Pub Date : 2024-12-02eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdae205
Iulian Emil Tampu, Tamara Bianchessi, Ida Blystad, Peter Lundberg, Per Nyman, Anders Eklund, Neda Haj-Hosseini
Purpose: To implement and evaluate deep learning-based methods for the classification of pediatric brain tumors (PBT) in magnetic resonance (MR) data.
Methods: A subset of the "Children's Brain Tumor Network" dataset was retrospectively used (n = 178 subjects, female = 72, male = 102, NA = 4, age range [0.01, 36.49] years) with tumor types being low-grade astrocytoma (n = 84), ependymoma (n = 32), and medulloblastoma (n = 62). T1w post-contrast (n = 94 subjects), T2w (n = 160 subjects), and apparent diffusion coefficient (ADC: n = 66 subjects) MR sequences were used separately. Two deep learning models were trained on transversal slices showing tumor. Joint fusion was implemented to combine image and age data, and 2 pre-training paradigms were utilized. Model explainability was investigated using gradient-weighted class-activation mapping (Grad-CAM), and the learned feature space was visualized using principal component analysis (PCA).
Results: The highest tumor-type classification performance was achieved when using a vision transformer model pre-trained on ImageNet and fine-tuned on ADC images with age fusion (Matthews correlation coefficient [MCC]: 0.77 ± 0.14, Accuracy: 0.87 ± 0.08), followed by models trained on T2w (MCC: 0.58 ± 0.11, Accuracy: 0.73 ± 0.08) and T1w post-contrast (MCC: 0.41 ± 0.11, Accuracy: 0.62 ± 0.08) data. Age fusion marginally improved the model's performance. Both model architectures performed similarly across the experiments, with no differences between the pre-training strategies. Grad-CAMs showed that the models' attention focused on the brain region. PCA of the feature space showed greater separation of the tumor-type clusters when using contrastive pre-training.
Conclusion: Classification of PBT on MR images could be accomplished using deep learning, with the top-performing model being trained on ADC data, which radiologists use for the clinical classification of these tumors.
{"title":"Pediatric brain tumor classification using deep learning on MR images with age fusion.","authors":"Iulian Emil Tampu, Tamara Bianchessi, Ida Blystad, Peter Lundberg, Per Nyman, Anders Eklund, Neda Haj-Hosseini","doi":"10.1093/noajnl/vdae205","DOIUrl":"https://doi.org/10.1093/noajnl/vdae205","url":null,"abstract":"<p><strong>Purpose: </strong>To implement and evaluate deep learning-based methods for the classification of pediatric brain tumors (PBT) in magnetic resonance (MR) data.</p><p><strong>Methods: </strong>A subset of the \"Children's Brain Tumor Network\" dataset was retrospectively used (<i>n</i> = 178 subjects, female = 72, male = 102, NA = 4, age range [0.01, 36.49] years) with tumor types being low-grade astrocytoma (<i>n</i> = 84), ependymoma (<i>n</i> = 32), and medulloblastoma (<i>n</i> = 62). T1w post-contrast (<i>n</i> = 94 subjects), T2w (<i>n</i> = 160 subjects), and apparent diffusion coefficient (ADC: <i>n</i> = 66 subjects) MR sequences were used separately. Two deep learning models were trained on transversal slices showing tumor. Joint fusion was implemented to combine image and age data, and 2 pre-training paradigms were utilized. Model explainability was investigated using gradient-weighted class-activation mapping (Grad-CAM), and the learned feature space was visualized using principal component analysis (PCA).</p><p><strong>Results: </strong>The highest tumor-type classification performance was achieved when using a vision transformer model pre-trained on ImageNet and fine-tuned on ADC images with age fusion (Matthews correlation coefficient [MCC]: 0.77 ± 0.14, Accuracy: 0.87 ± 0.08), followed by models trained on T2w (MCC: 0.58 ± 0.11, Accuracy: 0.73 ± 0.08) and T1w post-contrast (MCC: 0.41 ± 0.11, Accuracy: 0.62 ± 0.08) data. Age fusion marginally improved the model's performance. Both model architectures performed similarly across the experiments, with no differences between the pre-training strategies. Grad-CAMs showed that the models' attention focused on the brain region. PCA of the feature space showed greater separation of the tumor-type clusters when using contrastive pre-training.</p><p><strong>Conclusion: </strong>Classification of PBT on MR images could be accomplished using deep learning, with the top-performing model being trained on ADC data, which radiologists use for the clinical classification of these tumors.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdae205"},"PeriodicalIF":3.7,"publicationDate":"2024-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11701748/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142960796","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-30eCollection Date: 2024-01-01DOI: 10.1093/noajnl/vdae206
Manuela Vooijs, Faith C Robertson, Sarah E Blitz, Christine Jungk, Sandro M Krieg, Philippe Schucht, Steven De Vleeschouwer, Arnaud J P E Vincent, Mitchel S Berger, Brian V Nahed, Marike L D Broekman, Jasper K W Gerritsen
Background: Awake craniotomy (AC) is a technique that balances maximum resection and minimal postoperative deficits in patients with intracranial tumors. To aid in the comparability of functional outcomes after awake surgery, this study investigated its international practice and aimed to define categories of postoperative deficits.
Methods: A survey was distributed via neurosurgical networks in Europe (European Association of Neurosurgical Societies, EANS), the Netherlands (Nederlandse Vereniging voor Neurochirurgie, NVVN), Belgium (Belgian Society of Neurosurgery, BSN), and the United States (Congress of Neurological Surgeons, CNS) between April 2022 and April 2023. Questions involved decision-making, including patient selection, anxiety assessment, and termination of resection. Interpretation of "major" and "minor" deficits, respectively labeled "level I" and "level II," was assessed.
Results: Three hundred and ninety-five neurosurgeons from 46 countries completed the survey. Significant heterogeneity was found in the domains of indications, anxiety assessment, seizure management, and termination of resection. Moreover, the interpretation of "major" deficits mainly included language and motor impairments. Analysis across deficit categories showed significant overlap in the domains of executive function, social cognition, and vision. Secondly, "minor" deficits and "minor cognitive" deficits showed vast overlap.
Conclusions: This survey demonstrates high variability between neurosurgeons in AC practice across multiple domains, inviting international efforts to reach a consensus regarding the standardization and grading of postoperative deficits. The proposed categories of "level I" and "level II" deficits may aid in this standardization. It allows for systematic assessment of the benefit of surgery in neuro-oncology patients and allows for comparison of surgical outcomes between institutions and surgeons. This may help to optimize international guidelines for surgical neuro-oncology, including AC.
背景:清醒开颅手术(AC)是一种在颅内肿瘤患者中兼顾最大切除和最小术后功能障碍的技术。为了帮助比较清醒手术后的功能结果,本研究调查了清醒手术的国际惯例,旨在界定术后功能障碍的类别:在2022年4月至2023年4月期间,通过欧洲(欧洲神经外科协会,EANS)、荷兰(荷兰神经外科协会,NVVN)、比利时(比利时神经外科协会,BSN)和美国(神经外科医师大会,CNS)的神经外科网络分发了一份调查问卷。问题涉及决策,包括患者选择、焦虑评估和终止切除。对 "主要 "和 "次要 "缺陷(分别标为 "I级 "和 "II级")的解释进行了评估:来自 46 个国家的 395 名神经外科医生完成了调查。在适应症、焦虑评估、癫痫发作处理和终止切除术等方面发现了显著的异质性。此外,对 "主要 "缺陷的解释主要包括语言和运动障碍。对不同缺陷类别的分析表明,在执行功能、社会认知和视力领域存在明显重叠。其次,"轻微 "缺陷和 "轻微认知 "缺陷也有很大的重叠:这项调查显示,神经外科医生在 AC 实践中的多个领域存在很大差异,因此需要国际社会努力就术后缺陷的标准化和分级达成共识。所提出的 "I 级 "和 "II 级 "缺陷类别可能有助于实现标准化。它允许对神经肿瘤患者的手术获益进行系统评估,并允许对不同机构和外科医生的手术结果进行比较。这将有助于优化包括 AC 在内的国际神经肿瘤外科指南。
{"title":"Level I and II deficits-A clinical survey on international practice of awake craniotomy and definitions of postoperative \"major\" and \"minor\" deficits.","authors":"Manuela Vooijs, Faith C Robertson, Sarah E Blitz, Christine Jungk, Sandro M Krieg, Philippe Schucht, Steven De Vleeschouwer, Arnaud J P E Vincent, Mitchel S Berger, Brian V Nahed, Marike L D Broekman, Jasper K W Gerritsen","doi":"10.1093/noajnl/vdae206","DOIUrl":"10.1093/noajnl/vdae206","url":null,"abstract":"<p><strong>Background: </strong>Awake craniotomy (AC) is a technique that balances maximum resection and minimal postoperative deficits in patients with intracranial tumors. To aid in the comparability of functional outcomes after awake surgery, this study investigated its international practice and aimed to define categories of postoperative deficits.</p><p><strong>Methods: </strong>A survey was distributed via neurosurgical networks in Europe (European Association of Neurosurgical Societies, EANS), the Netherlands (Nederlandse Vereniging voor Neurochirurgie, NVVN), Belgium (Belgian Society of Neurosurgery, BSN), and the United States (Congress of Neurological Surgeons, CNS) between April 2022 and April 2023. Questions involved decision-making, including patient selection, anxiety assessment, and termination of resection. Interpretation of \"major\" and \"minor\" deficits, respectively labeled \"level I\" and \"level II,\" was assessed.</p><p><strong>Results: </strong>Three hundred and ninety-five neurosurgeons from 46 countries completed the survey. Significant heterogeneity was found in the domains of indications, anxiety assessment, seizure management, and termination of resection. Moreover, the interpretation of \"major\" deficits mainly included language and motor impairments. Analysis across deficit categories showed significant overlap in the domains of executive function, social cognition, and vision. Secondly, \"minor\" deficits and \"minor cognitive\" deficits showed vast overlap.</p><p><strong>Conclusions: </strong>This survey demonstrates high variability between neurosurgeons in AC practice across multiple domains, inviting international efforts to reach a consensus regarding the standardization and grading of postoperative deficits. The proposed categories of \"level I\" and \"level II\" deficits may aid in this standardization. It allows for systematic assessment of the benefit of surgery in neuro-oncology patients and allows for comparison of surgical outcomes between institutions and surgeons. This may help to optimize international guidelines for surgical neuro-oncology, including AC.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"6 1","pages":"vdae206"},"PeriodicalIF":3.7,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11647522/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142840570","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-29eCollection Date: 2024-01-01DOI: 10.1093/noajnl/vdae210
Norbert Galldiks, Jan-Michael Werner, Isabelle Stetter, Hannah C Puhr, Thomas S Nakuz, Gabriele Stoffels, Nathalie L Albert, Karl-Josef Langen, Philipp Lohmann, Matthias Preusser
The phase-3 INDIGO trial demonstrated that the isocitrate dehydrogenase (IDH) inhibitor vorasidenib significantly prolonged progression-free survival and delayed intervention in patients with CNS WHO grade 2 gliomas. However, conventional MRI showed limited response, with only 11% of patients having objective responses. Studies suggest that serial PET imaging with radiolabeled amino acids, such as O -(2-[18 F]-fluoroethyl)-L-tyrosine (FET) PET, may provide earlier and more informative assessments of treatment response than MRI. In an initial experience with FET PET, 3 out of 5 patients showed metabolic response to vorasidenib. This highlights FET PET's potential to guide decision-making, though further trials are needed to confirm outcome benefits.
{"title":"Evaluation of early metabolic changes following vorasidenib using FET PET in patients with <i>IDH</i>-mutant gliomas.","authors":"Norbert Galldiks, Jan-Michael Werner, Isabelle Stetter, Hannah C Puhr, Thomas S Nakuz, Gabriele Stoffels, Nathalie L Albert, Karl-Josef Langen, Philipp Lohmann, Matthias Preusser","doi":"10.1093/noajnl/vdae210","DOIUrl":"10.1093/noajnl/vdae210","url":null,"abstract":"<p><p>The phase-3 INDIGO trial demonstrated that the isocitrate dehydrogenase (<i>IDH</i>) inhibitor vorasidenib significantly prolonged progression-free survival and delayed intervention in patients with CNS WHO grade 2 gliomas. However, conventional MRI showed limited response, with only 11% of patients having objective responses. Studies suggest that serial PET imaging with radiolabeled amino acids, such as <i>O</i> -(2-[<sup>18</sup> F]-fluoroethyl)-L-tyrosine (FET) PET, may provide earlier and more informative assessments of treatment response than MRI. In an initial experience with FET PET, 3 out of 5 patients showed metabolic response to vorasidenib. This highlights FET PET's potential to guide decision-making, though further trials are needed to confirm outcome benefits.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"6 1","pages":"vdae210"},"PeriodicalIF":3.7,"publicationDate":"2024-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11683584/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142908154","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-28eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdae209
Farzaneh Rahmani, Garrett Camps, Olesya Mironchuk, Norman Atagu, David H Ballard, Tammie L S Benzinger, Vincent Tze Yang Chow, Sonika Dahiya, John Evans, Shama Jaswal, Sara Hosseinzadeh Kassani, Da Ma, Muhammad Naeem, Karteek Popuri, Cyrus A Raji, Marilyn J Siegel, Yifei Xu, Jingxia Liu, Mirza Faisal Beg, Michael R Chicoine, Joseph E Ippolito
Background: Alterations in cellular metabolism affect cancer survival and can manifest in metrics of body composition. We investigated the effects of various body composition metrics on survival in patients with glioblastoma (GBM).
Methods: We retrospectively analyzed patients who had an abdominal and pelvic computed tomography (CT) scan performed within 1 month of diagnosis of GBM (178 participants, 102 males, 76 females, median age: 62.1 years). Volumetric body composition metrics were derived using automated CT segmentation of adipose tissue, skeletal muscle, and aortic calcification from L1 to L5. Univariable and multivariable Cox proportional hazards models were performed separately in males and females using known predictors of GBM overall survival (OS) as covariates. A sex-specific composite score of predisposing and protective factors was constructed using the relative importance of each metric in GBM OS.
Results: Higher skeletal muscle volume and lower skeletal muscle fat fraction were associated with better OS in the entire dataset. A robust and independent effect on GBM OS was seen specifically for fraction of inter/intramuscular adipose tissue to total adipose tissue after correction for known survival predictors and comorbidities. Worse OS was observed with increased abdominal aortic calcification volume in both sexes. There was a significant difference in GBM OS among participants stratified into quartiles based on sex-specific composite predisposing and protective scores.
Conclusion: The relationship between body composition and GBM OS provides an actionable advancement toward precision medicine in GBM management, as lifestyle and dietary regimens can alter body composition and metabolism and from there GBM survival.
{"title":"Abdominal myosteatosis measured with computed tomography predicts poor outcomes in patients with glioblastoma.","authors":"Farzaneh Rahmani, Garrett Camps, Olesya Mironchuk, Norman Atagu, David H Ballard, Tammie L S Benzinger, Vincent Tze Yang Chow, Sonika Dahiya, John Evans, Shama Jaswal, Sara Hosseinzadeh Kassani, Da Ma, Muhammad Naeem, Karteek Popuri, Cyrus A Raji, Marilyn J Siegel, Yifei Xu, Jingxia Liu, Mirza Faisal Beg, Michael R Chicoine, Joseph E Ippolito","doi":"10.1093/noajnl/vdae209","DOIUrl":"10.1093/noajnl/vdae209","url":null,"abstract":"<p><strong>Background: </strong>Alterations in cellular metabolism affect cancer survival and can manifest in metrics of body composition. We investigated the effects of various body composition metrics on survival in patients with glioblastoma (GBM).</p><p><strong>Methods: </strong>We retrospectively analyzed patients who had an abdominal and pelvic computed tomography (CT) scan performed within 1 month of diagnosis of GBM (178 participants, 102 males, 76 females, median age: 62.1 years). Volumetric body composition metrics were derived using automated CT segmentation of adipose tissue, skeletal muscle, and aortic calcification from L1 to L5. Univariable and multivariable Cox proportional hazards models were performed separately in males and females using known predictors of GBM overall survival (OS) as covariates. A sex-specific composite score of predisposing and protective factors was constructed using the relative importance of each metric in GBM OS.</p><p><strong>Results: </strong>Higher skeletal muscle volume and lower skeletal muscle fat fraction were associated with better OS in the entire dataset. A robust and independent effect on GBM OS was seen specifically for fraction of inter/intramuscular adipose tissue to total adipose tissue after correction for known survival predictors and comorbidities. Worse OS was observed with increased abdominal aortic calcification volume in both sexes. There was a significant difference in GBM OS among participants stratified into quartiles based on sex-specific composite predisposing and protective scores.</p><p><strong>Conclusion: </strong>The relationship between body composition and GBM OS provides an actionable advancement toward precision medicine in GBM management, as lifestyle and dietary regimens can alter body composition and metabolism and from there GBM survival.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdae209"},"PeriodicalIF":3.7,"publicationDate":"2024-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11713020/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142960794","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: The EF-14 clinical trial demonstrated the safety and efficacy of tumor-treating fields (TTFields) for newly diagnosed glioblastoma. This study aimed to clarify the current status, safety, and efficacy of TTFields in Japanese patients who meet the EF-14 inclusion criteria.
Methods: This was a multicenter retrospective cohort study. Background, treatment, and outcome data of patients who satisfied the inclusion criteria of the EF-14 trial were collected from 45 institutions across Japan. The rate, determinants, and current status of TTField use, including its safety and efficacy in terms of progression and survival, were retrospectively investigated. This study was conducted in accordance with the STROBE checklist.
Results: Among the 607 patients enrolled, 70 were excluded due to progressive disease during radiation and temozolomide therapy, age > 80 years old, and Karnofsky Performance Status score of <70. Among the remaining 537 patients, 210 (39%) underwent TTField treatment. Multivariate analysis revealed younger age and spouse as a caregiver as significant factors for TTField use. The compliance rate of TTField use exceeded 75% in 60% of patients, with a median TTField usage duration of 11 months. Skin disorders requiring medical treatment occurred in 56% of patients. Multivariate Cox proportional hazards analysis in the whole series and propensity score-matched analysis revealed that TTField use was not a prognostic factor for progression-free survival (PFS) or overall survival (OS).
Conclusions: TTField use did not have a substantial effect on either PFS or OS in Japanese patients with glioblastoma, despite compliance rates comparable to those observed in the EF-14.
{"title":"Impact of tumor-treating fields on the survival of Japanese patients with newly diagnosed glioblastoma: A multicenter, retrospective cohort study.","authors":"Masayuki Kanamori, Shunsuke Tsuzuki, Ichiyo Shibahara, Kuniaki Saito, Yoshiteru Shimoda, Kazuhiro Tanaka, Shigeru Yamaguchi, Manabu Natsumeda, Tomoo Matsutani, Mitsuto Hanihara, Mitsutoshi Nakada, Jun-Ichiro Kuroda, Masahide Matsuda, Koji Yoshimoto, Ushio Yonezawa, Yukihiko Sonoda, Koji Takano, Hajime Yonezawa, Yoshihiro Otani, Yukiko Nakahara, Masashi Uchida, Masahiro Nonaka, Yohei Mineharu, Yohei Kitamura, Shinji Yamashita, Takahiro Yamauchi, Yohei Miyake, Shoichi Deguchi, Takaaki Beppu, Kaoru Tamura, Shinichiro Koizumi, Yuichi Hirose, Kenichiro Asano, Ryo Hiruta, Manabu Kinoshita, Keisuke Miyake, Noriyuki Nakayama, Akihiro Inoue, Takahiro Ono, Takahiro Sasaki, Yukinori Akiyama, Shinjiro Fukami, Atsuo Yoshino, Yu Kawanishi, Taku Asanome, Takuhiro Yamaguchi, Masamichi Takahashi, Fumiyuki Yamasaki, Yoshiki Arakawa, Yoshitaka Narita","doi":"10.1093/noajnl/vdae176","DOIUrl":"10.1093/noajnl/vdae176","url":null,"abstract":"<p><strong>Background: </strong>The EF-14 clinical trial demonstrated the safety and efficacy of tumor-treating fields (TTFields) for newly diagnosed glioblastoma. This study aimed to clarify the current status, safety, and efficacy of TTFields in Japanese patients who meet the EF-14 inclusion criteria.</p><p><strong>Methods: </strong>This was a multicenter retrospective cohort study. Background, treatment, and outcome data of patients who satisfied the inclusion criteria of the EF-14 trial were collected from 45 institutions across Japan. The rate, determinants, and current status of TTField use, including its safety and efficacy in terms of progression and survival, were retrospectively investigated. This study was conducted in accordance with the STROBE checklist.</p><p><strong>Results: </strong>Among the 607 patients enrolled, 70 were excluded due to progressive disease during radiation and temozolomide therapy, age > 80 years old, and Karnofsky Performance Status score of <70. Among the remaining 537 patients, 210 (39%) underwent TTField treatment. Multivariate analysis revealed younger age and spouse as a caregiver as significant factors for TTField use. The compliance rate of TTField use exceeded 75% in 60% of patients, with a median TTField usage duration of 11 months. Skin disorders requiring medical treatment occurred in 56% of patients. Multivariate Cox proportional hazards analysis in the whole series and propensity score-matched analysis revealed that TTField use was not a prognostic factor for progression-free survival (PFS) or overall survival (OS).</p><p><strong>Conclusions: </strong>TTField use did not have a substantial effect on either PFS or OS in Japanese patients with glioblastoma, despite compliance rates comparable to those observed in the EF-14.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"6 1","pages":"vdae176"},"PeriodicalIF":3.7,"publicationDate":"2024-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11629686/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142808973","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-22eCollection Date: 2024-01-01DOI: 10.1093/noajnl/vdae202
Joe S Mendez, Adam L Cohen, Midori Eckenstein, Randy L Jensen, Lindsay M Burt, Karen L Salzman, Marc Chamberlain, Henry H Hsu, Marguerite Hutchinson, Fabio Iwamoto, Keith L Ligon, Maciej M Mrugala, Michael Pelayo, Scott R Plotkin, Vinay K Puduvalli, Jeffrey Raizer, David A Reardon, Michael Sterba, Tobias Walbert, Brian L West, Eric T Wong, Chao Zhang, Howard Colman
Background: Glioblastoma (GBM) has a median survival of <2 years. Pexidartinib (PLX3397) is a small-molecule inhibitor of CSF1R, KIT, and oncogenic FTL3, which are implicated in GBM treatment resistance. Results from glioma models indicate that combining radiation therapy (RT) and pexidartinib reduces radiation resistance. We added pexidartinib to standard-of-care RT/temozolomide (TMZ) in patients with newly diagnosed GBM to assess the therapeutic benefit of altering the tumor microenvironment with pexidartinib.
Methods: In this open-label, dose-escalation, multicenter, Phase 1b/2 trial, pexidartinib was administered in combination with RT/TMZ followed by adjuvant pexidartinib + TMZ. During Phase 1b, pexidartinib was given 5 or 7 days/week at multiple dosing levels. The primary Phase 1b endpoint was the recommended Phase 2 dose (RP2D). Phase 2 patients received the RP2D with the primary endpoint of median progression-free survival (mPFS). Secondary objectives were median overall survival (mOS), pharmacokinetics, and safety.
Results: The RP2D of pexidartinib was 800 mg/day for 5 days/week during RT/TMZ, followed by 800 mg/day for 7 days/week with adjuvant TMZ. mPFS was 6.7 months (90% CI: 4.5, 11.5) for the modified intention-to-treat population. The actual mOS was 13.1 months (90% CI: 11.5, 24.5), and the mOS corrected for comparison with matched historical controls was 18.8 months (95% CI: 12.6, 28.0).
Conclusions: This trial established the RP2D of pexidartinib in combination with RT/TMZ and adjuvant TMZ. Pexidartinib was generally safe and well tolerated. Although the study regimen with pexidartinib was not efficacious, pharmacodynamic studies showed modulation of systemic markers that could lead to alteration of the tumor microenvironment.
{"title":"Phase 1b/2 study of orally administered pexidartinib in combination with radiation therapy and temozolomide in patients with newly diagnosed glioblastoma.","authors":"Joe S Mendez, Adam L Cohen, Midori Eckenstein, Randy L Jensen, Lindsay M Burt, Karen L Salzman, Marc Chamberlain, Henry H Hsu, Marguerite Hutchinson, Fabio Iwamoto, Keith L Ligon, Maciej M Mrugala, Michael Pelayo, Scott R Plotkin, Vinay K Puduvalli, Jeffrey Raizer, David A Reardon, Michael Sterba, Tobias Walbert, Brian L West, Eric T Wong, Chao Zhang, Howard Colman","doi":"10.1093/noajnl/vdae202","DOIUrl":"10.1093/noajnl/vdae202","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma (GBM) has a median survival of <2 years. Pexidartinib (PLX3397) is a small-molecule inhibitor of CSF1R, KIT, and oncogenic FTL3, which are implicated in GBM treatment resistance. Results from glioma models indicate that combining radiation therapy (RT) and pexidartinib reduces radiation resistance. We added pexidartinib to standard-of-care RT/temozolomide (TMZ) in patients with newly diagnosed GBM to assess the therapeutic benefit of altering the tumor microenvironment with pexidartinib.</p><p><strong>Methods: </strong>In this open-label, dose-escalation, multicenter, Phase 1b/2 trial, pexidartinib was administered in combination with RT/TMZ followed by adjuvant pexidartinib + TMZ. During Phase 1b, pexidartinib was given 5 or 7 days/week at multiple dosing levels. The primary Phase 1b endpoint was the recommended Phase 2 dose (RP2D). Phase 2 patients received the RP2D with the primary endpoint of median progression-free survival (mPFS). Secondary objectives were median overall survival (mOS), pharmacokinetics, and safety.</p><p><strong>Results: </strong>The RP2D of pexidartinib was 800 mg/day for 5 days/week during RT/TMZ, followed by 800 mg/day for 7 days/week with adjuvant TMZ. mPFS was 6.7 months (90% CI: 4.5, 11.5) for the modified intention-to-treat population. The actual mOS was 13.1 months (90% CI: 11.5, 24.5), and the mOS corrected for comparison with matched historical controls was 18.8 months (95% CI: 12.6, 28.0).</p><p><strong>Conclusions: </strong>This trial established the RP2D of pexidartinib in combination with RT/TMZ and adjuvant TMZ. Pexidartinib was generally safe and well tolerated. Although the study regimen with pexidartinib was not efficacious, pharmacodynamic studies showed modulation of systemic markers that could lead to alteration of the tumor microenvironment.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"6 1","pages":"vdae202"},"PeriodicalIF":3.7,"publicationDate":"2024-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11672110/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142904642","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-19eCollection Date: 2024-01-01DOI: 10.1093/noajnl/vdae187
Brittany Dewdney, Panimaya Jeffreena Miranda, Mani Kuchibhotla, Ranjith Palanisamy, Caitlyn Richworth, Carol J Milligan, Zi Ying Ng, Lauren Ursich, Steve Petrou, Emily V Fletcher, Roger J Daly, Terry C C Lim Kam Sian, Santosh Valvi, Raelene Endersby, Terrance G Johns
Background: Glioblastoma, a lethal high-grade glioma, has not seen improvements in clinical outcomes in nearly 30 years. Ion channels are increasingly associated with tumorigenesis, and there are hundreds of brain-penetrant drugs that inhibit ion channels, representing an untapped therapeutic resource. The aim of this exploratory drug study was to screen an ion channel drug library against patient-derived glioblastoma cells to identify new treatments for brain cancer.
Methods: Seventy-two ion channel inhibitors were screened in patient-derived glioblastoma cells, and cell viability was determined using the ViaLight Assay. Cell cycle and apoptosis analysis were determined with flow cytometry using PI and Annexin V staining, respectively. Protein and phosphoprotein expression was determined using mass spectrometry and analyzed using gene set enrichment analysis. Kaplan-Meier survival analyses were performed using intracranial xenograft models of GBM6 and WK1 cells.
Results: The voltage-gated sodium channel modulator, DPI-201-106, was revealed to reduce glioblastoma cell viability in vitro by inducing cell cycle arrest and apoptosis. Phosphoproteomics indicated that DPI-201-106 may impact DNA damage response (DDR) pathways. Combination treatment of DPI-201-106 with the CHK1 inhibitor prexasertib or the PARP inhibitor niraparib demonstrated synergistic effects in multiple patient-derived glioblastoma cells both in vitro and in intracranial xenograft mouse models, extending survival of glioblastoma-bearing mice.
Conclusions: DPI-201-106 enhances the efficacy of DDR inhibitors to reduce glioblastoma growth. As these drugs have already been clinically tested in humans, repurposing DPI-201-106 in novel combinatorial approaches will allow for rapid translation into the clinic.
{"title":"Ion channel modulator DPI-201-106 significantly enhances antitumor activity of DNA damage response inhibitors in glioblastoma.","authors":"Brittany Dewdney, Panimaya Jeffreena Miranda, Mani Kuchibhotla, Ranjith Palanisamy, Caitlyn Richworth, Carol J Milligan, Zi Ying Ng, Lauren Ursich, Steve Petrou, Emily V Fletcher, Roger J Daly, Terry C C Lim Kam Sian, Santosh Valvi, Raelene Endersby, Terrance G Johns","doi":"10.1093/noajnl/vdae187","DOIUrl":"10.1093/noajnl/vdae187","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma, a lethal high-grade glioma, has not seen improvements in clinical outcomes in nearly 30 years. Ion channels are increasingly associated with tumorigenesis, and there are hundreds of brain-penetrant drugs that inhibit ion channels, representing an untapped therapeutic resource. The aim of this exploratory drug study was to screen an ion channel drug library against patient-derived glioblastoma cells to identify new treatments for brain cancer.</p><p><strong>Methods: </strong>Seventy-two ion channel inhibitors were screened in patient-derived glioblastoma cells, and cell viability was determined using the ViaLight Assay. Cell cycle and apoptosis analysis were determined with flow cytometry using PI and Annexin V staining, respectively. Protein and phosphoprotein expression was determined using mass spectrometry and analyzed using gene set enrichment analysis. Kaplan-Meier survival analyses were performed using intracranial xenograft models of GBM6 and WK1 cells.</p><p><strong>Results: </strong>The voltage-gated sodium channel modulator, DPI-201-106, was revealed to reduce glioblastoma cell viability in vitro by inducing cell cycle arrest and apoptosis. Phosphoproteomics indicated that DPI-201-106 may impact DNA damage response (DDR) pathways. Combination treatment of DPI-201-106 with the CHK1 inhibitor prexasertib or the PARP inhibitor niraparib demonstrated synergistic effects in multiple patient-derived glioblastoma cells both in vitro and in intracranial xenograft mouse models, extending survival of glioblastoma-bearing mice.</p><p><strong>Conclusions: </strong>DPI-201-106 enhances the efficacy of DDR inhibitors to reduce glioblastoma growth. As these drugs have already been clinically tested in humans, repurposing DPI-201-106 in novel combinatorial approaches will allow for rapid translation into the clinic.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"6 1","pages":"vdae187"},"PeriodicalIF":3.7,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11630809/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142808976","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Following large resection, proposing a watch-and-wait strategy in selected grade 3 glioma, isocitrate dehydrogenase (IDH)-mutant patients is an emerging practice. We compared the watch-and-wait approach to the standard postoperative adjuvant oncological treatment for grade 3 gliomas, IDH-mutant.
Methods: Observational, retrospective, single-institution cohort (2011-2023) of 106 consecutive adult patients harboring supratentorial grade 3 gliomas, IDH-mutant treated by maximal awake resection and who received a watch-and-wait approach (surgery group) or an adjuvant oncological treatment (oncological group) postoperatively. Case-matched analysis (1:1) criteria between the surgery group and oncological group: extent of resection, tumor volume, Karnofsky Performance Status (KPS) score, tumor location and size, and age.
Results: Patients of the surgery group (n = 26) had significantly better KPS scores, less preoperative neurological and/or neurocognitive deficits, less hyperperfusion, less corpus callosum infiltration, smaller tumor volume, higher rate of total resection, and smaller residual tumor than patients of the oncological group (n = 80). The 5-year progression-free survival (66.2 vs. 77.9 months, P = .713) and the 5-year overall survival (88.9 vs. 83.9 months, P = .291) did not differ between surgery and oncological groups. In the whole series, a preoperative KPS score >70, a total resection, and the oligodendroglioma subtype were independent predictors of longer progression-free survival and overall survival. After case matching, no difference in survival was observed between watch-and-wait and oncological treatment both in astrocytomas (n = 14 per group) and oligodendrogliomas (n = 12 per group).
Conclusions: Watch-and-wait following radical resection appears to be feasible in highly selected grade 3 gliomas, IDH-mutant patients without impairing survival both in astrocytoma and in oligodendroglioma subgroups.
{"title":"Watch-and-wait approach versus adjuvant treatment after radical awake resection in selected adult-type grade 3 gliomas, <i>isocitrate dehydrogenase</i> mutant: A case-matched cohort.","authors":"Angela Elia, Alexandre Roux, Bénédicte Trancart, Alessandro Moiraghi, Maimiti Seneca, Edouard Dezamis, Pascale Varlet, Fabrice Chretien, Catherine Oppenheim, Marc Zanello, Johan Pallud","doi":"10.1093/noajnl/vdae189","DOIUrl":"https://doi.org/10.1093/noajnl/vdae189","url":null,"abstract":"<p><strong>Background: </strong>Following large resection, proposing a watch-and-wait strategy in selected grade 3 glioma, <i>isocitrate dehydrogenase</i> (<i>IDH)</i>-mutant patients is an emerging practice. We compared the watch-and-wait approach to the standard postoperative adjuvant oncological treatment for grade 3 gliomas, <i>IDH</i>-mutant.</p><p><strong>Methods: </strong>Observational, retrospective, single-institution cohort (2011-2023) of 106 consecutive adult patients harboring supratentorial grade 3 gliomas, <i>IDH</i>-mutant treated by maximal awake resection and who received a watch-and-wait approach (surgery group) or an adjuvant oncological treatment (oncological group) postoperatively. Case-matched analysis (1:1) criteria between the surgery group and oncological group: extent of resection, tumor volume, Karnofsky Performance Status (KPS) score, tumor location and size, and age.</p><p><strong>Results: </strong>Patients of the surgery group (<i>n</i> = 26) had significantly better KPS scores, less preoperative neurological and/or neurocognitive deficits, less hyperperfusion, less corpus callosum infiltration, smaller tumor volume, higher rate of total resection, and smaller residual tumor than patients of the oncological group (<i>n</i> = 80). The 5-year progression-free survival (66.2 vs. 77.9 months, <i>P</i> = .713) and the 5-year overall survival (88.9 vs. 83.9 months, <i>P</i> = .291) did not differ between surgery and oncological groups. In the whole series, a preoperative KPS score >70, a total resection, and the oligodendroglioma subtype were independent predictors of longer progression-free survival and overall survival. After case matching, no difference in survival was observed between watch-and-wait and oncological treatment both in astrocytomas (<i>n</i> = 14 per group) and oligodendrogliomas (<i>n</i> = 12 per group).</p><p><strong>Conclusions: </strong>Watch-and-wait following radical resection appears to be feasible in highly selected grade 3 gliomas, <i>IDH</i>-mutant patients without impairing survival both in astrocytoma and in oligodendroglioma subgroups.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"6 1","pages":"vdae189"},"PeriodicalIF":3.7,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11606645/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142775233","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-18eCollection Date: 2024-01-01DOI: 10.1093/noajnl/vdae196
Otto Mølby Henriksen, Aida Muhic, Michael Juncker Lundemann, Henrik Bo Wiberg Larsson, Ulrich Lindberg, Thomas Lund Andersen, Benedikte Hasselbalch, Søren Møller, Lisbeth Marner, Karine Madsen, Vibeke Andrée Larsen, Hans Skovgaard Poulsen, Adam Espe Hansen, Ian Law
Background: Magnetic resonance imaging (MRI) cerebral blood volume (CBV) measurements improve the diagnosis of recurrent gliomas. The study investigated the prognostic value of dynamic contrast-enhanced (DCE) CBV imaging in treated IDH wildtype glioblastoma when added to MRI or amino acid positron emission tomography (PET).
Methods: Hybrid [18F]FET PET/MRI with 2CXM (2-compartment exchange model) DCE from 86 adult patients with suspected recurrent or residual glioblastoma were retrospectively analyzed. High CBV tumor volume (VOLCBV), and contrast-enhancing (VOLCE) and [18F]FET active tumor (VOLFET) volumes were delineated. Absolute and fractional high CBV subvolumes within VOLCE and VOLFET were determined. Associations with overall survival (OS) were assessed by Cox analysis.
Results: Adjusted for methyltransferase gene status and steroid use all total tumor volumes were individually associated with shorter OS. Adding VOLCBV to VOLCE or VOLFET only the effect of VOLCBV was prognostic of OS (hazard ratio [HR] 1.327, P = .042 and 1.352, P = .011, respectively). High CBV subvolumes within both VOLCE and VOLFET were associated with shorter survival (HR 1.448, P = .042 and 1.416, P = .011, respectively), and the low CBV subvolumes with longer survival (HR 0.504, P = .002 and .365, P = .001, respectively). The fraction of VOLCE and VOLFET with high CBV was a strong predictor of OS with shorter median OS in upper versus lower tertiles (8.3 vs 14.5 months and 7.1 vs 15.6 months, respectively, both P < .001).
Conclusions: The high CBV tumor volume was a strong prognosticator of survival and allowed for the separation of high- and low-risk subvolumes underlining the heterogeneous physiological environment represented in the contrast-enhancing or metabolically active tumor volumes of treated glioblastoma.
{"title":"Added prognostic value of DCE blood volume imaging in patients with suspected recurrent or residual glioblastoma-A hybrid [<sup>18</sup>F]FET PET/MRI study.","authors":"Otto Mølby Henriksen, Aida Muhic, Michael Juncker Lundemann, Henrik Bo Wiberg Larsson, Ulrich Lindberg, Thomas Lund Andersen, Benedikte Hasselbalch, Søren Møller, Lisbeth Marner, Karine Madsen, Vibeke Andrée Larsen, Hans Skovgaard Poulsen, Adam Espe Hansen, Ian Law","doi":"10.1093/noajnl/vdae196","DOIUrl":"10.1093/noajnl/vdae196","url":null,"abstract":"<p><strong>Background: </strong>Magnetic resonance imaging (MRI) cerebral blood volume (CBV) measurements improve the diagnosis of recurrent gliomas. The study investigated the prognostic value of dynamic contrast-enhanced (DCE) CBV imaging in treated IDH wildtype glioblastoma when added to MRI or amino acid positron emission tomography (PET).</p><p><strong>Methods: </strong>Hybrid [<sup>18</sup>F]FET PET/MRI with 2CXM (2-compartment exchange model) DCE from 86 adult patients with suspected recurrent or residual glioblastoma were retrospectively analyzed. High CBV tumor volume (VOL<sub>CBV</sub>), and contrast-enhancing (VOL<sub>CE</sub>) and [<sup>18</sup>F]FET active tumor (VOL<sub>FET</sub>) volumes were delineated. Absolute and fractional high CBV subvolumes within VOL<sub>CE</sub> and VOL<sub>FET</sub> were determined. Associations with overall survival (OS) were assessed by Cox analysis.</p><p><strong>Results: </strong>Adjusted for methyltransferase gene status and steroid use all total tumor volumes were individually associated with shorter OS. Adding VOL<sub>CBV</sub> to VOL<sub>CE</sub> or VOL<sub>FET</sub> only the effect of VOL<sub>CBV</sub> was prognostic of OS (hazard ratio [HR] 1.327, <i>P</i> = .042 and 1.352, <i>P</i> = .011, respectively). High CBV subvolumes within both VOL<sub>CE</sub> and VOL<sub>FET</sub> were associated with shorter survival (HR 1.448, <i>P</i> = .042 and 1.416, <i>P</i> = .011, respectively), and the low CBV subvolumes with longer survival (HR 0.504, <i>P</i> = .002 and .365, <i>P</i> = .001, respectively). The fraction of VOL<sub>CE</sub> and VOL<sub>FET</sub> with high CBV was a strong predictor of OS with shorter median OS in upper versus lower tertiles (8.3 vs 14.5 months and 7.1 vs 15.6 months, respectively, both <i>P</i> < .001).</p><p><strong>Conclusions: </strong>The high CBV tumor volume was a strong prognosticator of survival and allowed for the separation of high- and low-risk subvolumes underlining the heterogeneous physiological environment represented in the contrast-enhancing or metabolically active tumor volumes of treated glioblastoma.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"6 1","pages":"vdae196"},"PeriodicalIF":3.7,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632823/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142815395","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-16eCollection Date: 2024-01-01DOI: 10.1093/noajnl/vdae200
Keyhan Najafian, Benjamin Rehany, Alexander Nowakowski, Saba Ghazimoghadam, Kevin Pierre, Rita Zakarian, Tariq Al-Saadi, Caroline Reinhold, Abbas Babajani-Feremi, Joshua K Wong, Marie-Christine Guiot, Marie-Constance Lacasse, Stephanie Lam, Peter M Siegel, Kevin Petrecca, Matthew Dankner, Reza Forghani
Background: Brain metastasis invasion pattern (BMIP) is an emerging biomarker associated with recurrence-free and overall survival in patients, and differential response to therapy in preclinical models. Currently, BMIP can only be determined from the histopathological examination of surgical specimens, precluding its use as a biomarker prior to therapy initiation. The aim of this study was to investigate the potential of machine learning (ML) approaches to develop a noninvasive magnetic resonance imaging (MRI)-based biomarker for BMIP determination.
Methods: From an initial cohort of 329 patients, a subset of 132 patients met the inclusion criteria for this retrospective study. We evaluated the ability of an expert neuroradiologist to reliably predict BMIP. Thereafter, the dataset was randomly divided into training/validation (80% of cases) and test subsets (20% of cases). The ground truth for BMIP was the histopathologic evaluation of resected specimens. Following MRI sequence co-registration, advanced feature extraction techniques deriving hand-crafted radiomic features with traditional ML classifiers and convolution-based deep learning (CDL) models were trained and evaluated. Different ML approaches were used individually or using ensembling techniques to determine the model with the best performance for BMIP prediction.
Results: Expert evaluation of brain MRI scans could not reliably predict BMIP, with an accuracy of 44%-59% depending on the semantic feature used. Among the different ML and CDL models evaluated, the best-performing model achieved an accuracy of 85% and an F1 score of 90%.
Conclusions: ML approaches can effectively predict BMIP, representing a noninvasive MRI-based approach to guide the management of patients with brain metastases.
背景:脑转移侵袭模式(BMIP)是一种新兴的生物标志物,它与患者的无复发生存率和总生存率以及临床前模型对治疗的不同反应相关。目前,BMIP只能通过手术标本的组织病理学检查来确定,因此无法在开始治疗前将其用作生物标志物。本研究旨在研究机器学习(ML)方法的潜力,以开发一种基于磁共振成像(MRI)的无创生物标记物,用于确定BMIP:在最初的 329 例患者中,有 132 例患者符合这项回顾性研究的纳入标准。我们评估了神经放射专家可靠预测 BMIP 的能力。之后,数据集被随机分为训练/验证子集(80% 的病例)和测试子集(20% 的病例)。BMIP 的基本事实是切除标本的组织病理学评估。在核磁共振成像序列共配准之后,通过传统的 ML 分类器和基于卷积的深度学习(CDL)模型训练和评估了先进的特征提取技术,这些特征提取技术可得出手工创建的放射学特征。对不同的 ML 方法单独使用或使用集合技术,以确定 BMIP 预测性能最佳的模型:专家对脑磁共振成像扫描的评估无法可靠地预测 BMIP,准确率为 44%-59%,具体取决于所使用的语义特征。在评估的不同 ML 和 CDL 模型中,表现最好的模型准确率达到 85%,F1 分数达到 90%:ML方法可有效预测BMIP,是一种基于磁共振成像的无创方法,可指导脑转移患者的治疗。
{"title":"Machine learning prediction of brain metastasis invasion pattern on brain magnetic resonance imaging scans.","authors":"Keyhan Najafian, Benjamin Rehany, Alexander Nowakowski, Saba Ghazimoghadam, Kevin Pierre, Rita Zakarian, Tariq Al-Saadi, Caroline Reinhold, Abbas Babajani-Feremi, Joshua K Wong, Marie-Christine Guiot, Marie-Constance Lacasse, Stephanie Lam, Peter M Siegel, Kevin Petrecca, Matthew Dankner, Reza Forghani","doi":"10.1093/noajnl/vdae200","DOIUrl":"10.1093/noajnl/vdae200","url":null,"abstract":"<p><strong>Background: </strong>Brain metastasis invasion pattern (BMIP) is an emerging biomarker associated with recurrence-free and overall survival in patients, and differential response to therapy in preclinical models. Currently, BMIP can only be determined from the histopathological examination of surgical specimens, precluding its use as a biomarker prior to therapy initiation. The aim of this study was to investigate the potential of machine learning (ML) approaches to develop a noninvasive magnetic resonance imaging (MRI)-based biomarker for BMIP determination.</p><p><strong>Methods: </strong>From an initial cohort of 329 patients, a subset of 132 patients met the inclusion criteria for this retrospective study. We evaluated the ability of an expert neuroradiologist to reliably predict BMIP. Thereafter, the dataset was randomly divided into training/validation (80% of cases) and test subsets (20% of cases). The ground truth for BMIP was the histopathologic evaluation of resected specimens. Following MRI sequence co-registration, advanced feature extraction techniques deriving hand-crafted radiomic features with traditional ML classifiers and convolution-based deep learning (CDL) models were trained and evaluated. Different ML approaches were used individually or using ensembling techniques to determine the model with the best performance for BMIP prediction.</p><p><strong>Results: </strong>Expert evaluation of brain MRI scans could not reliably predict BMIP, with an accuracy of 44%-59% depending on the semantic feature used. Among the different ML and CDL models evaluated, the best-performing model achieved an accuracy of 85% and an F1 score of 90%.</p><p><strong>Conclusions: </strong>ML approaches can effectively predict BMIP, representing a noninvasive MRI-based approach to guide the management of patients with brain metastases.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"6 1","pages":"vdae200"},"PeriodicalIF":3.7,"publicationDate":"2024-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11639946/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142831594","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}