1
|
Bakhtary M, Sharma P, Raspin S, Vemuri S, McCarthy M. Palliative Care for Children and Adolescents with Non-curative Brain Tumors: A Scoping Review. J Palliat Med 2025. [PMID: 40293742 DOI: 10.1089/jpm.2024.0203] [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: 04/30/2025] Open
Abstract
Children with non-curative brain tumors have a predictable and burdensome neurological decline. Care provision for these children falls in the gap where the acute inpatient services intersect with community-based providers. These children commonly receive care in the community; however, their neurological decline often outpaces engagement with community-based providers. Children with non-curative brain tumors are commonly referred to specialist palliative care services and need timely availability and access to this support. The purpose of this scoping review was to explore the evidence related to standards, guidelines, and models of palliative care provision for children with non-curative brain tumors, and to examine the facilitators and barriers to the implementation of, and access to, these services. Using a narrative synthesis method, a scoping review was conducted according to the Joanna Briggs Institute methodology using six electronic databases (Medline, Embase, Web of Science, Pubmed, CINAHL, and Cochrane Database of Systematic Reviews) from January 2013 to May 2023. A total of 2404 studies were screened for eligibility and 31 were selected for data extraction. There are no international models or standards of care for providing palliative care for children with non-curative brain tumors. Instead, palliative care is delivered at an institutional level, potentially leading to variability in the care that is provided. This variability can threaten the quality of life of these children and their families. Variability in care provision could be minimized by development of standardized palliative care provision. Any proposed standard for palliative care provision for children with non-curative brain tumors should include early integration of palliative care, and allocation of resources to enable training to operationalize referrals to palliative care teams and multidisciplinary care provision across settings, especially home-based care.
Collapse
Affiliation(s)
- Marmareen Bakhtary
- Melbourne Medical School, The University of Melbourne, Melbourne, Australia
| | - Pragati Sharma
- Department of Paediatrics, Monash University, Clayton, Australia
| | - Sarah Raspin
- Victorian Paediatric Palliative Care Program, The Royal Children's Hospital, Melbourne, Australia
| | - Sidharth Vemuri
- Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
- Victorian Paediatric Palliative Care Program, The Royal Children's Hospital, Melbourne, Australia
- Department of Paediatrics, Monash University, Clayton, Australia
| | - Maria McCarthy
- Department of Paediatrics, Monash University, Clayton, Australia
| |
Collapse
|
2
|
Zaghloul MS, Bishr MK, Refaat A, Hemaly AE, Ayadi MA, Ahmed S, Maher E, Todary ES. Re-Irradiation for the Progressive Pediatric Diffuse Intrinsic Pontine Glioma: A Report on 109 Children From a Single Center. Pediatr Blood Cancer 2025; 72:e31587. [PMID: 39905588 DOI: 10.1002/pbc.31587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/10/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Diffuse intrinsic pontine glioma (DIPG) is a challenging pediatric tumor that frequently progresses within the first year following local radiotherapy. However, several small studies have suggested that re-irradiation may improve quality of life and extend overall survival. PATIENTS AND METHODS This retrospective study included 109 children who experienced disease progression ≥3 months after their initial radiotherapy, and subsequently received re-irradiation at a single institution. These patients were compared with a cohort of 60 children, meeting the same criteria, who were treated before adopting the re-irradiation policy and received only the best supportive care (BSC). Most of the re-irradiated children (94%) received first radiation dose as hypofractionation (39 Gy/13 fractions). RESULTS The re-irradiation group demonstrated significantly higher overall survival (OS) rates after the first progression, with a 6-month OS of 42% (95% CI: 34%-53%) compared to 16% (95% CI: 8.9%-32%) in the BSC group (p < 0.001). Re-irradiation reduced the hazard of death by more than half (HR = 0.45, p < 0.001). Clinical response (p < 0.001) and radiological response (p = 0.016) were significant predictors of improved survival. While the time from initial radiotherapy to progression (p = 0.059) and higher re-irradiation doses (p = 0.074) were associated with improved OS, these factors did not reach statistical significance but may represent potential prognostic indicators. CONCLUSION Re-irradiation improved the OS in children with progression of DIPG and alleviated their signs and symptoms.
Collapse
Affiliation(s)
- Mohamed Saad Zaghloul
- Radiation Oncology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Radiation Oncology Department, Children's Cancer Hospital, Cairo, Egypt
| | - Mai K Bishr
- The Institute of Cancer Research, London, UK
| | - Amal Refaat
- Radiology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Radiology Department, Children's Cancer Hospital, Cairo, Egypt
| | - Ahmed El Hemaly
- Pediatric Oncology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Pediatric Oncology Department, Children's Cancer Hospital, Cairo, Egypt
| | - Moatssem Al Ayadi
- Pediatric Oncology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Pediatric Oncology Department, Children's Cancer Hospital, Cairo, Egypt
| | - Soha Ahmed
- Clinical Oncology Department, Suez University, Suez, Egypt
| | - Eslam Maher
- Clinical Research Department, Children's Cancer Hospital, Cairo, Egypt
| | - Engy S Todary
- Radiation Oncology Department, Children's Cancer Hospital, Cairo, Egypt
| |
Collapse
|
3
|
Pearson AD, Mueller S, Filbin MG, Grill J, Hawkins C, Jones C, Donoghue M, Drezner N, Weiner S, Russo M, Dun MD, Allen JE, Alonso M, Benaim E, Buenger V, de Rojas T, Desserich K, Fox E, Friend J, Glade Bender J, Hargrave D, Jensen M, Kholmanskikh O, Kieran MW, Knoderer H, Koschmann C, Lesa G, Ligas F, Lipsman N, Ludwinski D, Marshall L, McDonough J, McNicholl AG, Mirsky D, Monje M, Nysom K, Pappo A, Rosenfield A, Scobie N, Slaughter J, Smith M, Souweidane M, Straathof K, Ward L, Weigel B, Zamoryakhin D, Karres D, Vassal G. Paediatric strategy forum for medicinal product development in diffuse midline gliomas in children and adolescents ACCELERATE in collaboration with the European Medicines Agency with participation of the Food and Drug Administration. Eur J Cancer 2025; 217:115230. [PMID: 39854822 DOI: 10.1016/j.ejca.2025.115230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025]
Abstract
Fewer than 10 % of children with diffuse midline glioma (DMG) survive 2 years from diagnosis. Radiation therapy remains the cornerstone of treatment and there are no medicinal products with regulatory approval. Although the biology of DMG is better characterized, this has not yet translated into effective treatments. H3K27-alterations initiate the disease but additional drivers are required for malignant growth. Hence, there is an urgent unmet need to develop new multi-modality therapeutic strategies, including alternative methods of drug delivery. ONC201 (DRD2 antagonist and mitochondrial ClpP agonist) is the most widely evaluated investigational drug. Encouraging early data is emerging for CAR T-cells and oncolytic viruses. GD2, B7-H3 and PI3K signalling are ubiquitous targets across all subtypes and therapeutics directed to these targets would potentially benefit the largest number of children. PI3K, ACVR1, MAPK and PDGFRA pathways should be targeted in rational biological combinations. Drug discovery is a very high priority. New specific and potent epigenetic modifiers (PROTACS e.g. SMARCA4 degraders), with blood-brain penetrance are needed. Cancer neuroscience therapeutics are in early development. Overall survival is the preferred regulatory endpoint. However, the evaluation of this can be influenced by the use of re-irradiation at the time of progression. An efficient clinical trial design fit for regulatory purposes for the evaluation of new therapeutics would aid industry and facilitate more efficient therapy development. Challenges in conducting clinical trials such as the need for comparator data and defining endpoints, could be addressed through an international, first-in-child, randomised, complex innovative design trial. To achieve progress: i) drug discovery; ii) new multi-modality, efficient, collaborative, pre-clinical approaches, possibly including artificial intelligence and, iii) efficient clinical trial designs fit for regulatory purposes are required.
Collapse
Affiliation(s)
| | - Sabine Mueller
- Departments of Neurological Surgery, Pediatrics and, Neurology University of California, San Francisco, California, USA. Department of Oncology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Mariella G Filbin
- Broad Institute of Harvard and MIT, Cambridge, MA, USA; Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | | | - Cynthia Hawkins
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Chris Jones
- The Institute of Cancer Research, Sutton, Surrey, UK
| | | | - Nicole Drezner
- US Food and Drug Administration, Silver Springs, MD, USA
| | - Susan Weiner
- ACCELERATE, Europe; Children's Cancer Cause, Washington, DC, USA; Memorial Sloan Kettering Cancer Centre, New York, USA
| | | | - Matthew D Dun
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia; Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, Mark Hughes Foundation for Brain Cancer Research, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | | | - Marta Alonso
- Program of Solid Tumors, Center for the Applied Medical Research, Pamplona, Spain; Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Center for the Applied Medical Research, Pamplona, Spain
| | | | - Vickie Buenger
- Coalition Against Childhood Cancer (CAC2), Philadelphia, USA
| | | | | | | | | | | | - Darren Hargrave
- University College London Great Ormond Street Institute of Child Health, London, UK
| | | | | | | | | | | | - Giovanni Lesa
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Division, European Medicines Agency, The Netherlands
| | - Franca Ligas
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Division, European Medicines Agency, The Netherlands
| | - Nir Lipsman
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | - Lynley Marshall
- The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK
| | | | | | - David Mirsky
- University of Colorado, School of Medicine, CO, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA, Howard Hughes Medical Institute, Stanford, CA, USA
| | | | | | | | | | | | | | | | - Karin Straathof
- University College London Cancer Institute, Great Ormond Street Biomedical Research Centre, London, UK
| | - Lisa Ward
- DIPG-DMG Research Funding Alliance DDRFA /Tough2gether, Manhattan, KS, USA
| | | | | | - Dominik Karres
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Division, European Medicines Agency, The Netherlands
| | - Gilles Vassal
- ACCELERATE, Europe; Gustave Roussy Cancer Centre, Paris, France
| |
Collapse
|
4
|
Brown EJ, Balaguer-Lluna L, Cribbs AP, Philpott M, Campo L, Browne M, Wong JF, Oppermann U, Carcaboso ÁM, Bullock AN, Farnie G. PRMT5 inhibition shows in vitro efficacy against H3K27M-altered diffuse midline glioma, but does not extend survival in vivo. Sci Rep 2024; 14:328. [PMID: 38172189 PMCID: PMC10764357 DOI: 10.1038/s41598-023-48652-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 11/29/2023] [Indexed: 01/05/2024] Open
Abstract
H3K27-altered Diffuse Midline Glioma (DMG) is a universally fatal paediatric brainstem tumour. The prevalent driver mutation H3K27M creates a unique epigenetic landscape that may also establish therapeutic vulnerabilities to epigenetic inhibitors. However, while HDAC, EZH2 and BET inhibitors have proven somewhat effective in pre-clinical models, none have translated into clinical benefit due to either poor blood-brain barrier penetration, lack of efficacy or toxicity. Thus, there remains an urgent need for new DMG treatments. Here, we performed wider screening of an epigenetic inhibitor library and identified inhibitors of protein arginine methyltransferases (PRMTs) among the top hits reducing DMG cell viability. Two of the most effective inhibitors, LLY-283 and GSK591, were targeted against PRMT5 using distinct binding mechanisms and reduced the viability of a subset of DMG cells expressing wild-type TP53 and mutant ACVR1. RNA-sequencing and phenotypic analyses revealed that LLY-283 could reduce the viability, clonogenicity and invasion of DMG cells in vitro, representing three clinically important phenotypes, but failed to prolong survival in an orthotopic xenograft model. Together, these data show the challenges of DMG treatment and highlight PRMT5 inhibitors for consideration in future studies of combination treatments.
Collapse
Affiliation(s)
- Elizabeth J Brown
- Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Leire Balaguer-Lluna
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Adam P Cribbs
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, National Institute of Health Research Oxford Biomedical Research Unit (BRU), University of Oxford, Oxford, UK
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK
| | - Martin Philpott
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, National Institute of Health Research Oxford Biomedical Research Unit (BRU), University of Oxford, Oxford, UK
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK
| | - Leticia Campo
- Department of Oncology, Experimental Cancer Medicine Centre, University of Oxford, Oxford, UK
| | - Molly Browne
- Department of Oncology, Experimental Cancer Medicine Centre, University of Oxford, Oxford, UK
| | - Jong Fu Wong
- Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, UK
| | - Udo Oppermann
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, National Institute of Health Research Oxford Biomedical Research Unit (BRU), University of Oxford, Oxford, UK
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK
| | - Ángel M Carcaboso
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Alex N Bullock
- Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, UK.
| | - Gillian Farnie
- Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford, UK.
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK.
- Cancer Research Horizons, The Francis Crick Institute, London, UK.
| |
Collapse
|
5
|
Monje M, Cooney T, Glod J, Huang J, Peer CJ, Faury D, Baxter P, Kramer K, Lenzen A, Robison NJ, Kilburn L, Vinitsky A, Figg WD, Jabado N, Fouladi M, Fangusaro J, Onar-Thomas A, Dunkel IJ, Warren KE. Phase I trial of panobinostat in children with diffuse intrinsic pontine glioma: A report from the Pediatric Brain Tumor Consortium (PBTC-047). Neuro Oncol 2023; 25:2262-2272. [PMID: 37526549 PMCID: PMC10708931 DOI: 10.1093/neuonc/noad141] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Diffuse intrinsic pontine glioma (DIPG) is a lethal childhood cancer with median survival of less than 1 year. Panobinostat is an oral multihistone deacetylase inhibitor with preclinical activity in DIPG models. Study objectives were to determine safety, tolerability, maximum tolerated dose (MTD), toxicity profile, and pharmacokinetics of panobinostat in children with DIPG. PATIENTS AND METHODS In stratum 1, panobinostat was administered 3 days per week for 3 weeks on, 1 week off to children with progressive DIPG, with dose escalation following a two-stage continual reassessment method. After this MTD was determined, the study was amended to evaluate the MTD in children with nonprogressive DIPG/Diffuse midline glioma (DMG) (stratum 2) on an alternate schedule, 3 days a week every other week in an effort to escalate the dose. RESULTS For stratum 1, 19 subjects enrolled with 17/19 evaluable for dose-finding. The MTD was 10 mg/m2/dose. Dose-limiting toxicities included thrombocytopenia and neutropenia. Posterior reversible encephalopathy syndrome was reported in 1 patient. For stratum 2, 34 eligible subjects enrolled with 29/34 evaluable for dose finding. The MTD on this schedule was 22 mg/m2/dose. DLTs included thrombocytopenia, neutropenia, neutropenia with grade 4 thrombocytopenia, prolonged intolerable nausea, and increased ALT. CONCLUSIONS The MTD of panobinostat is 10 mg/m2/dose administered 3 times per week for 3 weeks on/1 week off in children with progressive DIPG/DMG and 22 mg/m2/dose administered 3 times per week for 1 week on/1 week off when administered in a similar population preprogression. The most common toxicity for both schedules was myelosuppression.
Collapse
Affiliation(s)
- Michelle Monje
- Department of Neurology, Stanford University and Lucile Packard Children’s Hospital, Palo Alto, CA, USA
| | - Tabitha Cooney
- Department of Pediatric Oncology, Dana Farber Cancer Institute/Boston Children’s Hospital, Boston, MA, USA
| | - John Glod
- Pediatric Oncology, Pediatric Oncology Branch, National Cancer Institute, Bethesda, MDUS
| | - Jie Huang
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Cody J Peer
- Center for Cancer Research, Clinical Pharmacology Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Damien Faury
- Research Institute of the McGill University Health Center, Montreal, QuebecCANADA
| | - Patricia Baxter
- Pediatric Oncology, Texas Children’s Cancer Center, Houston, TX, USA
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Alicia Lenzen
- Pediatric Hematology Oncology, Lurie Children’s Hospital, Chicago, IL, USA
| | - Nathan J Robison
- Department of Pediatrics, Children’s Hospital, Los Angeles, CA, USA
| | - Lindsay Kilburn
- Department of Oncology, Children’s National Hospital, Washington, DC, USA
| | - Anna Vinitsky
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - William D Figg
- Center for Cancer Research, Clinical Pharmacology Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Nada Jabado
- Research Institute of the McGill University Health Center, Montreal, QuebecCANADA
| | - Maryam Fouladi
- Pediatric Hematology Oncology, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Jason Fangusaro
- Department: Pediatric Hematology/Oncology and Stem Cell Transplantation, Atlanta, GA, USA
| | - Arzu Onar-Thomas
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Ira J Dunkel
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Katherine E Warren
- Department of Pediatric Oncology, Dana Farber Cancer Institute/Boston Children’s Hospital, Boston, MA, USA
- Pediatric Oncology, Pediatric Oncology Branch, National Cancer Institute, Bethesda, MDUS
| |
Collapse
|
6
|
Cacciotti C, Wright KD. Advances in Treatment of Diffuse Midline Gliomas. Curr Neurol Neurosci Rep 2023; 23:849-856. [PMID: 37921944 DOI: 10.1007/s11910-023-01317-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2023] [Indexed: 11/05/2023]
Abstract
PURPOSE OF REVIEW Diffuse midline gliomas (DMGs) generally carry a poor prognosis, occur during childhood, and involve midline structures of the central nervous system, including the thalamus, pons, and spinal cord. RECENT FINDINGS To date, irradiation has been shown to be the only beneficial treatment for DMG. Various genetic modifications have been shown to play a role in the pathogenesis of this disease. Current treatment strategies span targeting epigenetic dysregulation, cell cycle, specific genetic alterations, and the immune microenvironment. Herein, we review the complex features of this disease as it relates to current and past therapeutic approaches.
Collapse
Affiliation(s)
- Chantel Cacciotti
- Children's Hospital London Health Sciences/Western University, London, ON, Canada.
| | - Karen D Wright
- Dana Farber/Boston Children's Cancer and Blood Disorder Center, Boston, MA, USA
| |
Collapse
|
7
|
Ehteda A, Khan A, Rajakumar G, Vanniasinghe AS, Gopalakrishnan A, Liu J, Tsoli M, Ziegler DS. Microtubule-Targeting Combined with HDAC Inhibition Is a Novel Therapeutic Strategy for Diffuse Intrinsic Pontine Gliomas. Mol Cancer Ther 2023; 22:1413-1421. [PMID: 37683275 PMCID: PMC10690044 DOI: 10.1158/1535-7163.mct-23-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/30/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023]
Abstract
Diffuse intrinsic pontine gliomas (DIPG) are an incurable childhood brain cancer for which novel treatments are needed. DIPGs are characterized by a mutation in the H3 histone (H3K27M), resulting in loss of H3K27 methylation and global gene dysregulation. TRX-E-009-1 is a novel anticancer agent with preclinical activity demonstrated against a range of cancers. We examined the antitumor activity of TRX-E-009-1 against DIPG neurosphere cultures and observed tumor-specific activity with IC50s ranging from 20 to 100 nmol/L, whereas no activity was observed against normal human astrocyte cells. TRX-E-009-1 exerted its anti-proliferative effect through the induction of apoptotic pathways, with marked increases in cleaved caspase 3 and cleaved PARP levels, while also restoring histone H3K27me3 methylation. Co-administration of TRX-E-009-1 and the histone deacetylase (HDAC) inhibitor SAHA extended survival in DIPG orthotopic animal models. This antitumor effect was further enhanced with irradiation. Our findings indicate that TRX-E-009-1, combined with HDAC inhibition, represents a novel, potent therapy for children with DIPG.
Collapse
Affiliation(s)
- Anahid Ehteda
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| | - Aaminah Khan
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
| | - Gayathiri Rajakumar
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
| | - Anne S. Vanniasinghe
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
| | - Anjana Gopalakrishnan
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
| | - Jie Liu
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
| | - Maria Tsoli
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
| | - David S. Ziegler
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, High St, Randwick, Australia
| |
Collapse
|
8
|
Solst SR, Mapuskar KA, Graham CH, King SA, Rheem R, Current K, Allen BG, Caster JM, Spitz DR, Howard ME. Rapid Peroxide Removal Limits the Radiosensitization of Diffuse Intrinsic Pontine Glioma (DIPG) Cells by Pharmacologic Ascorbate. Radiat Res 2023; 200:456-461. [PMID: 37758035 PMCID: PMC10759934 DOI: 10.1667/rade-23-00006.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 08/14/2023] [Indexed: 10/03/2023]
Abstract
Diffuse intrinsic pontine gliomas (DIPG) are an aggressive type of pediatric brain tumor with a very high mortality rate. Surgery has a limited role given the tumor's location. Palliative radiation therapy alleviates symptoms and prolongs survival, but median survival remains less than 1 year. There is no clear role for chemotherapy in DIPGs as trials adding chemotherapy to palliative radiation therapy have failed to improve survival compared to radiation alone. Thus, there is a critical need to identify tissue-specific radiosensitizers to improve clinical outcomes for patients with DIPGs. Pharmacologic (high dose) ascorbate (P-AscH-) is a promising anticancer therapy that sensitizes human tumors, including adult high-grade gliomas, to radiation by acting selectively as a generator of hydrogen peroxide (H2O2) in cancer cells. In this study we demonstrate that in contrast to adult glioma models, P-AscH- does not radiosensitize DIPG. DIPG cells were sensitive to bolus of H2O2 but have faster H2O2 removal rates than GBM models which are radiosensitized by P-AscH-. These data support the hypothesis that P-AscH- does not enhance DIPG radiosensitivity, likely due to a robust capacity to detoxify and remove hydroperoxides.
Collapse
Affiliation(s)
- Shane R. Solst
- Free Radical and Radiation Biology Program, B180 Medical Laboratories, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242
| | - Kranti A. Mapuskar
- Free Radical and Radiation Biology Program, B180 Medical Laboratories, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242
| | - Claire H. Graham
- Free Radical and Radiation Biology Program, B180 Medical Laboratories, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242
| | - Sarah A. King
- Free Radical and Radiation Biology Program, B180 Medical Laboratories, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242
| | - Rana Rheem
- Free Radical and Radiation Biology Program, B180 Medical Laboratories, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242
| | - Kyle Current
- Free Radical and Radiation Biology Program, B180 Medical Laboratories, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, B180 Medical Laboratories, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242
| | - Joseph M. Caster
- Free Radical and Radiation Biology Program, B180 Medical Laboratories, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, B180 Medical Laboratories, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242
| | - Michelle E. Howard
- Free Radical and Radiation Biology Program, B180 Medical Laboratories, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
9
|
Lo Greco MC, Milazzotto R, Liardo RLE, Foti PV, Palmucci S, Basile A, Pergolizzi S, Spatola C. The Role of Reirradiation in Childhood Progressive Diffuse Intrinsic Pontine Glioma (DIPG): An Ongoing Challenge beyond Radiobiology. Brain Sci 2023; 13:1449. [PMID: 37891817 PMCID: PMC10605436 DOI: 10.3390/brainsci13101449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/26/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
To investigate the clinical impact of multiple courses of irradiation on pediatric patients with progressive diffuse intrinsic pontine glioma (DIPG), we conducted a retrospective case series on three children treated at our institution from 2018 to 2022. All children were candidates to receive systemic therapy with vinorelbine and nimotuzumab. Radiotherapy was administered to a total dose of 54 Gy. At any disease progression, our local tumor board evaluated the possibility of offering a new course of radiotherapy. To determine feasibility and assess toxicity rates, all children underwent clinical and hematological evaluation both during and after the treatment. To assess efficacy, all children performed contrast-enhanced MRI almost quarterly after the end of the treatment. In all children, following any treatment course, neurological improvement (>80%) was associated with a radiological response (41.7-46%). The longest overall survival (24 months) was observed in the child who underwent three courses of radiotherapy, without experiencing significant side effects. Even though it goes beyond the understanding of conventional radiobiology, first and second reirradiation in pediatric patients with progressive DIPG may represent a feasible and safe approach, capable of increasing overall survival and disease-free survival in selected patients and improving their quality of life.
Collapse
Affiliation(s)
- Maria Chiara Lo Greco
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy;
| | - Roberto Milazzotto
- Radiation Oncology Unit, Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (R.M.); (R.L.E.L.); (C.S.)
| | - Rocco Luca Emanuele Liardo
- Radiation Oncology Unit, Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (R.M.); (R.L.E.L.); (C.S.)
| | - Pietro Valerio Foti
- Radiology I Unit, Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (P.V.F.); (S.P.); (A.B.)
| | - Stefano Palmucci
- Radiology I Unit, Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (P.V.F.); (S.P.); (A.B.)
| | - Antonio Basile
- Radiology I Unit, Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (P.V.F.); (S.P.); (A.B.)
| | - Stefano Pergolizzi
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy;
| | - Corrado Spatola
- Radiation Oncology Unit, Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (R.M.); (R.L.E.L.); (C.S.)
| |
Collapse
|
10
|
Sánchez-Duffhues G, Hiepen C. Human iPSCs as Model Systems for BMP-Related Rare Diseases. Cells 2023; 12:2200. [PMID: 37681932 PMCID: PMC10487005 DOI: 10.3390/cells12172200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
Disturbances in bone morphogenetic protein (BMP) signalling contribute to onset and development of a number of rare genetic diseases, including Fibrodysplasia ossificans progressiva (FOP), Pulmonary arterial hypertension (PAH), and Hereditary haemorrhagic telangiectasia (HHT). After decades of animal research to build a solid foundation in understanding the underlying molecular mechanisms, the progressive implementation of iPSC-based patient-derived models will improve drug development by addressing drug efficacy, specificity, and toxicity in a complex humanized environment. We will review the current state of literature on iPSC-derived model systems in this field, with special emphasis on the access to patient source material and the complications that may come with it. Given the essential role of BMPs during embryonic development and stem cell differentiation, gain- or loss-of-function mutations in the BMP signalling pathway may compromise iPSC generation, maintenance, and differentiation procedures. This review highlights the need for careful optimization of the protocols used. Finally, we will discuss recent developments towards complex in vitro culture models aiming to resemble specific tissue microenvironments with multi-faceted cellular inputs, such as cell mechanics and ECM together with organoids, organ-on-chip, and microfluidic technologies.
Collapse
Affiliation(s)
- Gonzalo Sánchez-Duffhues
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), ISPA-HUCA, Avda. de Roma, s/n, 33011 Oviedo, Spain
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Christian Hiepen
- Department of Engineering and Natural Sciences, Westphalian University of Applied Sciences, August-Schmidt-Ring 10, 45665 Recklinghausen, Germany
| |
Collapse
|
11
|
Miguel Llordes G, Medina Pérez VM, Curto Simón B, Castells-Yus I, Vázquez Sufuentes S, Schuhmacher AJ. Epidemiology, Diagnostic Strategies, and Therapeutic Advances in Diffuse Midline Glioma. J Clin Med 2023; 12:5261. [PMID: 37629304 PMCID: PMC10456112 DOI: 10.3390/jcm12165261] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Object: Diffuse midline glioma (DMG) is a highly aggressive and lethal brain tumor predominantly affecting children and young adults. Previously known as diffuse intrinsic pontine glioma (DIPG) or grade IV brain stem glioma, DMG has recently been reclassified as "diffuse midline glioma" according to the WHO CNS5 nomenclature, expanding the DMG demographic. Limited therapeutic options result in a poor prognosis, despite advances in diagnosis and treatment. Radiotherapy has historically been the primary treatment modality to improve patient survival. Methods: This systematic literature review aims to comprehensively compile information on the diagnosis and treatment of DMG from 1 January 2012 to 31 July 2023. The review followed the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) statement and utilized databases such as PubMed, Cochrane Library, and SciELO. Results: Currently, molecular classification of DMG plays an increasingly vital role in determining prognosis and treatment options. Emerging therapeutic avenues, including immunomodulatory agents, anti-GD2 CAR T-cell and anti-GD2 CAR-NK therapies, techniques to increase blood-brain barrier permeability, isocitrate dehydrogenase inhibitors, oncolytic and peptide vaccines, are being explored based on the tumor's molecular composition. However, more clinical trials are required to establish solid guidelines for toxicity, dosage, and efficacy. Conclusions: The identification of the H3K27 genetic mutation has led to the reclassification of certain midline tumors, expanding the DMG demographic. The field of DMG research continues to evolve, with encouraging findings that underscore the importance of highly specific and tailored therapeutic strategies to achieve therapeutic success.
Collapse
Affiliation(s)
- Gloria Miguel Llordes
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Víctor Manuel Medina Pérez
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | | | - Irene Castells-Yus
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | | | - Alberto J. Schuhmacher
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Fundación Aragonesa para la Investigación y el Desarrollo (ARAID), 50018 Zaragoza, Spain
| |
Collapse
|
12
|
Rhodes A, Martin S, Toledo-Tamula MA, Loucas C, Glod J, Warren KE, Wolters PL. The neuropsychological profile of children with Diffuse Intrinsic Pontine Glioma (DIPG) before and after radiation therapy: A prospective longitudinal study. Child Neuropsychol 2023; 29:934-958. [PMID: 36369715 DOI: 10.1080/09297049.2022.2144189] [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: 05/05/2022] [Accepted: 11/01/2022] [Indexed: 11/15/2022]
Abstract
Children with Diffuse Intrinsic Pontine Gliomas (DIPG), a malignant brainstem tumor, experience poor prognosis. Because of the disease's rarity and highly aggressive course, there is a dearth of research on cognitive and psychosocial outcomes in this underserved, vulnerable population. However, evaluating effects of the disease and treatment on the cognitive and daily functioning of these patients is important to better understand their specific needs and improve their quality of life. The current longitudinal study administered prospective neuropsychological assessments to children diagnosed with CNS malignancies, including the largest sample of children with DIPG to date (n = 21, mean age = 7.86 years, range = 3-16) in neurocognitive, behavioral, social-emotional, and adaptive functioning at baseline, two weeks post-radiation, and six months later. The results describe population-based, cross-sectional characteristics and within-patient longitudinal changes. Prior to radiation, children with DIPG exhibited significant weaknesses compared to normative samples in both parent-report and performance-based measures of attention, and tests of processing speed and verbal learning/memory. Younger children demonstrated poorer inhibitory control on performance tests and worse parent-reported behavioral regulation, depression, and social withdrawal compared to older children. Six-months post-radiation, older children exhibited poorer socialization than younger children. Longitudinally, children with DIPG exhibited short-term improvements immediately post-radiation in performance-based attention tests and parent-reported behavior, including attention, hyperactivity, behavioral regulation, and executive function. However, these improvements did not persist and significant decline was documented on tests of attention by six months. Clinical implications for professionals working with children with DIPG and recommendations for cognitive remediation and quality of life interventions are provided.
Collapse
Affiliation(s)
- Amanda Rhodes
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Staci Martin
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Mary Anne Toledo-Tamula
- Clinical Research Directorate (CRD), Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Caitlyn Loucas
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - John Glod
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Katherine E Warren
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Department of Pediatric Neuro-Oncology, Dana Farber Cancer Institute/Boston Children's Hospital, Boston, MA, USA
| | - Pamela L Wolters
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
13
|
Huang B, Zhang Y, Mao Q, Ju Y, Liu Y, Su Z, Lei Y, Ren Y. Deep learning-based prediction of H3K27M alteration in diffuse midline gliomas based on whole-brain MRI. Cancer Med 2023; 12:17139-17148. [PMID: 37461358 PMCID: PMC10501256 DOI: 10.1002/cam4.6363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/02/2023] [Accepted: 07/08/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND H3K27M mutation status significantly affects the prognosis of patients with diffuse midline gliomas (DMGs), but this tumor presents a high risk of pathological acquisition. We aimed to construct a fully automated model for predicting the H3K27M alteration status of DMGs based on deep learning using whole-brain MRI. METHODS DMG patients from West China Hospital of Sichuan University (WCHSU; n = 200) and Chengdu Shangjin Nanfu Hospital (CSNH; n = 35) who met the inclusion and exclusion criteria from February 2016 to April 2022 were enrolled as the training and external test sets, respectively. To adapt the model to the human head MRI scene, we use normal human head MR images to pretrain the model. The classification and tumor segmentation tasks are naturally related, so we conducted cotraining for the two tasks to enable information interaction between them and improve the accuracy of the classification task. RESULTS The average classification accuracies of our model on the training and external test sets was 90.5% and 85.1%, respectively. Ablation experiments showed that pretraining and cotraining could improve the prediction accuracy and generalization performance of the model. In the training and external test sets, the average areas under the receiver operating characteristic curve (AUROCs) were 94.18% and 87.64%, and the average areas under the precision-recall curve (AUPRC) were 93.26% and 85.4%. CONCLUSIONS The developed model achieved excellent performance in predicting the H3K27M alteration status in DMGs, and its good reproducibility and generalization were verified in the external dataset.
Collapse
Affiliation(s)
- Bowen Huang
- Department of NeurosurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Yuekang Zhang
- Department of NeurosurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Qing Mao
- Department of NeurosurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Yan Ju
- Department of NeurosurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Yanhui Liu
- Department of NeurosurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Zhengzheng Su
- Department of PathologyWest China Hospital of Sichuan UniversityChengduChina
| | - Yinjie Lei
- College of Electronics and Information EngineeringSichuan UniversityChengduChina
| | - Yanming Ren
- Department of NeurosurgeryWest China Hospital of Sichuan UniversityChengduChina
| |
Collapse
|
14
|
Wang X, Sun Y, Zhang DY, Ming GL, Song H. Glioblastoma modeling with 3D organoids: progress and challenges. OXFORD OPEN NEUROSCIENCE 2023; 2:kvad008. [PMID: 38596241 PMCID: PMC10913843 DOI: 10.1093/oons/kvad008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Glioblastoma (GBM) is the most aggressive adult primary brain tumor with nearly universal treatment resistance and recurrence. The mainstay of therapy remains maximal safe surgical resection followed by concurrent radiation therapy and temozolomide chemotherapy. Despite intensive investigation, alternative treatment options, such as immunotherapy or targeted molecular therapy, have yielded limited success to achieve long-term remission. This difficulty is partly due to the lack of pre-clinical models that fully recapitulate the intratumoral and intertumoral heterogeneity of GBM and the complex tumor microenvironment. Recently, GBM 3D organoids originating from resected patient tumors, genetic manipulation of induced pluripotent stem cell (iPSC)-derived brain organoids and bio-printing or fusion with non-malignant tissues have emerged as novel culture systems to portray the biology of GBM. Here, we highlight several methodologies for generating GBM organoids and discuss insights gained using such organoid models compared to classic modeling approaches using cell lines and xenografts. We also outline limitations of current GBM 3D organoids, most notably the difficulty retaining the tumor microenvironment, and discuss current efforts for improvements. Finally, we propose potential applications of organoid models for a deeper mechanistic understanding of GBM and therapeutic development.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yusha Sun
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Y Zhang
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- GBM Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania Philadelphia, PA 19104, USA
| |
Collapse
|
15
|
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
|
16
|
Lyu Y, Guo Y, Okeoma CM, Yan Z, Hu N, Li Z, Zhou S, Zhao X, Li J, Wang X. Engineered extracellular vesicles (EVs): Promising diagnostic/therapeutic tools for pediatric high-grade glioma. Biomed Pharmacother 2023; 163:114630. [PMID: 37094548 DOI: 10.1016/j.biopha.2023.114630] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/29/2023] [Accepted: 03/29/2023] [Indexed: 04/26/2023] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a highly malignant brain tumor that mainly occurs in children with extremely low overall survival. Traditional therapeutic strategies, such as surgical resection and chemotherapy, are not feasible mostly due to the special location and highly diffused features. Radiotherapy turns out to be the standard treatment method but with limited benefits of overall survival. A broad search for novel and targeted therapies is in the progress of both preclinical investigations and clinical trials. Extracellular vesicles (EVs) emerged as a promising diagnostic and therapeutic candidate due to their distinct biocompatibility, excellent cargo-loading-delivery capacity, high biological barrier penetration efficiency, and ease of modification. The utilization of EVs in various diseases as biomarker diagnoses or therapeutic agents is revolutionizing modern medical research and practice. In this review, we will briefly talk about the research development of DIPG, and present a detailed description of EVs in medical applications, with a discussion on the application of engineered peptides on EVs. The possibility of applying EVs as a diagnostic tool and drug delivery system in DIPG is also discussed.
Collapse
Affiliation(s)
- Yuan Lyu
- Medical Research Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yupei Guo
- Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Chioma M Okeoma
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, NY 10595-1524, USA
| | - Zhaoyue Yan
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Nan Hu
- Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zian Li
- Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Shaolong Zhou
- Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xin Zhao
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Junqi Li
- Medical Research Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Xinjun Wang
- Henan Joint International Laboratory of Glioma Metabolism and Microenvironment Research, Henan Provincial Department of Science and Technology, Zhengzhou, Henan 450052, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Neurosurgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
17
|
Kim HJ, Suh CO. Radiotherapy for Diffuse Intrinsic Pontine Glioma: Insufficient but Indispensable. Brain Tumor Res Treat 2023; 11:79-85. [PMID: 37151149 PMCID: PMC10172015 DOI: 10.14791/btrt.2022.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/28/2022] [Accepted: 01/02/2023] [Indexed: 05/09/2023] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) account for 10%-20% of all central nervous system tumors in children and are the leading cause of death in children with brain tumors. Although many clinical trials have been conducted over the past decades, the survival outcome has remained unchanged. Over 90% of children die within 2 years of the diagnosis, and radiotherapy remains the standard treatment to date. To improve the prognosis, hyperfractionated and hypofractionated radiotherapy and/or addition of radiosensitizers have been investigated. However, none of the radiotherapy approaches have shown a survival benefit, and the overall survival of patients with DIPG is approximately 11 months. Here, we comprehensively review the management of DIPG with focus on radiotherapy.
Collapse
Affiliation(s)
- Hyun Ju Kim
- Department of Radiation Oncology, Gachon University Gil Hospital, Gachon University College of Medicine, Incheon, Korea
| | - Chang-Ok Suh
- Department of Radiation Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea.
| |
Collapse
|
18
|
Wagner MW, Namdar K, Napoleone M, Hainc N, Amirabadi A, Fonseca A, Laughlin S, Shroff MM, Bouffet E, Hawkins C, Khalvati F, Bartels U, Ertl-Wagner BB. Radiomic Features Based on MRI Predict Progression-Free Survival in Pediatric Diffuse Midline Glioma/Diffuse Intrinsic Pontine Glioma. Can Assoc Radiol J 2023; 74:119-126. [PMID: 35768942 DOI: 10.1177/08465371221109921] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Purpose: Biopsy-based assessment of H3 K27 M status helps in predicting survival, but biopsy is usually limited to unusual presentations and clinical trials. We aimed to evaluate whether radiomics can serve as prognostic marker to stratify diffuse intrinsic pontine glioma (DIPG) subsets. Methods: In this retrospective study, diagnostic brain MRIs of children with DIPG were analyzed. Radiomic features were extracted from tumor segmentations and data were split into training/testing sets (80:20). A conditional survival forest model was applied to predict progression-free survival (PFS) using training data. The trained model was validated on the test data, and concordances were calculated for PFS. Experiments were repeated 100 times using randomized versions of the respective percentage of the training/test data. Results: A total of 89 patients were identified (48 females, 53.9%). Median age at time of diagnosis was 6.64 years (range: 1-16.9 years) and median PFS was 8 months (range: 1-84 months). Molecular data were available for 26 patients (29.2%) (1 wild type, 3 K27M-H3.1, 22 K27M-H3.3). Radiomic features of FLAIR and nonenhanced T1-weighted sequences were predictive of PFS. The best FLAIR radiomics model yielded a concordance of .87 [95% CI: .86-.88] at 4 months PFS. The best T1-weighted radiomics model yielded a concordance of .82 [95% CI: .8-.84] at 4 months PFS. The best combined FLAIR + T1-weighted radiomics model yielded a concordance of .74 [95% CI: .71-.77] at 3 months PFS. The predominant predictive radiomic feature matrix was gray-level size-zone. Conclusion: MRI-based radiomics may predict progression-free survival in pediatric diffuse midline glioma/diffuse intrinsic pontine glioma.
Collapse
Affiliation(s)
- Matthias W Wagner
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| | - Khashayar Namdar
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| | - Marc Napoleone
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Nicolin Hainc
- Nicolin Hainc:Department of Neuroradiology, Clinical Neuroscience Center, 7979University Hospital Zurich,University of Zurich, Switzerland
| | - Afsaneh Amirabadi
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Adriana Fonseca
- Department of Neurooncology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Suzanne Laughlin
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| | - Manohar M Shroff
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| | - Eric Bouffet
- Department of Neurooncology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Cynthia Hawkins
- Department of Paediatric Laboratory Medicine, Division of Pathology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Farzad Khalvati
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Ute Bartels
- Department of Neurooncology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Birgit B Ertl-Wagner
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| |
Collapse
|
19
|
11C-methionine PET imaging characteristics in children with diffuse intrinsic pontine gliomas and relationship to survival and H3 K27M mutation status. Eur J Nucl Med Mol Imaging 2023; 50:1709-1719. [PMID: 36697961 DOI: 10.1007/s00259-022-06105-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/30/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE This study aimed to describe 11C-methionine (11C-MET) PET imaging characteristics in patients with paediatric diffuse intrinsic pontine glioma (DIPG) and correlate them with survival and H3 K27M mutation status. METHODS We retrospectively analysed 98 children newly diagnosed with DIPG who underwent 11C-MET PET. PET imaging characteristics evaluated included uptake intensity, uniformity, metabolic tumour volume (MTV), and total lesion methionine uptake (TLMU). The maximum, mean, and peak of the tumour-to-background ratio (TBR), calculated as the corresponding standardised uptake values (SUV) divided by the mean reference value, were also recorded. The associations between the PET imaging characteristics and clinical outcomes in terms of progression-free survival (PFS) and overall survival (OS) and H3 K27M mutation status were assessed, respectively. RESULTS In univariate analysis, imaging characteristics significantly associated with shorter PFS and OS included a higher uniformity grade, higher TBRs, larger MTV, and higher TLMU. In multivariate analysis, larger MTV at diagnosis, shorter symptom duration, and no treatment were significantly correlated with shorter PFS and OS. The PET imaging features were not correlated with H3 K27M mutation status. CONCLUSION Although several imaging features were significantly associated with PFS and OS, only MTV, indicating the size of the active tumour, was identified as a strong independent prognostic factor.
Collapse
|
20
|
Dalle Ore C, Coleman C, Gupta N, Mueller S. Advances and Clinical Trials Update in the Treatment of Diffuse Intrinsic Pontine Gliomas. Pediatr Neurosurg 2023; 58:259-266. [PMID: 36642062 PMCID: PMC10664325 DOI: 10.1159/000529099] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/12/2022] [Indexed: 01/14/2023]
Abstract
BACKGROUND Diffuse intrinsic pontine gliomas (DIPGs) are high-grade gliomas (HGGs) that occur primarily in children, and represent a leading cause of death in pediatric patients with brain tumors with a median overall survival of only 8-11 months. SUMMARY While these lesions were previously thought to behave similarly to adult HGG, emerging data have demonstrated that DIPG is a biologically distinct entity from adult HGG frequently driven by mutations in the histone genes H3.3 and H3.1 not found in adult glioma. While biopsy of DIPG was historically felt to confer unacceptable risk of morbidity and mortality, multiple studies have demonstrated that stereotactic biopsy of DIPG is safe, allowing not only for improved understanding of DIPG but also forming the basis for protocols for personalized medicine in DIPG. However, current options for personalized medicine in DIPG are limited by the lack of efficacious targeted therapies for the mutations commonly found in DIPG. Multiple treatment modalities including targeted therapies, immunotherapy, convection-enhanced delivery, and focused ultrasound are in various stages of investigation. KEY MESSAGE Increasing frequency of biopsy for DIPG has identified distinct driving mutations that may serve as therapeutic targets. Novel treatment modalities are under investigation.
Collapse
Affiliation(s)
- Cecilia Dalle Ore
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Christina Coleman
- Division of Hematology/Oncology, Montreal Children's Hospital, McGill University Health Centre, Montreal, Québec, Canada
| | - Nalin Gupta
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Sabine Mueller
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
21
|
Zhang M, Xiao X, Gu G, Zhang P, Wu W, Wang Y, Pan C, Wang L, Li H, Wu Z, Zhang J, Zhang L. Role of neuronavigation in the surgical management of brainstem gliomas. Front Oncol 2023; 13:1159230. [PMID: 37205194 PMCID: PMC10185888 DOI: 10.3389/fonc.2023.1159230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 03/20/2023] [Indexed: 05/21/2023] Open
Abstract
Objective NeuroNavigation (NN) is a widely used intraoperative imaging guidance technique in neurosurgical operations; however, its value in brainstem glioma (BSG) surgery is inadequately reported and lacks objective proof. This study aims to investigate the applicational value of NN in BSG surgery. Method A retrospective analysis was performed on 155 patients with brainstem gliomas who received craniotomy from May 2019 to January 2022 at Beijing Tiantan Hospital. Eighty-four (54.2%) patients received surgery with NN. Preoperative and postoperative cranial nerve dysfunctions, muscle strength, and Karnofsky (KPS) were evaluated. Patients' radiological features, tumor volume, and extent of resection (EOR) were obtained from conventional MRI data. Patients' follow-up data were also collected. Comparative analyses on these variables were made between the NN group and the non-NN group. Result The usage of NN is independently related to a higher EOR in diffuse intrinsic pontine glioma (DIPG) (p=0.005) and non-DIPG group (p<0.001). It was observed that fewer patients in the NN group suffered from deterioration of KPS (p=0.032) and cranial nerve function (p=0.017) in non-DIPG group, and deterioration of muscle strength (p=0.040) and cranial nerve function (p=0.038) in DIPG group. Moreover, the usage of NN is an independent protective factor for the deterioration of KPS (p=0.04) and cranial nerve function (p=0.026) in non-DIPG patients and the deterioration of muscle strength (p=0.009) in DIPG patients. Furthermore, higher EOR subgroups were found to be independently related to better prognoses in DIPG patients (p=0.008). Conclusion NN has significant value in BSG surgery. With the assistance of NN, BSG surgery achieved higher EOR without deteriorating patients' functions. In addition, DIPG patients may benefit from the appropriate increase of EOR.
Collapse
Affiliation(s)
- Mingxin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiong Xiao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guocan Gu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Peng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenhao Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Changcun Pan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Huan Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Junting Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing, China
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Liwei Zhang,
| |
Collapse
|
22
|
Estevez-Ordonez D, Gary SE, Atchley TJ, Maleknia PD, George JA, Laskay NMB, Gross EG, Devulapalli RK, Johnston JM. Immunotherapy for Pediatric Brain and Spine Tumors: Current State and Future Directions. Pediatr Neurosurg 2022; 58:313-336. [PMID: 36549282 PMCID: PMC10233708 DOI: 10.1159/000528792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Brain tumors are the most common solid tumors and the leading cause of cancer-related deaths in children. Incidence in the USA has been on the rise for the last 2 decades. While therapeutic advances in diagnosis and treatment have improved survival and quality of life in many children, prognosis remains poor and current treatments have significant long-term sequelae. SUMMARY There is a substantial need for the development of new therapeutic approaches, and since the introduction of immunotherapy by immune checkpoint inhibitors, there has been an exponential increase in clinical trials to adopt these and other immunotherapy approaches in children with brain tumors. In this review, we summarize the current immunotherapy landscape for various pediatric brain tumor types including choroid plexus tumors, embryonal tumors (medulloblastoma, AT/RT, PNETs), ependymoma, germ cell tumors, gliomas, glioneuronal and neuronal tumors, and mesenchymal tumors. We discuss the latest clinical trials and noteworthy preclinical studies to treat these pediatric brain tumors using checkpoint inhibitors, cellular therapies (CAR-T, NK, T cell), oncolytic virotherapy, radioimmunotherapy, tumor vaccines, immunomodulators, and other targeted therapies. KEY MESSAGES The current landscape for immunotherapy in pediatric brain tumors is still emerging, but results in certain tumors have been promising. In the age of targeted therapy, genetic tumor profiling, and many ongoing clinical trials, immunotherapy will likely become an increasingly effective tool in the neuro-oncologist armamentarium.
Collapse
Affiliation(s)
- Dagoberto Estevez-Ordonez
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA,
- Division of Pediatric Neurosurgery, Children's of Alabama, Birmingham, Alabama, USA,
| | - Sam E Gary
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Travis J Atchley
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Neurosurgery, Children's of Alabama, Birmingham, Alabama, USA
| | - Pedram D Maleknia
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jordan A George
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nicholas M B Laskay
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Neurosurgery, Children's of Alabama, Birmingham, Alabama, USA
| | - Evan G Gross
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rishi K Devulapalli
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James M Johnston
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Neurosurgery, Children's of Alabama, Birmingham, Alabama, USA
| |
Collapse
|
23
|
Zhao JP, Liu XJ, Lin HZ, Cui CX, Yue YJ, Gao S, Xu HZ. MRI comparative study of diffuse midline glioma, H3 K27-altered and glioma in the midline without H3 K27-altered. BMC Neurol 2022; 22:498. [PMID: 36550486 PMCID: PMC9773507 DOI: 10.1186/s12883-022-03026-0] [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: 03/23/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE The MRI features of Diffuse midline glioma, H3 K27-altered and glioma in the midline without H3 K27-altered were compared and analyzed, and the changes in the apparent diffusion coefficient (ADC) of the two groups were quantitatively analyzed. METHODS The MRI images of 35 patients with Diffuse midline gliomas, H3 K27-altered and gliomas in the midline without H3 K27-altered were analyzed retrospectively. The location, edge, signal, peritumoral edema and enhancement characteristics of the lesions were observed, and the changes in ADC values were analyzed. RESULTS In the H3 K27-altered group, 85.7% (12/14) of the tumors were located in the thalamus and brainstem compared with 28.6% (6/21) in the no H3 K27-altered group. In the H3 K27-altered group, for tumors only located in the midline area, only 14.3% (1/7) had irregular shapes and unclear boundaries, while for tumors also invaded the extramidline tissues 85.7% (6/7) had irregular shapes and unclear boundaries.The"basilar artery wrapped sign" was found in 6 patients with tumors located in the pons in the H3 K27-altered group, but none in the no H3 K27-altered group had this sign. In the H3 K27-altered group, only 14.3% (1/7) of the tumors confined to the midline area had small cystic degeneration and necrosis, while for tumors also invaded the extramidline tissues, 100% (7/7) of the tumors had cystic degeneration and necrosis, and the cystic degeneration and necrosis only located in the extramidline region of the tumor in 6 cases.A total of 78.6% (11/14) of tumors in the H3 K27-altered group showed mild to moderate enhancement, while 47.6% (10/21) of tumors in the no H3 K27-altered group showed mild to moderate enhancement. The average peritumoral edema index was 1.13 in the H3 K27-altered group and 1.75 in the no H3 K27-altered group. The average ADC value of tumor in the H3 K27-altered group was 7.83 × 10- 4 mm2/s, and the ratio to normal brain tissue was 0.844, while the values in the no H3 K27-altered group were 13.5 × 10- 4 mm2/s and 1.75, respectively. CONCLUSION Compared with gliomas in the midline without H3 K27-altered, The MRI findings and ADC value of Diffuse midline gliomas, H3K27-altered have some characteristics, which can help improve the diagnosis and differential diagnosis.
Collapse
Affiliation(s)
- Ji-ping Zhao
- grid.412521.10000 0004 1769 1119Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xue-jun Liu
- grid.412521.10000 0004 1769 1119Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hao-zhi Lin
- grid.412521.10000 0004 1769 1119Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chun-xiao Cui
- grid.412521.10000 0004 1769 1119Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ying-jie Yue
- grid.412521.10000 0004 1769 1119Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Song Gao
- grid.412521.10000 0004 1769 1119Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong-zhang Xu
- grid.477864.eDepartment of Radiology, Shidao People’s Hospital, Rongcheng, China
| |
Collapse
|
24
|
Del Baldo G, Carai A, Abbas R, Cacchione A, Vinci M, Di Ruscio V, Colafati GS, Rossi S, Diomedi Camassei F, Maestro N, Temelso S, Pericoli G, De Billy E, Giovannoni I, Carboni A, Rinelli M, Agolini E, Mackay A, Jones C, Chiesa S, Balducci M, Locatelli F, Mastronuzzi A. Targeted therapy for pediatric diffuse intrinsic pontine glioma: a single-center experience. Ther Adv Med Oncol 2022; 14:17588359221113693. [PMID: 36090803 PMCID: PMC9459464 DOI: 10.1177/17588359221113693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 06/28/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Diffuse intrinsic pontine glioma (DIPG) is a fatal disease with a median
overall survival (OS) of less than 12 months after diagnosis. Radiotherapy
(RT) still remains the mainstay treatment. Several other therapeutic
strategies have been attempted in the last years without a significant
effect on OS. Although radiological imaging is the gold standard for DIPG
diagnosis, the urgent need to improve the survival has led to the
reconsideration of biopsy with the aim to better understand the molecular
profile of DIPG and support personalized treatment. Methods: In this study, we present a single-center experience in treating DIPG
patients at disease progression combining targeted therapies with standard
of care. Biopsy was proposed to all patients at diagnosis or disease
progression. First-line treatment included RT and nimotuzumab/vinorelbine or
temozolomide. Immunohistochemistry-targeted research included study of
mTOR/p-mTOR pathway and BRAFv600E. Molecular analyses
included polymerase chain reaction, followed by Sanger sequences and/or
next-generation sequencing. Results: Based on the molecular profile, targeted therapy was administered in 9 out of
25 patients, while the remaining 16 patients were treated with standard of
care. Personalized treatment included inhibition of the PI3K/AKT/mTOR
pathway (5/9), PI3K/AKT/mTOR pathway and BRAFv600E (1/9),
ACVR1 (2/9) and PDGFRA (1/9); no
severe side effects were reported during treatment. Response to treatment
was evaluated according to Response Assessment in Pediatric Neuro-Oncology
criteria, and the overall response rate within the cohort was 66%. Patients
treated with targeted therapies were compared with the control cohort of 16
patients. Clinical and pathological characteristics of the two cohorts were
homogeneous. Median OS in the personalized treatment and control cohort was
20.26 and 14.18 months, respectively (p = 0.032). In our
experience, the treatment associated with the best OS was everolimus. Conclusion: Despite the small simple size of our study, our data suggest a prognostic
advantage and a safe profile of targeted therapies in DIPG patients, and we
strongly advocate to reconsider the role of biopsy for these patients.
Collapse
Affiliation(s)
- Giada Del Baldo
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Carai
- Neurosurgery Unit, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Piazza Sant'Onofrio 4, 00165 Rome, Italy
| | - Rachid Abbas
- CESP, INSERM, Université Paris Sud, Villejuif, France
| | - Antonella Cacchione
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Mara Vinci
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Valentina Di Ruscio
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giovanna Stefania Colafati
- Oncological Neuroradiology Unit, Imaging Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sabrina Rossi
- Pathology Unit, Department of Laboratories, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Nicola Maestro
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sara Temelso
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK.,Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Giulia Pericoli
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Emmanuel De Billy
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Isabella Giovannoni
- Pathology Unit, Department of Laboratories, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Alessia Carboni
- Oncological Neuroradiology Unit, Imaging Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Martina Rinelli
- Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Emanuele Agolini
- Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Alan Mackay
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK.,Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Chris Jones
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK.,Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Silvia Chiesa
- Department of Radiotherapy, Fondazione Policlinico Universitario "A. Gemelli," Catholic University of Sacred Heart, Rome, Italy
| | - Mario Balducci
- Department of Radiotherapy, Fondazione Policlinico Universitario "A. Gemelli," Catholic University of Sacred Heart, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Department of Life Sciences and Public Health, Fondazione Policlinico Universitario "A. Gemelli," Catholic University of Sacred Heart, Rome, Italy
| | - Angela Mastronuzzi
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
25
|
A microRNA Prognostic Signature in Patients with Diffuse Intrinsic Pontine Gliomas through Non-Invasive Liquid Biopsy. Cancers (Basel) 2022; 14:cancers14174307. [PMID: 36077842 PMCID: PMC9454461 DOI: 10.3390/cancers14174307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Diffuse intrinsic pontine glioma (DIPG) is a neuro-radiologically defined tumor of the brainstem, primarily affecting children, with most diagnoses occurring between 5 and 7 years of age. Surgical removal in DIPGs is not feasible. Subsequent tumor progression is almost universal and no biomarker for predicting the course of the disease has entered into clinical practice so far. Under these premises, it is essential to develop reliable biomarkers that are able to improve outcomes and stratify patients using non-invasive methods to determine tumor profiles. We designed a study assessing circulating miRNA expression by a high-throughput platform and divided patients into training and validation phases in order to disclose a potential signature with clinical impact. Our results for the first time have proved the usefulness of blood-circulating nucleic acids as powerful, easy-to-assay molecular markers of disease status in DIPG. Abstract Diffuse midline gliomas (DMGs) originate in the thalamus, brainstem, cerebellum and spine. This entity includes tumors that infiltrate the pons, called diffuse intrinsic pontine gliomas (DIPGs), with a rapid onset and devastating neurological symptoms. Since surgical removal in DIPGs is not feasible, the purpose of this study was to profile circulating miRNA expression in DIPG patients in an effort to identify a non-invasive prognostic signature with clinical impact. Using a high-throughput platform, miRNA expression was profiled in serum samples collected at the time of MRI diagnosis and prior to radiation and/or systemic therapy from 47 patients enrolled in clinical studies, combining nimotuzumab and vinorelbine with concomitant radiation. With progression-free survival as the primary endpoint, a semi-supervised learning approach was used to identify a signature that was also tested taking overall survival as the clinical endpoint. A signature comprising 13 circulating miRNAs was identified in the training set (n = 23) as being able to stratify patients by risk of disease progression (log-rank p = 0.00014; HR = 7.99, 95% CI 2.38–26.87). When challenged in a separate validation set (n = 24), it confirmed its ability to predict progression (log-rank p = 0.00026; HR = 5.51, 95% CI 2.03–14.9). The value of our signature was also confirmed when overall survival was considered (log-rank p = 0.0021, HR = 4.12, 95% CI 1.57–10.8). We have identified and validated a prognostic marker based on the expression of 13 circulating miRNAs that can shed light on a patient’s risk of progression. This is the first demonstration of the usefulness of nucleic acids circulating in the blood as powerful, easy-to-assay molecular markers of disease status in DIPG. This study provides Class II evidence that a signature based on 13 circulating miRNAs is associated with the risk of disease progression.
Collapse
|
26
|
Barragán-Pérez EJ, Alvarez-Amado DE, Dies-Suarez P, Tobón SH, García-Beristain JC, Peñaloza-González JG. Compassionate use of Quantum Magnetic Resonance Therapy for treatment of children with Diffuse Brainstem Glioma in Mexico City: a single institutional experience. J Neurooncol 2022; 157:377-382. [PMID: 35266065 DOI: 10.1007/s11060-022-03972-2] [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] [Received: 10/20/2021] [Accepted: 02/18/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE Diffuse Brainstem Glioma (DBG) is a catastrophic brain tumor with a survival rate of less than 10% two years after diagnosis despite the existence of different treatment protocols. Among the devices that use magnetic fields generated by Magnetic Resonance Imaging is Quantum Magnetic Resonance Therapy (QMRT). METHODS Five children diagnosed with DBG in our institution in Mexico City underwent treatment of compassionate use with QMRT between December 2018 and July 2019. A survival analysis was performed with previously reported historical data (n = 15). RESULTS Two patients (40%) survived after three years of follow-up; the log-rank test showed a statistically significant difference in overall survival between both groups (p = 0.032). All patients tolerated the treatment adequately without reporting any severe clinical or neuroradiological adverse effects. Of the patients included, all showed a decrease in the tumor one month after the end of the treatment, although there was great variability in the response and the difference was not statistically significant (p = 0.06). CONCLUSIONS Although future investigations are needed to confirm the findings reported in the present study, the improvement in survival is promising for a group of patients whose prognosis has been catastrophic over the years. Trial registration NCT03577600.
Collapse
Affiliation(s)
| | | | - Pilar Dies-Suarez
- Imagenology Department, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Silvia Hidalgo Tobón
- Imagenology Department, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
- Department of Physics, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | | | | |
Collapse
|
27
|
Madhavan K, Balakrishnan I, Lakshmanachetty S, Pierce A, Sanford B, Fosmire S, Elajaili HB, Walker F, Wang D, Nozik ES, Mitra SS, Dahl NA, Vibhakar R, Venkataraman S. Venetoclax cooperates with ionizing radiation to attenuate Diffuse Midline Glioma tumor growth. Clin Cancer Res 2022; 28:2409-2424. [PMID: 35344040 DOI: 10.1158/1078-0432.ccr-21-4002] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/10/2022] [Accepted: 03/24/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Tumor relapse after radiation therapy (RT) is a major hurdle in treating pediatric H3K27M-mutant diffuse midline gliomas (DMGs). RT-induced stress increases association of BCL2 family of proteins with BH3 pro-apoptotic activators preventing apoptosis. We hypothesized that inhibition of RT-induced BCL2 with a clinically relevant inhibitor, venetoclax, will block BCL2 activity leading to increased apoptosis. BCL2 has never been implicated in DMG as a RT-induced resistant mechanism. EXPERIMENTAL DESIGN We performed an integrated genomic analysis to determine genes responsible for radioresistance and a targeted drug screen to identify drugs that synergize with radiation in DMG. Effect of venetoclax on radiation-na�ve and 6Gy radiation on cells was evaluated by studying cell death, changes in BCL2 phosphorylation, reactive oxygen species (ROS), and apoptosis, as well as BCL2 association with BH3 apoptosis initiators. The efficacy of combining venetoclax with radiation was evaluated in vivo using orthotopic xenograft models. RESULTS BCL2 was identified as a key regulator of tumor growth after radiation in DMGs. Radiation sensitizes DMGs to venetoclax treatment independent of p53 status. Venetoclax as a monotherapy was not cytotoxic to DMG cells. Post-radiation venetoclax treatment significantly increased cell death, reduced BCL2-BIM association and augmented mitochondrial ROS leading to increased apoptosis. Combining venetoclax with RT significantly enhanced the survival of mice with DMG tumors. CONCLUSIONS This study shows that venetoclax impedes the anti-apoptotic function of radiation-induced BCL2 in DMG leading to increased apoptosis. Results from these pre-clinical studies demonstrate the potential use of the BCL2 inhibitor, venetoclax, combined with RT for pediatric DMG.
Collapse
Affiliation(s)
- Krishna Madhavan
- University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | | | | | - Angela Pierce
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Bridget Sanford
- University of Colorado Anschutz Medical Campus, United States
| | - Susan Fosmire
- University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Hanan B Elajaili
- University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Faye Walker
- University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Dong Wang
- University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Eva S Nozik
- University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Siddhartha S Mitra
- University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Nathan A Dahl
- University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | | | | |
Collapse
|
28
|
Elhemaly A, Refaey OE, Rizkallah RS, Zagulol MS. Palliative and end-of-life symptoms management for children with diffuse intrinsic pontine glioma. Future Oncol 2022; 18:1943-1950. [PMID: 35193393 DOI: 10.2217/fon-2021-1455] [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: 11/21/2022] Open
Abstract
Background: This study aimed to investigate diffuse intrinsic pontine glioma-specific symptoms in the last 12 weeks before death and to describe current palliative care. Materials & methods: A retrospective study included 80 pediatric diffuse intrinsic pontine glioma patients diagnosed between January 2018 and December 2019. Results: The most frequently encountered symptoms were headache, gait disturbance, vomiting, dysphagia, sensory loss, seizures and constipation. Steroids were used in 96% of patients with a high success rate, as well as analgesics (67.5%), antiemetics (59%), neuropathic medication (42.5%) and anticonvulsants (37.5%). Re-irradiation improved symptoms in 50% of patients. Conclusion: Steroids were efficient in managing many symptoms, with tolerated side effects. The symptomatic treatment succeeded in relieving end-of-life symptoms. Re-irradiation should be considered a good palliative tool in addition to regular symptomatic treatment.
Collapse
Affiliation(s)
- Ahmed Elhemaly
- Department of Pediatric Oncology, National Cancer Institute, Cairo University & Children's Cancer Hospital Egypt (CCHE-57357), Cairo, Egypt
| | - Osama El Refaey
- Department of Pediatric Oncology & Palliative Care, Children's Cancer Hospital Egypt (CCHE-57357), Cairo, Egypt
| | - Reda Samuel Rizkallah
- Department of Palliative Care, Children's Cancer Hospital Egypt (CCHE-57357), Cairo, Egypt
| | - Mohamed S Zagulol
- Department of Radiation Oncology, National Cancer Institute, Cairo University & Children's Cancer Hospital Egypt (CCHE-57357), Cairo, Egypt
| |
Collapse
|
29
|
Chorioallantoic membrane (CAM) assay to study treatment effects in diffuse intrinsic pontine glioma. PLoS One 2022; 17:e0263822. [PMID: 35157705 PMCID: PMC8843199 DOI: 10.1371/journal.pone.0263822] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 01/27/2022] [Indexed: 11/24/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a lethal pediatric brain tumor. While there are a number of in vivo rodent models for evaluating tumor biology and response to therapy, these models require significant time and resources. Here, we established the chick-embryo chorioallantoic (CAM) assay as an affordable and time efficient xenograft model for testing a variety of treatment approaches for DIPG. We found that patient-derived DIPG tumors develop in the CAM and maintain the same genetic and epigenetic characteristics of native DIPG tumors. We monitored tumor response to pharmaco- and radiation therapy by 3-D ultrasound volumetric and vasculature analysis. In this study, we established and validated the CAM model as a potential intermediate xenograft model for DIPG and its use for testing novel treatment approaches that include pharmacotherapy or radiation.
Collapse
|
30
|
He L, He D, Qi Y, Zhou J, Yuan C, Chang H, Wang Q, Li G, Shao Q. Stereotactic Biopsy for Brainstem Lesions: A Meta-analysis with Noncomparative Binary Data. Cancer Control 2021; 28:10732748211059858. [PMID: 34875878 PMCID: PMC8670786 DOI: 10.1177/10732748211059858] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Objectives To evaluate the diagnostic yield and safety of brainstem stereotactic biopsy
for brainstem lesions. Methods We performed a meta-analysis of English articles retrieved from the PubMed,
Web of Science, Cochrane Library, and APA psycInfo databases up to May 12,
2021. A binary fixed-effect model, the inverse variance method, or a binary
random-effect model, the Dersimonian Laird method, were utilized for pooling
the data. This meta-analysis was registered with INPLASY,
INPLASY202190034. Findings A total of 41 eligible studies with 2792 participants were included. The
weighted average diagnostic yield was 97.0% (95% confidential interval [CI],
96.0-97.9%). The weighted average proportions of temporary complications,
permanent deficits, and deaths were 6.2% (95% CI, 4.5–7.9%), .5% (95% CI,
.2–.8%), and .3% (95% CI, .1–.5%), respectively. The subgroup analysis
indicated a nearly identical weighted average diagnostic yield between
MRI-guided stereotactic biopsy and CT-guided stereotactic biopsy (95.9% vs
95.8%) but slightly increased proportions of temporary complications (7.9%
vs 6.0%), permanent deficits (1.9% vs .2%), and deaths (1.1% vs .4%) in the
former compared to the latter. Moreover, a greater weighted average
diagnostic yield (99.2% vs 97.6%) and lower proportions of temporary
complications (5.1% vs 6.8%) and deaths (.7% vs 1.5%) were shown in the
pediatric patient population than in the adult patient population. Conclusions Brainstem stereotactic biopsy demonstrates striking accuracy plus satisfying
safety in the diagnosis of brainstem lesions. The diagnostic yield,
morbidity, and mortality mildly vary based on the diversity of assistant
techniques and subject populations.
Collapse
Affiliation(s)
- Lin He
- Department of Radiotherapy, Tangdu Hospital, 56697Air Force Military Medical University, Xi'an, China
| | - Dongjie He
- Department of Radiotherapy, Tangdu Hospital, 56697Air Force Military Medical University, Xi'an, China
| | - Yuhong Qi
- Department of Radiotherapy, Tangdu Hospital, 56697Air Force Military Medical University, Xi'an, China
| | - Jiejing Zhou
- Department of Radiotherapy, Tangdu Hospital, 56697Air Force Military Medical University, Xi'an, China
| | - Canliang Yuan
- Department of Radiotherapy, Tangdu Hospital, 56697Air Force Military Medical University, Xi'an, China
| | - Hao Chang
- Department of Radiotherapy, Tangdu Hospital, 56697Air Force Military Medical University, Xi'an, China
| | - Qiming Wang
- Department of Radiotherapy, Tangdu Hospital, 56697Air Force Military Medical University, Xi'an, China
| | - Gaiyan Li
- Department of Radiotherapy, Tangdu Hospital, 56697Air Force Military Medical University, Xi'an, China
| | - Qiuju Shao
- Department of Radiotherapy, Tangdu Hospital, 56697Air Force Military Medical University, Xi'an, China
| |
Collapse
|
31
|
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: 17] [Impact Index Per Article: 4.3] [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
|
32
|
Ozerov SS, Ryzhova MV, Kumirova EV. [Diffuse brainstem tumors in children. Tumor biology and hope for a better outcome. Current state of the problem]. ZHURNAL VOPROSY NEĬROKHIRURGII IMENI N. N. BURDENKO 2021; 85:77-86. [PMID: 34463454 DOI: 10.17116/neiro20218504177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Diffuse brainstem tumor is a fatal disease and the main cause of child mortality from neoplasms of central nervous system. So far, no effective therapy has been found for this disease. The authors discuss the modern aspects of clinical data, biology, diagnosis and treatment of patients with diffuse brainstem tumors.
Collapse
Affiliation(s)
- S S Ozerov
- Dmitry Rogachev National Medical Research Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - M V Ryzhova
- Burdenko Neurosurgical Center, Moscow, Russia
| | - E V Kumirova
- Dmitry Rogachev National Medical Research Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| |
Collapse
|
33
|
Ruttens D, Messiaen J, Ferster A, Piette C, Schifflers S, Van Damme A, van der Werff Ten Bosch J, Verlooy J, Willems L, Jacobs S. Retrospective study of diffuse intrinsic pontine glioma in the Belgian population: a 25 year experience. J Neurooncol 2021; 153:293-301. [PMID: 33939103 DOI: 10.1007/s11060-021-03766-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/23/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Diffuse intrinsic pontine glioma is a rare disease with a high mortality. Our primary aim was to determine the incidence of this disease in Belgium. Secondly, we wanted to compare the treatment approach of Belgian pediatric oncology centres, to investigate possibilities for improvement. METHODS We retrospectively collected and analysed data on DIPG-patients diagnosed between 1994 and 2018 and recorded in the Belgian Cancer Registry. We included patients ≤ 18 years who were followed in one of the eight Belgian pediatric oncology centres. RESULTS We included 100 patients. Files were complete in 87 patients. We observed an increase in diagnoses with an incidence of 3.1 per 1,000,000 persons (aged 0-≤ 18) per year over the last 5 years compared to an overall incidence of 1.8. Biopsy was performed at diagnosis in 51.7% of patients. In one fifth this was study-related. Mutation analysis was known in eight patients, of which six showed the H3 K27M-mutation. 58.8% of patients received chemotherapy, without a significant survival benefit. 12.6% of patients were included in a clinical trial. Biopsy rate and the use of chemotherapy differed widely between centres. Mean OS and PFS were 10.49 and 4.87 months respectively. We observed an improved survival over time. CONCLUSIONS Over the past 25 years, we observed an increase of new DIPG-diagnoses. Outcome in our cohort is comparable with literature findings. We demonstrate an important heterogeneity in treatment approach between different centres and limited inclusion in clinical trials. Therefore, collaboration between centres and inclusion of patients in clinical trials is much needed.
Collapse
Affiliation(s)
- Dries Ruttens
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium.
| | - Julie Messiaen
- Department of Pediatric Hematology-Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Alina Ferster
- Department of Pediatric Hematology-Oncology, Queen Fabiola Children's University Hospital, Brussels, Belgium
| | - Caroline Piette
- Department of Pediatrics, Division of Hematology-Oncology, CHU of Liège, Liège, Belgium
| | - Stefan Schifflers
- Department of Pediatric Hematology-Oncology, CHC MontLégia, Liège, Belgium
| | - An Van Damme
- Department of Pediatric Hematology-Oncology, Saint-Luc University Hospital, Brussels, Belgium
| | | | - Joris Verlooy
- Department of Pediatric Hematology-Oncology, University Hospital of Antwerp, Edegem, Belgium
| | - Leen Willems
- Department of Pediatric Hematology-Oncology, University Hospital Ghent, Ghent, Belgium
| | - Sandra Jacobs
- Department of Pediatric Hematology-Oncology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
34
|
Baxter PA, Su JM, Onar-Thomas A, Billups CA, Li XN, Poussaint TY, Smith ER, Thompson P, Adesina A, Ansell P, Giranda V, Paulino A, Kilburn L, Quaddoumi I, Broniscer A, Blaney SM, Dunkel IJ, Fouladi M. A phase I/II study of veliparib (ABT-888) with radiation and temozolomide in newly diagnosed diffuse pontine glioma: a Pediatric Brain Tumor Consortium study. Neuro Oncol 2021; 22:875-885. [PMID: 32009149 DOI: 10.1093/neuonc/noaa016] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A Pediatric Brain Tumor Consortium (PBTC) phase I/II trial of veliparib and radiation followed by veliparib and temozolomide (TMZ) was conducted in children with newly diagnosed diffuse intrinsic pontine glioma (DIPG). The objectives were to: (i) estimate the recommended phase II dose (RP2D) of veliparib with concurrent radiation; (ii) evaluate the pharmacokinetic parameters of veliparib during radiation; (iii) evaluate feasibility of intrapatient TMZ dose escalation; (iv) describe toxicities of protocol therapy; and (v) estimate the overall survival distribution compared with historical series. METHODS Veliparib was given Monday through Friday b.i.d. during radiation followed by a 4-week rest. Patients then received veliparib at 25 mg/m2 b.i.d. and TMZ 135 mg/m2 daily for 5 days every 28 days. Intrapatient dose escalation of TMZ was investigated for patients experiencing minimal toxicity. RESULTS Sixty-six patients (65 eligible) were enrolled. The RP2D of veliparib was 65 mg/m2 b.i.d. with radiation. Dose-limiting toxicities during radiation with veliparib therapy included: grade 2 intratumoral hemorrhage (n = 1), grade 3 maculopapular rash (n = 2), and grade 3 nervous system disorder (generalized neurologic deterioration) (n = 1). Intrapatient TMZ dose escalation during maintenance was not tolerated. Following a planned interim analysis, it was concluded that this treatment did not show a survival benefit compared with PBTC historical controls, and accrual was stopped for futility. The 1- and 2-year overall survival rates were 37.2% (SE 7%) and 5.3% (SE 3%), respectively. CONCLUSION Addition of veliparib to radiation followed by TMZ and veliparib was tolerated but did not improve survival for patients with newly diagnosed DIPG. TRIAL REGISTRATION NCT01514201.
Collapse
Affiliation(s)
- Patricia A Baxter
- Texas Children's Hospital/Baylor College of Medicine, Houston, Texas
| | - Jack M Su
- Texas Children's Hospital/Baylor College of Medicine, Houston, Texas
| | | | | | - Xiao-Nan Li
- Texas Children's Hospital/Baylor College of Medicine, Houston, Texas
| | | | | | - Patrick Thompson
- University of North Carolina Children's Hospital, Chapel Hill, North Carolina
| | - Adekunle Adesina
- Texas Children's Hospital/Baylor College of Medicine, Houston, Texas
| | | | | | - Arnold Paulino
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | - Susan M Blaney
- Texas Children's Hospital/Baylor College of Medicine, Houston, Texas
| | - Ira J Dunkel
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
35
|
Dahl NA, Donson AM, Sanford B, Wang D, Walker FM, Gilani A, Foreman NK, Tinkle CL, Baker SJ, Hoffman LM, Venkataraman S, Vibhakar R. NTRK Fusions Can Co-Occur With H3K27M Mutations and May Define Druggable Subclones Within Diffuse Midline Gliomas. J Neuropathol Exp Neurol 2021; 80:345-353. [PMID: 33749791 PMCID: PMC7985828 DOI: 10.1093/jnen/nlab016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Diffuse midline gliomas (DMGs) are incurable pediatric tumors with extraordinarily limited treatment options. Decades of clinical trials combining conventional chemotherapies with radiation therapy have failed to improve these outcomes, demonstrating the need to identify and validate druggable biologic targets within this disease. NTRK1/2/3 fusions are found in a broad range of pediatric cancers, including high-grade gliomas and a subset of DMGs. Phase 1/2 studies of TRK inhibitors have demonstrated good tolerability, effective CNS penetration, and promising objective responses across all patients with TRK fusion-positive cancers, but their use has not been explored in TRK fusion-positive DMG. Here, we report 3 cases of NTRK fusions co-occurring within H3K27M-positive pontine diffuse midline gliomas. We employ a combination of single-cell and bulk transcriptome sequencing from TRK fusion-positive DMG to describe the phenotypic consequences of this co-occurring alteration. We then use ex vivo short-culture assays to evaluate the potential response to TRK inhibition in this disease. Together, these data highlight the importance of routine molecular characterization of these highly aggressive tumors and identify a small subset of patients that may benefit from currently available targeted therapies.
Collapse
Affiliation(s)
- Nathan A Dahl
- From the Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Andrew M Donson
- From the Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Bridget Sanford
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Dong Wang
- From the Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Faye M Walker
- From the Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ahmed Gilani
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Nicholas K Foreman
- From the Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado, Aurora, Colorado, USA
- Department of Neurosurgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Christopher L Tinkle
- Department of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Suzanne J Baker
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Lindsey M Hoffman
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, Arizona, USA
| | - Sujatha Venkataraman
- From the Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Rajeev Vibhakar
- From the Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado, Aurora, Colorado, USA
- Department of Neurosurgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
36
|
Advanced Spheroid, Tumouroid and 3D Bioprinted In-Vitro Models of Adult and Paediatric Glioblastoma. Int J Mol Sci 2021; 22:ijms22062962. [PMID: 33803967 PMCID: PMC8000246 DOI: 10.3390/ijms22062962] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 12/16/2022] Open
Abstract
The life expectancy of patients with high-grade glioma (HGG) has not improved in decades. One of the crucial tools to enable future improvement is advanced models that faithfully recapitulate the tumour microenvironment; they can be used for high-throughput screening that in future may enable accurate personalised drug screens. Currently, advanced models are crucial for identifying and understanding potential new targets, assessing new chemotherapeutic compounds or other treatment modalities. Recently, various methodologies have come into use that have allowed the validation of complex models—namely, spheroids, tumouroids, hydrogel-embedded cultures (matrix-supported) and advanced bioengineered cultures assembled with bioprinting and microfluidics. This review is designed to present the state of advanced models of HGG, whilst focusing as much as is possible on the paediatric form of the disease. The reality remains, however, that paediatric HGG (pHGG) models are years behind those of adult HGG. Our goal is to bring this to light in the hope that pGBM models can be improved upon.
Collapse
|
37
|
Chastkofsky MI, Pituch KC, Katagi H, Zannikou M, Ilut L, Xiao T, Han Y, Sonabend AM, Curiel DT, Bonner ER, Nazarian J, Horbinski CM, James CD, Saratsis AM, Hashizume R, Lesniak MS, Balyasnikova IV. Mesenchymal Stem Cells Successfully Deliver Oncolytic Virotherapy to Diffuse Intrinsic Pontine Glioma. Clin Cancer Res 2021; 27:1766-1777. [PMID: 33272983 PMCID: PMC7956061 DOI: 10.1158/1078-0432.ccr-20-1499] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 10/20/2020] [Accepted: 11/30/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Diffuse intrinsic pontine glioma (DIPG) is among the deadliest of pediatric brain tumors. Radiotherapy is the standard-of-care treatment for DIPG, but offers only transient relief of symptoms for patients with DIPG without providing significant survival benefit. Oncolytic virotherapy is an anticancer treatment that has been investigated for treating various types of brain tumors. EXPERIMENTAL DESIGN Here, we have explored the use of mesenchymal stem cells (MSC) for oncolytic virus (OV) delivery and evaluated treatment efficacy using preclinical models of DIPG. The survivin promoter drives the conditional replication of OV used in our studies. The efficiency of OV entry into the cells is mediated by fiber modification with seven lysine residues (CRAd.S.pK7). Patients' samples and cell lines were analyzed for the expression of viral entry proteins and survivin. The ability of MSCs to deliver OV to DIPG was studied in the context of a low dose of irradiation. RESULTS Our results show that DIPG cells and tumors exhibit robust expression of cell surface proteins and survivin that enable efficient OV entry and replication in DIPG cells. MSCs loaded with OV disseminate within a tumor and release OV throughout the DIPG brainstem xenografts in mice. Administration of OV-loaded MSCs with radiotherapy to mice bearing brainstem DIPG xenografts results in more prolonged survival relative to that conferred by either therapy alone (P < 0.01). CONCLUSIONS Our study supports OV, CRAd.S.pK7, encapsulated within MSCs as a therapeutic strategy that merits further investigation and potential translation for DIPG treatment.
Collapse
Affiliation(s)
- Michael I Chastkofsky
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Katarzyna C Pituch
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Hiroaki Katagi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Markella Zannikou
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Liliana Ilut
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Ting Xiao
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Yu Han
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - David T Curiel
- Department of Radiation Oncology, University of Washington, St. Louis, Missouri
| | - Erin R Bonner
- Center for Genomics and Precision Medicine, Children's National Medical Center, Washington, D.C
- Institute for Biomedical Sciences, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Javad Nazarian
- Center for Genomics and Precision Medicine, Children's National Medical Center, Washington, D.C
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, D.C
| | - Craig M Horbinski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - C David James
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Amanda M Saratsis
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Division of Neurosurgery, Department of Pediatric Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Rintaro Hashizume
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Irina V Balyasnikova
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| |
Collapse
|
38
|
Li D, Bonner ER, Wierzbicki K, Panditharatna E, Huang T, Lulla R, Mueller S, Koschmann C, Nazarian J, Saratsis AM. Standardization of the liquid biopsy for pediatric diffuse midline glioma using ddPCR. Sci Rep 2021; 11:5098. [PMID: 33658570 PMCID: PMC7930089 DOI: 10.1038/s41598-021-84513-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 02/05/2021] [Indexed: 01/15/2023] Open
Abstract
Diffuse midline glioma (DMG) is a highly morbid pediatric brain tumor. Up to 80% of DMGs harbor mutations in histone H3-encoding genes, associated with poor prognosis. We previously showed the feasibility of detecting H3 mutations in circulating tumor DNA (ctDNA) in the liquid biome of children diagnosed with DMG. However, detection of low levels of ctDNA is highly dependent on platform sensitivity and sample type. To address this, we optimized ctDNA detection sensitivity and specificity across two commonly used digital droplet PCR (ddPCR) platforms (RainDance and BioRad), and validated methods for detecting H3F3A c.83A > T (H3.3K27M) mutations in DMG CSF, plasma, and primary tumor specimens across three different institutions. DNA was extracted from H3.3K27M mutant and H3 wildtype (H3WT) specimens, including H3.3K27M tumor tissue (n = 4), CSF (n = 6), plasma (n = 4), and human primary pediatric glioma cells (H3.3K27M, n = 2; H3WT, n = 1). ctDNA detection was enhanced via PCR pre-amplification and use of distinct custom primers and fluorescent LNA probes for c.83 A > T H3F3A mutation detection. Mutation allelic frequency (MAF) was determined and validated through parallel analysis of matched H3.3K27M tissue specimens (n = 3). We determined technical nuances between ddPCR instruments, and optimized sample preparation and sequencing protocols for H3.3K27M mutation detection and quantification. We observed 100% sensitivity and specificity for mutation detection in matched DMG tissue and CSF across assays, platforms and institutions. ctDNA is reliably and reproducibly detected in the liquid biome using ddPCR, representing a clinically feasible, reproducible, and minimally invasive approach for DMG diagnosis, molecular subtyping and therapeutic monitoring.
Collapse
Affiliation(s)
- Daphne Li
- Department of Neurological Surgery, Loyola University Medical Center, Maywood, IL, USA
| | - Erin R Bonner
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Kyle Wierzbicki
- Department of Pediatric Hematology/Oncology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | | | - Tina Huang
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rishi Lulla
- Department of Pediatric Hematology/Oncology, Brown Alpert Medical School, Providence, Rhode Island, USA
| | - Sabine Mueller
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Carl Koschmann
- Department of Pediatric Hematology/Oncology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Javad Nazarian
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA.
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
- Department of Oncology, Children's Research Center, Diffuse Midline Glioma (DMG) Research Center, University Children's Hospital Zürich, Zürich, Switzerland.
- The Brain Tumor Institute, Children's National Health System, Washington, DC, USA.
| | - Amanda M Saratsis
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Division of Pediatric Neurosurgery, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E Chicago Ave Box 28, Chicago, IL, 60614, USA.
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
39
|
Khan A, Gamble LD, Upton DH, Ung C, Yu DMT, Ehteda A, Pandher R, Mayoh C, Hébert S, Jabado N, Kleinman CL, Burns MR, Norris MD, Haber M, Tsoli M, Ziegler DS. Dual targeting of polyamine synthesis and uptake in diffuse intrinsic pontine gliomas. Nat Commun 2021; 12:971. [PMID: 33579942 PMCID: PMC7881014 DOI: 10.1038/s41467-021-20896-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is an incurable malignant childhood brain tumor, with no active systemic therapies and a 5-year survival of less than 1%. Polyamines are small organic polycations that are essential for DNA replication, translation and cell proliferation. Ornithine decarboxylase 1 (ODC1), the rate-limiting enzyme in polyamine synthesis, is irreversibly inhibited by difluoromethylornithine (DFMO). Herein we show that polyamine synthesis is upregulated in DIPG, leading to sensitivity to DFMO. DIPG cells compensate for ODC1 inhibition by upregulation of the polyamine transporter SLC3A2. Treatment with the polyamine transporter inhibitor AMXT 1501 reduces uptake of polyamines in DIPG cells, and co-administration of AMXT 1501 and DFMO leads to potent in vitro activity, and significant extension of survival in three aggressive DIPG orthotopic animal models. Collectively, these results demonstrate the potential of dual targeting of polyamine synthesis and uptake as a therapeutic strategy for incurable DIPG.
Collapse
Affiliation(s)
- Aaminah Khan
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW 2052 Australia
| | - Laura D. Gamble
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW 2052 Australia
| | - Dannielle H. Upton
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW 2052 Australia
| | - Caitlin Ung
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW 2052 Australia
| | - Denise M. T. Yu
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW 2052 Australia
| | - Anahid Ehteda
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW 2052 Australia
| | - Ruby Pandher
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW 2052 Australia
| | - Chelsea Mayoh
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW 2052 Australia
| | - Steven Hébert
- grid.14709.3b0000 0004 1936 8649Lady Davis Institute for Medical Research, Jewish General Hospital, Department of Human Genetics, McGill University, 3999 Côte Ste-Catherine Road, Montreal, QC H4A 3J1 Canada
| | - Nada Jabado
- grid.63984.300000 0000 9064 4811Department of Pediatrics, McGill University Health Center, 1001 Decarie Boulevard, Montreal, QC H4A 3J1 Canada
| | - Claudia L. Kleinman
- grid.14709.3b0000 0004 1936 8649Lady Davis Institute for Medical Research, Jewish General Hospital, Department of Human Genetics, McGill University, 3999 Côte Ste-Catherine Road, Montreal, QC H4A 3J1 Canada
| | - Mark R. Burns
- Aminex Therapeutics Inc., Suite #364, 6947 Coal Creek Parkway SE, Newcastle, WA 98059 USA
| | - Murray D. Norris
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW 2052 Australia ,grid.1005.40000 0004 4902 0432Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW 2052 Australia
| | - Michelle Haber
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW 2052 Australia ,grid.1005.40000 0004 4902 0432Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW 2052 Australia
| | - Maria Tsoli
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW 2052 Australia
| | - David S. Ziegler
- grid.1005.40000 0004 4902 0432Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW 2052 Australia ,grid.414009.80000 0001 1282 788XKids Cancer Centre, Sydney Children’s Hospital, High St, Randwick, 2031 Australia
| |
Collapse
|
40
|
Exploiting Gangliosides for the Therapy of Ewing's Sarcoma and H3K27M-Mutant Diffuse Midline Glioma. Cancers (Basel) 2021; 13:cancers13030520. [PMID: 33572900 PMCID: PMC7866294 DOI: 10.3390/cancers13030520] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Osteosarcoma, Ewing’s sarcoma, and H3K27M-mutant diffuse midline glioma are rare but aggressive malignancies occurring mainly in children. Due to their rareness and often fatal course, drug development is challenging. Here, we repurposed the existing drugs dinutuximab and eliglustat and investigated their potential to directly target or indirectly modulate the tumor cell-specific ganglioside GD2. Our data suggest that targeting and/or modulating tumor cell-specific GD2 may offer a new therapeutic strategy for the above mentioned tumor entities. Abstract The ganglioside GD2 is an important target in childhood cancer. Nevertheless, the only therapy targeting GD2 that is approved to date is the monoclonal antibody dinutuximab, which is used in the therapy of neuroblastoma. The relevance of GD2 as a target in other tumor entities remains to be elucidated. Here, we analyzed the expression of GD2 in different pediatric tumor entities by flow cytometry and tested two approaches for targeting GD2. H3K27M-mutant diffuse midline glioma (H3K27M-mutant DMG) samples showed the highest expression of GD2 with all cells strongly positive for the antigen. Ewing’s sarcoma (ES) samples also showed high expression, but displayed intra- and intertumor heterogeneity. Osteosarcoma had low to intermediate expression with a high percentage of GD2-negative cells. Dinutuximab beta in combination with irinotecan and temozolomide was used to treat a five-year-old girl with refractory ES. Disease control lasted over 12 months until a single partially GD2-negative intracranial metastasis was detected. In order to target GD2 in H3K27M-mutant DMG, we blocked ganglioside synthesis via eliglustat, since dinutuximab cannot cross the blood–brain barrier. Eliglustat is an inhibitor of glucosylceramide synthase, and it is used for treating children with Gaucher’s disease. Eliglustat completely inhibited the proliferation of primary H3K27M-mutant DMG cells in vitro. In summary, our data provide evidence that dinutuximab might be effective in tumors with high GD2 expression. Moreover, disrupting the ganglioside metabolism in H3K27M-mutant DMG could open up a new therapeutic option for this highly fatal cancer.
Collapse
|
41
|
Deland K, Starr BF, Mercer JS, Byemerwa J, Crabtree DM, Williams NT, Luo L, Ma Y, Chen M, Becher OJ, Kirsch DG. Tumor genotype dictates radiosensitization after Atm deletion in primary brainstem glioma models. J Clin Invest 2021; 131:142158. [PMID: 32990677 PMCID: PMC7773366 DOI: 10.1172/jci142158] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/23/2020] [Indexed: 12/31/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) kills more children than any other type of brain tumor. Despite clinical trials testing many chemotherapeutic agents, palliative radiotherapy remains the standard treatment. Here, we utilized Cre/loxP technology to show that deleting Ataxia telangiectasia mutated (Atm) in primary mouse models of DIPG can enhance tumor radiosensitivity. Genetic deletion of Atm improved survival of mice with p53-deficient but not p53 wild-type gliomas after radiotherapy. Similar to patients with DIPG, mice with p53 wild-type tumors had improved survival after radiotherapy independent of Atm deletion. Primary p53 wild-type tumor cell lines induced proapoptotic genes after radiation and repressed the NRF2 target, NAD(P)H quinone dehydrogenase 1 (Nqo1). Tumors lacking p53 and Ink4a/Arf expressed the highest level of Nqo1 and were most resistant to radiation, but deletion of Atm enhanced the radiation response. These results suggest that tumor genotype may determine whether inhibition of ATM during radiotherapy will be an effective clinical approach to treat DIPGs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yan Ma
- Department of Radiation Oncology
| | - Mark Chen
- Department of Pharmacology & Cancer Biology
- Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA
| | - Oren J. Becher
- Department of Pediatrics and
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois, USA
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - David G. Kirsch
- Department of Radiation Oncology
- Department of Pharmacology & Cancer Biology
| |
Collapse
|
42
|
Robertson EG, Wakefield CE, Tsoli M, Kellie SJ, Alvaro F, Gifford AJ, Weber MA, Rodriguez M, Kirby M, Ziegler DS. Parents' experiences of postmortem tumor donation for high-grade gliomas: benefits and suggested improvements. Neurooncol Adv 2021; 3:vdab087. [PMID: 34458732 PMCID: PMC8386242 DOI: 10.1093/noajnl/vdab087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Pediatric high-grade glioma is a devastating diagnosis. There has been no improvement in outcomes for several decades, with few children surviving 2 years postdiagnosis. Research progress has been hampered by a lack of tumor samples, which can be used to develop and test novel therapies. Postmortem tumor donations are therefore a valuable opportunity to collect tissue. In this study, we explored Australian parents' experiences of donating their child's tumor for research after their child had died. METHODS We collected qualitative data from 11 bereaved parents who consented to donate samples of their child's high-grade glioma for research postmortem. We asked parents about their perceived benefits/burdens of the autopsy, recommendations for improving consent discussions, and decision regret. RESULTS Parents hoped that their donation would help to find a cure for future children with high-grade glioma. They described feeling comforted knowing that their child's suffering may help others. Some parents also felt that the donation would help them better understand their child's tumor. Although some parents described discomfort about procedures leading up to the autopsy, parents reported minimal regret regarding their decision to donate their child's tumor. Parents provided recommendations to improve consent discussions, such as providing more information about the autopsy logistics and why the donation was needed. CONCLUSION Parents consented to autopsy for altruistic reasons, although donation may also assist parents in their grieving. There is a strong need to improve access to tumor donations for any family who wishes to donate.
Collapse
Affiliation(s)
- Eden G Robertson
- School of Women’s and Children’s Health, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- Behavioural Sciences Unit, Kids Cancer Centre, Sydney Children’s Hospital, Sydney, New South Wales, Australia
| | - Claire E Wakefield
- School of Women’s and Children’s Health, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- Behavioural Sciences Unit, Kids Cancer Centre, Sydney Children’s Hospital, Sydney, New South Wales, Australia
| | - Maria Tsoli
- School of Women’s and Children’s Health, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- Children’s Cancer Institute, UNSW Sydney, Sydney, New South Wales, Australia
| | - Stewart J Kellie
- Children’s Cancer Centre, The Children’s Hospital at Westmead, Sydney, New South Wales, Australia
- Discipline of Child and Adolescent Health, University of Sydney, Sydney, New South Wales, Australia
| | - Frank Alvaro
- Children’s Cancer and Blood Disorders, John Hunter Children’s Hospital, Newcastle, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Andrew J Gifford
- School of Women’s and Children’s Health, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- Children’s Cancer Institute, UNSW Sydney, Sydney, New South Wales, Australia
- Anatomical Pathology, NSW Health Pathology East (Prince of Wales Hospital), Sydney, New South Wales, Australia
| | - Martin A Weber
- Anatomical Pathology, NSW Health Pathology East (Prince of Wales Hospital), Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Michael Rodriguez
- Anatomical Pathology, NSW Health Pathology East (Prince of Wales Hospital), Sydney, New South Wales, Australia
| | - Maria Kirby
- Department of Haematology and Oncology, Women’s and Children’s Hospital, Adelaide, South Australia, Australia
| | - David S Ziegler
- School of Women’s and Children’s Health, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- Children’s Cancer Institute, UNSW Sydney, Sydney, New South Wales, Australia
- Kids Cancer Centre, Sydney Children’s Hospital, Randwick, New South Wales, Australia
| |
Collapse
|
43
|
Perrone MG, Ruggiero A, Centonze A, Carrieri A, Ferorelli S, Scilimati A. Diffuse Intrinsic Pontine Glioma (DIPG): Breakthrough and Clinical Perspective. Curr Med Chem 2021; 28:3287-3317. [PMID: 32767913 DOI: 10.2174/0929867327666200806110206] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/16/2020] [Accepted: 07/23/2020] [Indexed: 11/22/2022]
Abstract
Diffuse intrinsic pontine glioma (DIPG) mainly affects children with a median age of 6-7 years old. It accounts for 10% of all pediatric tumors. Unfortunately, DIPG has a poor prognosis, and the median survival is generally less than 16-24 months independently from the treatment received. Up to now, children with DIPG are treated with focal radiotherapy alone or in combination with antitumor agents. In the last decade, ONC201 known as dopamine receptor antagonist was uncovered, by a high throughput screening of public libraries of compounds, to be endowed with cytotoxic activity against several cancer cell lines. Efforts were made to identify the real ONC201 target, responsible for its antiproliferative effect. The hypothesized targets were the Tumor necrosis factor-Related Apoptosis-Inducing Ligand stimulation (TRAIL), two oncogenic kinases (ERK/AKT system) that target the same tumor-suppressor gene (FOXO3a), dopamine receptors (DRD2 and DRD3 subtypes) and finally the mitochondrial Caseynolitic Protease P (ClpP). ONC201 structure-activity relationship is extensively discussed in this review, together with other two classes of compounds, namely ADEPs and D9, already known for their antibiotic activity but noteworthy to be discussed and studied as potential "leads" for the development of new drugs to be used in the treatment of DIPG. In this review, a detailed and critical description of ONC201, ADEPs, and D9 pro-apoptotic activity is made, with particular attention to the specific interactions established with its targets that also are intimately described. Pubmed published patents and clinical trial reports of the last ten years were used as the bibliographic source.
Collapse
Affiliation(s)
- Maria Grazia Perrone
- Department of Pharmacy and Pharmaceutical Sciences, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Antonella Centonze
- Department of Pharmacy and Pharmaceutical Sciences, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| | - Antonio Carrieri
- Department of Pharmacy and Pharmaceutical Sciences, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| | - Savina Ferorelli
- Department of Pharmacy and Pharmaceutical Sciences, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| | - Antonio Scilimati
- Department of Pharmacy and Pharmaceutical Sciences, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| |
Collapse
|
44
|
Bennett J, Erker C, Lafay-Cousin L, Ramaswamy V, Hukin J, Vanan MI, Cheng S, Coltin H, Fonseca A, Johnston D, Lo A, Zelcer S, Alvi S, Bowes L, Brossard J, Charlebois J, Eisenstat D, Felton K, Fleming A, Jabado N, Larouche V, Legault G, Mpofu C, Perreault S, Silva M, Sinha R, Strother D, Tsang DS, Wilson B, Crooks B, Bartels U. Canadian Pediatric Neuro-Oncology Standards of Practice. Front Oncol 2020; 10:593192. [PMID: 33415075 PMCID: PMC7783450 DOI: 10.3389/fonc.2020.593192] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/16/2020] [Indexed: 11/13/2022] Open
Abstract
Primary CNS tumors are the leading cause of cancer-related death in pediatrics. It is essential to understand treatment trends to interpret national survival data. In Canada, children with CNS tumors are treated at one of 16 tertiary care centers. We surveyed pediatric neuro-oncologists to create a national standard of practice to be used in the absence of a clinical trial for seven of the most prevalent brain tumors in children. This allowed description of practice across the country, along with a consensus. This had a multitude of benefits, including understanding practice patterns, allowing for a basis to compare in future research and informing Health Canada of the current management of patients. This also allows all children in Canada to receive equivalent care, regardless of location.
Collapse
Affiliation(s)
- Julie Bennett
- Division of Neuro-Oncology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Craig Erker
- Division of Pediatric Hematology/Oncology, IWK Health Centre, Halifax, NS, Canada
| | - Lucie Lafay-Cousin
- Department of Oncology, Alberta Children's Hospital, Calgary, AB, Canada
| | - Vijay Ramaswamy
- Division of Neuro-Oncology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Juliette Hukin
- Division of Hematology, Oncology and Bone Marrow Transplant, British Columbia Children's Hospital, Vancouver, BC, Canada
| | | | - Sylvia Cheng
- Division of Hematology, Oncology and Bone Marrow Transplant, British Columbia Children's Hospital, Vancouver, BC, Canada
| | - Hallie Coltin
- Division of Hematology/Oncology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Adriana Fonseca
- Division of Neuro-Oncology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Donna Johnston
- Division of Hematology/Oncology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Andrea Lo
- Division of Radiation Oncology and Developmental Radiotherapeutics, BC Cancer Centre, Vancouver, BC, Canada
| | - Shayna Zelcer
- Division of Pediatric Hematology/Oncology, London Health Sciences Centre, London, ON, Canada
| | - Saima Alvi
- Pediatric Oncology, Saskatchewan Cancer Agency, Regina, SK, Canada
| | - Lynette Bowes
- Division of Pediatrics, Memorial University, St. John's, NF, Canada
| | - Josée Brossard
- Division of Pediatric Hematology/Oncology, Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Janie Charlebois
- Division of Pediatric Hematology/Oncology, Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - David Eisenstat
- Division of Pediatric Hematology/Oncology & Palliative Care, Stollery Children's Hospital, Edmonton, AB, Canada
| | - Kathleen Felton
- Division of Pediatric Hematology/Oncology, Jim Pattison Children's Hospital, Saskatoon, SK, Canada
| | - Adam Fleming
- Division of Pediatric Hematology/Oncology, McMaster Children's Hospital, Hamilton, ON, Canada
| | - Nada Jabado
- Division of Hematology/Oncology, Montreal Children's Hospital, Montreal, QC, Canada
| | - Valérie Larouche
- Division of Hematology/Oncology, CHU de Quebec, Quebec City, QC, Canada
| | - Geneviève Legault
- Division of Hematology/Oncology, Montreal Children's Hospital, Montreal, QC, Canada
| | - Chris Mpofu
- Division of Pediatric Hematology/Oncology, Jim Pattison Children's Hospital, Saskatoon, SK, Canada
| | | | - Mariana Silva
- Division of Pediatrics, Queen's University, Kingston, ON, Canada
| | - Roona Sinha
- Division of Pediatric Hematology/Oncology, Jim Pattison Children's Hospital, Saskatoon, SK, Canada
| | - Doug Strother
- Department of Oncology, Alberta Children's Hospital, Calgary, AB, Canada
| | - Derek S Tsang
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Beverly Wilson
- Division of Pediatric Hematology/Oncology & Palliative Care, Stollery Children's Hospital, Edmonton, AB, Canada
| | - Bruce Crooks
- Division of Pediatric Hematology/Oncology, IWK Health Centre, Halifax, NS, Canada
| | - Ute Bartels
- Division of Neuro-Oncology, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
45
|
Thomas L, Smith N, Saunders D, Zalles M, Gulej R, Lerner M, Fung KM, Carcaboso AM, Towner RA. OKlahoma Nitrone-007: novel treatment for diffuse intrinsic pontine glioma. J Transl Med 2020; 18:424. [PMID: 33168005 PMCID: PMC7654606 DOI: 10.1186/s12967-020-02593-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diffuse intrinsic pontine glioma (DIPG) is the most common brainstem cancer in childhood. This rapidly progressing brainstem glioma holds a very dismal prognosis with median survival of less than 1 year. Despite extensive research, no significant therapeutic advancements have been made to improve overall survival in DIPG patients. METHODS Here, we used an orthotopic xenograft pediatric DIPG (HSJD-DIPG-007) mouse model to monitor the effects of anti-cancer agent, OKlahoma Nitrone-007 (OKN-007), as an inhibitor of tumor growth after 28 days of treatment. Using magnetic resonance imaging (MRI), we confirmed the previously described efficacy of LDN-193189, a known activin A receptor, type I (ACVR1) inhibitor, in decreasing tumor burden and found that OKN-007 was equally efficacious. RESULTS After 28 days of treatment, the tumor volumes were significantly decreased in OKN-007 treated mice (p < 0.01). The apparent diffusion coefficient (ADC), as a measure of tissue structural alterations, was significantly decreased in OKN-007 treated tumor-bearing mice (p < 0.0001). Histological analysis also showed a significant decrease in CD34 expression, essential for angiogenesis, of OKN-007 treated mice (p < 0.05) compared to LDN-193189 treated mice. OKN-007-treated mice also significantly decreased protein expression of the human nuclear antigen (HNA) (p < 0.001), ACVR1 (p < 0.0001), and c-MET (p < 0.05), as well as significantly increased expression of cleaved caspase 3 (p < 0.001) and histone H3 K27-trimethylation (p < 0.01), compared to untreated mouse tumors. CONCLUSIONS With the dismal prognosis and limited effective chemotherapy available for DIPG, there is significant room for continued research studies, and OKN-007 merits further exploration as a therapeutic agent.
Collapse
Affiliation(s)
- Lincy Thomas
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
- The Jimmy Everest Center for Cancer and Blood Disorders in Children, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- University of Texas Southwestern in the Division of Hematology and Oncology, Dallas, TX, USA
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Michelle Zalles
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
- Pharmaceutical Department, Medical University of Lodz, Lodz, Poland
| | - Megan Lerner
- Surgery Research Laboratory, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Angel M Carcaboso
- Department of Pediatric Hematology and Oncology, Hospital Sant Juan de Deu, Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA.
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
46
|
Giussani C, Guida L, Biassoni V, Schiavello E, Carrabba G, Trezza A, Sganzerla E, Massimino M. Retrospective analysis of the clinical and radiological features of 94 consecutive DIPGs patients to investigate the factors determining the development of hydrocephalus and its impact on clinical status and survival. Childs Nerv Syst 2020; 36:2701-2705. [PMID: 32222799 DOI: 10.1007/s00381-020-04589-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/23/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE There is no consensus in the literature about the impact of hydrocephalus on clinical course and overall survival of diffuse intrinsic pontine gliomas (DIPG) patients as well as about its specific treatment. Authors reviewed a series of DIPG patients to investigate factors related to the onset of hydrocephalus, its treatment, and its impact on clinical course and prognosis. METHODS A retrospective observational study was performed enrolling pediatric patients affected by DIPG from 2008 to 2018. Clinical and radiological charts were reviewed to find patients' demographic, pathologic and radiologic features in hydrocephalic and non-hydrocephalic patients. In the hydrocephalus cohort, treatment strategy and its effectiveness and complications were analyzed. RESULTS Ninety-four pediatric patients were enrolled in the study. Patients who developed hydrocephalus showed significantly lesser maximum axial tumor areas than patients without hydrocephalus (respectively 6.5 cm2 vs 16.45 cm2, p < 0.005). Hydrocephalus developed in 33 patients (35%) with an onset interval of 5.24 ± 1.21 months (range 3.2-7.3). The majority of hydrocephalic patients (28 cases, 90%) were treated by ventriculoperitoneal shunt, the remaining 3 patients being treated by endoscopic third ventriculostomy. Mean overall survival was 16.6 months ± 20 months without significative difference between the groups. CONCLUSION The onset of hydrocephalus occurs in the first moths of the disease story and found a negative correlation with tumor maximal axial diameter. Early treatment of hydrocephalus presents a very low complications rate with satisfying clinical outcome, as it allows the patients to continue the neurooncological therapies being a part of the treatment armamentarium instead of a palliative solution.
Collapse
Affiliation(s)
- Carlo Giussani
- Neurosurgery, School of Medicine, Ospedale San Gerardo, Università degli Studi di Milano Bicocca, Monza, Italy.
| | - Lelio Guida
- Neurosurgery, School of Medicine, Ospedale San Gerardo, Università degli Studi di Milano Bicocca, Monza, Italy
| | - Veronica Biassoni
- Pediatric Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Giorgio Carrabba
- Neurosurgery, School of Medicine, Fondazione IRCCS Ca' Granda, Università degli studi di Milano, Milan, Italy
| | - Andrea Trezza
- Neurosurgery, School of Medicine, Ospedale San Gerardo, Università degli Studi di Milano Bicocca, Monza, Italy
| | - Erik Sganzerla
- Neurosurgery, School of Medicine, Ospedale San Gerardo, Università degli Studi di Milano Bicocca, Monza, Italy
| | - Maura Massimino
- Pediatric Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
47
|
Balakrishnan I, Danis E, Pierce A, Madhavan K, Wang D, Dahl N, Sanford B, Birks DK, Davidson N, Metselaar DS, Meel MH, Lemma R, Donson A, Vijmasi T, Katagi H, Sola I, Fosmire S, Alimova I, Steiner J, Gilani A, Hulleman E, Serkova NJ, Hashizume R, Hawkins C, Carcaboso AM, Gupta N, Monje M, Jabado N, Jones K, Foreman N, Green A, Vibhakar R, Venkataraman S. Senescence Induced by BMI1 Inhibition Is a Therapeutic Vulnerability in H3K27M-Mutant DIPG. Cell Rep 2020; 33:108286. [PMID: 33086074 PMCID: PMC7574900 DOI: 10.1016/j.celrep.2020.108286] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 07/05/2020] [Accepted: 09/25/2020] [Indexed: 01/19/2023] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is an incurable brain tumor of childhood characterized by histone mutations at lysine 27, which results in epigenomic dysregulation. There has been a failure to develop effective treatment for this tumor. Using a combined RNAi and chemical screen targeting epigenomic regulators, we identify the polycomb repressive complex 1 (PRC1) component BMI1 as a critical factor for DIPG tumor maintenance in vivo. BMI1 chromatin occupancy is enriched at genes associated with differentiation and tumor suppressors in DIPG cells. Inhibition of BMI1 decreases cell self-renewal and attenuates tumor growth due to induction of senescence. Prolonged BMI1 inhibition induces a senescence-associated secretory phenotype, which promotes tumor recurrence. Clearance of senescent cells using BH3 protein mimetics co-operates with BMI1 inhibition to enhance tumor cell killing in vivo.
Collapse
Affiliation(s)
- Ilango Balakrishnan
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; The Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, USA
| | - Etienne Danis
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; The Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, USA
| | - Angela Pierce
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Krishna Madhavan
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; The Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, USA
| | - Dong Wang
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Nathan Dahl
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; The Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, USA
| | - Bridget Sanford
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Diane K Birks
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Nate Davidson
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Dennis S Metselaar
- Princess Máxima Center for Pediatric Oncology, Utrecht and Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Michaël Hananja Meel
- Princess Máxima Center for Pediatric Oncology, Utrecht and Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Rakeb Lemma
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew Donson
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; The Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, USA
| | - Trinka Vijmasi
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; The Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, USA
| | - Hiroaki Katagi
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ismail Sola
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Susan Fosmire
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Irina Alimova
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Jenna Steiner
- Departments of Radiology, Radiation Oncology, and Anesthesiology, Colorado Animal Imaging Shared Resource (AISR), University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ahmed Gilani
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, Utrecht and Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Natalie J Serkova
- Departments of Radiology, Radiation Oncology, and Anesthesiology, Colorado Animal Imaging Shared Resource (AISR), University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rintaro Hashizume
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Cynthia Hawkins
- Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Angel M Carcaboso
- Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain
| | - Nalin Gupta
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Michelle Monje
- Departments of Neurology, Neurosurgery, Pediatrics, and Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nada Jabado
- Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada; Department of Pediatrics, McGill University, and The Research Institute of the McGill University Health Center, Montreal, QC H4A 3J1, Canada
| | - Kenneth Jones
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Nicholas Foreman
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; The Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, USA
| | - Adam Green
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; The Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, USA
| | - Rajeev Vibhakar
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; The Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, USA.
| | - Sujatha Venkataraman
- Department of Pediatrics and Section of Pediatric Hematology/Oncology/BMT, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; The Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, USA.
| |
Collapse
|
48
|
Wang C, Sinha S, Jiang X, Fitch S, Wilson C, Caretti V, Ponnuswami A, Monje M, Grant G, Yang F. A comparative study of brain tumor cells from different age and anatomical locations using 3D biomimetic hydrogels. Acta Biomater 2020; 116:201-208. [PMID: 32911104 DOI: 10.1016/j.actbio.2020.09.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/19/2020] [Accepted: 09/03/2020] [Indexed: 12/27/2022]
Abstract
Brain tumors exhibit vast genotypic and phenotypic diversity depending on patient age and anatomical location. Hydrogels hold great promise as 3D in vitro models for studying brain tumor biology and drug screening, yet previous studies were limited to adult glioblastoma cells, and most studies used immortalized cell lines. Here we report a hydrogel platform that supports the proliferation and invasion of patient-derived brain tumor cell cultures (PDCs) isolated from different patient age groups and anatomical locations. Hydrogel stiffness was tuned by varying poly(ethylene-glycol) concentration. Cell adhesive peptide (CGRDS), hyaluronic acid, and MMP-cleavable crosslinkers were incorporated to facilitate cell adhesion and cell-mediated degradation. Three PDC lines were compared including adult glioblastoma cells (aGBM), pediatric glioblastoma cells (pGBM), and diffuse pontine intrinsic glioma (DIPG). A commonly used immortalized adult glioblastoma cell line U87 was included as a control. PDCs displayed stiffness-dependent behavior, with 40 Pa hydrogel promoting faster tumor proliferation and invasion. Adult GBM cells exhibited faster proliferation than pediatric GBM, and DIPG showed slowest proliferation. These results suggest both patient age and tumor location affects brain tumor behaviors. Adult GBM PDCs also exhibited very different cell proliferation and morphology from U87. The hydrogel reported here can provide a useful tool for future studies to better understand how age and anatomical locations impacts brain tumor progression using 3D in vitro models.
Collapse
|
49
|
Roux A, Boddaert N, Grill J, Castel D, Zanello M, Zah-Bi G, Chrétien F, Lefevre E, Ros VD, Zerah M, Puget S, Pallud J, Varlet P. High Prevalence of Developmental Venous Anomaly in Diffuse Intrinsic Pontine Gliomas: A Pediatric Control Study. Neurosurgery 2020; 86:517-523. [PMID: 31342064 DOI: 10.1093/neuros/nyz298] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 03/18/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND No link has been demonstrated between diffuse intrinsic pontine glioma and developmental venous anomaly in pediatric patients. OBJECTIVE To determine the prevalence of developmental venous anomaly in a pediatric cohort of diffuse intrinsic pontine glioma. METHODS We performed a retrospective cohort study (1998-2017) of consecutive pediatric patients harboring a diffuse intrinsic pontine glioma (experimental set, n = 162) or a craniopharyngioma (control set, n = 142) in a tertiary pediatric neurosurgical center. The inclusion criteria were the following: age <18 yr at diagnosis; histopathological diagnosis of diffuse intrinsic pontine glioma or craniopharyngioma according to the 2016 World Health Organization classification of tumors of the central nervous system; no previous oncological treatment; and available preoperative magnetic resonance imaging performed with similar acquisition protocol. RESULTS We found a significantly higher prevalence of developmental venous anomaly in the experimental set of 162 diffuse intrinsic pontine gliomas (24.1%) than in the control set of 142 craniopharyngiomas (10.6%; P = .001). The prevalence of developmental venous anomalies was not significantly impacted by demographic data (sex, age at diagnosis, and underlying pathological condition), biomolecular analysis (H3-K27M-mutant subgroup, H3.1-K27M-mutant subgroup, and H3.3-K27M-mutant subgroup), or imaging findings (anatomic location, anatomic extension, side, and obstructive hydrocephalus) of the studied diffuse intrinsic pontine gliomas. CONCLUSION We report a higher prevalence of developmental venous anomaly in pediatric diffuse intrinsic pontine glioma patients than in control patients, which suggests a potential underlying common predisposition or a causal relationship that will require deeper investigations.
Collapse
Affiliation(s)
- Alexandre Roux
- Department of Neurosurgery, Sainte-Anne Hospital, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Inserm UMR 1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| | - Nathalie Boddaert
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Paediatric Radiology, Necker Enfants-Malades Hospital, Paris, France.,Inserm UMR 1163, Institut Imagine, Inserm U1000, Paris, France
| | - Jacques Grill
- Department of Pediatric Oncology, Gustave-Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France.,UMR8203 "Vectorologie et Thérapeutiques Anticancéreuses," CNRS, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - David Castel
- Department of Pediatric Oncology, Gustave-Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France.,UMR8203 "Vectorologie et Thérapeutiques Anticancéreuses," CNRS, Gustave Roussy, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Marc Zanello
- Department of Neurosurgery, Sainte-Anne Hospital, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Inserm UMR 1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| | - Gilles Zah-Bi
- Department of Neurosurgery, Sainte-Anne Hospital, Paris, France
| | - Fabrice Chrétien
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Neuropathology, Sainte-Anne Hospital, Paris, France
| | - Etienne Lefevre
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Volodia Dangouloff Ros
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Paediatric Radiology, Necker Enfants-Malades Hospital, Paris, France.,Inserm UMR 1163, Institut Imagine, Inserm U1000, Paris, France
| | - Michel Zerah
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Pediatric Neurosurgery, Necker Enfants-Malades Hospital, Paris, France
| | - Stéphanie Puget
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Pediatric Neurosurgery, Necker Enfants-Malades Hospital, Paris, France
| | - Johan Pallud
- Department of Neurosurgery, Sainte-Anne Hospital, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Inserm UMR 1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| | - Pascale Varlet
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Inserm UMR 1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France.,Department of Neuropathology, Sainte-Anne Hospital, Paris, France
| |
Collapse
|
50
|
DeWire M, Fuller C, Hummel TR, Chow LML, Salloum R, de Blank P, Pater L, Lawson S, Zhu X, Dexheimer P, Carle AC, Kumar SS, Drissi R, Stevenson CB, Lane A, Breneman J, Witte D, Jones BV, Leach JL, Fouladi M. A phase I/II study of ribociclib following radiation therapy in children with newly diagnosed diffuse intrinsic pontine glioma (DIPG). J Neurooncol 2020; 149:511-522. [PMID: 33034839 DOI: 10.1007/s11060-020-03641-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/01/2020] [Indexed: 11/29/2022]
Abstract
PURPOSE Cyclin-dependent kinase-retinoblastoma (CDK-RB) pathway is dysregulated in some diffuse intrinsic pontine gliomas (DIPG). We evaluated safety, feasibility, and early efficacy of the CDK4/6-inhibitor ribociclib, administered following radiotherapy in newly-diagnosed DIPG patients. METHODS Following radiotherapy, eligible patients received ribociclib in 28-day cycles (350 mg/m2; 21 days on/7 days off). Feasibility endpoints included tolerability for at least 6 courses, and a less than 2-week delay in restarting therapy after 1 dose reduction. Early efficacy was measured by 1-year and median overall survival (OS). Patient/parent-by-proxy reported outcomes measurement information system (PROMIS) assessments were completed prospectively. RESULTS The study included 10 evaluable patients, 9 DIPG and 1 diffuse midline glioma (DMG)-all 3.7 to 19.8 years of age. The median number of courses was 8 (range 3-14). Three patients required dose reduction for grade-4 neutropenia, and 1 discontinued therapy for hematological toxicity following course 4. The most common grade-3/4 toxicity was myelosuppression. After 2 courses, MRI evaluations in 4 patients revealed increased necrotic volume, associated with new neurological symptoms in 3 patients. The 1-year and median OS for DIPG was 89% and 16.1 months (range 10-30), respectively; the DMG patient died at 6 months post-diagnosis. Five patients donated brain tissue and tumor; 3 were RB+ . CONCLUSIONS Ribociclib administered following radiotherapy is feasible in DIPG and DMG. Increased tumor necrosis may represent a treatment effect. These data warrant further prospective volumetric analyses of tumors with necrosis. Feasibility and stabilization findings support further investigation of ribociclib in combination therapies. TRIAL REGISTRATION NCT02607124.
Collapse
Affiliation(s)
- Mariko DeWire
- Division of Oncology, Department of Pediatrics College of Medicine, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, MLC 7015, Cincinnati, OH, 45209, USA.
| | - Christine Fuller
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Trent R Hummel
- Division of Oncology, Department of Pediatrics College of Medicine, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, MLC 7015, Cincinnati, OH, 45209, USA
| | - Lionel M L Chow
- Department of Hematology/Oncology, Dayton Children's Hospital, Dayton, OH, USA
| | - Ralph Salloum
- Division of Oncology, Department of Pediatrics College of Medicine, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, MLC 7015, Cincinnati, OH, 45209, USA
| | - Peter de Blank
- Division of Oncology, Department of Pediatrics College of Medicine, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, MLC 7015, Cincinnati, OH, 45209, USA
| | - Luke Pater
- Department of Radiation Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sarah Lawson
- Department of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaoting Zhu
- Department of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Phil Dexheimer
- Department of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Adam C Carle
- Department of Pediatrics, Department of Psychology, College of Medicine University of Cincinnati, College of Arts and Sciences University of Cincinnati, Anderson Center Health Systems Excellence, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Shiva Senthil Kumar
- Division of Oncology, Department of Pediatrics College of Medicine, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, MLC 7015, Cincinnati, OH, 45209, USA
| | - Rachid Drissi
- Division of Oncology, Department of Pediatrics College of Medicine, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, MLC 7015, Cincinnati, OH, 45209, USA
| | - Charles B Stevenson
- Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Adam Lane
- Department of Biostatistics, Cincinnati, OH, USA
| | - John Breneman
- Department of Radiation Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - David Witte
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Blaise V Jones
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - James L Leach
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Maryam Fouladi
- Division of Oncology, Department of Pediatrics College of Medicine, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, MLC 7015, Cincinnati, OH, 45209, USA
| |
Collapse
|