1
|
Sheikh SR, Patel NJ, Recinos VMR. Safety and Technical Efficacy of Pediatric Brainstem Biopsies: An Updated Meta-Analysis of 1000+ Children. World Neurosurg 2024; 189:428-438.e2. [PMID: 38968995 DOI: 10.1016/j.wneu.2024.06.163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Brainstem tumors represent ∼10% of pediatric brain tumors, ∼80% of these are diffuse midline glioma. Given invariably poor prognosis in diffuse midline glioma, there continues to be immense variation worldwide in performing biopsy of these lesions. Several contemporary studies in recent years have provided new data to elucidate the safety profile of biopsy and an updated meta-analysis is thus indicated. METHODS We found 29 studies of pediatric brainstem biopsy in the last 20 years (2003-2023, 1002 children). We applied meta-analysis of proportions using a random-effects model to generate point estimates, confidence intervals, and measures of heterogeneity. RESULTS Eighty-seven percent of procedures were stereotactic needle biopsies (of these, 62% with a frame, 14% without frame, and 24% robotic.) Biopsy resulted in a histological diagnosis ("technical yield") in 96.8% of cases (95% CI 95.4-98.2). Temporary complications were seen in 6% (95 CI 4-8), with the most common neurological complications being 1) cranial nerve dysfunction, 2) worsening or new ataxia, and 3) limb weakness. Permanent complications (excluding death) were seen in 1% (95% CI 0.5-2), most commonly including cranial nerve dysfunction and limb weakness. Five deaths were reported in the entire pooled cohort of 1002 children (0.5%). CONCLUSIONS When counseling families on the merits of brainstem biopsy in children, it is reasonable to state that permanent morbidity is rare (<2%). If biopsy is performed specifically to facilitate enrollment in clinical trials requiring a molecular diagnosis, the risks of biopsy outlined here should be weighed against potential benefits of trial enrollment.
Collapse
Affiliation(s)
| | - Neha J Patel
- Department of Pediatric Hematology-Oncology and Blood & Marrow Transplant, Cleveland Clinic, Cleveland, Ohio, USA
| | | |
Collapse
|
2
|
Furst LM, Roussel EM, Leung RF, George AM, Best SA, Whittle JR, Firestein R, Faux MC, Eisenstat DD. The Landscape of Pediatric High-Grade Gliomas: The Virtues and Pitfalls of Pre-Clinical Models. BIOLOGY 2024; 13:424. [PMID: 38927304 PMCID: PMC11200883 DOI: 10.3390/biology13060424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
Pediatric high-grade gliomas (pHGG) are malignant and usually fatal central nervous system (CNS) WHO Grade 4 tumors. The majority of pHGG consist of diffuse midline gliomas (DMG), H3.3 or H3.1 K27 altered, or diffuse hemispheric gliomas (DHG) (H3.3 G34-mutant). Due to diffuse tumor infiltration of eloquent brain areas, especially for DMG, surgery has often been limited and chemotherapy has not been effective, leaving fractionated radiation to the involved field as the current standard of care. pHGG has only been classified as molecularly distinct from adult HGG since 2012 through Next-Generation sequencing approaches, which have shown pHGG to be epigenetically regulated and specific tumor sub-types to be representative of dysregulated differentiating cells. To translate discovery research into novel therapies, improved pre-clinical models that more adequately represent the tumor biology of pHGG are required. This review will summarize the molecular characteristics of different pHGG sub-types, with a specific focus on histone K27M mutations and the dysregulated gene expression profiles arising from these mutations. Current and emerging pre-clinical models for pHGG will be discussed, including commonly used patient-derived cell lines and in vivo modeling techniques, encompassing patient-derived xenograft murine models and genetically engineered mouse models (GEMMs). Lastly, emerging techniques to model CNS tumors within a human brain environment using brain organoids through co-culture will be explored. As models that more reliably represent pHGG continue to be developed, targetable biological and genetic vulnerabilities in the disease will be more rapidly identified, leading to better treatments and improved clinical outcomes.
Collapse
Affiliation(s)
- Liam M. Furst
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia; (L.M.F.); (E.M.R.); (R.F.L.); (M.C.F.)
- Stem Cell Medicine, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia;
| | - Enola M. Roussel
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia; (L.M.F.); (E.M.R.); (R.F.L.); (M.C.F.)
- Stem Cell Medicine, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia;
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Ryan F. Leung
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia; (L.M.F.); (E.M.R.); (R.F.L.); (M.C.F.)
- Stem Cell Medicine, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia;
| | - Ankita M. George
- Stem Cell Medicine, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia;
| | - Sarah A. Best
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3010, Australia;
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - James R. Whittle
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia;
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3010, Australia;
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Ron Firestein
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia;
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Maree C. Faux
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia; (L.M.F.); (E.M.R.); (R.F.L.); (M.C.F.)
- Stem Cell Medicine, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia;
- Department of Surgery, University of Melbourne, Parkville, VIC 3010, Australia
| | - David D. Eisenstat
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia; (L.M.F.); (E.M.R.); (R.F.L.); (M.C.F.)
- Stem Cell Medicine, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia;
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Children’s Cancer Centre, The Royal Children’s Hospital Melbourne, 50 Flemington Road, Parkville, VIC 3052, Australia
| |
Collapse
|
3
|
Nduom EK, Glod J, Brown DA, Fagan M, Dalmage M, Heiss J, Steinberg SM, Peer C, Figg WD, Jackson S. Clinical protocol: Feasibility of evaluating abemaciclib neuropharmacokinetics of diffuse midline glioma using intratumoral microdialysis. PLoS One 2023; 18:e0291068. [PMID: 37682953 PMCID: PMC10490936 DOI: 10.1371/journal.pone.0291068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/20/2023] [Indexed: 09/10/2023] Open
Abstract
Diffuse midline gliomas (DMG) are the most aggressive brain tumors of childhood and young adults, with documented 2-year survival rates <10%. Treatment failure is due in part to the function of the BBB. Intratumoral microdialysis sampling is an effective tool to determine brain entry of varied agents and could help to provide a better understanding of the relationship of drug permeability to DMG treatment responsivity. This is a non-randomized, single-center, phase 1 clinical trial. Up to seven young adult (18-39 years) patients with recurrent high-grade or diffuse midline glioma will be enrolled with the goal of 5 patients completing the trial over an anticipated 24 months. All patients will take abemaciclib pre-operatively for 4.5 days at twice daily dosing. Patients will undergo resection or biopsy, placement of a microdialysis catheter, and 48 hours of dialysate sampling coupled with timed plasma collections. If intratumoral tumor or brain dialysate sampling concentrations are >10nmol/L, or tumor tissue studies demonstrate CDK inhibition, then restart of abemaciclib therapy along with temozolomide will be administered for maintenance therapy and discontinued with evidence of radiologic or clinical disease progression. The poor survival associated with diffuse midline gliomas underscore the need for improved means to evaluate efficacy of drug delivery to tumor and peritumoral tissue. The findings of this novel study, will provide real-time measurements of BBB function which have the potential to influence future prognostic and diagnostic decisions in such a lethal disease with limited treatment options. Trial registration: Clinicaltrials.gov, NCT05413304. Registered June 10, 2022, Abemaciclib Neuropharmacokinetics of Diffuse Midline Glioma Using Intratumoral Microdialysis.
Collapse
Affiliation(s)
- Edjah K. Nduom
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - John Glod
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Desmond A. Brown
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States of America
| | - Margaret Fagan
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Mahalia Dalmage
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States of America
| | - John Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States of America
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Cody Peer
- Clinical Pharmacology, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - William D. Figg
- Clinical Pharmacology, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Sadhana Jackson
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States of America
| |
Collapse
|
4
|
Miguel Llordes G, Medina Pérez VM, Curto Simón B, Castells-Yus I, Vázquez Sufuentes S, Schuhmacher AJ. Epidemiology, Diagnostic Strategies, and Therapeutic Advances in Diffuse Midline Glioma. J Clin Med 2023; 12:5261. [PMID: 37629304 PMCID: PMC10456112 DOI: 10.3390/jcm12165261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Object: Diffuse midline glioma (DMG) is a highly aggressive and lethal brain tumor predominantly affecting children and young adults. Previously known as diffuse intrinsic pontine glioma (DIPG) or grade IV brain stem glioma, DMG has recently been reclassified as "diffuse midline glioma" according to the WHO CNS5 nomenclature, expanding the DMG demographic. Limited therapeutic options result in a poor prognosis, despite advances in diagnosis and treatment. Radiotherapy has historically been the primary treatment modality to improve patient survival. Methods: This systematic literature review aims to comprehensively compile information on the diagnosis and treatment of DMG from 1 January 2012 to 31 July 2023. The review followed the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) statement and utilized databases such as PubMed, Cochrane Library, and SciELO. Results: Currently, molecular classification of DMG plays an increasingly vital role in determining prognosis and treatment options. Emerging therapeutic avenues, including immunomodulatory agents, anti-GD2 CAR T-cell and anti-GD2 CAR-NK therapies, techniques to increase blood-brain barrier permeability, isocitrate dehydrogenase inhibitors, oncolytic and peptide vaccines, are being explored based on the tumor's molecular composition. However, more clinical trials are required to establish solid guidelines for toxicity, dosage, and efficacy. Conclusions: The identification of the H3K27 genetic mutation has led to the reclassification of certain midline tumors, expanding the DMG demographic. The field of DMG research continues to evolve, with encouraging findings that underscore the importance of highly specific and tailored therapeutic strategies to achieve therapeutic success.
Collapse
Affiliation(s)
- Gloria Miguel Llordes
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Víctor Manuel Medina Pérez
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | | | - Irene Castells-Yus
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | | | - Alberto J. Schuhmacher
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Fundación Aragonesa para la Investigación y el Desarrollo (ARAID), 50018 Zaragoza, Spain
| |
Collapse
|
5
|
Noon A, Galban S. Therapeutic avenues for targeting treatment challenges of diffuse midline gliomas. Neoplasia 2023; 40:100899. [PMID: 37030112 PMCID: PMC10119952 DOI: 10.1016/j.neo.2023.100899] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
Diffuse midline glioma (DMG) is the leading cause of brain tumor-related deaths in children. DMG typically presents with variable neurologic symptoms between ages 3 and 10. Currently, radiation remains the standard therapy for DMG to halt progression and reduce tumor bulk to minimize symptoms. However, tumors recur in almost 100% of patients and thus, DMG is still considered an incurable cancer with a median survival of 9-12 months. Surgery is generally contraindicated due to the delicate organization of the brainstem, where DMG is located. Despite extensive research efforts, no chemotherapeutic agents, immune therapies, or molecularly targeted therapies have been approved to provide survival benefit. Furthermore, the efficacy of therapies is limited by poor blood-brain barrier penetration and inherent resistance mechanisms of the tumor. However, novel drug delivery approaches, along with recent advances in molecularly targeted therapies and immunotherapies, have advanced to clinical trials and may provide viable future treatment options for DMG patients. This review seeks to evaluate current therapeutics at the preclinical stage and those that have advanced to clinical trials and to discuss the challenges of drug delivery and inherent resistance to these therapies.
Collapse
Affiliation(s)
- Aleeha Noon
- College of Medicine, California Northstate University, 9700 W Taron Drive, Elk Grove, CA 95757, USA
| | - Stefanie Galban
- Center for Molecular Imaging, The University of Michigan Medical School, BSRB A502, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA; Department of Radiology, The University of Michigan Medical School, BSRB A502, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA; Rogel Cancer Center, The University of Michigan Medical School, 1500 E Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
6
|
Diffuse intrinsic pontine glioma: Insights into oncogenesis and opportunities for targeted therapy. PEDIATRIC HEMATOLOGY ONCOLOGY JOURNAL 2023. [DOI: 10.1016/j.phoj.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
|
7
|
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
|
8
|
Wagner MW, Namdar K, Napoleone M, Hainc N, Amirabadi A, Fonseca A, Laughlin S, Shroff MM, Bouffet E, Hawkins C, Khalvati F, Bartels U, Ertl-Wagner BB. Radiomic Features Based on MRI Predict Progression-Free Survival in Pediatric Diffuse Midline Glioma/Diffuse Intrinsic Pontine Glioma. Can Assoc Radiol J 2023; 74:119-126. [PMID: 35768942 DOI: 10.1177/08465371221109921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Purpose: Biopsy-based assessment of H3 K27 M status helps in predicting survival, but biopsy is usually limited to unusual presentations and clinical trials. We aimed to evaluate whether radiomics can serve as prognostic marker to stratify diffuse intrinsic pontine glioma (DIPG) subsets. Methods: In this retrospective study, diagnostic brain MRIs of children with DIPG were analyzed. Radiomic features were extracted from tumor segmentations and data were split into training/testing sets (80:20). A conditional survival forest model was applied to predict progression-free survival (PFS) using training data. The trained model was validated on the test data, and concordances were calculated for PFS. Experiments were repeated 100 times using randomized versions of the respective percentage of the training/test data. Results: A total of 89 patients were identified (48 females, 53.9%). Median age at time of diagnosis was 6.64 years (range: 1-16.9 years) and median PFS was 8 months (range: 1-84 months). Molecular data were available for 26 patients (29.2%) (1 wild type, 3 K27M-H3.1, 22 K27M-H3.3). Radiomic features of FLAIR and nonenhanced T1-weighted sequences were predictive of PFS. The best FLAIR radiomics model yielded a concordance of .87 [95% CI: .86-.88] at 4 months PFS. The best T1-weighted radiomics model yielded a concordance of .82 [95% CI: .8-.84] at 4 months PFS. The best combined FLAIR + T1-weighted radiomics model yielded a concordance of .74 [95% CI: .71-.77] at 3 months PFS. The predominant predictive radiomic feature matrix was gray-level size-zone. Conclusion: MRI-based radiomics may predict progression-free survival in pediatric diffuse midline glioma/diffuse intrinsic pontine glioma.
Collapse
Affiliation(s)
- Matthias W Wagner
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| | - Khashayar Namdar
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| | - Marc Napoleone
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Nicolin Hainc
- Nicolin Hainc:Department of Neuroradiology, Clinical Neuroscience Center, 7979University Hospital Zurich,University of Zurich, Switzerland
| | - Afsaneh Amirabadi
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Adriana Fonseca
- Department of Neurooncology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Suzanne Laughlin
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| | - Manohar M Shroff
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| | - Eric Bouffet
- Department of Neurooncology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Cynthia Hawkins
- Department of Paediatric Laboratory Medicine, Division of Pathology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Farzad Khalvati
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Ute Bartels
- Department of Neurooncology, 7979The Hospital for Sick Children, Toronto, Canada
| | - Birgit B Ertl-Wagner
- Department of Diagnostic Imaging, Division of Neuroradiology, 7979The Hospital for Sick Children, Toronto, Canada.,Department of Medical Imaging, 7938University of Toronto, Canada
| |
Collapse
|
9
|
Vallero SG, Bertero L, Morana G, Sciortino P, Bertin D, Mussano A, Ricci FS, Peretta P, Fagioli F. Pediatric diffuse midline glioma H3K27- altered: A complex clinical and biological landscape behind a neatly defined tumor type. Front Oncol 2023; 12:1082062. [PMID: 36727064 PMCID: PMC9885151 DOI: 10.3389/fonc.2022.1082062] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/23/2022] [Indexed: 01/18/2023] Open
Abstract
The 2021 World Health Organization Classification of Tumors of the Central Nervous System, Fifth Edition (WHO-CNS5), has strengthened the concept of tumor grade as a combination of histologic features and molecular alterations. The WHO-CNS5 tumor type "Diffuse midline glioma, H3K27-altered," classified within the family of "Pediatric-type diffuse high-grade gliomas," incarnates an ideally perfect integrated diagnosis in which location, histology, and genetics clearly define a specific tumor entity. It tries to evenly characterize a group of neoplasms that occur primarily in children and midline structures and that have a dismal prognosis. Such a well-defined pathological categorization has strongly influenced the pediatric oncology community, leading to the uniform treatment of most cases of H3K27-altered diffuse midline gliomas (DMG), based on the simplification that the mutation overrides the histological, radiological, and clinical characteristics of such tumors. Indeed, multiple studies have described pediatric H3K27-altered DMG as incurable tumors. However, in biology and clinical practice, exceptions are frequent and complexity is the rule. First of all, H3K27 mutations have also been found in non-diffuse gliomas. On the other hand, a minority of DMGs are H3K27 wild-type but have a similarly poor prognosis. Furthermore, adult-type tumors may rarely occur in children, and differences in prognosis have emerged between adult and pediatric H3K27-altered DMGs. As well, tumor location can determine differences in the outcome: patients with thalamic and spinal DMG have significantly better survival. Finally, other concomitant molecular alterations in H3K27 gliomas have been shown to influence prognosis. So, when such additional mutations are found, which one should we focus on in order to make the correct clinical decision? Our review of the current literature on pediatric diffuse midline H3K27-altered DMG tries to address such questions. Indeed, H3K27 status has become a fundamental supplement to the histological grading of pediatric gliomas; however, it might not be sufficient alone to exhaustively define the complex biological behavior of DMG in children and might not represent an indication for a unique treatment strategy across all patients, irrespective of age, additional molecular alterations, and tumor location.
Collapse
Affiliation(s)
- Stefano Gabriele Vallero
- Pediatric Oncohematology Division, Regina Margherita Children’s Hospital, Azienda Ospedaliera Universitaria (AOU) Città della Salute e della Scienza, Turin, Italy,*Correspondence: Stefano Gabriele Vallero,
| | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giovanni Morana
- Neuroradiology Unit, Department of Neuroscience, University of Turin, Turin, Italy
| | - Paola Sciortino
- Department of Neuroradiology, Azienda Ospedaliera Universitaria (AOU) Città della Salute e della Scienza, Turin, Italy
| | - Daniele Bertin
- Pediatric Oncohematology Division, Regina Margherita Children’s Hospital, Azienda Ospedaliera Universitaria (AOU) Città della Salute e della Scienza, Turin, Italy
| | - Anna Mussano
- Radiotherapy Unit, Regina Margherita Children’s Hospital, Azienda Ospedaliera Universitaria (AOU) Città della Salute e della Scienza, Turin, Italy
| | - Federica Silvia Ricci
- Child and Adolescent Neuropsychiatry Division, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Paola Peretta
- Pediatric Neurosurgery Division, Regina Margherita Children’s Hospital, Azienda Ospedaliera Universitaria (AOU) Città della Salute e della Scienza, Turin, Italy
| | - Franca Fagioli
- Pediatric Oncohematology Division, Regina Margherita Children’s Hospital, Azienda Ospedaliera Universitaria (AOU) Città della Salute e della Scienza, Turin, Italy,Department of Public Health and Pediatrics, University of Turin, Turin, Italy
| |
Collapse
|
10
|
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: 2.0] [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
|
11
|
Morota N, Deletis V. Brainstem Surgery: Functional Surgical Anatomy with the Use of an Advanced Modern Intraoperative Neurophysiological Procedure. Adv Tech Stand Neurosurg 2023; 48:21-55. [PMID: 37770680 DOI: 10.1007/978-3-031-36785-4_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Intraoperative neurophysiology (ION) in brainstem surgery evolved as brainstem surgery advanced.The original idea of brainstem mapping (BSM) is a neurophysiological procedure to locate cranial nerve motor nuclei (CNMN) on the floor of the fourth ventricle. With the introduction of various skull base approaches to the brainstem, BSM is carried out on any surface of the brainstem to expose the safe entry zone to the intrinsic brainstem lesion. It is the modern concept of BSM, a broader definition of BSM. BSM enables to avoid direct damage to the CNMN when approaching the brainstem through the negative mapping region.The corticobulbar tract (CBT) motor evoked potential (MEP) is another ION procedure in brainstem surgery. It enables monitoring of the functional integrity of the whole cranial motor pathway without interrupting surgical procedures. Combined application of both BSM and CBT-MEP monitoring is indispensable for the functional preservation of the CNMN and their supranuclear innervation during the brainstem surgery.In this paper, the neurophysiological aspect of BSM and the CBT-MEP was fully described. Normal anatomical background of the floor of the fourth ventricle and the detail of the CBT anatomy were demonstrated to better understand their clinical usefulness, limitations, and surgical implications derived from ION procedures. Finally, a future perspective in the role of ION procedures in brainstem surgery was presented. The latest magnetic resonance imaging (MRI) technology can allow surgeons to find an "on the image" safe entry zone to the brainstem. However, the role of BSM and the CBT-MEP monitoring in terms of safe brainstem surgery stays unshakable. Special attention was paid for the recent trend of management in diffuse intrinsic pontine gliomas. A new role of BSM during a stereotactic biopsy was discussed.It is the authors' expectation that the paper enhances the clinical application of a contemporary standard of the ION in brainstem surgery and supports safer brainstem surgery more than ever and in the future.
Collapse
Affiliation(s)
- Nobuhito Morota
- Department of Neurosurgery, Kitasato University Hospital, Sagamihara, Japan
| | - Vedran Deletis
- Department of Neurosurgery, University Hospital, Zagreb, Croatia
| |
Collapse
|
12
|
Biopsy of paediatric brainstem intrinsic tumours: Experience from a Singapore Children’s Hospital. J Clin Neurosci 2022; 106:8-13. [DOI: 10.1016/j.jocn.2022.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/30/2022] [Indexed: 11/15/2022]
|
13
|
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: 7.5] [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
|
14
|
Baquero-Herrera PE, Ardila-Martínez MA, Abdalá-Vargas NJ, Racedo J, Ordóñez-Rubiano EG. Treatment Limitations for Pediatric Diffuse Intrinsic Pontine Gliomas in a Middle-Income Country. J Neurosci Rural Pract 2022; 13:515-520. [PMID: 35945995 PMCID: PMC9357494 DOI: 10.1055/s-0042-1745714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Abstract
Objectives To evaluate the surgical management outcomes in pediatric patients with diffuse intrinsic pontine gliomas (DIPGs) who underwent intended biopsies and partial resections in a middle-income country, highlighting the barriers and challenges of these procedures for further investigation.
Methods A retrospective review of a prospective acquired series of patients who underwent biopsy or resection for DIPG between January 2012 and June 2018 at our institution was performed.
Results A total of 43 patients with posterior fossa tumors were identified. From these, seven pediatric DIPG cases were enrolled. Five were males. The median age was 5 years (range: 1–12 years). Only one patient (14.3%) had a ganglioglioma, while the others presented pilocytic and diffuse astrocytomas. Two (28.6%) patients had an intentional biopsy, and the other five (71.4%) had a partial resection. In the three (28.6%) patients who presented with associated hydrocephalus, the endoscopic third ventriculostomy was performed in the same surgical time. The median preoperative Lansky play-performance scale (LPPS) was 80 (range: 60–100), while the median postoperative LPPS was 23 (range: 7–52).
Conclusion A decrease in overall survival was noted compared with data reported in other series. Multifactorial barriers were discussed including the social, geographic, and economic features that may influence on final outcomes.
Collapse
Affiliation(s)
- Pablo E. Baquero-Herrera
- Department of Neurosurgery, Hospital Infantil Universitario de San José, Fundacion Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
| | - Manuel A. Ardila-Martínez
- Department of Neurosurgery, Hospital Infantil Universitario de San José, Fundacion Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
- Department of Neurosurgery, Hospital de San José – Sociedad de Cirugía de Bogotá, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
| | - Nadín J. Abdalá-Vargas
- Department of Neurosurgery, Hospital Infantil Universitario de San José, Fundacion Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
- Department of Neurosurgery, Hospital de San José – Sociedad de Cirugía de Bogotá, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
| | - Jorge Racedo
- microscopIA Research Group, microscopIA, Bogotá, Colombia
| | - Edgar G. Ordóñez-Rubiano
- Department of Neurosurgery, Hospital de San José – Sociedad de Cirugía de Bogotá, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
- Department of Biomedical Engineering, Universidad de Los Andes, Bogotá, Colombia
- Research Institute, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
| |
Collapse
|
15
|
Damodharan S, Lara-Velazquez M, Williamsen BC, Helgager J, Dey M. Diffuse Intrinsic Pontine Glioma: Molecular Landscape, Evolving Treatment Strategies and Emerging Clinical Trials. J Pers Med 2022; 12:840. [PMID: 35629262 PMCID: PMC9144327 DOI: 10.3390/jpm12050840] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 12/07/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a type of intrinsic brainstem glial tumor that occurs primarily in the pediatric population. DIPG is initially diagnosed based on clinical symptoms and the characteristic location on imaging. Histologically, these tumors are characterized by a heterogenous population of cells with multiple genetic mutations and high infiltrative capacity. The most common mutation seen in this group is a lysine to methionine point mutation seen at position 27 (K27M) within histone 3 (H3). Tumors with the H3 K27M mutation, are considered grade 4 and are now categorized within the H3 K27-altered diffuse midline glioma category by World Health Organization classification. Due to its critical location and aggressive nature, DIPG is resistant to the most eradicative treatment and is universally fatal; however, modern advances in the surgical techniques resulting in safe biopsy of the lesion have significantly improved our understanding of this disease at the molecular level. Genomic analysis has shown several mutations that play a role in the pathophysiology of the disease and can be targeted therapeutically. In this review, we will elaborate on DIPG from general aspects and the evolving molecular landscape. We will also review innovative therapeutic options that have been trialed along with new promising treatments on the horizon.
Collapse
Affiliation(s)
- Sudarshawn Damodharan
- Department of Pediatrics, Division of Pediatric Hematology, Oncology and Bone Marrow Transplant, School of Medicine & Public Health, University of Wisconsin, Madison, WI 53792, USA;
| | - Montserrat Lara-Velazquez
- Department of Neurosurgery, School of Medicine & Public Health, University of Wisconsin, UW Carbone Cancer Center, Madison, WI 53792, USA; (M.L.-V.); (B.C.W.)
| | - Brooke Carmen Williamsen
- Department of Neurosurgery, School of Medicine & Public Health, University of Wisconsin, UW Carbone Cancer Center, Madison, WI 53792, USA; (M.L.-V.); (B.C.W.)
| | - Jeffrey Helgager
- Department of Pathology, School of Medicine & Public Health, University of Wisconsin, UW Carbone Cancer Center, Madison, WI 53792, USA;
| | - Mahua Dey
- Department of Neurosurgery, School of Medicine & Public Health, University of Wisconsin, UW Carbone Cancer Center, Madison, WI 53792, USA; (M.L.-V.); (B.C.W.)
| |
Collapse
|
16
|
Ruttens D, Messiaen J, Ferster A, Piette C, Schifflers S, Van Damme A, van der Werff Ten Bosch J, Verlooy J, Willems L, Jacobs S. Retrospective study of diffuse intrinsic pontine glioma in the Belgian population: a 25 year experience. J Neurooncol 2021; 153:293-301. [PMID: 33939103 DOI: 10.1007/s11060-021-03766-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/23/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Diffuse intrinsic pontine glioma is a rare disease with a high mortality. Our primary aim was to determine the incidence of this disease in Belgium. Secondly, we wanted to compare the treatment approach of Belgian pediatric oncology centres, to investigate possibilities for improvement. METHODS We retrospectively collected and analysed data on DIPG-patients diagnosed between 1994 and 2018 and recorded in the Belgian Cancer Registry. We included patients ≤ 18 years who were followed in one of the eight Belgian pediatric oncology centres. RESULTS We included 100 patients. Files were complete in 87 patients. We observed an increase in diagnoses with an incidence of 3.1 per 1,000,000 persons (aged 0-≤ 18) per year over the last 5 years compared to an overall incidence of 1.8. Biopsy was performed at diagnosis in 51.7% of patients. In one fifth this was study-related. Mutation analysis was known in eight patients, of which six showed the H3 K27M-mutation. 58.8% of patients received chemotherapy, without a significant survival benefit. 12.6% of patients were included in a clinical trial. Biopsy rate and the use of chemotherapy differed widely between centres. Mean OS and PFS were 10.49 and 4.87 months respectively. We observed an improved survival over time. CONCLUSIONS Over the past 25 years, we observed an increase of new DIPG-diagnoses. Outcome in our cohort is comparable with literature findings. We demonstrate an important heterogeneity in treatment approach between different centres and limited inclusion in clinical trials. Therefore, collaboration between centres and inclusion of patients in clinical trials is much needed.
Collapse
Affiliation(s)
- Dries Ruttens
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium.
| | - Julie Messiaen
- Department of Pediatric Hematology-Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Alina Ferster
- Department of Pediatric Hematology-Oncology, Queen Fabiola Children's University Hospital, Brussels, Belgium
| | - Caroline Piette
- Department of Pediatrics, Division of Hematology-Oncology, CHU of Liège, Liège, Belgium
| | - Stefan Schifflers
- Department of Pediatric Hematology-Oncology, CHC MontLégia, Liège, Belgium
| | - An Van Damme
- Department of Pediatric Hematology-Oncology, Saint-Luc University Hospital, Brussels, Belgium
| | | | - Joris Verlooy
- Department of Pediatric Hematology-Oncology, University Hospital of Antwerp, Edegem, Belgium
| | - Leen Willems
- Department of Pediatric Hematology-Oncology, University Hospital Ghent, Ghent, Belgium
| | - Sandra Jacobs
- Department of Pediatric Hematology-Oncology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
17
|
Zhu X, Lazow MA, Schafer A, Bartlett A, Senthil Kumar S, Mishra DK, Dexheimer P, DeWire M, Fuller C, Leach JL, Fouladi M, Drissi R. A pilot radiogenomic study of DIPG reveals distinct subgroups with unique clinical trajectories and therapeutic targets. Acta Neuropathol Commun 2021; 9:14. [PMID: 33431066 PMCID: PMC7798248 DOI: 10.1186/s40478-020-01107-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/14/2020] [Indexed: 01/03/2023] Open
Abstract
An adequate understanding of the relationships between radiographic and genomic features in diffuse intrinsic pontine glioma (DIPG) is essential, especially in the absence of universal biopsy, to further characterize the molecular heterogeneity of this disease and determine which patients are most likely to respond to biologically-driven therapies. Here, a radiogenomics analytic approach was applied to a cohort of 28 patients with DIPG. Tumor size and imaging characteristics from all available serial MRIs were evaluated by a neuro-radiologist, and patients were divided into three radiographic response groups (partial response [PR], stable disease [SD], progressive disease [PD]) based on MRI within 2 months of radiotherapy (RT) completion. Whole genome and RNA sequencing were performed on autopsy tumor specimens. We report several key, therapeutically-relevant findings: (1) Certain radiologic features on first and subsequent post-RT MRIs are associated with worse overall survival, including PD following irradiation as well as present, new, and/or increasing peripheral ring enhancement, necrosis, and diffusion restriction. (2) Upregulation of EMT-related genes and distant tumor spread at autopsy are observed in a subset of DIPG patients who exhibit poorer radiographic response to irradiation and/or higher likelihood of harboring H3F3A mutations, suggesting possible benefit of upfront craniospinal irradiation. (3) Additional genetic aberrations were identified, including DYNC1LI1 mutations in a subgroup of patients with PR on post-RT MRI; further investigation into potential roles in DIPG tumorigenesis and/or treatment sensitivity is necessary. (4) Whereas most DIPG tumors have an immunologically “cold” microenvironment, there appears to be a subset which harbor a more inflammatory genomic profile and/or higher mutational burden, with a trend toward improved overall survival and more favorable radiographic response to irradiation, in whom immunotherapy should be considered. This study has begun elucidating relationships between post-RT radiographic response with DIPG molecular profiles, revealing radiogenomically distinct subgroups with unique clinical trajectories and therapeutic targets.
Collapse
|
18
|
Biery MC, Noll A, Myers C, Morris SM, Winter CA, Pakiam F, Cole BL, Browd SR, Olson JM, Vitanza NA. A Protocol for the Generation of Treatment-naïve Biopsy-derived Diffuse Intrinsic Pontine Glioma and Diffuse Midline Glioma Models. JOURNAL OF EXPERIMENTAL NEUROLOGY 2020. [PMID: 33768215 PMCID: PMC7990285 DOI: 10.33696//neurol.1.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a universally fatal tumor of the brainstem, most commonly affecting young children. Due to its location, surgical resection is not achievable, but consideration of a biopsy has become standard practice at children’s hospitals with the appropriate neurosurgical expertise. While the decision to obtain a biopsy should be directed by the presence of atypical radiographic features that call the diagnosis of DIPG into question or the requirement of biopsy tissue for clinical trial enrollment, once this precious tissue is available its use for research should be considered. The majority of DIPG and diffuse midline glioma, H3 K27M-mutant (DMG) models are autopsy-derived or genetically-engineered, each of which has limitations for translational studies, so the use of biopsy tissue for laboratory model development provides an opportunity to create unique model systems. Here, we present a detailed laboratory protocol for the generation of treatment-naïve biopsy-derived DIPG/DMG models.
Collapse
Affiliation(s)
- Matt C Biery
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Alyssa Noll
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Molecular and Cellular Biology Graduate Program and Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Carrie Myers
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Conrad A Winter
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Laboratories, Seattle Children's Hospital, Seattle, WA, USA
| | - Fiona Pakiam
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bonnie L Cole
- Department of Laboratories, Seattle Children's Hospital, Seattle, WA, USA.,Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Samuel R Browd
- Division of Neurosurgery, Department of Neurological Surgery, University of Washington, Seattle Children's Hospital, Seattle, WA, USA
| | - James M Olson
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Division of Hematology/Oncology, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Nicholas A Vitanza
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Division of Hematology/Oncology, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| |
Collapse
|
19
|
Bray DP, Buster BE, Boucher AB, Wrubel DM. Letter: High Prevalence of Developmental Venous Anomaly in Diffuse Intrinsic Pontine Gliomas: A Pediatric Control Study. Neurosurgery 2020; 87:E525-E526. [DOI: 10.1093/neuros/nyaa285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
20
|
Joud A, Stella I, Klein O. Diffuse infiltrative pontine glioma biopsy in children with neuronavigation, frameless procedure: A single center experience of 10 cases. Neurochirurgie 2020; 66:345-348. [PMID: 32860812 DOI: 10.1016/j.neuchi.2020.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/15/2020] [Accepted: 05/17/2020] [Indexed: 11/18/2022]
Abstract
INTRODUCTION This study presented pediatric DIPG 's biopsy with frameless Neuronavigation. PATIENTS AND METHODS We report our experience about 10 patients who had Diffuse Intrinsic Pontine Glioma between 2014 and 2018. All patients were biopsied with BrainLab Varioguide Neuronavigation®. We always used fusion between specific CT Scan and MRI to selected target, made planning and biopsies. All patients were included in BIOMEDE after scientific and ethic discussions. We always selected a trans-cerebellar trajectory and made same procedure (lot of biopsies at one level). All patients have MRI at J1 to verify site of biopsy and to eliminate complication. RESULTS The average age was 8.1 years. Symptoms were common with principally headaches and nystagmus. All biopsies were contributive for histopathological diagnosis and establish molecular profile for molecular study. We have no definitive morbidity and procedure duration was 93minutes in average. All MRI didn't showed intracranial complication after procedure and showed great precision of biopsy compared with the selected target. DISCUSSION We reviewed the literature and compare our results with series of DIPG biopsies using stereotactic frame or robotic assisted frameless. It was a safe, accuracy and easiness procedure. We always have histopathological and molecular result to proceed next step of treatment. This modality is an alternative possibility to biopsy very young patients with low morbidity.
Collapse
Affiliation(s)
- A Joud
- Department of Pediatric Neurosurgery, Nancy University Hospital, Hôpital d'Enfants, University of Lorraine, rue du Morvan, 54511 Vandoeuvre-les-Nancy cedex, France.
| | - I Stella
- Department of Pediatric Neurosurgery, Nancy University Hospital, Hôpital d'Enfants, University of Lorraine, rue du Morvan, 54511 Vandoeuvre-les-Nancy cedex, France
| | - O Klein
- Department of Pediatric Neurosurgery, Nancy University Hospital, Hôpital d'Enfants, University of Lorraine, rue du Morvan, 54511 Vandoeuvre-les-Nancy cedex, France
| |
Collapse
|
21
|
Variations in attitudes towards stereotactic biopsy of adult diffuse midline glioma patients: a survey of members of the AANS/CNS Tumor Section. J Neurooncol 2020; 149:161-170. [PMID: 32705457 PMCID: PMC7452882 DOI: 10.1007/s11060-020-03585-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/11/2020] [Indexed: 02/01/2023]
Abstract
Purpose Diffuse midline gliomas are rare midline CNS malignancies that primarily affect children but can also affect adults. While radiation is standard treatment, prognosis remains fatal. Furthermore, due to its sensitive anatomic location, many physicians have been reluctant to perform biopsies without potential for improved prognosis. However, recent advancements in molecular-targeted therapeutics have encouraged greater tissue sampling. While the literature reflects this progress, the landscape of how clinicians actually manage these patients remains unclear. Our goal was to assess the attitudes of current practicing neurosurgical oncologists towards management of adult diffuse midline gliomas, reasons behind their practices, and factors that might influence these practices. Methods We created and distributed a survey with 16 multiple choice and open-ended questions to members of the Tumor Section of the Congress of Neurological Surgeons. Results A total of 81 physicians responded to the survey. Although time since training and volume of glioma patients did not significantly affect the decision to consider clinical trials or to offer biopsy, those that operated on fewer gliomas (< 25/year) were more likely to cite surgical morbidity as the primary reason not to biopsy these midline locations. Further, surgeons with access to more advanced molecular testing were significantly more likely to consider clinical trial eligibility when offering biopsies. Conclusion Factors that affect the management of diffuse midline gliomas and the role of biopsy are relatively uniform across the field, however, there were a few notable differences that reflect the changes within the neuro-oncology field in response to clinical trials. Electronic supplementary material The online version of this article (10.1007/s11060-020-03585-7) contains supplementary material, which is available to authorized users.
Collapse
|
22
|
Convection Enhanced Delivery for Diffuse Intrinsic Pontine Glioma: Review of a Single Institution Experience. Pharmaceutics 2020; 12:pharmaceutics12070660. [PMID: 32674336 PMCID: PMC7407112 DOI: 10.3390/pharmaceutics12070660] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 01/24/2023] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are a pontine subtype of diffuse midline gliomas (DMGs), primary central nervous system (CNS) tumors of childhood that carry a terrible prognosis. Because of the highly infiltrative growth pattern and the anatomical position, cytoreductive surgery is not an option. An initial response to radiation therapy is invariably followed by recurrence; mortality occurs approximately 11 months after diagnosis. The development of novel therapeutics with great preclinical promise has been hindered by the tightly regulated blood-brain barrier (BBB), which segregates the tumor comportment from the systemic circulation. One possible solution to this obstacle is the use of convection enhanced delivery (CED), a local delivery strategy that bypasses the BBB by direct infusion into the tumor through a small caliber cannula. We have recently shown CED to be safe in children with DIPG (NCT01502917). In this review, we discuss our experience with CED, its advantages, and technical advancements that are occurring in the field. We also highlight hurdles that will likely need to be overcome in demonstrating clinical benefit with this therapeutic strategy.
Collapse
|
23
|
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: 6] [Impact Index Per Article: 1.5] [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
|
24
|
Rashed WM, Maher E, Adel M, Saber O, Zaghloul MS. Pediatric diffuse intrinsic pontine glioma: where do we stand? Cancer Metastasis Rev 2020; 38:759-770. [PMID: 31802357 DOI: 10.1007/s10555-019-09824-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pediatric diffuse intrinsic pontine glioma (DIPG) represents approximately 20% of all pediatric CNS tumors. However, disease outcomes are dismal with a median survival of less than 1 year and a 2-year overall survival rate of less than 10%. Despite extensive efforts to improve survival outcomes, progress towards clinical improvement has been largely stagnant throughout the last 4 decades. Focal radiotherapy remains the standard of care with no promising single-agent alternatives and no evidence for improvement with the addition of a long list of systemic therapies. A better understanding of the biology of DIPG, though not easy due to obstacles in obtaining pathological material to study, is promising for the development of specific individualized treatment for this fatal disease. Recent studies have found epigenetic mutations to be successful predictors and prognostic factors for developing future management policies. The aim of this review is to give a global overview about the epidemiology, diagnosis, and treatment of DIPG. We further examine the controversial biopsy and autopsy issue that is unique to DIPG and assess the subsequent impact this issue has on the research efforts and clinical management of DIPG.
Collapse
Affiliation(s)
- Wafaa M Rashed
- Research Department, Children's Cancer Hospital Egypt, Cairo, 57357, Egypt.
| | - Eslam Maher
- Research Department, Children's Cancer Hospital Egypt, Cairo, 57357, Egypt
| | - Mohamed Adel
- Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Ossama Saber
- Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Mohamed Saad Zaghloul
- Radiotherapy Department, National Cancer Institute, Cairo University & Children's Cancer Hospital, Cairo, 57357, Egypt.
| |
Collapse
|
25
|
Diffuse intrinsic pontine gliomas: Diagnostic approach and treatment strategies. J Clin Neurosci 2019; 72:15-19. [PMID: 31870682 DOI: 10.1016/j.jocn.2019.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 12/01/2019] [Indexed: 11/23/2022]
Abstract
Diffuse intrinsic pontine gliomas (DIPG) are high grade gliomas of the brainstem with fatal outcomes. Radiation is known to be partially effective to control the immediate flare but relapse is frequent. There has been ongoing research to study the role of molecular subgroups and identification of specific targets but this is not possible with histopathological diagnosis alone. The authors' objective is to highlight the need for and discuss ongoing molecular research. There is an inherent need for the availability of tumor tissue to be able to conduct research studies. The authors advocate the use of neuronavigation assisted stereotactic technique for tumor biopsy. The technique is feasible with a predefined surgical trajectory. After obtaining tissue diagnosis further work can be performed to isolate and identify histone protein genetic mutations and methylation changes responsible for DIPG molecular subgrouping. Moreover, convection enhanced delivery of therapeutic agents is being developed for better instillation of future drug agents. Despite identification of genetic/epigenetic mutations, growth factors, receptors, and tissue biomarkers, the oncogenesis of DIPG remains elusive. The authors' effort to provide a comprehensive review on DIPG to better understand the disease, need for tissue diagnosis, described surgical technique, and need for pre-clinical and clinical future research is novel.
Collapse
|
26
|
Diffuse Intrinsic Pontine Glioma: From Diagnosis to Next-Generation Clinical Trials. Curr Treat Options Neurol 2019; 21:37. [PMID: 31290035 DOI: 10.1007/s11940-019-0577-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE OF REVIEW This review of diffuse intrinsic pontine glioma (DIPG) provides clinical background, a systematic approach to diagnosis and initial care, and synthesizes historical, modern, and future directions for treatment. We present evidence supporting neurosurgical biopsy, early palliative care involvement, limitation of glucocorticoid use, and the leveraging of preclinical DIPG models as a pipeline to next-generation clinical trials. RECENT FINDINGS New molecular understanding of pediatric high-grade gliomas has led to the reclassification of DIPG as one member of a family of diffuse gliomas occurring in the midline of the central nervous system that exhibit pathognomonic mutations in genes encoding histone 3 (H3 K27M). DIPG remains a clinically relevant term, though diagnostically the 80% of DIPG cases that exhibit the H3 K27M mutation have been reclassified as diffuse midline glioma, H3 K27M-mutant. Re-irradiation has been shown to be well-tolerated and of potential benefit. Epigenetic targeting of transcriptional dependencies in preclinical models is fueling molecularly targeted clinical trials. Chimeric antigen receptor T cell immunotherapy has also demonstrated efficacy in preclinical models and provides a promising new clinical strategy. DIPG is a universally fatal, epigenetically driven tumor of the pons that is considered part of a broader class of diffuse midline gliomas sharing H3 K27M mutations. Radiation remains the standard of care, single-agent temozolomide is not recommended, and glucocorticoids should be used only sparingly. A rapid evolution of understanding in the chromatin, signaling, and immunological biology of DIPG may soon result in clinical breakthroughs.
Collapse
|
27
|
Mueller S, Jain P, Liang WS, Kilburn L, Kline C, Gupta N, Panditharatna E, Magge SN, Zhang B, Zhu Y, Crawford JR, Banerjee A, Nazemi K, Packer RJ, Petritsch CK, Truffaux N, Roos A, Nasser S, Phillips JJ, Solomon D, Molinaro A, Waanders AJ, Byron SA, Berens ME, Kuhn J, Nazarian J, Prados M, Resnick AC. A pilot precision medicine trial for children with diffuse intrinsic pontine glioma-PNOC003: A report from the Pacific Pediatric Neuro-Oncology Consortium. Int J Cancer 2019; 145:1889-1901. [PMID: 30861105 DOI: 10.1002/ijc.32258] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/21/2019] [Accepted: 02/15/2019] [Indexed: 12/13/2022]
Abstract
This clinical trial evaluated whether whole exome sequencing (WES) and RNA sequencing (RNAseq) of paired normal and tumor tissues could be incorporated into a personalized treatment plan for newly diagnosed patients (<25 years of age) with diffuse intrinsic pontine glioma (DIPG). Additionally, whole genome sequencing (WGS) was compared to WES to determine if WGS would further inform treatment decisions, and whether circulating tumor DNA (ctDNA) could detect the H3K27M mutation to allow assessment of therapy response. Patients were selected across three Pacific Pediatric Neuro-Oncology Consortium member institutions between September 2014 and January 2016. WES and RNAseq were performed at diagnosis and recurrence when possible in a CLIA-certified laboratory. Patient-derived cell line development was attempted for each subject. Collection of blood for ctDNA was done prior to treatment and with each MRI. A specialized tumor board generated a treatment recommendation including up to four FDA-approved agents based upon the genomic alterations detected. A treatment plan was successfully issued within 21 business days from tissue collection for all 15 subjects, with 14 of the 15 subjects fulfilling the feasibility criteria. WGS results did not significantly deviate from WES-based therapy recommendations; however, WGS data provided further insight into tumor evolution and fidelity of patient-derived cell models. Detection of the H3F3A or HIST1H3B K27M (H3K27M) mutation using ctDNA was successful in 92% of H3K27M mutant cases. A personalized treatment recommendation for DIPG can be rendered within a multicenter setting using comprehensive next-generation sequencing technology in a clinically relevant timeframe.
Collapse
Affiliation(s)
- Sabine Mueller
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.,Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.,Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Payal Jain
- Center for Data-Driven Discovery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Winnie S Liang
- Translational Genomic Research Institute (TGEN), Phoenix, AZ, USA
| | - Lindsay Kilburn
- Center for Cancer and Blood Disorders, Children's National Health System, Washington, DC, USA.,Brain Tumor Institute, Children's National Health System, Washington, DC, USA
| | - Cassie Kline
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.,Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Nalin Gupta
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.,Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Eshini Panditharatna
- Brain Tumor Institute, Children's National Health System, Washington, DC, USA.,Research Center for Genetic Medicine, Children's National Health System, Washington, DC, USA
| | - Suresh N Magge
- Division of Neurosurgery, Children's National Health System, Washington, DC, USA
| | - Bo Zhang
- Center for Data-Driven Discovery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yuankun Zhu
- Center for Data-Driven Discovery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Anu Banerjee
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.,Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Kellie Nazemi
- Doernbecher Children's Hospital, Oregon Health & Science University, Portland, OR, USA
| | - Roger J Packer
- Brain Tumor Institute, Children's National Health System, Washington, DC, USA
| | - Claudia K Petritsch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Nathalene Truffaux
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Alison Roos
- Translational Genomic Research Institute (TGEN), Phoenix, AZ, USA
| | - Sara Nasser
- Translational Genomic Research Institute (TGEN), Phoenix, AZ, USA
| | - Joanna J Phillips
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.,Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - David Solomon
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Annette Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Angela J Waanders
- Center for Data-Driven Discovery, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Children's Brain Tumor Tissue Consortium, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sara A Byron
- Translational Genomic Research Institute (TGEN), Phoenix, AZ, USA
| | - Michael E Berens
- Translational Genomic Research Institute (TGEN), Phoenix, AZ, USA
| | - John Kuhn
- College of Pharmacy, University of Texas Health Science Center, San Antonio, TX, USA
| | - Javad Nazarian
- Center for Data-Driven Discovery, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Center for Cancer and Blood Disorders, Children's National Health System, Washington, DC, USA.,Brain Tumor Institute, Children's National Health System, Washington, DC, USA.,Research Center for Genetic Medicine, Children's National Health System, Washington, DC, USA
| | - Michael Prados
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Adam C Resnick
- Center for Data-Driven Discovery, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Children's Brain Tumor Tissue Consortium, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
28
|
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: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
29
|
Tsoli M, Shen H, Mayoh C, Franshaw L, Ehteda A, Upton D, Carvalho D, Vinci M, Meel MH, van Vuurden D, Plessier A, Castel D, Drissi R, Farrell M, Cryan J, Crimmins D, Caird J, Pears J, Francis S, Ludlow LEA, Carai A, Mastronuzzi A, Liu B, Hansford J, Gottardo N, Hassall T, Kirby M, Fouladi M, Hawkins C, Monje M, Grill J, Jones C, Hulleman E, Ziegler DS. International experience in the development of patient-derived xenograft models of diffuse intrinsic pontine glioma. J Neurooncol 2019; 141:253-263. [PMID: 30446898 DOI: 10.1007/s11060-018-03038-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/24/2018] [Indexed: 10/27/2022]
Abstract
PURPOSE Diffuse intrinsic pontine glioma is the most aggressive form of high grade glioma in children with no effective therapies. There have been no improvements in survival in part due poor understanding of underlying biology, and lack of representative in vitro and in vivo models. Recently, it has been found feasible to use both biopsy and autopsy tumors to generate cultures and xenograft models. METHODS To further model development, we evaluated the collective international experience from 8 collaborating centers to develop DIPG pre-clinical models from patient-derived autopsies and biopsies. Univariate and multivariate analysis was performed to determine key factors associated with the success of in vitro and in vivo PDX development. RESULTS In vitro cultures were successfully established from 57% of samples (84.2% of biopsies and 38.2% of autopsies). Samples transferred in DMEM media were more likely to establish successful culture than those transported in Hibernate A. In vitro cultures were more successful from biopsies (84.2%) compared with autopsies (38.2%) and as monolayer on laminin-coated plates than as neurospheres. Primary cultures successfully established from autopsy samples were more likely to engraft in animal models than cultures established from biopsies (86.7% vs. 47.4%). Collectively, tumor engraftment was more successful when DIPG samples were directly implanted in mice (68%), rather than after culturing (40.7%). CONCLUSION This multi-center study provides valuable information on the success rate of establishing patient-derived pre-clinical models of DIPG. The results can lead to further optimization of DIPG model development and ultimately assist in the investigation of new therapies for this aggressive pediatric brain tumor.
Collapse
Affiliation(s)
- Maria Tsoli
- Children's Cancer Institute, Randwick, NSW, 2031, Australia
| | - Han Shen
- Children's Cancer Institute, Randwick, NSW, 2031, Australia
| | - Chelsea Mayoh
- Children's Cancer Institute, Randwick, NSW, 2031, Australia
| | - Laura Franshaw
- Children's Cancer Institute, Randwick, NSW, 2031, Australia
| | - Anahid Ehteda
- Children's Cancer Institute, Randwick, NSW, 2031, Australia
| | - Danielle Upton
- Children's Cancer Institute, Randwick, NSW, 2031, Australia
| | - Diana Carvalho
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Maria Vinci
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Michael H Meel
- Department of Pediatric Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Dannis van Vuurden
- Department of Pediatric Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Alexander Plessier
- Unite Mixte de Recherche 8203 du Centre National de la Recherche Scientifique (CNRS) et Departement de Cancerologie de l'Enfant et de l'Adolescent, Gustave Roussy et Universite Paris-Saclay, Villejuif, France
| | - David Castel
- Unite Mixte de Recherche 8203 du Centre National de la Recherche Scientifique (CNRS) et Departement de Cancerologie de l'Enfant et de l'Adolescent, Gustave Roussy et Universite Paris-Saclay, Villejuif, France
| | - Rachid Drissi
- Brain Tumor Center, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, USA
| | - Michael Farrell
- Histopathology Department, Beaumont Hospital, Dublin, Ireland
| | - Jane Cryan
- Histopathology Department, Beaumont Hospital, Dublin, Ireland
| | - Darach Crimmins
- Department of Neurosurgery, Temple Street Children's University Hospital, Dublin, Ireland
| | - John Caird
- Department of Neurosurgery, Temple Street Children's University Hospital, Dublin, Ireland
| | - Jane Pears
- Our Lady's Children's Hospital, Dublin, Ireland
| | - Stephanie Francis
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, 2052, Australia
| | - Louise E A Ludlow
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, 3052, Australia
| | - Andrea Carai
- Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, Rome, Italy
| | - Angela Mastronuzzi
- Neuro-Oncology Unit, Department of Hemato-Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy
| | - Bing Liu
- Children's Cancer Institute, Randwick, NSW, 2031, Australia
| | - Jordan Hansford
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, 3052, Australia
| | - Nick Gottardo
- Department of Oncology, Princess Margaret Hospital, Perth, WA, Australia
| | - Tim Hassall
- Lady Cilento Children's Hospital, Brisbane, Australia
| | - Maria Kirby
- Department of Haematology-Oncology, Women's and Children's Hospital, Adelaide, SA, Australia
| | - Maryam Fouladi
- Brain Tumor Center, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, USA
| | - Cynthia Hawkins
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Michelle Monje
- Stanford University and Lucile Packard Children's Hospital, Palo Alto, CA, USA
| | - Jacques Grill
- Unite Mixte de Recherche 8203 du Centre National de la Recherche Scientifique (CNRS) et Departement de Cancerologie de l'Enfant et de l'Adolescent, Gustave Roussy et Universite Paris-Saclay, Villejuif, France
| | - Chris Jones
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Esther Hulleman
- Unite Mixte de Recherche 8203 du Centre National de la Recherche Scientifique (CNRS) et Departement de Cancerologie de l'Enfant et de l'Adolescent, Gustave Roussy et Universite Paris-Saclay, Villejuif, France
| | - David S Ziegler
- Children's Cancer Institute, Randwick, NSW, 2031, Australia.
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, 2052, Australia.
| |
Collapse
|
30
|
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: 12.0] [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
|
31
|
Lin GL, Nagaraja S, Filbin MG, Suvà ML, Vogel H, Monje M. Non-inflammatory tumor microenvironment of diffuse intrinsic pontine glioma. Acta Neuropathol Commun 2018; 6:51. [PMID: 29954445 PMCID: PMC6022714 DOI: 10.1186/s40478-018-0553-x] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/07/2018] [Indexed: 01/05/2023] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a universally fatal malignancy of the childhood central nervous system, with a median overall survival of 9-11 months. We have previously shown that primary DIPG tissue contains numerous tumor-associated macrophages, and substantial work has demonstrated a significant pathological role for adult glioma-associated macrophages. However, work over the past decade has highlighted many molecular and genomic differences between pediatric and adult high-grade gliomas. Thus, we directly compared inflammatory characteristics of DIPG and adult glioblastoma (GBM). We found that the leukocyte (CD45+) compartment in primary DIPG tissue samples is predominantly composed of CD11b + macrophages, with very few CD3+ T-lymphocytes. In contrast, T-lymphocytes are more abundant in adult GBM tissue samples. RNA sequencing of macrophages isolated from primary tumor samples revealed that DIPG- and adult GBM-associated macrophages both express gene programs related to ECM remodeling and angiogenesis, but DIPG-associated macrophages express substantially fewer inflammatory factors than their adult GBM counterparts. Examining the secretome of glioma cells, we found that patient-derived DIPG cell cultures secrete markedly fewer cytokines and chemokines than patient-derived adult GBM cultures. Concordantly, bulk and single-cell RNA sequencing data indicates low to absent expression of chemokines and cytokines in DIPG. Together, these observations suggest that the inflammatory milieu of the DIPG tumor microenvironment is fundamentally different than adult GBM. The low intrinsic inflammatory signature of DIPG cells may contribute to the lack of lymphocytes and non-inflammatory phenotype of DIPG-associated microglia/macrophages. Understanding the glioma subtype-specific inflammatory milieu may inform the design and application of immunotherapy-based treatments.
Collapse
Affiliation(s)
- Grant L Lin
- Department of Neurology, Stanford University, Stanford, CA, 94305, USA
| | - Surya Nagaraja
- Department of Neurology, Stanford University, Stanford, CA, 94305, USA
| | - Mariella G Filbin
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA, USA
| | - Mario L Suvà
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of Harvard and Massachussetts Institute of Technology (MIT), Cambridge, MA, 02142, USA
| | - Hannes Vogel
- Department of Neurology, Stanford University, Stanford, CA, 94305, USA
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | - Michelle Monje
- Department of Neurology, Stanford University, Stanford, CA, 94305, USA.
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA.
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
32
|
Clymer J, Kieran MW. The Integration of Biology Into the Treatment of Diffuse Intrinsic Pontine Glioma: A Review of the North American Clinical Trial Perspective. Front Oncol 2018; 8:169. [PMID: 29868485 PMCID: PMC5968382 DOI: 10.3389/fonc.2018.00169] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/01/2018] [Indexed: 11/25/2022] Open
Abstract
Dramatic advances in the molecular analysis of diffuse intrinsic pontine glioma have occurred over the last decade and resulted in the identification of potential therapeutic targets. In spite of these advances, no significant improvement in the outcome has been achieved and median survival remains approximately 10 months. An understanding of the approaches that have been taken to date, why they failed, and how that information can lead the field forward is critical if we are to change the status quo. In this review, we will discuss the clinical trial landscape in North America with an overview of historical approaches that failed and what might account for this failure. We will then provide a discussion of how our understanding of the genotype of this disease has led to the development of a number of trials targeting the mutations and epigenome of diffuse intrinsic pontine gliomas and the issues related to these trials. Similarly, the introduction of methodologies to address penetration across the blood–brain barrier will be considered in the context of both targeted approaches, epigenetic modification, and immune surveillance of these tumors. The comprehensive analysis of these data, generated through cooperative groups, collaborative clinical trials, and pilot studies in North America will be the focus of the IVth Memorial Alicia Pueyo international symposium in Barcelona on March 12th, 2018 and will be compared and contrasted with a similar comprehensive analysis of the European data with the goal of bringing all of these data together to develop a uniform platform on which new rational trials can be based.
Collapse
Affiliation(s)
- Jessica Clymer
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA, United States
| | - Mark W Kieran
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| |
Collapse
|
33
|
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: 31] [Impact Index Per Article: 5.2] [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
|
34
|
Quick-Weller J, Tritt S, Behmanesh B, Mittelbronn M, Spyrantis A, Dinc N, Weise L, Seifert V, Marquardt G, Freiman TM. Biopsies of pediatric brainstem lesions display low morbidity but strong impact on further treatment decisions. J Clin Neurosci 2017; 44:254-259. [PMID: 28711290 DOI: 10.1016/j.jocn.2017.06.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/15/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The course of malignant brain stem gliomas in childhood is rarely positive. Because of limited therapeutic options and potentially hazardous biopsies oncologist often relay on MRI diagnoses only for further therapy decisions. In this study we show that brain stem biopsies display a low morbidity rate and neuropathological assessment has a considerable impact on further treatment decision. METHODS Within 18-months five children with brainstem symptoms and the radiological diagnosis of a malignant brainstem glioma, were identified. From this time point it was possible to analyze all samples with the 450K methylome analysis. Other neuropathological techniques included classical histology with immunohistochemistry. Surgery was performed as biopsy, either microsurgical, frame-guided (Leksell), robot-assisted (ROSA) or navigated (BrainLab, two children). RESULTS Mean age of the children was 7.5years (range: newborn to 12years). There was no biopsy-related morbidity or mortality. The mean number of taken samples was 12 (range: 1-25). Histologic diagnosis could be established in all children, however, 450K methylome diagnosis was positive in only two out of five patients. CONCLUSION Despite the technically difficult biopsies, all specimens were sufficient for immunohistochemical diagnosis, however, 450K methylome analysis could only be better established where multiple small samples were taken, instead of few larger ones. Based on the preoperative radiological diagnosis suggesting malignant brainstem glioma, all children would have been treated with combined radiation and temozolomid chemotherapy. Nevertheless, due to the availability of histology and molecular diagnostics, individualized therapy could be performed, preventing in two out of five children from unnecessary radiation and chemotherapy.
Collapse
Affiliation(s)
| | - Stephanie Tritt
- Institute for Neuroradiology, University Hospital Frankfurt, Germany
| | - Bedjan Behmanesh
- Department of Neurosurgery, University Hospital Frankfurt, Germany
| | - Michel Mittelbronn
- Luxembourg Centre of Neuropathology (LCNP), Luxembourg; Laboratoire National de Santè, Department of Pathology, Dudelange, Luxembourg; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg; Neurological Institute (Edinger Institute), Goethe University Frankfurt, Germany
| | - Andrea Spyrantis
- Department of Neurosurgery, University Hospital Frankfurt, Germany
| | - Nazife Dinc
- Department of Neurosurgery, University Hospital Frankfurt, Germany
| | - Lutz Weise
- Department of Neurosurgery, University Hospital Frankfurt, Germany; Division of Neurosurgery, Department of Surgery, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Volker Seifert
- Department of Neurosurgery, University Hospital Frankfurt, Germany
| | | | - Thomas M Freiman
- Department of Neurosurgery, University Hospital Frankfurt, Germany
| |
Collapse
|
35
|
Chen F, Li Z, Weng C, Li P, Tu L, Chen L, Xie W, Li L. Progressive multifocal exophytic pontine glioblastoma: a case report with literature review. CHINESE JOURNAL OF CANCER 2017; 36:34. [PMID: 28347331 PMCID: PMC5369214 DOI: 10.1186/s40880-017-0201-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 01/03/2017] [Indexed: 11/10/2022]
Abstract
Multifocal pontine glioblastoma exhibiting an exophytic growth pattern in the cerebello-pontine angle (CPA) is rare. We present a case of a 5-year-old girl with consecutive neurological imaging and other clinical findings indicating progressive multifocal exophytic pontine glioblastoma. Three lesions were reported, of which two were initially presented, and one was developed 2 months later. One lesion demonstrated a progressing exophytic extension in the cistern of the left side of the CPA. The other two lesions were located and confined within the pons. Initial magnetic resonance imaging and positron emission tomography–computed tomography indicated low-grade glioma or inflammatory disease. However, 2 and 3 months later, subsequent magnetic resonance spectroscopy (MRS) displayed elevated choline and depressed N-acetyl aspartate peaks compared with the peaks on the initial MRS, indicating a high-grade glioma. Subtotal resection was performed for the CPA lesion. Histopathologic examination showed discrepant features of different parts of the CPA lesion. The patient received no further chemotherapy or radiotherapy and died 2 months after surgery. The multifocal and exophytic features of this case and the heterogeneous manifestations on neurological images were rare and confusing for both diagnosis and surgical decision-making. Our case report may contribute knowledge and helpful guidance for other medical doctors.
Collapse
Affiliation(s)
- Fanfan Chen
- Neurosurgery Department, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong, P. R. China
| | - Zongyang Li
- Neurosurgery Department, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, 518000, Guangdong, P. R. China
| | - Chengyin Weng
- Oncology Department, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong, P. R. China
| | - Peng Li
- Neurosurgery Department, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong, P. R. China
| | - Lanbo Tu
- Neurosurgery Department, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong, P. R. China
| | - Lei Chen
- Neurosurgery Department, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, 518000, Guangdong, P. R. China
| | - Wei Xie
- Neurosurgery Department, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong, P. R. China
| | - Ling Li
- Record Department, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong, P. R. China.
| |
Collapse
|
36
|
Abstract
Diffuse Intrinsic Pontine Glioma (DIPG) is a childhood brainstem tumor that carries a universally fatal prognosis. Because surgical resection is not a viable treatment strategy and biopsy is not routinely performed, the availability of patient samples for research is limited. Consequently, efforts to study this disease have been challenged by a paucity of faithful disease models. To address this need, we describe here a protocol for the rapid processing of post-mortem autopsy tissue samples in order to generate durable patient-derived cell culture models that can be used in in vitro assays or in vivo orthotopic xenograft experiments. These models can be used to screen for potential drug targets and to study fundamental pathobiological processes within DIPG. This protocol can further be extended to analyze and isolate tumor and microenvironmental cells using Fluorescence-activated Cell Sorting (FACS), which enables subsequent analysis of gene expression, protein expression, or epigenetic modifications of DNA at the bulk cell or single cell level. Finally, this protocol can also be adapted to generate patient-derived cultures for other central nervous system tumors.
Collapse
Affiliation(s)
- Grant L Lin
- Graduate Program in Neuroscience, Department of Neurology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| | - Michelle Monje
- Departments of Neurology, Neurosurgery, Pathology and Pediatrics, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine;
| |
Collapse
|
37
|
Carai A, Mastronuzzi A, De Benedictis A, Messina R, Cacchione A, Miele E, Randi F, Esposito G, Trezza A, Colafati GS, Savioli A, Locatelli F, Marras CE. Robot-Assisted Stereotactic Biopsy of Diffuse Intrinsic Pontine Glioma: A Single-Center Experience. World Neurosurg 2017; 101:584-588. [PMID: 28254596 DOI: 10.1016/j.wneu.2017.02.088] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 02/16/2017] [Accepted: 02/17/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Diffuse intrinsic pontine glioma (DIPG) is a childhood tumor with a dismal prognosis. Emerging molecular signatures have paved the way for stereotactic biopsy in selected centers. We present our experience in DIPG stereotactic needle biopsy using the Robotic Stereotactic-Assisted system (ROSA) in a series of consecutive pediatric patients. METHODS All stereotactic biopsy procedures for DIPG performed during the last year at our institution were considered. All procedures were carried out using the ROSA surgical assistant through a precoronary approach. All children underwent a postoperative computed tomography scan to document possible surgical complications and confirm the site of biopsy. Postoperative clinical changes were recorded to test morbidity of the procedure. RESULTS In the last year, we performed 7 pontine needle biopsies. Specimens were diagnostic and useful for molecular analysis in all cases. No surgical complications were observed. One child showed a transient neurologic worsening related to the biopsy that resolved within 2 weeks. The combination of the precoronary approach and use of the stereotactic ROSA system allowed single-session surgeries in all cases. CONCLUSIONS Pontine biopsy for DIPG is a safe procedure in selected centers. The advantages of the single-session procedure we described might be of particular interest in the pediatric setting.
Collapse
Affiliation(s)
- Andrea Carai
- Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Angela Mastronuzzi
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Alessandro De Benedictis
- Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Raffaella Messina
- Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Antonella Cacchione
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Evelina Miele
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Franco Randi
- Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giacomo Esposito
- Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Trezza
- Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Neurosurgery, Department of Surgery and Translational Medicine, Milan Center for Neuroscience, University of Milano Bicocca, San Gerardo Hospital, Monza, Italy
| | | | - Alessandra Savioli
- Intensive Care Unit, Department of Emergency, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology and Stem Cell Transplantation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Pediatric Science, University of Pavia, Pavia, Italy
| | - Carlo Efisio Marras
- Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
38
|
Diffuse intrinsic pontine gliomas in children: Interest of robotic frameless assisted biopsy. A technical note. Neurochirurgie 2016; 62:327-331. [DOI: 10.1016/j.neuchi.2016.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 07/03/2016] [Accepted: 07/17/2016] [Indexed: 11/20/2022]
|
39
|
Bornhorst M, Hwang EI. Experimental Therapeutic Trial Design for Pediatric Brain Tumors. J Child Neurol 2016; 31:1421-32. [PMID: 26353880 DOI: 10.1177/0883073815604221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/11/2015] [Indexed: 11/17/2022]
Abstract
Pediatric brain tumors are the leading cause of cancer-related death during childhood. Since the first pediatric brain tumor clinical trials, the field has seen improved outcomes in some, but not all tumor types. In the past few decades, a number of promising new therapeutic agents have emerged, yet only a few of these agents have been incorporated into clinical trials for pediatric brain tumors. In this review, the authors discuss the process of and challenges in pediatric clinical trial design; this will allow for highly efficient and effective clinical trials with appropriate endpoints to ensure rapid and safe investigation of novel therapeutics for children with brain tumors.
Collapse
Affiliation(s)
- Miriam Bornhorst
- Department of Pediatric Hematology-Oncology, Children's National Medical Center, Washington, DC, USA Brain Tumor Institute, Washington, DC, USA
| | - Eugene I Hwang
- Department of Pediatric Hematology-Oncology, Children's National Medical Center, Washington, DC, USA Gilbert Family Neurofibromatosis Institute, Centers for Cancer and Immunology Research & Neuroscience Research, Children's National Medical Center, Washington, DC, USA
| |
Collapse
|
40
|
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: 2.8] [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
|
41
|
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: 3.0] [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
|
42
|
Kieran MW, Goumnerova LC, Prados M, Gupta N. Biopsy for diffuse intrinsic pontine glioma: a reappraisal. J Neurosurg Pediatr 2016; 18:390-1. [PMID: 27153377 DOI: 10.3171/2015.6.peds15374] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
43
|
Blumenthal DT, Yalon M, Vainer GW, Lossos A, Yust S, Tzach L, Cagnano E, Limon D, Bokstein F. Pembrolizumab: first experience with recurrent primary central nervous system (CNS) tumors. J Neurooncol 2016; 129:453-460. [PMID: 27377654 DOI: 10.1007/s11060-016-2190-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/23/2016] [Indexed: 12/26/2022]
Abstract
Patients with progressive primary brain tumors (PBT) are attracted to promising new treatments, even prior to convincing data. Anti-PD1 immunotherapies have been in the spotlight since publication of groundbreaking results for metastatic melanoma with pembrolizumab (PBL). Our objective was to report on the response and toxicity of PBL in patients with advanced PBT. We retrospectively reviewed the charts of 22 patients (17 adults and 5 children) with recurrent central nervous system tumors treated with PBL. We analyzed prior antineoplastic therapies, steroid usage, and outcomes. Patients received a median of two neoplastic therapies prior to PBL, and a median of three infusions of PBL in adults and four in children. Twelve patients (9 adults and 3 children) started PBL on steroids (median dose in adults 4 mg; range 2-8, and in children 1.5 mg, range 0.5-4) and five patients received steroids later during PBL treatment. Twelve patients (10 adults and 2 children) received concomitant bevacizumab with PBL. Side effects were minimal. All patients showed progressive tumor growth during therapy. Median OS from the start of PBL was 2.6 months in adults and 3.2 months in children. Two GB patients underwent tumor resection following treatment with PBL. Tumor-lymphocytic response in these cases was unremarkable, and PD-L1 immuno-staining was negative. In this series of 22 patients with recurrent primary brain tumors, PBL showed no clinical or histologic efficacy. We do not recommend further use of PBL for recurrent PBT unless convincing prospective clinical trial data are published.
Collapse
Affiliation(s)
- Deborah T Blumenthal
- Neuro-Oncology Service, Division of Oncology, Tel Aviv Sourasky Medical Center, 64239, Tel Aviv, Israel.
| | - Michal Yalon
- Pediatric Neuro-Oncology Service, Pediatric Hemato-Oncology Department, Chaim Sheba Medical Center, 52621, Tel HaShomer, Israel
| | - Gilad W Vainer
- Molecular Pathology Service, Department of Pathology, Tel Aviv Sourasky Medical Center, 64239, Tel Aviv, Israel
| | - Alexander Lossos
- Leslie and Michael Gaffin Center for Neuro-Oncology, Department of Oncology, Hadassah-Hebrew University Medical Center, 91120, Jerusalem, Israel
| | - Shlomit Yust
- Oncology Institute, Davidoff Center, Rabin Medical Center, 49100, Petah Tikva, Israel
| | - Lior Tzach
- Oncology Institute, Chaim Sheba Medical Center, 52621, Tel HaShomer, Israel
| | - Emanuela Cagnano
- Molecular Pathology Service, Department of Pathology, Tel Aviv Sourasky Medical Center, 64239, Tel Aviv, Israel
| | - Dror Limon
- Oncology Institute, Davidoff Center, Rabin Medical Center, 49100, Petah Tikva, Israel
| | - Felix Bokstein
- Neuro-Oncology Service, Division of Oncology, Tel Aviv Sourasky Medical Center, 64239, Tel Aviv, Israel
| |
Collapse
|
44
|
Abstract
INTRODUCTION Despite substantial improvements in standards of care, the most common aggressive pediatric and adult high-grade gliomas (HGG) carry uniformly fatal diagnoses due to unique treatment limitations, high recurrence rates and the absence of effective treatments following recurrence. Recent advancements in our understanding of the pathophysiology, genetics and epigenetics as well as mechanisms of immune surveillance during gliomagenesis have created new knowledge to design more effective and target-directed therapies to improve patient outcomes. AREAS COVERED In this review, the authors discuss the critical genetic, epigenetic and immunologic aberrations found in gliomas that appear rational and promising for therapeutic developments in the presence and future. The current state of the latest therapeutic developments including tumor-specific targeted drug therapies, metabolic targeting, epigenetic modulation and immunotherapy are summarized and suggestions for future directions are offered. Furthermore, they highlight contemporary issues related to the clinical development, such as challenges in clinical trials and toxicities. EXPERT OPINION The commitment to understanding the process of gliomagenesis has created a catalogue of aberrations that depict multiple mechanisms underlying this disease, many of which are suitable to therapeutic inhibition and are currently tested in clinical trials. Thus, future treatment endeavors will employ multiple treatment modalities that target disparate tumor characteristics personalized to the patient's individual tumor.
Collapse
Affiliation(s)
- Verena Staedtke
- a Department of Neurology , Johns Hopkins Medical Institutions , Baltimore , MD , USA
| | - Ren-Yuan Bai
- b Department of Neurosurgery , Johns Hopkins Medical Institutions , Baltimore , MD , USA
| | - John Laterra
- a Department of Neurology , Johns Hopkins Medical Institutions , Baltimore , MD , USA.,c Department of Oncology , Johns Hopkins Medical Institutions , Baltimore , MD , USA.,d Department of Neuroscience , Johns Hopkins Medical Institutions , Baltimore , MD , USA
| |
Collapse
|
45
|
Klimo P, Nesvick CL, Broniscer A, Orr BA, Choudhri AF. Malignant brainstem tumors in children, excluding diffuse intrinsic pontine gliomas. J Neurosurg Pediatr 2016; 17:57-65. [PMID: 26474099 DOI: 10.3171/2015.6.peds15166] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Malignant tumors of the brainstem, excluding classic diffuse intrinsic pontine gliomas (DIPGs), are a very rare, heterogeneous group of neoplasms that have been infrequently described in the literature. In this paper, the authors present their experiences with treating these unique cancers. METHODS A retrospective chart review was conducted to identify eligible cases over a 15-year period. All tumors involving the pons were, by consensus, felt not to be DIPGs based on their neuroimaging features. Demographic information, pathological specimens, neuroimaging characteristics, surgical and nonsurgical management plans, and survival data were gathered for analysis. RESULTS Between January 2000 and December 2014, 29 patients were identified. The mean age at diagnosis was 8.4 years (range 2 months to 25 years), and 17 (59%) patients were male. The most common presenting signs and symptoms were cranial neuropathies (n = 24; 83%), hemiparesis (n = 12; 41%), and ataxia or gait disturbance (n = 10; 34%). There were 18 glial and 11 embryonal tumors. Of the glial tumors, 5 were radiation-induced and 1 was a malignant transformation of a previously known low-grade tumor. Surgical intervention consisted of biopsy alone in 12 patients and some degree of resection in another 15 patients. Two tumors were diagnosed postmortem. The median overall survival for all patients was 196 days (range 15 to 3999 days). There are currently 5 (17%) patients who are still alive: 1 with an anaplastic astrocytoma and the remaining with embryonal tumors. CONCLUSIONS In general, malignant non-DIPG tumors of the brainstem carry a poor prognosis. However, maximal cytoreductive surgery may be an option for select patients with focal tumors. Long-term survival is possible in patients with nonmetastatic embryonal tumors after multimodal treatment, most importantly maximal resection.
Collapse
Affiliation(s)
- Paul Klimo
- Le Bonheur Neuroscience Institute, Le Bonheur Children's Hospital;,Departments of 2 Surgery.,Semmes-Murphey Neurologic & Spine Institute;,Departments of 4 Neurosurgery
| | - Cody L Nesvick
- University of Tennessee Health Science Center, Memphis, Tennessee
| | | | - Brent A Orr
- Pathology, St. Jude Children's Research Hospital
| | - Asim F Choudhri
- Le Bonheur Neuroscience Institute, Le Bonheur Children's Hospital;,Departments of 4 Neurosurgery.,Radiology, University of Tennessee Health Science Center; and
| |
Collapse
|
46
|
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: 130] [Impact Index Per Article: 14.4] [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
|
47
|
Abstract
Diffuse intrinsic pontine glioma (DIPG) is an aggressive tumor that is universally fatal, and to-date we are at a virtual standstill in improving its grim prognosis. Dearth of tissue due to rarity of biopsy has precluded understanding the elusive biology and frustration continues in reproducing faithful animal models for translational research. Furthermore the intricate anatomy of the pons has forestalled locoregional therapy and drug penetration. Over the last few years, biopsy-driven targeted therapy, development of vitro and xenograft animal models for therapeutic testing, profiling immunotherapeutic strategies and locoregional infusion of drugs in brain stem tumors, now provide a sense of hope in the years ahead. This review aims to discuss current status and advances in the management of these tumors.
Collapse
Affiliation(s)
- Soumen Khatua
- Pediatric Neuro-Oncology, Department of Pediatrics, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 87, Houston, TX 77030, USA
| | | |
Collapse
|
48
|
Buczkowicz P, Hawkins C. Pathology, Molecular Genetics, and Epigenetics of Diffuse Intrinsic Pontine Glioma. Front Oncol 2015; 5:147. [PMID: 26175967 PMCID: PMC4485076 DOI: 10.3389/fonc.2015.00147] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/16/2015] [Indexed: 11/13/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a devastating pediatric brain cancer with no effective therapy. Histological similarity of DIPG to supratentorial high-grade astrocytomas of adults has led to assumptions that these entities possess similar underlying molecular properties and therefore similar therapeutic responses to standard therapies. The failure of all clinical trials in the last 30 years to improve DIPG patient outcome has suggested otherwise. Recent studies employing next-generation sequencing and microarray technologies have provided a breadth of evidence highlighting the unique molecular genetics and epigenetics of this cancer, distinguishing it from both adult and pediatric cerebral high-grade astrocytomas. This review describes the most common molecular genetic and epigenetic signatures of DIPG in the context of molecular subgroups and histopathological diagnosis, including this tumor entity's unique mutational landscape, copy number alterations, and structural variants, as well as epigenetic changes on the global DNA and histone levels. The increased knowledge of DIPG biology and histopathology has opened doors to new diagnostic and therapeutic avenues.
Collapse
Affiliation(s)
- Pawel Buczkowicz
- Division of Pathology, The Hospital for Sick Children , Toronto, ON , Canada ; The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children , Toronto, ON , Canada
| | - Cynthia Hawkins
- Division of Pathology, The Hospital for Sick Children , Toronto, ON , Canada ; The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children , Toronto, ON , Canada ; Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto , Toronto, ON , Canada
| |
Collapse
|
49
|
Alken SP, D'Urso P, Saran FH. Managing teenage/young adult (TYA) brain tumors: a UK perspective. CNS Oncol 2015; 4:235-46. [PMID: 26118974 DOI: 10.2217/cns.15.14] [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] [Indexed: 01/27/2023] Open
Abstract
Tumors of the CNS are among the commonest malignancies occurring in teenage/young adult patients (i.e., those aged between 15 and 24 years). The treatment of this patient population is challenging. Adolescence and young adulthood are a turbulent period of life, with physical, emotional, social and cognitive changes. Best practice advocates their treatment in dedicated teenage/young adult units, with multidisciplinary team input and access to clinical trials. Treatment of CNS malignancies is dependent upon histological subtype and staging, with varying combinations of surgery, radiotherapy and chemotherapy used. Clinical trials directly targeted at this patient population are rare; treatments are based on pediatric protocols as studies have demonstrated improved outcomes in patients (with other malignancies) treated as such. Scope for improvement lies in minimizing patient risk of recurrence and long-term sequelae of treatment. Molecular characterization of tumors may provide further information.
Collapse
Affiliation(s)
- Scheryll P Alken
- Department of Neuro Oncology, Royal Marsden Hospital, Sutton, UK
| | - Pietro D'Urso
- Department of Neurosurgery, Salford Royal Hospital Foundation Trust, Salford, UK
| | - Frank H Saran
- Department of Neuro Oncology, Royal Marsden Hospital, Sutton, UK
| |
Collapse
|
50
|
Panditharatna E, Yaeger K, Kilburn LB, Packer RJ, Nazarian J. Clinicopathology of diffuse intrinsic pontine glioma and its redefined genomic and epigenomic landscape. Cancer Genet 2015. [PMID: 26206682 DOI: 10.1016/j.cancergen.2015.04.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is one of the most lethal pediatric central nervous system (CNS) cancers. Recently, a surge in molecular studies of DIPG has occurred, in large part due to the increased availability of tumor tissue through donation of post-mortem specimens. These new discoveries have established DIPGs as biologically distinct from adult gliomas, harboring unique genomic aberrations. Mutations in histone encoding genes are shown to be associated with >70% of DIPG cases. However, the exact molecular mechanisms of the tumorigenicity of these mutations remain elusive. Understanding the driving mutations and genomic landscape of DIPGs can now guide the development of targeted therapies for this incurable childhood cancer.
Collapse
Affiliation(s)
- Eshini Panditharatna
- Institute for Biomedical Sciences, George Washington University School of Medicine, Washington, DC, USA; Research Center for Genetic Medicine, Children's National Health System, Washington, DC, USA
| | - Kurt Yaeger
- Department of Neurosurgery, Georgetown University School of Medicine, Washington, DC, USA
| | - Lindsay B Kilburn
- Division of Oncology, Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, USA
| | - Roger J Packer
- Brain Tumor Institute, Center for Neuroscience and Behavioral Medicine, Children's National Health System, Washington, DC, USA
| | - Javad Nazarian
- Research Center for Genetic Medicine, Children's National Health System, Washington, DC, USA; Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|