Open Access

Glioma lateralization: Focus on the anatomical localization and the distribution of molecular alterations (Review)

  • Authors:
    • Nilgun Tuncel Cini
    • Manuela Pennisi
    • Sidika Genc
    • Demetrios A. Spandidos
    • Luca Falzone
    • Panayiotis D. Mitsias
    • Aristides Tsatsakis
    • Ali Taghizadehghalehjoughi
  • View Affiliations

  • Published online on: August 16, 2024     https://doi.org/10.3892/or.2024.8798
  • Article Number: 139
  • Copyright: © Cini et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It is well known how the precise localization of glioblastoma multiforme (GBM) predicts the direction of tumor spread in the surrounding neuronal structures. The aim of the present review is to reveal the lateralization of GBM by evaluating the anatomical regions where it is frequently located as well as the main molecular alterations observed in different brain regions. According to the literature, the precise or most frequent lateralization of GBM has yet to be determined. However, it can be said that GBM is more frequently observed in the frontal lobe. Tractus and fascicles involved in GBM appear to be focused on the corticospinal tract, superior longitudinal I, II and III fascicles, arcuate fascicle long segment, frontal strait tract, and inferior fronto‑occipital fasciculus. Considering the anatomical features of GBM and its brain involvement, it is logical that the main brain regions involved are the frontal‑temporal‑parietal‑occipital lobes, respectively. Although tumor volumes are higher in the right hemisphere, it has been determined that the prognosis of patients diagnosed with cancer in the left hemisphere is worse, probably reflecting the anatomical distribution of some detrimental alterations such as TP53 mutations, PTEN loss, EGFR amplification, and MGMT promoter methylation. There are theories stating that the right hemisphere is less exposed to external influences in its development as it is responsible for the functions necessary for survival while tumors in the left hemisphere may be more aggressive. To shed light on specific anatomical and molecular features of GBM in different brain regions, the present review article is aimed at describing the main lateralization pathways as well as gene mutations or epigenetic modifications associated with the development of brain tumors.

Introduction

Glioblastoma is the most common glioma-derived tumor in the central nervous system, especially in the brain (1). The World Health Organization (WHO) classifies central nervous system tumors according to histological, molecular, and prognostic factors (2). Every year, 3–5/100,000 individuals are diagnosed with a brain tumor of which glioblastoma multiforme (GBM) represents the most common form (3). In addition, GBM constitutes the most lethal form of primary brain tumor. According to the WHO, GBM is the Grade IV stage (highest grade) of astrocytoma. The WHO GBM grading was determined according to the core structure, mitotic activity, vascularization, necrosis, proliferation rate, clinical signs, and response to treatment (3,4). Due to its highly heterogeneous structure, radical resection is not possible, resulting in a shortened life expectancy in patients. Despite intensive treatment and surgery in patients with GBM, the overall life expectancy is 12–15 months (5). Rapid infiltrative growth of GBM cells into peripheral structures causes the disease to progress more aggressively (6).

Revealing the epidemiology, etiology, anatomy, molecular structure and spreading of GBM will provide us with more opportunities to create meaningful treatments and surgical approaches. The aim of the present review is to reveal the lateralization of GBM, the anatomical regions where it is frequently located, the main molecular features in order to associate these characteristics with tumor location and brain anatomy.

Lateralization of the brain and functional area

The right and left hemispheres, which are formed by the symmetrical division of the brain into two halves through the fissura longtidunalis, are connected to each other mainly by the corpus callosum and the comissura anterior. Interhemispheric connections occur between areas specialized for the same function in the contralateral cortex (7). However, morphological and physiological differences in the human brain cause significant asymmetries between hemispheres. In most individuals, the right hemisphere appears to be heavier than the left, while the left hemisphere tends to have a denser structure, which may signify that high-level control centers are located on the left (8). This indicates that one side is functionally dominant, and partially or completely responsible for that function, and this tendency is defined as lateralization. For example, in most individuals, speech function is dominant on the left side, while visual and spatial functions are specialized on the right side (9). From a different point of view, the fact that the speech function is dominant on the left side causes this hemisphere to be dominant in other verbal skills, which in most individuals is the left side dominant (8).

Frontal lobe

The frontal lobe is the largest lobe of the brain in terms of volume and contains numerous motor and cognitive control centers (10). It is reported that the systems located on the left side of the frontal lobe are responsible for the cognitive preferences and behaviors given by the existing memory, while the right side is mostly involved in the behaviors directed by the external environment. This indicates that the hemisphere plays a vital role in the healthy processing and assembly of new information cognitively (11).

Parietal lobe

The parietal lobe includes cortical regions related to sensory and language function at the subcortical level, and it is also accepted as an intersection point of white matter pathways related to motor, sensory, language, visuospatial and visual function (12). There is general agreement in the literature that visuospatially-oriented attention is dependent on a network of frontal and parietal areas in the right hemisphere. Visuspatially-oriented attention is also considered to be related to some functions of the right parietal lobe in the production of open-eye movements (13). It has been reported that the parietal cortex tends to be more lateralized on the left side than on the right side (14). Extra-personal and personal spatial neglect can be serious and is often observed in association with the right parietal lobe (15).

Occipital lobe

The occipital lobe is the part of the brain that processes visual data. It is associated with visual-spatial processing, distance and depth perception, color identification, object and face recognition, and memory formation (16). It has been reported that the brain regions involved in visual word processing are lateralized to the left hemisphere, which is considered logical considering that in the majority of individuals, language-related cortical structures are lateralized to the left hemisphere (17). Anatomically, what is called the Yakovlevian torque (occipital bending) is the right hemisphere's tendency to rotate slightly forward relative to the left, which can cause the right frontal lobe to be larger and wider, and the left occipital lobe to be wider and project to the right. This makes the left Sylvian fissure longer and straighter, resulting in a larger planum temporale (an extension of Wernicke's area) (18).

Temporal lobe

The temporal lobe is the part of the brain responsible for various cognitive functions including memory, senses, auditory, language processing, cognition, and semantics (19). Some studies report that while the temporal lobe has a larger area especially on the left side, on the contrary, the cortex thickness is higher on the right side (20,21). The presence of asymmetries in the morphological development of the temporal lobes is considered an important sign of lateralization. The most prominent asymmetry is observed in the peri-Sylvian region and superior temporal sulcus (21).

In general, when the whole brain is examined, it is reported that the total surface area and volume of the left hemisphere are higher, whereas the cortex thickness is higher on the right side (20). Although such lateralization is generally observed, it is also observed that there are differences between the subunits in each lobe (22).

All these data suggest how asymmetries can be found at different levels with different parameters such as regional volume, cortical thickness, connections, cellular and molecular organization, and surface area. The most clearly studied asymmetry is the speech function and it is known to be lateralized to the left side (8,23).

The right hemisphere is less exposed to external influences in its development as it is responsible for the functions necessary for survival. In addition, there is a general dominance of the right hemisphere for all functions except language, therefore the right hemisphere develops earlier (8).

Although genotype probably plays a role in the development of structural asymmetries, lesions in any hemisphere can trigger dominance of the unaffected side due to high plasticity. On the other hand, environmental and/or physiological factors can also cause asymmetries to occur, the best example of which is the right-hand preference due to cultural pressures (18,24).

Lateralization of the gliomas within the brain

Considering the complementary functions, the human brain usually has duality, and individuals are categorized as ‘left or right-brained’ according to the dominant hemisphere (25). It is known that a number of different functions, especially the preference for hand use, and lateralization of speech, are located on one side of the cerebrum (right or left) and it is accepted as the dominant hemisphere (26). While the right hemisphere is primarily associated with nonverbal abilities, the left one is reported to be responsible for verbal memory and language functions (27). It is suggested that evaluating the localization of GBM can be accepted as an indicator in determining the direction of spread (28). While magnetic resonance imaging (MRI) can reveal the characteristic structure of the disease by determining the volumetric information of the tumor and the determination of the anatomical structures that are or may be affected, these determinations are insufficient to reveal the pathophysiology and prognosis (29).

Inskip et al (30), in their study conducted on 489 patients with glioma (354 high-grade, 135 low-grade), 197 meningiomas, and 96 acoustic neuromas, did not find a statistical difference between the rates of incidence on the left and right sides, although there was no significant difference between them, in patients with low-grade glioma and meningioma. More specifically, in this study, a more common distribution on the right side compared with the left side was observed, although it was not significant in patients with high-grade and acoustic neuroma. These authors reported that aphasia and mental status changes are more commonly observed in patients with glioma and tumors affecting the left side of the brain. In a study by Jansma and Rutten it was reported that 30 tumors from patients with high-grade glioma were located on the left side and 26 were located on the right side, while 37 patients with low-grade glioma had tumors located on the left side and 16 had tumors located on the right side (31).

In a study by Coluccia et al (32) performed on 235 patients, the incidence of tumor spread was as follows: The frontal lobe (left, 29.8%; right, 43.0%), the temporal lobe (left, 42.1%; right, 43.8%), the occipital lobe (left, 15.8%; right, 9.9%), the parietal lobe (left, 35.1%; right, 29.8), the basal ganglia (left, 4.4%; right, 7.4%). These authors also observed that tumors located in the right hemisphere were larger. In the same study, it was reported that the patients with the tumor located on the right had paralysis in the extremities, and the patients with the tumor located on the left had more language problems (63.2% in the right hemisphere and 10.0% in the left hemisphere). There was a decrease in Karnofsky Performance Status after resection in patients with left-sided tumors compared with patients with right-sided lesions. While there was no difference in the overall survival (OS) of the patients regardless of the side, it was reported that there was a decrease in the progression-free survival (PFS) in left-sided patients (7.4 months vs. 10.1 months). The authors stated that the reason for this result was that total resection was performed with less success on the left side.

Appropriate surgical resection is one of the important points in achieving tumor control in GBM. The morbidity risks and reduced quality of life are taken into account when approaching tumors located in the vicinity of or directly within important anatomical regions (33). It has been observed that resection of the dominant hemisphere carries great surgical risks. It has been reported that patients with left temporal lobe glioma experience higher preoperative neurocognitive impairment and demonstrate a more frequent and severe decline in neurocognitive abilities after surgical resection compared with patients with right temporal lobe glioma (34).

In a study involving 507 patients by Ellingson et al (35), it was reported that tumors were located to a greater extent on the left side. A relationship between tumor lateralization and OS with an extended survival time of up to 36 months in patients with left-sided tumors compared with the survival time of 12 months observed in patients with tumors affecting the right side, was also oberved in this study. The authors also stated that the lesions were more inclined to be located in the frontal lobe in young patients compared with the elderly (35). In a study by Larjavaara et al (36), on 331 patients with glioma, it was reported that tumors were mostly located in the frontal lobe (40%), followed by the temporal lobe (29%), parietal lobe (14%), and occipital lobe (3%), respectively. The authors also stated that the tumor was located more frequently on the right side than on the left (51% on the right and 40% on the left).

One of the most comprehensive studies on lateralization in recent years is the study by Kommers et al (37), which was conducted on a total of 1,596 patients with GBM from 13 centers. In this study, the structure of the tumor, its volume, and the structures of the affected brain regions were examined by automatic and manual segmentation with MRI. In the automated segmentations, it was identified that 785 (49.2%) tumors were located on the left side, while 792 (49.6%) were on the right side. In manual segmentation, 794 (49.7%) patients had left-sided tumors, and 799 (50.1%) had right-sided tumors. It was also observed that 19 patients (1.2%) examined by automated segmentation and 3 patients (0.2%) examined by manual segmentation, exhibited no laterality. The authors of this study stated that automatic segmentation produced near-perfect results. In the continuation of the study, cortical and subcortical anatomical formations or anatomical regions in the affected hemisphere were determined in detail.

Based on the study by Kommers et al (37), data obtained through automated segmentation revealed that the tumors appeared to be most frequently localized in the insular lobe, frontal lobe and temporal lobe, respectively (37). According to this study, these tumors affected a number of areas of the frontal lobe and were frequently located in the precentral gyrus, with a lesser percentage in the cingulate anterior gyrus and frontal opercular cortex. In the parietal lobe, gliomas were mostly localized in the posterior division of the cingulate gyrus, posterior division of the marginal gyrus, and precuneus cortex. It was observed that the areas involved in the temporal lobe were several. Among these, tumors were often located in the superior temporal gyrus, planum temporale, and central opercular cortex. The incidence of the tumor was mainly observed to be located in the occipital cortex and in the superior and inferior divisions of the lateral occipital cortex. The incidence for each lobe was similar for the cortex structure affected on the right and left sides. Tractus and fascicles involved in subcortical structures appeared to be the corticospinal tract, superior longitudinal I, II and III fascicles, arcuate fascicle long segment, frontal strait tract, and inferior frontal-occipital fascicle. Overall, the eclipse rate of numerous of these structures was high on the right. It should be noted that, except for the arcuate fascicle long segment, the anterior segment was observed significantly higher on the right side than on the left side (37).

Mickevicius et al (38) retrospectively studied 113 patients with GBM, hypothesizing that the location of the white matter structures with the tumor was associated with survival. The authors found that OS times were reduced in patients with tumors located in the right anterior thalamic radiation (ATR), right lower inferior fronto-occipital fasciculus (IFOF), right and left cortico-spinal tract (CST), and corpus callosum (CC). It was also revealed that PFS times were decreased in patients with tumors located in the CST, CC body, right ATR, posterior IFOF, and inferior longitudinal fasciculus (ILF), and PFS times were increased in patients with tumors located in the right genu of CC and anterior IFOF. The main findings of the aforementioned studies are summarized in Table I.

Table I.

Studies reporting the lateralization of glioblastoma.

Table I.

Studies reporting the lateralization of glioblastoma.

Lateralization

First author(s), yearNo. of patientsBrain regionLeftRight(Refs.)
Inskip et al, 2003354 HGGEntire brainLeft (30)
135 LGGEntire brainRight
Ellingson et al, 2013507 Left (35)
Jansma and Rutten, 201756Entire brain53.6%46.4%(31)
53Entire brain69.8%30.2%
Coluccia et al, 2018235Frontal lobe29.8%43.0%(32)
Temporal lobe42.1%43.8%
Parietal lobe35.1%29.8%
Occipital lobe15.8%9.9%
Basal ganglia4.4%7.4%
Larjavaara et al, 2007116 (GBM)Entire brain44.5%55.5%(36)
Frontal lobe
Temporal lobe
Parietal lobe
Occipital lobe
Kommers et al, 20211,596 (Automated segmentation)Entire brain49.2%50.1%(37)
1,596 (Manual segmentation)Entire brain49.7%49.6%

[i] HGG, high grade glioma; LGG, low grade glioma; GBM, glioblastoma mutliforme.

Anatomy of brain regions affected by glioblastoma

CS

The CST is a complex system consisting of a series of projection fibers that control spinal cord functions by the brain, including the control of spinal reflexes and motor neuron activity. CST axons (75–90% of the axons) form a crossing at the level of the medulla oblongata and at the level of the midline, called the pyramidal decussation. This signifies that the left side of the brain controls the right side of the spinal cord and is critical for motor functions (39,40). Most CST axons originate from pyramidal neurons in layer V located in the primary motor and sensory cortex (M1 and S1). A pioneer study by Danks et al using diffusion tensor imaging (DTI) revealed that the CST originates mainly from the M1 and S1, but also receives input from the supplementary motor areas (SMA) and the ventral and dorsal premotor cortices (PMC). Leaving the neocortex, the CST reaches the brainstem by passing through the posterior limb of the internal capsule and cerebral peduncles before reaching the brainstem in the ventral position. Data obtained through the use of DTI revealed that the CST content originates from 37% M1, 32% S1, 25% SMA, and 7% PMC (41).

The control of complex motor functions such as the selection of movement, making the final decision, initiating the function, and monitoring the process is provided by the SMA. It is known that this area has connections with the precentral gyrus, prefrontal cortex, basal ganglia, limbic system, spinal cord, contralateral SMA, superior parietal cortex, and inferior frontal cortical areas, especially pars opercularis. When Salvati et al (42) compared 127 patients with GBM and SMA involvement (Group A) and patients with non-SMA but M1 and CST involvement (Group B), it was reported that group A patients had a higher volume, but there was no change in the OS and PFS durations of the patients.

Superior longitudinal fasciculus (SLF)

The primary function of the SLF is to provide communication between the frontal and parietal lobes and partial connections with the temporal lobe. It is accepted that there are two different paths due to its proximity to the arcuate fascicle that connects the posterior temporal lobe and the frontal lobe in the peri-Slyvian region (43,44). Makris et al (45) analyzed the SLF by dividing it into four anatomical subsections. SLF I is the dorsal part and provides the connection between the superior parietal and superior frontal lobes. SLF II starts from the angular gyrus, passes over the centrum semiovale on the insula, and ends in the caudal-lateral prefrontal region. SLF III is the ventral part of the pathway, and it travels between the anterior part of the angular gyrus, and the supramarginal gyrus, and the ventral premotor and prefrontal areas. The 4th subcomponent of the SLF, identified as SLF IV in earlier studies on non-human primates, corresponds to the arcuate fasciculus (AF). This segment connects the posterior part of the superior temporal gyrus to the lateral prefrontal cortex, running through the caudal extremity of the Sylvian fissure. Nonetheless, the designation of the AF as the 4th component of the SLF is not universally accepted (46,47).

In definitions by Catani and Thiebaut de Schotten, each arcuate fascicle is divided into three parts (long, anterior and posterior) connecting two regions of the Broca, Wernicke, or Geschwind region (inferior parietal lobule) (46). The anterior segment of AF appears to correspond to SLF III and the two terms are used interchangeably. Therefore, although these two bundles are separate structures, some of their subcomponents appear to overlap with each other (47).

Various studies have revealed that the SLF is markedly closer to the cingulum, while it cannot be separated by other SLF structures (SLF I–III and AF) (48,49); in addition, according to Thiebaut de Schotten, the SLF is symmetrical in both hemispheres (50). However, some studies in the literature are not concordant indicating that SLF is greater on the right side than on the left side (47,51), while other studies have claimed that SLF II is dominant on the left side through multiple components (49,52). This anatomical feature may explain the lateralization of the language in the left hemisphere (Fig. 1) (53).

Several studies have demonstrated the occurrence of gliomas and glioblastoma in the SLF region. More specifically, Davtian et al (54), presented a case report of a 59-year-old woman with recurrent GBM involving the left medial frontal and cingulate gyri with impairment of the SLF region (54). The authors described that the dissection of the tumor border in contact with the SLF resulted in recurrent speech arrest highlighting how the surgical resection of tumors in this region is not recommended for large lesions (54). Previously, Nakajima et al (55), observed visuospatial dysfunction in patients with glioma affecting the right dorsal SLF.

Similarly, Liu et al (56), observed cognitive deficits in patients with glioma located in the right SLF temporal part.

Overall, all these data are concordant in demonstrating the cognitive decline (in terms of visuospatial and speech abilities) associated with glioma development in the SLF region.

ILF

The ILF is the white matter structure that provides the reciprocal connection between the temporal-parietal-occipital lobes. It also functions in visual word recognition, connecting the occipital cortex to the posterior occipitotemporal cortices (57). Latini et al (58) reported that the main structure of the ILF is located more in the fusiform, lingual, and dorsolateral-occipital regions of the occipital lobe and that these are fixed components. It was revealed that there was no lateralization for the subcomponents of the ILF, but that they were located on the right in total volume (Fig. 2).

As regards the ILF, there are a few studies describing the localization of GBM in this region. Specifically, the postsurgical residual lesion volume located in the left ILF was associated with lexical retrieval impairments supporting the data already described in the aforemetioned section on SLF (59,60).

IFOF

The IFOF is the white matter structure that connects the occipital cortex, temporo-basal areas, superior parietal lobule, and precuneus to the frontal lobe. The region starting from the periphery of the caudate nucleus and extending to the temporal horn of the lateral ventricle is the sub-insular region, and the IFOF is located on the ventral third of this region and the outer side of the external capsule. This pathway is involved in mumerous functions such as language, non-verbal semantic processing, object identification, visual-spatial processing and planning, reading, facial expression recognition, and memory (Fig. 2) (61). Vassal et al (62) examined the cortical terminations of IFOF by tractography in 20 healthy individuals to determine individual variations and asymmetry. According to their data, IFOF terminations lateralize over the superior parietal lobule on the right and the inferior frontal gyrus on the left. Altieri et al (63) reported that of a total of 23 patients with 38% GBM, 33% oligodendroglioma, and 29% astrocytoma, 57% of tumors were located in the left hemisphere and 43% in the right henisphere. Moreover in this study, this pathway was divided into three parts anatomically: A vertical that runs along the frontal lobe, a horizontal segment that runs along the frontal lobe, and a horizontal segment that runs from the limen insulae. Caminis et al (64) reported that 33 lesions of patients with tumors involving the temporal lobe were located on the left side (97%), 14 were medial, 14 were lateral, and 6 were medial and lateral extensions. In total, it was reported that the lesions were directed to the insula in 22 cases.

Frontostriatal tract (FST)

The FST connects several cortical areas of the frontal lobe, especially the prefrontal cortex, with the striatum and the thalamus. It is responsible for a wide range of mental, motor, limbic, and cognitive functions (65). In particular, the connections between the caudate nucleus and the frontal cortex have a markedly rigid and clear topographic organization in regionally specific clusters. It is reported that these specific regions are located in the subregions of the ventrolateral, dorsolateral, and orbitofrontal cortex and that there is a similar organizational scheme in both hemispheres (66).

From a different anatomical point of view, two different association fiber structures originating from SMA and connecting with different regions are mentioned in the literature. One is the FST, which connects the pre-SMA to the anterior part of the caudate nucleus, and the other is the fronto-aslant tract, which connects the pre-SMA to the pars opercularis. The FST starts from the caudate nucleus, passes through the lateral ventricle's frontal horn, and terminates in the ipsilateral inferior frontal gyrus (67). The frontal aslant tract is a brain white matter pathway that connects the superior frontal gyrus (SMA, pre-SMA) to the pars opercularis and pars triangularis of the inferior frontal gyrus and the insula (68). Functionally, the left frontal aslant tract is responsible for language and the right for executive functions (69).

An in-depth analysis of the literature does not reveal any studies on glioblastoma involvement in the FST. However, Müller et al (70) questioned the possibility of performing neurosurgery for the removal of glioma affecting the caudate nucleus connected to the supplementary motor area through the FST. In this case, the authors discussed the functional role of both the caudate nucleus and the FST that can be indicative of the limits of resection in the case of supracomplete glioma resection (70).

A number of these pathways, which have a complex structure, are still the subject of research. A summary of these aforementioned pathways regarding GBM is included in Table II.

Table II.

White matter brain pathways.

Table II.

White matter brain pathways.

PathwayConnections
Corticospinal tractPrimary motor and sensory cortex to the spinal cord (lower motor neurons)
  SLF ISuperior parietal lobe to the superior frontal lobe
  SLF IIAngular gyrus to the caudal-lateral prefrontal region
  SLF IIIBetween the anterior part of the angular gyrus, supramarginal gyrus, ventral premotor and prefrontal areas
Inferior longitudinal fasciculusBetween the temporal, parietal and occipital lobes
Inferior fronto-occipital fascicleOccipital cortex, temporo-basal areas, superior parietal lobule and precuneus to th frontal lobe
Fronto-striatal tractBetween the prefrontal cortex, with the striatum and the thalamus

[i] SLF, superior longitudinal fasciculus.

Lateralization of molecular features in glioblastoma

As aforementioned, the lateralization of glioma and glioblastoma has profound anatomical implications that can limit the surgical approaches available due to the impairment of cognitive functions. In addition, it was widely demonstrated that GBM lateralization also has effects on tumor molecular features which influence the pathogenesis of the tumor and the clinical outcomes (71). Despite extensive research efforts, GBM remains a challenge due to its heterogeneity and resistance to treatment, and such heterogeneity is also reflected in the prevalence of molecular alterations observed in the right and left side of the brain (72).

Genomic studies have demonstrated profound differences existing between left and right brain tumors in terms of molecular profiles. Notably, mutations in the isocitrate dehydrogenase 1 (IDH1) gene are more prevalent in tumors located in the frontal lobe, whereas tumors located in the temporal lobe frequently exhibit epidermal growth factor receptor (EGFR) amplification and phosphatase and tensin homolog (PTEN) loss (73,74).

The lateralization of molecular features is not limited only to gene mutations. Notably, DNA methylation, histone modifications, and other epigenetic alterations have exhibited lateralization patterns in GBM (75,76). One of the most common epigenetic alterations observed in cancer, including GBM, is the alteration of DNA methylation (75). The analysis of DNA methylation patterns observed in GBM has revealed region-specific differences in the methylome of GBM, with distinct CpG island methylator phenotype (CIMP) subtypes associated with specific brain regions (75,77). Moreover, differential histone modifications have been observed in GBM located in different brain regions suggesting a putative role of epigenetics in regional tumor heterogeneity and specific subtypes such as the H3 G34 mutant type (78).

In addition to the anatomical and biological lateralization of GBM-associated molecular features, the lateralization of both genetic and epigenetic alterations also has important clinical and therapeutic implications. In this context, the lateralization of molecular features in GBM can be useful to improve the diagnosis and prognosis of this tumor. Indeed, the integration of molecular information with clinical and radio-imaging data could enhance the accuracy of tumor diagnosis and guide personalized treatment strategies. For example, the identification of specific molecular alterations associated with different brain regions could aid in the preoperative prediction of tumor location and facilitate surgical planning (79). In addition, the lateralization of molecular alterations may provide useful prognostic information correlating both side-specific genetic and epigenetic features with the survival outcomes of patients and the route of metastatization (80,81).

Some authors have also proposed to tailor the treatment depending on the lateralization of the molecular features in GBM (82,83). Due to the regional differences observed for the genetic and epigenetic alterations in GBM, targeted approaches could be designed to exploit specific vulnerabilities associated with each brain region. For instance, therapies targeting IDH1 mutations may be more effective in frontal lobe tumors, while targeting EGFR amplification could be more beneficial in temporal lobe tumors (84). In addition, the immune microenvironment and tumor-stromal cell interactions may have a role in the response to immunotherapies and other targeted interventions, however, more in-depth investigations should be performed to fully clarify these complex interactions.

Emerging evidence suggests that specific molecular alterations may exhibit a predilection for GBM located in the right side, left side, frontal lobe or other regions of the brain. This section aims to explore the molecular alterations frequently observed in different areas of the brain in patients with gliomas and glioblastomas, shedding light on their potential clinical and therapeutic implications (Fig. 3 and Table SI).

IDH1 mutations

IDH1 mutations represent one of the most observed molecular alterations in both gliomas and glioblastoma (85,86). A molecular and radio imaging study performed on patients with glioma revealed a higher prevalence of IDH1 mutations in tumors mainly affecting the right hemisphere compared with the left hemisphere (87). However, the precise mechanisms responsible for this lateralization pattern are not widely understood, but can be associated with GBM regional differences in IDH1 expression or genetic predispositions associated with brain asymmetry (88). As a limitation of these findings, the majority of the studies on the detection of IDH mutations do not report the right or left lateralization of the mutation but only the brain region affected.

EGFR amplification

Amplification of the EGFR gene is a common genetic alteration observed in glioblastoma (89). Notably, a previous study indicated a higher frequency of EGFR amplification in glioblastoma located on the left temporal lobe of the brain (35). The reasons for this specific lateralization are still under investigation, but could be attributed to variations in the microenvironment or signaling pathways specific to the left hemisphere (90). More importantly, the prevalence of EGFR mutation in GBM may have important prognostic implications as different targeted treatments for EGFR-positive tumors are currently available (91).

PTEN loss

PTEN is a tumor suppressor gene frequently mutated or deleted in glioblastoma. The loss-of-function of PTEN results in the dysregulation of multiple associated signaling pathways involved in cell proliferation, cell survival and apoptosis (92,93). Contrary to what was observed for IDH1 and EGFR, there are no studies describing the specific distribution of PTEN alterations in the two brain hemispheres, however, the study of Ellingson et al (35) revealed a higher incidence of PTEN loss in glioblastomas affecting the frontal lobe. However, with regard to this case, the underlying mechanisms for this lateralization pattern require further investigation.

O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation

As regards the lateralization of epigenetics alterations, the MGMT gene encodes a DNA repair protein that can be silenced through the hypermethylation of its promoter (94). MGMT promoter methylation is associated with increased sensitivity to alkylating chemotherapy, such as temozolomide, one of the main drugs used for the treatment of GBM (95,96). Studies have reported a higher frequency of MGMT promoter methylation in left-sided glioblastomas compared with the hypomethylation observed in tumors affecting the right side (97) suggesting how in the left hemisphere epigenetic alterations induced by stromal cells or the microenvironment are more evident compared with the right side (98).

TP53 mutations

A significant fraction of GBM harbor mutations affecting the TP53 gene, which encodes the tumor suppressor protein p53 and plays a key role in the regulation of DNA integrity (99). A previous study reported a higher prevalence of TP53 mutations in gliomas affecting the left medial temporal lobe and the right anterior temporal lobe (100). Another study revealed that p53-mutated glioblastomas were preferentially located in the frontal lobe near the rostral extension of the lateral ventricles (101). As for the other mutations observed in GBM, the mechanisms responsible for this lateralization remain unclear but it results in the alteration of DNA repair capacity or regional variations in the response to mutagenic factors (102).

Neurofibromin 1 (NF1) alterations

Alterations in the NF1 gene have been implicated in the pathogenesis of gliomas (103). Notably, NF1 acts as a negative regulator of the Ras signaling pathway thus playing a role in the modulation of cell proliferation and cell survival. As described for PTEN, NF1 alterations can affect GBM occurring in different parts of the brain, however, there is a slight preference for the left hemisphere as demonstrated by two independent studies where NF1-mutated gliomas affected the left side of the brainstem, left optic nerve and left cerebellum (104,105).

Platelet-derived growth factor receptor-α (PDGFRA) amplification

Amplification of the PDGFRA gene is a recurrent genetic alteration observed in glioblastoma (106). A previous study has indicated a higher frequency of PDGFRA amplification in H3 G34 diffuse hemispheric gliomas located on both the right and left frontal lobes of the brain (107). However, the specific mechanisms underlying this lateralization pattern remain to be fully elucidated.

ATRX chromatin remodeler (ATRX) mutations

ATRX is a gene involved in chromatin remodeling and it is often mutated in astrocytoma and low-grade glioma (108). Previous studies demonstrated that ATRX mutations can be found both in the right and left hemispheres affecting different brain regions including the frontal lobe, the temporal lobe, the parietal lobe, the occipital lobe, and in rare cases also the brainstem, and the thalamus (108,110).

Overall, understanding the specific right- or left-sided molecular alterations in glioma and glioblastoma has fundamental diagnostic, prognostic, and clinical implications. First of all, the molecular differences observed in right and left tumors could aid the clinical diagnosis performed through imaging techniques and identify potential hidden lesions. Secondly, the precise characterization of the lateralization of molecular alterations may contribute to the development of novel targeted therapies. In particular, the presence of EGFR and TP53 alterations, along with other molecular features, may drive the selection of the correct drug to be administered to the patients. Finally, the definition of the lateralization of a tumor from an anatomical and molecular point of view could improve the management of this tumor.

Notably, different studies are concordant in confirming a more common location of GBM within the frontal lobe and insula with a consequent poor prognosis in terms of PFS times when the tumor is located on the left side of the brain. It was also reported that the white matter structures on the right side are larger in volume, thus the tumors arising in this region are usually of a larger volume. Based on the data obtained in the study by Kommers et al it is demonstrated that tumors mainly affect the SLF, ILF, IFOF, AF, FST, and CST regions (however, no statistical comparison depending on the brain side was performed in the study) (37). Considering the anatomical features of these localizations and the areas they connect, it is logical that the major involvements are observed in the frontal-temporal-parietal-occipital lobes, respectively. This supports the hypothesis that the tumor follows the white matter structures during its spreading favored by both direct and indirect mechanisms (for example, mediated by GBM-derived exosomes which induce neurotoxicity in the surrounding structures) (111113). Although tumor volumes are higher on the right side, some sources state that the prognosis is worse in left-sided patients. However, it should be noted here that there is no difference in OS times, regardless of the localization of tumors. The aforementioned pathways related to GBM and the evaluation of the anatomy of the region for appropriate resection are especially emphasized by surgeons. Some sources state that establishing the location of tumors within brain regions could be useful to determine the prognosis of the disease as well as the surgical decisions (15). In the literature it is reported that regular radiological imaging can be used as a predictor in the evaluation of prognosis by detecting the anatomical and volumetric position of the tumor, necrosis or extent of edema, and pathophysiological changes in patients (16).

Overall, the anatomical data on GBM lateralization, coupled with the growing findings on molecular alterations affecting specific brain regions and epigenetics events involved in GBM development and progression (114,115), may improve the clinical management of patients with GBM and the positive effects on the outcomes of patients. In addition, a deep understanding of both anatomical and molecular lateralization is essential to propose novel effective treatment in GBM, including photoacoustic nanoprobes (116,117).

Conclusions

By analyzing the studies reported in the literature, it is not clear if a precise lateralization of brain tumors exists. However, there is strong evidence supporting the anatomical lateralization of both glioma and glioblastoma as well as the molecular lateralization of some key molecular alterations which influence the evolution of tumors and the prognosis of patients. Recently, a broad meta-analysis has collected all the studies reporting the anatomical lateralization of glioma and GBM revealing no specific lateralization patterns in the right or left hemispheres. However, a clear relationship between tumor location in specific brain regions and glioma-associated symptoms were established (118). As regards the lateralization of glioma and GBM molecular features, more robust investigations on this specific topic are needed to establish lateralization patterns of mutations and epigenetics alterations.

In conclusion, these data suggest that the specific evaluation of glioma and GBM localization from both an anatomical and molecular perspective can help clinicians in a number of areas. It may help develop more specific drugs and treatment methods based on different molecular changes in different brain regions. In addition, predicting the location of the tumor and the possibility of spread can provide the physician preliminary information regarding the prognosis of the disease. Another important issue is that technologies such as artificial intelligence can be used in this field. Evaluating radiological images with artificial intelligence, especially in the early stages, is a topic that can be evaluated in terms of prognosis.

Supplementary Material

Supporting Data

Acknowledgements

MP acknowledges the PIA.CE.RI Program of the University of Catania for its support in allowing young researchers to promote and participate in research activities (PIA.CE.RI ID: EBioCaSt).

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

ATs and ATa conceptualized the study. MP, LF, PDM, SG and NTC wrote the original draft of the manuscript. MP, LF, PDM, DAS and ATa provided critical revisions. LF, SG and NTC prepared the tables and figures, conducted the formal analysis, and critically analyzed the literature. Data authentication is not applicable. All authors contributed to the manuscript revision, as well as read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

DAS is the Editor-in-Chief for the journal, but had no personal involvement in the reviewing process, or any influence in terms of adjudicating on the final decision, for this article.

References

1 

Bouget D, Eijgelaar RS, Pedersen A, Kommers I, Ardon H, Barkhof F, Bello L, Berger MS, Nibali MC, Furtner J, et al: Glioblastoma surgery imaging-reporting and data system: Validation and performance of the automated segmentation task. Cancers (Basel). 13:46742021. View Article : Google Scholar : PubMed/NCBI

2 

Fontán-Lozano Á, Morcuende S, Davis-López de Carrizosa MA, Benítez-Temiño B, Mejías R and Matarredona ER: To Become or not to become tumorigenic: Subventricular zone versus hippocampal neural stem cells. Front Oncol. 10:6022172020. View Article : Google Scholar : PubMed/NCBI

3 

Xu H, Chen X, Sun Y, Hu X, Zhang X, Wang Y, Tang Q, Zhu Q, Song K, Chen H, et al: Comprehensive molecular characterization of long-term glioblastoma survivors. Cancer Lett. 593:2169382024. View Article : Google Scholar : PubMed/NCBI

4 

Taghizadehghalehjoughi A, Hacımüftüoglu A, Cetin M, Ugur AB, Galatenau B, Mezhuev Y, Okkay U, Taspinar N, Taspinar M, Uyanik A, et al: Effect of metformin/irinotecan-loaded poly-lactic-co-glycolic acid nanoparticles on glioblastoma: In vitro and in vivo studies. Nanomedicine. 13:1595–1606. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Grochans S, Cybulska AM, Simińska D, Korbecki J, Kojder K, Chlubek D and Baranowska-Bosiacka I: Epidemiology of glioblastoma multiforme-literature review. Cancers (Basel). 14:24122022. View Article : Google Scholar : PubMed/NCBI

6 

Seker-Polat F, Pinarbasi Degirmenci N, Solaroglu I and Bagci-Onder T: Tumor cell infiltration into the brain in glioblastoma: From mechanisms to clinical perspectives. Cancers (Basel). 14:4432022. View Article : Google Scholar : PubMed/NCBI

7 

Pittella JEH: The uniqueness of the human brain: A review. Dement Neuropsychol. 18:e202300782024. View Article : Google Scholar : PubMed/NCBI

8 

Bisiacchi P and Cainelli E: Structural and functional brain asymmetries in the early phases of life: A scoping review. Brain Struct Funct. 227:479–496. 2022. View Article : Google Scholar : PubMed/NCBI

9 

Spaccavento S, Caliendo S, Galetta R, Picciola E, Losavio E and Glueckauf R: Pragmatic communication deficit and functional outcome in patients with right- and left-brain damage: A pilot study. Brain Sci. 14:3872024. View Article : Google Scholar : PubMed/NCBI

10 

Catani M: The anatomy of the human frontal lobe. Handb Clin Neurol. 163:95–122. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Goldberg E, Podell K and Lovell M: Lateralization of frontal lobe functions and cognitive novelty. J Neuropsychiatry Clin Neurosci. 6:371–378. 1994. View Article : Google Scholar : PubMed/NCBI

12 

Wu Y, Wang J, Zhang Y, Zheng D, Zhang J, Rong M, Wu H, Wang Y, Zhou K and Jiang T: The neuroanatomical basis for posterior superior parietal lobule control lateralization of visuospatial attention. Front Neuroanat. 10:322016. View Article : Google Scholar : PubMed/NCBI

13 

Dziedzic TA, Bala A and Marchel A: Cortical and subcortical anatomy of the parietal lobe from the neurosurgical perspective. Front Neurol. 12:7270552021. View Article : Google Scholar : PubMed/NCBI

14 

Jeong SK and Xu Y: The impact of top-down spatial attention on laterality and hemispheric asymmetry in the human parietal cortex. J Vis. 16:22016. View Article : Google Scholar : PubMed/NCBI

15 

Kondo M: The laterality of parietal association areas: Hemispatial neglect, body images and body schema. Brain Nerve. 70:1059–1066. 2018.(In Japanese). PubMed/NCBI

16 

Rehman A and Al Khalili Y: Neuroanatomy, Occipital Lobe. StatPearls Treasure Island, FL: StatPearls Publishing; 2023, Available from:. https://www.ncbi.nlm.nih.gov/books/NBK544320/

17 

Vonk JMJ, Borghesani V, Battistella G, Younes K, DeLeon J, Welch A, Hubbard HI, Miller ZA, Miller BL and Gorno-Tempini ML: Verbal semantics and the left dorsolateral anterior temporal lobe: A longitudinal case of bilateral temporal degeneration. Aphasiology. 34:865–885. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Kuo F and Massoud TF: Structural asymmetries in normal brain anatomy: A brief overview. Ann Anat. 241:1518942022. View Article : Google Scholar : PubMed/NCBI

19 

Zhao F, Wu Z, Wang L, Lin W and Li G: Longitudinally consistent registration and parcellation of cortical surfaces using semi-supervised learning. Med Image Anal. 96:1031932024. View Article : Google Scholar : PubMed/NCBI

20 

Kong XZ, Mathias SR, Guadalupe T; ENIGMA Laterality Working Group, ; Glahn DC, Franke B, Crivello F, Tzourio-Mazoyer N, Fisher SE, Thompson PM, et al: Mapping cortical brain asymmetry in 17,141 healthy individuals worldwide via the ENIGMA Consortium. Proc Natl Acad Sci USA. 115:E5154–E5163. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Fu L, Wang Y, Fang H, Xiao X, Xiao T, Li Y, Li C, Wu Q, Chu K, Xiao C and Ke X: Longitudinal study of brain asymmetries in autism and developmental delays Aged 2–5 years. Neuroscience. 432:137–149. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Koelkebeck K, Miyata J, Kubota M, Kohl W, Son S, Fukuyama H, Sawamoto N, Takahashi H and Murai T: The contribution of cortical thickness and surface area to gray matter asymmetries in the healthy human brain. Hum Brain Mapp. 35:6011–6022. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Esteves M, Ganz E, Sousa N and Leite-Almeida H: Asymmetrical brain plasticity: Physiology and pathology. Neuroscience. 454:3–14. 2021. View Article : Google Scholar : PubMed/NCBI

24 

Grant JH, Parker AJ, Hodgson JC, Hudson JM and Bishop DVM: Testing the relationship between lateralization on sequence-based motor tasks and language laterality using an online battery. Laterality. 28:1–31. 2023. View Article : Google Scholar : PubMed/NCBI

25 

Corballis MC: Evolution of cerebral asymmetry. Prog Brain Res. 250:153–178. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Zhai Z and Feng J: Left-right asymmetry influenced the infarct volume and neurological dysfunction following focal middle cerebral artery occlusion in rats. Brain Behav. 8:e011662018. View Article : Google Scholar : PubMed/NCBI

27 

Edwards JD, Jacova C, Sepehry AA, Pratt B and Benavente OR: A quantitative systematic review of domain-specific cognitive impairment in lacunar stroke. Neurology. 80:315–322. 2013. View Article : Google Scholar : PubMed/NCBI

28 

De Luca C, Virtuoso A, Papa M, Certo F, Barbagallo GMV and Altieri R: Regional development of Glioblastoma: The anatomical conundrum of cancer biology and its surgical implication. Cells. 11:13492022. View Article : Google Scholar : PubMed/NCBI

29 

Nestler U, Lutz K, Pichlmeier U, Stummer W, Franz K, Reulen HJ and Bink A; 5-ALA Glioma Study Group, : Anatomic features of glioblastoma and their potential impact on survival. Acta Neurochir (Wien). 157:179–186. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Inskip PD, Tarone RE, Hatch EE, Wilcosky TC, Selker RG, Fine HA, Black PM, Loeffler JS, Shapiro WR and Linet MS: Laterality of brain tumors. Neuroepidemiology. 22:130–138. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Jansma JM and Rutten G: P04.11 effect of hemisphere and tumor grade on default mode deactivation in glioma patients. Neuro Onco. 19:iii422017. View Article : Google Scholar

32 

Coluccia D, Roth T, Marbacher S and Fandino J: Impact of laterality on surgical outcome of glioblastoma patients: A retrospective single-center study. World Neurosurg. 114:e121–e128. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Mazoyer B, Zago L, Jobard G, Crivello F, Joliot M, Perchey G, Mellet E, Petit L and Tzourio-Mazoyer N: Gaussian mixture modeling of hemispheric lateralization for language in a large sample of healthy individuals balanced for handedness. PLoS One. 9:e1011652014. View Article : Google Scholar : PubMed/NCBI

34 

Noll KR, Ziu M, Weinberg JD and Wefel J: Neurocognitive functioning in patients with glioma of the left and right temporal lobes. J Neurooncol. 128:323–331. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Ellingson BM, Lai A, Harris RJ, Selfridge JM, Yong WH, Das K, Pope WB, Nghiemphu PL, Vinters HV, Liau LM, et al: Probabilistic radiographic atlas of glioblastoma phenotypes. AJNR Am J Neuroradiol. 34:533–540. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Larjavaara S, Mäntylä R, Salminen T, Haapasalo H, Raitanen J, Jääskeläinen J and Auvinen A: Incidence of gliomas by anatomic location. Neuro Oncol. 9:319–325. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Kommers I, Bouget D, Pedersen A, Eijgelaar RS, Ardon H, Barkhof F, Bello L, Berger MS, Conti Nibali M, Furtner J, et al: Glioblastoma surgery imaging-reporting and data system: Standardized reporting of tumor volume, location, and resectability based on automated segmentations. Cancers (Basel). 13:28542021. View Article : Google Scholar : PubMed/NCBI

38 

Mickevicius NJ, Carle AB, Bluemel T, Santarriaga S, Schloemer F, Shumate D, Connelly J, Schmainda KM and LaViolette PS: Location of brain tumor intersecting white matter tracts predicts patient prognosis. J Neurooncol. 125:393–400. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Filippopulos FM, Brem C, Seelos K, Köglsperger T, Sonnenfeld S, Kellert L and Vollmar C: Uncrossed corticospinal tract in health and genetic disorders: Review, case report, and clinical implications. Eur J Neurol. 28:2804–2811. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Welniarz Q, Dusart I and Roze E: The corticospinal tract: Evolution, development, and human disorders. Devel Neurobio. 77:810–829. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Danks RA, Aglio LS, Gugino LD and Black PM: Craniotomy under local anesthesia and monitored conscious sedation for the resection of tumors involving eloquent cortex. J Neurooncol. 49:131–139. 2000. View Article : Google Scholar : PubMed/NCBI

42 

Salvati M, Armocida D, Pesce A, Palmieri M, Venditti E, D'Andrea G, Frati A and Santoro A: No prognostic differences between GBM-patients presenting with postoperative SMA-syndrome and GBM-patients involving cortico-spinal tract and primary motor cortex. J Neurol Sci. 419:1171882020. View Article : Google Scholar : PubMed/NCBI

43 

Frye RE, Hasan K, Malmberg B, Desouza L, Swank P, Smith K and Landry S: Superior longitudinal fasciculus and cognitive dysfunction in adolescents born preterm and at term. Dev Med Child Neurol. 52:760–766. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Nakajima R, Kinoshita M, Shinohara H and Nakada M: The superior longitudinal fascicle: Reconsidering the fronto-parietal neural network based on anatomy and function. Brain Imaging Behav. 14:2817–2830. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Makris N, Kennedy DN, McInerney S, Sorensen AG, Wang R, Caviness VS Jr and Pandya DN: Segmentation of subcomponents within the superior longitudinal fascicle in humans: A quantitative, in vivo, DT-MRI study. Cereb Cortex. 15:854–869. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Catani M and Thiebaut de Schotten M: Atlas of Human Brain Connections. Oxford; University Press: 2012, View Article : Google Scholar

47 

Janelle F, Iorio-Morin C, D'Amour S and Fortin D: Superior longitudinal fasciculus: A review of the anatomical descriptions with functional correlates. Front Neurol. 13:7946182022. View Article : Google Scholar : PubMed/NCBI

48 

Martino J, De Witt Hamer PC, Berger MS, Lawton MT, Arnold CM, de Lucas EM and Duffau H: Analysis of the subcomponents and cortical terminations of the perisylvian superior longitudinal fasciculus: A fiber dissection and DTI tractography study. Brain Struct Funct. 218:105–121. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Wang X, Pathak S, Stefaneanu L, Yeh FC, Li S and Fernandez-Miranda JC: Subcomponents and connectivity of the superior longitudinal fasciculus in the human brain. Brain Struct Funct. 221:2075–2092. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Thiebaut de Schotten M, Dell'Acqua F, Forkel SJ, Simmons A, Vergani F, Murphy DG and Catani M: A lateralized brain network for visuospatial attention. Nat Neurosci. 14:1245–1246. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Hecht EE, Gutman DA, Bradley BA, Preuss TM and Stout D: Virtual dissection and comparative connectivity of the superior longitudinal fasciculus in chimpanzees and humans. Neuroimage. 108:124–137. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Vernooij MW, Smits M, Wielopolski PA, Houston GC, Krestin GP and van der Lugt A: Fiber density asymmetry of the arcuate fasciculus in relation to functional hemispheric language lateralization in both right- and lefthanded healthy subjects: A combined fMRI and DTI study. Neuroimage. 35:1064–1076. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Netter FH: Atlas of Human Anatomy. 6th edition. Elsevier; Philedelphia: 2008

54 

Davtian M, Ulmer JL, Mueller WM, Gaggl W, Mulane MP and Krouwer HG: The superior longitudinal fasciculus and speech arrest. J Comput Assist Tomogr. 32:410–414. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Nakajima R, Kinoshita M, Miyashita K, Okita H, Genda R, Yahata T, Hayashi Y and Nakada M: Damage of the right dorsal superior longitudinal fascicle by awake surgery for glioma causes persistent visuospatial dysfunction. Sci Rep. 7:171582017. View Article : Google Scholar : PubMed/NCBI

56 

Liu D, Liu Y, Hu X, Hu G, Yang K, Xiao C, Hu J, Li Z, Zou Y, Chen J, et al: Alterations of white matter integrity associated with cognitive deficits in patients with glioma. Brain Behav. 10:e016392020. View Article : Google Scholar : PubMed/NCBI

57 

Tamai S, Kinoshita M, Nakajima R, Okita H and Nakada M: Two different subcortical language networks supporting distinct Japanese orthographies: Morphograms and phonograms. Brain Struct Funct. 227:1145–1154. 2022. View Article : Google Scholar : PubMed/NCBI

58 

Latini F, Mårtensson J, Larsson EM, Fredrikson M, Åhs F, Hjortberg M, Aldskogius H and Ryttlefors M: Segmentation of the inferior longitudinal fasciculus in the human brain: A white matter dissection and diffusion tensor tractography study. Brain Res. 1675:102–115. 2017. View Article : Google Scholar : PubMed/NCBI

59 

Duffau H: White matter tracts and diffuse Lower-grade gliomas: The pivotal role of myelin plasticity in the tumor pathogenesis, infiltration patterns, functional consequences and therapeutic management. Front Oncol. 12:8555872022. View Article : Google Scholar : PubMed/NCBI

60 

Herbet G, Moritz-Gasser S, Boiseau M, Duvaux S, Cochereau J and Duffau H: Converging evidence for a cortico-subcortical network mediating lexical retrieval. Brain. 139:3007–3021. 2016. View Article : Google Scholar : PubMed/NCBI

61 

DE Benedictis A, Marras CE, Petit L and Sarubbo S: The inferior Fronto-occipital fascicle: A century of controversies from anatomy theaters to operative neurosurgery. J Neurosurg Sci. 65:605–615. 2021.PubMed/NCBI

62 

Vassal F, Pommier B, Sontheimer A and Lemaire JJ: Inter-individual variations and hemispheric asymmetries in structural connectivity patterns of the inferior Fronto-occipital fascicle: A diffusion tensor imaging tractography study. Surg Radiol Anat. 40:129–137. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Altieri R, Melcarne A, Junemann C, Zeppa P, Zenga F, Garbossa D, Certo F and Barbagallo G: Inferior Fronto-occipital fascicle anatomy in brain tumor surgeries: From anatomy lab to surgical theater. J Clin Neurosci. 68:290–294. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Camins À, Naval-Baudin P, Majós C, Sierpowska J, Sanmillan JL, Cos M, Rodriguez-Fornells A and Gabarrós A: Inferior Fronto-occipital fascicle displacement in temporoinsular gliomas using diffusion tensor imaging. J Neuroimaging. 32:638–646. 2022. View Article : Google Scholar : PubMed/NCBI

65 

Heller C, Steinmann S, Levitt JJ, Makris N, Antshel KM, Fremont W, Coman IL, Schweinberger SR, Weiß T, Bouix S, et al: Abnormalities in white matter tracts in the Fronto-striatal-thalamic circuit are associated with verbal performance in 22q11.2DS. Schizophr Res. 224:141–150. 2020. View Article : Google Scholar : PubMed/NCBI

66 

Levitt JJ, Zhang F, Vangel M, Nestor PG, Rathi Y, Kubicki M, Shenton ME and O'Donnell LJ: The organization of frontostriatal brain wiring in healthy subjects using a novel diffusion imaging fiber cluster analysis. Cereb Cortex. 31:5308–5318. 2021. View Article : Google Scholar : PubMed/NCBI

67 

Kinoshita M, de Champfleur NM, Deverdun J, Moritz-Gasser S, Herbet G and Duffau H: Role of Fronto-striatal tract and frontal aslant tract in movement and speech: An axonal mapping study. Brain Struct Funct. 220:3399–3412. 2015. View Article : Google Scholar : PubMed/NCBI

68 

La Corte E, Eldahaby D, Greco E, Aquino D, Bertolini G, Levi V, Ottenhausen M, Demichelis G, Romito LM, Acerbi F, et al: The frontal aslant tract: A systematic review for neurosurgical applications. Front Neurol. 12:6415862021. View Article : Google Scholar : PubMed/NCBI

69 

Landers MJF, Meesters SPL, van Zandvoort M, de Baene W and Rutten GM: The frontal aslant tract and its role in executive functions: A quantitative tractography study in glioma patients. Brain Imaging Behav. 16:1026–1039. 2022. View Article : Google Scholar : PubMed/NCBI

70 

Müller DMJ, Robe PAJT, Eijgelaar RS, Witte MG, Visser M, de Munck JC, Broekman MLD, Seute T, Hendrikse J, Noske DP, et al: Comparing Glioblastoma surgery decisions between teams using brain maps of tumor locations, biopsies, and resections. JCO Clin Cancer Inform. 3:1–12. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Cancer Genome Atlas Research Network, . Brat DJ, Verhaak RG, Aldape KD, Yung WK, Salama SR, Cooper LA, Rheinbay E, Miller CR, Vitucci M, et al: Comprehensive, integrative genomic analysis of diffuse Lower-grade gliomas. N Engl J Med. 372:2481–2498. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Yabo YA, Niclou SP and Golebiewska A: Cancer cell heterogeneity and plasticity: A paradigm shift in glioblastoma. Neuro Oncol. 24:669–682. 2022. View Article : Google Scholar : PubMed/NCBI

73 

Qi S, Yu L, Li H, Ou Y, Qiu X, Ding Y, Han H and Zhang X: Isocitrate dehydrogenase mutation is associated with tumor location and magnetic resonance imaging characteristics in astrocytic neoplasms. Oncol Lett. 7:1895–1902. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Fathi Kazerooni A, Bakas S, Saligheh Rad H and Davatzikos C: Imaging signatures of glioblastoma molecular characteristics: A radiogenomics review. J Magn Reson Imaging. 52:54–69. 2020. View Article : Google Scholar : PubMed/NCBI

75 

J Dabrowski M and Wojtas B: Global DNA methylation patterns in human gliomas and their interplay with other epigenetic modifications. Int J Mol Sci. 20:34782019. View Article : Google Scholar : PubMed/NCBI

76 

Klughammer J, Kiesel B, Roetzer T, Fortelny N, Nemc A, Nenning KH, Furtner J, Sheffield NC, Datlinger P, Peter N, et al: The DNA methylation landscape of glioblastoma disease progression shows extensive heterogeneity in time and space. Nat Med. 24:1611–1624. 2018. View Article : Google Scholar : PubMed/NCBI

77 

de Souza CF, Sabedot TS, Malta TM, Stetson L, Morozova O, Sokolov A, Laird PW, Wiznerowicz M, Iavarone A, Snyder J, et al: A Distinct DNA methylation shift in a subset of Glioma CpG island methylator phenotypes during tumor recurrence. Cell Rep. 23:637–651. 2018. View Article : Google Scholar : PubMed/NCBI

78 

Lucas CG, Mueller S, Reddy A, Taylor JW, Oberheim Bush NA, Clarke JL, Chang SM, Gupta N, Berger MS, Perry A, et al: Diffuse hemispheric glioma, H3 G34-mutant: Genomic landscape of a new tumor entity and prospects for targeted therapy. Neuro Oncol. 23:1974–1976. 2021. View Article : Google Scholar : PubMed/NCBI

79 

Gatto L, Franceschi E, Tosoni A, Di Nunno V, Tonon C, Lodi R, Agati R, Bartolini S and Brandes AA: Beyond imaging and genetic signature in Glioblastoma: Radiogenomic holistic approach in Neuro-oncology. Biomedicines. 10:32052022. View Article : Google Scholar : PubMed/NCBI

80 

Cui M, Gao X, Chi Y, Zhang M, Lin H, Chen H, Sun C and Ma X: Molecular alterations and their correlation with the survival of glioblastoma patients with corpus callosum involvement. Front Neurosci. 15:7014262021. View Article : Google Scholar : PubMed/NCBI

81 

Kannan S, Murugan AK, Balasubramanian S, Munirajan AK and Alzahrani AS: Gliomas: Genetic alterations, mechanisms of metastasis, recurrence, drug resistance, and recent trends in molecular therapeutic options. Biochem Pharmacol. 201:1150902022. View Article : Google Scholar : PubMed/NCBI

82 

El Atat O, Naser R, Abdelkhalek M, Habib RA and El Sibai M: Molecular targeted therapy: A new avenue in glioblastoma treatment. Oncol Lett. 25:462022. View Article : Google Scholar : PubMed/NCBI

83 

Jiang H, Yu K, Cui Y, Ren X, Li M, Zhang G, Yang C, Zhao X, Zhu Q and Lin S: Differential predictors and clinical implications associated with long-term survivors in IDH Wildtype and mutant Glioblastoma. Front Oncol. 11:6326632021. View Article : Google Scholar : PubMed/NCBI

84 

Senhaji N, Louati S, Chbani L, El Fatemi H, Hammas N, Mikou K, Maaroufi M, Benzagmout M, Boujraf S, El Bardai S, et al: EGFR amplification and IDH mutations in glioblastoma patients of the northeast of morocco. Biomed Res Int. 2017:80458592017. View Article : Google Scholar : PubMed/NCBI

85 

Solomou G, Finch A, Asghar A and Bardella C. Mutant IDH in Gliomas: Role in cancer and treatment options. Cancers (Basel). 15:28832023. View Article : Google Scholar : PubMed/NCBI

86 

Sharma N, Mallela AN, Shi DD, Tang LW, Abou-Al-Shaar H, Gersey ZC, Zhang X, McBrayer SK and Abdullah KG: Isocitrate dehydrogenase mutations in gliomas: A review of current understanding and trials. Neurooncol Adv. 5:vdad0532023.PubMed/NCBI

87 

Kudulaiti N, Zhang H, Qiu T, Lu J, Aibaidula A, Zhang Z, Guan Y and Zhuang D: The Relationship between IDH1 mutation status and metabolic imaging in nonenhancing supratentorial diffuse gliomas: A 11C-MET PET study. Mol Imaging. 18:15360121198940872019. View Article : Google Scholar : PubMed/NCBI

88 

Kopal J, Kumar K, Shafighi K, Saltoun K, Modenato C, Moreau CA, Huguet G, Jean-Louis M, Martin CO, Saci Z, et al: Using rare genetic mutations to revisit structural brain asymmetry. bioRxiv. Apr 18–2023.(Epub ahead of print). doi: 10.1101/2023.04.17.537199.

89 

Xu H, Zong H, Ma C, Ming X, Shang M, Li K, He X, Du H and Cao L: Epidermal growth factor receptor in glioblastoma. Oncol Lett. 14:512–516. 2017. View Article : Google Scholar : PubMed/NCBI

90 

Oprita A, Baloi SC, Staicu GA, Alexandru O, Tache DE, Danoiu S, Micu ES and Sevastre AS: Updated insights on EGFR signaling pathways in glioma. Int J Mol Sci. 22:5872021. View Article : Google Scholar : PubMed/NCBI

91 

Ezzati S, Salib S, Balasubramaniam M and Aboud O: Epidermal growth factor receptor inhibitors in glioblastoma: Current status and future possibilities. Int J Mol Sci. 25:23162024. View Article : Google Scholar : PubMed/NCBI

92 

Moghaddam M, Vivarelli S, Falzone L, Libra M and Bonavida B: Cancer resistance via the downregulation of the tumor suppressors RKIP and PTEN expressions: Therapeutic implications. Explor Target Antitumor Ther. 4:170–207. 2023. View Article : Google Scholar : PubMed/NCBI

93 

Dogan E, Yildirim Z, Akalin T, Ozgiray E, Akinturk N, Aktan C, Solmaz AE, Biceroglu H, Caliskan KE, Ertan Y, et al: Investigating the effects of PTEN mutations on cGAS-STING pathway in glioblastoma tumours. J Neurooncol. 166:283–292. 2024. View Article : Google Scholar : PubMed/NCBI

94 

Della Monica R, Cuomo M, Buonaiuto M, Costabile D, Franca RA, Del Basso De Caro M, Catapano G, Chiariotti L and Visconti R: MGMT and Whole-genome DNA methylation impacts on diagnosis, prognosis and therapy of glioblastoma multiforme. Int J Mol Sci. 23:71482022. View Article : Google Scholar : PubMed/NCBI

95 

Falzone L, Bordonaro R and Libra M: SnapShot: Cancer chemotherapy. Cell. 186:18162023. View Article : Google Scholar

96 

Alnahhas I, Alsawas M, Rayi A, Palmer JD, Raval R, Ong S, Giglio P, Murad MH and Puduvalli V: Characterizing benefit from temozolomide in MGMT promoter unmethylated and methylated glioblastoma: A systematic review and meta-analysis. Neurooncol Adv. 2:vdaa0822020.PubMed/NCBI

97 

Ellingson BM, Cloughesy TF, Pope WB, Zaw TM, Phillips H, Lalezari S, Nghiemphu PL, Ibrahim H, Naeini KM, Harris RJ, et al: Anatomic localization of O6-methylguanine DNA methyltransferase (MGMT) promoter methylated and unmethylated tumors: A radiographic study in 358 de novo human glioblastomas. Neuroimage. 59:908–916. 2012. View Article : Google Scholar : PubMed/NCBI

98 

Li P, Ensink E, Lang S, Marshall L, Schilthuis M, Lamp J, Vega I and Labrie V: Hemispheric asymmetry in the human brain and in Parkinson's disease is linked to divergent epigenetic patterns in neurons. Genome Biol. 21:612020. View Article : Google Scholar : PubMed/NCBI

99 

Zhang Y, Dube C, Gibert M Jr, Cruickshanks N, Wang B, Coughlan M, Yang Y, Setiady I, Deveau C, Saoud K, et al: The p53 pathway in glioblastoma. Cancers (Basel). 10:2972018. View Article : Google Scholar : PubMed/NCBI

100 

Wang YY, Zhang T, Li SW, Qian TY, Fan X, Peng XX, Ma J, Wang L and Jiang T: Mapping p53 mutations in low-grade glioma: A voxel-based neuroimaging analysis. AJNR Am J Neuroradiol. 36:70–76. 2015. View Article : Google Scholar : PubMed/NCBI

101 

Zhang T, Wang Y, Fan X, Ma J, Li S, Jiang T and Wang L: Anatomical localization of p53 mutated tumors: A radiographic study of human glioblastomas. J Neurol Sci. 346:94–98. 2014. View Article : Google Scholar : PubMed/NCBI

102 

Marutani M, Tonoki H, Tada M, Takahashi M, Kashiwazaki H, Hida Y, Hamada J, Asaka M and Moriuchi T: Dominant-negative mutations of the tumor suppressor p53 relating to early onset of glioblastoma multiforme. Cancer Res. 59:4765–4769. 1999.PubMed/NCBI

103 

Scheer M, Leisz S, Sorge E, Storozhuk O, Prell J, Ho I and Harder A: Neurofibromatosis type 1 gene alterations define specific features of a subset of glioblastomas. Int J Mol Sci. 23:3522021. View Article : Google Scholar : PubMed/NCBI

104 

Costa AA and Gutmann DH: Brain tumors in neurofibromatosis type 1. Neurooncol Adv. 1:vdz0402019.PubMed/NCBI

105 

Lobbous M, Bernstock JD, Coffee E, Friedman GK, Metrock LK, Chagoya G, Elsayed G, Nakano I, Hackney JR, Korf BR, et al: An update on neurofibromatosis type 1-associated gliomas. Cancers (Basel). 12:1142020. View Article : Google Scholar : PubMed/NCBI

106 

Carlotto BS, Trevisan P, Provenzi VO, Soares FP, Rosa RFM, Varella-Garcia M and Zen PRG: PDGFRA, KIT, and KDR gene amplification in glioblastoma: Heterogeneity and clinical significance. Neuromolecular Med. 25:441–450. 2023. View Article : Google Scholar : PubMed/NCBI

107 

Hu W, Duan H, Zhong S, Zeng J and Mou Y: High frequency of PDGFRA and MUC family gene mutations in diffuse hemispheric glioma, H3 G34-mutant: A glimmer of hope? J Transl Med. 20:642022. View Article : Google Scholar : PubMed/NCBI

108 

Haase S, Garcia-Fabiani MB, Carney S, Altshuler D, Núñez FJ, Méndez FM, Núñez F, Lowenstein PR and Castro MG: Mutant ATRX: Uncovering a new therapeutic target for glioma. Expert Opin Ther Targets. 22:599–613. 2018. View Article : Google Scholar : PubMed/NCBI

109 

Meng L, Zhang R, Fa L, Zhang L, Wang L and Shao G: ATRX status in patients with gliomas: Radiomics analysis. Medicine (Baltimore). 101:e301892022. View Article : Google Scholar : PubMed/NCBI

110 

Silantyev AS, Falzone L, Libra M, Gurina OI, Kardashova KS, Nikolouzakis TK, Nosyrev AE, Sutton CW, Mitsias PD and Tsatsakis A: Current and future trends on diagnosis and prognosis of glioblastoma: From molecular biology to proteomics. Cells. 8:8632019. View Article : Google Scholar : PubMed/NCBI

111 

Zhang S, Su X, Kemp GJ, Yang X, Wan X, Tan Q, Yue Q and Gong Q: Two patterns of white matter connection in multiple gliomas: Evidence from probabilistic fiber tracking. J Clin Med. 11:36932022. View Article : Google Scholar : PubMed/NCBI

112 

Genc S, Pennisi M, Yeni Y, Yildirim S, Gattuso G, Altinoz MA, Taghizadehghalehjoughi A, Bolat I, Tsatsakis A, Hacımüftüoğlu A, et al: Potential neurotoxic effects of Glioblastoma-derived exosomes in primary cultures of cerebellar neurons via oxidant stress and glutathione depletion. Antioxidants (Basel). 11:12252022. View Article : Google Scholar : PubMed/NCBI

113 

Yeni Y, Taghizadehghalehjoughi A, Genc S, Hacimüftüoglu A, Yıldırım S and Bolat I: Glioblastoma cell-derived exosomes induce cell death and oxidative stress in primary cultures of olfactory neurons. Role of redox stress. Mol Biol Rep. 50:3999–4009. 2023. View Article : Google Scholar : PubMed/NCBI

114 

Stella M, Falzone L, Caponnetto A, Gattuso G, Barbagallo C, Battaglia R, Mirabella F, Broggi G, Altieri R, Certo F, et al: Serum extracellular Vesicle-derived circHIPK3 and circSMARCA5 Are two novel diagnostic biomarkers for glioblastoma multiforme. Pharmaceuticals (Basel). 14:6182021. View Article : Google Scholar : PubMed/NCBI

115 

Candido S, Lupo G, Pennisi M, Basile MS, Anfuso CD, Petralia MC, Gattuso G, Vivarelli S, Spandidos DA, Libra M, et al: The analysis of miRNA expression profiling datasets reveals inverse microRNA patterns in glioblastoma and Alzheimer's disease. Oncol Rep. 42:911–922. 2019.PubMed/NCBI

116 

Zeng F, Fan Z, Li S, Li L, Sun T, Qiu Y, Nie L and Huang G: Tumor microenvironment activated photoacoustic-fluorescence bimodal nanoprobe for precise Chemo-immunotherapy and immune response tracing of glioblastoma. ACS Nano. 17:19753–19766. 2023. View Article : Google Scholar : PubMed/NCBI

117 

Peng Y, Liu Y, Lu X, Wang S, Chen M, Huang W, Wu Z, Lu G and Nie L: Ag-hybridized plasmonic Au-triangular nanoplates: Highly sensitive photoacoustic/Raman evaluation and improved antibacterial/photothermal combination therapy. J Mater Chem B. 6:2813–2820. 2018. View Article : Google Scholar : PubMed/NCBI

118 

Valenzuela-Fuenzalida JJ, Moyano-Valarezo L, Silva-Bravo V, Milos-Brandenberg D, Orellana-Donoso M, Nova-Baeza P, Suazo-Santibáñez A, Rodríguez-Luengo M, Oyanedel-Amaro G, Sanchis-Gimeno J, et al: Association between the anatomical location of glioblastoma and its evaluation with clinical considerations: A systematic review and meta-analysis. J Clin Med. 13:34602024. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2024
Volume 52 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cini NT, Pennisi M, Genc S, Spandidos DA, Falzone L, Mitsias PD, Tsatsakis A and Taghizadehghalehjoughi A: Glioma lateralization: Focus on the anatomical localization and the distribution of molecular alterations (Review). Oncol Rep 52: 139, 2024.
APA
Cini, N.T., Pennisi, M., Genc, S., Spandidos, D.A., Falzone, L., Mitsias, P.D. ... Taghizadehghalehjoughi, A. (2024). Glioma lateralization: Focus on the anatomical localization and the distribution of molecular alterations (Review). Oncology Reports, 52, 139. https://doi.org/10.3892/or.2024.8798
MLA
Cini, N. T., Pennisi, M., Genc, S., Spandidos, D. A., Falzone, L., Mitsias, P. D., Tsatsakis, A., Taghizadehghalehjoughi, A."Glioma lateralization: Focus on the anatomical localization and the distribution of molecular alterations (Review)". Oncology Reports 52.4 (2024): 139.
Chicago
Cini, N. T., Pennisi, M., Genc, S., Spandidos, D. A., Falzone, L., Mitsias, P. D., Tsatsakis, A., Taghizadehghalehjoughi, A."Glioma lateralization: Focus on the anatomical localization and the distribution of molecular alterations (Review)". Oncology Reports 52, no. 4 (2024): 139. https://doi.org/10.3892/or.2024.8798