Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Biopharm Drug Dispos ; 44(1): 48-59, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36825693

ABSTRACT

PF-05212377 (SAM760) is a potent and selective 5-HT6 antagonist, previously under development for the treatment of Alzheimer's disease. In vitro, PF-05212377 was determined to be a P-gp/non-BCRP human transporter substrate. Species differences were observed in the in vivo brain penetration of PF-05212377 with a ratio of the unbound concentration in brain/unbound concentration in plasma (Cbu /Cpu ) of 0.05 in rat and 0.64 in non-human primates (NHP). Based on pre-clinical evidence, brain penetration and target engagement of PF-05212377 was confirmed in NHP using positron emission tomography (PET) measured 5-HT6 receptor occupancy (%RO). The NHP Cpu EC50 of PF-05212377 was 0.31 nM (consistent with the in vitro human 5HT6 Ki : 0.32 nM). P-gp has been reported to be expressed in higher abundance at the rat BBB and in similar abundance at the BBB of non-human primates and human; brain penetration of PF-05212377 in humans was postulated to be similar to that in non-human primates. In humans, PF-05212377 demonstrated dose and concentration dependent increases in 5-HT6 RO; maximal 5-HT6 RO of ∼80% was measured in humans at doses of ≥15 mg with an estimated unbound plasma EC50 of 0.37 nM (which was similar to the in vitro human 5HT6 binding Ki 0.32 nM). In conclusion, cumulative evidence from NHP and human PET RO assessments confirmed that NHP is more appropriate than the rat for the prediction of human brain penetration of PF-05212377, a P-gp/non-BCRP substrate. Clinical trial number: NCT01258751.


Subject(s)
Brain , Serotonin , Humans , Rats , Animals , Serotonin/metabolism , Brain/diagnostic imaging , Brain/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Primates/metabolism
2.
JCI Insight ; 4(20)2019 10 17.
Article in English | MEDLINE | ID: mdl-31619586

ABSTRACT

Intrathecal (IT) delivery and pharmacology of antisense oligonucleotides (ASOs) for the CNS have been successfully developed to treat spinal muscular atrophy. However, ASO pharmacokinetic (PK) and pharmacodynamic (PD) properties remain poorly understood in the IT compartment. We applied multimodal imaging techniques to elucidate the IT PK and PD of unlabeled, radioactively labeled, or fluorescently labeled ASOs targeting ubiquitously expressed or neuron-specific RNAs. Following lumbar IT bolus injection in rats, all ASOs spread rostrally along the neuraxis, adhered to meninges, and were partially cleared to peripheral lymph nodes and kidneys. Rapid association with the pia and arterial walls preceded passage of ASOs across the glia limitans, along arterial intramural basement membranes, and along white-matter axonal bundles. Several neuronal and glial cell types accumulated ASOs over time, with evidence of probable glial accumulation preceding neuronal uptake. IT doses of anti-GluR1 and anti-Gabra1 ASOs markedly reduced the mRNA and protein levels of their respective neurotransmitter receptor protein targets by 2 weeks and anti-Gabra1 ASOs also reduced binding of the GABAA receptor PET ligand 18F-flumazenil in the brain over 4 weeks. Our multimodal imaging approaches elucidate multiple transport routes underlying the CNS distribution, clearance, and efficacy of IT-dosed ASOs.


Subject(s)
Brain/metabolism , GABA-A Receptor Antagonists/pharmacokinetics , Muscular Atrophy, Spinal/drug therapy , Oligonucleotides, Antisense/pharmacokinetics , Animals , Arteries/diagnostic imaging , Arteries/metabolism , Brain/blood supply , Brain/cytology , Brain/diagnostic imaging , Flumazenil/administration & dosage , Flumazenil/analogs & derivatives , GABA-A Receptor Antagonists/administration & dosage , Gene Knockdown Techniques , Humans , Injections, Spinal , Intravital Microscopy , Male , Molecular Targeted Therapy/methods , Neuroglia/metabolism , Neurons/metabolism , Oligonucleotides, Antisense/administration & dosage , Pia Mater/diagnostic imaging , Pia Mater/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Receptors, AMPA/analysis , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/genetics , Receptors, GABA-A/analysis , Receptors, GABA-A/genetics , Single Photon Emission Computed Tomography Computed Tomography , Spatio-Temporal Analysis , Thionucleotides/administration & dosage , Thionucleotides/pharmacokinetics , Tissue Distribution
3.
J Med Chem ; 60(20): 8538-8551, 2017 10 26.
Article in English | MEDLINE | ID: mdl-28957634

ABSTRACT

As part of our effort in identifying phosphodiesterase (PDE) 4B-preferring inhibitors for the treatment of central nervous system (CNS) disorders, we sought to identify a positron emission tomography (PET) ligand to enable target occupancy measurement in vivo. Through a systematic and cost-effective PET discovery process, involving expression level (Bmax) and biodistribution determination, a PET-specific structure-activity relationship (SAR) effort, and specific binding assessment using a LC-MS/MS "cold tracer" method, we have identified 8 (PF-06445974) as a promising PET lead. Compound 8 has exquisite potency at PDE4B, good selectivity over PDE4D, excellent brain permeability, and a high level of specific binding in the "cold tracer" study. In subsequent non-human primate (NHP) PET imaging studies, [18F]8 showed rapid brain uptake and high target specificity, indicating that [18F]8 is a promising PDE4B-preferring radioligand for clinical PET imaging.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Phosphodiesterase Inhibitors/metabolism , Positron-Emission Tomography/methods , Animals , Cerebral Cortex/metabolism , Chromatography, Liquid , Drug Discovery , Macaca fascicularis , Radioligand Assay , Structure-Activity Relationship , Tandem Mass Spectrometry
4.
J Med Chem ; 60(18): 7764-7780, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28817277

ABSTRACT

We previously observed a cutaneous type IV immune response in nonhuman primates (NHP) with the mGlu5 negative allosteric modulator (NAM) 7. To determine if this adverse event was chemotype- or mechanism-based, we evaluated a distinct series of mGlu5 NAMs. Increasing the sp3 character of high-throughput screening hit 40 afforded a novel morpholinopyrimidone mGlu5 NAM series. Its prototype, (R)-6-neopentyl-2-(pyridin-2-ylmethoxy)-6,7-dihydropyrimido[2,1-c][1,4]oxazin-4(9H)-one (PF-06462894, 8), possessed favorable properties and a predicted low clinical dose (2 mg twice daily). Compound 8 did not show any evidence of immune activation in a mouse drug allergy model. Additionally, plasma samples from toxicology studies confirmed that 8 did not form any reactive metabolites. However, 8 caused the identical microscopic skin lesions in NHPs found with 7, albeit with lower severity. Holistically, this work supports the hypothesis that this unique toxicity may be mechanism-based although additional work is required to confirm this and determine clinical relevance.


Subject(s)
Allosteric Regulation/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Pyridines/pharmacology , Pyridines/pharmacokinetics , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Receptor, Metabotropic Glutamate 5/metabolism , Animals , Female , HEK293 Cells , Heterocyclic Compounds, 3-Ring/adverse effects , Heterocyclic Compounds, 3-Ring/chemistry , Humans , Male , Molecular Docking Simulation , Pyridines/adverse effects , Pyridines/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
5.
J Nucl Med ; 57(9): 1448-53, 2016 09.
Article in English | MEDLINE | ID: mdl-27199356

ABSTRACT

UNLABELLED: The enzyme phosphodiesterase 2A (PF-05270430) is a potential target for development of novel therapeutic agents for the treatment of cognitive impairments. The goal of the present study was to evaluate the PDE2A ligand (18)F-PF-05270430, 4-(3-fluoroazetidin-1-yl)-7-methyl-5-(1-methyl-5-(4-(trifluoromethyl)phenyl)-1H-pyrazol-4-yl)imidazo[1,5-f][1,2,4]triazine, in nonhuman primates. METHODS: (18)F-PF-05270430 was radiolabeled by 2 methods via nucleophilic substitution of its tosylate precursor. Tissue metabolite analysis in rodents and PET imaging in nonhuman primates under baseline and blocking conditions were performed to determine the pharmacokinetic and binding characteristics of the new radioligand. Various kinetic modeling approaches were assessed to select the optimal method for analysis of imaging data. RESULTS: (18)F-PF-05270430 was synthesized in greater than 98% radiochemical purity and high specific activity. In the nonhuman primate brain, uptake of (18)F-PF-05270430 was fast, with peak concentration (SUVs of 1.5-1.8 in rhesus monkeys) achieved within 7 min after injection. The rank order of uptake was striatum > neocortical regions > cerebellum. Regional time-activity curves were well fitted by the 2-tissue-compartment model and the multilinear analysis-1 (MA1) method to arrive at reliable estimates of regional distribution volume (VT) and binding potential (BPND) with 120 min of scan data. Regional VT values (MA1) ranged from 1.28 mL/cm(3) in the cerebellum to 3.71 mL/cm(3) in the putamen, with a BPND of 0.25 in the temporal cortex and 1.92 in the putamen. Regional BPND values estimated by the simplified reference tissue model (SRTM) were similar to those from MA1. Test-retest variability in high-binding regions (striatum) was 4% ± 6% for MA1 VT, 13% ± 6% for MA1 BPND, and 13% ± 7% SRTM BPND, respectively. Pretreatment of animals with the PDE2A inhibitor PF-05180999 resulted in a dose-dependent reduction of (18)F-PF-05270430 specific binding, with a half maximal effective concentration of 69.4 ng/mL in plasma PF-05180999 concentration. CONCLUSION: (18)F-PF-05270430 displayed fast and reversible kinetics in nonhuman primates, as well as specific binding blockable by a PDE2A inhibitor. This is the first PET tracer with desirable imaging properties and demonstrated ability to image and quantify PDE2A in vivo.


Subject(s)
Azabicyclo Compounds/pharmacokinetics , Azetidines/pharmacokinetics , Brain/diagnostic imaging , Brain/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Molecular Imaging/methods , Positron-Emission Tomography/methods , Animals , Macaca mulatta , Male , Metabolic Clearance Rate , Organ Specificity , Radiopharmaceuticals , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity , Species Specificity , Tissue Distribution
6.
J Med Chem ; 58(10): 4291-308, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25905800

ABSTRACT

A unique tetrahydrofuran ether class of highly potent α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor potentiators has been identified using rational and structure-based drug design. An acyclic lead compound, containing an ether-linked isopropylsulfonamide and biphenyl group, was pharmacologically augmented by converting it to a conformationally constrained tetrahydrofuran to improve key interactions with the human GluA2 ligand-binding domain. Subsequent replacement of the distal phenyl motif with 2-cyanothiophene to enhance its potency, selectivity, and metabolic stability afforded N-{(3S,4S)-4-[4-(5-cyano-2-thienyl)phenoxy]tetrahydrofuran-3-yl}propane-2-sulfonamide (PF-04958242, 3), whose preclinical characterization suggests an adequate therapeutic index, aided by low projected human oral pharmacokinetic variability, for clinical studies exploring its ability to attenuate cognitive deficits in patients with schizophrenia.


Subject(s)
Drug Evaluation, Preclinical/methods , Receptors, AMPA/metabolism , Sulfonamides/pharmacology , Thiophenes/pharmacology , Administration, Oral , Adolescent , Adult , Aged , Animals , Binding Sites , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Drug Discovery , Drug Stability , Female , Humans , Male , Memory, Short-Term/drug effects , Mice, Inbred C57BL , Middle Aged , Protein Conformation , Rats, Sprague-Dawley , Schizophrenia/drug therapy , Structure-Activity Relationship , Sulfonamides/chemistry , Thiophenes/chemistry , Young Adult
7.
J Med Chem ; 57(3): 861-77, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24392688

ABSTRACT

A novel series of pyrazolopyrazines is herein disclosed as mGluR5 negative allosteric modulators (NAMs). Starting from a high-throughput screen (HTS) hit (1), a systematic structure-activity relationship (SAR) study was conducted with a specific focus on balancing pharmacological potency with physicochemical and pharmacokinetic (PK) properties. This effort led to the discovery of 1-methyl-3-(4-methylpyridin-3-yl)-6-(pyridin-2-ylmethoxy)-1H-pyrazolo[3,4-b]pyrazine (PF470, 14) as a highly potent, selective, and orally bioavailable mGluR5 NAM. Compound 14 demonstrated robust efficacy in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-rendered Parkinsonian nonhuman primate model of l-DOPA-induced dyskinesia (PD-LID). However, the progression of 14 to the clinic was terminated because of a potentially mechanism-mediated finding consistent with a delayed-type immune-mediated type IV hypersensitivity in a 90-day NHP regulatory toxicology study.


Subject(s)
Pyrazines/chemical synthesis , Pyrazoles/chemical synthesis , Receptor, Metabotropic Glutamate 5/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Administration, Oral , Allosteric Regulation , Animals , Antiparkinson Agents/adverse effects , Biological Availability , Cell Membrane Permeability , Dogs , Dyskinesia, Drug-Induced/drug therapy , HEK293 Cells , Humans , Hypersensitivity, Delayed/chemically induced , Levodopa/adverse effects , Macaca fascicularis , Madin Darby Canine Kidney Cells , Male , Microsomes, Liver/metabolism , Models, Molecular , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Parkinson Disease/physiopathology , Pyrazines/pharmacology , Pyrazines/toxicity , Pyrazoles/pharmacology , Pyrazoles/toxicity , Radioligand Assay , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
8.
J Med Chem ; 56(11): 4568-79, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23651455

ABSTRACT

To accelerate the discovery of novel small molecule central nervous system (CNS) positron emission tomography (PET) ligands, we aimed to define a property space that would facilitate ligand design and prioritization, thereby providing a higher probability of success for novel PET ligand development. Toward this end, we built a database consisting of 62 PET ligands that have successfully reached the clinic and 15 radioligands that failed in late-stage development as negative controls. A systematic analysis of these ligands identified a set of preferred parameters for physicochemical properties, brain permeability, and nonspecific binding (NSB). These preferred parameters have subsequently been applied to several programs and have led to the successful development of novel PET ligands with reduced resources and timelines. This strategy is illustrated here by the discovery of the novel phosphodiesterase 2A (PDE2A) PET ligand 4-(3-[(18)F]fluoroazetidin-1-yl)-7-methyl-5-{1-methyl-5-[4-(trifluoromethyl)phenyl]-1H-pyrazol-4-yl}imidazo[5,1-f][1,2,4]triazine, [(18)F]PF-05270430 (5).


Subject(s)
Azabicyclo Compounds/chemical synthesis , Azetidines/chemical synthesis , Brain/diagnostic imaging , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Radiopharmaceuticals/chemical synthesis , Animals , Azabicyclo Compounds/chemistry , Azabicyclo Compounds/pharmacokinetics , Azetidines/chemistry , Azetidines/pharmacokinetics , Brain/enzymology , Computer Simulation , Databases, Factual , Dogs , Drug Design , Fluorine Radioisotopes , Humans , Ligands , Macaca fascicularis , Male , Models, Biological , Permeability , Positron-Emission Tomography , Protein Binding , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Wistar , Structure-Activity Relationship
9.
Nucl Med Biol ; 39(7): 1058-67, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22571907

ABSTRACT

INTRODUCTION: Fatty acid amide hydrolase (FAAH) is responsible for the enzymatic degradation of the fatty acid amide family of signaling lipids, including the endogenous cannabinoid (endocannabinoid) anandamide. The involvement of the endocannabinoid system in pain and other nervous system disorders has made FAAH an attractive target for drug development. Companion molecular imaging probes are needed, however, to assess FAAH inhibition in the nervous system in vivo. We report here the synthesis and in vivo evaluation of [(18)F]PF-9811, a novel PET ligand for non-invasive imaging of FAAH in the brain. METHODS: The potency and selectivity of unlabeled PF-9811 were determined by activity-based protein profiling (ABPP) both in vitro and in vivo. [(18)F]PF-9811 was synthesized in a 3-step, one-pot reaction sequence, followed by HPLC purification. Biological evaluation was performed by biodistribution and dynamic PET imaging studies in male rats. The specificity of [(18)F]PF-9811 uptake was evaluated by pre-administration of PF-04457845, a potent and selective FAAH inhibitor, 1h prior to radiotracer injection. RESULTS: Biodistribution studies show good uptake (SUV~0.8 at 90 min) of [(18)F]PF-9811 in rat brain, with significant reduction of the radiotracer in all brain regions (37%-73% at 90 min) in blocking experiments. Dynamic PET imaging experiments in rat confirmed the heterogeneous uptake of [(18)F]PF-9811 in brain regions with high FAAH enzymatic activity, as well as statistically significant reductions in signal following pre-administration of the blocking compound PF-04457845. CONCLUSIONS: [(18)F]PF-9811 is a promising PET imaging agent for FAAH. Biodistribution and PET imaging experiments show that the tracer has good uptake in brain, regional heterogeneity, and specific binding as determined by blocking experiments with the highly potent and selective FAAH inhibitor, PF-04457845.


Subject(s)
Amidohydrolases/metabolism , Brain/enzymology , Piperidines/chemical synthesis , Positron-Emission Tomography/methods , Pyridazines/chemical synthesis , Animals , Brain/diagnostic imaging , Chemistry Techniques, Synthetic , Ligands , Male , Piperidines/chemistry , Piperidines/pharmacokinetics , Pyridazines/chemistry , Pyridazines/pharmacokinetics , Radiochemistry , Rats
10.
Clin Cancer Res ; 18(5): 1303-12, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22170262

ABSTRACT

PURPOSE: [(18)F]FLT (3'-Fluoro-3' deoxythymidine)-PET imaging was proposed as a tool for measuring in vivo tumor cell proliferation. The aim of this article was to validate the use of [(18)F]FLT-PET imaging for measuring xenograft proliferation and subsequent monitoring of targeted therapy. EXPERIMENTAL DESIGN: In exponentially growing xenografts, factors that could impact the outcome of [(18)F]FLT-PET imaging, such as nucleoside transporters, thymidine kinase 1, the relative contribution of DNA salvage pathway, and the ratio of FLT to thymidine, were evaluated. The [(18)F]FLT tracer avidity was compared with other proliferation markers. RESULTS: In a panel of proliferating xenografts, [(18)F]FLT or [(3)H]thymidine tracer avidity failed to reflect the tumor growth rate across different tumor types, despite the high expressions of Ki67 and TK1. When FLT was injected at the same dose level as used in the preclinical [(18)F]FLT-PET imaging, the plasma exposure ratio of FLT to thymidine was approximately 1:200. Thymidine levels in different tumor types seemed to be variable and exhibited an inverse relationship with the FLT tracer avidity. In contrast, high-dose administration of bromdeoxyuridine (BrdUrd; 50 mg/kg) yielded a plasma exposure of more than 4-fold higher than thymidine and leads to a strong correlation between the BrdUrd uptake and the tumor proliferation rate. In FLT tracer-avid models, [(18)F]FLT-PET imaging as a surrogate biomarker predicted the therapeutic response of CDK4/6 inhibitor PD-0332991. CONCLUSIONS: Tumor thymidine level is one of the factors that impact the correlation between [(18)F]FLT uptake and tumor cell proliferation. With careful validation, [(18)F]FLT-PET imaging can be used to monitor antiproliferative therapies in tracer-avid malignancies.


Subject(s)
Dideoxynucleosides , Neoplasms/diagnostic imaging , Positron-Emission Tomography , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Bromodeoxyuridine/metabolism , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Dideoxynucleosides/pharmacokinetics , Disease Models, Animal , Humans , Mice , Mice, Nude , Mice, SCID , Neoplasms/drug therapy , Neoplasms/metabolism , Piperazines/administration & dosage , Piperazines/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Thymidine/metabolism , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 16(21): 5177-88, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20829331

ABSTRACT

PURPOSE: P-cadherin is a membrane glycoprotein that functionally mediates tumor cell adhesion, proliferation, and invasiveness. We characterized the biological properties of PF-03732010, a human monoclonal antibody against P-cadherin, in cell-based assays and tumor models. EXPERIMENTAL DESIGN: The affinity, selectivity, and cellular inhibitory activity of PF-03732010 were tested in vitro. Multiple orthotopic and metastatic tumor models were used for assessing the antitumor and antimetastatic activities of PF-03732010. Treatment-associated pharmacodynamic changes were also investigated. RESULTS: PF-03732010 selectively inhibits P-cadherin-mediated cell adhesion and aggregation in vitro. In the P-cadherin-overexpressing tumor models, including MDA-MB-231-CDH3, 4T1-CDH3, MDA-MB-435HAL-CDH3, HCT116, H1650, PC3M-CDH3, and DU145, PF-03732010 inhibited the growth of primary tumors and metastatic progression, as determined by bioluminescence imaging. Computed tomography imaging, H&E stain, and quantitative PCR analysis confirmed the antimetastatic activity of PF-03732010. In contrast, PF-03732010 did not show antitumor and antimetastatic efficacy in the counterpart tumor models exhibiting low P-cadherin expression. Mechanistic studies via immunofluorescence, immunohistochemical analyses, and 3'-[(18)F]fluoro-3'-deoxythymidine-positron emission tomography imaging revealed that PF-03732010 suppressed P-cadherin levels, caused degradation of membrane ß-catenin, and concurrently suppressed cytoplasmic vimentin, resulting in diminished metastatic capacity. Changes in the levels of Ki67, caspase-3, and 3'-[(18)F]fluoro-3'-deoxythymidine tracer uptake also indicated antiproliferative activity and increased apoptosis in the tested xenografts. CONCLUSIONS: These findings suggest that interrupting the P-cadherin signaling pathway may be a novel therapeutic approach for cancer therapy. PF-03732010 is presently undergoing evaluation in Phase 1 clinical trials.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cadherins/immunology , Neoplasm Metastasis/drug therapy , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasm Metastasis/prevention & control , Neoplasm Transplantation , Neoplasms/pathology , Transplantation, Heterotopic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
12.
Clin Cancer Res ; 15(14): 4630-40, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19584159

ABSTRACT

PURPOSE: Checkpoint kinase 1 (Chk1) plays a critical role in the activation of mitotic spindle checkpoint and DNA damage checkpoint. We examined the preclinical use of the Chk1 inhibitor PF-00477736 as a docetaxel-sensitizing agent. Specifically, we investigated the correlation between PF-00477736-mediated modulation of biomarkers and the sensitization of docetaxel efficacy. EXPERIMENTAL DESIGN: In vitro and in vivo studies using COLO205 and other cell lines were done to assess PF-00477736-induced enhancement of docetaxel efficacy and effects on associated biomarkers. RESULTS: PF-00477736 significantly enhanced the docetaxel-induced efficacy in tumor cells and xenografts. Docetaxel induced dose- and time-dependent increase in the levels of phosphorylated Chk1 (Ser(345)), phosphorylated histone H3 (Ser(10)), and gammaH2AX foci and promoted the cytoplasmic localization of phosphorylated Cdc25C (Ser(216)). PF-00477736 cotreatment suppressed docetaxel-induced changes in phosphorylated histone H3 and cytoplasmic phosphorylated Cdc25C (Ser(216)) levels and concurrently sensitized the docetaxel-induced apoptosis. Docetaxel alone or in combination with PF-00477736 induced significant antiproliferative activity in xenografts, shown via [18F]FLT-PET imaging. However, changes in [18F]FLT uptake did not reflect the potentiation of docetaxel efficacy. In contrast, bioluminescence imaging showed that PF-00477736 sensitized docetaxel-induced suppression of tumor survival. CONCLUSIONS: Docetaxel triggers mitotic spindle checkpoint activation at low concentrations and activates both the DNA damage checkpoint and the spindle checkpoint at high concentrations. In combination with docetaxel, PF-00477736 abrogates the mitotic checkpoint, as well as the DNA damage checkpoint, and results in sensitization to docetaxel. Chk1 inhibitor PF-00477736 offers a therapeutic potential for the enhancement of taxane therapy.


Subject(s)
Benzodiazepinones/pharmacology , Neoplasms/drug therapy , Protein Kinases/metabolism , Pyrazoles/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Benzodiazepinones/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Checkpoint Kinase 1 , Dideoxynucleosides , Docetaxel , Drug Synergism , Fluorine Radioisotopes , Histones/metabolism , Humans , Mice , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation/drug effects , Pyrazoles/administration & dosage , Taxoids/administration & dosage , Taxoids/pharmacology , Tomography, Emission-Computed , Tumor Burden/drug effects , cdc25 Phosphatases/metabolism
13.
J Vet Med Sci ; 70(1): 43-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18250571

ABSTRACT

Using microPET and (18)F-fluorodeoxyglucose ((18)F-FDG) as a tracer, we investigated regional brain activation in a rat model of visceral hypersensitivity induced by 2,4,6-trinitrobenzene sulfonic acid (TNBS). TNBS injection into the proximal colon through laparotomy resulted in a significant, sustained decrease in the pain threshold to mechanical distention of the distal colon, indicating a phenomenon referred to as visceral hypersensitivity. When TNBS-induced colonic hypersensitivity was fully developed, all the TNBS-treated rats presented characteristic pain behaviors in response to colonic distention at previously innocuous pressure (0-35 mmHg) that produced no abdominal pain in sham-operated control animals. In microPET study, colonic distention at the normally non-painful pressure produced significant increases in (18)F-FDG uptake in the thalamus and sensory cortex I of TNBS-treated rats. Since the increases in (18)F-FDG uptake in the brain regions were completely abolished by an analgesic dose of morphine (375 microg/kg, s.c.), it is most likely that the regional brain activation detected by microPET is a pain-related central event. The pharmacological and microPET data indicate that colonic distention at the normally non-painful pressure activates specific brain regions in rats with TNBS-induced visceral hypersensitivity, and the microPET protocol described here could provide an objective measure to test visceral analgesic compounds.


Subject(s)
Brain/metabolism , Colon/drug effects , Colon/physiology , Pain/metabolism , Positron-Emission Tomography/methods , Animals , Fluorodeoxyglucose F18/pharmacology , Male , Models, Animal , Morphine/pharmacology , Radiopharmaceuticals/pharmacology , Rats , Rats, Sprague-Dawley , Trinitrobenzenesulfonic Acid/pharmacology
14.
J Am Assoc Lab Anim Sci ; 46(5): 42-5, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17877327

ABSTRACT

The 18F isotope of fluoro-2-deoxy-D-glucose (FDG) is a radiotracer commonly used in positron emission tomography (PET) for determining regional metabolic activity in the brain. However, in rats and many other species with nictitating membranes, harderian glands located just behind the eyes aggressively incorporate 18F-FDG to the extent that PET images of the brain become obscured. This radioactive spillover, or 'partial volume error,' combined with the limited spatial resolution of microPET scanners (1.5 to 2 mm) may markedly reduce the ability to quantify neuronal activity in frontal brain structures. Theoretically, surgical removal of the harderian glands before 18F-FDG injection would eliminate the confounding uptake of the radioactive tracer and thereby permit visualization of glucose metabolism in the frontal brain. We conducted a pilot study of unilateral harderian gland adenectomy, leaving the contralateral gland intact for comparison. At 1 wk after surgery, each rat was injected intravenously with 18F-FDG, and 40 min later underwent brain microPET for 20 min. Review of the resulting images showed that the frontal cortex on the surgical side was defined more clearly, with only background 18F-FDG accumulation in the surgical bed. Activity in the frontal cortex on the intact side was obscured by intense accumulation of 18F-FDG in the harderian gland. By reducing partial volume error, this simple surgical procedure may become a valuable tool for visualization of the frontal cortex of rat brain by 18F-FDG microPET imaging.


Subject(s)
Brain/metabolism , Fluorodeoxyglucose F18 , Harderian Gland/surgery , Radiopharmaceuticals , Tomography, Emission-Computed/veterinary , Animals , Brain/diagnostic imaging , Frontal Lobe/diagnostic imaging , Frontal Lobe/metabolism , Harderian Gland/diagnostic imaging , Harderian Gland/metabolism , Rats , Rats, Sprague-Dawley , Tomography, Emission-Computed/methods
15.
J Nucl Med ; 46(5): 840-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15872359

ABSTRACT

UNLABELLED: (131)I radionuclide therapy studies have not shown a strong relationship between tumor absorbed dose and response, possibly due to inaccuracies in activity quantification and dose estimation. The goal of this work was to establish the accuracy of (131)I activity quantification and absorbed dose estimation when patient-specific, 3-dimensional (3D) methods are used for SPECT reconstruction and for absorbed dose calculation. METHODS: Clinically realistic voxel-phantom simulations were used in the evaluation of activity quantification and dosimetry. SPECT reconstruction was performed using an ordered-subsets expectation maximization (OSEM) algorithm with compensation for scatter, attenuation, and 3D detector response. Based on the SPECT image and a patient-specific density map derived from CT, 3D dosimetry was performed using a newly implemented Monte Carlo code. Dosimetry was evaluated by comparing mean absorbed dose estimates calculated directly from the defined phantom activity map with those calculated from the SPECT image of the phantom. Finally, the 3D methods were applied to a radioimmunotherapy patient, and the mean tumor absorbed dose from the new calculation was compared with that from conventional dosimetry obtained from conjugate-view imaging. RESULTS: Overall, the accuracy of the SPECT-based absorbed dose estimates in the phantom was >12% for targets down to 16 mL and up to 35% for the smallest 7-mL tumor. To improve accuracy in the smallest tumor, more OSEM iterations may be needed. The relative SD from multiple realizations was <3% for all targets except for the smallest tumor. For the patient, the mean tumor absorbed dose estimate from the new Monte Carlo calculation was 7% higher than that from conventional dosimetry. CONCLUSION: For target sizes down to 16 mL, highly accurate and precise dosimetry can be obtained with 3D methods for SPECT reconstruction and absorbed dose estimation. In the future, these methods can be applied to patients to potentially establish correlations between tumor regression and the absorbed dose statistics from 3D dosimetry.


Subject(s)
Abdominal Neoplasms/radiotherapy , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Iodine Radioisotopes/analysis , Iodine Radioisotopes/therapeutic use , Radiometry/methods , Radiotherapy Planning, Computer-Assisted/methods , Tomography, Emission-Computed, Single-Photon/methods , Abdominal Neoplasms/diagnostic imaging , Algorithms , Body Burden , Humans , Phantoms, Imaging , Radiotherapy Dosage , Radiotherapy, Computer-Assisted/methods , Relative Biological Effectiveness , Reproducibility of Results , Sensitivity and Specificity , Tomography, Emission-Computed, Single-Photon/instrumentation
16.
N Engl J Med ; 352(5): 441-9, 2005 Feb 03.
Article in English | MEDLINE | ID: mdl-15689582

ABSTRACT

BACKGROUND: Advanced-stage follicular B-cell lymphoma is considered incurable. Anti-CD20 radioimmunotherapy is effective in patients who have had a relapse after chemotherapy or who have refractory follicular lymphoma, but it has not been tested in previously untreated patients. METHODS: Seventy-six patients with stage III or IV follicular lymphoma received as initial therapy a single course of treatment with 131I-tositumomab therapy (registered as Tositumomab and Iodine I 131 Tositumomab [the Bexxar therapeutic regimen]). This consisted of a dosimetric dose of tositumomab and 131I-labeled tositumomab followed one week later by a therapeutic dose, delivering 75 cGy of radiation to the total body. RESULTS: Ninety-five percent of the patients had any response, and 75 percent had a complete response. The use of polymerase chain reaction (PCR) to detect rearrangement of the BCL2 gene showed molecular responses in 80 percent of assessable patients who had a clinical complete response. After a median follow-up of 5.1 years, the actuarial 5-year progression-free survival for all patients was 59 percent, with a median progression-free survival of 6.1 years. The annualized rate of relapse progressively decreased over time: 25 percent, 13 percent, and 12 percent during the first, second, and third years, respectively, and 4.4 percent per year after three years. Of 57 patients who had a complete response, 40 remained in remission for 4.3 to 7.7 years. Hematologic toxicity was moderate, with no patient requiring transfusions or hematopoietic growth factors. No cases of myelodysplastic syndrome have been observed. CONCLUSIONS: A single one-week course of 131I-tositumomab therapy as initial treatment can induce prolonged clinical and molecular remissions in patients with advanced follicular lymphoma.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD20/immunology , Lymphoma, Follicular/radiotherapy , Radioimmunotherapy , Adult , Aged , Antibodies, Monoclonal/adverse effects , B-Lymphocytes , Bone Marrow Examination , Disease-Free Survival , Female , Gene Rearrangement/drug effects , Genes, bcl-2 , Humans , Leukocyte Count , Lymphoma, Follicular/genetics , Lymphoma, Follicular/immunology , Male , Middle Aged , Radioimmunotherapy/adverse effects , Radiometry , Remission Induction , Survival Analysis , Thyrotropin/blood
18.
Mol Imaging Biol ; 5(2): 79-85, 2003.
Article in English | MEDLINE | ID: mdl-14499148

ABSTRACT

PURPOSE: This study evaluated in a phantom model and verified in patients with lung cancer whether the use of an internal positron-emitting labeled marker could localize a critical structure by positron emission tomography (PET) imaging and verify multimodality image registration. MATERIALS AND METHODS: An initial device and method were developed to demonstrate by dedicated PET the location of the normal esophagus in a phantom and in three patients using a column of 2-deoxy-2-[18F]fluoro-D-glucose (FDG) solution between proximal and distal gas phases in polyurethane tubing. The device was assessed for possible loss of radioactivity. PET, CT and PET-CT fusion imaging followed. RESULTS: X-rays of the marker device showed a continuous fluid column. No leakage of contents was detected. The internal marker in the phantom and in patients allowed visualization by PET of the esophagus, and verified an image registration algorithm. CONCLUSIONS: A positron-emitting internal marker was constructed, demonstrated to retain tracer, and shown to be capable of verifying an image registration algorithm and identifying a critical structure, the esophagus, by PET in a phantom and in patients.


Subject(s)
Esophagus/diagnostic imaging , Fluorodeoxyglucose F18 , Tomography, Emission-Computed/methods , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Humans , Image Processing, Computer-Assisted , Lung Neoplasms/diagnostic imaging , Phantoms, Imaging , Radiography , Radiopharmaceuticals , Reference Values , Reproducibility of Results
19.
J Nucl Med ; 44(3): 457-64, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12621015

ABSTRACT

UNLABELLED: A study of the use of (131)I-labeled tositumomab, preceded by an unlabeled tositumomab predose, for therapy of 76 previously untreated non-Hodgkin's lymphoma patients has been completed at the University of Michigan. Fifty-two of the 76 treated patients were imaged once during therapy with SPECT to assist in dosimetric estimation. In this article, the patient's average tumor dose, estimated by a hybrid method using that SPECT, is compared with the same statistic estimated by pretherapy conjugate views. METHODS: The SPECT activity-quantification procedure used 3-dimensional CT-to-SPECT image registration. Daily pretherapy conjugate-view images provided the shape of the time-activity curve for the hybrid dose estimation. RESULTS: With the hybrid method, the mean of the patient's average tumor dose over 8 patients using only their axillary tumors (162 cGy) was very significantly lower (P < 0.0001) than the mean over 47 patients using only their evaluated chest, abdominal, and pelvic tumors (624 cGy) for unknown reasons. Excluding axillary tumors as a best case for prediction, there still was considerable overlap in the distribution of a patient's average tumor dose over 38 patients who went on to a complete response (CR) and that from 9 patients who went on to a partial response (PR) using either method. However, a high value of the patient's average tumor dose was correctly associated with a CR for 15 of 16 patients (94%) with hybrid SPECT and for 9 of 12 patients (75%) with conjugate views. Also, the mean of the patient's average tumor dose for the CR patients was larger than the mean for PR patients; the P value was 0.18 with hybrid SPECT and 0.25 with conjugate views. A multiple logistic regression analysis combining the dose, tumor burden, and level of lactate dehydrogenase as explanatory variables for response did not yield statistical significance with either method. CONCLUSION: Patients with evaluated tumors that receive the highest tumor radiation dose are most likely to achieve a CR. Dosimetry based on a combination of pretherapy conjugate views and intratherapy SPECT provides somewhat better correspondence between the patient's average tumor dose and his or her degree of response compared with dosimetry from pretherapy conjugate views alone. Statistical significance for the correspondence is not reached either with the dosimetric method or with either method in combination with the tumor burden and level of lactate dehydrogenase.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Iodine Radioisotopes/therapeutic use , Lymphoma, Non-Hodgkin/diagnostic imaging , Lymphoma, Non-Hodgkin/radiotherapy , Radioimmunotherapy , Tomography, Emission-Computed, Single-Photon , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antigens, CD20/immunology , Antineoplastic Agents/therapeutic use , Female , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Radiotherapy Dosage , Tomography, X-Ray Computed
20.
Eur J Nucl Med Mol Imaging ; 30(2): 274-80, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12552346

ABSTRACT

The aim of this study was to determine the relationship between tumor blood flow and glucose utilization in women with untreated primary breast carcinomas. Noninvasive determinations of blood flow and glucose utilization with positron emission tomography (PET) were performed in 101 regions of tumor from nine women with untreated primary breast carcinoma. [(15)O]H(2)O PET scans of tumor blood flow were compared with fluorine-18 fluoro-2-deoxy- D-glucose (FDG) PET scans of tumor glucose metabolism. Modeling of multiple parameters was undertaken and flow and glucose utilization compared. Mean whole-tumor blood flow was 14.9 ml dl(-1) min(-1), but ranged from 7.6 to 29.2 ml dl(-1) min(-1). Mean whole-tumor standardized uptake value corrected for lean body mass, SUV-lean (50-60 min), was 2.32+/-0.19 while mean K(i) was 1.2 ml dl(-1) min(-1) for FDG. SUV-lean and blood flow were strongly correlated (r=0.82, P=0.007) as were K(1) for FDG and flow (r=0.84, P=0.004). In these women with untreated breast cancers, FDG uptake (SUV-lean) and tumor blood flow are strongly correlated. The slope of FDG uptake versus blood flow appears higher at low flow rates, suggesting the possible presence of areas of tumor hypoxia.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Fluorodeoxyglucose F18/pharmacokinetics , Glucose/metabolism , Models, Biological , Adult , Aged , Blood Flow Velocity , Breast Neoplasms/blood supply , Breast Neoplasms/diagnostic imaging , Female , Humans , Middle Aged , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Statistics as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...