Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Urology ; 154: 271-274, 2021 08.
Article in English | MEDLINE | ID: mdl-33581236

ABSTRACT

An identifiable genetic malformation or predisposition syndrome is present in 18% of Wilms tumor cases. Given this, children with conditions associated with a greater than 1% risk of developing Wilms tumor are recommended to have regular surveillance imaging with renal ultrasound until age 7. Seven years is the recommended screening duration because 95% of cases will occur by this age. We present a case of a child with isolated hemihypertrophy, associated with 5% risk of Wilms tumor, who presented with a tumor after the recommended screening, at age 9, brining into question the age cutoffs currently used.


Subject(s)
Hyperplasia/diagnosis , Kidney Neoplasms/diagnostic imaging , Wilms Tumor/diagnostic imaging , Abdominal Pain/etiology , Child , Early Detection of Cancer , Female , Humans , Infant , Kidney Neoplasms/complications , Magnetic Resonance Imaging , Vomiting/etiology , Wilms Tumor/complications
2.
Clin Cancer Res ; 22(17): 4391-404, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27012811

ABSTRACT

PURPOSE: Deregulated MYC drives oncogenesis in many tissues yet direct pharmacologic inhibition has proven difficult. MYC coordinately regulates polyamine homeostasis as these essential cations support MYC functions, and drugs that antagonize polyamine sufficiency have synthetic-lethal interactions with MYC Neuroblastoma is a lethal tumor in which the MYC homologue MYCN, and ODC1, the rate-limiting enzyme in polyamine synthesis, are frequently deregulated so we tested optimized polyamine depletion regimens for activity against neuroblastoma. EXPERIMENTAL DESIGN: We used complementary transgenic and xenograft-bearing neuroblastoma models to assess polyamine antagonists. We investigated difluoromethylornithine (DFMO; an inhibitor of Odc, the rate-limiting enzyme in polyamine synthesis), SAM486 (an inhibitor of Amd1, the second rate-limiting enzyme), and celecoxib (an inducer of Sat1 and polyamine catabolism) in both the preemptive setting and in the treatment of established tumors. In vitro assays were performed to identify mechanisms of activity. RESULTS: An optimized polyamine antagonist regimen using DFMO and SAM486 to inhibit both rate-limiting enzymes in polyamine synthesis potently blocked neuroblastoma initiation in transgenic mice, underscoring the requirement for polyamines in MYC-driven oncogenesis. Furthermore, the combination of DFMO with celecoxib was found to be highly active, alone, and combined with numerous chemotherapy regimens, in regressing established tumors in both models, including tumors harboring highest risk genetic lesions such as MYCN amplification, ALK mutation, and TP53 mutation with multidrug resistance. CONCLUSIONS: Given the broad preclinical activity demonstrated by polyamine antagonist regimens across diverse in vivo models, clinical investigation of such approaches in neuroblastoma and potentially other MYC-driven tumors is warranted. Clin Cancer Res; 22(17); 4391-404. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/drug effects , Neuroblastoma/etiology , Neuroblastoma/pathology , Polyamines/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Celecoxib/pharmacology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Drug Synergism , Eflornithine/pharmacology , Genes, myc , Homeostasis/drug effects , Humans , Mice , Mice, Transgenic , Neuroblastoma/drug therapy , Neuroblastoma/mortality , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Polyamines/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
3.
Pediatr Nephrol ; 29(5): 863-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24310825

ABSTRACT

BACKGROUND: Tyrosine kinase (TK) inhibitors are increasingly being used to treat a variety of pediatric malignancies. Reports in adult patients describe a range of effects of TK inhibitors on the kidney, including hypertension, proteinuria, acute kidney injury, and thrombotic microangiopathy (TMA); however, there are only a few reports of TK-inhibitor-associated nephrotic syndrome. METHODS: We report four pediatric patients with various malignancies (chronic myelogenous leukemia, acute lymphoblastic leukemia, and glioma/renal cell carcinoma) who developed nephrotic syndrome during treatment with TK inhibitors (imatinib, sunitinib, dasatinib, and quizartinib). One of the four patients also had clinical features of TMA. RESULTS: Three of the four patients achieved complete remission of nephrotic syndrome with discontinuation of the TK inhibitor and have had no additional nephrotic syndrome relapses to date. The temporal relationship of nephrotic syndrome onset to TK-inhibitor therapy and resolution of nephrotic syndrome with cessation of therapy strongly imply an association in these patients. CONCLUSIONS: TK inhibitors are important therapies in pediatric cancer, and their use is expanding. Nephrotic syndrome with or without features of TMA is a potential complication of these therapies in children.


Subject(s)
Antineoplastic Agents/adverse effects , Neoplasms/complications , Neoplasms/drug therapy , Nephrotic Syndrome/chemically induced , Protein Kinase Inhibitors/adverse effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Adolescent , Child , Female , Glioma/complications , Glioma/drug therapy , Humans , Infant , Leukemia, B-Cell/complications , Leukemia, B-Cell/drug therapy , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/drug therapy , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/therapeutic use
4.
Clin Cancer Res ; 15(19): 5956-61, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19789308

ABSTRACT

MYC genes are deregulated in a plurality of human cancers. Through direct and indirect mechanisms, the MYC network regulates the expression of > 15% of the human genome, including both protein-coding and noncoding RNAs. This complexity has complicated efforts to define the principal pathways mediating MYC's oncogenic activity. MYC plays a central role in providing for the bioenergetic and biomass needs of proliferating cells, and polyamines are essential cell constituents supporting many of these functions. The rate-limiting enzyme in polyamine biosynthesis, ODC, is a bona fide MYC target, as are other regulatory enzymes in this pathway. A wealth of data link enhanced polyamine biosynthesis to cancer progression, and polyamine depletion may limit the malignant transformation of preneoplastic lesions. Studies with transgenic cancer models also support the finding that the effect of MYC on tumor initiation and progression can be attenuated through the repression of polyamine production. High-risk neuroblastomas (an often lethal embryonal tumor in which MYC activation is paramount) deregulate numerous polyamine enzymes to promote the expansion of intracellular polyamine pools. Selective inhibition of key enzymes in this pathway, e.g., using DFMO and/or SAM486, reduces tumorigenesis and synergizes with chemotherapy to regress tumors in preclinical models. Here, we review the potential clinical application of these and additional polyamine depletion agents to neuroblastoma and other advanced cancers in which MYC is operative.


Subject(s)
Antineoplastic Agents/therapeutic use , Homeostasis/drug effects , Neuroblastoma/drug therapy , Polyamines/metabolism , Animals , Drug Delivery Systems/methods , Gene Expression Regulation, Neoplastic , Genes, myc/physiology , Humans , Mice , Mice, Transgenic , Models, Biological , Neuroblastoma/genetics , Neuroblastoma/metabolism
5.
Cancer Res ; 68(23): 9735-45, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047152

ABSTRACT

Neuroblastoma is a frequently lethal childhood tumor in which MYC gene deregulation, commonly as MYCN amplification, portends poor outcome. Identifying the requisite biopathways downstream of MYC may provide therapeutic opportunities. We used transcriptome analyses to show that MYCN-amplified neuroblastomas have coordinately deregulated myriad polyamine enzymes (including ODC1, SRM, SMS, AMD1, OAZ2, and SMOX) to enhance polyamine biosynthesis. High-risk tumors without MYCN amplification also overexpress ODC1, the rate-limiting enzyme in polyamine biosynthesis, when compared with lower-risk tumors, suggesting that this pathway may be pivotal. Indeed, elevated ODC1 (independent of MYCN amplification) was associated with reduced survival in a large independent neuroblastoma cohort. As polyamines are essential for cell survival and linked to cancer progression, we studied polyamine antagonism to test for metabolic dependence on this pathway in neuroblastoma. The Odc inhibitor alpha-difluoromethylornithine (DFMO) inhibited neuroblast proliferation in vitro and suppressed oncogenesis in vivo. DFMO treatment of neuroblastoma-prone genetically engineered mice (TH-MYCN) extended tumor latency and survival in homozygous mice and prevented oncogenesis in hemizygous mice. In the latter, transient Odc ablation permanently prevented tumor onset consistent with a time-limited window for embryonal tumor initiation. Importantly, we show that DFMO augments antitumor efficacy of conventional cytotoxics in vivo. This work implicates polyamine biosynthesis as an arbiter of MYCN oncogenesis and shows initial efficacy for polyamine depletion strategies in neuroblastoma, a strategy that may have utility for this and other MYC-driven embryonal tumors.


Subject(s)
Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Ornithine Decarboxylase/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biogenic Polyamines/biosynthesis , Cell Growth Processes/physiology , Cell Line, Tumor , Child , Cisplatin/administration & dosage , Cisplatin/pharmacology , Comparative Genomic Hybridization , Drug Synergism , Eflornithine/administration & dosage , Eflornithine/pharmacology , Gene Amplification , Gene Expression Profiling , Genes, myc , Humans , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neuroblastoma/drug therapy , Neuroblastoma/enzymology , Neuroblastoma/metabolism , Ornithine Decarboxylase/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...