1
|
Berthelot C, Huchedé P, Bertrand-Chapel A, Beuriat PA, Leblond P, Castets M. Bone Morphogenic Proteins in Pediatric Diffuse Midline Gliomas: How to Make New Out of Old? Int J Mol Sci 2024; 25:3361. [PMID: 38542334 PMCID: PMC10969837 DOI: 10.3390/ijms25063361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 11/11/2024] Open
Abstract
The BMP pathway is one of the major signaling pathways in embryonic development, ontogeny and homeostasis, identified many years ago by pioneers in developmental biology. Evidence of the deregulation of its activity has also emerged in many cancers, with complex and sometimes opposing effects. Recently, its role has been suspected in Diffuse Midline Gliomas (DMG), among which Diffuse Intrinsic Pontine Gliomas (DIPG) are one of the most complex challenges in pediatric oncology. Genomic sequencing has led to understanding part of their molecular etiology, with the identification of histone H3 mutations in a large proportion of patients. The epigenetic remodeling associated with these genetic alterations has also been precisely described, creating a permissive context for oncogenic transcriptional program activation. This review aims to describe the new findings about the involvement of BMP pathway activation in these tumors, placing their appearance in a developmental context. Targeting the oncogenic synergy resulting from this pathway activation in an H3K27M context could offer new therapeutic perspectives based on targeting treatment-resistant cell states.
Collapse
Affiliation(s)
- Clément Berthelot
- Childhood Cancer & Cell Death Team (C3 Team), LabEx DEVweCAN, Institut Convergence Plascan, Centre de Recherche en Cancérologie de Lyon (CRCL), Centre Léon Bérard, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, 69008 Lyon, France; (C.B.); (P.H.); (A.B.-C.); (P.L.); (M.C.)
- South-ROCK Pediatric Cancer Research Center, 69008 Lyon, France
| | - Paul Huchedé
- Childhood Cancer & Cell Death Team (C3 Team), LabEx DEVweCAN, Institut Convergence Plascan, Centre de Recherche en Cancérologie de Lyon (CRCL), Centre Léon Bérard, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, 69008 Lyon, France; (C.B.); (P.H.); (A.B.-C.); (P.L.); (M.C.)
- South-ROCK Pediatric Cancer Research Center, 69008 Lyon, France
| | - Adrien Bertrand-Chapel
- Childhood Cancer & Cell Death Team (C3 Team), LabEx DEVweCAN, Institut Convergence Plascan, Centre de Recherche en Cancérologie de Lyon (CRCL), Centre Léon Bérard, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, 69008 Lyon, France; (C.B.); (P.H.); (A.B.-C.); (P.L.); (M.C.)
- South-ROCK Pediatric Cancer Research Center, 69008 Lyon, France
| | - Pierre-Aurélien Beuriat
- South-ROCK Pediatric Cancer Research Center, 69008 Lyon, France
- Multisite Institute of Pathology, Groupement Hospitalier Est du CHU de Lyon, Hopital Femme-Mère-Enfant, 69677 Bron, France
| | - Pierre Leblond
- Childhood Cancer & Cell Death Team (C3 Team), LabEx DEVweCAN, Institut Convergence Plascan, Centre de Recherche en Cancérologie de Lyon (CRCL), Centre Léon Bérard, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, 69008 Lyon, France; (C.B.); (P.H.); (A.B.-C.); (P.L.); (M.C.)
- South-ROCK Pediatric Cancer Research Center, 69008 Lyon, France
- Department of Translational Research in Pediatric Oncology PROSPECT, Centre Léon Bérard, 69008 Lyon, France
- Department of Pediatric Oncology, Institut d’Hématologie et d’Oncologie Pédiatrique, Centre Léon Bérard, 69008 Lyon, France
| | - Marie Castets
- Childhood Cancer & Cell Death Team (C3 Team), LabEx DEVweCAN, Institut Convergence Plascan, Centre de Recherche en Cancérologie de Lyon (CRCL), Centre Léon Bérard, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, 69008 Lyon, France; (C.B.); (P.H.); (A.B.-C.); (P.L.); (M.C.)
- South-ROCK Pediatric Cancer Research Center, 69008 Lyon, France
- Department of Translational Research in Pediatric Oncology PROSPECT, Centre Léon Bérard, 69008 Lyon, France
| |
Collapse
|
2
|
Bruschi M, Midjek L, Ajlil Y, Vairy S, Lancien M, Ghermaoui S, Kergrohen T, Verreault M, Idbaih A, de Biagi CAO, Liu I, Filbin MG, Beccaria K, Blauwblomme T, Puget S, Tauziede-Espariat A, Varlet P, Dangouloff-Ros V, Boddaert N, Le Teuff G, Grill J, Montagnac G, Elkhatib N, Debily MA, Castel D. Diffuse midline glioma invasion and metastasis rely on cell-autonomous signaling. Neuro Oncol 2024; 26:553-568. [PMID: 37702430 PMCID: PMC10912010 DOI: 10.1093/neuonc/noad161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Diffuse midline gliomas (DMG) are pediatric tumors with negligible 2-year survival after diagnosis characterized by their ability to infiltrate the central nervous system. In the hope of controlling the local growth and slowing the disease, all patients receive radiotherapy. However, distant progression occurs frequently in DMG patients. Current clues as to what causes tumor infiltration circle mainly around the tumor microenvironment, but there are currently no known determinants to predict the degree of invasiveness. METHODS In this study, we use patient-derived glioma stem cells (GSCs) to create patient-specific 3D avatars to model interindividual invasion and elucidate the cellular supporting mechanisms. RESULTS We show that GSC models in 3D mirror the invasive behavior of the parental tumors, thus proving the ability of DMG to infiltrate as an autonomous characteristic of tumor cells. Furthermore, we distinguished 2 modes of migration, mesenchymal and ameboid-like, and associated the ameboid-like modality with GSCs derived from the most invasive tumors. Using transcriptomics of both organoids and primary tumors, we further characterized the invasive ameboid-like tumors as oligodendrocyte progenitor-like, with highly contractile cytoskeleton and reduced adhesion ability driven by crucial over-expression of bone morphogenetic pathway 7 (BMP7). Finally, we deciphered MEK, ERK, and Rho/ROCK kinases activated downstream of the BMP7 stimulation as actionable targets controlling tumor cell motility. CONCLUSIONS Our findings identify 2 new therapeutic avenues. First, patient-derived GSCs represent a predictive tool for patient stratification in order to adapt irradiation strategies. Second, autocrine and short-range BMP7-related signaling becomes a druggable target to prevent DMG spread and metastasis.
Collapse
Affiliation(s)
- Marco Bruschi
- Inserm U981, Molecular Predictors and New Targets in Oncology, Team Genomics and Oncogenesis of Pediatric Brain Tumors, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Lilia Midjek
- Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| | - Yassine Ajlil
- Inserm U981, Molecular Predictors and New Targets in Oncology, Team Genomics and Oncogenesis of Pediatric Brain Tumors, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Stephanie Vairy
- Inserm U981, Molecular Predictors and New Targets in Oncology, Team Genomics and Oncogenesis of Pediatric Brain Tumors, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Département de Cancérologie de l’Enfant et de l’Adolescent, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Manon Lancien
- Inserm U981, Molecular Predictors and New Targets in Oncology, Team Genomics and Oncogenesis of Pediatric Brain Tumors, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Samia Ghermaoui
- Inserm U981, Molecular Predictors and New Targets in Oncology, Team Genomics and Oncogenesis of Pediatric Brain Tumors, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Thomas Kergrohen
- Inserm U981, Molecular Predictors and New Targets in Oncology, Team Genomics and Oncogenesis of Pediatric Brain Tumors, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Maite Verreault
- Sorbonne Université, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, DMU Neurosciences, Service de Neurologie 2-Mazarin, Paris, France
| | - Ahmed Idbaih
- Sorbonne Université, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, DMU Neurosciences, Service de Neurologie 2-Mazarin, Paris, France
| | - Carlos Alberto Oliveira de Biagi
- Department of Pediatric Oncology, Dana-Farber Boston Children’s Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Ilon Liu
- Department of Pediatric Oncology, Dana-Farber Boston Children’s Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Mariella G Filbin
- Department of Pediatric Oncology, Dana-Farber Boston Children’s Cancer and Blood Disorders Center, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Kevin Beccaria
- Inserm U981, Molecular Predictors and New Targets in Oncology, Team Genomics and Oncogenesis of Pediatric Brain Tumors, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Department of Pediatric Neurosurgery, Necker Enfants Malades Hospital, APHP, Université Paris Cité, Paris, France
| | - Thomas Blauwblomme
- Department of Pediatric Neurosurgery, Necker Enfants Malades Hospital, APHP, Université Paris Cité, Paris, France
| | - Stephanie Puget
- Department of Pediatric Neurosurgery, Necker Enfants Malades Hospital, APHP, Université Paris Cité, Paris, France
| | - Arnault Tauziede-Espariat
- Department of Neuropathology, GHU Paris-Psychiatrie et Neurosciences, Sainte-Anne Hospital, ParisFrance
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR 1266, INSERM, IMA-BRAIN, Université de Paris, Paris, France
| | - Pascale Varlet
- Department of Neuropathology, GHU Paris-Psychiatrie et Neurosciences, Sainte-Anne Hospital, ParisFrance
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR 1266, INSERM, IMA-BRAIN, Université de Paris, Paris, France
| | - Volodia Dangouloff-Ros
- Paediatric Radiology Department, AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, ParisFrance
| | - Nathalie Boddaert
- Paediatric Radiology Department, AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, ParisFrance
| | - Gwenael Le Teuff
- Department of Biostatistics and Epidemiology, Gustave Roussy and Paris-Saclay University, Villejuif, France
| | - Jacques Grill
- Inserm U981, Molecular Predictors and New Targets in Oncology, Team Genomics and Oncogenesis of Pediatric Brain Tumors, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Département de Cancérologie de l’Enfant et de l’Adolescent, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Guillaume Montagnac
- Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| | - Nadia Elkhatib
- Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| | - Marie-Anne Debily
- Inserm U981, Molecular Predictors and New Targets in Oncology, Team Genomics and Oncogenesis of Pediatric Brain Tumors, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Département de Biologie, Université Evry Paris-Saclay, Evry, France
| | - David Castel
- Inserm U981, Molecular Predictors and New Targets in Oncology, Team Genomics and Oncogenesis of Pediatric Brain Tumors, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
3
|
Cacciotti C, Wright KD. Advances in Treatment of Diffuse Midline Gliomas. Curr Neurol Neurosci Rep 2023; 23:849-856. [PMID: 37921944 DOI: 10.1007/s11910-023-01317-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2023] [Indexed: 11/05/2023]
Abstract
PURPOSE OF REVIEW Diffuse midline gliomas (DMGs) generally carry a poor prognosis, occur during childhood, and involve midline structures of the central nervous system, including the thalamus, pons, and spinal cord. RECENT FINDINGS To date, irradiation has been shown to be the only beneficial treatment for DMG. Various genetic modifications have been shown to play a role in the pathogenesis of this disease. Current treatment strategies span targeting epigenetic dysregulation, cell cycle, specific genetic alterations, and the immune microenvironment. Herein, we review the complex features of this disease as it relates to current and past therapeutic approaches.
Collapse
Affiliation(s)
- Chantel Cacciotti
- Children's Hospital London Health Sciences/Western University, London, ON, Canada.
| | - Karen D Wright
- Dana Farber/Boston Children's Cancer and Blood Disorder Center, Boston, MA, USA
| |
Collapse
|
4
|
Cocito C, Martin B, Giantini-Larsen AM, Valcarce-Aspegren M, Souweidane MM, Szalontay L, Dahmane N, Greenfield JP. Leptomeningeal dissemination in pediatric brain tumors. Neoplasia 2023; 39:100898. [PMID: 37011459 PMCID: PMC10124141 DOI: 10.1016/j.neo.2023.100898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 04/03/2023]
Abstract
Leptomeningeal disease (LMD) in pediatric brain tumors (PBTs) is a poorly understood and categorized phenomenon. LMD incidence rates, as well as diagnosis, treatment, and screening practices, vary greatly depending on the primary tumor pathology. While LMD is encountered most frequently in medulloblastoma, reports of LMD have been described across a wide variety of PBT pathologies. LMD may be diagnosed simultaneously with the primary tumor, at time of recurrence, or as primary LMD without a primary intraparenchymal lesion. Dissemination and seeding of the cerebrospinal fluid (CSF) involves a modified invasion-metastasis cascade and is often the result of direct deposition of tumor cells into the CSF. Cells develop select environmental advantages to survive the harsh, nutrient poor and turbulent environment of the CSF and leptomeninges. Improved understanding of the molecular mechanisms that underlie LMD, along with improved diagnostic and treatment approaches, will help the prognosis of children affected by primary brain tumors.
Collapse
|
5
|
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
|
6
|
Park YW, Han K, Park JE, Ahn SS, Kim EH, Kim J, Kang SG, Chang JH, Kim SH, Lee SK. Leptomeningeal metastases in glioma revisited: incidence and molecular predictors based on postcontrast fluid-attenuated inversion recovery imaging. J Neurosurg 2022:1-11. [DOI: 10.3171/2022.9.jns221659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/22/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE
Leptomeningeal metastases (LMs) in glioma have been underestimated given their low incidence and the lack of reliable imaging. Authors of this study aimed to investigate the real-world incidence of LMs using cerebrospinal fluid (CSF)–sensitive imaging, namely postcontrast fluid-attenuated inversion recovery (FLAIR) imaging, and to analyze molecular predictors for LMs in the molecular era.
METHODS
A total of 1405 adult glioma (World Health Organization [WHO] grade 2–4) patients underwent postcontrast FLAIR imaging at initial diagnosis and during treatment monitoring between 2001 and 2021. Collected molecular data included isocitrate dehydrogenase (IDH) mutation, 1p/19q codeletion, H3 K27 alteration, and O6-methylguanine–DNA methyltransferase (MGMT) promoter methylation status. LM diagnosis was performed with MRI including postcontrast FLAIR sequences. Logistic regression analysis for LM development was performed with molecular, clinical, and imaging data. Overall survival (OS) was compared between patients with and those without LM.
RESULTS
LM was identified in 228 patients (16.2%), 110 (7.8%) at the initial diagnosis and 118 (8.4%) at recurrence. Among the molecular diagnostics, IDH-wildtype (OR 3.14, p = 0.001) and MGMT promoter unmethylation (OR 1.43, p = 0.034) were independent predictors of LM. WHO grade 4 (OR 10.52, p < 0.001) and nonlobar location (OR 1.56, p = 0.048) were associated with LM at initial diagnosis, whereas IDH-wildtype (OR 5.04, p < 0.001) and H3 K27 alteration (OR 3.39, p = 0.003) were associated with LM at recurrence. Patients with LM had a worse median OS than those without LM (16.7 vs 32.0 months, p < 0.001, log-rank test), which was confirmed as an independent factor on multivariable Cox analysis (p = 0.004).
CONCLUSIONS
CSF-sensitive imaging aids the diagnosis of LM, demonstrating a high incidence of LM in adult gliomas. Furthermore, molecular markers are associated with LM development in glioma, and patients with aggressive molecular markers warrant imaging surveillance for LM.
Collapse
Affiliation(s)
- Yae Won Park
- Departments of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science,
| | - Kyunghwa Han
- Departments of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science,
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung Soo Ahn
- Departments of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science,
| | | | - Jinna Kim
- Departments of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science,
| | | | | | - Se Hoon Kim
- Pathology, Yonsei University College of Medicine, Seoul; and
| | - Seung-Koo Lee
- Departments of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science,
| |
Collapse
|
7
|
Disseminated diffuse midline glioma associated with poorly differentiated orbital lesion and metastases in an 8-year-old girl: case report and literature review. Childs Nerv Syst 2022; 38:2005-2010. [PMID: 35460354 DOI: 10.1007/s00381-022-05530-7] [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: 12/06/2021] [Accepted: 04/13/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Disseminated diffuse midline glioma (DMG) is a devastating diagnosis. Molecular subtyping has increased our understanding of this tumor. CASE Here, we report the case of an 8-year-old girl who presented with symptoms of brainstem dysfunction and was found to have disseminated DMG with lesions in the pons, thalamus and bilateral temporal lobes. Molecular subtyping of the temporal lobe tumor tissue was consistent with H3 K27me3 loss and EZHIP overexpression, falling under the newly designated "H3 K27-altered" AQ5WHO subtype of DMG. Pathology from biopsy of the orbital lesion showed poorly differentiated rhabdoid-like disseminated tumor cells. The patient went on to develop lesions in the peritoneum, infratemporal fossa, and along the lumbosacral nerve roots. CONCLUSION This unique case illustrates the aggressive behavior of H3 K27-altered tumors and their potential to metastasize.
Collapse
|
8
|
Tomita Y, Shimazu Y, Somasundaram A, Tanaka Y, Takata N, Ishi Y, Gadd S, Hashizume R, Angione A, Pinero G, Hambardzumyan D, Brat DJ, Hoeman CM, Becher OJ. A novel mouse model of diffuse midline glioma initiated in neonatal oligodendrocyte progenitor cells highlights cell-of-origin dependent effects of H3K27M. Glia 2022; 70:1681-1698. [PMID: 35524725 PMCID: PMC9546478 DOI: 10.1002/glia.24189] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 11/13/2022]
Abstract
Diffuse midline glioma (DMG) is a type of lethal brain tumor that develops mainly in children. The majority of DMG harbor the K27M mutation in histone H3. Oligodendrocyte progenitor cells (OPCs) in the brainstem are candidate cells-of-origin for DMG, yet there is no genetically engineered mouse model of DMG initiated in OPCs. Here, we used the RCAS/Tv-a avian retroviral system to generate DMG in Olig2-expressing progenitors and Nestin-expressing progenitors in the neonatal mouse brainstem. PDGF-A or PDGF-B overexpression, along with p53 deletion, resulted in gliomas in both models. Exogenous overexpression of H3.3K27M had a significant effect on tumor latency and tumor cell proliferation when compared with H3.3WT in Nestin+ cells but not in Olig2+ cells. Further, the fraction of H3.3K27M-positive cells was significantly lower in DMGs initiated in Olig2+ cells relative to Nestin+ cells, both in PDGF-A and PDGF-B-driven models, suggesting that the requirement for H3.3K27M is reduced when tumorigenesis is initiated in Olig2+ cells. RNA-sequencing analysis revealed that the differentially expressed genes in H3.3K27M tumors were non-overlapping between Olig2;PDGF-B, Olig2;PDGF-A, and Nestin;PDGF-A models. GSEA analysis of PDGFA tumors confirmed that the transcriptomal effects of H3.3K27M are cell-of-origin dependent with H3.3K27M promoting epithelial-to-mesenchymal transition (EMT) and angiogenesis when Olig2 marks the cell-of-origin and inhibiting EMT and angiogenesis when Nestin marks the cell-of-origin. We did observe some overlap with H3.3K27M promoting negative enrichment of TNFA_Signaling_Via_NFKB in both models. Our study suggests that the tumorigenic effects of H3.3K27M are cell-of-origin dependent, with H3.3K27M being more oncogenic in Nestin+ cells than Olig2+ cells.
Collapse
Affiliation(s)
- Yusuke Tomita
- Department of PediatricsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
- Department of Neurosurgery and Neuroendovascular SurgeryHiroshima City Hiroshima Citizens HospitalHiroshimaJapan
| | - Yosuke Shimazu
- Department of PediatricsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Agila Somasundaram
- Division of Hematology, Oncology and Stem Cell TransplantAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
| | - Yoshihiro Tanaka
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Center for Arrhythmia Research, Department of CardiologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Nozomu Takata
- Center for Vascular and Developmental BiologyFeinberg Cardiovascular and Renal Research Institute (FCVRRI), Northwestern UniversityChicagoIllinoisUSA
- Simpson Querrey Institute for BioNanotechnologyNorthwestern UniversityChicagoIllinoisUSA
| | - Yukitomo Ishi
- Department of PediatricsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Samantha Gadd
- Department of PathologyAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
| | - Rintaro Hashizume
- Department of PediatricsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
- Division of Hematology, Oncology and Stem Cell TransplantAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Department of Biochemistry and Molecular GeneticsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Angelo Angione
- Department of Neurosurgery and Oncological SciencesMount Sinai School of MedicineNew YorkUSA
| | - Gonzalo Pinero
- Department of Neurosurgery and Oncological SciencesMount Sinai School of MedicineNew YorkUSA
| | - Dolores Hambardzumyan
- Department of Neurosurgery and Oncological SciencesMount Sinai School of MedicineNew YorkUSA
| | - Daniel J. Brat
- Department of PathologyFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Christine M. Hoeman
- Department of PediatricsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Oren J. Becher
- Department of PediatricsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
- Division of Hematology, Oncology and Stem Cell TransplantAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Department of Biochemistry and Molecular GeneticsFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
- Jack Martin Division of Pediatric Hematology‐oncologyMount Sinai Kravis Children's HospitalNew YorkUSA
| |
Collapse
|
9
|
Di Ruscio V, Del Baldo G, Fabozzi F, Vinci M, Cacchione A, de Billy E, Megaro G, Carai A, Mastronuzzi A. Pediatric Diffuse Midline Gliomas: An Unfinished Puzzle. Diagnostics (Basel) 2022; 12:2064. [PMID: 36140466 PMCID: PMC9497626 DOI: 10.3390/diagnostics12092064] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/15/2022] Open
Abstract
Diffuse midline glioma (DMG) is a heterogeneous group of aggressive pediatric brain tumors with a fatal prognosis. The biological hallmark in the major part of the cases is H3K27 alteration. Prognosis remains poor, with median survival ranging from 9 to 12 months from diagnosis. Clinical and radiological prognostic factors only partially change the progression-free survival but they do not improve the overall survival. Despite efforts, there is currently no curative therapy for DMG. Radiotherapy remains the standard treatment with only transitory benefits. No chemotherapeutic regimens were found to significantly improve the prognosis. In the new era of a deeper integration between histological and molecular findings, potential new approaches are currently under investigation. The entire international scientific community is trying to target DMG on different aspects. The therapeutic strategies involve targeting epigenetic alterations, such as methylation and acetylation status, as well as identifying new molecular pathways that regulate oncogenic proliferation; immunotherapy approaches too are an interesting point of research in the oncology field, and the possibility of driving the immune system against tumor cells has currently been evaluated in several clinical trials, with promising preliminary results. Moreover, thanks to nanotechnology amelioration, the development of innovative delivery approaches to overcross a hostile tumor microenvironment and an almost intact blood-brain barrier could potentially change tumor responses to different treatments. In this review, we provide a comprehensive overview of available and potential new treatments that are worldwide under investigation, with the intent that patient- and tumor-specific treatment could change the biological inauspicious history of this disease.
Collapse
Affiliation(s)
- Valentina Di Ruscio
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Giada Del Baldo
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Francesco Fabozzi
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
- Department of Pediatrics, University of Rome Tor Vergata, 00165 Rome, Italy
| | - Maria Vinci
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Antonella Cacchione
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Emmanuel de Billy
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Giacomina Megaro
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Andrea Carai
- Neurosurgery Unit, Department of Neurosciences, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Angela Mastronuzzi
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
- Faculty of Medicine and Surgery, Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| |
Collapse
|
10
|
Parenrengi MA, Prastikarunia R, Suryaningtyas W. Leptomeningeal and subependymal seeding of diffuse intrinsic pontine glioma: a case report. Childs Nerv Syst 2022; 38:1643-1645. [PMID: 35290487 DOI: 10.1007/s00381-022-05482-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/28/2022] [Indexed: 11/30/2022]
Abstract
DIPG (diffuse intrinsic pontine glioma) is a deadly cancerous tumor of the brainstem that spreads across the pons. The tumor's infiltrative nature, as well as the tumor's critical pathway and nuclei compression, contributes to the tumor's extremely poor prognosis and limited existing therapeutic options. A previous study revealed that in 40 patients with brainstem glioma, 13 (33%) patients had leptomeningeal spreading. In this paper, we reported a 7-year-old female patient who presented with a history of decreased consciousness and weakness of the right limb. Her magnetic resonance imaging (MRI) revealed a pontine mass. She was given 35 fractions of 54 Gy whole-brain radiotherapy. The post-radiotherapy MRI evaluation showed multiple nodules in periventricular region, and was suggestive of leptomeningeal and subependymal seeding of the pontine glioma in the lateral ventricles. This case report elucidated the leptomeningeal seeding in a pediatric patient with diffuse intrinsic pontine glioma.
Collapse
Affiliation(s)
- Muhammad Arifin Parenrengi
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia. .,Department of Neurosurgery, Soetomo General Academic Hospital, Dr, Surabaya, Indonesia.
| | - Resi Prastikarunia
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia.,Department of Neurosurgery, Soetomo General Academic Hospital, Dr, Surabaya, Indonesia
| | - Wihasto Suryaningtyas
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia.,Department of Neurosurgery, Soetomo General Academic Hospital, Dr, Surabaya, Indonesia
| |
Collapse
|
11
|
Massimino M, Vennarini S, Barretta F, Colombo F, Antonelli M, Pollo B, Pignoli E, Pecori E, Alessandro O, Schiavello E, Boschetti L, Podda M, Puma N, Gattuso G, Sironi G, Barzanò E, Nigro O, Bergamaschi L, Chiaravalli S, Luksch R, Meazza C, Spreafico F, Terenziani M, Casanova M, Ferrari A, Chisari M, Pellegrini C, Clerici CA, Modena P, Biassoni V. How ten-years of reirradiation for paediatric high-grade glioma may shed light on first line treatment. J Neurooncol 2022; 159:437-445. [PMID: 35809148 DOI: 10.1007/s11060-022-04079-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/25/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE Recurrence incidence for paediatric/adolescent high-grade glioma (HGG) exceeds 80%. Reirradiation (reRT) palliates symptoms and delays further progression. Strategies for reRT are scarce: we retrospectively analysed our series to develop rational future approaches. METHODS We re-evaluated MRI + RT plans of 21 relapsed HGG-patients, accrued 2010-2021, aged under 18 years. All underwent surgery and RT + chemotherapy at diagnosis. Pathologic/molecular re-evaluation allowed classification based on WHO 2021 criteria in 20/21 patients. Survival analyses and association with clinical parameters were performed. RESULTS Relapse after 1st RT was local in 12 (7 marginal), 4 disseminated, 5 local + disseminated. Re-RT obtained 8 SD, 1 PR, 1PsPD, 1 mixed response, 10 PD; neurological signs/symptoms improved in 8. Local reRT was given to 12, followed again by 6 local (2 marginal) and 4 local + disseminated second relapses in 10/12 re-evaluated. The 4 with dissemination had 1 whole brain, 2 craniospinal irradiation (CSI), 1 spine reRT and further relapsed with dissemination and local + dissemination in 3/four assessed. Five local + disseminated tumours had 3 CSI, 1 spine reRT, further progressing locally (2), disseminated (1), n.a. (1). Three had a third RT; three were alive at 19.4, 29, 50.3 months after diagnosis. Median times to progression/survival after re-RT were 3.7 months (0.6-16.2 months)/6.9 months (0.6-17.9 months), improved for longer interval between 1st RT and re-RT (P = 0.017) and for non-PD after reRT (P < 0.001). First marginal relapse showed potential association with dissemination after re-RT (P = 0.081). CONCLUSIONS This is the biggest series of re-RT in paediatric HGG. Considering the dissemination observed at relapse, our results could prompt the investigation of different first RT fields in a randomized trial.
Collapse
Affiliation(s)
- Maura Massimino
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy.
| | - Sabina Vennarini
- Pediatric Radiotherapy (SV, FC, EP, OA), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Francesco Barretta
- Medical Statistics, Biometry and Bioinformatics (FB), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | | | - Manila Antonelli
- Radiological, Oncological and Anatomo-Pathological Sciences (MA), Department of La Sapienza University, Rome, Italy
| | - Bianca Pollo
- Neuropathology (BP) Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Emanuele Pignoli
- Medical Physics (EP), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Emilia Pecori
- Pediatric Radiotherapy (SV, FC, EP, OA), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Ombretta Alessandro
- Pediatric Radiotherapy (SV, FC, EP, OA), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Elisabetta Schiavello
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Luna Boschetti
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Marta Podda
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Nadia Puma
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Giovanna Gattuso
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Giovanna Sironi
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Elena Barzanò
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Olga Nigro
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Luca Bergamaschi
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Stefano Chiaravalli
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Roberto Luksch
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Cristina Meazza
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Filippo Spreafico
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Monica Terenziani
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Michela Casanova
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Andrea Ferrari
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Marco Chisari
- Pain Therapy and Rehabilitation Units (MC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Chiara Pellegrini
- Pain Therapy and Rehabilitation Units (MC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Carlo Alfredo Clerici
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy.,Hemato-Oncology Department La Statale University, Milan, Italy
| | | | - Veronica Biassoni
- Pediatrics (MM, LB, VB, ES, CAC), Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| |
Collapse
|
12
|
Ikeda K, Kolakshyapati M, Takayasu T, Amatya VJ, Takano M, Yonezawa U, Taguchi A, Onishi S, Takeshima Y, Sugiyama K, Yamasaki F. Diffusion-weighted imaging-gadolinium enhancement mismatch sign in diffuse midline glioma. Eur J Radiol 2022; 147:110103. [DOI: 10.1016/j.ejrad.2021.110103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/04/2021] [Accepted: 12/08/2021] [Indexed: 11/03/2022]
|
13
|
Pan S, Ye D, Yue Y, Yang L, Pacia CP, DeFreitas D, Esakky P, Dahiya S, Limbrick DD, Rubin JB, Chen H, Strahle JM. Leptomeningeal disease and tumor dissemination in a murine diffuse intrinsic pontine glioma model: implications for the study of the tumor-cerebrospinal fluid-ependymal microenvironment. Neurooncol Adv 2022; 4:vdac059. [PMID: 35733516 PMCID: PMC9209751 DOI: 10.1093/noajnl/vdac059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Leptomeningeal disease and hydrocephalus are present in up to 30% of patients with diffuse intrinsic pontine glioma (DIPG), however there are no animal models of cerebrospinal fluid (CSF) dissemination. As the tumor-CSF-ependymal microenvironment may play an important role in tumor pathogenesis, we identified characteristics of the Nestin-tumor virus A (Nestin-Tva) genetically engineered mouse model that make it ideal to study the interaction of tumor cells with the CSF and its associated pathways with implications for the development of treatment approaches to address CSF dissemination in DIPG. Methods A Nestin-Tva model of DIPG utilizing the 3 most common DIPG genetic alterations (H3.3K27M, PDGF-B, and p53) was used for this study. All mice underwent MR imaging and a subset underwent histopathologic analysis with H&E and immunostaining. Results Tumor dissemination within the CSF pathways (ventricles, leptomeninges) from the subependyma was present in 76% (25/33) of mice, with invasion of the choroid plexus, disruption of the ciliated ependyma and regional subependymal fluid accumulation. Ventricular enlargement consistent with hydrocephalus was present in 94% (31/33). Ventricle volume correlated with region-specific transependymal CSF flow (periventricular T2 signal), localized anterior to the lateral ventricles. Conclusions This is the first study to report CSF pathway tumor dissemination associated with subependymal tumor in an animal model of DIPG and is representative of CSF dissemination seen clinically. Understanding the CSF-tumor-ependymal microenvironment has significant implications for treatment of DIPG through targeting mechanisms of tumor spread within the CSF pathways.
Collapse
Affiliation(s)
- Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Dezhuang Ye
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Yimei Yue
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Lihua Yang
- Department of Pediatrics, Washington University in St. Louis, St Louis, Missouri, USA
| | - Christopher P Pacia
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Dakota DeFreitas
- Department of Neurosurgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Prabagaran Esakky
- Department of Neurosurgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David D Limbrick
- Department of Neurosurgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Joshua B Rubin
- Department of Pediatrics, Washington University in St. Louis, St Louis, Missouri, USA
| | - Hong Chen
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Jennifer M Strahle
- Department of Neurosurgery, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
14
|
Pediatric midline H3K27M-mutant tumor with disseminated leptomeningeal disease and glioneuronal features: case report and literature review. Childs Nerv Syst 2021; 37:2347-2356. [PMID: 32989496 DOI: 10.1007/s00381-020-04892-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND H3K27M-mutant midline lesions were recently reclassified by the World Health Organization (WHO) as "diffuse midline glioma" (DMG) based entirely on their molecular signature. DMG is one of the most common and most lethal pediatric brain tumors; terminal progression is typically caused by local midbrain or brainstem progression, or secondary leptomeningeal dissemination. H3K27M mutations have also been infrequently associated with a histologically and prognostically diverse set of lesions, particularly spinal masses with early leptomeningeal spread. CASE PRESENTATION A 15-year-old girl after 1 week of symptoms was found to have a T2/FLAIR-hyperintense and contrast-enhancing thalamic mass accompanied by leptomeningeal enhancement along the entire neuraxis. Initial infectious workup was negative, and intracranial biopsy was inconclusive. Spinal arachnoid biopsy revealed an H3K27M-mutant lesion with glioneuronal features, classified thereafter as DMG. She received craniospinal irradiation with a boost to the thalamic lesion. Imaging 1-month post-radiation demonstrated significant treatment response with residual enhancement at the conus. CONCLUSIONS This case report describes the unique presentation of an H3K27M-mutant midline lesion with significant craniospinal leptomeningeal spread on admission and atypical glioneuronal histopathological markers. With such florid leptomeningeal disease, spinal dural biopsy should be considered earlier given its diagnostic yield in classifying the lesion as DMG. Consistent with similar prior reports, this lesion additionally demonstrated synaptophysin positivity-also potentially consistent with a diagnosis of diffuse leptomeningeal glioneuronal tumor (DLGNT). In atypical DMG cases, particularly with leptomeningeal spread, further consideration of clinical and histopathological context is necessary for accurate diagnosis and prognostication.
Collapse
|
15
|
Leach JL, Roebker J, Schafer A, Baugh J, Chaney B, Fuller C, Fouladi M, Lane A, Doughman R, Drissi R, DeWire-Schottmiller M, Ziegler DS, Minturn JE, Hansford JR, Wang SS, Monje-Deisseroth M, Fisher PG, Gottardo NG, Dholaria H, Packer R, Warren K, Leary SES, Goldman S, Bartels U, Hawkins C, Jones BV. MR imaging features of diffuse intrinsic pontine glioma and relationship to overall survival: report from the International DIPG Registry. Neuro Oncol 2021; 22:1647-1657. [PMID: 32506137 DOI: 10.1093/neuonc/noaa140] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND This study describes imaging features of diffuse intrinsic pontine glioma (DIPG) and correlates with overall survival (OS) and histone mutation status in the International DIPG Registry (IDIPGR). METHODS Four hundred cases submitted to the IDIPGR with a local diagnosis of DIPG and baseline MRI were evaluated by consensus review of 2 neuroradiologists; 43 cases were excluded (inadequate imaging or alternative diagnoses). Agreement between reviewers, association with histone status, and univariable and multivariable analyses relative to OS were assessed. RESULTS On univariable analysis imaging features significantly associated with worse OS included: extrapontine extension, larger size, enhancement, necrosis, diffusion restriction, and distant disease. On central review, 9.5% of patients were considered not to have DIPG. There was moderate mean agreement of MRI features between reviewers. On multivariable analysis, chemotherapy, age, and distant disease were predictors of OS. There was no difference in OS between wild-type and H3 mutated cases. The only imaging feature associated with histone status was the presence of ill-defined signal infiltrating pontine fibers. CONCLUSIONS Baseline imaging features are assessed in the IDIPGR. There was a 9.5% discordance in DIPG diagnosis between local and central review, demonstrating need for central imaging confirmation for prospective trials. Although several imaging features were significantly associated with OS (univariable), only age and distant disease were significant on multivariable analyses. There was limited association of imaging features with histone mutation status, although numbers are small and evaluation exploratory.
Collapse
Affiliation(s)
- James L Leach
- Department of Radiology and Medical Imaging, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - James Roebker
- Department of Radiology and Medical Imaging, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Austin Schafer
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joshua Baugh
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Neuro-oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Brooklyn Chaney
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Christine Fuller
- Department of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Maryam Fouladi
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Adam Lane
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Renee Doughman
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Rachid Drissi
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | | | - Jane E Minturn
- Division of Oncology, Children's Hospital of Philadelphia, Pennsylvania
| | - Jordan R Hansford
- Children's Cancer Centre, Royal Children's Hospital; Murdoch Children's Research Institute; University of Melbourne, Melbourne, Australia
| | - Stacie S Wang
- Children's Cancer Centre, Royal Children's Hospital; Murdoch Children's Research Institute; University of Melbourne, Melbourne, Australia
| | | | - Paul G Fisher
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, California
| | | | - Hetal Dholaria
- Department of Oncology, Perth Children's Hospital, Perth, AU
| | - Roger Packer
- Division of Oncology, Children's National Medical Center, Washington, DC
| | - Katherine Warren
- Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Harvard Cancer Center, Boston Massachusetts
| | - Sarah E S Leary
- Cancer and Blood Disorders Center, Seattle Children's, Seattle, Washington
| | - Stewart Goldman
- Division of Oncology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Ute Bartels
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, CA
| | - Cynthia Hawkins
- Division of Pathology, The Hospital for Sick Children, Toronto, CA
| | - Blaise V Jones
- Department of Radiology and Medical Imaging, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
16
|
Perez-Somarriba M, Santa-Maria V, Cruz O, Muchart J, Lavarino C, Mico S, Morales La Madrid A. Craniospinal irradiation as a salvage treatment for metastatic relapsed DIPG. Pediatr Blood Cancer 2021; 68:e28762. [PMID: 33063935 DOI: 10.1002/pbc.28762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/28/2020] [Indexed: 12/27/2022]
Affiliation(s)
| | - Vicente Santa-Maria
- Department of Pediatric Oncology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Ofelia Cruz
- Department of Pediatric Oncology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Jordi Muchart
- Department of Radiology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Cinzia Lavarino
- Department of Developmental Tumor Biology Laboratory, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Soraya Mico
- Department of Radiation Oncology, Hospital Vall d'Hebron, Barcelona, Spain
| | | |
Collapse
|
17
|
Rigamonti A, Simonetti G, Silvani A, Rudà R, Franchino F, Villani V, Pace A, Merli R, Servida M, Picca A, Berzero G, Cerase A, Chiarotti I, Spena G, Salmaggi A. Adult brainstem glioma: a multicentre retrospective analysis of 47 Italian patients. Neurol Sci 2020; 42:1879-1886. [PMID: 32954462 DOI: 10.1007/s10072-020-04725-7] [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: 06/21/2020] [Accepted: 09/15/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND AND PURPOSE Adult brainstem gliomas are rare primary brain tumours with heterogeneous clinical course. The low frequency of these tumours makes it difficult to achieve high-quality evidence regarding prognostic factors, adequate therapeutic approach and outcome in such patients. METHODS In this retrospective study, we analysed clinical, radiological, molecular, prognostic and therapeutic factors in a series of 47 histologically proven adult brainstem gliomas recruited over a 20-year period (1998-2018). RESULTS Twenty-two patients were male, 25 female with median age of 39 years. The tumour involved one brainstem segment in 20 cases and 2 or more segments in 27. Contrast enhancement was reported in 28 cases. Surgical procedures included biopsy in 26 cases and partial/total resection in the remaining 21. Histological diagnosis was of low-grade glioma in 23 patients, high-grade glioma in 22 and non-diagnostic in 2 cases. Data regarding molecular biology were available for 22 patients. Median overall survival was 35 months, in particular 16 months in high-grade glioma and 84 months in low-grade glioma. At univariate analysis, tumour grade was the only factor with a statistically significant impact on survival time (p = 0,003), whereas younger age, better performance status and total/subtotal resection showed a trend to more prolonged survival. This study also confirms safety of biopsy/surgery in adult brainstem glioma patients and shows a clear trend to a more frequent assessment of molecular biology data. CONCLUSIONS Further prospective multicentre efforts, and hopefully clinical trials, are necessary to improve outcome in this neglected glioma patient population.
Collapse
Affiliation(s)
- Andrea Rigamonti
- Neurosciences Department, Neurology Unit, Ospedale A. Manzoni, ASST Lecco, Via Dell'Eremo 9/11, 23900, Lecco, Italy.
| | - Giorgia Simonetti
- Clinical Neurooncology Unit, Neurooncology Department, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - Antonio Silvani
- Clinical Neurooncology Unit, Neurooncology Department, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - Roberta Rudà
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| | - Federica Franchino
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| | - Veronica Villani
- Neurooncology Unit, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Andrea Pace
- Neurooncology Unit, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | | | - Maura Servida
- Department of Neurology, ASST Ovest Milanese - Legnano Hospital, Legnano, Italy
| | - Alberto Picca
- Neurooncology Unit, IRCCS C. Mondino Foundation, Pavia, Italy
| | - Giulia Berzero
- Neurooncology Unit, IRCCS C. Mondino Foundation, Pavia, Italy
| | - Alfonso Cerase
- Unit of Neuroimaging - Diagnostic and Functional Neuroradiology Department of Neurological and Human Movement Sciences "Santa Maria alle Scotte", NHS & University General Hospital, Tuscany, Siena, Italy
| | - Ivano Chiarotti
- Unit of Neuroimaging - Diagnostic and Functional Neuroradiology Department of Neurological and Human Movement Sciences "Santa Maria alle Scotte", NHS & University General Hospital, Tuscany, Siena, Italy
| | - Giannantonio Spena
- Neurosciences Department, Neurology Unit, Ospedale A. Manzoni, ASST Lecco, Via Dell'Eremo 9/11, 23900, Lecco, Italy
| | - Andrea Salmaggi
- Neurosciences Department, Neurology Unit, Ospedale A. Manzoni, ASST Lecco, Via Dell'Eremo 9/11, 23900, Lecco, Italy
| | | |
Collapse
|
18
|
Erker C, Tamrazi B, Poussaint TY, Mueller S, Mata-Mbemba D, Franceschi E, Brandes AA, Rao A, Haworth KB, Wen PY, Goldman S, Vezina G, MacDonald TJ, Dunkel IJ, Morgan PS, Jaspan T, Prados MD, Warren KE. Response assessment in paediatric high-grade glioma: recommendations from the Response Assessment in Pediatric Neuro-Oncology (RAPNO) working group. Lancet Oncol 2020; 21:e317-e329. [PMID: 32502458 DOI: 10.1016/s1470-2045(20)30173-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/05/2020] [Accepted: 03/12/2020] [Indexed: 12/27/2022]
Abstract
Response criteria for paediatric high-grade glioma vary historically and across different cooperative groups. The Response Assessment in Neuro-Oncology working group developed response criteria for adult high-grade glioma, but these were not created to meet the unique challenges in children with the disease. The Response Assessment in Pediatric Neuro-Oncology (RAPNO) working group, consisting of an international panel of paediatric and adult neuro-oncologists, clinicians, radiologists, radiation oncologists, and neurosurgeons, was established to address issues and unique challenges in assessing response in children with CNS tumours. We established a subcommittee to develop response assessment criteria for paediatric high-grade glioma. Current practice and literature were reviewed to identify major challenges in assessing the response of paediatric high-grade gliomas to various treatments. For areas in which scientific investigation was scarce, consensus was reached through an iterative process. RAPNO response assessment recommendations include the use of MRI of the brain and the spine, assessment of clinical status, and the use of corticosteroids or antiangiogenics. Imaging standards for brain and spine are defined. Compared with the recommendations for the management of adult high-grade glioma, for paediatrics there is inclusion of diffusion-weighted imaging and a higher reliance on T2-weighted fluid-attenuated inversion recovery. Consensus recommendations and response definitions have been established and, similar to other RAPNO recommendations, prospective validation in clinical trials is warranted.
Collapse
Affiliation(s)
- Craig Erker
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Dalhousie University and IWK Health Centre, Halifax, NS, Canada.
| | - Benita Tamrazi
- Department of Radiology, Keck School of Medicine, University of Southern California and Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | - Tina Y Poussaint
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA
| | - Sabine Mueller
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA; Department of Neurosurgery, University of California San Francisco, San Francisco, CA, USA
| | - Daddy Mata-Mbemba
- Department of Diagnostic Imaging, Dalhousie University and IWK Health Centre, Halifax, NS, Canada
| | - Enrico Franceschi
- Department of Medical Oncology, Azienda USL, Bologna, Italy; IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Alba A Brandes
- Department of Medical Oncology, Azienda USL, Bologna, Italy; IRCCS Institute of Neurological Sciences, Bologna, Italy
| | - Arvind Rao
- Departments of Computational Medicine and Bioinformatics and Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Kellie B Haworth
- Division of Neuro-Oncology, Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stewart Goldman
- Department of Haematology, Oncology, Neuro-Oncology, and Stem Cell Transplantation, Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Gilbert Vezina
- Department of Radiology, Children's National Medical Center, Washington, DC, USA
| | - Tobey J MacDonald
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Ira J Dunkel
- Department of Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul S Morgan
- Department of Medical Physics and Clinical Engineering, Nottingham University Hospitals, Queen's Medical Centre, Nottingham, UK
| | - Tim Jaspan
- Department of Radiology, Nottingham University Hospitals, Queen's Medical Centre, Nottingham, UK
| | - Michael D Prados
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA, USA
| | - Katherine E Warren
- Department of Pediatric Oncology, Dana- Farber/Boston Children's Cancer and Blood Disorders Center, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
19
|
Cooney TM, Cohen KJ, Guimaraes CV, Dhall G, Leach J, Massimino M, Erbetta A, Chiapparini L, Malbari F, Kramer K, Pollack IF, Baxter P, Laughlin S, Patay Z, Young Poussaint T, Warren KE. Response assessment in diffuse intrinsic pontine glioma: recommendations from the Response Assessment in Pediatric Neuro-Oncology (RAPNO) working group. Lancet Oncol 2020; 21:e330-e336. [PMID: 32502459 DOI: 10.1016/s1470-2045(20)30166-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/25/2020] [Accepted: 03/04/2020] [Indexed: 12/20/2022]
Abstract
Optimising the conduct of clinical trials for diffuse intrinsic pontine glioma involves use of consistent, objective disease assessments and standardised response criteria. The Response Assessment in Pediatric Neuro-Oncology working group, consisting of an international panel of paediatric and adult neuro-oncologists, clinicians, radiologists, radiation oncologists, and neurosurgeons, was established to address issues and unique challenges in assessing response in children with CNS tumours. A working group was formed specifically to address response assessment in children and young adults with diffuse intrinsic pontine glioma and to develop a consensus on recommendations for response assessment. Response should be assessed using MRI of brain and spine, neurological examination, and anti-inflammatory or antiangiogenic drugs. Clinical imaging standards are defined. As with previous consensus recommendations, these recommendations will need to be validated in prospective clinical trials.
Collapse
Affiliation(s)
- Tabitha M Cooney
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kenneth J Cohen
- Departments of Pediatrics and Oncology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Girish Dhall
- Department of Pediatrics, Division of Hematology-Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James Leach
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Maura Massimino
- Department of Pediatric Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Erbetta
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Luisa Chiapparini
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fatema Malbari
- Department of Pediatrics, Section of Neurology and Developmental Neurosciences, Texas Children's Hospital, Houston, TX, USA
| | - Kim Kramer
- Department of Pediatric Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ian F Pollack
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Patricia Baxter
- Section of Pediatric Hematology-Oncology, Texas Children's Hospital, Houston, TX, USA
| | - Suzanne Laughlin
- Department of Medical Imaging, The Hospital for Sick Children, Toronto, ON, Canada
| | - Zoltán Patay
- Department of Radiology, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Katherine E Warren
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
20
|
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
|
21
|
Rodriguez Gutierrez D, Jones C, Varlet P, Mackay A, Warren D, Warmuth-Metz M, Aliaga ES, Calmon R, Hargrave DR, Cañete A, Massimino M, Azizi AA, Le Deley MC, Saran F, Rousseau RF, Zahlmann G, Garcia J, Vassal G, Grill J, Morgan PS, Jaspan T. Radiological Evaluation of Newly Diagnosed Non-Brainstem Pediatric High-Grade Glioma in the HERBY Phase II Trial. Clin Cancer Res 2020; 26:1856-1865. [PMID: 31924736 DOI: 10.1158/1078-0432.ccr-19-3154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/25/2019] [Accepted: 01/07/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE The HERBY trial evaluated the benefit of the addition of the antiangiogenic agent Bevacizumab (BEV) to radiotherapy/temozolomide (RT/TMZ) in pediatric patients with newly diagnosed non-brainstem high-grade glioma (HGG). The work presented here aims to correlate imaging characteristics and outcome measures with pathologic and molecular data. EXPERIMENTAL DESIGN Radiological, pathologic, and molecular data were correlated with trial clinical information to retrospectively re-evaluate event-free survival (EFS) and overall survival (OS). RESULTS One-hundred thirteen patients were randomized to the RT/TMZ arm (n = 54) or the RT/TMZ+BEV (BEV arm; n = 59). The tumor arose in the cerebral hemispheres in 68 patients (Cerebral group) and a midline location in 45 cases (Midline group). Pathologic diagnosis was available in all cases and molecular data in 86 of 113. H3 K27M histone mutations were present in 23 of 32 Midline cases and H3 G34R/V mutations in 7 of 54 Cerebral cases. Total/near-total resection occurred in 44 of 68 (65%) Cerebral cases but in only 5 of 45 (11%) Midline cases (P < 0.05). Leptomeningeal metastases (27 cases, 13 with subependymal spread) at relapse were more frequent in Midline (17/45) than in Cerebral tumors (10/68, P < 0.05). Mean OS (14.1 months) and EFS (9.0 months) in Midline tumors were significantly lower than mean OS (20.7 months) and EFS (14.9 months) in Cerebral tumors (P < 0.05). Pseudoprogression occurred in 8 of 111 (6.2%) cases. CONCLUSIONS This study has shown that the poor outcome of midline tumors (compared with cerebral) may be related to (1) lesser surgical resection, (2) H3 K27M histone mutations, and (3) higher leptomeningeal dissemination.
Collapse
Affiliation(s)
- Daniel Rodriguez Gutierrez
- Medical Physics and Clinical Engineering, Nottingham University Hospital Trust, Nottingham, United Kingdom.
- Division of Clinical Neuroscience, University of Nottingham, Nottingham, United Kingdom
| | - Chris Jones
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Pascale Varlet
- Anatomie et cytologie pathologiques, Centre Hospitalier Sainte Anne, Paris, France
| | - Alan Mackay
- Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Daniel Warren
- Department of Radiology, Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Monika Warmuth-Metz
- Institute for Diagnostic and Interventional Neuroradiology, Würzburg University, Würzburg, Germany
| | - Esther Sánchez Aliaga
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Raphael Calmon
- Pediatric Radiology, Necker Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Darren R Hargrave
- Haematology and Oncology Department, Great Ormond Street Hospital, London, United Kingdom
| | - Adela Cañete
- Pediatric Oncology and Hematology Unit, Hospital La Fe, Valencia, Spain
| | - Maura Massimino
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Amedeo A Azizi
- Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Medical University of Vienna, Vienna, Austria
| | - Marie-Cécile Le Deley
- Pediatric and Adolescent Oncology and Unite Mixte de Recherche, Gustave Roussy, Université Paris-Saclay, Université Paris-Sud, Villejuif, France
| | - Frank Saran
- Neuro-oncology Unit, Royal Marsden Hospital, London, United Kingdom
| | | | | | | | - Gilles Vassal
- Pediatric and Adolescent Oncology and Unite Mixte de Recherche, Gustave Roussy, Université Paris-Saclay, Université Paris-Sud, Villejuif, France
| | - Jacques Grill
- Pediatric and Adolescent Oncology and Unite Mixte de Recherche, Gustave Roussy, Université Paris-Saclay, Université Paris-Sud, Villejuif, France
| | - Paul S Morgan
- Medical Physics and Clinical Engineering, Nottingham University Hospital Trust, Nottingham, United Kingdom
- Division of Clinical Neuroscience, University of Nottingham, Nottingham, United Kingdom
| | - Tim Jaspan
- Department of Radiology, Nottingham University Hospital Trust, Nottingham, United Kingdom
| |
Collapse
|
22
|
Green AL, Flannery P, Hankinson TC, O'Neill B, Amani V, DeSisto J, Knox A, Chatwin H, Lemma R, Hoffman LM, Mulcahy Levy J, Raybin J, Hemenway M, Gilani A, Koschmann C, Dahl N, Handler M, Pierce A, Venkataraman S, Foreman N, Vibhakar R, Wempe MF, Dorris K. Preclinical and clinical investigation of intratumoral chemotherapy pharmacokinetics in DIPG using gemcitabine. Neurooncol Adv 2020; 2:vdaa021. [PMID: 32642682 PMCID: PMC7212907 DOI: 10.1093/noajnl/vdaa021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Background Hundreds of systemic chemotherapy trials in diffuse intrinsic pontine glioma (DIPG) have not improved survival, potentially due to lack of intratumoral penetration, which has not previously been assessed in humans. Methods We used gemcitabine as a model agent to assess DIPG intratumoral pharmacokinetics (PK) using mass spectrometry. Results In a phase 0 clinical trial of i.v. gemcitabine prior to biopsy in children newly diagnosed with DIPG by MRI, mean concentration in 4 biopsy cores in patient 1 (H3K27M diffuse midline glioma) was 7.65 µM. These compare favorably to levels for patient 2 (mean 3.85 µM, found to have an H3K27-wildtype low-grade glioma on histology), and from a similar study in adult glioblastoma (adjusted mean 3.48 µM). In orthotopic patient-derived xenograft (PDX) models of DIPG and H3K27M-wildtype pediatric glioblastoma, gemcitabine levels and clearance were similar in tumor, pons, and cortex and did not depend on H3K27 mutation status or tumor location. Normalized gemcitabine levels were similar in patient 1 and the DIPG PDX. Conclusions These findings, while limited to one agent, provide preliminary evidence for the hypotheses that lack of intratumoral penetration is not why systemic chemotherapy has failed in DIPG, and orthotopic PDX models can adequately model intratumoral PK in human DIPG.
Collapse
Affiliation(s)
- Adam L Green
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Patrick Flannery
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Todd C Hankinson
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Neurosurgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Brent O'Neill
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Department of Neurosurgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Vladimir Amani
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - John DeSisto
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Aaron Knox
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Hannah Chatwin
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Rakeb Lemma
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Lindsey M Hoffman
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jean Mulcahy Levy
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jennifer Raybin
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Molly Hemenway
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ahmed Gilani
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Nathan Dahl
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Michael Handler
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Neurosurgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Angela Pierce
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Sujatha Venkataraman
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Nicholas Foreman
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Rajeev Vibhakar
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Michael F Wempe
- University of Colorado School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA
| | - Kathleen Dorris
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Aurora, Colorado, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado, USA.,Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
23
|
Kuzan-Fischer CM, Souweidane MM. The intersect of neurosurgery with diffuse intrinsic pontine glioma. J Neurosurg Pediatr 2019; 24:611-621. [PMID: 31786541 DOI: 10.3171/2019.5.peds18376] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/02/2019] [Indexed: 11/06/2022]
Abstract
An invited article highlighting diffuse intrinsic pontine glioma (DIPG) to celebrate the 75th Anniversary of the Journal of Neurosurgery, a journal known to define surgical nuance and enterprise, is paradoxical since DIPG has long been relegated to surgical abandonment. More recently, however, the neurosurgeon is emerging as a critical stakeholder given our role in tissue sampling, collaborative scientific research, and therapeutic drug delivery. The foundation for this revival lies in an expanding reliance on tissue accession for understanding tumor biology, available funding to fuel research, and strides with interventional drug delivery.
Collapse
Affiliation(s)
| | - Mark M Souweidane
- Departments of1Neurological Surgery and
- 2Pediatrics, Weill Cornell Medical College, NewYork-Presbyterian Hospital, New York; and
- 3Department of Neurological Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
24
|
Lobon-Iglesias MJ, Santa-Maria Lopez V, Puerta Roldan P, Candela-Cantó S, Ramos-Albiac M, Gomez-Chiari M, Puget S, Bolle S, Goumnerova L, Kieran MW, Cruz O, Grill J, Morales La Madrid A. Tumor dissemination through surgical tracts in diffuse intrinsic pontine glioma. J Neurosurg Pediatr 2018; 22:678-683. [PMID: 30192215 DOI: 10.3171/2018.6.peds17658] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/12/2018] [Indexed: 11/06/2022]
Abstract
OBJECTIVEDiffuse intrinsic pontine glioma (DIPG) is a highly aggressive and lethal brainstem tumor in children. In the 1980s, routine biopsy at presentation was abandoned since it was claimed "unnecessary" for diagnosis. In the last decade, however, several groups have reincorporated this procedure as standard of care or in the context of clinical trials. Expert neurosurgical teams report no mortality and acceptable morbidity, and no relevant complications have been previously described. The aim of this study was to review needle tract dissemination as a potential complication in DIPG.METHODSThe authors retrospectively analyzed the incidence of dissemination through surgical tracts in DIPG patients who underwent biopsy procedures at diagnosis in 3 dedicated centers. Clinical records and images as well as radiation dosimetry from diagnosis to relapse were reviewed.RESULTSFour patients (2 boys and 2 girls, age range 6-12 years) had surgical tract dissemination: in 3 cases in the needle tract and in 1 case in the Ommaya catheter tract. The median time from biopsy to identification of dissemination was 5 months (range 4-6 months). The median overall survival was 11 months (range 7-12 months). Disseminated lesions were in the marginal radiotherapy field (n = 2), out of the field (n = 1), and in the radiotherapy field (n = 1).CONCLUSIONSAlthough surgical tract dissemination in DIPG is a rare complication (associated with 2.4% of procedures in this study), it should be mentioned to patients and family when procedures involving a surgical tract are proposed. The inclusion of the needle tract in the radiotherapy field may have only limited benefit. Future studies are warranted to explore the benefit of larger radiotherapy fields in patients with DIPG.
Collapse
Affiliation(s)
- Maria-Jesus Lobon-Iglesias
- 1Department of Pediatric and Adolescent Oncology and
- 2Team "Target Identification and Innovative Anticancer Therapies in Pediatric Cancers," Centre National de la Recherche Scientifique Unité Mixte de Recherche 8203, Villejuif
| | - Vicente Santa-Maria Lopez
- 3Department of Pediatric Hematology and Oncology
- 4Pediatric Neuro-Oncology, Department of Pediatric Hematology and Oncology
| | | | | | | | | | - Stephanie Puget
- 8Department of Pediatric Neurosurgery, Necker Sick Children's Hospital and University Paris-Descartes, Paris, France
| | - Stephanie Bolle
- 9Department of Radiation Therapy, Gustave Roussy and University Paris-Saclay, Villejuif
| | | | - Mark W Kieran
- 11The Pediatric Brain Tumor Program, Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Ofelia Cruz
- 3Department of Pediatric Hematology and Oncology
- 4Pediatric Neuro-Oncology, Department of Pediatric Hematology and Oncology
| | - Jacques Grill
- 1Department of Pediatric and Adolescent Oncology and
- 2Team "Target Identification and Innovative Anticancer Therapies in Pediatric Cancers," Centre National de la Recherche Scientifique Unité Mixte de Recherche 8203, Villejuif
| | - Andres Morales La Madrid
- 3Department of Pediatric Hematology and Oncology
- 4Pediatric Neuro-Oncology, Department of Pediatric Hematology and Oncology
| |
Collapse
|
25
|
Convection-enhanced delivery: chemosurgery in diffuse intrinsic pontine glioma. Lancet Oncol 2018; 19:1001-1003. [PMID: 29914797 DOI: 10.1016/s1470-2045(18)30408-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 05/22/2018] [Indexed: 02/03/2023]
|
26
|
Souweidane MM, Kramer K, Pandit-Taskar N, Zhou Z, Haque S, Zanzonico P, Carrasquillo JA, Lyashchenko SK, Thakur SB, Donzelli M, Turner RS, Lewis JS, Cheung NKV, Larson SM, Dunkel IJ. Convection-enhanced delivery for diffuse intrinsic pontine glioma: a single-centre, dose-escalation, phase 1 trial. Lancet Oncol 2018; 19:1040-1050. [PMID: 29914796 DOI: 10.1016/s1470-2045(18)30322-x] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 10/14/2022]
Abstract
BACKGROUND Diffuse intrinsic pontine glioma is one of the deadliest central nervous system tumours of childhood, with a median overall survival of less than 12 months. Convection-enhanced delivery has been proposed as a means to efficiently deliver therapeutic agents directly into the brainstem while minimising systemic exposure and associated toxic effects. We did this study to evaluate the safety of convection-enhanced delivery of a radioimmunotherapy agent targeting the glioma-associated B7-H3 antigen in children with diffuse intrinsic pontine glioma. METHODS We did a phase 1, single-arm, single-centre, dose-escalation study at the Memorial Sloan Kettering Cancer Center (New York, NY, USA). Eligible patients were aged 3-21 years and had diffuse intrinsic pontine glioma as diagnosed by consensus of a multidisciplinary paediatric neuro-oncology team; a Lansky (patients <16 years of age) or Karnofsky (patients ≥16 years) performance score of at least 50 at study entry; a minimum weight of 8 kg; and had completed external beam radiation therapy (54·0-59·4 Gy at 1·8 Gy per fraction over 30-33 fractions) at least 4 weeks but no more than 14 weeks before enrolment. Seven dose-escalation cohorts were planned based on standard 3 + 3 rules: patients received a single infusion of 9·25, 18·5, 27·75, 37, 92·5, 120·25, or 148 MBq, respectively, at a concentration of about 37 MBq/mL by convection-enhanced delivery of the radiolabelled antibody [124I]-8H9. The primary endpoint was identification of the maximum tolerated dose. The analysis of the primary endpoint was done in the per-protocol population (patients who received the full planned dose of treatment), and all patients who received any dose of study treatment were included in the safety analysis. This study is registered with ClinicalTrials.gov, number NCT01502917, and is ongoing with an expanded cohort. FINDINGS From April 5, 2012, to Oct 8, 2016, 28 children were enrolled and treated in the trial, of whom 25 were evaluable for the primary endpoint. The maximum tolerated dose was not reached as no dose-limiting toxicities were observed. One (4%) of 28 patients had treatment-related transient grade 3 hemiparesis and one (4%) had grade 3 skin infection. No treatment-related grade 4 adverse events or deaths occurred. Estimated volumes of distribution (Vd) were linearly dependent on volumes of infusion (Vi) and ranged from 1·5 to 20·1 cm3, with a mean Vd/Vi ratio of 3·4 (SD 1·2). The mean lesion absorbed dose was 0·39 Gy/MBq 124I (SD 0·20). Systemic exposure was negligible, with an average lesion-to-whole body ratio of radiation absorbed dose higher than 1200. INTERPRETATION Convection-enhanced delivery in the brainstem of children with diffuse intrinsic pontine glioma who have previously received radiation therapy seems to be a rational and safe therapeutic strategy. PET-based dosimetry of the radiolabelled antibody [124I]-8H9 validated the principle of using convection-enhanced delivery in the brain to achieve high intra-lesional dosing with negligible systemic exposure. This therapeutic strategy warrants further development for children with diffuse intrinsic pontine glioma. FUNDING National Institutes of Health, The Dana Foundation, The Cure Starts Now, Solving Kids' Cancer, The Lyla Nsouli Foundation, Cookies for Kids' Cancer, The Cristian Rivera Foundation, Battle for a Cure, Cole Foundation, Meryl & Charles Witmer Charitable Foundation, Tuesdays with Mitch Charitable Foundation, and Memorial Sloan Kettering Cancer Center.
Collapse
Affiliation(s)
- Mark M Souweidane
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Neurological Surgery, Weill Medical College of Cornell University, New York, USA; Department of Pediatrics, Weill Medical College of Cornell University, New York, USA.
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pediatrics, Weill Medical College of Cornell University, New York, USA
| | - Neeta Pandit-Taskar
- Department of Radiology, Molecular Imaging and Therapy (Nuclear Medicine) Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Targeted Radioimmunotherapy and Theranostics, Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, USA
| | - Zhiping Zhou
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Neurological Surgery, Weill Medical College of Cornell University, New York, USA
| | - Sofia Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, USA
| | - Pat Zanzonico
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge A Carrasquillo
- Department of Radiology, Molecular Imaging and Therapy (Nuclear Medicine) Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Targeted Radioimmunotherapy and Theranostics, Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, USA
| | - Serge K Lyashchenko
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Radiochemistry & Molecular Imaging Probes Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, USA
| | - Sunitha B Thakur
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Donzelli
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryan S Turner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Radiochemistry & Molecular Imaging Probes Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, USA; Pharmacology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Steven M Larson
- Department of Radiology, Molecular Imaging and Therapy (Nuclear Medicine) Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Targeted Radioimmunotherapy and Theranostics, Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA; Department of Radiology, Weill Medical College of Cornell University, New York, USA
| | - Ira J Dunkel
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pediatrics, Weill Medical College of Cornell University, New York, USA
| |
Collapse
|
27
|
Cohen KJ, Jabado N, Grill J. Diffuse intrinsic pontine gliomas-current management and new biologic insights. Is there a glimmer of hope? Neuro Oncol 2018; 19:1025-1034. [PMID: 28371920 DOI: 10.1093/neuonc/nox021] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) has proven to be one of the most challenging of all pediatric cancers. Owing to a historical reticence to obtain tumor tissue for study, and based on an erroneous assumption that the biology of DIPG would mirror that of supratentorial high-grade astrocytomas, innumerable studies have been undertaken-all of which have had a negligible impact on the natural history of this disease. More recently, improvements in neurosurgical techniques have allowed for the safe upfront biopsy of DIPG, which, together with a wider use of autopsy tissue, has led to an evolving understanding of the biology of this tumor. The discovery of a recurrent somatic gain-of-function mutation leading to lysine 27 to methionine (p.Lys27Met, K27M) substitution in histone 3 variants characterizes more than 85% of DIPG, suggesting for the first time the role of the epigenome and histones in the pathogenesis of this disease, and more unified diagnostic criteria. Along with further molecular insights into the pathogenesis of DIPG, rational targets are being identified and studied in the hopes of improving the otherwise dismal outcome for children with DIPG.
Collapse
Affiliation(s)
- Kenneth J Cohen
- Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland; Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Université Paris-Saclay & Gustave Roussy Unité Mixte de Recherche 8203 du Centre National de la Recherche Scientifique & Departement de Cancerologie de l'Enfant et de l'Adolescent, Villejuif, France
| | - Nada Jabado
- Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland; Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Université Paris-Saclay & Gustave Roussy Unité Mixte de Recherche 8203 du Centre National de la Recherche Scientifique & Departement de Cancerologie de l'Enfant et de l'Adolescent, Villejuif, France
| | - Jacques Grill
- Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland; Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Université Paris-Saclay & Gustave Roussy Unité Mixte de Recherche 8203 du Centre National de la Recherche Scientifique & Departement de Cancerologie de l'Enfant et de l'Adolescent, Villejuif, France
| |
Collapse
|
28
|
Spreafico F, Bongarzone I, Pizzamiglio S, Magni R, Taverna E, De Bortoli M, Ciniselli CM, Barzanò E, Biassoni V, Luchini A, Liotta LA, Zhou W, Signore M, Verderio P, Massimino M. Proteomic analysis of cerebrospinal fluid from children with central nervous system tumors identifies candidate proteins relating to tumor metastatic spread. Oncotarget 2018; 8:46177-46190. [PMID: 28526811 PMCID: PMC5542258 DOI: 10.18632/oncotarget.17579] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 04/11/2017] [Indexed: 12/11/2022] Open
Abstract
Central nervous system (CNS) tumors are the most common solid tumors in childhood. Since the sensitivity of combined cerebrospinal fluid (CSF) cytology and radiological neuroimaging in detecting meningeal metastases remains relatively low, we sought to characterize the CSF proteome of patients with CSF tumors to identify biomarkers predictive of metastatic spread. CSF samples from 27 children with brain tumors and 13 controls (extra-CNS non-Hodgkin lymphoma) were processed using core-shell hydrogel nanoparticles, and analyzed with reverse-phase liquid chromatography/electrospray tandem mass spectrometry (LC-MS/MS). Candidate proteins were identified with Fisher's exact test and/or a univariate logistic regression model. Reverse phase protein array (RPPA), Western blot (WB), and ELISA were used in the training set and in an independent set of CFS samples (60 cases, 14 controls) to validate our discovery findings. Among the 558 non-redundant proteins identified by LC-MS/MS, 147 were missing from the CSF database at http://www.biosino.org. Fourteen of the 26 final top-candidate proteins were chosen for validation with WB, RPPA and ELISA methods. Six proteins (type 1 collagen, insulin-like growth factor binding protein 4, procollagen C-endopeptidase enhancer 1, glial cell-line derived neurotrophic factor receptor α2, inter-alpha-trypsin inhibitor heavy chain 4, neural proliferation and differentiation control protein-1) revealed the ability to discriminate metastatic cases from controls. Combining a unique dataset of CSFs from pediatric CNS tumors with a novel enabling nanotechnology led us to identify CSF proteins potentially related to metastatic status.
Collapse
Affiliation(s)
- Filippo Spreafico
- Pediatric Oncology Unit, Department of Hematology and Pediatric Hematology-Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Italia Bongarzone
- Proteomics Laboratory, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Pizzamiglio
- Unit of Medical Statistics, Biometry and Bioinformatics, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ruben Magni
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Elena Taverna
- Proteomics Laboratory, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maida De Bortoli
- Proteomics Laboratory, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara M Ciniselli
- Unit of Medical Statistics, Biometry and Bioinformatics, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Barzanò
- Pediatric Oncology Unit, Department of Hematology and Pediatric Hematology-Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Veronica Biassoni
- Pediatric Oncology Unit, Department of Hematology and Pediatric Hematology-Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Luchini
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Lance A Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Michele Signore
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Paolo Verderio
- Unit of Medical Statistics, Biometry and Bioinformatics, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maura Massimino
- Pediatric Oncology Unit, Department of Hematology and Pediatric Hematology-Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
29
|
Nazarian J, Mason GE, Ho CY, Panditharatna E, Kambhampati M, Vezina LG, Packer RJ, Hwang EI. Histological and molecular analysis of a progressive diffuse intrinsic pontine glioma and synchronous metastatic lesions: a case report. Oncotarget 2018; 7:42837-42842. [PMID: 27329600 PMCID: PMC5173175 DOI: 10.18632/oncotarget.10034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 05/25/2016] [Indexed: 11/25/2022] Open
Abstract
There is no curative treatment for patients with diffuse intrinsic pontine glioma (DIPG). However, with the recent availability of biopsy and autopsy tissue, new data regarding the biologic behavior of this tumor have emerged, allowing greater molecular characterization and leading to investigations which may result in improved therapeutic options. Treatment strategies must address both primary disease sites as well as any metastatic deposits, which may be variably sensitive to a particular approach. In this case report, we present a patient with DIPG treated with irradiation and serial investigational agents. The clinical, pathological and molecular phenotypes of both the progressive primary tumor as well as concomitant metastatic deposits obtained at autopsy are discussed. While some mRNA differences were demonstrated, all analyzed sites of disease shared similar mutational arrangements, suggesting that targeting the mutations of the primary tumor may be effective for all sites of disease.
Collapse
Affiliation(s)
- Javad Nazarian
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA.,Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Gary E Mason
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cheng Ying Ho
- Department of Pathology, Children's National Medical Center, Washington, DC, USA
| | - Eshini Panditharatna
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA.,Institute for Biomedical Sciences, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Madhuri Kambhampati
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
| | - L Gilbert Vezina
- Division of Neuro-radiology, Children's National Medical Center, Washington, DC, USA
| | - Roger J Packer
- Brain Tumor Institute, Daniel and Jennifer Gilbert Neurofibromatosis Institute, Neuroscience and Behavioral Medicine, Children's National Medical Center, NW, Washington, DC, USA
| | - Eugene I Hwang
- Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC, USA
| |
Collapse
|
30
|
Wagner MW, Bell WR, Kern J, Bosemani T, Mhlanga J, Carson KA, Cohen KJ, Raabe EH, Rodriguez F, Huisman TAGM, Poretti A. Diffusion tensor imaging suggests extrapontine extension of pediatric diffuse intrinsic pontine gliomas. Eur J Radiol 2016; 85:700-6. [PMID: 26971411 DOI: 10.1016/j.ejrad.2016.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 01/12/2016] [Accepted: 02/03/2016] [Indexed: 11/30/2022]
Abstract
PURPOSE To apply DTI to detect early extrapontine extension of pediatric diffuse intrinsic pontine glioma along the corticospinal tracts. METHODS In children with diffuse intrinsic pontine glioma, low-grade brainstem glioma, and age-matched controls, DTI metrics were measured in the posterior limb of the internal capsule and posterior centrum semiovale. Histological examination was available in one patient. RESULTS 6 diffuse intrinsic pontine glioma, 8 low-grade brainstem glioma, and two groups of 25 controls were included. In diffuse intrinsic pontine glioma compared to controls, fractional anisotropy was lower in the bilateral posterior limb of the internal capsule, axial diffusivity was lower in the bilateral posterior centrum semiovale and posterior limb of the internal capsule, while radial diffusivity was higher in the bilateral posterior limb of the internal capsule. No significant differences were found between low-grade brainstem glioma and controls. In diffuse intrinsic pontine glioma compared to low-grade brainstem glioma, axial diffusivity was lower in the bilateral posterior limb of the internal capsule. Histological examination in one child showed tumor cells in the posterior limb of the internal capsule. CONCLUSION Reduction in fractional anisotropy and axial diffusivity and increase in radial diffusivity in diffuse intrinsic pontine glioma may reflect tumor extension along the corticospinal tracts as shown by histology. DTI may detect early extrapontine tumor extension in diffuse intrinsic pontine glioma before it becomes apparent on conventional MRI sequences.
Collapse
Affiliation(s)
- Matthias W Wagner
- Section of Pediatric Neuroradiology, Division of Pediatric Radiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - W Robert Bell
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jason Kern
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thangamadhan Bosemani
- Section of Pediatric Neuroradiology, Division of Pediatric Radiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joyce Mhlanga
- Section of Pediatric Neuroradiology, Division of Pediatric Radiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathryn A Carson
- Department of Epidemiology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Division of General Internal Medicine, Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Kenneth J Cohen
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric H Raabe
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fausto Rodriguez
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thierry A G M Huisman
- Section of Pediatric Neuroradiology, Division of Pediatric Radiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrea Poretti
- Section of Pediatric Neuroradiology, Division of Pediatric Radiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
31
|
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
|
32
|
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are a fairly common pediatric brain tumor, and children with these tumors have a dismal prognosis. They generally are diagnosed within the first decade of life, and due to their location within the pons, these tumors are not surgically resectable. The median survival for children with DIPGs is less than 1 year, in spite of decades of clinical trial development of unique approaches to radiation therapy and chemotherapy. Novel therapies are under investigation for these deadly tumors. As clinicians and researchers make a concerted effort to obtain tumor tissue, the molecular signals of these tumors are being investigated in an attempt to uncover targetable therapies for DIPGs. In addition, direct application of chemotherapies into the tumor (convection-enhanced delivery) is being investigated as a novel delivery system for treatment of DIPGs. Overall, DIPGs require creative thinking and a disciplined approach for development of a therapy that can improve the prognosis for these unfortunate children.
Collapse
Affiliation(s)
- Amy Lee Bredlau
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA; Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, USA.
| | - David N Korones
- Department of Pediatrics, University of Rochester, Rochester, New York, USA; Department of Palliative Care, University of Rochester, Rochester, New York, USA
| |
Collapse
|
33
|
Hargrave D. Pediatric diffuse intrinsic pontine glioma: can optimism replace pessimism? CNS Oncol 2015; 1:137-48. [PMID: 25057864 DOI: 10.2217/cns.12.15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pediatric diffuse intrinsic pontine glioma (DIPG) has a dismal prognosis that has not seen a change in outcome despite multiple clinical trials. Possible reasons for failure to make progress in this aggressive childhood brain tumor include: poor understanding of the underlying molecular biology due to lack of access to tumor material; absence of accurate and relevant DIPG preclinical models for drug development; ill-defined therapeutic targets for novel agents; and inadequate drug delivery to the brainstem. This review will demonstrate that systematic studies to identify solutions for each of these barriers is starting to deliver progress that can turn pessimism to optimism in DIPG.
Collapse
Affiliation(s)
- Darren Hargrave
- Department of Pediatric Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK.
| |
Collapse
|
34
|
Schindler G, Capper D, Korshunov A, Schmieder K, Brenke C. Spinal metastasis of gliosarcoma: Array-based comparative genomic hybridization for confirmation of metastatic spread. J Clin Neurosci 2014; 21:1945-50. [DOI: 10.1016/j.jocn.2014.03.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 03/19/2014] [Accepted: 03/29/2014] [Indexed: 10/25/2022]
|
35
|
Radiotherapy with concurrent and adjuvant temozolomide in children with newly diagnosed diffuse intrinsic pontine glioma. J Neurooncol 2011; 106:399-407. [PMID: 21858607 DOI: 10.1007/s11060-011-0681-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2011] [Accepted: 08/03/2011] [Indexed: 10/17/2022]
Abstract
The purpose of this study is to evaluate the efficacy and toxicity of radiation therapy (RT) with concurrent temozolomide (TMZ) chemotherapy followed by adjuvant TMZ in children with diffuse intrinsic pontine glioma (DIPG). Newly diagnosed patients younger than 18 years with histologically proven DIPG were treated with focal radiotherapy to a dose of 54 Gy in 30 fractions along with concurrent daily TMZ (75 mg/m(2)/day). Four weeks after completing the initial RT-TMZ schedule, adjuvant TMZ (200 mg/m(2)/day, days 1-5) was given every 28 days up to six cycles. Responses/progressions were assessed by clinical and 2-monthly MRI follow-up studies. Between September 2005 and September 2009, 21 patients with newly diagnosed histologically confirmed DIPG were eligible for this study. Median age at diagnosis was 6.4 years (range 4-16 years). At last update in August 2010, 17 children have died, 1 child was alive with progressive disease and 3 with stable disease. Metastatic relapse was documented in the cerebral site in two patients and in spinal cord in two cases. The median time to progression was 7.5 months (range 28 days-14.5 months) and the median survival was 11.7 months (range 26 days-17.5 months). The 1-year PFS and the 1-year OS were 33 and 50%, respectively. Five patients presented radiological findings compatible with pseudoprogression during the treatment. Haematological toxicity (Grade III/IV thrombocytopenia and leucopenia) was the most commonly found and led to dose reductions of TMZ in 58% of the patients. TMZ with radiation therapy has not yielded any significant improvement in outcome of children with DIPG and is associated with higher toxicity compared with radiotherapy alone. Novel treatment modalities are needed to improve the outcome of these patients.
Collapse
|
36
|
Wolff JE, Rytting ME, Vats TS, Zage PE, Ater JL, Woo S, Kuttesch J, Ketonen L, Mahajan A. Treatment of recurrent diffuse intrinsic pontine glioma: the MD Anderson Cancer Center experience. J Neurooncol 2011; 106:391-7. [PMID: 21858608 DOI: 10.1007/s11060-011-0677-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 07/30/2011] [Indexed: 01/27/2023]
Abstract
Recurrent diffuse intrinsic pontine gliomas (DIPG) are traditionally treated with palliative care since no effective treatments have been described for these tumors. Recently, clinical studies have been emerging, and individualized treatment is attempted more frequently. However, an informative way to compare the treatment outcomes has not been established, and historical control data are missing for recurrent disease. We conducted a retrospective chart review of patients with recurrent DIPG treated between 1998 and 2010. Response progression-free survival and possible influencing factors were evaluated. Thirty-one patients were identified who were treated in 61 treatment attempts using 26 treatment elements in 31 different regimens. The most frequently used drugs were etoposide (14), bevacizumab (13), irinotecan (13), nimotuzumab (13), and valproic acid (13). Seven patients had repeat radiation therapy to the primary tumor. Response was recorded after 58 treatment attempts and was comprised of 0 treatment attempts with complete responses, 7 with partial responses, 20 with stable diseases, and 31 with progressive diseases The median progression-free survival after treatment start was 0.16 years (2 months) and was found to be correlated to the prior time to progression but not to the number of previous treatment attempts. Repeat radiation resulted in the highest response rates (4/7), and the longest progression-free survival. These data provide a basis to plan future clinical trials for recurrent DIPG. Repeat radiation therapy should be tested in a prospective clinical study.
Collapse
Affiliation(s)
- Johannes E Wolff
- Department of Pediatrics, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Box 87, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Taylor M, Rössler J, Geoerger B, Vassal G, Farace F. New anti-angiogenic strategies in pediatric solid malignancies: agents and biomarkers of a near future. Expert Opin Investig Drugs 2010; 19:859-74. [PMID: 20470190 DOI: 10.1517/13543784.2010.487654] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IMPORTANCE OF THE FIELD Antiangiogenic strategies are affording considerable interest and have become a major milestone in therapeutics of various adult cancers. However, progress has been slow to expand such therapies to patients with pediatric solid malignancies. AREAS COVERED IN THIS REVIEW This review discusses the principal pathways for angiogenesis in pediatric solid malignancies and summarizes recent preclinical and clinical data on antiangiogenesis strategies in these tumors. WHAT THE READER WILL GAIN The reader will gain state-of-the-art knowledge in the current advancements of antiangiogenic therapies in pediatric clinical trials in regard to supporting preclinical data, and in the status of potential biomarkers investigated for monitoring angiogenesis inhibitors. Mechanisms of resistance to antiangiogenic therapy will also be discussed. Finally, we describe our experience in the monitoring of circulating endothelial cells and progenitors and their potential role as biomarkers of metastatic disease and resistance to antiangiogenic therapies. TAKE HOME MESSAGE Evaluation and development of antiangiogenesis protocols are starting and represent a crucial step in the management of pediatric solid malignancies today. Emphasis should be placed on the development of proper surrogate markers to monitor antiangiogenic activity and on the possible long-term effects of these therapies in a pediatric population.
Collapse
Affiliation(s)
- Melissa Taylor
- Translational Research Laboratory, Institut Gustave Roussy, Villejuif, France.
| | | | | | | | | |
Collapse
|
38
|
High dose methotrexate for pediatric high grade glioma: results of the HIT-GBM-D pilot study. J Neurooncol 2010; 102:433-42. [PMID: 20694800 DOI: 10.1007/s11060-010-0334-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 07/21/2010] [Indexed: 01/09/2023]
Abstract
We conducted a phase II study to test methotrexate (5 g/m(2)), as a single agent prior to radiochemotherapy for pediatric high-grade glioma and diffuse intrinsic pontine glioma. Thirty patients (19 male, median age 10.8) were enrolled. Tumors were located as follows: cortex 10, pons 7, other 13. Tumor resection was classified as gross total in 6, subtotal in 6, partial in 4, biopsy in 11 and not performed in 3. WHO grading of the histology was: IV: 11, III: 12 and II: 3. Patients received methotrexate 5 g/m(2) in 24-hour infusions on days 1 and 15. Subsequently 54 Gy radiation was administered with simultaneous chemotherapy including cisplatin, etoposide, vincristine and ifosfamide as previously described. Eight 6-weeks cycles of maintenance chemotherapy consisted of vincristine 1.5 mg/m(2) on days 1, 8 and 15; lomustine 100 mg/m(2) on day 2 and prednisone 40 mg/kg on days 1-17. Event-free survival rates in the whole group of 30 patients were: 43, 20, and 13% after 1, 2 and 5 years, respectively. The response evaluation after methotrexate was available in 19 of the 24 patients who started treatment with measurable disease: CR: 0, PR: 1, SD 18, PD: 0. After radiochemotherapy the response of 24 patients with measurable disease was CR: 1, PR 10, SD 12, PD 1. Both response and event-free survival were superior to the control group of 330 patients treated in various protocols of the same cooperative group. In subgroup analyses the use of dexamethasone during early treatment was linked to poor event free survival. Giving two cycles of high-dose methotrexate prior to radiochemotherapy was feasible, and the approach was taken forward to a randomized phase III trial.
Collapse
|
39
|
Sethi R, Allen J, Donahue B, Karajannis M, Gardner S, Wisoff J, Kunnakkat S, Mathew J, Zagzag D, Newman K, Narayana A. Prospective neuraxis MRI surveillance reveals a high risk of leptomeningeal dissemination in diffuse intrinsic pontine glioma. J Neurooncol 2010; 102:121-7. [PMID: 20623246 DOI: 10.1007/s11060-010-0301-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 06/28/2010] [Indexed: 01/28/2023]
Abstract
Prognosis of diffuse intrinsic pontine gliomas (DIPGs) remains poor. Failure has been predominantly local, with leptomeningeal dissemination (LD) occurring in 4-33% of patients in pre-MRI era series. Routine craniospinal imaging after initial treatment may reveal other relapse patterns relapse. Sixteen consecutive pediatric patients with DIPG treated between 2006 and 2009 were retrospectively reviewed. Treatment regimens, recurrence patterns, survival, and pathologic diagnosis were recorded. Fourteen patients received involved-field radiotherapy to 54 Gy, and two patients received craniospinal irradiation for LD at presentation. Neuraxis MRI was performed at diagnosis and at 4 month intervals following radiotherapy. Fifteen patients have had progression of disease (median progression-free survival 5.0 ± 1.2 months), and 13 patients have died (median survival 9.0 ± 1.4 months). Local failure occurred in 12 patients (75%). LD occurred in nine patients (56%). LD was present at diagnosis in three patients, after initial staging and treatment in six patients, and during autopsy in two patients. Median overall survival was 12.0 ± 3.3 months without LD and 8.0 ± 2.1 months with LD (P = 0.059, log rank test). Median progression-free survival was 9.5 ± 3.9 months without LD and 3.0 ± 2.1 months with LD (P = 0.012, log rank test). The high incidence of LD probably reflects liberal use of spine MRI surveillance. All patients should undergo routine craniospinal imaging at diagnosis and follow-up. Central nervous system prophylaxis should be considered in future clinical trials.
Collapse
Affiliation(s)
- Rajni Sethi
- Department of Radiation Oncology, New York University Langone Medical Center, 566 First Avenue, New York, NY 10014, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Symptomatic spinal metastases of intracranial glioblastoma: clinical characteristics and pathomechanism relating to GFAP expression. J Neurooncol 2010; 101:329-33. [PMID: 20549302 DOI: 10.1007/s11060-010-0257-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 05/24/2010] [Indexed: 10/19/2022]
Abstract
To demonstrate clinical characteristics of symptomatic spinal metastases of intracranial glioblastoma multiforme (GBM) and different spreading mechanisms relating to astrocytic cell differentiation, we present an extraordinary case of a 47-year-old patient with rapid progressive paraplegia due to coincident intramedullary and leptomeningeal dissemination of a supratentorial GBM. Serial biopsies of the intracranial, leptomeningeal, and intramedullary GBM lesions of our patient were analyzed for glial fibrillary acidic protein (GFAP). Furthermore, we present 19 additional cases of intracranial GBM with symptomatic spinal seeding, identified through literature review. GFAP expression was high in intracranial and intramedullary tumors, but low in leptomeningeal dissemination of our patient. Mean patient age was 45 years. Mean interval between identification of spinal metastases and death was 4.5 months. Mean overall survival was 18.6 months. Location of symptomatic spinal metastases was more frequently leptomeningeal (14 cases) than intramedullary (7 cases). The case presented herein supports the hypothesis of higher incidence of low GFAP expression in GBM cells in leptomeningeal manifestations after primary intracranial GBM. Because of the proposed tendency for early leptomeningeal spread from primary tumors with low astrocytic differentiation (low GFAP expression), patients with these tumors should be followed more closely to identify leptomeningeal tumor progression early on. Early identification of leptomeningeal spread could enable these patients to benefit from radiation therapy before they develop severe neurological deficits, which might translate into longer acceptable quality of life for these mostly young patients. This is an important finding, but further prospective studies are needed to verify our observations.
Collapse
|
41
|
Sato A, Sakurada K, Kumabe T, Sasajima T, Beppu T, Asano K, Ohkuma H, Ogawa A, Mizoi K, Tominaga T, Kitanaka C, Kayama T. Association of stem cell marker CD133 expression with dissemination of glioblastomas. Neurosurg Rev 2010; 33:175-83; discussion 183-4. [PMID: 20135187 DOI: 10.1007/s10143-010-0239-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 10/11/2009] [Accepted: 01/02/2010] [Indexed: 10/19/2022]
Abstract
Dissemination of glioblastoma was once considered rare but is now increasingly encountered with longer survival of glioblastoma patients. Despite the potential negative impact of dissemination on clinical outcome, however, molecular markers useful for prediction of dissemination risk still remains ill defined. We tested in this study for an association between the expression of stem cell marker CD133 and the risk of dissemination in 26 cases of glioblastoma (16 with dissemination and 10 without dissemination). The protein expression of CD133 was examined by western blot analysis of tumor specimens, and the CD133 expression levels were quantified by densitometry and normalized to beta-actin. The results indicated that CD133 expression levels are significantly higher in glioblastomas with dissemination (mean 10.3, range 0.20-27.8) than in those without (mean 1.18, range 0.07-3.58). The results suggest that CD133 could be a molecular predictor of glioblastoma dissemination, and also give rise to an intriguing idea that CD133-positive cancer stem cells may be implicated in the initiation of disseminated lesions.
Collapse
Affiliation(s)
- Atsushi Sato
- Department of Neurosurgery, Yamagata University School of Medicine, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Safety and toxicity of intrathecal liposomal cytarabine (Depocyte) in children and adolescents with recurrent or refractory brain tumors: a multi-institutional retrospective study. Anticancer Drugs 2009; 20:794-9. [PMID: 19617818 DOI: 10.1097/cad.0b013e32832f4abe] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
This retrospective study aimed to evaluate the safety and toxicity of intrathecal liposomal cytarabine (Depocyte) in children and adolescents with refractory or recurrent brain tumors. Nineteen heavily pretreated patients (males, n = 14; females, n = 5; median age at diagnosis 8.5 years; range, 1.4-22 years) were given intrathecal liposomal cytarabine on a compassionate use basis for recurrent refractory medulloblastoma (n = 12), mixed germ cell tumor (n = 2), central nervous system primitive neuroectodermal tumors of the pons (n = 1), anaplastic ependymoma (n = 1), anaplastic oligodendroglioma (n = 1), atypical teratoid rhabdoid tumor (n = 1), or rhabdoid papillary meningioma (n = 1). Eighteen patients received concomitant systemic radiochemotherapy. A total of 88 intrathecal injections of liposomal cytarabine (dose range, 20-50 mg) were administered with concomitant dexamethasone prophylaxis. The median number of doses per patient was four (range, 1-10). Duration of treatment ranged from (1/2) to 10 months. Eleven patients (57.9%) did not show any side effects, whereas eight patients (42.1%) developed side effects related to either chemical arachnoiditis (n = 4) or neurological progression (n = 2). Less typical treatment-related symptoms (e.g. lethargy, ataxia, and slurred speech) were observed in two patients. Treatment with intrathecal liposomal cytarabine was discontinued twice because of side effects. In conclusion, although intrathecal liposomal cytarabine was generally well tolerated, it should be used cautiously and only with dexamethasone prophylaxis in extensively pretreated patients with recurrent brain tumors. Proof of efficacy requires a prospective single-agent phase II study.
Collapse
|
43
|
Pytel P, Lukas RV. Update on diagnostic practice: tumors of the nervous system. Arch Pathol Lab Med 2009; 133:1062-77. [PMID: 19642733 DOI: 10.5858/133.7.1062] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2008] [Indexed: 11/06/2022]
Abstract
CONTEXT Changes in the practice of diagnosing brain tumors are formally reflected in the evolution of the World Health Organization classification. Beyond this classification, the practice of diagnostic pathology is also changing with the availability of new tests and the introduction of new treatment options. OBJECTIVE Glioblastomas, oligodendrogliomas, glioneuronal tumors, and primitive pediatric tumors are discussed in an exemplary way to illustrate these changes. DATA SOURCES Review of relevant publications through Medline database searches. CONCLUSIONS The example of glioblastomas shows how new predictive markers may help identify subgroups of tumors that respond to certain therapy regimens. The development of new treatment strategies also leads to different questions in the assessment of brain tumors, as seen in the example of pseudoprogression or the changes in tumor growth pattern in patients taking bevacizumab. Oligodendrogliomas illustrate how the identification of 1p/19q loss as a cytogenetic aberration aids our understanding of these tumors and changes diagnostic practice but also introduces new challenges in classification. Glioneuronal tumors are an evolving group of lesions. Besides a growing list of usually low-grade entities with well-defined morphologic features, these also include more poorly defined cases in which a component of infiltrating glioma is often associated with focal neuronal elements. The latter is biologically interesting but of uncertain clinical significance. Oligodendrogliomas and glioneuronal tumors both illustrate the importance of effective communication between the pathologist and the treating oncologist in the discussion of these patients. Finally, the discussion of primitive pediatric tumors stresses the clinical importance of the distinction between different entities, like atypical teratoid rhabdoid tumor, "central" (supratentorial) primitive neuroectodermal tumor, "peripheral" primitive neuroectodermal tumor, and medulloblastoma. In medulloblastomas, the recognition of different variants is emerging as a prognostic factor that may in the future also predict therapy responsiveness.
Collapse
Affiliation(s)
- Peter Pytel
- Department of Pathology, University of Chicago Medical Center, Chicago, Illinois 60637, USA.
| | | |
Collapse
|
44
|
Bibliography. Current world literature. Hematology and oncology. Curr Opin Pediatr 2008; 20:107-13. [PMID: 18197049 DOI: 10.1097/mop.0b013e3282f572b6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
45
|
Postovsky S, Eran A, Weyl Ben Arush M. Unusual case of leptomeningeal dissemination of a diffuse pontine high-grade astrocytoma in a child. Pediatr Neurosurg 2008; 44:208-11. [PMID: 18334845 DOI: 10.1159/000120152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2006] [Accepted: 04/11/2007] [Indexed: 11/19/2022]
Abstract
A 12-year-old girl was seen in our hospital and diagnosed with diffuse pontine glioma. Treatment with radiotherapy was suggested, but the parents rejected this plan and took the child to another hospital where partial resection of the tumor was performed. The histological diagnosis was anaplastic astrocytoma. Returning to our hospital, the parents agreed to radiotherapy which was given in a total dose of 54 Gy. However, 3 months after completion of radiotherapy, the child developed widespread leptomeningeal dissemination of her disease and died 12 months after the initial diagnosis. Since no effective curative methods of treatment exist for patients with diffuse pontine gliomas and surgical intervention is obsolete in this situation and may facilitate leptomeningeal dissemination of the disease, further innovative methods of treatment are needed to improve the bleak prognosis of these patients.
Collapse
Affiliation(s)
- Sergey Postovsky
- Department of Pediatric Hematology-Oncology, Rambam Health Care Campus and Faculty of Medicine, Technion-Israeli Institute of Technology, Haifa, Israel.
| | | | | |
Collapse
|
46
|
Vladimirova V, Denkhaus D, Soerensen N, Wagner S, Wolff JEA, Pietsch T. Low level of microsatellite instability in paediatric malignant astrocytomas. Neuropathol Appl Neurobiol 2007; 34:547-54. [PMID: 18053027 DOI: 10.1111/j.1365-2990.2007.00919.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIM Microsatellite instability (MSI) has been proposed as a possible mechanism in the development of cancer. The aim of the current study was to determine whether MSI is involved in the pathogenesis of paediatric malignant astrocytomas. METHODS We screened a cohort of 126 high-grade astrocytoma samples for MSI using a sensitive and precise method of DNA analysis including a panel of five mononucleotide repeats, in combination with immunohistochemistry for DNA mismatch repair (MMR) proteins. RESULTS We identified low level of MSI (MSI-L) in four of 126 (3.2%) paediatric malignant astrocytic tumours. To analyse the molecular profile associated with MSI-L positive tumours, we performed immunohistochemistry for protein expression of hMSH6 and p53 as well as mutational analysis of the K-ras gene. In MSI-L paediatric malignant astrocytic tumours we detected retained nuclear expression of hMSH6 protein and strong nuclear accumulation of p53 protein indicating possible mutations of TP53. There was no correlation between K-ras mutational status and frequency of MSI in this patient population. CONCLUSION Our results suggest that the MSI-L phenotype is associated with p53 accumulation and/or mutations. However, this represents only a small subgroup of paediatric gliomas with possible distinct biological features, and the deficiencies of DNA MMR genes do not play a main role in the tumourigenesis of the majority of paediatric malignant astrocytomas.
Collapse
Affiliation(s)
- V Vladimirova
- Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany.
| | | | | | | | | | | |
Collapse
|
47
|
Benesch M, Windelberg M, Sauseng W, Witt V, Fleischhack G, Lackner H, Gadner H, Bode U, Urban C. Compassionate use of bevacizumab (Avastin) in children and young adults with refractory or recurrent solid tumors. Ann Oncol 2007; 19:807-13. [PMID: 18056650 DOI: 10.1093/annonc/mdm510] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The aim of this study was to evaluate feasibility and toxicity of bevacizumab (Avastin), a monoclonal antibody directed against the vascular endothelial growth factor in children and young adults. PATIENTS AND METHODS Fifteen patients (male: n = 8; female: n = 7; median age, 14.6 years) received bevacizumab for recurrent or progressive solid tumors (carcinoma: n = 3; neuroblastoma: n = 2; astrocytoma grade III: n = 2; rhabdomyosarcoma: n = 2; nephroblastoma: n = 2; benign vascular tumors: n = 2; synovial sarcoma: n = 1; and malignant hemangiopericytoma: n = 1) on a compassionate basis. Bevacizumab was administered at 5-10 mg/kg body weight intravenously every 2-3 weeks. Most patients received chemotherapy in addition to bevacizumab. Duration of bevacizumab therapy ranged from 1.5 to 23 months. RESULTS Bevacizumab-related side-effects were mild and included hypertonia (n = 2), proteinuria/hematuria (n = 2), epistaxis (n = 2), local erythema (n = 1), and defective wound healing and ascites (n = 1). Radiographic objective responses (partial responses) were observed in two patients with astrocytoma grade III and in one patient each with neuroblastoma and pleomorphic rhabdomyosarcoma, respectively. CONCLUSIONS Bevacizumab seems to have a good acute safety profile and some antitumor activity in heavily pretreated children and young adults with recurrent solid tumors. Prospective clinical trials are urgently needed to further evaluate the safety and efficacy of bevacizumab in pediatric patients.
Collapse
Affiliation(s)
- M Benesch
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Austria.
| | | | | | | | | | | | | | | | | |
Collapse
|