1
|
Xiao W, Li S, Tong Z, Li L, Zhang Y. The Value of Diffusion Tensor Imaging in Differential Diagnosis of Embryonal Tumors Occurring in the Brainstem and Brainstem Gliomas in Pediatric Patients. Pediatr Neurol 2025; 163:58-65. [PMID: 39689574 DOI: 10.1016/j.pediatrneurol.2024.11.011] [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: 09/08/2023] [Revised: 11/03/2024] [Accepted: 11/24/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND There are no apparent distinctions in clinical presentation or conventional imaging findings between brainstem gliomas and embryonal tumors occurring in the brainstem. Our aim was to study the role of diffusion tensor imaging in differentiating embryonal tumors from gliomas of the brainstem. METHODS Three cases of embryonal tumors occurring in the brainstem and 19 cases of brainstem gliomas were analyzed retrospectively. RESULT The most common brainstem gliomas are diffuse intrinsic pontine gliomas. On the fiber tracking images, brainstem gliomas were associated with relatively intact projection fibers that continuously traversed the tumor and followed the trajectory of normal neural fibers, whereas embryonal tumors were associated with disruption of projection fibers. The close cellularity created tissues with significant directional properties in embryonal tumors, restricting the diffusion of water molecules. As a result, there were areas of high anisotropy within the embryonal tumors. Additionally, we observed that the apparent diffusion coefficient value of embryonal tumors occurring in the brainstem was lower than that of brainstem gliomas and the difference was statistically significant (P < 0.05). CONCLUSION Disruption of projection fibers within the tumor on diffusion tensor imaging may help differentiate embryonal pathology from glial.
Collapse
Affiliation(s)
- Wenjiao Xiao
- Department of Radiology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Shuang Li
- Department of Radiology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zanyong Tong
- Department of Radiology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lusheng Li
- Department of Neurosurgery Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China.
| | - Yuting Zhang
- Department of Radiology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China.
| |
Collapse
|
2
|
Boukaka RG, Beuriat PA, Di Rocco F, Vasiljevic A, Szathmari A, Mottolese C. Brainstem tumors in children: a monocentric series in the light of genetic and bio-molecular progress in pediatric neuro-oncology. Front Pediatr 2023; 11:1193474. [PMID: 37936887 PMCID: PMC10626527 DOI: 10.3389/fped.2023.1193474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/10/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Brainstem tumors represent a challenge. Their management and prognosis vary according to anatomopathological findings and genetic and bio-molecular fingerprints. We present our experience with pediatric brainstem tumors. Material and methods All patients admitted for a brainstem tumor at the Pediatric Neurosurgical Unit at Hôpital Femme Mère Enfant hospital between January 1997 and December 2019 were considered. Patients data were obtained through a retrospective review of the medical records; follow-up was from the last outpatient consultation. Results One hundred and twelve patients were included. Eighty-five patients (75.9%) had open surgery or stereotactic biopsy. Thirty-five patients were treated for hydrocephalus. Sixty-six received an adjuvant treatment. Several protocols were adopted according to the SFOP and SIOP during this time period. The overall survival rate was 45% with a median follow-up of five years (range 1-18 year). However, the survival rate was very different between the diffuse intrinsic pontine gliomas (DIPG) and the others tumor types. If we exclude the DIPG (59 patients), of which only 1 was alive at 3 years, the survival rate was 90.6% (only 5 deaths over 53 patients) with a median follow up of 5 years. Conclusions Our series confirms that benign tumors of the brainstem have a good survival when treated with surgical removal ± adjuvant therapy. Diffuse pontine gliomas continue to have a dismal prognosis. Individualized treatment based on molecular fingerprints may help to select the best adjuvant therapy and hence potentially improve survival.
Collapse
Affiliation(s)
- Rel Gerald Boukaka
- Department of Pediatric Neurosurgery, Hôpital Femme Mère Enfant, Hospices Civils de, Lyon, France
| | - Pierre-Aurélien Beuriat
- Department of Pediatric Neurosurgery, Hôpital Femme Mère Enfant, Hospices Civils de, Lyon, France
- Université Claude Bernard, Lyon 1, Lyon, France
| | - Federico Di Rocco
- Department of Pediatric Neurosurgery, Hôpital Femme Mère Enfant, Hospices Civils de, Lyon, France
- Université Claude Bernard, Lyon 1, Lyon, France
| | - Alexandre Vasiljevic
- Department of Pathology and Neuropathology, Groupement Hospitalier Est, Hospices Civils de Lyon, Lyon, France
| | - Alexandru Szathmari
- Department of Pediatric Neurosurgery, Hôpital Femme Mère Enfant, Hospices Civils de, Lyon, France
| | - Carmine Mottolese
- Department of Pediatric Neurosurgery, Hôpital Femme Mère Enfant, Hospices Civils de, Lyon, France
| |
Collapse
|
3
|
Foss A, Pathania M. Pediatric Glioma Models Provide Insights into Tumor Development and Future Therapeutic Strategies. Dev Neurosci 2023; 46:22-43. [PMID: 37231843 DOI: 10.1159/000531040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
In depth study of pediatric gliomas has been hampered due to difficulties in accessing patient tissue and a lack of clinically representative tumor models. Over the last decade, however, profiling of carefully curated cohorts of pediatric tumors has identified genetic drivers that molecularly segregate pediatric gliomas from adult gliomas. This information has inspired the development of a new set of powerful in vitro and in vivo tumor models that can aid in identifying pediatric-specific oncogenic mechanisms and tumor microenvironment interactions. Single-cell analyses of both human tumors and these newly developed models have revealed that pediatric gliomas arise from spatiotemporally discrete neural progenitor populations in which developmental programs have become dysregulated. Pediatric high-grade gliomas also harbor distinct sets of co-segregating genetic and epigenetic alterations, often accompanied by unique features within the tumor microenvironment. The development of these novel tools and data resources has led to insights into the biology and heterogeneity of these tumors, including identification of distinctive sets of driver mutations, developmentally restricted cells of origin, recognizable patterns of tumor progression, characteristic immune environments, and tumor hijacking of normal microenvironmental and neural programs. As concerted efforts have broadened our understanding of these tumors, new therapeutic vulnerabilities have been identified, and for the first time, promising new strategies are being evaluated in the preclinical and clinical settings. Even so, dedicated and sustained collaborative efforts are necessary to refine our knowledge and bring these new strategies into general clinical use. In this review, we will discuss the range of currently available glioma models, the way in which they have each contributed to recent developments in the field, their benefits and drawbacks for addressing specific research questions, and their future utility in advancing biological understanding and treatment of pediatric glioma.
Collapse
Affiliation(s)
- Amelia Foss
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Manav Pathania
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
| |
Collapse
|
4
|
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
|
5
|
Rechberger JS, Power BT, Power EA, Nesvick CL, Daniels DJ. H3K27-altered diffuse midline glioma: a paradigm shifting opportunity in direct delivery of targeted therapeutics. Expert Opin Ther Targets 2023; 27:9-17. [PMID: 36744399 PMCID: PMC10165636 DOI: 10.1080/14728222.2023.2177531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Despite much progress, the prognosis for H3K27-altered diffuse midline glioma (DMG), previously known as diffuse intrinsic pontine glioma when located in the brainstem, remains dark and dismal. AREAS COVERED A wealth of research over the past decade has revolutionized our understanding of the molecular basis of DMG, revealing potential targetable vulnerabilities for treatment of this lethal childhood cancer. However, obstacles to successful clinical implementation of novel therapies remain, including effective delivery across the blood-brain barrier (BBB) to the tumor site. Here, we review relevant literature and clinical trials and discuss direct drug delivery via convection-enhanced delivery (CED) as a promising treatment modality for DMG. We outline a comprehensive molecular, pharmacological, and procedural approach that may offer hope for afflicted patients and their families. EXPERT OPINION Challenges remain in successful drug delivery to DMG. While CED and other techniques offer a chance to bypass the BBB, the variables influencing successful intratumoral targeting are numerous and complex. We discuss these variables and potential solutions that could lead to the successful clinical implementation of preclinically promising therapeutic agents.
Collapse
Affiliation(s)
- Julian S Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Blake T Power
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Erica A Power
- Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Cody L Nesvick
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| |
Collapse
|
6
|
Frameless Robotic-Assisted Biopsy of Pediatric Brainstem Lesions: A Systematic Review and Meta-Analysis of Efficacy and Safety. World Neurosurg 2023; 169:87-93.e1. [PMID: 36307039 DOI: 10.1016/j.wneu.2022.10.071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Pediatric brainstem lesions are diagnoses that require tissue sampling to advance our understanding of them and their management. Frameless, robot-assisted biopsy of these lesions has emerged as a novel, viable biopsy approach. Correspondingly, the aim of this study was to quantitively and qualitatively summarize the contemporary literature regarding the likelihood of achieving tumor diagnosis and experiencing any postoperative complications. METHODS Searches of 7 electronic databases from inception to September 2022 were conducted following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Articles were screened against prespecified criteria. Outcomes were pooled by random-effects meta-analyses of proportions where possible. RESULTS A total of 8 cohort studies satisfied all criteria. They described 99 pediatric patients with brainstem lesions in whom frameless, robot-assisted biopsy was involved in their work-up. There were 62 (63%) male and 37 (37%) female patients with a median age of 9 years at time of biopsy. Overall, all patients had sufficient tissue obtained by initial biopsy for evaluation. Pooled estimate of achieving tumor diagnosis was 100% (95% confidence interval [CI] 97%-100%) across all studies with a high degree of certainty. Across all studies, there were no cases of procedure-related mortality. The pooled estimates of transient and permanent complications after biopsy were 10% (95% CI 4%-19%) and 0% (95% CI 0%-2%), respectively, of very low and low degrees of certainty each. CONCLUSIONS The contemporary metadata demonstrates the frameless, robot-assisted biopsy of pediatric brainstem lesions is both effective and safe when performed in an experienced setting. Further research is needed to augment robot and automated technologies into workup algorithms.
Collapse
|
7
|
Immunogenic Cell Death Enhances Immunotherapy of Diffuse Intrinsic Pontine Glioma: From Preclinical to Clinical Studies. Pharmaceutics 2022; 14:pharmaceutics14091762. [PMID: 36145510 PMCID: PMC9502387 DOI: 10.3390/pharmaceutics14091762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/02/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is the most lethal tumor involving the pediatric central nervous system. The median survival of children that are diagnosed with DIPG is only 9 to 11 months. More than 200 clinical trials have failed to increase the survival outcomes using conventional cytotoxic or myeloablative chemotherapy. Immunotherapy presents exciting therapeutic opportunities against DIPG that is characterized by unique and heterogeneous features. However, the non-inflammatory DIPG microenvironment greatly limits the role of immunotherapy in DIPG. Encouragingly, the induction of immunogenic cell death, accompanied by the release of damage-associated molecular patterns (DAMPs) shows satisfactory efficacy of immune stimulation and antitumor strategies. This review dwells on the dilemma and advances in immunotherapy for DIPG, and the potential efficacy of immunogenic cell death (ICD) in the immunotherapy of DIPG.
Collapse
|
8
|
Dubois FPB, Shapira O, Greenwald NF, Zack T, Wala J, Tsai JW, Crane A, Baguette A, Hadjadj D, Harutyunyan AS, Kumar KH, Blattner-Johnson M, Vogelzang J, Sousa C, Kang KS, Sinai C, Wang DK, Khadka P, Lewis K, Nguyen L, Malkin H, Ho P, O'Rourke R, Zhang S, Gold R, Deng D, Serrano J, Snuderl M, Jones C, Wright KD, Chi SN, Grill J, Kleinman CL, Goumnerova LC, Jabado N, Jones DTW, Kieran MW, Ligon KL, Beroukhim R, Bandopadhayay P. Structural variants shape driver combinations and outcomes in pediatric high-grade glioma. NATURE CANCER 2022; 3:994-1011. [PMID: 35788723 PMCID: PMC10365847 DOI: 10.1038/s43018-022-00403-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 05/23/2022] [Indexed: 12/13/2022]
Abstract
We analyzed the contributions of structural variants (SVs) to gliomagenesis across 179 pediatric high-grade gliomas (pHGGs). The most recurrent SVs targeted MYC isoforms and receptor tyrosine kinases (RTKs), including an SV amplifying a MYC enhancer in 12% of diffuse midline gliomas (DMG), indicating an underappreciated role for MYC in pHGG. SV signature analysis revealed that tumors with simple signatures were TP53 wild type (TP53WT) but showed alterations in TP53 pathway members PPM1D and MDM4. Complex signatures were associated with direct aberrations in TP53, CDKN2A and RB1 early in tumor evolution and with later-occurring extrachromosomal amplicons. All pHGGs exhibited at least one simple-SV signature, but complex-SV signatures were primarily restricted to subsets of H3.3K27M DMGs and hemispheric pHGGs. Importantly, DMGs with complex-SV signatures were associated with shorter overall survival independent of histone mutation and TP53 status. These data provide insight into the impact of SVs on gliomagenesis and the mechanisms that shape them.
Collapse
Affiliation(s)
- Frank P B Dubois
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ofer Shapira
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Noah F Greenwald
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Travis Zack
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jeremiah Wala
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jessica W Tsai
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Alexander Crane
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Audrey Baguette
- Quantitative Life Sciences, McGill University, Montreal, QC, Canada
| | - Djihad Hadjadj
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | | | - Kiran H Kumar
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mirjam Blattner-Johnson
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jayne Vogelzang
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Cecilia Sousa
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kyung Shin Kang
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Claire Sinai
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Dayle K Wang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Prasidda Khadka
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Lan Nguyen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hayley Malkin
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Patricia Ho
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ryan O'Rourke
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shu Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rose Gold
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Davy Deng
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Chris Jones
- Division of Cancer Therapeutics and Department of Molecular Pathology, Institute of Cancer Research 15 Cotswold Road, Sutton, London, UK
| | - Karen D Wright
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Susan N Chi
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Jacques Grill
- Department of Pediatric and Adolescent Oncology and INSERM Unit 981, Gustave Roussy Institute and University of Paris Saclay, Villejuif, France
| | - Claudia L Kleinman
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Lady Davis Research Institute, Jewish General Hospital, Montreal, QC, Canada
| | - Liliana C Goumnerova
- Department of Neurosurgery, Boston Children's Hospital; Dana Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- LCG: Tromboprotea, MWK: Day One Biopharmaceuticals, San Francisco, CA, USA
| | - Nada Jabado
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Division of Experimental Medicine, Department of Medicine and Department of Pediatrics, McGill University, and The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mark W Kieran
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- LCG: Tromboprotea, MWK: Day One Biopharmaceuticals, San Francisco, CA, USA
| | - Keith L Ligon
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham & Women's Hospital and Boston Children's Hospital, Boston, USA.
- Center for Patient Derived Models, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Rameen Beroukhim
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
| | - Pratiti Bandopadhayay
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA.
| |
Collapse
|
9
|
Lutz K, Jünger ST, Messing-Jünger M. Essential Management of Pediatric Brain Tumors. CHILDREN 2022; 9:children9040498. [PMID: 35455542 PMCID: PMC9031600 DOI: 10.3390/children9040498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 02/02/2023]
Abstract
Brain tumors are the most common solid tumors in children and are associated with high mortality. The most common childhood brain tumors are grouped as low-grade gliomas (LGG), high grade gliomas (HGG), ependymomas, and embryonal tumors, according to the World Health Organization (WHO). Advances in molecular genetics have led to a shift from pure histopathological diagnosis to integrated diagnosis. For the first time, these new criteria were included in the WHO classification published in 2016 and has been further updated in the 2021 edition. Integrated diagnosis is based on molecular genomic similarities of the tumor subclasses, and it can better explain the differences in clinical courses of previously histopathologically identical entities. Important advances have also been made in pediatric neuro-oncology. A growing understanding of the molecular-genetic background of tumorigenesis has improved the diagnostic accuracy. Re-stratification of treatment protocols and the development of targeted therapies will significantly affect overall survival and quality of life. For some pediatric tumors, these advances have significantly improved therapeutic management and prognosis in certain tumor subgroups. Some therapeutic approaches also have serious long-term consequences. Therefore, optimized treatments are greatly needed. Here, we discuss the importance of multidisciplinary collaboration and the role of (pediatric) neurosurgery by briefly describing the most common childhood brain tumors and their currently recognized molecular subgroups.
Collapse
Affiliation(s)
- Katharina Lutz
- Neurosurgery Department, Inselspital, 3010 Bern, Switzerland
- Pediatric Neurosurgery, Asklepios Children’s Hospital, 53757 Sankt Augustin, Germany;
- Correspondence:
| | - Stephanie T. Jünger
- Center for Neurosurgery, Department of General Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
| | | |
Collapse
|
10
|
Abstract
Pediatric midline gliomas are a uniformly fatal disease for which there is no cure. The location of these tumors makes surgical resection impossible, and so novel therapies are urgently needed to improve outcomes. The biology of these tumors is increasingly understood, with the histone H3K27M mutation playing a critical role in the pathogenesis of these tumors. Efforts to inhibit the growth of these tumors have also focused on inhibiting the Aurora kinase and Janus-associated kinase (JAK)/signal transducer and activator of transcription (STAT) pathway in order to disrupt tumor proliferation. A number of small molecule inhibitors of these kinases have shown promise in early studies. Screening and preclinical assessment of such inhibitors requires a functional assay to assess the degree of kinase inhibition. We detail here a luciferase-based reporter assay for STAT3 transcriptional activity that we have employed frequently in order to assess the efficacy of kinase inhibitors in pediatric gliomas. The assay we describe is specific to STAT3, but the overall methodology is generalizable to other downstream targets of the kinase of interest.
Collapse
Affiliation(s)
- Benjamin T Himes
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Liang Zhang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
11
|
Infratentorial Stereotactic Biopsy of Brainstem and Cerebellar Lesions. Brain Sci 2021; 11:brainsci11111432. [PMID: 34827431 PMCID: PMC8615913 DOI: 10.3390/brainsci11111432] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/15/2022] Open
Abstract
Stereotactic biopsy of posterior fossa lesions is often regarded as hazardous due to the critical structures in that area. Therefore, the aim of the study was to evaluate the diagnostic accuracy and safety of infratentorial stereotactic biopsy of brainstem or cerebellar lesions and its associations with other clinical, laboratory, and radiological parameters. From January 2000 to May 2021, 190 infratentorial stereotactic biopsies of posterior fossa tumors, including 108 biopsies of brainstem lesions, were performed. Moreover, 63 supratentorial biopsies of cerebral peduncle lesions were analyzed to compare the safety and efficacy of both approaches. Additionally, the presence of antibodies against Toxoplasma gondii and Epstein–Barr Virus (EBV) were documented in 67 and 66 patients, respectively, and magnetic resonance imaging (MRI) scans were evaluated in 114 patients. Only 4% of patients had minor complications and 1.5% had major complications, including one patient who died from intracranial bleeding. Nine (4.7%) biopsies were non-diagnostic. Isocitrate dehydrogenase 1 (IDH1) mutation, 1p/19q codeletion, and O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status were assessed in 29 patients, and were non-diagnostic in only 3 (10.3%) cases. Patients with high-grade gliomas (HGG) were more frequently seropositive for T. gondii than individuals with low-grade gliomas (LGG; p < 0.001). A total of 27% of HGG and 41% of LGG were non-enhancing on MRI. The infratentorial approach is generally safe and reliable for biopsy of brainstem and cerebellar lesions. In our study, the safety and efficacy of supratentorial biopsy of the cerebral peduncle and infratentorial biopsy of lesions below the cerebral peduncle were comparably high. Moreover, patients with HGG were more frequently seropositive for T. gondii than patients with LGG, and the relationship between toxoplasmosis and gliomagenesis requires further investigation.
Collapse
|
12
|
Patil N, Kelly ME, Yeboa DN, Buerki RA, Cioffi G, Balaji S, Ostrom QT, Kruchko C, Barnholtz-Sloan JS. Epidemiology of brainstem high-grade gliomas in children and adolescents in the United States, 2000-2017. Neuro Oncol 2021; 23:990-998. [PMID: 33346835 DOI: 10.1093/neuonc/noaa295] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Limited population-based data exist for the brainstem gliomas for children ages ≤19 years, which includes high-grade aggressively growing tumors such as diffuse intrinsic pontine glioma (DIPG). We examined the overall incidence and survival patterns in children with brainstem high-grade glioma (HGG) by age, sex, and race and ethnicity. METHODS We used data from Central Brain Tumor Registry of the United States (CBTRUS), obtained through data use agreements with the Centers for Disease Control (CDC) and the National Cancer Institute (NCI) from 2000 to 2017, and survival data from the CDCs National Program of Cancer Registries (NPCR), from 2001 to 2016 for malignant brainstem HGG for ages ≤19 years (per WHO ICD-O-3 codes). HGG was determined by established histologic and/or imaging criteria. Age-adjusted incidence rates and survival data were used to assess differences overall and by age, sex race, and ethnicity. RESULTS The incidence of brainstem HGG was higher among the female and Non-Hispanic population. Majority (69.8%) of these tumors were diagnosed radiographically. Incidence was higher in children aged 1-9 years compared to older children. Whites had a higher incidence compared to Blacks. However, the risk of death was higher among Blacks and Other race compared to Whites. There was no difference in survival by sex. CONCLUSIONS We report the most comprehensive incidence and survival data on these lethal brainstem HGGs. Incidence and survival among patients with brainstem HGGs differed significantly by race, ethnicity, age-groups, and grade.
Collapse
Affiliation(s)
- Nirav Patil
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois
| | - Michael E Kelly
- Department of Pediatrics, Northeast Ohio Medical University, Rootstown, Ohio
| | - Debra Nana Yeboa
- Department of Radiation Oncology at University of Texas, MD Anderson Cancer Center
| | - Robin A Buerki
- Department of Neurology, University Hospitals, and Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Gino Cioffi
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois.,The Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | | | - Quinn T Ostrom
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois.,Department of Medicine, Section of Epidemiology and Population Sciences, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Carol Kruchko
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois
| | - Jill S Barnholtz-Sloan
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois.,The Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Cleveland Center for Health Outcomes Research (CCHOR) Clevleand, Ohio
| |
Collapse
|
13
|
Use of intra-operative stimulation of brainstem lesion target sites for frameless stereotactic biopsies. Childs Nerv Syst 2021; 37:1515-1523. [PMID: 33683422 DOI: 10.1007/s00381-021-05101-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/24/2021] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Frameless stereotactic navigation is used to direct the trajectory and biopsy site of target lesions. We report on a novel intra-operative stimulating (IOS) probe that is integrated into a commercially available stereotactic biopsy needle with the rationale that stimulation of the intended biopsy site should predict functional tissue thus preventing inadvertent biopsy of eloquent tissue. METHODS Patients undergoing brainstem biopsies for atypical lesions were offered the additional stimulation procedure. The IOS probe was used to deliver stimulation in an attempt to determine the proximity of eloquent tissue. Once the desired location of the biopsy needle was achieved, the IOS probe was inserted down the centre of the biopsy needle and the stimulus applied. If no action potential was recorded, biopsies from four quadrants of the lesion were taken. If however a compound action potential was recorded, a new target was selected. RESULTS Nine patients had the biopsy and stimulation procedure performed. The median age was 36 months. A minimum of 8 samples were obtained from each patient. Biopsy material was adequate to obtain a diagnosis in all 9 patients. In 2 cases use of the device influenced the insertion trajectory or biopsy site. No patients experienced any complications directly attributable to either the biopsy procedure or application of the stimulation. CONCLUSIONS Use of the IOS probe for intra-operative stimulation of the intended brainstem biopsy site was found to be safe and feasible. The addition of stimulation using the IOS probe can be done with minimal change in workflow.
Collapse
|
14
|
Chen Z, Peng P, Zhang X, Mania-Farnell B, Xi G, Wan F. Advanced Pediatric Diffuse Pontine Glioma Murine Models Pave the Way towards Precision Medicine. Cancers (Basel) 2021; 13:cancers13051114. [PMID: 33807733 PMCID: PMC7961799 DOI: 10.3390/cancers13051114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) account for ~15% of pediatric brain tumors, which invariably present with poor survival regardless of treatment mode. Several seminal studies have revealed that 80% of DIPGs harbor H3K27M mutation coded by HIST1H3B, HIST1H3C and H3F3A genes. The H3K27M mutation has broad effects on gene expression and is considered a tumor driver. Determination of the effects of H3K27M on posttranslational histone modifications and gene regulations in DIPG is critical for identifying effective therapeutic targets. Advanced animal models play critical roles in translating these cutting-edge findings into clinical trial development. Here, we review current molecular research progress associated with DIPG. We also summarize DIPG animal models, highlighting novel genomic engineered mouse models (GEMMs) and innovative humanized DIPG mouse models. These models will pave the way towards personalized precision medicine for the treatment of DIPGs.
Collapse
Affiliation(s)
- Zirong Chen
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
| | - Peng Peng
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
| | - Xiaolin Zhang
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
| | - Barbara Mania-Farnell
- Department of Biological Science, Purdue University Northwest, Hammond, IN 46323, USA;
| | - Guifa Xi
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Correspondence: (G.X.); (F.W.); Tel.: +1-(312)5034296 (G.X.); +86-(027)-8366-5201 (F.W.)
| | - Feng Wan
- Department of Neurological Surgery, Tongji Hospital, Tongji Medical College, Huazhong University Science and Technology, Wuhan 430030, China; (Z.C.); (P.P.); (X.Z.)
- Correspondence: (G.X.); (F.W.); Tel.: +1-(312)5034296 (G.X.); +86-(027)-8366-5201 (F.W.)
| |
Collapse
|
15
|
Srikanthan D, Taccone MS, Van Ommeren R, Ishida J, Krumholtz SL, Rutka JT. Diffuse intrinsic pontine glioma: current insights and future directions. Chin Neurosurg J 2021; 7:6. [PMID: 33423692 PMCID: PMC7798267 DOI: 10.1186/s41016-020-00218-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a lethal pediatric brain tumor and the leading cause of brain tumor–related death in children. As several clinical trials over the past few decades have led to no significant improvements in outcome, the current standard of care remains fractionated focal radiation. Due to the recent increase in stereotactic biopsies, tumor tissue availabilities have enabled our advancement of the genomic and molecular characterization of this lethal cancer. Several groups have identified key histone gene mutations, genetic drivers, and methylation changes in DIPG, providing us with new insights into DIPG tumorigenesis. Subsequently, there has been increased development of in vitro and in vivo models of DIPG which have the capacity to unveil novel therapies and strategies for drug delivery. This review outlines the clinical characteristics, genetic landscape, models, and current treatments and hopes to shed light on novel therapeutic avenues and challenges that remain.
Collapse
Affiliation(s)
- Dilakshan Srikanthan
- Cell Biology Program, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Michael S Taccone
- Cell Biology Program, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Division of Neurosurgery, Department of Surgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Randy Van Ommeren
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Developmental and Stem Cell Biology Program, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada
| | - Joji Ishida
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada
| | - Stacey L Krumholtz
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada
| | - James T Rutka
- Cell Biology Program, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada. .,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada. .,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada. .,Division of Neurosurgery, Department of Surgery, The Hospital for Sick Children, Suite 1503, 555, University Avenue, Toronto, ON, M5G 1X8, Canada.
| |
Collapse
|
16
|
Bruzek AK, Ravi K, Muruganand A, Wadden J, Babila CM, Cantor E, Tunkle L, Wierzbicki K, Stallard S, Dickson RP, Wolfe I, Mody R, Schwartz J, Franson A, Robertson PL, Muraszko KM, Maher CO, Garton HJL, Qin T, Koschmann C. Electronic DNA Analysis of CSF Cell-free Tumor DNA to Quantify Multi-gene Molecular Response in Pediatric High-grade Glioma. Clin Cancer Res 2020; 26:6266-6276. [PMID: 33087334 DOI: 10.1158/1078-0432.ccr-20-2066] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/07/2020] [Accepted: 09/16/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Pediatric high-grade glioma (pHGG) diagnosis portends poor prognosis and therapeutic monitoring remains difficult. Tumors release cell-free tumor DNA (cf-tDNA) into cerebrospinal fluid (CSF), allowing for potential detection of tumor-associated mutations by CSF sampling. We hypothesized that direct, electronic analysis of cf-tDNA with a handheld platform (Oxford Nanopore MinION) could quantify patient-specific CSF cf-tDNA variant allele fraction (VAF) with improved speed and limit of detection compared with established methods. EXPERIMENTAL DESIGN We performed ultra-short fragment (100-200 bp) PCR amplification of cf-tDNA for clinically actionable alterations in CSF and tumor samples from patients with pHGG (n = 12) alongside nontumor CSF (n = 6). PCR products underwent rapid amplicon-based sequencing by Oxford Nanopore Technology (Nanopore) with quantification of VAF. Additional comparison to next-generation sequencing (NGS) and droplet digital PCR (ddPCR) was performed. RESULTS Nanopore demonstrated 85% sensitivity and 100% specificity in CSF samples (n = 127 replicates) with 0.1 femtomole DNA limit of detection and 12-hour results, all of which compared favorably with NGS. Multiplexed analysis provided concurrent analysis of H3.3A (H3F3A) and H3C2 (HIST1H3B) mutations in a nonbiopsied patient and results were confirmed by ddPCR. Serial CSF cf-tDNA sequencing by Nanopore demonstrated correlation of radiological response on a clinical trial, with one patient showing dramatic multi-gene molecular response that predicted long-term clinical response. CONCLUSIONS Nanopore sequencing of ultra-short pHGG CSF cf-tDNA fragments is feasible, efficient, and sensitive with low-input samples thus overcoming many of the barriers restricting wider use of CSF cf-tDNA diagnosis and monitoring in this patient population.
Collapse
Affiliation(s)
- Amy K Bruzek
- Department of Neurosurgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Karthik Ravi
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Ashwath Muruganand
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jack Wadden
- Department of Computer Engineering, University of Michigan, Ann Arbor, Michigan
| | - Clarissa May Babila
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Evan Cantor
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Leo Tunkle
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Kyle Wierzbicki
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Stefanie Stallard
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Robert P Dickson
- Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Ian Wolfe
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Rajen Mody
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | | | - Andrea Franson
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Patricia L Robertson
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Karin M Muraszko
- Department of Neurosurgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Cormac O Maher
- Department of Neurosurgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Hugh J L Garton
- Department of Neurosurgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Tingtin Qin
- Department of Computational Medicine and Bioinformatics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Carl Koschmann
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
17
|
Sun T, Xu Y, Pan C, Liu Y, Tian Y, Li C, Di F, Zhang L. Surgical treatment and prognosis of focal brainstem gliomas in children: A 7 year single center experience. Medicine (Baltimore) 2020; 99:e22029. [PMID: 32899058 PMCID: PMC7478697 DOI: 10.1097/md.0000000000022029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
This study aims to describe the role of open surgical treatment for focal brainstem gliomas (FBSGs) with the assistance of multimodal neuronavigation and intraoperative neurophysiological monitoring (IOM) in children to investigate the efficacy of microsurgical treatment in pediatric FBSGs. Also the prognostic factors related to the overall survival (OS) of FBSGs to describe the patient and tumor characteristics relevant to prognosis/outcome were focused on. Clinical data of 63 pediatric patients below 16 years of age with FBSGs admitted to the Neurosurgical Unit of Beijing Tiantan Hospital from January 2012 to December 2018 were retrospectively analyzed. All patients underwent initial surgical treatment, followed by magnetic resonance diffusion tensor imaging (DTI), neuronavigation and IOM. Gross or near total resection (GTR or NTR) was achieved in 57/63 (90.5%) cases, and subtotal resection (STR) was achieved in 6/63 (9.5%) cases. Postoperative adjuvant therapy was received by 27/63 (42.9%) cases. Postoperative pathological examination revealed that 36/63 (57.1%) cases had grade I gliomas, 22/63 (34.9%) had grade II, and 5/63 (8.0%) had grade III-IV gliomas according to the WHO classification. The mean Karnofsky score preoperatively was 60, and at the time of follow-up was 90. Consecutively, 6 cases demonstrated disease progression, and 5 of these were deceased. The OS in all patients was 81.2% at 5 years. Histological grade (P < .001) and age at diagnosis (P = .023) showed significant association with prolonged OS. Multimodal neuronavigation and IOM allow very precise intracranial surgery, contributing to a maximally safe resection that might decrease the postoperative disability and mortality rate. This study also showed that pediatric FBSGs were mostly low-grade tumors with excellent surgical outcomes. Consequently, it is suggested that microsurgery can be used to treat FBSGs in children in order to provide better prognosis and survival outcomes.
Collapse
Affiliation(s)
- Tao Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050
| | - Yan Xu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050
| | - Changcun Pan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050
| | - Yuhan Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050
| | - Yongji Tian
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050
| | - Chunde Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050
| | - Fei Di
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050
- Neuroscience Center, Zhangjiakou First Hospital, Zhangjiakou 075000, Hebei Province, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050
| |
Collapse
|
18
|
Smil D, Wong JF, Williams EP, Adamson RJ, Howarth A, McLeod DA, Mamai A, Kim S, Wilson BJ, Kiyota T, Aman A, Owen J, Poda G, Horiuchi KY, Kuznetsova E, Ma H, Hamblin JN, Cramp S, Roberts OG, Edwards AM, Uehling D, Al-Awar R, Bullock AN, O'Meara JA, Isaac MB. Leveraging an Open Science Drug Discovery Model to Develop CNS-Penetrant ALK2 Inhibitors for the Treatment of Diffuse Intrinsic Pontine Glioma. J Med Chem 2020; 63:10061-10085. [PMID: 32787083 DOI: 10.1021/acs.jmedchem.0c01199] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
There are currently no effective chemotherapeutic drugs approved for the treatment of diffuse intrinsic pontine glioma (DIPG), an aggressive pediatric cancer resident in the pons region of the brainstem. Radiation therapy is beneficial but not curative, with the condition being uniformly fatal. Analysis of the genomic landscape surrounding DIPG has revealed that activin receptor-like kinase-2 (ALK2) constitutes a potential target for therapeutic intervention given its dysregulation in the disease. We adopted an open science approach to develop a series of potent, selective, orally bioavailable, and brain-penetrant ALK2 inhibitors based on the lead compound LDN-214117. Modest structural changes to the C-3, C-4, and C-5 position substituents of the core pyridine ring afforded compounds M4K2009, M4K2117, and M4K2163, each with a superior potency, selectivity, and/or blood-brain barrier (BBB) penetration profile. Robust in vivo pharmacokinetic (PK) properties and tolerability mark these inhibitors as advanced preclinical compounds suitable for further development and evaluation in orthotopic models of DIPG.
Collapse
Affiliation(s)
- David Smil
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada
| | - Jong Fu Wong
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Eleanor P Williams
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Roslin J Adamson
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Alison Howarth
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - David A McLeod
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada
| | - Ahmed Mamai
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada
| | - Soyoung Kim
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada
| | - Brian J Wilson
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada
| | - Taira Kiyota
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada
| | - Ahmed Aman
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada.,Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Julie Owen
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada
| | - Gennady Poda
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada.,Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Kurumi Y Horiuchi
- Reaction Biology Corp., Suite 2, 1 Great Valley Parkway, Malvern, Pennsylvania 19355, United States
| | - Ekaterina Kuznetsova
- Reaction Biology Corp., Suite 2, 1 Great Valley Parkway, Malvern, Pennsylvania 19355, United States
| | - Haiching Ma
- Reaction Biology Corp., Suite 2, 1 Great Valley Parkway, Malvern, Pennsylvania 19355, United States
| | - J Nicole Hamblin
- Charles River Discovery, Chesterford Research Park, Saffron Waldon, Essex CB10 1XL, United Kingdom
| | - Sue Cramp
- Charles River Discovery, 8-9 Spire Green Centre, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Owen G Roberts
- M4K Pharma, 101 College Street, MaRS Centre, South Tower, Toronto, Ontario M5G 1L7, Canada
| | - Aled M Edwards
- M4K Pharma, 101 College Street, MaRS Centre, South Tower, Toronto, Ontario M5G 1L7, Canada.,Structural Genomics Consortium, University of Toronto, 101 College Street, MaRS Centre, South Tower, Toronto, Ontario M5G 1L7, Canada
| | - David Uehling
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada
| | - Rima Al-Awar
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Medical Sciences Building, Room 4207, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Alex N Bullock
- Structural Genomics Consortium, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Jeff A O'Meara
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada.,M4K Pharma, 101 College Street, MaRS Centre, South Tower, Toronto, Ontario M5G 1L7, Canada
| | - Methvin B Isaac
- Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, MaRS Centre, West Tower, Toronto, Ontario M5G 0A3, Canada
| |
Collapse
|
19
|
Luly KM, Choi J, Rui Y, Green JJ, Jackson EM. Safety considerations for nanoparticle gene delivery in pediatric brain tumors. Nanomedicine (Lond) 2020; 15:1805-1815. [PMID: 32698671 PMCID: PMC7441302 DOI: 10.2217/nnm-2020-0110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/01/2020] [Indexed: 12/26/2022] Open
Abstract
Current standard of care for many CNS tumors involves surgical resection followed by chemotherapy and/or radiation. Some pediatric brain tumor types are infiltrative and diffuse in nature, which reduces the role for surgery. Furthermore, children are extremely vulnerable to neurological sequelae from surgery and radiation therapy, thus alternative approaches are in critical need. As molecular targets underlying various cancers become more clearly defined, there is an increasing push for targeted gene therapies. Viral vectors and nonviral nanoparticles have been thoroughly investigated for gene delivery and show promise as vectors for gene therapy for pediatric brain cancer. Here, we review inorganic and organic materials in development for nanoparticle gene delivery to the brain with a particular focus on safety.
Collapse
Affiliation(s)
- Kathryn M Luly
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - John Choi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yuan Rui
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Institute for Nanobiotechnology & The Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Institute for Nanobiotechnology & The Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Ophthalmology & Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Materials Science & Engineering & Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21287, USA
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Eric M Jackson
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
20
|
Fonseca A, Afzal S, Bowes L, Crooks B, Larouche V, Jabado N, Perreault S, Johnston DL, Zelcer S, Fleming A, Scheinemann K, Silva M, Vanan MI, Mpofu C, Wilson B, Eisenstat DD, Lafay-Cousin L, Hukin J, Hawkins C, Bartels U, Bouffet E. Pontine gliomas a 10-year population-based study: a report from The Canadian Paediatric Brain Tumour Consortium (CPBTC). J Neurooncol 2020; 149:45-54. [PMID: 32632896 DOI: 10.1007/s11060-020-03568-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/22/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Diffuse intrinsic pontine gliomas (DIPG) are midline gliomas that arise from the pons and the majority are lethal within a few months after diagnosis. Due to the lack of histological diagnosis the epidemiology of DIPG is not completely understood. The aim of this report is to provide population-based data to characterize the descriptive epidemiology of this condition in Canadian children. PATIENTS AND METHODS A national retrospective study of children and adolescents diagnosed with DIPG between 2000 and 2010 was undertaken. All cases underwent central review to determine clinical and radiological diagnostic characteristics. Crude incidence figures were calculated using age-adjusted (0-17 year) population data from Statistics Canada. Survival analyses were performed using the Kaplan-Meier method. RESULTS A total of 163 patients with pontine lesions were identified. Central review determined one-hundred and forty-three patients who met clinical, radiological and/or histological criteria for diagnosis. We estimate an incidence rate of 1.9 DIPG/1,000,000 children/year in the Canadian population over a 10 years period. Median age at diagnosis was 6.8 years and 50.3% of patients were female. Most patients presented with cranial nerve palsies (76%) and ataxia (66%). Despite typical clinical and radiological characteristics, histological confirmation reported three lesions to be low-grade gliomas and three were diagnosed as CNS embryonal tumor not otherwise specified (NOS). CONCLUSIONS Our study highlights the challenges associated with epidemiology studies on DIPG and the importance of central review for incidence rate estimations. It emphasizes that tissue biopsies are required for accurate histological and molecular diagnosis in patients presenting with pontine lesions and reinforces the limitations of radiological and clinical diagnosis in DIPG. Likewise, it underscores the urgent need to increase the availability and accessibility to clinical trials.
Collapse
Affiliation(s)
- Adriana Fonseca
- Division of Haematology Oncology, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, M5G 1X8, Canada.
| | - Samina Afzal
- IWK Health Center, Dalhousie University, Halifax, B3K 6R8, Canada.,Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates
| | - Lynette Bowes
- Janeway Child Health Center, Memorial University, St. John's, A1B 3V6, Canada
| | - Bruce Crooks
- IWK Health Center, Dalhousie University, Halifax, B3K 6R8, Canada
| | - Valerie Larouche
- CHU de Québec- Université Laval, Laval University, Québec, G1V 4G2, Canada
| | - Nada Jabado
- Montreal Children's Hospital, McGill University, Montreal, H4A 3J1, Canada
| | - Sebastien Perreault
- Centre Hospitalier Universitaire Sainte, Justine, Université de Montreal, Montreal, H3T 1C5, Canada
| | - Donna L Johnston
- Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, K1H 8L1, Canada
| | - Shayna Zelcer
- London Health Sciences Children's Hospital, Western University, London, N6A 5A5, Canada
| | - Adam Fleming
- McMaster Children's Hospital, McMaster University, Hamilton, L8P 1H1, Canada
| | - Katrin Scheinemann
- McMaster Children's Hospital, McMaster University, Hamilton, L8P 1H1, Canada.,Department of Pediatrics, Kantonsspital Aarau, Aarau, Switzerland.,University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Mariana Silva
- Kingston General Hospital, Queens University, Kingston, K7L 2V7, Canada
| | - Magimairajan Issai Vanan
- Cancer Care Manitoba, Pediatrics and Child Health, University of Manitoba, Winnipeg, R3E 0V9, Canada
| | - Chris Mpofu
- Jim Pattison Children's Hospital, University of Saskatchewan, Saskatoon, S7N 0W8, Canada
| | - Beverly Wilson
- Stollery Children's Hospital, University of Alberta, Edmonton, T6G 2B7, Canada
| | - David D Eisenstat
- Stollery Children's Hospital, University of Alberta, Edmonton, T6G 2B7, Canada
| | - Lucie Lafay-Cousin
- Alberta Children's Hospital, University of Calgary, Calgary, T3B 6A8, Canada
| | - Juliette Hukin
- BC Children's Hospital, University of British Columbia, Vancouver, V6H 3N1, Canada
| | - Cynthia Hawkins
- Division of Haematology Oncology, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, M5G 1X8, Canada
| | - Ute Bartels
- Division of Haematology Oncology, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, M5G 1X8, Canada
| | - Eric Bouffet
- Division of Haematology Oncology, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, M5G 1X8, Canada
| |
Collapse
|
21
|
Impact of histological diagnosis on the treatment of atypical brainstem lesions. Sci Rep 2020; 10:11065. [PMID: 32632139 PMCID: PMC7338439 DOI: 10.1038/s41598-020-68063-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 06/04/2020] [Indexed: 11/29/2022] Open
Abstract
For atypical brainstem lesions, histological diagnosis can have an impact on treatment, especially in cases where diffuse glioma is not found. Since radiotherapy is the only therapeutic modality that has shown clinical and radiographic improvement in patients with diffuse glioma, the misdiagnosis of diffuse glioma can have drastic consequences, particularly in patients with nontumorous lesions. Thus, the purpose of this study was to evaluate the impact of histological diagnosis on the treatment of atypical brainstem lesions. This was a retrospective study of 31 patients who underwent biopsy of atypical brainstem lesions. The procedures were performed between January 2008 and December 2018 at the Life Center Hospital and Santa Casa de Belo Horizonte, MG, Brazil. A diagnosis was obtained in 26 (83.9%) cases. Three patients presented complications: one presented bleeding with no clinical repercussions and two showed worsening of neurological deficit, only one of which was definitive. No mortality occurred due to the procedure. The histological diagnosis was diffuse glioma in seven cases (22.6%) and not diffuse glioma in 19 cases (61.3%). Thus, the histological diagnosis had an impact on the treatment of 19 patients (treatment impact rate: 61.3%). The histological diagnosis of intrinsic brainstem lesions is a safe, efficient procedure with a high diagnosis rate, and as such, it should be considered in the management of atypical lesions.
Collapse
|
22
|
Van Gool SW, Makalowski J, Bonner ER, Feyen O, Domogalla MP, Prix L, Schirrmacher V, Nazarian J, Stuecker W. Addition of Multimodal Immunotherapy to Combination Treatment Strategies for Children with DIPG: A Single Institution Experience. MEDICINES 2020; 7:medicines7050029. [PMID: 32438648 PMCID: PMC7281768 DOI: 10.3390/medicines7050029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 01/29/2023]
Abstract
Background: The prognosis of children with diffuse intrinsic pontine glioma (DIPG) remains dismal despite radio- and chemotherapy or molecular-targeted therapy. Immunotherapy is a powerful and promising approach for improving the overall survival (OS) of children with DIPG. Methods: A retrospective analysis for feasibility, immune responsiveness, and OS was performed on 41 children treated in compassionate use with multimodal therapy consisting of Newcastle disease virus, hyperthermia, and autologous dendritic cell vaccines as part of an individualized combinatorial treatment approach for DIPG patients. Results: Patients were treated at diagnosis (n = 28) or at the time of progression (n = 13). In the case of 16 patients, histone H3K27M mutation was confirmed by analysis of biopsy (n = 9) or liquid biopsy (n = 9) specimens. PDL1 mRNA expression was detected in circulating tumor cells of ten patients at diagnosis. Multimodal immunotherapy was feasible as scheduled, until progression, in all patients without major toxicity. When immunotherapy was part of primary treatment, median PFS and OS were 8.4 m and 14.4 m from the time of diagnosis, respectively, with a 2-year OS of 10.7%. When immunotherapy was given at the time of progression, median PFS and OS were 6.5 m and 9.1 m, respectively. A longer OS was associated with a Th1 shift and rise in PanTum Detect test scores. Conclusions: Multimodal immunotherapy is feasible without major toxicity, and warrants further investigation as part of a combinatorial treatment approach for children diagnosed with DIPG.
Collapse
Affiliation(s)
- Stefaan W. Van Gool
- Immun-Onkologisches Zentrum Köln, Hohenstaufenring 30-32, 50674 Köln, Germany; (J.M.); (M.P.D.); (V.S.); (W.S.)
- Correspondence: ; Tel.: +49-221-420-39925
| | - Jennifer Makalowski
- Immun-Onkologisches Zentrum Köln, Hohenstaufenring 30-32, 50674 Köln, Germany; (J.M.); (M.P.D.); (V.S.); (W.S.)
| | - Erin R. Bonner
- Center for Genetic Medicine, Children’s National Health System, Washington, DC 20010, USA;
- Institute for Biomedical Sciences, The George Washington University School of Medicine and health Sciences, Washington, DC 20052, USA
| | - Oliver Feyen
- Zyagnum, Reißstrasse 1, 64319 Pfungstadt, Germany;
| | - Matthias P. Domogalla
- Immun-Onkologisches Zentrum Köln, Hohenstaufenring 30-32, 50674 Köln, Germany; (J.M.); (M.P.D.); (V.S.); (W.S.)
| | - Lothar Prix
- Biofocus, Berghäuser Strasse 295, 45659 Recklinghausen, Germany;
| | - Volker Schirrmacher
- Immun-Onkologisches Zentrum Köln, Hohenstaufenring 30-32, 50674 Köln, Germany; (J.M.); (M.P.D.); (V.S.); (W.S.)
| | - Javad Nazarian
- DIPG Research Institute, Universitäts-Kinderspital Zürich; Steinwiesstrasse 75, Ch-8032 Zürich, Switzerland;
| | - Wilfried Stuecker
- Immun-Onkologisches Zentrum Köln, Hohenstaufenring 30-32, 50674 Köln, Germany; (J.M.); (M.P.D.); (V.S.); (W.S.)
| |
Collapse
|
23
|
Mendez FM, Núñez FJ, Garcia-Fabiani MB, Haase S, Carney S, Gauss JC, Becher OJ, Lowenstein PR, Castro MG. Epigenetic reprogramming and chromatin accessibility in pediatric diffuse intrinsic pontine gliomas: a neural developmental disease. Neuro Oncol 2020; 22:195-206. [PMID: 32078691 PMCID: PMC7032633 DOI: 10.1093/neuonc/noz218] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a rare but deadly pediatric brainstem tumor. To date, there is no effective therapy for DIPG. Transcriptomic analyses have revealed DIPGs have a distinct profile from other pediatric high-grade gliomas occurring in the cerebral hemispheres. These unique genomic characteristics coupled with the younger median age group suggest that DIPG has a developmental origin. The most frequent mutation in DIPG is a lysine to methionine (K27M) mutation that occurs on H3F3A and HIST1H3B/C, genes encoding histone variants. The K27M mutation disrupts methylation by polycomb repressive complex 2 on histone H3 at lysine 27, leading to global hypomethylation. Histone 3 lysine 27 trimethylation is an important developmental regulator controlling gene expression. This review discusses the developmental and epigenetic mechanisms driving disease progression in DIPG, as well as the profound therapeutic implications of epigenetic programming.
Collapse
Affiliation(s)
- Flor M Mendez
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Felipe J Núñez
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Maria B Garcia-Fabiani
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Santiago Haase
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Stephen Carney
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jessica C Gauss
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Oren J Becher
- Department of Pediatrics, Northwestern University, Chicago, Illinois
- Ann & Robert Lurie Children’s Hospital of Chicago, Division of Hematology-Oncology and Stem Cell Transplant, Chicago, Illinois
| | - Pedro R Lowenstein
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Maria G Castro
- Department of Cell and Developmental Biology and Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
24
|
Fostering open collaboration in drug development for paediatric brain tumours. Biochem Soc Trans 2019; 47:1471-1479. [PMID: 31551357 PMCID: PMC6824674 DOI: 10.1042/bst20190315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 01/11/2023]
Abstract
Brain tumours have become the leading cause of child mortality from cancer. Indeed, aggressive brainstem tumours, such as diffuse intrinsic pontine glioma (DIPG), are nearly uniformly fatal. These tumours display a unique set of driver mutations that distinguish them from adult gliomas and define new opportunity for the development of precision medicines. The specific association of ACVR1 mutations with DIPG tumours suggests a direct link to neurodevelopment and highlights the encoded bone morphogenetic protein receptor kinase ALK2 as a promising drug target. Beneficial effects of ALK2 inhibition have now been observed in two different in vivo models of DIPG. Nonetheless, such tumours present a huge challenge for traditional economic models of drug development due to their small market size, high failure rate, tumour location and paediatric population. Moreover, a toolkit of different investigational drugs may be needed to fully address the heterogeneity of these tumours in clinical trials. One new business model is suggested by M4K Pharma, a recent virtual start up that aims to align diffuse academic and industry research into a collaborative open science drug discovery programme. Fostering scientific collaboration may offer hope in rare conditions of dire unmet clinical need and provide an alternative route to affordable medicines.
Collapse
|
25
|
Ebrahimi A, Skardelly M, Schuhmann MU, Ebinger M, Reuss D, Neumann M, Tabatabai G, Kohlhof-Meinecke P, Schittenhelm J. High frequency of H3 K27M mutations in adult midline gliomas. J Cancer Res Clin Oncol 2019; 145:839-850. [PMID: 30610375 DOI: 10.1007/s00432-018-02836-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/27/2018] [Indexed: 12/27/2022]
Abstract
PURPOSE Diffuse midline gliomas, H3 K27M-mutant were introduced as a new grade IV entity in WHO classification of tumors 2016. These tumors occur often in pediatric patients and show an adverse prognosis with a median survival less than a year. Most of the studies on these tumors, previously known as pediatric diffuse intrinsic pontine glioma, are on pediatric patients and its significance in adult patients is likely underestimated. METHODS We studied 165 cases of brain tumors of midline localization initially diagnosed as diffuse astrocytomas, oligodendrogliomas, pilocytic astrocytomas, supependymomas, ependymomas and medulloblastomas in patients with an age range of 2-85. RESULTS We identified 41 diffuse midline gliomas according WHO 2016, including 12 pediatric and 29 adult cases, among them two cases with histological features of low grade tumors: pilocytic astrocytoma and subependymoma. 49% (20/41) of the patients were above 30 years old by the first tumor manifestation including 29% (11/41) above 54 that signifies a broader age spectrum as previously reported. Our study confirms that H3 K27M mutations are associated with a poorer prognosis in pediatric patients compared to wild-type tumors, while in adult patients these mutations do not influence the survival significantly. The pattern of tumor growth was different in pediatric compared to adult patients; a diffuse growth along the brain axis was more evident in adult compared to pediatric patients (24% vs. 15%). CONCLUSION H3 K27M mutations are frequent in adult midline gliomas and have a prognostic role similar to H3 K27M wild-type high-grade tumors.
Collapse
Affiliation(s)
- Azadeh Ebrahimi
- Department of Neuropathology, Institute of Pathology and Neuropathology, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, 72076, Tuebingen, Germany.
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Center for CNS Tumors, Comprehensive Cancer Center Tuebingen-Stuttgart, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany.
- Department of Neuropathology, Institute of Pathology, University Hospital of Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany.
| | - Marco Skardelly
- Department of Neurosurgery, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, 72076, Tuebingen, Germany
- Interdisciplinary Division of Neurooncology, Departments of Vascular Neurology and Neurosurgery, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, 72076, Tuebingen, Germany
- Laboratory for Clinical and Experimental Neurooncology, Hertie-Institute for Clinical Brain Research, Tuebingen, Germany
- Center for CNS Tumors, Comprehensive Cancer Center Tuebingen-Stuttgart, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Martin U Schuhmann
- Department of Neurosurgery, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, 72076, Tuebingen, Germany
| | - Martin Ebinger
- Department of General Pediatrics, Hematology/Oncology, University Children's Hospital, 72076, Tuebingen, Germany
| | - David Reuss
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, University Hospital of Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Manuela Neumann
- Department of Neuropathology, Institute of Pathology and Neuropathology, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, 72076, Tuebingen, Germany
- Center for CNS Tumors, Comprehensive Cancer Center Tuebingen-Stuttgart, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Ghazaleh Tabatabai
- Interdisciplinary Division of Neurooncology, Departments of Vascular Neurology and Neurosurgery, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, 72076, Tuebingen, Germany
- Laboratory for Clinical and Experimental Neurooncology, Hertie-Institute for Clinical Brain Research, Tuebingen, Germany
- Center for Personalized Medicine, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- German Consortium for Translational Cancer Research (DKTK), DKFZ Partner Site Tuebingen, Tuebingen, Germany
- Center for CNS Tumors, Comprehensive Cancer Center Tuebingen-Stuttgart, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | | | - Jens Schittenhelm
- Department of Neuropathology, Institute of Pathology and Neuropathology, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, 72076, Tuebingen, Germany.
- Center for CNS Tumors, Comprehensive Cancer Center Tuebingen-Stuttgart, University Hospital of Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
26
|
Role of Radiation Therapy in the Management of Diffuse Intrinsic Pontine Glioma: A Systematic Review. Adv Radiat Oncol 2019; 4:520-531. [PMID: 31360809 PMCID: PMC6639749 DOI: 10.1016/j.adro.2019.03.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/20/2019] [Indexed: 01/05/2023] Open
Abstract
Purpose Diffuse intrinsic pontine glioma (DIPG) is the most aggressive primary pediatric brain tumor, with <10% of children surviving 2 years. Radiation therapy (RT) remains the mainstay of treatment, but there is a great clinical need for improvements and advancements in treatment strategies. The aim of this systematic review was to identify all available studies in which RT was used to treat patients with DIPG. Methods and Materials A literature search for studies published up to March 10, 2018 was conducted using the PubMed database. We identified 384 articles using search items “diffuse intrinsic pontine glioma” and 221 articles using search items “diffuse brainstem glioma radiotherapy.” Included studies were prospective and retrospective series that reported outcomes of DIPG treatment with RT. Results We identified 49 studies (1286 patients) using upfront conventionally fractionated RT, 5 studies (92 patients) using hypofractionated RT, and 8 studies (348 patients) using hyperfractionated RT. The mean median overall survival (OS) was 12.0 months, 10.2 months, and 7.9 months in patients who received conventional, hyperfractionated, and hypofractionated RT regimens, respectively. Patients undergoing radiosensitizing therapy had a mean median OS of 11.5 months, and patients who did not receive concomitant systemic therapy had an OS of 9.4 months. In patients who received salvage RT, the mean median OS from initial diagnosis was 16.3 months. Conclusions As one of the largest systematic reviews examining RT for DIPG, this report may serve as a useful tool to help clinicians choose the most appropriate treatment approach, while also providing a platform for future investigations into the utility of RT and systemic therapy.
Collapse
|
27
|
Pollack IF, Agnihotri S, Broniscer A. Childhood brain tumors: current management, biological insights, and future directions. J Neurosurg Pediatr 2019; 23:261-273. [PMID: 30835699 PMCID: PMC6823600 DOI: 10.3171/2018.10.peds18377] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 10/29/2018] [Indexed: 02/06/2023]
Abstract
Brain tumors are the most common solid tumors in children, and, unfortunately, many subtypes continue to have a suboptimal long-term outcome. During the last several years, however, remarkable advances in our understanding of the molecular underpinnings of these tumors have occurred as a result of high-resolution genomic, epigenetic, and transcriptomic profiling, which have provided insights for improved tumor categorization and molecularly directed therapies. While tumors such as medulloblastomas have been historically grouped into standard- and high-risk categories, it is now recognized that these tumors encompass four or more molecular subsets with distinct clinical and molecular characteristics. Likewise, high-grade glioma, which for decades was considered a single high-risk entity, is now known to comprise multiple subsets of tumors that differ in terms of patient age, tumor location, and prognosis. The situation is even more complex for ependymoma, for which at least nine subsets of tumors have been described. Conversely, the majority of pilocytic astrocytomas appear to result from genetic changes that alter a single, therapeutically targetable molecular pathway. Accordingly, the present era is one in which treatment is evolving from the historical standard of radiation and conventional chemotherapy to a more nuanced approach in which these modalities are applied in a risk-adapted framework and molecularly targeted therapies are implemented to augment or, in some cases, replace conventional therapy. Herein, the authors review advances in the categorization and treatment of several of the more common pediatric brain tumors and discuss current and future directions in tumor management that hold significant promise for patients with these challenging tumors.
Collapse
|
28
|
Welby JP, Kaptzan T, Wohl A, Peterson TE, Raghunathan A, Brown DA, Gupta SK, Zhang L, Daniels DJ. Current Murine Models and New Developments in H3K27M Diffuse Midline Gliomas. Front Oncol 2019; 9:92. [PMID: 30873381 PMCID: PMC6400847 DOI: 10.3389/fonc.2019.00092] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/31/2019] [Indexed: 01/24/2023] Open
Abstract
Diffuse Midline Gliomas with Histone 3-Lysine-27-Methionine (H3K27M) mutation constitute the majority of Diffuse Intrinsic Pontine Glioma (DIPG), which is the most aggressive form of pediatric glioma with a dire prognosis. DIPG are lethal tumors found in younger children with a median survival <1 year from diagnosis. Discovery of the characteristic H3K27M mutations offers opportunity and hope for development of targeted therapies for this deadly disease. The H3K27M mutation, likely through epigenetic alterations in specific H3 lysine trimethylation levels and subsequent gene expression, plays a significant role in pathogenesis of DIPG. Animal models accurately depicting molecular characteristics of H3K27M DIPG are important to elucidate underlying pathologic events and for preclinical drug evaluation. Here we review the past and present DIPG models and describe our efforts developing patient derived cell lines and xenografts from pretreated surgical specimens. Pre-treated surgical samples retain the characteristic genomic and phenotypic hallmarks of DIPG and establish orthotopic tumors in the mouse brainstem that recapitulate radiographic and morphological features of the original human DIPG tumor. These models that contain the H3K27M mutation constitute a valuable tool to further study this devastating disease and ultimately may uncover novel therapeutic vulnerabilities.
Collapse
Affiliation(s)
- John P Welby
- Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Tatiana Kaptzan
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Anton Wohl
- Department of Neurosurgery, Chaim Sheba Medical Center, Tel-HaShomer, Ramat-Gan, Israel
| | | | - Aditya Raghunathan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Desmond A Brown
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, United States
| | - Shiv K Gupta
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States
| | - Liang Zhang
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, United States
| | - David J Daniels
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
29
|
Gupta N, Goumnerova LC, Manley P, Chi SN, Neuberg D, Puligandla M, Fangusaro J, Goldman S, Tomita T, Alden T, DiPatri A, Rubin JB, Gauvain K, Limbrick D, Leonard J, Geyer JR, Leary S, Browd S, Wang Z, Sood S, Bendel A, Nagib M, Gardner S, Karajannis MA, Harter D, Ayyanar K, Gump W, Bowers DC, Weprin B, MacDonald TJ, Aguilera D, Brahma B, Robison NJ, Kiehna E, Krieger M, Sandler E, Aldana P, Khatib Z, Ragheb J, Bhatia S, Mueller S, Banerjee A, Bredlau AL, Gururangan S, Fuchs H, Cohen KJ, Jallo G, Dorris K, Handler M, Comito M, Dias M, Nazemi K, Baird L, Murray J, Lindeman N, Hornick JL, Malkin H, Sinai C, Greenspan L, Wright KD, Prados M, Bandopadhayay P, Ligon KL, Kieran MW. Prospective feasibility and safety assessment of surgical biopsy for patients with newly diagnosed diffuse intrinsic pontine glioma. Neuro Oncol 2018; 20:1547-1555. [PMID: 29741745 PMCID: PMC6176802 DOI: 10.1093/neuonc/noy070] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Diagnosis of diffuse intrinsic pontine glioma (DIPG) has relied on imaging studies, since the appearance is pathognomonic, and surgical risk was felt to be high and unlikely to affect therapy. The DIPG Biology and Treatment Study (DIPG-BATS) reported here incorporated a surgical biopsy at presentation and stratified subjects to receive FDA-approved agents chosen on the basis of specific biologic targets. Methods Subjects were eligible for the trial if the clinical features and imaging appearance of a newly diagnosed tumor were consistent with a DIPG. Surgical biopsies were performed after enrollment and prior to definitive treatment. All subjects were treated with conventional external beam radiotherapy with bevacizumab, and then stratified to receive bevacizumab with erlotinib or temozolomide, both agents, or neither agent, based on O6-methylguanine-DNA methyltransferase status and epidermal growth factor receptor expression. Whole-genome sequencing and RNA sequencing were performed but not used for treatment assignment. Results Fifty-three patients were enrolled at 23 institutions, and 50 underwent biopsy. The median age was 6.4 years, with 24 male and 29 female subjects. Surgical biopsies were performed with a specified technique and no deaths were attributed to the procedure. Two subjects experienced grade 3 toxicities during the procedure (apnea, n = 1; hypertension, n = 1). One subject experienced a neurologic deficit (left hemiparesis) that did not fully recover. Of the 50 tumors biopsied, 46 provided sufficient tissue to perform the study assays (92%, two-stage exact binomial 90% CI: 83%-97%). Conclusions Surgical biopsy of DIPGs is technically feasible, associated with acceptable risks, and can provide biologic data that can inform treatment decisions.
Collapse
Affiliation(s)
- Nalin Gupta
- UCSF Benioff Children’s Hospital & University of California San Francisco, San Francisco, California
| | - Liliana C Goumnerova
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Boston Children’s Hospital, Boston, Massachusetts
| | - Peter Manley
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Boston Children’s Hospital, Boston, Massachusetts
| | - Susan N Chi
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Boston Children’s Hospital, Boston, Massachusetts
| | | | | | - Jason Fangusaro
- Ann & Robert H. Lurie Children’s Hospital of Chicago & Northwestern University, Chicago, Illinois
| | - Stewart Goldman
- Ann & Robert H. Lurie Children’s Hospital of Chicago & Northwestern University, Chicago, Illinois
| | - Tadanori Tomita
- Ann & Robert H. Lurie Children’s Hospital of Chicago & Northwestern University, Chicago, Illinois
| | - Tord Alden
- Ann & Robert H. Lurie Children’s Hospital of Chicago & Northwestern University, Chicago, Illinois
| | - Arthur DiPatri
- Ann & Robert H. Lurie Children’s Hospital of Chicago & Northwestern University, Chicago, Illinois
| | - Joshua B Rubin
- Washington University Medical Center & St. Louis Children’s Hospital, St. Louis, Missouri
| | - Karen Gauvain
- Washington University Medical Center & St. Louis Children’s Hospital, St. Louis, Missouri
| | - David Limbrick
- Washington University Medical Center & St. Louis Children’s Hospital, St. Louis, Missouri
| | - Jeffrey Leonard
- Washington University Medical Center & St. Louis Children’s Hospital, St. Louis, Missouri
| | - J Russel Geyer
- Seattle Children’s Hospital & University of Washington, Seattle, Washington
| | - Sarah Leary
- Seattle Children’s Hospital & University of Washington, Seattle, Washington
| | - Samuel Browd
- Seattle Children’s Hospital & University of Washington, Seattle, Washington
| | - Zhihong Wang
- Children’s Hospital of Michigan & Wayne State University, Detroit, Michigan
| | - Sandeep Sood
- Children’s Hospital of Michigan & Wayne State University, Detroit, Michigan
| | - Anne Bendel
- Children’s Hospitals and Clinics of Minnesota, Minneapolis, Minnesota
| | - Mahmoud Nagib
- Children’s Hospitals and Clinics of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | - William Gump
- University of Louisville & Norton’s Children’s Hospital, Louisville, Kentucky
| | - Daniel C Bowers
- University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bradley Weprin
- University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tobey J MacDonald
- Children’s Healthcare of Atlanta & Emory University, Atlanta, Georgia
| | - Dolly Aguilera
- Children’s Healthcare of Atlanta & Emory University, Atlanta, Georgia
| | | | | | - Erin Kiehna
- Children’s Hospital Los Angeles, Los Angeles, California
| | - Mark Krieger
- Children’s Hospital Los Angeles, Los Angeles, California
| | - Eric Sandler
- Nemours Children’s Clinic, Wolfson’s Children’s Hospital & University of Florida, Jacksonville, Florida
| | - Philipp Aldana
- Nemours Children’s Clinic, Wolfson’s Children’s Hospital & University of Florida, Jacksonville, Florida
| | - Ziad Khatib
- Nicklaus Children’s Hospital, Miami, Florida
| | - John Ragheb
- Nicklaus Children’s Hospital, Miami, Florida
| | | | - Sabine Mueller
- UCSF Benioff Children’s Hospital & University of California San Francisco, San Francisco, California
| | - Anu Banerjee
- UCSF Benioff Children’s Hospital & University of California San Francisco, San Francisco, California
| | - Amy-Lee Bredlau
- Medical University of South Carolina, South Carolina, Charleston, South Carolina
| | - Sri Gururangan
- Preston Robert Tisch Brain Tumor Center & Duke University Medical Center, Durham, North Carolina
| | - Herbert Fuchs
- Preston Robert Tisch Brain Tumor Center & Duke University Medical Center, Durham, North Carolina
| | | | | | - Kathleen Dorris
- Children’s Hospital of Colorado & University of Colorado School of Medicine, Denver, Colorado
| | - Michael Handler
- Children’s Hospital of Colorado & University of Colorado School of Medicine, Denver, Colorado
| | - Melanie Comito
- Penn State Health Children’s Hospital, Hershey, Pennsylvania
| | - Mark Dias
- Penn State Health Children’s Hospital, Hershey, Pennsylvania
| | - Kellie Nazemi
- Oregon Health & Science University & Doernbecher Children’s Hospital, Portland, Oregon
| | - Lissa Baird
- Oregon Health & Science University & Doernbecher Children’s Hospital, Portland, Oregon
| | - Jeff Murray
- Cook Children’s Medical Center, Fort Worth, Texas
| | | | | | | | - Claire Sinai
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Karen D Wright
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Boston Children’s Hospital, Boston, Massachusetts
| | - Michael Prados
- UCSF Benioff Children’s Hospital & University of California San Francisco, San Francisco, California
| | - Pratiti Bandopadhayay
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Boston Children’s Hospital, Boston, Massachusetts
- Broad Institute, Cambridge, Massachusetts
| | - Keith L Ligon
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
| | - Mark W Kieran
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Boston Children’s Hospital, Boston, Massachusetts
| |
Collapse
|
30
|
Souweidane MM, Kramer K, Pandit-Taskar N, Zhou Z, Haque S, Zanzonico P, Carrasquillo JA, Lyashchenko SK, Thakur SB, Donzelli M, Turner RS, Lewis JS, Cheung NKV, Larson SM, Dunkel IJ. Convection-enhanced delivery for diffuse intrinsic pontine glioma: a single-centre, dose-escalation, phase 1 trial. Lancet Oncol 2018; 19:1040-1050. [PMID: 29914796 DOI: 10.1016/s1470-2045(18)30322-x] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 10/14/2022]
Abstract
BACKGROUND Diffuse intrinsic pontine glioma is one of the deadliest central nervous system tumours of childhood, with a median overall survival of less than 12 months. Convection-enhanced delivery has been proposed as a means to efficiently deliver therapeutic agents directly into the brainstem while minimising systemic exposure and associated toxic effects. We did this study to evaluate the safety of convection-enhanced delivery of a radioimmunotherapy agent targeting the glioma-associated B7-H3 antigen in children with diffuse intrinsic pontine glioma. METHODS We did a phase 1, single-arm, single-centre, dose-escalation study at the Memorial Sloan Kettering Cancer Center (New York, NY, USA). Eligible patients were aged 3-21 years and had diffuse intrinsic pontine glioma as diagnosed by consensus of a multidisciplinary paediatric neuro-oncology team; a Lansky (patients <16 years of age) or Karnofsky (patients ≥16 years) performance score of at least 50 at study entry; a minimum weight of 8 kg; and had completed external beam radiation therapy (54·0-59·4 Gy at 1·8 Gy per fraction over 30-33 fractions) at least 4 weeks but no more than 14 weeks before enrolment. Seven dose-escalation cohorts were planned based on standard 3 + 3 rules: patients received a single infusion of 9·25, 18·5, 27·75, 37, 92·5, 120·25, or 148 MBq, respectively, at a concentration of about 37 MBq/mL by convection-enhanced delivery of the radiolabelled antibody [124I]-8H9. The primary endpoint was identification of the maximum tolerated dose. The analysis of the primary endpoint was done in the per-protocol population (patients who received the full planned dose of treatment), and all patients who received any dose of study treatment were included in the safety analysis. This study is registered with ClinicalTrials.gov, number NCT01502917, and is ongoing with an expanded cohort. FINDINGS From April 5, 2012, to Oct 8, 2016, 28 children were enrolled and treated in the trial, of whom 25 were evaluable for the primary endpoint. The maximum tolerated dose was not reached as no dose-limiting toxicities were observed. One (4%) of 28 patients had treatment-related transient grade 3 hemiparesis and one (4%) had grade 3 skin infection. No treatment-related grade 4 adverse events or deaths occurred. Estimated volumes of distribution (Vd) were linearly dependent on volumes of infusion (Vi) and ranged from 1·5 to 20·1 cm3, with a mean Vd/Vi ratio of 3·4 (SD 1·2). The mean lesion absorbed dose was 0·39 Gy/MBq 124I (SD 0·20). Systemic exposure was negligible, with an average lesion-to-whole body ratio of radiation absorbed dose higher than 1200. INTERPRETATION Convection-enhanced delivery in the brainstem of children with diffuse intrinsic pontine glioma who have previously received radiation therapy seems to be a rational and safe therapeutic strategy. PET-based dosimetry of the radiolabelled antibody [124I]-8H9 validated the principle of using convection-enhanced delivery in the brain to achieve high intra-lesional dosing with negligible systemic exposure. This therapeutic strategy warrants further development for children with diffuse intrinsic pontine glioma. FUNDING National Institutes of Health, The Dana Foundation, The Cure Starts Now, Solving Kids' Cancer, The Lyla Nsouli Foundation, Cookies for Kids' Cancer, The Cristian Rivera Foundation, Battle for a Cure, Cole Foundation, Meryl & Charles Witmer Charitable Foundation, Tuesdays with Mitch Charitable Foundation, and Memorial Sloan Kettering Cancer Center.
Collapse
Affiliation(s)
- Mark M Souweidane
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Neurological Surgery, Weill Medical College of Cornell University, New York, USA; Department of Pediatrics, Weill Medical College of Cornell University, New York, USA.
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pediatrics, Weill Medical College of Cornell University, New York, USA
| | - Neeta Pandit-Taskar
- Department of Radiology, Molecular Imaging and Therapy (Nuclear Medicine) Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Targeted Radioimmunotherapy and Theranostics, Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, USA
| | - Zhiping Zhou
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Neurological Surgery, Weill Medical College of Cornell University, New York, USA
| | - Sofia Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, USA
| | - Pat Zanzonico
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge A Carrasquillo
- Department of Radiology, Molecular Imaging and Therapy (Nuclear Medicine) Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Targeted Radioimmunotherapy and Theranostics, Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, USA
| | - Serge K Lyashchenko
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Radiochemistry & Molecular Imaging Probes Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, USA
| | - Sunitha B Thakur
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Donzelli
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryan S Turner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Radiochemistry & Molecular Imaging Probes Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, USA; Pharmacology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Steven M Larson
- Department of Radiology, Molecular Imaging and Therapy (Nuclear Medicine) Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Targeted Radioimmunotherapy and Theranostics, Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, USA
| | - Ira J Dunkel
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pediatrics, Weill Medical College of Cornell University, New York, USA
| |
Collapse
|
31
|
Cohen KJ, Jabado N, Grill J. Diffuse intrinsic pontine gliomas-current management and new biologic insights. Is there a glimmer of hope? Neuro Oncol 2018; 19:1025-1034. [PMID: 28371920 DOI: 10.1093/neuonc/nox021] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) has proven to be one of the most challenging of all pediatric cancers. Owing to a historical reticence to obtain tumor tissue for study, and based on an erroneous assumption that the biology of DIPG would mirror that of supratentorial high-grade astrocytomas, innumerable studies have been undertaken-all of which have had a negligible impact on the natural history of this disease. More recently, improvements in neurosurgical techniques have allowed for the safe upfront biopsy of DIPG, which, together with a wider use of autopsy tissue, has led to an evolving understanding of the biology of this tumor. The discovery of a recurrent somatic gain-of-function mutation leading to lysine 27 to methionine (p.Lys27Met, K27M) substitution in histone 3 variants characterizes more than 85% of DIPG, suggesting for the first time the role of the epigenome and histones in the pathogenesis of this disease, and more unified diagnostic criteria. Along with further molecular insights into the pathogenesis of DIPG, rational targets are being identified and studied in the hopes of improving the otherwise dismal outcome for children with DIPG.
Collapse
Affiliation(s)
- Kenneth J Cohen
- Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland; Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Université Paris-Saclay & Gustave Roussy Unité Mixte de Recherche 8203 du Centre National de la Recherche Scientifique & Departement de Cancerologie de l'Enfant et de l'Adolescent, Villejuif, France
| | - Nada Jabado
- Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland; Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Université Paris-Saclay & Gustave Roussy Unité Mixte de Recherche 8203 du Centre National de la Recherche Scientifique & Departement de Cancerologie de l'Enfant et de l'Adolescent, Villejuif, France
| | - Jacques Grill
- Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland; Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Université Paris-Saclay & Gustave Roussy Unité Mixte de Recherche 8203 du Centre National de la Recherche Scientifique & Departement de Cancerologie de l'Enfant et de l'Adolescent, Villejuif, France
| |
Collapse
|
32
|
Mathew RK, Rutka JT. Diffuse Intrinsic Pontine Glioma : Clinical Features, Molecular Genetics, and Novel Targeted Therapeutics. J Korean Neurosurg Soc 2018; 61:343-351. [PMID: 29742880 PMCID: PMC5957322 DOI: 10.3340/jkns.2018.0008] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 01/21/2018] [Indexed: 12/18/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a deadly paediatric brain cancer. Transient response to radiation, ineffective chemotherapeutic agents and aggressive biology result in rapid progression of symptoms and a dismal prognosis. Increased availability of tumour tissue has enabled the identification of histone gene aberrations, genetic driver mutations and methylation changes, which have resulted in molecular and phenotypic subgrouping. However, many of the underlying mechanisms of DIPG oncogenesis remain unexplained. It is hoped that more representative in vitro and preclinical models–using both xenografted material and genetically engineered mice–will enable the development of novel chemotherapeutic agents and strategies for targeted drug delivery. This review provides a clinical overview of DIPG, the barriers to progress in developing effective treatment, updates on drug development and preclinical models, and an introduction to new technologies aimed at enhancing drug delivery.
Collapse
Affiliation(s)
- Ryan K Mathew
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada.,Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK.,Department of Neurosurgery, Leeds General Infirmary, Leeds, UK
| | - James T Rutka
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| |
Collapse
|
33
|
Tejada S, Díez-Valle R, Domínguez PD, Patiño-García A, González-Huarriz M, Fueyo J, Gomez-Manzano C, Idoate MA, Peterkin J, Alonso MM. DNX-2401, an Oncolytic Virus, for the Treatment of Newly Diagnosed Diffuse Intrinsic Pontine Gliomas: A Case Report. Front Oncol 2018; 8:61. [PMID: 29594041 PMCID: PMC5858123 DOI: 10.3389/fonc.2018.00061] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/22/2018] [Indexed: 12/15/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are aggressive glial brain tumors that primarily affect children, for which there is no curative treatment. Median overall survival is only one year. Currently, the scientific focus is on expanding the knowledge base of the molecular biology of DIPG, and identifying effective therapies. Oncolytic adenovirus DNX-2401 is a replication-competent, genetically modified virus capable of infecting and killing glioma cells, and stimulating an anti-tumor immune response. Clinical trials evaluating intratumoral DNX-2401 in adults with recurrent glioblastoma have demonstrated that the virus has a favorable safety profile and can prolong survival. Subsequently, these results have encouraged the transition of this biologically active therapy from adults into the pediatric population. To this aim, we have designed a clinical Phase I trial for newly diagnosed pediatric DIPG to investigate the feasibility, safety, and preliminary efficacy of delivering DNX-2401 into tumors within the pons following biopsy. This case report presents a pediatric patient enrolled in this ongoing Phase I trial for children and adolescents with newly diagnosed DIPG. The case involves an 8-year-old female patient with radiologically diagnosed DIPG who underwent stereotactic tumor biopsy immediately followed by intratumoral DNX-2401 in the same biopsy track. Because there were no safety concerns or new neurological deficits, the patient was discharged 3 days after the procedures. To our knowledge, this is the first report of intratumoral DNX-2401 for a patient with DIPG in a clinical trial. We plan to demonstrate that intratumoral delivery of an oncolytic virus following tumor biopsy for pediatric patients with DIPG is a novel and feasible approach and that DNX-2401 represents an innovative treatment for the disease.
Collapse
Affiliation(s)
- Sonia Tejada
- Department of Neurosurgery, University Hospital of Navarra, Pamplona, Spain.,The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
| | - Ricardo Díez-Valle
- Department of Neurosurgery, University Hospital of Navarra, Pamplona, Spain.,The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
| | - Pablo D Domínguez
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Radiology, University Hospital of Navarra, Pamplona, Spain
| | - Ana Patiño-García
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| | - Marisol González-Huarriz
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
| | - Juan Fueyo
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cande Gomez-Manzano
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | - Marta M Alonso
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| |
Collapse
|
34
|
Tinkle CL, Orr BA, Lucas JT, Klimo P, Patay Z, Baker SJ, Broniscer A, Qaddoumi I. Rapid and fulminant leptomeningeal spread following radiotherapy in diffuse intrinsic pontine glioma. Pediatr Blood Cancer 2017; 64:10.1002/pbc.26416. [PMID: 28084680 PMCID: PMC5796806 DOI: 10.1002/pbc.26416] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/10/2016] [Accepted: 11/25/2016] [Indexed: 11/09/2022]
Abstract
A 4-year-old male presented with rapid-onset cranial nerve palsy and ataxia. Brain magnetic resonance imaging (MRI) revealed a pontine mass lesion with discordant conventional and advanced imaging. A stereotactic core biopsy revealed glioblastoma with immunostaining suggestive of histone H3K27M and TP53 mutation, consistent with diffuse intrinsic pontine glioma. MRI 3 months after radiotherapy revealed extensive new leptomeningeal metastatic disease involving both the supra- and infratentorial brain, as well as the imaged portion of the spine. Tissue procured at the time of needle biopsy has undergone striking in vivo expansion as an orthotopic xenograft.
Collapse
Affiliation(s)
- Christopher L. Tinkle
- Department of Radiation Oncology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105
| | - Brent A. Orr
- Department of Pathology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105
| | - John T. Lucas
- Department of Radiation Oncology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105
| | - Paul Klimo
- Department of Neurosurgery, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105
| | - Zoltan Patay
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105
| | - Suzanne J. Baker
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105
| | - Alberto Broniscer
- Department of Oncology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105
| | - Ibrahim Qaddoumi
- Department of Oncology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105
| |
Collapse
|
35
|
Novel Use of the Leksell Gamma Frame for Stereotactic Biopsy of Posterior Fossa Lesions. World Neurosurg 2017; 107:1-5. [PMID: 28739517 DOI: 10.1016/j.wneu.2017.07.087] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/14/2017] [Accepted: 07/15/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Stereotactic transcerebellar biopsies of brainstem tumors have often been reported. The Leksell frame or Cosman-Roberts-Wells frame is often used in transcerebellar approaches. However, to access lesions via the cerebellum, these frames should be secured as inferiorly on the cranium as possible, which can require exaggerated neck flexion and limit the flexible trajectory to the target. To overcome these disadvantages, we have devised a new way to use the Leksell frame for transcerebellar approaches. METHODS The frame was fixed to the upper part of the head and arc support by attaching the frame upside down, which gives surgeons a wide operative field and permits flexible trajectory planning. RESULTS Under local anesthesia, the surgery was performed in a sitting position. Air was observed in the target site on postoperative computed tomography and magnetic resonance imaging, which confirmed that a specimen had been successfully sampled from the site as planned. CONCLUSIONS Our devised transcerebellar approach provided a generous operative field and a flexible trajectory, which enabled minimally invasive biopsy of a posterior fossa lesions to be performed in a short amount of time with the patient under local anesthesia.
Collapse
|
36
|
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a rare but uniformly fatal cancer of the brain, with peak incidence in children of 5–7 years of age. In contrast to most types of human cancer, there has been no significant improvement in treatment outcomes for patients with DIPG. Since DIPG occurs in the brainstem, a vital region of the brain, there are no surgical options for providing relief to patients, and chemotherapy as well as radiation therapy provide palliative relief at best. To date, more than 250 clinical trials evaluating radiotherapy along with conventional cytotoxic chemotherapy, as well as newer biologic agents, have failed to improve the dismal outcome when compared with palliative radiation alone. The recent discovery of somatic oncogenic histone gene mutations affecting chromatin regulation in DIPG has dramatically improved our understanding of the disease pathogenesis in DIPG, and these findings have stimulated the development of novel therapeutic approaches targeting epigenetic regulators for disease treatment. This review will discuss about the role of histone modification in chromatin machinery and epigenetic therapeutic strategies for the treatment of DIPG.
Collapse
Affiliation(s)
- Rintaro Hashizume
- Department of Neurological Surgery, Northwestern University.,Department of Biochemistry and Molecular Genetics, Northwestern University
| |
Collapse
|
37
|
Zukotynski KA, Vajapeyam S, Fahey FH, Kocak M, Brown D, Ricci KI, Onar-Thomas A, Fouladi M, Poussaint TY. Correlation of 18F-FDG PET and MRI Apparent Diffusion Coefficient Histogram Metrics with Survival in Diffuse Intrinsic Pontine Glioma: A Report from the Pediatric Brain Tumor Consortium. J Nucl Med 2017; 58:1264-1269. [PMID: 28360212 DOI: 10.2967/jnumed.116.185389] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 02/26/2017] [Indexed: 11/16/2022] Open
Abstract
The purpose of this study was to describe baseline 18F-FDG PET voxel characteristics in pediatric diffuse intrinsic pontine glioma (DIPG) and to correlate these metrics with baseline MRI apparent diffusion coefficient (ADC) histogram metrics, progression-free survival (PFS), and overall survival. Methods: Baseline brain 18F-FDG PET and MRI scans were obtained in 33 children from Pediatric Brain Tumor Consortium clinical DIPG trials. 18F-FDG PET images, postgadolinium MR images, and ADC MR images were registered to baseline fluid attenuation inversion recovery MR images. Three-dimensional regions of interest on fluid attenuation inversion recovery MR images and postgadolinium MR images and 18F-FDG PET and MR ADC histograms were generated. Metrics evaluated included peak number, skewness, and kurtosis. Correlation between PET and MR ADC histogram metrics was evaluated. PET pixel values within the region of interest for each tumor were plotted against MR ADC values. The association of these imaging markers with survival was described. Results: PET histograms were almost always unimodal (94%, vs. 6% bimodal). None of the PET histogram parameters (skewness or kurtosis) had a significant association with PFS, although a higher PET postgadolinium skewness tended toward a less favorable PFS (hazard ratio, 3.48; 95% confidence interval [CI], 0.75-16.28 [P = 0.11]). There was a significant association between higher MR ADC postgadolinium skewness and shorter PFS (hazard ratio, 2.56; 95% CI, 1.11-5.91 [P = 0.028]), and there was the suggestion that this also led to shorter overall survival (hazard ratio, 2.18; 95% CI, 0.95-5.04 [P = 0.067]). Higher MR ADC postgadolinium kurtosis tended toward shorter PFS (hazard ratio, 1.30; 95% CI, 0.98-1.74 [P = 0.073]). PET and MR ADC pixel values were negatively correlated using the Pearson correlation coefficient. Further, the level of PET and MR ADC correlation was significantly positively associated with PFS; tumors with higher values of ADC-PET correlation had more favorable PFS (hazard ratio, 0.17; 95% CI, 0.03-0.89 [P = 0.036]), suggesting that a higher level of negative ADC-PET correlation leads to less favorable PFS. A more significant negative correlation may indicate higher-grade elements within the tumor leading to poorer outcomes. Conclusion:18F-FDG PET and MR ADC histogram metrics in pediatric DIPG demonstrate different characteristics with often a negative correlation between PET and MR ADC pixel values. A higher negative correlation is associated with a worse PFS, which may indicate higher-grade elements within the tumor.
Collapse
Affiliation(s)
| | - Sridhar Vajapeyam
- Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Frederic H Fahey
- Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Mehmet Kocak
- University of Tennessee Health Science Center, Memphis, Tennessee.,St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Kelsey I Ricci
- Massachusetts General Hospital, Boston, Massachusetts; and
| | | | | | - Tina Young Poussaint
- Boston Children's Hospital, Boston, Massachusetts .,Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
38
|
Zhou Z, Singh R, Souweidane MM. Convection-Enhanced Delivery for Diffuse Intrinsic Pontine Glioma Treatment. Curr Neuropharmacol 2017; 15:116-128. [PMID: 27306036 PMCID: PMC5327456 DOI: 10.2174/1570159x14666160614093615] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 01/29/2015] [Accepted: 02/08/2016] [Indexed: 12/28/2022] Open
Abstract
Convection-enhanced delivery (CED) is a technique designed to deliver drugs directly into the brain or tumors. Its ability to bypass the blood-brain barrier (BBB), one of the major hurdles in delivering drugs to the brain, has made it a promising drug delivery method for the treatment of primary brain tumors. A number of clinical trials utilizing CED of various therapeutic agents have been conducted to treat patients with supratentorial high-grade gliomas. Significant responses have been observed in certain patients in all of these trials. However, the insufficient ability to monitor drug distribution and pharmacokinetics hampers CED from achieving its potentials on a larger scale. Brainstem CED for diffuse intrinsic pontine glioma (DIPG) treatment is appealing because this tumor is compact and has no definitive treatment. The safety of brainstem CED has been established in small and large animals, and recently in early stage clinical trials. There are a few current clinical trials of brainstem CED in treating DIPG patients using targeted macromolecules such as antibodies and immunotoxins. Future advances for CED in DIPG treatment will come from several directions including: choosing the right agents for infusion; developing better agents and regimen for DIPG infusion; improving instruments and technique for easier and accurate surgical targeting and for allowing multisession or prolonged infusion to implement optimal time sequence; and better understanding and control of drug distribution, clearance and time sequence. CED-based therapies for DIPG will continue to evolve with new understanding of the technique and the disease.
Collapse
|
39
|
Johung TB, Monje M. Diffuse Intrinsic Pontine Glioma: New Pathophysiological Insights and Emerging Therapeutic Targets. Curr Neuropharmacol 2017; 15:88-97. [PMID: 27157264 PMCID: PMC5327455 DOI: 10.2174/1570159x14666160509123229] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 01/27/2015] [Accepted: 02/08/2016] [Indexed: 01/04/2023] Open
Abstract
Abstract: Background Diffuse Intrinsic Pontine Glioma (DIPG) is the leading cause of brain tumor-related death in children, with median survival of less than one year. Despite decades of clinical trials, there has been no improvement in prognosis since the introduction of radiotherapy over thirty years ago. Objective To review the clinical features and current treatment challenges of DIPG, and discuss emerging insights into the unique genomic and epigenomic mechanisms driving DIPG pathogenesis that present new opportunities for the identification of therapeutic targets. Conclusion In recent years, an increased availability of biopsy and rapid autopsy tissue samples for preclinical investigation has combined with the advent of new genomic and epigenomic profiling tools to yield remarkable advancements in our understanding of DIPG disease mechanisms. As well, a deeper understanding of the developmental context of DIPG is shedding light on therapeutic targets in the microenvironment of the childhood brain.
Collapse
Affiliation(s)
| | - Michelle Monje
- Departments of Neurology, Pediatrics, Pathology, and Neurosurgery, Stanford University School of Medicine, 265 Campus Drive, Room G3077, Stanford, CA 94305, USA
| |
Collapse
|
40
|
Infinger LK, Stevenson CB. Re-Examining the Need for Tissue Diagnosis in Pediatric Diffuse Intrinsic Pontine Gliomas: A Review. Curr Neuropharmacol 2017; 15:129-133. [PMID: 27109746 PMCID: PMC5327458 DOI: 10.2174/1570159x14666160425114024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/18/2014] [Accepted: 02/08/2016] [Indexed: 01/24/2023] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a malignant brain tumor of childhood that carries an extremely poor prognosis. There are ~200-300 new cases diagnosed each year, [1, 2] and little progress has been made in changing the prognosis and outcome of the tumor since it was first documented in the literature in 1926 [3]. The median overall survival is 8-11 months [4], with an overall survival rate of 30% at 1 year, and less than 10% at 2 years [4]. This review will provide background information on DIPGs, a historical look at the trends in caring for DIPG, and current trends in diagnosis and treatment. By changing the way we care for these terminal tumors, we can work towards having a better understanding of the underlying molecular biology, and attempt to develop better chemotherapeutic tools to combat the disease.
Collapse
Affiliation(s)
| | - Charles B. Stevenson
- Cincinnati Children’s Hospital Medical Center, Division of Pediatric Neurosurgery, USA
| |
Collapse
|
41
|
Tisnado J, Young R, Peck KK, Haque S. Conventional and Advanced Imaging of Diffuse Intrinsic Pontine Glioma. J Child Neurol 2016; 31:1386-93. [PMID: 27071471 PMCID: PMC5659185 DOI: 10.1177/0883073816634855] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 01/12/2016] [Indexed: 12/11/2022]
Abstract
Diffuse intrinsic pontine glioma is the most common brainstem tumor in pediatric patients. This tumor remains one of the most deadly pediatric brain tumors. The diagnosis primarily relies on clinical symptoms and imaging findings. Conventional MRI provides a noninvasive accurate method of diagnosis of these tumors. Advanced MRI techniques are becoming more widely used and studied as additional noninvasive methods to assist clinicians in initial diagnosis and staging, monitoring disease, as well as in surgical and radiation planning. This article will provide an overview of DIPG and describe the typical imaging findings with a focus on advanced imaging techniques.
Collapse
Affiliation(s)
- Jamie Tisnado
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert Young
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kyung K Peck
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sofia Haque
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
42
|
Hennika T, Becher OJ. Diffuse Intrinsic Pontine Glioma: Time for Cautious Optimism. J Child Neurol 2016; 31:1377-85. [PMID: 26374787 PMCID: PMC6025797 DOI: 10.1177/0883073815601495] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/20/2015] [Indexed: 01/03/2023]
Abstract
Diffuse intrinsic pontine glioma is a lethal brain cancer that arises in the pons of children. The median survival for children with diffuse intrinsic pontine glioma is less than 1 year from diagnosis, and no improvement in survival has been realized in more than 30 years. Currently, the standard of care for diffuse intrinsic pontine glioma is focal radiation therapy, which provides only temporary relief. Recent genomic analysis of tumors from biopsies and autopsies, have resulted in the discovery of K27M H3.3/H3.1 mutations in 80% and ACVR1 mutations in 25% of diffuse intrinsic pontine gliomas, providing renewed hope for future success in identifying effective therapies. In addition, as stereotactic tumor biopsies at diagnosis at specialized centers have been demonstrated to be safe, biopsies have now been incorporated into several prospective clinical trials. This article summarizes the epidemiology, clinical presentation, diagnosis, prognosis, molecular genetics, current treatment, and future therapeutic directions for diffuse intrinsic pontine glioma.
Collapse
Affiliation(s)
- Tammy Hennika
- Department of Pediatrics Duke University Medical Center, Durham, NC, USA Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA
| | - Oren J Becher
- Department of Pediatrics Duke University Medical Center, Durham, NC, USA Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA Department of Pathology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
43
|
Pre-radiation chemotherapy improves survival in pediatric diffuse intrinsic pontine gliomas. Childs Nerv Syst 2016; 32:1415-23. [PMID: 27379495 DOI: 10.1007/s00381-016-3153-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/19/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND The median survival of patients with diffuse intrinsic pontine glioma (DIPG) remains less than 1 year. The BSG 98 pre-irradiation chemotherapy protocol showed a significant increase in overall survival. In contrast to current treatment strategies, patients did not have to undergo surgical stereotactic biopsy, which can sometimes lead to complications, to be included in this protocol. MATERIALS AND METHODS We retrospectively reviewed all the cases of DIPG that were treated in our department from September 15, 2004 to September 15, 2014. We compared the group of patients who followed our BSG 98 protocol to those who were treated with new targeted therapy protocols where systematic biopsy was required. RESULTS Patients in the BSG 98 protocol were treated with BCNU, cisplatin, and methotrexate, followed by radiation at disease progression. Targeted therapy protocols included radiation therapy along with treatment by erlotinib, cilengitide, or an association of nimotuzumab and vinblastine. Sixteen patients were treated with the BSG 98 protocol, and 9 patients were treated with new targeted therapy protocols. Median overall survival was significantly higher in the BSG 98 group compared to the targeted therapy group (16.1 months (95 % CI, 10.4-19.0) vs 8.8 months (95 % CI 1.4-12.3); p = 0.0003). An increase in the median progression-free survival was observed (respectively, 8.6 vs 3.0 months; p = 0.113). CONCLUSION The present study confirms that the BSG 98 protocol is one of the most effective current treatment strategies for DIPG. It may be used as the control arm in randomized trials investigating the use of innovative treatments and may be proposed to families who are averse to biopsy.
Collapse
|
44
|
Pierre-Aurélien B, Alexandru S, Federico DR, Justyna K, Carmine M, Didier F. Diffuse Intrinsic Pontine Glioma in Children: Document or Treat? World Neurosurg 2016; 93:485.e11-4. [PMID: 27422681 DOI: 10.1016/j.wneu.2016.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND The place of stereotactic biopsies in the management of diffuse intrinsic pontine gliomas (DIPGs) in children has changed over the years. Nonetheless, stereotactic biopsy remains a surgical procedure with its risks. One complication that has not been reported previously in case of a biopsy of a DIPG is metastatic seeding along the tract of the biopsy. We report the first 2 cases in the literature. CASE DESCRIPTION One 8-year-old and one 9-year-old boy were admitted for a typical DIPG. Parents choose to be included into a research protocol that required a stereotactic biopsy. The biopsy was performed in both cases without any intraoperative complications, and they both received their treatment according to protocol. Unfortunately, 3 and 1 months respectively after the biopsy, their clinical condition deteriorated. MRI showed a metastatic seeding along the tract of the biopsy, and both patients died of disease progression. CONCLUSIONS The era of targeted therapy with molecular and genomic discoveries has paved the way to a research protocol that requires a biopsy from the patient. The reported complications have never been described before. The purpose of this paper is not to suggest that no biopsy should be performed when a DIPG is suspected. For now, biopsy remains investigational, because no benefit in survival could be drawn so far for any patient. This subject deserves honest discussion with the children and their parents.
Collapse
Affiliation(s)
| | - Szathmari Alexandru
- Department of Pediatric Neurosurgery, Hôpital Femme Mère Enfant, Lyon Cedex, France
| | - Di Rocco Federico
- Department of Pediatric Neurosurgery, Hôpital Femme Mère Enfant, Lyon Cedex, France
| | - Kanold Justyna
- Pediatric Haematology and Oncology Unit, University Hospital, Clermont-Ferrand, France; INSERM-CIC 1405, CRECHE Unit, Clermont-Ferrand, France; Auvergne University, Clermont1 University, Faculty of Medicine, Clermont-Ferrand, France
| | - Mottolese Carmine
- Department of Pediatric Neurosurgery, Hôpital Femme Mère Enfant, Lyon Cedex, France
| | - Frappaz Didier
- Pediatric Institutes of Haematology-Oncology, Lyon, France
| |
Collapse
|
45
|
Abstract
Great progress has been made in many areas of pediatric oncology. However, tumors of the central nervous system (CNS) remain a significant challenge. A recent explosion of data has led to an opportunity to understand better the molecular basis of these diseases and is already providing a foundation for the pursuit of rationally chosen therapeutics targeting relevant molecular pathways. The molecular biology of pediatric brain tumors is shifting from a singular focus on basic scientific discovery to a platform upon which insights are being translated into therapies.
Collapse
|
46
|
Puget S, Blauwblomme T, Grill J. Is biopsy safe in children with newly diagnosed diffuse intrinsic pontine glioma? Am Soc Clin Oncol Educ Book 2016:629-33. [PMID: 24451809 DOI: 10.14694/edbook_am.2012.32.59] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Diffuse intrinsic pontine gliomas (DIPGs), with a median survival of 9 months, represent the biggest therapeutic challenge in pediatric neuro-oncology. Despite many clinical trials, no major improvements in treatment have been made over the past 30 years. In most cases, biopsy is not needed for diagnosis because DIPG diagnosis is based on a typical clinical picture with radiologic evidence on magnetic resonance imaging. Therefore, little data on newly diagnosed DIPG have been published and are confounded by including autopsy (i.e., postradiation therapy) cases. In most cancers, advancing to cure has been linked to the discovery of relevant biomarkers, only found by access to tissue. Therefore, to further understand the biology of DIPG, fresh tissue samples must be obtained at diagnosis. However, most neurosurgical teams are reluctant to perform biopsy in pediatric patients, citing potential risks and lack of direct benefit. Yet, in reviewing 90 patients with and the published data on brainstem biopsy, these procedures have a diagnostic yield and morbidity and mortality rates similar to those reported for other brain locations. In addition, the quality and quantity of the material obtained confirm the diagnosis and inform an extended molecular screen, including biomarker study-information important to designing next-generation trials with targeted agents. Stereotactic biopsies can be considered a safe procedure in well-trained neurosurgical teams and could be incorporated in well-defined protocols for patients with DIPG.
Collapse
Affiliation(s)
- Stephanie Puget
- From the Necker Enfants Malades Hospital, Université Paris Descartes, Sorbonne Paris Cité, France; Gustave Roussy Cancer Institute, Universite Paris Sud, Villejuif, France
| | - Thomas Blauwblomme
- From the Necker Enfants Malades Hospital, Université Paris Descartes, Sorbonne Paris Cité, France; Gustave Roussy Cancer Institute, Universite Paris Sud, Villejuif, France
| | - Jacques Grill
- From the Necker Enfants Malades Hospital, Université Paris Descartes, Sorbonne Paris Cité, France; Gustave Roussy Cancer Institute, Universite Paris Sud, Villejuif, France
| |
Collapse
|
47
|
Robison NJ, Kieran MW. Identification of novel biologic targets in the treatment of newly diagnosed diffuse intrinsic pontine glioma. Am Soc Clin Oncol Educ Book 2016:625-8. [PMID: 24451808 DOI: 10.14694/edbook_am.2012.32.190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) carry an extremely poor prognosis. Standard practice has been to base the diagnosis on classic imaging and clinical characteristics and to treat with focal radiation therapy, usually accompanied with experimental therapy. As a result of the desire to avoid upfront biopsy, little has been learned regarding the molecular features of this disease. Findings from several autopsy series have included loss of p53 and PTEN, and amplification of PDGFR. Based on these and other findings, murine models have been generated and provide a new tool for preclinical testing. DIPG biopsy at diagnosis has increasingly become incorporated into national protocols at several centers, bringing the prospect of a better understanding of DIPG biology in the future. Initial analyses of pretreatment tumors cast valuable new light and establish the importance of p53 inactivation and the RTK-PI3K pathway in this disease.
Collapse
Affiliation(s)
- Nathan J Robison
- From the Dana-Farber Children's Hospital Cancer Center, Boston, MA
| | - Mark W Kieran
- From the Dana-Farber Children's Hospital Cancer Center, Boston, MA
| |
Collapse
|
48
|
Foreman NK. Ethics of biopsy in children with newly diagnosed diffuse intrinsic pontine glioma. Am Soc Clin Oncol Educ Book 2016:634-5. [PMID: 24451810 DOI: 10.14694/edbook_am.2012.32.239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To understand the ethical dilemmas that beset this issue of biopsy in children with newly diagnosed diffuse intrinsic pontine glioma, one must understand both the history behind it and the current dominant interpretation of ethics through the medium of the institutional review boards. It is also important to understand that this article represents the author's personal viewpoint. At a consensus meeting to discuss the issue of biopsies in Diffuse Intrinsic Pontine Glioma (DIPG) at the National institutes of Health held in the fall of 2011, there were a variety of opinions expressed.
Collapse
|
49
|
Lam S, Lin Y, Auffinger B, Melkonian S. Analysis of survival in pediatric high-grade brainstem gliomas: A population-based study. J Pediatr Neurosci 2015; 10:199-206. [PMID: 26557158 PMCID: PMC4611886 DOI: 10.4103/1817-1745.165656] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: The purpose of this study was to use the National Cancer Institutes’ Surveillance, Epidemiology, and End Results (SEER) database to perform a large-scale analysis of brainstem anaplastic astrocytoma (AA) and glioblastoma multiforme (GBM). Use of the SEER database gave us a larger sample size of this rare tumor type, allowing for the analysis of the relationship between prognostic factors and survival. Materials and Methods: We selected pediatric patients (<18 years old) from the SEER database with histologically confirmed diagnoses of primary high-grade gliomas (World Health Organization Grade III/IV) of the brainstem. In univariate and multivariate analysis, we analyzed the relationship between demographic (age, gender, race, diagnosis date), histologic (AA, GBM), and treatment (surgery, radiation) factors on survival. Results: In our cohort of 124 patients, those with AA had a median survival of 13 months and those with GBM 9 months. Higher-grade tumors were associated with statistically significantly increased mortality (hazard ratio [HR]: 1.74, confidence intervals [CIs]: 1.17-2.60). Surgical intervention was associated with a significantly lower mortality, either alone (HR: 0.14, CI: 0.04-0.5) or in combination with radiation (HR: 0.35, CI: 0.15-0.82). Radiation therapy alone was significantly associated with decreased mortality within the first 9 months after diagnosis but not with overall mortality. No demographic characteristics were significantly associated with mortality. Conclusions: Outcome remains poor in the pediatric high-grade brainstem glioma population. Survival is correlated with lower-grade tumor histology, radiation therapy only in the first 9 months after diagnosis, and surgical resection.
Collapse
Affiliation(s)
- Sandi Lam
- Department of Neurosurgery, Texas Children's Hospital/Baylor College of Medicine, Houston, Texas, USA
| | - Yimo Lin
- Department of Neurosurgery, Texas Children's Hospital/Baylor College of Medicine, Houston, Texas, USA
| | - Brenda Auffinger
- Department of Neurosurgery, Texas Children's Hospital/Baylor College of Medicine, Houston, Texas, USA
| | - Stephanie Melkonian
- Department of Cancer Epidemiology, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
50
|
Vanan MI, Eisenstat DD. DIPG in Children - What Can We Learn from the Past? Front Oncol 2015; 5:237. [PMID: 26557503 PMCID: PMC4617108 DOI: 10.3389/fonc.2015.00237] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 10/08/2015] [Indexed: 02/02/2023] Open
Abstract
Brainstem tumors represent 10–15% of pediatric central nervous system tumors and diffuse intrinsic pontine glioma (DIPG) is the most common brainstem tumor of childhood. DIPG is almost uniformly fatal and is the leading cause of brain tumor-related death in children. To date, radiation therapy (RT) is the only form of treatment that offers a transient benefit in DIPG. Chemotherapeutic strategies including multi-agent neoadjuvant chemotherapy, concurrent chemotherapy with RT, and adjuvant chemotherapy have not provided any survival advantage. To overcome the restrictive ability of the intact blood–brain barrier (BBB) in DIPG, several alternative drug delivery strategies have been proposed but have met with minimal success. Targeted therapies either alone or in combination with RT have also not improved survival. Five decades of unsuccessful therapies coupled with recent advances in the genetics and biology of DIPG have taught us several important lessons (1). DIPG is a heterogeneous group of tumors that are biologically distinct from other pediatric and adult high grade gliomas (HGG). Adapting chemotherapy and targeted therapies that are used in pediatric or adult HGG for the treatment of DIPG should be abandoned (2). Biopsy of DIPG is relatively safe and informative and should be considered in the context of multicenter clinical trials (3). DIPG probably represents a whole brain disease so regular neuraxis imaging is important at diagnosis and during therapy (4). BBB permeability is of major concern in DIPG and overcoming this barrier may ensure that drugs reach the tumor (5). Recent development of DIPG tumor models should help us accurately identify and validate therapeutic targets and small molecule inhibitors in the treatment of this deadly tumor.
Collapse
Affiliation(s)
- Magimairajan Issai Vanan
- Department of Pediatrics and Child Health, University of Manitoba , Winnipeg, MB , Canada ; Department of Biochemistry and Medical Genetics, University of Manitoba , Winnipeg, MB , Canada
| | - David D Eisenstat
- Department of Pediatrics, University of Alberta , Edmonton, AB , Canada ; Department of Medical Genetics, University of Alberta , Edmonton, AB , Canada ; Department of Oncology, University of Alberta , Edmonton, AB , Canada
| |
Collapse
|