1
|
França Velo A, Giantini Larsen A, Humm JL, Zanzonico P, Haque S, Souweidane M, Pandit-Taskar N. Determination of the Intralesional Distribution of Theranostic 124I-Omburtamab Convection-Enhanced Delivery in Treatment of Diffuse Intrinsic Pontine Glioma. J Nucl Med 2025:jnumed.124.267995. [PMID: 40306974 DOI: 10.2967/jnumed.124.267995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 04/08/2025] [Indexed: 05/02/2025] Open
Abstract
This phase 1, dose-escalation study examined the use of the radiolabeled antibody 124I-Omburtamab delivered directly to brain-stem tumors via convection-enhanced delivery (CED). CED bypasses the blood-brain barrier by injecting the agent under the pressure of a peristaltic pump to convectively drive the therapeutic agent through the brain tissue and tumor compartment, enabling high concentrations at the target site. We evaluated 124I-Omburtamab's potential to deliver therapeutic radiation doses to diffuse intrinsic pontine glioma in children. PET and MRI were used to assess the alignment between the intended and actual distribution of the agent, with an analysis of tumor coverage and radiation absorbed doses. Methods: 124I-Omburtamab doses ranging from 9.25 to 370 MBq were administered to 36 patients. Tumor distribution volumes were derived from PET images by placing volumes of interest over lesions and overlaying them onto T2-weighted fluid-attenuated inversion recovery MRI-delineated tumor volumes. Dosimetry metrics evaluated after CED included dose-volume histograms, tumor coverage percentage, and the Dice similarity coefficient between the antibody distribution and tumor volume. A 4-quadrant scatter plot of Dice similarity versus tumor coverage was used to classify treatment variations among patients. Results: Serial PET scans showed 124I-Omburtamab localization in brain-stem lesions from 1 h to 7 d ± 1 d after dose administration. Coverage analysis revealed that 29 patients had tumor volume coverage greater than 50%, and 28 had a Dice similarity coefficient over 50%. The 4-quadrant statistical analysis-percentage of coverage versus Dice similarity coefficient-showed that 27 patients had acceptable coverage for treatment, and 4 patients experiencing suboptimal tumor coverage. Conclusion: CED of 124I-Omburtamab is a novel approach for delivering radiolabeled therapies into brain-stem tumors. Imaging enabled quantification of radiation dose coverage within the MRI-defined tumor target, highlighting the importance of precise alignment between therapeutic agent distribution and tumor volume.
Collapse
Affiliation(s)
- Alexandre França Velo
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexandra Giantini Larsen
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Neurological Surgery, Weil Medical College of Cornell University, New York, New York
| | - John L Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York;
| | - Pat Zanzonico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sofia Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Mark Souweidane
- Department of Neurological Surgery, Weil Medical College of Cornell University, New York, New York
| | - Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; and
- Weil Medical College of Cornell University, New York, New York
| |
Collapse
|
2
|
Stockwell CA, Thang M, Kram DE, Satterlee AB, Hingtgen S. Therapeutic approaches for targeting the pediatric brain tumor microenvironment. Drug Deliv Transl Res 2025:10.1007/s13346-025-01839-3. [PMID: 40257744 DOI: 10.1007/s13346-025-01839-3] [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] [Accepted: 03/14/2025] [Indexed: 04/22/2025]
Abstract
Central nervous system (CNS) tumors are the most frequent solid malignant tumors in pediatric patients and are the leading cause of tumor-related death in children. Treatment for this heterogeneous group of tumors consists of various combinations of safe maximal surgical resection, chemotherapy, and radiation therapy which offer a cure for some children but often cause debilitating adverse late effects in others. While therapies targeting the tumor microenvironment (TME) like immune checkpoint inhibition (ICI) have been successful in treating some cancers, these therapies failed to exhibit treatment efficacy in the majority of pediatric brain tumors in the clinic. Importantly, the pediatric TME is unique and distinct from adult brain tumors and designing therapies to effectively target these tumors requires understanding the unique biology of pediatric brain tumors and the use of translational models that recapitulate the TME. Here we describe the TME of medulloblastoma (MB) and diffuse midline glioma (DMG), specifically diffuse intrinsic pontine glioma (DIPG), and further present the current drug delivery approaches and clinical administration routes targeting the TME in these tumors, including preclinical and clinical studies.
Collapse
Affiliation(s)
- Caroline A Stockwell
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Morrent Thang
- Neuroscience Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David E Kram
- Division of Pediatric Hematology-Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew B Satterlee
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Eshelman Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shawn Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Gallitto M, Zhang X, De Los Santos G, Wei HJ, Fernández EC, Duan S, Sedor G, Yoh N, Kokossis D, Angel JC, Wang YF, White E, Kinslow CJ, Berg X, Tomassoni L, Zandkarimi F, Chio IIC, Canoll P, Bruce JN, Feldstein NA, Gartrell RD, Cheng SK, Garvin JH, Zacharoulis S, Wechsler-Reya RJ, Pavisic J, Califano A, Zhang Z, Wu CC. Targeted delivery of napabucasin with radiotherapy improves outcomes in diffuse midline glioma. Neuro Oncol 2025; 27:795-810. [PMID: 39394920 PMCID: PMC11889722 DOI: 10.1093/neuonc/noae215] [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: 04/30/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Diffuse midline glioma (DMG) is the most aggressive primary brain tumor in children. All previous studies examining the role of systemic agents have failed to demonstrate a survival benefit; the only standard of care is radiation therapy (RT). Successful implementation of radiosensitization strategies in DMG remains an essential and promising avenue of investigation. We explore the use of Napabucasin, an NAD(P)H quinone dehydrogenase 1 (NQO1)-bioactivatable reactive oxygen species (ROS)-inducer, as a potential therapeutic radiosensitizer in DMG. METHODS In this study, we conduct in vitro and in vivo assays using patient-derived DMG cultures to elucidate the mechanism of action of Napabucasin and its radiosensitizing properties. As penetration of systemic therapy through the blood-brain barrier (BBB) is a significant limitation to the success of DMG therapies, we explore focused ultrasound (FUS) and convection-enhanced delivery (CED) to overcome the BBB and maximize therapeutic efficacy. RESULTS Napabucasin is a potent ROS-inducer and radiosensitizer in DMG, and treatment-mediated ROS production and cytotoxicity are dependent on NQO1. In subcutaneous xenograft models, combination therapy with RT improves local control. After optimizing targeted drug delivery using CED in an orthotopic mouse model, we establish the novel feasibility and survival benefit of CED of Napabucasin concurrent with RT. CONCLUSIONS As nearly all DMG patients will receive RT as part of their treatment course, our validation of the efficacy of radiosensitizing therapy using CED to prolong survival in DMG opens the door for exciting novel studies of alternative radiosensitization strategies in this devastating disease while overcoming limitations of the BBB.
Collapse
Affiliation(s)
- Matthew Gallitto
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Xu Zhang
- Department of Genetics and Development, Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York, USA
| | - Genesis De Los Santos
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Hong-Jian Wei
- Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Roanoke, 24016, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Ester Calvo Fernández
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shoufu Duan
- Department of Genetics and Development, Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York, USA
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Geoffrey Sedor
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Nina Yoh
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Danae Kokossis
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - J Carlos Angel
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, New York, USA
- Department of Biomedical Informatics, Columbia University, New York, New York, USA
| | - Yi-Fang Wang
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Erin White
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Connor J Kinslow
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Xander Berg
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Lorenzo Tomassoni
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York, USA
- DarwinHealth Inc., New York, NYUSA
| | | | - Iok In Christine Chio
- Department of Genetics and Development, Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Neil A Feldstein
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Robyn D Gartrell
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Simon K Cheng
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - James H Garvin
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Stergios Zacharoulis
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Robert J Wechsler-Reya
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Jovana Pavisic
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrea Califano
- Chan Zuckerberg Biohub New York, New York, New York, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA
- Department of Medicine, Columbia University, New York, New YorkUSA
- Department of Biomedical Informatics, Columbia University, New York, New York, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Zhiguo Zhang
- Department of Genetics and Development, Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York, USA
| | - Cheng-Chia Wu
- Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Roanoke, 24016, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, 24061, USA
- The Brain Tumor Institute, Children's National Hospital, Washington, 20010, USA
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, 20010, USA
| |
Collapse
|
4
|
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; 172:219-227. [PMID: 39760796 PMCID: PMC11832582 DOI: 10.1007/s11060-024-04904-y] [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: 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
|
5
|
Pandit-Taskar N, Zanzonico PB, Grkovski M, Donzelli M, Vietri SM, Horan C, Serencsits B, Prasad K, Lyashchenko S, Kramer K, Dunkel IJ, Souweidane MM. Theranostic Intratumoral Convection-Enhanced Delivery of 124I-Omburtamab in Patients with Diffuse Intrinsic Pontine Glioma: Pharmacokinetics and Lesion Dosimetry. J Nucl Med 2024; 65:1364-1370. [PMID: 39142829 PMCID: PMC11372263 DOI: 10.2967/jnumed.123.266365] [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: 03/13/2024] [Accepted: 07/10/2024] [Indexed: 08/16/2024] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a rare childhood malignancy with poor prognosis. There are no effective treatment options other than external beam therapy. We conducted a pilot, first-in-human study using 124I-omburtamab imaging and theranostics as a therapeutic approach using a localized convection-enhanced delivery (CED) technique for administering radiolabeled antibody. We report the detailed pharmacokinetics and dosimetry results of intratumoral delivery of 124I-omburtamab. Methods: Forty-five DIPG patients who received 9.0-370.7 MBq of 124I-omburtamab intratumorally via CED underwent serial brain and whole-body PET/CT imaging at 3-5 time points after injection within 4, 24-48, 72-96, 120-144, and 168-240 h from the end of infusion. Serial blood samples were obtained for kinetic analysis. Whole-body, blood, lesion, and normal-tissue activities were measured, kinetic parameters (uptake and clearance half-life times) estimated, and radiation-absorbed doses calculated using the OLINDA software program. Results: All patients showed prominent activity within the lesion that was retained over several days and was detectable up to the last time point of imaging, with a mean 124I residence time in the lesion of 24.9 h and dose equivalent of 353 ± 181 mSv/MBq. Whole-body doses were low, with a dose equivalent of 0.69 ± 0.28 mSv/MBq. Systemic distribution and activities in normal organs and blood were low. Radiation dose to blood was very low, with a mean value of 0.27 ± 0.21 mGy/MBq. Whole-body clearance was monoexponential with a mean biologic half-life of 62.7 h and an effective half-life of 37.9 h. Blood clearance was biexponential, with a mean biologic half-life of 22.2 h for the rapid α phase and 155 h for the slower β phase. Conclusion: Intratumoral CED of 124I-omburtamab is a novel theranostics approach in DIPG. It allows for delivery of high radiation doses to the DIPG lesions, with high lesion activities and low systemic activities and high tumor-to-normal-tissue ratios and achieving a wide safety margin. Imaging of the actual therapeutic administration of 124I-omburtamab allows for direct estimation of the therapeutic lesion and normal-tissue-absorbed doses.
Collapse
Affiliation(s)
- Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Pat B Zanzonico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Milan Grkovski
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria Donzelli
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Scott M Vietri
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Radiochemistry & Molecular Imaging Probe Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christopher Horan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian Serencsits
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kavya Prasad
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Serge Lyashchenko
- Radiochemistry & Molecular Imaging Probe Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pediatrics, Weill Cornell Medical College, New York, New York
| | - Ira J Dunkel
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pediatrics, Weill Cornell Medical College, New York, New York
| | - Mark M Souweidane
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York; and
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York
| |
Collapse
|
6
|
de Camargo Magalhães ES, de Bont ESJM, Bruggeman SWM, Lima FRS. Targeting the reprogrammed metabolism in H3.3K27M pediatric high-grade gliomas. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167248. [PMID: 38777100 DOI: 10.1016/j.bbadis.2024.167248] [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: 02/23/2024] [Revised: 04/22/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Recent studies in Diffuse Midline Gliomas (DMG) demonstrated a strong connection between epigenome dysregulation and metabolic rewiring. Here, we evaluated the value of targeting a glycolytic protein named Phosphofructo-2-kinase/Fructose-2,6-bisphosphatase 3 (PFKFB3) in H3.3K27M DMG. We observed that the viability of H3.3K27M cells is dramatically reduced by PFK15, a potent inhibitor of PFKFB3. Furthermore, PFKFB3 inhibition induced apoptosis and G2/M arrest. Interestingly, CRISPR-Knockout of the K27M mutant allele has a synergistic effect on the observed phenotype. Altogether, we identified PFKFB3 as a new target for H3.3K27M DMG, making PFK15 a potential candidate for future animal studies and clinical trials.
Collapse
Affiliation(s)
- Eduardo S de Camargo Magalhães
- Department of Ageing Biology/ERIBA, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Glial Cell Biology Laboratory, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eveline S J M de Bont
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Division of Pediatric Oncology/Hematology, Groningen, the Netherlands
| | - Sophia W M Bruggeman
- Department of Ageing Biology/ERIBA, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Flavia R S Lima
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
7
|
Nonnenbroich LF, Bouchal SM, Millesi E, Rechberger JS, Khatua S, Daniels DJ. H3K27-Altered Diffuse Midline Glioma of the Brainstem: From Molecular Mechanisms to Targeted Interventions. Cells 2024; 13:1122. [PMID: 38994974 PMCID: PMC11240752 DOI: 10.3390/cells13131122] [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: 06/03/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024] Open
Abstract
Pediatric high-grade gliomas are a devastating subset of brain tumors, characterized by their aggressive pathophysiology and limited treatment options. Among them, H3 K27-altered diffuse midline gliomas (DMG) of the brainstem stand out due to their distinct molecular features and dismal prognosis. Recent advances in molecular profiling techniques have unveiled the critical role of H3 K27 alterations, particularly a lysine-to-methionine mutation on position 27 (K27M) of the histone H3 tail, in the pathogenesis of DMG. These mutations result in epigenetic dysregulation, which leads to altered chromatin structure and gene expression patterns in DMG tumor cells, ultimately contributing to the aggressive phenotype of DMG. The exploration of targeted therapeutic avenues for DMG has gained momentum in recent years. Therapies, including epigenetic modifiers, kinase inhibitors, and immunotherapies, are under active investigation; these approaches aim to disrupt aberrant signaling cascades and overcome the various mechanisms of therapeutic resistance in DMG. Challenges, including blood-brain barrier penetration and DMG tumor heterogeneity, require innovative approaches to improve drug delivery and personalized treatment strategies. This review aims to provide a comprehensive overview of the evolving understanding of DMG, focusing on the intricate molecular mechanisms driving tumorigenesis/tumor progression and the current landscape of emerging targeted interventions.
Collapse
Affiliation(s)
- Leo F. Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (L.F.N.); (J.S.R.)
- Hopp Children’s Cancer Center, Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - Samantha M. Bouchal
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (L.F.N.); (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Elena Millesi
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Research Laboratory of the Division of Plastic and Reconstructive Surgery, Department of Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Julian S. Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (L.F.N.); (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Soumen Khatua
- Department of Pediatric Hematology/Oncology, Section of Neuro-Oncology, Mayo Clinic, Rochester, MN 55905, USA;
| | - David J. Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (L.F.N.); (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
8
|
Arms LM, Duchatel RJ, Jackson ER, Sobrinho PG, Dun MD, Hua S. Current status and advances to improving drug delivery in diffuse intrinsic pontine glioma. J Control Release 2024; 370:835-865. [PMID: 38744345 DOI: 10.1016/j.jconrel.2024.05.018] [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: 12/05/2023] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
Diffuse midline glioma (DMG), including tumors diagnosed in the brainstem (diffuse intrinsic pontine glioma - DIPG), is the primary cause of brain tumor-related death in pediatric patients. DIPG is characterized by a median survival of <12 months from diagnosis, harboring the worst 5-year survival rate of any cancer. Corticosteroids and radiation are the mainstay of therapy; however, they only provide transient relief from the devastating neurological symptoms. Numerous therapies have been investigated for DIPG, but the majority have been unsuccessful in demonstrating a survival benefit beyond radiation alone. Although many barriers hinder brain drug delivery in DIPG, one of the most significant challenges is the blood-brain barrier (BBB). Therapeutic compounds must possess specific properties to enable efficient passage across the BBB. In brain cancer, the BBB is referred to as the blood-brain tumor barrier (BBTB), where tumors disrupt the structure and function of the BBB, which may provide opportunities for drug delivery. However, the biological characteristics of the brainstem's BBB/BBTB, both under normal physiological conditions and in response to DIPG, are poorly understood, which further complicates treatment. Better characterization of the changes that occur in the BBB/BBTB of DIPG patients is essential, as this informs future treatment strategies. Many novel drug delivery technologies have been investigated to bypass or disrupt the BBB/BBTB, including convection enhanced delivery, focused ultrasound, nanoparticle-mediated delivery, and intranasal delivery, all of which are yet to be clinically established for the treatment of DIPG. Herein, we review what is known about the BBB/BBTB and discuss the current status, limitations, and advances of conventional and novel treatments to improving brain drug delivery in DIPG.
Collapse
Affiliation(s)
- Lauren M Arms
- Therapeutic Targeting Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Ryan J Duchatel
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Evangeline R Jackson
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Pedro Garcia Sobrinho
- Therapeutic Targeting Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Matthew D Dun
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Susan Hua
- Therapeutic Targeting Research Group, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine & Wellbeing, University of Newcastle, Callaghan, NSW, Australia.
| |
Collapse
|
9
|
Tosi U, Souweidane M. Diffuse Midline Gliomas: Challenges and New Strategies in a Changing Clinical Landscape. Cancers (Basel) 2024; 16:219. [PMID: 38201646 PMCID: PMC10778507 DOI: 10.3390/cancers16010219] [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: 12/14/2023] [Revised: 12/29/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) was first described by Harvey Cushing, the father of modern neurosurgery, a century ago. Since then, the classification of this tumor changed significantly, as it is now part of the broader family of diffuse midline gliomas (DMGs), a heterogeneous group of tumors of midline structures encompassing the entire rostro-caudal space, from the thalamus to the spinal cord. DMGs are characterized by various epigenetic events that lead to chromatin remodeling similarities, as two decades of studies made possible by increased tissue availability showed. This new understanding of tumor (epi)biology is now driving novel clinical trials that rely on targeted agents, with finally real hopes for a change in an otherwise unforgiving prognosis. This biological discovery is being paralleled with equally exciting work in therapeutic drug delivery. Invasive and noninvasive platforms have been central to early phase clinical trials with a promising safety track record and anecdotal benefits in outcome.
Collapse
Affiliation(s)
- Umberto Tosi
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Neurological Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mark Souweidane
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Neurological Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
10
|
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
|
11
|
Shao X, Saito R, Sato A, Okuno S, Saigusa D, Saito R, Uruno A, Osada Y, Kanamori M, Tominaga T. Local Delivery of Nimustine Hydrochloride against Brain Tumors: Basic Characterization Study. TOHOKU J EXP MED 2023; 261:187-194. [PMID: 37635063 DOI: 10.1620/tjem.2023.j069] [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] [Indexed: 08/29/2023]
Abstract
Convection-enhanced delivery (CED) delivers agents directly into tumors and the surrounding parenchyma. Although a promising concept, clinical applications are often hampered by insufficient treatment efficacy. Toward developing an effective CED-based strategy for delivering drugs with proven clinical efficacy, we performed a basic characterization study to explore the locally delivered characteristics of the water soluble nitrosourea nimustine hydrochloride (ACNU). First, ACNU distribution after CED in rodent brain was studied using mass spectrometry imaging. Clearance of 14C-labeled ACNU after CED in striatum was also studied. ACNU was robustly distributed in rodent brain similar to the distribution of the hydrophilic dye Evans blue after CED, and locally delivered ACNU was observed for over 24 h at the delivery site. Subsequently, to investigate the potential of ACNU to induce an immunostimulative microenvironment, Fas and transforming growth factor-β1 (TGF-β1) was assessed in vitro. We found that ACNU significantly inhibited TGF-β1 secretion and reduced Fas expression. Further, after CED of ACNU in 9L-derived intracranial tumors, the infiltration of CD4/CD8 lymphocytes in tumors was evaluated by immunofluorescence.CED of ACNU in xenografted intracranial tumors induced tumor infiltration of CD4/CD8 lymphocytes. ACNU has a robust distribution in rodent brain by CED, and delayed clearance of the drug was observed at the local infusion site. Further, local delivery of ACNU affects the tumor microenvironment and induces immune cell migration in tumor. These characteristics make ACNU a promising agent for CED.
Collapse
Affiliation(s)
- Xiaodong Shao
- Department of Neurosurgery, Tohoku University Graduate School of Medicine
| | - Ryuta Saito
- Department of Neurosurgery, Tohoku University Graduate School of Medicine
- Department of Neurosurgery, Nagoya University Graduate School of Medicine
| | - Aya Sato
- Department of Neurosurgery, Tohoku University Graduate School of Medicine
| | - Saori Okuno
- Department of Neurosurgery, Tohoku University Graduate School of Medicine
| | - Daisuke Saigusa
- Faculty of Pharma-Science, Teikyo University
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University
| | - Ritsumi Saito
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University
| | - Akira Uruno
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University
| | - Yoshinari Osada
- Department of Neurosurgery, Tohoku University Graduate School of Medicine
| | - Masayuki Kanamori
- Department of Neurosurgery, Tohoku University Graduate School of Medicine
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine
| |
Collapse
|
12
|
Noon A, Galban S. Therapeutic avenues for targeting treatment challenges of diffuse midline gliomas. Neoplasia 2023; 40:100899. [PMID: 37030112 PMCID: PMC10119952 DOI: 10.1016/j.neo.2023.100899] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
Diffuse midline glioma (DMG) is the leading cause of brain tumor-related deaths in children. DMG typically presents with variable neurologic symptoms between ages 3 and 10. Currently, radiation remains the standard therapy for DMG to halt progression and reduce tumor bulk to minimize symptoms. However, tumors recur in almost 100% of patients and thus, DMG is still considered an incurable cancer with a median survival of 9-12 months. Surgery is generally contraindicated due to the delicate organization of the brainstem, where DMG is located. Despite extensive research efforts, no chemotherapeutic agents, immune therapies, or molecularly targeted therapies have been approved to provide survival benefit. Furthermore, the efficacy of therapies is limited by poor blood-brain barrier penetration and inherent resistance mechanisms of the tumor. However, novel drug delivery approaches, along with recent advances in molecularly targeted therapies and immunotherapies, have advanced to clinical trials and may provide viable future treatment options for DMG patients. This review seeks to evaluate current therapeutics at the preclinical stage and those that have advanced to clinical trials and to discuss the challenges of drug delivery and inherent resistance to these therapies.
Collapse
Affiliation(s)
- Aleeha Noon
- College of Medicine, California Northstate University, 9700 W Taron Drive, Elk Grove, CA 95757, USA
| | - Stefanie Galban
- Center for Molecular Imaging, The University of Michigan Medical School, BSRB A502, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA; Department of Radiology, The University of Michigan Medical School, BSRB A502, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA; Rogel Cancer Center, The University of Michigan Medical School, 1500 E Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
13
|
Straehla JP, Reardon DA, Wen PY, Agar NYR. The Blood-Brain Barrier: Implications for Experimental Cancer Therapeutics. ANNUAL REVIEW OF CANCER BIOLOGY 2023; 7:265-289. [PMID: 38323268 PMCID: PMC10846865 DOI: 10.1146/annurev-cancerbio-061421-040433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
The blood-brain barrier is critically important for the treatment of both primary and metastatic cancers of the central nervous system (CNS). Clinical outcomes for patients with primary CNS tumors are poor and have not significantly improved in decades. As treatments for patients with extracranial solid tumors improve, the incidence of CNS metastases is on the rise due to suboptimal CNS exposure of otherwise systemically active agents. Despite state-of-the art surgical care and increasingly precise radiation therapy, clinical progress is limited by the ability to deliver an effective dose of a therapeutic agent to all cancerous cells. Given the tremendous heterogeneity of CNS cancers, both across cancer subtypes and within a single tumor, and the range of diverse therapies under investigation, a nuanced examination of CNS drug exposure is needed. With a shared goal, common vocabulary, and interdisciplinary collaboration, the field is poised for renewed progress in the treatment of CNS cancers.
Collapse
Affiliation(s)
- Joelle P Straehla
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Szychot E, Bhagawati D, Sokolska MJ, Walker D, Gill S, Hyare H. Evaluating drug distribution in children and young adults with diffuse midline glioma of the pons (DIPG) treated with convection-enhanced drug delivery. FRONTIERS IN NEUROIMAGING 2023; 2:1062493. [PMID: 37554653 PMCID: PMC10406269 DOI: 10.3389/fnimg.2023.1062493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/08/2023] [Indexed: 08/10/2023]
Abstract
AIMS To determine an imaging protocol that can be used to assess the distribution of infusate in children with DIPG treated with CED. METHODS 13 children diagnosed with DIPG received between 3.8 and 5.7 ml of infusate, through two pairs of catheters to encompass tumor volume on day 1 of cycle one of treatment. Volumetric T2-weighted (T2W) and diffusion-weighted MRI imaging (DWI) were performed before and after day 1 of CED. Apparent diffusion coefficient (ADC) maps were calculated. The tumor volume pre and post CED was automatically segmented on T2W and ADC on the basis of signal intensity. The ADC maps pre and post infusion were aligned and subtracted to visualize the infusate distribution. RESULTS There was a significant increase (p < 0.001) in mean ADC and T2W signal intensity (SI) ratio and a significant (p < 0.001) increase in mean tumor volume defined by ADC and T2W SI post infusion (mean ADC volume pre: 19.8 ml, post: 24.4 ml; mean T2W volume pre: 19.4 ml, post: 23.4 ml). A significant correlation (p < 0.001) between infusate volume and difference in ADC/T2W SI defined tumor volume was observed (ADC, r = 0.76; T2W, r = 0.70). Finally, pixel-by-pixel subtraction of the ADC maps pre and post infusion demonstrated a volume of high signal intensity, presumed infusate distribution. CONCLUSIONS ADC and T2W MRI are proposed as a combined parameter method for evaluation of CED infusate distribution in brainstem tumors in future clinical trials.
Collapse
Affiliation(s)
- Elwira Szychot
- Department of Paediatric Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
- Department of Paediatric Oncology, Harley Street Children's Hospital, London, United Kingdom
- Department of Paediatrics, Paediatric Oncology and Immunology, Pomeranian Medical University, Szczecin, Poland
| | - Dolin Bhagawati
- Department of Paediatric Oncology, Harley Street Children's Hospital, London, United Kingdom
- Department of Neurosurgery, Charing Cross Hospital, Imperial College, London, United Kingdom
| | - Magdalena Joanna Sokolska
- Department of Medical Physics and Biomedical Engineering, Faculty of Engineering Sciences, University College London, London, United Kingdom
| | - David Walker
- Department of Paediatric Oncology, Harley Street Children's Hospital, London, United Kingdom
- Division of Child Health, School of Human Development, University of Nottingham, Nottingham, United Kingdom
| | - Steven Gill
- Department of Paediatric Oncology, Harley Street Children's Hospital, London, United Kingdom
- Department of Translational Health Sciences, Institute of Clinical Neurosciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Harpreet Hyare
- Department of Paediatric Oncology, Harley Street Children's Hospital, London, United Kingdom
- Department of Neuroradiology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
15
|
Tyrosine Kinase Inhibitors for Glioblastoma Multiforme: Challenges and Opportunities for Drug Delivery. Pharmaceutics 2022; 15:pharmaceutics15010059. [PMID: 36678688 PMCID: PMC9863099 DOI: 10.3390/pharmaceutics15010059] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain tumor with high mortality rates. Due to its invasiveness, heterogeneity, and incomplete resection, the treatment is very challenging. Targeted therapies such as tyrosine kinase inhibitors (TKIs) have great potential for GBM treatment, however, their efficacy is primarily limited by poor brain distribution due to the presence of the blood-brain barrier (BBB). This review focuses on the potential of TKIs in GBM therapy and provides an insight into the reasons behind unsuccessful clinical trials of TKIs in GBM despite the success in treating other cancer types. The main section is dedicated to the use of promising drug delivery strategies for targeted delivery to brain tumors. Use of brain targeted delivery strategies can help enhance the efficacy of TKIs in GBM. Among various drug delivery approaches used to bypass or cross BBB, utilizing nanocarriers is a promising strategy to augment the pharmacokinetic properties of TKIs and overcome their limitations. This is because of their advantages such as the ability to cross BBB, chemical stabilization of drug in circulation, passive or active targeting of tumor, modulation of drug release from the carrier, and the possibility to be delivered via non-invasive intranasal route.
Collapse
|
16
|
Di Ruscio V, Del Baldo G, Fabozzi F, Vinci M, Cacchione A, de Billy E, Megaro G, Carai A, Mastronuzzi A. Pediatric Diffuse Midline Gliomas: An Unfinished Puzzle. Diagnostics (Basel) 2022; 12:2064. [PMID: 36140466 PMCID: PMC9497626 DOI: 10.3390/diagnostics12092064] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/15/2022] Open
Abstract
Diffuse midline glioma (DMG) is a heterogeneous group of aggressive pediatric brain tumors with a fatal prognosis. The biological hallmark in the major part of the cases is H3K27 alteration. Prognosis remains poor, with median survival ranging from 9 to 12 months from diagnosis. Clinical and radiological prognostic factors only partially change the progression-free survival but they do not improve the overall survival. Despite efforts, there is currently no curative therapy for DMG. Radiotherapy remains the standard treatment with only transitory benefits. No chemotherapeutic regimens were found to significantly improve the prognosis. In the new era of a deeper integration between histological and molecular findings, potential new approaches are currently under investigation. The entire international scientific community is trying to target DMG on different aspects. The therapeutic strategies involve targeting epigenetic alterations, such as methylation and acetylation status, as well as identifying new molecular pathways that regulate oncogenic proliferation; immunotherapy approaches too are an interesting point of research in the oncology field, and the possibility of driving the immune system against tumor cells has currently been evaluated in several clinical trials, with promising preliminary results. Moreover, thanks to nanotechnology amelioration, the development of innovative delivery approaches to overcross a hostile tumor microenvironment and an almost intact blood-brain barrier could potentially change tumor responses to different treatments. In this review, we provide a comprehensive overview of available and potential new treatments that are worldwide under investigation, with the intent that patient- and tumor-specific treatment could change the biological inauspicious history of this disease.
Collapse
Affiliation(s)
- Valentina Di Ruscio
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Giada Del Baldo
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Francesco Fabozzi
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
- Department of Pediatrics, University of Rome Tor Vergata, 00165 Rome, Italy
| | - Maria Vinci
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Antonella Cacchione
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Emmanuel de Billy
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Giacomina Megaro
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Andrea Carai
- Neurosurgery Unit, Department of Neurosciences, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Angela Mastronuzzi
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
- Faculty of Medicine and Surgery, Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| |
Collapse
|
17
|
Lambride C, Vavourakis V, Stylianopoulos T. Convection-Enhanced Delivery In Silico Study for Brain Cancer Treatment. Front Bioeng Biotechnol 2022; 10:867552. [PMID: 35694227 PMCID: PMC9177080 DOI: 10.3389/fbioe.2022.867552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/02/2022] [Indexed: 12/02/2022] Open
Abstract
Brain cancer therapy remains a formidable challenge in oncology. Convection-enhanced delivery (CED) is an innovative and promising local drug delivery method for the treatment of brain cancer, overcoming the challenges of the systemic delivery of drugs to the brain. To improve our understanding about CED efficacy and drug transport, we present an in silico methodology for brain cancer CED treatment simulation. To achieve this, a three-dimensional finite element formulation is utilized which employs a brain model representation from clinical imaging data and is used to predict the drug deposition in CED regimes. The model encompasses biofluid dynamics and the transport of drugs in the brain parenchyma. Drug distribution is studied under various patho-physiological conditions of the tumor, in terms of tumor vessel wall pore size and tumor tissue hydraulic conductivity as well as for drugs of various sizes, spanning from small molecules to nanoparticles. Through a parametric study, our contribution reports the impact of the size of the vascular wall pores and that of the therapeutic agent on drug distribution during and after CED. The in silico findings provide useful insights of the spatio-temporal distribution and average drug concentration in the tumor towards an effective treatment of brain cancer.
Collapse
Affiliation(s)
- Chryso Lambride
- Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Vasileios Vavourakis
- Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
- *Correspondence: Vasileios Vavourakis, ; Triantafyllos Stylianopoulos,
| | - Triantafyllos Stylianopoulos
- Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
- *Correspondence: Vasileios Vavourakis, ; Triantafyllos Stylianopoulos,
| |
Collapse
|
18
|
Aquilina K, Chakrapani A, Carr L, Kurian MA, Hargrave D. Convection-Enhanced Delivery in Children: Techniques and Applications. Adv Tech Stand Neurosurg 2022; 45:199-228. [PMID: 35976451 DOI: 10.1007/978-3-030-99166-1_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Since its first description in 1994, convection-enhanced delivery (CED) has become a reliable method of administering drugs directly into the brain parenchyma. More predictable and effective than simple diffusion, CED bypasses the challenging boundary of the blood brain barrier, which has frustrated many attempts at delivering large molecules or polymers into the brain parenchyma. Although most of the clinical work with CED has been carried out on adults with incurable neoplasms, principally glioblastoma multiforme, an increasing number of studies have recognized its potential for paediatric applications, which now include treatment of currently incurable brain tumours such as diffuse intrinsic pontine glioma (DIPG), as well as metabolic and neurotransmitter diseases. The roadmap for the development of hardware and use of pharmacological agents in CED has been well-established, and some neurosurgical centres throughout the world have successfully undertaken clinical trials, admittedly mostly early phase, on the basis of in vitro, small animal and large animal pre-clinical foundations. However, the clinical efficacy of CED, although theoretically logical, has yet to be unequivocally demonstrated in a clinical trial; this applies particularly to neuro-oncology.This review aims to provide a broad description of the current knowledge of CED as applied to children. It reviews published studies of paediatric CED in the context of its wider history and developments and underlines the challenges related to the development of hardware, the selection of pharmacological agents, and gene therapy. It also reviews the difficulties related to the development of clinical trials involving CED and looks towards its potential disease-modifying opportunities in the future.
Collapse
Affiliation(s)
- K Aquilina
- Department of Neurosurgery, Great Ormond Street Hospital, London, UK.
| | - A Chakrapani
- Department of Metabolic Medicine, Great Ormond Street Hospital, London, UK
| | - L Carr
- Department of Neurology and Neurodisability, Great Ormond Street Hospital, London, UK
| | - M A Kurian
- Department of Neurology and Neurodisability, Great Ormond Street Hospital, London, UK
- Neurogenetics Group, Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL-Great Ormond Street Institute of Child Health, London, UK
| | - D Hargrave
- Cancer Group, UCL-Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
19
|
Childhood Malignant Brain Tumors: Balancing the Bench and Bedside. Cancers (Basel) 2021; 13:cancers13236099. [PMID: 34885207 PMCID: PMC8656510 DOI: 10.3390/cancers13236099] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 01/28/2023] Open
Abstract
Simple Summary Brain tumors remain the most common childhood solid tumors, accounting for approximately 25% of all pediatric cancers. They also represent the most common cause of cancer-related illness and death in this age group. Recent years have witnessed an evolution in our understanding of the biological underpinnings of many childhood brain tumors, potentially improving survival through both improved risk group allocation for patients to provide appropriate treatment intensity, and novel therapeutic breakthroughs. This review aims to summarize the molecular landscape, current trial-based standards of care, novel treatments being explored and future challenges for the three most common childhood malignant brain tumors—medulloblastomas, high-grade gliomas and ependymomas. Abstract Brain tumors are the leading cause of childhood cancer deaths in developed countries. They also represent the most common solid tumor in this age group, accounting for approximately one-quarter of all pediatric cancers. Developments in neuro-imaging, neurosurgical techniques, adjuvant therapy and supportive care have improved survival rates for certain tumors, allowing a future focus on optimizing cure, whilst minimizing long-term adverse effects. Recent times have witnessed a rapid evolution in the molecular characterization of several of the common pediatric brain tumors, allowing unique clinical and biological patient subgroups to be identified. However, a resulting paradigm shift in both translational therapy and subsequent survival for many of these tumors remains elusive, while recurrence remains a great clinical challenge. This review will provide an insight into the key molecular developments and global co-operative trial results for the most common malignant pediatric brain tumors (medulloblastoma, high-grade gliomas and ependymoma), highlighting potential future directions for management, including novel therapeutic options, and critical challenges that remain unsolved.
Collapse
|
20
|
Zagotto G, Bortoli M. Drug Design: Where We Are and Future Prospects. Molecules 2021; 26:7061. [PMID: 34834152 PMCID: PMC8622624 DOI: 10.3390/molecules26227061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 11/24/2022] Open
Abstract
Medicinal chemistry is facing new challenges in approaching precision medicine. Several powerful new tools or improvements of already used tools are now available to medicinal chemists to help in the process of drug discovery, from a hit molecule to a clinically used drug. Among the new tools, the possibility of considering folding intermediates or the catalytic process of a protein as a target for discovering new hits has emerged. In addition, machine learning is a new valuable approach helping medicinal chemists to discover new hits. Other abilities, ranging from the better understanding of the time evolution of biochemical processes to the comprehension of the biological meaning of the data originated from genetic analyses, are on their way to progress further in the drug discovery field toward improved patient care. In this sense, the new approaches to the delivery of drugs targeted to the central nervous system, together with the advancements in understanding the metabolic pathways for a growing number of drugs and relating them to the genetic characteristics of patients, constitute important progress in the field.
Collapse
Affiliation(s)
- Giuseppe Zagotto
- Department of Pharmaceutical Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Marco Bortoli
- Institute of Computational Chemistry and Catalysis (IQCC) and Department of Chemistry, Faculty of Sciences, University of Girona, C/M. A. Capmany 69, 17003 Girona, Spain;
| |
Collapse
|
21
|
Chatwin HV, Cruz Cruz J, Green AL. Pediatric high-grade glioma: moving toward subtype-specific multimodal therapy. FEBS J 2021; 288:6127-6141. [PMID: 33523591 DOI: 10.1111/febs.15739] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022]
Abstract
Pediatric high-grade gliomas (pHGG) comprise a deadly, heterogenous category of pediatric gliomas with a clear need for more effective treatment options. Advances in high-throughput molecular techniques have enhanced molecular understanding of these tumors, but outcomes are still poor, and treatments beyond resection and radiation have not yet been clearly established as standard of care. In this review, we first discuss the history of treatment approaches to pHGG to this point. We then review four distinct categories of pHGG, including histone 3-mutant, IDH-mutant, histone 3/IDH-wildtype, and radiation-induced pHGG. We discuss the molecular understanding of each subgroup and targeted treatment options in development. Finally, we look at the development and current status of two novel approaches to pHGG as a whole: localized convection-enhanced chemotherapy delivery and immunotherapy, including checkpoint inhibitors, vaccine therapy, and CAR-T cells. Through this review, we demonstrate the potential for rational, molecularly driven, subtype-specific therapy to be used with other novel approaches in combinations that could meaningfully improve the prognosis in pHGG.
Collapse
Affiliation(s)
- Hannah V Chatwin
- Department of Pediatrics, Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Joselyn Cruz Cruz
- Department of Pediatrics, Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Adam L Green
- Department of Pediatrics, Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine, Aurora, CO, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| |
Collapse
|