Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Am J Case Rep ; 25: e944869, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39348336

ABSTRACT

BACKGROUND Diffuse intrinsic pontine glioma represent approximately 10% to 20% of all pediatric central nervous system tumors. Classic brain stem symptoms are cranial nerve deficits, long tract signs, ataxia, alone or in combination. Focal radiotherapy has been the standard of care in patients with diffuse intrinsic pontine gliomas with minimum response. Here, we present an unusual case with excellent tumor regression with radiotherapy and good clinical outcome. CASE REPORT A 13-year-old girl presented with headache and imbalance during walking for the past 2-3 months, along with a deviation of the right eye in the last month. Brain magnetic resonance imaging (MRI) suggested a well-defined solid cystic altered-signal-intensity lesion involving the pons and medulla, causing its expansion up to the midbrain on the left side. The lesion was 4.6×3.7×3.6 cm. We applied the intensity-modulated radiotherapy technique (IMRT) using a 6-MV photon beam with the conventional dose fractionation of 54 Gy in 30 fractions (1.8 Gy/fraction). Three months later, MRI brain with spectroscopy and perfusion showed evidence of non-enhancing, altered-signal-intensity lesion in the pons and medulla, measuring 1.9×2.2×2.4 cm. CONCLUSIONS Early detection of symptoms of DIPG in a young patient along with effective radiological investigation with valid tumor board decision as definitive radiotherapy as a sole therapeutic treatment option and with robust radiotherapy planning resulted in an excellent response, with 80% reduction in gross tumor volume (GTV) as seen in pre-radiotherapy (RT) and post-RT MRI images.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Humans , Female , Adolescent , Brain Stem Neoplasms/radiotherapy , Brain Stem Neoplasms/diagnostic imaging , Diffuse Intrinsic Pontine Glioma/radiotherapy , Magnetic Resonance Imaging , Radiotherapy, Intensity-Modulated , Pons/diagnostic imaging , Pons/pathology , Glioma/radiotherapy , Glioma/diagnostic imaging
2.
J Nucl Med ; 65(9): 1364-1370, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39142829

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is a rare childhood malignancy with poor prognosis. There are no effective treatment options other than external beam therapy. We conducted a pilot, first-in-human study using 124I-omburtamab imaging and theranostics as a therapeutic approach using a localized convection-enhanced delivery (CED) technique for administering radiolabeled antibody. We report the detailed pharmacokinetics and dosimetry results of intratumoral delivery of 124I-omburtamab. Methods: Forty-five DIPG patients who received 9.0-370.7 MBq of 124I-omburtamab intratumorally via CED underwent serial brain and whole-body PET/CT imaging at 3-5 time points after injection within 4, 24-48, 72-96, 120-144, and 168-240 h from the end of infusion. Serial blood samples were obtained for kinetic analysis. Whole-body, blood, lesion, and normal-tissue activities were measured, kinetic parameters (uptake and clearance half-life times) estimated, and radiation-absorbed doses calculated using the OLINDA software program. Results: All patients showed prominent activity within the lesion that was retained over several days and was detectable up to the last time point of imaging, with a mean 124I residence time in the lesion of 24.9 h and dose equivalent of 353 ± 181 mSv/MBq. Whole-body doses were low, with a dose equivalent of 0.69 ± 0.28 mSv/MBq. Systemic distribution and activities in normal organs and blood were low. Radiation dose to blood was very low, with a mean value of 0.27 ± 0.21 mGy/MBq. Whole-body clearance was monoexponential with a mean biologic half-life of 62.7 h and an effective half-life of 37.9 h. Blood clearance was biexponential, with a mean biologic half-life of 22.2 h for the rapid α phase and 155 h for the slower ß phase. Conclusion: Intratumoral CED of 124I-omburtamab is a novel theranostics approach in DIPG. It allows for delivery of high radiation doses to the DIPG lesions, with high lesion activities and low systemic activities and high tumor-to-normal-tissue ratios and achieving a wide safety margin. Imaging of the actual therapeutic administration of 124I-omburtamab allows for direct estimation of the therapeutic lesion and normal-tissue-absorbed doses.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Iodine Radioisotopes , Radiometry , Humans , Male , Female , Child , Brain Stem Neoplasms/diagnostic imaging , Brain Stem Neoplasms/radiotherapy , Diffuse Intrinsic Pontine Glioma/diagnostic imaging , Diffuse Intrinsic Pontine Glioma/radiotherapy , Child, Preschool , Adolescent , Convection , Positron Emission Tomography Computed Tomography , Glioma/diagnostic imaging , Glioma/radiotherapy , Tissue Distribution , Infant , Young Adult
3.
Med J Malaysia ; 79(Suppl 4): 87-90, 2024 08.
Article in English | MEDLINE | ID: mdl-39215422

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is a highly aggressive paediatric brain tumour and nowadays has not had satisfactory result, with most patients do not survive within 1 year of diagnosis. Due to its proximity to critical organs, surgery is avoided, and radiation is the mainstay of treatment. In this case report, we present a case of DIPG treated with radiation and concurrent temozolomide. A 7- year-old child was admitted with complaints of weakness in the eyelid, upper and lower limbs 2 months ago. Physical examination showed tetra paresis and bilateral cranial nerve palsy. Magnetic resonance imaging (MRI) scan showed intracranial tumour consistent with DIPG. Diagnosis was made based on imaging as surgery or biopsy can lead to further morbidity. The patient underwent radiotherapy with concurrent chemotherapy of temozolomide. Radiation was given by dose of 54 Gy/30 fractions (30 × 1.8 Gy) with volumetric arc therapy (VMAT). Due to technical issue after the first five irradiations resulting in 2 weeks delay, boosting of dose by 5 × 1.8 Gy was then given, hence, the total dose was 63 Gy. The booster only targeted the gross tumour volume. Following radiation, the patient felt clinical improvement. Eyelid and limb movement improved since the 15th fraction. At the last fraction, the patient's condition improved symptomatically, but experienced complaints related to post radiation oedema including dizziness and nausea. These complaints were improved upon steroids administration. The MRI evaluation will be done after 8 to 12 weeks of radiation, considering the effects of acute radiation could still occur at this period. In conclusion, a combination of radiotherapy and temozolomide could be an option for DIPG management, with tolerable acute toxicity and possible clinical improvements.


Subject(s)
Brain Stem Neoplasms , Temozolomide , Humans , Child , Temozolomide/therapeutic use , Temozolomide/administration & dosage , Brain Stem Neoplasms/radiotherapy , Brain Stem Neoplasms/therapy , Brain Stem Neoplasms/drug therapy , Diffuse Intrinsic Pontine Glioma/therapy , Diffuse Intrinsic Pontine Glioma/drug therapy , Diffuse Intrinsic Pontine Glioma/radiotherapy , Male , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Agents, Alkylating/administration & dosage , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Magnetic Resonance Imaging , Chemoradiotherapy
4.
Strahlenther Onkol ; 200(9): 797-804, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38748214

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 2­Gy 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.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Disease Progression , Magnetic Resonance Imaging , Re-Irradiation , Humans , Brain Stem Neoplasms/radiotherapy , Brain Stem Neoplasms/diagnostic imaging , Male , Female , Child , Retrospective Studies , Prognosis , Child, Preschool , Diffuse Intrinsic Pontine Glioma/radiotherapy , Diffuse Intrinsic Pontine Glioma/diagnostic imaging , Adolescent , Proportional Hazards Models , Radiotherapy Dosage , Radiomics
5.
Pediatr Blood Cancer ; 71(5): e30929, 2024 May.
Article in English | MEDLINE | ID: mdl-38430472

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.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Re-Irradiation , Adolescent , Child , Child, Preschool , Humans , Brain Stem Neoplasms/radiotherapy , Diffuse Intrinsic Pontine Glioma/radiotherapy , Radiation Dose Hypofractionation , Steroids
6.
N Engl J Med ; 386(26): 2471-2481, 2022 06 30.
Article in English | MEDLINE | ID: mdl-35767439

ABSTRACT

BACKGROUND: Pediatric patients with diffuse intrinsic pontine glioma (DIPG) have a poor prognosis, with a median survival of less than 1 year. Oncolytic viral therapy has been evaluated in patients with pediatric gliomas elsewhere in the brain, but data regarding oncolytic viral therapy in patients with DIPG are lacking. METHODS: We conducted a single-center, dose-escalation study of DNX-2401, an oncolytic adenovirus that selectively replicates in tumor cells, in patients with newly diagnosed DIPG. The patients received a single virus infusion through a catheter placed in the cerebellar peduncle, followed by radiotherapy. The primary objective was to assess the safety and adverse-event profile of DNX-2401. The secondary objectives were to evaluate the effect of DNX-2401 on overall survival and quality of life, to determine the percentage of patients who have an objective response, and to collect tumor-biopsy and peripheral-blood samples for correlative studies of the molecular features of DIPG and antitumor immune responses. RESULTS: A total of 12 patients, 3 to 18 years of age, with newly diagnosed DIPG received 1×1010 (the first 4 patients) or 5×1010 (the subsequent 8 patients) viral particles of DNX-2401, and 11 received subsequent radiotherapy. Adverse events among the patients included headache, nausea, vomiting, and fatigue. Hemiparesis and tetraparesis developed in 1 patient each. Over a median follow-up of 17.8 months (range, 5.9 to 33.5), a reduction in tumor size, as assessed on magnetic resonance imaging, was reported in 9 patients, a partial response in 3 patients, and stable disease in 8 patients. The median survival was 17.8 months. Two patients were alive at the time of preparation of the current report, 1 of whom was free of tumor progression at 38 months. Examination of a tumor sample obtained during autopsy from 1 patient and peripheral-blood studies revealed alteration of the tumor microenvironment and T-cell repertoire. CONCLUSIONS: Intratumoral infusion of oncolytic virus DNX-2401 followed by radiotherapy in pediatric patients with DIPG resulted in changes in T-cell activity and a reduction in or stabilization of tumor size in some patients but was associated with adverse events. (Funded by the European Research Council under the European Union's Horizon 2020 Research and Innovation Program and others; EudraCT number, 2016-001577-33; ClinicalTrials.gov number, NCT03178032.).


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Oncolytic Virotherapy , Oncolytic Viruses , Adenoviridae , Adolescent , Astrocytoma/radiotherapy , Astrocytoma/therapy , Brain Stem Neoplasms/mortality , Brain Stem Neoplasms/pathology , Brain Stem Neoplasms/radiotherapy , Brain Stem Neoplasms/therapy , Child , Child, Preschool , Diffuse Intrinsic Pontine Glioma/mortality , Diffuse Intrinsic Pontine Glioma/radiotherapy , Diffuse Intrinsic Pontine Glioma/therapy , Glioma/radiotherapy , Glioma/therapy , Humans , Infusions, Intralesional , Oncolytic Virotherapy/adverse effects , Oncolytic Virotherapy/methods , Quality of Life , Tumor Microenvironment
7.
PLoS One ; 17(2): e0263822, 2022.
Article in English | MEDLINE | ID: mdl-35157705

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is a lethal pediatric brain tumor. While there are a number of in vivo rodent models for evaluating tumor biology and response to therapy, these models require significant time and resources. Here, we established the chick-embryo chorioallantoic (CAM) assay as an affordable and time efficient xenograft model for testing a variety of treatment approaches for DIPG. We found that patient-derived DIPG tumors develop in the CAM and maintain the same genetic and epigenetic characteristics of native DIPG tumors. We monitored tumor response to pharmaco- and radiation therapy by 3-D ultrasound volumetric and vasculature analysis. In this study, we established and validated the CAM model as a potential intermediate xenograft model for DIPG and its use for testing novel treatment approaches that include pharmacotherapy or radiation.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Stem Neoplasms/genetics , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/radiation effects , Diffuse Intrinsic Pontine Glioma/genetics , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/pathology , Brain Stem Neoplasms/radiotherapy , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/pathology , Diffuse Intrinsic Pontine Glioma/drug therapy , Diffuse Intrinsic Pontine Glioma/pathology , Diffuse Intrinsic Pontine Glioma/radiotherapy , Humans , Rats , Ultrasonography , Xenograft Model Antitumor Assays
8.
Int J Radiat Oncol Biol Phys ; 113(2): 360-368, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35150788

ABSTRACT

PURPOSE: Pediatric diffuse intrinsic pontine glioma is an orphen disease. This study aimed to confirm the noninferiority of hypofractionated (HF) radiation therapy. Identification of the prognostic factors that determine the overall survival (OS) and progression-free survival (PFS) was the secondary objective. METHODS AND MATERIALS: We randomized 253 patients into 3 arms of radiation therapy regimens: HF1, receiving 39 Gy in 13 fractions; HF2, receiving 45 Gy in 15 fractions; and conventional fractionation (CF), receiving 54 Gy in 30 fractions. The OS and PFS were calculated using Kaplan-Meier methods, and the noninferiority was estimated against the CF arm. RESULTS: The median OS for the HF1, HF2, and CF were 9.6, 8.2, and 8.7 months, respectively. The 1-, 1.5-, and 2-year OS were 34.6%, 17.9%, and 10.7% for HF1; 26.2%, 13.1%, and 4.8% for HF2; and 25.3%, 12.1%, and 8.4% for CF, respectively (P = .3). The hazard ratio was 0.776 and 1.124 for HF1 and HF2, respectively. Considering the noninferiority margin (Δ) of 15% and a power of 90%, the lower inferiority confidence interval for HF1 was -14.34% and for HF2 it was 11.37% (both below Δ), confirming its noninferiority at 18-months OS. Younger patients (2-5 years of age) had better median OS in the whole cohort (11.6 months), HF1 (13.5), and CF (12.1) but not HF2 (6.2) (P = .003). Furthermore, the OS rates at 1, 1.5, and 2 years for children 2 to 5 years of age in the HF2 arm were lower than those in the HF1 and CF arms. However, similar acute and late side effects were reported in the 3 arms. CONCLUSIONS: Two hypofractionated radiation therapy proved to be noninferior to conventional fractionation. Young age (2-5 years) is the only prognostic factor determining both OS and PFS. The young age superiority was lost with a higher hypofractionated radiation therapy dose, necessitating more caution in applying 45 Gy in 15 fractions in younger children (2-5 years of age).


Subject(s)
Astrocytoma , Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Brain Stem Neoplasms/radiotherapy , Child , Child, Preschool , Diffuse Intrinsic Pontine Glioma/radiotherapy , Dose Fractionation, Radiation , Humans , Infant , Radiation Dose Hypofractionation
9.
Jpn J Clin Oncol ; 51(5): 762-768, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33521824

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.


Subject(s)
Brain Stem Neoplasms/radiotherapy , Diffuse Intrinsic Pontine Glioma/radiotherapy , Quality of Life/psychology , Re-Irradiation/methods , Brain Stem Neoplasms/mortality , Child , Cohort Studies , Diffuse Intrinsic Pontine Glioma/mortality , Female , Humans , Male , Progression-Free Survival , Prospective Studies , Retrospective Studies , Survival Analysis
10.
World Neurosurg ; 148: e565-e571, 2021 04.
Article in English | MEDLINE | ID: mdl-33476781

ABSTRACT

OBJECTIVE: Diffuse intrinsic pontine glioma (DIPG) is a rare and devastating brainstem glioma that occurs predominately in children. To date, the prognostic impact of radiotherapy (RT) in conjunction with temozolomide (TMZ) in DIPG has not been thoroughly analyzed. The aim of this meta-analysis was to analyze the effectiveness of RT quantitatively and precisely in conjunction with TMZ in improving the prognosis of DIPG. METHODS: A systematic search of 8 electronic databases was conducted. Articles mainly discussing the prognostic impact of RT in conjunction with TMZ in DIPG were selected. The pooled 1- and 2-year overall survival (OS) and progression-free survival (PFS) were calculated. RESULTS: A total of 14 studies fulfilled our inclusion criteria, involving 283 cases of patients with DIPG who were treated with RT in conjunction with TMZ. The pooled 1- and 2-year OS of this treatment was 43% and 11%, respectively. The pooled 1- and 2-year PFS was 20% and 2%, respectively. Subgroup analysis revealed that the heterogeneity remained almost the same in all stratum. Egger's test demonstrated that the possibility of publication bias was low. CONCLUSIONS: Requirements of up-to-date evidence on evaluating the prognostic impact of this therapy are urgent.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Stem Neoplasms/therapy , Chemoradiotherapy/methods , Diffuse Intrinsic Pontine Glioma/therapy , Glioma/therapy , Temozolomide/therapeutic use , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/radiotherapy , Diffuse Intrinsic Pontine Glioma/drug therapy , Diffuse Intrinsic Pontine Glioma/radiotherapy , Glioma/drug therapy , Glioma/radiotherapy , Humans , Prognosis
11.
Acta Neuropathol Commun ; 9(1): 14, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431066

ABSTRACT

An adequate understanding of the relationships between radiographic and genomic features in diffuse intrinsic pontine glioma (DIPG) is essential, especially in the absence of universal biopsy, to further characterize the molecular heterogeneity of this disease and determine which patients are most likely to respond to biologically-driven therapies. Here, a radiogenomics analytic approach was applied to a cohort of 28 patients with DIPG. Tumor size and imaging characteristics from all available serial MRIs were evaluated by a neuro-radiologist, and patients were divided into three radiographic response groups (partial response [PR], stable disease [SD], progressive disease [PD]) based on MRI within 2 months of radiotherapy (RT) completion. Whole genome and RNA sequencing were performed on autopsy tumor specimens. We report several key, therapeutically-relevant findings: (1) Certain radiologic features on first and subsequent post-RT MRIs are associated with worse overall survival, including PD following irradiation as well as present, new, and/or increasing peripheral ring enhancement, necrosis, and diffusion restriction. (2) Upregulation of EMT-related genes and distant tumor spread at autopsy are observed in a subset of DIPG patients who exhibit poorer radiographic response to irradiation and/or higher likelihood of harboring H3F3A mutations, suggesting possible benefit of upfront craniospinal irradiation. (3) Additional genetic aberrations were identified, including DYNC1LI1 mutations in a subgroup of patients with PR on post-RT MRI; further investigation into potential roles in DIPG tumorigenesis and/or treatment sensitivity is necessary. (4) Whereas most DIPG tumors have an immunologically "cold" microenvironment, there appears to be a subset which harbor a more inflammatory genomic profile and/or higher mutational burden, with a trend toward improved overall survival and more favorable radiographic response to irradiation, in whom immunotherapy should be considered. This study has begun elucidating relationships between post-RT radiographic response with DIPG molecular profiles, revealing radiogenomically distinct subgroups with unique clinical trajectories and therapeutic targets.


Subject(s)
Brain Stem Neoplasms/diagnostic imaging , Brain Stem Neoplasms/genetics , Diffuse Intrinsic Pontine Glioma/diagnostic imaging , Diffuse Intrinsic Pontine Glioma/genetics , Imaging Genomics , Adolescent , Antineoplastic Agents/therapeutic use , Brain Stem Neoplasms/pathology , Brain Stem Neoplasms/radiotherapy , Chemotherapy, Adjuvant , Child , Child, Preschool , Cytoplasmic Dyneins/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Diffuse Intrinsic Pontine Glioma/pathology , Diffuse Intrinsic Pontine Glioma/radiotherapy , Disease Progression , Epithelial-Mesenchymal Transition/genetics , ErbB Receptors/genetics , Female , Histones/genetics , Humans , Magnetic Resonance Imaging , Male , Pilot Projects , Proton Therapy , Radiation-Sensitizing Agents/therapeutic use , Radiotherapy , Sequence Analysis, RNA , Survival Rate , Tumor Microenvironment/genetics , Tumor Suppressor Proteins/genetics , Whole Genome Sequencing , Young Adult
13.
Medicine (Baltimore) ; 99(42): e22721, 2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33080729

ABSTRACT

BACKGROUND: The standard treatment for diffuse intrinsic pontine glioma (DIPG) is radiotherapy, although conventional fractionated radiotherapy (CFRT) may not be in the best interest of the patient. Instead, hypofractionated radiotherapy (HFRT) may shorten the treatment period and reduce related costs for this treatment, which is typically palliative in nature. METHODS: This systematic review and meta-analysis evaluated survival outcomes among patients who received HFRT or CFRT for DIPG. The PubMed, Medline, EMBASE, Cochrane Central Register, and Scopus databases were searched to identify relevant studies. Overall survival was the primary outcome of interest and progression-free survival was the secondary outcome of interest. RESULTS: The search identified a total of 2376 reports, although only 4 reports were ultimately included in the meta-analysis. The studies included 88 patients who underwent HFRT and 96 patients who underwent CFRT. Relative to CFRT, HFRT provided comparable outcomes in terms of overall survival (hazard ratio [HR]: 1.07, 95% confidence interval [CI]: 0.77-1.47) and progression-free survival (HR: 1.04, 95% CI: 0.75-1.45). CONCLUSIONS: The results of this meta-analysis suggest that CFRT and HFRT provide similar survival outcomes for patients with DIPG.


Subject(s)
Brain Stem Neoplasms/radiotherapy , Diffuse Intrinsic Pontine Glioma/radiotherapy , Humans , Radiation Dose Hypofractionation
14.
J Neurooncol ; 147(1): 195-203, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32016716

ABSTRACT

PURPOSE: To estimate the rate and magnitude of neurologic symptom change during radiation therapy (RT) and impact of symptom change on survival outcomes in patients with diffuse intrinsic pontine glioma (DIPG). METHODS: From 2006 to 2014, 108 patients with newly diagnosed DIPG were treated with conventionally fractionated radiation therapy (RT) to 54 Gy (median) at our institution. The presence and severity of neurologic symptoms related to cranial neuropathy (CN) and cerebellar (CB) and long-tract (LT) signs was reviewed before and weekly during RT for each patient. The rate and magnitude of change for each symptom category was evaluated according to accumulated RT dose. The impact of clinical factors and radiation dose-volume parameters was determined using Cox proportional hazards models. RESULTS: Median dose to first sign of symptomatic improvement was 16.2 Gy (CN), 19.8 Gy (LT) and 21.6 Gy (CB). Most patients showed an improvement by 20 Gy. Larger uninvolved brainstem volume, alone or normalized to total brain (TB) or posterior fossa volume (PF), was associated with shorter time to LT sign improvement (P = 0.044, P = 0.033, and P = 0.05, respectively). Patients with any improvement in CN experienced significantly, yet modestly, prolonged progression-free survival (PFS) and overall survival (OS) (P = 0.002 and P = 0.008, respectively). Tumor volume, with or without normalization to TB or PF, was not significantly associated with PFS or OS. CONCLUSIONS: Low cumulative RT doses resulted in neurologic improvement in most patients with DIPG. The volume of brainstem spared by tumor influenced time to symptomatic improvement. Neurologic improvement during RT was associated with superior survival.


Subject(s)
Brain Stem Neoplasms/psychology , Brain Stem Neoplasms/radiotherapy , Diffuse Intrinsic Pontine Glioma/psychology , Diffuse Intrinsic Pontine Glioma/radiotherapy , Dose Fractionation, Radiation , Adolescent , Brain Stem Neoplasms/diagnosis , Child , Child, Preschool , Cohort Studies , Diffuse Intrinsic Pontine Glioma/diagnosis , Female , Humans , Kaplan-Meier Estimate , Male , Neuropsychology , Treatment Outcome
16.
Pediatr Int ; 62(1): 47-51, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31785177

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.


Subject(s)
Brain Stem Neoplasms/radiotherapy , Diffuse Intrinsic Pontine Glioma/radiotherapy , Adolescent , Child , Child, Preschool , Disease Progression , Female , Humans , Infant , Male , Quality of Life , Re-Irradiation , Retrospective Studies
17.
Int J Radiat Oncol Biol Phys ; 106(4): 838-847, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31785339

ABSTRACT

PURPOSE: Optimal radiation therapy (RT) target margins for diffuse intrinsic pontine glioma (DIPG) are unknown. We sought to define disease progression patterns in a contemporary cohort treated with conformal RT using different clinical target volume (CTV) margins. METHODS AND MATERIALS: We reviewed 105 patients with newly diagnosed DIPG treated with conformal conventionally fractionated RT at our institution from 2006 to 2014. CTV margins were classified as standard (1 cm) for 60 patients and extended (2-3 cm) for 45 patients. Survival and cumulative incidence of progression in treatment groups were compared by log-rank and Gray's tests, respectively. Cox proportional hazard models identified predictors of survival. RESULTS: For 97 patients evaluated with magnetic resonance imaging at progression, the cumulative incidences of isolated local, isolated distant, and synchronous disease progression at 1 year were 62.6%, 12.3%, and 7.2%, respectively, and did not differ significantly according to the CTV margin. Central dosimetric progression (Vprogression95% ≥95%) was observed in 80 of 81 evaluable patients. Median progression-free survival and overall survival (OS) were 7.6 months (95% confidence interval, 6.9-8.2) and 11.3 months (95% confidence interval, 10.0-12.8), respectively, and did not differ significantly according to margin status. DIPG survival prediction risk group (standard vs high, P = .02; intermediate vs high, P = .009) and development of distant metastasis (P = .003) were independent predictors of OS. For the 41 patients (39%) with a pathologic diagnosis, H3.3 K27M mutation was associated with shorter OS (hazard ratio [HR], 0.41; P =.02), whereas H3.1 K27M and ACVR1 mutations were associated with longer OS (HR, 3.56; P =.004 and HR, 2.58; P =.04, respectively). CONCLUSIONS: All patients who experienced local failure showed progression within the high-dose volume, and there was no apparent survival or tumor-control benefit to extending the CTV margins beyond 1 cm. Given the increasing use of reirradiation, standardizing the CTV margin to 1 cm may improve retreatment tolerance.


Subject(s)
Brain Stem Neoplasms/radiotherapy , Diffuse Intrinsic Pontine Glioma/radiotherapy , Radiotherapy, Conformal , Brain Stem Neoplasms/diagnostic imaging , Brain Stem Neoplasms/pathology , Child , Child, Preschool , Diffuse Intrinsic Pontine Glioma/diagnostic imaging , Diffuse Intrinsic Pontine Glioma/pathology , Disease Progression , Female , Humans , Male , Retrospective Studies , Survival Analysis , Tomography, X-Ray Computed
18.
Clin Cancer Res ; 25(22): 6788-6800, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31481512

ABSTRACT

PURPOSE: Diffuse intrinsic pontine gliomas (DIPG) are the most severe pediatric brain tumors. Although accepted as the standard therapeutic, radiotherapy is only efficient transiently and not even in every patient. The goal of the study was to identify the underlying molecular determinants of response to radiotherapy in DIPG. EXPERIMENTAL DESIGN: We assessed in vitro response to ionizing radiations in 13 different DIPG cellular models derived from treatment-naïve stereotactic biopsies reflecting the genotype variability encountered in patients at diagnosis and correlated it to their principal molecular alterations. Clinical and radiologic response to radiotherapy of a large cohort of 73 DIPG was analyzed according to their genotype. Using a kinome-wide synthetic lethality RNAi screen, we further identified target genes that can sensitize DIPG cells to ionizing radiations. RESULTS: We uncover TP53 mutation as the main driver of increased radioresistance and validated this finding in four isogenic pairs of TP53WT DIPG cells with or without TP53 knockdown. In an integrated clinical, radiological, and molecular study, we show that TP53MUT DIPG patients respond less to irradiation, relapse earlier after radiotherapy, and have a worse prognosis than their TP53WT counterparts. Finally, a kinome-wide synthetic lethality RNAi screen identifies CHK1 as a potential target, whose inhibition increases response to radiation specifically in TP53MUT cells. CONCLUSIONS: Here, we demonstrate that TP53 mutations are driving DIPG radioresistance both in patients and corresponding cellular models. We suggest alternative treatment strategies to mitigate radioresistance with CHK1 inhibitors. These findings will allow to consequently refine radiotherapy schedules in DIPG.


Subject(s)
Brain Stem Neoplasms/metabolism , Diffuse Intrinsic Pontine Glioma/metabolism , Radiation Tolerance , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/mortality , Brain Stem Neoplasms/radiotherapy , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Survival/radiation effects , Diffuse Intrinsic Pontine Glioma/genetics , Diffuse Intrinsic Pontine Glioma/mortality , Diffuse Intrinsic Pontine Glioma/radiotherapy , Dose-Response Relationship, Radiation , Gene Knockdown Techniques , Histones/genetics , Histones/metabolism , Humans , Mutation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Prognosis , RNA Interference , RNA, Small Interfering/genetics , Radiation Tolerance/genetics , Tumor Suppressor Protein p53/genetics
19.
Clin Cancer Res ; 25(18): 5572-5583, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31227500

ABSTRACT

PURPOSE: Radiotherapy (RT) has long been and remains the only treatment option for diffuse intrinsic pontine glioma (DIPG). However, all patients show evidence of disease progression within months of completing RT. No further clinical benefit has been achieved using alternative radiation strategies. Here, we tested the hypothesis that histone demethylase inhibition by GSK-J4 enhances radiation-induced DNA damage, making it a potential radiosensitizer in the treatment of DIPG.Experimental Design: We evaluated the effects of GSK-J4 on genes associated with DNA double-strand break (DSB) repair in DIPG cells by RNA sequence, ATAC sequence, and quantitative real-time PCR. Radiation-induced DNA DSB repair was analyzed by immunocytochemistry of DSB markers γH2AX and 53BP1, DNA-repair assay, and cell-cycle distribution. Clonogenic survival assay was used to determine the effect of GSK-J4 on radiation response of DIPG cells. In vivo response to radiation monotherapy and combination therapy of RT and GSK-J4 was evaluated in patient-derived DIPG xenografts. RESULTS: GSK-J4 significantly reduced the expression of DNA DSB repair genes and DNA accessibility in DIPG cells. GSK-J4 sustained high levels of γH2AX and 53BP1 in irradiated DIPG cells, thereby inhibiting DNA DSB repair through homologous recombination pathway. GSK-J4 reduced clonogenic survival and enhanced radiation effect in DIPG cells. In vivo studies revealed increased survival of animals treated with combination therapy of RT and GSK-J4 compared with either monotherapy. CONCLUSIONS: Together, these results highlight GSK-J4 as a potential radiosensitizer and provide a rationale for developing combination therapy with radiation in the treatment of DIPG.


Subject(s)
Diffuse Intrinsic Pontine Glioma/metabolism , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Animals , Benzazepines/pharmacology , Cell Line, Tumor , DNA Damage , DNA Repair/drug effects , Diffuse Intrinsic Pontine Glioma/genetics , Diffuse Intrinsic Pontine Glioma/mortality , Diffuse Intrinsic Pontine Glioma/radiotherapy , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Female , Homologous Recombination , Humans , Mice , Prognosis , Pyrimidines/pharmacology , Radiation Tolerance/genetics , Xenograft Model Antitumor Assays
20.
Acta Neuropathol Commun ; 7(1): 64, 2019 04 29.
Article in English | MEDLINE | ID: mdl-31036068

ABSTRACT

Pediatric high grade gliomas (pHGG), including diffuse intrinsic pontine gliomas (DIPGs), are aggressive tumors with a dismal outcome. Radiotherapy (RT) is part of the standard of care of these tumors; however, radiotherapy only leads to a transient clinical improvement. Delta-24-RGD is a genetically engineered tumor-selective adenovirus that has shown safety and clinical efficacy in adults with recurrent gliomas. In this work, we evaluated the feasibility, safety and therapeutic efficacy of Delta-24-RGD in combination with radiotherapy in pHGGs and DIPGs models. Our results showed that the combination of Delta-24-RGD with radiotherapy was feasible and resulted in a synergistic anti-glioma effect in vitro and in vivo in pHGG and DIPG models. Interestingly, Delta-24-RGD treatment led to the downregulation of relevant DNA damage repair proteins, further sensitizing tumors cells to the effect of radiotherapy. Additionally, Delta-24-RGD/radiotherapy treatment significantly increased the trafficking of immune cells (CD3, CD4+ and CD8+) to the tumor niche compared with single treatments. In summary, administration of the Delta-24-RGD/radiotherapy combination to pHGG and DIPG models is safe and significantly increases the overall survival of mice bearing these tumors. Our data offer a rationale for the combination Delta-24-RGD/radiotherapy as a therapeutic option for children with these tumors. SIGNIFICANCE: Delta-24-RGD/radiotherapy administration is safe and significantly increases the survival of treated mice. These positive data underscore the urge to translate this approach to the clinical treatment of children with pHGG and DIPGs.


Subject(s)
Brain Neoplasms/therapy , Brain Stem Neoplasms/therapy , Diffuse Intrinsic Pontine Glioma/therapy , Oncolytic Virotherapy/methods , Adenoviridae/physiology , Animals , Brain Neoplasms/complications , Brain Neoplasms/radiotherapy , Brain Stem Neoplasms/complications , Brain Stem Neoplasms/radiotherapy , Cell Line, Tumor , Combined Modality Therapy/methods , DNA Damage , Diffuse Intrinsic Pontine Glioma/complications , Diffuse Intrinsic Pontine Glioma/radiotherapy , Genetic Vectors , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL