1
|
Skouras P, Markouli M, Papadatou I, Piperi C. Targeting epigenetic mechanisms of resistance to chemotherapy in gliomas. Crit Rev Oncol Hematol 2024; 204:104532. [PMID: 39406277 DOI: 10.1016/j.critrevonc.2024.104532] [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: 07/11/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Glioma, an aggressive type of brain tumors of glial origin is highly heterogeneous, posing significant treatment challenges due to its intrinsic resistance to conventional therapeutic schemes. It is characterized by an interplay between epigenetic and genetic alterations in key signaling pathways which further endorse their resistance potential. Aberrant DNA methylation patterns, histone modifications and non-coding RNAs may alter the expression of genes associated with drug response and cell survival, induce gene silencing or deregulate key pathways contributing to glioma resistance. There is evidence that epigenetic plasticity enables glioma cells to adapt dynamically to therapeutic schemes and allow the formation of drug-resistant subpopulations. Furthermore, the tumor microenvironment adds an extra input on epigenetic regulation, increasing the complexity of resistance mechanisms. Herein, we discuss epigenetic changes conferring to drug resistance mechanisms in gliomas in order to delineate novel therapeutic targets and potential approaches that will enable personalized treatment.
Collapse
Affiliation(s)
- Panagiotis Skouras
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece; 1st Department of Neurosurgery, Evangelismos Hospital, National and Kapodistrian University of Athens, Greece.
| | - Mariam Markouli
- Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Ioanna Papadatou
- University Research Institute for the Study of Genetic & Malignant Disorders in Childhood, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece.
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece.
| |
Collapse
|
2
|
Xing YL, Grossauer S, Park JW, Nasajpour E, Bui B, Morales D, Panovska D, Nirschl JJ, Feng ZP, Wei R, Koeck K, Thomason W, Xiu J, Harter PN, Filipski K, Mahaney K, Ji X, Mulcahy Levy JM, Grant GA, Prolo LM, Walsh KM, Lim M, Hambardzumyan D, Petritsch CK. Dual MAPK Inhibition Triggers Pro-inflammatory Signals and Sensitizes BRAF V600E Glioma to T Cell-Mediated Checkpoint Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.03.526065. [PMID: 39416185 PMCID: PMC11482820 DOI: 10.1101/2023.02.03.526065] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
BRAF V600E pediatric low-grade gliomas frequently transform into high-grade gliomas (HGG) and poorly respond to chemotherapy, resulting in high mortality. Although combined BRAF and MEK inhibition (BRAFi+MEKi) outperforms chemotherapy, ∼70% of BRAF V600E HGG patients are therapy resistant and undergo unbridled tumor progression. BRAF V600E glioma have an immune-rich microenvironment suggesting that they could be responsive to immunotherapy but effects of BRAFi+MEKi on anti-tumor immunity are unclear. Using patient tumor tissue before and after BRAFi+MEKi, two novel syngeneic murine models of BRAF V600E HGG, and patient-derived cell lines, we examined the effects of clinically relevant BRAFi+MEKi with dabrafenib and trametinib on tumor growth, cell states, and tumor-infiltrating T cells. We find that BRAFi+MEKi treatment: i) upregulated programmed cell death protein-1 (PD-1) signaling genes and PD-1 ligand (PD-L1) protein expression in murine BRAF V600E HGG by stimulating IFNγ and IL-27, ii) attenuated T cell activity by IL-23, IL-27 and IL-32 production, which can promote the expansion of regulatory T cells, and iii) induced glial differentiation linked to a therapy-resistant PD-L1+ compartment through Galectin-3 secretion by tumor cells. Murine BRAF V600E HGG shrinkage by BRAFi+MEKi is associated with the upregulation of interferon-gamma response genes, MHC class I/II expression, and antigen presentation and processing programs, indicative of increased anti-tumor immunity. Combined BRAFi+MEKi with therapeutic antibodies inhibiting the PD-1 and cytotoxic T-lymphocyte associated protein 4 (CTLA-4) immune checkpoints re-activate T cells and provide a survival benefit over single therapy in a T cell-dependent manner. The quadruple treatment overcame BRAFi+MEKi resistance by invigorating T cell-mediated anti-tumor immunity in murine BRAF V600E HGG. PD-L1 expression was elevated in human BRAF-mutant versus BRAF-wildtype glioblastoma clinical specimen, complementing experimental findings and suggesting translational relevance for patient care.
Collapse
|
3
|
Bansal I, Merchant TE. Radiotherapy for pediatric low-grade glioma. Childs Nerv Syst 2024; 40:3277-3290. [PMID: 38775957 DOI: 10.1007/s00381-024-06460-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/13/2024] [Indexed: 10/26/2024]
Abstract
INTRODUCTION Radiotherapy is a highly effective treatment for pediatric low-grade glioma, serving as the standard for evaluating progression-free and overall survival, as well as vision preservation. Despite its proven efficacy, concerns about treatment complications have led to increased use of chemotherapy and targeted therapy, which are associated with poorer progression-free survival outcomes. METHODS This review by Indu Bansal and Thomas E. Merchant examines the indications, timing, and results of radiotherapy for pediatric low-grade glioma. The authors provide a comprehensive analysis of clinical management strategies, addressing the controversies surrounding the use and timing of radiotherapy compared to other therapies. RESULTS The review highlights that while radiotherapy is essential for certain patients, particularly those who are not candidates for complete resection due to the tumor's infiltrative nature or location, it is often deferred in favor of systemic therapies. This deferral can lead to significant morbidity, including poor visual outcomes. Reports indicate that systemic therapy negatively impacts progression-free survival in patients who eventually undergo radiotherapy. Newer radiotherapy techniques have been developed to minimize complications, offering potential benefits over traditional methods. DISCUSSION The evolving clinical management of pediatric low-grade glioma involves balancing the benefits of radiotherapy with concerns about its side effects. Although systemic therapies are increasingly favored, their associated inferior progression-free survival and potential for significant morbidity underscore the need for careful consideration of radiotherapy, particularly in older children, adolescents, or those with progressive disease post-systemic therapy. The emerging role of targeted therapy presents additional challenges, including uncertainties about long-term side effects and its interaction with radiotherapy. Further research is needed to optimize treatment strategies and improve outcomes for pediatric patients with low-grade glioma.
Collapse
Affiliation(s)
- Indu Bansal
- Department of Radiation Oncology at Paras Health, Gurugram, Haryana, India
| | - Thomas E Merchant
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
4
|
Jaimovich SG, Takeuchi K, Testa VT, Okumura E, Jaimovich R, Cinalli G. Cylinder tumor surgery in pediatric low-grade gliomas. Childs Nerv Syst 2024; 40:3051-3063. [PMID: 38644385 DOI: 10.1007/s00381-024-06417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 04/23/2024]
Abstract
BACKGROUND Periventricular pediatric low-grade gliomas (pLGG) present a surgical challenge due to their deep-seated location, accessibility, and relationship with the subcortical network connections. Minimally invasive parafascicular approaches with tubular brain retractors (port brain surgery) have emerged, in recent years, as an alternative to conventional microsurgical and endoscopic approaches for removal of periventricular tumors. OBJECTIVES To describe the minimally invasive approach with tubular brain retractors for periventricular pLGG, its technique, applications, safety, and efficacy. METHODS In this article, we describe the port brain surgery techniques for periventricular pLGG as performed in different centers, with different commercialized tubular retractor systems. Illustrative cases followed by a literature review are analyzed, with a detailed description of different approaches or techniques, comparing their advantages and disadvantages with contemporary microsurgical and endoscopic approaches. CONCLUSIONS The port brain surgery with micro-exoscopic vision and endoscopic assistance, for the treatment of deep-seated lesions such as periventricular pLGG, is an alternative for achieving a functionally safe-gross total or subtotal-tumor resection, obtaining adequate tissue for pathological examination. This technique could offer a new dimension for a less-invasive, safe, and effective access to deep-seated tumors, offering the possibility to lower morbidity in experienced hands.
Collapse
Affiliation(s)
- Sebastian Gaston Jaimovich
- Department of Neurosurgery, "Prof. Dr. Juan P. Garrahan" Pediatric Hospital, Buenos Aires, Argentina
- Department of Neurosurgery, FLENI, Buenos Aires, Argentina
| | - Kazuhito Takeuchi
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Aichi, Japan
| | | | - Eriko Okumura
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Roberto Jaimovich
- Department of Neurosurgery, "Prof. Dr. Juan P. Garrahan" Pediatric Hospital, Buenos Aires, Argentina
- Department of Neurosurgery, FLENI, Buenos Aires, Argentina
| | - Giuseppe Cinalli
- Department of Pediatric Neurosurgery, Santobono-Pausilipon Children's Hospital (AORN), Naples, Italy.
| |
Collapse
|
5
|
Zelt S, Cooney T, Yu S, Daral S, Krebs B, Markan R, Manley P, Kieran M, Govinda Raju S. Disease burden and healthcare utilization in pediatric low-grade glioma: A United States retrospective study of linked claims and electronic health records. Neurooncol Pract 2024; 11:583-592. [PMID: 39279771 PMCID: PMC11398936 DOI: 10.1093/nop/npae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024] Open
Abstract
Background Despite high long-term survival rates, pediatric low-grade gliomas (pLGGs) are linked with significant tumor- and treatment-associated morbidities that may persist throughout life. The aims of this descriptive cross-sectional pilot study were to characterize health conditions among a cohort of patients with pLGG and explore the feasibility of quantifying disease burden and healthcare resource utilization (HRU). Methods Optum® Market Clarity Data were used to identify patients aged ≤18 years with an ICD-10 code for brain neoplasm, ≥1 physician notes, and with evidence of pLGG recorded between January 1, 2017 and June 30, 2018. Outcomes including health characteristics, HRU, medications, and procedures were assessed at 6-month intervals over 36 months. Results One hundred and fifty-four patients were identified with pLGG and over half experienced headache/migraine, respiratory infection, pain, or behavioral issues during the 36-month study period. The most common comorbidities were ocular/visual (including blindness), mental health disorders, seizures, and behavioral/cognition disorders. Most symptoms and comorbidities persisted or increased during the study period, indicating long-term health deficits. HRU, including speciality care visits, filled prescriptions, and administered medications, was common; 74% of patients had prescriptions for anti-infectives, 56% antiemetics, and 52% required pain or fever relief. Sixty-five percent of patients underwent treatment to control their pLGG, the most common being brain surgery. Little decline was observed in medication use during the study period. Conclusions Patients with pLGG have complex healthcare needs requiring high HRU, often over a long time. Patients need to be optimally managed to minimize disease- and treatment-related burden and HRU.
Collapse
Affiliation(s)
- Susan Zelt
- Day One Biopharmaceuticals, Inc., Brisbane, California, USA (S.Z., T.C., S.Y., P.M., M.K., S.G.R.)
| | - Tabitha Cooney
- Day One Biopharmaceuticals, Inc., Brisbane, California, USA (S.Z., T.C., S.Y., P.M., M.K., S.G.R.)
| | - Sandie Yu
- Day One Biopharmaceuticals, Inc., Brisbane, California, USA (S.Z., T.C., S.Y., P.M., M.K., S.G.R.)
| | - Shailaja Daral
- Optum Lifesciences, Inc., Eden Prairie, Minnesota, USA (S.D., B.K., R.M.)
| | - Blake Krebs
- Optum Lifesciences, Inc., Eden Prairie, Minnesota, USA (S.D., B.K., R.M.)
| | - Riddhi Markan
- Optum Lifesciences, Inc., Eden Prairie, Minnesota, USA (S.D., B.K., R.M.)
| | - Peter Manley
- Day One Biopharmaceuticals, Inc., Brisbane, California, USA (S.Z., T.C., S.Y., P.M., M.K., S.G.R.)
| | - Mark Kieran
- Day One Biopharmaceuticals, Inc., Brisbane, California, USA (S.Z., T.C., S.Y., P.M., M.K., S.G.R.)
| | - Sandya Govinda Raju
- Day One Biopharmaceuticals, Inc., Brisbane, California, USA (S.Z., T.C., S.Y., P.M., M.K., S.G.R.)
| |
Collapse
|
6
|
Lu VM, Jallo GI, Shimony N. Intramedullary pediatric low-grade glioma of the spine. Childs Nerv Syst 2024; 40:3107-3117. [PMID: 38904769 DOI: 10.1007/s00381-024-06499-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/10/2024] [Indexed: 06/22/2024]
Abstract
PURPOSE Pediatric intramedullary spinal cord low-grade gliomas (pLGGs) are rare diagnoses among central nervous system (CNS) tumors in the pediatric population. The classic presentation of the patients includes some degree of neurologic deficit, although many times the symptoms are vague which leads to delayed diagnosis. MATERIAL AND METHODS The first step in the diagnosis includes special parameters in spinal imaging, particularly magnetic resonance imaging (MRI), and surgical resection remains the cornerstone for both diagnosis and treatment. Yet, recent years advancement in molecular and genetic understanding of CNS tumors allows for better adjustment of the treatment and follow-up regimens. Based on postoperative status, adjuvant therapy may provide additional therapeutic advantage for some types of tumors. CONCLUSION Ultimately, patients have a very promising prognosis when treated appropriately in most of the cases of pediatric spinal cord LGG with continued advances arising. This manuscript summarizes the most contemporary evidence regarding clinical and treatment features of intramedullary pLGGs.
Collapse
Affiliation(s)
- Victor M Lu
- Department of Neurological Surgery, University of Miami, Jackson Memorial Hospital, Miami, FL, USA
| | - George I Jallo
- Institute for Brain Protection Sciences, Johns Hopkins All Children's Hospital, St Petersburg, FL, USA.
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, USA.
| | - Nir Shimony
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, USA
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA
- Le Bonheur Neuroscience Institute, Le Bonheur Children's Hospital, Memphis, TN, USA
- Department of Neurosurgery, University of Tennessee Health Science Center, Memphis, TN, USA
- Semmes-Murphey Clinic, Memphis, TN, USA
| |
Collapse
|
7
|
Yaman Bajin I, Bouffet E. Advances in the Treatment of Pediatric Low-Grade Gliomas. Curr Neurol Neurosci Rep 2024; 24:527-535. [PMID: 39143379 DOI: 10.1007/s11910-024-01369-4] [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: 07/29/2024] [Indexed: 08/16/2024]
Abstract
PURPOSE OF REVIEW Pediatric low-grade gliomas (pLGGs) often result in significant long-term morbidities despite high overall survival rates. This review aims to consolidate the current understanding of pLGG biology and molecular features and provide an overview of current and emerging treatment strategies. RECENT FINDINGS Surgical resection remains a primary treatment modality, supplemented by chemotherapy and radiotherapy in specific cases. However, recent advances have elucidated the molecular underpinnings of pLGGs, revealing key genetic abnormalities such as BRAF fusions and mutations and the involvement of the RAS/MAPK and mTOR pathways. Novel targeted therapies, including MEK, BRAF and pan-RAF inhibitors, have shown promise in clinical trials, demonstrating significant efficacy and manageable toxicity. Understanding of pLGGs has significantly improved, leading to more personalized treatment approaches. Targeted therapies have emerged as effective alternatives, potentially reducing long-term toxicities. Future research should focus on optimizing therapy sequences, understanding long-term impacts, and ensuring global accessibility to advanced treatments.
Collapse
Affiliation(s)
- Inci Yaman Bajin
- Division of Paediatric Haematology-Oncology, Department of Paediatrics, Hospital for Sick Children, Toronto,, Canada.
| | - Eric Bouffet
- Division of Paediatric Haematology-Oncology, Department of Paediatrics, Hospital for Sick Children, Toronto,, Canada
| |
Collapse
|
8
|
Rufus P, Chatterjee S. Second-look surgery in postoperative pediatric low-grade glioma. Childs Nerv Syst 2024; 40:3135-3142. [PMID: 38970692 DOI: 10.1007/s00381-024-06516-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/20/2024] [Indexed: 07/08/2024]
Abstract
OBJECTIVE To review the literature on second-look surgery in pediatric low-grade gliomas (LGG) with a view to presenting both sides of the picture of re-exploration. METHODS Collection of material from recent literature on pediatric LGG. This was a retrospective review of these publications. RESULTS There are a number of publications recommending second-look surgery in selected cases, provided morbidity of the second surgery is minimum, and indeed some in which there is improvement in the neurodeficit after the second resection. CONCLUSION There seems a fair balance of articles recommending and dissuading the practice of second-look surgery, but in our limited experience we have found it useful in selected patients.
Collapse
|
9
|
Habibi MA, Rashidi F, Gharedaghi H, Arshadi MR, Kazemivand S. The safety and efficacy of bevacizumab in treatment of recurrent low-grade glioma: a systematic review and meta-analysis. Eur J Clin Pharmacol 2024; 80:1259-1270. [PMID: 38733390 DOI: 10.1007/s00228-024-03695-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Central nervous system (CNS) tumors are among the most common malignancies in various age ranges. Low-grade glioma (LGG) can account for nearly 30% of pediatric CNS malignancies. Progression or recurrence after the first-line treatments is common among these patients. Therefore, more treatments are required. Bevacizumab as an anti-VEGF antibody has come into the spotlight recently and is especially used in relapse or recurrence settings. This review aims to study the safety and efficacy of bevacizumab for patients with recurrent LGG. METHODS This study was conducted according to The Preferred Reporting Items for Systematic Reviews and Meta-Analyses. PubMed, Scopus, Web of Science, and Embase were comprehensively searched using the relevant key terms until 24th August 2023 to retrieve the studies that investigated clinical outcomes of bevacizumab in patients with recurrent LGG. All statistical analysis was performed by STATA v.17. RESULTS A total of 1306 papers were gathered, out of which 13 were incorporated in the meta-analysis. The pooled incidence rate of treatment according to the RANO scale was 70% (95% CI = 43-98%) for objective response rate, 26% (95% CI = 58-96%) for partial response, 21% (95% CI = 15-28%) for minor response, 14% (95% CI = 3-24%) for complete response, 48% (95% CI = 37-59%) for stable disease, and 8% (95% CI = 4-11%) for progressive disease. Furthermore, according to progressive survival after treatment, it was 4% (95% CI = -1 to 9%) for 6-month PFS, 41% (95% CI = 32-50%) for 2-year PFS, and 29% (95% CI = 22-35%) for 3-year PFS. CONCLUSION According to the RANO scale and PFS, clinicians should be aware that Bevacizumab could be a favorable alternative therapy for recurrent LGG. Furthermore, bevacizumab exhibits minimal toxicity and high tolerability in recurrent LGG.
Collapse
Affiliation(s)
- Mohammad Amin Habibi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Farhang Rashidi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Reza Arshadi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Sana Kazemivand
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Gupta RV, Welch JC, Kaul A, Prayson RA, Rong Y, Almefty RO. Rapid postradiation malignant transformation of a pilocytic astrocytoma in an adult with neurofibromatosis type 1: illustrative case. JOURNAL OF NEUROSURGERY. CASE LESSONS 2024; 8:CASE24241. [PMID: 39133944 PMCID: PMC11323849 DOI: 10.3171/case24241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/30/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Juvenile pilocytic astrocytoma (JPA) is the most common primary brain tumor of childhood and is rarely seen in adults. Neurofibromatosis type 1 (NF1), a common tumor predisposition syndrome, demonstrates a strong association with low-grade gliomas, most notably pilocytic astrocytoma, which are relatively indolent. Unlike its juvenile counterpart, reports of adult pilocytic astrocytoma (APA) vary widely in terms of disease progression from benign to much more malignant courses. Moreover, current studies discussing APA report different treatment approaches and outcomes (e.g., malignant transformation of JPA and APA with or without radiation), as little is known regarding the management of recurrent tumors and how adjuvant therapies may alter disease progression. OBSERVATIONS The authors report the unique case of an adult male with NF1 and APA who underwent rapid malignant conversion after intensity-modulated radiation therapy. LESSONS The authors demonstrate that caution should be taken in utilizing radiotherapy instead of resection in cases of APA and NF1, with close monitoring for posttreatment recurrence. https://thejns.org/doi/10.3171/CASE24241.
Collapse
Affiliation(s)
- Rohan V Gupta
- Departments of Neurosurgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Jonathan C Welch
- Departments of Neurosurgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Anand Kaul
- Departments of Neurosurgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Richard A Prayson
- Department of Pathology, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Yuan Rong
- Departments of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Rami O Almefty
- Departments of Neurosurgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Gorodezki D, Schuhmann MU, Ebinger M, Schittenhelm J. Dissecting the Natural Patterns of Progression and Senescence in Pediatric Low-Grade Glioma: From Cellular Mechanisms to Clinical Implications. Cells 2024; 13:1215. [PMID: 39056798 PMCID: PMC11274692 DOI: 10.3390/cells13141215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Pediatric low-grade gliomas (PLGGs) comprise a heterogeneous set of low-grade glial and glioneuronal tumors, collectively representing the most frequent CNS tumors of childhood and adolescence. Despite excellent overall survival rates, the chronic nature of the disease bears a high risk of long-term disease- and therapy-related morbidity in affected patients. Recent in-depth molecular profiling and studies of the genetic landscape of PLGGs led to the discovery of the paramount role of frequent upregulation of RAS/MAPK and mTOR signaling in tumorigenesis and progression of these tumors. Beyond, the subsequent unveiling of RAS/MAPK-driven oncogene-induced senescence in these tumors may shape the understanding of the molecular mechanisms determining the versatile progression patterns of PLGGs, potentially providing a promising target for novel therapies. Recent in vitro and in vivo studies moreover indicate a strong dependence of PLGG formation and growth on the tumor microenvironment. In this work, we provide an overview of the current understanding of the multilayered cellular mechanisms and clinical factors determining the natural progression patterns and the characteristic biological behavior of these tumors, aiming to provide a foundation for advanced stratification for the management of these tumors within a multimodal treatment approach.
Collapse
Affiliation(s)
- David Gorodezki
- Department of Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany;
| | - Martin U. Schuhmann
- Section of Pediatric Neurosurgery, Department of Neurosurgery, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Martin Ebinger
- Department of Hematology and Oncology, University Children’s Hospital Tübingen, 72076 Tübingen, Germany;
| | - Jens Schittenhelm
- Department of Neuropathology, Institute of Pathology, University Hospital Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
12
|
Tak D, Garomsa BA, Zapaishchykova A, Ye Z, Vajapeyam S, Mahootiha M, Climent Pardo JC, Smith C, Familiar AM, Chaunzwa T, Liu KX, Prabhu S, Bandopadhayay P, Nabavizadeh A, Mueller S, Aerts HJ, Haas-Kogan D, Poussaint TY, Kann BH. Longitudinal risk prediction for pediatric glioma with temporal deep learning. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.04.24308434. [PMID: 38978642 PMCID: PMC11230342 DOI: 10.1101/2024.06.04.24308434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Pediatric glioma recurrence can cause morbidity and mortality; however, recurrence pattern and severity are heterogeneous and challenging to predict with established clinical and genomic markers. Resultingly, almost all children undergo frequent, long-term, magnetic resonance (MR) brain surveillance regardless of individual recurrence risk. Deep learning analysis of longitudinal MR may be an effective approach for improving individualized recurrence prediction in gliomas and other cancers but has thus far been infeasible with current frameworks. Here, we propose a self-supervised, deep learning approach to longitudinal medical imaging analysis, temporal learning, that models the spatiotemporal information from a patient's current and prior brain MRs to predict future recurrence. We apply temporal learning to pediatric glioma surveillance imaging for 715 patients (3,994 scans) from four distinct clinical settings. We find that longitudinal imaging analysis with temporal learning improves recurrence prediction performance by up to 41% compared to traditional approaches, with improvements in performance in both low- and high-grade glioma. We find that recurrence prediction accuracy increases incrementally with the number of historical scans available per patient. Temporal deep learning may enable point-of-care decision-support for pediatric brain tumors and be adaptable more broadly to patients with other cancers and chronic diseases undergoing surveillance imaging.
Collapse
|
13
|
Joh-Carnella N, Bauman G, Yock TI, Zelcer S, Youkhanna S, Cacciotti C. Case report: Pediatric low-grade gliomas: a fine balance between treatment options, timing of therapy, symptom management and quality of life. Front Oncol 2024; 14:1366251. [PMID: 38912055 PMCID: PMC11190070 DOI: 10.3389/fonc.2024.1366251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/28/2024] [Indexed: 06/25/2024] Open
Abstract
Introduction Pediatric low-grade gliomas (pLGG) are the most common brain tumor in children and encompass a wide range of histologies. Treatment may pose challenges, especially in those incompletely resected or those with multiple recurrence or progression. Case description We report the clinical course of a girl diagnosed with pilocytic astrocytoma and profound hydrocephalus at age 12 years treated with subtotal resection, vinblastine chemotherapy, and focal proton radiotherapy. After radiotherapy the tumor increased in enhancement temporarily with subsequent resolution consistent with pseudoprogression. Despite improvement in imaging and radiographic local control, the patient continues to have challenges with headaches, visual and auditory concerns, stroke-like symptoms, and poor quality of life. Conclusion pLGG have excellent long-term survival; thus, treatments should focus on maintaining disease control and limiting long-term toxicities. Various treatment options exist including surgery, chemotherapy, targeted agents, and radiation therapy. Given the morbidity associated with pLGG, individualized treatment approaches are necessary, with a multi-disciplinary approach to care focused on minimizing treatment side effects, and promoting optimal quality of life for patients.
Collapse
Affiliation(s)
| | - Glenn Bauman
- Division of Radiation Oncology, Department of Oncology, London Health Sciences Centre & Western University, London, ON, Canada
| | - Torunn I. Yock
- Department of Pediatric Radiation Oncology, Massachusetts General Hospital, Boston, MA, United States
| | - Shayna Zelcer
- Division of Hematology/Oncology, Department of Pediatrics, London Health Sciences Centre & Western University, London, ON, Canada
| | - Sabin Youkhanna
- Department Radiation Oncology, London Regional Cancer Centre, London, ON, Canada
| | - Chantel Cacciotti
- Division of Hematology/Oncology, Department of Pediatrics, London Health Sciences Centre & Western University, London, ON, Canada
| |
Collapse
|
14
|
Fangusaro J, Avery RA, Fisher MJ, Packer RJ, Walsh KS, Schouten-van Meeteren A, Karres D, Bradford D, Bhatnagar V, Singh H, Kluetz PG, Donoghue M, Duke ES. Considering Functional Outcomes as Efficacy Endpoints in Pediatric Low-Grade Glioma Clinical Trials: An FDA Educational Symposium. Clin Cancer Res 2024; 30:2303-2308. [PMID: 38358393 PMCID: PMC11147731 DOI: 10.1158/1078-0432.ccr-23-3386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/22/2023] [Accepted: 02/13/2024] [Indexed: 02/16/2024]
Abstract
In October 2022, the FDA Oncology Center of Excellence hosted an educational symposium entitled, "Considering Functional Outcomes as Efficacy Endpoints in Pediatric Low-Grade Glioma (pLGG) Clinical Trials." The symposium brought together patient advocates, regulators from the FDA and the European Medicines Agency (EMA), and an international group of academic thought leaders in the field of pediatric neuro-oncology to discuss the potential role of functional outcomes, including visual acuity, motor function, and neurocognitive performance, as endpoints in clinical trials enrolling patients with pLGG. The panel discussed challenges and opportunities regarding the selection, implementation, and evaluation of clinical outcome assessments in these functional domains and outlined key considerations for their inclusion in future clinical trial design and role in new drug development.
Collapse
Affiliation(s)
- Jason Fangusaro
- Children's Hospital of Atlanta, Emory University and the Aflac Cancer Center, Atlanta, Georgia
| | - Robert A Avery
- The Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Michael J Fisher
- The Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Roger J Packer
- Center for Neuroscience and Behavioral Medicine and Brain Tumor Institute, Children's National Hospital, and The George Washington University School of Medicine, Washington, DC
| | - Karin S Walsh
- Center for Neuroscience and Behavioral Medicine and Brain Tumor Institute, Children's National Hospital, and The George Washington University School of Medicine, Washington, DC
| | | | - Dominik Karres
- Pediatric Medicines Office, Scientific Evidence Generation Department, Human Medicines Division, European Medicines Agency (EMA), Amsterdam, the Netherlands
| | - Diana Bradford
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Vishal Bhatnagar
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Harpreet Singh
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Paul G Kluetz
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Martha Donoghue
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Elizabeth S Duke
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
15
|
Obdeijn IV, Wiegers EC, Alic L, Plasschaert SLA, Kranendonk MEG, Hoogduin HM, Klomp DWJ, Wijnen JP, Lequin MH. Amide proton transfer weighted imaging in pediatric neuro-oncology: initial experience. NMR IN BIOMEDICINE 2024; 37:e5122. [PMID: 38369653 DOI: 10.1002/nbm.5122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/22/2023] [Accepted: 01/22/2024] [Indexed: 02/20/2024]
Abstract
Amide proton transfer weighted (APTw) imaging enables in vivo assessment of tissue-bound mobile proteins and peptides through the detection of chemical exchange saturation transfer. Promising applications of APTw imaging have been shown in adult brain tumors. As pediatric brain tumors differ from their adult counterparts, we investigate the radiological appearance of pediatric brain tumors on APTw imaging. APTw imaging was conducted at 3 T. APTw maps were calculated using magnetization transfer ratio asymmetry at 3.5 ppm. First, the repeatability of APTw imaging was assessed in a phantom and in five healthy volunteers by calculating the within-subject coefficient of variation (wCV). APTw images of pediatric brain tumor patients were analyzed retrospectively. APTw levels were compared between solid tumor tissue and normal-appearing white matter (NAWM) and between pediatric high-grade glioma (pHGG) and pediatric low-grade glioma (pLGG) using t-tests. APTw maps were repeatable in supratentorial and infratentorial brain regions (wCV ranged from 11% to 39%), except those from the pontine region (wCV between 39% and 50%). APTw images of 23 children with brain tumor were analyzed (mean age 12 years ± 5, 12 male). Significantly higher APTw values are present in tumor compared with NAWM for both pHGG and pLGG (p < 0.05). APTw values were higher in pLGG subtype pilocytic astrocytoma compared with other pLGG subtypes (p < 0.05). Non-invasive characterization of pediatric brain tumor biology with APTw imaging could aid the radiologist in clinical decision-making.
Collapse
Affiliation(s)
- Iris V Obdeijn
- Center for Image Sciences, High Field MR Research Group, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Evita C Wiegers
- Center for Image Sciences, High Field MR Research Group, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lejla Alic
- Magnetic Detection and Imaging Group, Technical Medical Center, University of Twente, Enschede, The Netherlands
| | - Sabine L A Plasschaert
- Department of Pediatric Neuro-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Mariëtte E G Kranendonk
- Department of Diagnostic Laboratory, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Hans M Hoogduin
- Center for Image Sciences, High Field MR Research Group, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dennis W J Klomp
- Center for Image Sciences, High Field MR Research Group, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jannie P Wijnen
- Center for Image Sciences, High Field MR Research Group, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maarten H Lequin
- Department of Pediatric Neuro-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Radiology and Nuclear Medicine, University of Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
16
|
Sathyakumar S, Martinez M, Perreault S, Legault G, Bouffet E, Jabado N, Larouche V, Renzi S. Advances in pediatric gliomas: from molecular characterization to personalized treatments. Eur J Pediatr 2024; 183:2549-2562. [PMID: 38558313 DOI: 10.1007/s00431-024-05540-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/12/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Pediatric gliomas, consisting of both pediatric low-grade (pLGG) and high-grade gliomas (pHGG), are the most frequently occurring brain tumors in children. Over the last decade, several milestone advancements in treatments have been achieved as a result of stronger understanding of the molecular biology behind these tumors. This review provides an overview of pLGG and pHGG highlighting their clinical presentation, molecular characteristics, and latest advancements in therapeutic treatments. Conclusion: The increasing understanding of the molecular biology characterizing pediatric low and high grade gliomas has revolutionized treatment options for these patients, especially in pLGG. The implementation of next generation sequencing techniques for these tumors is crucial in obtaining less toxic and more efficacious treatments. What is Known: • Pediatric Gliomas are the most common brain tumour in children. They are responsible for significant morbidity and mortality in this population. What is New: • Over the last two decades, there has been a significant increase in our global understanding of the molecular background of pediatric low and high grade gliomas. • The implementation of next generation sequencing techniques for these tumors is crucial in obtaining less toxic and more efficacious treatments, with the ultimate goal of improving both the survival and the quality of life of these patients.
Collapse
Affiliation(s)
| | - Matthew Martinez
- Department of Social Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Sébastien Perreault
- Division of Pediatric Neurology, Department of Neurosciences, CHU Sainte-Justine, Montreal, Québec, Canada
| | - Geneviève Legault
- Department of Pediatrics, Division of Neurology, Montreal Children's Hospital - McGill University Health Center, Montreal, Québec, Canada
- The Research Institute of the McGill University Health Centre, Montreal, Québec, Canada
| | - Eric Bouffet
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Haematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nada Jabado
- Division of Experimental Medicine, Montreal Children's Hospital, McGill University and McGill University Health Centre, Montreal, Québec, Canada
- Department of Pediatrics, McGill University, Montreal, Québec, Canada
| | - Valérie Larouche
- Division of Hemato-Oncology, Department of Pediatrics, CHU de Québec-Université Laval, 2705 Boulevard, Laurier, G1V 4G2, Québec, Canada
| | - Samuele Renzi
- Division of Hemato-Oncology, Department of Pediatrics, CHU de Québec-Université Laval, 2705 Boulevard, Laurier, G1V 4G2, Québec, Canada.
| |
Collapse
|
17
|
Ziegler DS, Lehmann R, Eisenstat DD. A paradigm shift in how we treat pediatric low-grade glioma-Targeting the molecular drivers. Neuro Oncol 2024; 26:593-595. [PMID: 38243845 PMCID: PMC10995501 DOI: 10.1093/neuonc/noae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Indexed: 01/22/2024] Open
Affiliation(s)
- David S Ziegler
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, New South Wales, Australia
- Kids Cancer Centre, Sydney Children’s Hospital, Randwick, New South Wales, Australia
| | - Rebecca Lehmann
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - David D Eisenstat
- Children’s Cancer Centre, Royal Children’s Hospital, Parkville, Victoria, Australia
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
18
|
Buckland ME, Sarkar C, Santosh V, Al‐Hussaini M, Park SH, Tihan T, Ng HK, Komori T. Announcing the Asian Oceanian Society of Neuropathology guidelines for Adapting Diagnostic Approaches for Practical Taxonomy in Resource-Restrained Regions (AOSNP-ADAPTR). Brain Pathol 2024; 34:e13201. [PMID: 37574221 PMCID: PMC10901611 DOI: 10.1111/bpa.13201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/24/2023] [Indexed: 08/15/2023] Open
Affiliation(s)
- M. E. Buckland
- Department of NeuropathologyRoyal Prince Alfred HospitalSydneyAustralia
| | - C. Sarkar
- Department of PathologyAll India Institute of Medical SciencesNew DelhiIndia
| | - V. Santosh
- Department of NeuropathologyNational Institute of Mental Health and Neuro SciencesBengaluruIndia
| | - M. Al‐Hussaini
- Department of Pathology and Laboratory MedicineKing Hussein Cancer CenterAmmanJordan
| | - S. H. Park
- Department of PathologySeoul National University, College of MedicineSeoulRepublic of Korea
| | - T. Tihan
- Department of PathologyUniversity of California San FranciscoSan FranciscoUSA
| | - H. K. Ng
- Department of Anatomical and Cellular PathologyChinese University of Hong KongHong KongChina
| | - T. Komori
- Department of Laboratory Medicine and PathologyTokyo Metropolitan Neurological Hospital, Tokyo Metropolitan Hospital OrganizationTokyoJapan
| |
Collapse
|
19
|
Yaman I, Bouffet E. How will tovorafenib change our treatment of pediatric low-grade glioma? Expert Opin Emerg Drugs 2024; 29:1-3. [PMID: 38293894 DOI: 10.1080/14728214.2024.2312817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/29/2024] [Indexed: 02/01/2024]
Affiliation(s)
- Inci Yaman
- The Hospital for Sick Children, 55 University Avenue, Toronto, Canada
| | - Eric Bouffet
- The Hospital for Sick Children, 55 University Avenue, Toronto, Canada
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
20
|
Mohamed AA, Alshaibi R, Faragalla S, Mohamed Y, Lucke-Wold B. Updates on management of gliomas in the molecular age. World J Clin Oncol 2024; 15:178-194. [PMID: 38455131 PMCID: PMC10915945 DOI: 10.5306/wjco.v15.i2.178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/06/2024] [Accepted: 01/25/2024] [Indexed: 02/20/2024] Open
Abstract
Gliomas are primary brain tumors derived from glial cells of the central nervous system, afflicting both adults and children with distinct characteristics and therapeutic challenges. Recent developments have ushered in novel clinical and molecular prognostic factors, reshaping treatment paradigms based on classification and grading, determined by histological attributes and cellular lineage. This review article delves into the diverse treatment modalities tailored to the specific grades and molecular classifications of gliomas that are currently being discussed and used clinically in the year 2023. For adults, the therapeutic triad typically consists of surgical resection, chemotherapy, and radiotherapy. In contrast, pediatric gliomas, due to their diversity, require a more tailored approach. Although complete tumor excision can be curative based on the location and grade of the glioma, certain non-resectable cases demand a chemotherapy approach usually involving, vincristine and carboplatin. Additionally, if surgery or chemotherapy strategies are unsuccessful, Vinblastine can be used. Despite recent advancements in treatment methodologies, there remains a need of exploration in the literature, particularly concerning the efficacy of treatment regimens for isocitrate dehydrogenase type mutant astrocytomas and fine-tuned therapeutic approaches tailored for pediatric cohorts. This review article explores into the therapeutic modalities employed for both adult and pediatric gliomas in the context of their molecular classification.
Collapse
Affiliation(s)
- Ali Ahmed Mohamed
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, United States
| | - Rakan Alshaibi
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, United States
| | - Steven Faragalla
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, United States
| | - Youssef Mohamed
- College of Osteopathic Medicine, Kansas City University, Joplin, MO 64804, United States
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, United States
| |
Collapse
|
21
|
Papini C, Mirzaei S. S, Xing M, Tonning Olsson I, de Blank PMK, Lange KR, Salloum R, Srivastava D, Leisenring WM, Howell RM, Oeffinger KC, Robison LL, Armstrong GT, Krull KR, Brinkman TM. Evolving therapies, neurocognitive outcomes, and functional independence in adult survivors of childhood glioma. J Natl Cancer Inst 2024; 116:288-298. [PMID: 37688569 PMCID: PMC10852618 DOI: 10.1093/jnci/djad190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/11/2023] [Accepted: 09/05/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Treatment of childhood glioma has evolved to reduce radiotherapy exposure with the goal of limiting late toxicity. However, the associations between treatment changes and neurocognition, and the contribution of neurocognition and chronic health conditions to attainment of adult independence, remain unknown. METHODS Adult survivors of childhood glioma diagnosed in 1970-1999 in the Childhood Cancer Survivor Study (n = 1284; median [minimum-maximum] 30 [18-51] years of age at assessment; 22 [15-34] years from diagnosis) self-reported neurocognitive impairment and chronic health conditions. Multivariable models evaluated associations between changes in treatment exposures (surgery only, chemotherapy [with or without surgery], cranial radiation [with or without chemotherapy and/or surgery]), and neurocognitive impairment. Latent class analysis with 5 indicators (employment, independent living, assistance with routine and/or personal care needs, driver's license, marital or partner status) identified classes of functional independence. Path analysis tested associations among treatment exposures, neurocognitive impairment, chronic health conditions, and functional independence. Statistical tests were 2-sided. RESULTS Cranial radiation exposure decreased over time (51%, 1970s; 46%, 1980s; 27%, 1990s]. However, compared with siblings, survivors with any treatment exposure were at elevated risk for neurocognitive impairment, including surgery only (eg, memory: relative risk = 2.22; task efficiency: relative risk = 1.88; both P < .001). Three classes of functional independence were identified: independent (58%), moderately independent (20%), and nonindependent (22%). Cranial radiation was associated with nonindependence through impaired task efficiency (β = 0.06), sensorimotor (β = 0.06), and endocrine (β = 0.10) chronic health conditions and through the associations between these conditions and task efficiency (each β = 0.04). Sensorimotor and endocrine chronic health conditions were associated with nonindependence through memory. CONCLUSION Most long-term glioma survivors achieve adult independence. However, functional nonindependence is associated with treatment-related neurocognitive impairment and chronic health conditions.
Collapse
Affiliation(s)
- Chiara Papini
- Department of Psychology and Biobehavioral Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Sedigheh Mirzaei S.
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Mengqi Xing
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Ingrid Tonning Olsson
- Department of Pediatrics, Skåne University Hospital, Lund, Sweden
- Department of Clinical Sciences, Pediatrics, Lund University, Lund, Sweden
| | - Peter M K de Blank
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, The Cure Starts Now Brain Tumor Center, Cincinnati, OH, USA
| | - Katharine R Lange
- Divison of Pediatric Oncology, Hackensack Meridian Children’s Health, Hackensack, NJ, USA
| | - Ralph Salloum
- Pediatric Brain Tumor Program, Division of Hematology, Oncology & Bone Marrow Transplant, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Deokumar Srivastava
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Wendy M Leisenring
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rebecca M Howell
- Division of Radiation Oncology, Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX, USA
| | | | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Gregory T Armstrong
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Kevin R Krull
- Department of Psychology and Biobehavioral Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Tara M Brinkman
- Department of Psychology and Biobehavioral Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
22
|
Haas-Kogan DA, Aboian MS, Minturn JE, Leary SE, Abdelbaki MS, Goldman S, Elster JD, Kraya A, Lueder MR, Ramakrishnan D, von Reppert M, Liu KX, Rokita JL, Resnick AC, Solomon DA, Phillips JJ, Prados M, Molinaro AM, Waszak SM, Mueller S. Everolimus for Children With Recurrent or Progressive Low-Grade Glioma: Results From the Phase II PNOC001 Trial. J Clin Oncol 2024; 42:441-451. [PMID: 37978951 PMCID: PMC10824388 DOI: 10.1200/jco.23.01838] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/09/2023] [Accepted: 10/17/2023] [Indexed: 11/19/2023] Open
Abstract
PURPOSE The PNOC001 phase II single-arm trial sought to estimate progression-free survival (PFS) associated with everolimus therapy for progressive/recurrent pediatric low-grade glioma (pLGG) on the basis of phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway activation as measured by phosphorylated-ribosomal protein S6 and to identify prognostic and predictive biomarkers. PATIENTS AND METHODS Patients, age 3-21 years, with progressive/recurrent pLGG received everolimus orally, 5 mg/m2 once daily. Frequency of driver gene alterations was compared among independent pLGG cohorts of newly diagnosed and progressive/recurrent patients. PFS at 6 months (primary end point) and median PFS (secondary end point) were estimated for association with everolimus therapy. RESULTS Between 2012 and 2019, 65 subjects with progressive/recurrent pLGG (median age, 9.6 years; range, 3.0-19.9; 46% female) were enrolled, with a median follow-up of 57.5 months. The 6-month PFS was 67.4% (95% CI, 60.0 to 80.0) and median PFS was 11.1 months (95% CI, 7.6 to 19.8). Hypertriglyceridemia was the most common grade ≥3 adverse event. PI3K/AKT/mTOR pathway activation did not correlate with clinical outcomes (6-month PFS, active 68.4% v nonactive 63.3%; median PFS, active 11.2 months v nonactive 11.1 months; P = .80). Rare/novel KIAA1549::BRAF fusion breakpoints were most frequent in supratentorial midline pilocytic astrocytomas, in patients with progressive/recurrent disease, and correlated with poor clinical outcomes (median PFS, rare/novel KIAA1549::BRAF fusion breakpoints 6.1 months v common KIAA1549::BRAF fusion breakpoints 16.7 months; P < .05). Multivariate analysis confirmed their independent risk factor status for disease progression in PNOC001 and other, independent cohorts. Additionally, rare pathogenic germline variants in homologous recombination genes were identified in 6.8% of PNOC001 patients. CONCLUSION Everolimus is a well-tolerated therapy for progressive/recurrent pLGGs. Rare/novel KIAA1549::BRAF fusion breakpoints may define biomarkers for progressive disease and should be assessed in future clinical trials.
Collapse
Affiliation(s)
- Daphne A. Haas-Kogan
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Mariam S. Aboian
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT
| | - Jane E. Minturn
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Sarah E.S. Leary
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA
| | - Mohamed S. Abdelbaki
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO
| | - Stewart Goldman
- Phoenix Children's Hospital, Phoenix, AZ
- University of Arizona College of Medicine, Phoenix, AZ
| | - Jennifer D. Elster
- Division of Hematology Oncology, Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, San Diego, CA
| | - Adam Kraya
- Division of Neurosurgery, Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Matthew R. Lueder
- Division of Neurosurgery, Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Divya Ramakrishnan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT
| | - Marc von Reppert
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT
- University of Leipzig, Leipzig, Germany
| | - Kevin X. Liu
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Jo Lynne Rokita
- Division of Neurosurgery, Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Adam C. Resnick
- Division of Neurosurgery, Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - David A. Solomon
- Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Joanna J. Phillips
- Department of Pathology, University of California, San Francisco, San Francisco, CA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Michael Prados
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA
| | - Annette M. Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Sebastian M. Waszak
- Laboratory of Computational Neuro-Oncology, Swiss Institute for Experimental Cancer Research, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Department of Neurology, University of California, San Francisco, San Francisco, CA
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Sabine Mueller
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA
- Department of Neurology, University of California, San Francisco, San Francisco, CA
- Department of Pediatrics, University of Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Barbato MI, Nashed J, Bradford D, Ren Y, Khasar S, Miller CP, Zolnik BS, Zhao H, Li Y, Bi Y, Shord SS, Amatya AK, Mishra-Kalyani PS, Scepura B, Al-Matari RA, Pazdur R, Kluetz PG, Donoghue M, Singh H, Drezner N. FDA Approval Summary: Dabrafenib in Combination with Trametinib for BRAFV600E Mutation-Positive Low-Grade Glioma. Clin Cancer Res 2024; 30:263-268. [PMID: 37610803 PMCID: PMC10841289 DOI: 10.1158/1078-0432.ccr-23-1503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/07/2023] [Accepted: 08/22/2023] [Indexed: 08/24/2023]
Abstract
On March 16, 2023, the FDA approved dabrafenib in combination with trametinib (Tafinlar, Mekinist; Novartis Pharmaceuticals Corporation) for the treatment of pediatric patients with low-grade glioma (LGG) with a BRAFV600E mutation who require systemic therapy. FDA also approved oral formulations of both drugs suitable for patients who cannot swallow pills. This approval was based on the LGG cohort from study CDRB436G2201 (NCT02684058), a multicenter, open-label trial in which pediatric patients with LGG with a BRAFV600E mutation were randomly assigned 2:1 to dabrafenib plus trametinib (D+T) or carboplatin plus vincristine (C+V). The overall response rate (ORR) by independent review based on Response Assessment in Neuro-oncology LGG (2017) criteria was assessed in 110 patients randomly assigned to D+T (n = 73) or C+V (n = 37). ORR was 47% [95% confidence interval (CI), 35-59] in the D+T arm and 11% (95% CI, 3.0-25) in the C+V arm. Duration of response (DOR) was 23.7 months (95% CI, 14.5-NE) in the D+T arm and not estimable (95% CI, 6.6- NE) in the C+V arm. Progression-free survival (PFS) was 20.1 months (95% CI: 12.8, NE) and 7.4 months (95% CI, 3.6- 11.8) [HR, 0.31 (95% CI, 0.17-0.55); P < 0.001] in the D+T and C+V arms, respectively. The most common (>20%) adverse reactions were pyrexia, rash, headache, vomiting, musculoskeletal pain, fatigue, diarrhea, dry skin, nausea, hemorrhage, abdominal pain, and dermatitis acneiform. This represents the first FDA approval of a systemic therapy for the first-line treatment of pediatric patients with LGG with a BRAFV600E mutation.
Collapse
Affiliation(s)
- Michael I. Barbato
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Jeannette Nashed
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Diana Bradford
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Yi Ren
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Sachia Khasar
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Claudia P. Miller
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Banu S. Zolnik
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Hong Zhao
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Yangbing Li
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Youwei Bi
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Stacy S. Shord
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | - Anup K. Amatya
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | | | - Barbara Scepura
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| | | | - Richard Pazdur
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
- Oncology Center of Excellence, U.S. Food and Drug Administration
| | - Paul G. Kluetz
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
- Oncology Center of Excellence, U.S. Food and Drug Administration
| | - Martha Donoghue
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
- Oncology Center of Excellence, U.S. Food and Drug Administration
| | - Harpreet Singh
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
- Oncology Center of Excellence, U.S. Food and Drug Administration
| | - Nicole Drezner
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration
| |
Collapse
|
24
|
Fangusaro J, Jones DT, Packer RJ, Gutmann DH, Milde T, Witt O, Mueller S, Fisher MJ, Hansford JR, Tabori U, Hargrave D, Bandopadhayay P. Pediatric low-grade glioma: State-of-the-art and ongoing challenges. Neuro Oncol 2024; 26:25-37. [PMID: 37944912 PMCID: PMC10768984 DOI: 10.1093/neuonc/noad195] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023] Open
Abstract
The most common childhood central nervous system (CNS) tumor is pediatric low-grade glioma (pLGG), representing 30%-40% of all CNS tumors in children. Although there is high associated morbidity, tumor-related mortality is relatively rare. pLGG is now conceptualized as a chronic disease, underscoring the importance of functional outcomes and quality-of-life measures. A wealth of data has emerged about these tumors, including a better understanding of their natural history and their molecular drivers, paving the way for the use of targeted inhibitors. While these treatments have heralded tremendous promise, challenges remain about how to best optimize their use, and the long-term toxicities associated with these inhibitors remain unknown. The International Pediatric Low-Grade Glioma Coalition (iPLGGc) is a global group of physicians and scientists with expertise in pLGG focused on addressing key pLGG issues. Here, the iPLGGc provides an overview of the current state-of-the-art in pLGG, including epidemiology, histology, molecular landscape, treatment paradigms, survival outcomes, functional outcomes, imaging response, and ongoing challenges. This paper also serves as an introduction to 3 other pLGG manuscripts on (1) pLGG preclinical models, (2) consensus framework for conducting early-phase clinical trials in pLGG, and (3) pLGG resistance, rebound, and recurrence.
Collapse
Affiliation(s)
- Jason Fangusaro
- Department of Hematology and Oncology, Children’s Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA
| | - David T Jones
- Translational Program, Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), University Hospital Heidelberg, Heidelberg, Germany
| | - Roger J Packer
- Brain Tumor Institute, Daniel and Jennifer Gilbert Neurofibromatosis Institute, Neuroscience and Behavioral Medicine, Children’s National Medical Center, Washington, District of Columbia, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Till Milde
- Translational Program, Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Olaf Witt
- Translational Program, Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Sabine Mueller
- Department of Neurological Surgery, University of California, San Francisco, California, USA
- Department of Pediatrics, University of California, San Francisco, California, USA
- Department of Neurology, University of California, San Francisco, California, USA
- Department of Oncology, University Children’s Hospital Zürich, Zürich, Switzerland
| | - Michael J Fisher
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jordan R Hansford
- Michael Rice Centre for Hematology and Oncology, Women’s and Children’s Hospital, Adelaide, South Australia, Australia
- South Australia Health and Medical Research Institute, Adelaide, South Australia, Australia
- South Australia ImmunoGENomics Cancer Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Uri Tabori
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Darren Hargrave
- University College London Great Ormond Street Institute of Child Health, London, UK
| | - Pratiti Bandopadhayay
- Department of Pediatric Oncology, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Kim EY, Vavere AL, Snyder SE, Chiang J, Li Y, Patni T, Qaddoumi I, Merchant TE, Robinson GW, Holtrop JL, Shulkin BL, Bag AK. [11C]-methionine positron emission tomography in the evaluation of pediatric low-grade gliomas. Neurooncol Adv 2024; 6:vdae056. [PMID: 38680989 PMCID: PMC11055465 DOI: 10.1093/noajnl/vdae056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024] Open
Abstract
Background [11C]-Methionine positron emission tomography (PET; [11C]-MET-PET) is principally used for the evaluation of brain tumors in adults. Although amino acid PET tracers are more commonly used in the evaluation of pediatric brain tumors, data on [11C]-MET-PET imaging of pediatric low-grade gliomas (pLGG) is scarce. This study aimed to investigate the roles of [11C]-MET-PET in the evaluation of pLGGs. Methods Eighteen patients with newly diagnosed pLGG and 26 previously treated pLGG patients underwent [11C]-MET-PET met the inclusion and exclusion criteria. Tumor-to-brain uptake ratio (TBR) and metabolic tumor volumes were assessed for diagnostic performances (newly diagnosed, 15; previously treated 26), change with therapy (newly diagnosed, 9; previously treated 7), and variability among different histology (n = 12) and molecular markers (n = 7) of pLGGs. Results The sensitivity of [11C]-MET-PET for diagnosing pLGG, newly diagnosed, and previously treated combined was 93% for both TBRmax and TBRpeak, 76% for TBRmean, and 95% for qualitative evaluation. TBRmax showed a statistically significant reduction after treatment, while other PET parameters showed a tendency to decrease. Median TBRmax, TBRpeak, and TBRmean values were slightly higher in the BRAFV600E mutated tumors compared to the BRAF fused tumors. Median TBRmax, and TBRpeak in diffuse astrocytomas were higher compared to pilocytic astrocytomas, but median TBRmean, was slightly higher in pilocytic astrocytomas. However, formal statistical analysis was not done due to the small sample size. Conclusions Our study shows that [11C]-MET-PET reliably characterizes new and previously treated pLGGs. Our study also shows that quantitative parameters tend to decrease with treatment, and differences may exist between various pLGG types.
Collapse
Affiliation(s)
- Emily Y Kim
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Amy L Vavere
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Scott E Snyder
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jason Chiang
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Yimei Li
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Tushar Patni
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Ibrahim Qaddoumi
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Thomas E Merchant
- Department of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Giles W Robinson
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Joseph L Holtrop
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Barry L Shulkin
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Asim K Bag
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
26
|
von Reppert M, Ramakrishnan D, Brüningk SC, Memon F, Abi Fadel S, Maleki N, Bahar R, Avesta AE, Jekel L, Sala M, Lost J, Tillmanns N, Kaur M, Aneja S, Fathi Kazerooni A, Nabavizadeh A, Lin M, Hoffmann KT, Bousabarah K, Swanson KR, Haas-Kogan D, Mueller S, Aboian MS. Comparison of volumetric and 2D-based response methods in the PNOC-001 pediatric low-grade glioma clinical trial. Neurooncol Adv 2024; 6:vdad172. [PMID: 38221978 PMCID: PMC10785766 DOI: 10.1093/noajnl/vdad172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
Background Although response in pediatric low-grade glioma (pLGG) includes volumetric assessment, more simplified 2D-based methods are often used in clinical trials. The study's purpose was to compare volumetric to 2D methods. Methods An expert neuroradiologist performed solid and whole tumor (including cyst and edema) volumetric measurements on MR images using a PACS-based manual segmentation tool in 43 pLGG participants (213 total follow-up images) from the Pacific Pediatric Neuro-Oncology Consortium (PNOC-001) trial. Classification based on changes in volumetric and 2D measurements of solid tumor were compared to neuroradiologist visual response assessment using the Brain Tumor Reporting and Data System (BT-RADS) criteria for a subset of 65 images using receiver operating characteristic (ROC) analysis. Longitudinal modeling of solid tumor volume was used to predict BT-RADS classification in 54 of the 65 images. Results There was a significant difference in ROC area under the curve between 3D solid tumor volume and 2D area (0.96 vs 0.78, P = .005) and between 3D solid and 3D whole volume (0.96 vs 0.84, P = .006) when classifying BT-RADS progressive disease (PD). Thresholds of 15-25% increase in 3D solid tumor volume had an 80% sensitivity in classifying BT-RADS PD included in their 95% confidence intervals. The longitudinal model of solid volume response had a sensitivity of 82% and a positive predictive value of 67% for detecting BT-RADS PD. Conclusions Volumetric analysis of solid tumor was significantly better than 2D measurements in classifying tumor progression as determined by BT-RADS criteria and will enable more comprehensive clinical management.
Collapse
Affiliation(s)
- Marc von Reppert
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neuroradiology, Leipzig University Hospital, Leipzig, Germany
| | - Divya Ramakrishnan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sarah C Brüningk
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
| | - Fatima Memon
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sandra Abi Fadel
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nazanin Maleki
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ryan Bahar
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Arman E Avesta
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, USA
- Center for Outcomes Research and Evaluation (CORE), Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neuroradiology, Harvard Medical School—Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Leon Jekel
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- University of Duisburg-Essen, Essen, Germany
| | - Matthew Sala
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- Tulane School of Medicine, New Orleans, Louisiana, USA
| | - Jan Lost
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Niklas Tillmanns
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Manpreet Kaur
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- Ludwig Maximilian University, Munich, Germany
| | - Sanjay Aneja
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, USA
- Center for Outcomes Research and Evaluation (CORE), Yale School of Medicine, New Haven, Connecticut, USA
| | - Anahita Fathi Kazerooni
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ali Nabavizadeh
- Center for Data-Driven Discovery in Biomedicine (D3b), Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - MingDe Lin
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- Visage Imaging, Inc., San Diego, California, USA
| | | | | | - Kristin R Swanson
- Mathematical Neuro-Oncology Lab, Department of Neurological Surgery, Mayo Clinic, Phoenix, Arizona, USA
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sabine Mueller
- Department of Neurology, Neurosurgery, and Pediatrics, UCSF, San Francisco, California, USA
- Children’s University Hospital Zürich, Zürich, Switzerland
| | - Mariam S Aboian
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
27
|
Malbari F. Pediatric Neuro-oncology. Continuum (Minneap Minn) 2023; 29:1680-1709. [PMID: 38085894 DOI: 10.1212/con.0000000000001360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
OBJECTIVE This article reviews the most common pediatric brain tumors, neurocutaneous syndromes, treatment-related neurotoxicities, and the long-term outcomes of survivors. LATEST DEVELOPMENTS In the era of molecular diagnostics, the classification, management, and prognostication of pediatric brain tumors and neurocutaneous syndromes has been refined, resulting in advancements in patient management. Molecular diagnostics have been incorporated into the most recent World Health Organization 2021 classification. This knowledge has allowed for novel therapeutic approaches targeting the biology of these tumors with the intent to improve overall survival, decrease treatment-related morbidity, and improve quality of life. Advances in management have led to better survival, but mortality remains high and significant morbidity persists. Current clinical trials focus on tumor biology targeted therapy, deescalation of therapy, and multimodal intensified approaches with targeted therapy in more high-risk tumors. ESSENTIAL POINTS Molecular diagnostics for pediatric brain tumors and neurocutaneous syndromes have led to novel therapeutic approaches targeting the biology of these tumors with the goals of improving overall survival and decreasing treatment-related morbidity. Further understanding will lead to continued refinement and improvement of tumor classification, management, and prognostication.
Collapse
|
28
|
Mastrangelo S. Special Issue: Childhood Brain Cancer Treatment. Cancers (Basel) 2023; 15:5278. [PMID: 37958448 PMCID: PMC10647487 DOI: 10.3390/cancers15215278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Brain cancer is the second most common childhood malignancy and is the leading cause of death among all pediatric cancers [...].
Collapse
Affiliation(s)
- Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Università Cattolica del Sacro Cuore di Roma, Largo A. Gemelli, 8, 00168 Rome, Italy
| |
Collapse
|
29
|
Hardin EC, Schmid S, Sommerkamp A, Bodden C, Heipertz AE, Sievers P, Wittmann A, Milde T, Pfister SM, von Deimling A, Horn S, Herz NA, Simon M, Perera AA, Azizi A, Cruz O, Curry S, Van Damme A, Garami M, Hargrave D, Kattamis A, Kotnik BF, Lähteenmäki P, Scheinemann K, Schouten-van Meeteren AYN, Sehested A, Viscardi E, Wormdal OM, Zapotocky M, Ziegler DS, Koch A, Hernáiz Driever P, Witt O, Capper D, Sahm F, Jones DTW, van Tilburg CM. LOGGIC Core BioClinical Data Bank: Added clinical value of RNA-Seq in an international molecular diagnostic registry for pediatric low-grade glioma patients. Neuro Oncol 2023; 25:2087-2097. [PMID: 37075810 PMCID: PMC10628936 DOI: 10.1093/neuonc/noad078] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND The international, multicenter registry LOGGIC Core BioClinical Data Bank aims to enhance the understanding of tumor biology in pediatric low-grade glioma (pLGG) and provide clinical and molecular data to support treatment decisions and interventional trial participation. Hence, the question arises whether implementation of RNA sequencing (RNA-Seq) using fresh frozen (FrFr) tumor tissue in addition to gene panel and DNA methylation analysis improves diagnostic accuracy and provides additional clinical benefit. METHODS Analysis of patients aged 0 to 21 years, enrolled in Germany between April 2019 and February 2021, and for whom FrFr tissue was available. Central reference histopathology, immunohistochemistry, 850k DNA methylation analysis, gene panel sequencing, and RNA-Seq were performed. RESULTS FrFr tissue was available in 178/379 enrolled cases. RNA-Seq was performed on 125 of these samples. We confirmed KIAA1549::BRAF-fusion (n = 71), BRAF V600E-mutation (n = 12), and alterations in FGFR1 (n = 14) as the most frequent alterations, among other common molecular drivers (n = 12). N = 16 cases (13%) presented rare gene fusions (eg, TPM3::NTRK1, EWSR1::VGLL1, SH3PXD2A::HTRA1, PDGFB::LRP1, GOPC::ROS1). In n = 27 cases (22%), RNA-Seq detected a driver alteration not otherwise identified (22/27 actionable). The rate of driver alteration detection was hereby increased from 75% to 97%. Furthermore, FGFR1 internal tandem duplications (n = 6) were only detected by RNA-Seq using current bioinformatics pipelines, leading to a change in analysis protocols. CONCLUSIONS The addition of RNA-Seq to current diagnostic methods improves diagnostic accuracy, making precision oncology treatments (MEKi/RAFi/ERKi/NTRKi/FGFRi/ROSi) more accessible. We propose to include RNA-Seq as part of routine diagnostics for all pLGG patients, especially when no common pLGG alteration was identified.
Collapse
Affiliation(s)
- Emily C Hardin
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK)
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Medical Faculty, University of Heidelberg, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Simone Schmid
- Department of Neuropathology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alexander Sommerkamp
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Paediatrics and Adolescent Medicine, The University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Carina Bodden
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK)
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Anna-Elisa Heipertz
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK)
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg Medical Faculty, University of Heidelberg, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Philipp Sievers
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Andrea Wittmann
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Till Milde
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK)
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas von Deimling
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Saint Luc University Hospital, Brussels, Belgium
| | - Svea Horn
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, HIT-LOGGIC German Registry for children and adolescents with low-grade glioma, Berlin, Germany
| | - Nina A Herz
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, HIT-LOGGIC German Registry for children and adolescents with low-grade glioma, Berlin, Germany
| | - Michèle Simon
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, HIT-LOGGIC German Registry for children and adolescents with low-grade glioma, Berlin, Germany
- Department of Pediatric Oncology/Hematology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ashwyn A Perera
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Heidelberg Medical Faculty, University of Heidelberg, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Amedeo Azizi
- Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Ofelia Cruz
- Neuro-Oncology Unit, Pediatric Cancer Center, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Sarah Curry
- Department of Haematology and Oncology, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - An Van Damme
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Saint Luc University Hospital, Brussels, Belgium
| | - Miklos Garami
- 2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Darren Hargrave
- Great Ormond Street Hospital for Children NHS Trust London, London, UK
| | - Antonis Kattamis
- Department of Neuropathology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- First Department of Paediatrics, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Barbara Faganel Kotnik
- Department of Haematology and Oncology, University Children’s Hospital, University Medical Centre Ljubljana (UMC), Ljubljana, Slovenia
| | - Päivi Lähteenmäki
- Turku University and University Hospital, Turku, Finland
- Swedish Childhood Cancer Registry, Karolinska Institutet, Stockholm, Sweden
| | - Katrin Scheinemann
- Division of Pediatric Oncology – Hematology, Department of Pediatrics, Kantonsspital Aarau, Aarau, Switzerland
- Department of Health Sciences and Medicine, University of Lucerne, Lucerne, Switzerland
- Department of Paediatrics, McMaster Children’s Hospital and McMaster University, Hamilton, Canada
| | | | - Astrid Sehested
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Paediatrics and Adolescent Medicine, The University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | - Ole Mikal Wormdal
- Section of Pediatric Oncology, UNN University Hospital of Northern Norway, Tromsø, Norway
| | - Michal Zapotocky
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - David S Ziegler
- Kids Cancer Centre, Sydney Children’s Hospital, High St, Randwick, NSW, Australia
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| | - Arend Koch
- Department of Neuropathology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- First Department of Paediatrics, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Pablo Hernáiz Driever
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, HIT-LOGGIC German Registry for children and adolescents with low-grade glioma, Berlin, Germany
- Department of Pediatric Oncology/Hematology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Olaf Witt
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK)
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - David Capper
- Department of Neuropathology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Sahm
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - David T W Jones
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cornelis M van Tilburg
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK)
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
30
|
Bouffet E, Hansford JR, Garrè ML, Hara J, Plant-Fox A, Aerts I, Locatelli F, van der Lugt J, Papusha L, Sahm F, Tabori U, Cohen KJ, Packer RJ, Witt O, Sandalic L, Bento Pereira da Silva A, Russo M, Hargrave DR. Dabrafenib plus Trametinib in Pediatric Glioma with BRAF V600 Mutations. N Engl J Med 2023; 389:1108-1120. [PMID: 37733309 DOI: 10.1056/nejmoa2303815] [Citation(s) in RCA: 75] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
BACKGROUND Detection of the BRAF V600E mutation in pediatric low-grade glioma has been associated with a lower response to standard chemotherapy. In previous trials, dabrafenib (both as monotherapy and in combination with trametinib) has shown efficacy in recurrent pediatric low-grade glioma with BRAF V600 mutations, findings that warrant further evaluation of this combination as first-line therapy. METHODS In this phase 2 trial, patients with pediatric low-grade glioma with BRAF V600 mutations who were scheduled to receive first-line therapy were randomly assigned in a 2:1 ratio to receive dabrafenib plus trametinib or standard chemotherapy (carboplatin plus vincristine). The primary outcome was the independently assessed overall response (complete or partial response) according to the Response Assessment in Neuro-Oncology criteria. Also assessed were the clinical benefit (complete or partial response or stable disease for ≥24 weeks) and progression-free survival. RESULTS A total of 110 patients underwent randomization (73 to receive dabrafenib plus trametinib and 37 to receive standard chemotherapy). At a median follow-up of 18.9 months, an overall response occurred in 47% of the patients treated with dabrafenib plus trametinib and in 11% of those treated with chemotherapy (risk ratio, 4.31; 95% confidence interval [CI], 1.7 to 11.2; P<0.001). Clinical benefit was observed in 86% of the patients receiving dabrafenib plus trametinib and in 46% receiving chemotherapy (risk ratio, 1.88; 95% CI, 1.3 to 2.7). The median progression-free survival was significantly longer with dabrafenib plus trametinib than with chemotherapy (20.1 months vs. 7.4 months; hazard ratio, 0.31; 95% CI, 0.17 to 0.55; P<0.001). Grade 3 or higher adverse events occurred in 47% of the patients receiving dabrafenib plus trametinib and in 94% of those receiving chemotherapy. CONCLUSIONS Among pediatric patients with low-grade glioma with BRAF V600 mutations, dabrafenib plus trametinib resulted in significantly more responses, longer progression-free survival, and a better safety profile than standard chemotherapy as first-line therapy. (Funded by Novartis; ClinicalTrials.gov number, NCT02684058.).
Collapse
Affiliation(s)
- Eric Bouffet
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Jordan R Hansford
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Maria Luisa Garrè
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Junichi Hara
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Ashley Plant-Fox
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Isabelle Aerts
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Franco Locatelli
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Jasper van der Lugt
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Ludmila Papusha
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Felix Sahm
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Uri Tabori
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Kenneth J Cohen
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Roger J Packer
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Olaf Witt
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Larissa Sandalic
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Ana Bento Pereira da Silva
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Mark Russo
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Darren R Hargrave
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| |
Collapse
|
31
|
Weiser A, Sanchez Bergman A, Machaalani C, Bennett J, Roth P, Reimann RR, Nazarian J, Guerreiro Stucklin AS. Bridging the age gap: a review of molecularly informed treatments for glioma in adolescents and young adults. Front Oncol 2023; 13:1254645. [PMID: 37781183 PMCID: PMC10533987 DOI: 10.3389/fonc.2023.1254645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/14/2023] [Indexed: 10/03/2023] Open
Abstract
Gliomas are the most common primary central nervous system (CNS) tumors and a major cause of cancer-related mortality in children (age <15 years), adolescents and young adults (AYA, ages 15-39 years), and adults (age >39 years). Molecular pathology has helped enhance the characterization of these tumors, revealing a heterogeneous and ever more complex group of malignancies. Recent molecular analyses have led to an increased appreciation of common genomic alterations prevalent across all ages. The 2021 World Health Organization (WHO) CNS tumor classification, 5th edition (WHO CNS5) brings forward a nomenclature distinguishing "pediatric-type" and "adult-type" gliomas. The spectrum of gliomas in AYA comprises both "pediatric-like" and "adult-like" tumor entities but remains ill-defined. With fragmentation of clinical management between pediatric and adult centers, AYAs face challenges related to gaps in medical care, lower rates of enrollment in clinical trials and additional psychosocial and economic challenges. This calls for a rethinking of diagnostic and therapeutic approaches, to improve access to appropriate testing and potentially beneficial treatments to patients of all ages.
Collapse
Affiliation(s)
- Annette Weiser
- Translational Brain Tumor Research Group, Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- Division of Oncology, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Astrid Sanchez Bergman
- Translational Brain Tumor Research Group, Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Charbel Machaalani
- Translational Brain Tumor Research Group, Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Julie Bennett
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Patrick Roth
- Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Regina R. Reimann
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Javad Nazarian
- Department of Pediatrics, Diffuse Midline Glioma (DMG) / Diffuse Intrinsic Pontine Glioma (DIPG) Center, Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- Research Center for Genetic Medicine, Children's National Hospital, Washington, DC, United States
| | - Ana S. Guerreiro Stucklin
- Translational Brain Tumor Research Group, Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- Division of Oncology, University Children’s Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Leary SES, Onar-Thomas A, Fangusaro J, Gottardo NG, Cohen K, Smith A, Huang A, Haas-Kogan D, Fouladi M. Children's Oncology Group's 2023 blueprint for research: Central nervous system tumors. Pediatr Blood Cancer 2023; 70 Suppl 6:e30600. [PMID: 37534382 PMCID: PMC10569820 DOI: 10.1002/pbc.30600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 08/04/2023]
Abstract
Tumors of the central nervous system (CNS) are a leading cause of morbidity and mortality in the pediatric population. Molecular characterization in the last decade has redefined CNS tumor diagnoses and risk stratification; confirmed the unique biology of pediatric tumors as distinct entities from tumors that occur in adulthood; and led to the first novel targeted therapies receiving Food and Drug Administration (FDA) approval for children with CNS tumors. There remain significant challenges to overcome: children with unresectable low-grade glioma may require multiple prolonged courses of therapy affecting quality of life; children with high-grade glioma have a dismal long-term prognosis; children with medulloblastoma may suffer significant short- and long-term morbidity from multimodal cytotoxic therapy, and approaches to improve survival in ependymoma remain elusive. The Children's Oncology Group (COG) is uniquely positioned to conduct the next generation of practice-changing clinical trials through rapid prospective molecular characterization and therapy evaluation in well-defined clinical and molecular groups.
Collapse
Affiliation(s)
- Sarah E. S. Leary
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s, Seattle, WA
| | - Arzu Onar-Thomas
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jason Fangusaro
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA
| | | | - Kenneth Cohen
- The Sidney Kimmel Comprehensive Cancer Center, John’s Hopkins, Baltimore, MD
| | - Amy Smith
- Division of Pediatric Hematology, Oncology and Bone Marrow Transplant, Orlando Health-Arnold Palmer Hospital, Orlando, FL
| | - Annie Huang
- Department of Hematology/Oncology, Hospital for Sick Children, Toronto, Canada
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Maryam Fouladi
- Division of Hematology, Oncology, and Bone Marrow Transplant, Nationwide Children’s Hospital, Columbus OH
| |
Collapse
|
33
|
van Schaik J, van Roessel IMAA, Bos ID, Claashen-van der Grinten HL, Clement SC, van Iersel L, Bakker B, Meijer L, Kremer L, Schouten-van Meeteren AYN, van Santen HM. Elevated IGF-1 concentrations in children with low grade glioma: A descriptive analysis in a retrospective national cohort. J Neuroendocrinol 2023; 35:e13317. [PMID: 37439273 DOI: 10.1111/jne.13317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/20/2023] [Accepted: 06/02/2023] [Indexed: 07/14/2023]
Abstract
Children with low grade glioma (LGG) may present with, or develop, elevated concentrations of insulin-like growth factor 1 (IGF-1). The prevalence, pathophysiology, or its possible clinical effects are poorly understood. Our aim was to evaluate the prevalence of such elevated IGF-1 concentrations and to describe its association with linear growth, body mass index (BMI), pituitary outcome, and tumor behavior in a large retrospective national cohort. From a nationwide retrospective cohort of pediatric brain tumor survivors diagnosed between 2002 and 2012, tumor, treatment, endocrine, and auxological data of children with LGG were collected (n = 358). Prevalence and risk factors for elevated IGF-1 concentrations, as well as the association between having elevated IGF-1 concentrations and receiving tumor treatment, were explored. IGF-1 concentrations had only been measured in 45.5% of cases (n = 163/358). In 18.4% of 163 children with available IGF-1 measurements, IGF-1 concentrations were found elevated. No association was described between having an elevated IGF-1 concentration and tumor behavior or height SDS at last moment of follow-up. Multivariate logistic regression identified posterior pituitary disorder (OR 6.14 95% CI: 2.21-17.09) and BMI SDS at follow-up (OR 1.56 95% CI: 1.09-2.20) to be significantly associated with elevated IGF-1 concentrations. In this retrospective cohort of children with LGG, IGF-1 was found elevated in 18.4% of children with available IGF-1 measurements. Elevated IGF-1 seems to be related to hypothalamic dysfunction worsening over time. Larger prospective cohort studies are needed.
Collapse
Affiliation(s)
- Jiska van Schaik
- Division of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Ichelle M A A van Roessel
- Division of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Iris D Bos
- Division of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Sarah C Clement
- Division of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Laura van Iersel
- Division of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Boudewijn Bakker
- Division of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lisethe Meijer
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Leontien Kremer
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Hanneke M van Santen
- Division of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
34
|
Damodharan S, Puccetti D. Pediatric Central Nervous System Tumor Overview and Emerging Treatment Considerations. Brain Sci 2023; 13:1106. [PMID: 37509034 PMCID: PMC10377074 DOI: 10.3390/brainsci13071106] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Pediatric central nervous system (CNS) tumors are the most common solid tumor in children, with the majority being glial in origin. These tumors are classified by the World Health Organization (WHO) as either being low grade (WHO grade 1 and 2) or high grade (WHO grade 3 and 4). Our knowledge of the molecular landscape of pediatric brain tumors has advanced over the last decade, which has led to newer categorizations along with an expansion of therapeutic targets and options. In this review, we will give an overview of common CNS tumors seen in children along with a focus on treatment options and future considerations.
Collapse
Affiliation(s)
- Sudarshawn Damodharan
- Department of Pediatrics, Division of Pediatric Hematology, Oncology and Bone Marrow Transplant, University of Wisconsin School of Medicine & Public Health, Madison, WI 53792, USA
| | - Diane Puccetti
- Department of Pediatrics, Division of Pediatric Hematology, Oncology and Bone Marrow Transplant, University of Wisconsin School of Medicine & Public Health, Madison, WI 53792, USA
| |
Collapse
|
35
|
Pinto SN, Chiang J, Qaddoumi I, Livingston D, Bag A. Pediatric diencephalic tumors: a constellation of entities and management modalities. Front Oncol 2023; 13:1180267. [PMID: 37519792 PMCID: PMC10374860 DOI: 10.3389/fonc.2023.1180267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
The diencephalon is a complex midline structure consisting of the hypothalamus, neurohypophysis, subthalamus, thalamus, epithalamus, and pineal body. Tumors arising from each of these diencephalic components differ significantly in terms of biology and prognosis. The aim of this comprehensive review is to describe the epidemiology, clinical symptoms, imaging, histology, and molecular markers in the context of the 2021 WHO classification of central nervous system neoplasms. We will also discuss the current management of each of these tumors.
Collapse
Affiliation(s)
- Soniya N. Pinto
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Jason Chiang
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Ibrahim Qaddoumi
- Departments of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - David Livingston
- Department of Radiology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Asim Bag
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
36
|
Sun F, Lv H, Feng B, Sun J, Zhang L, Dong B. Identification of natural killer cell-related characteristics to predict the clinical prognosis and immune microenvironment of patients with low-grade glioma. Aging (Albany NY) 2023; 15:6264-6291. [PMID: 37405952 PMCID: PMC10373982 DOI: 10.18632/aging.204850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Individuals with low-grade glioma (LGG) have a dismal prognosis, and most patients will eventually progress to high-grade disease. Therefore, it is crucial to accurately determine their prognoses. METHODS Seventy-nine NK cell genes were downloaded from the LM22 database and univariate Cox regression analysis was utilized to detect NK cell-related genes affecting prognosis. Molecular types were established for LGG using the "ConsensusClusterPlus" R package. The results from a functional enrichment analysis and the immune microenvironment were intensively explored to determine molecular heterogeneity and immune characteristics across distinct subtypes. Furthermore, a RiskScore model was developed and verified using expression profiles of NK cells, and a nomogram consisting of the RiskScore model and clinical traits was constructed. Moreover, pan-cancer traits of NK cells were also investigated. RESULTS The C1 subtype included the greatest amount of immune infiltration and the poorest prognosis among well-established subtypes. The majority of enriched pathways were those involved in tumor progression, including epithelial-mesenchymal transition and cell cycle pathways. Differentially expressed genes among distinct subtypes were determined and used to develop a novel RiskScore model. This model was able to distinguish low-risk patients with LGG from those with high-risk disease. An accurate nomogram including the RiskScore, disease grade and patient's age was constructed to predict clinical outcomes of LGG patients. Finally, a pan-cancer analysis further highlighted the crucial roles of NK cell-related genes in the tumor microenvironment. CONCLUSIONS An NK cell-related RiskScore model can accurately predict the prognoses of patients with LGG and provide valuable insights into personalized medicine.
Collapse
Affiliation(s)
- Fei Sun
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Neurosurgery, Xinhua Hospital Affiliated to Dalian University, Dalian, Liaoning, China
| | - Hongtao Lv
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Baozhi Feng
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jiaao Sun
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Linyun Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bin Dong
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
37
|
Muniz TP, Mason WP. BRAF Mutations in CNS Tumors-Prognostic Markers and Therapeutic Targets. CNS Drugs 2023; 37:587-598. [PMID: 37268805 DOI: 10.1007/s40263-023-01016-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/16/2023] [Indexed: 06/04/2023]
Abstract
Gliomas are a heterogeneous group of brain tumors with limited therapeutic options. However, identification of BRAF V600E mutations in a subset of gliomas has provided a genomic-targeted approach for management of these diseases. In this review, we aimed to review the role of BRAF V600E in gliomagenesis, to characterize concurrent genomic alterations and their potential prognostic implications, and to review comprehensively the efficacy data of BRAF inhibitors (combined or not with MEK inhibitors) for the treatment of low- and high-grade gliomas. We also provide a summary of the toxicity of these agents and describe resistance mechanisms that may be circumvented by alternative genomic approaches. Although the efficacy of targeted therapy for management of BRAF V600E-mutant gliomas has mostly been assessed in small retrospective and phase 2 studies with heterogeneous populations, the data generated so far are a proof of concept that genomic-directed therapies improve outcomes of patients with refractory/relapsed glioma and underpin the need of comprehensive genomic assessments for these difficult-to-treat diseases. In the future, the role of targeted therapy in the first-line setting and of genomic-directed therapies to overcome resistance mechanisms should be assessed in well-designed clinical trials.
Collapse
Affiliation(s)
- Thiago P Muniz
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada.
| | - Warren P Mason
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
38
|
Leblond P, Tresch-Bruneel E, Probst A, Néant N, Solas C, Sterin A, Boulanger T, Aerts I, Faure-Conter C, Bertozzi AI, Chastagner P, Entz-Werlé N, De Carli E, Deley MCL, Bouche G, André N. Phase I Study of a Combination of Fluvastatin and Celecoxib in Children with Relapsing/Refractory Low-Grade or High-Grade Glioma (FLUVABREX). Cancers (Basel) 2023; 15:cancers15072020. [PMID: 37046681 PMCID: PMC10093481 DOI: 10.3390/cancers15072020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Preclinical data support the activity of celecoxib and fluvastatin in high-grade (HGG) and low-grade gliomas (LGG). A phase I trial (NCT02115074) was designed to evaluate the safety of this combination in children with refractory/relapsed HGG and LGG using four dose levels of fluvastatin with a fixed daily dose of celecoxib. A Continual Reassessment Method was used for fluvastatin dose escalation. Dose-limiting toxicities (DLT) were determined on the first treatment cycle. Twenty patients were included. Ten LGG and ten HGG patients received a median of 3.5 treatment cycles. Two DLTs were reported: one grade 3 maculopapular rash (4 mg/kg dose level) and one grade 4 increase of Creatine Phospho-Kinase (6 mg/kg dose level). We identified the dose of 6 mg/kg/day as the recommended phase II dose (RP2D) of fluvastatin with celecoxib. Four patients with LGG continued treatment beyond 12 cycles because of stable disease, including one patient who received 23 treatment cycles. In children with refractory/relapsed glioma, the RP2D of fluvastatin with celecoxib is 6 mg/kg/day. The long-term stable diseases observed in LGG suggest a possible role of the combination in a maintenance setting, given its good tolerance and low cost for children living in low- and middle-income countries.
Collapse
|
39
|
Ronsley R, Lazow M, Henry RK. Growth hormone after CNS tumor diagnosis: the fundamentals, fears, facts, and future directions. Pediatr Hematol Oncol 2023; 40:786-799. [PMID: 36939305 DOI: 10.1080/08880018.2023.2190765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 03/21/2023]
Abstract
Growth hormone deficiency (GHD) may occur in pediatric patients with central nervous system (CNS) tumors at initial tumor presentation or later as treatment-related sequelae. While it is well recognized that growth hormone (GH) has beneficial effects on growth and endocrinopathies, there's often hesitancy by clinicians to initiate GH therapy for GHD after CNS tumor diagnosis due to the perceived increased risk of tumor recurrence. The available data is described here and based on this review, there is no evidence of increased risk of tumor recurrence or secondary malignancy in patients treated with GH after CNS tumor diagnosis. Further understanding of tumor biology and presence of downstream GH targets including insulin-like growth factor-1 (IGF-1) and insulin receptor activity is still needed.
Collapse
Affiliation(s)
- Rebecca Ronsley
- Section of Hematology, Oncology & BMT, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Section of Hematology, Oncology & BMT, Department of Pediatrics, Seattle Children's Hospital, The University of Washington, Seattle, Washington, USA
| | - Margot Lazow
- Section of Hematology, Oncology & BMT, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Rohan K Henry
- Section of Endocrinology, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
40
|
Sigaud R, Rösch L, Gatzweiler C, Benzel J, von Soosten L, Peterziel H, Selt F, Najafi S, Ayhan S, Gerloff XF, Hofmann N, Büdenbender I, Schmitt L, Foerster KI, Burhenne J, Haefeli WE, Korshunov A, Sahm F, van Tilburg CM, Jones DTW, Pfister SM, Knoerzer D, Kreider BL, Sauter M, Pajtler KW, Zuckermann M, Oehme I, Witt O, Milde T. The first-in-class ERK inhibitor ulixertinib shows promising activity in mitogen-activated protein kinase (MAPK)-driven pediatric low-grade glioma models. Neuro Oncol 2023; 25:566-579. [PMID: 35882450 PMCID: PMC10013652 DOI: 10.1093/neuonc/noac183] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Pediatric low-grade gliomas (pLGG) are the most common pediatric central nervous system tumors, with driving alterations typically occurring in the MAPK pathway. The ERK1/2 inhibitor ulixertinib (BVD-523) has shown promising responses in adult patients with mitogen-activated protein kinase (MAPK)-driven solid tumors. METHODS We investigated the antitumoral activity of ulixertinib monotherapy as well as in combination with MEK inhibitors (MEKi), BH3-mimetics, or chemotherapy in pLGG. Patient-derived pLGG models reflecting the two most common alterations in the disease, KIAA1549:BRAF-fusion and BRAFV600E mutation (DKFZ-BT66 and BT40, respectively) were used for in vitro and in vivo (zebrafish embryos and mice) efficacy testing. RESULTS Ulixertinib inhibited MAPK pathway activity in both models, and reduced cell viability in BT40 with clinically achievable concentrations in the low nanomolar range. Combination treatment of ulixertinib with MEKi or BH3-mimetics showed strong evidence of antiproliferative synergy in vitro. Ulixertinib showed on-target activity in all tested combinations. In vivo, sufficient penetrance of the drug into brain tumor tissue in concentrations above the in vitro IC50 and reduction of MAPK pathway activity was achieved. In a preclinical mouse trial, ulixertinib mono- and combined therapies slowed tumor growth and increased survival. CONCLUSIONS These data indicate a high clinical potential of ulixertinib for the treatment of pLGG and strongly support its first clinical evaluation in pLGG as single agent and in combination therapy in a currently planned international phase I/II umbrella trial.
Collapse
Affiliation(s)
- Romain Sigaud
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Lisa Rösch
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Charlotte Gatzweiler
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Julia Benzel
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laura von Soosten
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.,Preclinical Modeling Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heike Peterziel
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Florian Selt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,KiTZ Clinical Trial Unit, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sara Najafi
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,KiTZ Clinical Trial Unit, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Simay Ayhan
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,KiTZ Clinical Trial Unit, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Xenia F Gerloff
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,KiTZ Clinical Trial Unit, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Nina Hofmann
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Preclinical Modeling Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Isabel Büdenbender
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Lukas Schmitt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Pediatric Soft Tissue Sarcoma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kathrin I Foerster
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jürgen Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Walter E Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Andrey Korshunov
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cornelis M van Tilburg
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,KiTZ Clinical Trial Unit, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,KiTZ Clinical Trial Unit, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | - Max Sauter
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Kristian W Pajtler
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,KiTZ Clinical Trial Unit, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marc Zuckermann
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Preclinical Modeling Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ina Oehme
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,KiTZ Clinical Trial Unit, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Till Milde
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,KiTZ Clinical Trial Unit, Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
41
|
Dang H, Khan AB, Gadgil N, Sharma H, Trandafir C, Malbari F, Weiner HL. Behavioral Improvements following Lesion Resection for Pediatric Epilepsy: Pediatric Psychosurgery? Pediatr Neurosurg 2023; 58:80-88. [PMID: 36787706 PMCID: PMC10233708 DOI: 10.1159/000529683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/08/2023] [Indexed: 02/16/2023]
Abstract
INTRODUCTION Resection of brain lesions associated with refractory epilepsy to achieve seizure control is well accepted. However, concurrent behavioral effects of these lesions such as changes in mood, personality, and cognition and the effects of surgery on behavior have not been well characterized. We describe 5 such children with epileptogenic lesions and significant behavioral abnormalities which improved after surgery. CASE DESCRIPTIONS Five children (ages 3-14 years) with major behavioral abnormalities and lesional epilepsy were identified and treated at our center. Behavioral problems included academic impairment, impulsivity, self-injurious behavior, and decreased social interaction with diagnoses of ADHD, oppositional defiant disorder, and autism. Pre-operative neuropsychiatric testing was performed in 4/5 patients and revealed low-average cognitive and intellectual abilities for their age, attentional difficulties, and poor memory. Lesions were located in the temporal (2 gangliogliomas, 1 JPA, 1 cavernoma) and parietal (1 DNET) lobes. Gross total resection was achieved in all cases. At mean 1-year follow-up, seizure freedom (Engel 1a in 3 patients, Engel 1c in 2 patients) and significant behavioral improvements (academic performance, attention, socialization, and aggression) were achieved in all. Two patients manifested violence pre-operatively; one had extreme behavior with violence toward teachers and peers despite low seizure burden. Since surgery, his behavior has normalized. CONCLUSION We identified 5 patients with severe behavioral disorders in the setting of lesional epilepsy, all of whom demonstrated improvement after surgery. The degree of behavioral abnormality was disproportionate to epilepsy severity, suggesting a more complicated mechanism by which lesional epilepsy impacts behavior. We propose a novel paradigm in which lesionectomy may offer behavioral benefit even when seizures are not refractory. Thus, behavioral improvement may be an important novel goal for neurosurgical resection in children with epileptic brain lesions.
Collapse
Affiliation(s)
- Huy Dang
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA,
| | - Abdul Basit Khan
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
| | - Nisha Gadgil
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
- Division of Pediatric Neurosurgery, Department of Surgery, Texas Children's Hospital, Houston, Texas, USA
| | - Himanshu Sharma
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
| | - Cristina Trandafir
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Texas Children's Hospital, Houston, Texas, USA
| | - Fatema Malbari
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Texas Children's Hospital, Houston, Texas, USA
| | - Howard L Weiner
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
- Division of Pediatric Neurosurgery, Department of Surgery, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
42
|
Bevacizumab as Single Agent in Children and Teenagers with Optic Pathway Glioma. Cancers (Basel) 2023; 15:cancers15041036. [PMID: 36831379 PMCID: PMC9954224 DOI: 10.3390/cancers15041036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
This is a retrospective study conducted on patients with OPG, aged less than 19 years, treated with bevacizumab as a single agent, since 2010 at IHOPe (Institute of Pediatric Hematology and Oncology). Efficacy of the treatment was evaluated on the tumor response rate on MRI with a centralized review basing upon RAPNO criteria and with visual assessment basing upon a 0.2 log change in the logMAR scale. Thirty-one patients with OPG have been included. From a radiological point of view, best anytime responses were: 1 major response, 6 partial responses, 7 minor responses and 14 stable diseases; achieving disease control in 28 (96%) out of 29 patients. Ophthalmological response was evaluated in 25 patients and disease control was achieved in 22 (88%) out of 25, with 14 steady states and 8 significant improvements. Among patients treated with chemotherapy after the bevacizumab course, nine relapsed and have been retreated with objective responses. Bevacizumab used as single agent seems effective in children and adolescents with OPG. Our work paves the way for a phase II study in which bevacizumab alone could be used as frontline therapy.
Collapse
|
43
|
Kapadia T, Sahu A, Mahajan A, Ahuja A, Chatterjee A, Sahu A, Warade A, Padashetty S, Vijan A, Chidambaranthan N, Dasgupta A, Sridhar E, Sahay A, Shetty P, Prasad M, Chinnaswamy G, Moiyadi A, Gupta T, Gala F, Sankhe S. Imaging Guidelines and Recommendations for Diagnosis, Surveillance, and Management of Pediatric CNS and Spinal Tumors. Indian J Med Paediatr Oncol 2023. [DOI: 10.1055/s-0042-1759716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
AbstractCentral nervous system (CNS) tumors are the second most common cause of cancer in children when incidence rates of cancer are estimated according to the Indian population dynamics based on 2011 consensus. As per the estimates, CNS tumors account for 20.1% of cancer burden in children aged between 0 and 14 years and 16.8% when 0 to 19 years age group is considered. The most common pediatric brain tumors are astrocytoma and medulloblastoma followed by other embryonal tumors, craniopharyngioma, and ependymal tumors. The incidence of CNS tumors in children from India is similar to the western high-income countries, other than slightly higher incidence of craniopharyngioma in Indian children.
Collapse
Affiliation(s)
- Tejas Kapadia
- Children's X-Ray Department/Academic Unit of Paediatric Radiology, Royal Manchester Children's Hospital, United Kingdom
| | - Arpita Sahu
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, TMC &Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Abhishek Mahajan
- Department of Radiology, The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, United Kingdom
| | | | - Abhishek Chatterjee
- Department of Radiation Oncology, Tata Memorial Hospital; TMC &Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Amit Sahu
- Lilavati Hospital, Mumbai, Maharashtra, India
| | | | - Shubham Padashetty
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, TMC &Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Antariksh Vijan
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, TMC &Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | | | - Archya Dasgupta
- Department of Radiation Oncology, Tata Memorial Hospital; TMC &Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Epari Sridhar
- Department of Pathology, Tata Memorial Hospital; TMC &Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Ayushi Sahay
- Department of Pathology, Tata Memorial Hospital; TMC &Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Prakash Shetty
- Department of Neurosurgery, Tata Memorial Hospital; TMC &Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Maya Prasad
- Department of Paediatric Oncology, Tata Memorial Hospital; TMC &Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Girish Chinnaswamy
- Department of Paediatric Oncology, Tata Memorial Hospital; TMC &Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Aliasgar Moiyadi
- Department of Neurosurgery, Tata Memorial Hospital; TMC &Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Tejpal Gupta
- Department of Radiation Oncology, Tata Memorial Hospital; TMC &Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Foram Gala
- Bai Jerbai Wadia Hospital for Children, Parel, Mumbai, Maharashtra, India
| | - Shilpa Sankhe
- King Edward Memorial Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
44
|
Tsai JW, Choi JJ, Ouaalam H, Murillo EA, Yeo KK, Vogelzang J, Sousa C, Woods JK, Ligon KL, Warfield SK, Bandopadhayay P, Cooney TM. Integrated response analysis of pediatric low-grade gliomas during and after targeted therapy treatment. Neurooncol Adv 2023; 5:vdac182. [PMID: 36926246 PMCID: PMC10011805 DOI: 10.1093/noajnl/vdac182] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Pediatric low-grade gliomas (pLGGs) are the most common central nervous system tumor in children, characterized by RAS/MAPK pathway driver alterations. Genomic advances have facilitated the use of molecular targeted therapies, however, their long-term impact on tumor behavior remains critically unanswered. Methods We performed an IRB-approved, retrospective chart and imaging review of pLGGs treated with off-label targeted therapy at Dana-Farber/Boston Children's from 2010 to 2020. Response analysis was performed for BRAFV600E and BRAF fusion/duplication-driven pLGG subsets. Results Fifty-five patients were identified (dabrafenib n = 15, everolimus n = 26, trametinib n = 11, and vemurafenib n = 3). Median duration of targeted therapy was 9.48 months (0.12-58.44). The 1-year, 3-year, and 5-year EFS from targeted therapy initiation were 62.1%, 38.2%, and 31.8%, respectively. Mean volumetric change for BRAFV600E mutated pLGG on BRAF inhibitors was -54.11%; median time to best volumetric response was 8.28 months with 9 of 12 (75%) objective RAPNO responses. Median time to largest volume post-treatment was 2.86 months (+13.49%); mean volume by the last follow-up was -14.02%. Mean volumetric change for BRAF fusion/duplication pLGG on trametinib was +7.34%; median time to best volumetric response was 6.71 months with 3 of 7 (43%) objective RAPNO responses. Median time to largest volume post-treatment was 2.38 months (+71.86%); mean volume by the last follow-up was +39.41%. Conclusions Our integrated analysis suggests variability in response by pLGG molecular subgroup and targeted therapy, as well as the transience of some tumor growth following targeted therapy cessation.
Collapse
Affiliation(s)
- Jessica W Tsai
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Jungwhan John Choi
- Department of Radiology, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Hakim Ouaalam
- Department of Radiology, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Efrain Aguilar Murillo
- Department of Radiology, Division of Neuroradiology and Neurointervention, Boston, Massachusetts, USA
| | - Kee Kiat Yeo
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Jayne Vogelzang
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Cecilia Sousa
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Jared K Woods
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Keith L Ligon
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Pathology, Boston Children’s Hospital, Boston Massachusetts, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Simon K Warfield
- Department of Radiology, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Pratiti Bandopadhayay
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Tabitha M Cooney
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| |
Collapse
|
45
|
Single-agent Bevacizumab in the Treatment of Symptomatic Newly Diagnosed and Recurrent/Refractory Pediatric Cervicomedullary Brainstem Low-grade Gliomas: A Single Institutional Experience. J Pediatr Hematol Oncol 2023; 45:e21-e25. [PMID: 35426868 DOI: 10.1097/mph.0000000000002462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/08/2022] [Indexed: 02/03/2023]
Abstract
Bevacizumab-based therapies have been utilized as single or combination therapy of refractory/recurrent pediatric low-grade gliomas. Its efficacy for symptomatic cervicomedullary low-grade gliomas (cmLGGs) in the upfront and the recurrent setting is less known. We report our retrospective single institutional experience from 2015 to 2021 with single-agent bevacizumab for symptomatic cmLGG. Six consecutive patients (4 female, ages 2 to 12 y) with newly diagnosed (n=3) and recurrent/refractory (n=3) symptomatic nondisseminated cmLGG (5/6 biopsy-proven, 2 BRAFV600E, 2 BRAF-KIAA1549) were treated with single-agent bevacizumab. All demonstrated radiographic response most pronounced on post-gadolinium T1-weighted magnetic resonance imaging (2 complete, 4 partial) at a median of 8 weeks (range: 2 to 12 wk). Clinical response was seen in all patients with improvement in cranial nerve abnormalities (3 recurrent/refractory, 1 newly diagnosed), strength (2 recurrent/refractory, 2 newly diagnosed), pain (2 recurrent/refractory), and anorexia (1 newly diagnosed). Four patients (2 recurrent/refractory, 2 newly diagnosed) experienced disease progression on subsequent adjunct therapies, 2 of which (the 2 newly diagnosed patients) are currently being rechallenged. At a mean follow-up of 7 months, all patients are clinically stable without disease progression. Single-agent bevacizumab may be effective in the management of symptomatic newly diagnosed and recurrent/refractory cmLGG and warrants further evaluation in a clinical trial setting.
Collapse
|
46
|
Estevez-Ordonez D, Gary SE, Atchley TJ, Maleknia PD, George JA, Laskay NMB, Gross EG, Devulapalli RK, Johnston JM. Immunotherapy for Pediatric Brain and Spine Tumors: Current State and Future Directions. Pediatr Neurosurg 2022; 58:313-336. [PMID: 36549282 PMCID: PMC10233708 DOI: 10.1159/000528792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Brain tumors are the most common solid tumors and the leading cause of cancer-related deaths in children. Incidence in the USA has been on the rise for the last 2 decades. While therapeutic advances in diagnosis and treatment have improved survival and quality of life in many children, prognosis remains poor and current treatments have significant long-term sequelae. SUMMARY There is a substantial need for the development of new therapeutic approaches, and since the introduction of immunotherapy by immune checkpoint inhibitors, there has been an exponential increase in clinical trials to adopt these and other immunotherapy approaches in children with brain tumors. In this review, we summarize the current immunotherapy landscape for various pediatric brain tumor types including choroid plexus tumors, embryonal tumors (medulloblastoma, AT/RT, PNETs), ependymoma, germ cell tumors, gliomas, glioneuronal and neuronal tumors, and mesenchymal tumors. We discuss the latest clinical trials and noteworthy preclinical studies to treat these pediatric brain tumors using checkpoint inhibitors, cellular therapies (CAR-T, NK, T cell), oncolytic virotherapy, radioimmunotherapy, tumor vaccines, immunomodulators, and other targeted therapies. KEY MESSAGES The current landscape for immunotherapy in pediatric brain tumors is still emerging, but results in certain tumors have been promising. In the age of targeted therapy, genetic tumor profiling, and many ongoing clinical trials, immunotherapy will likely become an increasingly effective tool in the neuro-oncologist armamentarium.
Collapse
Affiliation(s)
- Dagoberto Estevez-Ordonez
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA,
- Division of Pediatric Neurosurgery, Children's of Alabama, Birmingham, Alabama, USA,
| | - Sam E Gary
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Travis J Atchley
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Neurosurgery, Children's of Alabama, Birmingham, Alabama, USA
| | - Pedram D Maleknia
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jordan A George
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nicholas M B Laskay
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Neurosurgery, Children's of Alabama, Birmingham, Alabama, USA
| | - Evan G Gross
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rishi K Devulapalli
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James M Johnston
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Pediatric Neurosurgery, Children's of Alabama, Birmingham, Alabama, USA
| |
Collapse
|
47
|
Leclair NK, Lambert W, Roche K, Gillan E, Gell JJ, Lau CC, Wrubel G, Knopf J, Amin S, Anderson M, Martin JE, Bookland MJ, Hersh DS. Early experience with targeted therapy as a first-line adjuvant treatment for pediatric low-grade glioma. Neurosurg Focus 2022; 53:E15. [PMID: 36455272 DOI: 10.3171/2022.9.focus22410] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/19/2022] [Indexed: 12/04/2022]
Abstract
OBJECTIVE Pediatric low-grade gliomas (pLGGs) frequently exhibit dysregulation of the mitogen-activated protein kinase (MAPK) pathway. Targeted therapies, including mutant BRAF inhibitors (dabrafenib) and MEK inhibitors (trametinib), have shown promise in patients in whom conventional chemotherapy has failed. However, few studies have investigated the use of targeted therapy as a first-line treatment for pLGG. Here, the authors reviewed their institutional experience with using a personalized medicine approach to patients with newly diagnosed pLGGs. METHODS All pediatric patients at the authors' institution who had been treated with dabrafenib or trametinib for pLGG without first receiving conventional chemotherapy or radiation were retrospectively reviewed. Demographic, clinical, and radiological data were collected. RESULTS Eight patients underwent targeted therapy as a first-line treatment for pLGG. Five patients had a BRAF alteration (1 with a BRAFV600E mutation, 4 with a KIAA1549:BRAF fusion), and 3 patients had an NF1 mutation. One of the 8 patients was initially treated with dabrafenib, and trametinib was added later. Seven patients were initially treated with trametinib; of these, 2 later transitioned to dual therapy, whereas 5 continued with trametinib monotherapy. Six patients (75%) demonstrated a partial response to therapy during their treatment course, whereas stable disease was identified in the remaining 2 patients (25%). One patient experienced mild disease progression after completing a course of trametinib monotherapy, but ultimately stabilized after a period of close observation. Another patient experienced tumor progression while on dabrafenib, but subsequently responded to dual therapy with dabrafenib and trametinib. The most common adverse reactions to targeted therapy were cutaneous toxicity (100%) and diarrhea (50%). CONCLUSIONS Targeted therapies have the potential to become a standard treatment option for pLGG due to their favorable toxicity profile and oral route of administration. This case series provides preliminary evidence that targeted therapies can induce an early disease response as a first-line adjuvant treatment; however, large-scale studies are required to assess long-term durability and safety.
Collapse
Affiliation(s)
| | | | - Kimberley Roche
- 2Division of Hematology and Oncology, Connecticut Children's, Hartford
| | - Eileen Gillan
- 2Division of Hematology and Oncology, Connecticut Children's, Hartford
| | - Joanna J Gell
- 2Division of Hematology and Oncology, Connecticut Children's, Hartford.,3The Jackson Laboratory for Genomic Medicine, Farmington.,4Department of Pediatrics, UConn School of Medicine; Farmington
| | - Ching C Lau
- 2Division of Hematology and Oncology, Connecticut Children's, Hartford.,3The Jackson Laboratory for Genomic Medicine, Farmington.,4Department of Pediatrics, UConn School of Medicine; Farmington
| | | | - Joshua Knopf
- 1School of Medicine, University of Connecticut, Farmington
| | - Shirali Amin
- 2Division of Hematology and Oncology, Connecticut Children's, Hartford
| | - Megan Anderson
- 6Division of Neurosurgery, Connecticut Children's, Hartford; and
| | - Jonathan E Martin
- 6Division of Neurosurgery, Connecticut Children's, Hartford; and.,7Department of Surgery, UConn School of Medicine, Farmington, Connecticut
| | - Markus J Bookland
- 6Division of Neurosurgery, Connecticut Children's, Hartford; and.,7Department of Surgery, UConn School of Medicine, Farmington, Connecticut
| | - David S Hersh
- 6Division of Neurosurgery, Connecticut Children's, Hartford; and.,7Department of Surgery, UConn School of Medicine, Farmington, Connecticut
| |
Collapse
|
48
|
Use of Trametinib in Children and Young Adults With Progressive Low-Grade Glioma and Glioneuronal Tumors. J Pediatr Hematol Oncol 2022; 45:e464-e470. [PMID: 36730221 DOI: 10.1097/mph.0000000000002598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 10/26/2022] [Indexed: 02/03/2023]
Abstract
Low-grade gliomas/glioneuronal tumors comprise one-third of all pediatric-type CNS tumors. These tumors are generally caused by activating mutations in the mitogen-activated protein kinase (MAPK) pathway. Targeted drugs, such as trametinib, have shown promise in other cancers and are being utilized in low-grade gliomas. A retrospective chart review was conducted to evaluate radiographic response, visual outcomes, tolerability, and durability of response in progressive circumscribed low-grade gliomas treated with trametinib. Eleven patients were treated with trametinib. The best radiographic response was 2/11 partial response, 3/11 minor response, 3/11 stable disease, and 3/13 progressive disease. In the patients with partial or minor response, the best response was seen after longer durations of therapy; 4 of 5 best responses occurred after at least 9 months of therapy with a median of 21 months. Patients with optic pathway tumors showed at least stable vision throughout treatment, with 3 having improved vision on treatment. Trametinib is effective and well-tolerated in patients with progressive low-grade glioma. Best responses were seen after a longer duration of therapy in those with a positive response. Patients with optic pathway lesions showed stable to improved vision while on treatment.
Collapse
|
49
|
Gao Y, Jing N, Teng X, Wang Y. Serine hydroxymethyltransferase 1 promotes low-grade glioma progression by activating mTORC1 signaling. Neurol Res 2022; 45:415-422. [PMID: 36417280 DOI: 10.1080/01616412.2022.2149516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVES This research aimed to explore the role and potential mechanism of serine hydroxymethyltransferase 1 (SHMT1) involvement in low-grade glioma (LGG). METHODS GEPIA were employed to analyze the expression and the correlation of LGG patient survival with the levels of SHMT1 in LGG based on the The Cancer Genome Atlas (TCGA) database. qRT-PCR and western blot were used to detect the expression of SHMT1 in LGG cells. Clone formation, EdU staining, MTT, Transwell and wound healing assays were conducted to analyze the proliferation, cell activity, migration and invasion of LGG cells. KEEG analysis was performed for enrichment pathways of SHMT1 in LGG. RESULTS SHMT1 was up-regulated in LGG tissues and cells, and SHMT1 level was negatively correlated with survival of patients with LGG. SHMT1 overexpression evidently promoted cell proliferation, migration and invasion, whereas SHMT1 silence obtained the opposite results. Next, KEEG analysis revealed that SHMT1 activated the mTORC1 pathway in LGG. SHMT1 overexpression significantly promoted the phosphorylation of downstream proteins (P70SK6 and S6) in LGG cells. Further, inhibition of the mTORC1 signaling pathway partially abolished the promotion of LGG progression by SHMT1 overexpression. CONCLUSION SHMT1 promoted proliferation, invasion and migration of LGG cells via activating mTORC1 signaling pathway. This provided a novel perspective for the treatment of LGG.
Collapse
Affiliation(s)
- Ye Gao
- Department of Neurosurgery, Zhangqiu District People’s Hospital, Jinan 250200, P.R. China
| | - Nianliang Jing
- Department of Neurosurgery, Zhangqiu District People’s Hospital, Jinan 250200, P.R. China
| | - Xukun Teng
- Department of Neurosurgery, Zhangqiu District People’s Hospital, Jinan 250200, P.R. China
| | - Yong Wang
- Department of Neurosurgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, P.R. China
| |
Collapse
|
50
|
Secondary Narcolepsy as Worsening Sign in a Pediatric Case of Optic Pathway Glioma. CHILDREN 2022; 9:children9101455. [PMID: 36291391 PMCID: PMC9600478 DOI: 10.3390/children9101455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 12/01/2022]
Abstract
Narcolepsy, a neurologic disorder that leads to excessive daytime sleepiness, may represent a rare consequence of neoplastic lesions involving the sellar/parasellar and hypothalamic regions, the anatomical areas responsible for wakefulness. Optic pathway gliomas represent the most common neoplasm of these regions and present an excellent overall survival, while long-term neurologic impairments, such as visual loss, endocrinopathies, or sleep disorders, are the principal causes of morbidity. In this case report, we describe a non-NF1 patient suffering from a very extensive optical pathway glioma, who several years after the diagnosis in a radiological condition of stable disease, presented with severe narcolepsy, a rare complication, that led to the death of the patient.
Collapse
|