1,356 results on '"Aghi, Manish K."'
Search Results
2. “De novo replication repair deficient glioblastoma, IDH-wildtype” is a distinct glioblastoma subtype in adults that may benefit from immune checkpoint blockade
- Author
-
Hadad, Sara, Gupta, Rohit, Oberheim Bush, Nancy Ann, Taylor, Jennie W, Villanueva-Meyer, Javier E, Young, Jacob S, Wu, Jasper, Ravindranathan, Ajay, Zhang, Yalan, Warrier, Gayathri, McCoy, Lucie, Shai, Anny, Pekmezci, Melike, Perry, Arie, Bollen, Andrew W, Phillips, Joanna J, Braunstein, Steve E, Raleigh, David R, Theodosopoulos, Philip, Aghi, Manish K, Chang, Edward F, Hervey-Jumper, Shawn L, Costello, Joseph F, de Groot, John, Butowski, Nicholas A, Clarke, Jennifer L, Chang, Susan M, Berger, Mitchel S, Molinaro, Annette M, and Solomon, David A
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Brain Cancer ,Cancer ,Precision Medicine ,Brain Disorders ,Genetics ,Human Genome ,Clinical Research ,Cancer Genomics ,Neurosciences ,Immunotherapy ,Orphan Drug ,Rare Diseases ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Adult ,Humans ,Child ,Middle Aged ,Aged ,Glioblastoma ,Immune Checkpoint Inhibitors ,Homozygote ,Prospective Studies ,Brain Neoplasms ,Sequence Deletion ,Mutation ,Isocitrate Dehydrogenase ,Giant cell glioblastoma ,Hypermutation ,Ultrahypermutation ,Mismatch repair deficiency ,POLE ,Lynch syndrome ,Immune checkpoint blockade ,Molecular neuropathology ,Molecular neuro-oncology ,Neurology & Neurosurgery - Abstract
Glioblastoma is a clinically and molecularly heterogeneous disease, and new predictive biomarkers are needed to identify those patients most likely to respond to specific treatments. Through prospective genomic profiling of 459 consecutive primary treatment-naïve IDH-wildtype glioblastomas in adults, we identified a unique subgroup (2%, 9/459) defined by somatic hypermutation and DNA replication repair deficiency due to biallelic inactivation of a canonical mismatch repair gene. The deleterious mutations in mismatch repair genes were often present in the germline in the heterozygous state with somatic inactivation of the remaining allele, consistent with glioblastomas arising due to underlying Lynch syndrome. A subset of tumors had accompanying proofreading domain mutations in the DNA polymerase POLE and resultant "ultrahypermutation". The median age at diagnosis was 50 years (range 27-78), compared with 63 years for the other 450 patients with conventional glioblastoma (p
- Published
- 2024
3. Single-cell RNA sequencing and spatial transcriptomics reveal cancer-associated fibroblasts in glioblastoma with protumoral effects
- Author
-
Jain, Saket, Rick, Jonathan W, Joshi, Rushikesh S, Beniwal, Angad, Spatz, Jordan, Gill, Sabraj, Chang, Alexander Chih-Chieh, Choudhary, Nikita, Nguyen, Alan T, Sudhir, Sweta, Chalif, Eric J, Chen, Jia-Shu, Chandra, Ankush, Haddad, Alexander F, Wadhwa, Harsh, Shah, Sumedh S, Choi, Serah, Hayes, Josie L, Wang, Lin, Yagnik, Garima, Costello, Joseph F, Diaz, Aaron, Heiland, Dieter Henrik, and Aghi, Manish K
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Human Genome ,Cancer Genomics ,Cancer ,Neurosciences ,Genetics ,Stem Cell Research - Nonembryonic - Human ,Brain Cancer ,Rare Diseases ,Brain Disorders ,Stem Cell Research ,Aetiology ,2.1 Biological and endogenous factors ,Humans ,Glioblastoma ,Cancer-Associated Fibroblasts ,Transcriptome ,DNA Copy Number Variations ,Endothelial Cells ,Sequence Analysis ,RNA ,Brain cancer ,Fibronectin ,Oncology ,Medical and Health Sciences ,Immunology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Cancer-associated fibroblasts (CAFs) were presumed absent in glioblastoma given the lack of brain fibroblasts. Serial trypsinization of glioblastoma specimens yielded cells with CAF morphology and single-cell transcriptomic profiles based on their lack of copy number variations (CNVs) and elevated individual cell CAF probability scores derived from the expression of 9 CAF markers and absence of 5 markers from non-CAF stromal cells sharing features with CAFs. Cells without CNVs and with high CAF probability scores were identified in single-cell RNA-Seq of 12 patient glioblastomas. Pseudotime reconstruction revealed that immature CAFs evolved into subtypes, with mature CAFs expressing actin alpha 2, smooth muscle (ACTA2). Spatial transcriptomics from 16 patient glioblastomas confirmed CAF proximity to mesenchymal glioblastoma stem cells (GSCs), endothelial cells, and M2 macrophages. CAFs were chemotactically attracted to GSCs, and CAFs enriched GSCs. We created a resource of inferred crosstalk by mapping expression of receptors to their cognate ligands, identifying PDGF and TGF-β as mediators of GSC effects on CAFs and osteopontin and HGF as mediators of CAF-induced GSC enrichment. CAFs induced M2 macrophage polarization by producing the extra domain A (EDA) fibronectin variant that binds macrophage TLR4. Supplementing GSC-derived xenografts with CAFs enhanced in vivo tumor growth. These findings are among the first to identify glioblastoma CAFs and their GSC interactions, making them an intriguing target.
- Published
- 2023
4. Recent advances in the molecular prognostication of meningiomas
- Author
-
Wang, Elaina J, Haddad, Alexander F, Young, Jacob S, Morshed, Ramin A, Wu, Joshua PH, Salha, Diana M, Butowski, Nicholas, and Aghi, Manish K
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Neurosciences ,Rare Diseases ,Brain Disorders ,Cancer ,Brain Cancer ,meningioma ,cytogenetics ,genomics ,epigenetics ,methylation ,atypical ,anaplastic ,Oncology and Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis - Abstract
Meningiomas are the most common primary intracranial neoplasm. While traditionally viewed as benign, meningiomas are associated with significant patient morbidity, and certain meningioma subgroups display more aggressive and malignant behavior with higher rates of recurrence. Historically, the risk stratification of meningioma recurrence has been primarily associated with the World Health Organization histopathological grade and surgical extent of resection. However, a growing body of literature has highlighted the value of utilizing molecular characteristics to assess meningioma aggressiveness and recurrence risk. In this review, we discuss preclinical and clinical evidence surrounding the use of molecular classification schemes for meningioma prognostication. We also highlight how molecular data may inform meningioma treatment strategies and future directions.
- Published
- 2023
5. CDKN2A/B co-deletion is associated with increased risk of local and distant intracranial recurrence after surgical resection of brain metastases.
- Author
-
Morshed, Ramin A, Nguyen, Minh P, Cummins, Daniel D, Saggi, Satvir, Young, Jacob S, Haddad, Alexander F, Goldschmidt, Ezequiel, Chang, Edward F, McDermott, Michael W, Berger, Mitchel S, Theodosopoulos, Philip V, Hervey-Jumper, Shawn L, Daras, Mariza, and Aghi, Manish K
- Subjects
CDKN2A ,CDKN2B ,brain metastasis ,distant recurrence ,local recurrence ,surgery ,Genetics ,Patient Safety ,Cancer ,Neurosciences ,Human Genome ,Clinical Research ,Rare Diseases ,Brain Disorders - Abstract
BackgroundWhile genetic alterations in brain metastases (BMs) have been previously explored, there are limited data examining their association with recurrence after surgical resection. This study aimed to identify genetic alterations within BMs associated with CNS recurrence after surgery across multiple cancer types.MethodsA retrospective, single-center study was conducted with patients who underwent resection of a BM with available clinical and gene sequencing data available. Local and remote CNS recurrence were the primary study outcomes. Next-generation sequencing of the coding regions in over 500 oncogenes was performed in brain metastasis specimens. Cox proportional hazards analyses were performed to identify clinical features and genomic alterations associated with CNS recurrence.ResultsA total of 90 patients undergoing resection of 91 BMs composed the cohort. Genes most frequently mutated in the cohort included TP53 (64%), CDKN2A (37%), TERT (29%), CDKN2B (23%), NF1 (14%), KRAS (14%), and PTEN (13%), all of which occurred across multiple cancer types. CDKN2A/B co-deletion was seen in 21 (23.1%) brain metastases across multiple cancer types. In multivariate Cox proportional hazard analyses including patient, tumor, and treatment factors, CDKN2A/B co-deletion in the brain metastasis was associated with increased risk of local (HR 4.07, 95% CI 1.32-12.54, P = 0.014) and remote (HR 2.28, 95% CI 1.11-4.69, P = 0.025) CNS progression. Median survival and length of follow-up were not different based on CDKN2A/B mutation status.ConclusionsCDKN2A/B co-deletion detected in BMs is associated with increased CNS recurrence after surgical resection. Additional work is needed to determine whether more aggressive treatment in patients with this mutation may improve outcomes.
- Published
- 2023
6. Resistance to immune checkpoint blockade: Mechanisms, counter-acting approaches, and future directions
- Author
-
Haddad, Alexander F, Young, Jacob S, Gill, Sabraj, and Aghi, Manish K
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Cancer ,Good Health and Well Being ,Humans ,Immune Checkpoint Inhibitors ,Immunotherapy ,Neoplasms ,Checkpoint inhibitors ,Resistance ,Biomarkers ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Immunotherapies seek to unleash the immune system against cancer cells. While a variety of immunotherapies exist, one of the most commonly used is immune checkpoint blockade, which refers to the use of antibodies to interfere with immunosuppressive signaling through immune checkpoint molecules. Therapies against various checkpoints have had success in the clinic across cancer types. However, the efficacy of checkpoint inhibitors has varied across different cancer types and non-responsive patient populations have emerged. Non-responders to these therapies have highlighted the importance of understanding underlying mechanisms of resistance in order to predict which patients will respond and to tailor individual treatment paradigms. In this review we discuss the literature surrounding tumor mediated mechanisms of immune checkpoint resistance. We also describe efforts to overcome resistance and combine checkpoint inhibitors with additional immunotherapies. Finally, we provide insight into the future of immune checkpoint blockade, including the need for improved preclinical modeling and predictive biomarkers to facilitate personalized cancer treatments for patients.
- Published
- 2022
7. Prospective genomically guided identification of “early/evolving” and “undersampled” IDH-wildtype glioblastoma leads to improved clinical outcomes
- Author
-
Zhang, Yalan, Lucas, Calixto-Hope G, Young, Jacob S, Morshed, Ramin A, McCoy, Lucie, Bush, Nancy Ann Oberheim, Taylor, Jennie W, Daras, Mariza, Butowski, Nicholas A, Villanueva-Meyer, Javier E, Cha, Soonmee, Wrensch, Margaret, Wiencke, John K, Lee, Julieann C, Pekmezci, Melike, Phillips, Joanna J, Perry, Arie, Bollen, Andrew W, Aghi, Manish K, Theodosopoulos, Philip, Chang, Edward F, Hervey-Jumper, Shawn L, Berger, Mitchel S, Clarke, Jennifer L, Chang, Susan M, Molinaro, Annette M, and Solomon, David A
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Rare Diseases ,Cancer ,Biotechnology ,Brain Disorders ,Neurosciences ,Clinical Research ,Human Genome ,Brain Cancer ,Genetics ,Detection ,screening and diagnosis ,4.2 Evaluation of markers and technologies ,Adult ,Astrocytoma ,Brain Neoplasms ,Glioblastoma ,Glioma ,Humans ,Isocitrate Dehydrogenase ,Mutation ,Prospective Studies ,genomic profiling ,glioblastoma ,molecular diagnostics ,molecular neuro-oncology ,precision medicine ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
BackgroundGenomic profiling studies of diffuse gliomas have led to new improved classification schemes that better predict patient outcomes compared to conventional histomorphology alone. One example is the recognition that patients with IDH-wildtype diffuse astrocytic gliomas demonstrating lower-grade histologic features but genomic and/or epigenomic profile characteristic of glioblastoma typically have poor outcomes similar to patients with histologically diagnosed glioblastoma. Here we sought to determine the clinical impact of prospective genomic profiling for these IDH-wildtype diffuse astrocytic gliomas lacking high-grade histologic features but with molecular profile of glioblastoma.MethodsClinical management and outcomes were analyzed for 38 consecutive adult patients with IDH-wildtype diffuse astrocytic gliomas lacking necrosis or microvascular proliferation on histologic examination that were genomically profiled on a prospective clinical basis revealing criteria for an integrated diagnosis of "diffuse astrocytic glioma, IDH-wildtype, with molecular features of glioblastoma, WHO grade IV" per cIMPACT-NOW criteria.ResultsWe identified that this diagnosis consists of two divergent clinical scenarios based on integration of radiologic, histologic, and genomic features that we term "early/evolving" and "undersampled" glioblastoma, IDH-wildtype. We found that prospective genomically guided identification of early/evolving and undersampled IDH-wildtype glioblastoma resulted in more aggressive patient management and improved clinical outcomes compared to a biologically matched historical control patient cohort receiving standard-of-care therapy based on histomorphologic diagnosis alone.ConclusionsThese results support routine use of genomic and/or epigenomic profiling to accurately classify glial neoplasms, as these assays not only improve diagnostic classification but critically lead to more appropriate patient management that can improve clinical outcomes.
- Published
- 2022
8. Impact of the COVID-19 Pandemic on Neurosurgical Transfers: A Single Tertiary Center Study.
- Author
-
Reihl, Sheantel J, Garcia, Joseph H, Morshed, Ramin A, Sankaran, Sujatha, DiGiorgio, Anthony, Chou, Dean, Theodosopoulos, Philip V, Aghi, Manish K, Berger, Mitchel S, Chang, Edward F, and Mummaneni, Praveen V
- Subjects
Humans ,Patient Transfer ,Retrospective Studies ,Neurosurgery ,Pandemics ,Tertiary Care Centers ,COVID-19 ,Catchment area ,Pandemic ,Transfers ,Patient Safety ,Clinical Research ,Clinical Sciences ,Neurosciences - Abstract
ObjectiveInterfacility transfers represent a large proportion of neurosurgical admissions to tertiary care centers each year. In this study, the authors examined the impact of the COVID-19 pandemic on the number of transfers, timing of transfers, demographic profile of transfer patients, and clinical outcomes including rates of surgical intervention.MethodsA retrospective review of neurosurgical transfer patients at a single tertiary center was performed. Patients transferred from April to November 2020 (the "COVID Era") were compared with an institutional database of transfer patients collected before the COVID-19 pandemic (the "Pre-COVID Era"). During the COVID Era, both emergent and nonemergent neurosurgical services had resumed. A comparison of demographic and clinical factors between the 2 cohorts was performed.ResultsA total of 674 patients were included in the study (331 Pre-COVID and 343 COVID-Era patients). Overall, there was no change in the average monthly number of transfers (P = 0.66) or in the catchment area of referral hospitals. However, COVID-Era patients were more likely to be uninsured (1% vs. 4%), had longer transfer times (COVID vs. Pre-COVID Era: 18 vs. 9 hours; P < 0.001), required higher rates of surgical intervention (63% vs. 50%, P = 0.001), had higher rates of spine pathology (17% vs. 10%), and less frequently were admitted to the intensive care unit (34% vs. 52%, P < 0.001). Overall, COVID-Era patients did not experience delays to surgical intervention (3.1 days vs. 3.6 days, P = 0.2). When analyzing the subgroup of COVID-Era patients, COVID infection status did not impact the time of transfer or rates of operation, although COVID-infected patients experienced a longer time to surgery after admission (14 vs. 2.9 days, P < 0.001).ConclusionThe COVID-19 pandemic did not reduce the number of monthly transfers, operation rates, or catchment area for transfer patients. Transfer rates of uninsured patients increased during the COVID Era, potentially reflecting changes in access to community neurosurgery care. Shorter time to surgery seen in COVID-Era patients possibly reflects institutional policies that improved operating room efficiency to compensate for surgical backlogs. COVID status affeted time to surgery, reflecting the preoperative care that these patients require before intervention.
- Published
- 2022
9. Pituitary adenoma or neuroendocrine tumour: the need for an integrated prognostic classification
- Author
-
Ho, Ken K. Y., Kaiser, Ursula B., Chanson, Phillippe, Gadelha, Monica, Wass, John, Nieman, Lynnette, Little, Andrew, Aghi, Manish K., Raetzman, Lori, Post, Kalmon, Raverot, Gerald, Borowsky, Alexander D., Erickson, Dana, Castaño, Justo P., Laws, Edward R., Zatelli, Maria Chiara, Sisco, Jill, Esserman, Laura, Yuen, Kevin C. J., Reincke, Martin, and Melmed, Shlomo
- Published
- 2023
- Full Text
- View/download PDF
10. Glioblastoma induces the recruitment and differentiation of dendritic-like “hybrid” neutrophils from skull bone marrow
- Author
-
Lad, Meeki, Beniwal, Angad S., Jain, Saket, Shukla, Poojan, Kalistratova, Venina, Jung, Jangham, Shah, Sumedh S., Yagnik, Garima, Saha, Atul, Sati, Ankita, Babikir, Husam, Nguyen, Alan T., Gill, Sabraj, Rios, Jennifer, Young, Jacob S., Lui, Austin, Salha, Diana, Diaz, Aaron, and Aghi, Manish K.
- Published
- 2024
- Full Text
- View/download PDF
11. Sarcopenia Diagnosed Using Masseter Muscle Diameter as a Survival Correlate in Elderly Patients with Glioblastoma
- Author
-
Morshed, Ramin A, Young, Jacob S, Casey, Megan, Wang, Elaina J, Aghi, Manish K, Berger, Mitchel S, and Hervey-Jumper, Shawn L
- Subjects
Neurosciences ,Clinical Research ,Brain Cancer ,Aging ,Patient Safety ,Brain Disorders ,Rare Diseases ,Cancer ,Detection ,screening and diagnosis ,Evaluation of treatments and therapeutic interventions ,6.4 Surgery ,4.2 Evaluation of markers and technologies ,Good Health and Well Being ,Aged ,Aged ,80 and over ,Glioblastoma ,Humans ,Masseter Muscle ,Patients ,Retrospective Studies ,Sarcopenia ,Biopsy ,Complications ,Elderly ,Masseter ,Octogenarian ,Resection ,Clinical Sciences - Abstract
BackgroundElderly patients with glioblastoma (GBM) have a worse prognosis than do younger patients. The present study aimed to identify the patient, treatment, and imaging features, including measures of sarcopenia, associated with worse survival and 90-day postoperative mortality for elderly patients with GBM.MethodsA single-center retrospective study was conducted of patients aged ≥79 years at surgery who had undergone biopsy or resection of a World Health Organization grade IV GBM at the initial diagnosis. Imaging features of sarcopenia were collected, including the masseter and temporalis muscle diameters. Multivariate analyses were performed to identify factors associated with survival and 30-day complications.ResultsThe cohort included 110 patients with a mean age of 82.8 years at surgery and a median preoperative Karnofsky performance scale score of 80. The majority of patients underwent a surgical resection (66.4%) while a minority underwent biopsy (33.6%). Adjuvant chemo- and/or radiation therapy were used in 72.5% of the cohort. On multivariate analysis, age (hazard ratio [HR], 7.97; 95% confidence interval [CI], 1.63-36.3), adjuvant therapy (RT or TMZ vs. none: HR, 0.12; 95% CI, 0.05-0.3; RT plus TMZ vs. none: HR, 0.05; 95% CI, 0.02-0.14), surgical resection (HR, 0.46; 95% CI, 0.24-0.9), multifocality (HR, 2.7; 95% CI, 1.14-6.4), and masseter diameter (HR, 0.12; 95% CI, 0.02-0.78) were associated with survival. Masseter diameter was the only factor associated with 90-day mortality after surgical resection (P = 0.044).ConclusionsGBM patients over the age of 79 have acceptable outcomes after resection, followed by adjuvant chemotherapy and RT. In addition to the treatment factors that predicted for survival, a decreased masseter diameter on preoperative imaging, a marker of sarcopenia, was associated with shorter overall survival and 90-day mortality after surgical resection.
- Published
- 2022
12. The immunology of low-grade gliomas.
- Author
-
Haddad, Alexander F, Young, Jacob S, Oh, Jun Yeop, Okada, Hideho, and Aghi, Manish K
- Subjects
Rare Diseases ,Brain Cancer ,Brain Disorders ,Vaccine Related ,Cancer ,Orphan Drug ,Immunization ,Good Health and Well Being ,Brain Neoplasms ,Glioma ,Humans ,Isocitrate Dehydrogenase ,Mutation ,Neoplasm Grading ,Prognosis ,Quality of Life ,Tumor Microenvironment ,low-grade glioma ,immunology ,microenvironment ,immune cell infiltration ,Clinical Sciences ,Neurosciences ,Neurology & Neurosurgery - Abstract
Low-grade gliomas (LGGs), which harbor an isocitrate dehydrogenase (IDH) mutation, have a better prognosis than their high-grade counterparts; nonetheless, they remain incurable and impart significant negative impacts on patients' quality of life. Although immunotherapies represent a novel avenue of treatment for patients with LGGs, they have not yet been successful. Accurately selecting and evaluating immunotherapies requires a detailed understanding of LGG tumor immunology and the underlying tumor immune phenotype. A growing body of literature suggests that LGGs significantly differ in their immunology from high-grade gliomas, highlighting the importance of investigation into LGG immunology specifically. In this review, the authors aimed to discuss relevant research surrounding the LGG tumor immune microenvironment, including immune cell infiltration, tumor immunogenicity, checkpoint molecule expression, the impact of an IDH mutation, and implications for immunotherapies, while also briefly touching on current immunotherapy trials and future directions for LGG immunology research.
- Published
- 2022
13. Pituitary adenomas and cerebrovascular disease: A review on pathophysiology, prevalence, and treatment
- Author
-
Osorio, Robert C, Oh, Jun Y, Choudhary, Nikita, Lad, Meeki, Savastano, Luis, and Aghi, Manish K
- Subjects
Paediatrics ,Biomedical and Clinical Sciences ,Brain Disorders ,Cardiovascular ,Prevention ,Rare Diseases ,Clinical Research ,Good Health and Well Being ,Humans ,Pituitary Neoplasms ,Acromegaly ,Prevalence ,Cardiovascular Diseases ,Adenoma ,Growth Hormone ,Cerebrovascular Disorders ,pituitary adenoma ,cerebrovascular disease ,cerebral infarct ,stroke ,pituitary neuroendocrine tumor ,Clinical Sciences ,Nutrition and Dietetics ,Clinical sciences - Abstract
Pituitary adenomas (PAs) have been shown to cause excess cardiovascular disease comorbidity and mortality. Cerebrovascular disease (CeVD) is a small subset of cardiovascular disease with high morbidity, and its risk in patients with pituitary adenomas has been sparingly explored. In this review, we examine what is known about the prevalence of cerebrovascular disease in patients with PAs, from its initial discovery in 1970 to present. An abundance of literature describes increased cerebrovascular mortality in patients with acromegaly, while research on other PA subtypes is less frequent but shows a similarly elevated CeVD mortality relative to healthy populations. We also review how cerebrovascular risk changes after PAs are treated, with PA treatment appearing to prevent further accumulation of cerebrovascular risk without reversing prior elevations. While acromegaly-associated CeVD appears to be caused by elevated growth hormone (GH) levels and Cushing disease's elevated glucocorticoids similarly cause durable alterations in cerebrovascular structure and function, less is known about the mechanisms behind CeVD in other PA subpopulations. Proposed pathophysiologies include growth hormone deficiency inducing vessel wall damage or other hormone deficits causing increased atherosclerotic disease. Early diagnosis and treatment of PAs may be the key to minimizing lifetime CeVD risk elevations. More research is needed to better understand the mechanisms behind the increased CeVD seen in patients with PAs. Physicians caring for PA patients must remain vigilant for signs and symptoms of cerebrovascular disease in this patient population.
- Published
- 2022
14. CDK 4/6 inhibitors for the treatment of meningioma
- Author
-
Young, Jacob S, Kidwell, Reilly L, Zheng, Allison, Haddad, Alex F, Aghi, Manish K, Raleigh, David R, Schulte, Jessica D, and Butowski, Nicholas A
- Subjects
Rare Diseases ,Brain Cancer ,Cancer ,Brain Disorders ,Neurosciences ,Clinical Trials and Supportive Activities ,Clinical Research ,CDK inhibitor ,meningioma ,cell cycle dysregulation ,clinical trials ,molecular profiling and subtyping ,Oncology and Carcinogenesis - Abstract
Meningiomas are the most common non-metastatic brain tumors, and although the majority are relatively slow-growing and histologically benign, a subset of meningiomas are aggressive and remain challenging to treat. Despite a standard of care that includes surgical resection and radiotherapy, and recent advances in meningioma molecular grouping, there are no systemic medical options for patients with meningiomas that are resistant to standard interventions. Misactivation of the cell cycle at the level of CDK4/6 is common in high-grade or molecularly aggressive meningiomas, and CDK4/6 has emerged as a potential target for systemic meningioma treatments. In this review, we describe the preclinical evidence for CDK4/6 inhibitors as a treatment for high-grade meningiomas and summarize evolving clinical experience with these agents. Further, we highlight upcoming clinical trials for patients meningiomas, and discuss future directions aimed at optimizing the efficacy of these therapies and selecting patients most likely to benefit from their use.
- Published
- 2022
15. ATRX regulates glial identity and the tumor microenvironment in IDH-mutant glioma
- Author
-
Babikir, Husam, Wang, Lin, Shamardani, Karin, Catalan, Francisca, Sudhir, Sweta, Aghi, Manish K, Raleigh, David R, Phillips, Joanna J, and Diaz, Aaron A
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Brain Cancer ,Genetics ,Neurosciences ,Cancer Genomics ,Orphan Drug ,Cancer ,Rare Diseases ,Human Genome ,Brain Disorders ,2.1 Biological and endogenous factors ,Animals ,Biomarkers ,Tumor ,Brain Neoplasms ,Glioma ,Humans ,Mice ,Mutation ,Prognosis ,Transcription Factors ,Transcriptome ,Tumor Microenvironment ,X-linked Nuclear Protein ,Environmental Sciences ,Information and Computing Sciences ,Bioinformatics - Abstract
BackgroundRecent single-cell transcriptomic studies report that IDH-mutant gliomas share a common hierarchy of cellular phenotypes, independent of genetic subtype. However, the genetic differences between IDH-mutant glioma subtypes are prognostic, predictive of response to chemotherapy, and correlate with distinct tumor microenvironments.ResultsTo reconcile these findings, we profile 22 human IDH-mutant gliomas using scATAC-seq and scRNA-seq. We determine the cell-type-specific differences in transcription factor expression and associated regulatory grammars between IDH-mutant glioma subtypes. We find that while IDH-mutant gliomas do share a common distribution of cell types, there are significant differences in the expression and targeting of transcription factors that regulate glial identity and cytokine elaboration. We knock out the chromatin remodeler ATRX, which suffers loss-of-function alterations in most IDH-mutant astrocytomas, in an IDH-mutant immunocompetent intracranial murine model. We find that both human ATRX-mutant gliomas and murine ATRX-knockout gliomas are more heavily infiltrated by immunosuppressive monocytic-lineage cells derived from circulation than ATRX-intact gliomas, in an IDH-mutant background. ATRX knockout in murine glioma recapitulates gene expression and open chromatin signatures that are specific to human ATRX-mutant astrocytomas, including drivers of astrocytic lineage and immune-cell chemotaxis. Through single-cell cleavage under targets and tagmentation assays and meta-analysis of public data, we show that ATRX loss leads to a global depletion in CCCTC-binding factor association with DNA, gene dysregulation along associated chromatin loops, and protection from therapy-induced senescence.ConclusionsThese studies explain how IDH-mutant gliomas from different subtypes maintain distinct phenotypes and tumor microenvironments despite a common lineage hierarchy.
- Published
- 2021
16. Survival outcomes and prognostic factors of infratentorial glioblastoma in the elderly
- Author
-
Chandra, Ankush, Lopez-Rivera, Victor, Ryba, Bryan, Chandran, Arjun S., Brandel, Michael G., Dono, Antonio, Sheinberg, Dallas L., Esquenazi, Yoshua L., and Aghi, Manish K.
- Published
- 2024
- Full Text
- View/download PDF
17. Correlation of Brain Metastasis Genomic Alterations with Preoperative Imaging Features
- Author
-
Uggerly, Amalie S.V., Cummins, Daniel D., Nguyen, Minh P., Saggi, Satvir, Aghi, Manish K., and Morshed, Ramin A.
- Published
- 2024
- Full Text
- View/download PDF
18. Modified RANO, Immunotherapy RANO, and Standard RANO Response to Convection-Enhanced Delivery of IL4R-Targeted Immunotoxin MDNA55 in Recurrent Glioblastoma
- Author
-
Ellingson, Benjamin M, Sampson, John, Achrol, Achal Singh, Aghi, Manish K, Bankiewicz, Krystof, Wang, Chencai, Bexon, Martin, Brem, Steven, Brenner, Andrew, Chowdhary, Sajeel, Floyd, John R, Han, Seunggu, Kesari, Santosh, Randazzo, Dina, Vogelbaum, Michael A, Vrionis, Frank, Zabek, Miroslaw, Butowski, Nicholas, Coello, Melissa, Merchant, Nina, and Merchant, Fahar
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunization ,Rare Diseases ,Cancer ,Adult ,Aged ,Disease-Free Survival ,Female ,Glioblastoma ,Humans ,Immunotherapy ,Immunotoxins ,Interleukin-4 Receptor alpha Subunit ,Male ,Middle Aged ,Neoplasm Recurrence ,Local ,Nervous System Neoplasms ,Prospective Studies ,Survival Rate ,Treatment Outcome ,Oncology & Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis - Abstract
PurposeThe current study compared the standard response assessment in neuro-oncology (RANO), immunotherapy RANO (iRANO), and modified RANO (mRANO) criteria as well as quantified the association between progression-free (PFS) and overall survival (OS) in an immunotherapy trial in recurrent glioblastoma (rGBM).Patients and methodsA total of 47 patients with rGBM were enrolled in a prospective phase II convection-enhanced delivery of an IL4R-targeted immunotoxin (MDNA55-05, NCT02858895). Bidirectional tumor measurements were created by local sites and centrally by an independent radiologic faculty, then standard RANO, iRANO, and mRANO criteria were applied.ResultsA total of 41 of 47 patients (mean age 56 ± 11.7) were evaluable for response. PFS was significantly shorter using standard RANO compared with iRANO (log-rank, P < 0.0001; HR = 0.3) and mRANO (P < 0.0001; HR = 0.3). In patients who died and had confirmed progression on standard RANO, no correlation was observed between PFS and OS (local, P = 0.47; central, P = 0.34). Using iRANO, a weak association was observed between confirmed PFS and OS via local site measurements (P = 0.017), but not central measurements (P = 0.18). A total of 24 of 41 patients (59%) were censored using iRANO and because they lacked confirmation of progression 3 months after initial progression. A strong correlation was observed between mRANO PFS and OS for both local (R2 = 0.66, P < 0.0001) and centrally determined reads (R2 = 0.57, P = 0.0007).ConclusionsNo correlation between radiographic PFS and OS was observed for standard RANO or iRANO, but a correlation was observed between PFS and OS using the mRANO criteria. Also, the iRANO criteria was difficult to implement due to need to confirm progression 3 months after initial progression, censoring more than half the patients.
- Published
- 2021
19. Transsphenoidal Surgery for Acromegaly
- Author
-
Phelps, Ryan R. L., Young, Jacob S., Gurrola, José, II, Aghi, Manish K., Poretsky, Leonid, Series Editor, Blevins Jr., Lewis S., editor, and Aghi, Manish K., editor
- Published
- 2022
- Full Text
- View/download PDF
20. The Molecular Biology and Pathology of Acromegaly
- Author
-
Carrete, Luis R., Aghi, Manish K., Poretsky, Leonid, Series Editor, Blevins Jr., Lewis S., editor, and Aghi, Manish K., editor
- Published
- 2022
- Full Text
- View/download PDF
21. Multiomic screening of invasive GBM cells reveals targetable transsulfuration pathway alterations
- Author
-
Garcia, Joseph H., Akins, Erin A., Jain, Saket, Wolf, Kayla J., Zhang, Jason, Choudhary, Nikita, Lad, Meeki, Shukla, Poojan, Rios, Jennifer, Seo, Kyounghee, Gill, Sabraj A., Carson, William H., Carette, Luis R., Zheng, Allison C., Raleigh, David R., Kumar, Sanjay, and Aghi, Manish K.
- Subjects
Thermo Fisher Scientific Inc. ,Scientific equipment and supplies industry -- Physiological aspects -- Analysis ,Ceramides -- Physiological aspects -- Analysis ,Genes -- Physiological aspects -- Analysis ,Biological products -- Physiological aspects -- Analysis ,Gliomas -- Prognosis ,Cysteine -- Physiological aspects -- Analysis ,Health care industry - Abstract
While the poor prognosis of glioblastoma arises from the invasion of a subset of tumor cells, little is known of the metabolic alterations within these cells that fuel invasion. We integrated spatially addressable hydrogel biomaterial platforms, patient site-directed biopsies, and multiomics analyses to define metabolic drivers of invasive glioblastoma cells. Metabolomics and lipidomics revealed elevations in the redox buffers cystathionine, hexosylceramides, and glucosyl ceramides in the invasive front of both hydrogel-cultured tumors and patient site-directed biopsies, with immunofluorescence indicating elevated reactive oxygen species (ROS) markers in invasive cells. Transcriptomics confirmed upregulation of ROS-producing and response genes at the invasive front in both hydrogel models and patient tumors. Among oncologic ROS, [H.sub.2][O.sub.2] specifically promoted glioblastoma invasion in 3D hydrogel spheroid cultures. A CRISPR metabolic gene screen revealed cystathionine [gamma]-lyase (CTH), which converts cystathionine to the nonessential amino acid cysteine in the transsulfuration pathway, to be essential for glioblastoma invasion. Correspondingly, supplementing CTH knockdown cells with exogenous cysteine rescued invasion. Pharmacologic CTH inhibition suppressed glioblastoma invasion, while CTH knockdown slowed glioblastoma invasion in vivo. Our studies highlight the importance of ROS metabolism in invasive glioblastoma cells and support further exploration of the transsulfuration pathway as a mechanistic and therapeutic target., Introduction Glioblastoma (GBM) is the most common and lethal adult brain tumor (1) and is characterized by an unparalleled invasive capacity (2). Current therapeutic strategies are insufficient to control the [...]
- Published
- 2024
- Full Text
- View/download PDF
22. Immunologic aspects of viral therapy for glioblastoma and implications for interactions with immunotherapies
- Author
-
Haddad, Alexander F, Young, Jacob S, Mummaneni, Nikhil V, Kasahara, Noriyuki, and Aghi, Manish K
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Genetics ,Vaccine Related ,Cancer ,Orphan Drug ,Immunization ,Brain Disorders ,Brain Cancer ,Rare Diseases ,Neurosciences ,Gene Therapy ,Development of treatments and therapeutic interventions ,5.2 Cellular and gene therapies ,5.1 Pharmaceuticals ,Animals ,Brain Neoplasms ,Combined Modality Therapy ,Glioblastoma ,Humans ,Immunotherapy ,Oncolytic Virotherapy ,Oncolytic viruses ,Combination ,Treatment ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
IntroductionThe treatment for glioblastoma (GBM) has remained unchanged for the past decade, with only minimal improvements in patient survival. As a result, novel treatments are needed to combat this devastating disease. Immunotherapies are treatments that stimulate the immune system to attack tumor cells and can be either local or systemically delivered. Viral treatments can lead to direct tumor cell death through their natural lifecycle or through the delivery of a suicide gene, with the potential to generate an anti-tumor immune response, making them interesting candidates for combinatorial treatment with immunotherapy.MethodsWe review the current literature surrounding the interactions between oncolytic viruses and the immune system as well as the use of oncolytic viruses combined with immunotherapies for the treatment of GBM.ResultsViral therapies have exhibited preclinical efficacy as single-agents and are being investigated in that manner in clinical trials. Oncolytic viruses have significant interactions with the immune system, although this can also vary depending on the strain of virus. Combinatorial treatments using both oncolytic viruses and immunotherapies have demonstrated promising preclinical findings.ConclusionsStudies combining viral and immunotherapeutic treatment modalities have provided exciting results thus far and hold great promise for patients with GBM. Additional studies assessing the clinical efficacy of these treatments as well as improved preclinical modeling systems, safety mechanisms, and the balance between treatment efficacy and immune-mediated viral clearance should be considered.
- Published
- 2021
23. Mouse Models of Glioblastoma for the Evaluation of Novel Therapeutic Strategies
- Author
-
Haddad, Alexander F, Young, Jacob S, Amara, Dominic, Berger, Mitchel S, Raleigh, David R, Aghi, Manish K, and Butowski, Nicholas A
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Brain Disorders ,Neurosciences ,Cancer ,Orphan Drug ,Brain Cancer ,Biotechnology ,Rare Diseases ,murine models ,glioblastoma ,tumor ,U87 ,GL261 - Abstract
Glioblastoma (GBM) is an incurable brain tumor with a median survival of approximately 15 months despite an aggressive standard of care that includes surgery, chemotherapy, and ionizing radiation. Mouse models have advanced our understanding of GBM biology and the development of novel therapeutic strategies for GBM patients. However, model selection is crucial when testing developmental therapeutics, and each mouse model of GBM has unique advantages and disadvantages that can influence the validity and translatability of experimental results. To shed light on this process, we discuss the strengths and limitations of 3 types of mouse GBM models in this review: syngeneic models, genetically engineered mouse models, and xenograft models, including traditional xenograft cell lines and patient-derived xenograft models.
- Published
- 2021
24. Are preoperative chlorhexidine gluconate showers associated with a reduction in surgical site infection following craniotomy? A retrospective cohort analysis of 3126 surgical procedures.
- Author
-
Ammanuel, Simon G, Edwards, Caleb S, Chan, Andrew K, Mummaneni, Praveen V, Kidane, Joseph, Vargas, Enrique, D'Souza, Sarah, Nichols, Amy D, Sankaran, Sujatha, Abla, Adib A, Aghi, Manish K, Chang, Edward F, Hervey-Jumper, Shawn L, Kunwar, Sandeep, Larson, Paul S, Lawton, Michael T, Starr, Philip A, Theodosopoulos, Philip V, Berger, Mitchel S, and McDermott, Michael W
- Subjects
Patient Safety ,surgical site infection ,chlorhexidine ,craniotomy ,antisepsis ,preoperative showers ,Clinical Sciences ,Neurosciences ,Neurology & Neurosurgery - Abstract
ObjectiveSurgical site infection (SSI) is a complication linked to increased costs and length of hospital stay. Prevention of SSI is important to reduce its burden on individual patients and the healthcare system. The authors aimed to assess the efficacy of preoperative chlorhexidine gluconate (CHG) showers on SSI rates following cranial surgery.MethodsIn November 2013, a preoperative CHG shower protocol was implemented at the authors' institution. A total of 3126 surgical procedures were analyzed, encompassing a time frame from April 2012 to April 2016. Cohorts before and after implementation of the CHG shower protocol were evaluated for differences in SSI rates.ResultsThe overall SSI rate was 0.6%. No significant differences (p = 0.11) were observed between the rate of SSI of the 892 patients in the preimplementation cohort (0.2%) and that of the 2234 patients in the postimplementation cohort (0.8%). Following multivariable analysis, implementation of preoperative CHG showers was not associated with decreased SSI (adjusted OR 2.96, 95% CI 0.67-13.1; p = 0.15).ConclusionsThis is the largest study, according to sample size, to examine the association between CHG showers and SSI following craniotomy. CHG showers did not significantly alter the risk of SSI after a cranial procedure.
- Published
- 2021
25. Immunotherapy Resistance in Glioblastoma
- Author
-
Wang, Elaina J, Chen, Jia-Shu, Jain, Saket, Morshed, Ramin A, Haddad, Alexander F, Gill, Sabraj, Beniwal, Angad S, and Aghi, Manish K
- Subjects
Biological Sciences ,Genetics ,Brain Cancer ,Vaccine Related ,Neurosciences ,Cancer ,Orphan Drug ,Biotechnology ,Brain Disorders ,Immunization ,Rare Diseases ,Evaluation of treatments and therapeutic interventions ,6.1 Pharmaceuticals ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Good Health and Well Being ,glioblastoma ,immunotherapy ,resistance ,immunoprivilege ,checkpoint inhibitors ,vaccine ,CAR (chimeric antigen receptor) T cells ,virotherapy ,Clinical Sciences ,Law - Abstract
Glioblastoma is the most common malignant primary brain tumor in adults. Despite treatment consisting of surgical resection followed by radiotherapy and adjuvant chemotherapy, survival remains poor at a rate of 26.5% at 2 years. Recent successes in using immunotherapies to treat a number of solid and hematologic cancers have led to a growing interest in harnessing the immune system to target glioblastoma. Several studies have examined the efficacy of various immunotherapies, including checkpoint inhibitors, vaccines, adoptive transfer of lymphocytes, and oncolytic virotherapy in both pre-clinical and clinical settings. However, these therapies have yielded mixed results at best when applied to glioblastoma. While the initial failures of immunotherapy were thought to reflect the immunoprivileged environment of the brain, more recent studies have revealed immune escape mechanisms created by the tumor itself and adaptive resistance acquired in response to therapy. Several of these resistance mechanisms hijack key signaling pathways within the immune system to create a protumoral microenvironment. In this review, we discuss immunotherapies that have been trialed in glioblastoma, mechanisms of tumor resistance, and strategies to sensitize these tumors to immunotherapies. Insights gained from the studies summarized here may help pave the way for novel therapies to overcome barriers that have thus far limited the success of immunotherapy in glioblastoma.
- Published
- 2021
26. Comprehensive analysis of diverse low-grade neuroepithelial tumors with FGFR1 alterations reveals a distinct molecular signature of rosette-forming glioneuronal tumor
- Author
-
Lucas, Calixto-Hope G, Gupta, Rohit, Doo, Pamela, Lee, Julieann C, Cadwell, Cathryn R, Ramani, Biswarathan, Hofmann, Jeffrey W, Sloan, Emily A, Kleinschmidt-DeMasters, Bette K, Lee, Han S, Wood, Matthew D, Grafe, Marjorie, Born, Donald, Vogel, Hannes, Salamat, Shahriar, Puccetti, Diane, Scharnhorst, David, Samuel, David, Cooney, Tabitha, Cham, Elaine, Jin, Lee-way, Khatib, Ziad, Maher, Ossama, Chamyan, Gabriel, Brathwaite, Carole, Bannykh, Serguei, Mueller, Sabine, Kline, Cassie N, Banerjee, Anu, Reddy, Alyssa, Taylor, Jennie W, Clarke, Jennifer L, Oberheim Bush, Nancy Ann, Butowski, Nicholas, Gupta, Nalin, Auguste, Kurtis I, Sun, Peter P, Roland, Jarod L, Raffel, Corey, Aghi, Manish K, Theodosopoulos, Philip, Chang, Edward, Hervey-Jumper, Shawn, Phillips, Joanna J, Pekmezci, Melike, Bollen, Andrew W, Tihan, Tarik, Chang, Susan, Berger, Mitchel S, Perry, Arie, and Solomon, David A
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Rare Diseases ,Genetics ,Human Genome ,Aetiology ,2.1 Biological and endogenous factors ,Adolescent ,Adult ,Aged ,Brain Neoplasms ,Child ,Female ,Humans ,Male ,Middle Aged ,Mutation ,Neoplasms ,Neuroepithelial ,Receptor ,Fibroblast Growth Factor ,Type 1 ,Spinal Cord Neoplasms ,Young Adult ,Rosette-forming glioneuronal tumor ,Extraventricular neurocytoma ,Dysembryoplastic neuroepithelial tumor ,Pilocytic astrocytoma ,FGFR1 ,PIK3CA ,Molecular neuropathology ,DNA methylation profiling ,PIK3R1 ,Biochemistry and Cell Biology ,Neurosciences ,Biochemistry and cell biology - Abstract
The FGFR1 gene encoding fibroblast growth factor receptor 1 has emerged as a frequently altered oncogene in the pathogenesis of multiple low-grade neuroepithelial tumor (LGNET) subtypes including pilocytic astrocytoma, dysembryoplastic neuroepithelial tumor (DNT), rosette-forming glioneuronal tumor (RGNT), and extraventricular neurocytoma (EVN). These activating FGFR1 alterations in LGNET can include tandem duplication of the exons encoding the intracellular tyrosine kinase domain, in-frame gene fusions most often with TACC1 as the partner, or hotspot missense mutations within the tyrosine kinase domain (either at p.N546 or p.K656). However, the specificity of these different FGFR1 events for the various LGNET subtypes and accompanying genetic alterations are not well defined. Here we performed comprehensive genomic and epigenomic characterization on a diverse cohort of 30 LGNET with FGFR1 alterations. We identified that RGNT harbors a distinct epigenetic signature compared to other LGNET with FGFR1 alterations, and is uniquely characterized by FGFR1 kinase domain hotspot missense mutations in combination with either PIK3CA or PIK3R1 mutation, often with accompanying NF1 or PTPN11 mutation. In contrast, EVN harbors its own distinct epigenetic signature and is characterized by FGFR1-TACC1 fusion as the solitary pathogenic alteration. Additionally, DNT and pilocytic astrocytoma are characterized by either kinase domain tandem duplication or hotspot missense mutations, occasionally with accompanying NF1 or PTPN11 mutation, but lacking the accompanying PIK3CA or PIK3R1 mutation that characterizes RGNT. The glial component of LGNET with FGFR1 alterations typically has a predominantly oligodendroglial morphology, and many of the pilocytic astrocytomas with FGFR1 alterations lack the biphasic pattern, piloid processes, and Rosenthal fibers that characterize pilocytic astrocytomas with BRAF mutation or fusion. Together, this analysis improves the classification and histopathologic stratification of LGNET with FGFR1 alterations.
- Published
- 2020
27. Phase 0 and window of opportunity clinical trial design in neuro-oncology: a RANO review
- Author
-
Vogelbaum, Michael A, Krivosheya, Daria, Borghei-Razavi, Hamid, Sanai, Nader, Weller, Michael, Wick, Wolfgang, Soffietti, Riccardo, Reardon, David A, Aghi, Manish K, Galanis, Evanthia, Wen, Patrick Y, van den Bent, Martin, and Chang, Susan
- Subjects
Rare Diseases ,Brain Cancer ,Neurosciences ,Cancer ,Clinical Trials and Supportive Activities ,Orphan Drug ,Brain Disorders ,Patient Safety ,Clinical Research ,Central Nervous System Neoplasms ,Clinical Trials as Topic ,Glioblastoma ,Humans ,Treatment Outcome ,clinical trial ,glioblastoma ,pharmacodynamics ,pharmacokinetics ,phase 0 ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
Glioblastoma is a devastating disease with poor prognosis. Few effective chemotherapeutics are currently available, and much effort has been expended to identify new drugs capable of slowing tumor progression. The phase 0 trial design was developed to facilitate early identification of promising agents for cancer that should undergo accelerated approval. This design features an early in-human study that enrolls a small number of patients who receive subtherapeutic doses of medication with the goals of describing pharmacokinetics through drug blood level measurements and determining intratumoral concentrations of the investigational compound as well as pharmacodynamics by studying the biochemical and physiological effects of drugs. In neuro-oncology, however, the presence of the blood-brain barrier and difficulty in obtaining brain tumor tissue warrant a separate set of considerations. In this paper, we critically reviewed the protocols used in all brain tumor related in-human phase 0 and phase 0-like ("window of opportunity") studies between 1993 and 2018, as well as ongoing clinical trials, and identified major challenges in trial design as applied to central nervous system tumors that include surgical specimen collection and storage, brain tumor drug level analysis, and confirmation of drug action. We therefore propose that phase 0 trials in neuro-oncology should include (i) only patients in whom a resection of the tumor is planned, (ii) use of clinical doses of an investigational agent, (iii) tissue sampling from enhancing and non-enhancing portions of the tumor, and (iv) assessment of drug-specific target effects. Standardization of clinical protocols for phase 0/window of opportunity studies can help accelerate the development of effective treatments for glioblastoma.
- Published
- 2020
28. Autophagy as a mechanism for anti-angiogenic therapy resistance
- Author
-
Chandra, Ankush, Rick, Jonathan, Yagnik, Garima, and Aghi, Manish K
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Angiogenesis Inhibitors ,Animals ,Autophagy ,Drug Resistance ,Neoplasm ,Humans ,Neoplasms ,Neovascularization ,Pathologic ,Tumor Microenvironment ,Angiogenesis ,Anti-angiogenesis ,VEGF ,Drugresistance ,Drug resistance ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Autophagy is a lysosomal-dependent degradation process that is highly conserved and maintains cellular homeostasis by sequestering cytosolic material for degradation either non-specifically by non-selective autophagy, or targeting specific proteins aggregates by selective autophagy. Autophagy serves as a protective mechanism defending the cell from stressors and also plays an important role in enabling tumor cells to overcome harsh conditions arising in their microenvironment during growth as well as oxidative and non-oxidative injuries secondary to therapeutic stressors. Recently, autophagy has been implicated to cause tumor resistance to anti-angiogenic therapy, joining an existing literature implicating autophagy in cancer resistance to conventional DNA damaging chemotherapy and ionizing radiation. In this review, we discuss the role of angiogenesis in malignancy, mechanisms of resistance to anti-angiogenic therapy in general, the role of autophagy in driving malignancy, and the current literature in autophagy-mediated anti-angiogenic therapy resistance. Finally, we provide future insight into the current challenges of using autophagy inhibitors in the clinic and provides tips for future studies to focus on to effectively target autophagy in overcoming resistance to anti-angiogenic therapy.
- Published
- 2020
29. Multiplatform genomic profiling and magnetic resonance imaging identify mechanisms underlying intratumor heterogeneity in meningioma.
- Author
-
Magill, Stephen T, Vasudevan, Harish N, Seo, Kyounghee, Villanueva-Meyer, Javier E, Choudhury, Abrar, John Liu, S, Pekmezci, Melike, Findakly, Sarah, Hilz, Stephanie, Lastella, Sydney, Demaree, Benjamin, Braunstein, Steve E, Bush, Nancy Ann Oberheim, Aghi, Manish K, Theodosopoulos, Philip V, Sneed, Penny K, Abate, Adam R, Berger, Mitchel S, McDermott, Michael W, Lim, Daniel A, Ullian, Erik M, Costello, Joseph F, and Raleigh, David R
- Subjects
Humans ,Brain Neoplasms ,Meningeal Neoplasms ,Cadherins ,Antigens ,CD ,Genetic Markers ,Magnetic Resonance Imaging ,Diffusion Magnetic Resonance Imaging ,Gene Expression Profiling ,Genomics ,Genetic Heterogeneity ,Aged ,Female ,Receptor-Like Protein Tyrosine Phosphatases ,Class 5 ,Epigenomics ,Transcriptome ,Rare Diseases ,Neurosciences ,Brain Cancer ,Biotechnology ,Brain Disorders ,Cancer ,Human Genome ,Genetics ,2.1 Biological and endogenous factors - Abstract
Meningiomas are the most common primary intracranial tumors, but the molecular drivers of meningioma tumorigenesis are poorly understood. We hypothesized that investigating intratumor heterogeneity in meningiomas would elucidate biologic drivers and reveal new targets for molecular therapy. To test this hypothesis, here we perform multiplatform molecular profiling of 86 spatially-distinct samples from 13 human meningiomas. Our data reveal that regional alterations in chromosome structure underlie clonal transcriptomic, epigenomic, and histopathologic signatures in meningioma. Stereotactic co-registration of sample coordinates to preoperative magnetic resonance images further suggest that high apparent diffusion coefficient (ADC) distinguishes meningioma regions with proliferating cells enriched for developmental gene expression programs. To understand the function of these genes in meningioma, we develop a human cerebral organoid model of meningioma and validate the high ADC marker genes CDH2 and PTPRZ1 as potential targets for meningioma therapy using live imaging, single cell RNA sequencing, CRISPR interference, and pharmacology.
- Published
- 2020
30. Endoscopic skull base and transoral surgery during COVID‐19 pandemic: Minimizing droplet spread with negative‐pressure otolaryngology viral isolation drape
- Author
-
David, Abel P, Jiam, Nicole T, Reither, Joshua M, Gurrola, Jose G, Aghi, Manish K, and El‐Sayed, Ivan H
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Patient Safety ,Dental/Oral and Craniofacial Disease ,Good Health and Well Being ,Air Pollutants ,Occupational ,Betacoronavirus ,COVID-19 ,Coronavirus Infections ,Fluorescein ,Fluorescent Dyes ,Humans ,Infection Control ,Infectious Disease Transmission ,Patient-to-Professional ,Occupational Exposure ,Otorhinolaryngologic Surgical Procedures ,Pandemics ,Pneumonia ,Viral ,Retrospective Studies ,SARS-CoV-2 ,Surgical Drapes ,Ultraviolet Rays ,aerosolization ,endoscopic surgery ,negative-pressure ,skull base ,Dentistry ,Otorhinolaryngology ,Clinical sciences - Abstract
BackgroundThe coronavirus disease (COVID-19) pandemic has raised concern of transmission of infectious organisms through aerosols formation in endonasal and transoral surgery.MethodsRetrospective review. We introduce the negative-pressure otolaryngology viral isolation drape (NOVID) system to reduce the risk of aerosol. NOVID consists of a plastic drape suspended above the patient's head and surgical field with a smoke evacuator suction placed inside the chamber.ResultsFour patients underwent endonasal (4) and endo-oral surgery (1). Fluorescein was applied to the surgical field. Black light examination of fluorescein-treated operative fields revealed minimal contamination distant to the surgical field. In two prolonged cases with high-speed drilling, droplets were identified under the barrier and on the tip of the smoke evacuator. Instruments and cottonoids appeared to be a greater contributor to field contamination.ConclusionsNegative-pressure aspiration of air under a chamber barrier, which appears to successfully keep aerosol and droplet contamination to a minimum.
- Published
- 2020
31. Cerebrospinal Fluid Leaks and Pseudomeningocele after Posterior Fossa Surgery: Effect of an Autospray Dural Sealant.
- Author
-
Lee, Young M, Ordaz, Angel, Durcanova, Beata, Viner, Jennifer A, Theodosopoulos, Philip V, Aghi, Manish K, and McDermott, Michael W
- Subjects
cost analysis ,dural sealant ,posterior fossa surgery ,pseudomeningocele ,Medical and Health Sciences - Abstract
Background Posterior fossa craniotomies can be complicated by cerebrospinal fluid (CSF) leaks, infection, meningitis, neurologic deficits, and intracranial hypotension caused by defective closure of the dura. Secondary dural closures such as pericranial graft, muscle graft, glue, sealants, or fat graft are used. However, there have been few studies examining the use of sealants with a polyethylene glycol and polyethylenimine component. Objective We studied the effect of one such sealant, Adherus® (HyperBranch Medical Technology, Durham, NC, USA), as an adjunct to secondary closure methods in the reduction of the use of abdominal fat grafting and lumbar puncture/drains. Methods We retrospectively reviewed the surgical records of all patients undergoing posterior fossa cranial surgery during a two-year period at a tertiary university affiliated medical center. Results Overall, data a total of 122 patients (62 in the no Adherus and 60 in the Adherus group) were collected. There was no statistically significant difference in the 30-day incisional CSF leak rate (4.1% vs. 6.5%; p=0.183), 30-day non-incisional CSF leak rate (11.3% vs. 5.0%; p=0.205), and 30-day pseudomeningocele rate (16.1% vs. 13.3%; p=0.663) in the no Adherus and Adherus groups, respectively. However, there was a significant reduction in the use of abdominal fat grafting (0% vs. 30.7%; p
- Published
- 2020
32. Clonal ZEB1-Driven Mesenchymal Transition Promotes Targetable Oncologic Antiangiogenic Therapy Resistance
- Author
-
Chandra, Ankush, Jahangiri, Arman, Chen, William, Nguyen, Alan T, Yagnik, Garima, Pereira, Matheus P, Jain, Saket, Garcia, Joseph H, Shah, Sumedh S, Wadhwa, Harsh, Joshi, Rushikesh S, Weiss, Jacob, Wolf, Kayla J, Lin, Jung-Ming G, Müller, Sören, Rick, Jonathan W, Diaz, Aaron A, Gilbert, Luke A, Kumar, Sanjay, and Aghi, Manish K
- Subjects
Stem Cell Research - Nonembryonic - Human ,Brain Cancer ,Brain Disorders ,Rare Diseases ,Biotechnology ,Stem Cell Research ,Cancer ,Aetiology ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,2.1 Biological and endogenous factors ,Adult ,Aged ,Angiogenesis Inhibitors ,Animals ,Antineoplastic Agents ,Phytogenic ,Antineoplastic Combined Chemotherapy Protocols ,Bevacizumab ,Biphenyl Compounds ,Brain ,Brain Neoplasms ,Cell Hypoxia ,Cell Line ,Tumor ,Chitinase-3-Like Protein 1 ,Drug Resistance ,Neoplasm ,Epithelial-Mesenchymal Transition ,Female ,Gene Expression Regulation ,Neoplastic ,Glioblastoma ,Human Umbilical Vein Endothelial Cells ,Humans ,Lignans ,Male ,Middle Aged ,Neoplasm Invasiveness ,Neoplastic Stem Cells ,Neovascularization ,Pathologic ,Tumor Microenvironment ,Up-Regulation ,Xenograft Model Antitumor Assays ,Young Adult ,Zinc Finger E-box-Binding Homeobox 1 ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
Glioblastoma (GBM) responses to bevacizumab are invariably transient with acquired resistance. We profiled paired patient specimens and bevacizumab-resistant xenograft models pre- and post-resistance toward the primary goal of identifying regulators whose targeting could prolong the therapeutic window, and the secondary goal of identifying biomarkers of therapeutic window closure. Bevacizumab-resistant patient specimens and xenografts exhibited decreased vessel density and increased hypoxia versus pre-resistance, suggesting that resistance occurs despite effective therapeutic devascularization. Microarray analysis revealed upregulated mesenchymal genes in resistant tumors correlating with bevacizumab treatment duration and causing three changes enabling resistant tumor growth in hypoxia. First, perivascular invasiveness along remaining blood vessels, which co-opts vessels in a VEGF-independent and neoangiogenesis-independent manner, was upregulated in novel biomimetic 3D bioengineered platforms modeling the bevacizumab-resistant microenvironment. Second, tumor-initiating stem cells housed in the perivascular niche close to remaining blood vessels were enriched. Third, metabolic reprogramming assessed through real-time bioenergetic measurement and metabolomics upregulated glycolysis and suppressed oxidative phosphorylation. Single-cell sequencing of bevacizumab-resistant patient GBMs confirmed upregulated mesenchymal genes, particularly glycoprotein YKL-40 and transcription factor ZEB1, in later clones, implicating these changes as treatment-induced. Serum YKL-40 was elevated in bevacizumab-resistant versus bevacizumab-naïve patients. CRISPR and pharmacologic targeting of ZEB1 with honokiol reversed the mesenchymal gene expression and associated stem cell, invasion, and metabolic changes defining resistance. Honokiol caused greater cell death in bevacizumab-resistant than bevacizumab-responsive tumor cells, with surviving cells losing mesenchymal morphology. Employing YKL-40 as a resistance biomarker and ZEB1 as a target to prevent resistance could fulfill the promise of antiangiogenic therapy. SIGNIFICANCE: Bevacizumab resistance in GBM is associated with mesenchymal/glycolytic shifts involving YKL-40 and ZEB1. Targeting ZEB1 reduces bevacizumab-resistant GBM phenotypes. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/7/1498/F1.large.jpg.
- Published
- 2020
33. WHO Grade I Meningioma Recurrence: Identifying High Risk Patients Using Histopathological Features and the MIB-1 Index
- Author
-
Haddad, Alexander F, Young, Jacob S, Kanungo, Ishan, Sudhir, Sweta, Chen, Jia-Shu, Raleigh, David R, Magill, Stephen T, McDermott, Michael W, and Aghi, Manish K
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Prevention ,meningioma ,WHO grade I ,recurrence ,MIB-1 ,benign ,early recurrence ,late recurrence ,Oncology and Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis - Abstract
Objective: In this study, we identify clinical, radiographic, and histopathologic prognosticators of overall, early, and post-median recurrence in World Health Organization (WHO) grade I meningiomas. We also determine a clinically relevant cutoff for MIB-1 to identify patients at high risk for recurrence. Method: A retrospective review of WHO grade I meningioma patients with available MIB-1 index data who underwent treatment at our institution from 2007 to 2017 was performed. Univariate and multivariate analyses, and recursive partitioning analysis (RPA), were used to identify risk factors for overall, early (within 24 months), and post-median (>24 months post-treatment) recurrence. Result: A total of 239 patients were included. The mean age was 60.0 years, and 69.5% of patients were female. The average follow-up was 41.1 months. All patients received surgery and 2 patients each received either adjuvant radiotherapy (2/239) or gamma knife treatment (2/239). The incidence of recurrence was 10.9% (26/239 patients), with an average time to recurrence of 33.2 months (6-105 months). Posterior fossa tumor location (p = 0.004), MIB-1 staining (p = 0.008), nuclear atypia (p = 0.003), and STR (p < 0.001) were independently associated with an increased risk of recurrence on cox-regression analysis. RPA for overall recurrence highlighted extent of resection, and after gross total resection (GTR), a MIB-1 index cutoff of 4.5% as key prognostic factors for recurrence. Patients with a GTR and MIB-1 >4.5% had a similar incidence of recurrence as those with STR (18.8 vs. 18.6%). Variables independently associated with early recurrence on binary logistic regression modeling included STR (p = 0.002) and nuclear atypia (p = 0.019). RPA confirmed STR as associated with early recurrence. Conclusion: STR, posterior fossa location, nuclear atypia, and elevated MIB-1 index are prognostic factors for WHO grade I meningioma recurrence. Moreover, MIB-1 index >4.5% is prognostic for recurrence in patients with GTR. Verification of our findings in larger, multi-institutional studies could enable risk stratification and recommendations for adjuvant radiotherapy following resection of WHO grade I meningiomas.
- Published
- 2020
34. Clinical, radiologic, and genetic characteristics of histone H3 K27M-mutant diffuse midline gliomas in adults
- Author
-
Schulte, Jessica D, Buerki, Robin A, Lapointe, Sarah, Molinaro, Annette M, Zhang, Yalan, Villanueva-Meyer, Javier E, Perry, Arie, Phillips, Joanna J, Tihan, Tarik, Bollen, Andrew W, Pekmezci, Melike, Butowski, Nicholas, Bush, Nancy Ann Oberheim, Taylor, Jennie W, Chang, Susan M, Theodosopoulos, Philip, Aghi, Manish K, Hervey-Jumper, Shawn L, Berger, Mitchel S, Solomon, David A, and Clarke, Jennifer L
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Pediatric Cancer ,Brain Disorders ,Pediatric ,Brain Cancer ,Cancer Genomics ,Neurosciences ,Cancer ,Human Genome ,Rare Diseases ,Genetics ,Orphan Drug ,2.1 Biological and endogenous factors ,adult ,diffuse midline glioma ,genetics ,H3 K27M ,survival - Abstract
Background"Diffuse midline glioma (DMG), H3 K27M-mutant" is a new tumor entity established in the 2016 WHO classification of Tumors of the Central Nervous System that comprises a set of diffuse gliomas arising in midline structures and is molecularly defined by a K27M mutation in genes encoding the histone 3 variants H3.3 or H3.1. While this tumor entity is associated with poor prognosis in children, clinical experience in adults remains limited.MethodsPatient demographics, radiologic and pathologic characteristics, treatment course, progression, and patient survival were collected for 60 adult patients with DMG, H3 K27M-mutant. A subset of tumors also underwent next-generation sequencing. Analysis of progression-free survival and overall survival was conducted using Kaplan-Meier modeling, and univariate and multivariate analysis.ResultsMedian patient age was 32 years (range 18-71 years). Tumors were centered in the thalamus (n = 34), spinal cord (10), brainstem (5), cerebellum (4), or other midline sites (4), or were multifocal (3). Genomic profiling revealed p.K27M mutations exclusively in the H3F3A gene and an absence of mutations in HIST1H3B or HIST1H3C, which are present in approximately one-third of pediatric DMGs. Accompanying mutations in TP53, PPM1D, FGFR1, NF1, and ATRX were frequently found. The overall survival of this adult cohort was 27.6 months, longer than historical averages for both H3 K27M-mutant DMG in children and IDH-wildtype glioblastoma in adults.ConclusionsTogether, these findings indicate that H3 K27M-mutant DMG represents a heterogeneous disease with regard to outcomes, sites of origin, and molecular pathogenesis in adults versus children.
- Published
- 2020
35. The Phenotypes of Proliferating Glioblastoma Cells Reside on a Single Axis of Variation
- Author
-
Wang, Lin, Babikir, Husam, Müller, Sören, Yagnik, Garima, Shamardani, Karin, Catalan, Francisca, Kohanbash, Gary, Alvarado, Beatriz, Di Lullo, Elizabeth, Kriegstein, Arnold, Shah, Sumedh, Wadhwa, Harsh, Chang, Susan M, Phillips, Joanna J, Aghi, Manish K, and Diaz, Aaron A
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Neurosciences ,Stem Cell Research ,Genetics ,Stem Cell Research - Nonembryonic - Non-Human ,Cancer Genomics ,Rare Diseases ,Brain Cancer ,Cancer ,Stem Cell Research - Nonembryonic - Human ,Brain Disorders ,Human Genome ,Precision Medicine ,Brain Neoplasms ,Cell Line ,Tumor ,Cell Lineage ,Cell Proliferation ,Gene Expression Profiling ,Gene Expression Regulation ,Neoplastic ,Gene Regulatory Networks ,Glioblastoma ,Humans ,Neoplastic Stem Cells ,Sequence Analysis ,RNA ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Although tumor-propagating cells can be derived from glioblastomas (GBM) of the proneural and mesenchymal subtypes, a glioma stem-like cell (GSC) of the classic subtype has not been identified. It is unclear whether mesenchymal GSCs (mGSC) and/or proneural GSCs (pGSC) alone are sufficient to generate the heterogeneity observed in GBM. We performed single-cell/single-nucleus RNA sequencing of 28 gliomas, and single-cell ATAC sequencing for 8 cases. We found that GBM GSCs reside on a single axis of variation, ranging from proneural to mesenchymal. In silico lineage tracing using both transcriptomics and genetics supports mGSCs as the progenitors of pGSCs. Dual inhibition of pGSC-enriched and mGSC-enriched growth and survival pathways provides a more complete treatment than combinations targeting one GSC phenotype alone. This study sheds light on a long-standing debate regarding lineage relationships among GSCs and presents a paradigm by which personalized combination therapies can be derived from single-cell RNA signatures, to overcome intratumor heterogeneity. SIGNIFICANCE: Tumor-propagating cells can be derived from mesenchymal and proneural glioblastomas. However, a stem cell of the classic subtype has yet to be demonstrated. We show that classic-subtype gliomas are comprised of proneural and mesenchymal cells. This study sheds light on a long-standing debate regarding lineage relationships between glioma cell types.See related commentary by Fine, p. 1650.This article is highlighted in the In This Issue feature, p. 1631.
- Published
- 2019
36. Presence of Histopathological Treatment Effects at Resection of Recurrent Glioblastoma: Incidence and Effect on Outcome
- Author
-
Ore, Cecilia L Dalle, Chandra, Ankush, Rick, Jonathan, Lau, Darryl, Shahin, Maryam, Nguyen, Alan T, McDermott, Michael, Berger, Mitchel S, and Aghi, Manish K
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Brain Disorders ,Brain Cancer ,Cancer ,Clinical Trials and Supportive Activities ,Clinical Research ,Rare Diseases ,Evaluation of treatments and therapeutic interventions ,6.4 Surgery ,6.1 Pharmaceuticals ,Adult ,Aged ,Brain Neoplasms ,Female ,Glioblastoma ,Humans ,Incidence ,Male ,Middle Aged ,Neoplasm Recurrence ,Local ,Reoperation ,Retrospective Studies ,Treatment Outcome ,Young Adult ,Bevacizumab ,Radiation necrosis ,Recurrent ,Treatment effect ,Neurosciences ,Neurology & Neurosurgery ,Clinical sciences ,Biological psychology - Abstract
BackgroundResection may be appropriate for select patients with recurrent glioblastoma. The incidence of histopathological findings related to prior treatment and their prognostic implications are incompletely characterized.ObjectiveTo quantify the incidence and survival outcomes associated with treatment effect at resection of recurrent glioblastoma (GBM).MethodsPatients who underwent resection for recurrent GBM were retrospectively reviewed, and pathology, treatment history, and survival data were collected. Treatment effect was defined as any component of treatment-related changes on pathology.ResultsIn total, 110 patients underwent 146 reoperations. Median age at first reoperation was 57.2 yr and overall survival from reoperation was 10.8 mo. Treatment effect of any kind was noted in 81 of 146 reoperations (55%). Increased treatment effect was observed closer to radiotherapy; by quartile of time from radiotherapy, the rates of treatment effect were 77.8%, 55.6%, 40.7%, and 44.4% (P = .028). Treatment effect was associated with earlier reoperation (8.9 vs 13.8 mo after radiotherapy, P = .003), and the presence of treatment effect did not impact survival from primary surgery (25.4 vs 24.3 mo, P = .084). Patients treated with bevacizumab prior to reoperation were less likely to have treatment effect (20% vs 65%, P
- Published
- 2019
37. Dissecting and rebuilding the glioblastoma microenvironment with engineered materials
- Author
-
Wolf, Kayla J, Chen, Joseph, Coombes, Jason D, Aghi, Manish K, and Kumar, Sanjay
- Subjects
Brain Disorders ,Nanotechnology ,Biotechnology ,Cancer ,Neurosciences ,Bioengineering ,Brain Cancer ,Rare Diseases ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Good Health and Well Being - Abstract
Glioblastoma (GBM) is the most aggressive and common form of primary brain cancer. Several decades of research have provided great insight into GBM progression; however, the prognosis remains poor with a median patient survival time of ~ 15 months. The tumour microenvironment (TME) of GBM plays a crucial role in mediating tumour progression and thus is being explored as a therapeutic target. Progress in the development of treatments targeting the TME is currently limited by a lack of model systems that can accurately recreate the distinct extracellular matrix composition and anatomic features of the brain, such as the blood-brain barrier and axonal tracts. Biomaterials can be applied to develop synthetic models of the GBM TME to mimic physiological and pathophysiological features of the brain, including cellular and ECM composition, mechanical properties, and topography. In this Review, we summarize key features of the GBM microenvironment and discuss different strategies for the engineering of GBM TME models, including 2D and 3D models featuring chemical and mechanical gradients, interfaces and fluid flow. Finally, we highlight the potential of engineered TME models as platforms for mechanistic discovery and drug screening as well as preclinical testing and precision medicine.
- Published
- 2019
38. Systemic therapy for brain metastases
- Author
-
Rick, Jonathan W, Shahin, Maryam, Chandra, Ankush, Dalle Ore, Cecilia, Yue, John K, Nguyen, Alan, Yagnik, Garima, Sagar, Soumya, Arfaie, Saman, and Aghi, Manish K
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Breast Cancer ,Neurosciences ,Rare Diseases ,Brain Cancer ,Lung ,Brain Disorders ,Cancer ,Brain Neoplasms ,Breast Neoplasms ,Humans ,Lung Neoplasms ,Melanoma ,Systemic therapy ,Metastasis ,Lung cancer ,Breast cancer ,Cardiorespiratory Medicine and Haematology ,Oncology & Carcinogenesis ,Cardiovascular medicine and haematology ,Oncology and carcinogenesis - Abstract
Metastases from cells outside of the central nervous system are the most common cancer found in the brain and are commonly associated with poor prognosis. Although cancer treatment is improving overall, central nervous system metastases are becoming more prevalent and require finesse to properly treat. Physicians must consider the biology of the primary tumor and the complex neurological environment that the metastasis resides in. This can be further complicated by the fact that the practice of cancer management is constantly evolving and therapy that works outside of the blood-brain barrier may not be effective inside of it. Therefore, this review seeks to update the reader on recent advancements made on the three most common sources of brain metastases: lung cancer, breast cancer, and melanoma. Each of these malignancies has been the subject of intriguing and novel avenues of therapy which are reviewed here.
- Published
- 2019
39. Immunotherapy for High-Grade Gliomas: A Clinical Update and Practical Considerations for Neurosurgeons.
- Author
-
Young, Jacob S, Dayani, Fara, Morshed, Ramin A, Okada, Hideho, and Aghi, Manish K
- Subjects
Adoptive T cell therapy ,Cerebral edema ,Checkpoint inhibitors ,Corticosteroids ,Dendritic cell vaccines ,Peptide vaccines ,Clinical Research ,Brain Cancer ,Immunization ,Rare Diseases ,Cancer ,Vaccine Related ,Neurosciences ,Orphan Drug ,Clinical Trials and Supportive Activities ,Brain Disorders ,Good Health and Well Being ,Clinical Sciences - Abstract
BackgroundThe current standard of care for patients with high-grade gliomas includes surgical resection, chemotherapy, and radiation; but even still most patients experience disease progression and succumb to their illness within a few years of diagnosis. Immunotherapy, which stimulates an anti-tumor immune response, has been revolutionary in the treatment of some hematologic and solid malignancies, generating substantial excitement for its potential for patients with glioblastoma. However, to date, the preclinical success of these approaches against high-grade glioma models has not been replicated in human clinical trials. Moreover, the complex response to these biologically active treatments can complicate management decisions, and the neurosurgical oncology community needs to be actively involved in and up to date on the use of these agents in patients with high-grade glioma. In this review, we discuss the challenges immunotherapy faces for high-grade gliomas, the completed and ongoing clinical trials for the major immunotherapies, and the nuances in management for patients being actively treated with one of these agents.MethodsWe reviewed the literature to summarize the current immunotherapy strategies for high-grade gliomas.ResultsPreclinical and clinical trials investigating dendritic cell and peptide vaccines, checkpoint inhibitors, and adoptive T cell therapy are highlighted in this review.ConclusionsAlthough immunotherapy has yet to fully fulfill its promise for patients with glioblastoma and improve patient outcomes, there is still excitement that these approaches will eventually lead to durable anti-tumor responses. As neurosurgeons, an understanding of the complex interactions between the standard of care therapies and the other medications used in the treatment arsenal for patients with high-grade brain tumors is crucial to the management of these patients.
- Published
- 2019
40. Surgical Outcomes, Complications, and Management Strategies for Foramen Magnum Meningiomas.
- Author
-
Magill, Stephen T, Shahin, Maryam N, Lucas, Calixto-Hope G, Yen, Adam J, Lee, David S, Raleigh, David R, Aghi, Manish K, Theodosopoulos, Philip V, and McDermott, Michael W
- Subjects
approach ,complications ,foramen magnum ,management ,meningioma ,outcomes ,surgical technique ,vertebral artery ,Rare Diseases ,Brain Disorders ,Cancer ,Neurosciences ,Clinical Sciences ,Neurology & Neurosurgery - Abstract
Objectives Foramen magnum meningiomas (FMM) are complex lesions because of their proximity to the brain stem and posterior cerebrovasculature. The objective of this study is to report surgical outcomes and complications after resection of FMM. Methods A retrospective chart review was conducted on patients with FMM from 1998 to 2015. Univariate logistic regression and recursive partitioning analysis were used to identify risk factors associated with complications and extent of resection (EOR). Results We identified 28 patients with FMM. Median follow-up was 5.9 years. Tumors were World Health Organization grade I (92.9%) or grade II (7.1%). The vertebral artery was completely encased (25%), partially encased (11%), or not encased (64%). Median size was 11.9 cm 3 . EOR was gross total (39%) and subtotal (61%). The observed recurrence rate was 4% ( n = 1). There were 38 complications in 12 patients (43%), and 6 patients (21%) had complications requiring additional surgery. Complications included cerebrospinal fluid leak/hydrocephalus ( n = 7, 25%), weakness ( n = 4, 14%), numbness ( n = 4, 14%), and cranial nerve deficits: IX, X ( n = 4, 14%), XI ( n = 2, 7%), XII ( n = 5, 18%). Medical complications included pneumonia ( n = 1, 4%) and meningitis ( n = 1, 4%). Tumor volume greater than 14 cm 3 (odds ratio [OR] = 21.7, p = 0.0010), any vertebral artery encasement (OR 6.1, p = 0.0386), and subtotal resection (OR 6.4, p = 0.0398) were significantly associated with complications. Tumor volume greater than 14 cm 3 was also significantly associated with subtotal resection (OR 8.3, p = 0.0201). Conclusions Resection of FMM carries perioperative morbidity that increases with larger tumor size. Despite the morbidity, long-term recurrence-free survival is achievable with maximal safe resection and adjuvant radiation.
- Published
- 2019
41. Surgery for Control of Brain Metastases After Previous Checkpoint Inhibitor Immunotherapy
- Author
-
Morshed, Ramin A., Chung, Jason E., Cummins, Daniel D., Sudhakar, Vivek, Young, Jacob S., Daras, Mariza, Hervey-Jumper, Shawn L., Theodosopoulos, Philip V., and Aghi, Manish K.
- Published
- 2022
- Full Text
- View/download PDF
42. Lateral orientation of Rathke cleft cysts may be associated with high rates of recurrence after surgery
- Author
-
Carrete, Luis R. and Aghi, Manish K.
- Published
- 2022
- Full Text
- View/download PDF
43. Petrous Face Meningiomas: Classification, Clinical Syndromes, and Surgical Outcomes
- Author
-
Magill, Stephen T, Rick, Jonathan W, Chen, William C, Haase, David A, Raleigh, David R, Aghi, Manish K, Theodosopoulos, Philip V, and McDermott, Michael W
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Cancer ,Neurosciences ,Brain Disorders ,Rare Diseases ,Adult ,Aged ,Aged ,80 and over ,Cranial Nerves ,Facial Neoplasms ,Female ,Follow-Up Studies ,Humans ,Male ,Meningeal Neoplasms ,Meningioma ,Middle Aged ,Petrous Bone ,Retrospective Studies ,Treatment Outcome ,Cerebellopontine angle ,Complications ,Outcomes ,Petrous ,Petrous face ,Clinical sciences - Abstract
BackgroundPetrous face meningiomas (PFMs) are challenging tumors because of their proximity to the cranial nerves, brainstem, and critical vasculature. The objective of this study is to present surgical outcomes and support an anatomic classification for PFM based on clinical presentation.MethodsA retrospective chart review was performed, and 51 PFMs were identified. Tumors were classified by location along the petrous face into anterior, middle, and posterior. Presentation and outcomes were analyzed with logistic regression.ResultsThe median follow-up was 31.6 months. Tumors were World Health Organization grade I (n = 50), with 1 World Health Organization grade II tumor. Location was anterior (22%), middle (14%), posterior (53%), and overlapping (12%). Median tumor diameter was 3.0 cm (range, 0.8-6.2 cm). Anterior location was associated with facial pain/numbness on presentation (P < 0.0001), middle location with hearing loss/vestibular dysfunction (P = 0.0035), and posterior with hydrocephalus (P = 0.0190), headache (P = 0.0039), and vertigo (P = 0.0265). Extent of resection was gross total (63%), near total (14%), and subtotal (25%). The observed radiographic recurrence rate was 15%. Mean progression-free survival after diagnosis was 9.1 years with 2-year, 5-year, and 10-year progression-free survival of 91.8%, 78.6%, and 62.9%, respectively. The complication rate was 27%. Age, location, and approach were not associated with complications.ConclusionsPFMs present with distinct clinical syndromes based on their location along the petrous face: anterior with trigeminal symptoms, middle with auditory/vestibular symptoms, and posterior with symptoms of mass effect/hydrocephalous. Surgical resection is associated with excellent long-term survival and a low rate of recurrence, which can be managed with radiotherapy.
- Published
- 2018
44. Disparities in health care determine prognosis in newly diagnosed glioblastoma.
- Author
-
Chandra, Ankush, Rick, Jonathan W, Dalle Ore, Cecilia, Lau, Darryl, Nguyen, Alan T, Carrera, Diego, Bonte, Alexander, Molinaro, Annette M, Theodosopoulos, Philip V, McDermott, Michael W, Berger, Mitchel S, and Aghi, Manish K
- Subjects
Humans ,Glioblastoma ,Brain Neoplasms ,Prognosis ,Retrospective Studies ,Adolescent ,Adult ,Aged ,Aged ,80 and over ,Middle Aged ,Medically Uninsured ,Health Services Accessibility ,Female ,Male ,Healthcare Disparities ,Young Adult ,CCI = Charlson Comorbidity Index ,GBM = glioblastoma ,HR = hazard ratio ,PCP ,PCP = primary care physician ,SEER = Surveillance ,Epidemiology ,and End Results ,TMZ = temozolomide ,XRT = radiation therapy ,glioblastoma ,health care ,insurance ,primary care physician ,prognosis ,socioeconomic ,Prevention ,Brain Disorders ,Brain Cancer ,Neurosciences ,Cancer ,Health Services ,Rare Diseases ,Clinical Research ,Good Health and Well Being ,Clinical Sciences ,Neurology & Neurosurgery - Abstract
OBJECTIVE Glioblastoma (GBM) is an aggressive brain malignancy with a short overall patient survival, yet there remains significant heterogeneity in outcomes. Although access to health care has previously been linked to impact on prognosis in several malignancies, this question remains incompletely answered in GBM. METHODS This study was a retrospective analysis of 354 newly diagnosed patients with GBM who underwent first resection at the authors' institution (2007-2015). RESULTS Of the 354 patients (median age 61 years, and 37.6% were females), 32 (9.0%) had no insurance, whereas 322 (91.0%) had insurance, of whom 131 (40.7%) had Medicare, 45 (14%) had Medicaid, and 146 (45.3%) had private insurance. On average, insured patients survived almost 2-fold longer (p < 0.0001) than those who were uninsured, whereas differences between specific insurance types did not influence survival. The adjusted hazard ratio (HR) for death was higher in uninsured patients (HR 2.27 [95% CI 1.49-3.33], p = 0.0003). Age, mean household income, tumor size at diagnosis, and extent of resection did not differ between insured and uninsured patients, but there was a disparity in primary care physician (PCP) status-none of the uninsured patients had PCPs, whereas 72% of insured patients had PCPs. Postoperative adjuvant treatment rates with temozolomide (TMZ) and radiation therapy (XRT) were significantly less in uninsured (TMZ in 56.3%, XRT in 56.3%) than in insured (TMZ in 75.2%, XRT in 79.2%; p = 0.02 and p = 0.003) patients. Insured patients receiving both agents had better prognosis than uninsured patients receiving the same treatment (9.1 vs 16.34 months; p = 0.025), suggesting that the survival effect in insured patients could only partly be explained by higher treatment rates. Moreover, having a PCP increased survival among the insured cohort (10.7 vs 16.1 months, HR 1.65 [95% CI 1.27-2.15]; p = 0.0001), which could be explained by significant differences in tumor diameter at initial diagnosis between patients with and without PCPs (4.3 vs 4.8 cm, p = 0.003), and a higher rate of clinical trial enrollment, suggesting a critical role of PCPs for a timelier diagnosis of GBM and proactive cancer care management. CONCLUSIONS Access to health care is a strong determinant of prognosis in newly diagnosed patients with GBM. Any type of insurance coverage and having a PCP improved prognosis in this patient cohort. Higher rates of treatment with TMZ plus XRT, clinical trial enrollment, fewer comorbidities, and early diagnosis may explain survival disparities. Lack of health insurance or a PCP are major challenges within the health care system, which, if improved upon, could favorably impact the prognosis of patients with GBM.
- Published
- 2018
45. A Glial Signature and Wnt7 Signaling Regulate Glioma-Vascular Interactions and Tumor Microenvironment
- Author
-
Griveau, Amelie, Seano, Giorgio, Shelton, Samuel J, Kupp, Robert, Jahangiri, Arman, Obernier, Kirsten, Krishnan, Shanmugarajan, Lindberg, Olle R, Yuen, Tracy J, Tien, An-Chi, Sabo, Jennifer K, Wang, Nancy, Chen, Ivy, Kloepper, Jonas, Larrouquere, Louis, Ghosh, Mitrajit, Tirosh, Itay, Huillard, Emmanuelle, Alvarez-Buylla, Arturo, Oldham, Michael C, Persson, Anders I, Weiss, William A, Batchelor, Tracy T, Stemmer-Rachamimov, Anat, Suvà, Mario L, Phillips, Joanna J, Aghi, Manish K, Mehta, Shwetal, Jain, Rakesh K, and Rowitch, David H
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Neurosciences ,Rare Diseases ,Cancer ,Brain Cancer ,Brain Disorders ,2.1 Biological and endogenous factors ,Aetiology ,Animals ,Bevacizumab ,Blood-Brain Barrier ,Brain Neoplasms ,Cell Line ,Tumor ,Gene Expression Regulation ,Neoplastic ,Glioma ,Humans ,Mice ,Neoplasm Transplantation ,Oligodendrocyte Transcription Factor 2 ,Oligodendroglia ,Temozolomide ,Tumor Cells ,Cultured ,Tumor Microenvironment ,Wnt Proteins ,Wnt Signaling Pathway ,Olig2 ,Wnt ,angiogenesis ,astrocyte ,blood-brain barrier ,glioma ,invasiveness ,oligodendrocyte precursor ,p53 ,vessel co-option ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Gliomas comprise heterogeneous malignant glial and stromal cells. While blood vessel co-option is a potential mechanism to escape anti-angiogenic therapy, the relevance of glial phenotype in this process is unclear. We show that Olig2+ oligodendrocyte precursor-like glioma cells invade by single-cell vessel co-option and preserve the blood-brain barrier (BBB). Conversely, Olig2-negative glioma cells form dense perivascular collections and promote angiogenesis and BBB breakdown, leading to innate immune cell activation. Experimentally, Olig2 promotes Wnt7b expression, a finding that correlates in human glioma profiling. Targeted Wnt7a/7b deletion or pharmacologic Wnt inhibition blocks Olig2+ glioma single-cell vessel co-option and enhances responses to temozolomide. Finally, Olig2 and Wnt7 become upregulated after anti-VEGF treatment in preclinical models and patients. Thus, glial-encoded pathways regulate distinct glioma-vascular microenvironmental interactions.
- Published
- 2018
46. Genomic analysis of the origins and evolution of multicentric diffuse lower-grade gliomas
- Author
-
Hayes, Josie, Yu, Yao, Jalbert, Llewellyn E, Mazor, Tali, Jones, Lindsey E, Wood, Matthew D, Walsh, Kyle M, Bengtsson, Henrik, Hong, Chibo, Oberndorfer, Stefan, Roetzer, Thomas, Smirnov, Ivan V, Clarke, Jennifer L, Aghi, Manish K, Chang, Susan M, Nelson, Sarah J, Woehrer, Adelheid, Phillips, Joanna J, Solomon, David A, and Costello, Joseph F
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Brain Disorders ,Orphan Drug ,Brain Cancer ,Genetics ,Biotechnology ,Cancer Genomics ,Cancer ,Neurosciences ,Human Genome ,Rare Diseases ,Clinical Research ,Adult ,Biomarkers ,Tumor ,Brain Neoplasms ,Clonal Evolution ,Female ,Genomics ,Glioma ,Humans ,Male ,Middle Aged ,Mutation ,Neoplasm Grading ,Phylogeny ,Young Adult ,astrocytoma ,bilateral ,IDH1 ,multicentric ,oligodendroglioma ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
BackgroundRare multicentric lower-grade gliomas (LGGs) represent a unique opportunity to study the heterogeneity among distinct tumor foci in a single patient and to infer their origins and parallel patterns of evolution.MethodsIn this study, we integrate clinical features, histology, and immunohistochemistry for 4 patients with multicentric LGG, arising both synchronously and metachronously. For 3 patients we analyze the phylogeny of the lesions using exome sequencing, including one case with a total of 8 samples from the 2 lesions.ResultsOne patient was diagnosed with multicentric isocitrate dehydrogenase 1 (IDH1) mutated diffuse astrocytomas harboring distinct IDH1 mutations, R132H and R132C; the latter mutation has been associated with Li-Fraumeni syndrome, which was subsequently confirmed in the patient's germline DNA and shown in additional cases with The Cancer Genome Atlas data. In another patient, phylogenetic analysis of synchronously arising grade II and grade III diffuse astrocytomas demonstrated a single shared mutation, IDH1 R132H, and revealed convergent evolution via non-overlapping mutations in ATRX and TP53. In 2 cases, there was divergent evolution of IDH1-mutated and 1p/19q-codeleted oligodendroglioma and IDH1-mutated and 1p/19q-intact diffuse astrocytoma, occurring synchronously in one case and metachronously in a second.ConclusionsEach tumor in multicentric LGG cases may arise independently or may diverge very early in their development, presenting as genetically and histologically distinct tumors. Comprehensive sampling of these lesions can therefore significantly alter diagnosis and management. Additionally, somatic IDH1 R132C mutation in either multicentric or solitary LGG identifies unsuspected germline TP53 mutation, validating the limited number of published cases.
- Published
- 2018
47. CD97 is associated with mitogenic pathway activation, metabolic reprogramming, and immune microenvironment changes in glioblastoma
- Author
-
Safaee, Michael M., Wang, Elaina J., Jain, Saket, Chen, Jia-Shu, Gill, Sabraj, Zheng, Allison C., Garcia, Joseph H., Beniwal, Angad S., Tran, Y., Nguyen, Alan T., Trieu, Melissa, Leung, Kevin, Wells, Jim, Maclean, James M., Wycoff, Keith, and Aghi, Manish K.
- Published
- 2022
- Full Text
- View/download PDF
48. Acute hyponatremia post craniotomy resulting in a unilateral fixed and dilated pupil: A case study on diagnosis and management
- Author
-
Caldwell, David J., primary, Scheer, Justin K., additional, Umbach, Gray, additional, and Aghi, Manish K., additional
- Published
- 2024
- Full Text
- View/download PDF
49. Geographic landscape of foreign medical graduates in US neurosurgery training programs from 2007 to 2017
- Author
-
Chandra, Ankush, Brandel, Michael G., Wadhwa, Harsh, Pereira, Matheus P., Lewis, Cole T., Haddad, Yazeed W., Lu, Victor M., Almeida, Neil D., Nuru, Mohammed O., Esquenazi, Yoshua, and Aghi, Manish K.
- Published
- 2021
- Full Text
- View/download PDF
50. Plurihormonal PIT-1–Positive Pituitary Adenomas: A Systematic Review and Single-Center Series
- Author
-
Andrews, John P., Joshi, Rushikesh S., Pereira, Matheus P., Oh, Taemin, Haddad, Alexander F., Pereira, Kaitlyn M., Osorio, Robert C., Donohue, Kevin C., Peeran, Zain, Sudhir, Sweta, Jain, Saket, Beniwal, Angad, Chopra, Ashley S., Sandhu, Narpal S., Tihan, Tarik, Blevins, Lewis, and Aghi, Manish K.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.