Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Neurooncol ; 165(1): 161-169, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37878192

ABSTRACT

BACKGROUND: Neurocognitive deficits are common in pediatric brain tumor survivors. The use of single nucleotide polymorphism (SNP) analysis in DNA repair genes may identify children treated with radiation therapy for brain tumors at increased risk for treatment toxicity and adverse neurocognitive outcomes. MATERIALS: The Human 660W-Quad v1.0 DNA BeadChip analysis (Illumina) was used to evaluate 1048 SNPs from 59 DNA repair genes in 46 subjects. IQ testing was measured by the Wechsler Intelligence Scale for Children. Linear regression was used to identify the 10 SNPs with the strongest association with IQ scores while adjusting for radiation type. RESULTS: The low vs high IQ patient cohorts were well matched for time from first treatment to most recent IQ, first treatment age, sex, and treatments received. 5 SNPs on 3 different genes (CYP29, XRCC1, and BRCA1) and on 3 different chromosomes (10, 19, and 17) had the strongest association with most recent IQ score that was not modified by radiation type. Furthermore, 5 SNPs on 4 different genes (WRN, NR3C1, ERCC4, RAD51L1) on 4 different chromosomes (8, 5, 16, 14) had the strongest association with change in IQ independent of radiation type, first IQ, and years between IQ measures. CONCLUSIONS: SNPs offer the potential to predict adverse neurocognitive outcomes in pediatric brain tumor survivors. Our results require validation in a larger patient cohort. Improving the ability to identify children at risk of treatment related neurocognitive deficits could allow for better treatment stratification and early cognitive interventions.


Subject(s)
Brain Neoplasms , Child , Humans , Brain Neoplasms/complications , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , Intelligence Tests , Survivors , Cranial Irradiation/adverse effects , Neuropsychological Tests , X-ray Repair Cross Complementing Protein 1
2.
Adv Radiat Oncol ; 7(4): 100945, 2022.
Article in English | MEDLINE | ID: mdl-35814855

ABSTRACT

Purpose: : The role of peri-transplant radiation therapy (RT) in children with primary brain tumors is unclear. We characterized our institutional practice patterns and patient outcomes. Methods and Materials: The cohort included all patients treated with high-dose chemotherapy and autologous stem cell transplant for primary brain tumors at our institution from 2011 to 2017. Rates of local control, progression-free survival, overall survival, and radiation-associated injury were assessed. Results: Of the 37 eligible patients, 29 (78%) received peri-transplant RT. Patients treated with RT were more likely to have metastatic (P = .0121) and incompletely resected (P = .056) disease. Of those treated with RT, 13 (45%) received craniospinal irradiation (CSI) and 16 (55%) received focal RT. The median CSI dose was 23.4 Gy (interquartile range [IQR], 18-36 Gy; boost: median, 54 Gy [IQR, 53.7-55.8 Gy]) and focal RT dose was 50.4 Gy [IQR, 50.4-54.5 Gy]). Compared with the focal RT group, patients treated with CSI were older (P = .0499) and more likely to have metastatic disease (P = .0004). For the complete cohort, 2-year local control was 82% (95% confidence interval [CI], 70%-96%), progression-free survival 63% (95% CI, 49%-81%), and overall survival 65% (95% CI, 51%-82%). These rates did not differ significantly between patients treated with and without peri-transplant RT. Two cases of fatal myelopathy were observed after spinal cord doses within the highest tertile (41.4 cobalt Gy equivalent and 36 Gy). Conclusions: Peri-transplant RT was used for high-risk disease. Oncologic outcomes after RT were encouraging. However, 2 cases of grade 5 myelopathy were observed. If used cautiously, RT may contribute to durable remission in patients at high risk of relapse.

3.
Pediatr Blood Cancer ; 68(1): e28705, 2021 01.
Article in English | MEDLINE | ID: mdl-32985070

ABSTRACT

Radiation necrosis is a potentially debilitating side effect of therapy necessary to treat pediatric central nervous system tumors. Clinical signs of cerebral radiation necrosis (CRN) are similar to symptoms of disease progression and require close monitoring. The case of an infant diagnosed with a malignant rhabdoid tumor is presented to describe the medical and rehabilitation interventions implemented to address CRN. Rehabilitation providers should routinely be consulted in children with CRN as they fill a critical role in treatment, neurological symptom monitoring, and intervention planning to address family-centered functional goals.


Subject(s)
Central Nervous System Neoplasms/therapy , Chemoradiotherapy/adverse effects , Radiation Injuries/rehabilitation , Central Nervous System Neoplasms/pathology , Female , Humans , Infant , Prognosis , Radiation Injuries/etiology , Radiation Injuries/pathology
4.
Clin Case Rep Rev ; 6(2)2020 Jun.
Article in English | MEDLINE | ID: mdl-32999736

ABSTRACT

Gangliogliomas are predominantly low-grade primary brain tumors comprised of neuronal and glial components that are found in both pediatric and young adult populations. In the majority of cases, surgical resection of these tumors is curative. However, tumor location in eloquent centers of the brain can make surgical intervention inappropriate. Additionally, a subset of tumors progress to anaplastic ganglioglioma which carries a poor prognosis, despite resection. Activating mutations in the MAPK pathway, such as BRAF V600E, have been identified in many of these tumors. Tumors carrying such mutations have demonstrated susceptibility to MEK inhibition therapy. However, there remains a subset of ganglioglioma that do not contain a known mutation in the MAPK pathway and thus have not been targeted with MEK inhibition therapy. Here, we present a young adult ganglioglioma patient without identified MAPK pathway activation mutations who demonstrated a significant and sustained response to MEK inhibition with trametinib.

5.
Oncogene ; 39(12): 2641, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31969682

ABSTRACT

The original version of this Article omitted the following from the Acknowledgements: This work was supported by the Luke's Army Pediatric Cancer Research Fund St. Baldrick's Scholar Award. This has now been corrected in both the PDF and HTML versions of the Article.

6.
Oncogene ; 39(11): 2305-2327, 2020 03.
Article in English | MEDLINE | ID: mdl-31844250

ABSTRACT

High-grade gliomas (HGG) afflict both children and adults and respond poorly to current therapies. Epigenetic regulators have a role in gliomagenesis, but a broad, functional investigation of the impact and role of specific epigenetic targets has not been undertaken. Using a two-step, in vitro/in vivo epigenomic shRNA inhibition screen, we determine the chromatin remodeler BPTF to be a key regulator of adult HGG growth. We then demonstrate that BPTF knockdown decreases HGG growth in multiple pediatric HGG models as well. BPTF appears to regulate tumor growth through cell self-renewal maintenance, and BPTF knockdown leads these glial tumors toward more neuronal characteristics. BPTF's impact on growth is mediated through positive effects on expression of MYC and MYC pathway targets. HDAC inhibitors synergize with BPTF knockdown against HGG growth. BPTF inhibition is a promising strategy to combat HGG through epigenetic regulation of the MYC oncogenic pathway.

7.
Mol Cancer Ther ; 17(9): 1984-1994, 2018 09.
Article in English | MEDLINE | ID: mdl-29925527

ABSTRACT

Children with ependymoma (EPN) are cured in less than 50% of cases, with little improvement in outcome over the last several decades. Chemotherapy has not affected survival in EPN, due in part to a lack of preclinical models that has precluded comprehensive drug testing. We recently developed two human EPN cell lines harboring high-risk phenotypes which provided us with an opportunity to execute translational studies. EPN and other pediatric brain tumor cell lines were subject to a large-scale comparative drug screen of FDA-approved oncology drugs for rapid clinical application. The results of this in vitro study were combined with in silico prediction of drug sensitivity to identify EPN-selective compounds, which were validated by dose curve and time course modeling. Mechanisms of EPN-selective antitumor effect were further investigated using transcriptome and proteome analyses. We identified three classes of oncology drugs that showed EPN-selective antitumor effect, namely, (i) fluorinated pyrimidines (5-fluorouracil, carmofur, and floxuridine), (ii) retinoids (bexarotene, tretinoin and isotretinoin), and (iii) a subset of small-molecule multireceptor tyrosine kinase inhibitors (axitinib, imatinib, and pazopanib). Axitinib's antitumor mechanism in EPN cell lines involved inhibition of PDGFRα and PDGFRß and was associated with reduced mitosis-related gene expression and cellular senescence. The clinically available, EPN-selective oncology drugs identified by our study have the potential to critically inform design of upcoming clinical studies in EPN, in particular for those children with recurrent EPN who are in the greatest need of novel therapeutic approaches. Mol Cancer Ther; 17(9); 1984-94. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/genetics , Drug Screening Assays, Antitumor/methods , Ependymoma/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Child , Computer Simulation , Drug Approval , Ependymoma/drug therapy , Ependymoma/pathology , Humans , Risk Factors
8.
Pediatr Blood Cancer ; 65(5): e26960, 2018 05.
Article in English | MEDLINE | ID: mdl-29350470

ABSTRACT

BACKGROUND: A desperate need for novel therapies in pediatric ependymoma (EPN) exists, as chemotherapy remains ineffective and radiotherapy often fails. EPN have significant infiltration of immune cells, which correlates with outcome. Immune checkpoint inhibitors provide an avenue for new treatments. This study characterizes tumor-infiltrating immune cells in EPN and aims at predicting candidates for clinical trials using checkpoint inhibitors targeting PD-L1/PD-1 (programmed death ligand 1/programmed death 1). METHODS: The transcriptomic profiles of the primary study cohort of EPN and other pediatric brain tumors were interrogated to identify PD-L1 expression levels. Transcriptomic findings were validated using the western blotting, immunohistochemistry and flow cytometry. RESULTS: We evaluated PD-L1 mRNA expression across four intracranial subtypes of EPN in two independent cohorts and found supratentorial RELA fusion (ST-RELA) tumors to have significantly higher levels. There was a correlation between high gene expression and protein PD-L1 levels in ST-RELA tumors by both the western blot and immunohistochemisty. The investigation of EPN cell populations revealed PD-L1 was expressed on both tumor and myeloid cells in ST-RELA. Other subtypes had little PD-L1 in either tumor or myeloid cell compartments. Lastly, we measured PD-1 levels on tumor-infiltrating T cells and found ST-RELA tumors express PD-1 in both CD4 and CD8 T cells. A functional T-cell exhaustion assay found ST-RELA T cells to be exhausted and unable to secrete IFNγ on stimulation. CONCLUSIONS: These findings in ST-RELA suggest tumor evasion and immunsuppression due to PD-L1/PD-1-mediated T-cell exhaustion. Trials of checkpoint inhibitors in EPN should be enriched for ST-RELA tumors.


Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Ependymoma/metabolism , Supratentorial Neoplasms/metabolism , Transcription Factor RelA/metabolism , Adolescent , Adult , B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Child , Child, Preschool , Cohort Studies , Ependymoma/genetics , Ependymoma/pathology , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Infant , Male , Molecular Targeted Therapy , Prognosis , Supratentorial Neoplasms/genetics , Supratentorial Neoplasms/pathology , T-Lymphocytes/metabolism , Transcription Factor RelA/genetics , Young Adult
9.
Mol Diagn Ther ; 22(1): 25-39, 2018 02.
Article in English | MEDLINE | ID: mdl-28900876

ABSTRACT

Pediatric brain tumors are the primary cause of cancer-related death during childhood. Unfortunately, the number of primary and metastatic brain tumors is steadily increasing while the mortality rates for many central nervous system (CNS) lesions have remained stagnant. Molecularly defined tumor classes have been added to the most recent 2016 World Health Organization (WHO) Classification System of Central Nervous System Brain Tumors, driving potential new treatments and identifying targets to improve survival for these patients. Focusing on the genetic mutations most commonly seen in the pediatric CNS tumor population provides the ability to better define tumors based on shared molecular characteristics. Consequently, there is the potential for greater efficacy in targeted therapy to treat these identified genetic aberrations. Understanding the growing importance of molecular diagnosis in pediatric CNS tumors is vital to successfully using novel targeted therapies and improving patient outcomes.


Subject(s)
Brain Neoplasms/diagnosis , Gene Regulatory Networks , Brain Neoplasms/genetics , Child , Genetic Predisposition to Disease , Humans , Mutation , Oncogene Fusion
10.
Oncol Rep ; 39(2): 455-464, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29207163

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is an incurable childhood brain tumor. The mechanistic target of rapamycin (MTOR), a key oncogene, functions as two distinct signaling complexes, MTORC1 and MTORC2. We set out to determine the preclinical efficacy and mechanism of action of MTOR inhibitors in DIPG. We evaluated the MTORC1 inhibitor everolimus and the MTORC1/2 inhibitor AZD2014 in three patient-derived DIPG cell lines using cell culture models. We created dose-response curves for both compounds. We measured phenotypic effects on cell self-renewal, apoptosis, cell cycle, differentiation, senescence, and autophagy. We assessed the effects of each compound on the AKT pathway. Finally, we measured the efficacy of AZD2014 in combination with radiation therapy (RT) and a panel of FDA-approved chemotherapy drugs. While everolimus showed minimal antitumor efficacy, AZD2014 revealed IC50 levels of 410-552 nM and IC90 levels of 1.30-8.86 µM in the three cell lines. AZD2014 demonstrated increased inhibition of cell self-renewal compared to everolimus. AZD2014 decreased expression of phospho-AKT, while no such effect was noted with everolimus. Direct AKT inhibition showed similar efficacy to AZD2014, and induction of constitutive AKT activity rescued DIPG cells from the effects of AZD2014. AZD2014 exhibited synergistic relationships with both RT and various chemotherapy agents across classes, including the multikinase inhibitor ponatinib. MTORC1/2 inhibition shows antitumor activity in cell culture models of DIPG due to the effect of MTORC2 inhibition on AKT. This strategy should be further assessed for potential incorporation into combinatorial approaches to the treatment of DIPG.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Stem Neoplasms/metabolism , Everolimus/pharmacology , Glioma/metabolism , Morpholines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Benzamides , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/radiotherapy , Cell Culture Techniques , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Glioma/drug therapy , Glioma/genetics , Glioma/radiotherapy , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Phosphorylation/drug effects , Phosphorylation/radiation effects , Pyrimidines
11.
Nat Rev Cancer ; 17(9): 528-542, 2017 09.
Article in English | MEDLINE | ID: mdl-28751651

ABSTRACT

Autophagy is a mechanism by which cellular material is delivered to lysosomes for degradation, leading to the basal turnover of cell components and providing energy and macromolecular precursors. Autophagy has opposing, context-dependent roles in cancer, and interventions to both stimulate and inhibit autophagy have been proposed as cancer therapies. This has led to the therapeutic targeting of autophagy in cancer to be sometimes viewed as controversial. In this Review, we suggest a way forwards for the effective targeting of autophagy by understanding the context-dependent roles of autophagy and by capitalizing on modern approaches to clinical trial design.


Subject(s)
Autophagy/drug effects , Chloroquine/therapeutic use , Hydroxychloroquine/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Autophagy/genetics , Biomarkers, Tumor/metabolism , Clinical Trials as Topic , Humans , Microtubule-Associated Proteins/metabolism , Sequestosome-1 Protein/metabolism , Tumor Escape
12.
BMC Cancer ; 16: 647, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27538997

ABSTRACT

BACKGROUND: Diffuse intrinsic pontine gliomas (DIPGs) are highly aggressive, fatal, childhood tumors that arise in the brainstem. DIPGs have no effective treatment, and their location and diffuse nature render them inoperable. Radiation therapy remains the only standard of care for this devastating disease. New therapeutic targets are needed to develop novel therapy for DIPG. METHODS: We examined the expression of PLK1 mRNA in DIPG tumor samples through microarray analysis and found it to be up regulated versus normal pons. Using the DIPG tumor cells, we inhibited PLK1 using a clinically relevant specific inhibitor BI 6727 and evaluated the effects on, proliferation, apoptosis, induction of DNA damage and radio sensitization of the DIPG tumor cells. RESULTS: Treatment of DIPG cell lines with BI 6727, a new generation, highly selective inhibitor of PLK1, resulted in decreased cell proliferation and a marked increase in cellular apoptosis. Cell cycle analysis showed a significant arrest in G2-M phase and a substantial increase in cell death. Treatment also resulted in an increased γH2AX expression, indicating induction of DNA damage. PLK1 inhibition resulted in radiosensitization of DIPG cells. CONCLUSION: These findings suggest that targeting PLK1 with small-molecule inhibitors, in combination with radiation therapy, will hold a novel strategy in the treatment of DIPG that warrants further investigation.


Subject(s)
Brain Neoplasms/genetics , Cell Cycle Proteins/genetics , Glioma/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Pteridines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oligonucleotide Array Sequence Analysis/methods , Sequence Analysis, DNA/methods , Up-Regulation/drug effects , Polo-Like Kinase 1
13.
Cell Res ; 25(6): 707-25, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25952668

ABSTRACT

Receptor-interacting protein kinase-3 (RIP3 or RIPK3) is an essential part of the cellular machinery that executes "programmed" or "regulated" necrosis. Here we show that programmed necrosis is activated in response to many chemotherapeutic agents and contributes to chemotherapy-induced cell death. However, we show that RIP3 expression is often silenced in cancer cells due to genomic methylation near its transcriptional start site, thus RIP3-dependent activation of MLKL and downstream programmed necrosis during chemotherapeutic death is largely repressed. Nevertheless, treatment with hypomethylating agents restores RIP3 expression, and thereby promotes sensitivity to chemotherapeutics in a RIP3-dependent manner. RIP3 expression is reduced in tumors compared to normal tissue in 85% of breast cancer patients, suggesting that RIP3 deficiency is positively selected during tumor growth/development. Since hypomethylating agents are reasonably well-tolerated in patients, we propose that RIP3-deficient cancer patients may benefit from receiving hypomethylating agents to induce RIP3 expression prior to treatment with conventional chemotherapeutics.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , DNA Methylation , Necrosis/drug therapy , Necrosis/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Animals , Breast Neoplasms/genetics , Cell Survival/drug effects , DNA Methylation/drug effects , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Necrosis/genetics , Necrosis/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Structure-Activity Relationship , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Cancer Discov ; 4(7): 773-80, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24823863

ABSTRACT

UNLABELLED: Autophagy inhibition is a potential therapeutic strategy in cancer, but it is unknown which tumors will benefit. The BRAF(V600E) mutation has been identified as important in pediatric central nervous system (CNS) tumors and is known to affect autophagy in other tumor types. We evaluated CNS tumor cells with BRAF(V600E) and found that mutant (but not wild-type) cells display high rates of induced autophagy, are sensitive to pharmacologic and genetic autophagy inhibition, and display synergy when the clinically used autophagy inhibitor chloroquine was combined with the RAF inhibitor vemurafenib or standard chemotherapeutics. Importantly, we also demonstrate that chloroquine can improve vemurafenib sensitivity in a resistant ex vivo primary culture and provide the first demonstration in a patient harboring the V600E mutation treated with vemurafenib that the addition of chloroquine can improve clinical outcomes. These findings suggest that CNS tumors with BRAF(V600E) are autophagy-dependent and should be targeted with autophagy inhibition in combination with other therapeutic strategies. SIGNIFICANCE: Autophagy inhibition may improve cancer therapy, but it is unclear which tumors will benefit. We found that BRAF mutations cause brain tumor cells to depend on autophagy and display selective chemosensitization with autophagy inhibition. We present a pediatric case in which deliberate autophagy inhibition halted tumor growth and overcame acquired BRAF-inhibition resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Brain Neoplasms/drug therapy , Central Nervous System/drug effects , Chloroquine/pharmacology , Indoles/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/pharmacology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Central Nervous System/pathology , Child , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Mutation , Vemurafenib
15.
Brain Pathol ; 24(2): 173-83, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24238153

ABSTRACT

Brainstem gangliogliomas (GGs), often cannot be resected, have a much poorer prognosis than those located in more common supratentorial sites and may benefit from novel therapeutic approaches. Therapeutically targetable BRAF c.1799T>A (p.V600E) (BRAF(V600E) ) mutations are harbored in roughly 50% of collective GGs taken from all anatomical sites. Large numbers of pediatric brainstem GGs, however, have not been specifically assessed and anatomic-and age-restricted assessment of genetic and biological factors are becoming increasingly important. Pediatric brainstem GGs (n = 13), non-brainstem GGs (n = 11) and brainstem pilocytic astrocytomas (PAs) (n = 8) were screened by standard Sanger DNA sequencing of BRAF exon 15. Five of 13 (38%) pediatric GG harbored a definitive BRAF(V600E) mutation, with two others exhibiting an equivocal result by this method. BRAF(V600E) was also seen in five of 11 (45%) non-brainstem GGs and one of eight (13%) brainstem PAs. VE1 immunostaining for BRAF(V600E) showed concordance with sequencing in nine of nine brainstem GGs including the two cases equivocal by Sanger. The equivocal brainstem GGs were subsequently shown to harbor BRAF(V600E) using a novel, more sensitive, RNA-sequencing approach, yielding a final BRAF(V600E) mutation frequency of 54% (seven of 13) in brainstem GGs. BRAF(V600E) -targeted therapeutics should be a consideration for the high percentage of pediatric brainstem GGs refractory to conventional therapies.


Subject(s)
Brain Stem Neoplasms/genetics , Ganglioglioma/genetics , Proto-Oncogene Proteins B-raf/genetics , Adolescent , Brain Stem Neoplasms/pathology , Child , Child, Preschool , Exons , Female , Ganglioglioma/pathology , Humans , Infant , Infant, Newborn , Male , Mutation , Retrospective Studies , Young Adult
16.
J Neurooncol ; 106(2): 281-90, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21842312

ABSTRACT

Brain and spinal tumors are the second most common malignancies in childhood after leukemia, and they remain the leading cause of death from childhood cancer. Autophagy is a catabolic cellular process that is thought to regulate chemosensitivity, however its role in pediatric tumors is unknown. Here we present studies in pediatric medulloblastoma cell lines (DAOY, ONS76) and atypical teratoid/rhabdoid tumor cell lines (BT-16, BT-12) to test this role. Autophagy was inhibited using siRNA against autophagy-related genes ATG12 and ATG7 or pharmacologically induced or inhibited using rapamycin and chloroquine to test the effect of autophagy on chemosensitivity. Autophagic flux was measured using Western blot analysis of LC3-II and p62 and cell viability was determined using MTS assays and clonogenic growth. We found that when pediatric brain tumor cells under starvation stress, exposed to known autophagy inducers such as rapamycin, or treated with current chemotherapeutics (lomustine, cisplatin), all stimulate autophagy. Silencing ATG12 and ATG7 or exposure to a known autophagy inhibitor, chloroquine, could inhibit this autophagy increase; however, the effect of autophagy on tumor cell killing was small. These results may have clinical relevance in the future planning of therapeutic regimens for pediatric brain tumors.


Subject(s)
Autophagy/drug effects , Autophagy/physiology , Brain Neoplasms/pathology , Drug Resistance, Neoplasm/physiology , Antineoplastic Agents/pharmacology , Autophagy-Related Protein 12 , Autophagy-Related Protein 7 , Blotting, Western , Brain Neoplasms/physiopathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Child , Humans , RNA, Small Interfering , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Transfection , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism
17.
Pharmacol Ther ; 131(1): 130-41, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21440002

ABSTRACT

Autophagy is a catabolic process that turns over long-lived proteins and organelles and contributes to cell and organism survival in times of stress. Current cancer therapies including chemotherapy and radiation are known to induce autophagy within tumor cells. This is therefore an attractive process to target during cancer therapy as there are safe, clinically available drugs known to both inhibit and stimulate autophagy. However, there are conflicting positive and negative effects of autophagy and no current consensus on how to manipulate autophagy to improve clinical outcomes. Careful and rigorous evaluation of autophagy with a focus on how to translate laboratory findings into relevant clinical therapies remains an important aspect of improving clinical outcomes in patients with malignant disease.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Humans , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...