Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Publication year range
1.
Pediatr Blood Cancer ; 68(9): e29065, 2021 09.
Article in English | MEDLINE | ID: mdl-33881209

ABSTRACT

BACKGROUND: Prexasertib (LY2606368) is a novel, second-generation, selective dual inhibitor of checkpoint kinase proteins 1 (CHK1) and 2 (CHK2). We conducted a phase 1 trial of prexasertib to estimate the maximum-tolerated dose (MTD) and/or recommended phase 2 dose (RP2D), to define and describe the toxicities, and to characterize the pharmacokinetics (PK) of prexasertib in pediatric patients with recurrent or refractory solid and central nervous system (CNS) tumors. METHODS: Prexasertib was administered intravenously (i.v.) on days 1 and 15 of a 28-day cycle. Four dose levels, 80, 100, 125, and 150 mg/m2 , were evaluated using a rolling-six design. PK analysis was performed during cycle 1. Tumor tissue was examined for biomarkers (CHK1 and TP53) of prexasertib activity. RESULTS: Thirty patients were enrolled; 25 were evaluable. The median age was 9.5 years (range: 2-20) and 21 (70%) were male. Twelve patients (40%) had solid tumors and 18 patients (60%) had CNS tumors. There were no cycle 1 or later dose-limiting toxicities. Common cycle 1, drug-related grade 3/4 toxicities (> 10% of patients) included neutropenia (100%), leukopenia (68%), thrombocytopenia (24%), lymphopenia (24%), and anemia (12%). There were no objective responses; best overall response was stable disease in three patients for five cycles (hepatocellular carcinoma), three cycles (ependymoma), and five cycles (undifferentiated sarcoma). The PK appeared dose proportional across the 80-150 mg/m2 dose range. CONCLUSIONS: Although the MTD of prexasertib was not defined by this study, 150 mg/m2 administered i.v. on days 1 and 15 of a 28-day cycle was determined to be the RP2D.


Subject(s)
Central Nervous System Neoplasms , Neoplasms , Protein Kinase Inhibitors/administration & dosage , Pyrazines/administration & dosage , Pyrazoles/administration & dosage , Adolescent , Central Nervous System Neoplasms/drug therapy , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 2/antagonists & inhibitors , Child , Child, Preschool , Female , Humans , Leukopenia , Male , Maximum Tolerated Dose , Neoplasm Recurrence, Local , Neoplasms/drug therapy , Neutropenia , Protein Kinase Inhibitors/pharmacokinetics , Pyrazines/pharmacokinetics , Pyrazoles/pharmacokinetics , Thrombocytopenia , Young Adult
2.
Support Care Cancer ; 29(3): 1549-1555, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32734390

ABSTRACT

PURPOSE: Two of the most common acute side effects of chemotherapy are nausea and vomiting. Nausea and vomiting impact quality of life, nutritional status, and ability to tolerate further chemotherapy. Parents of pediatric oncology patients rank nausea as one of the most bothersome treatment-related symptoms. METHODS: Utilizing Quality Improvement methodology, we developed a dashboard interface to facilitate extraction of data from the electronic medical record (EMR), which is presented via a visual display that summarizes the type of chemotherapy and antiemetic medications, use of as needed medications, and number of episodes of emesis. RESULTS: This dashboard interface allows for rapid and efficient identification of patients whose antiemetic regimen is mismatched for the emetogenicity of ordered chemotherapy, thus providing a timely opportunity to modify the antiemetic regimen based on published guidelines before administration of chemotherapy drugs. It also allows measurement of the effectiveness of the antiemetic regimen in terms of the number of break through emesis and the need for as needed medications. CONCLUSIONS: A novel CINV dashboard was created, which visually conveys complex information about antiemetics, chemotherapy emetogenicity, as needed medications, and breakthrough vomiting for inpatient pediatric oncology patients.


Subject(s)
Antiemetics/therapeutic use , Antineoplastic Agents/adverse effects , Nausea/chemically induced , Neoplasms/complications , Quality of Life/psychology , Vomiting/chemically induced , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Neoplasms/drug therapy , Young Adult
3.
Pediatr Blood Cancer ; 67(4): e28134, 2020 04.
Article in English | MEDLINE | ID: mdl-31876107

ABSTRACT

BACKGROUND/PURPOSE: To determine the maximum tolerated dose, toxicities, and response of sirolimus combined with oral metronomic therapy in pediatric patients with recurrent and refractory solid and brain tumors. PROCEDURE: Patients younger than 30 years of age with recurrent, refractory, or high-risk solid and brain tumors were eligible. Patients received six-week cycles of sirolimus with twice daily celecoxib, and alternating etoposide and cyclophosphamide every three weeks, with Bayesian dose escalation over four dose levels (NCT01331135). RESULTS: Eighteen patients were enrolled: four on dose level (DL) 1, four on DL2, eight on DL3, and two on DL4. Diagnoses included solid tumors (Ewing sarcoma, osteosarcoma, malignant peripheral nerve sheath tumor, rhabdoid tumor, retinoblastoma) and brain tumors (glioblastoma multiforme [GBM], diffuse intrinsic pontine glioma, high-grade glioma [HGG], medulloblastoma, ependymoma, anaplastic astrocytoma, low-grade infiltrative astrocytoma, primitive neuroectodermal tumor, nongerminomatous germ cell tumor]. One dose-limiting toxicity (DLT; grade 4 neutropenia) was observed on DL2, two DLTs (grade 3 abdominal pain and grade 3 mucositis) on DL3, and two DLTs (grade 3 dehydration and grade 3 mucositis) on DL4. The recommended phase II dose of sirolimus was 2 mg/m2 (DL3). Best response was stable disease (SD) in eight patients, and partial response (PR) in one patient with GBM. A patient with HGG was removed from the study with SD and developed PR without further therapy. Western blot analysis showed inhibition of phospho-S6 kinase in all patients during the first cycle of therapy. CONCLUSION: The combination of sirolimus with metronomic chemotherapy is well tolerated in children. A phase II trial of this combination is ongoing.


Subject(s)
Administration, Metronomic , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , Sirolimus/administration & dosage , Adolescent , Brain Neoplasms/drug therapy , Celecoxib/administration & dosage , Child , Child, Preschool , Cyclophosphamide/administration & dosage , Dose-Response Relationship, Drug , Etoposide/administration & dosage , Female , Humans , Male , Maximum Tolerated Dose , Young Adult
5.
Acta Neuropathol ; 137(4): 637-655, 2019 04.
Article in English | MEDLINE | ID: mdl-30770999

ABSTRACT

Histone H3 K27M mutation is the defining molecular feature of the devastating pediatric brain tumor, diffuse intrinsic pontine glioma (DIPG). The prevalence of histone H3 K27M mutations indicates a critical role in DIPGs, but the contribution of the mutation to disease pathogenesis remains unclear. We show that knockdown of this mutation in DIPG xenografts restores K27M-dependent loss of H3K27me3 and delays tumor growth. Comparisons of matched DIPG xenografts with and without K27M knockdown allowed identification of mutation-specific effects on the transcriptome and epigenome. The resulting transcriptional changes recapitulate expression signatures from K27M primary DIPG tumors and are strongly enriched for genes associated with nervous system development. Integrated analysis of ChIP-seq and expression data showed that genes upregulated by the mutation are overrepresented in apparently bivalent promoters. Many of these targets are associated with more immature differentiation states. Expression profiles indicate K27M knockdown decreases proliferation and increases differentiation within lineages represented in DIPG. These data suggest that K27M-mediated loss of H3K27me3 directly regulates a subset of genes by releasing poised promoters, and contributes to tumor phenotype and growth by limiting differentiation. The delayed tumor growth associated with knockdown of H3 K27M provides evidence that this highly recurrent mutation is a relevant therapeutic target.


Subject(s)
Brain Stem Neoplasms/genetics , Cell Differentiation/genetics , Diffuse Intrinsic Pontine Glioma/genetics , Histones/genetics , Mutation , Animals , Brain Stem Neoplasms/pathology , Cell Line, Tumor , Diffuse Intrinsic Pontine Glioma/pathology , Disease Models, Animal , Gene Knockdown Techniques , Mice
6.
BMC Cancer ; 19(1): 1251, 2019 Dec 27.
Article in English | MEDLINE | ID: mdl-31881855

ABSTRACT

BACKGROUND: Acute Myeloid Leukemia (AML) is a malignancy of myeloid precursor cells that arise from genomic alterations in the expression of key growth regulatory genes causing cells to assume an undifferentiated state and continue to proliferate. Recent efforts have focused on developing therapies that target specific protein products of aberrantly expressed genes. However, many of the identified proteins are difficult to target and thought to be "undrugable" because of structural challenges, protein overexpression, or mutations that confer resistance to therapy. A novel technology that circumvents some of these issues is the use of small molecules that stabilize secondary DNA structures present in the promoters of many potential oncogenes and modulate their transcription. METHODS: This study characterizes the in vitro activity of the G-quadruplex-stabilizing small molecule GQC-05 in AML cells. The effect of GQC-05 on three AML cell lines was analyzed using viability and apoptosis assays. GQC-05 has been shown to down-regulate MYC through G-quadruplex stabilization in Burkitt's lymphoma cell lines. MYC expression was evaluated through qPCR and immunoblotting in the three AML cell lines following the treatment of GQC-05. In order to identify other therapeutic agents that potentiate the activity of GQC-05, combination drug screening was performed. The drug combinations were validated using in vitro cytotoxicity assays and compared to other commonly used chemotherapeutic agents. RESULTS: GQC-05 treatment of KG-1a, CMK and TF-1 cells decreased cell viability and resulted in increased DNA damage and apoptosis. Additionally, treatment of KG-1a, CMK and TF-1 with GQC-05 resulted in decreased expression of MYC mRNA and protein, with a more pronounced effect in KG-1a cells. Combination drug screening identified the Bcl-2/Bcl-XL inhibitor Navitoclax as a compound that potentiated GQC-05 activity. Co-treatment with GQC-05 and Navitoclax showed a synergistic decrease in cell viability of AML cells as determined by Chou-Talalay analysis, and induced more DNA damage, apoptosis, and rapid cytotoxicity. The cytotoxicity induced by GQC-05 and Navitoclax was more potent than that of Navitoclax combined with either cytarabine or doxorubicin. CONCLUSION: These results suggest that the G-quadruplex stabilizing small molecule GQC-05 induces down regulated MYC expression and DNA damage in AML cells. Treatment with both GQC-05 with a Bcl-2/Bcl-XL inhibitor Navitoclax results in increased cytotoxic activity, which is more pronounced than Navitoclax or GQC-05 alone, and more significant than Navitoclax in combination with cytarabine and doxorubicin that are currently being used clinically.


Subject(s)
Aniline Compounds/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Ellipticines/pharmacology , G-Quadruplexes/drug effects , Leukemia, Myeloid, Acute/drug therapy , Sulfonamides/therapeutic use , Apoptosis , Cell Line, Tumor , DNA Damage , Ellipticines/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , Proto-Oncogene Proteins c-myc/genetics , Treatment Outcome
8.
J Neurooncol ; 123(1): 85-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25859842

ABSTRACT

Co-expression of ERBB2 and ERBB4, reported in 75% of pediatric ependymomas, correlates with worse overall survival. Lapatinib, a selective ERBB1 and ERBB2 inhibitor has produced prolonged disease stabilization in patients with ependymoma in a phase I study. Bevacizumab exposure in ependymoma xenografts leads to ablation of tumor self-renewing cells, arresting growth. Thus, we conducted an open-label, phase II study of bevacizumab and lapatinib in children with recurrent ependymomas. Patients ≤ 21 years of age with recurrent ependymoma received lapatinib orally twice daily (900 mg/m(2)/dose to the first 10 patients, and then 700 mg/m(2)/dose) and bevacizumab 10 mg/kg intravenously on days 1 and 15 of a 28-day course. Lapatinib serum trough levels were analyzed prior to each course. Total and phosphorylated VEGFR2 expression was measured in peripheral blood mononuclear cells (PBMCs) before doses 1 and 2 of bevacizumab and 24-48 h following dose 2 of bevacizumab. Twenty-four patients with a median age of 10 years (range 2-21 years) were enrolled; 22 were eligible and 20 evaluable for response. Thirteen had anaplastic ependymoma. There were no objective responses; 4 patients had stable disease for ≥ 4 courses (range 4-14). Grade 3 toxicities included rash, elevated ALT, and diarrhea. Grade 4 toxicities included peri-tracheostomy hemorrhage (n = 1) and elevated creatinine phosphokinase (n = 1). The median lapatinib pre-dose trough concentration was 3.72 µM. Although the combination of bevacizumab and lapatinib was well tolerated in children with recurrent ependymoma, it proved ineffective.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/drug effects , Ependymoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Salvage Therapy , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Bevacizumab/administration & dosage , Child , Child, Preschool , Ependymoma/pathology , Female , Follow-Up Studies , Humans , Lapatinib , Male , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Quinazolines/administration & dosage , Survival Rate , Tissue Distribution , Young Adult
9.
Pediatr Blood Cancer ; 62(2): 329-334, 2015 02.
Article in English | MEDLINE | ID: mdl-25327609

ABSTRACT

BACKGROUND: Female survivors of central nervous system (CNS) tumors are at an increased risk for gonadal damage and variations in the timing of puberty following radiotherapy and alkylating agent-based chemotherapy. PROCEDURE: Clinical and laboratory data were obtained from 30 evaluable female patients with newly diagnosed embryonal CNS tumors treated on a prospective protocol (SJMB 96) at St. Jude Children's Research Hospital (SJCRH). Pubertal development was evaluated by Tanner staging. Primary ovarian insufficiency (POI) was determined by Tanner staging and FSH level. Females with Tanner stage I-II and FSH > 15 mIU/ml, or Tanner stage III-V, FSH > 25 mIU/ml and FSH greater than LH were defined to have ovarian insufficiency. Recovery of ovarian function was defined as normalization of FSH without therapeutic intervention. RESULTS: Median length of follow-up post completion of therapy was 7.2 years (4.0-10.8 years). The cumulative incidence of pubertal onset was 75.6% by the age of 13. Precocious puberty was observed in 11.1% and delayed puberty in 11.8%. The cumulative incidence of POI was 82.8%, though recovery was observed in 38.5%. CONCLUSIONS: Treatment for primary CNS embryonal tumors may cause variations in the timing of pubertal development, impacting physical and psychosocial development. Female survivors are at risk for POI, a subset of whom will recover function over time. Further refinement of therapies is needed in order to reduce late ovarian insufficiency. Pediatr Blood Cancer 2015;62:329-334. © 2014 Wiley Periodicals, Inc.


Subject(s)
Brain Neoplasms/radiotherapy , Craniospinal Irradiation/adverse effects , Neoplasms, Germ Cell and Embryonal/radiotherapy , Primary Ovarian Insufficiency/diagnosis , Puberty, Delayed/diagnosis , Puberty, Precocious/diagnosis , Adolescent , Adult , Alkylating Agents/therapeutic use , Child , Child, Preschool , Female , Follicle Stimulating Hormone/blood , Humans , Young Adult
10.
Childs Nerv Syst ; 31(8): 1283-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25930724

ABSTRACT

PURPOSE: Amplification and high levels of NOTCH ligand expression have been identified in several types of pediatric brain tumors. A phase I trial of weekly MK-0752, an oral inhibitor of gamma-secretase, was conducted in children with recurrent central nervous system (CNS) malignancies to estimate the maximum tolerated dose, dose-limiting toxicities (DLT), pharmacokinetics (PK), and pharmacodynamics of weekly MK-0752. METHODS: MK-0752 was administered once weekly at 1000 and 1400 mg/m(2) using a rolling-6 design. PK analysis was performed during the first course. NOTCH and HES expression was assessed by immunohistochemistry and Western blot. RESULTS: Ten eligible patients were enrolled (median age 8.8 years; range 3.1-19.2) with diagnoses of brain stem glioma (n = 3), ependymoma (n = 2), anaplastic astrocytoma (n = 1), choroid plexus carcinoma (n = 2), medulloblastoma (n = 1), and primitive neuroectodermal tumor (n = 1). Nine were evaluable for toxicity. One DLT of fatigue occurred in the six evaluable patients enrolled at 1000 mg/m(2)/dose. No DLTs were experienced by three patients treated at 1400 mg/m(2)/dose. Non-dose-limiting grade 3 toxicities included lymphopenia, neutropenia, and anemia. Median number of treatment courses was 2 (range 1-10). Two patients continued on therapy for at least 6 months. The median (range) C(max) of MK-0752 was 88.2 µg/mL (40.6 to 109 µg/mL) and 60.3 µg/mL (59.2 to 91.9 µg/mL) in patients receiving 1000 and 1400 mg/m(2)/week, respectively. NOTCH expression was decreased in six of seven patients for whom tissue was available at 24 h post-MK-0752. CONCLUSION: MK-0752 is well tolerated and exhibits target inhibition at 1000 and 1400 mg/m(2)/week in children with recurrent CNS malignancies.


Subject(s)
Benzene Derivatives/therapeutic use , Brain Neoplasms/drug therapy , Central Nervous System Diseases/drug therapy , Enzyme Inhibitors/therapeutic use , Propionates/therapeutic use , Sulfones/therapeutic use , Administration, Oral , Adolescent , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Area Under Curve , Basic Helix-Loop-Helix Transcription Factors/metabolism , Benzene Derivatives/blood , Central Nervous System Diseases/blood , Child , Child, Preschool , Dose-Response Relationship, Drug , Enzyme Inhibitors/blood , Female , Follow-Up Studies , Gene Expression Regulation/drug effects , Homeodomain Proteins/metabolism , Humans , Male , Propionates/blood , Receptor, Notch1/metabolism , Repressor Proteins/metabolism , Sulfones/blood , Time Factors , Transcription Factor HES-1 , Young Adult
11.
J Biol Chem ; 288(17): 11786-94, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23482563

ABSTRACT

Although the biological role of KRAS is clearly established in carcinogenesis, the molecular mechanisms underlying this phenomenon are not completely understood. In this study, we provide evidence of a novel signaling network regulated by the transcription factor GLI1 mediating KRAS-induced carcinogenesis. Using pancreatic cancer (a disease with high prevalence of KRAS mutations) as a model, we show that loss of GLI1 blocks the progression of KRAS-induced pancreatic preneoplastic lesions in mice with pancreas-specific Cre-activated oncogenic mutant kras. Mice lacking GLI1 develop only low-grade lesions at low frequency, and in most cases, the pancreata are histologically normal. Further characterization of the phenotype showed a decrease in the activation of STAT3 in pancreatic preneoplastic lesions; STAT3 is a transcription factor required for the development of premalignant lesions and their progression into pancreatic cancer. Analysis of the mechanisms revealed a key role for GLI1 in maintaining the levels of activated STAT3 through the modulation of IL-6 signaling. GLI1 binds to the IL-6 mouse promoter and regulates the activity and expression of this cytokine. This newly identified GLI1/IL-6 axis is active in fibroblasts, a known source of IL-6 in the tumor microenvironment. Sonic hedgehog induces GLI1 binding to the IL-6 promoter and increases IL-6 expression in fibroblasts in a paracrine manner. Finally, we demonstrate that mutant KRAS initiates this cascade by inducing the expression of Sonic hedgehog in cancer cells. Collectively, these results define a novel role for GLI1 in carcinogenesis acting as a downstream effector of oncogenic KRAS in the tumor microenvironment.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/biosynthesis , Tumor Microenvironment , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Interleukin-6/biosynthesis , Interleukin-6/genetics , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Zinc Finger Protein GLI1
12.
Cancer ; 120(23): 3731-7, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25080363

ABSTRACT

BACKGROUND: Patients with recurrent medulloblastoma (MB) have a dismal prognosis. There has been a reluctance to use radiation in the salvage therapy regimens for these patients because of concerns about toxicity and unknown efficacy. Comparing survival outcomes and toxicities in relapsed patients treated with and without radiation may help to define its role. METHODS: A retrospective review was conducted that included 38 patients with recurrent MB treated with similar risk-adapted therapy at initial diagnosis; reirradiation was a component of salvage therapy in 14. Overall survival (OS) and toxicity were evaluated according to the use of radiation, prior risk stratification, and other factors. RESULTS: For relapsed standard-risk patients, the use of additional irradiation resulted in a statistically significant improvement in OS from initial diagnosis (P = .036), with 5- and 10-year OS rates of 55% ± 14% and 33% ± 16% versus 46% ± 14% and 0% for reirradiated patients versus others, respectively. Similar improvement was observed in high-risk patients (P = .003). There was an association between the use of additional irradiation and an increased rate of necrosis as determined by neuroimaging (P = .0468). CONCLUSIONS: The use of irradiation as a component of salvage therapy for relapsed MB may prolong survival. The benefit appears to be greatest for relapsed standard-risk patients.


Subject(s)
Medulloblastoma/radiotherapy , Neoplasm Recurrence, Local/radiotherapy , Adolescent , Child , Child, Preschool , Cohort Studies , Female , Humans , Male , Medulloblastoma/mortality , Retreatment/methods , Retrospective Studies , Risk Assessment , Salvage Therapy/methods , Treatment Outcome , Young Adult
13.
J Pediatr Psychol ; 39(4): 450-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24369366

ABSTRACT

OBJECTIVE: To investigate the feasibility of a computer-based reading intervention completed by patients diagnosed with a brain tumor. METHODS: Patients were randomized to the intervention (n = 43) or standard of care group (n = 38). The intervention consisted of 30 sessions using Fast ForWord® exercises in a game-like format. Change in reading decoding scores over time since diagnosis was examined. Gender, race, parent education, parent marital status, and age at diagnosis were examined as covariates. RESULTS: 17 patients (39.5%) were able to complete the target goal of 30 intervention sessions. Females had significantly greater training time than males (p = .022). Age at diagnosis was associated with average training time/session for females (r = .485, p = .041). No significant differences were found in reading scores between the randomized groups. CONCLUSIONS: The study was well accepted by families and adherence by patients undergoing radiation therapy for medulloblastoma was moderate. Suggestions for improved methodology are discussed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain Neoplasms/therapy , Cranial Irradiation/adverse effects , Dyslexia, Acquired/prevention & control , Medulloblastoma/therapy , Therapy, Computer-Assisted , Adolescent , Age Factors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/psychology , Brain Neoplasms/radiotherapy , Child , Child, Preschool , Combined Modality Therapy , Dyslexia, Acquired/etiology , Feasibility Studies , Female , Humans , Male , Medulloblastoma/drug therapy , Medulloblastoma/psychology , Medulloblastoma/radiotherapy , Quality of Life , Reading , Sex Factors , Treatment Outcome , Young Adult
14.
Neuro Oncol ; 25(2): 386-397, 2023 02 14.
Article in English | MEDLINE | ID: mdl-35652336

ABSTRACT

BACKGROUND: Recurrent atypical teratoid/rhabdoid tumor (AT/RT) is, most often, a fatal pediatric malignancy with limited curative options. METHODS: We conducted a phase II study of Aurora kinase A inhibitor alisertib in patients aged <22 years with recurrent AT/RT. Patients received alisertib once daily (80 mg/m2 as enteric-coated tablets or 60 mg/m2 as liquid formulation) on Days 1-7 of a 21-day cycle until progressive disease (PD) occurred. Alisertib plasma concentrations were measured in cycle 1 on Days 1 (single dose) and 7 (steady state) and analyzed with noncompartmental pharmacokinetics. Trial efficacy end point was ≥10 participants with stable disease (SD) or better at 12 weeks. RESULTS: SD (n = 8) and partial response (PR) (n = 1) were observed among 30 evaluable patients. Progression-free survival (PFS) was 30.0% ± 7.9% at 6 months and 13.3% ± 5.6% at 1 year. One-year overall survival (OS) was 36.7% ± 8.4%. Two patients continued treatment for >12 months. PFS did not differ by AT/RT molecular groups. Neutropenia was the most common adverse effect (n = 23/30, 77%). The 22 patients who received liquid formulation had a higher mean maximum concentration (Cmax) of 10.1 ± 3.0 µM and faster time to Cmax (Tmax = 1.2 ± 0.7 h) than those who received tablets (Cmax = 5.7 ± 2.4 µM, Tmax = 3.4 ± 1.4 h). CONCLUSIONS: Although the study did not meet predetermined efficacy end point, single-agent alisertib was well tolerated by children with recurrent AT/RT, and SD or PR was observed in approximately a third of the patients.


Subject(s)
Antineoplastic Agents , Central Nervous System Neoplasms , Rhabdoid Tumor , Child , Humans , Antineoplastic Agents/therapeutic use , Rhabdoid Tumor/drug therapy , Azepines/therapeutic use , Pyrimidines/therapeutic use , Central Nervous System Neoplasms/drug therapy , Aurora Kinase A , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/adverse effects
15.
16.
Acta Neuropathol Commun ; 10(1): 80, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35642016

ABSTRACT

The majority of diffuse midline gliomas, H3 K27-altered (DMG-H3 K27-a), are infiltrating pediatric brain tumors that arise in the pons with no effective treatment. To understand how clonal evolution contributes to the tumor's invasive spread, we performed exome sequencing and SNP array profiling on 49 multi-region autopsy samples from 11 patients with pontine DMG-H3 K27-a enrolled in a phase I clinical trial of PDGFR inhibitor crenolanib. For each patient, a phylogenetic tree was constructed by testing multiple possible clonal evolution models to select the one consistent with somatic mutations and copy number variations across all tumor regions. The tree was then used to deconvolute subclonal composition and prevalence at each tumor region to study convergent evolution and invasion patterns. Somatic variants in the PI3K pathway, a late event, are enriched in our cohort, affecting 70% of patients. Convergent evolution of PI3K at distinct phylogenetic branches was detected in 40% of the patients. 24 (~ 50%) of tumor regions were occupied by subclones of mixed lineages with varying molecular ages, indicating multiple waves of invasion across the pons and extrapontine. Subclones harboring a PDGFRA amplicon, including one that amplified a PDGRFAY849C mutant allele, were detected in four patients; their presence in extrapontine tumor and normal brain samples imply their involvement in extrapontine invasion. Our study expands the current knowledge on tumor invasion patterns in DMG-H3 K27-a, which may inform the design of future clinical trials.


Subject(s)
DNA Copy Number Variations , Glioma , Child , Glioma/drug therapy , Glioma/genetics , Glioma/pathology , Histones/genetics , Humans , Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , Phylogeny , Protein Kinase Inhibitors
17.
Pediatrics ; 150(4)2022 10 01.
Article in English | MEDLINE | ID: mdl-36180615

ABSTRACT

Bruising or bleeding in a child can raise the concern for child abuse. Assessing whether the findings are the result of trauma and/or whether the child has a bleeding disorder is critical. Many bleeding disorders are rare, and not every child with bruising/bleeding that may raise a concern for abuse requires an evaluation for bleeding disorders. However, in some instances, bleeding disorders can present in a manner similar to child abuse. Bleeding disorders cannot be ruled out solely on the basis of patient and family history, no matter how extensive. The history and clinical evaluation can be used to determine the necessity of an evaluation for a possible bleeding disorder, and prevalence and known clinical presentations of individual bleeding disorders can be used to guide the extent of laboratory testing. This clinical report provides guidance to pediatricians and other clinicians regarding the evaluation for bleeding disorders when child abuse is suspected.


Subject(s)
Blood Coagulation Disorders , Child Abuse , Contusions , Child , Child Abuse/diagnosis , Contusions/diagnosis , Contusions/etiology , Hemorrhage/diagnosis , Hemorrhage/etiology , Humans , Prevalence
18.
J Neurooncol ; 105(2): 191-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21499912

ABSTRACT

Fractionated cranial irradiation is an essential part of treatment in the management of cohorts of pediatric brain tumor and leukemia patients. Ionizing radiation damages normal brain parenchyma through a variety of poorly understood mechanisms and results in cognitive dysfunction and significant life-long disability. The goal of our study was to establish and characterize a mouse model of radiation-induced damage to the developing nervous system. Male C57BL/6 mice were exposed to a total dose of 20 Gy of fractionated cranial irradiation at 1 month of age to assess the early and late effects of clinically relevant irradiation doses on the young mouse brain. Compared to age-matched controls, an acute and prolonged decrease in proliferation and the number of immature neurons in the stem cell niche of the hippocampal subgranular zone within the dentate gyrus at 72 h and 1 month following cranial irradiation was noted. Behavioral characterization at 1 and 5 months post-radiation demonstrated significant, persistent and progressive hippocampus-dependent learning deficits. Our study characterizes a clinically relevant mouse model of radiation-induced damage that serves as a platform for future evaluation of therapeutic interventions that may mitigate such cognitive damage. Our research also emphasizes the need for targeted treatment strategies that protect regions of neurogenesis while maximizing therapeutic effects in pediatric cancer patients.


Subject(s)
Cell Proliferation/radiation effects , Cognition Disorders/etiology , Cranial Irradiation/adverse effects , Gamma Rays/adverse effects , Hippocampus/radiation effects , Maze Learning/radiation effects , Neurons/radiation effects , Animals , Cesium Radioisotopes , Cognition Disorders/pathology , Dose-Response Relationship, Radiation , Male , Mice , Mice, Inbred C57BL , Motor Activity/radiation effects , Neurons/cytology
19.
Pediatr Neurosurg ; 47(4): 279-83, 2011.
Article in English | MEDLINE | ID: mdl-22456272

ABSTRACT

A 5-year-old female presented to the emergency department with a 24-hour history of nausea, vomiting and mental status changes. Imaging demonstrated hemorrhage from a suprasellar mass consistent with an optic nerve glioma. The patient was taken to surgery for an open biopsy and hematoma evacuation. Pathology revealed a grade 3 fibrillary astrocytoma. She was subsequently treated with carboplatin and vincristine and is now 2 years out without evidence of recurrence. Apoplexy is a rare presentation for an optic pathway glioma. To the authors' knowledge, this is the first reported case in the pediatric population. Clinical presentation appears similar to the symptoms observed in pituitary apoplexy. Timely surgery should be directed at obtaining diagnostic tissue, prevention of long-term nervous system damage and evacuation of the hematoma where feasible.


Subject(s)
Astrocytoma/diagnosis , Astrocytoma/surgery , Optic Chiasm/surgery , Optic Nerve Neoplasms/diagnosis , Stroke/etiology , Astrocytoma/pathology , Child, Preschool , Female , Humans , Magnetic Resonance Imaging , Neurosurgery/methods , Optic Chiasm/pathology , Optic Chiasm/physiopathology , Optic Nerve Neoplasms/pathology , Optic Nerve Neoplasms/surgery , Tomography, X-Ray Computed
20.
Eur J Drug Metab Pharmacokinet ; 46(3): 343-352, 2021 May.
Article in English | MEDLINE | ID: mdl-33852135

ABSTRACT

BACKGROUND AND OBJECTIVE: Population pharmacokinetic analysis explored the pharmacokinetics of sunitinib and its primary active metabolite, SU012662, in children and evaluated the sunitinib dose(s) that produce comparable plasma exposures to adults receiving the approved daily dose. METHODS: Data were from 65 children with gastrointestinal stromal tumors (GIST) or solid tumors. Pharmacokinetic models of sunitinib and SU012662 were developed using a systematic multi-step approach employing nonlinear mixed-effects modeling. The effect of predefined covariates on pharmacokinetic parameters was assessed. Final models were validated using visual predictive check and statistical techniques. RESULTS: The final dataset comprised 439 sunitinib and 417 SU012662 post-baseline plasma observations. Base models were characterized by two-compartment models with first-order absorption and lag time. Body surface area (BSA) was the only covariate that affected (P < 0.001) pharmacokinetic parameters for sunitinib and SU012662 and was incorporated into the final models. Bootstrap results indicated that the final models represented the final dataset adequately. Based on the final models, a sunitinib dose of ~ 20mg/m2/day in children with GIST aged 6-17 years would be expected to lead to similar total plasma exposures of sunitinib and SU012661 as a dose of 50 mg/day in an adult with GIST on schedule 4/2. CONCLUSIONS: In children with GIST or solid tumors receiving sunitinib, population pharmacokinetic analysis identified BSA as the only covariate that affected pharmacokinetic parameters and predicted a dose of ~ 20 mg/m2/day as achieving equivalent exposure to 50 mg/day in adults with GIST on schedule 4/2. TRIAL REGISTRATION: ClinicalTrials.gov identifiers (date registered): NCT01396148 (July 2011); NCT01462695 (October 2011); NCT00387920 (October 2006).


Subject(s)
Gastrointestinal Stromal Tumors/drug therapy , Models, Biological , Neoplasms/drug therapy , Sunitinib/pharmacokinetics , Adolescent , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/pathology , Humans , Indoles/pharmacokinetics , Infant , Male , Pyrroles/pharmacokinetics , Sunitinib/administration & dosage , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL