1
|
Pedder JH, Sonabend AM, Cearns MD, Michael BD, Zakaria R, Heimberger AB, Jenkinson MD, Dickens D. Crossing the blood-brain barrier: emerging therapeutic strategies for neurological disease. Lancet Neurol 2025:S1474-4422(24)00476-9. [PMID: 39862873 DOI: 10.1016/s1474-4422(24)00476-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/20/2024] [Accepted: 11/12/2024] [Indexed: 01/27/2025]
Abstract
The blood-brain barrier is a physiological barrier that can prevent both small and complex drugs from reaching the brain to exert a pharmacological effect. For treatment of neurological diseases, drug concentrations at the target site are a fundamental parameter for therapeutic effect; thus, the blood-brain barrier is a major obstacle to overcome. Novel strategies have been developed to circumvent the blood-brain barrier, including CSF delivery, intracranial delivery, ultrasound-based methods, membrane transporters, receptor-mediated transcytosis, and nanotherapeutics. These approaches each have their advantages and disadvantages. CSF delivery and intracranial delivery are direct but invasive techniques that have not yet shown efficacy in clinical trials, although development of novel delivery devices might improve these approaches. Ultrasound-based disruption has shown some efficacy in clinical trials, but it can require invasive procedures. Approaches using membrane transporters and receptor-mediated transcytosis are less invasive than are other techniques, but they can have off-target effects. Nanotherapeutics have shown promise, but these strategies are in early stages of development. Advancements in drug delivery across the blood-brain barrier will require appropriately designed and powered clinical studies, with a focus on the timing of treatment, demographic and genetic considerations, head-to-head comparison with other treatment strategies (rather than a placebo), and relevant primary and secondary outcome measures.
Collapse
Affiliation(s)
- Josephine H Pedder
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Adam M Sonabend
- Department of Neurological Surgery, Malnati Brain Tumor Institute of the Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michael D Cearns
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - Benedict D Michael
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK; Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - Rasheed Zakaria
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - Amy B Heimberger
- Department of Neurological Surgery, Malnati Brain Tumor Institute of the Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michael D Jenkinson
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - David Dickens
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
2
|
Pinheiro Lopes B, O’Neill L, Bourke P, Boehm D. Enhanced Cytotoxic Effects of Cold Plasma Deposition of Topotecan: A Novel Approach for Local Cancer Drug Delivery to Glioblastoma Cells. Cancers (Basel) 2025; 17:201. [PMID: 39857983 PMCID: PMC11764045 DOI: 10.3390/cancers17020201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Despite the numerous advances in glioblastoma multiforme (GBM) treatment, GBM remains as the most malignant and aggressive form of brain cancer, characterized by a very poor outcome, highlighting the ongoing need for the development of new therapeutic strategies. A novel intervention using plasma-assisted local delivery of oncology drugs was developed to mediate the drug delivery, which might improve drug uptake and/or chemotherapeutic action. Topotecan (TPT), a water-soluble topoisomerase I inhibitor with major cytotoxic effects during the S-phase of the cell cycle, was selected as the candidate drug because despite its potent antitumor activity, the systemic administration to the brain is limited due to low crossing of the blood-brain barrier. For these reasons, TPT may be repurposed for local combined therapies. METHODS We aimed to explore options for the local treatment of GBM where systematic delivery is challenging, using a combination between plasma-based technologies and TPT on a human brain cancer cell line (U-251mg). RESULTS The evaluation of direct TPT plasma deposition using a helium plasma jet (J-Plasma, Apyx Medical) with a nebulizer onto U-251mg cells grown in 2D or 3D culture showed a reduction in the metabolic activity and cell mass and decreased long-term survival, indicating synergistic effects between the drug and the plasma treatment. The plasma-assisted approach was confirmed using temozolomide (TMZ) as a standard drug for glioblastoma treatment, as well as with two skin cancer cell lines. CONCLUSIONS These results revealed a pathway for new combinations and approaches to local drug application for a range of cancers.
Collapse
Affiliation(s)
- Beatriz Pinheiro Lopes
- School of Chemical and Bioprocess Engineering, University College Dublin, D04 V1W8 Dublin, Ireland
- Sustainability and Health Research Hub and School of Food Science and Environmental Health, Technological University Dublin, D07 H6K8 Dublin, Ireland
| | - Liam O’Neill
- TheraDep Ltd., QUESTUM Innovation Centre, E91 V329 Clonmel, Ireland
| | - Paula Bourke
- School of Chemical and Bioprocess Engineering, University College Dublin, D04 V1W8 Dublin, Ireland
- Plasma Research Group, School of Biosystems and Food Engineering, University College Dublin, D04 V1W8 Dublin, Ireland
- Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Daniela Boehm
- School of Chemical and Bioprocess Engineering, University College Dublin, D04 V1W8 Dublin, Ireland
- Sustainability and Health Research Hub and School of Food Science and Environmental Health, Technological University Dublin, D07 H6K8 Dublin, Ireland
| |
Collapse
|
3
|
Narsinh KH, Kumar K, Bankiewicz K, Martin AJ, Berger M, Clarke J, Taylor J, Bush NAO, Molinaro AM, Aghi M, Butowski N. A phase I study of convection-enhanced delivery (CED) of liposomal-irinotecan using real-time magnetic resonance imaging in patients with recurrent high-grade glioma. J Neurooncol 2025:10.1007/s11060-024-04904-y. [PMID: 39760796 DOI: 10.1007/s11060-024-04904-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Irinotecan demonstrates anti-tumor efficacy in preclinical glioma models but clinical results are modest due to drug delivery limitations. Convection enhanced delivery (CED) improves drug delivery by increasing intratumoral drug concentration. Real-time magnetic resonance imaging of infusate delivery during CED may optimize tumor coverage. This phase 1 trial examines the safety and tolerability of liposomal irinotecan and gadolinium delivered via CED using real-time MRI guidance in recurrent high-grade glioma patients. METHODS Initially, a 3 + 3 dose-escalating, single dose trial was planned with 4 cohorts based on a fixed drug dose and volume. After 9 patients, a protocol amendment allowed for variable volume and dose of the study agent based on tumor size. The amended design specified 'personalized' drug volume but fixed concentration of 20 mg/mL of liposomal irinotecan in the first cohort escalating to 40 mg/mL in the second cohort. RESULTS Eighteen patients with recurrent WHO grade 3 or 4 gliomas (diameter 1-4 cm) were treated. Based on the tumor volume, the total dose of liposomal irinotecan was 20-680 mg in a total volume of 2-17 ml. Technical challenges were overcome by real-time MRI guidance and protocol amendment. The only dose-limiting toxicity (DLT) was a grade 3 stroke. Safety and survival information is presented. CONCLUSIONS CED of liposomal irinotecan using real-time MRI in patients with recurrent high-grade glioma is feasible. Image-guidance allowed for improved placement of CED cannulas and optimal tumor coverage. Our results warrant further study with repeat CED dosing.
Collapse
Affiliation(s)
- Kazim H Narsinh
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
- Department of Radiology & Biomedical Imaging, San Francisco, CA, USA.
| | - Karishma Kumar
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Krystof Bankiewicz
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, Ohio State University, Columbus, OH, USA
| | - Alastair J Martin
- Department of Radiology & Biomedical Imaging, San Francisco, CA, USA
| | - Mitchell Berger
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer Clarke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Jennie Taylor
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Nancy Ann Oberheim Bush
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Manish Aghi
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
4
|
Giantini-Larsen AM, Pandey A, Garton ALA, Rampichini M, Winston G, Goldberg JL, Magge R, Stieg PE, Souweidane MM, Ramakrishna R. Therapeutic manipulation and bypass of the blood-brain barrier: powerful tools in glioma treatment. Neurooncol Adv 2025; 7:vdae201. [PMID: 39877748 PMCID: PMC11773386 DOI: 10.1093/noajnl/vdae201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025] Open
Abstract
The blood-brain barrier (BBB) remains an obstacle for delivery of chemotherapeutic agents to gliomas. High grade and recurrent gliomas continue to portend a poor prognosis. Multiple methods of bypassing or manipulating the BBB have been explored, including hyperosmolar therapy, convection-enhanced delivery (CED), laser-guided interstitial thermal therapy (LITT), and Magnetic Resonance Guided Focused Ultrasound (MRgFUS) to enhance delivery of chemotherapeutic agents to glial neoplasms. Here, we review these techniques, currently ongoing clinical trials to disrupt or bypass the BBB in gliomas, and the results of completed trials.
Collapse
Affiliation(s)
- Alexandra M Giantini-Larsen
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Abhinav Pandey
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Andrew L A Garton
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Margherita Rampichini
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Graham Winston
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Jacob L Goldberg
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Rajiv Magge
- Department of Neurology, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Philip E Stieg
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Mark M Souweidane
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Rohan Ramakrishna
- Corresponding Author: Rohan Ramakrishna, MD, Chief, Neurological Surgery, New York Presbyterian Brooklyn Methodist Hospital, Weill Cornell Medical Center, 525 East 68 Street, New York, NY 10065, USA ()
| |
Collapse
|
5
|
Kinslow CJ, Mehta MP. Future Directions in the Treatment of Low-Grade Gliomas. Cancer J 2025; 31:e0759. [PMID: 39841425 DOI: 10.1097/ppo.0000000000000759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
ABSTRACT There is major interest in deintensifying therapy for isocitrate dehydrogenase-mutant low-grade gliomas, including with single-agent cytostatic isocitrate dehydrogenase inhibitors. These efforts need head-to-head comparisons with proven modalities, such as chemoradiotherapy. Ongoing clinical trials now group tumors by intrinsic molecular subtype, rather than classic clinical risk factors. Advances in imaging, surgery, and radiotherapy have improved outcomes in low-grade gliomas. Emerging biomarkers, targeted therapies, immunotherapy, radionuclides, and novel medical devices are a promising frontier for future treatment. Diverse representation in glioma research and clinical trials will help to ensure that advancements in care are realized by all groups.
Collapse
Affiliation(s)
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| |
Collapse
|
6
|
Gordon CR, Perez CF. Review of Functional Cranioplasty and Implantable Neurotechnology. J Craniofac Surg 2024:00001665-990000000-02303. [PMID: 39718390 DOI: 10.1097/scs.0000000000011028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/06/2024] [Indexed: 12/25/2024] Open
Abstract
Cranioplasty for secondary reconstruction of cranial defects has historically focused on simply replacing the missing cranial bone to restore cerebral protection and fluid dynamics, but recent innovations have led to the development of customized cranial implants that address both bone and soft tissue deficits while avoiding postoperative complications such as temporal hollowing. In addition, customized cranial implants have incorporated implantable neurotechnology like ventriculostomy shunts, intracranial pressure monitoring devices, and medicine delivery systems within low-profile designs to convert previously "basic" implants into "smart" implants for added functionality. These "smart" implants aim to reduce complications and improve patient outcomes by leveraging the cranial space to house advanced technologies, providing benefits such as real-time biosensing, and treatment of chronic neurological conditions. This review outlines the progression of cranioplasty from basic bone replacement to functional implants with embedded neurotechnologies, highlighting the multidisciplinary approaches that enhance surgical outcomes and patient quality of life.
Collapse
Affiliation(s)
- Chad R Gordon
- Section of Neuroplastic and Reconstructive Surgery, Departments of Plastic and Reconstructive Surgery and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Colleen F Perez
- Department of Otolaryngology-Head and Neck Surgery, Neuroplastic and Reconstructive Surgery, Naval Medical Center Portsmouth, Portsmouth, VA
| |
Collapse
|
7
|
Semreen AM, Alsoud LO, Semreen MH, Ahmed M, Al-Hroub HM, El-Awady R, Ramadan WS, Abuhelwa A, Bustanji Y, Soares NC, Alzoubi KH. Multi-omics analysis revealed significant metabolic changes in brain cancer cells treated with paclitaxel and/or topotecan. Heliyon 2024; 10:e39420. [PMID: 39524752 PMCID: PMC11550653 DOI: 10.1016/j.heliyon.2024.e39420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Glioblastoma (GBM) stands as the most common primary malignant brain tumor. Despite the best standard therapies, GBM survivors have a brief survival time, about 24 months on average. The treatment is troublesome because the cancer cells may not respond well to specific therapies as they grow within an extensive network of blood vessels. Our study aims to evaluate the impact of paclitaxel 5.3 μg/mL and topotecan 0.26 μM solely and in pairwise combination on the resultant metabolic and proteomic signatures of the U87 cell line while using the precise ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF) analytical technology. The U87 cells wear treated with DMSO, paclitaxel 5.3 μM, topotecan 0.26 μM, and their combinations. Using One-way ANOVA, we observed 14 significantly altered metabolites compared to those cells treated with DMSO. For combination treatment (paclitaxel and topotecan), 11 metabolites were significantly dysregulated. Sparse partial least squares-discriminant analysis (sPLS-DA) revealed minimal overlap, highlighting distinctions among the four groups. While for proteomics, a total of 79 proteins were significantly dysregulated among the groups. These findings can aid in identifying new biomarkers associated with the utilized drugs and creating a map for targeted therapy. EIF3F, GNB2L1, HINT2, and RPA3 were shown to be significantly upregulated in the combination group relative to the control. Moreover, ribosome, apoptosis, HIF-1 signaling, arginine and proline, glutathione, purine metabolism, apelin signaling pathway, and glycolysis were significantly altered in the combination group. Overall, this study underscores the effectiveness of multi-omics approaches in revealing the molecular mechanisms driving chemotherapy responses in cancer cells. Additionally, this work generates a comprehensive list of molecular alterations that can serve as a foundation for further investigations and inform personalized healthcare strategies to enhance patient outcomes.
Collapse
Affiliation(s)
- Ahlam M. Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Leen Oyoun Alsoud
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Mohammad H. Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Munazza Ahmed
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hamza M. Al-Hroub
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Raafat El-Awady
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Wafaa S. Ramadan
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Ahmad Abuhelwa
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Yasser Bustanji
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman, 11942, Jordan
- Department of Basic and Clinical Pharmacology, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Nelson C. Soares
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Center for Applied and Translational Genomics (CATG), Mohamed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai Health, Dubai, United Arab Emirates
- College of Medicine, Mohamed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai Health, Dubai, United Arab Emirates
| | - Karem H. Alzoubi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| |
Collapse
|
8
|
Wang H, Argenziano MG, Yoon H, Boyett D, Save A, Petridis P, Savage W, Jackson P, Hawkins-Daarud A, Tran N, Hu L, Singleton KW, Paulson L, Dalahmah OA, Bruce JN, Grinband J, Swanson KR, Canoll P, Li J. Biologically informed deep neural networks provide quantitative assessment of intratumoral heterogeneity in post treatment glioblastoma. NPJ Digit Med 2024; 7:292. [PMID: 39427044 PMCID: PMC11490546 DOI: 10.1038/s41746-024-01277-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024] Open
Abstract
Intratumoral heterogeneity poses a significant challenge to the diagnosis and treatment of recurrent glioblastoma. This study addresses the need for non-invasive approaches to map heterogeneous landscape of histopathological alterations throughout the entire lesion for each patient. We developed BioNet, a biologically-informed neural network, to predict regional distributions of two primary tissue-specific gene modules: proliferating tumor (Pro) and reactive/inflammatory cells (Inf). BioNet significantly outperforms existing methods (p < 2e-26). In cross-validation, BioNet achieved AUCs of 0.80 (Pro) and 0.81 (Inf), with accuracies of 80% and 75%, respectively. In blind tests, BioNet achieved AUCs of 0.80 (Pro) and 0.76 (Inf), with accuracies of 81% and 74%. Competing methods had AUCs lower or around 0.6 and accuracies lower or around 70%. BioNet's voxel-level prediction maps reveal intratumoral heterogeneity, potentially improving biopsy targeting and treatment evaluation. This non-invasive approach facilitates regular monitoring and timely therapeutic adjustments, highlighting the role of ML in precision medicine.
Collapse
Affiliation(s)
- Hairong Wang
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael G Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Hyunsoo Yoon
- Department of Industrial Engineering, Yonsei University, Seoul, South Korea
| | - Deborah Boyett
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Akshay Save
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Petros Petridis
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Psychiatry, New York University, New York, NY, USA
| | - William Savage
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Pamela Jackson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Andrea Hawkins-Daarud
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Nhan Tran
- Department of Cancer Biology, Mayo Clinic, Phoenix, AZ, USA
| | - Leland Hu
- Department of Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Kyle W Singleton
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Lisa Paulson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Osama Al Dalahmah
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Jack Grinband
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kristin R Swanson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jing Li
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
9
|
Yonk MG, Lim MA, Thompson CM, Tora MS, Lakhina Y, Du Y, Hoang KB, Molinaro AM, Boulis NM, Hassaneen W, Lei K. Improving glioma drug delivery: A multifaceted approach for glioma drug development. Pharmacol Res 2024; 208:107390. [PMID: 39233056 PMCID: PMC11440560 DOI: 10.1016/j.phrs.2024.107390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/16/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Glioma is one of the most common central nervous system (CNS) cancers that can be found within the brain and the spinal cord. One of the pressing issues plaguing the development of therapeutics for glioma originates from the selective and semipermeable CNS membranes: the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB). It is difficult to bypass these membranes and target the desired cancerous tissue because the purpose of the BBB and BSCB is to filter toxins and foreign material from invading CNS spaces. There are currently four varieties of Food and Drug Administration (FDA)-approved drug treatment for glioma; yet these therapies have limitations including, but not limited to, relatively low transmission through the BBB/BSCB, despite pharmacokinetic characteristics that allow them to cross the barriers. Steps must be taken to improve the development of novel and repurposed glioma treatments through the consideration of pharmacological profiles and innovative drug delivery techniques. This review addresses current FDA-approved glioma treatments' gaps, shortcomings, and challenges. We then outline how incorporating computational BBB/BSCB models and innovative drug delivery mechanisms will help motivate clinical advancements in glioma drug delivery. Ultimately, considering these attributes will improve the process of novel and repurposed drug development in glioma and the efficacy of glioma treatment.
Collapse
Affiliation(s)
- Marybeth G Yonk
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA; College of Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Megan A Lim
- Carle Illinois College of Medicine, University of Illinois Urbana Champaign, Champaign, IL, USA; Department of Neurosurgery, Carle Foundation Hospital, Urbana, IL, USA
| | - Charee M Thompson
- Carle Illinois College of Medicine, University of Illinois Urbana Champaign, Champaign, IL, USA; College of Liberal Arts & Sciences, University of Illinois Urbana Champaign, Champaign, IL, USA
| | - Muhibullah S Tora
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Yuliya Lakhina
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Kimberly B Hoang
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Nicholas M Boulis
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Wael Hassaneen
- Carle Illinois College of Medicine, University of Illinois Urbana Champaign, Champaign, IL, USA; Department of Neurosurgery, Carle Foundation Hospital, Urbana, IL, USA.
| | - Kecheng Lei
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
10
|
Banu MA, Dovas A, Argenziano MG, Zhao W, Sperring CP, Cuervo Grajal H, Liu Z, Higgins DM, Amini M, Pereira B, Ye LF, Mahajan A, Humala N, Furnari JL, Upadhyayula PS, Zandkarimi F, Nguyen TT, Teasley D, Wu PB, Hai L, Karan C, Dowdy T, Razavilar A, Siegelin MD, Kitajewski J, Larion M, Bruce JN, Stockwell BR, Sims PA, Canoll P. A cell state-specific metabolic vulnerability to GPX4-dependent ferroptosis in glioblastoma. EMBO J 2024; 43:4492-4521. [PMID: 39192032 PMCID: PMC11480389 DOI: 10.1038/s44318-024-00176-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/12/2024] [Accepted: 07/01/2024] [Indexed: 08/29/2024] Open
Abstract
Glioma cells hijack developmental programs to control cell state. Here, we uncover a glioma cell state-specific metabolic liability that can be therapeutically targeted. To model cell conditions at brain tumor inception, we generated genetically engineered murine gliomas, with deletion of p53 alone (p53) or with constitutively active Notch signaling (N1IC), a pathway critical in controlling astrocyte differentiation during brain development. N1IC tumors harbored quiescent astrocyte-like transformed cell populations while p53 tumors were predominantly comprised of proliferating progenitor-like cell states. Further, N1IC transformed cells exhibited increased mitochondrial lipid peroxidation, high ROS production and depletion of reduced glutathione. This altered mitochondrial phenotype rendered the astrocyte-like, quiescent populations more sensitive to pharmacologic or genetic inhibition of the lipid hydroperoxidase GPX4 and induction of ferroptosis. Treatment of patient-derived early-passage cell lines and glioma slice cultures generated from surgical samples with a GPX4 inhibitor induced selective depletion of quiescent astrocyte-like glioma cell populations with similar metabolic profiles. Collectively, these findings reveal a specific therapeutic vulnerability to ferroptosis linked to mitochondrial redox imbalance in a subpopulation of quiescent astrocyte-like glioma cells resistant to standard forms of treatment.
Collapse
Affiliation(s)
- Matei A Banu
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Athanassios Dovas
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael G Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Wenting Zhao
- Department of System Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Colin P Sperring
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Zhouzerui Liu
- Department of System Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Dominique Mo Higgins
- Department of Neurological Surgery, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Misha Amini
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Brianna Pereira
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ling F Ye
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Aayushi Mahajan
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Nelson Humala
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Julia L Furnari
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Pavan S Upadhyayula
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Fereshteh Zandkarimi
- Department of Biological Sciences, Department of Chemistry and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Trang Tt Nguyen
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Damian Teasley
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter B Wu
- Department of Neurological Surgery, UCLA Geffen School of Medicine, Los Angeles, CA, USA
| | - Li Hai
- Sulzberger Columbia Genome Center, Columbia University, New York, NY, USA
| | - Charles Karan
- Sulzberger Columbia Genome Center, Columbia University, New York, NY, USA
| | | | - Aida Razavilar
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jan Kitajewski
- University of Illinois Cancer Center, Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, USA
| | | | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Brent R Stockwell
- Department of Biological Sciences, Department of Chemistry and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Peter A Sims
- Department of System Biology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Peter Canoll
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
11
|
Dadario NB, Boyett DM, Teasley DE, Chabot PJ, Winans NJ, Argenziano MG, Sperring CP, Canoll P, Bruce JN. Unveiling the Inflammatory Landscape of Recurrent Glioblastoma through Histological-Based Assessments. Cancers (Basel) 2024; 16:3283. [PMID: 39409905 PMCID: PMC11476027 DOI: 10.3390/cancers16193283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
The glioblastoma (GBM) tumor microenvironment consists of a heterogeneous mixture of neoplastic and non-neoplastic cells, including immune cells. Tumor recurrence following standard-of-care therapy results in a rich landscape of inflammatory cells throughout the glioma-infiltrated cortex. Immune cells consisting of glioma-associated macrophages and microglia (GAMMs) overwhelmingly constitute the bulk of the recurrent glioblastoma (rGBM) microenvironment, in comparison to the highly cellular and proliferative tumor microenvironment characteristic of primary GBM. These immune cells dynamically interact within the tumor microenvironment and can contribute to disease progression and therapy resistance while also providing novel targets for emerging immunotherapies. Within these varying contexts, histological-based assessments of immune cells in rGBM, including immunohistochemistry (IHC) and immunofluorescence (IF), offer a critical way to visualize and examine the inflammatory landscape. Here, we exhaustively review the available body of literature on the inflammatory landscape in rGBM as identified through histological-based assessments. We highlight the heterogeneity of immune cells throughout the glioma-infiltrated cortex with a focus on microglia and macrophages, drawing insights from canonical and novel immune-cell histological markers to estimate cell phenotypes and function. Lastly, we discuss opportunities for immunomodulatory treatments aiming to harness the inflammatory landscape in rGBM.
Collapse
Affiliation(s)
- Nicholas B. Dadario
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Deborah M. Boyett
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Damian E. Teasley
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Peter J. Chabot
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Nathan J. Winans
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Michael G. Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Colin P. Sperring
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| | - Peter Canoll
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA
| | - Jeffrey N. Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, NY-Presbyterian Hospital, New York, NY 10032, USA; (D.M.B.); (D.E.T.); (P.J.C.); (N.J.W.); (M.G.A.); (C.P.S.); (P.C.)
| |
Collapse
|
12
|
Piper K, Kumar JI, Domino J, Tuchek C, Vogelbaum MA. Consensus review on strategies to improve delivery across the blood-brain barrier including focused ultrasound. Neuro Oncol 2024; 26:1545-1556. [PMID: 38770775 PMCID: PMC11376463 DOI: 10.1093/neuonc/noae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Indexed: 05/22/2024] Open
Abstract
Drug delivery to the central nervous system (CNS) has been a major challenge for CNS tumors due to the impermeability of the blood-brain barrier (BBB). There has been a multitude of techniques aimed at overcoming the BBB obstacle aimed at utilizing natural transport mechanisms or bypassing the BBB which we review here. Another approach that has generated recent interest in the recently published literature is to use new technologies (Laser Interstitial Thermal Therapy, LITT; or Low-Intensity Focused Ultrasound, LIFU) to temporarily increase BBB permeability. This review overviews the advantages, disadvantages, and major advances of each method. LIFU has been a major area of research to allow for chemotherapeutics to cross the BBB which has a particular emphasis in this review. While most of the advances remain in animal studies, there are an increasing number of translational clinical trials that will have results in the next few years.
Collapse
Affiliation(s)
- Keaton Piper
- Department of Neurosurgery, University of South Florida, Tampa, Florida, USA
| | - Jay I Kumar
- Department of Neurosurgery, University of South Florida, Tampa, Florida, USA
| | - Joseph Domino
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Chad Tuchek
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Michael A Vogelbaum
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| |
Collapse
|
13
|
Zhang TQ, Lv QY, Jin WL. The cellular-centered view of hypoxia tumor microenvironment: Molecular mechanisms and therapeutic interventions. Biochim Biophys Acta Rev Cancer 2024; 1879:189137. [PMID: 38880161 DOI: 10.1016/j.bbcan.2024.189137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/01/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Cancer is a profoundly dynamic, heterogeneous and aggressive systemic ailment, with a coordinated evolution of various types of tumor niches. Hypoxia plays an indispensable role in the tumor micro-ecosystem, drastically enhancing the plasticity of cancer cells, fibroblasts and immune cells and orchestrating intercellular communication. Hypoxia-induced signals, particularly hypoxia-inducible factor-1α (HIF-1α), drive the reprogramming of genetic, transcriptional, and proteomic profiles. This leads to a spectrum of interconnected processes, including augmented survival of cancer cells, evasion of immune surveillance, metabolic reprogramming, remodeling of the extracellular matrix, and the development of resistance to conventional therapeutic modalities like radiotherapy and chemotherapy. Here, we summarize the latest research on the multifaceted effects of hypoxia, where a multitude of cellular and non-cellular elements crosstalk with each other and co-evolve in a synergistic manner. Additionally, we investigate therapeutic approaches targeting hypoxic niche, encompassing hypoxia-activated prodrugs, HIF inhibitors, nanomedicines, and combination therapies. Finally, we discuss some of the issues to be addressed and highlight the potential of emerging technologies in the treatment of cancer.
Collapse
Affiliation(s)
- Tian-Qi Zhang
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; The Second Hospital of Jilin University, Changchun 130041, China
| | - Qian-Yu Lv
- The Second Hospital of Jilin University, Changchun 130041, China
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
14
|
Ghosh S, Bhaskar R, Mishra R, Arockia Babu M, Abomughaid MM, Jha NK, Sinha JK. Neurological insights into brain-targeted cancer therapy and bioinspired microrobots. Drug Discov Today 2024; 29:104105. [PMID: 39029869 DOI: 10.1016/j.drudis.2024.104105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/03/2024] [Accepted: 07/12/2024] [Indexed: 07/21/2024]
Abstract
Cancer, a multifaceted and pernicious disease, continuously challenges medicine, requiring innovative treatments. Brain cancers pose unique and daunting challenges due to the intricacies of the central nervous system and the blood-brain barrier. In this era of precision medicine, the convergence of neurology, oncology, and cutting-edge technology has given birth to a promising avenue - targeted cancer therapy. Furthermore, bioinspired microrobots have emerged as an ingenious approach to drug delivery, enabling precision and control in cancer treatment. This Keynote review explores the intricate web of neurological insights into brain-targeted cancer therapy and the paradigm-shifting world of bioinspired microrobots. It serves as a critical and comprehensive overview of these evolving fields, aiming to underscore their integration and potential for revolutionary cancer treatments.
Collapse
Affiliation(s)
- Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | - Richa Mishra
- Department of Computer Science and Engineering, Parul University, Vadodara, Gujrat 391760, India
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Niraj Kumar Jha
- Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, Punjab, India; Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | | |
Collapse
|
15
|
Luo Y, Niu M, Liu Y, Zhang M, Deng Y, Mu D, Xu J, Hong S. Oncoproteins E6 and E7 upregulate topoisomerase I to activate the cGAS-PD-L1 pathway in cervical cancer development. Front Pharmacol 2024; 15:1450875. [PMID: 39156107 PMCID: PMC11327024 DOI: 10.3389/fphar.2024.1450875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
Background: Cervical cancer (CC) stands as a significant health threat to women globally, with high-risk human papillomaviruses as major etiologic agents. The DNA damage repair (DDR) protein topoisomerase I (TOP1) has been linked to various cancers, yet its distinct roles and mechanisms in CC are not fully elucidated. Methods: We investigated TOP1 expression in cervical intraepithelial neoplasia (CIN) and CC tissues utilizing qRT-PCR and IHC, correlating findings with patient prognosis. Subsequent knockdown studies were performed in vitro and in vivo to evaluate the influence of TOP1 on tumor growth, DNA repair, and inflammatory responses. Results: TOP1 was highly expressed in CIN and CC, negatively correlating with patient prognosis. Inhibition of TOP1 impeded CC cell growth and disrupted DNA repair. TOP1 was shown to regulate tumor-promoting inflammation and programmed death-ligand 1 (PD-L1) production in a cGAS-dependent manner. HPV oncoproteins E6 and E7 upregulated TOP1 and activated the cGAS-PD-L1 pathway. Conclusions: TOP1 acts as a DNA repair mediator, promoting CC development and immune evasion. Targeting the TOP1-cGAS-PD-L1 axis could be a potential therapeutic strategy for CC.
Collapse
Affiliation(s)
- Ying Luo
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Mengda Niu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yanfei Liu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Miaochang Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yuanyuan Deng
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Dan Mu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Junfen Xu
- Department of Gynecologic Oncology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shiyuan Hong
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Di Francesco V, Chua AJ, Davoudi E, Kim J, Bleier BS, Amiji MM. Minimally invasive nasal infusion (MINI) approach for CNS delivery of protein therapeutics: A case study with ovalbumin. J Control Release 2024; 372:674-681. [PMID: 38909700 DOI: 10.1016/j.jconrel.2024.06.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/08/2024] [Accepted: 06/21/2024] [Indexed: 06/25/2024]
Abstract
One of the primary obstacles in treating central nervous system (CNS) disorders lies in the limited ability of disease-modifying drugs to cross the blood-brain barrier (BBB). Our previously described Minimally Invasive Nasal Depot (MIND) technique has proven successful in delivering various drugs to the brain in rat models via a trans-olfactory mucosal approach. In this study, we introduce a novel Minimally Invasive Nasal Infusion (MINI) delivery approach for administering ovalbumin, a model protein, utilizing a programmable infusion pump (iPRECIO SMP-310R) in a mouse model. This research highlights the significant role of olfactory mucosa in nose-to-brain delivery, with an efficacy of nearly 45% compared to intracerebroventricular (ICV) administration. This demonstrates its potential as an alternative procedure for treating CNS diseases, offering a greater safety profile relative to the highly invasive clinical routes traditionally adopted for CNS drug delivery.
Collapse
Affiliation(s)
- Valentina Di Francesco
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, 140 The Fenway Building, MA 02115., USA; Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114., USA
| | - Andy J Chua
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, 140 The Fenway Building, MA 02115., USA; Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114., USA; Department of Otorhinolaryngology - Head and Neck Surgery, Sengkang General Hospital, 110, Sengkang, E Way, Singapore 544886
| | - Elham Davoudi
- Department of Biomedical and Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts at Lowell, Lowell, MA, USA
| | - Jonghan Kim
- Department of Biomedical and Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts at Lowell, Lowell, MA, USA
| | - Benjamin S Bleier
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114., USA.
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, 140 The Fenway Building, MA 02115., USA; Department of Chemical Engineering, College of Engineering, Northeastern University, 360 Huntington Avenue, 140 The Fenway Building, Boston, MA 02115., USA.
| |
Collapse
|
17
|
Jain S, Griffith JI, Porath KA, Rathi S, Le J, Pasa TI, Decker PA, Gupta SK, Hu Z, Carlson BL, Bakken K, Burgenske DM, Feldsien TM, Lefebvre DR, Vaubel RA, Eckel-Passow JE, Reilly EB, Elmquist WF, Sarkaria JN. Bystander Effects, Pharmacokinetics, and Linker-Payload Stability of EGFR-Targeting Antibody-Drug Conjugates Losatuxizumab Vedotin and Depatux-M in Glioblastoma Models. Clin Cancer Res 2024; 30:3287-3297. [PMID: 38743766 PMCID: PMC11292202 DOI: 10.1158/1078-0432.ccr-24-0426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/05/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
PURPOSE Antibody-drug conjugates (ADC) are targeted therapies with robust efficacy in solid cancers, and there is intense interest in using EGFR-specific ADCs to target EGFR-amplified glioblastoma (GBM). Given GBM's molecular heterogeneity, the bystander activity of ADCs may be important for determining treatment efficacy. In this study, the activity and toxicity of two EGFR-targeted ADCs with similar auristatin toxins, Losatuxizumab vedotin (ABBV-221) and Depatuxizumab mafodotin (Depatux-M), were compared in GBM patient-derived xenografts (PDX) and normal murine brain following direct infusion by convection-enhanced delivery (CED). EXPERIMENTAL DESIGN EGFRviii-amplified and non-amplified GBM PDXs were used to determine in vitro cytotoxicity, in vivo efficacy, and bystander activities of ABBV-221 and Depatux-M. Nontumor-bearing mice were used to evaluate the pharmacokinetics (PK) and toxicity of ADCs using LC-MS/MS and immunohistochemistry. RESULTS CED improved intracranial efficacy of Depatux-M and ABBV-221 in three EGFRviii-amplified GBM PDX models (Median survival: 125 to >300 days vs. 20-49 days with isotype control AB095). Both ADCs had comparable in vitro and in vivo efficacy. However, neuronal toxicity and CD68+ microglia/macrophage infiltration were significantly higher in brains infused with ABBV-221 with the cell-permeable monomethyl auristatin E (MMAE), compared with Depatux-M with the cell-impermeant monomethyl auristatin F. CED infusion of ABBV-221 into the brain or incubation of ABBV-221 with normal brain homogenate resulted in a significant release of MMAE, consistent with linker instability in the brain microenvironment. CONCLUSIONS EGFR-targeting ADCs are promising therapeutic options for GBM when delivered intratumorally by CED. However, the linker and payload for the ADC must be carefully considered to maximize the therapeutic window.
Collapse
Affiliation(s)
- Sonia Jain
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Jessica I. Griffith
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota.
| | - Kendra A. Porath
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Sneha Rathi
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota.
| | - Jiayan Le
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota.
| | - Tugce I. Pasa
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota.
| | - Paul A. Decker
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota.
| | - Shiv K. Gupta
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Zeng Hu
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Brett L. Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Katrina Bakken
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| | | | | | | | - Rachael A. Vaubel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota.
| | | | | | - William F. Elmquist
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota.
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
18
|
Savage WM, Yeary MD, Tang AJ, Sperring CP, Argenziano MG, Adapa AR, Yoh N, Canoll P, Bruce JN. Biomarkers of immunotherapy in glioblastoma. Neurooncol Pract 2024; 11:383-394. [PMID: 39006524 PMCID: PMC11241363 DOI: 10.1093/nop/npae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
Glioblastoma (GBM) is the most common primary brain cancer, comprising half of all malignant brain tumors. Patients with GBM have a poor prognosis, with a median survival of 14-15 months. Current therapies for GBM, including chemotherapy, radiotherapy, and surgical resection, remain inadequate. Novel therapies are required to extend patient survival. Although immunotherapy has shown promise in other cancers, including melanoma and non-small lung cancer, its efficacy in GBM has been limited to subsets of patients. Identifying biomarkers of immunotherapy response in GBM could help stratify patients, identify new therapeutic targets, and develop more effective treatments. This article reviews existing and emerging biomarkers of clinical response to immunotherapy in GBM. The scope of this review includes immune checkpoint inhibitor and antitumoral vaccination approaches, summarizing the variety of molecular, cellular, and computational methodologies that have been explored in the setting of anti-GBM immunotherapies.
Collapse
Affiliation(s)
- William M Savage
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Mitchell D Yeary
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Anthony J Tang
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Colin P Sperring
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Michael G Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Arjun R Adapa
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Nina Yoh
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
19
|
Rolfe NW, Dadario NB, Canoll P, Bruce JN. A Review of Therapeutic Agents Given by Convection-Enhanced Delivery for Adult Glioblastoma. Pharmaceuticals (Basel) 2024; 17:973. [PMID: 39204078 PMCID: PMC11357193 DOI: 10.3390/ph17080973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 09/03/2024] Open
Abstract
Glioblastoma remains a devastating disease with a bleak prognosis despite continued research and numerous clinical trials. Convection-enhanced delivery offers researchers and clinicians a platform to bypass the blood-brain barrier and administer drugs directly to the brain parenchyma. While not without significant technological challenges, convection-enhanced delivery theoretically allows for a wide range of therapeutic agents to be delivered to the tumoral space while preventing systemic toxicities. This article provides a comprehensive review of the antitumor agents studied in clinical trials of convection-enhanced delivery to treat adult high-grade gliomas. Agents are grouped by classes, and preclinical evidence for these agents is summarized, as is a brief description of their mechanism of action. The strengths and weaknesses of each clinical trial are also outlined. By doing so, the difficulty of untangling the efficacy of a drug from the technological challenges of convection-enhanced delivery is highlighted. Finally, this article provides a focused review of some therapeutics that might stand to benefit from future clinical trials for glioblastoma using convection-enhanced delivery.
Collapse
Affiliation(s)
- Nathaniel W. Rolfe
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, NY 10032, USA;
| | - Nicholas B. Dadario
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, NY 10032, USA;
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, NY 10032, USA;
| | - Jeffrey N. Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, NY 10032, USA;
| |
Collapse
|
20
|
Amirrashedi M, Jensen AI, Tang Q, Straathof NJW, Ravn K, Pedersen CG, Langhorn L, Poulsen FR, Woolley M, Johnson D, Williams J, Kidd C, Thisgaard H, Halle B. The Influence of Size on the Intracranial Distribution of Biomedical Nanoparticles Administered by Convection-enhanced Delivery in Minipigs. ACS NANO 2024; 18:17869-17881. [PMID: 38925630 PMCID: PMC11238734 DOI: 10.1021/acsnano.4c04159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/25/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
Because of the blood-brain barrier (BBB), successful drug delivery to the brain has long been a key objective for the medical community, calling for pioneering technologies to overcome this challenge. Convection-enhanced delivery (CED), a form of direct intraparenchymal microinfusion, shows promise but requires optimal infusate design and real-time distribution monitoring. The size of the infused substances appears to be especially critical, with current knowledge being limited. Herein, we examined the intracranial administration of polyethylene glycol (PEG)-coated nanoparticles (NPs) of various sizes using CED in groups of healthy minipigs (n = 3). We employed stealth liposomes (LIPs, 130 nm) and two gold nanoparticle designs (AuNPs) of different diameters (8 and 40 nm). All were labeled with copper-64 for quantitative and real-time monitoring of the infusion via positron emission tomography (PET). NPs were infused via two catheters inserted bilaterally in the putaminal regions of the animals. Our results suggest CED with NPs holds promise for precise brain drug delivery, with larger LIPs exhibiting superior distribution volumes and intracranial retention over smaller AuNPs. PET imaging alongside CED enabled dynamic visualization of the process, target coverage, timely detection of suboptimal infusion, and quantification of distribution volumes and concentration gradients. These findings may augment the therapeutic efficacy of the delivery procedure while mitigating unwarranted side effects associated with nonvisually monitored delivery approaches. This is of vital importance, especially for chronic intermittent infusions through implanted catheters, as this information enables informed decisions for modulating targeted infusion volumes on a catheter-by-catheter, patient-by-patient basis.
Collapse
Affiliation(s)
- Mahsa Amirrashedi
- Department
of Nuclear Medicine, Odense University Hospital, Odense 5000, Denmark
- Department
of Applied Mathematics and Computer Science, Technical University of Denmark, Lyngby 2800, Denmark
- Danish
Research Centre for Magnetic Resonance, Centre for Functional and
Diagnostic Imaging and Research, Copenhagen
University Hospital Amager and Hvidovre, Copenhagen 2650, Denmark
| | - Andreas Ingemann Jensen
- The
Hevesy Laboratory, Department of Health Technology, Technical University of Denmark, Roskilde 4000, Denmark
| | - Qing Tang
- The
Hevesy Laboratory, Department of Health Technology, Technical University of Denmark, Roskilde 4000, Denmark
| | | | - Katharina Ravn
- The
Hevesy Laboratory, Department of Health Technology, Technical University of Denmark, Roskilde 4000, Denmark
| | | | - Louise Langhorn
- Biomedical
Laboratory, University of Southern Denmark, Odense 5000, Denmark
| | - Frantz Rom Poulsen
- Department
of Clinical Research and BRIDGE (Brain Research - Interdisciplinary
Guided Excellence), University of Southern
Denmark, Odense 5230, Denmark
- Department
of Neurosurgery, Odense University Hospital, Odense 5000, Denmark
| | - Max Woolley
- Renishaw
Neuro Solutions Ltd (RNS), Gloucestershire GL12 8SP, United Kingdom
| | - David Johnson
- Renishaw
Neuro Solutions Ltd (RNS), Gloucestershire GL12 8SP, United Kingdom
| | - Julia Williams
- Renishaw
Neuro Solutions Ltd (RNS), Gloucestershire GL12 8SP, United Kingdom
| | - Charlotte Kidd
- Renishaw
Neuro Solutions Ltd (RNS), Gloucestershire GL12 8SP, United Kingdom
| | - Helge Thisgaard
- Department
of Nuclear Medicine, Odense University Hospital, Odense 5000, Denmark
- Department
of Clinical Research and BRIDGE (Brain Research - Interdisciplinary
Guided Excellence), University of Southern
Denmark, Odense 5230, Denmark
| | - Bo Halle
- Department
of Clinical Research and BRIDGE (Brain Research - Interdisciplinary
Guided Excellence), University of Southern
Denmark, Odense 5230, Denmark
- Department
of Neurosurgery, Odense University Hospital, Odense 5000, Denmark
| |
Collapse
|
21
|
Nguyen TTT, Greene LA, Mnatsakanyan H, Badr CE. Revolutionizing Brain Tumor Care: Emerging Technologies and Strategies. Biomedicines 2024; 12:1376. [PMID: 38927583 PMCID: PMC11202201 DOI: 10.3390/biomedicines12061376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive forms of brain tumor, characterized by a daunting prognosis with a life expectancy hovering around 12-16 months. Despite a century of relentless research, only a select few drugs have received approval for brain tumor treatment, largely due to the formidable barrier posed by the blood-brain barrier. The current standard of care involves a multifaceted approach combining surgery, irradiation, and chemotherapy. However, recurrence often occurs within months despite these interventions. The formidable challenges of drug delivery to the brain and overcoming therapeutic resistance have become focal points in the treatment of brain tumors and are deemed essential to overcoming tumor recurrence. In recent years, a promising wave of advanced treatments has emerged, offering a glimpse of hope to overcome the limitations of existing therapies. This review aims to highlight cutting-edge technologies in the current and ongoing stages of development, providing patients with valuable insights to guide their choices in brain tumor treatment.
Collapse
Affiliation(s)
- Trang T. T. Nguyen
- Ronald O. Perelman Department of Dermatology, Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Lloyd A. Greene
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Hayk Mnatsakanyan
- Department of Neurology, Massachusetts General Hospital, Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA; (H.M.); (C.E.B.)
| | - Christian E. Badr
- Department of Neurology, Massachusetts General Hospital, Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA; (H.M.); (C.E.B.)
| |
Collapse
|
22
|
Narsinh KH, Perez E, Haddad AF, Young JS, Savastano L, Villanueva-Meyer JE, Winkler E, de Groot J. Strategies to Improve Drug Delivery Across the Blood-Brain Barrier for Glioblastoma. Curr Neurol Neurosci Rep 2024; 24:123-139. [PMID: 38578405 PMCID: PMC11016125 DOI: 10.1007/s11910-024-01338-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2024] [Indexed: 04/06/2024]
Abstract
PURPOSE OF REVIEW Glioblastoma remains resistant to most conventional treatments. Despite scientific advances in the past three decades, there has been a dearth of effective new treatments. New approaches to drug delivery and clinical trial design are needed. RECENT FINDINGS We discuss how the blood-brain barrier and tumor microenvironment pose challenges for development of effective therapies for glioblastoma. Next, we discuss treatments in development that aim to overcome these barriers, including novel drug designs such as nanoparticles and antibody-drug conjugates, novel methods of drug delivery, including convection-enhanced and intra-arterial delivery, and novel methods to enhance drug penetration, such as blood-brain barrier disruption by focused ultrasound and laser interstitial thermal therapy. Lastly, we address future opportunities, positing combination therapy as the best strategy for effective treatment, neoadjuvant and window-of-opportunity approaches to simultaneously enhance therapeutic effectiveness with interrogation of on-treatment biologic endpoints, and adaptive platform and basket trials as imperative for future trial design. New approaches to GBM treatment should account for the blood-brain barrier and immunosuppression by improving drug delivery, combining treatments, and integrating novel clinical trial designs.
Collapse
Affiliation(s)
- Kazim H Narsinh
- Department of Neurologic Surgery, University of California, San Francisco, CA, USA.
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, CA, USA.
| | - Edgar Perez
- Department of Neurologic Surgery, University of California, San Francisco, CA, USA
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Alexander F Haddad
- Department of Neurologic Surgery, University of California, San Francisco, CA, USA
| | - Jacob S Young
- Department of Neurologic Surgery, University of California, San Francisco, CA, USA
| | - Luis Savastano
- Department of Neurologic Surgery, University of California, San Francisco, CA, USA
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Javier E Villanueva-Meyer
- Department of Neurologic Surgery, University of California, San Francisco, CA, USA
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Ethan Winkler
- Department of Neurologic Surgery, University of California, San Francisco, CA, USA
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, CA, USA
| | - John de Groot
- Department of Neurologic Surgery, University of California, San Francisco, CA, USA
| |
Collapse
|
23
|
Wang H, Argenziano MG, Yoon H, Boyett D, Save A, Petridis P, Savage W, Jackson P, Hawkins-Daarud A, Tran N, Hu L, Al Dalahmah O, Bruce JN, Grinband J, Swanson KR, Canoll P, Li J. Biologically-informed deep neural networks provide quantitative assessment of intratumoral heterogeneity in post-treatment glioblastoma. RESEARCH SQUARE 2024:rs.3.rs-3891425. [PMID: 38585856 PMCID: PMC10996806 DOI: 10.21203/rs.3.rs-3891425/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Intratumoral heterogeneity poses a significant challenge to the diagnosis and treatment of glioblastoma (GBM). This heterogeneity is further exacerbated during GBM recurrence, as treatment-induced reactive changes produce additional intratumoral heterogeneity that is ambiguous to differentiate on clinical imaging. There is an urgent need to develop non-invasive approaches to map the heterogeneous landscape of histopathological alterations throughout the entire lesion for each patient. We propose to predictively fuse Magnetic Resonance Imaging (MRI) with the underlying intratumoral heterogeneity in recurrent GBM using machine learning (ML) by leveraging image-localized biopsies with their associated locoregional MRI features. To this end, we develop BioNet, a biologically-informed neural network model, to predict regional distributions of three tissue-specific gene modules: proliferating tumor, reactive/inflammatory cells, and infiltrated brain tissue. BioNet offers valuable insights into the integration of multiple implicit and qualitative biological domain knowledge, which are challenging to describe in mathematical formulations. BioNet performs significantly better than a range of existing methods on cross-validation and blind test datasets. Voxel-level prediction maps of the gene modules by BioNet help reveal intratumoral heterogeneity, which can improve surgical targeting of confirmatory biopsies and evaluation of neuro-oncological treatment effectiveness. The non-invasive nature of the approach can potentially facilitate regular monitoring of the gene modules over time, and making timely therapeutic adjustment. These results also highlight the emerging role of ML in precision medicine.
Collapse
Affiliation(s)
- Hairong Wang
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael G Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Hyunsoo Yoon
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ, USA
| | - Deborah Boyett
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Akshay Save
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Petros Petridis
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Psychiatry, New York University, New York, NY, USA
| | - William Savage
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Pamela Jackson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Andrea Hawkins-Daarud
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Nhan Tran
- Department of Cancer Biology, Mayo Clinic, Phoenix, AZ, USA
| | - Leland Hu
- Department of Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Osama Al Dalahmah
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jeffrey N. Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Jack Grinband
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kristin R Swanson
- Mathematical NeuroOncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jing Li
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
24
|
Perera A, Brock O, Ahmed A, Shaw C, Ashkan K. Taking the knife to neurodegeneration: a review of surgical gene therapy delivery to the CNS. Acta Neurochir (Wien) 2024; 166:136. [PMID: 38483631 PMCID: PMC10940433 DOI: 10.1007/s00701-024-06028-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/28/2024] [Indexed: 03/17/2024]
Abstract
Gene supplementation and editing for neurodegenerative disorders has emerged in recent years as the understanding of the genetic mechanisms underlying several neurodegenerative disorders increases. The most common medium to deliver genetic material to cells is via viral vectors; and with respect to the central nervous system, adeno-associated viral (AAV) vectors are a popular choice. The most successful example of AAV-based gene therapy for neurodegenerative disorders is Zolgensma© which is a transformative intravenous therapy given to babies with spinal muscular atrophy. However, the field has stalled in achieving safe drug delivery to the central nervous system in adults for which treatments for disorders such as amyotrophic lateral sclerosis are desperately needed. Surgical gene therapy delivery has been proposed as a potential solution to this problem. While the field of the so-called regenerative neurosurgery has yielded pre-clinical optimism, several challenges have emerged. This review seeks to explore the field of regenerative neurosurgery with respect to AAV-based gene therapy for neurodegenerative diseases, its progress so far and the challenges that need to be overcome.
Collapse
Affiliation(s)
- Andrea Perera
- Maurice Wohl Institute of Neuroscience, Department of Basic Clinical Neuroscience, King's College London, Cutcombe Road, Denmark Hill, London, SE5 9RS, UK.
- Department of Neurosurgery, King's College Hospital NHS Trust, London, UK.
| | - Olivier Brock
- Maurice Wohl Institute of Neuroscience, Department of Basic Clinical Neuroscience, King's College London, Cutcombe Road, Denmark Hill, London, SE5 9RS, UK
| | - Aminul Ahmed
- Department of Neurosurgery, King's College Hospital NHS Trust, London, UK
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Chris Shaw
- Maurice Wohl Institute of Neuroscience, Department of Basic Clinical Neuroscience, King's College London, Cutcombe Road, Denmark Hill, London, SE5 9RS, UK
- Centre for Brain Research, University of Auckland, 85 Park Road Grafton, Auckland, 1023, New Zealand
| | - Keyoumars Ashkan
- Maurice Wohl Institute of Neuroscience, Department of Basic Clinical Neuroscience, King's College London, Cutcombe Road, Denmark Hill, London, SE5 9RS, UK
- Department of Neurosurgery, King's College Hospital NHS Trust, London, UK
| |
Collapse
|
25
|
Kreatsoulas D, Damante M, Cua S, Lonser RR. Adjuvant convection-enhanced delivery for the treatment of brain tumors. J Neurooncol 2024; 166:243-255. [PMID: 38261143 PMCID: PMC10834622 DOI: 10.1007/s11060-023-04552-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND Malignant gliomas are a therapeutic challenge and remain nearly uniformly fatal. While new targeted chemotherapeutic agentsagainst malignant glioma have been developed in vitro, these putative therapeutics have not been translated into successful clinical treatments. The lack of clinical effectiveness can be the result of ineffective biologic strategies, heterogeneous tumor targets and/or the result of poortherapeutic distribution to malignant glioma cells using conventional nervous system delivery modalities (intravascular, cerebrospinal fluid and/orpolymer implantation), and/or ineffective biologic strategies. METHODS The authors performed a review of the literature for the terms "convection enhanced delivery", "glioblastoma", and "glioma". Selectclinical trials were summarized based on their various biological mechanisms and technological innovation, focusing on more recently publisheddata when possible. RESULTS We describe the properties, features and landmark clinical trials associated with convection-enhanced delivery for malignant gliomas.We also discuss future trends that will be vital to CED innovation and improvement. CONCLUSION Efficacy of CED for malignant glioma to date has been mixed, but improvements in technology and therapeutic agents arepromising.
Collapse
Affiliation(s)
- Daniel Kreatsoulas
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, The Ohio State University, N1019 Doan Hall, 410 W 10Th Avenue, Columbus, OH, 43210, USA.
| | - Mark Damante
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, The Ohio State University, N1019 Doan Hall, 410 W 10Th Avenue, Columbus, OH, 43210, USA
| | - Santino Cua
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, The Ohio State University, N1019 Doan Hall, 410 W 10Th Avenue, Columbus, OH, 43210, USA
| | - Russell R Lonser
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, The Ohio State University, N1019 Doan Hall, 410 W 10Th Avenue, Columbus, OH, 43210, USA
| |
Collapse
|
26
|
Sugii N, Ninomiya Y, Akimoto Y, Tsurubuchi T, Ishikawa E. H3 K27-altered diffuse midline glioma in adults arising from atypical regions: Two case reports and literature review. Radiol Case Rep 2024; 19:200-206. [PMID: 38028289 PMCID: PMC10651424 DOI: 10.1016/j.radcr.2023.10.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/06/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Diffuse midline glioma (DMG), H3 K27-altered, is a newly defined "pediatric-type," diffuse, high-grade glioma under current WHO classifications (updated in 2021). An essential diagnostic criteria of DMG is its occurrence in the midline structures; most intracranial DMG occurs in the brainstem or thalamus but can also occur in other midline structures. We experienced 2 adult cases of intracranial DMGs in areas other than the brainstem and thalamus that were initially difficult to diagnose. Case 1 was a 49-year-old man with extensive T2 high-signal lesions in the bilateral frontal lobes and corpus callosum on brain MRI. A Gd-based contrast medium partially enhanced the lesion and showed marked diffusion restriction, mimicking malignant lymphoma. Case 2 was a 24-year-old man who presented with paroxysmal olfactory abnormalities. The tumor extended mainly to the right temporal lobe, the right basal forebrain, and the bilateral hypothalamus, showing a T2/FLAIR mismatch sign suggestive of IDH-mutant astrocytoma without 1p/19q co-deletion. After a biopsy, both cases were properly diagnosed as DMG, H3 K27-altered (K27M-mutant). Diagnosing adult cases involving atypical midline structures is sometimes challenging before surgery; we discuss this phenomenon with both case details and a literature review.
Collapse
Affiliation(s)
- Narushi Sugii
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575 Japan
| | - Yuki Ninomiya
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575 Japan
| | - Yu Akimoto
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575 Japan
| | - Takao Tsurubuchi
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575 Japan
| | - Eiichi Ishikawa
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575 Japan
| |
Collapse
|
27
|
Xi J, Liu K, Peng Z, Dai X, Wang Y, Cai C, Yang D, Yan C, Li X. Toxic warhead-armed antibody for targeted treatment of glioblastoma. Crit Rev Oncol Hematol 2024; 193:104205. [PMID: 38036153 DOI: 10.1016/j.critrevonc.2023.104205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 12/02/2023] Open
Abstract
Glioblastoma is a fatal intracranial tumor with a poor prognosis, exhibiting uninterrupted malignant progression, widespread invasion throughout the brain leading to the destruction of normal brain tissue and inevitable death. Monoclonal antibodies alone or conjugated with cytotoxic payloads to treat patients with different solid tumors showed effective. This treatment strategy is being explored for patients with glioblastoma (GBM) to obtain meaningful clinical responses and offer new drug options for the treatment of this devastating disease. In this review, we summarize clinical data (from pubmed.gov database and clinicaltrial.gov database) on the efficacy and toxicity of naked antibodies and antibody-drug conjugates (ADCs) against multiple targets on GBM, elucidate the mechanisms that ADCs act at the site of GBM lesions. Finally, we discuss the potential strategies for ADC therapies currently used to treat GBM patients.
Collapse
Affiliation(s)
- Jingjing Xi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Kai Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhaolei Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaolin Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yulin Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chunyan Cai
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dejun Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chunmei Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
28
|
Dhanawat M, Garima, Wilson K, Gupta S, Chalotra R, Gupta N. Convection-enhanced Diffusion: A Novel Tactics to Crack the BBB. Curr Drug Deliv 2024; 21:1515-1528. [PMID: 38275045 DOI: 10.2174/0115672018266501231207095127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/20/2023] [Accepted: 11/20/2023] [Indexed: 01/27/2024]
Abstract
Although the brain is very accessible to nutrition and oxygen, it can be difficult to deliver medications to malignant brain tumours. To get around some of these issues and enable the use of therapeutic pharmacological substances that wouldn't typically cross the blood-brain barrier (BBB), convection-enhanced delivery (CED) has been developed. It is a cutting-edge strategy that gets beyond the blood-brain barrier and enables targeted drug administration to treat different neurological conditions such as brain tumours, Parkinson's disease, and epilepsy. Utilizing pressure gradients to spread the medicine across the target area is the main idea behind this diffusion mechanism. Through one to several catheters positioned stereotactically directly within the tumour mass, around the tumour, or in the cavity created by the resection, drugs are given. This method can be used in a variety of drug classes, including traditional chemotherapeutics and cutting-edge investigational targeted medications by using positive-pressure techniques. The drug delivery volume must be optimized for an effective infusion while minimizing backflow, which causes side effects and lowers therapeutic efficacy. Therefore, this technique provides a promising approach for treating disorders of the central nervous system (CNS).
Collapse
Affiliation(s)
- Meenakshi Dhanawat
- Amity Institute of Pharmacy, Amity University Haryana, Amity Education Valley, Panchgaon, Manesar, Gurugram, Haryana, 122413, India
| | - Garima
- M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana- Ambala, Haryana, 133207, India
| | - Kashish Wilson
- M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana- Ambala, Haryana, 133207, India
| | - Sumeet Gupta
- M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana- Ambala, Haryana, 133207, India
| | - Rishabh Chalotra
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Nidhi Gupta
- M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana- Ambala, Haryana, 133207, India
| |
Collapse
|
29
|
Aquilanti E, Goldoni S, Baker A, Kotynkova K, Andersen S, Bozinov V, Gao GF, Cherniack AD, Lange M, Lesche R, Kopitz C, Lienau P, Lewis TA, Garrido M, Gradl S, Seidel H, Tseng YY, Ligon KL, Wen PY, Meyerson M, Greulich H. Velcrin molecular glues induce apoptosis in glioblastomas with high PDE3A and SLFN12 expression. Neurooncol Adv 2024; 6:vdae115. [PMID: 39166256 PMCID: PMC11333922 DOI: 10.1093/noajnl/vdae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024] Open
Abstract
Background Velcrins are molecular glues that kill cells by inducing the formation of a protein complex between the RNase SLFN12 and the phosphodiesterase PDE3A. Formation of the complex activates SLFN12, which cleaves tRNALeu(TAA) and induces apoptosis. Velcrins such as the clinical investigational compound, BAY 2666605, were found to have activity across multiple solid tumor cell lines from the cancer cell line encyclopedia, including glioblastoma cell lines. We therefore aim to characterize velcrins as novel therapeutic agents in glioblastoma. Materials and Methods PDE3A and SLFN12 expression levels were measured in glioblastoma cell lines, the Cancer Genome Atlas (TCGA) tumor samples, and tumor neurospheres. Velcrin-treated cells were assayed for viability, induction of apoptosis, cell cycle phases, and global changes in translation. Transcriptional profiling of the cells was obtained. Xenograft-harboring mice treated with velcrins were also monitored for survival. Results We identified several velcrin-sensitive glioblastoma cell lines and 4 velcrin-sensitive glioblastoma patient-derived models. We determined that BAY 2666605 crosses the blood-brain barrier and elicits full tumor regression in an orthotopic xenograft model of GB1 cells. We also determined that the velcrins BAY 2666605 and BRD3800 induce tumor regression in subcutaneous glioblastoma PDX models. Conclusions Velcrins have antitumor activity in preclinical models of glioblastoma, warranting further investigation as potential therapeutic agents.
Collapse
Affiliation(s)
- Elisa Aquilanti
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
- Division of Neuro-Oncology, Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Silvia Goldoni
- Bayer Pharmaceuticals, Research and Early Development Oncology, Cambridge, Massachusetts, USA
| | - Andrew Baker
- Department of Pediatric Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | - Sawyer Andersen
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
| | - Vincent Bozinov
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
| | - Galen F Gao
- School of Medicine, University of Texas Southwestern, Dallas, Texas, USA
| | - Andrew D Cherniack
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Martin Lange
- Nuvisan ICB GmbH, Therapeutic Research, Berlin, Germany
- Research and Development, Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Ralf Lesche
- Nuvisan ICB GmbH, Therapeutic Research, Berlin, Germany
- Research and Development, Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Charlotte Kopitz
- Nuvisan ICB GmbH, Therapeutic Research, Berlin, Germany
- Research and Development, Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Philip Lienau
- Research and Development, Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Timothy A Lewis
- Center for the Development of Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
| | - Marine Garrido
- Bayer Consumer Care AG, Research and Early Development Oncology, Basel, Switzerland
| | - Stefan Gradl
- Research and Development, Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Henrik Seidel
- Research and Development, Pharmaceuticals, Bayer AG, Berlin, Germany
| | - Yuen-Yi Tseng
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
| | - Keith L Ligon
- Department of Pathology, Brigham and Women’s Hospital, Boston Children’s Hospital, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Division of Neuro-Oncology, Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Matthew Meyerson
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Heidi Greulich
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
30
|
Young JS, Morshed RA, Hervey-Jumper SL, Berger MS. The surgical management of diffuse gliomas: Current state of neurosurgical management and future directions. Neuro Oncol 2023; 25:2117-2133. [PMID: 37499054 PMCID: PMC10708937 DOI: 10.1093/neuonc/noad133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Indexed: 07/29/2023] Open
Abstract
After recent updates to the World Health Organization pathological criteria for diagnosing and grading diffuse gliomas, all major North American and European neuro-oncology societies recommend a maximal safe resection as the initial management of a diffuse glioma. For neurosurgeons to achieve this goal, the surgical plan for both low- and high-grade gliomas should be to perform a supramaximal resection when feasible based on preoperative imaging and the patient's performance status, utilizing every intraoperative adjunct to minimize postoperative neurological deficits. While the surgical approach and technique can vary, every effort must be taken to identify and preserve functional cortical and subcortical regions. In this summary statement on the current state of the field, we describe the tools and technologies that facilitate the safe removal of diffuse gliomas and highlight intraoperative and postoperative management strategies to minimize complications for these patients. Moreover, we discuss how surgical resections can go beyond cytoreduction by facilitating biological discoveries and improving the local delivery of adjuvant chemo- and radiotherapies.
Collapse
Affiliation(s)
- Jacob S Young
- Department of Neurological Surgery, University of California, San Francisco, USA
| | - Ramin A Morshed
- Department of Neurological Surgery, University of California, San Francisco, USA
| | | | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, USA
| |
Collapse
|
31
|
Levitin HM, Zhao W, Bruce JN, Canoll P, Sims PA. Consensus scHPF Identifies Cell Type-Specific Drug Responses in Glioma by Integrating Large-Scale scRNA-seq. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570193. [PMID: 38105955 PMCID: PMC10723271 DOI: 10.1101/2023.12.05.570193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Single-cell transcriptomic analyses now frequently involve elaborate study designs including samples from multiple individuals, experimental conditions, perturbations, and batches from complex tissues. Dimensionality reduction is required to facilitate integration, interpretation, and statistical analysis. However, these datasets often include subtly different cellular subpopulations or state transitions, which are poorly described by clustering. We previously reported a Bayesian matrix factorization algorithm called single-cell hierarchical Poisson factorization (scHPF) that identifies gene co-expression patterns directly from single-cell RNA-seq (scRNA-seq) count matrices while accounting for transcript drop-out and noise. Here, we describe consensus scHPF, which analyzes scHPF models from multiple random initializations to identify the most robust gene signatures and automatically determine the number of factors for a given dataset. Consensus scHPF facilitates integration of complex datasets with highly multi-modal posterior distributions, resulting in factors that can be uniformly analyzed across individuals and conditions. To demonstrate the utility of consensus scHPF, we performed a meta-analysis of a large-scale scRNA-seq dataset from drug-treated, human glioma slice cultures generated from surgical specimens across three major cell types, 19 patients, 10 drug treatment conditions, and 52 samples. In addition to recapitulating previously reported cell type-specific drug responses from smaller studies, consensus scHPF identified disparate effects of the topoisomerase poisons etoposide and topotecan that are highly consistent with the distinct roles and expression patterns of their respective protein targets.
Collapse
Affiliation(s)
- Hanna Mendes Levitin
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Wenting Zhao
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter Canoll
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
32
|
Pinheiro Lopes B, O’Neill L, Bourke P, Boehm D. Combined Effect of Plasma-Activated Water and Topotecan in Glioblastoma Cells. Cancers (Basel) 2023; 15:4858. [PMID: 37835552 PMCID: PMC10571909 DOI: 10.3390/cancers15194858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/16/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
The increase in cancer diagnoses and cancer deaths, severe side effects of existing treatments and resistance to traditional treatments have generated a need for new anticancer treatments. Glioblastoma multiforme (GBM) is the most common, malignant and aggressive brain cancer. Despite many innovations regarding GBM treatment, the final outcome is still very poor, making it necessary to develop new therapeutic approaches. Cold atmospheric plasma (CAP) as well as plasma-activated liquids (PAL) are being studied as new possible approaches against cancer. The anticancer activity of PAL such as "plasma-activated water" (PAW) is dependent on the reactive chemical compounds present in the solution. Possible combinatory effects with conventional therapies, such as chemotherapeutics, may expand the potential of PAL for cancer treatment. We aim to explore the therapeutic properties of a combination of PAW and topotecan (TPT), an antineoplastic agent with major cytotoxic effects during the S phase of the cell cycle, on a GBM cancer cell line (U-251mg). Combined treatments with PAW and TPT showed a reduction in the metabolic activity and cell mass, an increase in apoptotic cell death and a reduction in the long-term survival. Single applications of PAW+TPT treatments showed a cytotoxic effect in the short term and an antiproliferative effect in the long term, warranting future exploration of combining PAW with chemotherapeutic agents as new therapeutic approaches.
Collapse
Affiliation(s)
- Beatriz Pinheiro Lopes
- School of Chemical and Bioprocess Engineering, University College Dublin, D04 V1W8 Dublin, Ireland;
- Environmental Sustainability and Health Institute and School of Food Science and Environmental Health, Technological University Dublin, D07 H6K8 Dublin, Ireland;
| | - Liam O’Neill
- TheraDep Ltd., QUESTUM Innovation Centre, Limerick Institute of Technology, E91 V329 Clonmel, Ireland;
| | - Paula Bourke
- Environmental Sustainability and Health Institute and School of Food Science and Environmental Health, Technological University Dublin, D07 H6K8 Dublin, Ireland;
- Plasma Research Group, School of Biosystems and Food Engineering, University College Dublin, D04 V1W8 Dublin, Ireland
- Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Daniela Boehm
- School of Chemical and Bioprocess Engineering, University College Dublin, D04 V1W8 Dublin, Ireland;
- Environmental Sustainability and Health Institute and School of Food Science and Environmental Health, Technological University Dublin, D07 H6K8 Dublin, Ireland;
| |
Collapse
|
33
|
Tosi U, Souweidane M. Fifty years of DIPG: looking at the future with hope. Childs Nerv Syst 2023; 39:2675-2686. [PMID: 37382660 DOI: 10.1007/s00381-023-06037-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a primary brainstem tumor of childhood that carries a dismal prognosis, with median survival of less than 1 year. Because of the brain stem location and pattern of growth within the pons, Dr. Harvey Cushing, the father of modern neurosurgery, urged surgical abandonment. Such a dismal prognosis remained unchanged for decades, coupled with a lack of understanding of tumor biology and an unchanging therapeutic panorama. Beyond palliative external beam radiation therapy, no therapeutic approach has been widely accepted. In the last one to two decades, however, increased tissue availability, an improving understanding of biology, genetics, and epigenetics have led to the development of novel therapeutic targets. In parallel with this biological revolution, new methods intended to enhance drug delivery into the brain stem are contributing to a surge of exciting experimental therapeutic strategies.
Collapse
Affiliation(s)
- Umberto Tosi
- Department of Neurosurgery, Weill Cornell Medicine, 525 E 68th St Box 99, New York, NY, 10021, USA
| | - Mark Souweidane
- Department of Neurosurgery, Weill Cornell Medicine, 525 E 68th St Box 99, New York, NY, 10021, USA.
| |
Collapse
|
34
|
Ferrada L, Barahona MJ, Vera M, Stockwell BR, Nualart F. Dehydroascorbic acid sensitizes cancer cells to system x c- inhibition-induced ferroptosis by promoting lipid droplet peroxidation. Cell Death Dis 2023; 14:637. [PMID: 37752118 PMCID: PMC10522586 DOI: 10.1038/s41419-023-06153-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023]
Abstract
Since the discovery of ferroptosis, it has been postulated that this type of cell death could be utilized in treatments for cancer. Unfortunately, several highly aggressive tumor models are resistant to the pharmacological induction of ferroptosis. However, with the use of combined therapies, it is possible to recover sensitivity to ferroptosis in certain cellular models. Here, we discovered that co-treatment with the metabolically stable ferroptosis inducer imidazole ketone erastin (IKE) and the oxidized form of vitamin C, dehydroascorbic acid (DHAA), is a powerful therapy that induces ferroptosis in tumor cells previously resistant to IKE-induced ferroptosis. We determined that DHAA and IKE + DHAA delocalize and deplete GPX4 in tumor cells, specifically inducing lipid droplet peroxidation, which leads to ferroptosis. Moreover, in vivo, IKE + DHAA has high efficacy with regard to the eradication of highly aggressive tumors such as glioblastomas. Thus, the use of IKE + DHAA could be an effective and safe therapy for the eradication of difficult-to-treat cancers.
Collapse
Affiliation(s)
- Luciano Ferrada
- Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile.
| | - María José Barahona
- Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Matías Vera
- Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
- Department of Biological Sciences, Columbia University, New York, NY, 10027, USA
| | - Francisco Nualart
- Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| |
Collapse
|
35
|
Bagley SJ. Phase II trials in the era of glioblastoma immunotherapy: New mechanisms of action, familiar challenges in trial design and tumor response assessment. Neuro Oncol 2023; 25:1098-1099. [PMID: 36789741 PMCID: PMC10237420 DOI: 10.1093/neuonc/noad043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Indexed: 02/16/2023] Open
Affiliation(s)
- Stephen J Bagley
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
36
|
Padmakumar S, Amiji MM. Long-Acting Therapeutic Delivery Systems for the Treatment of Gliomas. Adv Drug Deliv Rev 2023; 197:114853. [PMID: 37149040 DOI: 10.1016/j.addr.2023.114853] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/13/2023] [Accepted: 04/23/2023] [Indexed: 05/08/2023]
Abstract
Despite the emergence of cutting-edge therapeutic strategies and tremendous progress in research, a complete cure of glioma remains elusive. The heterogenous nature of tumor, immunosuppressive state and presence of blood brain barrier are few of the major obstacles in this regard. Long-acting depot formulations such as injectables and implantables are gaining attention for drug delivery to brain owing to their ease in administration and ability to elute drug locally for extended durations in a controlled manner with minimal toxicity. Hybrid matrices fabricated by incorporating nanoparticulates within such systems help to enhance pharmaceutical advantages. Utilization of long-acting depots as monotherapy or in conjunction with existing strategies rendered significant survival benefits in many preclinical studies and some clinical trials. The discovery of novel targets, immunotherapeutic strategies and alternative drug administration routes are now coupled with several long-acting systems with an ultimate aim to enhance patient survival and prevent glioma recurrences.
Collapse
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115; Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, 02115.
| |
Collapse
|
37
|
Yalamarty SSK, Filipczak N, Li X, Subhan MA, Parveen F, Ataide JA, Rajmalani BA, Torchilin VP. Mechanisms of Resistance and Current Treatment Options for Glioblastoma Multiforme (GBM). Cancers (Basel) 2023; 15:cancers15072116. [PMID: 37046777 PMCID: PMC10093719 DOI: 10.3390/cancers15072116] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive form of brain cancer that is difficult to treat due to its resistance to both radiation and chemotherapy. This resistance is largely due to the unique biology of GBM cells, which can evade the effects of conventional treatments through mechanisms such as increased resistance to cell death and rapid regeneration of cancerous cells. Additionally, the blood–brain barrier makes it difficult for chemotherapy drugs to reach GBM cells, leading to reduced effectiveness. Despite these challenges, there are several treatment options available for GBM. The standard of care for newly diagnosed GBM patients involves surgical resection followed by concurrent chemoradiotherapy and adjuvant chemotherapy. Emerging treatments include immunotherapy, such as checkpoint inhibitors, and targeted therapies, such as bevacizumab, that attempt to attack specific vulnerabilities in GBM cells. Another promising approach is the use of tumor-treating fields, a type of electric field therapy that has been shown to slow the growth of GBM cells. Clinical trials are ongoing to evaluate the safety and efficacy of these and other innovative treatments for GBM, intending to improve with outcomes for patients.
Collapse
Affiliation(s)
- Satya Siva Kishan Yalamarty
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Xiang Li
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Farzana Parveen
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- Department of Pharmacy Services, DHQ Hospital, Jhang 35200, Pakistan
| | - Janaína Artem Ataide
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas 13083-871, Brazil
| | - Bharat Ashok Rajmalani
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Vladimir P. Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
38
|
Banu MA, Dovas A, Argenziano MG, Zhao W, Grajal HC, Higgins DM, Sperring CP, Pereira B, Ye LF, Mahajan A, Humala N, Furnari JL, Upadhyayula PS, Zandkarimi F, Nguyen TTT, Wu PB, Hai L, Karan C, Razavilar A, Siegelin MD, Kitajewski J, Bruce JN, Stockwell BR, Sims PA, Canoll PD. A cell state specific metabolic vulnerability to GPX4-dependent ferroptosis in glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.22.529581. [PMID: 36865302 PMCID: PMC9980114 DOI: 10.1101/2023.02.22.529581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Glioma cells hijack developmental transcriptional programs to control cell state. During neural development, lineage trajectories rely on specialized metabolic pathways. However, the link between tumor cell state and metabolic programs is poorly understood in glioma. Here we uncover a glioma cell state-specific metabolic liability that can be leveraged therapeutically. To model cell state diversity, we generated genetically engineered murine gliomas, induced by deletion of p53 alone (p53) or with constitutively active Notch signaling (N1IC), a pathway critical in controlling cellular fate. N1IC tumors harbored quiescent astrocyte-like transformed cell states while p53 tumors were predominantly comprised of proliferating progenitor-like cell states. N1IC cells exhibit distinct metabolic alterations, with mitochondrial uncoupling and increased ROS production rendering them more sensitive to inhibition of the lipid hydroperoxidase GPX4 and induction of ferroptosis. Importantly, treating patient-derived organotypic slices with a GPX4 inhibitor induced selective depletion of quiescent astrocyte-like glioma cell populations with similar metabolic profiles.
Collapse
Affiliation(s)
- Matei A. Banu
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Athanassios Dovas
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael G. Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Wenting Zhao
- Department of System Biology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Dominique M.O. Higgins
- Department of Neurological Surgery, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Colin P. Sperring
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Brianna Pereira
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ling F. Ye
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Aayushi Mahajan
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Nelson Humala
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Julia L. Furnari
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Pavan S. Upadhyayula
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Fereshteh Zandkarimi
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA
| | - Trang T. T. Nguyen
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter B. Wu
- Department of Neurological Surgery, UCLA Geffen School of Medicine, Los Angeles, CA, USA
| | - Li Hai
- Sulzberger Columbia Genome Center, Columbia University, New York, NY, USA
| | - Charles Karan
- Sulzberger Columbia Genome Center, Columbia University, New York, NY, USA
| | - Aida Razavilar
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Markus D. Siegelin
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jan Kitajewski
- University of Illinois Cancer Center, Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, USA
| | - Jeffrey N. Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Brent R. Stockwell
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA
| | - Peter A. Sims
- Department of System Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter D. Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
39
|
Wang Y, Malik S, Suh HW, Xiao Y, Deng Y, Fan R, Huttner A, Bindra RS, Singh V, Saltzman WM, Bahal R. Anti-seed PNAs targeting multiple oncomiRs for brain tumor therapy. SCIENCE ADVANCES 2023; 9:eabq7459. [PMID: 36753549 PMCID: PMC9908025 DOI: 10.1126/sciadv.abq7459] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 01/06/2023] [Indexed: 06/18/2023]
Abstract
Glioblastoma (GBM) is one of the most lethal malignancies with poor survival and high recurrence rates. Here, we aimed to simultaneously target oncomiRs 10b and 21, reported to drive GBM progression and invasiveness. We designed short (8-mer) γ-modified peptide nucleic acids (sγPNAs), targeting the seed region of oncomiRs 10b and 21. We entrapped these anti-miR sγPNAs in nanoparticles (NPs) formed from a block copolymer of poly(lactic acid) and hyperbranched polyglycerol (PLA-HPG). The surface of the NPs was functionalized with aldehydes to produce bioadhesive NPs (BNPs) with superior transfection efficiency and tropism for tumor cells. When combined with temozolomide, sγPNA BNPs administered via convection-enhanced delivery (CED) markedly increased the survival (>120 days) of two orthotopic (intracranial) mouse models of GBM. Hence, we established that BNPs loaded with anti-seed sγPNAs targeting multiple oncomiRs are a promising approach to improve the treatment of GBM, with a potential to personalize treatment based on tumor-specific oncomiRs.
Collapse
Affiliation(s)
- Yazhe Wang
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Shipra Malik
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Hee-Won Suh
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Yong Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Yanxiang Deng
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Anita Huttner
- Department of Pathology, Yale University, New Haven, CT 06510, USA
| | - Ranjit S. Bindra
- Department of Therapeutic Radiology, Yale University, New Haven, CT 06510, USA
| | - Vijender Singh
- Computational Biology Core, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - W. Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Raman Bahal
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
40
|
Crockett C, Lorimer C. OncoFlash January 2023. Clin Oncol (R Coll Radiol) 2023; 35:3-5. [PMID: 36586727 DOI: 10.1016/j.clon.2022.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- C Crockett
- Northern Ireland Cancer Centre, Belfast City Hospital, Belfast, UK
| | - C Lorimer
- University Hospitals Sussex NHS Foundation Trust, West Sussex, UK
| |
Collapse
|
41
|
Sperring CP, Argenziano MG, Savage WM, Teasley DE, Upadhyayula PS, Winans NJ, Canoll P, Bruce JN. Convection-enhanced delivery of immunomodulatory therapy for high-grade glioma. Neurooncol Adv 2023; 5:vdad044. [PMID: 37215957 PMCID: PMC10195574 DOI: 10.1093/noajnl/vdad044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
The prognosis for glioblastoma has remained poor despite multimodal standard of care treatment, including temozolomide, radiation, and surgical resection. Further, the addition of immunotherapies, while promising in a number of other solid tumors, has overwhelmingly failed in the treatment of gliomas, in part due to the immunosuppressive microenvironment and poor drug penetrance to the brain. Local delivery of immunomodulatory therapies circumvents some of these challenges and has led to long-term remission in select patients. Many of these approaches utilize convection-enhanced delivery (CED) for immunological drug delivery, allowing high doses to be delivered directly to the brain parenchyma, avoiding systemic toxicity. Here, we review the literature encompassing immunotherapies delivered via CED-from preclinical model systems to clinical trials-and explore how their unique combination elicits an antitumor response by the immune system, decreases toxicity, and improves survival among select high-grade glioma patients.
Collapse
Affiliation(s)
- Colin P Sperring
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Michael G Argenziano
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - William M Savage
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Damian E Teasley
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Pavan S Upadhyayula
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Nathan J Winans
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center/NY-Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
42
|
Chronic convection-enhanced intratumoural delivery of chemotherapy for glioblastoma. Lancet Oncol 2022; 23:1347-1348. [DOI: 10.1016/s1470-2045(22)00626-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022]
|