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.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34369389

ABSTRACT

Angelman syndrome (AS) is a severe neurodevelopmental disorder for which only symptomatic treatment with limited benefits is available. AS is caused by mutations affecting the maternally inherited ubiquitin protein ligase E3A (UBE3A) gene. Previous studies showed that the silenced paternal Ube3a gene can be activated by targeting the antisense Ube3a-ATS transcript. We investigated antisense oligonucleotide-induced (ASO-induced) Ube3a-ATS degradation and its ability to induce UBE3A reinstatement and rescue of AS phenotypes in an established Ube3a mouse model. We found that a single intracerebroventricular injection of ASOs at postnatal day 1 (P1) or P21 in AS mice resulted in potent and specific UBE3A reinstatement in the brain, with levels up to 74% of WT levels in the cortex and a full rescue of sensitivity to audiogenic seizures. AS mice treated with ASO at P1 also showed rescue of established AS phenotypes, such as open field and forced swim test behaviors, and significant improvement on the reversed rotarod. Hippocampal plasticity of treated AS mice was comparable to WT but not significantly different from PBS-treated AS mice. No rescue was observed for the marble burying and nest building phenotypes. Our findings highlight the promise of ASO-mediated reactivation of UBE3A as a disease-modifying treatment for AS.


Subject(s)
Angelman Syndrome , Oligonucleotides, Antisense/therapeutic use , Ubiquitin-Protein Ligases/metabolism , Angelman Syndrome/genetics , Angelman Syndrome/metabolism , Animals , Biological Variation, Population , Disease Models, Animal , Gene Expression Profiling , Gene Silencing , Mice , Targeted Gene Repair/methods , Treatment Outcome
2.
BMC Cancer ; 13: 168, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23548101

ABSTRACT

BACKGROUND: Belinostat is a histone deacetylase inhibitor with anti-tumor effect in several pre-clinical tumor models and clinical trials. The aim of the study was to evaluate changes in cell proliferation and glucose uptake by use of 3'-deoxy-3'-[(18)F]fluorothymidine ([18F]FLT) and 2-deoxy-2-[(18)F]fluoro-D-glucose ([18F]FDG) positron emission tomography (PET) following treatment with belinostat in ovarian cancer in vivo models. METHODS: In vivo uptake of [18F]FLT and [18F]FDG in human ovary cancer xenografts in mice (A2780) were studied after treatment with belinostat. Mice were divided in 2 groups receiving either belinostat (40 mg/kg ip twice daily Day 0-4 and 6-10) or vehicle. Baseline [18F]FLT or [18F]FDG scans were made before treatment (Day 0) and repeated at Day 3, 6 and 10. Tracer uptake was quantified using small animal PET/CT. RESULTS: Tumors in the belinostat group had volumes that were 462 ± 62% (640 mm(3)) at Day 10 relative to baseline which was significantly different (P = 0.011) from the control group 769 ± 74% (926 mm(3)). [18F]FLT SUVmax increased from baseline to Day 10 (+30 ± 9%; P = 0.048) in the control group. No increase was observed in the treatment group. [18F]FDG SUVmean was significantly different in the treatment group compared to the control group (P = 0.0023) at Day 10. Within treatment groups [18F]FDG uptake and to a lesser extent [18F]FLT uptake at Day 3 were significantly correlated with tumor growth at Day 10. CONCLUSIONS: [18F]FDG uptake early following treatment initiation predicted tumor sizes at Day 10, suggesting that [18F]FDG may be a valuable biomarker for non-invasive assessment of anti-tumor activity of belinostat.


Subject(s)
Dideoxynucleosides , Fluorodeoxyglucose F18 , Ovarian Neoplasms/diagnosis , Positron-Emission Tomography , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Female , Glucose Transporter Type 1/genetics , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/therapeutic use , Ki-67 Antigen/genetics , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Radiopharmaceuticals , Sulfonamides/administration & dosage , Sulfonamides/therapeutic use , Thymidine Kinase/genetics , Tumor Burden
3.
PLoS One ; 8(1): e53410, 2013.
Article in English | MEDLINE | ID: mdl-23308217

ABSTRACT

INTRODUCTION: APO866 is a new anti-tumor compound inhibiting nicotinamide phosphoribosyltransferase (NAMPT). APO866 has an anti-tumor effect in several pre-clinical tumor models and is currently in several clinical phase II studies. 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT) is a tracer used to assess cell proliferation in vivo. The aim of this study was non-invasively to study effect of APO866 treatment on [18F]FLT and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) uptake. METHODS: In vivo uptake of [18F]FLT and [18F]FDG in human ovary cancer xenografts in mice (A2780) was studied at various time points after APO866 treatment. Baseline [18F]FLT or [18F]FDG scans were made before treatment and repeated after 24 hours, 48 hours and 7 days. Tumor volume was followed with computed tomography (CT). Tracer uptake was quantified using small animal PET/CT. One hour after iv injection of tracer, static PET scans were performed. Imaging results were compared with Ki67 immunohistochemistry. RESULTS: Tumors treated with APO866 had volumes that were 114% (24 h), 128% (48 h) and 130% (Day 7) relative to baseline volumes at Day 0. In the control group tumor volumes were 118% (24 h), 145% (48 h) and 339% (Day 7) relative to baseline volumes Day 0. Tumor volume between the treatment and control group was significantly different at Day 7 (P = 0.001). Compared to baseline, [18F]FLT SUVmax was significantly different at 24 h (P<0.001), 48 h (P<0.001) and Day 7 (P<0.001) in the APO866 group. Compared to baseline, [18F]FDG SUVmax was significantly different at Day 7 (P = 0.005) in the APO866 group. CONCLUSIONS: APO866 treatment caused a significant decrease in [18F]FLT uptake 24 and 48 hours after treatment initiation. The early reductions in tumor cell proliferation preceded decrease in tumor volume. The results show the possibility to use [18F]FLT and [18F]FDG to image treatment effect early following treatment with APO866 in future clinical studies.


Subject(s)
Acrylamides/pharmacology , Antineoplastic Agents/pharmacology , Dideoxynucleosides/metabolism , Drug Monitoring/methods , Fluorodeoxyglucose F18/metabolism , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/drug therapy , Piperidines/pharmacology , Radiopharmaceuticals/metabolism , Animals , Biological Transport/drug effects , Cell Proliferation/drug effects , Clinical Trials, Phase II as Topic , Female , Fluorine Radioisotopes , Gene Expression/drug effects , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Mice , Mice, Nude , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Positron-Emission Tomography , Radiography , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
PLoS One ; 8(12): e85126, 2013.
Article in English | MEDLINE | ID: mdl-24386456

ABSTRACT

INTRODUCTION: A combination of carboplatin and paclitaxel is often used as first line chemotherapy for treatment of ovarian cancer. Therefore the use of imaging biomarkers early after initiation of treatment to determine treatment sensitivity would be valuable in order to identify responders from non-responders. In this study we describe the non-invasive PET imaging of glucose uptake and cell proliferation using 2-deoxy-2-[(18)F]fluoro-D-glucose (FDG) and 3'-deoxy-3'-[(18)F]fluorothymidine (FLT) for early assessment of treatment response in a pre-clinical mouse model of human ovarian cancer treated with carboplatin and paclitaxel. METHODS: In vivo uptake of FLT and FDG in human ovarian cancer xenografts in mice (A2780) was determined before treatment with carboplatin and paclitaxel (CaP) and repeated day 1, 4 and 8 after treatment start. Tracer uptake was quantified using small animal PET/CT. Tracer uptake was compared with gene expression of Ki67, TK1, GLUT1, HK1 and HK2. RESULTS: Tumors in the CaP group was significantly smaller than in the control group (p=0.03) on day 8. On day 4 FDG SUVmax ratio was significantly lower in the CaP group compared to the control group (105 ± 4% vs 138 ± 9%; p=0.002) and on day 8 the FDG SUVmax ratio was lower in the CaP compared to the control group (125 ± 13% vs 167 ± 13%; p=0.05). On day 1 the uptake of FLT SUVmax ratio was 89 ± 9% in the CaP group and 109 ± 6% in the control group; however the difference was not statistically significant (p=0.08). CONCLUSIONS: Our data suggest that both FDG and FLT PET may be used for the assessment of anti-tumor effects of a combination of carboplatin and paclitaxel in the treatment of ovarian cancer. FLT provides an early and transient signal and FDG a later and more prolonged response. This underscores the importance of optimal timing between treatment and FLT or FDG imaging since treatment response may otherwise be overlooked.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Fluorodeoxyglucose F18/pharmacology , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/drug therapy , Positron-Emission Tomography , Radiopharmaceuticals/pharmacology , Animals , Carboplatin/pharmacology , Female , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Paclitaxel/pharmacology , Radiography , Time Factors
5.
PLoS One ; 7(11): e50618, 2012.
Article in English | MEDLINE | ID: mdl-23226334

ABSTRACT

AIM: 3'-deoxy-3'-[¹8F]fluorothymidine ([18F]FLT) is a tracer used to assess cell proliferation in vivo. The aim of the study was to use [18F]FLT positron emission tomography (PET) to study non-invasively early anti-proliferative effects of the experimental chemotherapeutic agent TP202377 in both sensitive and resistant tumors. METHODS: Xenografts in mice from 3 human cancer cell lines were used: the TP202377 sensitive A2780 ovary cancer cell line (n = 8-16 tumors/group), the induced resistant A2780/Top216 cell line (n = 8-12 tumors/group) and the natural resistant SW620 colon cancer cell line (n = 10 tumors/group). In vivo uptake of [18F]FLT was studied at baseline and repeated 6 hours, Day 1, and Day 6 after TP202377 treatment (40 mg/kg i.v.) was initiated. Tracer uptake was quantified using small animal PET/CT. RESULTS: TP202377 (40 mg/kg at 0 hours) caused growth inhibition at Day 6 in the sensitive A2780 tumor model compared to the control group (P<0.001). In the A2780 tumor model TP202377 treatment caused significant decrease in uptake of [18F]FLT at 6 hours (-46%; P<0.001) and Day 1 (-44%; P<0.001) after treatment start compared to baseline uptake. At Day 6 uptake was comparable to baseline. Treatment with TP202377 did not influence tumor growth or [18F]FLT uptake in the resistant A2780/Top216 and SW620 tumor models. In all control groups uptake of [18F]FLT did not change. Ki67 gene expression paralleled [18F]FLT uptake. CONCLUSION: Treatment of A2780 xenografts in mice with TP202377 (single dose i.v.) caused a significant decrease in cell proliferation assessed by [18F]FLT PET after 6 hours. Inhibition persisted at Day 1; however, cell proliferation had returned to baseline at Day 6. In the resistant A2780/Top216 and SW620 tumor models uptake of [18F]FLT did not change after treatment. With [18F]FLT PET it was possible to distinguish non-invasively between sensitive and resistant tumors already 6 hours after treatment initiation.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Dideoxynucleosides , Indoles/pharmacology , Indoles/therapeutic use , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/drug therapy , Positron-Emission Tomography , Xenograft Model Antitumor Assays , Animals , Biological Transport , Cell Line, Tumor , Cell Proliferation/drug effects , Dideoxynucleosides/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ki-67 Antigen/genetics , Mice , Ovarian Neoplasms/pathology , Thymidine Kinase/genetics , Time Factors , Tomography, X-Ray Computed , Treatment Failure , Tumor Burden/drug effects
6.
Cancer Chemother Pharmacol ; 68(1): 127-38, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20852860

ABSTRACT

BACKGROUND: Rapidly dividing tumor cells have an increased demand for nutrients to support their characteristic unabated growth; this demand is met by an increased availability of nutrients such as amino acids through vasculogenesis and by the enhanced cellular entry of nutrients through the upregulation of specific transporters. Deprivation of intracellular amino acids or block of amino acid uptake has been shown to be cytotoxic to many established human cancer cell lines in vitro and in human cancer xenograft models. RESULTS: In this paper, we provide evidence that the two small molecule oxyphenisatine analogs TOP001 and TOP216 exert their anti-cancer effect by affecting tumor cell metabolism and inducing intracellular amino acid deprivation, leading to a block of cell proliferation. GCN2-mediated phosphorylation of eIF2α as well as mTOR pathway inhibition supports the above notion. In addition, these novel anti-cancer compounds inhibit DNA and protein synthesis and induce apoptosis in a broad spectrum of cancer cell lines. In vivo, the compounds induce tumor stasis and regression in mouse xenograft models of human breast, prostate, ovarian and pancreatic cancer, both when administered intravenously and orally. CONCLUSION: In conclusion, these small molecules, built on a 1,3-dihydroindole-2-one scaffold, elicit strong anti-proliferative and cytotoxic activity, and importantly, a strong anti-tumorigenicity is observed in in vivo xenograft models of human breast, ovary, prostate and pancreatic cancers encouraging the translation of this class of compounds into the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Neoplasms/drug therapy , Oxyphenisatin Acetate/analogs & derivatives , AMP-Activated Protein Kinase Kinases , Amino Acids/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Energy Metabolism/drug effects , Female , Humans , Male , Mice , Mice, Nude , Neoplasms/metabolism , Oxyphenisatin Acetate/chemistry , Oxyphenisatin Acetate/pharmacology , Protein Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
7.
PLoS One ; 5(9): e12965, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20885974

ABSTRACT

BACKGROUND: 3'-Deoxy-3'-[(18)F]fluorothymidine ((18)F-FLT) is a tracer used to assess cell proliferation in vivo. The aim of the study was to use (18)F-FLT positron emission tomography (PET) to study treatment responses to a new anti-cancer compound. To do so, we studied early anti-proliferative effects of the experimental chemotherapy Top216 non-invasively by PET. METHODOLOGY/PRINCIPAL FINDINGS: In vivo uptake of (18)F-FLT in human ovary cancer xenografts in mice (A2780) was studied at various time points after Top216 treatment (50 mg/kg i.v. at 0 and 48 hours) was initiated. Baseline (18)F-FLT scans were made before either Top216 (n = 7-10) or vehicle (n = 5-7) was injected and repeated after 2 and 6 hours and 1 and 5 days of treatment. A parallel study was made with 2'-deoxy-2'-[(18)F]fluoro-D-glucose ((18)F-FDG) (n = 8). Tracer uptake was quantified using small animal PET/CT. Imaging results were validated by tumor volume changes and gene-expression of Ki67 and TK1. Top216 (50 mg/kg 0 and 48 hours) inhibited the growth of the A2780 tumor compared to the control group (P<0.001). (18)F-FLT uptake decreased significantly at 2 hours (-52%; P<0.001), 6 hours (-49%; P = 0.002) and Day 1 (-47%; P<0.001) after Top216 treatment. At Day 5 (18)F-FLT uptake was comparable to uptake in the control group. Uptake of (18)F-FLT was unchanged in the control group during the experiment. In the treatment group, uptake of (18)F-FDG was significantly decreased at 6 hours (-21%; P = 0.003), Day 1 (-29%; P<0.001) and Day 5 (-19%; P = 0.05) compared to baseline. CONCLUSIONS/SIGNIFICANCE: One injection with Top216 initiated a fast and significant decrease in cell-proliferation assessable by (18)F-FLT after 2 hours. The early reductions in tumor cell proliferation preceded changes in tumor size. Our data indicate that (18)F-FLT PET is promising for the early non-invasive assessment of chemotherapy effects in both drug development and for tailoring therapy in patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Monitoring/methods , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/drug therapy , Positron-Emission Tomography/methods , Animals , Cell Proliferation/drug effects , Dideoxynucleosides , Disease Models, Animal , Drug Monitoring/instrumentation , Female , Fluorodeoxyglucose F18 , Humans , Mice , Mice, Nude , Ovarian Neoplasms/physiopathology , Positron-Emission Tomography/instrumentation , Radiopharmaceuticals , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...