Next Article in Journal
Exercise Intervention for Alzheimer’s Disease: Unraveling Neurobiological Mechanisms and Assessing Effects
Next Article in Special Issue
From Dormant Collections to Repositories for the Study of Habitat Changes: The Importance of Herbaria in Modern Life Sciences
Previous Article in Journal
Imaging of Strictures in Crohn’s Disease
Previous Article in Special Issue
Review of Predicting Synergistic Drug Combinations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Domestic Animal Models of Central Nervous System Tumors: Focus on Meningiomas

1
Department of Experimental Medicine, University of Genova, 16132 Genova, Italy
2
Pharmacology Section, Department of Internal Medicine (DIMI), University of Genova, 16126 Genova, Italy
3
IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
4
National Reference Center for Veterinary and Comparative Oncology, Veterinary Medical Research Institute for Piemonte, Liguria and Valle d’Aosta, 10154 Torino, Italy
*
Authors to whom correspondence should be addressed.
Life 2023, 13(12), 2284; https://doi.org/10.3390/life13122284
Submission received: 13 October 2023 / Accepted: 9 November 2023 / Published: 30 November 2023
(This article belongs to the Collection Feature Review Papers for Life)

Abstract

:
Intracranial primary tumors (IPTs) are aggressive forms of malignancies that cause high mortality in both humans and domestic animals. Meningiomas are frequent adult IPTs in humans, dogs, and cats, and both benign and malignant forms cause a decrease in life quality and survival. Surgery is the primary therapeutic approach to treat meningiomas, but, in many cases, it is not resolutive. The chemotherapy and targeted therapy used to treat meningiomas also display low efficacy and many side effects. Therefore, it is essential to find novel pharmacological approaches to increase the spectrum of therapeutic options for meningiomas. This review analyzes the similarities between human and domestic animal (dogs and cats) meningiomas by evaluating the molecular and histological characteristics, diagnosis criteria, and treatment options and highlighting possible research areas to identify novel targets and pharmacological approaches, which are useful for the diagnosis and therapy of this neoplasia to be used in human and veterinary medicine.

1. Introduction

Intracranial primary tumors (IPTs) constitute a heterogeneous class of neoplasms characterized by a high mortality rate in humans. According to the International Classification of Oncology Diseases by the World Health Organization (WHO), IPTs are classified into more than 100 different tumor types [1]. They can manifest at any age and include both malignant and non-malignant tumors. In infants and adolescents, malignant tumors are the most frequently diagnosed IPTs, occurring at a rate of 3.55 cases per 100,000 individuals. Instead, non-malignant forms are less frequently diagnosed, with an incidence of 2.60 cases per 100,000 individuals [2]. The most common malignant IPT in infants is glioma, whereas pituitary tumor stands out as the most frequently occurring non-malignant form. The mortality rates exhibit variability but, on the whole, signify a substantial cause of death within this population group. In adults, IPTs rank as the eighth most common cancer. Although non-malignant tumors are more common in adults, with an incidence of 16.33 cases per 100,000 individuals, than in children, malignant tumors reach a noteworthy incidence of 7.08/100,000 [3]. Additionally, in adults, gliomas represent the most diagnosed form among malignant IPTs, whereas meningiomas and pituitary tumors are the most frequent non-malignant forms.
IPTs have been associated with fatal outcomes in various animal species, with extensive research conducted in dogs and cats. The most frequently occurring IPTs in dogs and cats include gliomas, meningiomas, and choroid plexus tumors. The incidence of these tumors appears to be higher in dogs compared to cats: about 14.5 cases per 100,000 dogs and an estimated 3.5 cases per 100,000 cats [4,5]. The incidence is strongly correlated with the age of the animal, with IPTs occurring in middle-aged to older dogs and in older cats. Most reports indicate that meningioma diagnoses occur in dogs over 7 years of age and in cats over 9 years of age [6]. In juvenile dogs, intracranial tumors are more commonly identified as neuroepithelial tumors of glial, neuronal, or embryonal origin [4]. One study found a statistically significant linear relationship between age and body weight and the occurrence of IPT in dogs [7]. Although meningiomas are the most common IPTs in dogs, oligodendrogliomas and gliomas are also very frequent [4,8]. Feline meningiomas have the highest incidence with respect to other brain tumor forms [9]. Canine and feline IPT incidence also seems to depend on the breed [7,9,10,11]. A genetic predisposition to canine glioma exists, with the protein expression of TP53, MDM2, P21, AKT, PTEN, RB1, P16, MTOR, and MAPK correlated to the specific glioma subtype [12]; moreover, a genome-wide association study followed by regional mapping [13] identified single nucleotide variants in three neighboring genes, DENR, CAMKK2, and P2RX7, which are highly associated with glioma susceptibility [13]. Intracranial tumors can also manifest as secondary tumors in dogs, representing about 50% of all intracranial tumor diagnoses and exhibiting a higher frequency than primary tumors. These metastases primarily originate from hemangiosarcoma (about 30%), lymphoma (about 20%), and metastatic carcinomas (about 20%) [11]. Intracranial tumors account for 2.2% of all tumors diagnosed in cats. It has been reported that meningiomas are the most prevalent primary brain tumors, comprising 58.1%, while lymphomas are the most frequently occurring secondary tumors, accounting for 14.4%.
In this review, we report comparative aspects of the etiology, histology, and diagnosis of meningiomas in humans, dogs, and cats. Additionally, we delve into the molecular aspects that characterize tumors in these three species, along with therapeutic approaches in order to summarize the similarities and differences, which can be useful in the study of these pathologies under a “One Health” perspective.
Following the “One Health” approach, various animal model validations were carried out for numerous types of cancer, including CNS tumors, to improve the medical practices in both the human and veterinary fields [14].
The focus of this review on meningiomas stems from their significance; despite the benign nature of a large percentage of these tumors, their growth can lead to dementia and clinical symptoms, severely compromising the quality of life. Moreover, a high rate of relapse is present in about 20% of histologically benign meningiomas, even after a radical excision, determining a poor prognosis. In some cases, meningiomas are diagnosed in multiple forms that cannot be treated by surgery [15,16,17,18,19]. However, surgery is not always curative and, under specific conditions, may pose risks to health. Therefore, the exploration of alternative therapeutic approaches, leveraging insights from animal models, could provide novel valid options for this type of neoplasm.
The literature predominantly features animal models of meningiomas involving murine and domestic species. Nonetheless, studies on meningiomas have also been conducted in horses, cattle, and pigs [20]. Murine models of meningiomas have facilitated the examination of mutations that characterize human tumors through primary cultures and xenografts. These models permitted the evaluation of the role of stem cells in tumor biology [21,22,23,24]. The use of murine models, mainly Genetically Engineered Mice (GEM) and Patient-Derived Xenograft (PDX), in oncology is particularly important due to their wide availability and economic feasibility. However, translating data from these models to humans presents several limitations, as discussed in previous reviews by Xu, C. et al. (2019) and Cho, S.Y. et al. (2020) [25,26]. GEMs offer a valuable tool for studying the response to immunotherapy, due to their intact immune systems, and to observe the physiological tumor environment, but other studies presented variability in the clinical presentation caused by altered phenotypes [26,27]. On the other hand, PDX models reflect the cellular complexity and histological architecture of real tumors, but, unlike GEM, they lack immune system information. Consequently, they are not suitable for studying immunotherapy [26].
In general, manipulating murine models at the central nervous system (CNS) level poses challenges due to their small size. The most notable limitation in mouse models lies in tumor induction. Induced models lack many important features found in spontaneously occurring models [27,28]. Several naturally occurring inherited and acquired neurological diseases, including meningiomas, that occur in dogs and cats have analogous counterparts in humans [29]. The genetic and environmental characteristics of tumors have been studied, revealing several analogies with human cancer [30,31,32,33]. Moreover, due to the close sharing of the environment, domestic animals and humans are exposed to similar environmental and risk factors that promote the spontaneous onset of tumors. This condition allows the use of domestic animals not only as models but also as a sentinel to predict an environmental risk for humans [34]. Some cases of sentinel dogs are reported in the literature [18,19,20,21,22,23]. However, it is important to remark the different incidence of specific tumors between domestic animals and humans. This aspect suggests an important role of biological risk factors present in both species in the onset of tumors and not only environmental factors [35,36].
Domestic animal models overcome some limitations observed in murine models when studying tumors. Consequently, domestic animals have been recognized as suitable pre-clinical models for investigating various types of tumors, including brain tumors [4,28,29,37]. In this context, the use of domestic animals as a model for meningiomas may allow comparative studies on a larger size of samples with histological, molecular, and neuro-imaging aspects similar to humans within immunocompetent organisms.
Moreover, the validation of the canine model may have an additional advantage, considering that the analysis of the canine genome reported nucleotide sequences and rearrangements more similar to the human genome than that of rodents and the sharing of similar gene expression profiles [37,38].
At the same time, domestic animals also have limitations in their use as a model for human cancer studies. The use of domestic animals implies higher efforts and costs for sample collection compared to the mouse model. The heterogeneity of the tumors can be variable between different species, and it could be a disadvantage during the study of specific molecular pathways if no in-depth knowledge exists. The incidence of the tumors is different between humans and pets for some cancer types [39]. Another obstacle can be the ethics and oversights of possible trials with pets. Every kind of trial (even the use of collected specimens for research) need an informed consent process [40] and the emotional state of the owners could affect the decision. Moreover, the execution of additional diagnostics procedures or invasive exams could be not accepted by the owners [40].
Table 1 summarizes the advantages and disadvantages of using domestic animals as translational cancer models.
In 2004, the Canine Comparative Oncology and Genomics Consortium (CCOGC) was established, creating an extensive biospecimen repository of canine cancers and tissues. The consortium strengthened connections among veterinary oncologists, researchers, and clinicians and initiated non-clinical trials employing dogs for the development of novel paths for new cancer drugs [41].
Brain tumors in dogs and humans arise spontaneously with similar incidence [4]. The existence of species in which tumors arise spontaneously represents the best translational model. An important advantage of the comparative studies of CNS cancer in companion animals is determined by the possibility of evaluating the role of the blood–brain barrier (BBB) in the pharmacokinetics and pharmacodynamics of the drugs; in fact, the physical size and vascular biology of the BBB are similar in pets and humans [42,43,44].
When considering brain cancers, it is important to highlight that these tumors represent a low percentage of canine cancer diagnoses. Therefore, the number of dogs that can be recruited for studies is low [40]. Conducting potential clinical trials involving dogs may take approximately 1–3 years, a duration that may be financially challenging for small biotechnology companies to support.
To improve the employment of companion animals as translational models for brain cancer in humans, a “Comparative Brain Tumor Consortium” (CBTC) was organized. The CBTC includes the comparative oncology program and the neuro-oncology branch by the National Cancer Institute (NIH) in association with an external community of radiologists, neuro-oncologists, radiation oncologists, and surgeons. The functions of CBTC include the study of the pathology and molecular markers, drug discovery, tumor biology, and immunology. Moreover, CBTC tries to simplify the organization of clinical trials regarding the recruitment of animals [32].

2. Meningiomas

Meningiomas frequently occur in humans and domestic animals. Although they are mostly benign, they can also exhibit malignant characteristics. In humans, meningiomas are the most common type of IPT, accounting for approximately 40% of all brain cancers, with a median age of diagnosis at 67 years [45,46,47]. Meningiomas represent around 30% of all primary CNS tumors in adults but only 0.4–4.6% in pediatric patients, with a higher proportion observed in females [48].
In humans, the most frequent site of origin for meningiomas is the arachnoid cells in the dura mater. Additional sites include the arachnoid associated with cranial nerves or choroid plexus [49]. Despite being often diagnosed as benign lesions due to their histological features, these tumors can show a clinical behavior similar to malignant neoplasia, with a high recurrence rate and poor prognosis. The 10-year overall survival for non-malignant meningiomas is approximately 81.4%, whereas for malignant forms, it decreases to 57.1% [45,46].
Meningiomas are also diagnosed in canine and feline species displaying a strong relationship with age and breeds and accounting for from 22.3% up to 50% of all brain tumors in dogs [4,11,50] and 58% in cats [51]. In a comparative study between canine and feline meningiomas, Wada et al. (2020) highlighted the development of tumors at median ages of 11.7 years and 14.1 years, respectively. According to the same study, the canine breeds most affected were Miniature Dachshund, Toy Poodle, Beagle, Shetland Sheepdog, Labrador Retriever, Flat-coated Retriever, Shiba Inu, Jack Russel Terrier, Welsh Corgi, and mixed breed [52]. A Japanese study that analyzed data from 186 canine intracranial tumors showed a breed predisposition for meningiomas in Rough Collie, Golden Retriever, Miniature Schnauzer, and Scottish Terrier [53]. Other studies associated a frequent meningioma diagnosis with canine dolichocephalic breeds [7,54,55,56], while a female sex predisposition was not confirmed in more recent studies [11,57]. In cats, Domestic Shorthair seems to be the most predisposed breed to meningioma and no significant difference between sexes exists [57]. Adamo et al. (2003) reported a higher onset of meningiomas in Persian, Domestic Shorthair, and Domestic Longhair cats [6]. In dogs, meningiomas originate in the calvarium-adjacent region, involving the olfactory and frontal regions, cranial cavity, optic chiasm, and suprasellar and parasellar regions, although it has been rarely diagnosed in other regions [58,59]. The sites in which feline meningiomas arise are mainly the tela choroidea of the third ventricle, the supratentorial meninges, and less frequently the cerebellar meninges [51,60,61]. Conversely to what happens in dogs, multiple meningiomas are frequently found in cats (about 17% of all meningioma cases) [51,57,62,63]. Three theories have been proposed to explain the occurrence of multiple meningiomas: multicentric dural foci, metastasis by blood-borne spread, and metastasis via the cerebrospinal fluid. The first hypothesis seems most plausible considering the tumor’s histologically benign nature and the reports showing histological variants in the same patient [64]. However, more research is needed to confirm them.
In about 15% of cases in cats and 20% of cases in dogs, meningiomas are diagnosed in the presence of other neurological disorders [51,57], including depression, stupor, coma, ataxia, lethargy, inappetence, and anorexia in cats [51] and menace response deficits, other cranial nerve deficits, ataxia, and reduced postural reactions in dogs [65].

3. Pathogenesis

The factors contributing to the development of meningiomas in humans and domestic animals need to be further studied. Currently, several hypotheses and mechanisms have been proposed.
Ionizing radiation stands out as the primary environmental risk factor consistently linked to the development of meningiomas. Human exposure to ionizing radiation leads to a 6- to 10-fold incidence increase in this condition [66]. Furthermore, this heightened risk is notably evident among survivors of the atomic bombings in Hiroshima and Nagasaki, where a substantial increase in meningioma cases has been documented [67,68]. In addition to radiation exposure, occupational contact with herbicides and pesticides also appears to elevate the likelihood of developing meningiomas [69]. Moreover, obesity has been identified as a significant positive risk factor for tumor development, likely due to its association with chronic inflammation and the signaling of insulin or insulin-like growth factors [70,71,72,73,74].
Several receptors are over-expressed in meningiomas, particularly somatostatin receptors (SSTRs) and intracellular receptors for sexual steroid hormones, such as androgens, progesterone, and estrogen, suggesting a role for these systems in tumor pathogenesis [67,75,76,77,78]. Among SSTRs, although all the subtypes are expressed in meningiomas [79], SSTR2 expression was associated with a poor prognosis, while SSTR1 expression was associated with reduced incidence of relapses, with less strong evidence. Nevertheless, the in vitro activation of SSTRs demonstrates antiproliferative effects. This evidence led to the development of clinical trials exploring the use of selective agonists, such as octreotide and pasireotide, although, to date, no conclusive results have been reported [80]. Receptors for the steroidal hormones estrogen (ERα) and progesterone (PR) have been reported to be expressed in most meningiomas: ERα presence has been associated with increased proliferation and the development of high-grade tumors, while the high expression of PRs has been correlated with Grade 1 tumors according to the WHO grading system [81]. It is now assumed that PR expression alone represents a favorable prognostic factor in meningiomas, while its loss or the association with ER expression correlates with a worse clinical outcome. However, pharmacovigilance data indicated that the prolonged use of androgen receptor antagonists and/or progesterone receptor agonists (cyproterone acetate, nomegestrol acetate) results in increased meningioma incidence [82,83], making rather complex the prognostic or therapeutic evaluation of these receptors.
Feline and canine meningiomas also express PR [6,84]. Adamo et al. (2003) reported a high proportion of PRs and the absence of ERs in feline meningiomas. Furthermore, a high number of cells with PRs and a significantly lower number of cells with ERs in canine meningiomas were observed. According to this study, the proportion of PR-positive cells in canine benign meningiomas was >80%, while in malignant meningioma only 32% of cells were PR-positive; in cats, the percentages were >80% and 38%, respectively. In dogs, the number of PRs correlated to more aggressive progression (with nuclear pleomorphism, severe necrosis, and histological subtypes), while in cats, such a correlation was not observed [6].
In humans, meningiomas occur in several forms, sometimes associated with other syndromes. Table 2 shows the syndromes associated with high-frequency meningioma and the gene assumed to be involved.
Some of the syndromes listed in Table 2 have also been modeled in animals or have been observed in other species. A murine model was developed to accurately replicate the human NF2-related schwannoma phenotype, including the deficit in hearing and balance [98]. Mice with mutations in PTC, an orthologue of human PTCH1, develop many of the characteristics of Gorlin syndrome and exhibit a high incidence of rhabdomyosarcomas [99]. PtenM3M4 missense knock-in mutant mice present megalencephalic brains and elevated nuclear proteasome activity, also observed in patients with Cowden syndrome-related mutations in PTEN [100]. Multiple endocrine neoplasia type-I-like syndrome was reported in two male Domestic Shorthair cats that developed symmetric alopecia, insulin-resistant diabetes mellitus, and pituitary-dependent hyperadrenocorticism at 12 and 13 years of age [101] and in a crossbred 12-year-old male dog with abdominal enlargement, seborrhoea, and polypnea [102]. Nevertheless, the listed studies did not highlight associations with meningioma.
Few studies reported contemporaneous and unrelated neoplasms in 3–23% of dogs with IPTs, mainly in the thoracic or abdominal cavity [4,11,103].
As reported in a review by Motta et al. (2012), intracranial meningiomas in dogs as well as in cats have been diagnosed with concurrent neural (oligodendroglioma and meningioangiomatosis) [11,51,104,105] or extra-neural disorders (mucopolysaccharidosis type 1 and thymic lymphoma) [106,107]. Moreover, it has been reported that 13.9% of cats and 19% of dogs develop a meningioma in addition to another intracranial neoplasm [11,51,104,108].
In humans, the malignant forms of meningiomas increase the tumor cell invasion processes and the risk of metastasis is higher compared to non-malignant meningiomas [109]. Metastases developments are reported mainly in the lung, pleura, bone, and liver. In domestic animals, meningioma metastases have been described almost uniquely in dogs, and mainly pulmonary metastases were observed [110,111]. A study performed in cats highlighted skull osteolysis. The authors hypothesized that metastases could be responsible for osteolysis [112].

4. Histopathological Classification

The WHO classifies meningiomas in humans and domestic animals with similar criteria.
In humans, the WHO classifies meningiomas into 15 subtypes, reflecting a broad heterogeneity [1]. These are clustered into three groups, differentiated by their histological components. The first group is composed of benign forms classified in different variants: meningothelial, fibrous, and transitional, which are the most common forms. Psammomatous, angiomatous, microcystic, secretory, lymphoplasmacyte-rich, and metaplastic variants are also included in group 1, but their incidence is significantly lower. In group 2, three additional classes are clustered: atypical, choroid, and clear cell meningiomas. The third group encompasses anaplastic, papillary, and rhabdoid meningiomas, often diagnosed as a single histological type, but including different biological and oncological aspects associated with different documented genetic mutations [113,114].
In domestic animals, meningiomas are divided into subtypes, according to the morphological characteristics of the cells [5]. Initially, WHO classified domestic animal meningiomas into two categories: benign and malignant. The benign meningiomas included eight subtypes: meningotheliomatous, fibrous, transitional, psammomatous, angiomatous, papillary, granular cell, and myxoid. Malignant meningioma was classified as anaplastic. However, this classification presents some limitations, and considering the similarities between human and domestic animal meningiomas in pathological, immunological, molecular, and MRI aspects, an improved classification was defined. The benign meningioma classification must be used with caution as histological aspects sometimes lead to considering benign neoplasia that does not match with biological/oncological characteristics [1,115,116,117]. In a study on feline meningiomas, concurrent benign and malignant forms were diagnosed [108].
Dogs show a rare form of meningioma, known as cystic meningioma, characterized by cysts originating through tumoral processes, such as necrosis or release of fluids. The size of the cyst depends on the fluid volume and causes increased intracranial pressure [118].
The observed similarities in pathological, immunological, and MRI aspects between human and canine meningiomas allowed for the classification of canine meningiomas according to three grades of the 2016 WHO human histological grading system [115]. In order to evaluate the possibility of translating the human grading system to canine tumors in terms of accuracy and reproducibility, Belluco et al. (2022) evaluated veterinary neuropathologists’ inter-observer agreement with the application of a human grading system to canine meningioma [115]. The reproducibility of each histologic criterion was evaluated to identify a possible disagreement. The authors proposed amendments to increase reproducibility in canine meningioma [115]. In their study, Belluco et al. (2022) proposed a criterion for canine meningioma classification (Table 3) based on mitotic grade in a specific area (2.37 mm2) of tumor tissue [115].
Commonly, canine meningiomas present characteristics similar to the group 1 WHO classification of human meningiomas. The group 1 meningioma subtypes include meningotheliomatous, fibrous (fibroblastic), transitional (mixed), psammomatous, angiomatous (angioblastic), papillary, granular cell, myxoid, and anaplastic (malignant) [119]. Canine meningiomas usually display transitional, meningothelial, microcystic, and psammonas histological aspects [20]. In some cases, canine meningiomas present chondroid, osseous, myxoid, and xanthomatous-like areas in meningotheliomatous and transitional subtypes. Another aspect highlighted in meningotheliomatous and transitional subtypes was polymorphic infiltration with or without tumor cells necrosis area. Feline meningiomas are commonly classified into the transitional and fibroblastic subtypes. However, feline meningioma histology is cytologically bland and uniform; for this reason, it is very difficult to adapt to the human WHO guidelines [20]. The Comparative Brain Tumors Consortium (CBTC) tried to establish the translational aspects of canine brain tumors as a model for their human counterparts [120]. The work of CBTC provided the foundation for a histologic atlas of canine glioma that included astrocytoma, oligodendroglioma, and undefined glioma [121]. The samples were collected from several institutions and analyzed with immunohistochemistry to evaluate the expression of specific markers [121]. Other criteria studied were infiltrations, necrosis, mitosis, and vascularization. A grading classification was built reporting these criteria [121]. The grading was compared with human tumor counterparts. We suggest repeating the same studies for meningioma. Belluco et al. (2022) evaluated the reproducibility of criteria used in the human meningioma grading when applied to canine meningioma [115].

5. Diagnosis

Meningiomas are diagnosed in both humans and domestic animals through a combination of clinical evaluations, imaging techniques, and histopathological analysis. The primary diagnostic tools employed to assess the presence of meningiomas are magnetic resonance imaging (MRI), computed tomography (CT), and positron emission tomography (PET) [122,123].
Specific conditions such as edema, cyst formation, change in vascularity, and necrosis are well detected by MRI [124], and novel imaging techniques are improving the diagnosis of brain lesions. The use of MRI contrast agents permits the distinction of the tumor from the normal tissues. Furthermore, neurosurgeons can use MRI to minimize the size of craniotomy, maximize tumor removal, and minimize damage to the surrounding brain. Moreover, diffusion-weighted imaging, magnetic resonance spectroscopy, and dynamic contrast-enhanced MRI are entering clinical practice [125,126,127,128,129], whereas cerebrospinal fluid (CSF) analysis can provide clinical parameters, such as altered protein content and leukocyte count, useful for diagnosing pathology [130]. Motta et al. (2012) reported the main MRI characteristics of dog and cat meningiomas [57]. Table 4 summarizes meningioma MRI features observed in canine, feline, and human meningiomas [128,131,132,133].
Immunohistochemistry (IHC) plays a significant role in the diagnosis of meningiomas. IHC can be performed in human, canine, and feline meningiomas, using similar markers (Table 5). Saito et al. (2021) described about 39 cases of feline meningiomas of various grades in which specific markers were observed to classify the tumors [134]. The study analyzed markers such as Cytokeratin, Vimentin, E-cadherin, β-catenin, N-cadherin, and Ki-67. In feline meningiomas, Cytokeratin was only recognized in particular histological phenotypes (fibrous and transitional types) and showed high immunoreactivity in some studies involving dogs and humans [134]. Vimentin tested positive in some cases of feline meningiomas. According to Saito et al., E-cadherin remained stable in all subtypes of meningiomas, making it a reliable marker for feline meningiomas. In some studies, N-cadherin was detected in many human and canine meningiomas [135,136]. β-catenin was analyzed in both canine and feline meningiomas. In feline meningiomas, while β-catenin was detected in over half of the analyzed meningiomas, its translocation to the nucleus (indicating the active form) was observed only in specific tumor types; in particular, translocation was not evaluated in atypical and anaplastic subtypes. In dogs, β-catenin was detected at the nuclear level, mainly in anaplastic meningiomas [137]. Ki-67 is an important proliferation index associated with many tumors, as well as in meningioma [138]. In the human literature, KI-67 expression correlates with tumor aggressiveness in meningiomas [139,140,141,142]. Matiasek et al. (2009) evaluated the role of KI-67 in dog meningiomas [143] considering about 70 canine meningiomas. The samples were analyzed via immunohistochemistry and 64 cases tested positive for KI-67 [143]. Some studies reported KI-67 be predictive for the survival of dogs with non-nervous tissue tumors [144,145,146,147]. Matiasek et al. (2009) did not find the same prediction; however, this was a retrospective study performed with a small sample size [143]. Janssen et al. (2023) studied the expression of KI-67 in 68 canine meningiomas to correlate KI-67 expression with the WHO grading of meningioma [148]. Many samples positive for KI-67 were classified as WHO grade I and the authors hypothesized a possible role of KI-67 in meningioma development, in particular during the early stage of these tumors [148]. Saito et al. (2021) did not correlate Ki-67 with specific subtypes of feline meningiomas [134].
Therefore, the above-mentioned observations highlight common characteristics in meningioma classification and similar markers in IHC among the three species.

6. Molecular Characteristics

Meningiomas shows numerous molecular alterations in both human and domestic animal tumors, and some similarities and differences have been reported.
In humans, many genetic alterations have been associated with meningioma development. Deletions of NF2 seem to be a common characteristic condition of meningiomas [150]. Indeed, alterations in NF2 have been recognized as meningioma’s driver mutation, being present in about 50% of sporadic meningiomas [150]. Neurofibromatosis type II determines the inactivation of merlin gene NF2, a tumor suppressor involved in cytoskeleton dynamics, tumor-associated increased motility, and the regulation of cell proliferation [151]. Another pivotal player in meningioma’s development is EPB41L3, also known as DAL-1 or 4.1B, a tumor-suppressor gene that encodes erythrocyte membrane protein-band 4.1-like 3, involved in cell–cell interaction and having an important role in the control of motility [152,153,154]. Decreased EPB41L3 expression is observed in about 70% of meningiomas.
Alterations in chromosome 1 were frequently found in meningiomas [113,155] and include mutations in TP73, CDKN2C, RAD54, EPB41, GADD45A, and ALPL [156,157,158,159].
Loss of function in chromosome 14 is commonly found in high-grade meningiomas and includes inactivation in NDRG family protein 2 and maternally expressed gene 3 (MEG3), which were associated with a poor prognosis [113,160,161]
Loss of function in chromosome 9 due to the deletions of the cyclin-dependent kinase inhibitors 2A (CDKN2A) and 2B (CDKN2B) has been associated with the progression from Grade 2 to anaplastic meningioma (Grade 3) [113,162].
Whole-genome sequencing approaches have identified mutations occurring in TRAF7, AKT1, KLF4, PIK3CA, and SMO, although these mutations seem to be mutually exclusive with those associated with NRF-2 [163,164,165].
TRAF7 is mutated in about 20% of all meningiomas and is frequently associated with KLF4, AKT1, and PIK3CA mutations [113,164].
AKT1 mutations are found in about 12% of Grade 1 meningiomas and, although less frequently, in Grade 2 and 3. Interestingly, about 50% of all AKT1-mutated meningiomas also show alterations in TRAF7 [113,164,166]. PIK3CA mutations occur in about 7% of all meningiomas; they are mutually exclusive with NF2 and AKT1 and often associated with TRAF7 mutations [163]. Other somatic mutations associated with meningiomas include BAP1, SMARCB1, SMARCE1, BRAF-V600E, NOTCH2, CHEK2, PTEN, CDKN2A, CDKN2B, and DMD [92,167,168,169,170,171,172,173,174].
In humans, telomerase alterations are frequently associated with an increased risk of meningioma development [175]. Telomerase mutations occur in all grades of meningiomas, the frequency being associated with the tumor grade [176,177,178]. The main somatic mutations occur in the promoter region at two specific hotspots, C228T and C250T, resulting in up-regulation of the protein and the increased survival of cancer cells [177]. Slavik et al. (2022) performed RNA-seq in 64 meningiomas to identify novel prognostic markers for these tumors. This study found the dysregulation of many transcripts involved in the WNT signaling pathway, highlighting the importance of the WNT pathway in meningioma development, as already reported for dogs [179].
Initially, meningiomas were studied in athymic nude mouse models, after PDX meningioma cell inoculation. The study by Rath et al. (2011) demonstrated that meningioma PDX retained the characteristics of the original tumors and that a stabilized cell line of meningiomas exhibited similar features to the tumors of origin [180]. A limitation of the xenograft models is the absence of the tumor microenvironment, which restricts the study of the aspects of meningiomas, including drug resistance. GEM overcomes this limit by having an intact tumor microenvironment. Peyre et al. (2018) utilized a model of transgenic mice in which alterations in NF-2 and CDKN2AB were induced [181]. Nevertheless, as above reported, studying meningiomas in mice models does not yield the same results obtained with spontaneous animal models and, in general, the existence of spontaneous models is a fundamental tool for studying cancer and other pathologies. Indeed, they mimic key aspects in the investigation of novel therapeutical approaches that are absent in non-spontaneous models, i.e., the role of the microenvironment and of the immune system [182].
Few available studies explored the genetic factors involved in the formation of canine and feline meningiomas. However, canine and feline models are reported to be strong translational models for CNS diseases, like stroke, epilepsy, movement disorders, lysosomal storage diseases, Alzheimer’s and cognitive disorders, and neuro-oncology [29]. Differential expression regulation in orthologue genes of Homo sapiens in canine and feline meningiomas has been reported (Table 6). Partridge et al. (2020) highlighted the advantages of using canine and feline models to study meningiomas since the larger size of the animals allows for surgical handling of the CNS; moreover, the presence of an intact immune system and the molecular, histological, and neuroimaging characteristics make them comparable to human neoplasia [29].
Besides genetics, it is important to consider the role of epigenetics in meningioma development. Many studies aimed at the identification of specific methylation profiles that could explain the mechanisms of oncogenesis, and some have demonstrated an epigenetic role in human meningioma onset [113,190,191]. These epigenetics mechanisms include DNA methylation, defective chromatin remodeling, alterations in microRNAs, and the hypermethylation of TIMP3. Alterations in the methylation of the TIMP3 promoter determine inhibition of the metalloproteinases and can be associated with a poor prognosis [113,192,193]. TP73 inactivation by hypermethylation has been investigated as a possible risk factor for malignant meningiomas [192,194]. Interestingly, experimental evidence suggests that methylation profiles might be suitable to predict the clinical outcome of patients. Indeed, some studies showed that an altered methylation profile is associated with a worse prognosis [178].
Few studies analyzed the molecular alterations driving canine and feline meningioma pathogenesis [189]. Different genes resulted in down-regulation, for instance MYOC, ALP, PRKD1, FHL5, TIE1, MCAM and PECAM1. Courtay-Cahen et al. (2008) highlighted the following alterations in canine meningiomas: histone acetyltransferase p300, PDGF-β, thioredoxin reductase 1, mutS homolog 2 and 6, Dal-1, Clusterin-like 1 (Retinal), B-cell lymphoma, T-cell differentiation protein, BCL-2-like 11, IL-1α, and IL-1β [195]. An RNA-seq transcriptome analysis performed in canine meningioma determined a series of over-expressed genes, for example, FOSB, KLF5, WNT, GEM, EGR1, and DACT2 [189]. The entire altered gene list is reported in Table 6 and includes also genes altered in both humans and dogs: FOS, KLF5, BMPR1B, NR4A1, WNT5A, PDPN, MYRF, MYC, ALP, COL14A1, THSB1, MMRN2, FHL5, SFRP1, MCAM, DAAM2, AQP1, KITLG, PECAM1, OLFML3, ADCY2, NOTCH3, FBLN2, APOD, PTCH2, IGFBP6, GAS6, COL16A1, PIK3R1, and FMN2 [196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222]. A case-report study speculated about the relationship between mucopolysaccharidosis I (caused by alpha-L-iduronidase deletions) and the onset of meningiomas in cats [106].
The role of PR receptors in the pathogenesis of human and animal meningiomas has been already mentioned [6,84,223,224,225,226,227,228,229]. Other studies reported the important role of matrix metalloproteinases MMP-2 and MMP-9 in canine and human meningiomas, where they are involved in extracellular matrix degradation, required for tumor progression and recurrence [230,231,232,233]. Mandara et al. (2007) evaluated the expression of MMP-2 and MMP-9 in canine meningiomas, establishing correlations with TIMP-1 and TIMP-2 expression [234]. The authors found that TIMP-1 levels were elevated in Grade 1 and Grade 2 meningiomas, but not in Grade 3 cases and hypothesized different pathways in which TIMP-1 can be involved in the progression of meningiomas [174]. Cyclooxygenase-2 (COX-2) is reported to have an impact on canine meningiomas [235] and is over-expressed in some feline meningiomas [236]. However, only in humans, COX-2 seems to be correlated with meningioma grade and local invasion [29] and is a marker used to classify the tumors [237]. Vascular Endothelial Growth Factor (VEGF) has been proposed as a prognostic marker in canine, where it seems to be inversely correlated with survival time [29], while in human meningiomas, it is a tumor recurrence marker [238]. The role of E-cadherin, N-cadherin, β-catenin and doublecortin (DCX) have been evaluated in both canine and human meningiomas. In canine tumors, N-cadherin, β-catenin, and DCX seem to have a positive correlation with invasion and anaplastic subtypes of meningiomas, instead in human meningiomas, it has not demonstrated a correlation and this role is still debated [29,136,239,240]. Alterations of NF-2 in chromosome 22 do not seem to play an important role in the onset of canine meningiomas [183], as opposed to human meningiomas, where they have been reported to predispose to the development of the tumors [239].
Glucose transporter (Glut-1) is expressed at high levels in malignant meningiomas both in dogs and humans [29]. Boozer et al. (2012) evaluated the immunological infiltrate in canine meningiomas [241], reporting a prevalence of CD18+ microglia and macrophages surrounding and infiltrating the tumors; CD11d+ cells were also present. Lymphocyte infiltrate included mainly CD3+ T-cells and a sparse number of CD79a+ B-cells. In human meningiomas, similar T-cell and B-cell infiltrate have been found, but in both cases, the biological role of this infiltrate was not determined [242,243,244,245]. The proteins altered in human and dog meningiomas are reported in Table 7.

7. Role of Mitochondria in Meningiomas

The human mitochondrial DNA (mtDNA) genome counts 16,569 bp and is characterized by a high copy number per cell. This genome is constituted of 37 genes, including 13 genes encoding proteins involved in the oxidative phosphorylation system, 2 rRNA, and 22 tRNAs. Noncoding sequences are present in mtDNA only in a small segment called D-loop, which is characterized by regulatory elements for replication and transcription of mtDNA. Point mutations, insertions, deletions, and nucleotide substitution in D-loop have been associated to the development of some tumors [246,247,248] including brain neoplasia [249,250,251]. MtDNA is especially prone to mutations because mitochondria are the major site of the reactive oxygen species (ROS) production; therefore, mtDNA is constantly exposed to their mutagenic activity. In CNS tumors, including meningiomas, a considerable number of mutations have been found in mtDNA [251]. Moreover, alterations in levels of complex II and IV of the respiratory chain involved in oxidative phosphorylation were highlighted in meningiomas. These anomalies seem to be linked to HIF-1α deregulation [252].
The role of p53 in the regulation of mitochondrial respiration is well-known [253]. Being TP53 mutated with high frequency in meningiomas, its involvement in mitochondrial dysfunction could be a key event [253,254]. In many cancer types, a frequent mtDNA molecular alteration is the deletion between nucleotides 8470 and 13447, called mtDNA4977. This mutation can alter the oxidative phosphorylation and biogenesis processes. A case study highlighted the frequent presence of this mutation in patients with meningiomas [250]. There is currently no available research regarding molecular modifications of mtDNA in canine and feline meningioma, but several studies have been performed in other tumor types and disorders, also of the CNS [255,256,257,258].

8. Role of miRNA in Meningiomas

MicroRNAs (miRNAs) are short non-coding RNA of about 22 nucleotides, distributed in the genome to modulate protein expression, by inducing the degradation/instability of the target mRNA or by inhibiting its translation. MiRNAs can interact with the 3′UTR—sequence or directly with mRNA causing protein translation arrest or mRNA degradation [259,260,261,262]. MiRNA deregulation has been demonstrated to have a role in tumor cell growth, proliferation, and migration in many cancers, including meningiomas. In the last 20 years, many studies have been focused on the identification of miRNAs playing a function in cancer [259,260,262,263,264,265,266,267]. Nowadays, miRNAs have been proposed as molecular biomarkers for diagnosis and prognosis in meningiomas. Considering type 1 and type 2 human meningiomas, an important role of miR-21, miR-34a, miR-143, miR-193b, miR-218, and miR-451 deregulation compared to normal tissues have been highlighted [268]. Kopkova et al. (2019) analyzed CSF samples from meningioma patients and found high expression of miR-196a-5p, miR-140-5p, miR-10a-5p in neoplastic tissues [269]. Zhi et al. (2016) studied a cohort of Chinese patients to identify circulating miRNAs as novel possible biomarkers for meningiomas, identifying deregulation of miR-106a-5p, miR-219-5p, miR-375, miR-409-3p miR-197, and miR-224 [270]. Slavik et al. (2020) demonstrated that miR-15a, miR-146a-5p, and miR-331-3p expression in meningiomas was correlated to the activity of important intracellular pathways such as NF-kB, VEGF, STAT3, PTEN/AKT/mTOR [271].
The main miRNAs involved in meningioma biology are summarized in Table 8.
The identification of miRNAs biomarkers in meningiomas could represent an important advancement also in the veterinary field. The diagnostic or prognostic role of miRNAs has been investigated in different animal tumors, as well as in meningioma. Foiani et al. (2021) studied a panel of 14 miRNAs, with high sequence identity between human and canine, in 41 canine meningioma biopsy samples [277]. Five miRNAs resulted down-regulated (miR-96, miR-145, miR-200a, miR-29c and miR-335), while three were up-regulated (miR-136, miR-146a and miR-155) when compared to normal arachnoid tissue. Some of the miRNAs listed in Table 8 play a role in canine and feline neoplasms, also. In particular, the role of miRNAs was highlighted in mammary gland tumors, melanomas, osteosarcomas, urothelial carcinomas, leukemia, hemangiosarcomas [278,279,280,281]. For a detailed review see Varvil and dos Santos (2023) [282]. The miRNAs listed in Table 8, which have been investigated in animal neoplasms are reported in Table 9. MiRNAs’ involvement in IPTs is a little explored field in veterinary medicine and in the future could be important to study their role also in these types of tumors.

9. Therapeutic Approaches

Therapeutic strategies to treat meningiomas in humans and domestic animals depend on the grade of advancement of the tumors.
The basic therapeutic strategy for meningiomas is surgery to remove the tumor and surrounding tissues to make sure that all the cancer cells are eliminated. In some cases, surgery is sufficient to eradicate the disease, but in others further treatment is needed. Surgery is classified according to the Simpson grading, from 1 to 5 [303]. This classification helps to predict an eventual recurrence at 10 years of overall free survival. Grade 1 is determined by complete tumor removal, including the resection of the underlying bone and associated dura. The symptomatic recurrence at 10 years is about 9% [303]. Grade 2 is characterized by complete removal associated with coagulation of dural attachment. Symptomatic recurrence at 10 years is estimated at around 19% [303]. Grade 3 is associated with complete tumor removal without resection of dura or coagulation. In these cases, symptomatic recurrence at 10 years is about 29% [303]. Grade 4 is associated with subtotal resection; symptomatic recurrence exceeds 40% [303]. Grade 5 is characterized by simple decompression with or without biopsy. These cases have a 100% of recurrence risk at 10 years [303,304,305,306].
Radiation therapy is used as a first-line option for meningiomas characterized by the involvement of nerve sheath or the cavernous sinus [307]. Stereotactic radiotherapy provides significant benefits in patients, increasing 5-year survival [308]. Brachytherapy and radionuclide therapy are used in the treatment of atypical and malignant meningiomas, providing in few studies advantages in the survival [308,309,310].
Although several studies showed limited efficacy, systemic therapies have been proposed as alternative approaches to treat meningiomas [311,312,313,314,315]. In this context, the availability of novel pharmacological agents is a priority. Drugs used for systemic therapy include: temozolomide, bevacizumab, irinotecan, everolimus, sunitinib, mifepristone, imatinib, doxorubicin, and vincristine. An important randomized, multicentric study (EORTC-1320-BTG) is ongoing to evaluate the efficacy in meningiomas of trabectedin, an inhibitor of active transcription acting on RNA polymerase II [316].
Since molecular alterations of NF2 are frequent in meningiomas, molecular pathways NF2-linked have been studied as possible targets for pharmacotherapy [150,317,318,319]. In particular, the role of merlin, whose function is related to the control of the mammalian target of the rapamycin (mTOR) pathway, has been considered, and in mouse models specific mTOR inhibitors (temsirolimus and everolimus) suppress meningioma growth [312]. Other studies investigated the association of the inhibition of mTOR with the activity of other molecular targets. In this context, AZD2014, a dual-inhibitor of mTOR1 and 2, is currently in 2 phase-II trials which include the treatment of different forms of meningiomas [316].
Anti-angiogenic molecules, such as bevacizumab, have not demonstrated high efficacy in the treatment of meningiomas [320,321].
Massive parallel sequencing technologies increased the knowledge of meningioma genomic landscape, highlighted the heterogeneity of these tumors and opening novel scenarios of possible target therapy in the future [166,316]. Researchers identified in the genome of a set of Grade 1 meningiomas, somatic copy-number alterations (SCNAs), rearrangements, mutations, and insertions and/or deletions in a total of 645 known cancer-associated genes. Several clinical trials are now testing the validity of compounds that target epigenetic alterations. In this field, a particularly promising compound is KDOAM 25, a KDM5 inhibitor [322,323,324]. KDM5 comprises a histone demethylase family that removes tri- and di-methylations of lysine form on histone H3. In a study conducted on myeloma KDOAM25, altered the methylation profile of the tumor, and increased cell-cycle arrest in G1 phase and the apoptotic processes [322]. Olar et al. (2017) studied the methylation profile of the patients to create predictions on the recurrence of the meningiomas. These criteria could add up to histological and clinical predictors [323].
In NF2 wild-type tumors, experimental drugs showed the ability to inhibit smoothened (SMO), a member of the hedgehog pathway and AKT1 [163,325,326]. Boetto et al. (2017) showed that SMO-mutated meningiomas had a higher recurrence rate with respect to AKT1 mutated-meningiomas and that, overall, SMO-mutated meningiomas had a poor prognosis as compared to AKT1-mutated ones [327]. An important phase II clinical trial is currently testing the efficacy of SMO, AKT1, and focal adhesion kinase (FAK) inhibitors in patients with malignant meningiomas [328].
FAK has synthetic lethality with NF-2 loss; therefore, Brastianos et al. (2023) tested GSK2256098 (FAK inhibitor) in NF-2 meningiomas, observing an increase in progression- free survival (PFS) in the presence of a good safety of the drug [328].
Considering the high levels of PD-L1 expressing cells in some meningiomas, in which correlated to poor prognosis, immunotherapy has been considered as treatment also for this neoplasia [329]. Grade 2 and Grade 3 meningiomas seem to have high levels of PDL-1, but the correlation of PDL-1 expression with treatment response is still not clear [330]. Garzon-Muvdi’s work illustrated five-clinical trials, conducted in the United States, starting in 2020 to evaluate the response of meningioma patients treated with the immune checkpoint inhibitor antibodies nivolumab, ipilimumab, pembrolizumab, and avelumab [330].
The role of anti-progesterone drugs in therapy is under investigation in human meningiomas [331,332,333]. The use of the progesterone antagonist RU486 has been evaluated in humans to treat meningiomas [332]. The results demonstrated a reduction in tumor size, which was evaluated through MRI analysis. Andersen et al. (2013) showed that the use of hormone therapy did not increase the risk of developing meningiomas, as speculated in other studies [333].
In dogs, treatment of choice for meningiomas is surgery, and the median overall survival time after excision is about 7 months [334]. Radiation therapy is another important approach to treat dogs. In some cases, the therapeutic protocols provide an association between radiotherapy and corticosteroids [184,334,335]. Radiotherapy, usually fractionated in 5 weeks [184,336] increases survival time with respect to dogs treated with surgery only, while the association between surgery and consolidation radiotherapy increases the survival of dogs with intracranial meningiomas of about 8 months [334]. Adjuvant radiation therapy is typically reserved for atypical or anaplastic meningiomas in a similar approach as in human patients [337].
In domestic animals, chemotherapy has been used with non-effective results. In particular, a study reported the comparative use of carmustine and lomustine treatment in association with corticosteroids and anticonvulsants [338]. Some case reports showed a low efficacy of alkylating agents in increasing dog survival as compared to corticosteroids and anticonvulsants alone [338].
Other studies reported the possibility of treating meningiomas with hydroxyurea [339,340], an antineoplastic drug frequently used in veterinary practice, for example for the treatment of leukemias or mast-cell tumors. Marconato et al. (2007) reported two cases of dog meningiomas treated with hydroxyurea, in which a survival increase of 4 months was observed [339]. Moreover, hydroxyurea treatment demonstrated efficacy in meningioma cell culture, showing a reduction in proliferation rate and cell-cycle arrest. However, in a veterinary case, the use of hydroxyurea was associated to dermatological side effects [339]. In another case report, hydroxyurea was used in combination with phenobarbital and prednisolone, but this drug association did not produce noteworthy results [341]. The combination between hydroxyurea and imatinib-mesylate showed effectiveness in dogs [342,343,344]. Imatinib-mesylate use in meningiomas derives from its inhibition of PDGFR, a tyrosine kinase receptor which plays a role in meningiomas progression. In humans, the association between these two drugs produced a prolonged survival as compared to imatinib alone and was well tolerated by the patients [343,344]. The same treatment was evaluated also in veterinary practice on Belgian Malinois dogs, causing a decrease in tumor size; however, the dogs died due to anesthesia issues, and results about survival and efficacy of the treatment are not available [342].
Gene therapy for meningiomas has been proposed in one study, but to date, the road to applying it in clinics appears still long [345].
The evidence that canine meningiomas express high levels of PR prompted to study the possibility of treating animals with antiprogestinic drugs [6].
Another tested approach was the possibility of adopting vaccinations that can increase the activation of the immune response toward meningiomas [346]. The vaccination involves the administration of peptides containing defined T-cell epitopes to stimulate immunity response. In this study, although it was evaluated the possibility of enhancing T-cell response to the tumor, a major B-cell activation was highlighted instead [346].
The gold-standard therapeutical approach for feline meningiomas is also represented by surgery [347,348,349] and the median survival after surgery is 26 months. It is worth highlighting that about 80% of the cats after surgical removal of the tumor, did not develop recurrence [63,347]. In a study of 17 cases, the treatment with radiotherapy after the removal of the tumor nodule has been proposed, but no evidence of better efficacy of this approach has been reported [347]. In general, cat meningiomas are treated with the same panel of drugs used in dogs. Yun et al. (2021) highlighted an advantage of the combination between hydroxyurea and prednisolone [350]. This additive effect allowed the reduction in the tumor size. Palliative therapies remain important for seizure control, edema, and intracranial pressure, using anti-epileptic drugs (phenobarbital and potassium bromide) and glucocorticoids (prednisone and dexamethasone). The effects of palliative therapies are studied to stabilize a therapeutic regimen [184,351,352].

10. Discussion

IPTs represent a heterogeneous group of malignancies that occur in both humans and domestic animals, such as dogs and cats [57,353]. These tumors develop in both malignant and non-malignant forms leading to a high mortality rate, mainly due to the lack of effective pharmacological approaches. Meningiomas are the most frequent forms of IPTs in humans, dogs, and cats. Although these tumors mainly occur in benign forms, several related syndromes, such as dementia, depression, stupor, coma, ataxia, lethargy, inappetence, and anorexia result in a significant reduction in the quality of life [16,354,355]. However, meningiomas can also assume malignant characteristics determining a poor prognosis. A recent study characterized a population of tumor stem-like cells regulated by chemokine receptors in a large series of human meningiomas, which were proposed as determinants of the development of the more malignant clinical course [24].
A number of inherited pathologies have been identified in dogs and cats, and many of these share similarities with human counterparts [30,31]. Various aspects, including phenotypes, genotypes, pathophysiology, progression and factors influencing disease onset exhibit similarities in human, canine, and feline diseases. These analogies enhance the potential for discovering novel therapeutical approaches [29,356,357]. Domestic animal models serve as valuable translational models for humans, as they share similar lifestyles, food intake, and environmental conditions. Furthermore, advancements in genomic, diagnostic, and molecular biology techniques have contributed to the enhancement of veterinary practice [32], facilitating comparisons with analogous disciplines commonly employed in the medical field.
In humans, meningioma diagnosis primarily relies on MRI techniques, with specific criteria established to classify the tumor’s developmental grade [358,359,360,361]. Surgery remains the optimal approach for treating these tumors across all three considered species, but it is not always curative. Clinical pharmacological strategies predominantly involve chemotherapy and targeted therapy [311,362,363,364,365,366]. Chemotherapy’s efficacy is limited and often accompanied by significant side effects, while targeted therapy presents a promising but relatively novel option, with numerous ongoing clinical studies. Despite extensive exploration of these aspects in human medicine, there is still significant potential for research in animal models [315,367,368,369,370]. A particularly intriguing area of investigation is the role of steroid hormones in meningioma pathogenesis. The high expression of ERα and PR has been linked to meningioma proliferation in both humans and domestic animals. Consequently, hormone therapy represents a crucial field for exploration, leveraging the observed similarities among species [229,371,372,373,374]. Additional research is still needed to elucidate the role of sex hormones, especially progesterone, and their receptors in both tumorigenesis and the progression of meningioma [375]. Saito et al. (2021) explored this aspect, but no correlation was found between castration or sterilization and meningiomas. However, it is worth noting a potential study bias due to the limited number of females considered [134]. Rzechorzek et al. (2019) suggested a potential protective role of estrogens in the onset of brain tumors and brain aging based on a study involving 281 dogs. The results confirmed the beneficial effects of estrogen on the brain [376]. Understanding this role could enable the utilization of estrogens in primary preventive, secondary preventive, and therapeutic strategies for meningiomas. The role of immunotherapy remains controversial, as the evaluation of PD-L1 levels in these tumors does not seem to correlate with a response to checkpoint inhibitors [329,377,378]. In this field, a successful strategy might be determined by the use of vaccination aimed at stimulating the activation of T-cells and B-cells against the tumor [346]. While gene therapy is a well-studied strategy studied for many tumors in clinical settings, it has not been extensively explored for meningiomas. Nonetheless, some works have speculated about its potential implementation in treating meningiomas in dogs and cat [379,380].
The genetics and molecular characteristics of human meningiomas are well understood, [113,181,317,381,382,383,384,385,386]. Gene expression profiles and molecular alterations have been more comprehensively studied in humans compared to dogs or cats. In this review, we report the primary molecular characteristics in the three species that reflect the current state of art in meningiomas. While not all the molecular pathways studied in humans have been explored in domestic animal meningiomas, preliminary studies have identified similarities in the molecular pathways underlying pathogenesis and in gene expression or alterations. These findings, combined with the observed similarities in genome sequences and molecular pathways among humans, dogs, and cats, as demonstrated in other studies and neoplasms, give hope for the identification of shared molecular alterations to focus on.
The role of mtDNA and miRNA in the pathogenesis and biological response of human meningiomas has recently been investigated. To date, alterations in mtDNA have not been assessed in canine meningioma. However, this aspect has been explored in other canine tumors and SNC disorders; therefore, future studies on meningioma in canines could provide novel insights, and knowledge developed in humans could serve as a promising starting point for veterinary research. The role of miRNA as biomarkers has been studied in various canine tumors, including meningiomas [248], but the field is more extensively explored in human tumors.
Another aspect to be explored in the veterinary field is the challenge in classifying and diagnosing animal meningiomas [387,388,389]. The WHO classification of meningiomas acknowledges similarities across the three species but also differences. The difficulty in identifying analogies between human and domestic animal subtypes, along with histological variations, could be attributed to a higher number of studies in humans compared to animals, or to less comprehensive histological investigations on meningiomas in the veterinary field. In this context, a future approach similar to that adopted by the Comparative Brain Tumor Consortium (CBTC) for the classification of canine glioma is desirable. The CBTC involved a panel of expert physician and veterinary neuropathologists to assess the features of canine glioma, determine the best classification from both human and veterinary perspectives, and establish a common ground at the most basic level of tumor diagnosis [120].
While surgical approaches and radiotherapy remain the primary treatments of choice, understanding the molecular alterations present in meningiomas across the three species could be valuable in assessing the potential use of specific targeted therapies, in addition to traditional approaches. Adopting a “One Health” approach and amalgamating aspects incompletely studied in one species compared to in others may contribute to completing the puzzle represented by issues related to diagnosis, classification, therapy, and the validation of prognostic markers. Such an integrated approach holds mutual benefit for both human and veterinary medicine in the fight against this pathology [14,390].

11. Conclusions

Meningiomas represent the most common non-malignant tumors of the central nervous system in humans, dogs, and cats, displaying comparable frequencies and molecular features. Despite their prevalence and the significant clinical challenges associated with meningioma in humans, dogs, and cats, alternative therapies have not been fully explored. Consequently, surgery remains the treatment of choice, albeit with potential disadvantages and adverse effects.
Here, we reported an overview of the current knowledge regarding meningiomas in humans, dogs, and cats with the goal of identifying potential similarities or discrepancies. The objective was to highlight areas that could benefit from a knowledge exchange facilitated by a “One Health” approach.
Specific molecular pathways have been highlighted, demonstrating their role in meningiomas across different species. These pathways, including COX-2, PR, ER, MMP-2, MMP-9, GLUT-1, VEGF, FOS, KLF5, BMPR1B, NR4A1, WNT5A, PDPN, MYRF, MYC, ALP, COL14A1, THSB1, MMRN2, FHL5, SFRP1, MCAM, DAAM2, AQP1, KITLG, PECAM1, OLFML3, ADCY2, NOTCH3, FBLN2, APOD, PTCH2, IGFBP6, GAS6, COL16A1, PIK3R1, and FMN2, could be assessed in the context of novel targeted therapies. Simultaneously, certain miRNAs (miR-200a, miR-335, miR-146a, miR-29c, miR-155, miR-96) could be further investigated as potential prognostic markers or therapeutic targets.

Author Contributions

Conceptualization, M.T.; writing—original draft preparation, M.T.; writing—review and editing, T.F., P.M., G.C.V. and A.P.; visualization, M.T.; supervision, P.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The Associazione Italiana per la Lotta al Neuroblastoma/Fondazione Neuroblastoma (Genova, Italy) is gratefully acknowledged for their support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef] [PubMed]
  2. Ostrom, Q.T.; Francis, S.S.; Barnholtz-Sloan, J.S. Epidemiology of Brain and Other CNS Tumors. Curr. Neurol. Neurosci. Rep. 2021, 21, 68. [Google Scholar] [CrossRef] [PubMed]
  3. Ostrom, Q.T.; Cioffi, G.; Gittleman, H.; Patil, N.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2012–2016. Neuro Oncol. 2019, 21, v1–v100. [Google Scholar] [CrossRef] [PubMed]
  4. Miller, A.D.; Miller, C.R.; Rossmeisl, J.H. Canine Primary Intracranial Cancer: A Clinicopathologic and Comparative Review of Glioma, Meningioma, and Choroid Plexus Tumors. Front. Oncol. 2019, 9, 1151. [Google Scholar] [CrossRef] [PubMed]
  5. Pintus, D.; Marruchella, G.; Masia, M.; Maestrale, C.; Cancedda, M.G.; Contu, C.; Macciocu, S.; Ligios, C. Glioblastoma with oligodendroglioma component in a ewe. J. Vet. Diagn. Investig. 2016, 28, 449–454. [Google Scholar] [CrossRef] [PubMed]
  6. Adamo, P.F.; Cantile, C.; Steinberg, H. Evaluation of progesterone and estrogen receptor expression in 15 meningiomas of dogs and cats. Am. J. Vet. Res. 2003, 64, 1310–1318. [Google Scholar] [CrossRef]
  7. Song, R.B.; Vite, C.H.; Bradley, C.W.; Cross, J.R. Postmortem evaluation of 435 cases of intracranial neoplasia in dogs and relationship of neoplasm with breed, age, and body weight. J. Vet. Intern. Med. 2013, 27, 1143–1152. [Google Scholar] [CrossRef]
  8. Amphimaque, B.; Durand, A.; Oevermann, A.; Vidondo, B.; Schweizer, D. Grading of oligodendroglioma in dogs based on magnetic resonance imaging. J. Vet. Intern. Med. 2022, 36, 2104–2112. [Google Scholar] [CrossRef]
  9. Dickinson, P.J. Advances in diagnostic and treatment modalities for intracranial tumors. J. Vet. Intern. Med. 2014, 28, 1165–1185. [Google Scholar] [CrossRef]
  10. Priester, W.A.; Mantel, N. Occurrence of tumors in domestic animals. Data from 12 United States and Canadian colleges of veterinary medicine. J. Natl. Cancer Inst. 1971, 47, 1333–1344. [Google Scholar]
  11. Snyder, J.M.; Shofer, F.S.; Van Winkle, T.J.; Massicotte, C. Canine intracranial primary neoplasia: 173 cases (1986–2003). J. Vet. Intern. Med. 2006, 20, 669–675. [Google Scholar] [CrossRef] [PubMed]
  12. Boudreau, C.E.; York, D.; Higgins, R.J.; LeCouteur, R.A.; Dickinson, P.J. Molecular signalling pathways in canine gliomas. Vet. Comp. Oncol. 2017, 15, 133–150. [Google Scholar] [CrossRef] [PubMed]
  13. Truve, K.; Dickinson, P.; Xiong, A.; York, D.; Jayashankar, K.; Pielberg, G.; Koltookian, M.; Muren, E.; Fuxelius, H.H.; Weishaupt, H.; et al. Utilizing the Dog Genome in the Search for Novel Candidate Genes Involved in Glioma Development-Genome Wide Association Mapping followed by Targeted Massive Parallel Sequencing Identifies a Strongly Associated Locus. PLoS Genet. 2016, 12, e1006000. [Google Scholar] [CrossRef] [PubMed]
  14. Mackenzie, J.S.; Jeggo, M. The One Health Approach-Why Is It So Important? Trop. Med. Infect. Dis. 2019, 4, 88. [Google Scholar] [CrossRef] [PubMed]
  15. Fisher, F.L.; Zamanipoor Najafabadi, A.H.; van der Meer, P.B.; Boele, F.W.; Peerdeman, S.M.; Peul, W.C.; Taphoorn, M.J.B.; Dirven, L.; van Furth, W.R. Long-term health-related quality of life and neurocognitive functioning after treatment in skull base meningioma patients. J. Neurosurg. 2022, 136, 1077–1089. [Google Scholar] [CrossRef]
  16. Haider, S.; Taphoorn, M.J.B.; Drummond, K.J.; Walbert, T. Health-related quality of life in meningioma. Neurooncol. Adv. 2021, 3, vdab089. [Google Scholar] [CrossRef] [PubMed]
  17. Timmer, M.; Seibl-Leven, M.; Wittenstein, K.; Grau, S.; Stavrinou, P.; Rohn, G.; Krischek, B.; Goldbrunner, R. Long-Term Outcome and Health-Related Quality of Life of Elderly Patients After Meningioma Surgery. World Neurosurg. 2019, 125, e697–e710. [Google Scholar] [CrossRef]
  18. Van Nieuwenhuizen, D.; Klein, M.; Stalpers, L.J.; Leenstra, S.; Heimans, J.J.; Reijneveld, J.C. Differential effect of surgery and radiotherapy on neurocognitive functioning and health-related quality of life in WHO grade I meningioma patients. J. Neurooncol. 2007, 84, 271–278. [Google Scholar] [CrossRef]
  19. Zamanipoor Najafabadi, A.H.; Peeters, M.C.M.; Dirven, L.; Lobatto, D.J.; Groen, J.L.; Broekman, M.L.D.; Peerdeman, S.M.; Peul, W.C.; Taphoorn, M.J.B.; van Furth, W.R. Impaired health-related quality of life in meningioma patients—A systematic review. Neuro Oncol. 2017, 19, 897–907. [Google Scholar] [CrossRef]
  20. Meuten, D.J. Tumors in Domestic Animals, 5th ed.; Wiley-Blackwell, Ed.: Hoboken, NJ, USA, 2020. [Google Scholar]
  21. Mawrin, C. Animal models of meningiomas. Chin. Clin. Oncol. 2017, 6, S6. [Google Scholar] [CrossRef]
  22. Boetto, J.; Peyre, M.; Kalamarides, M. Mouse Models in Meningioma Research: A Systematic Review. Cancers 2021, 13, 3712. [Google Scholar] [CrossRef] [PubMed]
  23. Kalamarides, M.; Peyre, M.; Giovannini, M. Meningioma mouse models. J. Neurooncol. 2010, 99, 325–331. [Google Scholar] [CrossRef] [PubMed]
  24. Barbieri, F.; Bajetto, A.; Dellacasagrande, I.; Solari, A.; Wurth, R.; Fernandez, V.; Rancati, S.; Ceresa, D.; Appolloni, I.; De Luca, G.; et al. Stem-like signatures in human meningioma cells are under the control of CXCL11/CXCL12 chemokine activity. Neuro Oncol. 2023, 25, 1775–1787. [Google Scholar] [CrossRef] [PubMed]
  25. Xu, C.; Li, X.; Liu, P.; Li, M.; Luo, F. Patient-derived xenograft mouse models: A high fidelity tool for individualized medicine. Oncol. Lett. 2019, 17, 3–10. [Google Scholar] [CrossRef] [PubMed]
  26. Cho, S.Y. Patient-derived xenografts as compatible models for precision oncology. Lab. Anim. Res. 2020, 36, 14. [Google Scholar] [CrossRef]
  27. Candolfi, M.; Curtin, J.F.; Nichols, W.S.; Muhammad, A.G.; King, G.D.; Pluhar, G.E.; McNiel, E.A.; Ohlfest, J.R.; Freese, A.B.; Moore, P.F.; et al. Intracranial glioblastoma models in preclinical neuro-oncology: Neuropathological characterization and tumor progression. J. Neurooncol. 2007, 85, 133–148. [Google Scholar] [CrossRef] [PubMed]
  28. Stoica, G.; Kim, H.T.; Hall, D.G.; Coates, J.R. Morphology, immunohistochemistry, and genetic alterations in dog astrocytomas. Vet. Pathol. 2004, 41, 10–19. [Google Scholar] [CrossRef] [PubMed]
  29. Partridge, B.; Rossmeisl, J.H., Jr. Companion animal models of neurological disease. J. Neurosci. Methods 2020, 331, 108484. [Google Scholar] [CrossRef]
  30. Christopher, M.M. One health, one literature: Weaving together veterinary and medical research. Sci. Transl. Med. 2015, 7, 303fs336. [Google Scholar] [CrossRef]
  31. Starkey, M.P.; Scase, T.J.; Mellersh, C.S.; Murphy, S. Dogs really are man’s best friend—Canine genomics has applications in veterinary and human medicine! Brief. Funct. Genom. 2005, 4, 112–128. [Google Scholar] [CrossRef]
  32. LeBlanc, A.K.; Mazcko, C.; Brown, D.E.; Koehler, J.W.; Miller, A.D.; Miller, C.R.; Bentley, R.T.; Packer, R.A.; Breen, M.; Boudreau, C.E.; et al. Creation of an NCI comparative brain tumor consortium: Informing the translation of new knowledge from canine to human brain tumor patients. Neuro Oncol. 2016, 18, 1209–1218. [Google Scholar] [CrossRef] [PubMed]
  33. Parker, H.G.; Shearin, A.L.; Ostrander, E.A. Man’s best friend becomes biology’s best in show: Genome analyses in the domestic dog. Annu. Rev. Genet. 2010, 44, 309–336. [Google Scholar] [CrossRef] [PubMed]
  34. Reif, J.S.; Cohen, D. The environmental distribution of canine respiratory tract neoplasms. Arch. Environ. Health 1971, 22, 136–140. [Google Scholar] [CrossRef] [PubMed]
  35. Sarver, A.L.; Makielski, K.M.; DePauw, T.A.; Schulte, A.J.; Modiano, J.F. Increased risk of cancer in dogs and humans: A consequence of recent extension of lifespan beyond evolutionarily-determined limitations? Aging Cancer 2022, 3, 3–19. [Google Scholar] [CrossRef] [PubMed]
  36. Davis, B.W.; Ostrander, E.A. Domestic dogs and cancer research: A breed-based genomics approach. ILAR J. 2014, 55, 59–68. [Google Scholar] [CrossRef] [PubMed]
  37. Khanna, C.; Lindblad-Toh, K.; Vail, D.; London, C.; Bergman, P.; Barber, L.; Breen, M.; Kitchell, B.; McNeil, E.; Modiano, J.F.; et al. The dog as a cancer model. Nat. Biotechnol. 2006, 24, 1065–1066. [Google Scholar] [CrossRef] [PubMed]
  38. Thomson, S.A.; Kennerly, E.; Olby, N.; Mickelson, J.R.; Hoffmann, D.E.; Dickinson, P.J.; Gibson, G.; Breen, M. Microarray analysis of differentially expressed genes of primary tumors in the canine central nervous system. Vet. Pathol. 2005, 42, 550–558. [Google Scholar] [CrossRef]
  39. Giuliano, A. Companion Animal Model in Translational Oncology; Feline Oral Squamous Cell Carcinoma and Canine Oral Melanoma. Biology 2021, 11, 54. [Google Scholar] [CrossRef]
  40. Kimmelman, J.; Nalbantoglu, J. Faithful companions: A proposal for neurooncology trials in pet dogs. Cancer Res. 2007, 67, 4541–4544. [Google Scholar] [CrossRef]
  41. Mazcko, C.; Thomas, R. The Establishment of the Pfizer-Canine Comparative Oncology and Genomics Consortium Biospecimen Repository. Vet. Sci. 2015, 2, 127–130. [Google Scholar] [CrossRef]
  42. Dickinson, P.J.; LeCouteur, R.A.; Higgins, R.J.; Bringas, J.R.; Larson, R.F.; Yamashita, Y.; Krauze, M.T.; Forsayeth, J.; Noble, C.O.; Drummond, D.C.; et al. Canine spontaneous glioma: A translational model system for convection-enhanced delivery. Neuro Oncol. 2010, 12, 928–940. [Google Scholar] [CrossRef] [PubMed]
  43. Platt, S.; Nduom, E.; Kent, M.; Freeman, C.; Machaidze, R.; Kaluzova, M.; Wang, L.; Mao, H.; Hadjipanayis, C.G. Canine model of convection-enhanced delivery of cetuximab-conjugated iron-oxide nanoparticles monitored with magnetic resonance imaging. Clin. Neurosurg. 2012, 59, 107–113. [Google Scholar] [CrossRef] [PubMed]
  44. Dickinson, P.J.; LeCouteur, R.A.; Higgins, R.J.; Bringas, J.R.; Roberts, B.; Larson, R.F.; Yamashita, Y.; Krauze, M.; Noble, C.O.; Drummond, D.; et al. Canine model of convection-enhanced delivery of liposomes containing CPT-11 monitored with real-time magnetic resonance imaging: Laboratory investigation. J. Neurosurg. 2008, 108, 989–998. [Google Scholar] [CrossRef] [PubMed]
  45. Ostrom, Q.T.; Gittleman, H.; Fulop, J.; Liu, M.; Blanda, R.; Kromer, C.; Wolinsky, Y.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2008–2012. Neuro Oncol. 2015, 17 (Suppl. S4), iv1–iv62. [Google Scholar] [CrossRef] [PubMed]
  46. Ostrom, Q.T.; Gittleman, H.; Truitt, G.; Boscia, A.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2011–2015. Neuro Oncol. 2018, 20, iv1–iv86. [Google Scholar] [CrossRef] [PubMed]
  47. Ostrom, Q.T.; Price, M.; Neff, C.; Cioffi, G.; Waite, K.A.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2015–2019. Neuro Oncol. 2022, 24, v1–v95. [Google Scholar] [CrossRef]
  48. Kotecha, R.S.; Pascoe, E.M.; Rushing, E.J.; Rorke-Adams, L.B.; Zwerdling, T.; Gao, X.; Li, X.; Greene, S.; Amirjamshidi, A.; Kim, S.K.; et al. Meningiomas in children and adolescents: A meta-analysis of individual patient data. Lancet Oncol. 2011, 12, 1229–1239. [Google Scholar] [CrossRef]
  49. Huntoon, K.; Toland, A.M.S.; Dahiya, S. Meningioma: A Review of Clinicopathological and Molecular Aspects. Front. Oncol. 2020, 10, 579599. [Google Scholar] [CrossRef]
  50. Hayes, H.M.; Priester, W.A., Jr.; Pendergrass, T.W. Occurrence of nervous-tissue tumors in cattle, horses, cats and dogs. Int. J. Cancer 1975, 15, 39–47. [Google Scholar] [CrossRef]
  51. Troxel, M.T.; Vite, C.H.; Van Winkle, T.J.; Newton, A.L.; Tiches, D.; Dayrell-Hart, B.; Kapatkin, A.S.; Shofer, F.S.; Steinberg, S.A. Feline intracranial neoplasia: Retrospective review of 160 cases (1985–2001). J. Vet. Intern. Med. 2003, 17, 850–859. [Google Scholar] [CrossRef]
  52. Wada, M.; Hasegawa, D.; Hamamoto, Y.; Yu, Y.; Asada, R.; Fujiwara-Igarashi, A.; Fujita, M. Comparison of Canine and Feline Meningiomas Using the Apparent Diffusion Coefficient and Fractional Anisotropy. Front. Vet. Sci. 2020, 7, 614026. [Google Scholar] [CrossRef] [PubMed]
  53. Kishimoto, T.E.; Uchida, K.; Chambers, J.K.; Kok, M.K.; Son, N.V.; Shiga, T.; Hirabayashi, M.; Ushio, N.; Nakayama, H. A retrospective survey on canine intracranial tumors between 2007 and 2017. J. Vet. Med. Sci. 2020, 82, 77–83. [Google Scholar] [CrossRef] [PubMed]
  54. Withrow, S.J.; Page, R.; Vail, D.M. Small Animal Clinical Oncology; Elsevier: Amsterdam, The Netherlands, 2013. [Google Scholar]
  55. Bagley, R.S.; Gavin, P.R. Seizures as a complication of brain tumors in dogs. Clin. Tech. Small Anim. Pract. 1998, 13, 179–184. [Google Scholar] [CrossRef] [PubMed]
  56. Gavin, P.R.; Fike, J.R.; Hoopes, P.J. Central nervous system tumors. Semin. Vet. Med. Surg. Small Anim. 1995, 10, 180–189. [Google Scholar]
  57. Motta, L.; Mandara, M.T.; Skerritt, G.C. Canine and feline intracranial meningiomas: An updated review. Vet. J. 2012, 192, 153–165. [Google Scholar] [CrossRef]
  58. Sturges, B.K.; Dickinson, P.J.; Bollen, A.W.; Koblik, P.D.; Kass, P.H.; Kortz, G.D.; Vernau, K.M.; Knipe, M.F.; Lecouteur, R.A.; Higgins, R.J. Magnetic resonance imaging and histological classification of intracranial meningiomas in 112 dogs. J. Vet. Intern. Med. 2008, 22, 586–595. [Google Scholar] [CrossRef]
  59. Patnaik, A.K.; Lieberman, P.H.; Erlandson, R.A.; Shaker, E.; Hurvitz, A.I. Paranasal meningioma in the dog: A clinicopathologic study of ten cases. Vet. Pathol. 1986, 23, 362–368. [Google Scholar] [CrossRef]
  60. Kaldrymidou, E.; Polizopoulou, Z.S.; Koutinas, A.F.; Papaioannou, N.; Papadopoulos, G.; Poutahidis, T. Papillary meningioma in the cerebellum of a cat. J. Comp. Pathol. 2000, 123, 222–225. [Google Scholar] [CrossRef]
  61. Mandara, M.T.; Ricci, G.; Sforna, M. A cerebral granular cell tumor in a cat. Vet. Pathol. 2006, 43, 797–800. [Google Scholar] [CrossRef]
  62. Forterre, F.; Tomek, A.; Konar, M.; Vandevelde, M.; Howard, J.; Jaggy, A. Multiple meningiomas: Clinical, radiological, surgical, and pathological findings with outcome in four cats. J. Feline Med. Surg. 2007, 9, 36–43. [Google Scholar] [CrossRef]
  63. Gordon, L.E.; Thacher, C.; Matthiesen, D.T.; Joseph, R.J. Results of craniotomy for the treatment of cerebral meningioma in 42 cats. Vet. Surg. 1994, 23, 94–100. [Google Scholar] [CrossRef] [PubMed]
  64. Song, K.; Lee, H.; Jeong, J.; Roh, Y. Multiple Meningioma Resection by Bilateral Extended Rostrotentorial Craniotomy with a 3D-Print Guide in a Cat. Vet. Sci. 2022, 9, 512. [Google Scholar] [CrossRef] [PubMed]
  65. Forward, A.K.; Volk, H.A.; Cherubini, G.B.; Harcourt-Brown, T.; Plessas, I.N.; Garosi, L.; De Decker, S. Clinical presentation, diagnostic findings and outcome of dogs undergoing surgical resection for intracranial meningioma: 101 dogs. BMC Vet. Res. 2022, 18, 88. [Google Scholar] [CrossRef] [PubMed]
  66. Wiemels, J.; Wrensch, M.; Claus, E.B. Epidemiology and etiology of meningioma. J. Neurooncol. 2010, 99, 307–314. [Google Scholar] [CrossRef] [PubMed]
  67. Ogasawara, C.; Philbrick, B.D.; Adamson, D.C. Meningioma: A Review of Epidemiology, Pathology, Diagnosis, Treatment, and Future Directions. Biomedicines 2021, 9, 319. [Google Scholar] [CrossRef]
  68. Sadamori, N.; Shibata, S.; Mine, M.; Miyazaki, H.; Miyake, H.; Kurihara, M.; Tomonaga, M.; Sekine, I.; Okumura, Y. Incidence of intracranial meningiomas in Nagasaki atomic-bomb survivors. Int. J. Cancer 1996, 67, 318–322. [Google Scholar] [CrossRef]
  69. Samanic, C.M.; De Roos, A.J.; Stewart, P.A.; Rajaraman, P.; Waters, M.A.; Inskip, P.D. Occupational exposure to pesticides and risk of adult brain tumors. Am. J. Epidemiol. 2008, 167, 976–985. [Google Scholar] [CrossRef]
  70. Shao, C.; Bai, L.P.; Qi, Z.Y.; Hui, G.Z.; Wang, Z. Overweight, obesity and meningioma risk: A meta-analysis. PLoS ONE 2014, 9, e90167. [Google Scholar] [CrossRef]
  71. Lichtor, T.; Kurpakus, M.A.; Gurney, M.E. Expression of insulin-like growth factors and their receptors in human meningiomas. J. Neurooncol. 1993, 17, 183–190. [Google Scholar] [CrossRef]
  72. Glick, R.P.; Gettleman, R.; Patel, K.; Lakshman, R.; Tsibris, J.C. Insulin and insulin-like growth factor I in brain tumors: Binding and in vitro effects. Neurosurgery 1989, 24, 791–797. [Google Scholar] [CrossRef]
  73. Antoniades, H.N.; Galanopoulos, T.; Neville-Golden, J.; Maxwell, M. Expression of insulin-like growth factors I and II and their receptor mRNAs in primary human astrocytomas and meningiomas; in vivo studies using in situ hybridization and immunocytochemistry. Int. J. Cancer 1992, 50, 215–222. [Google Scholar] [CrossRef] [PubMed]
  74. Kurihara, M.; Tokunaga, Y.; Tsutsumi, K.; Kawaguchi, T.; Shigematsu, K.; Niwa, M.; Mori, K. Characterization of insulin-like growth factor I and epidermal growth factor receptors in meningioma. J. Neurosurg. 1989, 71, 538–544. [Google Scholar] [CrossRef] [PubMed]
  75. Korhonen, K.; Salminen, T.; Raitanen, J.; Auvinen, A.; Isola, J.; Haapasalo, H. Female predominance in meningiomas can not be explained by differences in progesterone, estrogen, or androgen receptor expression. J. Neurooncol. 2006, 80, 1–7. [Google Scholar] [CrossRef] [PubMed]
  76. Baldi, I.; Engelhardt, J.; Bonnet, C.; Bauchet, L.; Berteaud, E.; Gruber, A.; Loiseau, H. Epidemiology of meningiomas. Neurochirurgie 2018, 64, 5–14. [Google Scholar] [CrossRef] [PubMed]
  77. Wu, W.; Zhou, Y.; Wang, Y.; Liu, L.; Lou, J.; Deng, Y.; Zhao, P.; Shao, A. Clinical Significance of Somatostatin Receptor (SSTR) 2 in Meningioma. Front. Oncol. 2020, 10, 1633. [Google Scholar] [CrossRef] [PubMed]
  78. Delgado-Lopez, P.D.; Cubo-Delgado, E.; Gonzalez-Bernal, J.J.; Martin-Alonso, J. A Practical Overview on the Molecular Biology of Meningioma. Curr. Neurol. Neurosci. Rep. 2020, 20, 62. [Google Scholar] [CrossRef]
  79. Arena, S.; Barbieri, F.; Thellung, S.; Pirani, P.; Corsaro, A.; Villa, V.; Dadati, P.; Dorcaratto, A.; Lapertosa, G.; Ravetti, J.L.; et al. Expression of somatostatin receptor mRNA in human meningiomas and their implication in in vitro antiproliferative activity. J. Neurooncol. 2004, 66, 155–166. [Google Scholar] [CrossRef]
  80. Miyagishima, D.F.; Moliterno, J.; Claus, E.; Gunel, M. Hormone therapies in meningioma-where are we? J. Neurooncol. 2022, 161, 297–308. [Google Scholar] [CrossRef]
  81. Pravdenkova, S.; Al-Mefty, O.; Sawyer, J.; Husain, M. Progesterone and estrogen receptors: Opposing prognostic indicators in meningiomas. J. Neurosurg. 2006, 105, 163–173. [Google Scholar] [CrossRef]
  82. Samoyeau, T.; Provost, C.; Roux, A.; Legrand, L.; Dezamis, E.; Plu-Bureau, G.; Pallud, J.; Oppenheim, C.; Benzakoun, J. Meningioma in patients exposed to progestin drugs: Results from a real-life screening program. J. Neurooncol. 2022, 160, 127–136. [Google Scholar] [CrossRef]
  83. Hoisnard, L.; Laanani, M.; Passeri, T.; Duranteau, L.; Coste, J.; Zureik, M.; Froelich, S.; Weill, A. Risk of intracranial meningioma with three potent progestogens: A population-based case-control study. Eur. J. Neurol. 2022, 29, 2801–2809. [Google Scholar] [CrossRef] [PubMed]
  84. Mandara, M.T.; Ricci, G.; Rinaldi, L.; Sarli, G.; Vitellozzi, G. Immunohistochemical identification and image analysis quantification of oestrogen and progesterone receptors in canine and feline meningioma. J. Comp. Pathol. 2002, 127, 214–218. [Google Scholar] [CrossRef]
  85. Abaza, M.M.; Makariou, E.; Armstrong, M.; Lalwani, A.K. Growth rate characteristics of acoustic neuromas associated with neurofibromatosis type 2. Laryngoscope 1996, 106, 694–699. [Google Scholar] [CrossRef] [PubMed]
  86. Goutagny, S.; Bah, A.B.; Henin, D.; Parfait, B.; Grayeli, A.B.; Sterkers, O.; Kalamarides, M. Long-term follow-up of 287 meningiomas in neurofibromatosis type 2 patients: Clinical, radiological, and molecular features. Neuro Oncol. 2012, 14, 1090–1096. [Google Scholar] [CrossRef] [PubMed]
  87. Albrecht, S.; Goodman, J.C.; Rajagopolan, S.; Levy, M.; Cech, D.A.; Cooley, L.D. Malignant meningioma in Gorlin’s syndrome: Cytogenetic and p53 gene analysis. Case report. J. Neurosurg. 1994, 81, 466–471. [Google Scholar] [CrossRef]
  88. Kimonis, V.E.; Mehta, S.G.; Digiovanna, J.J.; Bale, S.J.; Pastakia, B. Radiological features in 82 patients with nevoid basal cell carcinoma (NBCC or Gorlin) syndrome. Genet. Med. 2004, 6, 495–502. [Google Scholar] [CrossRef]
  89. Lopes, N.N.; Caran, E.M.; Lee, M.L.; Silva, N.S.; Rocha, A.C.; Macedo, C.R. Gorlin-Goltz syndrome and neoplasms: A case study. J. Clin. Pediatr. Dent. 2010, 35, 203–206. [Google Scholar] [CrossRef]
  90. Narang, A.; Maheshwari, C.; Aggarwal, V.; Bansal, P.; Singh, P. Gorlin-Goltz Syndrome with Intracranial Meningioma: Case Report and Review of Literature. World Neurosurg. 2020, 133, 324–330. [Google Scholar] [CrossRef]
  91. Yakubov, E.; Ghoochani, A.; Buslei, R.; Buchfelder, M.; Eyupoglu, I.Y.; Savaskan, N. Hidden association of Cowden syndrome, PTEN mutation and meningioma frequency. Oncoscience 2016, 3, 149–155. [Google Scholar] [CrossRef]
  92. Shankar, G.M.; Abedalthagafi, M.; Vaubel, R.A.; Merrill, P.H.; Nayyar, N.; Gill, C.M.; Brewster, R.; Bi, W.L.; Agarwalla, P.K.; Thorner, A.R.; et al. Germline and somatic BAP1 mutations in high-grade rhabdoid meningiomas. Neuro Oncol. 2017, 19, 535–545. [Google Scholar] [CrossRef]
  93. Marini, F.; Falchetti, A.; Del Monte, F.; Carbonell Sala, S.; Gozzini, A.; Luzi, E.; Brandi, M.L. Multiple endocrine neoplasia type 1. Orphanet J. Rare Dis. 2006, 1, 38. [Google Scholar] [CrossRef] [PubMed]
  94. Asgharian, B.; Chen, Y.J.; Patronas, N.J.; Peghini, P.L.; Reynolds, J.C.; Vortmeyer, A.; Zhuang, Z.; Venzon, D.J.; Gibril, F.; Jensen, R.T. Meningiomas may be a component tumor of multiple endocrine neoplasia type 1. Clin. Cancer Res. 2004, 10, 869–880. [Google Scholar] [CrossRef] [PubMed]
  95. Lauper, J.M.; Krause, A.; Vaughan, T.L.; Monnat, R.J., Jr. Spectrum and risk of neoplasia in Werner syndrome: A systematic review. PLoS ONE 2013, 8, e59709. [Google Scholar] [CrossRef]
  96. Verstegen, M.J.; van den Munckhof, P.; Troost, D.; Bouma, G.J. Multiple meningiomas in a patient with Rubinstein-Taybi syndrome. Case report. J. Neurosurg. 2005, 102, 167–168. [Google Scholar] [CrossRef] [PubMed]
  97. Boot, M.V.; van Belzen, M.J.; Overbeek, L.I.; Hijmering, N.; Mendeville, M.; Waisfisz, Q.; Wesseling, P.; Hennekam, R.C.; de Jong, D. Benign and malignant tumors in Rubinstein-Taybi syndrome. Am. J. Med. Genet. A 2018, 176, 597–608. [Google Scholar] [CrossRef] [PubMed]
  98. Gehlhausen, J.R.; Park, S.J.; Hickox, A.E.; Shew, M.; Staser, K.; Rhodes, S.D.; Menon, K.; Lajiness, J.D.; Mwanthi, M.; Yang, X.; et al. A murine model of neurofibromatosis type 2 that accurately phenocopies human schwannoma formation. Hum. Mol. Genet. 2015, 24, 1–8. [Google Scholar] [CrossRef]
  99. Hahn, H.; Wojnowski, L.; Zimmer, A.M.; Hall, J.; Miller, G.; Zimmer, A. Rhabdomyosarcomas and radiation hypersensitivity in a mouse model of Gorlin syndrome. Nat. Med. 1998, 4, 619–622. [Google Scholar] [CrossRef]
  100. He, X.; Arrotta, N.; Radhakrishnan, D.; Wang, Y.; Romigh, T.; Eng, C. Cowden syndrome-related mutations in PTEN associate with enhanced proteasome activity. Cancer Res. 2013, 73, 3029–3040. [Google Scholar] [CrossRef]
  101. Roccabianca, P.; Rondena, M.; Paltrinieri, S.; Pocacqua, V.; Scarpa, P.; Faverzani, S.; Scanziani, E.; Caniatti, M. Multiple endocrine neoplasia type-I-like syndrome in two cats. Vet. Pathol. 2006, 43, 345–352. [Google Scholar] [CrossRef]
  102. Walker, M.C.; Jones, B.R.; Guildford, W.G.; Burbidge, H.M.; Alley, M.R. Multiple endocrine neoplasia type 1 in a crossbred dog. J. Small Anim. Pract. 2000, 41, 67–70. [Google Scholar] [CrossRef]
  103. Bigio Marcello, A.; Gieger, T.L.; Jimenez, D.A.; Granger, L.A. Detection of comorbidities and synchronous primary tumours via thoracic radiography and abdominal ultrasonography and their influence on treatment outcome in dogs with soft tissue sarcomas, primary brain tumours and intranasal tumours. Vet. Comp. Oncol. 2015, 13, 433–442. [Google Scholar] [CrossRef] [PubMed]
  104. Stacy, B.A.; Stevenson, T.L.; Lipsitz, D.; Higgins, R.J. Simultaneously occurring oligodendroglioma and meningioma in a dog. J. Vet. Intern. Med. 2003, 17, 357–359. [Google Scholar] [CrossRef] [PubMed]
  105. Ginel, P.J.; Novales, M.; Blanco, B.; Perez, J.; Zafra, R.; Perez-Ecija, R.A. Meningioangiomatosis associated with fibrous meningioma in a dog. Vet. Rec. 2009, 164, 756–758. [Google Scholar] [CrossRef] [PubMed]
  106. Haskins, M.E.; Aguirre, G.D.; Jezyk, P.F.; Desnick, R.J.; Patterson, D.F. The pathology of the feline model of mucopolysaccharidosis I. Am. J. Pathol. 1983, 112, 27–36. [Google Scholar] [PubMed]
  107. Lobetti, R.G.; Nesbit, J.W.; Miller, D.B. Multiple malignant meningiomas in a young cat. J. S. Afr. Vet. Assoc. 1997, 68, 62–65. [Google Scholar] [CrossRef] [PubMed]
  108. Lu, D.; Pocknell, A.; Lamb, C.R.; Targett, M.P. Concurrent benign and malignant multiple meningiomas in a cat: Clinical, MRI and pathological findings. Vet. Rec. 2003, 152, 780–782. [Google Scholar] [CrossRef] [PubMed]
  109. Enam, S.A.; Abdulrauf, S.; Mehta, B.; Malik, G.M.; Mahmood, A. Metastasis in meningioma. Acta Neurochir. 1996, 138, 1172–1177; discussion 1177–1178. [Google Scholar] [CrossRef]
  110. Mutnuru, P.C.; Ahmed, S.F.; Uppin, S.G.; Lachi, P.K. Pulmonary metastases from intracranial meningioma. Lung India 2015, 32, 661–663. [Google Scholar] [CrossRef]
  111. Schulman, F.Y.; Ribas, J.L.; Carpenter, J.L.; Sisson, A.F.; LeCouteur, R.A. Intracranial meningioma with pulmonary metastasis in three dogs. Vet. Pathol. 1992, 29, 196–202. [Google Scholar] [CrossRef]
  112. Dussaux, A.; Gomes, E.; Hurion, M.; Rattez, E.; Mayousse, V. Cerebral meningioma associated with extensive calvarium osteolysis and presumed intratumoral carcinoma metastasis in a cat. JFMS Open Rep. 2022, 8, 20551169221122849. [Google Scholar] [CrossRef]
  113. Lee, Y.S.; Lee, Y.S. Molecular characteristics of meningiomas. J. Pathol. Transl. Med. 2020, 54, 45–63. [Google Scholar] [CrossRef] [PubMed]
  114. Shibuya, M. Pathology and Molecular Genetics of Meningioma: Recent Advances. Neurol. Med. Chir. 2015, 55 (Suppl. S1), 14–27. [Google Scholar] [CrossRef] [PubMed]
  115. Belluco, S.; Marano, G.; Baiker, K.; Beineke, A.; Oevermann, A.; Seehusen, F.; Boracchi, P.; Pumarola, M.; Mandara, M.T. Standardisation of canine meningioma grading: Inter-observer agreement and recommendations for reproducible histopathologic criteria. Vet. Comp. Oncol. 2022, 20, 509–520. [Google Scholar] [CrossRef] [PubMed]
  116. Vernau, K.M.; Higgins, R.J.; Bollen, A.W.; Jimenez, D.F.; Anderson, J.V.; Koblik, P.D.; LeCouteur, R.A. Primary canine and feline nervous system tumors: Intraoperative diagnosis using the smear technique. Vet. Pathol. 2001, 38, 47–57. [Google Scholar] [CrossRef] [PubMed]
  117. Fages, J.; Oura, T.J.; Sutherland-Smith, J.; Jennings, S.H. Atypical and malignant canine intracranial meningiomas may have lower apparent diffusion coefficient values than benign tumors. Vet. Radiol. Ultrasound 2020, 61, 40–47. [Google Scholar] [CrossRef] [PubMed]
  118. Kitagawa, M.; Kanayama, K.; Sakai, T. Cystic meningioma in a dog. J. Small Anim. Pract. 2002, 43, 272–274. [Google Scholar] [CrossRef]
  119. Koestner, A. Histological Classification of Tumors of the Nervous System of Domestic Animals; Armed Forces Institute of Pathology: Washington, DC, USA, 1999.
  120. Comparative Brain Tumors Consortium. Available online: https://ccr.cancer.gov/comparative-oncology-program/pathology-and-molecular-markers (accessed on 6 September 2023).
  121. Koehler, J.W.; Miller, A.D.; Miller, C.R.; Porter, B.; Aldape, K.; Beck, J.; Brat, D.; Cornax, I.; Corps, K.; Frank, C.; et al. A Revised Diagnostic Classification of Canine Glioma: Towards Validation of the Canine Glioma Patient as a Naturally Occurring Preclinical Model for Human Glioma. J. Neuropathol. Exp. Neurol. 2018, 77, 1039–1054. [Google Scholar] [CrossRef]
  122. Goldbrunner, R.; Minniti, G.; Preusser, M.; Jenkinson, M.D.; Sallabanda, K.; Houdart, E.; von Deimling, A.; Stavrinou, P.; Lefranc, F.; Lund-Johansen, M.; et al. EANO guidelines for the diagnosis and treatment of meningiomas. Lancet Oncol. 2016, 17, e383–e391. [Google Scholar] [CrossRef]
  123. Nowosielski, M.; Galldiks, N.; Iglseder, S.; Kickingereder, P.; von Deimling, A.; Bendszus, M.; Wick, W.; Sahm, F. Diagnostic challenges in meningioma. Neuro Oncol. 2017, 19, 1588–1598. [Google Scholar] [CrossRef]
  124. Turrel, J.M.; Fike, J.R.; LeCouteur, R.A.; Higgins, R.J. Computed tomographic characteristics of primary brain tumors in 50 dogs. J. Am. Vet. Med. Assoc. 1986, 188, 851–856. [Google Scholar]
  125. Sutherland, T.; Temple, F.; Galvin, A.; Hennessy, O. Contrast-enhanced ultrasound of the spleen: An introduction and pictorial essay. Insights Imaging 2011, 2, 515–524. [Google Scholar] [CrossRef] [PubMed]
  126. Vite, C.H.; Cross, J.R. Correlating magnetic resonance findings with neuropathology and clinical signs in dogs and cats. Vet. Radiol. Ultrasound 2011, 52, S23–S31. [Google Scholar] [CrossRef] [PubMed]
  127. Zhao, W.; Zheng, X.; Liu, Y.; Yang, W.; Amirbekian, V.; Diaz, L.E.; Huang, X. An MRI study of symptomatic adhesive capsulitis. PLoS ONE 2012, 7, e47277. [Google Scholar] [CrossRef] [PubMed]
  128. Zhao, M.; Zhou, Y.; Chang, J.; Hu, J.; Liu, H.; Wang, S.; Si, D.; Yuan, Y.; Li, H. The accuracy of MRI in the diagnosis of anterior cruciate ligament injury. Ann. Transl. Med. 2020, 8, 1657. [Google Scholar] [CrossRef] [PubMed]
  129. Zhao, J.P.; Liu, X.J.; Lin, H.Z.; Cui, C.X.; Yue, Y.J.; Gao, S.; Xu, H.Z. MRI comparative study of diffuse midline glioma, H3 K27-altered and glioma in the midline without H3 K27-altered. BMC Neurol. 2022, 22, 498. [Google Scholar] [CrossRef] [PubMed]
  130. Latouche, E.L.; Arena, C.B.; Ivey, J.W.; Garcia, P.A.; Pancotto, T.E.; Pavlisko, N.; Verbridge, S.S.; Davalos, R.V.; Rossmeisl, J.H. High-Frequency Irreversible Electroporation for Intracranial Meningioma: A Feasibility Study in a Spontaneous Canine Tumor Model. Technol. Cancer Res. Treat. 2018, 17, 1533033818785285. [Google Scholar] [CrossRef]
  131. Filippi, C.G.; Edgar, M.A.; Ulug, A.M.; Prowda, J.C.; Heier, L.A.; Zimmerman, R.D. Appearance of meningiomas on diffusion-weighted images: Correlating diffusion constants with histopathologic findings. AJNR Am. J. Neuroradiol. 2001, 22, 65–72. [Google Scholar]
  132. Elster, A.D.; Challa, V.R.; Gilbert, T.H.; Richardson, D.N.; Contento, J.C. Meningiomas: MR and histopathologic features. Radiology 1989, 170, 857–862. [Google Scholar] [CrossRef]
  133. Volckaert, V.; Royaux, E. Peritumoral (pseudo)cystic meningioma in a cat. Vlaams Diergeneeskd. Tijdschr. 2020, 89, 215–220. [Google Scholar] [CrossRef]
  134. Saito, R.; Chambers, J.K.; Kishimoto, T.E.; Uchida, K. Pathological and immunohistochemical features of 45 cases of feline meningioma. J. Vet. Med. Sci. 2021, 83, 1219–1224. [Google Scholar] [CrossRef]
  135. Figarella-Branger, D.; Pellissier, J.F.; Bouillot, P.; Bianco, N.; Mayan, M.; Grisoli, F.; Rougon, G. Expression of neural cell-adhesion molecule isoforms and epithelial cadherin adhesion molecules in 47 human meningiomas: Correlation with clinical and morphological data. Mod. Pathol. 1994, 7, 752–761. [Google Scholar]
  136. Ide, T.; Uchida, K.; Suzuki, K.; Kagawa, Y.; Nakayama, H. Expression of cell adhesion molecules and doublecortin in canine anaplastic meningiomas. Vet. Pathol. 2011, 48, 292–301. [Google Scholar] [CrossRef] [PubMed]
  137. Kepes, J.J. Presidential Address: The Histopathology of Meningiomas. A Reflection of Origins and Expected Behavior? J. Neuropathol. Exp. Neurol. 1986, 45, 95–107. [Google Scholar] [CrossRef] [PubMed]
  138. Liu, N.; Song, S.Y.; Jiang, J.B.; Wang, T.J.; Yan, C.X. The prognostic role of Ki-67/MIB-1 in meningioma: A systematic review with meta-analysis. Medicine 2020, 99, e18644. [Google Scholar] [CrossRef]
  139. Devaprasath, A.; Chacko, G. Diagnostic validity of the Ki-67 labeling index using the MIB-1 monoclonal antibody in the grading of meningiomas. Neurol. India 2003, 51, 336–340. [Google Scholar] [PubMed]
  140. Takahashi, J.A.; Ueba, T.; Hashimoto, N.; Nakashima, Y.; Katsuki, N. The combination of mitotic and Ki-67 indices as a useful method for predicting short-term recurrence of meningiomas. Surg. Neurol. 2004, 61, 149–155; discussion 155–146. [Google Scholar] [CrossRef] [PubMed]
  141. Ferraraccio, F.; Accardo, M.; Giangaspero, F.; Cuccurullo, L. Recurrent and atypical meningiomas--a multiparametric study using Ki67 labelling index, AgNOR and DNA Feulgen staining. Clin. Neuropathol. 2003, 22, 187–192. [Google Scholar] [PubMed]
  142. Hsu, D.W.; Efird, J.T.; Hedley-Whyte, E.T. MIB-1 (Ki-67) index and transforming growth factor-alpha (TGF alpha) immunoreactivity are significant prognostic predictors for meningiomas. Neuropathol. Appl. Neurobiol. 1998, 24, 441–452. [Google Scholar] [CrossRef]
  143. Matiasek, L.A.; Platt, S.R.; Adams, V.; Scase, T.J.; Keys, D.; Miller, J.; Adamo, F.; Long, S.; Matiasek, K. Ki-67 and vascular endothelial growth factor expression in intracranial meningiomas in dogs. J. Vet. Intern. Med. 2009, 23, 146–151. [Google Scholar] [CrossRef]
  144. Roels, S.; Tilmant, K.; Ducatelle, R. PCNA and Ki67 proliferation markers as criteria for prediction of clinical behaviour of melanocytic tumours in cats and dogs. J. Comp. Pathol. 1999, 121, 13–24. [Google Scholar] [CrossRef]
  145. Castagnaro, M.; De Maria, R.; Bozzetta, E.; Ru, G.; Casalone, C.; Biolatti, B.; Caramelli, M. Ki-67 index as indicator of the post-surgical prognosis in feline mammary carcinomas. Res. Vet. Sci. 1998, 65, 223–226. [Google Scholar] [CrossRef] [PubMed]
  146. Peña, L.L.; Nieto, A.I.; Pérez-Alenza, D.; Cuesta, P.; Castaño, M. Immunohistochemical detection of Ki-67 and PCNA in canine mammary tumors: Relationship to clinical and pathologic variables. J. Vet. Diagn. Investig. 1998, 10, 237–246. [Google Scholar] [CrossRef] [PubMed]
  147. Abadie, J.J.; Amardeilh, M.A.; Delverdier, M.E. Immunohistochemical detection of proliferating cell nuclear antigen and Ki-67 in mast cell tumors from dogs. J. Am. Vet. Med. Assoc. 1999, 215, 1629–1634. [Google Scholar] [PubMed]
  148. Janssen, J.; Oevermann, A.; Walter, I.; Tichy, A.; Kummer, S.; Gradner, G. Osteopontin and Ki-67 expression in World Health Organization graded canine meningioma. J. Comp. Pathol. 2023, 201, 41–48. [Google Scholar] [CrossRef] [PubMed]
  149. Artlich, A.; Schmidt, D. Immunohistochemical profile of meningiomas and their histological subtypes. Hum. Pathol. 1990, 21, 843–849. [Google Scholar] [CrossRef] [PubMed]
  150. Ruttledge, M.H.; Sarrazin, J.; Rangaratnam, S.; Phelan, C.M.; Twist, E.; Merel, P.; Delattre, O.; Thomas, G.; Nordenskjold, M.; Collins, V.P.; et al. Evidence for the complete inactivation of the NF2 gene in the majority of sporadic meningiomas. Nat. Genet. 1994, 6, 180–184. [Google Scholar] [CrossRef] [PubMed]
  151. Sato, T.; Sekido, Y. NF2/Merlin Inactivation and Potential Therapeutic Targets in Mesothelioma. Int. J. Mol. Sci. 2018, 19, 988. [Google Scholar] [CrossRef]
  152. Gerber, M.A.; Bahr, S.M.; Gutmann, D.H. Protein 4.1B/Differentially Expressed in Adenocarcinoma of the Lung-1 Functions as a Growth Suppressor in Meningioma Cells by Activating Rac1-Dependent c-Jun-NH2-kinase Signaling. Cell Tumor Stem Cell Biol. 2016, 66, 5295–5303. [Google Scholar] [CrossRef]
  153. Pecina-Slaus, N.; Kafka, A.; Lechpammer, M. Molecular Genetics of Intracranial Meningiomas with Emphasis on Canonical Wnt Signalling. Cancers 2016, 8, 67. [Google Scholar] [CrossRef]
  154. Wang, Z.; Zhang, J.; Ye, M.; Zhu, M.; Zhang, B.; Roy, M.; Liu, J.; An, X. Tumor suppressor role of protein 4.1B/DAL-1. Cell Mol. Life Sci. 2014, 71, 4815–4830. [Google Scholar] [CrossRef]
  155. Lamszus, K. Meningioma pathology, genetics, and biology. J. Neuropathol. Exp. Neurol. 2004, 63, 275–286. [Google Scholar] [CrossRef] [PubMed]
  156. Bostrom, J.; Meyer-Puttlitz, B.; Wolter, M.; Blaschke, B.; Weber, R.G.; Lichter, P.; Ichimura, K.; Collins, V.P.; Reifenberger, G. Alterations of the tumor suppressor genes CDKN2A (p16(INK4a)), p14(ARF), CDKN2B (p15(INK4b)), and CDKN2C (p18(INK4c)) in atypical and anaplastic meningiomas. Am. J. Pathol. 2001, 159, 661–669. [Google Scholar] [CrossRef] [PubMed]
  157. Mendiola, M.; Bello, M.J.; Alonso, J.; Leone, P.E.; Vaquero, J.; Sarasa, J.L.; Kusak, M.E.; De Campos, J.M.; Pestana, A.; Rey, J.A. Search for mutations of the hRAD54 gene in sporadic meningiomas with deletion at 1p32. Mol. Carcinog. 1999, 24, 300–304. [Google Scholar] [CrossRef]
  158. Lomas, J.; Bello, M.J.; Arjona, D.; Alonso, M.E.; Martinez-Glez, V.; Lopez-Marin, I.; Aminoso, C.; de Campos, J.M.; Isla, A.; Vaquero, J.; et al. Genetic and epigenetic alteration of the NF2 gene in sporadic meningiomas. Genes. Chromosomes Cancer 2005, 42, 314–319. [Google Scholar] [CrossRef] [PubMed]
  159. Piaskowski, S.; Rieske, P.; Szybka, M.; Wozniak, K.; Bednarek, A.; Pluciennik, E.; Jaskolski, D.; Sikorska, B.; Liberski, P.P. GADD45A and EPB41 as tumor suppressor genes in meningioma pathogenesis. Cancer Genet. Cytogenet. 2005, 162, 63–67. [Google Scholar] [CrossRef] [PubMed]
  160. Lusis, E.A.; Watson, M.A.; Chicoine, M.R.; Lyman, M.; Roerig, P.; Reifenberger, G.; Gutmann, D.H.; Perry, A. Integrative genomic analysis identifies NDRG2 as a candidate tumor suppressor gene frequently inactivated in clinically aggressive meningioma. Cancer Res. 2005, 65, 7121–7126. [Google Scholar] [CrossRef]
  161. Zhang, X.; Gejman, R.; Mahta, A.; Zhong, Y.; Rice, K.A.; Zhou, Y.; Cheunsuchon, P.; Louis, D.N.; Klibanski, A. Maternally expressed gene 3, an imprinted noncoding RNA gene, is associated with meningioma pathogenesis and progression. Cancer Res. 2010, 70, 2350–2358. [Google Scholar] [CrossRef]
  162. Buschges, R.; Ichimura, K.; Weber, R.G.; Reifenberger, G.; Collins, V.P. Allelic gain and amplification on the long arm of chromosome 17 in anaplastic meningiomas. Brain Pathol. 2002, 12, 145–153. [Google Scholar] [CrossRef]
  163. Abedalthagafi, M.; Bi, W.L.; Aizer, A.A.; Merrill, P.H.; Brewster, R.; Agarwalla, P.K.; Listewnik, M.L.; Dias-Santagata, D.; Thorner, A.R.; Van Hummelen, P.; et al. Oncogenic PI3K mutations are as common as AKT1 and SMO mutations in meningioma. Neuro Oncol. 2016, 18, 649–655. [Google Scholar] [CrossRef]
  164. Clark, V.E.; Erson-Omay, E.Z.; Serin, A.; Yin, J.; Cotney, J.; Ozduman, K.; Avsar, T.; Li, J.; Murray, P.B.; Henegariu, O.; et al. Genomic analysis of non-NF2 meningiomas reveals mutations in TRAF7, KLF4, AKT1, and SMO. Science 2013, 339, 1077–1080. [Google Scholar] [CrossRef]
  165. Yuzawa, S.; Nishihara, H.; Tanaka, S. Genetic landscape of meningioma. Brain Tumor Pathol. 2016, 33, 237–247. [Google Scholar] [CrossRef] [PubMed]
  166. Brastianos, P.K.; Horowitz, P.M.; Santagata, S.; Jones, R.T.; McKenna, A.; Getz, G.; Ligon, K.L.; Palescandolo, E.; Van Hummelen, P.; Ducar, M.D.; et al. Genomic sequencing of meningiomas identifies oncogenic SMO and AKT1 mutations. Nat. Genet. 2013, 45, 285–289. [Google Scholar] [CrossRef]
  167. Baia, G.S.; Stifani, S.; Kimura, E.T.; McDermott, M.W.; Pieper, R.O.; Lal, A. Notch activation is associated with tetraploidy and enhanced chromosomal instability in meningiomas. Neoplasia 2008, 10, 604–612. [Google Scholar] [CrossRef] [PubMed]
  168. Basu, S.; Totty, N.F.; Irwin, M.S.; Sudol, M.; Downward, J. Akt phosphorylates the Yes-associated protein, YAP, to induce interaction with 14-3-3 and attenuation of p73-mediated apoptosis. Mol. Cell 2003, 11, 11–23. [Google Scholar] [CrossRef] [PubMed]
  169. Juratli, T.A.; McCabe, D.; Nayyar, N.; Williams, E.A.; Silverman, I.M.; Tummala, S.S.; Fink, A.L.; Baig, A.; Martinez-Lage, M.; Selig, M.K.; et al. DMD genomic deletions characterize a subset of progressive/higher-grade meningiomas with poor outcome. Acta Neuropathol. 2018, 136, 779–792. [Google Scholar] [CrossRef]
  170. Lino, M.M.; Merlo, A. PI3Kinase signaling in glioblastoma. J. Neurooncol. 2011, 103, 417–427. [Google Scholar] [CrossRef]
  171. Loo, E.; Khalili, P.; Beuhler, K.; Siddiqi, I.; Vasef, M.A. BRAF V600E Mutation Across Multiple Tumor Types: Correlation Between DNA-based Sequencing and Mutation-specific Immunohistochemistry. Appl. Immunohistochem. Mol. Morphol. 2018, 26, 709–713. [Google Scholar] [CrossRef]
  172. Perry, A.; Banerjee, R.; Lohse, C.M.; Kleinschmidt-DeMasters, B.K.; Scheithauer, B.W. A role for chromosome 9p21 deletions in the malignant progression of meningiomas and the prognosis of anaplastic meningiomas. Brain Pathol. 2002, 12, 183–190. [Google Scholar] [CrossRef]
  173. Schmitz, U.; Mueller, W.; Weber, M.; Sevenet, N.; Delattre, O.; von Deimling, A. INI1 mutations in meningiomas at a potential hotspot in exon 9. Br. J. Cancer 2001, 84, 199–201. [Google Scholar] [CrossRef]
  174. Yang, H.W.; Kim, T.M.; Song, S.S.; Shrinath, N.; Park, R.; Kalamarides, M.; Park, P.J.; Black, P.M.; Carroll, R.S.; Johnson, M.D. Alternative splicing of CHEK2 and codeletion with NF2 promote chromosomal instability in meningioma. Neoplasia 2012, 14, 20–28. [Google Scholar] [CrossRef]
  175. Maier, A.D.; Stenman, A.; Svahn, F.; Mirian, C.; Bartek, J., Jr.; Juhler, M.; Zedenius, J.; Broholm, H.; Mathiesen, T. TERT promoter mutations in primary and secondary WHO grade III meningioma. Brain Pathol. 2021, 31, 61–69. [Google Scholar] [CrossRef] [PubMed]
  176. Goutagny, S.; Nault, J.C.; Mallet, M.; Henin, D.; Rossi, J.Z.; Kalamarides, M. High incidence of activating TERT promoter mutations in meningiomas undergoing malignant progression. Brain Pathol. 2014, 24, 184–189. [Google Scholar] [CrossRef] [PubMed]
  177. Koelsche, C.; Sahm, F.; Capper, D.; Reuss, D.; Sturm, D.; Jones, D.T.; Kool, M.; Northcott, P.A.; Wiestler, B.; Bohmer, K.; et al. Distribution of TERT promoter mutations in pediatric and adult tumors of the nervous system. Acta Neuropathol. 2013, 126, 907–915. [Google Scholar] [CrossRef] [PubMed]
  178. Sahm, F.; Schrimpf, D.; Stichel, D.; Jones, D.T.W.; Hielscher, T.; Schefzyk, S.; Okonechnikov, K.; Koelsche, C.; Reuss, D.E.; Capper, D.; et al. DNA methylation-based classification and grading system for meningioma: A multicentre, retrospective analysis. Lancet Oncol. 2017, 18, 682–694. [Google Scholar] [CrossRef] [PubMed]
  179. Slavik, H.; Balik, V.; Kokas, F.Z.; Slavkovsky, R.; Vrbkova, J.; Rehulkova, A.; Lausova, T.; Ehrmann, J.; Gurska, S.; Uberall, I.; et al. Transcriptomic Profiling Revealed Lnc-GOLGA6A-1 as a Novel Prognostic Biomarker of Meningioma Recurrence. Neurosurgery 2022, 91, 360–369. [Google Scholar] [CrossRef] [PubMed]
  180. Rath, P.; Miller, D.C.; Litofsky, N.S.; Anthony, D.C.; Feng, Q.; Franklin, C.; Pei, L.; Free, A.; Liu, J.; Ren, M.; et al. Isolation and characterization of a population of stem-like progenitor cells from an atypical meningioma. Exp. Mol. Pathol. 2011, 90, 179–188. [Google Scholar] [CrossRef] [PubMed]
  181. Peyre, M.; Kalamarides, M. Molecular genetics of meningiomas: Building the roadmap towards personalized therapy. Neurochirurgie 2018, 64, 22–28. [Google Scholar] [CrossRef]
  182. Suppiah, S.; Nassiri, F.; Bi, W.L.; Dunn, I.F.; Hanemann, C.O.; Horbinski, C.M.; Hashizume, R.; James, C.D.; Mawrin, C.; Noushmehr, H.; et al. Molecular and translational advances in meningiomas. Neuro Oncol. 2019, 21, i4–i17. [Google Scholar] [CrossRef]
  183. Dickinson, P.J.; Surace, E.I.; Cambell, M.; Higgins, R.J.; Leutenegger, C.M.; Bollen, A.W.; LeCouteur, R.A.; Gutmann, D.H. Expression of the tumor suppressor genes NF2, 4.1B, and TSLC1 in canine meningiomas. Vet. Pathol. 2009, 46, 884–892. [Google Scholar] [CrossRef]
  184. Platt, S.R.; Scase, T.J.; Adams, V.; Wieczorek, L.; Miller, J.; Adamo, F.; Long, S. Vascular endothelial growth factor expression in canine intracranial meningiomas and association with patient survival. J. Vet. Intern. Med. 2006, 20, 663–668. [Google Scholar] [CrossRef]
  185. Mandara, M.T.; Pavone, S.; Mandrioli, L.; Bettini, G.; Falzone, C.; Baroni, M. Matrix metalloproteinase-2 and matrix metalloproteinase-9 expression in canine and feline meningioma. Vet. Pathol. 2009, 46, 836–845. [Google Scholar] [CrossRef] [PubMed]
  186. Foiani, G.; Guelfi, G.; Chiaradia, E.; Mancini, F.; Trivelli, C.; Vitellozzi, G.; Lepri, E.; Mandara, M.T. Somatostatin Receptor 2 Expression in Canine Meningioma. J. Comp. Pathol. 2019, 166, 59–68. [Google Scholar] [CrossRef] [PubMed]
  187. Dickinson, P.J.; Roberts, B.N.; Higgins, R.J.; Leutenegger, C.M.; Bollen, A.W.; Kass, P.H.; LeCouteur, R.A. Expression of receptor tyrosine kinases VEGFR-1 (FLT-1), VEGFR-2 (KDR), EGFR-1, PDGFRalpha and c-Met in canine primary brain tumours. Vet. Comp. Oncol. 2006, 4, 132–140. [Google Scholar] [CrossRef] [PubMed]
  188. Pierce, J.T.; Cho, S.S.; Nag, S.; Zeh, R.; Jeon, J.; Holt, D.; Durham, A.; Nasrallah, M.P.; Singhal, S.; Lee, J.Y.K. Folate Receptor Overexpression in Human and Canine Meningiomas-Immunohistochemistry and Case Report of Intraoperative Molecular Imaging. Neurosurgery 2019, 85, 359–368. [Google Scholar] [CrossRef] [PubMed]
  189. Grenier, J.K.; Foureman, P.A.; Sloma, E.A.; Miller, A.D. RNA-seq transcriptome analysis of formalin fixed, paraffin-embedded canine meningioma. PLoS ONE 2017, 12, e0187150. [Google Scholar] [CrossRef] [PubMed]
  190. He, S.; Pham, M.H.; Pease, M.; Zada, G.; Giannotta, S.L.; Wang, K.; Mack, W.J. A review of epigenetic and gene expression alterations associated with intracranial meningiomas. Neurosurg. Focus 2013, 35, E5. [Google Scholar] [CrossRef]
  191. Galani, V.; Lampri, E.; Varouktsi, A.; Alexiou, G.; Mitselou, A.; Kyritsis, A.P. Genetic and epigenetic alterations in meningiomas. Clin. Neurol. Neurosurg. 2017, 158, 119–125. [Google Scholar] [CrossRef] [PubMed]
  192. Barski, D.; Wolter, M.; Reifenberger, G.; Riemenschneider, M.J. Hypermethylation and transcriptional downregulation of the TIMP3 gene is associated with allelic loss on 22q12.3 and malignancy in meningiomas. Brain Pathol. 2010, 20, 623–631. [Google Scholar] [CrossRef]
  193. Lyu, Y.; Lou, J.; Yang, Y.; Feng, J.; Hao, Y.; Huang, S.; Yin, L.; Xu, J.; Huang, D.; Ma, B.; et al. Dysfunction of the WT1-MEG3 signaling promotes AML leukemogenesis via p53-dependent and -independent pathways. Leukemia 2017, 31, 2543–2551. [Google Scholar] [CrossRef]
  194. Nakane, Y.; Natsume, A.; Wakabayashi, T.; Oi, S.; Ito, M.; Inao, S.; Saito, K.; Yoshida, J. Malignant transformation-related genes in meningiomas: Allelic loss on 1p36 and methylation status of p73 and RASSF1A. J. Neurosurg. 2007, 107, 398–404. [Google Scholar] [CrossRef]
  195. Courtay-Cahen, C.; Platt, S.R.; De Risio, L.; Starkey, M.P. Preliminary analysis of genomic abnormalities in canine meningiomas. Vet. Comp. Oncol. 2008, 6, 182–192. [Google Scholar] [CrossRef] [PubMed]
  196. Du, Z.; Brewster, R.; Merrill, P.H.; Chmielecki, J.; Francis, J.; Aizer, A.; Abedalthagafi, M.; Sholl, L.M.; Geffers, L.; Alexander, B.; et al. Meningioma transcription factors link cell lineage with systemic metabolic cues. Neuro Oncol. 2018, 20, 1331–1343. [Google Scholar] [CrossRef] [PubMed]
  197. Riva, P.; Larizza, L. Expression of c-sis and c-fos genes in human meningiomas and neurinomas. Int. J. Cancer 1992, 51, 873–877. [Google Scholar] [CrossRef] [PubMed]
  198. Aarhus, M.; Bruland, O.; Bredholt, G.; Lybaek, H.; Husebye, E.S.; Krossnes, B.K.; Vedeler, C.; Wester, K.; Lund-Johansen, M.; Knappskog, P.M. Microarray analysis reveals down-regulation of the tumour suppressor gene WWOX and up-regulation of the oncogene TYMS in intracranial sporadic meningiomas. J. Neurooncol. 2008, 88, 251–259. [Google Scholar] [CrossRef] [PubMed]
  199. Wang, A.Z.; Bowman-Kirigin, J.A.; Desai, R.; Kang, L.I.; Patel, P.R.; Patel, B.; Khan, S.M.; Bender, D.; Marlin, M.C.; Liu, J.; et al. Single-cell profiling of human dura and meningioma reveals cellular meningeal landscape and insights into meningioma immune response. Genome Med. 2022, 14, 49. [Google Scholar] [CrossRef]
  200. Medici, G.; Freudenmann, L.K.; Velz, J.; Wang, S.S.; Kapolou, K.; Paramasivam, N.; Mühlenbruch, L.; Kowalewski, D.J.; Vasella, F.; Bilich, T.; et al. A T-cell antigen atlas for meningioma: Novel options for immunotherapy. Acta Neuropathol. 2023, 146, 173–190. [Google Scholar] [CrossRef]
  201. Shintaku, M.; Honda, T.; Sakai, T. Expression of podoplanin and calretinin in meningioma: An immunohistochemical study. Brain Tumor Pathol. 2010, 27, 23–27. [Google Scholar] [CrossRef]
  202. Maier, A.D.; Meddis, A.; Mirian, C.; Haslund-Vinding, J.; Bartek, J.; Krog, S.M.; Nguyen, T.U.P.; Areškevičiūtė, A.; Melchior, L.C.; Heegaard, S.; et al. Gene expression analysis during progression of malignant meningioma compared to benign meningioma. J. Neurosurg. 2023, 138, 1302–1312. [Google Scholar] [CrossRef]
  203. Kazumoto, K.; Tamura, M.; Hoshino, H.; Yuasa, Y. Enhanced expression of the sis and c-myc oncogenes in human meningiomas. J. Neurosurg. 1990, 72, 786–791. [Google Scholar] [CrossRef]
  204. Ogashiwa, M.; Nishiyama, F.; Fukai, K.; Takeuchi, K.; Hirano, H. Cytochemical study of Mg2(+)-ATPase and ALPase activity in human meningiomas. No Shinkei 1990, 42, 497–503. [Google Scholar]
  205. Zhang, L.; Wang, L.; Tan, Y.; Li, C.; Fang, C. Identification of key genes of anti-programmed death ligand 1 for meningioma immunotherapy by bioinformatic analysis. Med. Oncol. 2022, 40, 54. [Google Scholar] [CrossRef] [PubMed]
  206. Sahm, F.; Bissel, J.; Koelsche, C.; Schweizer, L.; Capper, D.; Reuss, D.; Böhmer, K.; Lass, U.; Göck, T.; Kalis, K.; et al. AKT1E17K mutations cluster with meningothelial and transitional meningiomas and can be detected by SFRP1 immunohistochemistry. Acta Neuropathol. 2013, 126, 757–762. [Google Scholar] [CrossRef] [PubMed]
  207. Serna, E.; Morales, J.M.; Mata, M.; Gonzalez-Darder, J.; San Miguel, T.; Gil-Benso, R.; Lopez-Gines, C.; Cerda-Nicolas, M.; Monleon, D. Gene expression profiles of metabolic aggressiveness and tumor recurrence in benign meningioma. PLoS ONE 2013, 8, e67291. [Google Scholar] [CrossRef] [PubMed]
  208. Tabernero, M.D.; Maillo, A.; Gil-Bellosta, C.J.; Castrillo, A.; Sousa, P.; Merino, M.; Orfao, A. Gene expression profiles of meningiomas are associated with tumor cytogenetics and patient outcome. Brain Pathol. 2009, 19, 409–420. [Google Scholar] [CrossRef] [PubMed]
  209. Marton, E.; Feletti, A.; Basaldella, L.; Dei Tos, A.P.; Bendini, M.; Longatti, P. Atypical cystic meningioma overexpressing AQP1 in early infancy: Case report with literature review. Acta Paediatr. 2008, 97, 1145–1149. [Google Scholar] [CrossRef] [PubMed]
  210. Saini, M.; Jha, A.N.; Abrari, A.; Ali, S. Expression of proto-oncogene KIT is up-regulated in subset of human meningiomas. BMC Cancer 2012, 12, 212. [Google Scholar] [CrossRef] [PubMed]
  211. Atukeren, P.; Turk, O.; Yanar, K.; Kemerdere, R.; Sayyahmelli, S.; Eren, B.; Tanriverdi, T. Evaluation of ALCAM, PECAM-1 and selectin levels in intracranial meningiomas. Clin. Neurol. Neurosurg. 2017, 160, 21–26. [Google Scholar] [CrossRef] [PubMed]
  212. Schulten, H.J.; Hussein, D. Array expression meta-analysis of cancer stem cell genes identifies upregulation of PODXL especially in DCC low expression meningiomas. PLoS ONE 2019, 14, e0215452. [Google Scholar] [CrossRef]
  213. Zhang, H.; Qi, L.; Du, Y.; Huang, L.F.; Braun, F.K.; Kogiso, M.; Zhao, Y.; Li, C.; Lindsay, H.; Zhao, S.; et al. Patient-Derived Orthotopic Xenograft (PDOX) Mouse Models of Primary and Recurrent Meningioma. Cancers 2020, 12, 1478. [Google Scholar] [CrossRef]
  214. Choudhury, A.; Cady, M.A.; Lucas, C.G.; Najem, H.; Phillips, J.J.; Palikuqi, B.; Zakimi, N.; Joseph, T.; Birrueta, J.O.; Chen, W.C.; et al. NOTCH3 drives meningioma tumorigenesis and resistance to radiotherapy. bioRxiv 2023. [Google Scholar] [CrossRef]
  215. Sofela, A.A.; Hilton, D.A.; Ammoun, S.; Baiz, D.; Adams, C.L.; Ercolano, E.; Jenkinson, M.D.; Kurian, K.M.; Teo, M.; Whitfield, P.C.; et al. Fibulin-2: A Novel Biomarker for Differentiating Grade II from Grade I Meningiomas. Int. J. Mol. Sci. 2021, 22, 560. [Google Scholar] [CrossRef] [PubMed]
  216. Fornelli, A.; Bacci, A.; Collina, G.; Eusebi, V. Breast carcinoma metastatic to meningioma: Review of the literature and description of 2 new cases. Pathologica 1995, 87, 506–512. [Google Scholar] [PubMed]
  217. Sulman, E.P.; White, P.S.; Brodeur, G.M. Genomic annotation of the meningioma tumor suppressor locus on chromosome 1p34. Oncogene 2004, 23, 1014–1020. [Google Scholar] [CrossRef] [PubMed]
  218. Nordqvist, A.C.; Mathiesen, T. Expression of IGF-II, IGFBP-2, -5, and -6 in meningiomas with different brain invasiveness. J. Neurooncol. 2002, 57, 19–26. [Google Scholar] [CrossRef] [PubMed]
  219. Hilton, D.A.; Shivane, A.; Kirk, L.; Bassiri, K.; Enki, D.G.; Hanemann, C.O. Activation of multiple growth factor signalling pathways is frequent in meningiomas. Neuropathology 2016, 36, 250–261. [Google Scholar] [CrossRef] [PubMed]
  220. Mukherjee, S.; Biswas, D.; Gadre, R.; Jain, P.; Syed, N.; Stylianou, J.; Zeng, Q.; Mahadevan, A.; Epari, S.; Shetty, P.; et al. Comprehending Meningioma Signaling Cascades Using Multipronged Proteomics Approaches & Targeted Validation of Potential Markers. Front. Oncol. 2020, 10, 1600. [Google Scholar] [CrossRef]
  221. Gill, C.M.; Loewenstern, J.; Rutland, J.W.; Arib, H.; Pain, M.; Umphlett, M.; Kinoshita, Y.; McBride, R.B.; Bederson, J.; Donovan, M.; et al. SWI/SNF chromatin remodeling complex alterations in meningioma. J. Cancer Res. Clin. Oncol. 2021, 147, 3431–3440. [Google Scholar] [CrossRef]
  222. Iglesias Gómez, J.C.; Mosquera Orgueira, A. An integrative analysis of meningioma tumors reveals the determinant genes and pathways of malignant transformation. Front. Oncol. 2014, 4, 147. [Google Scholar] [CrossRef]
  223. Carroll, R.S.; Zhang, J.; Dashner, K.; Black, P.M. Progesterone and glucocorticoid receptor activation in meningiomas. Neurosurgery 1995, 37, 92–97. [Google Scholar] [CrossRef]
  224. Black, P.; Carroll, R.; Zhang, J. The molecular biology of hormone and growth factor receptors in meningiomas. Acta Neurochir. Suppl. 1996, 65, 50–53. [Google Scholar] [CrossRef]
  225. Hsu, D.W.; Efird, J.T.; Hedley-Whyte, E.T. Progesterone and estrogen receptors in meningiomas: Prognostic considerations. J. Neurosurg. 1997, 86, 113–120. [Google Scholar] [CrossRef] [PubMed]
  226. Briscoe, K.; Barrs, V.R.; Foster, D.F.; Beatty, J.A. Hyperaldosteronism and hyperprogesteronism in a cat. J. Feline Med. Surg. 2009, 11, 758–762. [Google Scholar] [CrossRef] [PubMed]
  227. DeClue, A.E.; Breshears, L.A.; Pardo, I.D.; Kerl, M.E.; Perlis, J.; Cohn, L.A. Hyperaldosteronism and hyperprogesteronism in a cat with an adrenal cortical carcinoma. J. Vet. Intern. Med. 2005, 19, 355–358. [Google Scholar] [CrossRef] [PubMed]
  228. Shayanfar, N.; Mashayekh, M.; Mohammadpour, M. Expression of progestrone receptor and proliferative marker ki 67 in various grades of meningioma. Acta Med. Iran. 2010, 48, 142–147. [Google Scholar]
  229. Maiuri, F.; Mariniello, G.; de Divitiis, O.; Esposito, F.; Guadagno, E.; Teodonno, G.; Barbato, M.; Del Basso De Caro, M. Progesterone Receptor Expression in Meningiomas: Pathological and Prognostic Implications. Front. Oncol. 2021, 11, 611218. [Google Scholar] [CrossRef]
  230. von Randow, A.J.; Schindler, S.; Tews, D.S. Expression of extracellular matrix-degrading proteins in classic, atypical, and anaplastic meningiomas. Pathol. Res. Pract. 2006, 202, 365–372. [Google Scholar] [CrossRef]
  231. Barresi, V.; Vitarelli, E.; Tuccari, G.; Barresi, G. MMP-9 expression in meningiomas: A prognostic marker for recurrence risk? J. Neurooncol. 2011, 102, 189–196. [Google Scholar] [CrossRef]
  232. Pei, J.; Jung, S.; Jin, S.G.; Moon, K.S.; Wen, M.; Li, S.Y.; Jang, W.Y.; Ryu, H.H.; Lee, K.H.; Kim, I.Y.; et al. Possible role of matrix metalloproteinases (MMPs) in hyperostosis of intracranial meningiomas. Acta Neurochir. 2012, 154, 611–620; discussion 620. [Google Scholar] [CrossRef]
  233. Iwado, E.; Ichikawa, T.; Kosaka, H.; Otsuka, S.; Kambara, H.; Tamiya, T.; Kondo, S.; Date, I. Role of VEGF and matrix metalloproteinase-9 in peritumoral brain edema associated with supratentorial benign meningiomas. Neuropathology 2012, 32, 638–646. [Google Scholar] [CrossRef]
  234. Mandara, M.T.; Reginato, A.; Foiani, G.; De Luca, S.; Guelfi, G. Gene Expression of Matrix Metalloproteinases and their Inhibitors (TIMPs) in Meningiomas of Dogs. J. Vet. Intern. Med. 2017, 31, 1816–1821. [Google Scholar] [CrossRef]
  235. Hicks, J.; Platt, S.; Kent, M.; Haley, A. Canine brain tumours: A model for the human disease? Vet. Comp. Oncol. 2017, 15, 252–272. [Google Scholar] [CrossRef] [PubMed]
  236. Samarani, F.; de la Fuente, C.; Parodi, A.; Mandara, M.T.; Pumarola, M.; Anor, S. Immunohistochemical expression of cyclooxygenase-2 (COX-2) is not associated with tumor grade in feline meningiomas. Vet. J. 2018, 241, 20–23. [Google Scholar] [CrossRef] [PubMed]
  237. Lin, C.C.; Kenyon, L.; Hyslop, T.; Hammond, E.; Andrews, D.W.; Curran, W.J., Jr.; Dicker, A.P. Cyclooxygenase-2 (COX-2) expression in human meningioma as a function of tumor grade. Am. J. Clin. Oncol. 2003, 26, S98–S102. [Google Scholar] [CrossRef] [PubMed]
  238. Preusser, M.; Hassler, M.; Birner, P.; Rudas, M.; Acker, T.; Plate, K.H.; Widhalm, G.; Knosp, E.; Breitschopf, H.; Berger, J.; et al. Microvascularization and expression of VEGF and its receptors in recurring meningiomas: Pathobiological data in favor of anti-angiogenic therapy approaches. Clin. Neuropathol. 2012, 31, 352–360. [Google Scholar] [CrossRef] [PubMed]
  239. Tabernero, M.; Jara-Acevedo, M.; Nieto, A.B.; Caballero, A.R.; Otero, A.; Sousa, P.; Gonçalves, J.; Domingues, P.H.; Orfao, A. Association between mutation of the NF2 gene and monosomy 22 in menopausal women with sporadic meningiomas. BMC Med. Genet. 2013, 14, 114. [Google Scholar] [CrossRef]
  240. Shimada, S.; Ishizawa, K.; Hirose, T. Expression of E-cadherin and catenins in meningioma: Ubiquitous expression and its irrelevance to malignancy. Pathol. Int. 2005, 55, 1–7. [Google Scholar] [CrossRef]
  241. Boozer, L.B.; Davis, T.W.; Borst, L.B.; Zseltvay, K.M.; Olby, N.J.; Mariani, C.L. Characterization of immune cell infiltration into canine intracranial meningiomas. Vet. Pathol. 2012, 49, 784–795. [Google Scholar] [CrossRef]
  242. Rossi, M.L.; Cruz Sanchez, F.; Hughes, J.T.; Esiri, M.M.; Coakham, H.B. Immunocytochemical study of the cellular immune response in meningiomas. J. Clin. Pathol. 1988, 41, 314–319. [Google Scholar] [CrossRef]
  243. Asai, J.; Suzuki, R.; Fujimoto, T.; Suzuki, T.; Nakagawa, N.; Nagashima, G.; Miyo, T.; Hokaku, H.; Takei, A. Fluorescence automatic cell sorter and immunohistochemical investigation of CD68-positive cells in meningioma. Clin. Neurol. Neurosurg. 1999, 101, 229–234. [Google Scholar] [CrossRef]
  244. Stavrou, D.; Anzil, A.P.; Weidenbach, W.; Rodt, H. Immunofluorescence study of lymphocytic infiltration in gliomas. Identification of T-lymphocytes. J. Neurol. Sci. 1977, 33, 275–282. [Google Scholar] [CrossRef]
  245. Bo, L.; Mork, S.J.; Nyland, H. An immunohistochemical study of mononuclear cells in meningiomas. Neuropathol. Appl. Neurobiol. 1992, 18, 548–558. [Google Scholar] [CrossRef]
  246. Fliss, M.S.; Usadel, H.; Caballero, O.L.; Wu, L.; Buta, M.R.; Eleff, S.M.; Jen, J.; Sidransky, D. Facile detection of mitochondrial DNA mutations in tumors and bodily fluids. Science 2000, 287, 2017–2019. [Google Scholar] [CrossRef]
  247. Habano, W.; Nakamura, S.; Sugai, T. Microsatellite instability in the mitochondrial DNA of colorectal carcinomas: Evidence for mismatch repair systems in mitochondrial genome. Oncogene 1998, 17, 1931–1937. [Google Scholar] [CrossRef] [PubMed]
  248. Ksiezakowska-Lakoma, K.; Zyla, M.; Wilczynski, J.R. Mitochondrial dysfunction in cancer. Prz. Menopauzalny 2014, 13, 136–144. [Google Scholar] [CrossRef] [PubMed]
  249. Kirches, E.; Michael, M.; Woy, C.; Schneider, T.; Warich-Kirches, M.; Schneider-Stock, R.; Winkler, K.; Wittig, H.; Dietzmann, K. Loss of heteroplasmy in the displacement loop of brain mitochondrial DNA in astrocytic tumors. Genes. Chromosomes Cancer 1999, 26, 80–83. [Google Scholar] [CrossRef]
  250. Mohamed Yusoff, A.A.; Mohd Khair, S.Z.N.; Abd Radzak, S.M.; Idris, Z.; Lee, H.C. Prevalence of mitochondrial DNA common deletion in patients with gliomas and meningiomas: A first report from a Malaysian study group. J. Chin. Med. Assoc. 2020, 83, 838–844. [Google Scholar] [CrossRef] [PubMed]
  251. Vega, A.; Salas, A.; Gamborino, E.; Sobrido, M.J.; Macaulay, V.; Carracedo, A. mtDNA mutations in tumors of the central nervous system reflect the neutral evolution of mtDNA in populations. Oncogene 2004, 23, 1314–1320. [Google Scholar] [CrossRef] [PubMed]
  252. Feichtinger, R.G.; Weis, S.; Mayr, J.A.; Zimmermann, F.A.; Bogner, B.; Sperl, W.; Kofler, B. Alterations of oxidative phosphorylation in meningiomas and peripheral nerve sheath tumors. Neuro Oncol. 2016, 18, 184–194. [Google Scholar] [CrossRef]
  253. Ma, W.; Sung, H.J.; Park, J.Y.; Matoba, S.; Hwang, P.M. A pivotal role for p53: Balancing aerobic respiration and glycolysis. J. Bioenerg. Biomembr. 2007, 39, 243–246. [Google Scholar] [CrossRef]
  254. Blandino, G.; Valenti, F.; Sacconi, A.; Di Agostino, S. Wild type- and mutant p53 proteins in mitochondrial dysfunction: Emerging insights in cancer disease. Semin. Cell Dev. Biol. 2020, 98, 105–117. [Google Scholar] [CrossRef]
  255. Tkaczyk-Wlizlo, A.; Kowal, K.; Slaska, B. Mitochondrial DNA alterations in the domestic dog (Canis lupus familiaris) and their association with development of diseases: A review. Mitochondrion 2022, 63, 72–84. [Google Scholar] [CrossRef] [PubMed]
  256. Jackson, M.; Serada, N.; Sheehan, M.; Srinivasan, S.; Mason, N.; Guha, M.; Avadhani, N. Mitochondrial genome and functional defects in osteosarcoma are associated with their aggressive phenotype. PLoS ONE 2018, 13, e0209489. [Google Scholar] [CrossRef] [PubMed]
  257. Kowal, K.; Tkaczyk-Wlizlo, A.; Pierzchala, M.; Gawor, J.; Slaska, B. Molecular differences in mitochondrial DNA genomes of dogs with malignant mammary tumours. Vet. Comp. Oncol. 2022, 20, 256–264. [Google Scholar] [CrossRef]
  258. Smiech, A.; Slaska, B.; Surdyka, M.; Grzybowska-Szatkowska, L.; Lopuszynski, W.; Rozanska, D. Identification of additional mitochondrial DNA mutations in canine mast cell tumours. Acta Vet. Scand. 2016, 58, 28. [Google Scholar] [CrossRef] [PubMed]
  259. Cowland, J.B.; Hother, C.; Gronbaek, K. MicroRNAs and cancer. APMIS 2007, 115, 1090–1106. [Google Scholar] [CrossRef] [PubMed]
  260. Dalmay, T.; Edwards, D.R. MicroRNAs and the hallmarks of cancer. Oncogene 2006, 25, 6170–6175. [Google Scholar] [CrossRef]
  261. Saliminejad, K.; Khorram Khorshid, H.R.; Soleymani Fard, S.; Ghaffari, S.H. An overview of microRNAs: Biology, functions, therapeutics, and analysis methods. J. Cell Physiol. 2019, 234, 5451–5465. [Google Scholar] [CrossRef] [PubMed]
  262. Ruvkun, G. Clarifications on miRNA and cancer. Science 2006, 311, 36–37. [Google Scholar] [CrossRef]
  263. Peng, Y.; Croce, C.M. The role of MicroRNAs in human cancer. Signal Transduct. Target. Ther. 2016, 1, 15004. [Google Scholar] [CrossRef]
  264. Si, W.; Shen, J.; Zheng, H.; Fan, W. The role and mechanisms of action of microRNAs in cancer drug resistance. Clin. Epigenetics 2019, 11, 25. [Google Scholar] [CrossRef]
  265. Visone, R.; Croce, C.M. MiRNAs and cancer. Am. J. Pathol. 2009, 174, 1131–1138. [Google Scholar] [CrossRef] [PubMed]
  266. Medina, P.P.; Slack, F.J. microRNAs and cancer: An overview. Cell Cycle 2008, 7, 2485–2492. [Google Scholar] [CrossRef] [PubMed]
  267. Dalmay, T. MicroRNAs and cancer. J. Intern. Med. 2008, 263, 366–375. [Google Scholar] [CrossRef] [PubMed]
  268. El-Gewely, M.R.; Andreassen, M.; Walquist, M.; Ursvik, A.; Knutsen, E.; Nystad, M.; Coucheron, D.H.; Myrmel, K.S.; Hennig, R.; Johansen, S.D. Differentially Expressed MicroRNAs in Meningiomas Grades I and II Suggest Shared Biomarkers with Malignant Tumors. Cancers 2016, 8, 31. [Google Scholar] [CrossRef] [PubMed]
  269. Kopkova, A.; Sana, J.; Machackova, T.; Vecera, M.; Radova, L.; Trachtova, K.; Vybihal, V.; Smrcka, M.; Kazda, T.; Slaby, O.; et al. Cerebrospinal Fluid MicroRNA Signatures as Diagnostic Biomarkers in Brain Tumors. Cancers 2019, 11, 1546. [Google Scholar] [CrossRef]
  270. Zhi, F.; Shao, N.; Li, B.; Xue, L.; Deng, D.; Xu, Y.; Lan, Q.; Peng, Y.; Yang, Y. A serum 6-miRNA panel as a novel non-invasive biomarker for meningioma. Sci. Rep. 2016, 6, 32067. [Google Scholar] [CrossRef]
  271. Slavik, H.; Balik, V.; Vrbkova, J.; Rehulkova, A.; Vaverka, M.; Hrabalek, L.; Ehrmann, J.; Vidlarova, M.; Gurska, S.; Hajduch, M.; et al. Identification of Meningioma Patients at High Risk of Tumor Recurrence Using MicroRNA Profiling. Neurosurgery 2020, 87, 1055–1063. [Google Scholar] [CrossRef]
  272. Katar, S.; Baran, O.; Evran, S.; Cevik, S.; Akkaya, E.; Baran, G.; Antar, V.; Hanimoglu, H.; Kaynar, M.Y. Expression of miRNA-21, miRNA-107, miRNA-137 and miRNA-29b in meningioma. Clin. Neurol. Neurosurg. 2017, 156, 66–70. [Google Scholar] [CrossRef]
  273. Zhi, F.; Zhou, G.; Wang, S.; Shi, Y.; Peng, Y.; Shao, N.; Guan, W.; Qu, H.; Zhang, Y.; Wang, Q.; et al. A microRNA expression signature predicts meningioma recurrence. Int. J. Cancer 2013, 132, 128–136. [Google Scholar] [CrossRef]
  274. Dagli, H.; Gülbahar, Ö.; Saadet, T.; Bulut, D.; Şahin, M.Ç. Investıgatıon of the Dıagnostıc and Prognostıc Values of Some Specıfıc mıcroRNAs in Menıngıom Tumors. Res. Sq. 2022. [Google Scholar] [CrossRef]
  275. Urbschat, S.; Landau, B.; Bewersdorf, N.C.; Schuster, C.; Wagenpfeil, G.; Schulz-Schaeffer, W.J.; Oertel, J.; Ketter, R. MicroRNA 200a as a histologically independent marker for meningioma recurrence: Results of a four microRNA panel analysis in meningiomas. Cancer Med. 2022, 12, 8433–8444. [Google Scholar] [CrossRef] [PubMed]
  276. Song, L.R.; Li, D.; Weng, J.C.; Li, C.B.; Wang, L.; Wu, Z.; Zhang, J.T. MicroRNA-195 Functions as a Tumor Suppressor by Directly Targeting Fatty Acid Synthase in Malignant Meningioma. World Neurosurg. 2020, 136, e355–e364. [Google Scholar] [CrossRef] [PubMed]
  277. Foiani, G.; Guelfi, G.; Mandara, M.T. MicroRNA Dysregulation in Canine Meningioma: RT-qPCR Analysis of Formalin-Fixed Paraffin-Embedded Samples. J. Neuropathol. Exp. Neurol. 2021, 80, 769–775. [Google Scholar] [CrossRef]
  278. Sahabi, K.; Selvarajah, G.T.; Abdullah, R.; Cheah, Y.K.; Tan, G.C. Comparative aspects of microRNA expression in canine and human cancers. J. Vet. Sci. 2018, 19, 162–171. [Google Scholar] [CrossRef] [PubMed]
  279. Agarwal, P.; Crepps, M.P.; Stahr, N.A.; Kretzschmar, W.P.; Harris, H.C.; Prasad, N.; Levy, S.E.; Smith, B.F. Identification of canine circulating miRNAs as tumor biospecific markers using Next-Generation Sequencing and Q-RT-PCR. Biochem. Biophys. Rep. 2021, 28, 101106. [Google Scholar] [CrossRef] [PubMed]
  280. Wagner, S.; Willenbrock, S.; Nolte, I.; Murua Escobar, H. Comparison of non-coding RNAs in human and canine cancer. Front. Genet. 2013, 4, 46. [Google Scholar] [CrossRef]
  281. Kim, E.P.; Jang, G.; Kim, J.W.; Kim, S.W.; Chung, H.; Yang, Y.J.; Kim, W.H.; Kim, G.A. MicroRNA and Messenger RNA Expression Profiles in Canine Mammary Gland Tumor. Int. J. Mol. Sci. 2023, 24, 2618. [Google Scholar] [CrossRef]
  282. Varvil, M.S.; dos Santos, A.P. A review on microRNA detection and expression studies in dogs. Front. Vet. Sci. 2023, 10, 1261085. [Google Scholar] [CrossRef]
  283. Sarver, A.L.; Thayanithy, V.; Scott, M.C.; Cleton-Jansen, A.M.; Hogendoorn, P.C.; Modiano, J.F.; Subramanian, S. MicroRNAs at the human 14q32 locus have prognostic significance in osteosarcoma. Orphanet J. Rare Dis. 2013, 8, 7. [Google Scholar] [CrossRef]
  284. Albonico, F.; Mortarino, M.; Avallone, G.; Gioia, G.; Comazzi, S.; Roccabianca, P. The expression ratio of miR-17-5p and miR-155 correlates with grading in canine splenic lymphoma. Vet. Immunol. Immunopathol. 2013, 155, 117–123. [Google Scholar] [CrossRef]
  285. Craig, K.K.L.; Wood, G.A.; Keller, S.M.; Mutsaers, A.J.; Wood, R.D. MicroRNA profiling in canine multicentric lymphoma. PLoS ONE 2019, 14, e0226357. [Google Scholar] [CrossRef] [PubMed]
  286. Zamarian, V.; Stefanello, D.; Ferrari, R.; Chiti, L.E.; Grieco, V.; DallaCosta, E.; Ceciliani, F.; Lecchi, C. Salivary miR-21 is a potential biomarker for canine mast cell tumors. Vet. Pathol. 2023, 60, 47–51. [Google Scholar] [CrossRef] [PubMed]
  287. Ramadan, E.S.; Salem, N.Y.; Emam, I.A.; AbdElKader, N.A.; Farghali, H.A.; Khattab, M.S. MicroRNA-21 expression, serum tumor markers, and immunohistochemistry in canine mammary tumors. Vet. Res. Commun. 2022, 46, 377–388. [Google Scholar] [CrossRef] [PubMed]
  288. Fish, E.J.; Martinez-Romero, E.G.; DeInnocentes, P.; Koehler, J.W.; Prasad, N.; Smith, A.N.; Bird, R.C. Circulating microRNA as biomarkers of canine mammary carcinoma in dogs. J. Vet. Intern. Med. 2020, 34, 1282–1290. [Google Scholar] [CrossRef] [PubMed]
  289. Bulkowska, M.; Rybicka, A.; Senses, K.M.; Ulewicz, K.; Witt, K.; Szymanska, J.; Taciak, B.; Klopfleisch, R.; Hellmen, E.; Dolka, I.; et al. MicroRNA expression patterns in canine mammary cancer show significant differences between metastatic and non-metastatic tumours. BMC Cancer 2017, 17, 728. [Google Scholar] [CrossRef] [PubMed]
  290. Lopez, C.M.; Yu, P.Y.; Zhang, X.; Yilmaz, A.S.; London, C.A.; Fenger, J.M. MiR-34a regulates the invasive capacity of canine osteosarcoma cell lines. PLoS ONE 2018, 13, e0190086. [Google Scholar] [CrossRef] [PubMed]
  291. Elshafie, N.O.; Nascimento, N.C.D.; Lichti, N.I.; Kasinski, A.L.; Childress, M.O.; Santos, A.P.D. MicroRNA Biomarkers in Canine Diffuse Large B-Cell Lymphoma. Vet. Pathol. 2021, 58, 34–41. [Google Scholar] [CrossRef] [PubMed]
  292. Kang, L.; Mao, J.; Tao, Y.; Song, B.; Ma, W.; Lu, Y.; Zhao, L.; Li, J.; Yang, B.; Li, L. MicroRNA-34a suppresses the breast cancer stem cell-like characteristics by downregulating Notch1 pathway. Cancer Sci. 2015, 106, 700–708. [Google Scholar] [CrossRef]
  293. Ren, X.; Fan, Y.; Shi, D.; Xu, E.; Liu, Y. MicroRNA-124 inhibits canine mammary carcinoma cell proliferation, migration and invasion by targeting CDH2. Res. Vet. Sci. 2022, 146, 5–14. [Google Scholar] [CrossRef]
  294. Lujambio, A.; Ropero, S.; Ballestar, E.; Fraga, M.F.; Cerrato, C.; Setien, F.; Casado, S.; Suarez-Gauthier, A.; Sanchez-Cespedes, M.; Git, A.; et al. Genetic unmasking of an epigenetically silenced microRNA in human cancer cells. Cancer Res. 2007, 67, 1424–1429. [Google Scholar] [CrossRef]
  295. Noguchi, S.; Mori, T.; Hoshino, Y.; Yamada, N.; Nakagawa, T.; Sasaki, N.; Akao, Y.; Maruo, K. Comparative study of anti-oncogenic microRNA-145 in canine and human malignant melanoma. J. Vet. Med. Sci. 2012, 74, 1–8. [Google Scholar] [CrossRef] [PubMed]
  296. Ushio, N.; Rahman, M.M.; Maemura, T.; Lai, Y.C.; Iwanaga, T.; Kawaguchi, H.; Miyoshi, N.; Momoi, Y.; Miura, N. Identification of dysregulated microRNAs in canine malignant melanoma. Oncol. Lett. 2019, 17, 1080–1088. [Google Scholar] [CrossRef]
  297. Boggs, R.M.; Wright, Z.M.; Stickney, M.J.; Porter, W.W.; Murphy, K.E. MicroRNA expression in canine mammary cancer. Mamm. Genome 2008, 19, 561–569. [Google Scholar] [CrossRef] [PubMed]
  298. Yoshikawa, R.; Mori, T.; Noguchi, S.; Akao, Y.; Maruo, K.; Kitade, Y. Synthetic microRNA-205 exhibited tumour suppression in spontaneous canine malignant melanoma by intratumoral injection. Vet. Comp. Oncol. 2019, 17, 407–412. [Google Scholar] [CrossRef] [PubMed]
  299. Ferrari, E.; Gandellini, P. Unveiling the ups and downs of miR-205 in physiology and cancer: Transcriptional and post-transcriptional mechanisms. Cell Death Dis. 2020, 11, 980. [Google Scholar] [CrossRef] [PubMed]
  300. von Deetzen, M.C.; Schmeck, B.T.; Gruber, A.D.; Klopfleisch, R. Malignancy Associated MicroRNA Expression Changes in Canine Mammary Cancer of Different Malignancies. ISRN Vet. Sci. 2014, 2014, 148597. [Google Scholar] [CrossRef] [PubMed]
  301. Petrouskova, P.; Hudakova, N.; Maloveska, M.; Humenik, F.; Cizkova, D. Non-Exosomal and Exosome-Derived miRNAs as Promising Biomarkers in Canine Mammary Cancer. Life 2022, 12, 524. [Google Scholar] [CrossRef]
  302. Kobayashi, M.; Saito, A.; Tanaka, Y.; Michishita, M.; Kobayashi, M.; Irimajiri, M.; Kaneda, T.; Ochiai, K.; Bonkobara, M.; Takahashi, K.; et al. MicroRNA expression profiling in canine prostate cancer. J. Vet. Med. Sci. 2017, 79, 719–725. [Google Scholar] [CrossRef]
  303. Simpson, D. The recurrence of intracranial meningiomas after surgical treatment. J. Neurol. Neurosurg. Psychiatry 1957, 20, 22–39. [Google Scholar] [CrossRef]
  304. Oya, S.; Kawai, K.; Nakatomi, H.; Saito, N. Significance of Simpson grading system in modern meningioma surgery: Integration of the grade with MIB-1 labeling index as a key to predict the recurrence of WHO Grade I meningiomas. J. Neurosurg. 2012, 117, 121–128. [Google Scholar] [CrossRef]
  305. Behling, F.; Fodi, C.; Gepfner-Tuma, I.; Machetanz, K.; Renovanz, M.; Skardelly, M.; Bornemann, A.; Honegger, J.; Tabatabai, G.; Tatagiba, M.; et al. CNS Invasion in Meningioma-How the Intraoperative Assessment Can Improve the Prognostic Evaluation of Tumor Recurrence. Cancers 2020, 12, 3620. [Google Scholar] [CrossRef] [PubMed]
  306. Nanda, A.; Bir, S.C.; Konar, S.; Maiti, T.; Kalakoti, P.; Jacobsohn, J.A.; Guthikonda, B. Outcome of resection of WHO Grade II meningioma and correlation of pathological and radiological predictive factors for recurrence. J. Clin. Neurosci. 2016, 31, 112–121. [Google Scholar] [CrossRef] [PubMed]
  307. Prabhu, V.C.; Melian, E.; Germanwala, A.V.; Solanki, A.A.; Borys, E.; Barton, K.; Anderson, D.E. Cranial Base Meningiomas. World Neurosurg. 2018, 109, 258–262. [Google Scholar] [CrossRef] [PubMed]
  308. Patibandla, M.R.; Lee, C.C.; Sheehan, J. Stereotactic Radiosurgery of Central Skull Base Meningiomas-Volumetric Evaluation and Long-Term Outcomes. World Neurosurg. 2017, 108, 176–184. [Google Scholar] [CrossRef] [PubMed]
  309. Mooney, M.A.; Essayed, W.; Patel, V.; Devlin, P.M.; Al-Mefty, O. Brachytherapy as Salvage Treatment for Meningioma With Malignant Progression After Exhausting Other Treatment Options: 2-Dimensional Operative Video. Oper. Neurosurg. 2022, 22, e215. [Google Scholar] [CrossRef]
  310. Walcott, B.P.; Nahed, B.V.; Brastianos, P.K.; Loeffler, J.S. Radiation Treatment for WHO Grade II and III Meningiomas. Front. Oncol. 2013, 3, 227. [Google Scholar] [CrossRef] [PubMed]
  311. Shahbandi, A.; Shah, D.S.; Hadley, C.C.; Patel, A.J. The Role of Pharmacotherapy in Treatment of Meningioma: A Systematic Review. Cancers 2023, 15, 483. [Google Scholar] [CrossRef]
  312. Pachow, D.; Andrae, N.; Kliese, N.; Angenstein, F.; Stork, O.; Wilisch-Neumann, A.; Kirches, E.; Mawrin, C. mTORC1 inhibitors suppress meningioma growth in mouse models. Clin. Cancer Res. 2013, 19, 1180–1189. [Google Scholar] [CrossRef]
  313. Mair, M.J.; Berghoff, A.S.; Brastianos, P.K.; Preusser, M. Emerging systemic treatment options in meningioma. J. Neurooncol 2023, 161, 245–258. [Google Scholar] [CrossRef]
  314. Sharma, S.; Rana, R.; Prakash, P.; Ganguly, N.K. Drug target therapy and emerging clinical relevance of exosomes in meningeal tumors. Mol. Cell Biochem. 2023, 1–44. [Google Scholar] [CrossRef]
  315. Drappatz, J. How useful is chemotherapy for atypical and anaplastic meningiomas? Expert Opin. Pharmacother. 2022, 23, 1559–1561. [Google Scholar] [CrossRef]
  316. Brastianos, P.K.; Galanis, E.; Butowski, N.; Chan, J.W.; Dunn, I.F.; Goldbrunner, R.; Herold-Mende, C.; Ippen, F.M.; Mawrin, C.; McDermott, M.W.; et al. Advances in multidisciplinary therapy for meningiomas. Neuro Oncol. 2019, 21, i18–i31. [Google Scholar] [CrossRef]
  317. Choy, W.; Kim, W.; Nagasawa, D.; Stramotas, S.; Yew, A.; Gopen, Q.; Parsa, A.T.; Yang, I. The molecular genetics and tumor pathogenesis of meningiomas and the future directions of meningioma treatments. Neurosurg. Focus 2011, 30, E6. [Google Scholar] [CrossRef]
  318. Lekanne Deprez, R.H.; Bianchi, A.B.; Groen, N.A.; Seizinger, B.R.; Hagemeijer, A.; van Drunen, E.; Bootsma, D.; Koper, J.W.; Avezaat, C.J.; Kley, N.; et al. Frequent NF2 gene transcript mutations in sporadic meningiomas and vestibular schwannomas. Am. J. Hum. Genet. 1994, 54, 1022–1029. [Google Scholar]
  319. Wellenreuther, R.; Kraus, J.A.; Lenartz, D.; Menon, A.G.; Schramm, J.; Louis, D.N.; Ramesh, V.; Gusella, J.F.; Wiestler, O.D.; von Deimling, A. Analysis of the neurofibromatosis 2 gene reveals molecular variants of meningioma. Am. J. Pathol. 1995, 146, 827–832. [Google Scholar]
  320. Nunes, F.P.; Merker, V.L.; Jennings, D.; Caruso, P.A.; di Tomaso, E.; Muzikansky, A.; Barker, F.G., II; Stemmer-Rachamimov, A.; Plotkin, S.R. Bevacizumab treatment for meningiomas in NF2: A retrospective analysis of 15 patients. PLoS ONE 2013, 8, e59941. [Google Scholar] [CrossRef]
  321. Lou, E.; Sumrall, A.L.; Turner, S.; Peters, K.B.; Desjardins, A.; Vredenburgh, J.J.; McLendon, R.E.; Herndon, J.E., II; McSherry, F.; Norfleet, J.; et al. Bevacizumab therapy for adults with recurrent/progressive meningioma: A retrospective series. J. Neurooncol. 2012, 109, 63–70. [Google Scholar] [CrossRef]
  322. Tumber, A.; Nuzzi, A.; Hookway, E.S.; Hatch, S.B.; Velupillai, S.; Johansson, C.; Kawamura, A.; Savitsky, P.; Yapp, C.; Szykowska, A.; et al. Potent and Selective KDM5 Inhibitor Stops Cellular Demethylation of H3K4me3 at Transcription Start Sites and Proliferation of MM1S Myeloma Cells. Cell Chem. Biol. 2017, 24, 371–380. [Google Scholar] [CrossRef]
  323. Olar, A.; Wani, K.M.; Wilson, C.D.; Zadeh, G.; DeMonte, F.; Jones, D.T.; Pfister, S.M.; Sulman, E.P.; Aldape, K.D. Global epigenetic profiling identifies methylation subgroups associated with recurrence-free survival in meningioma. Acta Neuropathol. 2017, 133, 431–444. [Google Scholar] [CrossRef]
  324. Vasudevan, H.N.; Braunstein, S.E.; Phillips, J.J.; Pekmezci, M.; Tomlin, B.A.; Wu, A.; Reis, G.F.; Magill, S.T.; Zhang, J.; Feng, F.Y.; et al. Comprehensive Molecular Profiling Identifies FOXM1 as a Key Transcription Factor for Meningioma Proliferation. Cell Rep. 2018, 22, 3672–3683. [Google Scholar] [CrossRef]
  325. Strickland, M.R.; Gill, C.M.; Nayyar, N.; D’Andrea, M.R.; Thiede, C.; Juratli, T.A.; Schackert, G.; Borger, D.R.; Santagata, S.; Frosch, M.P.; et al. Targeted sequencing of SMO and AKT1 in anterior skull base meningiomas. J. Neurosurg. 2017, 127, 438–444. [Google Scholar] [CrossRef]
  326. Silapunt, S.; Chen, L.; Migden, M.R. Hedgehog pathway inhibition in advanced basal cell carcinoma: Latest evidence and clinical usefulness. Ther. Adv. Med. Oncol. 2016, 8, 375–382. [Google Scholar] [CrossRef]
  327. Boetto, J.; Bielle, F.; Sanson, M.; Peyre, M.; Kalamarides, M. SMO mutation status defines a distinct and frequent molecular subgroup in olfactory groove meningiomas. Neuro Oncol. 2017, 19, 345–351. [Google Scholar] [CrossRef]
  328. Brastianos, P.K.; Twohy, E.L.; Gerstner, E.R.; Kaufmann, T.J.; Iafrate, A.J.; Lennerz, J.; Jeyapalan, S.; Piccioni, D.E.; Monga, V.; Fadul, C.E.; et al. Alliance A071401: Phase II Trial of Focal Adhesion Kinase Inhibition in Meningiomas with Somatic NF2 Mutations. J. Clin. Oncol. 2023, 41, 618–628. [Google Scholar] [CrossRef]
  329. Han, S.J.; Reis, G.; Kohanbash, G.; Shrivastav, S.; Magill, S.T.; Molinaro, A.M.; McDermott, M.W.; Theodosopoulos, P.V.; Aghi, M.K.; Berger, M.S.; et al. Expression and prognostic impact of immune modulatory molecule PD-L1 in meningioma. J. Neurooncol. 2016, 130, 543–552. [Google Scholar] [CrossRef]
  330. Garzon-Muvdi, T.; Bailey, D.D.; Pernik, M.N.; Pan, E. Basis for Immunotherapy for Treatment of Meningiomas. Front. Neurol. 2020, 11, 945. [Google Scholar] [CrossRef]
  331. Sharif, S.; Brennan, P.; Rawluk, D. Non-surgical treatment of meningioma: A case report and review. Br. J. Neurosurg. 1998, 12, 369–372. [Google Scholar] [CrossRef]
  332. Grunberg, S.M.; Weiss, M.H.; Spitz, I.M.; Ahmadi, J.; Sadun, A.; Russell, C.A.; Lucci, L.; Stevenson, L.L. Treatment of unresectable meningiomas with the antiprogesterone agent mifepristone. J. Neurosurg. 1991, 74, 861–866. [Google Scholar] [CrossRef]
  333. Andersen, L.; Friis, S.; Hallas, J.; Ravn, P.; Schroder, H.D.; Gaist, D. Hormone replacement therapy increases the risk of cranial meningioma. Eur. J. Cancer 2013, 49, 3303–3310. [Google Scholar] [CrossRef]
  334. Axlund, T.W.; McGlasson, M.L.; Smith, A.N. Surgery alone or in combination with radiation therapy for treatment of intracranial meningiomas in dogs: 31 cases (1989–2002). J. Am. Vet. Med. Assoc. 2002, 221, 1597–1600. [Google Scholar] [CrossRef]
  335. Nakaichi, M.; Taura, Y.; Nakama, S.; Takeuchi, A.; Matsunaga, N.; Ebe, K.; Amimoto, A. Primary brain tumors in two dogs treated by surgical resection in combination with postoperative radiation therapy. J. Vet. Med. Sci. 1996, 58, 773–775. [Google Scholar] [CrossRef]
  336. Brearley, M.J.; Jeffery, N.D.; Phillips, S.M.; Dennis, R. Hypofractionated radiation therapy of brain masses in dogs: A retrospective analysis of survival of 83 cases (1991–1996). J. Vet. Intern. Med. 1999, 13, 408–412. [Google Scholar] [CrossRef]
  337. Apra, C.; Peyre, M.; Kalamarides, M. Current treatment options for meningioma. Expert Rev. Neurother. 2018, 18, 241–249. [Google Scholar] [CrossRef]
  338. Berg, J. Canine meningiomas: Other approaches: Hydroxyurea and ultrasonic aspiration (CUSA). ACVIM Forum 2004, 340, 342. [Google Scholar]
  339. Marconato, L.; Bonfanti, U.; Fileccia, I. Unusual dermatological toxicity of hydroxyurea in two dogs with spontaneously occurring tumours. J. Small Anim. Pract. 2007, 48, 514–517. [Google Scholar] [CrossRef]
  340. Tamura, S.; Tamura, Y.; Ohoka, A.; Hasegawa, T.; Uchida, K. A canine case of skull base meningioma treated with hydroxyurea. J. Vet. Med. Sci. 2007, 69, 1313–1315. [Google Scholar] [CrossRef]
  341. Jung, D. Long-term Chemotherapy with Hydroxyurea in a Dog with Suspected Intracranial Meningioma. J. Vet. Clin. 2008, 25, 514–517. [Google Scholar]
  342. Jung, H.W.; Lee, H.C.; Kim, J.H.; Jang, H.M.; Moon, J.H.; Sur, J.H.; Ha, J.; Jung, D.I. Imatinib mesylate plus hydroxyurea chemotherapy for cerebellar meningioma in a Belgian Malinois dog. J. Vet. Med. Sci. 2014, 76, 1545–1548. [Google Scholar] [CrossRef]
  343. Wen, P.Y.; Yung, W.K.; Lamborn, K.R.; Norden, A.D.; Cloughesy, T.F.; Abrey, L.E.; Fine, H.A.; Chang, S.M.; Robins, H.I.; Fink, K.; et al. Phase II study of imatinib mesylate for recurrent meningiomas (North American Brain Tumor Consortium study 01–08). Neuro Oncol. 2009, 11, 853–860. [Google Scholar] [CrossRef]
  344. Reardon, D.A.; Norden, A.D.; Desjardins, A.; Vredenburgh, J.J.; Herndon, J.E., II; Coan, A.; Sampson, J.H.; Gururangan, S.; Peters, K.B.; McLendon, R.E.; et al. Phase II study of Gleevec(R) plus hydroxyurea (HU) in adults with progressive or recurrent meningioma. J. Neurooncol. 2012, 106, 409–415. [Google Scholar] [CrossRef]
  345. Chauvet, A.E.; Kesava, P.P.; Goh, C.S.; Badie, B. Selective intraarterial gene delivery into a canine meningioma. J. Neurosurg. 1998, 88, 870–873. [Google Scholar] [CrossRef]
  346. Andersen, B.M.; Pluhar, G.E.; Seiler, C.E.; Goulart, M.R.; SantaCruz, K.S.; Schutten, M.M.; Meints, J.P.; O’Sullivan, M.G.; Bentley, R.T.; Packer, R.A.; et al. Vaccination for invasive canine meningioma induces in situ production of antibodies capable of antibody-dependent cell-mediated cytotoxicity. Cancer Res. 2013, 73, 2987–2997. [Google Scholar] [CrossRef]
  347. Gallagher, J.G.; Berg, J.; Knowles, K.E.; Williams, L.L.; Bronson, R.T. Prognosis after surgical excision of cerebral meningiomas in cats: 17 cases (1986–1992). J. Am. Vet. Med. Assoc. 1993, 203, 1437–1440. [Google Scholar]
  348. Lawson, D.C. Cerebral meningioma in the cat: Diagnosis and surgical treatment in ten cases. J. Am. Anim. Hosp. Assoc. 1984, 20, 333–342. [Google Scholar]
  349. LeCouteur, R.A. Current concepts in the diagnosis and treatment of brain tumours in dogs and cats. J. Small Anim. Pract. 1999, 40, 411–416. [Google Scholar] [CrossRef]
  350. Yun, T.; Koo, Y.; Kim, H.; Lee, W.; Kim, S.; Jung, D.I.; Yang, M.P.; Kang, B.T. Case Report: Long-Term Chemotherapy with Hydroxyurea and Prednisolone in a Cat With a Meningioma: Correlation of FDG Uptake and Tumor Grade Assessed by Histopathology and Expression of Ki-67 and p53. Front. Vet. Sci. 2021, 8, 576839. [Google Scholar] [CrossRef]
  351. Coates, J.R.; Johnson, G.C. Nervous system neoplasia. In Cancer Management in Small Animal Practice; Elsevier: Amsterdam, The Netherlands, 2010. [Google Scholar]
  352. Foster, E.S.; Carrillo, J.M.; Patnaik, A.K. Clinical signs of tumors affecting the rostral cerebrum in 43 dogs. J. Vet. Intern. Med. 1988, 2, 71–74. [Google Scholar] [CrossRef]
  353. Lapointe, S.; Perry, A.; Butowski, N.A. Primary brain tumours in adults. Lancet 2018, 392, 432–446. [Google Scholar] [CrossRef]
  354. Solheim, O.; Jakola, A.S. Quality of life outcomes in meningioma surgery. Handb. Clin. Neurol. 2020, 170, 311–321. [Google Scholar] [CrossRef]
  355. Benz, L.S.; Wrensch, M.R.; Schildkraut, J.M.; Bondy, M.L.; Warren, J.L.; Wiemels, J.L.; Claus, E.B. Quality of life after surgery for intracranial meningioma. Cancer 2018, 124, 161–166. [Google Scholar] [CrossRef]
  356. Gardner, H.L.; Fenger, J.M.; London, C.A. Dogs as a Model for Cancer. Annu. Rev. Anim. Biosci. 2016, 4, 199–222. [Google Scholar] [CrossRef]
  357. Marconato, L.; Gelain, M.E.; Comazzi, S. The dog as a possible animal model for human non-Hodgkin lymphoma: A review. Hematol. Oncol. 2013, 31, 1–9. [Google Scholar] [CrossRef]
  358. Yu, J.; Chen, F.F.; Zhang, H.W.; Zhang, H.; Luo, S.P.; Huang, G.D.; Lin, F.; Lei, Y.; Luo, L. Comparative Analysis of the MRI Characteristics of Meningiomas According to the 2016 WHO Pathological Classification. Technol. Cancer Res. Treat. 2020, 19, 1533033820983287. [Google Scholar] [CrossRef]
  359. Wang, M.; Wang, Z.; Ren, P.; Zhang, X.; Liu, S. Meningioma with ring enhancement on MRI: A rare case report. BMC Med. Imaging 2021, 21, 22. [Google Scholar] [CrossRef]
  360. Yao, Y.; Xu, Y.; Liu, S.; Xue, F.; Wang, B.; Qin, S.; Sun, X.; He, J. Predicting the grade of meningiomas by clinical-radiological features: A comparison of precontrast and postcontrast MRI. Front. Oncol. 2022, 12, 1053089. [Google Scholar] [CrossRef]
  361. Watts, J.; Box, G.; Galvin, A.; Brotchie, P.; Trost, N.; Sutherland, T. Magnetic resonance imaging of meningiomas: A pictorial review. Insights Imaging 2014, 5, 113–122. [Google Scholar] [CrossRef]
  362. Sherman, W.J.; Raizer, J.J. Medical management of meningiomas. CNS Oncol. 2013, 2, 161–170. [Google Scholar] [CrossRef]
  363. Zhao, L.; Zhao, W.; Hou, Y.; Wen, C.; Wang, J.; Wu, P.; Guo, Z. Corrigendum: An Overview of Managements in Meningiomas. Front. Oncol. 2020, 10, 599431. [Google Scholar] [CrossRef]
  364. Preusser, M.; Berghoff, A.S.; Hottinger, A.F. High-grade meningiomas: New avenues for drug treatment? Curr. Opin. Neurol. 2013, 26, 708–715. [Google Scholar] [CrossRef]
  365. Ahsan, S.A.; Chendeb, K.; Profyris, C.; Teo, C.; Sughrue, M.E. Pharmacotherapeutic options for atypical meningiomas. Expert. Opin. Pharmacother. 2019, 20, 1831–1836. [Google Scholar] [CrossRef]
  366. Preusser, M.; Brastianos, P.K.; Mawrin, C. Advances in meningioma genetics: Novel therapeutic opportunities. Nat. Rev. Neurol. 2018, 14, 106–115. [Google Scholar] [CrossRef] [PubMed]
  367. Graillon, T.; Tabouret, E.; Chinot, O. Chemotherapy and targeted therapies for meningiomas: What is the evidence? Curr. Opin. Neurol. 2021, 34, 857–867. [Google Scholar] [CrossRef] [PubMed]
  368. Kyritsis, A.P. Chemotherapy for meningiomas. J. Neurooncol. 1996, 29, 269–272. [Google Scholar] [CrossRef] [PubMed]
  369. Moazzam, A.A.; Wagle, N.; Zada, G. Recent developments in chemotherapy for meningiomas: A review. Neurosurg. Focus 2013, 35, E18. [Google Scholar] [CrossRef] [PubMed]
  370. Sioka, C.; Kyritsis, A.P. Chemotherapy, hormonal therapy, and immunotherapy for recurrent meningiomas. J. Neurooncol. 2009, 92, 1–6. [Google Scholar] [CrossRef] [PubMed]
  371. Agopiantz, M.; Carnot, M.; Denis, C.; Martin, E.; Gauchotte, G. Hormone Receptor Expression in Meningiomas: A Systematic Review. Cancers 2023, 15, 980. [Google Scholar] [CrossRef] [PubMed]
  372. Carroll, R.S.; Zhang, J.; Black, P.M. Expression of estrogen receptors alpha and beta in human meningiomas. J. Neurooncol. 1999, 42, 109–116. [Google Scholar] [CrossRef]
  373. Blitshteyn, S.; Crook, J.E.; Jaeckle, K.A. Is there an association between meningioma and hormone replacement therapy? J. Clin. Oncol. 2008, 26, 279–282. [Google Scholar] [CrossRef]
  374. Telugu, R.B.; Chowhan, A.K.; Rukmangadha, N.; Patnayak, R.; Phaneendra, B.V.; Mowliswara Prasad, B.C.; Reddy, M.K. Estrogen and progesterone receptor in meningiomas: An immunohistochemical analysis. J. Cancer Res. Ther. 2020, 16, 1482–1487. [Google Scholar] [CrossRef]
  375. Ichwan, S.; Santoso, F.; Aman, R.A.; Tandian, D.; Fachniadin, A.; Nugroho, S.W. Estrogen and progesterone in meningioma: Bridging the gap of knowledge. Neurol. Asia 2023, 28, 1–11. [Google Scholar] [CrossRef]
  376. Rzechorzek, N.M.; Saunders, O.M.; Hiscox, L.V.; Schwarz, T.; Marioni-Henry, K.; Argyle, D.J.; Schoenebeck, J.J.; Freeman, T.C. Network analysis of canine brain morphometry links tumour risk to oestrogen deficiency and accelerated brain ageing. Sci. Rep. 2019, 9, 12506. [Google Scholar] [CrossRef] [PubMed]
  377. Johnson, M.D. PD-L1 expression in meningiomas. J. Clin. Neurosci. 2018, 57, 149–151. [Google Scholar] [CrossRef] [PubMed]
  378. Karimi, S.; Mansouri, S.; Nassiri, F.; Bunda, S.; Singh, O.; Brastianos, P.K.; Dunn, I.F.; Zadeh, G. Clinical significance of checkpoint regulator “Programmed death ligand-1 (PD-L1)” expression in meningioma: Review of the current status. J. Neurooncol. 2021, 151, 443–449. [Google Scholar] [CrossRef] [PubMed]
  379. Dirven, C.M.; Grill, J.; Lamfers, M.L.; Van der Valk, P.; Leonhart, A.M.; Van Beusechem, V.W.; Haisma, H.J.; Pinedo, H.M.; Curiel, D.T.; Vandertop, W.P.; et al. Gene therapy for meningioma: Improved gene delivery with targeted adenoviruses. J. Neurosurg. 2002, 97, 441–449. [Google Scholar] [CrossRef] [PubMed]
  380. De La Garza-Ramos, R.; Flores-Rodriguez, J.V.; Martinez-Gutierrez, J.C.; Ruiz-Valls, A.; Caro-Osorio, E. Current standing and frontiers of gene therapy for meningiomas. Neurosurg. Focus 2013, 35, E4. [Google Scholar] [CrossRef] [PubMed]
  381. Riemenschneider, M.J.; Perry, A.; Reifenberger, G. Histological classification and molecular genetics of meningiomas. Lancet Neurol. 2006, 5, 1045–1054. [Google Scholar] [CrossRef] [PubMed]
  382. Ragel, B.T.; Jensen, R.L. Molecular genetics of meningiomas. Neurosurg. Focus 2005, 19, E9. [Google Scholar] [CrossRef] [PubMed]
  383. Dezamis, E.; Sanson, M. The molecular genetics of meningiomas and genotypic/phenotypic correlations. Rev. Neurol. 2003, 159, 727–738. [Google Scholar]
  384. Mawrin, C.; Perry, A. Pathological classification and molecular genetics of meningiomas. J. Neurooncol. 2010, 99, 379–391. [Google Scholar] [CrossRef]
  385. Youngblood, M.W.; Gunel, M. Molecular genetics of meningiomas. Handb. Clin. Neurol. 2020, 169, 101–119. [Google Scholar] [CrossRef]
  386. Pham, M.H.; Zada, G.; Mosich, G.M.; Chen, T.C.; Giannotta, S.L.; Wang, K.; Mack, W.J. Molecular genetics of meningiomas: A systematic review of the current literature and potential basis for future treatment paradigms. Neurosurg. Focus 2011, 30, E7. [Google Scholar] [CrossRef]
  387. Gareev, I.; Beylerli, O.; Liang, Y.; Xiang, H.; Liu, C.; Xu, X.; Yuan, C.; Ahmad, A.; Yang, G. The Role of MicroRNAs in Therapeutic Resistance of Malignant Primary Brain Tumors. Front. Cell Dev. Biol. 2021, 9, 740303. [Google Scholar] [CrossRef] [PubMed]
  388. Wang, L.; Chen, S.; Liu, Y.; Zhang, H.; Ren, N.; Ma, R.; He, Z. The biological and diagnostic roles of MicroRNAs in meningiomas. Rev. Neurosci. 2020, 31, 771–778. [Google Scholar] [CrossRef] [PubMed]
  389. Carneiro, V.; Cirino, M.; Panepucci, R.; Peria, F.; Tirapelli, D.; Colli, B.; Carlotti, C.G., Jr. The Role of MicroRNA 181d as a Possible Biomarker Associated with Tumor Progression in Meningiomas. Cureus 2021, 13, e19158. [Google Scholar] [CrossRef] [PubMed]
  390. One Health High-Level Expert Panel; Adisasmito, W.B.; Almuhairi, S.; Behravesh, C.B.; Bilivogui, P.; Bukachi, S.A.; Casas, N.; Cediel Becerra, N.; Charron, D.F.; Chaudhary, A.; et al. One Health: A new definition for a sustainable and healthy future. PLoS Pathog. 2022, 18, e1010537. [Google Scholar] [CrossRef]
Table 1. Advantages and disadvantages of using domestic animals instead of murine models as a model for human cancer.
Table 1. Advantages and disadvantages of using domestic animals instead of murine models as a model for human cancer.
AdvantagesDisadvantages
Spontaneously occurring tumorsHigher effort and costs for sample collection compared to mouse model
Shared environmental and risk factorsTumor heterogeneity between species
Intact immune systemDifferent incidence between human and pets
Similar clinical, molecular, histopathological, genetic characteristics Ethics and oversights of trials with pets
Large sizeInvasive exams could be needed
Similar diagnostic techniques
Genome and involved pathways are very similar to those of humans
Table 2. Syndromes associated with a considerable frequency of meningiomas.
Table 2. Syndromes associated with a considerable frequency of meningiomas.
Associated
Syndromes
Meningioma
Frequency
Associated GenesReferences
Neurofibromatosis type 2, schwannomatosis, spinal tumors, spinal ependymoma35%NF2[49,85,86]
Gorlin–Goltz syndrome1–5%PTCH1, P53[87,88,89,90]
Cowden syndrome8.25%PTEN[91]
BAP1 Tumor Predisposition Syndrome<1%BAP-1[92]
Multiple Endocrine Neoplasia Type 1-MEN1[93,94]
Werner syndrome10.9%WRN[95]
Rubinstein–Taybi syndromeCase report [96,97]
Table 3. Canine meningioma classification criteria proposed by Belluco et al. (2022) [115].
Table 3. Canine meningioma classification criteria proposed by Belluco et al. (2022) [115].
Number of
Mitoses (n)
Mitotic Grade
<4 mitoses in 2.37 mm2Grade 1
4 ≤ n ≤ 20 in 2.37 mm2Grade 2:
tumors with sheeting architecture, small cells, hypercellularity, macronuclei and spontaneous necrosis
>20 mitoses in 2.37 mm2Grade 3:
high anaplasia
Table 4. Common MRI features observed in canine, feline, and human meningiomas.
Table 4. Common MRI features observed in canine, feline, and human meningiomas.
MRI FeaturesDogCatsHuman
MarginsDefinedDefinedIrregular
Pre-contrastIsodense to hyperdense (T1 and T2)Isodense to hyperdense (T1 and T2)Isodense to hyperdense (T1 and T2)
ContrastMarked and uniformMarked and uniformNonhomogeneous
Tumor associated edemaMildMildSevere
HyperostosisRareFrequentOccasionally
Dura tailFrequentFrequentFrequent
Mass observationCorrectly observedCorrectly observedCorrectly observed
Table 5. Immunohistochemical markers evaluated in human, canine and feline meningiomas.
Table 5. Immunohistochemical markers evaluated in human, canine and feline meningiomas.
SpeciesMarkersReference
HumanVimentin, KL-1, Claudin-1, NSE, S-100, EMA, Leu-7, Cytokeratin, N-Cadherin, KI-67[149]
DogVimentin, EMA, Claudin-1, CD18, CD1c, CD11d, CD3, IBA-1, Cytokeratin, β-Catenin, N-Cadherin, S100, Pancytokeratin, PGP9.5, CD34[20,29]
CatVimentin, E-Cadherin, β-Catenin[134]
Table 6. Reported differentially expressed genes in canine and feline meningiomas.
Table 6. Reported differentially expressed genes in canine and feline meningiomas.
SpeciesGene NameReference
DogCADM1 (also known as TSLC1)[183]
DogNF2[183]
DogVEGF[143,184]
DogMMP-2 and MMP-9[185]
CatMMP-2 and MMP-9[185]
DogSSTR2[186]
DogPDGFR[187]
DogFOLR1[188]
DogFOSB, FOS, CTSE, ZC2HC1C, KLF5, BMPR1B,
MYBL1, NR4A1, PAMR1, GJB2, GEM, UPK3B, EGR1, ZFP36, CHGA, NOL4, DACT2, AMPD3,
MEDAG, KLKB1, TF, AMDHD1, GPR133, WNT5A, RIMS1, PDPN, COMP, LPAR3, NEAT1_2, STXBP6, MYRF, PERP, BMPER, IRF6, VWA5A, MYC, ERMP1, DMXL2, WFCD2, NEAT1_1, FAM210B, CTPS1
[189]
DogMYOC, ALP, CILP2, COL14A1, THBS1, MMRN2, ADAMTSL1, PRKD1, COL8A1, SPTBN5, FHL5, MYH11, SFRP1, SCN7A, ARHGEF15, FRZB, SYNPO2, ACTC, PTGIR, TIE1, CD93, MCAM, FLT1, EMCN, MYCT1, DAAM2, KANK3, LAMC3, CALCRL, CPZ, DCP1/ACE, AQP1, KITLG, TINAGL1, PECAM1, CRISPLD2, OLFML3, ESAM, ADCY2, NOTCH3, FBLN2, FAM180A, STC2, APOD, PTCH2, APOE, CACNA2D2, PTPRB, FAM198B, MGLL, PDGFRL, TEK, IGFBP6, GAS6, ABCA8, TLL1, COL16A1, PALD1, PIK3R1, FMN2, MCF2L, NOS3, ARAP3, FMOD, DYSF, CYYR1, PLVAP, DRP2, ZFHX4, ECE1, GM2A, RCN3, AMOT, LEPREL2, TM4SF18, KALRN, PREX2, HECW, CCDC80, LIMCH1, ETS1, MYO1E, SHE[189]
Table 7. Proteins frequently altered in human and dog Meningiomas.
Table 7. Proteins frequently altered in human and dog Meningiomas.
GenesSpeciesReference
MMP-2Human, Dog[185]
MMP-9Human, Dog[185]
TIMP-1Human, Dog[185]
TIMP-2Human, Dog[185]
COX-2Human[208]
VEGFHuman[211]
N-CadherinDog[212,213]
β-CateninDog[239,240]
DCXDog[239,240]
NF-2Human[183]
Glut-1Human, Dog[29]
CD18+Dog[242,243,244,245]
CD11d+Dog[242,243,244,245]
CD3+Dog[242,243,244,245]
CD79a+Dog[242,243,244,245]
CD34Dog[4]
NSEDog[4]
PGP9.5Dog[4]
PancytokeratinDog[4]
S100Dog[4]
Table 8. Differentially regulated miRNAs in human meningiomas from different tissues.
Table 8. Differentially regulated miRNAs in human meningiomas from different tissues.
Cases-StudiesmiRNA Up RegulatedmiRNA Down RegulatedmiRNA
Status Not Specified
Reference
Grade I and II meningiomas biopsiesmiR-34a, miR-218 miR-21,
miR-143
miR-193b
miR-451
not specified[268]
Cerebrospinal fluid from meningioma patientsmiR-10a-5p, miR-140-5p, miR-196a-5p, miR-196b-5p not specified[269]
Circulating miRNAsmiR-106a-5p, miR-219-5p, miR-375, miR-409-3p, miR-1275, miR-657, and miR-224miR-107, miR-129-3p, miR-1285-3p, miR-197-3pnot specified[270]
Variable Grade of Meningiomas patientsmiR-146a-5p, miR-155-5p, miR-96-5pnot specifiedmiR-15a, miR-331-3p, miR-21a, miR-107, miR-137 and miR-29b, miR-29c, miR-200a, miR-335[271,272,273]
Variable Grade of Meningiomas patientsnot specifiednot specifiedmiR-197, miR-34a, miR-375, miR-219a, miR-224, miR-21,
miR-200a, miR-409
[274,275]
Grade I and grade III meningiomas biopsiesnot specifiedmiR-195 [276]
Table 9. miRNAs differentially regulated in human meningiomas, which have a role in different canine malignancies.
Table 9. miRNAs differentially regulated in human meningiomas, which have a role in different canine malignancies.
miRNAFunction/Mutational StatusReference
miR-cluster 14q32Important role in c-myc regulation[278,280,283]
miR-17-5pOver-expressed in canine lymphomas[278,284,285]
miR-21Up-regulated in canine mammary tumors[278,281,286,287,288]
miR-34Deleted in canine mammary tumors[278,289,290,291,292]
miR-124aRole in apoptosis control in canine hemangiosarcoma[278,293,294]
miR-145Inhibits cell growth in melanoma cells[277,278,295,296,297]
miR-200Functions in EMT regulation[277,278,288]
miR-203Reduces cell proliferation in canine lymphoma cells[278,289,296]
miR-205Down-regulated in melanoma [278,298,299]
miR-210Over-expressed in canine mammary tumor[278,300,301]
miR-221, miR-222Up-regulated in canine prostate cancer[278,302]
miR-96, miR-145,
miR-200a, miR-29c miR-335
Down-regulated in canine meningioma[277]
miR-136, miR-155, miR-146aUp-regulated in canine meningioma[277]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tomanelli, M.; Florio, T.; Vargas, G.C.; Pagano, A.; Modesto, P. Domestic Animal Models of Central Nervous System Tumors: Focus on Meningiomas. Life 2023, 13, 2284. https://doi.org/10.3390/life13122284

AMA Style

Tomanelli M, Florio T, Vargas GC, Pagano A, Modesto P. Domestic Animal Models of Central Nervous System Tumors: Focus on Meningiomas. Life. 2023; 13(12):2284. https://doi.org/10.3390/life13122284

Chicago/Turabian Style

Tomanelli, Michele, Tullio Florio, Gabriela Coronel Vargas, Aldo Pagano, and Paola Modesto. 2023. "Domestic Animal Models of Central Nervous System Tumors: Focus on Meningiomas" Life 13, no. 12: 2284. https://doi.org/10.3390/life13122284

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop