.

57.

Stache
 
C
,
Hölsken
 
A
,
Schlaffer
 
S
, et al.  
Insights into the infiltrative behavior of adamantinomatous craniopharyngioma in a new xenotransplant mouse model
.
Brain Pathol
.
2015
;
25
(
1
):
1
10
.

58.

Vidal
 
S
,
Kovacs
 
K
,
Lloyd
 
RV
,
Meyer
 
FB
,
Scheithauer
 
BW.
 
Angiogenesis in patients with craniopharyngiomas: correlation with treatment and outcome
.
Cancer.
 
2002
;
94
(
3
):
738
745
.

59.

Xia
 
Z
,
Liu
 
W
,
Li
 
S
, et al.  
Expression of matrix metalloproteinase-9, type IV collagen and vascular endothelial growth factor in adamantinous craniopharyngioma
.
Neurochem Res.
 
2011
;
36
(
12
):
2346
2351
.

60.

Jia
 
Y
,
Ma
 
L
,
Cai
 
K
, et al.  
Immune infiltration in aggressive papillary craniopharyngioma: high infiltration but low action
.
Front Immunol.
 
2022
;
13
:
995655
.

61.

Donson
 
AM
,
Apps
 
J
,
Griesinger
 
AM
, et al. ;
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
.

62.

Grob
 
S
,
Mirsky
 
DM
,
Donson
 
AM
, et al.  
Targeting IL-6 is a potential treatment for primary cystic craniopharyngioma
.
Front Oncol.
 
2019
;
9
:
791
.

63.

Gonzalez-Meljem
 
JM
,
Haston
 
S
,
Carreno
 
G
, et al.  
Stem cell senescence drives age-attenuated induction of pituitary tumours in mouse models of paediatric craniopharyngioma
.
Nat Commun.
 
2017
;
8
(
1
):
1819
.

64.

Prince
 
EW
,
Apps
 
JR
,
Jeang
 
J
, et al.  
Unraveling the complexity of the senescence-associated secretory phenotype in adamantinomatous craniopharyngioma using multimodal machine learning analysis
.
Neuro-Oncology.
 
2024
;
26
(
6
):
1109
1123
.

65.

Wang
 
X
,
Lin
 
J
,
Liu
 
H
, et al.  
Single-cell and spatial sequencing identifies senescent and germinal tumor cells in adamantinomatous craniopharyngiomas
.
Cell Biosci
.
2024
;
14
(
1
):
112
.

66.

Wang
 
B
,
Kohli
 
J
,
Demaria
 
M.
 
Senescent cells in cancer therapy: friends or foes
?
Trends Cancer
.
2020
;
6
(
10
):
838
857
.

67.

Mortini
 
P
,
Losa
 
M
,
Pozzobon
 
G
, et al.  
Neurosurgical treatment of craniopharyngioma in adults and children: early and long-term results in a large case series
.
J Neurosurg.
 
2011
;
114
(
5
):
1350
1359
.

68.

Castle-Kirszbaum
 
M
,
Shi
 
MDY
,
Goldschlager
 
T.
 
Quality of life in craniopharyngioma: a systematic review
.
World Neurosurg
.
2022
;
164
:
424
435.e2
.

69.

Puget
 
S
,
Garnett
 
M
,
Wray
 
A
, et al.  
Pediatric craniopharyngiomas: classification and treatment according to the degree of hypothalamic involvement
.
J Neurosurg.
 
2007
;
106
(
1 Suppl
):
3
12
.

70.

Prieto
 
R
,
Pascual
 
JM
,
Rosdolsky
 
M
, et al.  
Craniopharyngioma adherence: a comprehensive topographical categorization and outcome-related risk stratification model based on the methodical examination of 500 tumors
.
Neurosurg Focus.
 
2016
;
41
(
6
):
E13
.

71.

Hori
 
T.
 
What is the role of radiosurgery in the management of sellar tumors
?
Acta Neurochir Suppl.
 
2013
;
116
:
45
48
.

72.

Pascual
 
JM
,
Prieto
 
R
,
Rosdolsky
 
M.
 
Craniopharyngiomas primarily affecting the hypothalamus
.
Handb Clin Neurol
.
2021
;
181
(
7
):
75
115
.

73.

Calandrelli
 
R
,
D’Apolito
 
G
,
Martucci
 
M
, et al.  
Topography and radiological variables as ancillary parameters for evaluating tissue adherence, hypothalamic-pituitary dysfunction, and recurrence in craniopharyngioma: an integrated multidisciplinary overview
.
Cancers (Basel)
.
2024
;
16
(
14
):
2532
.

74.

Müller
 
HL.
 
The diagnosis and treatment of craniopharyngioma
.
Neuroendocrinology.
 
2020
;
110
(
9-10
):
753
766
.

75.

Ciric
 
IS
,
Cozzens
 
JW.
 
Craniopharyngiomas: transsphenoidal method of approach—for the virtuoso only
?
Clin Neurosurg.
 
1980
;
27
:
169
187
.

76.

Steno
 
J.
 
Microsurgical topography of craniopharyngiomas
.
Acta Neurochir Suppl (Wien).
 
1985
;
35
:
94
100
.

77.

Yaşargil
 
MG
,
Curcic
 
M
,
Kis
 
M
, et al.  
Total removal of craniopharyngiomas. Approaches and long-term results in 144 patients
.
J Neurosurg.
 
1990
;
73
(
1
):
3
11
.

78.

Hoffman
 
HJ.
 
Surgical management of craniopharyngioma
.
Pediatr Neurosurg.
 
1994
;
21
(
Suppl 1
):
44
49
.

79.

Samii
 
M
,
Tatagiba
 
M.
 
Surgical management of craniopharyngiomas: a review
.
Neurol Med Chir (Tokyo).
 
1997
;
37
(
2
):
141
149
.

80.

Pascual
 
JM
,
González-Llanos
 
F
,
Barrios
 
L
,
Roda
 
JM.
 
Intraventricular craniopharyngiomas: topographical classification and surgical approach selection based on an extensive overview
.
Acta Neurochir (Wien).
 
2004
;
146
(
8
):
785
802
.

81.

Wang
 
KC
,
Hong
 
SH
,
Kim
 
SK
,
Cho
 
BK.
 
Origin of craniopharyngiomas: implication on the growth pattern
.
Childs Nerv Syst.
 
2005
;
21
(
8-9
):
628
634
.

82.

Kassam
 
AB
,
Gardner
 
PA
,
Snyderman
 
CH
, et al.  
Expanded endonasal approach, a fully endoscopic transnasal approach for the resection of midline suprasellar craniopharyngiomas: a new classification based on the infundibulum
.
J Neurosurg.
 
2008
;
108
(
4
):
715
728
.

83.

Tang
 
B
,
Xie
 
S
,
Huang
 
G
, et al.  
Clinical features and operative technique of transinfundibular craniopharyngioma
.
J Neurosurg.
 
2019
;
133
(
1
):
119
128
.

84.

Fatemi
 
N
,
Dusick
 
JR
,
de Paiva Neto
 
MA
,
Malkasian
 
D
,
Kelly
 
DF.
 
Endonasal versus supraorbital keyhole removal of craniopharyngiomas and tuberculum sellae meningiomas
.
Neurosurgery.
 
2009
;
64
(
5
):
ons269
ons287
.

85.

Yamada
 
S
,
Fukuhara
 
N
,
Oyama
 
K
, et al.  
Surgical outcome in 90 patients with craniopharyngioma: an evaluation of transsphenoidal surgery
.
World Neurosurg
.
2010
;
74
(
2-3
):
320
330
.

86.

Qi
 
S
,
Lu
 
Y
,
Pan
 
J
, et al.  
Anatomic relations of the arachnoidea around the pituitary stalk: relevance for surgical removal of craniopharyngiomas
.
Acta Neurochir (Wien).
 
2011
;
153
(
4
):
785
796
.

87.

Hu
 
W
,
Qiu
 
B
,
Mei
 
F
, et al.  
Clinical impact of craniopharyngioma classification based on location origin: a multicenter retrospective study
.
Ann Transl Med
.
2021
;
9
(
14
):
1164
.

88.

Matsuo
 
T
,
Kamada
 
K
,
Izumo
 
T
,
Nagata
 
I.
 
Indication and limitations of endoscopic extended transsphenoidal surgery for craniopharyngioma
.
Neurol Med Chir (Tokyo).
 
2014
;
54
(
12
):
974
982
.

89.

Morisako
 
H
,
Goto
 
T
,
Goto
 
H
, et al.  
Aggressive surgery based on an anatomical subclassification of craniopharyngiomas
.
Neurosurg Focus.
 
2016
;
41
(
6
):
E10
.

90.

Jeswani
 
S
,
Nuño
 
M
,
Wu
 
A
, et al.  
Comparative analysis of outcomes following craniotomy and expanded endoscopic endonasal transsphenoidal resection of craniopharyngioma and related tumors: a single-institution study
.
J Neurosurg.
 
2016
;
124
(
3
):
627
638
.

91.

Tang
 
B
,
Xie
 
SH
,
Xiao
 
LM
, et al.  
A novel endoscopic classification for craniopharyngioma based on its origin
.
Sci Rep.
 
2018
;
8
(
1
):
10215
.

92.

Prieto
 
R
,
Rosdolsky
 
M
,
Hofecker
 
V
,
Barrios
 
L
,
Pascual
 
JM.
 
Craniopharyngioma treatment: an updated summary of important clinicopathological concepts
.
Expert Rev Endocrinol Metab
.
2020
;
15
(
4
):
261
282
.

93.

Prieto
 
R
,
Pascual
 
JM
,
Castro-Dufourny
 
I
,
Carrasco
 
R
,
Barrios
 
LC.
 
Surgical outcome as related to the degree of hypothalamic involvement
.
World Neurosurg
.
2017
;
104
:
1006
1010
.

94.

Almeida
 
JP
,
Workewych
 
A
,
Takami
 
H
, et al.  
Surgical anatomy applied to the resection of craniopharyngiomas: anatomic compartments and surgical classifications
.
World Neurosurg
.
2020
;
142
:
611
625
.

95.

Lei
 
C
,
Chuzhong
 
L
,
Chunhui
 
L
, et al.  
Approach selection and outcomes of craniopharyngioma resection: a single-institute study
.
Neurosurg Rev.
 
2021
;
44
(
3
):
1737
1746
.

96.

Massimi
 
L
,
Palombi
 
D
,
Musarra
 
A
, et al.  
Adamantinomatous craniopharyngioma: evolution in the management
.
Childs Nerv Syst.
 
2023
;
39
(
10
):
2613
2632
.

97.

Wu
 
J
,
Pan
 
C
,
Xie
 
S
, et al.  
A propensity-adjusted comparison of endoscopic endonasal surgery versus transcranial microsurgery for pediatric craniopharyngioma: a single-center study
.
J Neurosurg Pediatr
.
2022
;
29
(
3
):
325
334
.

98.

Na
 
MK
,
Jang
 
B
,
Choi
 
KS
, et al.  
Craniopharyngioma resection by endoscopic endonasal approach versus transcranial approach: a systematic review and meta-analysis of comparative studies
.
Front Oncol.
 
2022
;
12
:
1058329
.

99.

Müller
 
HL
,
Tauber
 
M
,
Lawson
 
EA
, et al.  
Hypothalamic syndrome
.
Nat Rev Dis Primers.
 
2022
;
8
(
1
):
24
.

100.

Otte
 
A
,
Müller
 
HL.
 
Childhood-onset craniopharyngioma
.
J Clin Endocrinol Metab.
 
2021
;
106
(
10
):
e3820
e3836
.

101.

Jiang
 
Y
,
Yang
 
J
,
Liang
 
R
, et al.  
Single-cell RNA sequencing highlights intratumor heterogeneity and intercellular network featured in adamantinomatous craniopharyngioma
.
Sci Adv.
 
2023
;
9
(
15
):
eadc8933
.

102.

Pang
 
JC
,
Chung
 
DD
,
Wang
 
J
, et al.  
Characteristics and outcomes in pediatric versus adult craniopharyngiomas: a systematic review and meta-analysis
.
Neurosurgery.
 
2023
;
92
(
6
):
1112
1129
.

103.

Alboqami
 
MN
,
Khalid S Albaiahy
 
A
,
Bukhari
 
BH
, et al.  
Craniopharyngioma: a comprehensive review of the clinical presentation, radiological findings, management, and future Perspective
.
Heliyon
.
2024
;
10
(
11
):
e32112
.

104.

Alexandraki
 
KI
,
Xekouki
 
P.
 
Medical therapy for craniopharyngiomas.
touchREV Endocrinol.
2021
;
17
(
2
):
121
132
.

105.

Paleari
 
L.
 
Cancer prevention with molecular targeted therapies
.
Int J Mol Sci .
 
2022
;
23
(
15
):
8429
.

106.

Skálová
 
A
,
Bradová
 
M
,
Michal
 
M
, et al.  
Molecular pathology in diagnosis and prognostication of head and neck tumors
.
Virchows Arch.
 
2024
;
484
(
2
):
215
231
.

107.

Sekine
 
S
,
Shibata
 
T
,
Kokubu
 
A
, et al.  
Craniopharyngiomas of adamantinomatous type harbor β-Catenin gene mutations
.
Am J Pathol.
 
2002
;
161
(
6
):
1997
2001
.

108.

Mota
 
JIS
,
Silva-Júnior
 
RMP
,
Martins
 
CS
, et al.  
Telomere length and Wnt/β-catenin pathway in adamantinomatous craniopharyngiomas
.
Eur J Endocrinol.
 
2022
;
187
(
2
):
219
230
.

109.

Hölsken
 
A
,
Kreutzer
 
J
,
Hofmann
 
BM
, et al.  
Target gene activation of the Wnt signaling pathway in nuclear beta-catenin accumulating cells of adamantinomatous craniopharyngiomas
.
Brain Pathol.
 
2009
;
19
(
3
):
357
364
.

110.

Marrero-Gutiérrez
 
J
,
Bueno
 
AC
,
Martins
 
CS
, et al.  
Epigenetic control of adamantinomatous craniopharyngiomas
.
J Clin Endocrinol Metab.
 
2024
;
109
(
10
):
e1867
e1880
.

111.

Chen
 
Y
,
Liu
 
X
,
Ainiwan
 
Y
, et al.  
Axl as a potential therapeutic target for adamantinomatous craniopharyngiomas: based on single nucleus RNA-seq and spatial transcriptome profiling
.
Cancer Lett.
 
2024
;
592
:
216905
.

112.

Wu
 
J
,
Qin
 
C
,
Fang
 
G
, et al.  
Machine learning approach to screen new diagnostic features of adamantinomatous craniopharyngioma and explore personalised treatment strategies
.
Transl Pediatr
.
2023
;
12
(
5
):
947
966
.

113.

Wang
 
Y
,
Deng
 
J
,
Guo
 
G
, et al.  
Clinical and prognostic role of annexin A2 in adamantinomatous craniopharyngioma
.
J Neurooncol.
 
2017
;
131
(
1
):
21
29
.

114.

Zhao
 
C
,
Wang
 
Y
,
Liu
 
H
, et al.  
Molecular biological features of cyst wall of adamantinomatous craniopharyngioma
.
Sci Rep.
 
2023
;
13
(
1
):
3049
.

115.

Yuan
 
F
,
Cai
 
X
,
Zhu
 
J
, et al.  
A novel immune classification for predicting immunotherapy responsiveness in patients with adamantinomatous craniopharyngioma
.
Front Neurol.
 
2021
;
12
:
704130
.

116.

Omay
 
SB
,
Chen
 
YN
,
Almeida
 
JP
, et al.  
Do craniopharyngioma molecular signatures correlate with clinical characteristics
?
J Neurosurg.
 
2018
;
128
(
5
):
1473
1478
.

117.

Zhang
 
H
,
Wang
 
C
,
Fan
 
J
, et al.  
Factor analysis and subtyping significance of CTNNB1 gene mutation detection in adamantinomatous craniopharyngioma
.
Genes Dis
.
2024
;
11
(
6
):
101188
.

118.

Coy
 
S
,
Rashid
 
R
,
Lin
 
JR
, et al.  
Multiplexed immunofluorescence reveals potential PD-1/PD-L1 pathway vulnerabilities in craniopharyngioma
.
Neuro Oncol
.
2018
;
20
(
8
):
1101
1112
.

119.

Wang
 
Y
,
Deng
 
J
,
Wang
 
L
, et al.  
Expression and clinical significance of PD-L1, B7-H3, B7-H4 and VISTA in craniopharyngioma
.
J ImmunoTher Cancer.
 
2020
;
8
(
2
):
e000406
.

120.

Karpathiou
 
G
,
Hamlat
 
M
,
Dridi
 
M
, et al.  
Autophagy and immune microenvironment in craniopharyngioma and ameloblastoma
.
Exp Mol Pathol.
 
2021
;
123
:
104712
.

121.

Whelan
 
R
,
Hengartner
 
A
,
Folzenlogen
 
Z
,
Prince
 
E
,
Hankinson
 
TC.
 
Adamantinomatous craniopharyngioma in the molecular age and the potential of targeted therapies: a review
.
Childs Nerv Syst.
 
2020
;
36
(
8
):
1635
1642
.

122.

Prince
 
E
,
Whelan
 
R
,
Donson
 
A
, et al. ;
Advancing Treatment for Pediatric Craniopharyngioma Consortium
.
Transcriptional analyses of adult and pediatric adamantinomatous craniopharyngioma reveals similar expression signatures regarding potential therapeutic targets
.
Acta Neuropathol Commun
.
2020
;
8
(
1
):
68
.

123.

Petralia
 
F
,
Tignor
 
N
,
Reva
 
B
, et al. ;
Children’s Brain Tumor Network
.
Integrated proteogenomic characterization across major histological types of pediatric brain cancer
.
Cell.
 
2020
;
183
(
7
):
1962
1985.e31
.

124.

Patel
 
K
,
Allen
 
J
,
Zagzag
 
D
, et al.  
Radiologic response to MEK inhibition in a patient with a WNT-activated craniopharyngioma
.
Pediatr Blood Cancer.
 
2021
;
68
(
3
):
e28753
.

125.

Massimi
 
L
,
Martelli
 
C
,
Caldarelli
 
M
,
Castagnola
 
M
,
Desiderio
 
C.
 
Proteomics in pediatric cystic craniopharyngioma
.
Brain Pathol.
 
2017
;
27
(
3
):
370
376
.

126.

Agosti
 
E
,
Zeppieri
 
M
,
Antonietti
 
S
, et al.  
Advancing craniopharyngioma management: a systematic review of current targeted therapies and future perspectives
.
Int J Mol Sci .
 
2024
;
25
(
2
):
723
.

127.

Hengartner
 
AC
,
Prince
 
E
,
Vijmasi
 
T
,
Hankinson
 
TC.
 
Adamantinomatous craniopharyngioma: moving toward targeted therapies
.
Neurosurg Focus.
 
2020
;
48
(
1
):
E7
.

128.

Jakacki
 
RI
,
Cohen
 
BH
,
Jamison
 
C
, et al.  
Phase II evaluation of interferon α-2a for progressive or recurrent craniopharyngiomas
.
J Neurosurg.
 
2000
;
92
(
2
):
255
260
.

129.

Yeung
 
JT
,
Pollack
 
IF
,
Panigrahy
 
A
,
Jakacki
 
RI.
 
Pegylated interferon-α-2b for children with recurrent craniopharyngioma
.
J Neurosurg Pediatr
.
2012
;
10
(
6
):
498
503
.

130.

Kilday
 
JP
,
Caldarelli
 
M
,
Massimi
 
L
, et al.  
Intracystic interferon-alpha in pediatric craniopharyngioma patients: an international multicenter assessment on behalf of SIOPE and ISPN
.
Neuro Oncol
.
2017
;
19
(
10
):
1398
1407
.

131.

Goldman
 
S
,
Pollack
 
IF
,
Jakacki
 
RI
, et al.  
Phase II study of peginterferon alpha-2b for patients with unresectable or recurrent craniopharyngiomas: a pediatric brain tumor consortium report
.
Neuro Oncol
.
2020
;
22
(
11
):
1696
1704
.

132.

De Vos-Kerkhof
 
E
,
Buis
 
DR
,
Lequin
 
MH
, et al.  
Tocilizumab for the fifth progression of cystic childhood craniopharyngioma—a case report
.
Front Endocrinol
.
2023
;
14
:
1225734
.

133.

ClinicalTrials.gov
. ACTEMRA® for the treatment of pediatric adamantinomatous craniopharyngioma (NCT05233397).
n.d
. https://clinicaltrials.gov/study/NCT05233397. Retrieved
December 12, 2024
.

134.

De Rosa
 
A
,
Calvanese
 
F
,
Ducray
 
F
, et al.  
First evidence of anti-VEGF efficacy in an adult case of adamantinomatous craniopharyngioma: case report and illustrative review
.
Ann Endocrinol (Paris).
 
2023
;
84
(
6
):
727
733
.

135.

ClinicalTrials.gov
. MEKTOVI® for the treatment of pediatric adamantinomatous craniopharyngioma (NCT05286788).
n.d
. https://clinicaltrials.gov/study/NCT05286788. Retrieved
December 12, 2024
.

136.

ClinicalTrials.gov
. Nivolumab and Tovorafenib for treatment of craniopharyngioma in children and young adults (PNOC029) (NCT05465174).
n.d
. https://clinicaltrials.gov/study/NCT05465174. Retrieved
December 12, 2024
.

137.

Ma
 
G
,
Kang
 
J
,
Qiao
 
N
, et al.  
Non-invasive radiomics approach predict invasiveness of adamantinomatous craniopharyngioma before surgery
.
Front Oncol.
 
2020
;
10
:
599888
.

138.

Wahida
 
A
,
Buschhorn
 
L
,
Fröhling
 
S
, et al.  
The coming decade in precision oncology: six riddles
.
Nat Rev Cancer.
 
2023
;
23
(
1
):
43
54
.

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact [email protected] for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact [email protected].