1
|
Shariff N, Moreno AS, Bennett J, Ramaswamy V, Das A, Liu AP, Huang A, Tabori U, Hawkins C, Dirks P, Bouffet E, Keilty DM, Millar BA, Hodgson DC, Tsang DS. Re-irradiation for children with diffuse intrinsic pontine glioma and diffuse midline glioma. Radiother Oncol 2025; 207:110865. [PMID: 40139463 DOI: 10.1016/j.radonc.2025.110865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND AND PURPOSE Diffuse intrinsic pontine glioma (DIPG) and diffuse midline glioma (DMG) are incurable brain malignancies. In this study, we report one of the largest known single-institution cohorts of DIPG/DMG patients undergoing re-irradiation (RT2) to evaluate its effect on survival. MATERIALS AND METHODS Children aged less than 18 years treated for DIPG/DMG with initial fractionated photon radiotherapy (RT1) and had subsequent recurrence were retrospectively reviewed. Patients treated with or without RT2 were compared. The primary outcomes were overall survival (OS) from time of recurrence after RT1, and from start of RT2 (for the RT2 group). RESULTS A total of 118 children were included, 39 of whom received RT2. Children treated with RT2 had superior OS, with 6-month OS of 66 % vs 22 % in those who did not undergo RT2 (p < 0.0001). Median survivals were 6.9 months for the RT2 group vs 2.7 months for RT1 only. Median time from RT1 to RT2 was 7.7 months; patients with a greater than 1-year latent time between RT1 and RT2 had longer OS from start of RT2 (median 10.9 months vs 5.5 months, p = 0.023). 61 % of those treated with RT2 experienced improvement of neurologic symptoms post-RT2. Multivariate analysis identified younger age, adverse imaging findings on the 4-week post-RT1 reassessment MRI (including pseudoprogression), and the absence of RT2 as poor prognostic factors for OS. CONCLUSION Re-irradiation was associated with improved survival and neurological recovery in children with recurrent DIPG and DMG. There is a need to identify novel biomarkers to better select patients who respond best to RT2.
Collapse
Affiliation(s)
- Nisha Shariff
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Canada; Department of Clinical Oncology, University Malaya Medical Centre, Malaysia
| | - Alejandro S Moreno
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Canada
| | - Julie Bennett
- Division of Haematology/Oncology, Hospital for Sick Children, Canada
| | - Vijay Ramaswamy
- Division of Haematology/Oncology, Hospital for Sick Children, Canada
| | - Anirban Das
- Division of Haematology/Oncology, Hospital for Sick Children, Canada
| | - Anthony P Liu
- Division of Haematology/Oncology, Hospital for Sick Children, Canada
| | - Annie Huang
- Division of Haematology/Oncology, Hospital for Sick Children, Canada
| | - Uri Tabori
- Division of Haematology/Oncology, Hospital for Sick Children, Canada
| | - Cynthia Hawkins
- Department of Paediatric Laboratory Medicine, Hospital for Sick Children, Canada
| | - Peter Dirks
- Division of Neurosurgery, Hospital for Sick Children, Canada
| | - Eric Bouffet
- Division of Haematology/Oncology, Hospital for Sick Children, Canada
| | - Dana M Keilty
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Canada
| | - Barbara-Ann Millar
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Canada
| | - David C Hodgson
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Canada
| | - Derek S Tsang
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Canada.
| |
Collapse
|
2
|
Chen XS, Zhang L, Ajithkumar T, Butala AA, Kim MM, Mayo C, Rosen BS, Shen CJ, Murray L. Practice Patterns of Reirradiation for Brain and Spinal Tumors-An International Survey From the Reirradiation Collaborative Group (ReCOG). Pract Radiat Oncol 2025:S1879-8500(25)00100-6. [PMID: 40280482 DOI: 10.1016/j.prro.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/03/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025]
Abstract
PURPOSE An international workshop was convened by the Reirradiation Collaborative Group. We conducted a survey among the invited attendants to assess practice patterns of reirradiation for central nervous system tumors. METHODS AND MATERIALS A web-based survey regarding central nervous system reirradiation was distributed to an international group of radiation oncologists and medical physicists via email. RESULTS Sixty-six respondents from 20 countries completed at least one section of the survey. The most important clinical considerations were treatment goal, degree of overlap, and cumulative dose. Among technical challenges, uncertainties in tolerance of organs at risk (OARs), tissue recovery factors (TRFs) and dose accumulation ranked the highest. Most respondents (68%) used a planning OAR volume with 0 to 3 mm margin. Highly conformal radiation techniques were preferred, including stereotactic body radiation therapy for spine (85%), intensity modulated radiation therapy for adult primary brain tumors (93%), and intensity modulated radiation therapy (100%) and proton therapy (83%) for pediatric cases. Most performed dose accumulation (65%) and evaluated cumulative biological (ie, equieffective) dose (88%). Sixty-one percent preferred rigid registration, whereas 35% used deformable registration, most commonly in pediatric cases (67%). The most frequently used α/β value for OARs was 2 Gy (76%). There was no clear consensus on OAR tolerance for any disease site. Different dose metrics were used for evaluation, including Dmax (48%) and D0.1cc (48%). Most (79%) considered time intervals between radiation courses. For adult primary brain tumors and brain metastasis, 50% and 46% recommended against reirradiation within a short interval (3-6 months). Most respondents (52%) used time dependent TRFs. CONCLUSIONS Among respondents, there are substantial variations in approaches to reirradiation (eg, addition of systemic therapy) and uncertainties in technical implementation (eg, OAR tolerance, TRF, and dose accumulation). Future collaborative registry-based and prospective studies should help address these uncertainties.
Collapse
Affiliation(s)
- Xuguang Scott Chen
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| | - Lei Zhang
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Thankamma Ajithkumar
- Department of Oncology, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Anish A Butala
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Charles Mayo
- Department of Radiation Oncology, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Benjamin S Rosen
- Department of Radiation Oncology, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Colette J Shen
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Louise Murray
- Department of Clinical Oncology, Leeds Cancer Centre, Leeds, United Kingdom; Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
3
|
McVeigh L, Patel T, Miclea M, Schwark K, Ajaero D, Momen F, Clausen M, Adam T, Aittaleb R, Wadden J, Lau B, Franson AT, Koschmann C, Marupudi NI. Updates in Diagnostic Techniques and Experimental Therapies for Diffuse Intrinsic Pontine Glioma. Cancers (Basel) 2025; 17:931. [PMID: 40149267 PMCID: PMC11940218 DOI: 10.3390/cancers17060931] [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: 01/25/2025] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a rare but extremely malignant central nervous system tumor primarily affecting children that is almost universally fatal with a devastating prognosis of 8-to-12-month median survival time following diagnosis. Traditionally, DIPG has been diagnosed via MR imaging alone and treated with palliative radiation therapy. While performing surgical biopsies for these patients has been controversial, in recent years, advancements have been made in the safety and efficacy of surgical biopsy techniques, utilizing stereotactic, robotics, and intraoperative cranial nerve monitoring as well as the development of liquid biopsies that identify tumor markers in either cerebrospinal fluid or serum. With more molecular data being collected from these tumors due to more frequent biopsies being performed, multiple treatment modalities including chemotherapy, radiation therapy, immunotherapy, and epigenetic modifying agents continue to be developed. Numerous recent clinical trials have been completed or are currently ongoing that have shown promise in extending survival for patients with DIPG. Focused ultrasound (FUS) has also emerged as an additional promising adjunct invention used to increase the effectiveness of therapeutic agents. In this review, we discuss the current evidence to date for these advancements in the diagnosis and treatment of DIPG.
Collapse
Affiliation(s)
- Luke McVeigh
- Department of Neurosurgery, Michigan Medicine, Ann Arbor, MI 48109, USA;
| | - Tirth Patel
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Madeline Miclea
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Kallen Schwark
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Diala Ajaero
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Fareen Momen
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Madison Clausen
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Tiffany Adam
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Rayan Aittaleb
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Jack Wadden
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Benison Lau
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Andrea T. Franson
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Carl Koschmann
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI 48109, USA; (T.P.); (M.M.); (K.S.); (D.A.); (F.M.); (M.C.); (T.A.); (R.A.); (J.W.); (B.L.); (C.K.)
| | - Neena I. Marupudi
- Department of Neurosurgery, Michigan Medicine, Ann Arbor, MI 48109, USA;
| |
Collapse
|
4
|
Pearson AD, Mueller S, Filbin MG, Grill J, Hawkins C, Jones C, Donoghue M, Drezner N, Weiner S, Russo M, Dun MD, Allen JE, Alonso M, Benaim E, Buenger V, de Rojas T, Desserich K, Fox E, Friend J, Glade Bender J, Hargrave D, Jensen M, Kholmanskikh O, Kieran MW, Knoderer H, Koschmann C, Lesa G, Ligas F, Lipsman N, Ludwinski D, Marshall L, McDonough J, McNicholl AG, Mirsky D, Monje M, Nysom K, Pappo A, Rosenfield A, Scobie N, Slaughter J, Smith M, Souweidane M, Straathof K, Ward L, Weigel B, Zamoryakhin D, Karres D, Vassal G. Paediatric strategy forum for medicinal product development in diffuse midline gliomas in children and adolescents ACCELERATE in collaboration with the European Medicines Agency with participation of the Food and Drug Administration. Eur J Cancer 2025; 217:115230. [PMID: 39854822 DOI: 10.1016/j.ejca.2025.115230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025]
Abstract
Fewer than 10 % of children with diffuse midline glioma (DMG) survive 2 years from diagnosis. Radiation therapy remains the cornerstone of treatment and there are no medicinal products with regulatory approval. Although the biology of DMG is better characterized, this has not yet translated into effective treatments. H3K27-alterations initiate the disease but additional drivers are required for malignant growth. Hence, there is an urgent unmet need to develop new multi-modality therapeutic strategies, including alternative methods of drug delivery. ONC201 (DRD2 antagonist and mitochondrial ClpP agonist) is the most widely evaluated investigational drug. Encouraging early data is emerging for CAR T-cells and oncolytic viruses. GD2, B7-H3 and PI3K signalling are ubiquitous targets across all subtypes and therapeutics directed to these targets would potentially benefit the largest number of children. PI3K, ACVR1, MAPK and PDGFRA pathways should be targeted in rational biological combinations. Drug discovery is a very high priority. New specific and potent epigenetic modifiers (PROTACS e.g. SMARCA4 degraders), with blood-brain penetrance are needed. Cancer neuroscience therapeutics are in early development. Overall survival is the preferred regulatory endpoint. However, the evaluation of this can be influenced by the use of re-irradiation at the time of progression. An efficient clinical trial design fit for regulatory purposes for the evaluation of new therapeutics would aid industry and facilitate more efficient therapy development. Challenges in conducting clinical trials such as the need for comparator data and defining endpoints, could be addressed through an international, first-in-child, randomised, complex innovative design trial. To achieve progress: i) drug discovery; ii) new multi-modality, efficient, collaborative, pre-clinical approaches, possibly including artificial intelligence and, iii) efficient clinical trial designs fit for regulatory purposes are required.
Collapse
Affiliation(s)
| | - Sabine Mueller
- Departments of Neurological Surgery, Pediatrics and, Neurology University of California, San Francisco, California, USA. Department of Oncology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Mariella G Filbin
- Broad Institute of Harvard and MIT, Cambridge, MA, USA; Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | | | - Cynthia Hawkins
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Chris Jones
- The Institute of Cancer Research, Sutton, Surrey, UK
| | | | - Nicole Drezner
- US Food and Drug Administration, Silver Springs, MD, USA
| | - Susan Weiner
- ACCELERATE, Europe; Children's Cancer Cause, Washington, DC, USA; Memorial Sloan Kettering Cancer Centre, New York, USA
| | | | - Matthew D Dun
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia; Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, Mark Hughes Foundation for Brain Cancer Research, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | | | - Marta Alonso
- Program of Solid Tumors, Center for the Applied Medical Research, Pamplona, Spain; Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Center for the Applied Medical Research, Pamplona, Spain
| | | | - Vickie Buenger
- Coalition Against Childhood Cancer (CAC2), Philadelphia, USA
| | | | | | | | | | | | - Darren Hargrave
- University College London Great Ormond Street Institute of Child Health, London, UK
| | | | | | | | | | | | - Giovanni Lesa
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Division, European Medicines Agency, The Netherlands
| | - Franca Ligas
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Division, European Medicines Agency, The Netherlands
| | - Nir Lipsman
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | - Lynley Marshall
- The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK
| | | | | | - David Mirsky
- University of Colorado, School of Medicine, CO, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA, Howard Hughes Medical Institute, Stanford, CA, USA
| | | | | | | | | | | | | | | | - Karin Straathof
- University College London Cancer Institute, Great Ormond Street Biomedical Research Centre, London, UK
| | - Lisa Ward
- DIPG-DMG Research Funding Alliance DDRFA /Tough2gether, Manhattan, KS, USA
| | | | | | - Dominik Karres
- Paediatric Medicines Office, Scientific Evidence Generation Department, Human Division, European Medicines Agency, The Netherlands
| | - Gilles Vassal
- ACCELERATE, Europe; Gustave Roussy Cancer Centre, Paris, France
| |
Collapse
|
5
|
Østergaard DE, Embring A, Sehested A, Magelssen H, Vogelius IR, Kjærsgaard M, Nysom K, Mathiasen R, Lukacova S, Maraldo MV. REMIT: Reirradiation of Diffuse Midline Glioma Patients -A Nordic Society of Paediatric Haematology and Oncology Feasibility Study. Clin Oncol (R Coll Radiol) 2025; 37:103682. [PMID: 39626445 DOI: 10.1016/j.clon.2024.103682] [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: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 01/20/2025]
Abstract
Diffuse midline glioma (DMG) continues to be an aggressive brain stem cancer among children and young adults. It has a dismal prognosis, with less than 10% of patients alive two years after diagnosis. Radiotherapy has been demonstrated to be effective, albeit transient. Hence, radiotherapy is considered a cornerstone in the treatment. Reirradiation has, in retrospective studies, shown promising overall survival and palliative effect, but no pan-European consensus for reirradiation exists. The REMIT (Reirradiation of diffuse Midline glioma paTients) protocol evaluates safety and the palliative efficacy of reirradiation of patients with DMG (clinicaltrials.gov NCT06093165). Patients included in the protocol will be followed with 1) performance status (Karnofsky or Lansky), 2) toxicity monitored with Common Terminology Criteria for Adverse Events (CTCAE), 3) motor and functioning skill with PEDI-CAT (The Pediatric Evaluation of Disability Inventory) and 4) quantification of corticosteroid use. Furthermore, the impact on quality of life and well-being will be assessed qualitatively with interviews as well as with the Pediatric Quality of Life Inventory (PedsQl) Cancer Module questionnaire. The protocol also includes dose accumulation and contouring studies to assess standardization as well as a prescreening log to address selection bias of patients. The safety and palliative efficacy of reirradiation in DMG will be prospectively evaluated, including qualitative patient reported outcomes, through the REMIT protocol. REMIT is planned to open for inclusion in 2024.
Collapse
Affiliation(s)
- D E Østergaard
- Section of Radiotherapy, Department of Oncology, Centre for Cancer and Organ Diseases, Copenhagen University Hospital, Copenhagen, Denmark; Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark.
| | - A Embring
- Department of Oncology, Karolinska University Hospital, Sweden; Department of Oncology-Pathology, Karolinska Institute, Sweden
| | - A Sehested
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital, Copenhagen, Denmark
| | - H Magelssen
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - I R Vogelius
- Section of Radiotherapy, Department of Oncology, Centre for Cancer and Organ Diseases, Copenhagen University Hospital, Copenhagen, Denmark; Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| | - M Kjærsgaard
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital, Copenhagen, Denmark
| | - K Nysom
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital, Copenhagen, Denmark
| | - R Mathiasen
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital, Copenhagen, Denmark
| | - S Lukacova
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - M V Maraldo
- Section of Radiotherapy, Department of Oncology, Centre for Cancer and Organ Diseases, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Wawrzuta D, Chojnacka M, Drogosiewicz M, Pędziwiatr K, Dembowska-Bagińska B. Reirradiation for diffuse intrinsic pontine glioma: prognostic radiomic factors at progression. Strahlenther Onkol 2024; 200:797-804. [PMID: 38748214 PMCID: PMC11343881 DOI: 10.1007/s00066-024-02241-7] [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: 12/19/2023] [Accepted: 04/23/2024] [Indexed: 08/24/2024]
Abstract
PURPOSE Diffuse intrinsic pontine glioma (DIPG) is a lethal pediatric brain tumor. Radiation therapy (RT) is the standard treatment, with reirradiation considered in case of progression. However, the prognostic factors for reirradiation are not well understood. This study aims to investigate the outcomes of DIPG patients undergoing reirradiation and identify clinical and radiomic prognostic factors. METHODS We conducted a retrospective analysis of patients with DIPG who underwent reirradiation at our institution between January 2016 and December 2023. Using PyRadiomics, we extracted radiomic features of tumors at the time of progression from FLAIR MRI images and collected clinical data. We used the least absolute shrinkage and selection operator (lasso) for Cox's proportional hazard model with leave-one-out cross-validation to select optimal prognostic factors for survival after reirradiation. RESULTS The study included 18 patients who underwent reirradiation at first progression, receiving a total dose of 20 Gy or 24 Gy in 2‑Gy fractions. Reirradiation was well tolerated, with no severe toxicity. Most patients (78%) showed neurological improvement after treatment. Median survival after progression was 29.2 weeks. The Cox model demonstrated a concordance of 0.81 (95% CI: 0.75-0.88), revealing that tumor sphericity and structural gray-level heterogeneity in FLAIR MRI images were associated with longer survival of reirradiated patients. CONCLUSION Reirradiation is a safe and effective approach for patients with DIPG. MRI-based radiomic models could be helpful in predicting survival after reirradiation.
Collapse
Affiliation(s)
- Dominik Wawrzuta
- Department of Radiation Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Wawelska 15B, 02-034, Warsaw, Poland.
| | - Marzanna Chojnacka
- Department of Radiation Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Wawelska 15B, 02-034, Warsaw, Poland
| | - Monika Drogosiewicz
- Department of Oncology, Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730, Warsaw, Poland
| | - Katarzyna Pędziwiatr
- Department of Radiation Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Wawelska 15B, 02-034, Warsaw, Poland
| | - Bożenna Dembowska-Bagińska
- Department of Oncology, Children's Memorial Health Institute, Al. Dzieci Polskich 20, 04-730, Warsaw, Poland
| |
Collapse
|
7
|
Akdemir EY, Odia Y, Hall MD, Mehta MP, Kotecha R. An Update on H3K27M-altered Diffuse Midline Glioma: Diagnostic and Therapeutic Challenges in Clinical Practice. Pract Radiat Oncol 2024; 14:443-451. [PMID: 38704025 DOI: 10.1016/j.prro.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
H3K27-altered diffuse midline glioma (DMG H3K27-altered) is a relatively newly-designated WHO entity which primarily affects the midline structures of the central nervous system (CNS), including the brainstem (predominantly pontine region), thalamus, midbrain, or spinal cord, and primarily affects children and young adults. Despite the proximity of these tumors to eloquent areas in the CNS, novel stereotactic approaches have facilitated the ability to obtain tissue diagnoses without significant morbidity, providing molecular diagnostic information in more than half of patients. Conventionally fractionated radiation therapy to a total dose of 54-60 Gy in 27-30 fractions and 24 Gy in 12 fractions play a crucial role in the definitive treatment of these tumors in the primary and salvage settings, respectively. Hypofractionated regimens may allow for accelerated treatment courses in selected patients without jeopardizing disease control or survival. The decision to add concurrent or adjuvant systemic therapy mainly relies on the physicians' experience without solid evidence in the literature in favor of any particular regimen. Recently, novel agents, such as ONC201 have demonstrated promising oncologic outcomes in progressive/recurrent tumors and are currently under investigation in ongoing randomized trials. Given the scarcity of data and well-established guidelines due to the rare nature of the disease, we provide a contemporary overview on the molecular underpinnings of this disease entity, describe the role of radiotherapy and systemic therapy, and present practice management principles based on the published literature.
Collapse
Affiliation(s)
- Eyub Yasar Akdemir
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida
| | - Yazmin Odia
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida; Department of Neuro-Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida
| | - Matthew D Hall
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida; Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida; Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Rupesh Kotecha
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida; Herbert Wertheim College of Medicine, Florida International University, Miami, Florida.
| |
Collapse
|
8
|
Pasqualetti F, Lombardi G, Gadducci G, Giannini N, Montemurro N, Feletti A, Zeppieri M, Somma T, Caffo M, Bertolotti C, Ius T. Brain Stem Glioma Recurrence: Exploring the Therapeutic Frontiers. J Pers Med 2024; 14:899. [PMID: 39338153 PMCID: PMC11433503 DOI: 10.3390/jpm14090899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
Gliomas of the brainstem represent a small percentage of central nervous system gliomas in adults. Due to the proximity of the tumor to critical structures, radical surgery is highly challenging and limited to selected cases. In addition, postoperative treatments, which become exclusive to non-operable patients, do not guarantee satisfactory disease control, making the progression of the disease inevitable. Currently, there is a lack of therapeutic options to control tumor growth after the diagnosis of recurrence. The rarity of these tumors, their distinct behavioral characteristics, and the limited availability of tumor tissue necessary for the development of prognostic and predictive biomarkers contribute to the absence of a standardized approach for treating recurrent brainstem gliomas. A salvage radiotherapy (RT) retreatment could represent a promising approach for recurrent brainstem gliomas. However, to date, it has been mainly evaluated in pediatric cases, with few experiences available to assess the most appropriate RT dose, safety, and clinical responses in adult patients. This comprehensive review aims to identify instances of adult patients with recurrent brainstem gliomas subjected to a secondary course of RT, with a specific focus on the analysis of treatment-related toxicity and outcomes. Through this investigation, we endeavor to contribute valuable insights into the viability and efficacy of salvage RT retreatment in managing recurrent brainstem gliomas in the adult population.
Collapse
Affiliation(s)
- Francesco Pasqualetti
- Division of Radiation Oncology, Azienda Ospedaliero Universitaria Pisana, 56100 Pisa, Italy; (F.P.)
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
| | - Giovanni Gadducci
- Division of Radiation Oncology, Azienda Ospedaliero Universitaria Pisana, 56100 Pisa, Italy; (F.P.)
| | - Noemi Giannini
- Division of Radiation Oncology, Azienda Ospedaliero Universitaria Pisana, 56100 Pisa, Italy; (F.P.)
| | - Nicola Montemurro
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana, 56100 Pisa, Italy
| | - Alberto Feletti
- Department of Neurosciences, Biomedicine, and Movement Sciences, Institute of Neurosurgery, University of Verona, 37126 Verona, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, Piazzale S. Maria della Misericordia 15, 33100 Udine, Italy
| | - Teresa Somma
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, 80134 Naples, Italy
| | - Maria Caffo
- Unit of Neurosurgery, Department of Biomorphology and Dental Science, and Morphofunctional Imaging, Università degli Studi di Messina, 98125 Messina, Italy
| | - Chiara Bertolotti
- Department of Neuroradiology, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy
| | - Tamara Ius
- Neurosurgery Unit, Head-Neck and NeuroScience Department, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy
| |
Collapse
|
9
|
Yu X, Lai M, Li J, Wang L, Ye K, Zhang D, Hu Q, Li S, Hu X, Wang Q, Ma M, Xiao Z, Zhou J, Shi C, Luo L, Cai L. The relationship between imaging features, therapeutic response, and overall survival in pediatric diffuse intrinsic pontine glioma. Neurosurg Rev 2024; 47:212. [PMID: 38727935 PMCID: PMC11087318 DOI: 10.1007/s10143-024-02435-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/24/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024]
Abstract
We aimed to evaluate the relationship between imaging features, therapeutic responses (comparative cross-product and volumetric measurements), and overall survival (OS) in pediatric diffuse intrinsic pontine glioma (DIPG). A total of 134 patients (≤ 18 years) diagnosed with DIPG were included. Univariate and multivariate analyses were performed to evaluate correlations of clinical and imaging features and therapeutic responses with OS. The correlation between cross-product (CP) and volume thresholds in partial response (PR) was evaluated by linear regression. The log-rank test was used to compare OS patients with discordant therapeutic response classifications and those with concordant classifications. In univariate analysis, characteristics related to worse OS included lower Karnofsky, larger extrapontine extension, ring-enhancement, necrosis, non-PR, and increased ring enhancement post-radiotherapy. In the multivariate analysis, Karnofsky, necrosis, extrapontine extension, and therapeutic response can predict OS. A 25% CP reduction (PR) correlated with a 32% volume reduction (R2 = 0.888). Eight patients had discordant therapeutic response classifications according to CP (25%) and volume (32%). This eight patients' median survival time was 13.0 months, significantly higher than that in the non-PR group (8.9 months), in which responses were consistently classified as non-PR based on CP (25%) and volume (32%). We identified correlations between imaging features, therapeutic responses, and OS; this information is crucial for future clinical trials. Tumor volume may represent the DIPG growth pattern more accurately than CP measurement and can be used to evaluate therapeutic response.
Collapse
Affiliation(s)
- Xiaojun Yu
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Mingyao Lai
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Juan Li
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Lichao Wang
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Kunlin Ye
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Dong Zhang
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Qingjun Hu
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Shaoqun Li
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Xinpeng Hu
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Qiong Wang
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Mengjie Ma
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Zeyu Xiao
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China
| | - Jiangfen Zhou
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China
| | - Changzheng Shi
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China.
| | - Liangping Luo
- Department of Medical Imaging Center, Jinan University First Affiliated Hospital, No. 613, Huangpu Road West, Tianhe District, Guangzhou, 510630, Guangdong Province, China.
- Department of Medical Imaging Center, The Fifth Affiliated Hospital of Jinan University, Yingke Avenue, Heyuan City, 517000, China.
| | - Linbo Cai
- Department of Oncology, Guangdong sanjiu Brain Hospital, No. 578, Shatai South Road, Baiyun District, Guangzhou, 510510, Guangdong Province, China.
| |
Collapse
|
10
|
van den Bent M, Saratsis AM, Geurts M, Franceschi E. H3 K27M-altered glioma and diffuse intrinsic pontine glioma: Semi-systematic review of treatment landscape and future directions. Neuro Oncol 2024; 26:S110-S124. [PMID: 38102230 PMCID: PMC11066941 DOI: 10.1093/neuonc/noad220] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Indexed: 12/17/2023] Open
Abstract
H3 K27M-mutant diffuse glioma is a recently identified brain tumor associated with poor prognosis. As of 2016, it is classified by the World Health Organization as a distinct form of grade IV glioma. Despite recognition as an important prognostic and diagnostic feature in diffuse glioma, radiation remains the sole standard of care and no effective systemic therapies are available for H3K27M mutant tumors. This review will detail treatment interventions applied to diffuse midline glioma and diffuse intrinsic pontine glioma (DIPG) prior to the identification of the H3 K27M mutation, the current standard-of-care for H3 K27M-mutant diffuse glioma treatment, and ongoing clinical trials listed on www.clinicaltrials.gov evaluating novel therapeutics in this population. Current clinical trials were identified using clinicaltrials.gov, and studies qualifying for this analysis were active or ongoing interventional trials that evaluated a therapy in at least 1 treatment arm or cohort comprised exclusively of patients with DIPG and H3 K27M-mutant glioma. Forty-one studies met these criteria, including trials evaluating H3 K27M vaccination, chimeric antigen receptor T-cell therapy, and small molecule inhibitors. Ongoing evaluation of novel therapeutics is necessary to identify safe and effective interventions in this underserved patient population.
Collapse
Affiliation(s)
- Martin van den Bent
- Brain Tumor Center at Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Amanda M Saratsis
- Department of Neurosurgery, Advocate Children’s Hospital, Park Ridge, Illinois, USA
| | - Marjolein Geurts
- Brain Tumor Center at Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Enrico Franceschi
- Department of Nervous System Medical Oncology, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
11
|
Mankuzhy NP, Tringale KR, Dunkel IJ, Farouk Sait S, Souweidane MM, Khakoo Y, Karajannis MA, Wolden S. Hypofractionated re-irradiation for diffuse intrinsic pontine glioma. Pediatr Blood Cancer 2024; 71:e30929. [PMID: 38430472 PMCID: PMC11791744 DOI: 10.1002/pbc.30929] [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: 12/16/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Re-irradiation (reRT) increases survival in locally recurrent diffuse intrinsic pontine glioma (DIPG). There is no standard dose and fractionation for reRT, but conventional fractionation (CF) is typically used. We report our institutional experience of reRT for DIPG, which includes hypofractionation (HF). METHODS We reviewed pediatric patients treated with brainstem reRT for DIPG at our institution from 2012 to 2022. Patients were grouped by HF or CF. Outcomes included steroid use, and overall survival (OS) was measured from both diagnosis and start of reRT. RESULTS Of 22 patients who received reRT for DIPG, two did not complete their course due to clinical decline. Of the 20 who completed reRT, the dose was 20-30 Gy in 2-Gy fractions (n = 6) and 30-36 Gy in 3-Gy fractions (n = 14). Median age was 5 years (range: 3-14), median interval since initial RT was 8 months (range: 3-20), and 12 received concurrent bevacizumab. Median OS from diagnosis was 18 months [95% confidence interval: 17-24]. Median OS from start of reRT for HF versus CF was 8.2 and 7.5 months, respectively (p = .20). Thirteen (93%) in the HF group and three (75%) in the CF group tapered pre-treatment steroid dose down or off within 2 months after reRT due to clinical improvement. There was no significant difference in steroid taper between HF and CF (p = .4). No patients developed radionecrosis. CONCLUSION reRT with HF achieved survival duration comparable to published outcomes and effectively palliated symptoms. Future investigation of this regimen in the context of new systemic therapies and upfront HF is warranted.
Collapse
Affiliation(s)
- Nikhil P Mankuzhy
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kathryn R Tringale
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ira J Dunkel
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sameer Farouk Sait
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mark M Souweidane
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Yasmin Khakoo
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Matthias A Karajannis
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Suzanne Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
12
|
Ius T, Montemurro N, Lombardi G, Berardinelli J, Romano A, Barresi V, Cerretti G, Guarnera A, Tel A, Cavallo LM, Pasqualetti F, Feletti A. Decoding the puzzle: A multidisciplinary systematic review of adult brainstem glioma. Crit Rev Oncol Hematol 2024; 196:104261. [PMID: 38395241 DOI: 10.1016/j.critrevonc.2024.104261] [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: 08/06/2023] [Revised: 12/26/2023] [Accepted: 01/04/2024] [Indexed: 02/25/2024] Open
Abstract
Adult brainstem gliomas (BSGs) are a group of rare central nervous system tumors with varying prognoses and controversial standard treatment strategies. To provide an overview of current trends, a systematic review using the PRISMA guidelines, Class of evidence (CE) and strength of recommendation (SR), was conducted. The review identified 27 studies. Surgery was found to have a positive impact on survival, particularly for focal lesions with CE II SR C. Stereotactic image-guided biopsy was recommended when resective surgery was not feasible with CE II and SR B. The role of systemic treatments remains unclear. Eight studies provided molecular biology data. This review gathers crucial literature on diagnosis and management of adult BSGs. It provides evidence-based guidance with updated recommendations for diagnosing and treating, taking into account recent molecular and genetic advancements. The importance of brain biopsy is emphasized to optimize treatment using emerging genetic-molecular findings and explore potential targeted therapies.
Collapse
Affiliation(s)
- Tamara Ius
- Neurosurgery Unit, Head-Neck and NeuroScience Department University Hospital of Udine, Italy.
| | - Nicola Montemurro
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Jacopo Berardinelli
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Andrea Romano
- Department of Neuroradiology, NESMOS S. Andrea Hospital, University Sapienza, Rome, Italy
| | - Valeria Barresi
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Giulia Cerretti
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Alessia Guarnera
- Department of Neuroradiology, NESMOS S. Andrea Hospital, University Sapienza, Rome, Italy
| | - Alessandro Tel
- Clinic of Maxillofacial Surgery, Head-Neck and NeuroScience Department University Hospital of Udine, Italy
| | - Luigi Maria Cavallo
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Francesco Pasqualetti
- Division of Radiation Oncology, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Alberto Feletti
- Department of Neurosciences, Biomedicine, and Movement Sciences, Institute of Neurosurgery, University of Verona, Italy
| |
Collapse
|
13
|
Di Nunno V, Lombardi G, Simonelli M, Minniti G, Mastronuzzi A, Di Ruscio V, Corrà M, Padovan M, Maccari M, Caccese M, Simonetti G, Berlendis A, Farinotti M, Pollo B, Antonelli M, Di Muzio A, Dipasquale A, Asioli S, De Biase D, Tosoni A, Silvani A, Franceschi E. The role of adjuvant chemotherapy in patients with H3K27 altered diffuse midline gliomas: a multicentric retrospective study. J Neurooncol 2024; 167:145-154. [PMID: 38457090 DOI: 10.1007/s11060-024-04589-3] [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: 12/30/2023] [Accepted: 01/25/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE Adult Diffuse midline glioma (DMG) is a very rare disease. DMGs are currently treated with radiotherapy and chemotherapy even if only a few retrospective studies assessed the impact on overall survival (OS) of these approaches. METHODS We carried out an Italian multicentric retrospective study of adult patients with H3K27-altered DMG to assess the effective role of systemic therapy in the treatment landscape of this rare tumor type. RESULTS We evaluated 49 patients from 6 Institutions. The median age was 37.3 years (range 20.1-68.3). Most patients received biopsy as primary approach (n = 30, 61.2%) and radiation therapy after surgery (n = 39, 79.6%). 25 (51.0%) of patients received concurrent chemotherapy and 26 (53.1%) patients received adjuvant temozolomide. In univariate analysis, concurrent chemotherapy did not result in OS improvement while adjuvant temozolomide was associated with longer OS (21.2 vs. 9.0 months, HR 0.14, 0.05-0.41, p < 0.001). Multivariate analysis confirmed the role of adjuvant chemotherapy (HR 0.1, 95%CI: 0.03-0.34, p = 0.003). In patients who progressed after radiation and/or chemotherapy the administration of a second-line systemic treatment had a significantly favorable impact on survival (8.0 vs. 3.2 months, HR 0.2, 95%CI 0.1-0.65, p = 0.004). CONCLUSION In our series, adjuvant treatment after radiotherapy can be useful in improving OS of patients with H3K27-altered DMG. When feasible another systemic treatment after treatment progression could be proposed.
Collapse
Affiliation(s)
- Vincenzo Di Nunno
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, 40139, Italy
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Matteo Simonelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milano, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milano, Italy
| | - Giuseppe Minniti
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University, Policlinico Umberto I, Rome, Italy
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | - Angela Mastronuzzi
- Onco-Hematology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Valentina Di Ruscio
- Onco-Hematology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Martina Corrà
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Marta Padovan
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Marta Maccari
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Mario Caccese
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Giorgia Simonetti
- Neuro-Oncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Arianna Berlendis
- Unit of Immunotherapy of Brain Tumors, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, 20133, Italy
| | - Mariangela Farinotti
- Unit of Neuroepidemiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy
| | - Bianca Pollo
- Unit of Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy
| | - Manila Antonelli
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University, Policlinico Umberto I, Rome, Italy
| | | | | | - Sofia Asioli
- IRCCS-Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences (DIBINEM)-Surgical Pathology Section, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Dario De Biase
- Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, 40139, Italy
| | - Antonio Silvani
- Neuro-Oncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, 40139, Italy.
| |
Collapse
|
14
|
Cacciotti C, Wright KD. Advances in Treatment of Diffuse Midline Gliomas. Curr Neurol Neurosci Rep 2023; 23:849-856. [PMID: 37921944 DOI: 10.1007/s11910-023-01317-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2023] [Indexed: 11/05/2023]
Abstract
PURPOSE OF REVIEW Diffuse midline gliomas (DMGs) generally carry a poor prognosis, occur during childhood, and involve midline structures of the central nervous system, including the thalamus, pons, and spinal cord. RECENT FINDINGS To date, irradiation has been shown to be the only beneficial treatment for DMG. Various genetic modifications have been shown to play a role in the pathogenesis of this disease. Current treatment strategies span targeting epigenetic dysregulation, cell cycle, specific genetic alterations, and the immune microenvironment. Herein, we review the complex features of this disease as it relates to current and past therapeutic approaches.
Collapse
Affiliation(s)
- Chantel Cacciotti
- Children's Hospital London Health Sciences/Western University, London, ON, Canada.
| | - Karen D Wright
- Dana Farber/Boston Children's Cancer and Blood Disorder Center, Boston, MA, USA
| |
Collapse
|
15
|
Østergaard DE, Wahlstedt I, Jørgensen M, Kjærsgaard M, Mathiasen R, Nysom K, Sehested A, Vogelius IR, Maraldo MV. Dose-accumulation analysis of target and organs at risk with clinical outcome after re-irradiation of diffuse midline glioma. Acta Oncol 2023; 62:1526-1530. [PMID: 37733582 DOI: 10.1080/0284186x.2023.2258271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/08/2023] [Indexed: 09/23/2023]
Affiliation(s)
- Daniella Elisabet Østergaard
- Section of Radiotherapy, Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Isak Wahlstedt
- Section of Radiotherapy, Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Morten Jørgensen
- Section of Radiotherapy, Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mimi Kjærsgaard
- Department of Paediatric Haematology and Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rene Mathiasen
- Department of Paediatric Haematology and Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Karsten Nysom
- Department of Paediatric Haematology and Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Astrid Sehested
- Department of Paediatric Haematology and Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ivan Richter Vogelius
- Section of Radiotherapy, Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Maja Vestmø Maraldo
- Section of Radiotherapy, Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
16
|
Lo Greco MC, Milazzotto R, Liardo RLE, Foti PV, Palmucci S, Basile A, Pergolizzi S, Spatola C. The Role of Reirradiation in Childhood Progressive Diffuse Intrinsic Pontine Glioma (DIPG): An Ongoing Challenge beyond Radiobiology. Brain Sci 2023; 13:1449. [PMID: 37891817 PMCID: PMC10605436 DOI: 10.3390/brainsci13101449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/26/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
To investigate the clinical impact of multiple courses of irradiation on pediatric patients with progressive diffuse intrinsic pontine glioma (DIPG), we conducted a retrospective case series on three children treated at our institution from 2018 to 2022. All children were candidates to receive systemic therapy with vinorelbine and nimotuzumab. Radiotherapy was administered to a total dose of 54 Gy. At any disease progression, our local tumor board evaluated the possibility of offering a new course of radiotherapy. To determine feasibility and assess toxicity rates, all children underwent clinical and hematological evaluation both during and after the treatment. To assess efficacy, all children performed contrast-enhanced MRI almost quarterly after the end of the treatment. In all children, following any treatment course, neurological improvement (>80%) was associated with a radiological response (41.7-46%). The longest overall survival (24 months) was observed in the child who underwent three courses of radiotherapy, without experiencing significant side effects. Even though it goes beyond the understanding of conventional radiobiology, first and second reirradiation in pediatric patients with progressive DIPG may represent a feasible and safe approach, capable of increasing overall survival and disease-free survival in selected patients and improving their quality of life.
Collapse
Affiliation(s)
- Maria Chiara Lo Greco
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy;
| | - Roberto Milazzotto
- Radiation Oncology Unit, Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (R.M.); (R.L.E.L.); (C.S.)
| | - Rocco Luca Emanuele Liardo
- Radiation Oncology Unit, Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (R.M.); (R.L.E.L.); (C.S.)
| | - Pietro Valerio Foti
- Radiology I Unit, Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (P.V.F.); (S.P.); (A.B.)
| | - Stefano Palmucci
- Radiology I Unit, Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (P.V.F.); (S.P.); (A.B.)
| | - Antonio Basile
- Radiology I Unit, Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (P.V.F.); (S.P.); (A.B.)
| | - Stefano Pergolizzi
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy;
| | - Corrado Spatola
- Radiation Oncology Unit, Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (R.M.); (R.L.E.L.); (C.S.)
| |
Collapse
|
17
|
Al Sharie S, Abu Laban D, Al-Hussaini M. Decoding Diffuse Midline Gliomas: A Comprehensive Review of Pathogenesis, Diagnosis and Treatment. Cancers (Basel) 2023; 15:4869. [PMID: 37835563 PMCID: PMC10571999 DOI: 10.3390/cancers15194869] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Diffuse midline gliomas (DMGs) are a group of aggressive CNS tumors, primarily affecting children and young adults, which have historically been associated with dismal outcomes. As the name implies, they arise in midline structures in the CNS, primarily in the thalamus, brainstem, and spinal cord. In more recent years, significant advances have been made in our understanding of DMGs, including molecular features, with the identification of potential therapeutic targets. We aim to provide an overview of the most recent updates in the field of DMGs, including classification, molecular subtypes, diagnostic techniques, and emerging therapeutic strategies including a review of the ongoing clinical trials, thus providing the treating multidisciplinary team with a comprehensive understanding of the current landscape and potential therapeutic strategies for this devastating group of tumors.
Collapse
Affiliation(s)
- Sarah Al Sharie
- Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan;
| | - Dima Abu Laban
- Department of Radiology, King Hussein Cancer Center, Amman 11941, Jordan;
| | - Maysa Al-Hussaini
- Department of Pathology and Laboratory Medicine, King Hussein Cancer Center, Amman 11941, Jordan
| |
Collapse
|
18
|
Liu KX, Haas-Kogan DA, Elhalawani H. Radiotherapy for Primary Pediatric Central Nervous System Malignancies: Current Treatment Paradigms and Future Directions. Pediatr Neurosurg 2023; 58:356-366. [PMID: 37703864 DOI: 10.1159/000533777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 08/21/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Central nervous system tumors are the most common solid tumors in childhood. Treatment paradigms for pediatric central nervous system malignancies depend on elements including tumor histology, age of patient, and stage of disease. Radiotherapy is an important modality of treatment for many pediatric central nervous system malignancies. SUMMARY While radiation contributes to excellent overall survival rates for many patients, radiation also carries significant risks of long-term side effects including neurocognitive decline, hearing loss, growth impairment, neuroendocrine dysfunction, strokes, and secondary malignancies. In recent decades, clinical trials have demonstrated that with better imaging and staging along with more sophisticated radiation planning and treatment set-up verification, smaller treatment volumes can be utilized without decrement in survival. Furthermore, the development of intensity-modulated radiotherapy and proton-beam radiotherapy has greatly improved conformality of radiation. KEY MESSAGES Recent changes in radiation treatment paradigms have decreased risks of short- and long-term toxicity for common histologies and in different age groups. Future studies will continue to develop novel radiation regimens to improve outcomes in aggressive central nervous system tumors, integrate molecular subtypes to tailor radiation treatment, and decrease radiation-associated toxicity for long-term survivors.
Collapse
Affiliation(s)
- Kevin X Liu
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - 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, Massachusetts, USA
| | - Hesham Elhalawani
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
19
|
De Pietro R, Zaccaro L, Marampon F, Tini P, De Felice F, Minniti G. The evolving role of reirradiation in the management of recurrent brain tumors. J Neurooncol 2023; 164:271-286. [PMID: 37624529 PMCID: PMC10522742 DOI: 10.1007/s11060-023-04407-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023]
Abstract
Despite aggressive management consisting of surgery, radiation therapy (RT), and systemic therapy given alone or in combination, a significant proportion of patients with brain tumors will experience tumor recurrence. For these patients, no standard of care exists and management of either primary or metastatic recurrent tumors remains challenging.Advances in imaging and RT technology have enabled more precise tumor localization and dose delivery, leading to a reduction in the volume of health brain tissue exposed to high radiation doses. Radiation techniques have evolved from three-dimensional (3-D) conformal RT to the development of sophisticated techniques, including intensity modulated radiation therapy (IMRT), volumetric arc therapy (VMAT), and stereotactic techniques, either stereotactic radiosurgery (SRS) or stereotactic radiotherapy (SRT). Several studies have suggested that a second course of RT is a feasible treatment option in patients with a recurrent tumor; however, survival benefit and treatment related toxicity of reirradiation, given alone or in combination with other focal or systemic therapies, remain a controversial issue.We provide a critical overview of the current clinical status and technical challenges of reirradiation in patients with both recurrent primary brain tumors, such as gliomas, ependymomas, medulloblastomas, and meningiomas, and brain metastases. Relevant clinical questions such as the appropriate radiation technique and patient selection, the optimal radiation dose and fractionation, tolerance of the brain to a second course of RT, and the risk of adverse radiation effects have been critically discussed.
Collapse
Affiliation(s)
- Raffaella De Pietro
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | - Lucy Zaccaro
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | - Francesco Marampon
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | - Paolo Tini
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Francesca De Felice
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | - Giuseppe Minniti
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Policlinico Umberto I, Rome, Italy.
- IRCCS Neuromed, Pozzilli (IS), Isernia, Italy.
| |
Collapse
|
20
|
Zaghloul MS, Hunter A, Mostafa AG, Parkes J. Re-irradiation for recurrent/progressive pediatric brain tumors: from radiobiology to clinical outcomes. Expert Rev Anticancer Ther 2023; 23:709-717. [PMID: 37194207 DOI: 10.1080/14737140.2023.2215439] [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: 01/29/2023] [Accepted: 05/15/2023] [Indexed: 05/18/2023]
Abstract
INTRODUCTION Brain tumors are the most common solid tumors in children. Neurosurgical excision, radiotherapy, and/or chemotherapy represent the standard of care in most histopathological types of pediatric central nervous system (CNS) tumors. Even though the successful cure rate is reasonable, some patients may develop recurrence locally or within the neuroaxis. AREA COVERED The management of these recurrences is not easy; however, significant advances in neurosurgery, radiation techniques, radiobiology, and the introduction of newer biological therapies, have improved the results of their salvage treatment. In many cases, salvage re-irradiation is feasible and has achieved encouraging results. The results of re-irradiation depend upon several factors. These factors include tumor type, extent of the second surgery, tumor volume, location of the recurrence, time that elapses between the initial treatment, the combination with other treatment agents, relapse, and the initial response to radiotherapy. EXPERT OPINION Reviewing the radiobiological basis and clinical outcome of pediatric brain re-irradiation revealed that re-irradiation is safe, feasible, and indicated for recurrent/progressive different tumor types such as; ependymoma, medulloblastoma, diffuse intrinsic pontine glioma (DIPG) and glioblastoma. It is now considered part of the treatment armamentarium for these patients. The challenges and clinical results in treating recurrent pediatric brain tumors were highly documented.
Collapse
Affiliation(s)
- Mohamed S Zaghloul
- Radiation Oncology department. National Cancer Institute, Cairo University & Children's Cancer Hospital, Cairo, Egypt
| | - Alistair Hunter
- Division of Radiobiology, Radiation Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Ayatullah G Mostafa
- Department of Radiology, Faculty of Medicine, Egypt and Department of Diagnostic Imaging, Cairo University, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jeannette Parkes
- Radiation Oncology Department, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
21
|
Huang WH, Huang TY, Lin CM, Mu PF, Lee YY, Liu SH, Hsu SM, Chen YW. Salvage boron neutron capture therapy for pediatric patients with recurrent diffuse midline glioma. Childs Nerv Syst 2023; 39:1529-1536. [PMID: 36821007 DOI: 10.1007/s00381-023-05850-2] [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: 12/06/2022] [Accepted: 01/14/2023] [Indexed: 02/24/2023]
Abstract
PURPOSE Pediatric diffuse malignant glioma located in the brainstem was officially named "diffuse midline glioma" (DMG) by the World Health Organization in 2016. For this disease, radical surgery is not beneficial, and the only major treatment strategy is radiotherapy. However, the dose limitations to brainstem tissue mean that treatment by radiotherapy can only control and not eradicate the tumors, and there is no effective treatment for recurrence, resulting in short overall survival of 6-12 months. This paper reports our experience with boron neutron capture therapy (BNCT), a new treatment process, and its efficacy in treating children with recurrent DMG. METHODS From September 2019 to July 2022, we treated 6 children affected by recurrent DMG. With the collaboration of Taipei Veteran General Hospital (TVGH) and National Tsing-Hua University (NTHU), each patient received two sessions of BNCT within 1 month. RESULTS Among the six patients, three showed partial response and the rest had stable disease after the treatment. The overall survival and recurrence-free survival duration after treatment were 6.39 and 4.35 months, respectively. None of the patients developed severe side effects, and only one patient developed brain necrosis, which was most likely resulted from previous hypofractionated radiotherapy received. CONCLUSION BNCT elicited sufficient tumor response with low normal tissue toxicity; it may benefit vulnerable pediatric patients with DMG.
Collapse
Affiliation(s)
- Wei-Hsuan Huang
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei City, Taiwan
- Medical Physics and Radiation Measurements Laboratory, National Yang Ming Chiao Tung University, Taipei City, Taiwan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Ting-Yu Huang
- Department of Nursing, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Chun-Mei Lin
- Department of Nursing, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Pei-Fan Mu
- College of Nursing, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Yi-Yen Lee
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Shih-Hua Liu
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei City, Taiwan
| | - Shih-Ming Hsu
- Medical Physics and Radiation Measurements Laboratory, National Yang Ming Chiao Tung University, Taipei City, Taiwan.
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei City, Taiwan.
| | - Yi-Wei Chen
- Division of Radiotherapy, Department of Oncology, Taipei Veterans General Hospital, Taipei City, Taiwan.
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan.
- Department of Medical Imaging and Radiological Technology, Yuanpei University of Medical Technology, Hsinchu City, Taiwan.
- School of Medicine, National Tsing Hua University, Hsinchu City, Taiwan.
| |
Collapse
|
22
|
Kim HJ, Suh CO. Radiotherapy for Diffuse Intrinsic Pontine Glioma: Insufficient but Indispensable. Brain Tumor Res Treat 2023; 11:79-85. [PMID: 37151149 PMCID: PMC10172015 DOI: 10.14791/btrt.2022.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/28/2022] [Accepted: 01/02/2023] [Indexed: 05/09/2023] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) account for 10%-20% of all central nervous system tumors in children and are the leading cause of death in children with brain tumors. Although many clinical trials have been conducted over the past decades, the survival outcome has remained unchanged. Over 90% of children die within 2 years of the diagnosis, and radiotherapy remains the standard treatment to date. To improve the prognosis, hyperfractionated and hypofractionated radiotherapy and/or addition of radiosensitizers have been investigated. However, none of the radiotherapy approaches have shown a survival benefit, and the overall survival of patients with DIPG is approximately 11 months. Here, we comprehensively review the management of DIPG with focus on radiotherapy.
Collapse
Affiliation(s)
- Hyun Ju Kim
- Department of Radiation Oncology, Gachon University Gil Hospital, Gachon University College of Medicine, Incheon, Korea
| | - Chang-Ok Suh
- Department of Radiation Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea.
| |
Collapse
|
23
|
Farrukh S, Habib S, Rafaqat A, Sarfraz Z, Sarfraz A, Sarfraz M, Robles-Velasco K, Felix M, Cherrez-Ojeda I. Emerging Therapeutic Strategies for Diffuse Intrinsic Pontine Glioma: A Systematic Review. Healthcare (Basel) 2023; 11:healthcare11040559. [PMID: 36833093 PMCID: PMC9956230 DOI: 10.3390/healthcare11040559] [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: 11/04/2022] [Revised: 01/19/2023] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Of all central nervous systems tumors, 10-20% are located in the brainstem; diffuse intrinsic pontine glioma (DIPG) is diagnosed in 80% of them. With over five decades of clinical trial testing, there are no established therapeutic options for DIPG. This research article aims to collate recent clinical trial data and provide a landscape for the most promising therapies that have emerged in the past five years. METHODS PubMed/MEDLINE, Web of Science, Scopus, and Cochrane were systematically searched using the following keywords: Diffuse intrinsic pontine glioma, Pontine, Glioma, Treatment, Therapy, Therapeutics, curative, and/or Management. Both adult and pediatric patients with newly diagnosed or progressive DIPG were considered in the clinical trial setting. The risk of bias was assessed using the ROBINS-I tool. RESULTS A total of 22 trials were included reporting the efficacy and safety outcomes among patients. First, five trials reported outcomes of blood-brain barrier bypass via single or repeated-dose intra-arterial therapy or convection-enhanced delivery. Second, external beam radiation regimens were assessed for safety and efficacy in three trials. Third, four trials administered intravenous treatment without using chemotherapeutic regimens. Fourth, eight trials reported the combinations of one or more chemotherapeutic agents. Fifth, immunotherapy was reported in two trials in an adjuvant monotherapy in the post-radiotherapy setting. CONCLUSION This research article captures a clinical picture of the last five years of the direction toward which DIPG research is heading. The article finds that re-irradiation may prolong survival in patients with progressive DIPG; it also instills that insofar palliative radiotherapy has been a key prognostic choice.
Collapse
Affiliation(s)
- Shahrukh Farrukh
- Department of Research, Khawaja Muhammad Safdar Medical College, Sialkot 51310, Pakistan
| | - Shagufta Habib
- Department of Research, University Medical and Dental College Faisalabad, Faisalabad 38800, Pakistan
| | - Amna Rafaqat
- Department of Research and Publications, Fatima Jinnah Medical University, Lahore 54000, Pakistan
| | - Zouina Sarfraz
- Department of Research and Publications, Fatima Jinnah Medical University, Lahore 54000, Pakistan
| | - Azza Sarfraz
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi 74000, Pakistan
- Correspondence: (A.S.); (I.C.-O.)
| | | | - Karla Robles-Velasco
- Department of Allergy, Immunology and Pulmonary Medicine, Universidad Espíritu Santo, Samborondón 092301, Ecuador
| | - Miguel Felix
- Department of Internal Medicine, New York City Health + Hospitals, Lincoln, The Bronx, NY 10451, USA
| | - Ivan Cherrez-Ojeda
- Department of Allergy, Immunology and Pulmonary Medicine, Universidad Espíritu Santo, Samborondón 092301, Ecuador
- Correspondence: (A.S.); (I.C.-O.)
| |
Collapse
|
24
|
Di Nunno V, Franceschi E, Gatto L, Tosoni A, Bartolini S, Brandes AA. How to treat histone 3 altered gliomas: molecular landscape and therapeutic developments. Expert Rev Clin Pharmacol 2023; 16:17-26. [PMID: 36576307 DOI: 10.1080/17512433.2023.2163385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Diffuse midline gliomas (DMG) and diffuse hemispheric glioma (DHG) are both rare tumors characterized and recognized for specific alterations of histone 3 including H3K27 (DMG) and H3G34 (DHG). Despite these tumors arising from alterations of the same gene their clinical, radiological, and molecular behaviors strongly diverge, requiring a personalized therapeutic approach. AREAS COVERED We performed a review on Medline/PudMed aiming to search papers relative to prospective trials, retrospective studies, case series, and case reports of interest in order to investigate current knowledge toward the main clinical and molecular characteristics, radiology, and diagnosis, loco-regional and systemic treatments of these tumors. Moreover, we also evaluated the novel treatments under investigation. EXPERT OPINION Thanks to an increased knowledge of the genomic landscape of these rare tumors, there are novels promising therapeutic targets for these malignancies. However, the majority of available trials allowed enrollment only in DMG, while few studies are focused on or allow the inclusion of DHG patients.
Collapse
Affiliation(s)
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Italy
| | - Lidia Gatto
- Department of Oncology, AUSL Bologna, Bologna, Italy
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Italy
| | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Italy
| | - Alba Ariela Brandes
- Nervous System Medical Oncology Department, IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Italy
| |
Collapse
|
25
|
Coleman C, Chen K, Lu A, Seashore E, Stoller S, Davis T, Braunstein S, Gupta N, Mueller S. Interdisciplinary care of children with diffuse midline glioma. Neoplasia 2022; 35:100851. [PMID: 36410226 PMCID: PMC9676429 DOI: 10.1016/j.neo.2022.100851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/10/2022] [Accepted: 10/22/2022] [Indexed: 11/19/2022] Open
Abstract
Diffuse Midline Glioma (DMG) which includes Diffuse Intrinsic Pontine Glioma (DIPG) is an infiltrative tumor of the midline structures of the central nervous system that demonstrates an aggressive pattern of growth and has no known curative treatment. As these tumors progress, children experience ongoing neurological decline including inability to ambulate, swallow and communicate effectively. We propose that optimal care for patients with DMG should involve a specialized team experienced in caring for the multifaceted needs of these patients and their families. Herein we review the roles and evidence to support early involvement of a specialized interdisciplinary team and outline our views on best practices for these challenging tumors.
Collapse
Affiliation(s)
- Christina Coleman
- Department of Pediatrics, University of California, San Francisco, United States
| | - Katherine Chen
- Department of Radiation Oncology, University of California, San Francisco, United States
| | - Alex Lu
- Department of Neurological Surgery, University of California, San Francisco, United States
| | - Elizabeth Seashore
- Department of Pediatrics, University of California, San Francisco, United States
| | - Schuyler Stoller
- Department of Neurology, University of California, San Francisco, United States
| | - Taron Davis
- Department of Orthopedic Surgery, University of California, San Francisco, United States
| | - Steve Braunstein
- Department of Radiation Oncology, University of California, San Francisco, United States
| | - Nalin Gupta
- Department of Pediatrics, University of California, San Francisco, United States,Department of Neurological Surgery, University of California, San Francisco, United States
| | - Sabine Mueller
- Department of Pediatrics, University of California, San Francisco, United States,Department of Neurological Surgery, University of California, San Francisco, United States,Department of Neurology, University of California, San Francisco, United States,Department of Pediatrics, University of Zurich, Zurich, Switzerland,Corresponding author at: Departments of Neurology, Neurosurgery and Pediatrics, University of California, San Francisco, Sandler Neuroscience Building, 675 Nelson Rising Lane, San Francisco, CA 94148, United States.
| |
Collapse
|
26
|
Bernstock JD, Hoffman SE, Kappel AD, Valdes PA, Essayed WI, Klinger NV, Kang KD, Totsch SK, Olsen HE, Schlappi CW, Filipski K, Gessler FA, Baird L, Filbin MG, Hashizume R, Becher OJ, Friedman GK. Immunotherapy approaches for the treatment of diffuse midline gliomas. Oncoimmunology 2022; 11:2124058. [PMID: 36185807 PMCID: PMC9519005 DOI: 10.1080/2162402x.2022.2124058] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 12/14/2022] Open
Abstract
Diffuse midline gliomas (DMG) are a highly aggressive and universally fatal subgroup of pediatric tumors responsible for the majority of childhood brain tumor deaths. Median overall survival is less than 12 months with a 90% mortality rate at 2 years from diagnosis. Research into the underlying tumor biology and numerous clinical trials have done little to change the invariably poor prognosis. Continued development of novel, efficacious therapeutic options for DMGs remains a critically important area of active investigation. Given that DMGs are not amenable to surgical resection, have only limited response to radiation, and are refractory to traditional chemotherapy, immunotherapy has emerged as a promising alternative treatment modality. This review summarizes the various immunotherapy-based treatments for DMG as well as their specific limitations. We explore the use of cell-based therapies, oncolytic virotherapy or immunovirotherapy, immune checkpoint inhibition, and immunomodulatory vaccination strategies, and highlight the recent clinical success of anti-GD2 CAR-T therapy in diffuse intrinsic pontine glioma (DIPG) patients. Finally, we address the challenges faced in translating preclinical and early phase clinical trial data into effective standardized treatment for DMG patients.
Collapse
Affiliation(s)
- Joshua D. Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Samantha E. Hoffman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Children’s Hospital Cancer Center, Boston, MA, USA
| | - Ari D. Kappel
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Pablo A. Valdes
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Walid Ibn Essayed
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Neil V. Klinger
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kyung-Don Kang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stacie K. Totsch
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hannah E. Olsen
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Charles W. Schlappi
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Children’s Hospital Cancer Center, Boston, MA, USA
| | - Katharina Filipski
- Neurological Institute (Edinger Institute), University Hospital, Frankfurt Am Main, Germany
- German Cancer Consortium (DKTK), Germany and German Cancer Research Center (DFKZ), Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt, Germany
- University Cancer Center (UCT), Frankfurt, Germany
| | - Florian A. Gessler
- Department of Neurosurgery, University Medicine Rostock, Rostock, Germany
| | - Lissa Baird
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Mariella G. Filbin
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Children’s Hospital Cancer Center, Boston, MA, USA
| | - Rintaro Hashizume
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Oren J. Becher
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, the Mount Sinai Hospital, NY, NY, USA
| | - Gregory K. Friedman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
27
|
Chavaz L, Janssens GO, Bolle S, Mandeville H, Ramos-Albiac M, Van Beek K, Benghiat H, Hoeben B, Morales La Madrid A, Seidel C, Kortmann RD, Hargrave D, Gandola L, Pecori E, van Vuurden DG, Biassoni V, Massimino M, Kramm CM, von Bueren AO. Neurological Symptom Improvement After Re-Irradiation in Patients With Diffuse Intrinsic Pontine Glioma: A Retrospective Analysis of the SIOP-E-HGG/DIPG Project. Front Oncol 2022; 12:926196. [PMID: 35814457 PMCID: PMC9259094 DOI: 10.3389/fonc.2022.926196] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/09/2022] [Indexed: 11/19/2022] Open
Abstract
Purpose The aim of this study is to investigate the spectrum of neurological triad improvement in patients with diffuse intrinsic pontine glioma (DIPG) treated by re-irradiation (re-RT) at first progression. Methods We carried out a re-analysis of the SIOP-E retrospective DIPG cohort by investigating the clinical benefits after re-RT with a focus on the neurological triad (cranial nerve deficits, ataxia, and long tract signs). Patients were categorized as “responding” or “non-responding” to re-RT. To assess the interdependence between patients’ characteristics and clinical benefits, we used a chi-square or Fisher’s exact test. Survival according to clinical response to re-RT was calculated by the Kaplan–Meier method. Results As earlier reported, 77% (n = 24/31) of patients had any clinical benefit after re-RT. Among 25/31 well-documented patients, 44% (n = 11/25) had improvement in cranial nerve palsies, 40% (n = 10/25) had improvement in long-tract signs, and 44% (11/25) had improvement in cerebellar signs. Clinical benefits were observed in at least 1, 2, or 3 out of 3 symptoms of the DIPG triad, in 64%, 40%, and 24%, respectively. Patients irradiated with a dose ≥20 Gy versus <20 Gy may improve slightly better with regard to ataxia (67% versus 23%; p-value = 0.028). The survival from the start of re-RT to death was not different between responding and non-responding DIPG patients (p-value = 0.871). Conclusion A median re-irradiation dose of 20 Gy provides a neurological benefit in two-thirds of patients with an improvement of at least one symptom of the triad. DIPG patients receiving ≥20 Gy appear to improve slightly better with regard to ataxia; however, we need more data to determine whether dose escalation up to 30 Gy provides additional benefits.
Collapse
Affiliation(s)
- Lara Chavaz
- Department of Pediatrics, Gynecology and Obstetrics, Division of Pediatric Hematology and Oncology, University Hospital of Geneva, Geneva, Switzerland
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
| | - Geert O. Janssens
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, Netherlands
- Princess Maxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Stephanie Bolle
- Department of Radiation Oncology, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Henry Mandeville
- Department of Radiotherapy, The Royal Marsden Hospital and Institute of Cancer Research, Sutton, United Kingdom
| | | | - Karen Van Beek
- Department of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Helen Benghiat
- Department of Clinical Oncology, University Hospital Birmingham, Birmingham, United Kingdom
| | - Bianca Hoeben
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, Netherlands
- Princess Maxima Center for Pediatric Oncology, Utrecht, Netherlands
| | | | - Clemens Seidel
- Department of Radiation-Oncology, University Hospital Leipzig, Leipzig, Germany
| | | | - Darren Hargrave
- Pediatric Oncology Unit, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Lorenza Gandola
- Pediatric Radiotherapy Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale dei Tumori, Milan, Italy
| | - Emilia Pecori
- Pediatric Radiotherapy Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale dei Tumori, Milan, Italy
| | - Dannis G. van Vuurden
- Princess Maxima Center for Pediatric Oncology, Utrecht, Netherlands
- Department of Pediatric Oncology, Emma Children’s Hospital, Amsterdam University Medical Centers (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Veronica Biassoni
- Pediatrics Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale dei Tumori, Milan, Italy
| | - Maura Massimino
- Pediatrics Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale dei Tumori, Milan, Italy
| | - Christof M. Kramm
- Division of Pediatric Hematology and Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Andre O. von Bueren
- Department of Pediatrics, Gynecology and Obstetrics, Division of Pediatric Hematology and Oncology, University Hospital of Geneva, Geneva, Switzerland
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
- *Correspondence: Andre O. von Bueren,
| |
Collapse
|
28
|
Bronk JK, Hou P, Amsbaugh MJ, Khatua S, Mahajan A, Ketonen L, McGovern SL. Sequential Diffusion Tensor Imaging and Magnetic Resonance Spectroscopy in Patients Undergoing Reirradiation for Progressive Diffuse Intrinsic Pontine Glioma. Adv Radiat Oncol 2022; 7:100847. [PMID: 35071836 PMCID: PMC8763636 DOI: 10.1016/j.adro.2021.100847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 10/31/2021] [Indexed: 11/29/2022] Open
Abstract
Purpose Diffusion tensor imaging for evaluation of white matter tracts is used with magnetic resonance spectroscopy (MRS) to improve management of diffuse intrinsic pontine glioma (DIPG). Changes in the apparent diffusion coefficient (ADC), fractional anisotropy (FA), and tumor metabolite ratios have been reported after initial radiation for DIPG, but these markers have not been studied sequentially in patients undergoing reirradiation for progressive DIPG. Here, we report a case series of 4 patients who received reirradiation for progressive DIPG on a prospective clinical trial in which we evaluated quantitative changes in FA, ADC, and tumor metabolites and qualitative changes in white matter tracts. Methods and Materials The median reirradiation dose was 25.2 Gy (24-30.8 Gy). Fiber tracking was performed using standard tractography analysis. The FA and ADC values for the corticospinal and medial lemniscus tracts were calculated before and after reirradiation. Multivoxel MRS was performed. Findings were correlated with clinical features and conventional MRI of tumors. Results All patients had an initial response to reirradiation as shown by a decrease in tumor size. In general, FA increased with disease response and decreased with progression, whereas ADC decreased with disease response and increased with progression. At second progression, the FA fold change relative to values during disease response decreased in both patients with available imaging at second progression. Visualization of tracts demonstrated robust reconstitution of previously disrupted paths during tumor response; conversely, there was increased fiber tract disruption and infiltration during tumor progression. The MRS analysis revealed a decrease in choline:creatinine and choline:N-acetylaspartate ratios during tumor response and increase during progression. Conclusions Distinct changes in white matter tracts and tumor metabolism were observed in patients with DIPG undergoing reirradiation on a prospective clinical trial. Changes related to tumor response and progression were observed after 24 to 30.8 Gy reirradiation.
Collapse
Affiliation(s)
- Julianna K. Bronk
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ping Hou
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark J. Amsbaugh
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Soumen Khatua
- Departments of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anita Mahajan
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Leena Ketonen
- Departments of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Susan L. McGovern
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Corresponding author: Susan L. McGovern, MD, PhD
| |
Collapse
|
29
|
Fan H, Sievert W, Hofmann J, Keppler SJ, Steiger K, Puig-Bosch X, Haller B, Rammes G, Multhoff G. Partial-Brain Radiation-Induced Microvascular Cognitive Impairment in Juvenile Murine Unilateral Hippocampal Synaptic Plasticity. Int J Radiat Oncol Biol Phys 2021; 112:747-758. [PMID: 34619330 DOI: 10.1016/j.ijrobp.2021.09.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE Radiation-induced cognitive deficits have a severe negative impact on pediatric brain tumor patients. The severity of cognitive symptoms is related to the age of the child when radiation was applied, with the most severe effects seen in the youngest. Previous studies using whole-brain irradiation in mice confirmed these findings. To understand ipsilateral and contralateral changes in the hippocampus after partial-brain radiation therapy (PBRT) of the left hemisphere, we assessed the neuroplasticity and changes in the microvasculature of the irradiated and nonirradiated hippocampus in juvenile mice. METHODS AND MATERIALS The left hemispheres of 5-week-old mice were irradiated with 2, 8, and 20 Gy and a fractionated dose of 8 Gy in 2 fractions using a computed tomography image guided small animal radiation research platform. Long-term potentiation (LTP) has been monitored ex vivo in the hippocampal cornu ammonis 1 (CA1) region and was assessed 3 days and 5 and 10 weeks after PBRT in both hemispheres and compared to a sham group. Irradiation effects on the hippocampus microvasculature were quantified by efficient tissue clearing and multiorgan volumetric imaging. RESULTS LTP in irradiated hippocampal slices of juvenile mice declines 3 days after radiation, lasts up to 10 weeks in the irradiated part of the hippocampus, and correlates with a significantly reduced microvasculature length. Specifically, LTP inhibition is sustained in the irradiated (20 Gy, 8 Gy in 2 fractions, 8 Gy, 2 Gy) hippocampus, whereas the contralateral hippocampus remains unaffected after PBRT. LTP inhibition in the irradiated hemisphere after PBRT might be associated with an impaired microvascular network. CONCLUSION PBRT induces a long-lasting impairment in neuroplasticity and the microvessel network of the irradiated hippocampus, whereas the contralateral hippocampus remains unaffected. These findings provide insight into the design of PBRT strategies to better protect the young developing brain from cognitive decline.
Collapse
Affiliation(s)
- Hengyi Fan
- Department of Radiation Oncology, Klinikum rechts der Isar; Central Institute for Translational Cancer Research, TranslaTUM, Klinikum rechts der Isar
| | - Wolfgang Sievert
- Department of Radiation Oncology, Klinikum rechts der Isar; Central Institute for Translational Cancer Research, TranslaTUM, Klinikum rechts der Isar
| | - Julian Hofmann
- Central Institute for Translational Cancer Research, TranslaTUM, Klinikum rechts der Isar; Inflammation and Immunity Lab, Institute for Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar
| | - Selina J Keppler
- Central Institute for Translational Cancer Research, TranslaTUM, Klinikum rechts der Isar; Inflammation and Immunity Lab, Institute for Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar
| | - Katja Steiger
- Comparative Experimental Pathology, Institute Pathology
| | - Xènia Puig-Bosch
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar
| | - Bernhard Haller
- Institute of Medical Informatics, Statistics and Epidemiology, Technische Universität München, Munich, Germany
| | - Gerhard Rammes
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar; Central Institute for Translational Cancer Research, TranslaTUM, Klinikum rechts der Isar.
| |
Collapse
|
30
|
García-Cabezas S, Rivin del Campo E, Solivera-Vela J, Palacios-Eito A. Re-irradiation for high-grade gliomas: Has anything changed? World J Clin Oncol 2021; 12:767-786. [PMID: 34631441 PMCID: PMC8479348 DOI: 10.5306/wjco.v12.i9.767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/21/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
Optimal management after recurrence or progression of high-grade gliomas is still undefined and remains a challenge for neuro-oncology multidisciplinary teams. Improved radiation therapy techniques, new imaging methods, published experience, and a better radiobiological knowledge of brain tissue have positioned re-irradiation (re-RT) as an option for many of these patients. Decisions must be individualized, taking into account the pattern of relapse, previous treatment, and functional status, as well as the patient’s preferences and expected quality of life. Many questions remain unanswered with respect to re-RT: Who is the most appropriate candidate, which dose and fractionation are most effective, how to define the target volume, which imaging technique is best for planning, and what is the optimal timing? This review will focus on describing the most relevant studies that include re-RT as salvage therapy, with the aim of simplifying decision-making and designing the best available therapeutic strategy.
Collapse
Affiliation(s)
- Sonia García-Cabezas
- Department of Radiation Oncology, Reina Sofia University Hospital, Cordoba 14004, Spain
| | | | - Juan Solivera-Vela
- Department of Neurosurgery, Reina Sofia University Hospital, Cordoba 14004, Spain
| | - Amalia Palacios-Eito
- Department of Radiation Oncology, Reina Sofia University Hospital, Cordoba 14004, Spain
| |
Collapse
|
31
|
Zamora PL, Miller SR, Kovoor JJ. Single institution experience in re-irradiation of biopsy-proven diffuse intrinsic pontine gliomas. Childs Nerv Syst 2021; 37:2539-2543. [PMID: 33973056 DOI: 10.1007/s00381-021-05195-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/28/2021] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Diffuse intrinsic pontine glioma (DIPG) is the leading cause of death from CNS tumors in children. Multiple clinical trials have failed to show any benefit from systemic therapy in DIPG, and radiation therapy (RT) alone remains the standard of care. Re-irradiation (rRT) for symptomatic relief is an option at disease progression. However, published data on treatment details and outcomes are limited. The objective of this study was to review and report our institutional experience with re-irradiation of patients with biopsy-proven DIPG. METHODS We identified a cohort of pediatric patients with biopsy-proven DIPG with clinical disease progression after initial radiotherapy who received a second course of radiotherapy at our institution. We reviewed patient and treatment characteristics and outcomes. RESULTS Between January 2014 and July 2018, we identified five patients with progressive DIPG who received re-irradiation. Re-irradiation was well tolerated with no serious adverse events reported and all patients experiencing stable to improved neurologic function during treatment. Median survival from completion of re-irradiation was 116 days (range 62 to 159 days). Median overall survival from time of diagnosis was 16.3 months (range 13.0 to 18.0 months), which is longer than the historical average of less than 12 months. In patients with available postmortem neuropathology, common findings were Wallerian degeneration and necrosis. CONCLUSIONS In our experience, re-irradiation is safe and feasible for patients with DIPG with symptomatic disease progression following initial radiotherapy treatment.
Collapse
Affiliation(s)
- Pedro L Zamora
- Department of Radiation Oncology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Steven R Miller
- Department of Radiation Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Division of Radiation Oncology, Karmanos Cancer Institute, Detroit, MI, USA
| | - Joshua J Kovoor
- Department of Radiation Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
32
|
Krishnatry R, Manjali JJ, Chinnaswamy G, Chatterjee A, Goda JS, Janu A, Sahu A, Jalali R, Gupta T. Clinical approach to re-irradiation for recurrent diffuse intrinsic pontine glioma. Jpn J Clin Oncol 2021; 51:762-768. [PMID: 33521824 DOI: 10.1093/jjco/hyab006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND We present our institutional approach for re-irradiation in diffuse intrinsic pontine glioma and their outcomes. METHODS Consecutive patients of recurrent diffuse intrinsic pontine glioma treated with re-irradiation (January 2015-September 2019) were reviewed retrospectively to describe the clinical-response-based approach followed for the dose and volume decision. Outcomes were defined with clinical and steroid response criteria and survival endpoints included progression-free survival and overall survival as cumulative(c) overall survival and re-irradiation overall survival (re-irradiation starting to death). The Kaplan-Meier method and log-rank test were used for survival analysis. RESULTS Twenty-patient cohort with a median (m) age of 7.5 years, m-progression-free survival of 8.4 months and m-Lansky performance score of 50 received re-irradiation of which 17 (85%) were called clinical responders. The median re-irradiation-overall survival with 39.6-41.4, 43.2 and 45 Gy were 5.8, 7 and 5.3 months, respectively. One-month post-re-irradiation steroid independent status was a significant predictor of better survival outcomes (overall survival, P≤0.004). No ≥ grade 3 toxicities were noticed. Two patients succumbed to intra-tumoral hemorrhage. CONCLUSIONS Higher doses of re-irradiation based on a clinical-response-based approach show improvement in survival and steroid dependence rates with acceptable toxicity. Steroid independent status at 1-month post-re-irradiation predicts better outcomes. Prospective studies may validate this with quality of life data.
Collapse
Affiliation(s)
- Rahul Krishnatry
- Department of Radiation Oncology, Tata Memorial Centre (TMH/ACTREC), Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Jifmi Jose Manjali
- Department of Radiation Oncology, Tata Memorial Centre (TMH/ACTREC), Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Girish Chinnaswamy
- Department of Pediatric Oncology, Tata Memorial Centre (TMH/ACTREC), Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Abhishek Chatterjee
- Department of Radiation Oncology, Tata Memorial Centre (TMH/ACTREC), Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Jayant Sastri Goda
- Department of Radiation Oncology, Tata Memorial Centre (TMH/ACTREC), Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Amit Janu
- Department of Radiology, Tata Memorial Centre (TMH/ACTREC), Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Arpita Sahu
- Department of Radiology, Tata Memorial Centre (TMH/ACTREC), Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Rakesh Jalali
- Department of Radiation Oncology, Tata Memorial Centre (TMH/ACTREC), Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Tejpal Gupta
- Department of Radiation Oncology, Tata Memorial Centre (TMH/ACTREC), Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
33
|
A phase I/II study of bevacizumab, irinotecan and erlotinib in children with progressive diffuse intrinsic pontine glioma. J Neurooncol 2021; 153:263-271. [PMID: 33963476 PMCID: PMC8211596 DOI: 10.1007/s11060-021-03763-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/20/2021] [Indexed: 12/26/2022]
Abstract
INTRODUCTION This study investigates the safety, tolerability, and preliminary efficacy of combined treatment with VEGF inhibitor bevacizumab, topoisomerase I inhibitor irinotecan, and EGFR inhibitor erlotinib in children with progressive diffuse intrinsic pontine glioma (DIPG). METHODS Biweekly bevacizumab (10 mg/kg) and irinotecan (125 mg/m2) were combined with daily erlotinib. Two cohorts received increasing doses of erlotinib (65 and 85 mg/m2) following a 3 + 3 dose-escalation schedule, until disease progression with a maximum of one year. Dose-limiting toxicities (DLT) were monitored biweekly. Secondary progression free survival (sPFS) and overall survival (OS) were determined based on clinical and radiological response measurements. Quality of life (QoL) during treatment was also assessed. RESULTS Between November 2011 and March 2018, nine patients with disease progression after initial radiotherapy were enrolled. Median PFS at start of the study was 7.3 months (range 3.5-10.0). In the first dose cohort, one patient experienced a DLT (grade III acute diarrhea), resulting in enrollment of three additional patients in this cohort. No additional DLTs were observed in consecutive patients receiving up to a maximum dose of 85 mg/m2. Median sPFS was 3.2 months (range 1.0-10.9), and median OS was 13.8 months (range 9.3-33.0). Overall QoL was stable during treatment. CONCLUSIONS Daily erlotinib is safe and well tolerated in doses up to 85 mg/m2 when combined with biweekly bevacizumab and irinotecan in children with progressive DIPG. Median OS of the study patients was longer than known form literature.
Collapse
|
34
|
Prasad D, Vern-Gross T, Wolden S. Radiosurgery, reirradiation, and brachytherapy. Pediatr Blood Cancer 2021; 68 Suppl 2:e28531. [PMID: 33818888 DOI: 10.1002/pbc.28531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 06/07/2020] [Accepted: 06/08/2020] [Indexed: 11/05/2022]
Abstract
Radiosurgery and brachytherapy are potentially useful treatment techniques that are sparingly applied in pediatric oncology. They are often used in the setting of reirradiation for recurrent or metastatic tumors. Reirradiation in children with recurrent tumors is complicated by the tolerance of critical organs and the potential risks for overall long-term dose-dependent complications. We review the current literature available in support of reirradiation and the use of radiosurgery and brachytherapy in pediatric patients.
Collapse
Affiliation(s)
- Dheerendra Prasad
- Department of Radiation Oncology and Neurosurgery, Roswell Park Comprehensive Cancer Center and Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York
| | | | - Suzanne Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
35
|
Yoon HI, Wee CW, Kim YZ, Seo Y, Im JH, Dho YS, Kim KH, Hong JB, Park JS, Choi SH, Kim MS, Moon J, Hwang K, Park JE, Cho JM, Yoon WS, Kim SH, Kim YI, Kim HS, Sung KS, Song JH, Lee MH, Han MH, Lee SH, Chang JH, Lim DH, Park CK, Lee YS, Gwak HS. The Korean Society for Neuro-Oncology (KSNO) Guideline for Adult Diffuse Midline Glioma: Version 2021.1. Brain Tumor Res Treat 2021; 9:1-8. [PMID: 33913265 PMCID: PMC8082289 DOI: 10.14791/btrt.2021.9.e8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 02/15/2021] [Accepted: 03/03/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND There have been no guidelines for the management of adult patients with diffuse midline glioma (DMG), H3K27M-mutant in Korea since the 2016 revised WHO classification newly defined this disease entity. Thus, the Korean Society for Neuro-Oncology (KSNO), a multidisciplinary academic society, had begun preparing guidelines for DMG since 2019. METHODS The Working Group was composed of 27 multidisciplinary medical experts in Korea. References were identified through searches of PubMed, MEDLINE, EMBASE, and Cochrane CENTRAL using specific and sensitive keywords as well as combinations of keywords. As 'diffuse midline glioma' was recently defined, and there was no international guideline, trials and guidelines of 'diffuse intrinsic pontine glioma' or 'brain stem glioma' were thoroughly reviewed first. RESULTS The core contents are as follows. The DMG can be diagnosed when all of the following three criteria are satisfied: the presence of the H3K27M mutation, midline location, and infiltrating feature. Without identification of H3K27M mutation by diagnostic biopsy, DMG cannot be diagnosed. For the primary treatment, maximal safe resection should be considered for tumors when feasible. Radiotherapy is the primary option for tumors in case the total resection is not possible. A total dose of 54 Gy to 60 Gy with conventional fractionation prescribed at 1-2 cm plus gross tumor volume is recommended. Although no chemotherapy has proven to be effective in DMG, concurrent chemoradiotherapy (± maintenance chemotherapy) with temozolomide following WHO grade IV glioblastoma's protocol is recommended. CONCLUSION The detection of H3K27M mutation is the most important diagnostic criteria for DMG. Combination of surgery (if amenable to surgery), radiotherapy, and chemotherapy based on comprehensive multidisciplinary discussion can be considered as the treatment options for DMG.
Collapse
Affiliation(s)
- Hong In Yoon
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Chan Woo Wee
- Department of Radiation Oncology, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Young Zoon Kim
- Division of Neurooncology and Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Youngbeom Seo
- Department of Neurosurgery, Yeungnam University Hospital, Yeungnam University College of Medicine, Daegu, Korea
| | - Jung Ho Im
- Department of Radiation Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| | - Yun Sik Dho
- Department of Neurosurgery, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Kyung Hwan Kim
- Department of Neurosurgery, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Korea
| | - Je Beom Hong
- Department of Neurosurgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae Sung Park
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seo Hee Choi
- Department of Radiation Oncology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Min Sung Kim
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jangsup Moon
- Department of Genomic Medicine, Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Kihwan Hwang
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jin Mo Cho
- Department of Neurosurgery, Catholic Kwandong University, International St. Mary's Hospital, Incheon, Korea
| | - Wan Soo Yoon
- Department of Neurosurgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young Il Kim
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyoung Su Sung
- Department of Neurosurgery, Dong-A University Hospital, Dong-A University College of Medicine, Busan, Korea
| | - Jin Ho Song
- Department of Radiation Oncology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Min Ho Lee
- Department of Neurosurgery, Uijeongbu St. Mary's Hospital, The Catholic University of Korea, Uijeongbu, Korea
| | - Myung Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Se Hoon Lee
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chul Kee Park
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
| | - Youn Soo Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| | - Ho Shin Gwak
- Department of Cancer Control, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Korea.
| |
Collapse
|
36
|
Cacciotti C, Liu KX, Haas-Kogan DA, Warren KE. Reirradiation practices for children with diffuse intrinsic pontine glioma. Neurooncol Pract 2021; 8:68-74. [PMID: 33664971 DOI: 10.1093/nop/npaa063] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Background Diffuse intrinsic pontine gliomas (DIPGs) are a leading cause of brain tumor deaths in children. Current standard of care includes focal radiation therapy (RT). Despite clinical improvement in most patients, the effect is temporary and median survival is less than 1 year. The use and benefit of reirradiation have been reported in progressive DIPG, yet standardized approaches are lacking. We conducted a survey to assess reirradiation practices for DIPG in North America. Methods A 14-question REDCap survey was disseminated to 396 North American physicians who care for children with CNS tumors. Results The response rate was 35%. Participants included radiation-oncologists (63%; 85/135) and pediatric oncologists/neuro-oncologists (37%; 50/135). Most physicians (62%) treated 1 to 5 DIPG patients per year, with 10% treating more than 10 patients per year. Reirradiation was considered a treatment option by 88% of respondents. Progressive disease and worsening clinical status were the most common reasons to consider reirradiation. The majority (84%) surveyed considered reirradiation a minimum of 6 months following initial RT. Doses varied, with median total dose of 2400 cGy (range, 1200-6000 cGy) and fraction size of 200 cGy (range, 100-900 cGy). Concurrent use of systemic agents with reirradiation was considered in 46%, including targeted agents (37%), biologics (36%), or immunotherapy (25%). One-time reirradiation was the most common practice (71%). Conclusion Although the vast majority of physicians consider reirradiation as a treatment for DIPG, total doses and fractionation varied. Further clinical trials are needed to determine the optimal radiation dose and fractionation for reirradiation in children with progressive DIPG.
Collapse
Affiliation(s)
- Chantel Cacciotti
- Dana Farber/Boston Children's Cancer and Blood Disorder Center, Boston, Massachusetts
| | - Kevin X Liu
- Department of Radiation-Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Daphne A Haas-Kogan
- Department of Radiation-Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Katherine E Warren
- Dana Farber/Boston Children's Cancer and Blood Disorder Center, Boston, Massachusetts
| |
Collapse
|
37
|
Perez-Somarriba M, Santa-Maria V, Cruz O, Muchart J, Lavarino C, Mico S, Morales La Madrid A. Craniospinal irradiation as a salvage treatment for metastatic relapsed DIPG. Pediatr Blood Cancer 2021; 68:e28762. [PMID: 33063935 DOI: 10.1002/pbc.28762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/28/2020] [Indexed: 12/27/2022]
Affiliation(s)
| | - Vicente Santa-Maria
- Department of Pediatric Oncology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Ofelia Cruz
- Department of Pediatric Oncology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Jordi Muchart
- Department of Radiology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Cinzia Lavarino
- Department of Developmental Tumor Biology Laboratory, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Soraya Mico
- Department of Radiation Oncology, Hospital Vall d'Hebron, Barcelona, Spain
| | | |
Collapse
|
38
|
Yamasaki F, Nishibuchi I, Karakawa S, Kaichi Y, Kolakshyapati M, Takano M, Yonezawa U, Imano N, Taguchi A, Shimomura M, Taniguchi M, Onishi S, Okada S, Awai K, Sugiyama K, Nagata Y. T2-FLAIR Mismatch Sign and Response to Radiotherapy in Diffuse Intrinsic Pontine Glioma. Pediatr Neurosurg 2021; 56:1-9. [PMID: 33535215 DOI: 10.1159/000513360] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/23/2020] [Indexed: 11/19/2022]
Abstract
PURPOSE The T2-fluid-attenuated inversion recovery (FLAIR) mismatch sign was previously reported as a diagnostic indicator of diffuse astrocytoma, isocitrate dehydrogenase-mutant, and 1p/19q noncodeletion. Subsequently, it was reported that the same findings were observed in diffuse intrinsic pontine glioma (DIPG). We investigated the clinical significance of T2-FLAIR mismatch sign in DIPG. METHODS Twenty-one patients with DIPG (Male: Female = 12:9) were treated at our institute between 2004 and 2019. All patients were treated with local radiotherapy of 54 Gy/30 fractions. The positive T2-FLAIR mismatch sign was defined if it fulfilled the following criteria: (1) T2-FLAIR mismatch volume was >50% of T2 high volume at nonenhanced area, (2) the FLAIR low lesion is not associated with gadolinium enhancement (inside of enhancement or just outside of enhancement defined as edema), and (3) signal-intensity of FLAIR lowest lesion at tumor is lower than the normal cerebellar cortex. RESULTS In our patient series, T2-FLAIR mismatch sign was found in 5 out of 21 patients. Objective response rate of radiotherapy was 100% in patients positive for T2-FLAIR mismatch, while it was 25.0% in patients negative for T2-FLAIR mismatch, and this difference was statistically significant (p < 0.01, Fisher's exact test). In patients under the age of 18-years, T2-FLAIR mismatch positive had a slightly better prognosis (p < 0.05, Wilcoxon test). CONCLUSION T2-FLAIR mismatch sign in DIPG may be an indicator for better response to radiotherapy and a better prognostic factor.
Collapse
Affiliation(s)
- Fumiyuki Yamasaki
- Department of Neurosurgery, Hiroshima University Hospital, Hiroshima, Japan,
| | - Ikuno Nishibuchi
- Department of Radiation Oncology, Hiroshima University Hospital, Hiroshima, Japan
| | - Shuhei Karakawa
- Department of Pediatrics, Hiroshima University Hospital, Hiroshima, Japan
| | - Yoko Kaichi
- Department of Diagnostic Radiology, Hiroshima University Hospital, Hiroshima, Japan
| | | | - Motoki Takano
- Department of Neurosurgery, Hiroshima University Hospital, Hiroshima, Japan
| | - Ushio Yonezawa
- Department of Neurosurgery, Hiroshima University Hospital, Hiroshima, Japan
| | - Nobuki Imano
- Department of Radiation Oncology, Hiroshima University Hospital, Hiroshima, Japan
| | - Akira Taguchi
- Department of Neurosurgery, Hiroshima University Hospital, Hiroshima, Japan
| | - Maiko Shimomura
- Department of Pediatrics, Hiroshima University Hospital, Hiroshima, Japan
| | - Maki Taniguchi
- Department of Pediatrics, Hiroshima University Hospital, Hiroshima, Japan
| | - Shumpei Onishi
- Department of Neurosurgery, Hiroshima University Hospital, Hiroshima, Japan
| | - Satoshi Okada
- Department of Pediatrics, Hiroshima University Hospital, Hiroshima, Japan
| | - Kazuo Awai
- Department of Diagnostic Radiology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kazuhiko Sugiyama
- Department of Clinical Oncology and Neuro-Oncology Program, Hiroshima University Hospital, Hiroshima, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
39
|
Lin R, Zhou Y, Yan F, Li D, Yuan Y. BOIN12: Bayesian Optimal Interval Phase I/II Trial Design for Utility-Based Dose Finding in Immunotherapy and Targeted Therapies. JCO Precis Oncol 2020; 4:2000257. [PMID: 33283133 DOI: 10.1200/po.20.00257] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2020] [Indexed: 12/15/2022] Open
Abstract
PURPOSE For immunotherapy, such as checkpoint inhibitors and chimeric antigen receptor T-cell therapy, where the efficacy does not necessarily increase with the dose, the maximum tolerated dose may not be the optimal dose for treating patients. For these novel therapies, the objective of dose-finding trials is to identify the optimal biologic dose (OBD) that optimizes patients' risk-benefit trade-off. METHODS We propose a simple and flexible Bayesian optimal interval phase I/II (BOIN12) trial design to find the OBD that optimizes the risk-benefit trade-off. The BOIN12 design makes the decision of dose escalation and de-escalation by simultaneously taking account of efficacy and toxicity and adaptively allocates patients to the dose that optimizes the toxicity-efficacy trade-off. We performed simulation studies to evaluate the performance of the BOIN12 design. RESULTS Compared with existing phase I/II dose-finding designs, the BOIN12 design is simpler to implement, has higher accuracy to identify the OBD, and allocates more patients to the OBD. One of the most appealing features of the BOIN12 design is that its adaptation rule can be pretabulated and included in the protocol. During the trial conduct, clinicians can simply look up the decision table to allocate patients to a dose without complicated computation. CONCLUSION The BOIN12 design is simple to implement and yields desirable operating characteristics. It overcomes the computational and implementation complexity that plagues existing Bayesian phase I/II dose-finding designs and provides a useful design to optimize the dose of immunotherapy and targeted therapy. User-friendly software is freely available to facilitate the application of the BOIN12 design.
Collapse
Affiliation(s)
- Ruitao Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yanhong Zhou
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Fangrong Yan
- China Pharmaceutical University, Nanjing, People's Republic of China
| | - Daniel Li
- Juno Therapeutics, a Bristol Myers Squibb Company, Seattle, WA
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
40
|
Hayashi A, Ito E, Omura M, Aida N, Tanaka M, Tanaka Y, Sato H, Miyagawa N, Yokosuka T, Iwasaki F, Hamanoue S, Goto H. Hypofractionated radiotherapy in children with diffuse intrinsic pontine glioma. Pediatr Int 2020; 62:47-51. [PMID: 31785177 PMCID: PMC7027509 DOI: 10.1111/ped.14070] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/10/2019] [Accepted: 11/27/2019] [Indexed: 12/03/2022]
Abstract
BACKGROUND Overall survival (OS) of patients with diffuse intrinsic pontine glioma (DIPG) is poor, with radiation therapy (RT) the only intervention that transiently delays tumor progression. Hypofractionated RT and re-irradiation at first progression have gained popularity in improving the quality of life of such patients. METHODS We performed a retrospective review of children with DIPG treated at Kanagawa Children's Medical Center from 2000 to 2018. RESULTS A total of 24 cases were reviewed. Median age at diagnosis was 6.3 years (1.6-14.0). Twenty patients received RT only once. Thirteen patients received conventionally fractionated RT, and seven patients received hypofractionated RT as up-front RT. Severe toxicities were not observed in patients who received hypofractionated RT. Median OS and time to progression were similar between conventionally fractionated and hypofractionated RT groups.(9.7 [95% confidence interval(CI): 7.1-11.2] versus 11.0[95% CI: 5.2-13.6] months, P = 0.60; 4.2[95% CI: 1.8-8.3] versus 7.1 [95% CI:4.5-8.7] months, P = 0.38). Four patients received re-irradiation at first progression and all patients showed transient neurological improvement and survival more than a year after diagnosis. A 4-year-old boy was re-irradiated 5-and-a-half months after the first re-irradiation; following transient neurological improvement. He survived a further 5 months. CONCLUSION Hypofractionated RT for children with newly diagnosed DIPG is well tolerated and feasible from the viewpoint of reducing a patient's burden of treatment. Re-irradiation at first progression is suggested to be beneficial.
Collapse
Affiliation(s)
- Akiko Hayashi
- Division of Hematology/Oncology, Department of Pediatrics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Eiko Ito
- Department of Radiology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Motoko Omura
- Department of Radiology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Noriko Aida
- Department of Radiology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Mio Tanaka
- Department of Pathology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yukichi Tanaka
- Department of Pathology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hironobu Sato
- Department of Neurosurgery, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Naoyuki Miyagawa
- Division of Hematology/Oncology, Department of Pediatrics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Tomoko Yokosuka
- Division of Hematology/Oncology, Department of Pediatrics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Fuminori Iwasaki
- Division of Hematology/Oncology, Department of Pediatrics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Satoshi Hamanoue
- Division of Hematology/Oncology, Department of Pediatrics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hiroaki Goto
- Division of Hematology/Oncology, Department of Pediatrics, Kanagawa Children's Medical Center, Yokohama, Japan
| |
Collapse
|
41
|
Tsang DS, Oliveira C, Bouffet E, Hawkins C, Ramaswamy V, Yee R, Tabori U, Bartels U, Huang A, Millar BA, Crooks B, Bowes L, Zelcer S, Laperriere N. Repeat irradiation for children with supratentorial high-grade glioma. Pediatr Blood Cancer 2019; 66:e27881. [PMID: 31207154 DOI: 10.1002/pbc.27881] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/10/2019] [Accepted: 05/31/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND There are very few studies about the role of repeat irradiation (RT2) for children with recurrent supratentorial high-grade glioma (HGG). It was the aim of this study to assess the effectiveness and safety of RT2 in this population. PROCEDURE This was a retrospective cohort study of 40 children age 18 years and under with recurrent supratentorial HGG who had received at least one course of RT. In-field reirradiation volumes included focal or whole brain RT, with doses ranging from 30 to 54 Gy. The primary endpoint was overall survival (OS) from the first day of RT2. RESULTS Fourteen patients underwent RT2. The median survival of these patients was 6.5 months. Patients with ≥12 months elapsed time between RT1 and RT2 experienced longer OS than patients who had < 12 months (P = 0.009). There was no difference in OS between patients with or without germline mutations (e.g., Lynch, Li-Fraumeni, or constitutional mismatch-repair deficiency, P = 0.20). Ten patients received RT2 that overlapped with RT1 volumes for locally recurrent disease. Of this group, 80% experienced clinical benefit from in-field RT2, defined as clinical/radiologic response or stable disease. Ninety-three percent completed the prescribed course of RT2, with one patient developing grade 3 radiation necrosis four months after RT2. When compared with 26 patients who were not offered reirradiation, those selected for RT2 had improved median survival from the time of first disease progression (9.4 vs 3.8 months, P = 0.005). CONCLUSIONS Reirradiation for children with recurrent supratentorial HGG is a safe, effective treatment that provides short-term disease control.
Collapse
Affiliation(s)
- Derek S Tsang
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Carol Oliveira
- Division of Radiation Oncology, Queen's University, Kingston, Ontario, Canada
| | - Eric Bouffet
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Cynthia Hawkins
- Paediatric Laboratory Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Vijay Ramaswamy
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ryan Yee
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Uri Tabori
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ute Bartels
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Annie Huang
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Barbara-Ann Millar
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Bruce Crooks
- Division of Hematology-Oncology, IWK Health Centre, Halifax, Nova Scotia, Canada
| | - Lynette Bowes
- Janeway Child Health Centre, St. John's, Newfoundland, Canada
| | - Shayna Zelcer
- London Health Sciences Centre, London, Ontario, Canada
| | - Normand Laperriere
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Haematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
42
|
Krishnatry R, Manjali JJ. In Regard to Amsbaugh et al. Int J Radiat Oncol Biol Phys 2019; 104:467-468. [DOI: 10.1016/j.ijrobp.2019.02.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 01/14/2019] [Accepted: 02/14/2019] [Indexed: 11/28/2022]
|
43
|
Amsbaugh MJ, Mahajan A, Thall PF, McAleer MF, Paulino AC, Grosshans D, Khatua S, Ketonen L, Fontanilla H, McGovern SL. In Reply to Krishnatry and Manjali. Int J Radiat Oncol Biol Phys 2019; 104:468-469. [DOI: 10.1016/j.ijrobp.2019.02.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 11/27/2022]
|
44
|
Lu VM, Welby JP, Mahajan A, Laack NN, Daniels DJ. Reirradiation for diffuse intrinsic pontine glioma: a systematic review and meta-analysis. Childs Nerv Syst 2019; 35:739-746. [PMID: 30879125 DOI: 10.1007/s00381-019-04118-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 03/08/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Diffuse intrinsic pontine glioma (DIPG) is a pediatric brain tumor with dismal prognosis despite initial radiation therapy (RT). The clinical consequences of attempting reirradiation (reRT) in these patients to alleviate both symptomatology and improve prognosis are currently unclear. Thus, the aim of this systematic review and meta-analysis was to clarify the efficacy and safety of reRT in DIPG. METHODS Searches of seven electronic databases from inception to January 2019 were conducted following the appropriate guidelines. Articles were screened against prespecified criteria. The incidence and duration of clinical outcomes were then extracted and pooled by means of meta-analysis from the included studies. RESULTS A total of 7 studies satisfied all criteria, describing 90 cases of DIPG in which reRT was attempted 11.8-14 months after initial RT. Based on a random-effects model, the incidences of clinical improvement and radiologic response following reRT were 87% (95% CI, 78-95%) and 69% (95% CI, 52-84%), respectively. The incidence of acute serious toxicity was 0% (95% CI, 0-4%). Pooled overall survivals from initial diagnosis and time of reRT were 18.0 months (95% CI, 14.2-21.7) and 6.2 months (95% CI, 5.5-7.0), respectively. CONCLUSIONS Based on these results, the clinical consequences of reRT for DIPG when administered appropriately and safely at first progression appear acceptable, and potentially favorable, based on the limited evidence in the current literature. Concerns regarding acute serious toxicity were not realized. It is likely that a subcohort of all DIPG diagnoses will be most amenable to improve prognosis with reRT, and greater investigation is required to identify their characteristics.
Collapse
Affiliation(s)
- Victor M Lu
- Department of Neurosurgery, Mayo Clinic, 200 First St. SW, Rochester, MN, USA.
| | - John P Welby
- Department of Neurosurgery, Mayo Clinic, 200 First St. SW, Rochester, MN, USA
| | - Anita Mahajan
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Nadia N Laack
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - David J Daniels
- Department of Neurosurgery, Mayo Clinic, 200 First St. SW, Rochester, MN, USA.
| |
Collapse
|