Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Pediatr Hematol Oncol ; 43(8): e1217-e1219, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-33031159

ABSTRACT

Extramedullary hematopoiesis (EMH) is hematopoiesis occurring outside of the bone marrow. It has been reported to develop in abdominal organs or lymph nodes after chemotherapy. Here, the authors describe a patient with a localized central nervous system embryonal tumor who, during intensive chemotherapy, developed dural nodules. Biopsy revealed these nodules to be EMH. Without a pathologic diagnosis, this may have been considered disease progression, altering the patient's treatment plan. This report intends to serve as a reminder that EMH should be included in the differential diagnosis of suspicious lesions and highlights the importance of their biopsy because of potential management implications.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Central Nervous System Neoplasms/drug therapy , Dura Mater/pathology , Hematopoiesis, Extramedullary , Neoplasms, Germ Cell and Embryonal/drug therapy , Central Nervous System Neoplasms/pathology , Child, Preschool , Disease Progression , Dura Mater/drug effects , Humans , Male , Neoplasms, Germ Cell and Embryonal/pathology , Prognosis
2.
Pediatr Radiol ; 51(4): 649-657, 2021 04.
Article in English | MEDLINE | ID: mdl-33231717

ABSTRACT

BACKGROUND: Intra-arterial chemotherapy (IAC) represents a mainstay of retinoblastoma treatment in children. Patients with retinoblastoma are uniquely at risk for secondary malignancies and are sensitive to the ionizing effects of radiation. OBJECTIVE: To retrospectively review a single institution's experience with IAC for retinoblastoma and the effect of variable intra-procedural imaging techniques on radiation exposure. MATERIALS AND METHODS: Twenty-four consecutive patients, with a mean age of 30.8±16.3 months (range: 3.2-83.4 months), undergoing IAC for retinoblastoma between May 2014 and May 2020 (72 months) were included. No patients were excluded. The primary outcome was radiation exposure and secondary outcomes included technical success and procedural adverse events. Technical success was defined as catheterization of the ophthalmic or meningolacrimal artery and complete delivery of chemotherapy. Each procedure was retrospectively reviewed and categorized as one of five imaging protocol types. Protocol types were characterized by uniplanar versus multiplanar imaging and digital subtraction angiographic versus roadmap angiographic techniques. Radiation exposure, protocol utilization, the association of protocol and radiation exposure were assessed. RESULTS: During 96 consecutive interventions, 109 ocular treatments were performed. Thirteen of the 96 (15.5%) treatments were bilateral. Ocular technical success was 106 of 109 (97.2%). All three treatment failures were successfully repeated within a week. Mean fluoroscopy time was 6.4±6.2 min (range: 0.7-31.1 min). Mean air kerma was 36.2±52.2 mGy (range: 1.4-215.0 mGy). There were two major (1.8%) complications and four (3.7%) minor complications. Of the 96 procedures, 10 (10.4%), 9 (9.4%), 13 (13.5%), 28 (29.2%) and 36 (37.5%) were performed using protocol types A, B, C, D and E, respectively. For protocol type A, mean fluoroscopy time was 10.3±6.8 min (range: 3.0-25.4 min) and mean air kerma was 118.2±61.2 mGy (range: 24.5-167.3 mGy). For protocol type E, mean fluoroscopy time was 3.1±3.2 min (range: 0.7-15.1 min) and mean air kerma was 5.4±4.2 mGy (range: 1.4-19.5 mGy). Fluoroscopy time and air kerma decreased over time, corresponding to the reduced use of multiplanar imaging and digital subtraction angiography. In the first quartile (procedures 1-24), 8 (33.3%), 7 (29.2%), 2 (8.3%), 6 (25.0%) and 1 (4.2%) were performed using protocol types A, B, C, D and E, respectively. Mean fluoroscopy time was 10.5±8.2 min (range: 2.4-28.1 min) and mean air kerma was 84.2±71.6 mGy (range: 12.8-215.0 mGy). In the final quartile (procedures 73-96), 24 (100%) procedures were performed using protocol type E. Mean fluoroscopy time was 3.5±4.0 min (range: 0.7-15.1 min) and mean air kerma was 5.0±4.3 mGy (range: 1.4-18.0 mGy), representing 66.7% and 94.1% reductions from the first quartile, respectively. Technical success in the second half of the experience was 100%. CONCLUSION: Sequence elimination, consolidation from biplane imaging to lateral-only imaging, and replacing digital subtraction with roadmap angiography dramatically reduced radiation exposure during IAC for retinoblastoma without adversely affecting technical success or safety.


Subject(s)
Radiation Exposure , Retinal Neoplasms , Retinoblastoma , Angiography, Digital Subtraction , Child , Child, Preschool , Drug Tapering , Fluoroscopy , Humans , Infant , Radiation Dosage , Retinal Neoplasms/diagnostic imaging , Retinal Neoplasms/drug therapy , Retinoblastoma/diagnostic imaging , Retinoblastoma/drug therapy , Retrospective Studies
3.
J Neurooncol ; 148(3): 607-617, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32556862

ABSTRACT

INTRODUCTION: Beyond focal radiation, there is no consensus standard therapy for pediatric high-grade glioma (pHGG) and outcomes remain dismal. We describe the largest molecularly-characterized cohort of children with pHGG treated with a 3-drug maintenance regimen of temozolomide, irinotecan, and bevacizumab (TIB) following radiation. METHODS: We retrospectively reviewed 36 pediatric patients treated with TIB at Seattle Children's Hospital from 2009 to 2018 and analyzed survival using the Kaplan-Meier method. Molecular profiling was performed by targeted DNA sequencing and toxicities, steroid use, and palliative care utilization were evaluated. RESULTS: Median age at diagnosis was 10.9 years (18 months-18 years). Genetic alterations were detected in 26 genes and aligned with recognized molecular subgroups including H3 K27M-mutant (12), H3F3A G34-mutant (2), IDH-mutant (4), and hypermutator profiles (4). Fifteen patients (42%) completed 12 planned cycles of maintenance. Side effects associated with chemotherapy delays or modifications included thrombocytopenia (28%) and nausea/vomiting (19%), with temozolomide dosing most frequently modified. Median event-free survival (EFS) and overall survival (OS) was 16.2 and 20.1 months, with shorter survival seen in DIPG (9.3 and 13.3 months, respectively). Survival at 1, 2, and 5 years was 80%, 10% and 0% for DIPG and 85%, 38%, and 16% for other pHGG. CONCLUSION: Our single-center experience demonstrates tolerability of this 3-drug regimen, with prolonged survival in DIPG compared to historical single-agent temozolomide. pHGG survival was comparable to analogous 3-drug regimens and superior to historical agents; however, cure was rare. Children with pHGG remain excellent candidates for the study of novel therapeutics combined with standard therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Stem Neoplasms/drug therapy , Diffuse Intrinsic Pontine Glioma/drug therapy , Glioma/drug therapy , Adolescent , Bevacizumab/administration & dosage , Brain Stem Neoplasms/pathology , Child , Child, Preschool , Diffuse Intrinsic Pontine Glioma/pathology , Female , Follow-Up Studies , Glioma/pathology , Humans , Infant , Irinotecan/administration & dosage , Male , Neoplasm Grading , Retrospective Studies , Survival Rate , Temozolomide/administration & dosage
4.
Curr Oncol Rep ; 16(7): 393, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24838613

ABSTRACT

Intracranial germ cell tumors represent a relatively small portion of pediatric central nervous system tumors, with a reported incidence ranging from 3.6% in North America to 15.3% in parts of Asia. They are known to arise in midline locations, most frequently the pineal gland and suprasellar regions, and affect males twice to three times more frequently than females, especially in the case of pineal-region tumors. The median age of diagnosis is 10-12 years. The treatment of intracranial germ cells tumors is complicated by their relative rarity, histologic heterogeneity, and a lack of widespread consensus on their optimal management. Despite these challenges, important progress continues to be made, with a move toward multicenter and international collaborative efforts that offer potential for clinical advancements. This review focuses on recent developments in the management of intracranial germ cell tumors, including updates in diagnosis, surveillance, biology, treatment, and outcomes.


Subject(s)
Brain Neoplasms/therapy , Neoplasms, Germ Cell and Embryonal/therapy , Antineoplastic Agents/therapeutic use , Brain Neoplasms/diagnosis , Chemotherapy, Adjuvant/methods , Child , Humans , Neoplasms, Germ Cell and Embryonal/diagnosis , Radiotherapy, Adjuvant/methods
5.
Clin Cancer Res ; 28(8): 1614-1627, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35078859

ABSTRACT

PURPOSE: Therapy-related myelodysplastic syndrome and acute leukemias (t-MDS/AL) are a major cause of nonrelapse mortality among pediatric cancer survivors. Although the presence of clonal hematopoiesis (CH) in adult patients at cancer diagnosis has been implicated in t-MDS/AL, there is limited published literature describing t-MDS/AL development in children. EXPERIMENTAL DESIGN: We performed molecular characterization of 199 serial bone marrow samples from 52 patients treated for high-risk neuroblastoma, including 17 with t-MDS/AL (transformation), 14 with transient cytogenetic abnormalities (transient), and 21 without t-MDS/AL or cytogenetic alterations (neuroblastoma-treated control). We also evaluated for CH in a cohort of 657 pediatric patients with solid tumor. RESULTS: We detected at least one disease-defining alteration in all cases at t-MDS/AL diagnosis, most commonly TP53 mutations and KMT2A rearrangements, including involving two novel partner genes (PRDM10 and DDX6). Backtracking studies identified at least one t-MDS/AL-associated mutation in 13 of 17 patients at a median of 15 months before t-MDS/AL diagnosis (range, 1.3-32.4). In comparison, acquired mutations were infrequent in the transient and control groups (4/14 and 1/21, respectively). The relative risk for development of t-MDS/AL in the presence of an oncogenic mutation was 8.8 for transformation patients compared with transient. Unlike CH in adult oncology patients, TP53 mutations were only detectable after initiation of cancer therapy. Last, only 1% of pediatric patients with solid tumor evaluated had CH involving myeloid genes. CONCLUSIONS: These findings demonstrate the clinical relevance of identifying molecular abnormalities in predicting development of t-MDS/AL and should guide the formation of intervention protocols to prevent this complication in high-risk pediatric patients.


Subject(s)
Cancer Survivors , Leukemia, Myeloid, Acute , Neuroblastoma , Adult , Bone Marrow/pathology , Child , Clone Cells , Humans , Leukemia, Myeloid, Acute/genetics , Neuroblastoma/pathology
6.
PLoS One ; 12(6): e0178593, 2017.
Article in English | MEDLINE | ID: mdl-28582410

ABSTRACT

The PI3K/Akt/mTOR signaling pathway is aberrantly activated in various pediatric tumors. We conducted a phase I study of the Akt inhibitor perifosine in patients with recurrent/refractory pediatric CNS and solid tumors. This was a standard 3+3 open-label dose-escalation study to assess pharmacokinetics, describe toxicities, and identify the MTD for single-agent perifosine. Five dose levels were investigated, ranging from 25 to 125 mg/m2/day for 28 days per cycle. Twenty-three patients (median age 10 years, range 4-18 years) with CNS tumors (DIPG [n = 3], high-grade glioma [n = 5], medulloblastoma [n = 2], ependymoma [n = 3]), neuroblastoma (n = 8), Wilms tumor (n = 1), and Ewing sarcoma (n = 1) were treated. Only one DLT occurred (grade 4 hyperuricemia at dose level 4). The most common grade 3 or 4 toxicity at least possibly related to perifosine was neutropenia (8.7%), with the remaining grade 3 or 4 toxicities (fatigue, hyperglycemia, fever, hyperuricemia, and catheter-related infection) occurring in one patient each. Pharmacokinetics was dose-saturable at doses above 50 mg/m2/day with significant inter-patient variability, consistent with findings reported in adult studies. One patient with DIPG (dose level 5) and 4 of 5 patients with high-grade glioma (dose levels 2 and 3) experienced stable disease for two months. Five subjects with neuroblastoma (dose levels 1 through 4) achieved stable disease which was prolonged (≥11 months) in three. No objective responses were noted. In conclusion, the use of perifosine was safe and feasible in patients with recurrent/refractory pediatric CNS and solid tumors. An MTD was not defined by the 5 dose levels investigated. Our RP2D is 50 mg/m2/day.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Central Nervous System Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Neuroblastoma/drug therapy , Phosphorylcholine/analogs & derivatives , Adolescent , Antineoplastic Agents/adverse effects , Central Nervous System Neoplasms/pathology , Child , Child, Preschool , Drug Administration Schedule , Ependymoma/drug therapy , Ependymoma/pathology , Female , Glioma/drug therapy , Glioma/pathology , Humans , Hyperuricemia/chemically induced , Hyperuricemia/diagnosis , Male , Medulloblastoma/drug therapy , Medulloblastoma/pathology , Neuroblastoma/pathology , Neutropenia/chemically induced , Neutropenia/diagnosis , Phosphorylcholine/adverse effects , Phosphorylcholine/pharmacokinetics , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/pathology , Treatment Outcome , Wilms Tumor/drug therapy , Wilms Tumor/pathology
7.
J Child Neurol ; 31(12): 1341-53, 2016 10.
Article in English | MEDLINE | ID: mdl-26336203

ABSTRACT

Medulloblastoma accounts for nearly 10% of all childhood brain tumors. These tumors occur exclusively in the posterior fossa and have the potential for leptomeningeal spread. Treatment includes a combination of surgery, radiation therapy (in patients >3 years old). Patients >3 years old are stratified based on the volume of postoperative residual tumor and the presence or absence of metastases into "standard risk" and "high risk" categories with long-term survival rates of approximately 85% and 70%, respectively. Outcomes are inferior in infants and children younger than 3 years with exception of those patients with the medulloblastoma with extensive nodularity histologic subtype. Treatment for medulloblastoma is associated with significant morbidity, especially in the youngest patients. Recent molecular subclassification of medulloblastoma has potential prognostic and therapeutic implications. Future incorporation of molecular subgroups into treatment protocols will hopefully improve both survival outcomes and posttreatment quality of life.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Medulloblastoma/diagnosis , Medulloblastoma/therapy , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Child , Child, Preschool , Humans , Infant , Medulloblastoma/metabolism , Medulloblastoma/mortality
SELECTION OF CITATIONS
SEARCH DETAIL