Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 117(31): 18401-18411, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32690709

ABSTRACT

Disparities in cancer patient responses have prompted widespread searches to identify differences in sensitive vs. nonsensitive populations and form the basis of personalized medicine. This customized approach is dependent upon the development of pathway-specific therapeutics in conjunction with biomarkers that predict patient responses. Here, we show that Cdk5 drives growth in subgroups of patients with multiple types of neuroendocrine neoplasms. Phosphoproteomics and high throughput screening identified phosphorylation sites downstream of Cdk5. These phosphorylation events serve as biomarkers and effectively pinpoint Cdk5-driven tumors. Toward achieving targeted therapy, we demonstrate that mouse models of neuroendocrine cancer are responsive to selective Cdk5 inhibitors and biomimetic nanoparticles are effective vehicles for enhanced tumor targeting and reduction of drug toxicity. Finally, we show that biomarkers of Cdk5-dependent tumors effectively predict response to anti-Cdk5 therapy in patient-derived xenografts. Thus, a phosphoprotein-based diagnostic assay combined with Cdk5-targeted therapy is a rational treatment approach for neuroendocrine malignancies.


Subject(s)
Neoplasms/drug therapy , Neoplasms/metabolism , Neuroectodermal Tumors/drug therapy , Phosphoproteins/metabolism , Protein Kinase Inhibitors/administration & dosage , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Heterografts , Humans , Mice , Neoplasms/genetics , Neuroectodermal Tumors/genetics , Neuroectodermal Tumors/metabolism , Phosphoproteins/analysis , Phosphoproteins/genetics , Phosphorylation
2.
Dev Period Med ; 22(4): 358-363, 2018.
Article in English | MEDLINE | ID: mdl-30636233

ABSTRACT

The aim of this paper is a clinical and anatomopathological demonstration of a malignant lesion, a gastrointestinal neuroectodermal tumor (GNET), as an exceedingly rare cause of ileus in the pediatric population. Specifically, we present the case of a 12-year-old boy who showed dramatic weight loss, hypochromic anemia, fever, dehydration, exaggerated granulation of the terminal ileum, and mechanical ileus due to the obstruction by an intramural tumor of the small intestine. A 50cm-long part of the small intestine with pathological stricture was surgically removed, sampled and routinely fixed and stained with hematoxylin and eosin. The additional immunostains that were preformed were: PAS, S-100, HMB-45, NSE, LCA, CK AE1 / AE3, desmin, SMA, vimentin, CD99, NSE, synaptophysin, WT-1, calretinin, and DOG-1. Moreover, fluorescent in situ hybridization (FISH) with the EWSR1 Break Apart FISH Probe was applied. The neoplasm was composed of nests and alveolar patterns of frankly malignant clear cells with immunoreactivity to S-100, vimentin, and CD 99. The FISH technique detected chromosomal breaking at 22q12. The tumor metastasized to both the mesenteric lymph nodes and a number of hepatic segments. With several chemotherapy protocols, repeat laparotomies, and liver thermal ablations, the patient had a 1.5-year-long survival from the moment of diagnosis. The diagnosis of this malignancy requires both histopathological evaluation and molecular analysis, and the follow-up is based on careful clinical imaging of the neoplastic spread in order to apply proper surgical and oncological treatments. In conclusion, the clinical course of GNET was highly aggressive.


Subject(s)
Antineoplastic Agents/therapeutic use , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/drug therapy , Neuroectodermal Tumors/diagnosis , Neuroectodermal Tumors/drug therapy , Sarcoma, Clear Cell/diagnosis , Sarcoma, Clear Cell/drug therapy , Biopsy , Child , Endometrial Ablation Techniques , Gastrointestinal Neoplasms/surgery , Humans , In Situ Hybridization, Fluorescence , Male , Neuroectodermal Tumors/surgery , Poland , Rare Diseases/diagnosis , Rare Diseases/drug therapy , Rare Diseases/surgery , Sarcoma, Clear Cell/surgery , Treatment Outcome
3.
J Craniofac Surg ; 28(3): 781-784, 2017 May.
Article in English | MEDLINE | ID: mdl-28468165

ABSTRACT

INTRODUCTION: Primitive neuroectodermal tumors of peripheral origin are very rare, and orbital neuroectodermal tumors are even more uncommon. Only 25 patients with primary orbital involvement in the pediatric age group have been reported. METHODS: In this article, the authors describe their experience in the multimodality treatment approach to treat neuroectodermal tumor of the orbit. The authors also present a male patient 3-year old presenting with a neuroectodermal tumor of the right orbit causing rapidly progressive proptosis. The patient underwent an upper and lateral orbital marginotomy. The entire bone defect was reconstructed with a bone graft, allowing for the reconstruction of the floor and the lateral wall of the middle cranial fossa, the floor of the anterior cranial fossa, the upper and lateral orbital frame, and the right zygomatic bone. Over a period of 16 months, the patient was subjected to chemotherapy. RESULTS: In the postoperative period, a favorable evolution of the disease was observed, with growth in the reconstructed structures, good projection of the orbit and the eyeball, and stable results without tumor recurrence. CONCLUSIONS: The authors present the clinical analysis, surgical management, as well as the chemotherapy treatment established, with follow-ups at 1 and 2 and a half years. This experience shows the effectiveness of multimodality therapy in the treatment of rare tumors of difficult handling.


Subject(s)
Bone Transplantation/methods , Cranial Fossa, Middle/surgery , Neuroectodermal Tumors/surgery , Orbit/surgery , Orbital Neoplasms/surgery , Plastic Surgery Procedures/methods , Prosthesis Implantation/methods , Antineoplastic Agents/therapeutic use , Child, Preschool , Combined Modality Therapy , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Neuroectodermal Tumors/diagnosis , Neuroectodermal Tumors/drug therapy , Orbital Neoplasms/diagnosis , Orbital Neoplasms/drug therapy , Time Factors , Tomography, X-Ray Computed , Zygoma/surgery
4.
Oncology ; 91(6): 348-353, 2016.
Article in English | MEDLINE | ID: mdl-27764830

ABSTRACT

Malignant gastrointestinal neuroectodermal tumor (GNET) is an aggressive rare tumor, primarily occurring in young adults with frequent local-regional metastases and recurrence after local control. The tumor is characterized by the presence of EWSR1-ATF1 or EWSR1-CREB1 and immunohistochemical positivity for S-100 protein without melanocytic marker positivity. Due to poor responses to standard sarcoma regimens, GNET has a poor prognosis, and development of effective systemic therapy is desperately needed to treat these patients. Herein, we present a patient with a small bowel GNET who experienced recurrent hepatic and skeletal metastases after a primary resection. Comprehensive genomic profiling (CGP) in the course of clinical care with DNA and RNA sequencing demonstrated the presence of an exon 7 to exon 6 EWSR1-CREB1 fusion in the context of a diploid genome with no other genomic alterations. In a clinical trial, the patient received a combination of 250 mg crizotinib with 600 mg pazopanib quaque die and achieved partial response and durable clinical benefit for over 2.8 years, and with minimal toxicity from therapy. Using a CGP database of over 50,000 samples, we identified 11 additional cases that harbor EWSR1-CREB1 and report clinicopathologic characteristics, as these patients may also benefit from such a regimen.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/drug therapy , Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/genetics , Liver Neoplasms/drug therapy , Neuroectodermal Tumors/drug therapy , Neuroectodermal Tumors/genetics , Oncogene Proteins, Fusion/genetics , Adolescent , Adult , Anaplastic Lymphoma Kinase , Bone Neoplasms/secondary , Crizotinib , Female , Humans , Indazoles , Intestinal Neoplasms/pathology , Liver Neoplasms/secondary , Male , Middle Aged , Neuroectodermal Tumors/secondary , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrazoles/administration & dosage , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Response Evaluation Criteria in Solid Tumors , Sulfonamides/administration & dosage , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Young Adult
5.
J Neurooncol ; 106(3): 441-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21850536

ABSTRACT

Supratentorial primitive neuroectodermal tumors (sPNET) are rare childhood brain tumors. There is no standard strategy for treating relapsed sPNETs. The role of high dose chemotherapy with hematopoietic stem cell rescue (HDC with HSCR) in treating relapsed sPNET is controversial. A systematic review of the literature regarding outcome of patients with relapsed sPNET treated with HDC and HSCR was performed to examine the potential predictive factors that would justify its use in this subset of patients. Forty-six patients were identified fulfilling the inclusion criteria. Of those, 15 patients were infants and 15 were pineoblastomas. With a median follow-up of 40 months (range 3-123 months) 15 patients were reported alive. Thirteen patients out of the 15 survivors did not receive craniospinal irradiation (CSRT). The 12 month overall survival (OS) of the cohort was 44.2 ± 7.5 months. Twelve-month OS for children less than 36 months was 66.7 ± 12.2 months while for older children it was 27.8 ± 10.6 (P = 0.003). Twelve-month OS was 20.0 ± 10.3 for those patients with pineoblastoma versus 54.6 ± 9.0 for those with non-pineal sPNETs (P < 0.001). Cox regression analysis revealed pineal location as the only independent adverse prognostic factor. In conclusion high dose chemotherapy with HSCR might lead to survival primarily in younger children with relapsed sPNET even in the absence of concomitant use of radiotherapy, whereas the outcome in older children and/or in pineal location is extremely poor with this modality.


Subject(s)
Drug Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Neuroectodermal Tumors/drug therapy , Neuroectodermal Tumors/surgery , Supratentorial Neoplasms/drug therapy , Supratentorial Neoplasms/surgery , Combined Modality Therapy , Databases, Bibliographic/statistics & numerical data , Follow-Up Studies , Humans , Neuroectodermal Tumors/mortality , Proportional Hazards Models , Retrospective Studies , Supratentorial Neoplasms/mortality , Survival Analysis
6.
J Biol Chem ; 285(9): 6091-100, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20022965

ABSTRACT

The neuroectodermal tumors neuroblastoma and melanoma represent biologically aggressive and chemoresistant cancers. The chemotherapeutic agents fenretinide and bortezomib induce apoptosis through endoplasmic reticulum (ER) stress in these tumor types. The aim of this study was to test the hypothesis that the early events of ER stress signaling and response pathways induced by fenretinide and bortezomib are mediated by the eukaryotic initiation factor 2alpha (eIF2alpha)-ATF4 signaling pathway. Treatment of neuroblastoma and melanoma cell lines with fenretinide, bortezomib, or thapsigargin resulted in induction of eIF2alpha signaling, characterized by increased expression of phosphorylated eIF2alpha, ATF4, ATF3, and GADD34. These events correlated with induction of the pro-apoptotic protein Noxa. The cytotoxic response, characterized by up-regulation of Noxa and cell death, was dependent on ATF4, but not the ER-related pro-death signaling pathways involving GADD153 or IRE1. Although PERK-dependent phosphorylation of eIF2alpha enhanced ATF4 protein levels during ER stress, cell death in response to fenretinide, bortezomib, or thapsigargin was not abrogated by inhibition of eIF2alpha phosphorylation through PERK knockdown or overexpression of wild-type eIF2alpha. Furthermore, ATF4 induction in response to ER stress was dependent primarily on transcriptional activation, which occurred in a PERK- and phosphorylated eIF2alpha-independent manner. These results demonstrate that ATF4 mediates ER stress-induced cell death of neuroectodermal tumor cells in response to fenretinide or bortezomib. Understanding the complex regulation of cell death pathways in response to ER stress-inducing drugs has the potential to reveal novel therapeutic targets, thus allowing the development of improved treatment strategies to overcome chemoresistance.


Subject(s)
Activating Transcription Factor 4/physiology , Cell Death , Endoplasmic Reticulum/pathology , Neuroectodermal Tumors/pathology , Stress, Physiological/drug effects , Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Fenretinide/pharmacology , Humans , Neuroectodermal Tumors/drug therapy , Pyrazines/pharmacology , Signal Transduction
7.
Clin Dev Immunol ; 2011: 245181, 2011.
Article in English | MEDLINE | ID: mdl-21941577

ABSTRACT

The N-glycolylated ganglioside NeuGc-GM3 has been described in solid tumors such as breast carcinoma, nonsmall cell lung cancer, and melanoma, but is usually not detected in normal human cells. Our aim was to evaluate the presence of NeuGc-GM3 in pediatric neuroectodermal tumors by immunohistochemistry. Twenty-seven archival cases of neuroblastoma and Ewing sarcoma family of tumors (ESFT) were analyzed. Formalin-fixed, paraffin-embedded tumor samples were cut into 5 µm sections. The monoclonal antibody 14F7, a mouse IgG1 that specifically recognizes NeuGc-GM3, and a peroxidase-labeled polymer conjugated to secondary antibodies were used. Presence of NeuGc-GM3 was evident in 23 of 27 cases (85%), with an average of about 70% of positive tumors cells. Immunoreactivity was moderate to intense in most tumors, showing a diffuse cytoplasmic and membranous staining, although cases of ESFT demonstrated a fine granular cytoplasmic pattern. No significant differences were observed between neuroblastoma with and without NMYC oncogene amplification, suggesting that expression of NeuGc-GM3 is preserved in more aggressive cancers. Until now, the expression of N-glycolylated gangliosides in pediatric neuroectodermal tumors has not been investigated. The present study evidenced the expression of NeuGc-GM3 in a high proportion of neuroectodermal tumors, suggesting its potential utility as a specific target of immunotherapy.


Subject(s)
G(M3) Ganglioside/analogs & derivatives , Neuroectodermal Tumors/chemistry , Adolescent , Cancer Vaccines/immunology , Child , Child, Preschool , G(M3) Ganglioside/analysis , G(M3) Ganglioside/immunology , Gene Expression Regulation, Neoplastic , Genes, myc , Humans , Immunohistochemistry , Infant , Ki-67 Antigen/metabolism , Neuroectodermal Tumors/drug therapy , Neuroectodermal Tumors/pathology
8.
Cells ; 10(6)2021 06 06.
Article in English | MEDLINE | ID: mdl-34204116

ABSTRACT

Compared to pancreatic adenocarcinoma (PDAC), pancreatic neuroendocrine tumors (PanNET) represent a rare and heterogeneous tumor entity. In addition to surgical resection, several therapeutic approaches, including biotherapy, targeted therapy or chemotherapy are applicable. However, primary or secondary resistance to current therapies is still challenging. Recent genome-wide sequencing efforts in PanNET identified a large number of mutations in pathways involved in epigenetic modulation, including acetylation. Therefore, targeting epigenetic modulators in neuroendocrine cells could represent a new therapeutic avenue. Detailed information on functional effects and affected signaling pathways upon epigenetic targeting in PanNETs, however, is missing. The primary human PanNET cells NT-3 and NT-18 as well as the murine insulinoma cell lines beta-TC-6 (mouse) and RIN-T3 (rat) were treated with the non-selective histone-deacetylase (HDAC) inhibitor panobinostat (PB) and analyzed for functional effects and affected signaling pathways by performing Western blot, FACS and qPCR analyses. Additionally, NanoString analysis of more than 500 potentially affected targets was performed. In vivo immunohistochemistry (IHC) analyses on tumor samples from xenografts and the transgenic neuroendocrine Rip1Tag2-mouse model were investigated. PB dose dependently induced cell cycle arrest and apoptosis in neuroendocrine cells in human and murine species. HDAC inhibition stimulated redifferentiation of human primary PanNET cells by increasing mRNA-expression of somatostatin receptors (SSTRs) and insulin production. In addition to hyperacetylation of known targets, PB mediated pleitropic effects via targeting genes involved in the cell cycle and modulation of the JAK2/STAT3 axis. The HDAC subtypes are expressed ubiquitously in the existing cell models and in human samples of metastatic PanNET. Our results uncover epigenetic HDAC modulation using PB as a promising new therapeutic avenue in PanNET, linking cell-cycle modulation and pathways such as JAK2/STAT3 to epigenetic targeting. Based on our data demonstrating a significant impact of HDAC inhibition in clinical relevant in vitro models, further validation in vivo is warranted.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Neoplasm Proteins , Neuroectodermal Tumors , Pancreatic Neoplasms , Panobinostat/pharmacology , Animals , Cell Line, Tumor , Humans , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neuroectodermal Tumors/drug therapy , Neuroectodermal Tumors/enzymology , Neuroectodermal Tumors/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Rats
9.
Mol Cancer Ther ; 20(9): 1680-1691, 2021 09.
Article in English | MEDLINE | ID: mdl-34158347

ABSTRACT

We analyzed the efficacy and mechanistic interactions of PARP inhibition (PARPi; olaparib) and CDK4/6 inhibition (CDK4/6i; palbociclib or abemaciclib) combination therapy in castration-resistant prostate cancer (CRPC) and neuroendocrine prostate cancer (NEPC) models. We demonstrated that combined olaparib and palbociblib or abemaciclib treatment resulted in synergistic suppression of the p-Rb1-E2F1 signaling axis at the transcriptional and posttranslational levels, leading to disruption of cell-cycle progression and inhibition of E2F1 gene targets, including genes involved in DDR signaling/damage repair, antiapoptotic BCL-2 family members (BCL-2 and MCL-1), CDK1, and neuroendocrine differentiation (NED) markers in vitro and in vivo In addition, olaparib + palbociclib or olaparib + abemaciclib combination treatment resulted in significantly greater growth inhibition and apoptosis than either single agent alone. We further showed that PARPi and CDK4/6i combination treatment-induced CDK1 inhibition suppressed p-S70-BCL-2 and increased caspase cleavage, while CDK1 overexpression effectively prevented the downregulation of p-S70-BCL-2 and largely rescued the combination treatment-induced cytotoxicity. Our study defines a novel combination treatment strategy for CRPC and NEPC and demonstrates that combination PARPi and CDK4/6i synergistically promotes suppression of the p-Rb1-E2F1 axis and E2F1 target genes, including CDK1 and NED proteins, leading to growth inhibition and increased apoptosis in vitro and in vivo Taken together, our results provide a molecular rationale for PARPi and CDK4/6i combination therapy and reveal mechanism-based clinical trial opportunities for men with NEPC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Differentiation , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Neuroectodermal Tumors/drug therapy , Poly(ADP-ribose) Polymerases/chemistry , Prostatic Neoplasms/drug therapy , Aminopyridines/administration & dosage , Animals , Apoptosis , Benzimidazoles/administration & dosage , Cell Cycle , Cell Proliferation , Humans , Male , Mice , Mice, Nude , Neuroectodermal Tumors/metabolism , Neuroectodermal Tumors/pathology , Phthalazines/administration & dosage , Piperazines/administration & dosage , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Pyridines/administration & dosage , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Skeletal Radiol ; 39(6): 595-600, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20151121

ABSTRACT

Extraskeletal Ewing's sarcoma (ES) and primitive neuroectodermal tumor (PNET) are widely regarded as clinically and histologically identical tumors which consist of small blue round cells. Extraskeletal ESs/PNETs usually occur in the deep soft tissues of the paraspinal region, chest wall, or lower extremities. However, superficially located cases, so-called cutaneous ESs/PNETs, are exceedingly rare, and the vast majority of the reported cases present as a single small mass. We present magnetic resonance imaging (MRI) findings and clinical course of a unique case of primary cutaneous ES/PNET presenting as numerous huge masses with severe ulceration on them.


Subject(s)
Antineoplastic Agents/administration & dosage , Magnetic Resonance Imaging/methods , Neoplasms, Multiple Primary/diagnosis , Neuroectodermal Tumors/diagnosis , Sarcoma, Ewing/diagnosis , Skin Neoplasms/diagnosis , Ulcer/diagnosis , Humans , Male , Middle Aged , Neoplasms, Multiple Primary/complications , Neoplasms, Multiple Primary/drug therapy , Neuroectodermal Tumors/complications , Neuroectodermal Tumors/drug therapy , Sarcoma, Ewing/complications , Sarcoma, Ewing/drug therapy , Skin Neoplasms/complications , Skin Neoplasms/drug therapy , Treatment Outcome , Ulcer/etiology , Ulcer/prevention & control
11.
FEBS Lett ; 594(22): 3602-3618, 2020 11.
Article in English | MEDLINE | ID: mdl-32860713

ABSTRACT

Aberrant expression of glycosphingolipids (GSLs) is a unique feature of cancer and stromal cells in tumor microenvironments. Although the impact of GSLs on tumor progression remains largely unclear, anticancer immunotherapies directed against GSLs are attracting growing attention. Here, we focus on GD2, a disialoganglioside expressed in tumors of neuroectodermal origin, and Globo H ceramide (GHCer), the most prevalent cancer-associated GSL overexpressed in a variety of epithelial cancers. We first summarize recent advances on our understanding of GD2 and GHCer biology and then discuss the clinical development of the first immunotherapeutic agent targeting a glycolipid, the GD2-specific antibody dinutuximab, its approved indications, and new strategies to improve its efficacy for neuroblastoma. Next, we review ongoing clinical trials on Globo H-targeted immunotherapeutics. We end with highlighting how these studies provide sound scientific rationales for targeting GSLs in cancer and may facilitate a rational design of new GSL-targeted anticancer therapeutics.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Glycosphingolipids/metabolism , Neoplasms, Glandular and Epithelial/drug therapy , Neuroectodermal Tumors/drug therapy , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antigens, Tumor-Associated, Carbohydrate/metabolism , Antineoplastic Agents, Immunological/pharmacology , Clinical Trials as Topic , Gangliosides/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy , Neoplasms, Glandular and Epithelial/metabolism , Neuroectodermal Tumors/metabolism , Tumor Microenvironment/drug effects
12.
Am J Surg Pathol ; 44(4): 456-466, 2020 04.
Article in English | MEDLINE | ID: mdl-31651526

ABSTRACT

A malignant gastrointestinal neuroectodermal tumor (GNET) is rare, and it is therefore yet to be completely understood. This study aimed to present the clinicopathologic features of GNET, including treatment information. We included 19 patients with GNET with a mean tumor size of 4.2 cm. The most common site of tumor origin was the small intestine (57.9%), followed by the stomach (15.8%), colon (10.5%), ileocecal junction (5.3%), lower esophagus (5.3%), and anal canal (5.3%). Microscopically, the tumors were composed of epithelioid cells with eosinophilic or clear cytoplasm arranged in nest, sheet-like, papillary, or pseudoalveolar patterns and/or spindle tumor cells with eosinophilic cytoplasm arranged in a fascicular pattern. Immunohistochemically, the tumor cells stained positively for S100 (19/19,100%), SOX10 (14/15, 93.3%), vimentin (17/17, 100%), synaptophysin (Syn) (7/17, 41.2%), CD56 (4/13, 30.8%), CD99 (1/5, 20%), and CD117 (1/15, 6.7%), and negatively for HMB45, Melan A, DOG1, CD34, AE1/AE3, CAM5.2, chromogranin A, smooth muscle actin, and desmin. In total, 14/15 (93.3%) cases showed split Ewing sarcoma breakpoint region 1 gene (EWSR1) signals consistent with a chromosomal translocation involving EWSR1. Within a mean follow-up of 29.7 months (range: 3 to 63 mo), 2/15 (13.3%) patients died of disease, 5 (33.3%) were alive with disease, and 8 (53.3%) had no evidence of disease. Two and 1 patients showed partial response to apatinib and anlotinib, respectively. In conclusion, GNET has distinctive morphologic, immunohistochemical, and molecular genetic features and should be distinguished from other gastrointestinal tract malignancies. Apatinib and anlotinib might be effective for the treatment of advanced GNET and could prolong patient survival.


Subject(s)
Biomarkers, Tumor , Gastrointestinal Neoplasms , Immunohistochemistry , In Situ Hybridization, Fluorescence , Neuroectodermal Tumors , RNA-Binding Protein EWS/genetics , Translocation, Genetic , Adult , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Female , Gastrointestinal Neoplasms/chemistry , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Genetic Predisposition to Disease , Humans , Indoles/therapeutic use , Male , Middle Aged , Neuroectodermal Tumors/chemistry , Neuroectodermal Tumors/drug therapy , Neuroectodermal Tumors/genetics , Neuroectodermal Tumors/pathology , Predictive Value of Tests , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Quinolines/therapeutic use , Treatment Outcome , Tumor Burden
13.
Tumori ; 94(2): 246-53, 2008.
Article in English | MEDLINE | ID: mdl-18564613

ABSTRACT

Disseminated neuroectoderma-derived tumors, mainly neuroblastoma in childhood and melanoma in the adulthood, are refractory to most current therapeutic regimens and hence the prognosis remains very poor. Preclinical research studies have indicated several agents that show promising therapeutic potential for these neoplasms. However, there appears to be a limitation to their in vivo applicability, mainly due to unfavorable pharmacokinetic properties that lead to insufficient drug delivery to the tumor or metastatic sites or to high systemic or organ-specific toxicity. In this scenario, the focus is on targeted cancer therapy. Encapsulating anticancer drugs in liposomes enables targeted drug delivery to tumor tissue and prevents damage to the normal surrounding tissue. Indeed, sterically stabilized liposomes have been shown to enhance the selective localization of entrapped drugs to solid tumors, with improvements in therapeutic indices. The identification of tumor-associated antigens and/or genes and the relative ease of manipulating the physicochemical features of liposome hold promise for the development of novel therapeutic strategies that selectively target tumor cells. Combined targeting is still investigated, especially the availability to simultaneously target and kill both the cancer cells and the tumor vasculature. Animal models make it possible to link molecular genetics and biochemistry information to the physiological basis of disease and are important predictive tools that offer a frontline testing system for studying the involvement of specific genes and the efficacy of novel therapeutics approaches. Relevant experimental models of human neuroblastoma and melanoma, which better reflect the tumor behavior in patients, are required to evaluate the effectiveness of the various targeted liposomal formulations and their possible systemic and organ-specific toxicity. The most multifunctional targeted liposomes are herein described, with primary attention on testing their efficacy in clinically relevant animal models for the treatment of neuroblastoma and melanoma.


Subject(s)
Antineoplastic Agents/administration & dosage , Antisense Elements (Genetics)/administration & dosage , Liposomes , Neuroectodermal Tumors/drug therapy , Animals , Drug Delivery Systems , Humans , Neuroblastoma/drug therapy
14.
Mol Cancer Ther ; 6(3): 979-86, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17363491

ABSTRACT

Resistance to anticancer drugs and consequent failure of chemotherapy is a complex problem severely limiting therapeutic options in metastatic cancer. Many studies have shown a role for drug efflux pumps of the ATP-binding cassette transporters family in the development of drug resistance. ClC-3, a member of the CLC family of chloride channels and transporters, is expressed in intracellular compartments of neuronal cells and involved in vesicular acidification. It has previously been suggested that acidification of intracellular organelles can promote drug resistance by increasing drug sequestration. Therefore, we hypothesized a role for ClC-3 in drug resistance. Here, we show that ClC-3 is expressed in neuroendocrine tumor cell lines, such as BON, LCC-18, and QGP-1, and localized in intracellular vesicles co-labeled with the late endosomal/lysosomal marker LAMP-1. ClC-3 overexpression increased the acidity of intracellular vesicles, as assessed by acridine orange staining, and enhanced resistance to the chemotherapeutic drug etoposide by almost doubling the IC(50) in either BON or HEK293 cell lines. Prevention of organellar acidification, by inhibition of the vacuolar H(+)-ATPase, reduced etoposide resistance. No expression of common multidrug resistance transporters, such as P-glycoprotein or multidrug-related protein-1, was detected in either the BON parental cell line or the derivative clone overexpressing ClC-3. The probable mechanism of enhanced etoposide resistance can be attributed to the increase of vesicular acidification as consequence of ClC-3 overexpression. This study therefore provides first evidence for a role of intracellular CLC proteins in the modulation of cancer drug resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Chloride Channels/metabolism , Drug Resistance, Neoplasm , Endosomes/drug effects , Etoposide/pharmacology , Neuroectodermal Tumors/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Acridine Orange , Cell Proliferation/drug effects , Cells, Cultured/drug effects , Drug Resistance, Multiple , Endosomes/metabolism , Genes, MDR/physiology , Humans , Immunoenzyme Techniques , Kidney/drug effects , Muscle Proteins/pharmacology , Neuroectodermal Tumors/drug therapy , Neuroectodermal Tumors/pathology
15.
Eur J Cancer ; 42(12): 1875-80, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16806903

ABSTRACT

The aims of this study were reviewing our experience regarding the pulmonary toxicity of the mammalian target of rapamycin (mTOR) inhibitor temsirolimus, discussing potential pathogenic mechanisms and proposing management strategies. Medical records and radiological reports of 22 patients treated with weekly doses of temsirolimus 25 mg were reviewed. Eight (36%) out of 22 patients developed pulmonary abnormalities compatible with drug-induced pneumonitis. Half were asymptomatic and in those with symptoms, dyspnea and dry cough were the most common. Radiologically two different patterns, ground glass opacities and lung parenchymal consolidation, were described. The management of this toxicity was variable, ranging from no intervention to discontinuation of the drug. In our experience temsirolimus may cause drug-induced pneumonitis at a higher incidence than that previously reported. The presentation and its severity are variable. The risk of developing this toxicity may be increased among subjects with abnormal pre-treatment pulmonary functions or history of lung disease.


Subject(s)
Antineoplastic Agents/adverse effects , Endometrial Neoplasms/drug therapy , Lung Diseases/chemically induced , Neuroectodermal Tumors/drug therapy , Sirolimus/analogs & derivatives , Adult , Aged , Endometrial Neoplasms/diagnostic imaging , Female , Humans , Male , Middle Aged , Neuroectodermal Tumors/diagnostic imaging , Sirolimus/adverse effects , Tomography, X-Ray Computed
16.
Int J Radiat Oncol Biol Phys ; 65(1): 210-21, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16472938

ABSTRACT

PURPOSE: Model the effects of radiation dosimetry on IQ among pediatric patients with central nervous system (CNS) tumors. METHODS AND MATERIALS: Pediatric patients with CNS embryonal tumors (n = 39) were prospectively evaluated with serial cognitive testing, before and after treatment with postoperative, risk-adapted craniospinal irradiation (CSI) and conformal primary-site irradiation, followed by chemotherapy. Differential dose-volume data for 5 brain volumes (total brain, supratentorial brain, infratentorial brain, and left and right temporal lobes) were correlated with IQ after surgery and at follow-up by use of linear regression. RESULTS: When the dose distribution was partitioned into 2 levels, both had a significantly negative effect on longitudinal IQ across all 5 brain volumes. When the dose distribution was partitioned into 3 levels (low, medium, and high), exposure to the supratentorial brain appeared to have the most significant impact. For most models, each Gy of exposure had a similar effect on IQ decline, regardless of dose level. CONCLUSIONS: Our results suggest that radiation dosimetry data from 5 brain volumes can be used to predict decline in longitudinal IQ. Despite measures to reduce radiation dose and treatment volume, the volume that receives the highest dose continues to have the greatest effect, which supports current volume-reduction efforts.


Subject(s)
Central Nervous System Neoplasms/radiotherapy , Cognition Disorders/etiology , Intelligence/radiation effects , Models, Psychological , Neoplasms, Germ Cell and Embryonal/radiotherapy , Central Nervous System Neoplasms/drug therapy , Child , Child, Preschool , Cranial Irradiation/methods , Dose-Response Relationship, Radiation , Female , Humans , Intelligence Tests , Male , Medulloblastoma/drug therapy , Medulloblastoma/radiotherapy , Neoplasms, Germ Cell and Embryonal/drug therapy , Neuroectodermal Tumors/drug therapy , Neuroectodermal Tumors/radiotherapy , Prospective Studies , Radiotherapy Dosage , Rhabdoid Tumor/drug therapy , Rhabdoid Tumor/radiotherapy , Supratentorial Neoplasms/drug therapy , Supratentorial Neoplasms/radiotherapy , Temporal Lobe/radiation effects
17.
Pediatr Infect Dis J ; 25(5): 465-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16645519

ABSTRACT

Primary invasive aspergillosis of the gut is a rare event and is associated with high mortality. We report for the first time on a patient who had isolated aspergillosis of the small bowel after autologous stem cell transplantation. Diagnosis of invasive aspergillosis of the gut was based on abdominal pain, galactomannan antigenemia and isolation of Aspergillus fumigatus from the stool and was later confirmed by pathohistologic examination. No other site of invasive aspergillosis was evident. The patient was successfully treated with early surgery and combination antifungal therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Aspergillosis/diagnosis , Aspergillus fumigatus/isolation & purification , Intestines/microbiology , Stem Cell Transplantation/adverse effects , Transplantation, Autologous/adverse effects , Central Nervous System Neoplasms/drug therapy , Child , Humans , Intestinal Diseases/microbiology , Male , Neuroectodermal Tumors/drug therapy
18.
Cancer Res ; 60(11): 2800-4, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10850417

ABSTRACT

Here we report that aloe-emodin (AE), a hydroxyanthraquinone present in Aloe vera leaves, has a specific in vitro and in vivo antineuroectodermal tumor activity. The growth of human neuroectodermal tumors is inhibited in mice with severe combined immunodeficiency without any appreciable toxic effects on the animals. The compound does not inhibit the proliferation of normal fibroblasts nor that of hemopoietic progenitor cells. The cytotoxicity mechanism consists of the induction of apoptosis, whereas the selectivity against neuroectodermal tumor cells is founded on a specific energy-dependent pathway of drug incorporation. Taking into account its unique cytotoxicity profile and mode of action, AE might represent a conceptually new lead antitumor drug.


Subject(s)
Emodin/analogs & derivatives , Emodin/pharmacology , Emodin/therapeutic use , Neuroectodermal Tumors/drug therapy , Animals , Anthraquinones , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Cycle/drug effects , Dose-Response Relationship, Drug , Emodin/chemistry , Emodin/toxicity , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Female , Flow Cytometry , HeLa Cells , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, SCID , Microscopy, Electron , Models, Chemical , Time Factors , Tumor Cells, Cultured
20.
BMJ Case Rep ; 20152015 Oct 29.
Article in English | MEDLINE | ID: mdl-26516248

ABSTRACT

Primary neuroectodermal renal tumours (PNET) are rare and aggressive neoplasms; thrombosis of the inferior vena cava (IVC) is associated with this entity. We report here the case of a 19-year-old man who experienced a new onset of abdominal pain. A CT scan revealed a large left renal mass, perirenal haematoma and IVC thrombosis. Owing to an acute drop in haemoglobin and subsegmentary pulmonary embolism, he underwent emergency selective renal artery angiography and embolisation of bleeding vessels and IVC filter (IVCF) placement. Once stable, he underwent a left radical nephrectomy and IVC thrombectomy; the pathology report confirmed PNET. 6 months later, imaging revealed a residual tumoral thrombus in the IVCF located in the retrohepatic IVC. The patient underwent removal of this device and the thrombus via a right thoracoabdominal approach. He recovered well and at 4 months, he continues his chemotherapy cycles.


Subject(s)
Device Removal , Kidney Neoplasms/surgery , Neuroectodermal Tumors/surgery , Thrombosis/surgery , Vena Cava, Inferior/surgery , Humans , Kidney Neoplasms/drug therapy , Male , Neoplasm, Residual , Neuroectodermal Tumors/drug therapy , Radiography , Reoperation , Thrombectomy , Thrombosis/diagnostic imaging , Vena Cava Filters , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL