Next Article in Journal
Early Detection and Age-Comparative Analysis of Colorectal Cancer Screening: Insights from the Turkish Population
Previous Article in Journal
Real-World Treatment Patterns, Healthcare Resource Utilization, and Healthcare Costs in the First-Line Treatment of Metastatic Non-Small Cell Lung Cancer in the US
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cognitive Decline in Glioblastoma (GB) Patients with Different Treatment Modalities and Insights on Untreated Cases

Department of Neurological Surgery, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Curr. Oncol. 2025, 32(3), 152; https://doi.org/10.3390/curroncol32030152
Submission received: 28 January 2025 / Revised: 28 February 2025 / Accepted: 4 March 2025 / Published: 6 March 2025
(This article belongs to the Section Neuro-Oncology)

Abstract

:
Background: Cognitive decline is common in patients with Glioblastoma (GB), occurring in both treated and untreated cases. It frequently presents as impairments in memory, attention, language, or other cognitive functions. In addition, these cognitive deficits can affect quality of life, functional independence, and overall survival, and they are associated with psychiatric conditions such as anxiety and depression. Methods: This narrative review evaluates cognitive deficits in GB patients, both with and without treatment. It also explores the impact of tumor features such as size, location, and histology, along with patient characteristics such as age and education, and discusses the effects of standard therapies, such as surgery, chemotherapy, and radiotherapy, on cognitive outcomes. Results: Cognitive impairment in GB is influenced by tumor- and patient-specific factors, as well as treatment modalities. Initially, combination therapies such as surgery, radiotherapy, and chemotherapy may improve cognitive domains by reducing tumor burden, relieving cerebral edema, and reducing mass effects, subsequently bringing indirect effects of improved mental health and mood. While certain treatments like radiotherapy and chemotherapy carry risks of delayed neurotoxicity, studies indicate that, on balance, treated patients generally show better preservation or improvement in cognitive function than those who go untreated. However, excessive treatment aggressiveness and cumulative neurotoxic effects may diminish cognitive benefits. Conclusion: Cognitive function is an independent factor in GB, which could affect survival in GB patients, therefore making routine cognitive assessments essential for prognosis, treatment planning, and rehabilitation. Neuroprotective agents, cognitive rehabilitation, and personalized, multidisciplinary strategies can help optimize both survival and cognitive preservation.

1. Introduction

Glioblastoma (GB) is the most common primary brain tumor in adults, accounting for approximately 47.7% of all primary brain tumors [1]. GB has an incidence of about 3.21 per 100,000 people, is more common in males, and has a median onset age of around 64 years [2]. Because GB is known for rapid growth and poor prognosis, it often requires a combination of aggressive interventions (e.g., surgery, radiotherapy), which may in turn affect the patient’s cognitive abilities [3].
GB is classified by the World Health Organization (WHO) as a grade 4 astrocytoma, given its rapid cellular proliferation, diffuse infiltration, robust angiogenesis, and necrosis regions [3,4,5]. It remains a major clinical challenge due to its invasive nature, tumor heterogeneity, and resistance to available therapies [6].

1.1. Pathophysiology

The exact etiology of GB remains incompletely understood, though known risk factors include TP53 mutation, EGFR amplification, PTEN deletion, exposure to ionizing radiation, and various hereditary syndromes [7].
GB is characterized by rapid growth, high invasiveness, and marked genetic heterogeneity [8,9,10]. Molecular profiling differentiates IDH-wildtype grade 4 astrocytomas (known as glioblastomas) from IDH-mutant, WHO grade 4 astrocytomas, which were historically labeled as ‘secondary glioblastomas’. These tumors often harbor IDH1 mutations or EGFR overexpression, contributing to unchecked proliferation and apoptosis resistance [10,11]. Under the 2021 WHO classification, IDH-mutant tumors are now classified as ‘astrocytoma, IDH-mutant, WHO grade 4’ rather than ‘glioblastoma’, although prior studies frequently group them under the broader term glioblastoma.

1.2. Prognosis and Survival Rates

Patients with GB generally face a poor prognosis: the median survival is around three months without treatment [12]. With the current standard of care—maximal safe surgical resection (MSR) plus chemoradiotherapy (e.g., Temozolomide)—the median overall survival can increase to roughly 14.6 months [13]. Despite aggressive therapy, the five-year survival rate remains under 5% [14]. Owing to its infiltrative behavior, complete surgical excision is nearly impossible, and the blood–brain barrier (BBB) further limits the efficacy of systemic chemotherapy [15]. Recurrence is common, typically arising near the original tumor site [16].

1.3. Aim and Objectives

A cognitive deficit is a significant impairment in mental abilities such as memory, attention, reasoning, language processing, or executive function that often hinders daily functioning and reduces the quality of life [17].
Notably, cognitive decline is common among patients with GB before treatment and can affect up to 90% of these patients [18,19]. Such deficits pose significant challenges for both clinicians and patients, making accurate diagnosis more difficult while reducing the quality of life, functional independence, and decision-making capacity [20,21,22].
Therefore, these cognitive impairments strongly affect lifestyle and overall well-being; particularly in the context of a brain tumor, it is crucial to investigate whether treatment exacerbates or mitigates such deficits. Accordingly, this review examines cognitive outcomes in GB patients who receive various therapeutic interventions compared to those who remain untreated.

2. Materials and Methods

We conducted a narrative review evaluating the cognitive outcomes of GB patients before and after treatment. We searched PubMed, MEDLINE, and Web of Science from 2000 to 2024 using the keywords ‘glioblastoma’, ‘cognitive function’, ‘treatment effects’, and ‘quality of life’. Boolean operators (AND, OR) were applied to refine and optimize these search results.
Inclusion criteria included studies of adult patients (≥18 years at diagnosis) with GB that reported cognitive outcomes before and after treatment (surgery, chemotherapy, radiotherapy), or in untreated patients.
Exclusion criteria were pediatric patients (<18 years), absence of cognitive assessments or lack of essential data, and non-English publication.
We extracted data on study design, patient demographics, diagnostic data, treatment modalities, cognitive assessment methods, primary outcomes, tumor characteristics (e.g., size, location), and patient-specific variables (e.g., age, education).
Quality assessment of articles was performed using standardized tools (e.g., CONSORT guidelines) based on the study design, and discrepancies in inclusion criteria were resolved by discussion among reviewers.

3. Cognitive Deficits

3.1. Prevalence and Nature of Cognitive Deficits

Research indicates that GB-related cognitive deficits can affect various domains such as memory, attention, executive functions, language, and visuospatial skills [23,24]. Tucha et al. [24] found that many GB patients already manifest difficulties in attention, memory, and organization before treatment. Likewise, a systematic review by Acevedo-Vergara et al. [17] highlighted that high-grade gliomas often cause substantial cognitive changes in language, attention, memory, empathy, and executive functions (Table 1).

3.2. Impact of Tumor Location on Cognition

Tumor location strongly influences the type and severity of cognitive impairments in GB. Lesions in the frontal or prefrontal lobes may cause deficits in executive functions, such as planning, decision-making, and inhibitory control [22,27]. In untreated patients, frontal tumors often lead to marked executive dysfunction as mass effect and tumor growth proceed unchecked. In contrast, treated patients may fare better, with milder impairments, particularly when surgical resection spares critical areas [28].
Temporal lobe involvement often results in memory impairment and language comprehension issues [20]. Untreated temporal lobe lesions can rapidly degrade cognitive function, especially in verbal memory. Treated patients, however, may retain more capabilities, aided by compensatory neuroplasticity and radiotherapy-induced tumor control [20].
Parietal and occipital lobe tumors can compromise spatial orientation, attention, and visual processing [29]. Untreated disease in these regions can lead to neglect syndromes or cortical blindness, whereas treatment may slow the decline or partially preserve function, due to neurorehabilitation efforts [24]. Additionally, subcortical white matter involvement disrupts neural networks, often causing broad deficits in processing speed and connectivity [30].

3.3. Factors Influencing Cognitive Function

Multiple factors contribute to cognitive impairments in GB, including the following:
Tumor Characteristics: Tumor size, histology, WHO grade, and location correlate strongly with cognitive function [22,31,32]. Larger, more infiltrative tumors worsen outcomes for untreated patients due to a greater mass effect and disruption of surrounding tissue. In contrast, patients with smaller residual tumors post-resection tend to perform better cognitively [24]. Also, IDH-mutant, WHO grade 4 astrocytoma (formerly called secondary GB) often portends longer survival and better cognitive outcomes, irrespective of treatment [33].
Patient Characteristics: Variables such as age, education level, and IDH status influence the extent of cognitive deficits [22,34,35]. Younger treated patients typically display better cognitive resilience than untreated individuals, who may show swifter, more global declines [36].
Treatment Modalities: Maximal safe resection (MSR) remains essential to GB management and is linked to better cognitive outcomes than subtotal or no resection. MSR can reduce tumor burden, ease mass effect, and lower edema, but it carries a risk of damaging healthy tissue in eloquent areas [37,38].
Radiotherapy is standard adjuvant therapy but can cause delayed neurotoxicity, including hippocampus damage that impairs memory [13,39]. Chemotherapy (particularly Temozolomide) boosts survival but may also cause executive and memory deficits [13]. Combination therapy often yields the best survival gains, although cumulative neurotoxic effects may undermine cognition [40,41].
Medication Use: Some medications improve cognition by reducing tumor-related symptoms; others can exacerbate deficits [42,43]. Antiepileptic drugs (AEDs) and corticosteroids can help manage edema and seizures, temporarily improving cognition. Older AEDs (e.g., phenytoin) cause sedation and memory problems, while long-term corticosteroid use can produce mood changes and memory impairment [44,45].
Clinical Symptoms: Depression, anxiety, and neuropsychiatric complications can worsen cognitive function [26,46]. Untreated patients often experience intense psychological distress that magnifies cognitive deficits, whereas treated patients may benefit from better mood and mental health [20,47].

4. Cognitive Impairment and Survival

Cognitive function contributes significantly as a dependent factor to quality of life and independently predicts survival in GB [48,49]. Multiple studies have demonstrated that poorer cognitive performance is linked to reduced survival, independent of other clinical factors [50,51]. Evidence increasingly underscores cognitive function’s prognostic significance, as it often reflects tumor burden, treatment complications, or disease progression. For instance, Meyers et al. observed that verbal memory correlated strongly with survival in recurrent malignant glioma [48]. Johnson et al. showed that early deficits in attention and executive functions predict survival in newly diagnosed GB [35]. Similarly, Klein et al. reported decreased survival in high-grade glioma patients with cognitive impairment, irrespective of therapy (relative risk: 4.099) [50]. These findings suggest that routine baseline and follow-up cognitive assessment can guide therapeutic intensity and balance aggressive treatment with functional preservation.
Hence, cognitive assessments should be integral to GB management, aiding prognosis, guiding therapy, and influencing overall outcomes [15,35].

5. Follow-Up Timing for Patients with GB After Treatment Options

An important component of post-treatment management of GB is timely follow-up to detect cognitive deficits. Current data suggest that impairment may develop within ~3 months after surgery, underlining the need for early cognitive evaluation, as it can be an independent predictor of survival [52]. Because GB typically requires multimodal therapy (surgery, chemotherapy, radiotherapy), follow-up scheduling can be complex. Radiotherapy and chemotherapy both carry a risk of long-term cognitive impairments. Studies recommend follow-up assessments of 6 to 12 months for patients receiving combined modalities [53,54]. Regarding the frequency of evaluations, both immediate and delayed cognitive changes can occur. Thus, monthly check-ups during the first three months are advisable to monitor early decline, followed by evaluations every three months for the next year [52,54].

6. Treatment Options and Their Cognitive Effects

The main goals of GB therapy are prolonging survival, alleviating symptoms, and sustaining or enhancing neurological function via surgery, chemotherapy, radiotherapy, or combined approaches [15]. Although such interventions can improve cognition by reducing mass effects and limiting progression, they also carry risks of further cognitive decline [55].

6.1. Surgical Interventions

6.1.1. Benefits and Risks

MSR is central to GB management [56], often improving cognition by lowering intracranial pressure and mass effect [34]. However, surgery risks harming healthy tissue, leading to postoperative edema and anesthesia-related complications, any of which can worsen cognitive declines [36,57,58].

6.1.2. Techniques to Preserve Cognition

Neurosurgeons employ various techniques to optimize resection while safeguarding cognition. Awake craniotomy enables real-time monitoring of language and motor areas, reducing postoperative deficits [59,60]. Intraoperative brain mapping identifies critical brain areas to avoid [61], and neuronavigation enhances surgical precision [62]. Though these methods often improve survival and preserve neurological function, some risk of cognitive decline remains [37].

6.2. Chemotherapy

6.2.1. Common Agents Used

Temozolomide, the standard chemotherapeutic agent for GB, is typically given alongside radiotherapy [63]. It crosses the BBB and induces DNA damage in tumor cells [64]. Nevertheless, the BBB still limits drug penetration into the cerebrospinal fluid (CSF), promoting investigations into BBB destruction techniques to enhance delivery [64].

6.2.2. Neurotoxic Effects

Temozolomide and related agents improve overall and progression-free survival but can trigger fatigue, concentration difficulties, and other cognitive impairments [65,66]. As an alkylating agent, Temozolomide methylates DNA to kill rapidly dividing tumor cells [64], yet it may also affect non-dividing or slow-dividing cells (including neurons and glia), causing neurotoxicity and cognitive decline [19]. Higher doses correlate with a greater neurotoxic risk [67].
Other chemotherapeutic agents, such as Nitrosoureas, have higher neurotoxicity profiles and are therefore less commonly used [68].

6.3. Radiotherapy

6.3.1. Radiotherapy Approaches

Radiotherapy, often used post-surgically, targets and destroys residual tumor cells. Techniques include external beam radiotherapy, intensity-modulated radiotherapy, and stereotactic radiosurgery [69,70].

6.3.2. Cognitive Side Effects

Radiation may produce both acute and delayed cognitive impairment via white matter necrosis, vascular injury, and neuroinflammation [71,72,73]. Chronic effects, such as memory impairment, can emerge months or even years post-treatment [74]. Fractionation schedules and hippocampal-sparing techniques aim to mitigate these impacts, yet radiotherapy still carries an inherent neurotoxic risk [75,76].

6.4. Combination Therapies

Combining treatments typically yields better survival than monotherapy but may heighten cognitive deficit [36,77]. Therefore, balancing treatment efficacy with quality-of-life goals is critical [78] (Table 2).

6.5. Immunotherapy

Approaches such as immune checkpoint inhibitors, vaccines, and CAR T-cell therapy aim to harness the immune system against GB [79]. Early trials show potential for enhanced efficacy with possibly fewer cognitive side effects [80]. However, recent studies indicate that immune effector cell-associated neurotoxicity syndrome (ICANS) and cytokine release syndrome (CRS), observed in hematologic malignancies treated with CAR T-cells, can lead to delayed or mild cognitive deficits [81,82]. While the incidence of severe persistent impairment is relatively low, patients with more severe ICANS may report worse long-term perceived cognition. Further large-scale trials focusing on primary or metastatic brain tumors are needed to assess the true extent of immunotherapy-related cognitive changes [82,83].
Table 2. Treatment modalities for glioblastoma and their cognitive effects.
Table 2. Treatment modalities for glioblastoma and their cognitive effects.
Treatment ModalityPotential Cognitive BenefitsPotential Cognitive RisksStrategies to Mitigate RisksReferences
SURGICAL RESECTIONReduces mass effect, alleviates symptomsRisk of damage to eloquent brain areasAwake craniotomy, intraoperative mapping[34]
RADIOTHERAPYControls residual tumor growthWhite matter damage, neuroinflammationFractionation schedules, hippocampal-sparing techniques[69,70]
CHEMOTHERAPY
(TEMOZOLOMIDE)
Crosses BBB, prolongs survivalFatigue, concentration difficultiesDose management, supportive care[64]
COMBINED MODALITY THERAPYIncreased efficacy against tumor cellsCompounded neurotoxicityPersonalized treatment plans[36,77]
EXPERIMENTAL THERAPIESPotential for targeted treatmentUnknown long-term cognitive effectsClinical trials, close monitoring[79]
COGNITIVE REHABILITATIONImproves specific cognitive deficitsRequires sustained patient engagementPersonalized rehabilitation programs[84]

7. Comparative Analysis of Cognitive Outcomes

Comparing cognitive outcomes in treated and untreated GB patients is vital for guiding therapy and counseling. Such comparisons provide insights into how treatment modalities might affect cognitive function over time.

7.1. Treated vs. Untreated Patients

Research indicates that treated GB patients frequently experience early stabilization or improvement in cognitive function, attributed to tumor debulking and symptom management [24]. However, neurotoxicity effects from radiation and chemotherapy can contribute to cognitive decline over time, sometimes months or even years after treatment completion [48].
By contrast, in untreated patients, cognitive decline is typically much more rapid and progressive due to the tumor’s aggressive nature, leading to significant tumor growth and quick invasion of the adjacent parenchyma [85]. Acevedo-Vergara et al. reported that high-grade gliomas cause significant alterations in cognitive domains, and patients require neuropsychological evaluation to determine the grade of cognitive dysfunction. Cognitive impairments are generally more severe in high-grade gliomas compared to low-grade gliomas, where brain plasticity processes are faster and more effective, potentially allowing for greater adaptation and less pronounced cognitive difficulties [17].

7.2. Factors Influencing Outcomes

Age, baseline cognition, tumor size, location (especially frontal lobe), and molecular markers all shape cognitive outcomes in GB. While more extensive resection often improves survival, it may heighten cognitive risks. Additionally, more aggressive therapies can escalate side effects [20,43,56,66].

7.3. Importance of Neuropsychological Evaluation

Standard neuropsychological assessments are essential in the management of patients with brain tumors before and after surgery to monitor cognitive function, guide rehabilitation programs, and evaluate treatment outcomes [48]. Performing assessments helps identify specific cognitive deficits and inform interventions aimed at improving quality of life [84].

8. Mitigation Strategies for Cognitive Decline

Addressing the cognitive decline in GB patients involves a comprehensive approach, including advanced medical techniques and supportive therapies.

8.1. Advanced Surgical Techniques

Awake Craniotomy: Reduces the risk of postoperative cognitive deficits by allowing for real-time monitoring [60,61].
Functional Brain Mapping: Guides surgical planning to minimize cognitive risks [59].
Intraoperative Technologies: Neuronavigation and intraoperative imaging enhance surgical precision [37,62].

8.2. Pharmacological Interventions

Research is exploring neuroprotective agents to prevent cognitive decline:
Memantine: Shown to reduce cognitive decline during radiotherapy [86].
Donepezil: May improve cognitive function in irradiated patients [87].
Moreover, a recent systematic review by Kirkman et al. [88] indicates that a variety of pharmacological strategies (e.g., methylphenidate, modafinil, Ginkgo biloba, and Shenqi fuzheng) have been studied in adult brain tumor populations, with some evidence of cognitive benefits. However, many of these studies suffered from methodological limitations or a high risk of bias, and it remains unclear whether improvements persist after cessation of therapy. Future large-scale, randomized trials are needed to confirm durable cognitive gains.

8.3. Rehabilitation Programs

8.3.1. Cognitive Rehabilitation

Structured programs aim to improve specific cognitive deficits through targeted exercises and other strategies [84]. Interventions may focus on the following:
Memory Training: Techniques to enhance recall and retention [18].
Attention and Concentration: Exercises to improve focus [89].
Executive Function: Problem-solving tasks and organizational skills [90].
Studies have demonstrated that cognitive rehabilitation can lead to significant improvements in cognitive performance and daily functioning [84]. For instance, Kirkman et al. [88] identified interventions such as general cognitive rehabilitation, working memory training, goal management training, and aerobic exercise as potentially beneficial, though long-term durability remains uncertain.

8.3.2. Multidisciplinary Support

Comprehensive care includes the involvement of occupational therapists, speech-language pathologists, and neuropsychologists [91]. Support groups and counseling services address emotional and psychological needs [92].

9. Quality-of-Life Considerations

Quality of life (QoL) is a multifaceted construct encompassing physiological, psychological, social, and functional well-being. These domains exhibit intricate interdependencies and are integral to an individual’s overall health status [93]. In the context of GB, cognitive impairment is a critical factor that significantly affects the patient’s ability to perform daily activities, maintain relationships, and engage in meaningful pursuits, ultimately diminishing their overall QoL [94].

9.1. Psychological Impact

Cognitive deficits can lead to depression and anxiety, social isolation, and emotional instability [95]. These issues contribute to social withdrawal and feelings of helplessness, increasing isolation and worsening emotional distress [40].
Providing psychological support, including cognitive–behavioral therapy (CBT) and counseling, is essential to help patients and families cope [96].

9.2. Caregiver Burden

Caregivers of patients with GB often experience significant stress, as they must adapt to the disease progression and the patient’s changing needs and behaviors. The burden includes constant monitoring, assistance with daily tasks, and navigating emotional complexities [97]. Support services and respite care can help alleviate the caregiver’s burden by offering practical assistance and providing emotional support, thereby improving the caregiver’s well-being and ensuring long-term care for the patient [96].

10. Future Directions and Research Opportunities

Advancements in understanding GB biology and cognitive neuroscience offer hope for improved outcomes.

10.1. Novel Therapies

10.1.1. Targeted Molecular Therapies

Drugs targeting specific genetic mutations and pathways in GB cells may improve treatment specificity and reduce toxicity [98]. Examples include the following:
EGFR Inhibitors: Target overexpressed receptors in GB [99].
VEGF Inhibitors: Reduce angiogenesis, e.g., Bevacizumab [100].

10.1.2. Gene Therapy

Gene editing technologies like CRISPR/Cas9 offer avenues for correcting genetic abnormalities in tumor cells [18,101].

10.2. Personalized Medicine

Integrating genomic and molecular profiling into clinical practice enables tailored treatments based on individual tumor characteristics [102]. This approach aims to maximize efficacy and minimize adverse effects, including cognitive decline [103].

10.3. Neuroprotective Strategies

Ongoing research is focused on identifying agents and interventions that can protect neural tissue during treatment [104]. Investigations into the mechanisms of radiation-induced cognitive decline may lead to novel protective measures [105].

10.4. Rehabilitation Innovations

Advancements in neurorehabilitation, such as computerized cognitive training and virtual reality therapies, offer new modalities for cognitive improvement [106]. Research into neuroplasticity may uncover ways to promote brain recovery [107].

10.5. Clinical Trials and Collaborative Research

Participation in clinical trials provides access to innovative therapies and contributes to the collective understanding of GB [108]. Collaborative efforts across institutions enhance research quality and accelerate progress [13].

11. Conclusions

Available evidence confirms that GB—whether treated or untreated—poses a substantial threat to both survival and neurological function. Cognitive decline remains a major concern, undermining independence, social relationships, and overall well-being. While surgical resection, radiotherapy, and chemotherapy can extend survival and may preserve selected cognitive domains, these modalities also carry risks of neurotoxicity, particularly when used in combination or at higher doses. Similarly, immunotherapies show promise in controlling tumor progression, yet immune-related neurotoxicity can occur and may have longer-term implications for cognition.
Recent systematic reviews highlight that various pharmacological and nonpharmacological interventions can help mitigate cognitive deficits in brain tumor populations, although high-quality evidence is still limited. Likewise, studies examining CAR T-cell therapy in hematological malignancies provide preliminary insights into late cognitive changes and suggest close monitoring for neuropsychiatric symptoms. Therefore, a comprehensive, multidisciplinary approach that integrates tumor-directed treatments with neuroprotective strategies, cognitive rehabilitation, and psychosocial support appears essential to optimize outcomes.
Future research should continue pursuing more effective and less neurotoxic treatments, including newer immunotherapies and neuroprotective agents, while standardizing cognitive assessments to refine prognosis and personalize care. In particular, large-scale, collaborative clinical trials with robust neurocognitive endpoints are necessary to confirm the durability of these interventions and better characterize risk factors for long-term impairment.

Author Contributions

Conceptualization, K.G. and E.N.E.; methodology, K.G.; validation, A.K. and I.A.; formal analysis, I.A.; investigation, K.G.; resources, A.K.; writing—original draft preparation, K.G. and A.K.; writing—review and editing, K.G., A.K., I.A., C.C., E.N.E. and A.J.K.; visualization, C.C.; supervision, A.J.K. and E.N.E.; project administration, K.G. and I.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

This study did not create or analyze new data, and data sharing does not apply to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
GBGlioblastoma
WHOWorld Health Organization
MSRMaximal safe surgical resection
BBBBlood–brain barrier
CSFCerebrospinal fluid
QoLQuality of life

References

  1. Grochans, S.; Cybulska, A.M.; Simińska, D.; Korbecki, J.; Kojder, K.; Chlubek, D.; Baranowska-Bosiacka, I. Epidemiology of Glioblastoma Multiforme–Literature Review. Cancers 2022, 14, 2412. [Google Scholar] [CrossRef] [PubMed]
  2. Thakkar, J.P.; Peruzzi, P.P.; Prabhu, V.C. Glioblastoma Multiforme; AANS: Rolling Meadows, IL, USA, 2021. [Google Scholar]
  3. Ostrom, Q.T.; Gittleman, H.; Truitt, G.; Boscia, A.; Kruchko, C.; Barnholtz-Sloan, J.S. Cbtrus Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2011–2015. Neuro-Oncology 2018, 20, iv1–iv86. [Google Scholar] [CrossRef] [PubMed]
  4. Torp, S.H.; Solheim, O.; Skjulsvik, A.J. The WHO 2021 Classification of Central Nervous System Tumours: A Practical Update on What Neurosurgeons Need to Know-a Minireview. Acta Neurochir. 2022, 164, 2453–2464. [Google Scholar] [CrossRef]
  5. Louis, D.N.; Perry, A.; Reifenberger, G.; Von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A Summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef] [PubMed]
  6. Omuro, A.; DeAngelis, L.M. Glioblastoma and Other Malignant Gliomas: A Clinical Review. JAMA 2013, 310, 1842–1850. [Google Scholar] [CrossRef]
  7. Thakkar, J.P.; Dolecek, T.A.; Horbinski, C.; Ostrom, Q.T.; Lightner, D.D.; Barnholtz-Sloan, J.S.; Villano, J.L. Epidemiologic and Molecular Prognostic Review of Glioblastoma. Cancer Epidemiol. Biomark. Prev. 2014, 23, 1985–1996. [Google Scholar] [CrossRef]
  8. Brennan, C.W.; Verhaak, R.G.; McKenna, A.; Campos, B.; Noushmehr, H.; Salama, S.R.; Zheng, S.; Chakravarty, D.; Sanborn, J.Z.; Berman, S.H. The Somatic Genomic Landscape of Glioblastoma. Cell 2013, 155, 462–477. [Google Scholar] [CrossRef]
  9. Ohgaki, H.; Kleihues, P. The Definition of Primary and Secondary Glioblastoma. Clin. Cancer Res. 2013, 19, 764–772. [Google Scholar] [CrossRef]
  10. Furnari, F.B.; Fenton, T.; Bachoo, R.M.; Mukasa, A.; Stommel, J.M.; Stegh, A.; Hahn, W.C.; Ligon, K.L.; Louis, D.N.; Brennan, C. Malignant Astrocytic Glioma: Genetics, Biology, and Paths to Treatment. Genes Dev. 2007, 21, 2683–2710. [Google Scholar] [CrossRef]
  11. Umphlett, M.; Bilal, K.H.; Martini, M.L.; Suwala, A.K.; Ahuja, S.; Rashidipour, O.; Germano, I.; Snuderl, M.; Morgenstern, P.; Tsankova, N.M. Idh-Mutant Astrocytoma with Egfr Amplification-Genomic Profiling in Four Cases and Review of Literature. Neurooncol. Adv. 2022, 4, vdac067. [Google Scholar] [CrossRef]
  12. DeAngelis, L.M. Brain Tumors. N. Engl. J. Med. 2001, 344, 114–123. [Google Scholar] [CrossRef] [PubMed]
  13. Stupp, R.; Mason, W.P.; Van Den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U. Radiotherapy Plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef]
  14. Johnson, D.R.; O’Neill, B.P. Glioblastoma Survival in the United States before and During the Temozolomide Era. J. Neuro-Oncol. 2012, 107, 359–364. [Google Scholar] [CrossRef] [PubMed]
  15. Weller, M.; Van Den Bent, M.; Hopkins, K.; Tonn, J.C.; Stupp, R.; Falini, A.; Cohen-Jonathan-Moyal, E.; Frappaz, D.; Henriksson, R.; Balana, C. EANO Guideline for the Diagnosis and Treatment of Anaplastic Gliomas and Glioblastoma. Lancet Oncol. 2014, 15, e395–e403. [Google Scholar] [CrossRef]
  16. Reardon, D.A.; Wen, P.Y. Therapeutic Advances in the Treatment of Glioblastoma: Rationale and Potential Role of Targeted Agents. Oncology 2006, 11, 152–164. [Google Scholar] [CrossRef]
  17. Acevedo-Vergara, K.; Perez-Florez, M.; Ramirez, A.; Torres-Bayona, S.; Dau, A.; Salva, S.; Maloof, D.; Garcia, C.; Luque, M.; Guillen-Burgos, H.F. Cognitive Deficits in Adult Patients with High-Grade Glioma: A Systematic Review. Clin. Neurol. Neurosurg. 2022, 219, 107296. [Google Scholar] [CrossRef]
  18. Zucchella, C.; Bartolo, M.; Di Lorenzo, C.; Villani, V.; Pace, A. Cognitive Impairment in Primary Brain Tumors Outpatients: A Prospective Cross-Sectional Survey. J. Neuro-Oncol. 2013, 112, 455–460. [Google Scholar] [CrossRef]
  19. Johnson, D.R.; Wefel, J.S. Relationship between Cognitive Function and Prognosis in Glioblastoma. CNS Oncol. 2013, 2, 195–201. [Google Scholar] [CrossRef] [PubMed]
  20. Noll, K.R.; Bradshaw, M.E.; Weinberg, J.S.; Wefel, J.S. Neurocognitive Functioning Is Associated with Functional Independence in Newly Diagnosed Patients with Temporal Lobe Glioma. Neuro-Oncol. Pract. 2018, 5, 184–193. [Google Scholar] [CrossRef]
  21. Guariglia, L.; Ieraci, S.; Villani, V.; Tanzilli, A.; Benincasa, D.; Sperati, F.; Terrenato, I.; Pace, A. Coping Style in Glioma Patients and Their Caregiver: Evaluation During Disease Trajectory. Front. Neurol. 2021, 12, 709132. [Google Scholar] [CrossRef]
  22. Rijnen, S.J.M.; Kaya, G.; Gehring, K.; Verheul, J.B.; Wallis, O.C.; Sitskoorn, M.M.; Rutten, G.-J.M. Cognitive Functioning in Patients with Low-Grade Glioma: Effects of Hemispheric Tumor Location and Surgical Procedure. J. Neurosurg. 2019, 133, 1671–1682. [Google Scholar] [CrossRef] [PubMed]
  23. Barzilai, O.; Moshe, S.B.; Sitt, R.; Sela, G.; Shofty, B.; Ram, Z. Improvement in Cogitive Function after Surgery for Low-Grade Glioma. J. Neurosurg. 2018, 130, 426–434. [Google Scholar] [CrossRef] [PubMed]
  24. Tucha, O.; Smely, C.; Preier, M.; Lange, K.W. Cognitive Deficits before Treatment among Patients with Brain Tumors. Neurosurgery 2000, 47, 324–334. [Google Scholar] [CrossRef] [PubMed]
  25. Aabedi, A.A.; Lipkin, B.; Kaur, J.; Kakaizada, S.; Valdivia, C.; Reihl, S.; Young, J.S.; Lee, A.T.; Krishna, S.; Berger, M.S. Functional Alterations in Cortical Processing of Speech in Glioma-Infiltrated Cortex. Proc. Natl. Acad. Sci. USA 2021, 118, e2108959118. [Google Scholar] [CrossRef]
  26. Armstrong, C.L.; Goldstein, B.; Shera, D.; Ledakis, G.E.; Tallent, E.M. The Predictive Value of Longitudinal Neuropsychologic Assessment in the Early Detection of Brain Tumor Recurrence. Cancer Interdiscip. Int. J. Am. Cancer Soc. 2003, 97, 649–656. [Google Scholar] [CrossRef]
  27. Duffau, H. A New Concept of Diffuse (Low-Grade) Glioma Surgery. Adv. Tech. Stand. Neurosurg. 2012, 38, 3–27. [Google Scholar]
  28. Duffau, H. Brain Plasticity: From Pathophysiological Mechanisms to Therapeutic Applications. J. Clin. Neurosci. 2006, 13, 885–897. [Google Scholar] [CrossRef]
  29. Hart, M.G.; Romero-Garcia, R.; Price, S.J.; Suckling, J. Global Effects of Focal Brain Tumors on Functional Complexity and Network Robustness: A Prospective Cohort Study. Neurosurgery 2019, 84, 1201–1213. [Google Scholar] [CrossRef]
  30. van Kessel, E.; Baumfalk, A.E.; van Zandvoort, M.J.; Robe, P.A.; Snijders, T.J. Tumor-Related Neurocognitive Dysfunction in Patients with Diffuse Glioma: A Systematic Review of Neurocognitive Functioning Prior to Anti-Tumor Treatment. J. Neuro-Oncol. 2017, 134, 9–18. [Google Scholar] [CrossRef]
  31. Wang, Q.; Xiao, F.; Qi, F.; Song, X.; Yu, Y. Risk Factors for Cognitive Impairment in High-Grade Glioma Patients Treated with Postoperative Radiochemotherapy. Cancer Res. Treat. Off. J. Korean Cancer Assoc. 2020, 52, 586–593. [Google Scholar] [CrossRef]
  32. Coomans, M.B.; Dirven, L.; Taphoorn, M.J. Quality of Life and Cognition. In Oncology of CNS Tumors; Springer: Berlin/Heidelberg, Germany, 2019; pp. 769–786. [Google Scholar]
  33. Liu, Y.; Gao, D.; Chen, H.; Zhang, J.; Yao, K.; Wu, C.; Li, S.; Yan, W.; Qiu, X. Idh-Mutant Grade 4 Astrocytoma: A Comparison Integrating the Clinical, Pathological, and Survival Features between Primary and Secondary Patients. J. Neurosurg. 2024, 140, 94–103. [Google Scholar] [CrossRef] [PubMed]
  34. Duffau, H. Surgery of Low-Grade Gliomas: Towards a ‘Functional Neurooncology’. Curr. Opin. Oncol. 2009, 21, 543–549. [Google Scholar] [CrossRef]
  35. Johnson, D.R.; Sawyer, A.M.; Meyers, C.A.; O’Neill, B.P.; Wefel, J.S. Early Measures of Cognitive Function Predict Survival in Patients with Newly Diagnosed Glioblastoma. Neuro-Oncology 2012, 14, 808–816. [Google Scholar] [CrossRef] [PubMed]
  36. Taphoorn, M.J.; Klein, M. Cognitive Deficits in Adult Patients with Brain Tumours. Lancet Neurol. 2004, 3, 159–168. [Google Scholar] [CrossRef] [PubMed]
  37. Sanai, N.; Berger, M.S. Surgical Oncology for Gliomas: The State of the Art. Nat. Rev. Clin. Oncol. 2018, 15, 112–125. [Google Scholar] [CrossRef]
  38. Hardesty, D.A.; Sanai, N. The Value of Glioma Extent of Resection in the Modern Neurosurgical Era. Front. Neurol. 2012, 3, 140. [Google Scholar] [CrossRef]
  39. Brown, P.D.; Gondi, V.; Pugh, S.; Tome, W.A.; Wefel, J.S.; Armstrong, T.S.; Bovi, J.A.; Robinson, C.; Konski, A.; Khuntia, D. Hippocampal Avoidance During Whole-Brain Radiotherapy Plus Memantine for Patients with Brain Metastases: Phase III Trial Nrg Oncology CC001. J. Clin. Oncol. 2020, 38, 1019–1029. [Google Scholar] [CrossRef]
  40. Cavers, D.; Hacking, B.; Erridge, S.E.; Kendall, M.; Morris, P.G.; Murray, S.A. Social, Psychological and Existential Well-Being in Patients with Glioma and Their Caregivers: A Qualitative Study. CMAJ 2012, 184, E373–E382. [Google Scholar] [CrossRef]
  41. Weller, M.; Wick, W.; Aldape, K.; Brada, M.; Berger, M.; Pfister, S.M.; Nishikawa, R.; Rosenthal, M.; Wen, P.Y.; Stupp, R. Glioma. Nat. Rev. Dis. Primers 2015, 1, 15017. [Google Scholar] [CrossRef]
  42. Wefel, J.S.; Noll, K.R.; Scheurer, M.E. Neurocognitive Functioning and Genetic Variation in Patients with Primary Brain Tumours. Lancet Oncol. 2016, 17, e97–e108. [Google Scholar] [CrossRef]
  43. Klein, M.; Engelberts, N.H.; van der Ploeg, H.M.; Kasteleijn-Nolst Trenité, D.G.; Aaronson, N.K.; Taphoorn, M.J.B.; Baaijen, H.; Vandertop, W.P.; Muller, M.; Postma, T.J. Epilepsy in Low-Grade Gliomas: The Impact on Cognitive Function and Quality of Life. Ann. Neurol. Off. J. Am. Neurol. Assoc. Child Neurol. Soc. 2003, 54, 514–520. [Google Scholar] [CrossRef] [PubMed]
  44. Bootsma, H.P.R.; Aldenkamp, A.P.; Diepman, L.; Hulsman, J.; Lambrechts, D.; Leenen, L.; Majoie, M.; Schellekens, A.; De Krom, M. The Effect of Antiepileptic Drugs on Cognition: Patient Perceived Cognitive Problems of Topiramate Versus Levetiracetam in Clinical Practice. Epilepsia 2006, 47, 24–27. [Google Scholar] [CrossRef] [PubMed]
  45. Weller, M.; Cloughesy, T.; Perry, J.R.; Wick, W. Standards of Care for Treatment of Recurrent Glioblastoma—Are We There Yet? Neuro-Oncology 2013, 15, 4–27. [Google Scholar] [CrossRef]
  46. Boele, F.W.; Klein, M.; Reijneveld, J.C.; Verdonck-de Leeuw, I.M.; Heimans, J.J. Symptom Management and Quality of Life in Glioma Patients. CNS Oncol. 2014, 3, 37–47. [Google Scholar] [CrossRef] [PubMed]
  47. Renovanz, M.; Maurer, D.; Lahr, H.; Weimann, E.; Deininger, M.; Wirtz, C.R.; Ringel, F.; Singer, S.; Coburger, J. Supportive Care Needs in Glioma Patients and Their Caregivers in Clinical Practice: Results of a Multicenter Cross-Sectional Study. Front. Neurol. 2018, 9, 763. [Google Scholar] [CrossRef]
  48. Meyers, C.A.; Hess, K.R.; Yung, W.A.; Levin, V.A. Cognitive Function as a Predictor of Survival in Patients with Recurrent Malignant Glioma. J. Clin. Oncol. 2000, 18, 646. [Google Scholar] [CrossRef]
  49. Gorlia, T.; van den Bent, M.J.; Hegi, M.E.; Mirimanoff, R.O.; Weller, M.; Cairncross, J.G.; Eisenhauer, E.; Belanger, K.; Brandes, A.A.; Allgeier, A. Nomograms for Predicting Survival of Patients with Newly Diagnosed Glioblastoma: Prognostic Factor Analysis of Eortc and Ncic trial 26981-22981/CE. 3. Lancet Oncol. 2008, 9, 29–38. [Google Scholar] [CrossRef]
  50. Klein, M.; Postma, T.; Taphoorn, M.; Aaronson, N.; Vandertop, W.; Muller, M.; Van der Ploeg, H.; Heimans, J. The Prognostic Value of Cognitive Functioning in the Survival of Patients with High-Grade Glioma. Neurology 2003, 61, 1796–1798. [Google Scholar] [CrossRef]
  51. Li, J.; Bentzen, S.M.; Li, J.; Renschler, M.; Mehta, M.P. Relationship between Neurocognitive Function and Quality of Life after Whole-Brain Radiotherapy in Patients with Brain Metastasis. Int. J. Radiat. Oncol. Biol. Phys. 2008, 71, 64–70. [Google Scholar] [CrossRef]
  52. Butterbrod, E.; Synhaeve, N.; Rutten, G.-J.; Schwabe, I.; Gehring, K.; Sitskoorn, M. Cognitive Impairment Three Months after Surgery Is an Independent Predictor of Survival Time in Glioblastoma Patients. J. Neuro-Oncol. 2020, 149, 103–111. [Google Scholar] [CrossRef]
  53. Sharma, P.; Medhi, P.P.; Kalita, A.K.; Bhattacharyya, M.; Nath, J.; Sarma, G.; Yanthan, Y. Factors Associated with Neurocognitive Impairment Following Chemoradiotherapy in Patients with High-Grade Glioma: Results of a Prospective Trial. Brain Tumor Res. Treat. 2023, 11, 183–190. [Google Scholar] [CrossRef]
  54. Schagen, S.B.; Wefel, J.S. Chemotherapy-Related Changes in Cognitive Functioning. Eur. J. Cancer Suppl. 2013, 11, 225–232. [Google Scholar] [CrossRef]
  55. Douw, L.; Klein, M.; Fagel, S.S.; van den Heuvel, J.; Taphoorn, M.J.; Aaronson, N.K.; Postma, T.J.; Vandertop, W.P.; Mooij, J.J.; Boerman, R.H. Cognitive and Radiological Effects of Radiotherapy in Patients with Low-Grade Glioma: Long-Term Follow-Up. Lancet Neurol. 2009, 8, 810–818. [Google Scholar] [CrossRef] [PubMed]
  56. Sanai, N.; Berger, M.S. Glioma Extent of Resection and Its Impact on Patient Outcome. Neurosurgery 2008, 62, 753–766. [Google Scholar] [CrossRef]
  57. Meskal, I.; Gehring, K.; Rutten, G.-J.M.; Sitskoorn, M.M. Cognitive Functioning in Meningioma Patients: A Systematic Review. J. Neuro-Oncol. 2016, 128, 195–205. [Google Scholar] [CrossRef] [PubMed]
  58. Gempt, J.; Förschler, A.; Buchmann, N.; Pape, H.; Ryang, Y.-M.; Krieg, S.M.; Zimmer, C.; Meyer, B.; Ringel, F. Postoperative Ischemic Changes Following Resection of Newly Diagnosed and Recurrent Gliomas and Their Clinical Relevance. J. Neurosurg. 2013, 118, 801–808. [Google Scholar] [CrossRef] [PubMed]
  59. Berger, M.S.; Hadjipanayis, C.G. Surgery of Intrinsic Cerebral Tumors. Neurosurgery 2007, 61, 305. [Google Scholar] [CrossRef]
  60. Duffau, H. Awake Surgery for Nonlanguage Mapping. Neurosurgery 2010, 66, 523–529. [Google Scholar] [CrossRef]
  61. De Witt Hamer, P.C.; Robles, S.G.; Zwinderman, A.H.; Duffau, H.; Berger, M.S. Impact of Intraoperative Stimulation Brain Mapping on Glioma Surgery Outcome: A Meta-Analysis. J. Clin. Oncol. 2012, 30, 2559–2565. [Google Scholar] [CrossRef]
  62. Nimsky, C.; Ganslandt, O.; Buchfelder, M.; Fahlbusch, R. Intraoperative Visualization for Resection of Gliomas: The Role of Functional Neuronavigation and Intraoperative 1.5 T MRI. Neurol. Res. 2006, 28, 482–487. [Google Scholar] [CrossRef]
  63. Stupp, R.; Hegi, M.E.; Mason, W.P.; Van Den Bent, M.J.; Taphoorn, M.J.; Janzer, R.C.; Ludwin, S.K.; Allgeier, A.; Fisher, B.; Belanger, K. Effects of Radiotherapy with Concomitant and Adjuvant Temozolomide Versus Radiotherapy Alone on Survival in Glioblastoma in a Randomised Phase III Study: 5-year Analysis of the Eortc-Ncic Trial. Lancet Oncol. 2009, 10, 459–466. [Google Scholar] [CrossRef] [PubMed]
  64. Hegi, M.E.; Diserens, A.-C.; Gorlia, T.; Hamou, M.-F.; De Tribolet, N.; Weller, M.; Kros, J.M.; Hainfellner, J.A.; Mason, W.; Mariani, L. MGMT Gene Silencing and Benefit from Temozolomide in Glioblastoma. N. Engl. J. Med. 2005, 352, 997–1003. [Google Scholar] [CrossRef] [PubMed]
  65. Aygun, C.; Altınok, A.Y.; Çakır, A.; Agan, A.F.; Balaban, Y. Acute Temozolomide Induced Liver Injury: Mixed Type Hepatocellular and Cholestatic Toxicity. Acta Gastro-Enterol. Belg. 2016, 79, 363–365. [Google Scholar]
  66. Batchelor, T.T.; Betensky, R.A.; Esposito, J.M.; Pham, L.-D.D.; Dorfman, M.V.; Piscatelli, N.; Jhung, S.; Rhee, D.; Louis, D.N. Age-Dependent Prognostic Effects of Genetic Alterations in Glioblastoma. Clin. Cancer Res. 2004, 10, 228–233. [Google Scholar] [CrossRef]
  67. Chamberlain, M.C.; Kormanik, P.A. Practical Guidelines for the Treatment of Malignant Gliomas. West. J. Med. 1998, 168, 114. [Google Scholar]
  68. Walker, M.D.; Green, S.B.; Byar, D.P.; Alexander, E., Jr.; Batzdorf, U.; Brooks, W.H.; Hunt, W.E.; MacCarty, C.S.; Mahaley, M.S., Jr.; Mealey, J., Jr. Randomized Comparisons of Radiotherapy and Nitrosoureas for the Treatment of Malignant Glioma after Surgery. N. Engl. J. Med. 1980, 303, 1323–1329. [Google Scholar] [CrossRef]
  69. Brown, P.D.; Buckner, J.C.; O’Fallon, J.R.; Iturria, N.L.; Brown, C.A.; O’Neill, B.P.; Scheithauer, B.W.; Dinapoli, R.P.; Arusell, R.M.; Curran, W.J. Effects of Radiotherapy on Cognitive Function in Patients with low-Grade Glioma Measured by the Folstein Mini-Mental State Examination. J. Clin. Oncol. 2003, 21, 2519–2524. [Google Scholar] [CrossRef]
  70. Giordano, F.A.; Brehmer, S.; Abo-Madyan, Y.; Welzel, G.; Sperk, E.; Keller, A.; Schneider, F.; Clausen, S.; Herskind, C.; Schmiedek, P. Intrago: Intraoperative Radiotherapy in Glioblastoma Multiforme–A Phase I/II Dose Escalation Study. BMC Cancer 2014, 14, 992. [Google Scholar] [CrossRef]
  71. Monje, M.L.; Palmer, T. Radiation Injury and Neurogenesis. Curr. Opin. Neurol. 2003, 16, 129–134. [Google Scholar] [CrossRef]
  72. Greene-Schloesser, D.; Robbins, M.E. Radiation-Induced Cognitive Impairment-from Bench to Bedside. Neuro-Oncol. 2012, 14, iv37–iv44. [Google Scholar] [CrossRef]
  73. Makale, M.T.; McDonald, C.R.; Hattangadi-Gluth, J.A.; Kesari, S. Mechanisms of Radiotherapy-Associated Cognitive Disability in Patients with Brain Tumours. Nat. Rev. Neurol. 2017, 13, 52–64. [Google Scholar] [CrossRef] [PubMed]
  74. Crossen, J.R.; Garwood, D.; Glatstein, E.; Neuwelt, E.A. Neurobehavioral Sequelae of Cranial Irradiation in Adults: A Review of Radiation-Induced Encephalopathy. J. Clin. Oncol. 1994, 12, 627–642. [Google Scholar] [CrossRef] [PubMed]
  75. Gondi, V.; Pugh, S.L.; Tome, W.A.; Caine, C.; Corn, B.; Kanner, A.; Rowley, H.; Kundapur, V.; DeNittis, A.; Greenspoon, J.N. Preservation of Memory with Conformal Avoidance of the Hippocampal Neural Stem-Cell Compartment During Whole-Brain Radiotherapy for Brain Metastases (Rtog 0933): A Phase Ii Multi-Institutional Trial. J. Clin. Oncol. 2014, 32, 3810–3816. [Google Scholar] [CrossRef]
  76. Redmond, K.J.; Mehta, M. Stereotactic Radiosurgery for Glioblastoma. Cureus 2015, 7, e413. [Google Scholar] [CrossRef] [PubMed]
  77. Bae, S.H.; Park, M.-J.; Lee, M.M.; Kim, T.M.; Lee, S.-H.; Cho, S.Y.; Kim, Y.-H.; Kim, Y.J.; Park, C.-K.; Kim, C.-Y. Toxicity Profile of Temozolomide in the Treatment of 300 Malignant Glioma Patients in Korea. J. Korean Med. Sci. 2014, 29, 980–984. [Google Scholar] [CrossRef]
  78. Armstrong, T.S.; Wefel, J.S.; Wang, M.; Gilbert, M.R.; Won, M.; Bottomley, A.; Mendoza, T.R.; Coens, C.; Werner-Wasik, M.; Brachman, D.G. Net Clinical Benefit Analysis of Radiation Therapy Oncology Group 0525: A Phase III Trial Comparing Conventional Adjuvant Temozolomide with Dose-Intensive Temozolomide in Patients with Newly Diagnosed Glioblastoma. J. Clin. Oncol. 2013, 31, 4076–4084. [Google Scholar] [CrossRef]
  79. Reardon, D.A.; Brandes, A.A.; Omuro, A.; Mulholland, P.; Lim, M.; Wick, A.; Baehring, J.; Ahluwalia, M.S.; Roth, P.; Bähr, O. Effect of Nivolumab vs Bevacizumab in Patients with Recurrent Glioblastoma: The Checkmate 143 phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1003–1010. [Google Scholar] [CrossRef]
  80. Jackson, C.M.; Lim, M.; Drake, C.G. Immunotherapy for Brain Cancer: Recent Progress and Future Promise. Clin. Cancer Res. 2014, 20, 3651–3659. [Google Scholar] [CrossRef]
  81. Ruark, J.; Mullane, E.; Cleary, N.; Cordeiro, A.; Bezerra, E.D.; Wu, V.; Voutsinas, J.; Shaw, B.E.; Flynn, K.E.; Lee, S.J. Patient-Reported Neuropsychiatric Outcomes of Long-Term Survivors after Chimeric Antigen Receptor T Cell Therapy. Biol. Blood Marrow Transplant. 2020, 26, 34–43. [Google Scholar] [CrossRef]
  82. Barata, A.; Hoogland, A.I.; Kommalapati, A.; Logue, J.; Welniak, T.; Hyland, K.A.; Eisel, S.L.; Small, B.J.; Jayani, R.V.; Booth-Jones, M. Change in Patients’ Perceived Cognition Following Chimeric Antigen Receptor T-Cell Therapy for Lymphoma. Transplant. Cell Ther. 2022, 28, e401–e407. [Google Scholar] [CrossRef]
  83. Kazzi, C.; Kuznetsova, V.; Siriratnam, P.; Griffith, S.; Wong, S.; Tam, C.S.; Alpitsis, R.; Spencer, A.; O’Brien, T.J.; Malpas, C.B. Cognition Following Chimeric Antigen Receptor T-Cell Therapy: A Systematic Review. J. Autoimmun. 2023, 140, 103126. [Google Scholar] [CrossRef] [PubMed]
  84. Gehring, K.; Sitskoorn, M.M.; Aaronson, N.K.; Taphoorn, M.J. Interventions for Cognitive Deficits in Adults with Brain Tumours. Lancet Neurol. 2008, 7, 548–560. [Google Scholar] [CrossRef]
  85. Deutsch, M.B.; DeAngelis, L.M. Neurologic Complications of Chemotherapy and Radiation Therapy. Aminoff’s Neurol. Gen. Med. 2014, 21, 591–609. [Google Scholar]
  86. Brown, P.D.; Pugh, S.; Laack, N.N.; Wefel, J.S.; Khuntia, D.; Meyers, C.; Choucair, A.; Fox, S.; Suh, J.H.; Roberge, D. Memantine for the Prevention of Cognitive Dysfunction in Patients Receiving Whole-Brain Radiotherapy: A Randomized, Double-Blind, Placebo-Controlled Trial. Neuro-Oncology 2013, 15, 1429–1437. [Google Scholar] [CrossRef] [PubMed]
  87. Shaw, E.G.; Rosdhal, R.; D’Agostino, R.B., Jr.; Lovato, J.; Naughton, M.J.; Robbins, M.E.; Rapp, S.R. Phase II Study of Donepezil in Irradiated Brain Tumor Patients: Effect on Cognitive Function, Mood, and Quality of Life. J. Clin. Oncol. 2006, 24, 1415–1420. [Google Scholar] [CrossRef]
  88. Kirkman, M.A.; Ekert, J.O.; Hunn, B.H.; Thomas, M.S.; Tolmie, A.K. A Systematic Review of Cognitive Interventions for Adult Patients with Brain Tumours. Cancer Med. 2023, 12, 11191–11210. [Google Scholar] [CrossRef]
  89. Kesler, S.R.; Lacayo, N.J.; Jo, B. A Pilot Study of an Online Cognitive Rehabilitation Program for Executive Function Skills in Children with Cancer-Related Brain Injury. Brain Inj. 2011, 25, 101–112. [Google Scholar] [CrossRef]
  90. Locke, D.; Cerhan, J.H.; Wu, W.; Malec, J.F.; Clark, M.M.; Rummans, T.A.; Brown, P.D. Cognitive Rehabilitation and Problem-Solving to Improve Quality of Life of Patients with Primary Brain Tumors: A Pilot Study. J. Support Oncol. 2008, 6, 383–391. [Google Scholar]
  91. Bartolo, M.; Zucchella, C.; Pace, A.; Lanzetta, G.; Vecchione, C.; Bartolo, M.; Grillea, G.; Serrao, M.; Tassorelli, C.; Sandrini, G. Early Rehabilitation after Surgery Improves Functional Outcome in Inpatients with Brain Tumours. J. Neuro-Oncol. 2012, 107, 537–544. [Google Scholar] [CrossRef]
  92. Moore, G.; Collins, A.; Brand, C.; Gold, M.; Lethborg, C.; Murphy, M.; Sundararajan, V.; Philip, J. Palliative and Supportive Care Needs of Patients with High-Grade Glioma and Their Carers: A Systematic Review of Qualitative Literature. Patient Educ. Couns. 2013, 91, 141–153. [Google Scholar] [CrossRef]
  93. Aaronson, N.K.; Taphoorn, M.J.; Heimans, J.J.; Postma, T.J.; Gundy, C.M.; Beute, G.N.; Slotman, B.J.; Klein, M. Compromised Health-Related Quality of Life in Patients with Low-Grade Glioma. J. Clin. Oncol. 2011, 29, 4430–4435. [Google Scholar] [CrossRef] [PubMed]
  94. Armstrong, T.; Mendoza, T.; Gring, I.; Coco, C.; Cohen, M.; Eriksen, L.; Hsu, M.-A.; Gilbert, M.; Cleeland, C. Validation of the Md Anderson Symptom Inventory Brain Tumor Module (Mdasi-Bt). J. Neuro-Oncol. 2006, 80, 27–35. [Google Scholar] [CrossRef] [PubMed]
  95. Rooney, A.G.; Carson, A.; Grant, R. Depression in Cerebral Glioma Patients: A Systematic Review of Observational Studies. J. Natl. Cancer Inst. 2011, 103, 61–76. [Google Scholar] [CrossRef] [PubMed]
  96. Sherwood, P.R.; Given, B.A.; Given, C.W.; Schiffman, R.F.; Murman, D.L.; Lovely, M.; Von Eye, A.; Rogers, L.R.; Remer, S. Predictors of Distress in Caregivers of Persons with a Primary Malignant Brain Tumor. Res. Nurs. Health 2006, 29, 105–120. [Google Scholar] [CrossRef]
  97. Peeters, M.; Ottenheijm, G.; Bienfait, P.; Eekers, D.; Gijtenbeek, A.; Hanse, M.; Koekkoek, J.; van Leeuwen, L.; Tijssen, C.; Dirven, L. Glioma Patient-Reported Outcomes: Patients and Clinicians. BMJ Support. Palliat. Care 2023, 13, e205–e212. [Google Scholar] [CrossRef]
  98. Cloughesy, T.; Finocchiaro, G.; Belda-Iniesta, C.; Recht, L.; Brandes, A.A.; Pineda, E.; Mikkelsen, T.; Chinot, O.L.; Balana, C.; Macdonald, D.R. Randomized, Double-Blind, Placebo-Controlled, Multicenter Phase Ii Study of Onartuzumab Plus Bevacizumab Versus Placebo Plus Bevacizumab in Patients with Recurrent Glioblastoma: Efficacy, Safety, and Hepatocyte Growth Factor and O6-Methylguanine–DNA Methyltransferase Biomarker Analyses. J. Clin. Oncol. 2017, 35, 343–351. [Google Scholar]
  99. Touat, M.; Idbaih, A.; Sanson, M.; Ligon, K. Glioblastoma Targeted Therapy: Updated Approaches from Recent Biological Insights. Ann. Oncol. 2017, 28, 1457–1472. [Google Scholar] [CrossRef]
  100. Friedman, H.S.; Prados, M.D.; Wen, P.Y.; Mikkelsen, T.; Schiff, D.; Abrey, L.E.; Yung, W.A.; Paleologos, N.; Nicholas, M.K.; Jensen, R. Bevacizumab Alone and in Combination with Irinotecan in Recurrent Glioblastoma. J. Clin. Oncol. 2009, 27, 4733–4740. [Google Scholar] [CrossRef]
  101. Zucchella, C.; Capone, A.; Codella, V.; De Nunzio, A.M.; Vecchione, C.; Sandrini, G.; Pace, A.; Pierelli, F.; Bartolo, M. Cognitive Rehabilitation for Early Post-Surgery Inpatients Affected by Primary Brain Tumor: A Randomized, Controlled Trial. J. Neuro-Oncol. 2013, 114, 93–100. [Google Scholar] [CrossRef]
  102. Verhaak, R.G.; Hoadley, K.A.; Purdom, E.; Wang, V.; Qi, Y.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Mesirov, J.P. Integrated Genomic Analysis Identifies Clinically Relevant Subtypes of Glioblastoma Characterized by Abnormalities in Pdgfra, Idh1, Egfr, and Nf1. Cancer Cell 2010, 17, 98–110. [Google Scholar] [CrossRef]
  103. Beroukhim, R.; Mermel, C.H.; Porter, D.; Wei, G.; Raychaudhuri, S.; Donovan, J.; Barretina, J.; Boehm, J.S.; Dobson, J.; Urashima, M. The Landscape of Somatic Copy-Number Alteration across Human Cancers. Nature 2010, 463, 899–905. [Google Scholar] [CrossRef] [PubMed]
  104. Dietrich, J.; Rao, K.; Pastorino, S.; Kesari, S. Corticosteroids in Brain Cancer Patients: Benefits and Pitfalls. Expert Rev. Clin. Pharmacol. 2011, 4, 233–242. [Google Scholar] [CrossRef] [PubMed]
  105. Greene-Schloesser, D.; Moore, E.; Robbins, M.E. Molecular Pathways: Radiation-Induced Cognitive Impairment. Clin. Cancer Res. 2013, 19, 2294–2300. [Google Scholar] [CrossRef]
  106. Willard, V.W.; Cox, L.E.; Russell, K.M.; Kenney, A.; Jurbergs, N.; Molnar, A.E., Jr.; Harman, J.L. Cognitive and Psychosocial Functioning of Preschool-Aged Children with Cancer. J. Dev. Behav. Pediatr. 2017, 38, 638–645. [Google Scholar] [CrossRef] [PubMed]
  107. Kannan, S.; Murugan, A.K.; Balasubramanian, S.; Munirajan, A.K.; Alzahrani, A.S. Gliomas: Genetic Alterations, Mechanisms of Metastasis, Recurrence, Drug Resistance, and Recent Trends in Molecular Therapeutic Options. Biochem. Pharmacol. 2022, 201, 115090. [Google Scholar] [CrossRef]
  108. O’connor, J.; Rose, C.; Jackson, A.; Watson, Y.; Cheung, S.; Maders, F.; Whitcher, B.; Roberts, C.; Buonaccorsi, G.; Thompson, G. Dce-Mri Biomarkers of Tumour Heterogeneity Predict Crc Liver Metastasis Shrinkage Following Bevacizumab and Folfox-6. Br. J. Cancer 2011, 105, 139–145. [Google Scholar] [CrossRef]
Table 1. Cognitive domains affected in glioblastoma patients and influencing factors.
Table 1. Cognitive domains affected in glioblastoma patients and influencing factors.
Cognitive Domain Common Impairments Influencing Factors References
MEMORYShort-term memory loss, difficulty recallingTemporal lobe involvement, tumor size[24]
ATTENTIONReduced concentration, distractibilityFrontal and parietal lobe involvement[22]
EXECUTIVE FUNCTIONSImpaired planning, decision-making, inhibitory controlFrontal lobe tumors, patient age, treatment effects[25]
LANGUAGEAphasia, word-finding difficultiesLeft hemisphere tumors, surgical impact[22]
VISUOSPATIAL SKILLSDifficulty with spatial orientation and perceptionParietal and occipital lobe involvement[24]
PROCESSING SPEEDSlowed cognitive processingTreatment effects, overall disease burden[24]
EMOTIONAL PROCESSINGDepression, anxiety, altered affectTumor location, corticosteroid use[26]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ghadimi, K.; Abbas, I.; Karandish, A.; Crisman, C.; Eskandar, E.N.; Kobets, A.J. Cognitive Decline in Glioblastoma (GB) Patients with Different Treatment Modalities and Insights on Untreated Cases. Curr. Oncol. 2025, 32, 152. https://doi.org/10.3390/curroncol32030152

AMA Style

Ghadimi K, Abbas I, Karandish A, Crisman C, Eskandar EN, Kobets AJ. Cognitive Decline in Glioblastoma (GB) Patients with Different Treatment Modalities and Insights on Untreated Cases. Current Oncology. 2025; 32(3):152. https://doi.org/10.3390/curroncol32030152

Chicago/Turabian Style

Ghadimi, Keyvan, Imane Abbas, Alireza Karandish, Celina Crisman, Emad N. Eskandar, and Andrew J. Kobets. 2025. "Cognitive Decline in Glioblastoma (GB) Patients with Different Treatment Modalities and Insights on Untreated Cases" Current Oncology 32, no. 3: 152. https://doi.org/10.3390/curroncol32030152

APA Style

Ghadimi, K., Abbas, I., Karandish, A., Crisman, C., Eskandar, E. N., & Kobets, A. J. (2025). Cognitive Decline in Glioblastoma (GB) Patients with Different Treatment Modalities and Insights on Untreated Cases. Current Oncology, 32(3), 152. https://doi.org/10.3390/curroncol32030152

Article Metrics

Back to TopTop