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.
Oncotarget ; 8(24): 38337-38350, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28418885

ABSTRACT

Over the last decade kinase inhibitors have witnessed tremendous growth as anti-cancer drugs. Unfortunately, despite their promising clinical successes, a large portion of patients does not benefit from these targeted therapeutics. Vemurafenib is a serine/threonine kinase inhibitor approved for the treatment of melanomas specifically expressing the BRAFV600E mutation. The aim of this study was to develop vemurafenib as PET tracer to determine its potential for identification of tumors sensitive to vemurafenib treatment. Therefore, vemurafenib was labeled with carbon-11 and analyzed for its tumor targeting potential in melanoma xenografts Colo829 (BRAFV600E) and MeWo (BRAFwt) using autoradiography on tissue sections, in vitro tumor cell uptake studies and biodistribution studies in xenografted athymic nu/nu mice. [11C]vemurafenib was synthesized in 21 ± 4% yield (decay corrected, calculated from [11C]CO) in > 99% radiochemical purity and a specific activity of 55 ± 18 GBq/µmol. Similar binding of [11C]vemurafenib was shown during autoradiography and cellular uptake studies in both cell lines. Plasma metabolite analysis demonstrated > 95% intact [11C]vemurafenib in vivo at 45 minutes after injection, indicating excellent stability. Biodistribution studies confirmed the in vitro results, showing similar tumor-to-background ratios in both xenografts models. These preliminary results suggest that identification of BRAFV600E mutations in vivo using PET with [11C]vemurafenib will be challenging.


Subject(s)
Indoles/pharmacokinetics , Melanoma/genetics , Proto-Oncogene Proteins B-raf/genetics , Radiopharmaceuticals/pharmacokinetics , Sulfonamides/pharmacokinetics , Animals , Carbon Radioisotopes/pharmacokinetics , Cell Line, Tumor , Heterografts , Humans , Melanoma/diagnostic imaging , Mice , Mice, Nude , Mutation , Patient Selection , Positron-Emission Tomography , Radioactive Tracers , Tissue Distribution , Vemurafenib
2.
Nucl Med Biol ; 43(10): 612-24, 2016 10.
Article in English | MEDLINE | ID: mdl-27497236

ABSTRACT

INTRODUCTION: Tyrosine kinase inhibitors (TKIs) are very attractive targeted drugs, although a large portion of patients remains unresponsive. PET imaging with EGFR targeting TKIs ([(11)C]erlotinib and [(18)F]afatinib) showed promise in identifying treatment sensitive tumors. The aim of this study was to synthesize two anti-angiogenic TKI tracers, [(11)C]axitinib and [(11)C]nintedanib, and to evaluate their potential for PET. METHODS: Following successful tracer synthesis, biodistribution studies in VU-SCC-OE and FaDu xenograft bearing mice were performed. Furthermore, tracer stability studies in mice were performed employing (radio-)HPLC and LC-MS/MS techniques. For [(11)C]nintedanib an LC-MS/MS method was developed to detect the primary carboxylic acid metabolite, resulting from methylester cleavage, in plasma and tumors, because this metabolite is postulated to be important for nintedanib efficacy. LC-MS/MS was also explored to assess the metabolic fate of [(11)C]axitinib in vivo, since axitinib has an isomerizable double bond. RESULTS: [(11)C]axitinib and [(11)C]nintedanib were successfully synthesized with 10.5±2.6% and 25.6±3.3% radiochemical yield (corrected for decay), respectively. Biodistribution studies only demonstrated tumor uptake of [(11)C]nintedanib in FaDu xenografts of 1.66±0.02% ID/g at 60min p.i. In vivo stability analysis of [(11)C]axitinib at 45min p.i. revealed the formation of predominantly non-polar metabolites (36.6±6.8% vs 47.1±8.4% of parent tracer and 16.3±2.1% of polar metabolites), while for [(11)C]nintedanib mostly polar metabolites were found (70.9±4.1 vs 26.7±3.9% of parent tracer and only 2.4±1.6 of a non-polar metabolites). No isomerization of [(11)C]axtinib was observed in vivo; however, a sulfoxide metabolite could be detected using LC-MS/MS. For [(11)C]nintedanib, LC-MS/MS revealed formation of the reported primary carboxylic acid metabolite when in vitro plasma incubations were performed, with large differences in plasmas from different species. In vivo metabolite analysis, however, did not demonstrate the presence of the carboxylic acid in plasma or tumor tissue. CONCLUSIONS: Reliable syntheses of [(11)C]axitinib and [(11)C]nintedanib were successfully developed. Tumor uptake was observed for [(11)C]nintedanib, albeit modest. The metabolic profiles of the tracers suggest that rapid metabolism is partly responsible for the modest tumor targeting observed.


Subject(s)
Carbon Radioisotopes , Imidazoles/chemical synthesis , Imidazoles/metabolism , Indazoles/chemical synthesis , Indazoles/metabolism , Indoles/chemical synthesis , Indoles/metabolism , Positron-Emission Tomography/methods , Protein-Tyrosine Kinases/antagonists & inhibitors , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacokinetics , Animals , Axitinib , Cell Line, Tumor , Cell Transformation, Neoplastic , Humans , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Indazoles/chemistry , Indazoles/pharmacokinetics , Indoles/chemistry , Indoles/pharmacokinetics , Mice , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Radioactive Tracers , Radiochemistry , Rats , Tissue Distribution
3.
EJNMMI Res ; 5: 14, 2015.
Article in English | MEDLINE | ID: mdl-25853020

ABSTRACT

BACKGROUND: Tyrosine kinase inhibitors (TKIs) have experienced a tremendous boost in the last decade, where more than 15 small molecule TKIs have been approved by the FDA. Unfortunately, despite their promising clinical successes, a large portion of patients remain unresponsive to these targeted drugs. For non-small cell lung cancer (NSCLC), the effectiveness of TKIs is dependent on the mutational status of epidermal growth factor receptor (EGFR). The exon 19 deletion as well as the L858R point mutation lead to excellent sensitivity to TKIs such as erlotinib and gefitinib; however, despite initial good response, most patients invariably develop resistance against these first-generation reversible TKIs, e.g., via T790M point mutation. Second-generation TKIs that irreversibly bind to EGFR wild-type and mutant isoforms have therefore been developed and one of these candidates, afatinib, has now reached the market. Whether irreversible TKIs differ from reversible TKIs in their in vivo tumor-targeting properties is, however, not known and is the subject of the present study. METHODS: Erlotinib was labeled with carbon-11 and afatinib with fluorine-18 without modifying the structure of these compounds. A preclinical positron emission tomography (PET) study was performed in mice bearing NSCLC xenografts with a representative panel of mutations: an EGFR-WT xenograft cell line (A549), an acquired treatment-resistant L858R/T790M mutant (H1975), and a treatment-sensitive exon 19 deleted mutant (HCC827). PET imaging was performed in these xenografts with both tracers. Additionally, the effect of drug efflux transporter permeability glycoprotein (P-gp) on the tumor uptake of tracers was explored by therapeutic blocking with tariquidar. RESULTS: Both tracers only demonstrated selective tumor uptake in the HCC827 xenograft line (tumor-to-background ratio, [(11)C]erlotinib 1.9 ± 0.5 and [(18)F]afatinib 2.3 ± 0.4), thereby showing the ability to distinguish sensitizing mutations in vivo. No major differences were observed in the kinetics of the reversible and the irreversible tracers in each of the xenograft models. Under P-gp blocking conditions, no significant changes in tumor-to-background ratio were observed; however, [(18)F]afatinib demonstrated better tumor retention in all xenograft models. CONCLUSIONS: TKI-PET provides a method to image sensitizing mutations and can be a valuable tool to compare the distinguished targeting properties of TKIs in vivo.

4.
Nucl Med Biol ; 41(9): 749-57, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25066021

ABSTRACT

INTRODUCTION: Afatinib is an irreversible ErbB family blocker that was approved for the treatment of EGFR mutated non-small cell lung cancer in 2013. Positron emission tomography (PET) with fluorine-18 labeled afatinib provides a means to obtain improved understanding of afatinib tumor disposition in vivo. PET imaging with [(18)F]afatinib may also provide a method to select treatment responsive patients. The aim of this study was to label afatinib with fluorine-18 and evaluate its potential as TKI-PET tracer in tumor bearing mice. METHODS: A radiochemically novel coupling, using peptide coupling reagent BOP, was explored and optimized to synthesize [(18)F]afatinib, followed by a metabolite analysis and biodistribution studies in two clinically relevant lung cancer cell lines, xenografted in nude mice. RESULTS: A reliable [(18)F]afatinib radiosynthesis was developed and the tracer could be produced in yields of 17.0 ± 2.5% calculated from [(18)F]F(-) and >98% purity. The identity of the product was confirmed by co-injection on HPLC with non-labeled afatinib. Metabolite analysis revealed a moderate rate of metabolism, with >80% intact tracer in plasma at 45 min p.i. Biodistribution studies revealed rapid tumor accumulation and good retention for a period of at least 2 hours, while background tissues showed rapid clearance of the tracer. CONCLUSION: We have developed a method to synthesize [(18)F]afatinib and related fluorine-18 labeled 4-anilinoquinazolines. [(18)F]Afatinib showed good stability in vivo, justifying further evaluation as a TKI-PET tracer.


Subject(s)
ErbB Receptors/metabolism , Fluorine Radioisotopes/pharmacokinetics , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , Quinazolines/pharmacokinetics , Afatinib , Animals , Cell Line, Tumor , Fluorine Radioisotopes/chemistry , Isotope Labeling/methods , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Mice, Nude , Organ Specificity , Positron-Emission Tomography/methods , Quinazolines/chemistry , Radiopharmaceuticals/chemical synthesis , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
5.
J Org Chem ; 79(11): 5219-26, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24820129

ABSTRACT

A convenient trans-selective one-pot synthesis of tetrafunctionalized 2-imidazolines is described. Our approach to these valuable heterocyclic scaffolds involves a formal 1,3-dipolar cycloaddition between nitrile ylides or nitrilium triflates and imines. A detailed experimental study in combination with a high-level computational exploration of reaction routes reveals a plausible reaction pathway that accounts for the observed diastereoselectivity.


Subject(s)
Heterocyclic Compounds/chemistry , Imidazoles/chemical synthesis , Imidazolines/chemical synthesis , Nitriles/chemistry , Catalysis , Crystallography, X-Ray , Cycloaddition Reaction , Imidazoles/chemistry , Imidazolines/chemistry , Molecular Structure , Stereoisomerism
6.
Chembiochem ; 13(14): 2056-60, 2012 Sep 24.
Article in English | MEDLINE | ID: mdl-22927162

ABSTRACT

A litter of pups: The synthesis and in vitro evaluation of new Pup-based fluorogenic substrates for Dop, the mycobacterial depupylase, are described. A full-length Pup-amidomethylcoumarin conjugate as well as an amino-terminus-truncated analogue exhibited high sensitivity and specificity towards hydrolysis by Dop. The substrates developed here might find application as high-throughput screening assay reagents for the identification of Dop inhibitors.


Subject(s)
Amidohydrolases/metabolism , Bacterial Proteins/metabolism , Fluorescent Dyes/chemistry , Mycobacterium tuberculosis/drug effects , Ubiquitins/metabolism , Virulence Factors/metabolism , Amidohydrolases/antagonists & inhibitors , Amino Acid Sequence , Bacterial Proteins/antagonists & inhibitors , Biocatalysis , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Hydrolysis , Kinetics , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/chemistry , Peptides/metabolism , Substrate Specificity , Ubiquitins/antagonists & inhibitors , Virulence Factors/antagonists & inhibitors
7.
Drug Discov Today ; 17(21-22): 1175-87, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22766374

ABSTRACT

The discovery and increased understanding of tumor targets has led to the development and approval of 12 small molecule tyrosine kinase inhibitors (TKIs). Despite tremendous efforts in TKI development, treatment efficacies with these therapeutics are still too low and improvements require a personalized medicine approach. Positron emission tomography (PET) with radiolabeled TKIs (TKI-PET) is a tracking, quantification and imaging method, which provides a unique understanding of the behavior of these drugs in vivo and of the interaction with their target(s). In this article we provide an overview of tracer synthesis and development because each TKI requires a tailor made approach. Moreover, we describe current preclinical work and the first proof-of-principle clinical studies on the application of TKI-PET, illustrating the potential of this approach for improving therapy efficacy and personalized cancer treatment.


Subject(s)
Drug Design , Positron-Emission Tomography/methods , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Precision Medicine/methods , Protein Kinase Inhibitors/chemical synthesis , Protein-Tyrosine Kinases/antagonists & inhibitors , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL