1
|
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
|
2
|
Verbeek TCAI, Vrenken KS, Arentsen-Peters STCJM, Castro PG, van de Ven M, van Tellingen O, Pieters R, Stam RW. Selective inhibition of HDAC class IIA as therapeutic intervention for KMT2A-rearranged acute lymphoblastic leukemia. Commun Biol 2024; 7:1257. [PMID: 39362994 PMCID: PMC11450098 DOI: 10.1038/s42003-024-06916-w] [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: 03/12/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024] Open
Abstract
KMT2A-rearranged acute lymphoblastic leukemia (ALL) is characterized by deregulation of the epigenome and shows susceptibility towards histone deacetylase (HDAC) inhibition. Most broad-spectrum HDAC inhibitors simultaneously target multiple human HDAC isoforms. Consequently, they often induce toxicity and especially in combination with other therapeutic agents. Therefore, more specifically targeting HDAC isoforms may represent a safer therapeutic strategy. Here we show that shRNA-mediated knock-down of the class IIA HDAC isoforms HDAC4, HDAC5, and HDAC7 results in apoptosis induction and cell cycle arrest in KMT2A-rearranged ALL cells. In concordance, the HDAC4/5 selective small molecule inhibitor LMK-235 effectively eradicates KMT2A-rearranged ALL cell lines as well as primary patient samples in vitro. However, using a xenograft mouse model of KMT2A-rearranged ALL we found that the maximum achievable dose of LMK-235 was insufficient to induce anti-leukemic effects in vivo. Similar results were obtained for the specific class IIA HDAC inhibitors MC1568 and TMP195. Finally, LMK-235 appeared to exert minimal anti-leukemic effects in vivo in combination with the BCL-2 inhibitor venetoclax, but not enough to prolong survival in treated mice. In conclusion, class IIA HDAC isoforms represent attractive therapeutic target in KMT2A-rearranged ALL, although clinical applications require the development of more stable and efficient specific HDAC inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rob Pieters
- Princess Máxima Center, Utrecht, The Netherlands
| | | |
Collapse
|
3
|
Zhang W, Oh JH, Zhang W, Rathi S, Larson JD, Wechsler-Reya RJ, Sirianni RW, Elmquist WF. Central Nervous System Distribution of Panobinostat in Preclinical Models to Guide Dosing for Pediatric Brain Tumors. J Pharmacol Exp Ther 2023; 387:315-327. [PMID: 37827699 PMCID: PMC10658912 DOI: 10.1124/jpet.123.001826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/14/2023] Open
Abstract
Achieving adequate exposure of the free therapeutic agent at the target is a critical determinant of efficacious chemotherapy. With this in mind, a major challenge in developing therapies for central nervous system (CNS) tumors is to overcome barriers to delivery, including the blood-brain barrier (BBB). Panobinostat is a nonselective pan-histone deacetylase inhibitor that is being tested in preclinical and clinical studies, including for the treatment of pediatric medulloblastoma, which has a propensity for leptomeningeal spread and diffuse midline glioma, which can infiltrate into supratentorial brain regions. In this study, we examined the rate, extent, and spatial heterogeneity of panobinostat CNS distribution in mice. Transporter-deficient mouse studies show that panobinostat is a dual substrate of P-glycoprotein (P-gp) and breast cancer resistant protein (Bcrp), which are major efflux transporters expressed at the BBB. The CNS delivery of panobinostat was moderately limited by P-gp and Bcrp, and the unbound tissue-to-plasma partition coefficient of panobinostat was 0.32 and 0.21 in the brain and spinal cord in wild-type mice. In addition, following intravenous administration, panobinostat demonstrated heterogeneous distribution among brain regions, indicating that its efficacy would be influenced by tumor location or the presence and extent of leptomeningeal spread. Simulation using a compartmental BBB model suggests inadequate exposure of free panobinostat in the brain following a recommended oral dosing regimen in patients. Therefore, alternative approaches to CNS delivery may be necessary to have adequate exposure of free panobinostat for the treatment of a broad range of pediatric brain tumors. SIGNIFICANCE STATEMENT: This study shows that the central nervous system (CNS) penetration of panobinostat is limited by P-gp and Bcrp, and its efficacy may be limited by inadequate distribution to the tumor. Panobinostat has heterogeneous distribution into various brain regions, indicating that its efficacy might depend on the anatomical location of the tumors. These distributional parameters in the mouse CNS can inform both preclinical and clinical trial study design and may guide treatment for these devastating brain tumors in children.
Collapse
Affiliation(s)
- Wenqiu Zhang
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (Wenq.Z, J.-H.O., Wenj.Z., S.R., W.F.E.); Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California (J.D.L.); Herbert Irving Comprehensive Cancer Center, Columbia University Medical, New York, New York (R.J.W.-R.); and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| | - Ju-Hee Oh
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (Wenq.Z, J.-H.O., Wenj.Z., S.R., W.F.E.); Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California (J.D.L.); Herbert Irving Comprehensive Cancer Center, Columbia University Medical, New York, New York (R.J.W.-R.); and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| | - Wenjuan Zhang
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (Wenq.Z, J.-H.O., Wenj.Z., S.R., W.F.E.); Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California (J.D.L.); Herbert Irving Comprehensive Cancer Center, Columbia University Medical, New York, New York (R.J.W.-R.); and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| | - Sneha Rathi
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (Wenq.Z, J.-H.O., Wenj.Z., S.R., W.F.E.); Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California (J.D.L.); Herbert Irving Comprehensive Cancer Center, Columbia University Medical, New York, New York (R.J.W.-R.); and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| | - Jon D Larson
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (Wenq.Z, J.-H.O., Wenj.Z., S.R., W.F.E.); Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California (J.D.L.); Herbert Irving Comprehensive Cancer Center, Columbia University Medical, New York, New York (R.J.W.-R.); and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| | - Robert J Wechsler-Reya
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (Wenq.Z, J.-H.O., Wenj.Z., S.R., W.F.E.); Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California (J.D.L.); Herbert Irving Comprehensive Cancer Center, Columbia University Medical, New York, New York (R.J.W.-R.); and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| | - Rachael W Sirianni
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (Wenq.Z, J.-H.O., Wenj.Z., S.R., W.F.E.); Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California (J.D.L.); Herbert Irving Comprehensive Cancer Center, Columbia University Medical, New York, New York (R.J.W.-R.); and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| | - William F Elmquist
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (Wenq.Z, J.-H.O., Wenj.Z., S.R., W.F.E.); Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California (J.D.L.); Herbert Irving Comprehensive Cancer Center, Columbia University Medical, New York, New York (R.J.W.-R.); and Department of Neurologic Surgery, UMass Chan Medical School, Worcester, Massachusetts (R.W.S.)
| |
Collapse
|
4
|
Malik JR, Podany AT, Khan P, Shaffer CL, Siddiqui JA, Baranowska‐Kortylewicz J, Le J, Fletcher CV, Ether SA, Avedissian SN. Chemotherapy in pediatric brain tumor and the challenge of the blood-brain barrier. Cancer Med 2023; 12:21075-21096. [PMID: 37997517 PMCID: PMC10726873 DOI: 10.1002/cam4.6647] [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: 07/19/2023] [Revised: 09/18/2023] [Accepted: 10/12/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Pediatric brain tumors (PBT) stand as the leading cause of cancer-related deaths in children. Chemoradiation protocols have improved survival rates, even for non-resectable tumors. Nonetheless, radiation therapy carries the risk of numerous adverse effects that can have long-lasting, detrimental effects on the quality of life for survivors. The pursuit of chemotherapeutics that could obviate the need for radiotherapy remains ongoing. Several anti-tumor agents, including sunitinib, valproic acid, carboplatin, and panobinostat, have shown effectiveness in various malignancies but have not proven effective in treating PBT. The presence of the blood-brain barrier (BBB) plays a pivotal role in maintaining suboptimal concentrations of anti-cancer drugs in the central nervous system (CNS). Ongoing research aims to modulate the integrity of the BBB to attain clinically effective drug concentrations in the CNS. However, current findings on the interaction of exogenous chemical agents with the BBB remain limited and do not provide a comprehensive explanation for the ineffectiveness of established anti-cancer drugs in PBT. METHODS We conducted our search for chemotherapeutic agents associated with the blood-brain barrier (BBB) using the following keywords: Chemotherapy in Cancer, Chemotherapy in Brain Cancer, Chemotherapy in PBT, BBB Inhibition of Drugs into CNS, Suboptimal Concentration of CNS Drugs, PBT Drugs and BBB, and Potential PBT Drugs. We reviewed each relevant article before compiling the information in our manuscript. For the generation of figures, we utilized BioRender software. FOCUS We focused our article search on chemical agents for PBT and subsequently investigated the role of the BBB in this context. Our search criteria included clinical trials, both randomized and non-randomized studies, preclinical research, review articles, and research papers. FINDING Our research suggests that, despite the availability of potent chemotherapeutic agents for several types of cancer, the effectiveness of these chemical agents in treating PBT has not been comprehensively explored. Additionally, there is a scarcity of studies examining the role of the BBB in the suboptimal outcomes of PBT treatment, despite the effectiveness of these drugs for other types of tumors.
Collapse
Affiliation(s)
- Johid Reza Malik
- Antiviral Pharmacology LaboratoryCollege of Pharmacy, University of Nebraska Medical CenterOmahaNebraskaUSA
| | - Anthony T. Podany
- Antiviral Pharmacology LaboratoryCollege of Pharmacy, University of Nebraska Medical CenterOmahaNebraskaUSA
- Pediatric Clinical Pharmacology ProgramChild Health Research Institute, University of Nebraska Medical CenterOmahaNebraskaUSA
| | - Parvez Khan
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Christopher L. Shaffer
- Pediatric Clinical Pharmacology ProgramChild Health Research Institute, University of Nebraska Medical CenterOmahaNebraskaUSA
| | - Jawed A. Siddiqui
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | | | - Jennifer Le
- University of California San Diego Skaggs School of Pharmacy and Pharmaceutical SciencesSan DiegoCaliforniaUSA
| | - Courtney V. Fletcher
- Antiviral Pharmacology LaboratoryCollege of Pharmacy, University of Nebraska Medical CenterOmahaNebraskaUSA
| | - Sadia Afruz Ether
- Antiviral Pharmacology LaboratoryCollege of Pharmacy, University of Nebraska Medical CenterOmahaNebraskaUSA
| | - Sean N. Avedissian
- Antiviral Pharmacology LaboratoryCollege of Pharmacy, University of Nebraska Medical CenterOmahaNebraskaUSA
- Pediatric Clinical Pharmacology ProgramChild Health Research Institute, University of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
5
|
Mueller S, Kline C, Stoller S, Lundy S, Christopher L, Reddy AT, Banerjee A, Cooney TM, Raber S, Hoffman C, Luks T, Wembacher-Schroeder E, Lummel N, Zhang Y, Bonner ER, Nazarian J, Molinaro AM, Prados M, Villanueva-Meyer JE, Gupta N. PNOC015: Repeated convection-enhanced delivery of MTX110 (aqueous panobinostat) in children with newly diagnosed diffuse intrinsic pontine glioma. Neuro Oncol 2023; 25:2074-2086. [PMID: 37318058 PMCID: PMC10628948 DOI: 10.1093/neuonc/noad105] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND The objective of this study was to determine the safety, tolerability, and distribution of MTX110 (aqueous panobinostat) delivered by convection-enhanced delivery (CED) in patients with newly diagnosed diffuse intrinsic pontine glioma (DIPG) who completed focal radiation therapy (RT). METHODS Patients with DIPG (2-21 years) were enrolled after RT. CED of MTX110 combined with gadoteridol was completed across 7 dose levels (DL) (30-90 µM; volumes ranging from 3 mL to 2 consecutive doses of 6 mL). An accelerated dose escalation design was used. Distribution of infusate was monitored with real-time MR imaging. Repeat CED was performed every 4-8 weeks. Quality-of-life (QoL) assessments were obtained at baseline, every 3 months on therapy, and end of therapy. RESULTS Between May 2018 and March 2020, 7 patients who received a total of 48 CED infusions, were enrolled (median age 8 years, range 5-21). Three patients experienced dose-limited toxicities. Four grade 3 treatment-related adverse events were observed. Most toxicities were transient new or worsening neurologic function. Median overall survival (OS) was 26.1 months (95% confidence interval: 14.8-not reached). Progression-free survival was 4-14 months (median, 7). Cumulative percentage of tumor coverage for combined CED infusions per patient ranged from 35.6% to 81.0%. Increased CED infusions were negatively associated with self-reported QoL assessments. CONCLUSION Repeat CED of MTX110 with real-time imaging with gadoteridol is tolerable for patients with DIPG. Median OS of 26.1 months compares favorably with historical data for children with DIPG. The results support further investigation of this strategy in a larger cohort.
Collapse
Affiliation(s)
- Sabine Mueller
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
- Department of Pediatrics, University of Zurich, Zurich, Switzerland
| | - Cassie Kline
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Schuyler Stoller
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Shannon Lundy
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Lauren Christopher
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Alyssa T Reddy
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Anu Banerjee
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Tabitha M Cooney
- Department of Pediatric Oncology, Dana-Farber/Boston Children’s Cancer and Blood Disorder Center, Boston, Massachusetts, USA
| | - Shannon Raber
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Carly Hoffman
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Tracy Luks
- Department of Radiology, University of California, San Francisco, San Francisco, California, USA
| | | | | | - Yalan Zhang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Erin R Bonner
- Center for Genetic Medicine Research, Children’s National Medical Center, Washington, DC, USA
| | - Javad Nazarian
- Department of Pediatrics, University of Zurich, Zurich, Switzerland
- Center for Genetic Medicine Research, Children’s National Medical Center, Washington, DC, USA
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Michael Prados
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Javier E Villanueva-Meyer
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
- Department of Radiology, University of California, San Francisco, San Francisco, California, USA
| | - Nalin Gupta
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
6
|
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
|
7
|
Jane EP, Reslink MC, Gatesman TA, Halbert ME, Miller TA, Golbourn BJ, Casillo SM, Mullett SJ, Wendell SG, Obodo U, Mohanakrishnan D, Dange R, Michealraj A, Brenner C, Agnihotri S, Premkumar DR, Pollack IF. Targeting mitochondrial energetics reverses panobinostat- and marizomib-induced resistance in pediatric and adult high-grade gliomas. Mol Oncol 2023; 17:1821-1843. [PMID: 37014128 PMCID: PMC10483615 DOI: 10.1002/1878-0261.13427] [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: 11/08/2022] [Revised: 02/22/2023] [Accepted: 04/03/2023] [Indexed: 04/05/2023] Open
Abstract
In previous studies, we demonstrated that panobinostat, a histone deacetylase inhibitor, and bortezomib, a proteasomal inhibitor, displayed synergistic therapeutic activity against pediatric and adult high-grade gliomas. Despite the remarkable initial response to this combination, resistance emerged. Here, in this study, we aimed to investigate the molecular mechanisms underlying the anticancer effects of panobinostat and marizomib, a brain-penetrant proteasomal inhibitor, and the potential for exploitable vulnerabilities associated with acquired resistance. RNA sequencing followed by gene set enrichment analysis (GSEA) was employed to compare the molecular signatures enriched in resistant compared with drug-naïve cells. The levels of adenosine 5'-triphosphate (ATP), nicotinamide adenine dinucleotide (NAD)+ content, hexokinase activity, and tricarboxylic acid (TCA) cycle metabolites required for oxidative phosphorylation to meet their bioenergetic needs were analyzed. Here, we report that panobinostat and marizomib significantly depleted ATP and NAD+ content, increased mitochondrial permeability and reactive oxygen species generation, and promoted apoptosis in pediatric and adult glioma cell lines at initial treatment. However, resistant cells exhibited increased levels of TCA cycle metabolites, which required for oxidative phosphorylation to meet their bioenergetic needs. Therefore, we targeted glycolysis and the electron transport chain (ETC) with small molecule inhibitors, which displayed substantial efficacy, suggesting that resistant cell survival is dependent on glycolytic and ETC complexes. To verify these observations in vivo, lonidamine, an inhibitor of glycolysis and mitochondrial function, was chosen. We produced two diffuse intrinsic pontine glioma (DIPG) models, and lonidamine treatment significantly increased median survival in both models, with particularly dramatic effects in panobinostat- and marizomib-resistant cells. These data provide new insights into mechanisms of treatment resistance in gliomas.
Collapse
Affiliation(s)
- Esther P. Jane
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePAUSA
- John G. Rangos Sr. Research CenterChildren's Hospital of PittsburghPAUSA
| | - Matthew C. Reslink
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePAUSA
| | - Taylor A. Gatesman
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePAUSA
- John G. Rangos Sr. Research CenterChildren's Hospital of PittsburghPAUSA
| | - Matthew E. Halbert
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePAUSA
- John G. Rangos Sr. Research CenterChildren's Hospital of PittsburghPAUSA
| | - Tracy A. Miller
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePAUSA
| | - Brian J. Golbourn
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePAUSA
| | - Stephanie M. Casillo
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePAUSA
- John G. Rangos Sr. Research CenterChildren's Hospital of PittsburghPAUSA
| | - Steven J. Mullett
- Department of Pharmacology and Chemical BiologyUniversity of PittsburghPAUSA
| | - Stacy G. Wendell
- Department of Pharmacology and Chemical BiologyUniversity of PittsburghPAUSA
| | - Udochukwu Obodo
- Department of Diabetes & Cancer MetabolismCity of Hope Medical CenterDuarteCAUSA
| | | | - Riya Dange
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePAUSA
| | - Antony Michealraj
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePAUSA
| | - Charles Brenner
- Department of Diabetes & Cancer MetabolismCity of Hope Medical CenterDuarteCAUSA
| | - Sameer Agnihotri
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePAUSA
- John G. Rangos Sr. Research CenterChildren's Hospital of PittsburghPAUSA
- UPMC Hillman Cancer CenterPittsburghPAUSA
| | - Daniel R. Premkumar
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePAUSA
- John G. Rangos Sr. Research CenterChildren's Hospital of PittsburghPAUSA
- UPMC Hillman Cancer CenterPittsburghPAUSA
| | - Ian F. Pollack
- Department of NeurosurgeryUniversity of Pittsburgh School of MedicinePAUSA
- John G. Rangos Sr. Research CenterChildren's Hospital of PittsburghPAUSA
- UPMC Hillman Cancer CenterPittsburghPAUSA
| |
Collapse
|
8
|
Rechberger JS, Bouchal SM, Power EA, Nonnenbroich LF, Nesvick CL, Daniels DJ. Bench-to-bedside investigations of H3 K27-altered diffuse midline glioma: drug targets and potential pharmacotherapies. Expert Opin Ther Targets 2023; 27:1071-1086. [PMID: 37897190 PMCID: PMC11079776 DOI: 10.1080/14728222.2023.2277232] [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: 08/30/2023] [Accepted: 10/26/2023] [Indexed: 10/29/2023]
Abstract
INTRODUCTION H3 K27-altered diffuse midline glioma (DMG) is the most common malignant brainstem tumor in the pediatric population. Despite enormous preclinical and clinical efforts, the prognosis remains dismal, with fewer than 10% of patients surviving for two years after diagnosis. Fractionated radiation remains the only standard treatment options for DMG. Developing novel treatments and therapeutic delivery methods is critical to improving outcomes in this devastating disease. AREAS COVERED This review addresses recent advances in molecularly targeted pharmacotherapy and immunotherapy in DMG. The clinical presentation, diagnostic workup, unique pathological challenges, and current clinical trials are highlighted throughout. EXPERT OPINION Promising pharmacotherapies targeting various components of DMG pathology and the application of immunotherapies have the potential to improve patient outcomes. However, novel approaches are needed to truly revolutionize treatment for this tumor. First, combinational therapy should be employed, as DMG can develop resistance to single-agent approaches and many therapies are susceptible to rapid clearance from the brain. Second, drug-tumor residence time, i.e. the time for which a therapeutic is present at efficacious concentrations within the tumor, must be maximized to facilitate a durable treatment response. Engineering extended drug delivery methods with minimal off-tumor toxicity should be a focus of future studies.
Collapse
Affiliation(s)
- Julian S. Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Samantha M. Bouchal
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Erica A. Power
- Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Leo F. Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Cody L. Nesvick
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - David J. Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| |
Collapse
|
9
|
Vanbilloen WJF, Rechberger JS, Anderson JB, Nonnenbroich LF, Zhang L, Daniels DJ. Nanoparticle Strategies to Improve the Delivery of Anticancer Drugs across the Blood-Brain Barrier to Treat Brain Tumors. Pharmaceutics 2023; 15:1804. [PMID: 37513992 PMCID: PMC10383584 DOI: 10.3390/pharmaceutics15071804] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Primary brain and central nervous system (CNS) tumors are a diverse group of neoplasms that occur within the brain and spinal cord. Although significant advances in our understanding of the intricate biological underpinnings of CNS neoplasm tumorigenesis and progression have been made, the translation of these discoveries into effective therapies has been stymied by the unique challenges presented by these tumors' exquisitely sensitive location and the body's own defense mechanisms (e.g., the brain-CSF barrier and blood-brain barrier), which normally protect the CNS from toxic insult. These barriers effectively prevent the delivery of therapeutics to the site of disease. To overcome these obstacles, new methods for therapeutic delivery are being developed, with one such approach being the utilization of nanoparticles. Here, we will cover the current state of the field with a particular focus on the challenges posed by the BBB, the different nanoparticle classes which are under development for targeted CNS tumor therapeutics delivery, and strategies which have been developed to bypass the BBB and enable effective therapeutics delivery to the site of disease.
Collapse
Affiliation(s)
- Wouter J. F. Vanbilloen
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Neurology, Elisabeth-Tweesteden Hospital, 5022 GC Tilburg, The Netherlands
| | - Julian S. Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jacob B. Anderson
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
- Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Leo F. Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (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
| | - Liang Zhang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
| | - David J. Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
10
|
Diffuse intrinsic pontine glioma: Insights into oncogenesis and opportunities for targeted therapy. PEDIATRIC HEMATOLOGY ONCOLOGY JOURNAL 2023. [DOI: 10.1016/j.phoj.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
|
11
|
Everix L, Seane EN, Ebenhan T, Goethals I, Bolcaen J. Introducing HDAC-Targeting Radiopharmaceuticals for Glioblastoma Imaging and Therapy. Pharmaceuticals (Basel) 2023; 16:227. [PMID: 37259375 PMCID: PMC9967489 DOI: 10.3390/ph16020227] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 09/29/2023] Open
Abstract
Despite recent advances in multimodality therapy for glioblastoma (GB) incorporating surgery, radiotherapy, chemotherapy and targeted therapy, the overall prognosis remains poor. One of the interesting targets for GB therapy is the histone deacetylase family (HDAC). Due to their pleiotropic effects on, e.g., DNA repair, cell proliferation, differentiation, apoptosis and cell cycle, HDAC inhibitors have gained a lot of attention in the last decade as anti-cancer agents. Despite their known underlying mechanism, their therapeutic activity is not well-defined. In this review, an extensive overview is given of the current status of HDAC inhibitors for GB therapy, followed by an overview of current HDAC-targeting radiopharmaceuticals. Imaging HDAC expression or activity could provide key insights regarding the role of HDAC enzymes in gliomagenesis, thus identifying patients likely to benefit from HDACi-targeted therapy.
Collapse
Affiliation(s)
- Liesbeth Everix
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, 2610 Antwerpen, Belgium
| | - Elsie Neo Seane
- Department of Medical Imaging and Therapeutic Sciences, Cape Peninsula University of Technology, Cape Town 7530, South Africa
| | - Thomas Ebenhan
- Pre-Clinical Imaging Facility (PCIF), (NuMeRI) NPC, Pretoria 0001, South Africa
- Department of Science and Technology/Preclinical Drug Development Platform (PCDDP), North West University, Potchefstroom 2520, South Africa
- Nuclear Medicine, University of Pretoria, Pretoria 0001, South Africa
| | - Ingeborg Goethals
- Department of Nuclear Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Julie Bolcaen
- Radiation Biophysics Division, SSC laboratory, iThemba LABS, Cape Town 7131, South Africa
| |
Collapse
|
12
|
Rechberger JS, Power BT, Power EA, Nesvick CL, Daniels DJ. H3K27-altered diffuse midline glioma: a paradigm shifting opportunity in direct delivery of targeted therapeutics. Expert Opin Ther Targets 2023; 27:9-17. [PMID: 36744399 PMCID: PMC10165636 DOI: 10.1080/14728222.2023.2177531] [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: 02/07/2023]
Abstract
INTRODUCTION Despite much progress, the prognosis for H3K27-altered diffuse midline glioma (DMG), previously known as diffuse intrinsic pontine glioma when located in the brainstem, remains dark and dismal. AREAS COVERED A wealth of research over the past decade has revolutionized our understanding of the molecular basis of DMG, revealing potential targetable vulnerabilities for treatment of this lethal childhood cancer. However, obstacles to successful clinical implementation of novel therapies remain, including effective delivery across the blood-brain barrier (BBB) to the tumor site. Here, we review relevant literature and clinical trials and discuss direct drug delivery via convection-enhanced delivery (CED) as a promising treatment modality for DMG. We outline a comprehensive molecular, pharmacological, and procedural approach that may offer hope for afflicted patients and their families. EXPERT OPINION Challenges remain in successful drug delivery to DMG. While CED and other techniques offer a chance to bypass the BBB, the variables influencing successful intratumoral targeting are numerous and complex. We discuss these variables and potential solutions that could lead to the successful clinical implementation of preclinically promising therapeutic agents.
Collapse
Affiliation(s)
- Julian S Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Blake T Power
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Erica A Power
- Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Cody L Nesvick
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| |
Collapse
|
13
|
Power EA, Rechberger JS, Zhang L, Oh JH, Anderson JB, Nesvick CL, Ge J, Hinchcliffe EH, Elmquist WF, Daniels DJ. Overcoming translational barriers in H3K27-altered diffuse midline glioma: Increasing the drug-tumor residence time. Neurooncol Adv 2023; 5:vdad033. [PMID: 37128506 PMCID: PMC10148679 DOI: 10.1093/noajnl/vdad033] [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: 03/29/2023] Open
Abstract
Background H3K27-altered diffuse midline glioma (DMG) is the deadliest pediatric brain tumor; despite intensive research efforts, every clinical trial to date has failed. Is this because we are choosing the wrong drugs? Or are drug delivery and other pharmacokinetic variables at play? We hypothesize that the answer is likely a combination, where optimization may result in a much needed novel therapeutic approach. Methods We used in vitro drug screening, patient samples, and shRNA knockdown models to identify an upregulated target in DMG. A single small molecule protein kinase inhibitor with translational potential was selected for systemic and direct, loco-regional delivery to patient-derived xenografts (PDX) and genetically engineered mouse models (GEMM). Pharmacokinetic studies were conducted in non-tumor bearing rats. Results Aurora kinase (AK) inhibitors demonstrated strong antitumor effects in DMG drug screens. Additional in vitro studies corroborated the importance of AK to DMG survival. Systemic delivery of alisertib showed promise in subcutaneous PDX but not intracranial GEMM and PDX models. Repeated loco-regional drug administration into the tumor through convection-enhanced delivery (CED) was equally inefficacious, and pharmacokinetic studies revealed rapid clearance of alisertib from the brain. In an effort to increase the drug to tumor residence time, continuous CED over 7 days improved drug retention in the rodent brainstem and significantly extended survival in both orthotopic PDXs and GEMMs. Conclusions These studies provide evidence for increasing drug-tumor residence time of promising targeted therapies via extended CED as a valuable treatment strategy for DMG.
Collapse
Affiliation(s)
- Erica A Power
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Julian S Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Liang Zhang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ju-Hee Oh
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jacob B Anderson
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Cody L Nesvick
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Jizhi Ge
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - William F Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
14
|
Groves A, Cooney TM. Epigenetic programming of pediatric high-grade glioma: Pushing beyond proof of concept to clinical benefit. Front Cell Dev Biol 2022; 10:1089898. [PMID: 36589742 PMCID: PMC9795020 DOI: 10.3389/fcell.2022.1089898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022] Open
Abstract
Pediatric high-grade gliomas (pHGG) are a molecularly diverse group of malignancies, each incredibly aggressive and in dire need of treatment advancements. Genomic analysis has revolutionized our understanding of these tumors, identifying biologically relevant subgroups with differing canonical mutational profiles that vary based on tumor location and age. In particular, the discovery of recurrent histone H3 mutations (H3K27M in diffuse midline glioma, H3G34R/V in hemispheric pediatric high-grade gliomas) as unique "oncohistone" drivers revealed epigenetic dysregulation as a hallmark of pediatric high-grade gliomas oncogenesis. While reversing this signature through epigenetic programming has proven effective in several pre-clinical survival models, early results from pediatric high-grade gliomas clinical trials suggest that epigenetic modifier monotherapy will likely not provide long-term disease control. In this review we summarize the genetic, epigenetic, and cellular heterogeneity of pediatric high-grade gliomas, and highlight potential paths forward for epigenetic programming in this devastating disease.
Collapse
Affiliation(s)
- Andrew Groves
- Division of Hematology/Oncology, University of Iowa Stead Family Children’s Hospital, Iowa City, IA, United States
| | - Tabitha M. Cooney
- Dana Farber/Boston Children’s Cancer and Blood Disorder Center, Boston, MA, United States
| |
Collapse
|
15
|
Shen J, Liu J. Bruton's tyrosine kinase inhibitors in the treatment of primary central nervous system lymphoma: A mini-review. Front Oncol 2022; 12:1034668. [PMID: 36465385 PMCID: PMC9713408 DOI: 10.3389/fonc.2022.1034668] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/31/2022] [Indexed: 09/19/2023] Open
Abstract
Primary central nervous system lymphoma (PCNSL) is a highly aggressive brain tumor with poor prognosis if no treatment. The activation of the NF-κB (nuclear factor kappa-B) is the oncogenic hallmark of PCNSL, and it was driven by B cell receptor (BCR) and Toll-like receptor (TLR) signaling pathways. The emergence of Bruton's tyrosine kinase inhibitors (BTKis) has brought the dawn of life to patients with PCNSL. This review summarizes the management of PCNSL with BTKis and potential molecular mechanisms of BTKi in the treatment of PCNSL. And the review will focus on the clinical applications of BTKi in the treatment of PCNSL including the efficacy and adverse events, the clinical trials currently being carried out, the underlying mechanisms of resistance to BTKi and possible solutions to drug resistance.
Collapse
Affiliation(s)
- Jing Shen
- Department of Hematology, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Jinghua Liu
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Hematology, Northern Theater General Hospital, Shenyang, China
| |
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
|
Gleason JM, Klass SH, Huang P, Ozawa T, Santos RA, Fogarty MM, Raleigh DR, Berger MS, Francis MB. Intrinsically Disordered Protein Micelles as Vehicles for Convection-Enhanced Drug Delivery to Glioblastoma Multiforme. ACS APPLIED BIO MATERIALS 2022; 5:3695-3702. [PMID: 35857070 DOI: 10.1021/acsabm.2c00215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lipid and micelle-based nanocarriers have been explored for anticancer drug delivery to improve accumulation and uptake in tumor tissue. As an experimental opportunity in this area, our lab has developed a protein-based micelle nanocarrier consisting of a hydrophilic intrinsically disordered protein (IDP) domain bound to a hydrophobic tail, termed IDP-2Yx2A. This construct can be used to encapsulate hydrophobic chemotherapeutics that would otherwise be too insoluble in water to be administered. In this study, we evaluate the in vivo efficacy of IDP-2Yx2A by delivering a highly potent but water-insoluble cancer drug, SN38, into glioblastoma multiforme (GBM) tumors via convection-enhanced delivery (CED). The protein carriers alone are shown to elicit minimal toxicity effects in mice; furthermore, they can encapsulate and deliver concentrations of SN38 that would otherwise be lethal without the carriers. CED administration of these drug-loaded micelles into mice bearing U251-MG GBM xenografts resulted in slowed tumor growth and significant increases in median survival times compared to nonencapsulated SN38 and PBS controls.
Collapse
Affiliation(s)
- Jamie M Gleason
- Department of Chemistry, University of California, Berkeley, California 94720, United States
| | - Sarah H Klass
- Department of Chemistry, University of California, Berkeley, California 94720, United States
| | - Paul Huang
- Department of Chemistry, University of California, Berkeley, California 94720, United States
| | - Tomoko Ozawa
- Department of Neurological Surgery, University of California, San Francisco, California 94158, United States
| | - Raquel A Santos
- Department of Neurological Surgery, University of California, San Francisco, California 94158, United States
| | - Miko M Fogarty
- Department of Neurological Surgery, University of California, San Francisco, California 94158, United States
| | - David R Raleigh
- Department of Neurological Surgery, University of California, San Francisco, California 94158, United States.,Department of Radiation Oncology, University of California, San Francisco, California 94518, United States
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, California 94158, United States
| | - Matthew B Francis
- Department of Chemistry, University of California, Berkeley, California 94720, United States.,Materials Sciences Division, Lawrence Berkeley National Laboratories, Berkeley, California 94720, United States
| |
Collapse
|
18
|
Rechberger JS, Daniels DJ. Locoregional infusion of IL13Rα2-specific immunotoxins in children and adults with high-grade glioma. Ther Deliv 2022; 13:385-389. [PMID: 35872639 PMCID: PMC9756106 DOI: 10.4155/tde-2022-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/18/2022] [Indexed: 08/03/2023] Open
Affiliation(s)
- Julian S Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| |
Collapse
|
19
|
Shan S, Chen J, Sun Y, Wang Y, Xia B, Tan H, Pan C, Gu G, Zhong J, Qing G, Zhang Y, Wang J, Wang Y, Wang Y, Zuo P, Xu C, Li F, Guo W, Xu L, Chen M, Fan Y, Zhang L, Liang X. Functionalized Macrophage Exosomes with Panobinostat and PPM1D-siRNA for Diffuse Intrinsic Pontine Gliomas Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200353. [PMID: 35585670 PMCID: PMC9313473 DOI: 10.1002/advs.202200353] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/01/2022] [Indexed: 05/05/2023]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a rare and fatal pediatric brain tumor. Mutation of p53-induced protein phosphatase 1 (PPM1D) in DIPG cells promotes tumor cell proliferation, and inhibition of PPM1D expression in DIPG cells with PPM1D mutation effectively reduces the proliferation activity of tumor cells. Panobinostat effectively kills DIPG tumor cells, but its systemic toxicity and low blood-brain barrier (BBB) permeability limits its application. In this paper, a nano drug delivery system based on functionalized macrophage exosomes with panobinostat and PPM1D-siRNA for targeted therapy of DIPG with PPM1D mutation is prepared. The nano drug delivery system has higher drug delivery efficiency and better therapeutic effect than free drugs. In vivo and in vitro experimental results show that the nano drug delivery system can deliver panobinostat and siRNA across the BBB and achieve a targeted killing effect of DIPG tumor cells, resulting in the prolonged survival of orthotopic DIPG mice. This study provides new ideas for the delivery of small molecule drugs and gene drugs for DIPG therapy.
Collapse
Affiliation(s)
- Shaobo Shan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationBeijing Advanced Innovation Center for Biomedical EngineeringSchool of Biological Science and Medical Engineering & School of Engineering Medicine & Shenzhen Institute of Beihang UniversityBeihang UniversityBeijing100083P. R. China
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijing100050P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| | - Junge Chen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationBeijing Advanced Innovation Center for Biomedical EngineeringSchool of Biological Science and Medical Engineering & School of Engineering Medicine & Shenzhen Institute of Beihang UniversityBeihang UniversityBeijing100083P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| | - Yu Sun
- Pediatric Epilepsy CenterPeking University First HospitalNo.1 Xi'an Men Street, Xicheng DistrictBeijing100034P. R. China
| | - Yongchao Wang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijing100050P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| | - Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| | - Hong Tan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| | - Changcun Pan
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijing100050P. R. China
| | - Guocan Gu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijing100050P. R. China
| | - Jie Zhong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| | - Guangchao Qing
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| | - Yuxuan Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| | - Jinjin Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| | - Yufei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| | - Yi Wang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijing100050P. R. China
| | - Pengcheng Zuo
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijing100050P. R. China
| | - Cheng Xu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijing100050P. R. China
| | - Fangzhou Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| | - Weisheng Guo
- Department of Minimally Invasive Interventional RadiologyCollege of Biomedical Engineering & The Second Affiliated HospitalGuangzhou Medical UniversityGuangzhou510260P. R. China
| | - Lijun Xu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationBeijing Advanced Innovation Center for Biomedical EngineeringSchool of Biological Science and Medical Engineering & School of Engineering Medicine & Shenzhen Institute of Beihang UniversityBeihang UniversityBeijing100083P. R. China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacau999078P. R. China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationBeijing Advanced Innovation Center for Biomedical EngineeringSchool of Biological Science and Medical Engineering & School of Engineering Medicine & Shenzhen Institute of Beihang UniversityBeihang UniversityBeijing100083P. R. China
| | - Liwei Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationBeijing Advanced Innovation Center for Biomedical EngineeringSchool of Biological Science and Medical Engineering & School of Engineering Medicine & Shenzhen Institute of Beihang UniversityBeihang UniversityBeijing100083P. R. China
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijing100050P. R. China
- China National Clinical Research Center for Neurological Diseases (NCRC‐ND)Beijing100070P. R. China
| | - Xing‐Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and Technology of ChinaBeijing100190P. R. China
| |
Collapse
|
20
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
21
|
Power EA, Rechberger JS, Gupta S, Schwartz JD, Daniels DJ, Khatua S. Drug delivery across the blood-brain barrier for the treatment of pediatric brain tumors - An update. Adv Drug Deliv Rev 2022; 185:114303. [PMID: 35460714 DOI: 10.1016/j.addr.2022.114303] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 12/14/2022]
Abstract
Even though the last decade has seen a surge in the identification of molecular targets and targeted therapies in pediatric brain tumors, the blood brain barrier (BBB) remains a significant challenge in systemic drug delivery. This continues to undermine therapeutic efficacy. Recent efforts have identified several strategies that can facilitate enhanced drug delivery into pediatric brain tumors. These include invasive methods such as intra-arterial, intrathecal, and convection enhanced delivery and non-invasive technologies that allow for transient access across the BBB, including focused ultrasound and nanotechnology. This review discusses current strategies that are being used to enhance delivery of different therapies across the BBB to the tumor site - a major unmet need in pediatric neuro-oncology.
Collapse
Affiliation(s)
- Erica A Power
- Mayo Clinic Graduate School of Biomedical Sciences, 200 First Street SW, Rochester, MN 55905, United States; Department of Neurologic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States
| | - Julian S Rechberger
- Mayo Clinic Graduate School of Biomedical Sciences, 200 First Street SW, Rochester, MN 55905, United States; Department of Neurologic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States
| | - Sumit Gupta
- Department of Pediatric Hematology/Oncology, Roseman University of Health Sciences, Las Vegas, NV 89118, United States
| | - Jonathan D Schwartz
- Department of Pediatric Hematology/Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States
| | - Soumen Khatua
- Department of Pediatric Hematology/Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States.
| |
Collapse
|
22
|
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
|
23
|
Rechberger JS, Porath KA, Zhang L, Nesvick CL, Schrecengost RS, Sarkaria JN, Daniels DJ. IL-13Rα2 Status Predicts GB-13 (IL13.E13K-PE4E) Efficacy in High-Grade Glioma. Pharmaceutics 2022; 14:922. [PMID: 35631512 PMCID: PMC9143740 DOI: 10.3390/pharmaceutics14050922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/14/2022] [Accepted: 04/22/2022] [Indexed: 02/05/2023] Open
Abstract
High-grade gliomas (HGG) are devastating diseases in children and adults. In the pediatric population, diffuse midline gliomas (DMG) harboring H3K27 alterations are the most aggressive primary malignant brain tumors. With no effective therapies available, children typically succumb to disease within one year of diagnosis. In adults, glioblastoma (GBM) remains largely intractable, with a median survival of approximately 14 months despite standard clinical care of radiation and temozolomide. Therefore, effective therapies for these tumors remain one of the most urgent and unmet needs in modern medicine. Interleukin 13 receptor subunit alpha 2 (IL-13Rα2) is a cell-surface transmembrane protein upregulated in many HGGs, including DMG and adult GBM, posing a potentially promising therapeutic target for these tumors. In this study, we investigated the pharmacological effects of GB-13 (also known as IL13.E13K-PE4E), a novel peptide-toxin conjugate that contains a targeting moiety designed to bind IL-13Rα2 with high specificity and a point-mutant cytotoxic domain derived from Pseudomonas exotoxin A. Glioma cell lines demonstrated a spectrum of IL-13Rα2 expression at both the transcript and protein level. Anti-tumor effects of GB-13 strongly correlated with IL-13Rα2 expression and were reflected in apoptosis induction and decreased cell proliferation in vitro. Direct intratumoral administration of GB-13 via convection-enhanced delivery (CED) significantly decreased tumor burden and resulted in prolonged survival in IL-13Rα2-upregulated orthotopic xenograft models of HGG. In summary, administration of GB-13 demonstrated a promising pharmacological response in HGG models both in vitro and in vivo in a manner strongly associated with IL-13Rα2 expression, underscoring the potential of this IL-13Rα2-targeted therapy in a subset of HGG with increased IL-13Rα2 levels.
Collapse
Affiliation(s)
- Julian S. Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (J.S.R.); (L.Z.); (C.L.N.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Kendra A. Porath
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.P.); (J.N.S.)
| | - Liang Zhang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (J.S.R.); (L.Z.); (C.L.N.)
| | - Cody L. Nesvick
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (J.S.R.); (L.Z.); (C.L.N.)
| | | | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA; (K.A.P.); (J.N.S.)
| | - David J. Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (J.S.R.); (L.Z.); (C.L.N.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| |
Collapse
|
24
|
Zhou Q, Xu Y, Zhou Y, Wang J. Promising Chemotherapy for Malignant Pediatric Brain Tumor in Recent Biological Insights. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092685. [PMID: 35566032 PMCID: PMC9104915 DOI: 10.3390/molecules27092685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/16/2022]
Abstract
Brain tumors are the most widespread malignancies in children around the world. Chemotherapy plays a critical role in the treatment of these tumors. Although the current chemotherapy process has a remarkable outcome for a certain subtype of brain tumor, improving patient survival is still a major challenge. Further intensive treatment with conventional non-specific chemotherapy could cause additional adverse reactions without significant advancement in survival. Recently, patient derived brain tumor, xenograft, and whole genome analysis using deep sequencing technology has made a significant contribution to our understanding of cancer treatment. This realization has changed the focus to new agents, targeting the molecular pathways that are critical to tumor survival or proliferation. Thus, many novel drugs targeting epigenetic regulators or tyrosine kinase have been developed. These selective drugs may have less toxicity in normal cells and are expected to be more effective than non-specific chemotherapeutics. This review will summarize the latest novel targets and corresponding candidate drugs, which are promising chemotherapy for brain tumors according to the biological insights.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Pharmacy, Hangzhou Medical College, Hangzhou 310053, China; (Q.Z.); (Y.Z.)
| | - Yichen Xu
- Department of Biological Sciences, University of Southern California (Main Campus), Los Angeles, CA 90007, USA;
| | - Yan Zhou
- Department of Pharmacy, Hangzhou Medical College, Hangzhou 310053, China; (Q.Z.); (Y.Z.)
| | - Jincheng Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Correspondence:
| |
Collapse
|
25
|
Gatto L, Franceschi E, Tosoni A, Di Nunno V, Bartolini S, Brandes AA. Molecular Targeted Therapies: Time for a Paradigm Shift in Medulloblastoma Treatment? Cancers (Basel) 2022; 14:333. [PMID: 35053495 PMCID: PMC8773620 DOI: 10.3390/cancers14020333] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/11/2022] Open
Abstract
Medulloblastoma is a rare malignancy of the posterior cranial fossa. Although until now considered a single disease, according to the current WHO classification, it is a heterogeneous tumor that comprises multiple molecularly defined subgroups, with distinct gene expression profiles, pathogenetic driver alterations, clinical behaviors and age at onset. Adult medulloblastoma, in particular, is considered a rarer "orphan" entity in neuro-oncology practice because while treatments have progressively evolved for the pediatric population, no practice-changing prospective, randomized clinical trials have been performed in adults. In this scenario, the toughest challenge is to transfer the advances in cancer genomics into new molecularly targeted therapeutics, to improve the prognosis of this neoplasm and the treatment-related toxicities. Herein, we focus on the recent advances in targeted therapy of medulloblastoma based on the new and deeper knowledge of disease biology.
Collapse
Affiliation(s)
- Lidia Gatto
- Medical Oncology Department, Azienda Unità Sanitaria Locale, 40139 Bologna, Italy; (L.G.); (V.D.N.)
| | - Enrico Franceschi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Oncologia Medica del Sistema Nervoso, 40139 Bologna, Italy; (A.T.); (S.B.); (A.A.B.)
| | - Alicia Tosoni
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Oncologia Medica del Sistema Nervoso, 40139 Bologna, Italy; (A.T.); (S.B.); (A.A.B.)
| | - Vincenzo Di Nunno
- Medical Oncology Department, Azienda Unità Sanitaria Locale, 40139 Bologna, Italy; (L.G.); (V.D.N.)
| | - Stefania Bartolini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Oncologia Medica del Sistema Nervoso, 40139 Bologna, Italy; (A.T.); (S.B.); (A.A.B.)
| | - Alba Ariela Brandes
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Oncologia Medica del Sistema Nervoso, 40139 Bologna, Italy; (A.T.); (S.B.); (A.A.B.)
| |
Collapse
|
26
|
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
|
27
|
Rechberger JS, Thiele F, Daniels DJ. Status Quo and Trends of Intra-Arterial Therapy for Brain Tumors: A Bibliometric and Clinical Trials Analysis. Pharmaceutics 2021; 13:pharmaceutics13111885. [PMID: 34834300 PMCID: PMC8625566 DOI: 10.3390/pharmaceutics13111885] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022] Open
Abstract
Intra-arterial drug delivery circumvents the first-pass effect and is believed to increase both efficacy and tolerability of primary and metastatic brain tumor therapy. The aim of this update is to report on pertinent articles and clinical trials to better understand the research landscape to date and future directions. Elsevier's Scopus and ClinicalTrials.gov databases were reviewed in August 2021 for all possible articles and clinical trials of intra-arterial drug injection as a treatment strategy for brain tumors. Entries were screened against predefined selection criteria and various parameters were summarized. Twenty clinical trials and 271 articles satisfied all inclusion criteria. In terms of articles, 201 (74%) were primarily clinical and 70 (26%) were basic science, published in a total of 120 different journals. Median values were: publication year, 1986 (range, 1962-2021); citation count, 15 (range, 0-607); number of authors, 5 (range, 1-18). Pertaining to clinical trials, 9 (45%) were phase 1 trials, with median expected start and completion years in 2011 (range, 1998-2019) and 2022 (range, 2008-2025), respectively. Only one (5%) trial has reported results to date. Glioma was the most common tumor indication reported in both articles (68%) and trials (75%). There were 215 (79%) articles investigating chemotherapy, while 13 (65%) trials evaluated targeted therapy. Transient blood-brain barrier disruption was the commonest strategy for articles (27%) and trials (60%) to optimize intra-arterial therapy. Articles and trials predominately originated in the United States (50% and 90%, respectively). In this bibliometric and clinical trials analysis, we discuss the current state and trends of intra-arterial therapy for brain tumors. Most articles were clinical, and traditional anti-cancer agents and drug delivery strategies were commonly studied. This was reflected in clinical trials, of which only a single study had reported outcomes. We anticipate future efforts to involve novel therapeutic and procedural strategies based on recent advances in the field.
Collapse
Affiliation(s)
- Julian S. Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA;
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence:
| | - Frederic Thiele
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA;
| | - David J. Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
28
|
Sokolov AV, Dostdar SA, Attwood MM, Krasilnikova AA, Ilina AA, Nabieva AS, Lisitsyna AA, Chubarev VN, Tarasov VV, Schiöth HB. Brain Cancer Drug Discovery: Clinical Trials, Drug Classes, Targets, and Combinatorial Therapies. Pharmacol Rev 2021; 73:1-32. [PMID: 34663683 DOI: 10.1124/pharmrev.121.000317] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Brain cancer is a formidable challenge for drug development, and drugs derived from many cutting-edge technologies are being tested in clinical trials. We manually characterized 981 clinical trials on brain tumors that were registered in ClinicalTrials.gov from 2010 to 2020. We identified 582 unique therapeutic entities targeting 581 unique drug targets and 557 unique treatment combinations involving drugs. We performed the classification of both the drugs and drug targets based on pharmacological and structural classifications. Our analysis demonstrates a large diversity of agents and targets. Currently, we identified 32 different pharmacological directions for therapies that are based on 42 structural classes of agents. Our analysis shows that kinase inhibitors, chemotherapeutic agents, and cancer vaccines are the three most common classes of agents identified in trials. Agents in clinical trials demonstrated uneven distribution in combination approaches; chemotherapy agents, proteasome inhibitors, and immune modulators frequently appeared in combinations, whereas kinase inhibitors, modified immune effector cells did not as was shown by combination networks and descriptive statistics. This analysis provides an extensive overview of the drug discovery field in brain cancer, shifts that have been happening in recent years, and challenges that are likely to come. SIGNIFICANCE STATEMENT: This review provides comprehensive quantitative analysis and discussion of the brain cancer drug discovery field, including classification of drug, targets, and therapies.
Collapse
Affiliation(s)
- Aleksandr V Sokolov
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Samira A Dostdar
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Misty M Attwood
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Aleksandra A Krasilnikova
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anastasia A Ilina
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Amina Sh Nabieva
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anna A Lisitsyna
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vladimir N Chubarev
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vadim V Tarasov
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
29
|
Leszczynska KB, Jayaprakash C, Kaminska B, Mieczkowski J. Emerging Advances in Combinatorial Treatments of Epigenetically Altered Pediatric High-Grade H3K27M Gliomas. Front Genet 2021; 12:742561. [PMID: 34646308 PMCID: PMC8503186 DOI: 10.3389/fgene.2021.742561] [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: 07/16/2021] [Accepted: 08/17/2021] [Indexed: 01/27/2023] Open
Abstract
Somatic mutations in histone encoding genes result in gross alterations in the epigenetic landscape. Diffuse intrinsic pontine glioma (DIPG) is a pediatric high-grade glioma (pHGG) and one of the most challenging cancers to treat, with only 1% surviving for 5 years. Due to the location in the brainstem, DIPGs are difficult to resect and rapidly turn into a fatal disease. Over 80% of DIPGs confer mutations in genes coding for histone 3 variants (H3.3 or H3.1/H3.2), with lysine to methionine substitution at position 27 (H3K27M). This results in a global decrease in H3K27 trimethylation, increased H3K27 acetylation, and widespread oncogenic changes in gene expression. Epigenetic modifying drugs emerge as promising candidates to treat DIPG, with histone deacetylase (HDAC) inhibitors taking the lead in preclinical and clinical studies. However, some data show the evolving resistance of DIPGs to the most studied HDAC inhibitor panobinostat and highlight the need to further investigate its mechanism of action. A new forceful line of research explores the simultaneous use of multiple inhibitors that could target epigenetically induced changes in DIPG chromatin and enhance the anticancer response of single agents. In this review, we summarize the therapeutic approaches against H3K27M-expressing pHGGs focused on targeting epigenetic dysregulation and highlight promising combinatorial drug treatments. We assessed the effectiveness of the epigenetic drugs that are already in clinical trials in pHGGs. The constantly expanding understanding of the epigenetic vulnerabilities of H3K27M-expressing pHGGs provides new tumor-specific targets, opens new possibilities of therapy, and gives hope to find a cure for this deadly disease.
Collapse
Affiliation(s)
- Katarzyna B Leszczynska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Chinchu Jayaprakash
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Jakub Mieczkowski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland.,3P-Medicine Laboratory, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
30
|
Atypical Teratoid Rhabdoid Tumours Are Susceptible to Panobinostat-Mediated Differentiation Therapy. Cancers (Basel) 2021; 13:cancers13205145. [PMID: 34680294 PMCID: PMC8534272 DOI: 10.3390/cancers13205145] [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: 08/30/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Atypical teratoid rhabdoid tumour (ATRT) is an aggressive undifferentiated malignancy of the central nervous system in children. A defining feature of ATRT is the loss of the SMARCB1 gene that is essential for regulating gene expression required for normal developmental processes. We show that treatment of human ATRT cell models with the histone deacetylate inhibitor, panobinostat, inhibits tumour growth, reactivates the expression of developmental genes, and drives neuronal differentiation. These results demonstrate the therapeutic potential of panobinostat for the treatment of ATRT. Abstract Atypical teratoid rhabdoid tumour (ATRT) is a rare but highly aggressive undifferentiated solid tumour arising in the central nervous system and predominantly affecting infants and young children. ATRT is exclusively characterized by the inactivation of SMARCB1, a member of the SWI/SNF chromatin remodelling complex that is essential for the regulation of large sets of genes required for normal development and differentiation. Histone deacetylase inhibitors (HDACi) are a promising anticancer therapy and are able to mimic the normal acetylation functions of SMARCB1 in SMARCB1-deficient cells and drive multilineage differentiation in extracranial rhabdoid tumours. However, the potential efficacy of HDACi in ATRT is unknown. Here, we show that human ATRT cells are highly responsive to the HDACi panobinostat and that sustained treatment leads to growth arrest, increased cell senescence, decreased clonogenicity and induction of a neurogenesis gene-expression profile. Furthermore, in an orthotopic ATRT xenograft model, continuous panobinostat treatment inhibits tumour growth, increases survival and drives neuronal differentiation as shown by the expression of the neuronal marker, TUJ1. Collectively, this preclinical study supports the therapeutic potential of panobinostat-mediated differentiation therapy for ATRT.
Collapse
|
31
|
Alghamdi M, Gumbleton M, Newland B. Local delivery to malignant brain tumors: potential biomaterial-based therapeutic/adjuvant strategies. Biomater Sci 2021; 9:6037-6051. [PMID: 34357362 DOI: 10.1039/d1bm00896j] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is the most aggressive malignant brain tumor and is associated with a very poor prognosis. The standard treatment for newly diagnosed patients involves total tumor surgical resection (if possible), plus irradiation and adjuvant chemotherapy. Despite treatment, the prognosis is still poor, and the tumor often recurs within two centimeters of the original tumor. A promising approach to improving the efficacy of GBM therapeutics is to utilize biomaterials to deliver them locally at the tumor site. Local delivery to GBM offers several advantages over systemic administration, such as bypassing the blood-brain barrier and increasing the bioavailability of the therapeutic at the tumor site without causing systemic toxicity. Local delivery may also combat tumor recurrence by maintaining sufficient drug concentrations at and surrounding the original tumor area. Herein, we critically appraised the literature on local delivery systems based within the following categories: polymer-based implantable devices, polymeric injectable systems, and hydrogel drug delivery systems. We also discussed the negative effect of hypoxia on treatment strategies and how one might utilize local implantation of oxygen-generating biomaterials as an adjuvant to enhance current therapeutic strategies.
Collapse
Affiliation(s)
- Majed Alghamdi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK. and Faculty of Pharmacy, King Abdulaziz University, Jeddah, 22522, Kingdom of Saudi Arabia
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK. and Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| |
Collapse
|
32
|
Heravi Shargh V, Luckett J, Bouzinab K, Paisey S, Turyanska L, Singleton WGB, Lowis S, Gershkovich P, Bradshaw TD, Stevens MFG, Bienemann A, Coyle B. Chemosensitization of Temozolomide-Resistant Pediatric Diffuse Midline Glioma Using Potent Nanoencapsulated Forms of a N(3)-Propargyl Analogue. ACS APPLIED MATERIALS & INTERFACES 2021; 13:35266-35280. [PMID: 34310112 DOI: 10.1021/acsami.1c04164] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The lack of clinical response to the alkylating agent temozolomide (TMZ) in pediatric diffuse midline/intrinsic pontine glioma (DIPG) has been associated with O6-methylguanine-DNA-methyltransferase (MGMT) expression and mismatch repair deficiency. Hence, a potent N(3)-propargyl analogue (N3P) was derived, which not only evades MGMT but also remains effective in mismatch repair deficient cells. Due to the poor pharmacokinetic profile of N3P (t1/2 < 1 h) and to bypass the blood-brain barrier, we proposed convection enhanced delivery (CED) as a method of administration to decrease dose and systemic toxicity. Moreover, to enhance N3P solubility, stability, and sustained distribution in vivo, either it was incorporated into an apoferritin (AFt) nanocage or its sulfobutyl ether β-cyclodextrin complex was loaded into nanoliposomes (Lip). The resultant AFt-N3P and Lip-N3P nanoparticles (NPs) had hydrodynamic diameters of 14 vs 93 nm, icosahedral vs spherical morphology, negative surface charge (-17 vs -34 mV), and encapsulating ∼630 vs ∼21000 N3P molecules per NP, respectively. Both NPs showed a sustained release profile and instant uptake within 1 h incubation in vitro. In comparison to the naked drug, N3P NPs demonstrated stronger anticancer efficacy against 2D TMZ-resistant DIPG cell cultures [IC50 = 14.6 (Lip-N3P) vs 32.8 μM (N3P); DIPG-IV) and (IC50 = 101.8 (AFt-N3P) vs 111.9 μM (N3P); DIPG-VI)]. Likewise, both N3P-NPs significantly (P < 0.01) inhibited 3D spheroid growth compared to the native N3P in MGMT+ DIPG-VI (100 μM) and mismatch repair deficient DIPG-XIX (50 μM) cultures. Interestingly, the potency of TMZ was remarkably enhanced when encapsulated in AFt NPs against DIPG-IV, -VI, and -XIX spheroid cultures. Dynamic PET scans of CED-administered zirconium-89 (89Zr)-labeled AFt-NPs in rats also demonstrated substantial enhancement over free 89Zr radionuclide in terms of localized distribution kinetics and retention within the brain parenchyma. Overall, both NP formulations of N3P represent promising approaches for treatment of TMZ-resistant DIPG and merit the next phase of preclinical evaluation.
Collapse
Affiliation(s)
| | | | | | - Stephen Paisey
- Wales Research and Diagnostic PET Imaging Centre, School of Medicine, Cardiff University, Cardiff, CF14 4XN, United Kingdom
| | - Lyudmila Turyanska
- Faculty of Engineering, University of Nottingham, Nottingham, Nottinghamshire NG7 2RD, United Kingdom
| | - William G B Singleton
- Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | | | | | | | | | - Alison Bienemann
- Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | | |
Collapse
|
33
|
Van Laar AD, Van Laar VS, San Sebastian W, Merola A, Bradley Elder J, Lonser RR, Bankiewicz KS. An Update on Gene Therapy Approaches for Parkinson's Disease: Restoration of Dopaminergic Function. JOURNAL OF PARKINSONS DISEASE 2021; 11:S173-S182. [PMID: 34366374 PMCID: PMC8543243 DOI: 10.3233/jpd-212724] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
At present there is a significant unmet need for clinically available treatments for Parkinson’s disease (PD) patients to stably restore balance to dopamine network function, leaving patients with inadequate management of symptoms as the disease progresses. Gene therapy is an attractive approach to impart a durable effect on neuronal function through introduction of genetic material to reestablish dopamine levels and/or functionally recover dopaminergic signaling by improving neuronal health. Ongoing clinical gene therapy trials in PD are focused on enzymatic enhancement of dopamine production and/or the restoration of the nigrostriatal pathway to improve dopaminergic network function. In this review, we discuss data from current gene therapy trials for PD and recent advances in study design and surgical approaches.
Collapse
Affiliation(s)
- Amber D Van Laar
- Asklepios BioPharmaceutical, Inc., Columbus, OH, USA.,Department of Neurology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Victor S Van Laar
- Department of Neurological Surgery, Ohio State University College of Medicine, Columbus, OH, USA
| | - Waldy San Sebastian
- Asklepios BioPharmaceutical, Inc., Columbus, OH, USA.,Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Aristide Merola
- Department of Neurology, College of Medicine, the Ohio State University, Columbus, OH, USA
| | - J Bradley Elder
- Department of Neurological Surgery, Ohio State University College of Medicine, Columbus, OH, USA
| | - Russell R Lonser
- Department of Neurological Surgery, Ohio State University College of Medicine, Columbus, OH, USA
| | - Krystof S Bankiewicz
- Department of Neurological Surgery, Ohio State University College of Medicine, Columbus, OH, USA.,Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
34
|
Antibody-drug conjugates for H3K27M-mutant diffuse midline gliomas: prospects and challenges. Ther Deliv 2021; 12:553-557. [PMID: 34286602 DOI: 10.4155/tde-2021-0045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
35
|
Harttrampf AC, da Costa MEM, Renoult A, Daudigeos-Dubus E, Geoerger B. Histone deacetylase inhibitor panobinostat induces antitumor activity in epithelioid sarcoma and rhabdoid tumor by growth factor receptor modulation. BMC Cancer 2021; 21:833. [PMID: 34281526 PMCID: PMC8290558 DOI: 10.1186/s12885-021-08579-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 07/10/2021] [Indexed: 01/06/2023] Open
Abstract
Background Epithelioid sarcomas and rhabdoid tumors are rare, aggressive malignancies with poor prognosis. Both are characterized by INI1 alterations and deregulation of growth factor receptors albeit their interaction has not been elucidated. Methods In this study, we investigated the activity of a panel of epigenetic modulators and receptor tyrosine kinase inhibitors in vitro on respective cell lines as well as on primary patient-derived epithelioid sarcoma cells, and in vivo on xenografted mice. Focusing on histone deacetylase (HDAC) inhibitors, we studied the mechanism of action of this class of agents, its effect on growth factor receptor regulation, and changes in epithelial-to-mesenchymal transition by using cell- and RT-qPCR-based assays. Results Pan-HDAC inhibitor panobinostat exhibited potent anti-proliferative activity at low nanomolar concentrations in A204 rhabdoid tumor, and VAESBJ/GRU1 epithelioid sarcoma cell lines, strongly induced apoptosis, and resulted in significant tumor growth inhibition in VAESBJ xenografts. It differentially regulated EGFR, FGFR1 and FGFR2, leading to downregulation of EGFR in epithelioid sarcoma and to mesenchymal-to-epithelial transition whereas in rhabdoid tumor cells, EGFR was strongly upregulated and reinforced the mesenchymal phenotype. All three cell lines were rendered more susceptible towards combination with EGFF inhibitor erlotinib, further enhancing apoptosis. Conclusions HDAC inhibitors exhibit significant anticancer activity due to their multifaceted actions on cytotoxicity, differentiation and drug sensitization. Our data suggest that the tailored, tissue-specific combination of HDAC inhibitors with therapeutics which target cellular salvage mechanisms might increase their therapeutic relevance. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08579-w.
Collapse
Affiliation(s)
- Anne Catherine Harttrampf
- Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France.,Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | | | - Aline Renoult
- Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France.,Present address: Institute of Research in Immunology and Cancer, Dr Trang Hoang Laboratory, Université de Montréal, Montreal, Québec, Canada
| | - Estelle Daudigeos-Dubus
- Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France.,Present address: AP-HP Nord, DMU Neurosciences, Service de Neurologie, FHU NeuroVasc, Université de Paris, Paris, France
| | - Birgit Geoerger
- Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France. .,Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, 114 Rue Edouard Vaillant, 94805, Villejuif, France.
| |
Collapse
|
36
|
Patel JP, Spiller SE, Barker ED. Drug penetration in pediatric brain tumors: Challenges and opportunities. Pediatr Blood Cancer 2021; 68:e28983. [PMID: 33719183 DOI: 10.1002/pbc.28983] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/23/2022]
Abstract
Larger clinical trial enrollments and a greater understanding of biological heterogeneity have led to improved survival rates for children diagnosed with brain tumors in the last 50 years. However, reducing long-term morbidities and improving survival rates of high-risk tumors remain major challenges. Chemotherapy can reduce tumor burden, but effective drug penetration at the tumor site is limited by barriers in the route of drug administration and within the tumor microenvironment. Bioavailability of drugs is impeded by the blood-brain barrier, plasma protein binding, and structural components by the tumor including the matrix and vasculature contributing to increased interstitial fluid pressure, hypoxia, and acidity. Designing drug delivery systems to circumvent these barriers could lead to improved drug penetration at the tumor site and reduce adverse systemic side effects. In this review, we expand on how systemic and local barriers limit drug penetration and present potential methods to enhance drug penetration in pediatric brain tumors.
Collapse
Affiliation(s)
- Jenny P Patel
- Department of Mechanical, Aerospace, and Biomedical Engineering, The University of Tennessee at Knoxville, Knoxville, Tennessee
| | - Susan E Spiller
- Pediatric Hematology/Oncology, East Tennessee Children's Hospital, Knoxville, Tennessee
| | - Elizabeth D Barker
- Department of Mechanical, Aerospace, and Biomedical Engineering, The University of Tennessee at Knoxville, Knoxville, Tennessee
| |
Collapse
|
37
|
Chaudhuri S, Fowler MJ, Baker C, Stopka SA, Regan MS, Sablatura L, Broughton CW, Knight BE, Stabenfeldt SE, Agar NYR, Sirianni RW. β-Cyclodextrin-poly (β-Amino Ester) Nanoparticles Are a Generalizable Strategy for High Loading and Sustained Release of HDAC Inhibitors. ACS APPLIED MATERIALS & INTERFACES 2021; 13:20960-20973. [PMID: 33905245 PMCID: PMC8153536 DOI: 10.1021/acsami.0c22587] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Therapeutic development of histone deacetylase inhibitors (HDACi) has been hampered by a number of barriers to drug delivery, including poor solubility and inadequate tissue penetration. Nanoparticle encapsulation could be one approach to improve the delivery of HDACi to target tissues; however, effective and generalizable loading of HDACi within nanoparticle systems remains a long-term challenge. We hypothesized that the common terminally ionizable moiety on many HDACi molecules could be capitalized upon for loading in polymeric nanoparticles. Here, we describe the simple, efficient formulation of a novel library of β-cyclodextrin-poly (β-amino ester) networks (CDN) to achieve this goal. We observed that network architecture was a critical determinant of CDN encapsulation of candidate molecules, with a more hydrophobic core enabling effective self-assembly and a PEGylated surface enabling high loading (up to ∼30% w/w), effective self-assembly of the nanoparticle, and slow release of drug into aqueous media (up to 24 days) for the model HDACi panobinostat. We next constructed a library of CDNs to encapsulate various small, hydrophobic, terminally ionizable molecules (panobinostat, quisinostat, dacinostat, givinostat, bortezomib, camptothecin, nile red, and cytarabine), which yielded important insights into the structural requirements for effective drug loading and CDN self-assembly. Optimized CDN nanoparticles were taken up by GL261 cells in culture and a released panobinostat was confirmed to be bioactive. Panobinostat-loaded CDNs were next administered by convection-enhanced delivery (CED) to mice bearing intracranial GL261 tumors. These studies confirm that CDN encapsulation enables a higher deliverable dose of drug to effectively slow tumor growth. Matrix-assisted laser desorption/ionization (MALDI) analysis on tissue sections confirms higher exposure of tumor to drug, which likely accounts for the therapeutic effects. Taken in sum, these studies present a novel nanocarrier platform for encapsulation of HDACi via both ionic and hydrophobic interactions, which is an important step toward better treatment of disease via HDACi therapy.
Collapse
Affiliation(s)
- Sauradip Chaudhuri
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center at Houston, Houston, Texas 77030, United States
| | - Martha J. Fowler
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center at Houston, Houston, Texas 77030, United States
| | - Cassandra Baker
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center at Houston, Houston, Texas 77030, United States
| | - Sylwia A. Stopka
- Department
of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Michael S. Regan
- Department
of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Lindsey Sablatura
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center at Houston, Houston, Texas 77030, United States
| | - Colton W. Broughton
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center at Houston, Houston, Texas 77030, United States
| | - Brandon E. Knight
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center at Houston, Houston, Texas 77030, United States
| | - Sarah E. Stabenfeldt
- School
of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85281, United States
| | - Nathalie Y. R. Agar
- Department
of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Rachael W. Sirianni
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center at Houston, Houston, Texas 77030, United States
- School
of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85281, United States
| |
Collapse
|
38
|
Convection-enhanced delivery for H3K27M diffuse midline glioma: how can we efficaciously modulate the blood-brain barrier? Ther Deliv 2021; 12:419-422. [PMID: 33949200 DOI: 10.4155/tde-2021-0026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Graphical abstract [Formula: see text].
Collapse
|
39
|
Vitanza NA, Biery MC, Myers C, Ferguson E, Zheng Y, Girard EJ, Przystal JM, Park G, Noll A, Pakiam F, Winter CA, Morris SM, Sarthy J, Cole BL, Leary SES, Crane C, Lieberman NAP, Mueller S, Nazarian J, Gottardo R, Brusniak MY, Mhyre AJ, Olson JM. Optimal therapeutic targeting by HDAC inhibition in biopsy-derived treatment-naïve diffuse midline glioma models. Neuro Oncol 2021; 23:376-386. [PMID: 33130903 DOI: 10.1093/neuonc/noaa249] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Diffuse midline gliomas (DMGs), including diffuse intrinsic pontine gliomas (DIPGs), have a dismal prognosis, with less than 2% surviving 5 years postdiagnosis. The majority of DIPGs and all DMGs harbor mutations altering the epigenetic regulatory histone tail (H3 K27M). Investigations addressing DMG epigenetics have identified a few promising drugs, including the HDAC inhibitor (HDACi) panobinostat. Here, we use clinically relevant DMG models to identify and validate other effective HDACi and their biomarkers of response. METHODS HDAC inhibitors were tested across biopsy-derived treatment-naïve in vitro and in vivo DMG models with biologically relevant radiation resistance. RNA sequencing was performed to define and compare drug efficacy and to map predictive biomarkers of response. RESULTS Quisinostat and romidepsin showed efficacy with low nanomolar half-maximal inhibitory concentration (IC50) values (~50 and ~5 nM, respectively). Comparative transcriptome analyses across quisinostat, romidepsin, and panobinostat showed a greater degree of shared biological effects between quisinostat and panobinostat, and less overlap with romidepsin. However, some transcriptional changes were consistent across all 3 drugs at similar biologically effective doses, such as overexpression of troponin T1 slow skeletal type (TNNT1) and downregulation of collagen type 20 alpha 1 chain (COL20A1), identifying these as potential vulnerabilities or on-target biomarkers in DMG. Quisinostat and romidepsin significantly (P < 0.0001) inhibited in vivo tumor growth. CONCLUSIONS Our data highlight the utility of treatment-naïve biopsy-derived models; establishes quisinostat and romidepsin as effective in vivo; illuminates potential mechanisms and/or biomarkers of DMG cell lethality due to HDAC inhibition; and emphasizes the need for brain tumor-penetrant versions of potentially efficacious agents.
Collapse
Affiliation(s)
- Nicholas A Vitanza
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, Washington, USA
| | - Matt C Biery
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Carrie Myers
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Eric Ferguson
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Ye Zheng
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Emily J Girard
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Giulia Park
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Alyssa Noll
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Molecular and Cellular Biology Graduate Program and Medical Scientist Training Program, University of Washington, Seattle, Washington, USA
| | - Fiona Pakiam
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Conrad A Winter
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Shelli M Morris
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jay Sarthy
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Bonnie L Cole
- Department of Laboratories, Seattle Children's Hospital, Seattle, Washington, USA.,Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Sarah E S Leary
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, Washington, USA
| | - Courtney Crane
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Nicole A P Lieberman
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Sabine Mueller
- University Children's Hospital Zurich, Zurich, Switzerland.,University of California San Francisco, San Francisco, California, USA
| | - Javad Nazarian
- University Children's Hospital Zurich, Zurich, Switzerland.,Department of Genetic Medicine Research, Children's National Medical Center, Washington DC, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Statistics, University of Washington, Seattle, Washington, USA
| | - Mi-Youn Brusniak
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Andrew J Mhyre
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - James M Olson
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Division of Pediatric Hematology/Oncology, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, Washington, USA
| |
Collapse
|
40
|
Wei X, Xiao B, Wang L, Zang L, Che F. Potential new targets and drugs related to histone modifications in glioma treatment. Bioorg Chem 2021; 112:104942. [PMID: 33965781 DOI: 10.1016/j.bioorg.2021.104942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023]
Abstract
Glioma accounts for 40-50% of craniocerebral tumors, whose outcome rarely improves after standard treatment. The development of new therapeutic targets for glioma treatment has important clinical significance. With the deepening of research on gliomas, recent researchers have found that the occurrence and development of gliomas is closely associated with histone modifications, including methylation, acetylation, phosphorylation, and ubiquitination. Additionally, evidence has confirmed the close relationship between histone modifications and temozolomide (TMZ) resistance. Therefore, histone modification-related proteins have been widely recognized as new therapeutic targets for glioma treatment. In this review, we summarize the potential histone modification-associated targets and related drugs for glioma treatment. We have further clarified how histone modifications regulate the pathogenesis of gliomas and the mechanism of drug action, providing novel insights for the current clinical glioma treatment. Herein, we have also highlighted the limitations of current clinical therapies and have suggested future research directions and expected advances in potential areas of disease prognosis. Due to the complicated glioma pathogenesis, in the present review, we have acknowledged the limitations of histone modification applications in the related clinical treatment.
Collapse
Affiliation(s)
- Xiuhong Wei
- Graduate School, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, China; Department of Neurology, Linyi People's Hospital, Shandong University, Linyi, Shandong, China
| | - Bolian Xiao
- Central Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong, China; Key Laboratory of Neurophysiology, Key Laboratory of Tumor Biology, Linyi, Shandong, China
| | - Liying Wang
- Department of Neurology, Linyi People's Hospital, Shandong University, Linyi, Shandong, China; Department of Neurology, the Clinical Medical College of Weifang Medical College, Weifang, Shandong, China
| | - Lanlan Zang
- Central Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong, China; Key Laboratory of Neurophysiology, Key Laboratory of Tumor Biology, Linyi, Shandong, China; Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, China.
| | - Fengyuan Che
- Graduate School, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, China; Department of Neurology, Linyi People's Hospital, Shandong University, Linyi, Shandong, China; Central Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong, China; Key Laboratory of Neurophysiology, Key Laboratory of Tumor Biology, Linyi, Shandong, China.
| |
Collapse
|
41
|
Ung C, Tsoli M, Liu J, Cassano D, Pocoví-Martínez S, Upton DH, Ehteda A, Mansfeld FM, Failes TW, Farfalla A, Katsinas C, Kavallaris M, Arndt GM, Vittorio O, Cirillo G, Voliani V, Ziegler DS. Doxorubicin-Loaded Gold Nanoarchitectures as a Therapeutic Strategy against Diffuse Intrinsic Pontine Glioma. Cancers (Basel) 2021; 13:1278. [PMID: 33805713 PMCID: PMC7999568 DOI: 10.3390/cancers13061278] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 01/11/2023] Open
Abstract
Diffuse Intrinsic Pontine Gliomas (DIPGs) are highly aggressive paediatric brain tumours. Currently, irradiation is the only standard treatment, but is palliative in nature and most patients die within 12 months of diagnosis. Novel therapeutic approaches are urgently needed for the treatment of this devastating disease. We have developed non-persistent gold nano-architectures (NAs) functionalised with human serum albumin (HSA) for the delivery of doxorubicin. Doxorubicin has been previously reported to be cytotoxic in DIPG cells. In this study, we have preclinically evaluated the cytotoxic efficacy of doxorubicin delivered through gold nanoarchitectures (NAs-HSA-Dox). We found that DIPG neurospheres were equally sensitive to doxorubicin and doxorubicin-loaded NAs. Colony formation assays demonstrated greater potency of NAs-HSA-Dox on colony formation compared to doxorubicin. Western blot analysis indicated increased apoptotic markers cleaved Parp, cleaved caspase 3 and phosphorylated H2AX in NAs-HSA-Dox treated DIPG neurospheres. Live cell content and confocal imaging demonstrated significantly higher uptake of NAs-HSA-Dox into DIPG neurospheres compared to doxorubicin alone. Despite the potency of the NAs in vitro, treatment of an orthotopic model of DIPG showed no antitumour effect. This disparate outcome may be due to the integrity of the blood-brain barrier and highlights the need to develop therapies to enhance penetration of drugs into DIPG.
Collapse
Affiliation(s)
- Caitlin Ung
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
| | - Maria Tsoli
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Jie Liu
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
| | - Domenico Cassano
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy; (D.C.); (S.P.-M.); (V.V.)
| | - Salvador Pocoví-Martínez
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy; (D.C.); (S.P.-M.); (V.V.)
| | - Dannielle H. Upton
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Anahid Ehteda
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
| | - Friederike M. Mansfeld
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Royal Parade, Parkville, VIC 3052, Australia
| | - Timothy W. Failes
- ACRF Drug Discovery Centre, Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (T.W.F.); (G.M.A.)
| | - Annafranca Farfalla
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy; (A.F.); (G.C.)
| | - Christopher Katsinas
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
| | - Maria Kavallaris
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Greg M. Arndt
- ACRF Drug Discovery Centre, Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (T.W.F.); (G.M.A.)
| | - Orazio Vittorio
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Giuseppe Cirillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy; (A.F.); (G.C.)
| | - Valerio Voliani
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy; (D.C.); (S.P.-M.); (V.V.)
| | - David S. Ziegler
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- Kids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW 2052, Australia
| |
Collapse
|
42
|
Bander ED, Tizi K, Wembacher-Schroeder E, Thomson R, Donzelli M, Vasconcellos E, Souweidane MM. Deformational changes after convection-enhanced delivery in the pediatric brainstem. Neurosurg Focus 2021; 48:E3. [PMID: 31896089 DOI: 10.3171/2019.10.focus19679] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 10/07/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE In the brainstem, there are concerns regarding volumetric alterations following convection-enhanced delivery (CED). The relationship between distribution volume and infusion volume is predictably greater than one. Whether this translates into deformational changes and influences clinical management is unknown. As part of a trial using CED for diffuse intrinsic pontine glioma (DIPG), the authors measured treatment-related volumetric alterations in the brainstem and ventricles. METHODS Enrolled patients underwent a single infusion of radioimmunotherapy. Between 2012 and 2019, 23 patients with volumetric pre- and postoperative day 1 (POD1) and day 30 (POD30) MRI scans were analyzed using iPlan® Flow software for semiautomated volumetric measurements of the ventricles and pontine segment of the brainstem. RESULTS Children in the study had a mean age of 7.7 years (range 2-18 years). The mean infusion volume was 3.9 ± 1.7 ml (range 0.8-8.8 ml). Paired t-tests demonstrated a significant increase in pontine volume immediately following infusion (p < 0.0001), which trended back toward baseline by POD30 (p = 0.046; preoperative 27.6 ± 8.4 ml, POD1 30.2 ± 9.0 ml, POD30 29.5 ± 9.4 ml). Lateral ventricle volume increased (p = 0.02) and remained elevated on POD30 (p = 0.04; preoperative 23.5 ± 15.4 ml, POD1 26.3 ± 16.0, POD30 28.6 ± 21.2). Infusion volume had a weak, positive correlation with pontine and lateral ventricle volume change (r2 = 0.22 and 0.27, respectively). Four of the 23 patients had an increase in preoperative neurological deficits at POD30. No patients required shunt placement within 90 days. CONCLUSIONS CED infusion into the brainstem correlates with immediate but self-limited deformation changes in the pons. The persistence of increased ventricular volume and no need for CSF diversion post-CED are inconsistent with obstructive hydrocephalus. Defining the degree and time course of these deformational changes can assist in the interpretation of neuroimaging along the DIPG disease continuum when CED is incorporated into the treatment algorithm.
Collapse
Affiliation(s)
- Evan D Bander
- 1Department of Neurological Surgery, Weill Medical College of Cornell University, New York, New York.,2Department of Neurological Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Karima Tizi
- 3Department of Neurosurgery, Hôpitaux Universitaires de Genève, Geneva, Switzerland; and
| | | | | | - Maria Donzelli
- 2Department of Neurological Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Mark M Souweidane
- 1Department of Neurological Surgery, Weill Medical College of Cornell University, New York, New York.,2Department of Neurological Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
43
|
Rechberger JS, Power EA, Lu VM, Zhang L, Sarkaria JN, Daniels DJ. Evaluating infusate parameters for direct drug delivery to the brainstem: a comparative study of convection-enhanced delivery versus osmotic pump delivery. Neurosurg Focus 2021; 48:E2. [PMID: 31896090 DOI: 10.3171/2019.10.focus19703] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Convection-enhanced delivery (CED) and osmotic pump delivery both have been promoted as promising techniques to deliver drugs to pediatric diffuse intrinsic pontine gliomas (DIPGs). Correspondingly, the aim of this study was to understand how infusate molecular weight (MW), duration of delivery, and mechanism of delivery (CED or osmotic pump) affect volume of distribution (Vd) in the brainstem, to better inform drug selection and delivery in future DIPG investigations. METHODS A series of in vivo experiments were conducted using rat models. CED and osmotic pump delivery systems were surgically implanted in the brainstem, and different MW fluorescent dextran beads were infused either once (acute) or daily for 5 days (chronic) in a volume infused (Vi). Brainstems were harvested after the last infusion, and Vd was quantified using serial sectioning and fluorescence imaging. RESULTS Fluorescence imaging showed infusate uptake within the brainstem for both systems without complication. A significant inverse relationship was observed between infusate MW and Vd in all settings, which was distinctly exponential in nature in the setting of acute delivery across the 570-Da to 150-kDa range. Chronic duration and CED technique resulted in significantly greater Vd compared to acute duration or osmotic pump delivery, respectively. When accounting for Vi, acute infusion yielded significantly greater Vd/Vi than chronic infusion. The distribution in CED versus osmotic pump delivery was significantly affected by infusate MW at higher weights. CONCLUSIONS Here the authors demonstrate that infusate MW, duration of infusion, and infusion mechanism all impact the Vd of an infused agent and should be considered when selecting drugs and infusion parameters for novel investigations to treat DIPGs.
Collapse
Affiliation(s)
| | - Erica A Power
- 1Department of Neurologic Surgery, Mayo Clinic.,2Mayo Clinic Graduate School of Biomedical Sciences
| | - Victor M Lu
- 1Department of Neurologic Surgery, Mayo Clinic
| | - Liang Zhang
- 1Department of Neurologic Surgery, Mayo Clinic
| | | | - David J Daniels
- 1Department of Neurologic Surgery, Mayo Clinic.,4Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
44
|
Wummer B, Woodworth D, Flores C. Brain stem gliomas and current landscape. J Neurooncol 2021; 151:21-28. [PMID: 33398531 DOI: 10.1007/s11060-020-03655-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 10/24/2020] [Indexed: 11/24/2022]
Abstract
PURPOSE CNS malignancies are currently the most common cause of disease related deaths in children. Although brainstem gliomas are invariably fatal cancers in children, clinical studies against this disease are limited. This review is to lead to a succinct collection of knowledge of known biological mechanisms of this disease and discuss available therapeutics. METHODS A hallmark of brainstem gliomas are mutations in the histone H3.3 with the majority of cases expressing the mutation K27M on histone 3.3. Recent studies using whole genome sequencing have revealed other mutations associated with disease. Current standard clinical practice may merely involve radiation and/or chemotherapy with little hope for long term survival. Here we discuss the potential of new therapies. CONCLUSION Despite the lack of treatment options using frequently practiced clinical techniques, immunotherapeutic strategies have recently been developed to target brainstem gliomas. To target brainstem gliomas, investigators are evaluating the use of broad non-targeted therapy with immune checkpoint inhibitors. Alternatively, others have begun to explore adoptive T cell strategies against these fatal malignancies.
Collapse
Affiliation(s)
- Brandon Wummer
- Lillian S. Wells Department of Neurosurgery, University of Florida Health Center, Gainesville, FL, 32610, USA
| | - Delaney Woodworth
- Lillian S. Wells Department of Neurosurgery, University of Florida Health Center, Gainesville, FL, 32610, USA
| | - Catherine Flores
- Lillian S. Wells Department of Neurosurgery, University of Florida Health Center, Gainesville, FL, 32610, USA.
| |
Collapse
|
45
|
Griffith JI, Rathi S, Zhang W, Zhang W, Drewes LR, Sarkaria JN, Elmquist WF. Addressing BBB Heterogeneity: A New Paradigm for Drug Delivery to Brain Tumors. Pharmaceutics 2020; 12:E1205. [PMID: 33322488 PMCID: PMC7763839 DOI: 10.3390/pharmaceutics12121205] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
Effective treatments for brain tumors remain one of the most urgent and unmet needs in modern oncology. This is due not only to the presence of the neurovascular unit/blood-brain barrier (NVU/BBB) but also to the heterogeneity of barrier alteration in the case of brain tumors, which results in what is referred to as the blood-tumor barrier (BTB). Herein, we discuss this heterogeneity, how it contributes to the failure of novel pharmaceutical treatment strategies, and why a "whole brain" approach to the treatment of brain tumors might be beneficial. We discuss various methods by which these obstacles might be overcome and assess how these strategies are progressing in the clinic. We believe that by approaching brain tumor treatment from this perspective, a new paradigm for drug delivery to brain tumors might be established.
Collapse
Affiliation(s)
- Jessica I. Griffith
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| | - Sneha Rathi
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| | - Wenqiu Zhang
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| | - Wenjuan Zhang
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| | - Lester R. Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School—Duluth, Duluth, MN 55812, USA;
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55902, USA;
| | - William F. Elmquist
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (S.R.); (W.Z.); (W.Z.)
| |
Collapse
|
46
|
Bellat V, Alcaina Y, Tung CH, Ting R, Michel AO, Souweidane M, Law B. A combined approach of convection-enhanced delivery of peptide nanofiber reservoir to prolong local DM1 retention for diffuse intrinsic pontine glioma treatment. Neuro Oncol 2020; 22:1495-1504. [PMID: 32301996 PMCID: PMC7566426 DOI: 10.1093/neuonc/noaa101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Diffuse intrinsic pontine glioma (DIPG) is a highly lethal malignancy that occurs predominantly in children. DIPG is inoperable and post-diagnosis survival is less than 1 year, as conventional chemotherapy is ineffective. The intact blood-brain barrier (BBB) blocks drugs from entering the brain. Convection-enhanced delivery (CED) is a direct infusion technique delivering drugs to the brain, but it suffers from rapid drug clearance. Our goal is to overcome the delivery barrier via CED and maintain a therapeutic concentration at the glioma site with a payload-adjustable peptide nanofiber precursor (NFP) that displays a prolonged retention property as a drug carrier. METHODS The post-CED retention of 89Zr-NFP was determined in real time using PET/CT imaging. Emtansine (DM1), a microtubule inhibitor, was conjugated to NFP. The cytotoxicity of the resulting DM1-NFP was tested against patient-derived DIPG cell lines. The therapeutic efficacy was evaluated in animals bearing orthotopic DIPG, according to glioma growth (measured using bioluminescence imaging) and the long-term survival. RESULTS DM1-NFP demonstrated potency against multiple glioma cell lines. The half-maximal inhibitory concentration values were in the nanomolar range. NFP remained at the infusion site (pons) for weeks, with a clearance half-life of 60 days. DM1-NFP inhibited glioma progression in animals, and offered a survival benefit (median survival of 62 days) compared with the untreated controls (28 days) and DM1-treated animal group (26 days). CONCLUSIONS CED, in combination with DM1-NFP, complementarily functions to bypass the BBB, prolong drug retention at the fusion site, and maintain an effective therapeutic effect against DIPG to improve treatment outcome.
Collapse
Affiliation(s)
- Vanessa Bellat
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Yago Alcaina
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Ching-Hsuan Tung
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Richard Ting
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Adam O Michel
- Laboratory of Comparative Pathology, Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, New York
| | - Mark Souweidane
- Department of Neurological Surgery, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, New York
| | - Benedict Law
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
| |
Collapse
|
47
|
Radio-Resistance and DNA Repair in Pediatric Diffuse Midline Gliomas. Cancers (Basel) 2020; 12:cancers12102813. [PMID: 33007840 PMCID: PMC7600397 DOI: 10.3390/cancers12102813] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Malignant gliomas (MG) are among the most prevalent and lethal primary intrinsic brain tumors. Although radiotherapy (RT) is the most effective nonsurgical therapy, recurrence is universal. Dysregulated DNA damage response pathway (DDR) signaling, rampant genomic instability, and radio-resistance are among the hallmarks of MGs, with current therapies only offering palliation. A subgroup of pediatric high-grade gliomas (pHGG) is characterized by H3K27M mutation, which drives global loss of di- and trimethylation of histone H3K27. Here, we review the most recent literature and discuss the key studies dissecting the molecular biology of H3K27M-mutated gliomas in children. We speculate that the aberrant activation and/or deactivation of some of the key components of DDR may be synthetically lethal to H3K27M mutation and thus can open novel avenues for effective therapeutic interventions for patients suffering from this deadly disease.
Collapse
|
48
|
Lospinoso Severini L, Ghirga F, Bufalieri F, Quaglio D, Infante P, Di Marcotullio L. The SHH/GLI signaling pathway: a therapeutic target for medulloblastoma. Expert Opin Ther Targets 2020; 24:1159-1181. [PMID: 32990091 DOI: 10.1080/14728222.2020.1823967] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Medulloblastoma (MB) is a heterogeneous tumor of the cerebellum that is divided into four main subgroups with distinct molecular and clinical features. Sonic Hedgehog MB (SHH-MB) is the most genetically understood and occurs predominantly in childhood. Current therapies consist of aggressive and non-targeted multimodal approaches that are often ineffective and cause long-term complications. These problems intensify the need to develop molecularly targeted therapies to improve outcome and reduce treatment-related morbidities. In this scenario, Hedgehog (HH) signaling, a developmental pathway whose deregulation is involved in the pathogenesis of several malignancies, has emerged as an attractive druggable pathway for SHH-MB therapy. AREAS COVERED This review provides an overview of the advancements in the HH antagonist research field. We place an emphasis on Smoothened (SMO) and glioma-associated oncogene homolog (GLI) inhibitors and immunotherapy approaches that are validated in preclinical SHH-MB models and that have therapeutic potential for MB patients. Literature from Pubmed and data reported on ClinicalTrial.gov up to August 2020 were considered. EXPERT OPINION Extensive-omics analysis has enhanced our knowledge and has transformed the way that MB is studied and managed. The clinical use of SMO antagonists has yet to be determined, however, future GLI inhibitors and multitargeting approaches are promising.
Collapse
Affiliation(s)
| | - Francesca Ghirga
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia , 00161, Rome, Italy
| | - Francesca Bufalieri
- Department of Molecular Medicine, University of Rome La Sapienza , 00161, Rome, Italy
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs, University of Rome La Sapienza, 00185 , Rome, Italy
| | - Paola Infante
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia , 00161, Rome, Italy
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, University of Rome La Sapienza , 00161, Rome, Italy.,Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome La Sapienza , 00161, Rome, Italy
| |
Collapse
|
49
|
Elleaume H, Barth RF, Rousseau J, Bobyk L, Balosso J, Yang W, Huo T, Nakkula R. Radiation therapy combined with intracerebral convection-enhanced delivery of cisplatin or carboplatin for treatment of the F98 rat glioma. J Neurooncol 2020; 149:193-208. [PMID: 32809095 DOI: 10.1007/s11060-020-03600-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/08/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND The purpose of this review is to summarize our own experimental studies carried out over a 13-year period of time using the F98 rat glioma as model for high grade gliomas. We evaluated a binary chemo-radiotherapeutic modality that combines either cisplatin (CDDP) or carboplatin, administered intracerebrally (i.c.) by means of convection-enhanced delivery (CED) or osmotic pumps, in combination with either synchrotron or conventional X-irradiation. METHODS F98 glioma cells were implanted stereotactically into the brains of syngeneic Fischer rats. Approximately 14 days later, either CDDP or carboplatin was administered i.c. by CED, followed 24 h later by radiotherapy using either a synchrotron or, subsequently, megavoltage linear accelerators (LINAC). RESULTS CDDP was administered at a dose of 3 µg in 5 µL, followed 24 h later with an irradiation dose of 15 Gy or carboplatin at a dose of 20 µg in 10 µL, followed 24 h later with 3 fractions of 8 Gy each, at the source at the European Synchrotron Radiation Facility (ESRF). This resulted in a median survival time (MeST) > 180 days with 33% long term survivors (LTS) for CDDP and a MeST > 60 days with 8 to 22% LTS, for carboplatin. Subsequently it became apparent that comparable survival data could be obtained with megavoltage X-irradiation using a LINAC source. The best survival data were obtained with a dose of 72 µg of carboplatin administered by means of Alzet® osmotic pumps over 7 days. This resulted in a MeST of > 180 days, with 55% LTS. Histopathologic examination of all the brains of the surviving rats revealed no residual tumor cells or evidence of significant radiation related effects. CONCLUSIONS The results obtained using this combination therapy has, to the best of our knowledge, yielded the most promising survival data ever reported using the F98 glioma model.
Collapse
Affiliation(s)
- Hélène Elleaume
- INSERM UA07 Team STROBE, ESRF, 71 Avenue des Martyrs, 38000, Grenoble, France.
- European Synchrotron Radiation Facility, ID17 Medical Beamline, 71 Avenue Martyrs, 38000, Grenoble, France.
| | - Rolf F Barth
- Department of Pathology, The Ohio State University, 4132 Graves Hall - 333 W. 10th Avenue, Columbus, OH, 43210, USA.
| | - Julia Rousseau
- INSERM UA07 Team STROBE, ESRF, 71 Avenue des Martyrs, 38000, Grenoble, France
- European Synchrotron Radiation Facility, ID17 Medical Beamline, 71 Avenue Martyrs, 38000, Grenoble, France
| | - Laure Bobyk
- INSERM UA07 Team STROBE, ESRF, 71 Avenue des Martyrs, 38000, Grenoble, France
- European Synchrotron Radiation Facility, ID17 Medical Beamline, 71 Avenue Martyrs, 38000, Grenoble, France
| | - Jacques Balosso
- INSERM UA07 Team STROBE, ESRF, 71 Avenue des Martyrs, 38000, Grenoble, France
- Service de Radiothérapie, Centre Hospitalier Universitaire Grenoble-Alpes, 38700, La Tronche, France
- Centre de lutte contre le Cancer F. Baclesse, 3 avenue du général Harris, 14000, Caen, France
| | - Weilian Yang
- Department of Pathology, The Ohio State University, 4132 Graves Hall - 333 W. 10th Avenue, Columbus, OH, 43210, USA
- Department of Neurosurgery, Suzhou Medical College, Suzhou, China
| | - Tianyao Huo
- Department of Pathology, The Ohio State University, 4132 Graves Hall - 333 W. 10th Avenue, Columbus, OH, 43210, USA
- Department of Health Outcomes and Policy, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Robin Nakkula
- Department of Pathology, The Ohio State University, 4132 Graves Hall - 333 W. 10th Avenue, Columbus, OH, 43210, USA
- Research Institute, Nationwide Children's Hospital, Columbus, OH, 43205, USA
| |
Collapse
|
50
|
Diffuse midline glioma: review of epigenetics. J Neurooncol 2020; 150:27-34. [DOI: 10.1007/s11060-020-03553-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/01/2020] [Indexed: 10/23/2022]
|