Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Front Neuroimaging ; 3: 1355402, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38606196

RESUMO

Purpose: We evaluated the impact of partial volume correction (PVC) methods on the quantification of longitudinal [18F]GTP1 tau positron-emission tomography (PET) in Alzheimer's disease and the suitability of describing the tau pathology burden temporal trajectories using linear mixed-effects models (LMEM). Methods: We applied van Cittert iterative deconvolution (VC), 2-compartment, and 3-compartment, and the geometric transfer matrix plus region-based voxelwise methods to data acquired in an Alzheimer's disease natural history study over 18 months at a single imaging site. We determined the optimal PVC method by comparing the standardized uptake value ratio change (%ΔSUVR) between diagnostic and tau burden-level groups and the longitudinal repeatability derived from the LMEM. The performance of LMEM analysis for calculating %ΔSUVR was evaluated in a natural history study and in a multisite clinical trial of semorinemab in prodromal to mild Alzheimer's disease by comparing results to traditional per-visit estimates. Results: The VC, 2-compartment, and 3-compartment PVC methods had similar performance, whereas region-based voxelwise overcorrected regions with a higher tau burden. The lowest within-subject variability and acceptable group separation scores were observed without PVC. The LMEM-derived %ΔSUVR values were similar to the per-visit estimates with lower variability. Conclusion: The results indicate that the tested PVC methods do not offer a clear advantage or improvement over non-PVC images for the quantification of longitudinal [18F]GTP1 PET data. LMEM offers a robust framework for the longitudinal tau PET quantification with low longitudinal test-retest variability. Clinical trial registration: NCT02640092 and NCT03289143.

2.
Neurology ; 101(14): e1391-e1401, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37643887

RESUMO

BACKGROUND AND OBJECTIVES: Accumulation of tau pathology in Alzheimer disease (AD) correlates with cognitive decline. Anti-tau immunotherapies were proposed as potential interventions in AD. While antibodies targeting N-terminal tau failed to demonstrate clinical efficacy in prodromal-to-mild AD, their utility at other disease stages was not evaluated in prior studies. Lauriet is a phase 2 study of an anti-tau monoclonal antibody, semorinemab, in patients with mild-to-moderate AD. METHODS: The phase 2 Lauriet study included a randomized, placebo-controlled, double-blind period, during which participants with mild-to-moderate AD received 4,500 mg of IV semorinemab or placebo every 4 weeks for 48 or 60 weeks. Participants who chose to continue in the subsequent optional open-label extension received 4,500 mg of semorinemab every 4 weeks for up to 96 weeks. Coprimary efficacy endpoints were change from baseline to week 49 or 61 on the 11-item version of the Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog11) and the Alzheimer's Disease Cooperative Study-Activities of Daily Living (ADCS-ADL) scale. Secondary efficacy endpoints included change from baseline on the Mini-Mental State Examination (MMSE) and Clinical Dementia Rating-Sum of Boxes (CDR-SB). Safety, pharmacokinetics, and pharmacodynamic effects were also evaluated. RESULTS: Between December 3, 2018, and February 27, 2020, 624 individuals were screened, 272 participants were randomized, and 238 were included in the modified intent-to-treat population (received ≥1 dose(s) of study medication and underwent baseline and ≥1 postbaseline assessment(s)). Baseline characteristics were well balanced. At week 49, the semorinemab arm demonstrated a 42.2% reduction (-2.89 points, 95% CI -4.56 to -1.21, p = 0.0008) in decline on the ADAS-Cog11 (coprimary endpoint) relative to the placebo arm. However, no treatment effects were observed on the ADCS-ADL scale (coprimary endpoint; absolute difference between the 2 treatment arms in the ADCS-ADL score change from baseline of -0.83 points, 95% CI -3.39 to 1.72, p = 0.52) or on the MMSE or CDR-SB (secondary endpoints). Semorinemab was safe and well tolerated. DISCUSSION: Based on the results of the prespecified coprimary endpoints, this study was negative. While semorinemab had a significant effect on cognition measured by the ADAS-Cog11, this effect did not extend to improved functional or global outcomes. These results may warrant further exploration of semorinemab or other anti-tau therapies in mild-to-moderate AD. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that semorinemab does not slow functional decline in patients with mild-to-moderate AD. TRIAL REGISTRATION INFORMATION: The Lauriet study is registered on ClinicalTrials.gov, NCT03828747, and EudraCT 2018-003398-87.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Humanos , Doença de Alzheimer/psicologia , Atividades Cotidianas , Resultado do Tratamento , Anticorpos Monoclonais/uso terapêutico , Método Duplo-Cego
3.
Nat Med ; 28(12): 2601-2610, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36471036

RESUMO

Immune checkpoint inhibitors (ICIs), by reinvigorating CD8+ T cell mediated immunity, have revolutionized cancer therapy. Yet, the systemic CD8+ T cell distribution, a potential biomarker of ICI response, remains poorly characterized. We assessed safety, imaging dose and timing, pharmacokinetics and immunogenicity of zirconium-89-labeled, CD8-specific, one-armed antibody positron emission tomography tracer 89ZED88082A in patients with solid tumors before and ~30 days after starting ICI therapy (NCT04029181). No tracer-related side effects occurred. Positron emission tomography imaging with 10 mg antibody revealed 89ZED88082A uptake in normal lymphoid tissues, and tumor lesions across the body varying within and between patients two days after tracer injection (n = 38, median patient maximum standard uptake value (SUVmax) 5.2, IQI 4.0-7.4). Higher SUVmax was associated with mismatch repair deficiency and longer overall survival. Uptake was higher in lesions with stromal/inflamed than desert immunophenotype. Tissue radioactivity was localized to areas with immunohistochemically confirmed CD8 expression. Re-imaging patients on treatment showed no change in average (geometric mean) tumor tracer uptake compared to baseline, but individual lesions showed diverse changes independent of tumor response. The imaging data suggest enormous heterogeneity in CD8+ T cell distribution and pharmacodynamics within and between patients. In conclusion, 89ZED88082A can characterize the complex dynamics of CD8+ T cells in the context of ICIs, and may inform immunotherapeutic treatments.


Assuntos
Imunoconjugados , Neoplasias , Humanos , Linfócitos T CD8-Positivos , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Tomografia por Emissão de Pósitrons/métodos , Imunoterapia/efeitos adversos , Imunoterapia/métodos
4.
JAMA Neurol ; 79(11): 1113-1121, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36121669

RESUMO

Importance: Alzheimer disease (AD), a neurodegenerative disease characterized by ß-amyloid plaques and τ tangles in the brain, represents an unmet medical need with no fully approved therapeutics to modify disease progression. Objective: To investigate the safety and efficacy of crenezumab, a humanized monoclonal immunoglobulin G4 antibody targeting ß-amyloid oligomers, in participants with prodromal to mild (early) AD. Design, Setting, and Participants: Two phase 3 multicenter randomized double-blind placebo-controlled parallel-group efficacy and safety studies of crenezumab in participants with early AD, CREAD and CREAD2, were initiated in 2016 and 2017, respectively, and were designed to evaluate the efficacy and safety of crenezumab in participants with early AD. CREAD (194 sites in 30 countries) and CREAD2 (209 sites in 27 countries) were global multicenter studies. A total of 3736 and 3664 participants were screened in CREAD and CREAD2, respectively. A total of 3736 and 3664 participants were screened in CREAD and CREAD2, respectively. Both trials enrolled individuals aged 50 to 85 years with early AD. Participants with some comorbidities and evidence of cerebral infarction or more than 4 microbleeds or areas of leptomeningeal hemosiderosis on magnetic resonance imaging were excluded. After 2923 and 2858 were excluded, respectively, 813 participants in CREAD and 806 in CREAD2 were randomly assigned in a 1:1 ratio to either placebo or crenezumab. In the final analysis, there were 409 participants in the placebo group and 404 in the crenezumab group in CREAD and 399 in the placebo group and 407 in the crenezumab group in CREAD2. Data were analyzed up until January 2019 and August 2019, respectively. Interventions: Participants received placebo or 60 mg/kg crenezumab intravenously every 4 weeks for up to 100 weeks. Main Outcomes and Measures: The primary outcome was change from baseline to week 105 in Clinical Dementia Rating-Sum of Boxes (CDR-SB) score. Results: There were 813 participants in CREAD (mean [SD] age, 70.7 [8.2] years; 483 female and 330 male) and 806 in CREAD2 (mean [SD] age, 70.9 [7.7] years; 456 female and 350 male). Baseline characteristics were balanced between both groups. The between-group difference in mean change from baseline in CDR-SB score (placebo minus crenezumab) was -0.17 (95% CI, -0.86 to 0.53; P = .63) at week 105 in the CREAD study (88 placebo; 86 crenezumab). Compared with previous trials, no new safety signals were identified, and amyloid-related imaging abnormalities with edema were rare, mild, and transient. No meaningful changes in AD biomarkers were observed. Both studies were discontinued following a preplanned interim analysis indicating that CREAD was unlikely to meet the primary end point. Conclusions and Relevance: Crenezumab was well tolerated but did not reduce clinical decline in participants with early AD. Trial Registration: ClinicalTrials.gov Identifiers: CREAD, NCT02670083; CREAD2, NCT03114657.


Assuntos
Doença de Alzheimer , Anticorpos Monoclonais Humanizados , Adulto , Idoso , Feminino , Humanos , Masculino , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides , Método Duplo-Cego , Placa Amiloide , Resultado do Tratamento , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico
5.
Clin Cancer Res ; 26(6): 1229-1236, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31848189

RESUMO

PURPOSE: ERK1/2 signaling can be dysregulated in cancer. GDC-0994 is an oral inhibitor of ERK1/2. A first-in-human, phase I dose escalation study of GDC-0994 was conducted in patients with locally advanced or metastatic solid tumors. PATIENTS AND METHODS: GDC-0994 was administered once daily on a 21-day on/7-day off schedule to evaluate safety, pharmacokinetics, and preliminary signs of efficacy. Patients with pancreatic adenocarcinoma and BRAF-mutant colorectal cancer were enrolled in the expansion stage. RESULTS: Forty-seven patients were enrolled in six successive cohorts (50-800 mg). A single DLT of grade 3 rash occurred at 600 mg. The most common drug-related adverse events (AE) were diarrhea, rash, nausea, fatigue, and vomiting. Pharmacokinetic data showed dose-proportional increases in exposure, with a mean half-life of 23 hours, supportive of once daily dosing. In evaluable paired biopsies, MAPK pathway inhibition ranged from 19% to 51%. Partial metabolic responses by FDG-PET were observed in 11 of 20 patients across dose levels in multiple tumor types. Overall, 15 of 45 (33%) patients had a best overall response of stable disease and 2 patients with BRAF-mutant colorectal cancer had a confirmed partial response. CONCLUSIONS: GDC-0994 had an acceptable safety profile and pharmacodynamic effects were observed by FDG-PET and in serial tumor biopsies. Single-agent activity was observed in 2 patients with BRAF-mutant colorectal cancer.


Assuntos
Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/uso terapêutico , Piridonas/farmacocinética , Piridonas/uso terapêutico , Pirimidinas/farmacocinética , Pirimidinas/uso terapêutico , Adulto , Idoso , Relação Dose-Resposta a Droga , Fadiga/induzido quimicamente , Feminino , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Náusea/induzido quimicamente , Neoplasias/induzido quimicamente , Neoplasias/patologia , Segurança do Paciente , Distribuição Tecidual , Vômito/induzido quimicamente
6.
Cancer Discov ; 7(7): 704-715, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28331003

RESUMO

Taselisib is a potent and selective tumor growth inhibitor through PI3K pathway suppression. Thirty-four patients with locally advanced or metastatic solid tumors were treated (phase I study, modified 3+3 dose escalation; 5 cohorts; 3-16 mg taselisib once-daily capsule). Taselisib pharmacokinetics were dose-proportional; mean half-life was 40 hours. Frequent dose-dependent, treatment-related adverse events included diarrhea, hyperglycemia, decreased appetite, nausea, rash, stomatitis, and vomiting. At 12 and 16 mg dose levels, dose-limiting toxicities (DLT) were observed, with an accumulation of higher-grade adverse events after the cycle 1 DLT assessment window. Pharmacodynamic findings showed pathway inhibition at ≥3 mg in patient tumor samples, consistent with preclinical PIK3CA-mutant tumor xenograft models. Confirmed response rate was 36% for PIK3CA-mutant tumor patients with measurable disease [5/14: 4 breast cancer (3 patients at 12 mg); 1 non-small cell lung cancer], where responses started at 3 mg, and 0% in patients with tumors without known PIK3CA hotspot mutations (0/15).Significance: Preliminary data consistent with preclinical data indicate increased antitumor activity of taselisib in patients with PIK3CA-mutant tumors (in comparison with patients with tumors without known activating PIK3CA hotspot mutations) starting at the lowest dose tested of 3 mg, thereby supporting higher potency for taselisib against PIK3CA-mutant tumors. Cancer Discov; 7(7); 704-15. ©2017 AACR.See related commentary by Rodon and Tabernero, p. 666This article is highlighted in the In This Issue feature, p. 653.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Imidazóis/administração & dosagem , Neoplasias/tratamento farmacológico , Oxazepinas/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Adulto , Idoso , Animais , Classe I de Fosfatidilinositol 3-Quinases/genética , Relação Dose-Resposta a Droga , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/classificação , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Humanos , Imidazóis/efeitos adversos , Masculino , Camundongos , Pessoa de Meia-Idade , Mutação , Neoplasias/genética , Neoplasias/patologia , Oxazepinas/efeitos adversos , Inibidores de Proteínas Quinases/efeitos adversos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cancer Discov ; 7(1): 102-113, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27872130

RESUMO

Activation of AKT signaling by PTEN loss or PIK3CA mutations occurs frequently in human cancers, but targeting AKT has been difficult due to the mechanism-based toxicities of inhibitors that target the inactive conformation of AKT. Ipatasertib (GDC-0068) is a novel selective ATP-competitive small-molecule inhibitor of AKT that preferentially targets active phosphorylated AKT (pAKT) and is potent in cell lines with evidence of AKT activation. In this phase I study, ipatasertib was well tolerated; most adverse events were gastrointestinal and grade 1-2 in severity. The exposures of ipatasertib ≥200 mg daily in patients correlated with preclinical TGI90, and pharmacodynamic studies confirmed that multiple targets (i.e., PRAS40, GSK3ß, and mTOR) were inhibited in paired on-treatment biopsies. Preliminary antitumor activity was observed; 16 of 52 patients (30%), with diverse solid tumors and who progressed on prior therapies, had radiographic stable disease, and many of their tumors had activation of AKT. SIGNIFICANCE: Potent inhibition of AKT signaling with ipatasertib was associated with a tolerable safety profile and meaningful disease control in a subgroup of patients. Targeting pAKT with an ATP-competitive inhibitor provides a greater therapeutic window than allosteric inhibitors. Further investigation with ipatasertib is ongoing in phase II studies. Cancer Discov; 7(1); 102-13. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 1.


Assuntos
Neoplasias/tratamento farmacológico , Piperazinas/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Pirimidinas/administração & dosagem , Administração Oral , Adulto , Idoso , Linhagem Celular Tumoral , Esquema de Medicação , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/genética , Piperazinas/efeitos adversos , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Pirimidinas/efeitos adversos , Pirimidinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Clin Cancer Res ; 22(7): 1642-52, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26589435

RESUMO

PURPOSE: Mesothelin (MSLN) is frequently overexpressed in pancreatic and ovarian cancers, making it a potential drug target. We performed an (89)Zr-PET imaging study with MMOT0530A, a MSLN antibody, in conjunction with a phase I study with the antibody-drug conjugate DMOT4039A, containing MMOT0530A bound to MMAE. The aim was to study antibody tumor uptake, whole-body distribution, and relation between uptake, response to treatment, and MSLN expression. EXPERIMENTAL DESIGN: Before DMOT4039A treatment, patients received 37 MBq (89)Zr-MMOT0530A followed by PET/CT imaging 2, 4, and 7 days postinjection. Tracer uptake was expressed as standardized uptake value (SUV). MSLN expression was determined with immunohistochemistry (IHC) on archival tumor tissue. RESULTS: Eleven patients were included, 7 with pancreatic and 4 with ovarian cancer. IHC MSLN expression varied from absent to strong. Suitable tracer antibody dose was 10 mg MMOT0530A and optimal imaging time was 4 and 7 days postinjection. Tumor tracer uptake occurred in 37 lesions with mean SUVmax of 13.1 (±7.5) on PET 4 days postinjection, with 11.5 (±7.5) in (N= 17) pancreatic and 14.5 (±8.7) in (N= 20) ovarian cancer lesions. Within patients, a mean 2.4-fold (±1.10) difference in uptake between tumor lesions existed. Uptake in blood, liver, kidneys, spleen, and intestine reflected normal antibody distribution. Tracer tumor uptake was correlated to IHC. Best response to DMOT4039A was partial response in one patient. CONCLUSIONS: With (89)Zr-MMOT0530A-PET, pancreatic and ovarian cancer lesions as well as antibody biodistribution could be visualized. This technique can potentially guide individualized antibody-based treatment.


Assuntos
Anticorpos Monoclonais , Imunoconjugados/uso terapêutico , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/tratamento farmacológico , Tomografia por Emissão de Pósitrons/métodos , Adulto , Idoso , Anticorpos Monoclonais/imunologia , Antígenos de Neoplasias/imunologia , Feminino , Proteínas Ligadas por GPI/imunologia , Humanos , Imunoconjugados/imunologia , Masculino , Mesotelina , Pessoa de Meia-Idade , Neoplasias Ovarianas/imunologia , Neoplasias Pancreáticas/imunologia , Tomografia Computadorizada por Raios X , Resultado do Tratamento , Zircônio
9.
Cancer Imaging ; 15: 9, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26231380

RESUMO

BACKGROUND: Several reproducibility studies have established good test-retest reliability of FDG-PET in various oncology settings. However, these studies are based on relatively short inter-scan periods of 1-3 days while, in contrast, response assessments based on FDG-PET in early phase drug trials are typically made over an interval of 2-3 weeks during the first treatment cycle. With focus on longer, on-treatment scan intervals, we develop a data-driven approach to calculate baseline-specific cutoff values to determine patient-level changes in glucose uptake that are unlikely to be explained by random variability. Our method takes into account the statistical nature of natural fluctuations in SUV as well as potential bias effects. METHODS: To assess variability in SUV over clinically relevant scan intervals for clinical trials, we analyzed baseline and follow-up FDG-PET scans with a median scan interval of 21 days from 53 advanced stage cancer patients enrolled in a Phase 1 trial. The 53 patients received a sub-pharmacologic drug dose and the trial data is treated as a 'test-retest' data set. A simulation-based tool is presented which takes as input baseline lesion SUVmax values, the variance of spurious changes in SUVmax between scans, the desired Type I error rate, and outputs lesion and patient based cut-off values. Bias corrections are included to account for variations in tracer uptake time. RESULTS: In the training data, changes in SUVmax follow an approximately zero-mean Gaussian distribution with constant variance across levels of the baseline measurements. Because of constant variance, the coefficient of variation is a decreasing function of the magnitude of baseline SUVmax. This finding is consistent with published results, but our data shows greater variability. Application of our method to NSCLC patients treated with erlotinib produces results distinct from those based on the EORTC criteria. Based on data presented here as well as previous repeatability studies, the proposed method has greater statistical power to detect a significant %-decrease on SUVmax compared to published criteria relying on symmetric thresholds. CONCLUSIONS: Defining patient-specific, baseline dependent cut-off values based on the (null) distribution of naturally occurring fluctuations in glucose uptake enable identification of statistically significant changes in SUVmax. For lower baseline values, the produced cutoff values are notably asymmetric with relatively large changes (e.g. >50 %) required for statistical significance. For use with prospectively defined endpoints, the developed method enables the use of one-armed trials to detect pharmacodynamic drug effects based on FDG-PET. The clinical importance of changes in SUVmax is likely to remain dependent on both tumor biology and the type of treatment.


Assuntos
Algoritmos , Neoplasias/terapia , Tomografia por Emissão de Pósitrons/normas , Tomografia Computadorizada por Raios X/normas , Biomarcadores Farmacológicos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Fluordesoxiglucose F18 , Glucose/metabolismo , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias/metabolismo , Distribuição Normal , Valor Preditivo dos Testes , Compostos Radiofarmacêuticos , Resultado do Tratamento
10.
Synapse ; 69(1): 33-40, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25196464

RESUMO

Decreased glutamatergic neurotransmission is hypothesized to be involved in the pathophysiology of schizophrenia. Inhibition of glycine transporter Type-1 (GlyT1) reuptake is expected to increase the glutamatergic neurotransmission and may serve as treatment for cognitive and negative symptoms of schizophrenia. In this article, we present human data from a novel GlyT1 PET tracer, [(18) F]MK-6577. In the process of developing a GlyT1 inhibitor therapeutic, a PET tracer can assist in determining the dose with a high probability of sufficiently testing the mechanism of action. This article reports the human PET studies with [(18) F]MK-6577 for measuring GlyT1 receptor availability at baseline in normal human subjects and occupancy with a GlyT1 inhibitor, MK-2637. Studies were also performed to measure radiation burden and the baseline test-retest (T-RT) variability of the tracer. The effective dose from sequential whole-body dosimetry scans in three male subjects was estimated to be 24.5 ± 2.9 µSV/MBq (mean ± SD). The time-activity curves from T-RT scans modeled satisfactorily using a two tissue compartmental model. The tracer uptake was highest in the pons (VT = 6.7 ± 0.9, BPND = 4.1 ± 0.43) and lowest in the cortex (VT = 2.1 ± 0.5, BPND = 0.60 ± 0.23). VT T-RT variability measured in three subjects was <12% on average. The occupancy scans performed in a cohort of 15 subjects indicated absence of a reference region. The in vivo potency (Occ50 ) of MK-2637 was determined using two methods: A: Lassen plot with a population input function (Occ50 = 106 nM, SE = 20 nM) and B: pseudo reference tissue model using cortex as the pseudo reference region (Occ50 = 141 nM, SE = 21 nM).


Assuntos
Benzamidas , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Proteínas da Membrana Plasmática de Transporte de Glicina/metabolismo , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Sulfonamidas , Adulto , Benzamidas/farmacocinética , Encéfalo/efeitos dos fármacos , Mapeamento Encefálico , Estudos de Coortes , Proteínas da Membrana Plasmática de Transporte de Glicina/antagonistas & inibidores , Humanos , Cinética , Masculino , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacocinética , Reprodutibilidade dos Testes , Sulfonamidas/farmacocinética , Adulto Jovem
11.
J Nucl Med ; 55(1): 65-72, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24263088

RESUMO

UNLABELLED: The histamine 3 (H3) receptor is a presynaptic autoreceptor in the central nervous system that regulates the synthesis and release of histamine and modulates the release of other major neurotransmitters. H3 receptor inverse agonists (IAs) may be efficacious in the treatment of various central nervous system disorders, including excessive daytime sleepiness, attention deficit hyperactivity disorder, Alzheimer disease, ethanol addiction, and obesity. METHODS: Using PET and a novel high-affinity and selective radioligand (11)C-MK-8278, we studied the tracer biodistribution, quantification, and brain H3 receptor occupancy (RO) of MK-0249 and MK-3134, 2 potential IA drugs targeting cerebral H3 receptors, in 6 healthy male subjects (age, 19-40 y). The relationship among H3 IA dose, time on target, and peripheral pharmacokinetics was further investigated in 15 healthy male volunteers (age, 18-40 y) with up to 3 PET scans and 3 subjects per dose level. RESULTS: The mean effective dose for (11)C-MK-8278 was 5.4 ± 1.1 µSv/MBq. Human brain kinetics showed rapid high uptake and fast washout. Binding potential values can be assessed using the pons as a reference region, with a test-retest repeatability of 7%. Drug RO data showed low interindividual variability per dose (mean RO SD, 2.1%), and a targeted 90% RO can be reached for both IAs at clinically feasible doses. CONCLUSION: (11)C-MK-8278 is a useful novel PET radioligand for determination of human cerebral H3 receptor binding and allows highly reproducible in vivo brain occupancy of H3-targeting drugs, hereby enabling the evaluation of novel compounds in early development to select doses and schedules.


Assuntos
Benzofuranos/farmacologia , Encéfalo/diagnóstico por imagem , Radioisótopos de Carbono/farmacologia , Agonistas dos Receptores Histamínicos/química , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/farmacologia , Receptores Histamínicos H3/química , Compostos de Espiro/farmacologia , Administração Oral , Adulto , Animais , Humanos , Imageamento Tridimensional , Ligantes , Macaca mulatta , Masculino , Radiometria , Reprodutibilidade dos Testes , Adulto Jovem
12.
Psychopharmacology (Berl) ; 231(3): 511-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24051602

RESUMO

Reduced NMDA receptor functioning is hypothesized to underlie the cognitive and negative symptoms associated with schizophrenia. However, because direct activation of the NMDA receptor is accompanied by neurotoxicity, mechanisms that activate the glycine co-agonist site on the NMDA receptor could carry greater therapeutic potential. In the current study, the effects of two glycine transporter 1 (GlyT1) inhibitors, RG1678 and ORG25935, were characterized in the object-retrieval detour (ORD) task in scopolamine-impaired rhesus monkeys and, using positron emission tomography (PET), the GlyT1 occupancy to efficacy relationship of each compound was established. Scopolamine exerted a significant decrease in accuracy in the ORD task. Lower doses of RG1678 (0.3 and 1.0 mg/kg, p.o.) significantly attenuated the impact of scopolamine, whereas the highest dose tested (1.8 mg/kg) did not. The predicted GlyT1 occupancies of RG1678 at the effective doses were ~10 and 30 %. ORG25935 (0.1, 0.3, and 1 mg/kg, p.o.) also significantly attenuated the impact of scopolamine on the ORD task, whereas 3 mg/kg did not. The predicted GlyT1 occupancies of ORG25935 at the effective doses ranged from 16 to 80 %. These data suggest that GlyT1 inhibitors have the potential to improve performance on prefrontal cortex-dependent tests such as the ORD task, but that efficacy is lost when higher occupancies are achieved. Importantly, recent Ph2B data published by Roche suggests that low but not high doses of RG1678 improved negative symptoms in patients with schizophrenia, highlighting the potential translational nature of the current preclinical findings.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Glicina/antagonistas & inibidores , Proteínas da Membrana Plasmática de Transporte de Glicina/metabolismo , Antagonistas Muscarínicos/efeitos adversos , Piperazinas/farmacologia , Escopolamina/efeitos adversos , Sulfonas/farmacologia , Tetra-Hidronaftalenos/farmacologia , Animais , Fármacos do Sistema Nervoso Central/farmacocinética , Fármacos do Sistema Nervoso Central/farmacologia , Relação Dose-Resposta a Droga , Macaca mulatta , Masculino , Atividade Motora/efeitos dos fármacos , Piperazinas/farmacocinética , Tomografia por Emissão de Pósitrons , Sulfonas/farmacocinética , Análise e Desempenho de Tarefas , Tetra-Hidronaftalenos/farmacocinética
13.
Neuroimage ; 59(3): 2589-99, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21930214

RESUMO

BACKGROUND: Glycine transporter 1 (GlyT1) inhibitors have emerged as potential treatments for schizophrenia due to their potentiation of NMDA receptor activity by modulating the local concentrations of the NMDA co-agonist glycine. [18F]MK-6577 is a potent and selective GlyT1 inhibitor PET tracer. Although differences in ligand kinetics can be expected between non-human primates and humans, the tracer pre-clinical evaluation can provide valuable information supporting protocol design and quantification in the clinical space. The main objective of this work was to evaluate the in vivo kinetics of [18F]MK-6577 in rhesus monkey brain. Additionally, a method for estimating the tracer input function from the tracer brain tissue kinetics and venous sampling was validated. This technique was applied for determination of the dose-occupancy relationship of a GlyT1 inhibitor in monkey brain. METHODS: Compartmental and Logan graphical analysis were utilized for quantification of the [18F]MK-6577 binding using the measured tracer arterial input function. The stability of the tracer volume of distribution relative to scan length was assessed. The proposed model-based input function method takes advantage of the agreement between the tracer concentration in arterial and venous plasma from ~5 min. The approach estimates the initial peak of the input curve by adding a gamma like function term to the measured venous curve. The parameters of the model function were estimated by simultaneously fitting several brain time activity curves to a compartmental model. RESULTS: Good agreement was found between the model-based and the measured arterial plasma curve and the corresponding distribution volumes. The Logan analysis was the preferred method of analysis providing reliable and stable volume of distribution and occupancy results using a 90 and possibly 60 min scan length. CONCLUSION: The model-based input function method and Logan analysis are well suited for quantification of [18F]MK-6577 binding and GlyT1 occupancy in monkey brain.


Assuntos
Química Encefálica/fisiologia , Encéfalo/diagnóstico por imagem , Agonistas de Aminoácidos Excitatórios , Proteínas da Membrana Plasmática de Transporte de Glicina/metabolismo , Algoritmos , Animais , Corpo Estriado/diagnóstico por imagem , Relação Dose-Resposta a Droga , Agonistas de Aminoácidos Excitatórios/farmacocinética , Radioisótopos de Flúor , Proteínas da Membrana Plasmática de Transporte de Glicina/análise , Processamento de Imagem Assistida por Computador , Macaca mulatta , Modelos Neurológicos , Modelos Estatísticos , Tomografia por Emissão de Pósitrons , Receptores de N-Metil-D-Aspartato/fisiologia , Tálamo/diagnóstico por imagem
14.
Synapse ; 65(4): 261-70, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20687108

RESUMO

Two positron emission tomography radiotracers for the glycine transporter 1 (GlyT1) are reported here. Each radiotracer is a propylsulfonamide-containing benzamide and was labeled with either carbon-11 or fluorine-18. [¹¹C]CMPyPB was synthesized by the alkylation of a 3-hydroxypyridine precursor using [¹¹C]MeI, and [¹8F]MK-6577 was synthesized by a nucleophilic aromatic substitution reaction using a 2-chloropyridine precursor. Each tracer shows good uptake into rhesus monkey brain with the expected distribution of highest uptake in the pons, thalamus, and cerebellum and lower uptake in the striatum and gray matter of the frontal cortex. In vivo blockade and chase studies of [¹8F]MK-6577 showed a large specific signal and reversible binding. In vitro autoradiographic studies with [¹8F]MK-6577 showed a large specific signal in both rhesus monkey and human brain slices and a distribution consistent with the in vivo results and those reported in the literature. In vivo metabolism studies in rhesus monkeys demonstrated that only more-polar metabolites are formed for each tracer. Of these two tracers, [¹8F]MK-6577 was more extensively characterized and is a promising clinical positron emission tomography tracer for imaging GlyT1 and for measuring GlyT1 occupancy of therapeutic compounds.


Assuntos
Benzamidas/síntese química , Radioisótopos de Carbono , Radioisótopos de Flúor , Proteínas da Membrana Plasmática de Transporte de Glicina/sangue , Tomografia por Emissão de Pósitrons/métodos , Piridinas/síntese química , Sulfonamidas/síntese química , Animais , Benzamidas/sangue , Radioisótopos de Carbono/sangue , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos/métodos , Radioisótopos de Flúor/sangue , Proteínas da Membrana Plasmática de Transporte de Glicina/metabolismo , Humanos , Macaca mulatta , Piridinas/sangue , Sulfonamidas/sangue
16.
Biol Psychiatry ; 64(2): 153-61, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18339360

RESUMO

BACKGROUND: A novel, high-affinity (.7-2.0 nmol) compound that selectively activates the alpha2, alpha 3, and alpha 5 (but not alpha1) gamma-aminobutyric acid-A (GABA(A)) receptor subtypes, TPA023B (2',6-difluoro-5'-[3-(1-hydroxy-1-methylethyl) imidazo[1,2-b][1,2,4]triazin-7-yl][1,1'-biphenyl]-2-carbonitrile) was pharmacologically characterized and studied by means of positron emission tomography (PET) to determine dynamic occupancies of the benzodiazepine binding site of human brain GABA(A) receptors after a single oral dose. METHODS: Four healthy male volunteers were studied in a double-blind, randomized placebo-controlled study of which three were given a single dose of 1.5 mg TPA023B and the fourth received placebo. The time course of GABA(A) receptor occupancy was determined with multiple dynamic [(11)C]flumazenil PET studies at pre-dose baseline and 5 and 24 hours after dose. Arterial sampling and full kinetic modeling with a two-compartment model was used to calculate parametric maps of receptor availability (distribution volume V(T)) and of occupancy. RESULTS: The GABA(A) receptor occupancy as determined from [(11)C]flumazenil V(T) values in all brain regions was reduced homogeneously, on average by 52.5 +/- 1.2% after 5 hours and 46.4 +/- 6.0% after 24 hours. No serious adverse events were encountered in humans. CONCLUSIONS: Single oral doses of 1.5 mg of TPA023B correspond to average receptor occupancies in neocortical regions of 52% and 46% after 5 and 24 hours, respectively. Provided suitable ligands and quantification methods are available for the appropriate target, quantitative PET offers a unique tool for dynamic in vivo measurement of relevant on-site receptor occupancy.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/diagnóstico por imagem , Agonistas GABAérgicos/farmacologia , Agonistas GABAérgicos/farmacocinética , Tomografia por Emissão de Pósitrons , Piridazinas/farmacologia , Piridazinas/farmacocinética , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Triazóis/farmacologia , Triazóis/farmacocinética , Administração Oral , Adulto , Disponibilidade Biológica , Radioisótopos de Carbono , Método Duplo-Cego , Flumazenil , Humanos , Cinética , Masculino
17.
J Nucl Med ; 49(3): 439-45, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18287275

RESUMO

UNLABELLED: The cannabinoid type-1 (CB1) receptor is one of the most abundant G-coupled protein receptors in the human body and is responsible for signal transduction of both endogenous and exogenous cannabinoids. The endocannabinoid system is strongly implicated in regulation of homeostasis and several neuropsychiatric disorders, obesity, and associated comorbidities, such as dyslipidemia and metabolic syndrome. We have used whole-body PET/CT to characterize the biodistribution and dosimetry of a novel high-affinity, subtype-selective radioligand, (18)F-MK-9470, in healthy male and female subjects. METHODS: Eight nonobese subjects (5 men, 3 women; age, 22-54 y) underwent serial whole-body PET/CT for 6 h after a bolus injection of 251 +/- 25 MBq (18)F-MK-9470 (N-[2-(3-cyano-phenyl)-3-(4-(2-(18)F-fluorethoxy)phenyl)-1-methylpropyl]-2-(5-methyl-2-pyridyloxy)-2-methylproponamide). Source organs were delineated 3-dimensionally using the combined morphologic and functional data. Residence times were derived from time-activity profiles using both the trapezoid rule and curve fitting. Individual organ doses and effective doses were determined using the OLINDA software package, with different approaches for gastrointestinal and urinary excretion modeling. RESULTS: (18)F-MK-9470 is taken up slowly in the brain, reaching a plateau at approximately 90-120 min after bolus injection and is excreted predominantly through the hepatobiliary system. The gallbladder, upper large intestine, small intestine, and liver are the organs with the highest absorbed dose (average: 159, 98, 87, and 86 microGy/MBq, respectively). The mean effective dose (ED) was 22.8 +/- 4.3 microSv/MBq, indicating relatively low intersubject variability and a mean value in the range of many commercially available (18)F-labeled radiopharmaceuticals. Brain uptake was relatively high compared with that of existing central nervous system ligands for other receptors, between 3.2% and 4.9% of the injected dose. CONCLUSION: The estimated radiation burden of (18)F-MK-9470 for PET CB1 receptor imaging shows relatively low variability between subjects and has an acceptable ED, which allows multiple serial cerebral scans of good image quality, while remaining within the risk category class II-b defined by the World Health Organization and the International Commission for Radiation Protection for a standard injected activity (185-370 MBq).


Assuntos
Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Piridinas/farmacocinética , Receptor CB1 de Canabinoide/metabolismo , Contagem Corporal Total , Adulto , Carga Corporal (Radioterapia) , Feminino , Humanos , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Especificidade de Órgãos , Doses de Radiação , Cintilografia , Compostos Radiofarmacêuticos/análise , Compostos Radiofarmacêuticos/farmacocinética , Valores de Referência , Eficiência Biológica Relativa , Distribuição Tecidual
18.
Cell Metab ; 7(1): 68-78, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18177726

RESUMO

Cannabinoid 1 receptor (CB1R) inverse agonists are emerging as a potential obesity therapy. However, the physiological mechanisms by which these agents modulate human energy balance are incompletely elucidated. Here, we describe a comprehensive clinical research study of taranabant, a structurally novel acyclic CB1R inverse agonist. Positron emission tomography imaging using the selective CB1R tracer [(18)F]MK-9470 confirmed central nervous system receptor occupancy levels ( approximately 10%-40%) associated with energy balance/weight-loss effects in animals. In a 12-week weight-loss study, taranabant induced statistically significant weight loss compared to placebo in obese subjects over the entire range of evaluated doses (0.5, 2, 4, and 6 mg once per day) (p < 0.001). Taranabant treatment was associated with dose-related increased incidence of clinical adverse events, including mild to moderate gastrointestinal and psychiatric effects. Mechanism-of-action studies suggest that engagement of the CB1R by taranabant leads to weight loss by reducing food intake and increasing energy expenditure and fat oxidation.


Assuntos
Amidas/farmacologia , Ingestão de Energia/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Piridinas/farmacologia , Receptor CB1 de Canabinoide/agonistas , Redução de Peso/efeitos dos fármacos , Adulto , Idoso , Amidas/uso terapêutico , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Método Duplo-Cego , Gorduras/metabolismo , Humanos , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons , Piridinas/uso terapêutico
19.
Cell Metab ; 4(4): 275-82, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17011500

RESUMO

Neuropeptide Y (NPY) is a potent orexigenic neuropeptide, and antagonism of NPY Y1 and NPY Y5 receptors (NPYxR) is considered a potentially important anti-obesity drug target. We tested the hypothesis that blockade of the NPY5R will lead to weight loss in humans using MK-0557, a potent, highly selective, orally active NPY5R antagonist. The initial series of experiments reported herein, including a multiple-dose positron-emission tomography study and a 12 week proof-of concept/dose-ranging study, suggested an optimal MK-0557 dose of 1 mg/day. The hypothesis was then tested in a 52 week, multicenter, randomized, double-blind, placebo-controlled trial involving 1661 overweight and obese patients. Although statistically significant at 52 weeks, the magnitude of induced weight loss was not clinically meaningful. These observations provide the first clinical insight into the human NPY-energy homeostatic pathway and suggest that solely targeting the NPY5R in future drug development programs is unlikely to produce therapeutic efficacy.


Assuntos
Fármacos Antiobesidade/uso terapêutico , Cicloexanos/uso terapêutico , Obesidade/tratamento farmacológico , Pirazóis/uso terapêutico , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Compostos de Espiro/uso terapêutico , Administração Oral , Adolescente , Adulto , Idoso , Fármacos Antiobesidade/administração & dosagem , Peso Corporal , Cicloexanos/administração & dosagem , Relação Dose-Resposta a Droga , Método Duplo-Cego , Humanos , Pessoa de Meia-Idade , Estrutura Molecular , Placebos , Tomografia por Emissão de Pósitrons/métodos , Pirazóis/administração & dosagem , Receptores de Neuropeptídeo Y/metabolismo , Sensibilidade e Especificidade , Compostos de Espiro/administração & dosagem , Relação Estrutura-Atividade , Resultado do Tratamento
20.
Mol Imaging Biol ; 5(2): 72-8, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14499147

RESUMO

PURPOSE: We describe a method for analysis of [11C]flumazenil data using an input curve directly derived from the positron emission tomography (PET) images. PROCEDURE: The shape of the tracer plasma curve was obtained from the product of the intact flumazenil fraction in plasma in six arterial samples and the internal carotid artery time-activity curve (TAC). The resulting curve was calibrated using the [11C]flumazenil concentration in three of the six samples. The curve peak was recovered by adding an exponential function to the scaled curve whose parameters were estimated from simultaneous fittings of several tissue TACs assuming that all regions share the same input. RESULTS: Good agreement was found between the image-derived and the experimental plasma curves in six subjects. Distribution volumes were highly correlated with linear regression slope and intercept values between [0.94, 1.03] and [-0.10, 0.16], respectively. CONCLUSION: The proposed method is suitable for benzodiazepine receptor quantification requiring only a few blood samples.


Assuntos
Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Flumazenil/farmacocinética , Antagonistas da Serotonina/farmacocinética , Tomografia Computadorizada de Emissão/métodos , Radioisótopos de Carbono , Artérias Carótidas/diagnóstico por imagem , Artérias Carótidas/metabolismo , Flumazenil/sangue , Humanos , Processamento de Imagem Assistida por Computador , Cinética , Compostos Radiofarmacêuticos/farmacocinética , Antagonistas da Serotonina/sangue , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA