Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Pediatr Blood Cancer ; 68(6): e28983, 2021 06.
Article in English | MEDLINE | ID: mdl-33719183

ABSTRACT

Larger clinical trial enrollments and a greater understanding of biological heterogeneity have led to improved survival rates for children diagnosed with brain tumors in the last 50 years. However, reducing long-term morbidities and improving survival rates of high-risk tumors remain major challenges. Chemotherapy can reduce tumor burden, but effective drug penetration at the tumor site is limited by barriers in the route of drug administration and within the tumor microenvironment. Bioavailability of drugs is impeded by the blood-brain barrier, plasma protein binding, and structural components by the tumor including the matrix and vasculature contributing to increased interstitial fluid pressure, hypoxia, and acidity. Designing drug delivery systems to circumvent these barriers could lead to improved drug penetration at the tumor site and reduce adverse systemic side effects. In this review, we expand on how systemic and local barriers limit drug penetration and present potential methods to enhance drug penetration in pediatric brain tumors.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Drug Delivery Systems/methods , Biological Availability , Blood-Brain Barrier/physiology , Brain Neoplasms/mortality , Humans , Survival Rate , Tumor Microenvironment/drug effects
2.
J Pediatr Hematol Oncol ; 35(3): e123-6, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23425999

ABSTRACT

Prognosis for children with glioblastoma is unacceptably poor. Modest improvements in progression-free survival were seen in adults with glioblastoma by combining temozolomide and bevacizumab with conformal radiation. We retrospectively reviewed 3 cases of glioblastoma in children treated using upfront bevacizumab and temozolomide during radiation, followed by 12 cycles of maintenance therapy. All patients completed therapy with minimal toxicity and no delays in treatment. Two patients remain disease free at 38 and 49 months from diagnosis. One patient recurred 14 months off therapy and currently receives salvage therapy 48 months from diagnosis. These results support further investigation of this regimen.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/therapy , Chemoradiotherapy , Glioblastoma/therapy , Adolescent , Antibodies, Monoclonal, Humanized/administration & dosage , Bevacizumab , Brain Neoplasms/pathology , Child , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Female , Glioblastoma/pathology , Humans , Magnetic Resonance Imaging , Male , Prognosis , Retrospective Studies , Temozolomide
3.
BMC Cancer ; 11: 136, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21492457

ABSTRACT

BACKGROUND: Medulloblastoma is a highly malignant pediatric brain tumor that requires surgery, whole brain and spine irradiation, and intense chemotherapy for treatment. A more sophisticated understanding of the pathophysiology of medulloblastoma is needed to successfully reduce the intensity of treatment and improve outcomes. Nuclear factor kappa-B (NFκB) is a signaling pathway that controls transcriptional activation of genes important for tight regulation of many cellular processes and is aberrantly expressed in many types of cancer. METHODS: To test the importance of NFκB to medulloblastoma cell growth, the effects of multiple drugs that inhibit NFκB, pyrrolidine dithiocarbamate, diethyldithiocarbamate, sulfasalazine, curcumin and bortezomib, were studied in medulloblastoma cell lines compared to a malignant glioma cell line and normal neurons. Expression of endogenous NFκB was investigated in cultured cells, xenograft flank tumors, and primary human tumor samples. A dominant negative construct for the endogenous inhibitor of NFκB, IκB, was prepared from medulloblastoma cell lines and flank tumors were established to allow specific pathway inhibition. RESULTS: We report high constitutive activity of the canonical NFκB pathway, as seen by Western analysis of the NFκB subunit p65, in medulloblastoma tumors compared to normal brain. The p65 subunit of NFκB is extremely highly expressed in xenograft tumors from human medulloblastoma cell lines; though, conversely, the same cells in culture have minimal expression without specific stimulation. We demonstrate that pharmacological inhibition of NFκB in cell lines halts proliferation and leads to apoptosis. We show by immunohistochemical stain that phosphorylated p65 is found in the majority of primary tumor cells examined. Finally, expression of a dominant negative form of the endogenous inhibitor of NFκB, dnIκB, resulted in poor xenograft tumor growth, with average tumor volumes 40% smaller than controls. CONCLUSIONS: These data collectively demonstrate that NFκB signaling is important for medulloblastoma tumor growth, and that inhibition can reduce tumor size and viability in vivo. We discuss the implications of NFκB signaling on the approach to managing patients with medulloblastoma in order to improve clinical outcomes.


Subject(s)
Cerebellar Neoplasms/metabolism , I-kappa B Proteins/metabolism , Medulloblastoma/metabolism , NF-kappa B/metabolism , Signal Transduction , Animals , Boronic Acids/pharmacology , Bortezomib , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , Cell Line, Tumor , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/physiopathology , Curcumin/pharmacology , Ditiocarb/pharmacology , Humans , I-kappa B Proteins/genetics , Medulloblastoma/genetics , Medulloblastoma/pathology , Medulloblastoma/physiopathology , Mice , Mice, Inbred C57BL , Mice, Nude , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Pyrazines/pharmacology , Pyrrolidines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfasalazine/pharmacology , Thiocarbamates/pharmacology , Transgenes/genetics , Tumor Burden/genetics , Xenograft Model Antitumor Assays
4.
J Membr Biol ; 237(2-3): 59-69, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20931182

ABSTRACT

Medulloblastoma is a pediatric high-grade cerebellar malignancy derived from neuronal precursors. Although electrophysiologic characteristics of cerebellar granule neurons at all stages of cell development have been well described, such characterization has not been reported for medulloblastoma. In this study we attempt to characterize important electrophysiologic features of medulloblastoma that may distinguish it from the surrounding cerebellum. Using patient-derived cell lines and tumor tissues, we show that medulloblastoma cells have no inward Na+ current or transient K+ current involved in action potential generation and propagation, typically seen in granule neurons. Expression and function of calcium-activated, large-conductance K+ channels are diminished in medulloblastoma, judged by electrophysiology and Western analysis. The resting membrane potential of medulloblastoma cells in culture is quite depolarized compared to granule neurons. Interestingly, medulloblastoma cells express small, fast-inactivating calcium currents consistent with T-type calcium channels, but these channels are activated only from hyperpolarized potentials, which are unlikely to occur. Additionally, a background acid-sensitive K+ current is present with features characteristic of TASK1 or TASK3 channels, such as inhibition by ruthenium red. Western analysis confirms expression of TASK1 and TASK3. In describing the electrophysiologic characteristics of medulloblastoma, one can see features that resemble other high-grade malignancies as opposed to normal cerebellar granule neurons. This supports the notion that the malignant phenotype of medulloblastoma is characterized by unique changes in ion channel expression.


Subject(s)
Medulloblastoma/metabolism , Action Potentials/physiology , Blotting, Western , Cells/drug effects , Cells, Cultured , Electrophysiology , Humans , Membrane Potentials/physiology , Mibefradil/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Peptides/pharmacology , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Potassium Channels, Calcium-Activated/metabolism , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/metabolism , Ruthenium Red/pharmacology
5.
J Neurooncol ; 87(2): 133-41, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18060600

ABSTRACT

PURPOSE: Current medulloblastoma therapy, surgery, radiation, and chemotherapy, is unacceptably toxic. However, 13-cis retinoic acid (RA) and SAHA, a histone deacetylase inhibitor, have each been shown to induce apoptosis in medulloblastoma cultures and mouse models. Both drugs cross the blood brain barrier, have been given safely to children, and achieve brain concentrations that are at or near therapeutic levels. Retinoic acid acts by transcriptionally activating bone morphogenetic protein-2 (BMP-2) and SAHA facilitates transcriptional activity through chromatin accessibility. We tested the hypothesis that these drugs additively induce BMP-2 transcription and apoptosis. EXPERIMENTAL DESIGN: RA + SAHA induction of BMP-2 transcription and apoptosis in medulloblastoma cultures was evaluated. Subsequently the response of mouse medulloblastomas to these two agents in the presence and absence of cisplatin was evaluated. RESULTS: BMP-2 transcription multiplied 3-fold with addition of RA to culture, and 7-fold with both agents. The IC50 of SAHA was reduced by 40% when low dose RA was added. Interestingly, a p38 MAP kinase inhibitor that partially blocks RA-induced apoptosis did not inhibit the activity of RA + SAHA. Flank D283 tumors in athymic mice had slower growth in the RA + SAHA arm than single drug or control arms. Intracranial tumors in ND2:SmoA1 mice treated with RA + SAHA + cisplatin showed a 4-fold increase in apoptosis over controls, and a 2-fold increase over animals receiving only SAHA or RA + SAHA. CONCLUSIONS: RA + SAHA additively induce BMP-2 transcription and medulloblastoma apoptosis. The combination may act through a p38 MAPK independent mechanism. Efficacy increased with cisplatin, which has implications for clinical trial design.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cerebellar Neoplasms/drug therapy , Hydroxamic Acids/administration & dosage , Isotretinoin/administration & dosage , Medulloblastoma/drug therapy , Animals , Apoptosis/drug effects , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/drug effects , Cell Line, Tumor , Cisplatin/administration & dosage , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects , Transforming Growth Factor beta/drug effects , Vorinostat
6.
J Neurooncol ; 79(3): 259-70, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16645722

ABSTRACT

PURPOSE: Suberoylanilide hydroxamic acid (SAHA) has been studied in adult solid and hematologic malignancies. However, little information has been reported on the effects of SAHA on central nervous system (CNS) tumors including medulloblastoma, the most common malignant brain tumor in children. We investigated SAHA in preclinical medulloblastoma models to determine its anti-cancer efficacy as well as its ability to affect intracranial lesions when administered systemically. EXPERIMENTAL DESIGN AND RESULTS: Tissue culture studies were performed treating primary human fibroblasts, established medulloblastoma cell lines, and primary human medulloblastoma tumors with SAHA. At 10 microM concentration, SAHA had little effect on normal fibroblasts but caused >90% apoptosis in cultured medulloblastoma cells. Primary medulloblastomas from patients were sensitive to SAHA compared to vehicle alone in ex vivo studies. In athymic mice with medulloblastoma xenograft tumors, oral SAHA resulted in apoptosis of tumor tissue and significantly slowed tumor growth. In the ND2:Smo transgenic mouse medulloblastoma model, SAHA treatment caused significant apoptosis in these cerebellar tumors. CONCLUSIONS: SAHA effectively induces cell death in established medulloblastoma cell lines, human patient primary tumor cultures, medulloblastoma xenografts and intracranial spontaneous medulloblastomas. Fibroblasts in culture and mice treated with SAHA did not reveal prohibitive toxicity profiles. These findings support the advancement of SAHA to pediatric clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Cerebellar Neoplasms/drug therapy , Hydroxamic Acids/pharmacology , Medulloblastoma/drug therapy , Animals , Apoptosis/drug effects , Cells, Cultured , Child , Fibroblasts/drug effects , Humans , In Situ Nick-End Labeling , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Vorinostat
7.
Pediatr Blood Cancer ; 45(1): 60-3, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15800910

ABSTRACT

Malignant melanoma is rare in childhood and has never been reported to cause pancytopenia due to bone marrow metastases in a child. We report a 3-year-old boy with a large congenital melanocytic nevus who presented with bone pain and pancytopenia due to diffuse bone and bone marrow infiltration with metastatic melanoma without an identifiable primary site. Despite treatment with imatinib mesylate there was no response and the patient died with progressive disease. This case illustrates an unusual presentation of bone marrow failure secondary to malignant melanoma in a young child with symptomatic metastatic marrow infiltration, a rarely reported site of melanoma involvement in adults or children.


Subject(s)
Bone Marrow Neoplasms/secondary , Melanoma/secondary , Nevus, Pigmented/congenital , Pancytopenia/etiology , Skin Neoplasms/pathology , Bone Marrow Neoplasms/complications , Bone Marrow Neoplasms/pathology , Child, Preschool , Fatal Outcome , Humans , Male , Melanoma/complications , Melanoma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...