Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 12(10)2020 Oct 01.
Article in English | MEDLINE | ID: mdl-33019710

ABSTRACT

The number of genomic aberrations known to be relevant in making therapeutic decisions for non-small cell lung cancer patients has increased in the past decade. Multiple molecular tests are required to reliably establish the presence of these aberrations, which is challenging because available tissue specimens are generally small. To optimize diagnostic testing, we developed a transcriptome-based next-generation sequencing (NGS) assay based on single primed enrichment technology. We interrogated 11 cell lines, two patient-derived frozen biopsies, nine pleural effusion, and 29 formalin-fixed paraffin-embedded (FFPE) samples. All clinical samples were selected based on previously identified mutations at the DNA level in EGFR, KRAS, ALK, PIK3CA, BRAF, AKT1, MET, NRAS, or ROS1 at the DNA level, or fusion genes at the chromosome level, or by aberrant protein expression of ALK, ROS1, RET, and NTRK1. A successful analysis is dependent on the number of unique reads and the RNA quality, as indicated by the DV200 value. In 27 out of 51 samples with >50 K unique reads and a DV200 >30, all 19 single nucleotide variants (SNVs)/small insertions and deletions (INDELs), three MET exon 14 skipping events, and 13 fusion gene transcripts were detected at the RNA level, giving a test accuracy of 100%. In summary, this lung-cancer-specific all-in-one transcriptome-based assay for the simultaneous detection of mutations and fusion genes is highly sensitive.

2.
Med Res Rev ; 38(6): 1713-1768, 2018 09.
Article in English | MEDLINE | ID: mdl-29528513

ABSTRACT

Successful treatment of cancer patients requires balancing of the dose, timing, and type of therapeutic regimen. Detection of increased cell death may serve as a predictor of the eventual therapeutic success. Imaging of cell death may thus lead to early identification of treatment responders and nonresponders, and to "patient-tailored therapy." Cell death in organs and tissues of the human body can be visualized, using positron emission tomography or single-photon emission computed tomography, although unsolved problems remain concerning target selection, tracer pharmacokinetics, target-to-nontarget ratio, and spatial and temporal resolution of the scans. Phosphatidylserine exposure by dying cells has been the most extensively studied imaging target. However, visualization of this process with radiolabeled Annexin A5 has not become routine in the clinical setting. Classification of death modes is no longer based only on cell morphology but also on biochemistry, and apoptosis is no longer found to be the preponderant mechanism of cell death after antitumor therapy, as was earlier believed. These conceptual changes have affected radiochemical efforts. Novel probes targeting changes in membrane permeability, cytoplasmic pH, mitochondrial membrane potential, or caspase activation have recently been explored. In this review, we discuss molecular changes in tumors which can be targeted to visualize cell death and we propose promising biomarkers for future exploration.


Subject(s)
Apoptosis , Molecular Imaging , Neoplasms/diagnostic imaging , Neoplasms/pathology , Animals , DNA Damage , DNA Repair , Humans , Membrane Potential, Mitochondrial
3.
Mol Imaging Biol ; 18(4): 588-97, 2016 08.
Article in English | MEDLINE | ID: mdl-26637208

ABSTRACT

PURPOSE: Sigma-1 receptor ligands modulate the release of several neurotransmitters and intracellular calcium signaling. We examined the binding of a radiolabeled sigma-1 agonist in the aging rat brain with positron emission tomography (PET). PROCEDURES: Time-dependent uptake of [(11)C]SA4503 was measured in the brain of young (1.5 to 3 months) and aged (18 to 32 months) Wistar Hannover rats, and tracer-kinetic models were fitted to this data, using metabolite-corrected plasma radioactivity as input function. RESULTS: In aged animals, the injected probe was less rapidly metabolized and cleared. Logan graphical analysis and a 2-tissue compartment model (2-TCM) fit indicated changes of total distribution volume (V T) and binding potential (BP ND) of the tracer. BP ND was reduced particularly in the (hypo)thalamus, pons, and medulla. CONCLUSIONS: Some areas showed reductions of ligand binding with aging whereas binding in other areas (cortex) was not significantly affected.


Subject(s)
Aging/metabolism , Brain/metabolism , Piperazines/pharmacokinetics , Positron-Emission Tomography/methods , Receptors, sigma/agonists , Animals , Brain/diagnostic imaging , Brain/pathology , Carbon Radioisotopes , Imaging, Three-Dimensional , Kinetics , Male , Metabolome , Models, Biological , Piperazines/blood , Rats, Wistar , Receptors, sigma/metabolism , Tissue Distribution , Sigma-1 Receptor
4.
Biochim Biophys Acta ; 1848(10 Pt B): 2703-14, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25173780

ABSTRACT

Sigma receptors (sigma-1 and sigma-2) represent two independent classes of proteins. Their endogenous ligands may include the hallucinogen N,N-dimethyltryptamine (DMT) and sphingolipid-derived amines which interact with sigma-1 receptors, besides steroid hormones (e.g., progesterone) which bind to both sigma receptor subpopulations. The sigma-1 receptor is a ligand-regulated molecular chaperone with various ion channels and G-protein-coupled membrane receptors as clients. The sigma-2 receptor was identified as the progesterone receptor membrane component 1 (PGRMC1). Although sigma receptors are over-expressed in tumors and up-regulated in rapidly dividing normal tissue, their ligands induce significant cell death only in tumor tissue. Sigma ligands may therefore be used to selectively eradicate tumors. Multiple mechanisms appear to underlie cell killing after administration of sigma ligands, and the signaling pathways are dependent both on the type of ligand and the type of tumor cell. Recent evidence suggests that the sigma-2 receptor is a potential tumor and serum biomarker for human lung cancer and an important target for inhibiting tumor invasion and cancer progression. Current radiochemical efforts are focused on the development of subtype-selective radioligands for positron emission tomography (PET) imaging. Right now, the mostpromising tracers are [18F]fluspidine and [18F]FTC-146 for sigma-1 receptors and [11C]RHM-1 and [18F]ISO-1 for the sigma-2 subtype. Nanoparticles coupled to sigma ligands have shown considerable potential for targeted delivery of antitumor drugs in animal models of cancer, but clinical studies exploring this strategy in cancer patients have not yet been reported. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.


Subject(s)
Antineoplastic Agents , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/diagnosis , Receptors, sigma/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Contrast Media/chemistry , Contrast Media/therapeutic use , Fluorine Radioisotopes/chemistry , Fluorine Radioisotopes/therapeutic use , Humans , Ligands , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/radiotherapy , Positron-Emission Tomography , Prognosis , Receptors, sigma/genetics , Sigma-1 Receptor
5.
Mol Cancer ; 13: 85, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24741998

ABSTRACT

BACKGROUND: Stimulation of CD40 can augment anti-cancer T cell immune responses by triggering effective activation and maturation of antigen-presenting cells (APCs). Although CD40 agonists have clinical activity in humans, the associated systemic activation of the immune system triggers dose-limiting side-effects. METHODS: To increase the tumor selectivity of CD40 agonist-based therapies, we developed an approach in which soluble trimeric CD40L (sCD40L) is genetically fused to tumor targeting antibody fragments, yielding scFv:CD40L fusion proteins. We hypothesized that scFv:CD40L fusion proteins would have reduced CD40 agonist activity similar to sCD40L but will be converted to a highly agonistic membrane CD40L-like form of CD40L upon anchoring to cell surface exposed antigen via the scFv domain. RESULTS: Targeted delivery of CD40L to the carcinoma marker EpCAM on carcinoma cells induced dose-dependent paracrine maturation of DCs ~20-fold more effective than a non-targeted control scFv:CD40L fusion protein. Similarly, targeted delivery of CD40L to the B cell leukemia marker CD20 induced effective paracrine maturation of DCs. Of note, the CD20-selective delivery of CD40L also triggered loss of cell viability in certain B cell leukemic cell lines as a result of CD20-induced apoptosis. CONCLUSIONS: Targeted delivery of CD40L to cancer cells is a promising strategy that may help to trigger cancer-localized activation of CD40 and can be modified to exert additional anti-cancer activity via the targeting domain.


Subject(s)
B-Lymphocytes/drug effects , CD40 Ligand/genetics , Dendritic Cells/drug effects , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/genetics , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, CD20/genetics , Antigens, CD20/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Antineoplastic Agents/pharmacology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , CD40 Ligand/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dendritic Cells/metabolism , Dendritic Cells/pathology , Epithelial Cell Adhesion Molecule , Gene Expression , HEK293 Cells , Humans , Molecular Targeted Therapy , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rituximab , Single-Chain Antibodies/metabolism
6.
Nucl Med Biol ; 41(2): 203-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24480269

ABSTRACT

UNLABELLED: Cerebral ß-adrenergic receptors (ß-ARs) play important roles in normal brain and changes of ß-AR expression are associated with several neuropsychiatric illnesses. Given the high density of ß-AR in several brain regions, quantification of ß-AR levels using PET is feasible. However, there is a lack of radiotracers with suitable biological properties and meeting safety requirements for use in humans. We developed a PET tracer for ß-AR by (18)F-fluorination of 1-((9H-carbazol-4-yl)oxy)-3-4(4-((2-(2-(fluoromethoxy)-ethoxy)methyl)-1H-1,2,3-triazol-1-yl)propan-2-ol ((18)F-FPTC). METHODS: [(18)F] FPTC was synthesized by Cu(I)-catalyzed alkyne-azide cycloaddition. First, (18)F-PEGylated alkyne was prepared by (18)F-fluorination of the corresponding tosylate. Next (18)F-PEGylated alkyne was reacted with an azidoalcohol derivative of 4-hydroxycarbazol in the presence of the phosphoramidite Monophos as a ligand and Cu(I) as a catalyst. After purification with radio-HPLC, the binding properties of [(18)F FPTC were tested in ß-AR-expressing C6-glioma cells in vitro and in Wistar rats in vivo using microPET. RESULTS: The radiochemical yield of (18)F-PEGylated alkyne was 74%-89%. The click reaction to prepare [(18)F]FPTC proceeded in 10min with a conversion efficiency of 96%. The total synthesis time was 55min from the end of bombardment. Specific activities were >120GBq/µmol. Propranolol strongly and dose-dependently inhibited the binding of both [(125)I]-ICYP and [(18)F]FPTC to C6 glioma cells, with IC50 values in the 50-60 nM range. However, although both FPTC and propranolol inhibited cellular [(125)I]ICYP binding, FPTC decreased [(125)I]ICYP uptake by only 25%, whereas propranolol reduced it by 83%. [(18)F]FPTC has the appropriate lipophilicity to penetrate the blood brain barrier (logP +2.48). The brain uptake reached a maximum within 2min after injection of 20-25MBq [(18)F]FPTC. SUV values ranged from 0.4 to 0.6 and were not reduced by propranolol. Cerebral distribution volume of the tracer (calculated from a Logan plot) was increased rather than decreased after propranolol treatment. CONCLUSION: 'Click chemistry' was successfully applied to the synthesis of [(18)F]FPTC resulting in high radiochemical yields. [(18)F]FPTC showed specific binding in vitro, but not in vivo. Based on the logP value and its ability to block [(125)I]ICYP binding to C6 cells, FPTC may be a lead to suitable cerebral ß-AR ligands.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Carbazoles/metabolism , Positron-Emission Tomography/methods , Receptors, Adrenergic, beta/metabolism , Triazoles/metabolism , Animals , Carbazoles/pharmacokinetics , Cell Line, Tumor , Kinetics , Ligands , Male , Rats , Substrate Specificity , Triazoles/pharmacokinetics
7.
J Nucl Med ; 54(9): 1613-20, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23940303

ABSTRACT

UNLABELLED: σ-ligands can kill tumor cells. Previously we have shown that a short in vitro incubation of C6 tumor cells with σ-ligands (24 h) results in a dose-dependent increase of cellular (18)F-FDG uptake and that the magnitude of this increase is predictive of subsequent cell death. Here, we aimed to assess whether the σ-ligand rimcazole inhibits growth of A375M melanoma xenografts in nude mice and whether rimcazole treatment changes (18)F-FDG uptake in vivo. METHODS: Athymic mice were inoculated with A375M melanoma cells. After 2 wk, tumors had reached a size of 41 ± 6 mm(3). We then started a 14-d treatment schedule with daily drug dosing. Control animals were injected with water and treated animals with rimcazole (26 mg/kg) in water. Three small-animal PET scans with (18)F-FDG were obtained: on days 0, 7, and 14 of treatment. After the last scan, animals were terminated, and a biodistribution study was performed. RESULTS: Rimcazole treatment resulted in a greater than 4-fold reduction of tumor weight in comparison to controls at day 14 (100 ± 26 vs. 436 ± 117 mg, respectively, P < 0.03). Treatment did not affect the levels of (nonradioactive) glucose in blood, σ-1 and σ-2 receptor expression in the tumor, animal weight, behavior, or appearance. Antitumor activity of rimcazole was accompanied by a transient increase of the tumor uptake of (18)F-FDG (measured at day 7). Significant increases of (18)F-FDG uptake at day 14 were observed in the liver and pancreas. CONCLUSION: Rimcazole strongly inhibited the growth of A375M melanoma xenografts. This growth inhibition is accompanied by an early increase of (18)F-FDG uptake in the tumor.


Subject(s)
Carbazoles/administration & dosage , Fluorodeoxyglucose F18/pharmacokinetics , Melanoma/diagnostic imaging , Melanoma/metabolism , Positron-Emission Tomography/methods , Receptors, sigma/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Humans , Metabolic Clearance Rate/drug effects , Mice , Mice, Nude , Organ Specificity , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution , Treatment Outcome
8.
J Nucl Med ; 54(8): 1377-83, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23785170

ABSTRACT

UNLABELLED: Pituitary tumors are often detected only after death or at late stages of the disease when they are macroadenomas with a low surgical cure rate. Spontaneous pituitary tumors occur in rats over 1 y of age. In an ongoing study of changes in σ-1 agonist binding related to aging, several of our rats developed such tumors. The aim of the current study was to assess the kinetics of (11)C-SA4503 ((11)C-labeled 1-[2-(3,4-dimethoxyphenthyl)]-4-(3-phenylpropyl)-piperazine dihydrochloride) in tumor and brain and to evaluate the utility of this tracer in the detection of pituitary tumors. METHODS: Small-animal PET scans of the brain region of male Wistar Hannover rats (age, 18-32 mo) were acquired using the σ-1 agonist tracer (11)C-SA4503. The time-dependent uptake of (11)C in the entire brain, tumor or normal pituitary, and thyroid was measured. A 2-tissue-compartment model was fitted to the PET data, using metabolite-corrected plasma radioactivity as the input function. RESULTS: Pituitary tumors showed up as bright hot spots in the scans. The total distribution volume (VT) of the tracer was significantly higher in the tumor than in the normal pituitary. Surprisingly, a higher VT was also seen in the brain and thyroid tissue of animals with pituitary tumors than in healthy rats. The increase in VT in the brain and thyroid was not related to a change in nondisplaceable binding potential (BPND) but rather to an increase in the partition coefficient (K1/k2) of (11)C-SA4503. The increase in VT in the tumor on the other hand was accompanied by a significant increase in BPND. Western blotting analysis indicated that pituitary tumors overexpressed σ-1 receptors. CONCLUSION: The overexpression of σ-1 receptors in spontaneous pituitary tumors is detected as an increase in uptake and BPND of (11)C-SA4503. Therefore, this tracer may have promise for the detection of pituitary adenomas, using PET.


Subject(s)
Aging , Piperazines/metabolism , Pituitary Neoplasms/diagnostic imaging , Positron-Emission Tomography/methods , Receptors, sigma/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Kinetics , Ligands , Male , Piperazines/blood , Piperazines/pharmacokinetics , Pituitary Neoplasms/metabolism , Rats , Rats, Wistar , Tissue Distribution , Sigma-1 Receptor
9.
J Nucl Med ; 52(8): 1293-300, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21764782

ABSTRACT

UNLABELLED: Activation of adenosine A(1) receptors (A(1)R) in the brain causes sedation, reduces anxiety, inhibits seizures, and promotes neuroprotection. Cerebral A(1)R can be visualized using 8-dicyclopropylmethyl-1-(11)C-methyl-3-propyl-xanthine ((11)C-MPDX) and PET. This study aims to test whether (11)C-MPDX can be used for quantitative studies of cerebral A(1)R in rodents. METHODS: (11)C-MPDX was injected (intravenously) into isoflurane-anesthetized male Wistar rats (300 g). A dynamic scan of the central nervous system was obtained, using a small-animal PET camera. A cannula in a femoral artery was used for blood sampling. Three groups of animals were studied: group 1, controls (saline-treated); group 2, animals pretreated with the A(1)R antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX, 1 mg, intraperitoneally); and group 3, animals pretreated (intraperitoneally) with a 20% solution of ethanol in saline (2 mL) plus the adenosine kinase inhibitor 4-amino-5-(3-bromophenyl)-7-(6-morpholino-pyridin-3-yl)pyrido[2,3-d] pyrimidine dihydrochloride (ABT-702) (1 mg). DPCPX is known to occupy cerebral A(1)R, whereas ethanol and ABT-702 increase extracellular adenosine. RESULTS: In groups 1 and 3, the brain was clearly visualized. High uptake of (11)C-MPDX was noted in striatum, hippocampus, and cerebellum. In group 2, tracer uptake was strongly suppressed and regional differences were abolished. The treatment of group 3 resulted in an unexpected 40%-45% increase of the cerebral uptake of radioactivity as indicated by increases of PET standardized uptake value, distribution volume from Logan plot, nondisplaceable binding potential from 2-tissue-compartment model fit, and standardized uptake value from a biodistribution study performed after the PET scan. The partition coefficient of the tracer (K(1)/k(2) from the model fit) was not altered under the study conditions. CONCLUSION: (11)C-MPDX shows a regional distribution in rat brain consistent with binding to A(1)R. Tracer binding is blocked by the selective A(1)R antagonist DPCPX. Pretreatment of animals with ethanol and adenosine kinase inhibitor increases (11)C-MPDX uptake. This increase may reflect an increased availability of A(1)R after acute exposure to ethanol.


Subject(s)
Adenosine/metabolism , Brain/metabolism , Positron-Emission Tomography/methods , Receptor, Adenosine A1/biosynthesis , Adenosine Kinase/metabolism , Animals , Carbon Isotopes/pharmacology , Ethanol/chemistry , Kinetics , Ligands , Male , Rats , Rats, Wistar , Tissue Distribution , Xanthines/pharmacology
10.
Behav Brain Res ; 221(2): 543-54, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-20060423

ABSTRACT

This article provides an overview of present knowledge regarding the relationship between the cholinergic system and sigma-1 receptors, and discusses potential applications of sigma-1 receptor agonists in the treatment of memory deficits and cognitive disorders. Sigma-1 receptors, initially considered as a subtype of the opioid family, are unique ligand-regulated molecular chaperones in the endoplasmatic reticulum playing a modulatory role in intracellular calcium signaling and in the activity of several neurotransmitter systems, particularly the cholinergic and glutamatergic pathways. Several central nervous system (CNS) drugs show high to moderate affinities for sigma-1 receptors, including acetylcholinesterase inhibitors (donepezil), antipsychotics (haloperidol, rimcazole), selective serotonin reuptake inhibitors (fluvoxamine, sertraline) and monoamine oxidase inhibitors (clorgyline). These compounds can influence cognitive functions both via their primary targets and by activating sigma-1 receptors in the CNS. Sigma-1 agonists show powerful anti-amnesic and neuroprotective effects in a large variety of animal models of cognitive dysfunction involving, among others (i) pharmacologic target blockade (with muscarinic or NMDA receptor antagonists or p-chloroamphetamine); (ii) selective lesioning of cholinergic neurons; (iii) CNS administration of ß-amyloid peptides; (iv) aging-induced memory loss, both in normal and senescent-accelerated rodents; (v) neurodegeneration induced by toxic compounds (CO, trimethyltin, cocaine), and (vi) prenatal restraint stress.


Subject(s)
Cholinergic Fibers/physiology , Cognition/physiology , Neurodegenerative Diseases/drug therapy , Psychotropic Drugs/therapeutic use , Receptors, sigma/physiology , Acetylcholine/physiology , Aging/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/physiology , Calcium/metabolism , Cell Differentiation/physiology , Cognition/drug effects , Disease Models, Animal , Glutamic Acid/metabolism , Humans , N-Methylaspartate/metabolism , Neurodegenerative Diseases/metabolism , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/metabolism , Psychotropic Drugs/pharmacology , Receptors, sigma/agonists , Receptors, sigma/metabolism , Sigma-1 Receptor
11.
Mol Cancer ; 9: 301, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21092273

ABSTRACT

BACKGROUND: Advanced melanoma is characterized by a pronounced resistance to therapy leading to a limited patient survival of ~6 - 9 months. Here, we report on a novel bifunctional therapeutic fusion protein, designated anti-MCSP:TRAIL, that is comprised of a melanoma-associated chondroitin sulfate proteoglycan (MCSP)-specific antibody fragment (scFv) fused to soluble human TRAIL. MCSP is a well-established target for melanoma immunotherapy and has recently been shown to provide important tumorigenic signals to melanoma cells. TRAIL is a highly promising tumoricidal cytokine with no or minimal toxicity towards normal cells. Anti-MCSP:TRAIL was designed to 1. selectively accrete at the cell surface of MCSP-positive melanoma cells and inhibit MCSP tumorigenic signaling and 2. activate apoptotic TRAIL-signaling. RESULTS: Treatment of a panel of MCSP-positive melanoma cell lines with anti-MCSP:TRAIL induced TRAIL-mediated apoptotic cell death within 16 h. Of note, treatment with anti-MCSP:sTRAIL was also characterized by a rapid dephosphorylation of key proteins, such as FAK, implicated in MCSP-mediated malignant behavior. Importantly, anti-MCSP:TRAIL treatment already inhibited anchorage-independent growth by 50% at low picomolar concentrations, whereas > 100 fold higher concentrations of non-targeted TRAIL failed to reduce colony formation. Daily i.v. treatment with a low dose of anti-MCSP:TRAIL (0.14 mg/kg) resulted in a significant growth retardation of established A375 M xenografts. Anti-MCSP:TRAIL activity was further synergized by co-treatment with rimcazole, a σ-ligand currently in clinical trials for the treatment of various cancers. CONCLUSIONS: Anti-MCSP:TRAIL has promising pre-clinical anti-melanoma activity that appears to result from combined inhibition of tumorigenic MCSP-signaling and concordant activation of TRAIL-apoptotic signaling. Anti-MCSP:TRAIL alone, or in combination with rimcazole, may be of potential value for the treatment of malignant melanoma.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Melanoma/drug therapy , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Animals , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Cells, Cultured , Chondroitin Sulfate Proteoglycans/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Melanocytes/drug effects , Melanocytes/metabolism , Melanoma/metabolism , Mice , Mice, Nude , Proteome/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Xenograft Model Antitumor Assays
12.
Curr Pharm Des ; 16(31): 3519-37, 2010.
Article in English | MEDLINE | ID: mdl-21050178

ABSTRACT

Sigma receptors (subtypes sigma-1 and sigma-2) are a unique class of binding sites expressed throughout the mammalian body. The endogenous ligand for these sites has not been identified, but steroid hormones (particularly progesterone), sphingolipid-derived amines and N,N-dimethyltryptamine can bind with fairly high affinity. Sigma receptors are overexpressed in rapidly proliferating cells, like cancer cells. Particularly the sigma-2 subtype is upregulate when cells divide and down regulated when they become quiescent. Sigma ligands, especially sigma-2 agonists, can inhibit proliferation and induce apoptosis by a mechanism involving changes in cytosolic Ca(2+), ceramide and sphingolipid levels. Tumor cells are much more sensitive to such treatment than cells from their tissue of origin. Sigma ligands induce apoptosis not only in drug-sensitive but also in drug-resistant cancer cells (e.g., cells with p53 mutations, or caspase dysfunction). Moreover, sigma ligands may abrogate P-glycoprotein-mediated drug resistance and at subtoxic doses, they can potentiate the effect of conventional cytostatics. Thus, sigma-2 agonists may be developed as antineoplastic agents for the treatment of drug-resistant tumors. A large number of radiolabeled sigma ligands has been prepared for SPECT (single-photon emission computed tomography) and PET (positron emission tomography) imaging. Such radiopharmaceuticals can be used for tumor detection, tumor staging, and evaluation of anti-tumor therapy. There is still a need for the development of ligands with (1) high selectivity for the sigma-2 subtype, (2) defined action (agonist or antagonist) and (3) optimal pharmacokinetics (low affinity for P-glycoprotein, high and specific tumor uptake, and rapid washout from non-target tissues).


Subject(s)
Ligands , Neoplasms/diagnosis , Neoplasms/metabolism , Receptors, sigma/metabolism , Receptors, sigma/physiology , Animals , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor/metabolism , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Neoplasms/diagnostic imaging , Positron-Emission Tomography/methods , Radiopharmaceuticals , Receptors, sigma/agonists , Tomography, Emission-Computed, Single-Photon/methods
13.
J Med Chem ; 52(14): 4524-32, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19530699

ABSTRACT

UNLABELLED: P-glycoprotein (P-gp) is an ATP-dependent efflux pump protecting the body against xenobiotics. A P-gp substrate (7) and an inhibitor (6) were labeled with (11)C, resulting in potential tracers of P-gp function and expression. METHODS: 6 and 7 were labeled using (11)CH(3)I. (11)C-verapamil was prepared as published previously, using (11)C-methyl triflate. MicroPET scans (with arterial sampling) and biodistribution studies were performed in rats pretreated with saline, cyclosporin A (CsA, 50 mg/kg), or cold 6 (15 mg/kg). RESULTS: The radiochemical yields of (11)C-6 and (11)C-7 were approximately 30% with a total synthesis time of 45 min. Cerebral distribution volumes (DV) of (11)C-6 (2.35 +/- 0.11) and (11)C-7 (1.86 +/- 0.15) in saline-treated rats were higher than of (11)C-verapamil (0.64 +/- 0.12). DVs of (11)C-7 and (11)C-verapamil were significantly increased by CsA (to 5.26 +/- 0.14 and 5.85 +/- 0.32, respectively). The DV of (11)C-6 was reduced by cold 6 (to 1.65 +/- 0.03). Its uptake was also reduced (up to 67%) in several peripheral organs that express P-gp. CONCLUSIONS: (11)C-7 is a novel tracer of P-gp function with higher baseline uptake than (11)C-verapamil. Upregulation of P-gp function in response to treatment (which is hard to detect with (11)C-verapamil) may be detectable using (11)C-7 and PET. Because (11)C-6 shows specific binding in target organs, this compound is the first PET tracer allowing measurement of P-gp expression.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Gene Expression Regulation , Isoquinolines , Animals , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes , Drug Evaluation, Preclinical , Isoquinolines/chemical synthesis , Isoquinolines/chemistry , Isoquinolines/pharmacokinetics , Kinetics , Male , Positron-Emission Tomography , Radioactive Tracers , Radioactivity , Rats , Rats, Wistar , Staining and Labeling , Tissue Distribution
14.
Eur J Nucl Med Mol Imaging ; 36(7): 1167-75, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19247652

ABSTRACT

PURPOSE: Sigma receptors are implicated in memory and cognitive functions, drug addiction, depression and schizophrenia. In addition, sigma receptors are strongly overexpressed in many tumours. Although the natural ligands are still unknown, steroid hormones are potential candidates. Here, we examined changes in binding of the sigma-1 agonist (11)C-SA4503 in C6 glioma cells and in living rats after modification of endogenous steroid levels. METHODS: (11)C-SA4503 binding was assessed in C6 monolayers by gamma counting and in anaesthetized rats by microPET scanning. C6 cells were either repeatedly washed and incubated in steroid-free medium or exposed to five kinds of exogenous steroids (1 h or 5 min before tracer addition, respectively). Tumour-bearing male rats were repeatedly treated with pentobarbital (a condition known to result in reduction of endogenous steroid levels) or injected with progesterone. RESULTS: Binding of (11)C-SA4503 to C6 cells was increased (approximately 50%) upon removal and decreased (approximately 60%) upon addition of steroid hormones (rank order of potency: progesterone > allopregnanolone = testosterone = androstanolone > dehydroepiandrosterone-3-sulphate, IC(50) progesterone 33 nM). Intraperitoneally administered progesterone reduced tumour uptake and tumour-to-muscle contrast (36%). Repeated treatment of animals with pentobarbital increased the PET standardized uptake value of (11)C-SA4503 in tumour (16%) and brain (27%), whereas the kinetics of blood pool radioactivity was unaffected. CONCLUSIONS: The binding of (11)C-SA4503 is sensitive to steroid competition. Since not only increases but also decreases of steroid levels affect ligand binding, a considerable fraction of the sigma-1 receptor population in cultured tumour cells or tumour-bearing animals is normally occupied by endogenous steroids.


Subject(s)
Hormones/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Piperazines/metabolism , Receptors, sigma/metabolism , Steroids/metabolism , Animals , Binding, Competitive , Cell Line, Tumor , Disease Models, Animal , Ligands , Neoplasms/diagnostic imaging , Piperazines/pharmacokinetics , Piperazines/pharmacology , Positron-Emission Tomography , Rats , Receptors, sigma/agonists , Tissue Distribution
15.
J Nucl Med ; 49(12): 2049-56, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18997057

ABSTRACT

UNLABELLED: Tumor cells can be selectively killed by application of sigma-ligands; high concentrations (20-100 muM) are often required, however, because either diffusion barriers must be passed to reach intracellular sites or the entire sigma-receptor population should be occupied to induce cell death. We measured receptor occupancies associated with the cytotoxic effect and dose-dependent changes of cellular metabolism in a tumor cell line. METHODS: C6 cells (rat glioma) were grown in monolayers and exposed to (+)-pentazocine (sigma-1 agonist), AC915 (sigma-1 antagonist), rimcazole (sigma-1/sigma-2 antagonist), or haloperidol (sigma-1/sigma-2 antagonist). Occupancy of sigma-receptors by the test drugs was measured by studying the competition of the test drugs with cellular binding of the ligand (11)C-SA4503. Metabolic changes were quantified by measuring cellular uptake of (18)F-FDG, (18)F-FLT, (11)C-choline, or (11)C-methionine. Cytotoxicity was assessed by cellular morphology observation and cell counting after 24 h. RESULTS: IC(50) values (drug concentrations resulting in 50% occupancy of the available binding sites) of the test drugs for inhibition of cellular (11)C-SA4503 binding were 6.5, 7.4, 0.36, and 0.27 muM for (+)-pentazocine, AC915, rimcazole, and haloperidol, respectively. EC(50) values (dose required for a 50% reduction of cell number after 24 h) were 710, 819, 31, and 58 muM, for pentazocine, AC915, rimcazole, and haloperidol, respectively. Cytotoxic doses of sigma-ligands were generally associated with increased uptake of (18)F-FDG, decreased uptake of (18)F-FLT and (11)C-choline, and little change in (11)C-methionine uptake per viable cell. CONCLUSION: IC(50) values of the test drugs reflect their in vitro affinities to sigma-2 rather than to sigma-1 receptors. Because cytotoxicity occurred at concentrations 2 orders of magnitude higher than IC(50) values for inhibition of cellular (11)C-SA4503 binding, high (99%) occupancy of sigma-2 receptors is associated with loss of cell viability. (18)F-FLT, (11)C-choline, and (18)F-FDG responded most strongly to drug treatment and showed changes corresponding to the cytotoxicity of the test compounds.


Subject(s)
Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Glioma/metabolism , Glioma/pathology , Receptors, sigma/antagonists & inhibitors , Receptors, sigma/metabolism , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Lethal Dose 50 , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...