Brain tumor
A brain tumor is the growth of abnormal cells in the tissues of the brain, which can be benign (noncancerous) or malignant (cancerous).[1] These tumors may originate in the brain as primary tumors or spread from other parts of the body as secondary (metastatic) tumors.[2] Brain tumors vary widely in type, location, and behavior; common primary types include gliomas (arising from glial cells), meningiomas (from meninges covering the brain), and medulloblastomas (embryonal tumors often in children).[3] They can occur in both adults and children, with an estimated lifetime risk of diagnosis around 0.6% for brain and other nervous system cancers.[4] Symptoms of brain tumors depend on the tumor's size, location, and growth rate, but often include persistent headaches, seizures, vision or hearing changes, balance problems, and cognitive or personality alterations.[5] Weakness or numbness on one side of the body, nausea, and vomiting may also occur, particularly if the tumor increases pressure within the skull.[6] Diagnosis typically involves imaging such as MRI or CT scans, followed by biopsy to determine the tumor type and grade.[3] The exact causes of most brain tumors remain unknown, though risk factors include prior exposure to ionizing radiation (especially in childhood), certain genetic syndromes like neurofibromatosis or Li-Fraumeni syndrome, and, for some subtypes, specific genetic mutations.[3] Unlike many cancers, lifestyle factors such as smoking or diet play little established role.[7] Treatment is multidisciplinary and tailored to the tumor's characteristics; options include surgical resection to remove as much tumor as possible, radiation therapy (including stereotactic radiosurgery), chemotherapy, and targeted therapies for specific molecular features.[8] Prognosis varies significantly, with benign tumors often curable by surgery alone, while aggressive malignant types like glioblastoma have lower survival rates despite advances in care.[3]Overview
Definition
A brain tumor is defined as the growth of abnormal cells in the tissues of the brain, forming a mass that can disrupt normal brain function. These tumors originate from various cellular components within or around the brain, including glial cells that support neurons, neurons themselves (though rarely), or structures like the meninges that cover the brain and spinal cord. Primary brain tumors arise directly in the brain, while secondary tumors spread from other parts of the body.[1][9][10] Brain tumors are broadly classified into benign and malignant categories based on their behavior and potential to cause harm. Benign tumors are non-cancerous, typically slow-growing, and do not invade surrounding tissues, though they can exert pressure on adjacent brain structures; examples include certain meningiomas. In contrast, malignant tumors are cancerous, aggressive, and invasive, often spreading within the brain or to other areas, leading to more severe complications; these are graded by the World Health Organization from 1 (least aggressive) to 4 (most aggressive).[1][9][10] As intracranial neoplasms, brain tumors are distinguished from spinal cord tumors, which occur in the lower central nervous system; this entry focuses on those affecting the brain. They can develop in key anatomical compartments, such as the cerebrum (responsible for higher functions like thought and movement), the cerebellum (coordinating balance and posture), the brainstem (controlling vital functions like breathing), or the meninges (protective layers). The location influences the tumor's impact, though specific effects are detailed in subsequent sections.[10][9] Evidence of brain tumors dates back to ancient times, with paleopathological findings of meningiomas—benign tumors of the meninges—identified in Egyptian and Peruvian skulls from thousands of years ago, suggesting early human encounters with these growths. Modern understanding and classification advanced significantly in the 1920s through the work of neurosurgeons Percival Bailey and Harvey Cushing, who established a foundational histological system for categorizing gliomas and other brain tumors based on cellular origins and characteristics.[11][12]Common types
Brain tumors are broadly classified into primary and secondary types based on their origin. Primary brain tumors originate within the brain or its surrounding tissues, such as the meninges or cranial nerves, and do not spread from other parts of the body.[13] These account for the majority of tumors in children but a smaller proportion in adults. Common examples include gliomas, which arise from glial cells and encompass subtypes like astrocytomas (including the aggressive glioblastoma) and oligodendrogliomas; meningiomas, which develop from the meninges and are often benign; pituitary adenomas, originating in the pituitary gland; schwannomas, which form on nerve sheaths, particularly the vestibular schwannoma on the eighth cranial nerve; and medulloblastomas (embryonal tumors common in children).[9][14][3] Secondary, or metastatic, brain tumors occur when cancer cells from elsewhere in the body, such as the lungs, breast, or skin (melanoma), spread to the brain via the bloodstream. These are the most common type of brain tumor in adults, outnumbering primary brain tumors by a ratio of approximately 10:1, and often present as multiple lesions.[15][16][17] Brain tumors can also be categorized by location, which influences symptoms and treatment approaches. Supratentorial tumors are located above the tentorium cerebelli in the cerebrum and are more common in adults, while infratentorial tumors occur below the tentorium in the cerebellum or brainstem and predominate in children. Sellar tumors arise in or near the sella turcica, typically involving the pituitary gland.[18][19] Less common primary brain tumors include primary central nervous system (CNS) lymphoma, which affects the brain, spinal cord, or eyes and is often associated with immunosuppression; craniopharyngiomas, benign growths near the pituitary that can cause hormonal disruptions; and choroid plexus tumors, which develop in the ventricles and produce cerebrospinal fluid, more frequently seen in children.[20][9][21] The World Health Organization (WHO) grading system classifies most primary brain tumors into four grades (I-IV) based on histological features like cell atypia, mitosis, vascular proliferation, and necrosis, indicating their potential for malignancy and aggressiveness: grade I tumors are benign and slow-growing, grade II are low-grade with possible progression, grade III are malignant and anaplastic, and grade IV are highly malignant with rapid growth.[22][23]Signs and symptoms
General symptoms
Brain tumors often present with general symptoms resulting from increased intracranial pressure (ICP) and mass effect, which can affect brain function broadly regardless of tumor location. These symptoms arise as the tumor growth leads to swelling or obstruction of cerebrospinal fluid flow, elevating pressure within the skull.[9] Headaches are among the most common initial complaints, typically described as persistent, dull, and pressure-like, often worsening in the morning due to higher ICP during sleep when lying flat. They may intensify with activities that further increase pressure, such as coughing, sneezing, or bending over, and are frequently accompanied by nausea. This occurs because the elevated ICP stretches pain-sensitive structures like the dura mater (meninges) and blood vessels.[24][25][26] Nausea and vomiting frequently accompany headaches in brain tumor patients, often manifesting as projectile vomiting without preceding nausea, particularly in cases of acute ICP elevation. These symptoms result from the pressure affecting the brainstem's vomiting center or disrupting normal cerebrospinal fluid dynamics.[27][28][29] Seizures, which can be focal (affecting one part of the body) or generalized (involving the whole body), occur in a significant proportion of cases and may serve as the presenting symptom, especially in adults. They stem from the tumor's irritation of surrounding brain tissue or secondary effects like edema.[30][31][32] Cognitive impairments, such as memory loss and confusion, can emerge from the diffuse impact of ICP or mass effect on neural networks, leading to broader disruptions in attention, executive function, and information processing.[33][34][35] Headaches occur in about 50% of patients at diagnosis, while seizures are reported in 20-50% of cases and may be the initial symptom in many adults, highlighting their role as key indicators prompting medical evaluation. While these symptoms are nonspecific and can vary by tumor site, they underscore the need for prompt neuroimaging in persistent cases.[9][36]Location-dependent effects
The symptoms of brain tumors vary significantly based on their anatomical location, as the tumor disrupts specific brain functions associated with that region. While general symptoms such as headaches or seizures may occur due to increased intracranial pressure, location-specific effects often manifest as focal neurological deficits, including motor, sensory, or autonomic impairments.[9][37] Frontal lobe tumors can lead to motor weakness, such as hemiparesis or difficulty with voluntary movements, as well as aphasia affecting speech production or comprehension. Balance problems and trouble walking may also arise from involvement of motor planning areas. Personality changes, including forgetfulness or altered behavior, can occur but are often intertwined with broader neurological shifts.[9][37][38] Temporal lobe tumors frequently cause memory disturbances, such as difficulty forming new memories or recalling recent events, due to disruption of hippocampal and related structures. Auditory hallucinations, including hearing sounds or voices that are not present, may emerge, alongside complex partial seizures characterized by altered consciousness and automatisms. Speech impairments and unusual sensory perceptions, like unpleasant tastes or smells, can also result.[9][37] Parietal lobe tumors often produce sensory loss, including deficits in touch, pain, or temperature sensation on the contralateral side of the body. Spatial disorientation, such as difficulty navigating spaces or judging distances, is common, along with challenges in identifying objects by touch (astereognosis) or integrating sensory inputs for body position awareness. Vision or hearing problems may occur if adjacent areas are affected.[9][37] Occipital lobe tumors primarily result in visual field defects, such as hemianopia (loss of half the visual field) or quadrantanopia, due to interference with primary visual processing. Visual hallucinations, often simple geometric patterns or flashes of light, can arise from irritation of cortical areas. Complete vision loss in one or both eyes may develop in advanced cases.[9][37] Cerebellar tumors lead to ataxia, characterized by unsteady gait and limb incoordination, as well as general coordination problems affecting fine motor tasks like writing or buttoning clothes. Nystagmus, involuntary eye movements causing blurred vision or dizziness, is a frequent finding, often accompanied by balance issues that worsen with head position changes.[9][37] Brainstem tumors commonly cause cranial nerve palsies, resulting in facial weakness, double vision (diplopia), swallowing difficulties (dysphagia), or hoarseness from involvement of nerves V through XII. Respiratory issues, such as irregular breathing patterns or apnea, may occur due to compression of vital centers, potentially leading to life-threatening failure. Motor deficits like tetraparesis and sensory disturbances can also manifest.[37][39][40] Pituitary tumors, located at the base of the brain, often produce hormonal imbalances by compressing or invading the gland. For instance, adrenocorticotropic hormone (ACTH)-secreting tumors can cause Cushing's syndrome, with symptoms including central obesity, purple striae, hypertension, and proximal muscle weakness from excess cortisol. Other imbalances may lead to growth abnormalities or reproductive dysfunction, though mass effects can also cause headaches or visual field loss.[41]Neurological and behavioral changes
Brain tumors can induce a range of neurological and behavioral changes due to direct tumor invasion, surrounding edema, or secondary effects like hydrocephalus, often manifesting as subtle alterations that progress over time. These changes may include irritability, apathy, and disinhibition, particularly when tumors affect frontal regions or cause diffuse pressure. For instance, frontal lobe involvement or peritumoral edema can lead to emotional lability and reduced impulse control, disrupting daily social interactions.[38][33] Psychiatric symptoms are prevalent in brain tumor patients, affecting approximately 20-30% with conditions such as depression, anxiety, and psychosis. Depression occurs in 2.5-44% of cases, often linked to frontal or temporal tumors, while anxiety frequently coexists and may exacerbate cognitive fog. Psychosis, including delusions or hallucinations, is reported in about 22% of patients, commonly with temporal or pituitary tumors, and can present as the initial symptom in up to 18% of cases. These manifestations arise from disrupted neurotransmitter pathways or limbic system involvement, sometimes mimicking primary psychiatric disorders.[38][42][43] Advanced neurological alterations, such as those from hydrocephalus secondary to tumor obstruction, include gait instability and urinary incontinence as part of the classic triad alongside cognitive slowing. Hydrocephalus elevates intracranial pressure, leading to apraxic gait disturbances and bladder control loss, which can develop over weeks to months in tumor-related cases. Behavioral correlates include increased irritability and apathy due to frontal compression.[44][38] Symptom progression in brain tumors typically worsens gradually over weeks to months, often simulating dementia through progressive memory loss and executive dysfunction or stroke via acute focal deficits from mass effect. This insidious onset contributes to diagnostic delays often lasting several months, particularly when psychiatric features predominate.[42][45] In pediatric patients, brain tumors frequently cause developmental delays and heightened irritability, especially in infants where nonverbal cues like excessive crying signal underlying pressure. Adolescents exhibit elevated rates of depression and anxiety compared to peers, with personality shifts such as aggression or withdrawal in up to 44% of cases at some point during illness. These differences stem from tumors impacting developing neural circuits, leading to broader emotional dysregulation.[46][47]Causes and risk factors
Genetic predispositions
Genetic predispositions to brain tumors involve inherited germline mutations that increase susceptibility, though most cases are sporadic without a clear hereditary component. Certain hereditary syndromes confer elevated risks for specific tumor types, often through disruptions in tumor suppressor genes or signaling pathways. These syndromes account for a minority of brain tumors but highlight the role of genetics in tumorigenesis.[48] Neurofibromatosis type 1 (NF1), caused by mutations in the NF1 gene on chromosome 17, predisposes individuals to optic pathway gliomas and other low-grade gliomas, affecting approximately 1 in 3,000 people with an autosomal dominant inheritance pattern. Neurofibromatosis type 2 (NF2), resulting from NF2 gene mutations on chromosome 22, is associated with schwannomas, meningiomas, and ependymomas, occurring in about 1 in 40,000 individuals and also following autosomal dominant transmission. Li-Fraumeni syndrome, driven by germline TP53 mutations on chromosome 17, significantly raises the risk of gliomas and other brain tumors, with affected individuals facing a lifetime cancer risk exceeding 90%. Turcot syndrome, particularly type 2 linked to APC gene mutations in the Wnt pathway, is characterized by medulloblastomas alongside colorectal cancers, inherited in an autosomal dominant manner.[49][48][49][50] Familial clustering of gliomas is observed in 5-10% of cases, suggesting polygenic or low-penetrance genetic factors rather than single high-impact mutations. Tuberous sclerosis complex (TSC), caused by TSC1 or TSC2 gene mutations on chromosomes 9 and 16 respectively, leads to subependymal giant cell astrocytomas and other hamartomatous brain lesions in up to 80% of patients, with autosomal dominant inheritance and a prevalence of 1 in 6,000. Somatic mutations, such as those in IDH1 and IDH2 genes, are common in low-grade gliomas (occurring in over 70% of cases) and may arise postzygotically, contributing to tumor initiation but primarily serving as diagnostic markers rather than inherited risks.[51][52][53] For the majority of sporadic brain tumors, no single causative gene has been identified, with risk likely stemming from complex interactions of multiple low-effect variants and environmental influences.[54]Environmental and lifestyle factors
Exposure to ionizing radiation is the most well-established environmental risk factor for brain tumors, particularly from therapeutic sources such as cranial radiotherapy for childhood cancers like leukemia. This exposure can increase the risk of meningiomas by 10- to 40-fold, depending on the dose and age at irradiation, with risks persisting for decades post-treatment.[55][56] Data from atomic bomb survivors in Hiroshima and Nagasaki demonstrate a statistically significant linear dose-response relationship for the development of gliomas and meningiomas, with excess relative risks observed even at moderate doses received during the 1945 bombings.[57][58] Electromagnetic fields, particularly from mobile phone use, have been extensively studied but show inconclusive associations with brain tumor risk. The INTERPHONE study, a large international case-control analysis conducted in 2010, found no overall increased risk of glioma or meningioma from regular cell phone use, though it noted a potential elevated risk in the highest exposure categories that may reflect recall bias or other confounders.[59] More recent analyses up to 2025, including trend studies on brain tumor incidence alongside rising mobile phone adoption, continue to support no strong causal link, with stable or declining rates of malignant brain tumors despite widespread exposure.[60][61] Certain chemical exposures have been linked to modest increases in brain tumor risk, though evidence varies by agent. Occupational exposure to pesticides, as observed in farming populations through the Agricultural Health Study, shows suggestive associations with gliomas, particularly among applicators of herbicides like glyphosate or insecticides, with odds ratios around 1.5-2.0 for high-exposure groups.[62][63] Vinyl chloride, a chemical used in plastics manufacturing, has been associated with a possible excess risk of brain tumors in highly exposed workers, based on cohort studies showing standardized mortality ratios up to 2-3 times higher, though confounding by other occupational factors cannot be fully ruled out.[64][65] The relationship with asbestos remains debated, with some older cohort studies of insulation workers reporting slightly elevated brain tumor mortality (e.g., 24 observed vs. 18 expected deaths), but larger reviews finding insufficient evidence for a causal link after adjusting for lifestyle confounders.[66][67] Lifestyle factors generally show weak or no associations with brain tumor development. Smoking and alcohol consumption have not been consistently linked to increased risk, with large prospective studies and meta-analyses reporting no significant elevation in glioma or meningioma incidence among current or former smokers (odds ratios near 1.0) or heavy drinkers.[68][69] Obesity, however, may represent a minor modifiable risk factor, particularly for meningiomas in women, where body mass index greater than 30 kg/m² is associated with a 20-50% higher risk, potentially mediated by hormonal or inflammatory pathways.[70][71] Viral infections play a rare role in brain tumor etiology, with the JC virus (a polyomavirus) implicated in occasional cases through its association with progressive multifocal leukoencephalopathy (PML), a demyelinating condition that can mimic tumor-like lesions on imaging. Experimental and epidemiological evidence suggests JC virus DNA integration in some brain tumors, such as gliomas and medulloblastomas, potentially contributing to oncogenesis via T-antigen expression that disrupts cell cycle control, though human causation remains unproven and limited to immunocompromised individuals.[72][73][74]Pathophysiology
Tumor growth mechanisms
Brain tumors grow through a series of interconnected cellular and molecular processes that enable neoplastic cells to proliferate, invade surrounding tissue, and sustain their expansion despite the brain's constrained environment. These mechanisms include uncontrolled cell division driven by genetic alterations, the formation of new blood vessels to secure nutrients, enzymatic degradation of barriers for infiltration, disruption of vascular integrity leading to swelling, and adaptive responses to oxygen deprivation that promote survival and aggressiveness. In primary brain tumors like gliomas, these processes are particularly pronounced, contributing to the tumors' infiltrative nature and resistance to interventions.[75] Neoplastic transformation in brain tumors begins with genetic changes that confer uncontrolled proliferation, often involving oncogene activation such as epidermal growth factor receptor (EGFR) amplification in glioblastomas. EGFR amplification occurs in approximately 40-57% of primary glioblastomas, leading to overexpression of the receptor on tumor cells—up to 10^6 molecules per cell compared to 10^4-10^5 in normal cells—and constitutive signaling through pathways like PI3K/AKT and RAS/MAPK. This drives cell cycle progression and survival, exemplified by the EGFRvIII mutant variant, which creates autocrine loops with ligands like TGF-α, further amplifying proliferation and neoplastic growth. Such alterations transform normal glial cells into malignant ones, establishing the foundation for tumor expansion.[76][76][77] To support this rapid proliferation, brain tumors induce angiogenesis, primarily through vascular endothelial growth factor (VEGF) secretion, which stimulates the formation of irregular new vessels to deliver oxygen and nutrients. In glioblastomas, VEGF-A expression is upregulated in hypoxic regions via hypoxia-inducible factor-1α (HIF-1α) activation, binding to VEGFR-2 on endothelial cells to promote their proliferation, migration, and tube formation. This results in a hypervascular tumor microenvironment, though the vessels are often leaky and inefficient, perpetuating hypoxia; for instance, VEGF levels increase with glioma grade, correlating with higher vessel density and poorer prognosis.[78][78][78] Invasion into the brain parenchyma is facilitated by matrix metalloproteinases (MMPs), enzymes that degrade the extracellular matrix and allow tumor cells to migrate beyond the primary mass. In gliomas, MMP-2 and MMP-9 are overexpressed, breaking down type IV collagen in basement membranes and tight junctions, with MMP-9 activity significantly elevated in high-grade tumors (p < 0.001) and linked to increased invasiveness. MMP-14 (MT1-MMP) further activates pro-MMP-2, enabling pericellular proteolysis that supports diffuse infiltration characteristic of glioblastomas, contributing to their recurrence despite surgical resection.[79][79][79] Peritumoral edema arises from blood-brain barrier (BBB) disruption, where tumor-secreted factors compromise endothelial integrity, leading to leakage of plasma proteins and fluid into surrounding tissue. In brain tumors like gliomas, VEGF and MMP-9 degrade tight junction proteins such as claudin-5 and occludin, increasing vascular permeability and causing vasogenic edema that can elevate intracranial pressure. Aquaporin-4 upregulation on astrocytes exacerbates water influx, with edema volume correlating with tumor grade and contributing to neurological symptoms.[80][80][80] Hypoxia adaptation in aggressive brain tumors is mediated by HIF-1α, which accumulates in low-oxygen tumor cores to transcriptionally activate genes promoting survival and progression. Under hypoxia, prolyl hydroxylases are inhibited, stabilizing HIF-1α for nuclear translocation and binding to hypoxia-responsive elements, upregulating glycolytic enzymes, VEGF for angiogenesis, and MMPs for invasion. In glioblastomas, HIF-1α expression at the invasive front correlates with higher grade and vessel density, fueling a cycle of hypoxia-driven aggressiveness that limits treatment efficacy.[75][75][75]Impact on brain structures
Brain tumors exert significant physical and functional impacts on surrounding brain structures through their growth and associated physiological alterations. The mass effect generated by tumor expansion compresses adjacent neural tissue, potentially displacing critical structures and increasing intracranial pressure. This compression can lead to life-threatening herniation syndromes, such as subfalcine herniation where the cingulate gyrus shifts under the falx cerebri, or tonsillar herniation involving downward displacement of the cerebellar tonsils through the foramen magnum, both of which risk compressing vital brainstem areas.[81][82] Tumors located near cerebrospinal fluid (CSF) pathways frequently obstruct flow, resulting in hydrocephalus characterized by ventricular enlargement due to accumulated fluid. Obstructive hydrocephalus arises when neoplasms block key sites like the aqueduct of Sylvius or foramina of Monro, impeding the normal circulation of CSF from production in the choroid plexus to absorption in the arachnoid granulations. This leads to dilation of upstream ventricles and secondary compression of periventricular white matter, exacerbating local tissue damage.[44][83] Infiltration or compression by tumors disrupts white matter tracts, which are bundles of myelinated axons facilitating inter-regional communication. Diffusion tensor imaging (DTI), a magnetic resonance technique, reveals these disruptions by quantifying fractional anisotropy—a measure of water diffusion directionality along fiber tracts—showing reduced values in peritumoral regions indicative of tract integrity loss. Such alterations impair neural connectivity, particularly in eloquent areas like the corticospinal tract or optic radiations.[84][85] Breakdown of the blood-brain barrier (BBB), a selective endothelial interface protecting the central nervous system, occurs in response to tumor-secreted factors, allowing plasma leakage and vasogenic edema formation. This edema manifests as extracellular fluid accumulation in the brain parenchyma surrounding the tumor, increasing tissue volume and contributing to further mass effect. The disrupted BBB also facilitates unintended drug extravasation but primarily drives peritumoral swelling that compresses viable neurons and glia.[86][87] Hypoxic regions within rapidly proliferating tumors undergo metabolic shifts, including anaerobic glycolysis and lactate accumulation, which acidify the local microenvironment. In these oxygen-deprived areas, cells rely on lactate dehydrogenase to convert pyruvate to lactate, sustaining energy production but promoting necrosis and inflammation in adjacent brain tissue. This metabolic dysregulation not only supports tumor survival under low oxygen but also indirectly exacerbates structural damage through pH changes and oxidative stress.[88][89]Diagnosis
Imaging modalities
Magnetic resonance imaging (MRI) serves as the primary modality for detecting and characterizing brain tumors due to its superior soft-tissue contrast and multi-parametric capabilities. T1-weighted sequences provide anatomical detail, distinguishing gray and white matter, while T2-weighted and fluid-attenuated inversion recovery (FLAIR) images reveal peritumoral edema and non-enhancing tumor infiltration.[90] Gadolinium contrast enhancement on T1-weighted images evaluates blood-brain barrier disruption, with heterogeneous or ring-like enhancement common in high-grade gliomas indicating neovascularization and necrosis.[90] Diffusion-weighted imaging (DWI) quantifies cellularity through apparent diffusion coefficient (ADC) values, typically lower in high-grade tumors due to restricted water motion in dense cell populations, while perfusion MRI assesses vascular proliferation via elevated relative cerebral blood volume (rCBV) in aggressive lesions.[91] Computed tomography (CT) is often the initial imaging choice in acute settings for its speed and availability, effectively identifying hemorrhage, calcifications (e.g., in oligodendrogliomas), mass effect, or hydrocephalus that may require urgent intervention.[90] However, CT's lower resolution for soft tissues limits its role in detailed tumor characterization, positioning it as a complementary tool to MRI rather than a standalone method.[91] Positron emission tomography (PET) complements anatomical imaging by revealing metabolic activity. 18F-fluorodeoxyglucose (FDG)-PET measures glucose uptake, which is elevated in most tumors but challenged by high physiologic brain background, reducing sensitivity for low-grade lesions.[90] Amino acid tracers like O-(2-[18F]fluoroethyl)-L-tyrosine (FET)-PET provide superior tumor delineation with low background uptake, enabling accurate assessment of tumor extent, differentiation of recurrence from radiation necrosis, and guidance for targeted therapies.[91] Magnetic resonance spectroscopy (MRS) offers biochemical profiling without tissue sampling, focusing on metabolite alterations. Elevated choline (Cho) signals membrane synthesis in proliferating cells, while decreased N-acetylaspartate (NAA) reflects neuronal damage; the Cho/NAA ratio exceeds 1.5 in high-grade gliomas, aiding in grading and monitoring treatment response.[92] Functional MRI (fMRI) utilizes blood-oxygen-level-dependent (BOLD) contrast to map eloquent cortical areas, preserving critical functions during tumor resection planning.[90] By 2025, artificial intelligence (AI) integration has transformed imaging workflows, enabling faster detection through deep learning models that segment and classify tumors on MRI with accuracies up to 98.9% and inference times under 90 seconds.[93] These AI tools also predict molecular features like IDH mutations from standard sequences, supporting non-invasive tumor classification with areas under the curve (AUC) of 0.70 or higher.[94]Biopsy and histopathological analysis
Biopsy procedures are essential for obtaining tissue samples from suspected brain tumors, enabling definitive diagnosis through pathological examination, often guided by preoperative imaging such as MRI or CT scans.[95] These invasive techniques target lesions that cannot be fully characterized by imaging alone, providing material for microscopic and molecular analysis to identify tumor type and guide treatment. Stereotactic biopsy is the preferred method for deep-seated or inaccessible tumors, involving a frame or frameless system to precisely guide a needle to the lesion under imaging guidance.[95] The procedure typically uses a small burr hole in the skull, through which a biopsy needle extracts cylindrical tissue cores, minimizing disruption to surrounding brain tissue.[95] Risks include intracranial hemorrhage in approximately 1-2% of cases and infection in less than 1%.[95][96] For superficial or more accessible tumors, an open biopsy may be performed, involving a craniotomy to directly expose and sample the lesion.[97] This approach allows for larger tissue samples but carries higher risks due to the surgical exposure, though it is often integrated into planned resections.[97] Once obtained, tissue samples undergo histopathological analysis, beginning with hematoxylin and eosin (H&E) staining to evaluate cell morphology, such as nuclear atypia, mitotic activity, and architectural patterns indicative of malignancy.[98] Immunohistochemistry (IHC) enhances specificity; for instance, glial fibrillary acidic protein (GFAP) staining confirms astrocytic differentiation in gliomas by highlighting intermediate filament expression in tumor cells.[99] Molecular testing on biopsy samples further informs prognosis and therapy, particularly assessment of O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status, which predicts response to alkylating chemotherapy like temozolomide in glioblastoma.[100] Methylated MGMT promoters correlate with reduced enzyme activity, enhancing chemotherapy efficacy and improving survival outcomes.[100] Potential complications from biopsy include infection, cerebral edema, and hemorrhage, which can exacerbate neurological deficits and occur in up to 3% of procedures.[101] Intraoperative frozen section analysis mitigates some risks by providing rapid preliminary diagnosis during surgery, allowing immediate adjustments to sampling or procedure extent with high diagnostic accuracy.[102]Tumor classification
Brain tumors are systematically classified using the World Health Organization (WHO) framework, with the 2021 fifth edition marking a pivotal shift by integrating molecular diagnostics with traditional histopathology to define tumor entities more accurately. This approach reorganizes CNS tumors into 11 families, such as adult-type diffuse gliomas and ependymal tumors, emphasizing genetic and epigenetic profiles for diagnosis.[103] A key innovation is the incorporation of molecular markers to differentiate subtypes, exemplified by gliomas: IDH-wildtype glioblastoma, defined by the absence of IDH1/IDH2 mutations and often harboring EGFR amplification or TERT promoter mutations, is uniformly graded as CNS WHO grade 4, whereas IDH-mutant astrocytomas range from grades 2 to 4 based on additional features like CDKN2A/B homozygous deletion for grade 4 escalation. Similarly, oligodendrogliomas require both IDH mutation and complete 1p/19q codeletion for diagnosis, highlighting how these genetic alterations refine classification and inform prognosis within the glioma family.[103] Classification distinguishes primary CNS tumors, which arise de novo within the brain or spinal cord, from secondary tumors, which are metastatic lesions from extracranial primaries such as lung or breast cancer; the latter are not integrated into the CNS WHO schema but graded according to the originating tumor's system. The WHO grading scale, specific to CNS neoplasms, ranges from grade I (slow-growing, often curable by resection) to grade IV (rapidly proliferating, invasive, and associated with necrosis), differing from broader oncologic grading by prioritizing CNS-relevant criteria like microvascular proliferation and molecular aberrations over generic mitotic rates.[103][104] Pediatric brain tumors warrant distinct categorization due to their unique biology, with the 2021 edition establishing separate families like pediatric-type diffuse gliomas and embryonal tumors to reflect age-specific molecular drivers. Embryonal tumors, comprising about 10-20% of pediatric CNS malignancies, include atypical teratoid/rhabdoid tumors (ATRT), aggressive grade 4 entities driven by biallelic inactivation of SMARCB1 or SMARCA4 and subdivided into molecular groups (ATRT-SHH, ATRT-TYR, ATRT-MYC) based on epigenetic profiles that influence location and outcome. This pediatric focus underscores divergences from adult classifications, prioritizing entities like H3 G34-mutant high-grade gliomas over adult-centric IDH subtypes.[105][103]Treatment options
Surgical interventions
Surgical interventions play a central role in the management of brain tumors, primarily aimed at achieving maximal safe resection to alleviate symptoms, confirm diagnosis, and potentially improve survival outcomes. The primary goal is to remove as much tumor tissue as possible while preserving neurological function, particularly in tumors located near eloquent brain areas such as those controlling language, movement, or sensation. Preoperative planning often incorporates imaging modalities to map tumor boundaries and critical structures.[106] Craniotomy remains the standard open surgical approach for brain tumor resection, involving the temporary removal of a portion of the skull to access the tumor. This technique allows for direct visualization and removal of tumors in various locations, including supratentorial and infratentorial regions. For tumors in eloquent areas, awake craniotomy is employed, where the patient is conscious during parts of the procedure to enable real-time functional mapping through tasks like speaking or moving, minimizing postoperative deficits. Intraoperative MRI (iMRI) is frequently integrated with awake craniotomy to provide updated imaging during surgery, enhancing the precision of resection and reducing the risk of incomplete removal.[107][108][109] The extent of resection is a critical determinant of outcomes, with gross total resection (GTR), defined as removal of all visible tumor, compared to subtotal resection (STR), where residual tumor remains. In glioblastoma, achieving greater than 90% resection has been associated with improved survival, with studies showing a stepwise increase in median survival from 11-12 months for STR to 15-18 months or more for GTR. Supramaximal resection, extending beyond visible tumor margins while preserving function, is increasingly pursued in select cases to further enhance prognosis.[110][111][112] Minimally invasive techniques are preferred for deep-seated or ventricular tumors to reduce tissue disruption and recovery time. Endoscopic surgery utilizes a small endoscope inserted through a burr hole to resect tumors in the ventricles or near the skull base, offering visualization without large incisions. Laser interstitial thermal therapy (LITT) involves inserting a laser probe under stereotactic guidance to ablate tumor tissue with heat, achieving cytoreduction with lower morbidity than traditional craniotomy, particularly for recurrent or inoperable lesions.[106][113][114] Surgical risks include infection, neurological deficits, and intraoperative seizures, which can impact patient recovery. Postoperative infection rates following craniotomy average around 6%, often due to factors like prolonged surgery duration or posterior fossa location. New neurological deficits occur in 10-20% of cases, more commonly with resections near eloquent areas, though many resolve over time. Intraoperative seizures affect up to 15-20% of patients during awake procedures but are typically managed with anticonvulsants.[115][116][117] Adjunct technologies enhance surgical accuracy and safety. Neuronavigation systems provide real-time 3D guidance based on preoperative imaging, allowing precise tumor localization and avoidance of vital structures. Fluorescence-guided surgery using 5-aminolevulinic acid (5-ALA), administered orally preoperatively, causes malignant cells to fluoresce under blue light, improving visualization of tumor margins and increasing the rate of complete resection in high-grade gliomas.[118][119][120]Radiation and chemotherapy
Radiation therapy is a cornerstone of non-surgical treatment for brain tumors, particularly for high-grade gliomas and tumors that are inoperable or residual after surgery. External beam radiation therapy (EBRT), including intensity-modulated radiation therapy (IMRT), delivers targeted high-energy beams to the tumor while minimizing exposure to surrounding healthy tissue. Stereotactic radiosurgery, such as Gamma Knife, provides precise, high-dose radiation in a single or few sessions for smaller tumors or metastases. Standard doses for primary brain tumors typically range from 50 to 60 Gy, fractionated over 5 to 6 weeks to balance efficacy and toxicity. Common side effects include fatigue, which often peaks 1 to 2 weeks post-treatment and resolves gradually; headaches; nausea; and skin irritation. Long-term risks encompass radionecrosis, a delayed tissue death in irradiated areas occurring in 5-10% of cases, and cognitive impairments such as memory loss. Chemotherapy employs alkylating agents to damage tumor DNA, with temozolomide (TMZ) as the primary agent for gliomas due to its ability to cross the blood-brain barrier. TMZ methylates DNA at the O6 position of guanine, leading to cell death, but resistance frequently develops via the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT), which removes methyl groups and restores DNA integrity. Tumors with methylated MGMT promoters exhibit greater sensitivity to TMZ, correlating with improved outcomes. The Stupp protocol, established in a landmark phase III trial, integrates concurrent TMZ (75 mg/m² daily) with radiation followed by adjuvant TMZ (150-200 mg/m² for 5 days every 28 days for 6-12 cycles) as the standard for newly diagnosed glioblastoma, extending median survival from 12.1 to 14.6 months compared to radiation alone. Combination regimens enhance efficacy for specific subtypes; for high-grade gliomas, concurrent and adjuvant TMZ with radiation remains the benchmark, while procarbazine, lomustine (CCNU), and vincristine (PCV) chemotherapy is preferred for 1p/19q codeleted anaplastic oligodendrogliomas, yielding a 40% reduction in mortality risk when added to radiation. In codeleted tumors, PCV post-radiation improves median overall survival to over 14 years versus 7.8 years with radiation alone. These approaches are typically sequenced after maximal surgical resection to target residual microscopic disease. Chemotherapeutic agents for brain tumors are administered orally (e.g., TMZ capsules), intravenously (e.g., for PCV components), or intrathecally via lumbar puncture or reservoir for leptomeningeal spread, ensuring direct cerebrospinal fluid delivery to circumvent the blood-brain barrier. Toxicity profiles include myelosuppression, manifesting as low blood counts and increased infection risk, particularly with alkylating agents; nausea and vomiting, managed with antiemetics; and long-term neurocognitive decline, such as deficits in executive function and memory, observed in up to 50% of survivors receiving combined radiation and chemotherapy.Targeted and emerging therapies
Targeted therapies for brain tumors aim to disrupt specific molecular pathways driving tumor growth, offering precision over traditional cytotoxic approaches. Bevacizumab, a monoclonal antibody targeting vascular endothelial growth factor (VEGF), is FDA-approved for recurrent glioblastoma and effectively reduces peritumoral edema by inhibiting angiogenesis, thereby improving neurological symptoms and decreasing corticosteroid dependence in patients with poor performance status.[121][122] Clinical studies have shown radiographic response rates of up to 38% and median progression-free survival of 4 months when combined with chemotherapy for recurrent cases.[123] Epidermal growth factor receptor (EGFR) inhibitors, such as erlotinib, target activating mutations in the EGFR gene, which are present in approximately 40-50% of glioblastomas. These tyrosine kinase inhibitors have demonstrated intracranial activity in EGFR-mutant tumors, particularly in non-small cell lung cancer metastases to the brain, where erlotinib achieves partial responses in 60-80% of cases with exon 19 or 21 mutations.[124] In primary gliomas, efficacy is more limited due to the blood-brain barrier, but select patients with amplified EGFR show tumor stabilization, with phase II trials reporting median progression-free survival of 2-3 months.[125][126] Tumor-treating fields (TTFields) represent a non-invasive device-based therapy that delivers low-intensity alternating electric fields (200 kHz) via a portable scalp array (Optune) to disrupt mitosis in dividing tumor cells. In the phase III EF-14 trial for newly diagnosed glioblastoma, TTFields combined with temozolomide extended median overall survival to 20.9 months from 16.0 months with temozolomide alone, with a hazard ratio of 0.63 for survival benefit.[127] For recurrent glioblastoma, the EF-11 phase III trial showed TTFields monotherapy improved median overall survival to 6.6 months versus 5.2 months with chemotherapy, alongside a favorable safety profile limited mainly to skin irritation.[128][129][130] Hormone therapies are particularly relevant for pituitary adenomas, which often secrete excess hormones. Somatostatin analogs, such as octreotide and lanreotide, bind to somatostatin receptors on tumor cells to suppress growth hormone and thyrotropin secretion, achieving tumor volume reduction in 50-75% of acromegaly and thyrotropinoma cases after 12-24 months of treatment.[131][132] These agents are well-tolerated, with primary side effects including gastrointestinal upset, and serve as first-line medical management for functioning pituitary tumors.[133] Emerging therapies focus on overcoming delivery barriers and enhancing specificity. Nanomedicine approaches, including liposomes and polymeric nanoparticles, facilitate targeted drug delivery across the blood-brain barrier, enabling sustained release of chemotherapeutics like temozolomide directly into tumor tissue and improving efficacy in preclinical glioma models by 2-5 fold compared to free drugs.[134][135] Clinical trials of chimeric antigen receptor (CAR)-T cell therapies targeting antigens like IL13Rα2 or GD2 have shown preliminary tumor regression in 40-60% of glioblastoma and diffuse midline glioma patients, with phase I studies in 2025 reporting durable responses in pediatric cases lasting up to 12 months post-infusion.[136][137] Advances in 2025 have highlighted BRAF inhibitors for tumors harboring BRAF V600E mutations, such as gangliogliomas, which occur in 20-60% of these low-grade glioneuronal tumors. Inhibitors like dabrafenib and vemurafenib, often combined with MEK inhibitors, induce rapid tumor shrinkage in recurrent cases, with case reports documenting over 50% volume reduction within 2-4 weeks and progression-free survival exceeding 12 months in pediatric and adult patients.[138][139] These targeted agents are transforming management of BRAF-altered gliomas, particularly when integrated with standard therapies.[140]Prognosis and outcomes
Prognostic factors
Prognostic factors in brain tumors encompass a range of tumor, patient, and treatment characteristics that influence survival outcomes and quality of life. These factors help clinicians stratify patients for personalized management, with higher tumor grades and adverse molecular profiles generally associated with poorer prognosis.[8] Tumor-related factors play a central role in determining prognosis. The World Health Organization (WHO) grade is a primary determinant, where low-grade tumors (grades I-II) confer a more favorable outlook compared to high-grade ones (grades III-IV), such as glioblastoma, which exhibit aggressive behavior and limited curability. Larger tumor size is linked to worse survival, as larger lesions complicate complete removal and increase recurrence risk. Location also significantly impacts outcomes; tumors in eloquent areas like the brainstem or those crossing the midline are associated with poorer prognosis due to challenges in surgical access and higher risks of neurological deficits.[8][8][141] Molecular markers provide critical prognostic insights, particularly in gliomas. Mutations in IDH1 or IDH2 genes are favorable indicators, correlating with improved survival in diffuse gliomas by altering tumor metabolism and growth patterns. Similarly, 1p/19q codeletion, common in oligodendrogliomas, is a strong positive prognostic factor, often leading to better response to therapy and extended survival. In glioblastoma, MGMT promoter methylation enhances prognosis, with median overall survival reaching 18.2 months versus 12.2 months in unmethylated cases, as it predicts better responsiveness to alkylating agents like temozolomide.[8][8][8] Patient-related factors include age and performance status. Younger age at diagnosis, typically under 40 years, is associated with better survival across tumor types, likely due to greater physiological reserve and tolerance to aggressive treatments. The Karnofsky Performance Status (KPS) score is a validated metric, where scores above 70 indicate good functional status and correlate with prolonged survival, while scores below 70 predict shorter life expectancy and higher treatment complications.[8][141][142] Treatment-related factors, such as the extent of surgical resection, directly affect prognosis. Gross total resection, where feasible, improves survival compared to subtotal resection or biopsy alone by reducing tumor burden and enabling adjuvant therapies. Positive response to radiation or chemotherapy further enhances outcomes, with multimodal approaches extending median survival in high-grade tumors. For glioblastoma, standard therapy yields a median survival of 12-15 months, while low-grade gliomas often achieve over 10 years with appropriate intervention.[8] Quality of life serves as an important prognostic endpoint, particularly regarding neurocognitive function. Post-treatment neurocognitive decline, influenced by tumor location and therapies like whole-brain radiation, can impair daily functioning and correlate with reduced survival; however, targeted agents like bevacizumab may delay such deterioration in glioblastoma patients. Assessments of neurocognitive status and health-related quality of life help predict long-term functional independence.[8][143][144]Survival rates by tumor type
Survival rates for brain tumors vary significantly depending on the histological type, grade, molecular features, and treatment received, with some benign tumors offering excellent long-term outcomes while aggressive malignancies like glioblastoma confer poor prognosis.[4] These rates are derived from large-scale registries and clinical studies, reflecting standard multimodal therapies including surgery, radiation, and chemotherapy.[145] Glioblastoma (WHO grade IV), the most common and aggressive primary brain tumor in adults, has a median overall survival of 12-15 months following maximal safe resection, concurrent temozolomide chemotherapy, and radiotherapy.[145] The 5-year survival rate remains below 10%, with only about 2-5% of patients achieving long-term survival beyond this period, even with optimal treatment.[146] Factors such as MGMT promoter methylation can modestly improve outcomes, but recurrence is nearly universal.[147] Meningioma (predominantly WHO grade I), often benign and arising from the meninges, demonstrates favorable survival, with 5-year overall survival exceeding 90% and 10-year rates around 80-85% for completely resected tumors.[148][149] Recurrence is uncommon after gross total resection, contributing to these high rates, though incomplete removal or higher grades reduce long-term survival to 70-80% at 10 years.[150] Oligodendroglioma, particularly the IDH-mutant and 1p/19q-codeleted subtype (often WHO grade 2 or 3), is associated with relatively good prognosis among diffuse gliomas, with median survival exceeding 15 years for grade 2 tumors and 10-year survival rates over 70% in favorable molecular cases treated with procarbazine, lomustine, and vincristine chemotherapy plus radiation.[151] Without the 1p/19q codeletion or IDH mutation, outcomes worsen significantly, with median survival dropping to under 10 years and 10-year survival below 50%.[152] Metastatic brain tumors, secondary to primaries like lung cancer, have survival influenced by the originating cancer and systemic control, with median overall survival of 6-12 months for non-small cell lung cancer metastases despite whole-brain radiation therapy or stereotactic radiosurgery.[153] For small cell lung cancer brain metastases, median survival is shorter at around 5-7 months, though targeted therapies for driver mutations can extend this in select patients.[154] In pediatric cases, medulloblastoma—the most common malignant brain tumor in children—yields 5-year overall survival rates of 70-80% with multimodal therapy including surgery, craniospinal radiation, and chemotherapy, particularly in average-risk groups without metastasis.[155] High-risk features, such as metastasis or anaplastic histology, reduce this to 50-60%, but molecular subtyping (e.g., WNT-activated) identifies subgroups with over 90% survival.[156]| Tumor Type | Median Survival | 5-Year Survival Rate | Key Reference |
|---|---|---|---|
| Glioblastoma (WHO IV) | 12-15 months | <10% | PMC11719842 |
| Meningioma (WHO I) | >10 years (post-resection) | >90% | PMC10876080 |
| Oligodendroglioma (IDH-mutant/1p19q codeleted) | >15 years | >70% (10-year) | PMC10462563 |
| Brain Metastases (lung primary) | 6-12 months | 20-30% (1-year) | PMC6128200 |
| Pediatric Medulloblastoma | 7-10 years | 70-80% | PubMed32564147 |