Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38722383

RESUMEN

PURPOSE: Mu-opioid receptors (MORs) are widely expressed in the central nervous system (CNS), peripheral organs, and immune system. This study measured the whole body distribution of MORs in rhesus macaques using the MOR selective radioligand [11C]carfentanil ([11C]CFN) on the PennPET Explorer. Both baseline and blocking studies were conducted using either naloxone or GSK1521498 to measure the effect of the antagonists on MOR binding in both CNS and peripheral organs. METHODS: The PennPET Explorer was used for MOR total-body PET imaging in four rhesus macaques using [11C]CFN under baseline, naloxone pretreatment, and naloxone or GSK1521498 displacement conditions. Logan distribution volume ratio (DVR) was calculated by using a reference model to quantitate brain regions, and the standard uptake value ratios (SUVRs) were calculated for peripheral organs. The percent receptor occupancy (%RO) was calculated to establish the blocking effect of 0.14 mg/kg naloxone or GSK1521498. RESULTS: The %RO in MOR-abundant brain regions was 75-90% for naloxone and 72-84% for GSK1521498 in blocking studies. A higher than 90% of %RO were observed in cervical spinal cord for both naloxone and GSK1521498. It took approximately 4-6 min for naloxone or GSK1521498 to distribute to CNS and displace [11C]CFN from the MOR. A smaller effect was observed in heart wall in the naloxone and GSK1521498 blocking studies. CONCLUSION: [11C]CFN total-body PET scans could be a useful approach for studying mechanism of action of MOR drugs used in the treatment of acute and chronic opioid use disorder and their effect on the biodistribution of synthetic opioids such as CFN. GSK1521498 could be a potential naloxone alternative to reverse opioid overdose.

2.
Int J Mol Sci ; 25(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38732162

RESUMEN

The synucleinopathies are a diverse group of neurodegenerative disorders characterized by the accumulation of aggregated alpha-synuclein (aSyn) in vulnerable populations of brain cells. Oxidative stress is both a cause and a consequence of aSyn aggregation in the synucleinopathies; however, noninvasive methods for detecting oxidative stress in living animals have proven elusive. In this study, we used the reactive oxygen species (ROS)-sensitive positron emission tomography (PET) radiotracer [18F]ROStrace to detect increases in oxidative stress in the widely-used A53T mouse model of synucleinopathy. A53T-specific elevations in [18F]ROStrace signal emerged at a relatively early age (6-8 months) and became more widespread within the brain over time, a pattern which paralleled the progressive development of aSyn pathology and oxidative damage in A53T brain tissue. Systemic administration of lipopolysaccharide (LPS) also caused rapid and long-lasting elevations in [18F]ROStrace signal in A53T mice, suggesting that chronic, aSyn-associated oxidative stress may render these animals more vulnerable to further inflammatory insult. Collectively, these results provide novel evidence that oxidative stress is an early and chronic process during the development of synucleinopathy and suggest that PET imaging with [18F]ROStrace holds promise as a means of detecting aSyn-associated oxidative stress noninvasively.


Asunto(s)
Encéfalo , Modelos Animales de Enfermedad , Estrés Oxidativo , Tomografía de Emisión de Positrones , Sinucleinopatías , alfa-Sinucleína , Animales , Sinucleinopatías/diagnóstico por imagen , Sinucleinopatías/metabolismo , Sinucleinopatías/patología , Tomografía de Emisión de Positrones/métodos , Ratones , alfa-Sinucleína/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/patología , Radioisótopos de Flúor , Masculino , Ratones Transgénicos , Radiofármacos , Especies Reactivas de Oxígeno/metabolismo
3.
J Med Chem ; 66(17): 12185-12202, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37651366

RESUMEN

Abnormal α-synuclein (α-syn) aggregation characterizes α-synucleinopathies, including Parkinson's disease (PD) and multiple system atrophy (MSA). However, no suitable positron emission tomography (PET) radiotracer for imaging α-syn in PD and MSA exists currently. Our structure-activity relationship studies identified 4-methoxy-N-(4-(3-(pyridin-2-yl)-3,8-diazabicyclo[3.2.1]octan-8-yl)phenyl)benzamide (4i) as a PET radiotracer candidate for imaging α-syn. In vitro assays revealed high binding of 4i to recombinant α-syn fibrils (inhibition constant (Ki) = 6.1 nM) and low affinity for amyloid beta (Aß) fibrils in Alzheimer's disease (AD) homogenates. However, [3H]4i also exhibited high specific binding to AD, progressive supranuclear palsy, and corticobasal degeneration tissues as well as PD and MSA tissues, suggesting notable affinity to tau. Nevertheless, the specific binding to pathologic α-syn aggregates in MSA post-mortem brain tissues was significantly higher than in PD tissues. This finding demonstrated the potential use of [11C]4i as a PET tracer for imaging α-syn in MSA patients. Nonhuman primate PET studies confirmed good brain uptake and rapid washout for [11C]4i.


Asunto(s)
Enfermedad de Alzheimer , Atrofia de Múltiples Sistemas , Enfermedad de Parkinson , Animales , alfa-Sinucleína , Atrofia de Múltiples Sistemas/diagnóstico por imagen , Péptidos beta-Amiloides , Tomografía de Emisión de Positrones , Encéfalo/diagnóstico por imagen
4.
Mol Imaging Biol ; 25(4): 704-719, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36991273

RESUMEN

PURPOSE: Previous studies from our lab utilized an ultra-high throughput screening method to identify compound 1 as a small molecule that binds to alpha-synuclein (α-synuclein) fibrils. The goal of the current study was to conduct a similarity search of 1 to identify structural analogs having improved in vitro binding properties for this target that could be labeled with radionuclides for both in vitro and in vivo studies for measuring α-synuclein aggregates. METHODS: Using 1 as a lead compound in a similarity search, isoxazole derivative 15 was identified to bind to α-synuclein fibrils with high affinity in competition binding assays. A photocrosslinkable version was used to confirm binding site preference. Derivative 21, the iodo-analog of 15, was synthesized, and subsequently radiolabeled isotopologs [125I]21 and [11C]21 were successfully synthesized for use in in vitro and in vivo studies, respectively. [125I]21 was used in radioligand binding studies in post-mortem Parkinson's disease (PD) and Alzheimer's disease (AD) brain homogenates. In vivo imaging of an α-synuclein mouse model and non-human primates was performed with [11C]21. RESULTS: In silico molecular docking and molecular dynamic simulation studies for a panel of compounds identified through a similarity search, were shown to correlate with Ki values obtained from in vitro binding studies. Improved affinity of isoxazole derivative 15 for α-synuclein binding site 9 was indicated by photocrosslinking studies with CLX10. Design and successful (radio)synthesis of iodo-analog 21 of isoxazole derivative 15 enabled further in vitro and in vivo evaluation. Kd values obtained in vitro with [125I]21 for α-synuclein and Aß42 fibrils were 0.48 ± 0.08 nM and 2.47 ± 1.30 nM, respectively. [125I]21 showed higher binding in human postmortem PD brain tissue compared with AD tissue, and low binding in control brain tissue. Lastly, in vivo preclinical PET imaging showed elevated retention of [11C]21 in PFF-injected mouse brain. However, in PBS-injected control mouse brain, slow washout of the tracer indicates high non-specific binding. [11C]21 showed high initial brain uptake in a healthy non-human primate, followed by fast washout that may be caused by rapid metabolic rate (21% intact [11C]21 in blood at 5 min p.i.). CONCLUSION: Through a relatively simple ligand-based similarity search, we identified a new radioligand that binds with high affinity (<10 nM) to α-synuclein fibrils and PD tissue. Although the radioligand has suboptimal selectivity for α-synuclein towards Aß and high non-specific binding, we show here that a simple in silico approach is a promising strategy to identify novel ligands for target proteins in the CNS with the potential to be radiolabeled for PET neuroimaging studies.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Parkinson , Ratones , Animales , Humanos , alfa-Sinucleína/metabolismo , Simulación del Acoplamiento Molecular , Radioisótopos de Yodo , Enfermedad de Parkinson/diagnóstico por imagen , Enfermedad de Alzheimer/metabolismo , Neuroimagen , Ligandos , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Tomografía de Emisión de Positrones/métodos
6.
Commun Biol ; 5(1): 1260, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36396952

RESUMEN

Astatine-211-parthanatine ([211At]PTT) is an alpha-emitting radiopharmaceutical therapeutic that targets poly(adenosine-diphosphate-ribose) polymerase 1 (PARP1) in cancer cells. High-risk neuroblastomas exhibit among the highest PARP1 expression across solid tumors. In this study, we evaluated the efficacy of [211At]PTT using 11 patient-derived xenograft (PDX) mouse models of high-risk neuroblastoma, and assessed hematological and marrow toxicity in a CB57/BL6 healthy mouse model. We observed broad efficacy in PDX models treated with [211At]PTT at the maximum tolerated dose (MTD 36 MBq/kg/fraction x4) administered as a fractionated regimen. For the MTD, complete tumor response was observed in 81.8% (18 of 22) of tumors and the median event free survival was 72 days with 30% (6/20) of mice showing no measurable tumor >95 days. Reversible hematological and marrow toxicity was observed 72 hours post-treatment at the MTD, however full recovery was evident by 4 weeks post-therapy. These data support clinical development of [211At]PTT for high-risk neuroblastoma.


Asunto(s)
Neuroblastoma , Humanos , Animales , Ratones , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Modelos Animales de Enfermedad
7.
Clin Cancer Res ; 28(18): 4146-4157, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35861867

RESUMEN

PURPOSE: [131I]meta-iodobenzylguanidine ([131I]MIBG) is a targeted radiotherapeutic administered systemically to deliver beta particle radiation in neuroblastoma. However, relapses in the bone marrow are common. [211At]meta-astatobenzylguanidine ([211At] MABG) is an alpha particle emitter with higher biological effectiveness and short path length which effectively sterilizes microscopic residual disease. Here we investigated the safety and antitumor activity [211At]MABG in preclinical models of neuroblastoma. EXPERIMENTAL DESIGN: We defined the maximum tolerated dose (MTD), biodistribution, and toxicity of [211At]MABG in immunodeficient mice in comparison with [131I]MIBG. We compared the antitumor efficacy of [211At]MABG with [131I]MIBG in three murine xenograft models. Finally, we explored the efficacy of [211At]MABG after tail vein xenografting designed to model disseminated neuroblastoma. RESULTS: The MTD of [211At]MABG was 66.7 MBq/kg (1.8 mCi/kg) in CB17SC scid-/- mice and 51.8 MBq/kg (1.4 mCi/kg) in NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice. Biodistribution of [211At]MABG was similar to [131I]MIBG. Long-term toxicity studies on mice administered with doses up to 41.5 MBq/kg (1.12 mCi/kg) showed the radiotherapeutic to be well tolerated. Both 66.7 MBq/kg (1.8 mCi/kg) single dose and fractionated dosing 16.6 MBq/kg/fraction (0.45 mCi/kg) × 4 over 11 days induced marked tumor regression in two of the three models studied. Survival was significantly prolonged for mice treated with 12.9 MBq/kg/fraction (0.35 mCi/kg) × 4 doses over 11 days [211At]MABG in the disseminated disease (IMR-05NET/GFP/LUC) model (P = 0.003) suggesting eradication of microscopic disease. CONCLUSIONS: [211At]MABG has significant survival advantage in disseminated models of neuroblastoma. An alpha particle emitting radiopharmaceutical may be effective against microscopic disseminated disease, warranting clinical development.


Asunto(s)
Astato , Neuroblastoma , 3-Yodobencilguanidina/efectos adversos , Partículas alfa/uso terapéutico , Animales , Astato/uso terapéutico , Guanidinas/uso terapéutico , Humanos , Radioisótopos de Yodo/uso terapéutico , Ratones , Ratones Endogámicos NOD , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/radioterapia , Radiofármacos/efectos adversos , Distribución Tisular , Células Tumorales Cultivadas
8.
EJNMMI Res ; 12(1): 43, 2022 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-35895177

RESUMEN

BACKGROUND: Oxidative stress is implicated in the pathogenesis of the most common neurodegenerative diseases, such as Alzheimer's disease (AD). However, tracking oxidative stress in the brain has proven difficult and impeded its use as a biomarker. Herein, we investigate the utility of a novel positron emission tomography (PET) tracer, [18F]ROStrace, as a biomarker of oxidative stress throughout the course of AD in the well-established APP/PS1 double-mutant mouse model. PET imaging studies were conducted in wild-type (WT) and APP/PS1 mice at 3 different time points, representing early (5 mo.), middle (10 mo.), and advanced (16 mo.) life (n = 6-12, per sex). Semi-quantitation SUVRs of the plateau phase (40-60 min post-injection; SUVR40-60) of ten brain subregions were designated by the Mirrione atlas and analyzed by Pmod. Statistical parametric mapping (SPM) was used to distinguish brain regions with elevated ROS in APP/PS1 relative to WT in both sexes. The PET studies were validated by ex vivo autoradiography and immunofluorescence with the parent compound, dihydroethidium. RESULTS: [18F]ROStrace retention was increased in the APP/PS1 brain compared to age-matched controls by 10 mo. of age (p < 0.0001) and preceded the accumulation of oxidative damage in APP/PS1 neurons at 16 mo. (p < 0.005). [18F]ROStrace retention and oxidative damages were higher and occurred earlier in female APP/PS1 mice as measured by PET (p < 0.001), autoradiography, and immunohistochemistry (p < 0.05). [18F]ROStrace differences emerged midlife, temporally and spatially correlating with increased Aß burden (r2 = 0.36; p = 0.0003), which was also greatest in the female brain (p < 0.001). CONCLUSIONS: [18F]ROStrace identifies increased oxidative stress and neuroinflammation in APP/PS1 female mice, concurrent with increased amyloid burden midlife. Differences in oxidative stress during this crucial time may partially explain the sexual dimorphism in AD. [18F]ROStrace may provide a long-awaited tool to stratify at-risk patients who may benefit from antioxidant therapy prior to irreparable neurodegeneration.

9.
Mol Ther ; 28(1): 42-51, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31668558

RESUMEN

Cell-based therapeutics have considerable promise across diverse medical specialties; however, reliable human imaging of the distribution and trafficking of genetically engineered cells remains a challenge. We developed positron emission tomography (PET) probes based on the small-molecule antibiotic trimethoprim (TMP) that can be used to image the expression of the Escherichia coli dihydrofolate reductase enzyme (eDHFR) and tested the ability of [18F]-TMP, a fluorine-18 probe, to image primary human chimeric antigen receptor (CAR) T cells expressing the PET reporter gene eDHFR, yellow fluorescent protein (YFP), and Renilla luciferase (rLuc). Engineered T cells showed an approximately 50-fold increased bioluminescent imaging signal and 10-fold increased [18F]-TMP uptake compared to controls in vitro. eDHFR-expressing anti-GD2 CAR T cells were then injected into mice bearing control GD2- and GD2+ tumors. PET/computed tomography (CT) images acquired on days 7 and 13 demonstrated early residency of CAR T cells in the spleen followed by on-target redistribution to the GD2+ tumors. This was corroborated by autoradiography and anti-human CD8 immunohistochemistry. We found a high sensitivity of detection for identifying tumor-infiltrating CD8 CAR T cells, ∼11,000 cells per mm3. These data suggest that the [18F]-TMP/eDHFR PET pair offers important advantages that could better allow investigators to monitor immune cell trafficking to tumors in patients.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Escherichia coli/enzimología , Genes Reporteros , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Receptores Quiméricos de Antígenos/metabolismo , Tetrahidrofolato Deshidrogenasa/genética , Animales , Linfocitos T CD8-positivos/metabolismo , Femenino , Radioisótopos de Flúor , Gangliósidos/metabolismo , Células HCT116 , Voluntarios Sanos , Xenoinjertos/diagnóstico por imagen , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Bazo/diagnóstico por imagen , Bazo/metabolismo , Tetrahidrofolato Deshidrogenasa/metabolismo , Trimetoprim
10.
Biochem Biophys Res Commun ; 516(2): 397-401, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31221481

RESUMEN

Reactive oxygen species (ROS) are believed to play an important role in the proinflammatory form of neuroinflammation. Therefore, the availability of a radiotracer labeled with a positron-emitting radionuclide that can measure levels of ROS in tissue could provide a valuable method for imaging neuroinflammation in vivo with the functional imaging technique positron emission tomography (PET). We previously reported the synthesis and in vivo evaluation of [18F]ROStrace, a radiotracer for imaging ROS in vivo with PET, in an LPS model of neuroinflammation. In the current study, we conducted additional validation studies aimed at determining the cellular localization of this radiotracer in the same model. Our results indicate that [18F]ROStrace is primarily localized in microglia/macrophages and neurons in LPS-treated animals, and provide further support in the use of this radiotracer as a PET-based probe for imaging the proinflammatory form of neuroinflammation.


Asunto(s)
Autorradiografía , Etidio/análogos & derivados , Radioisótopos de Flúor/metabolismo , Lipopolisacáridos/farmacología , Imagen Óptica , Especies Reactivas de Oxígeno/metabolismo , Animales , Anticuerpos/metabolismo , Etidio/metabolismo , Femenino , Ratones Endogámicos BALB C
11.
Mol Cancer Ther ; 18(7): 1195-1204, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31072830

RESUMEN

Alpha-emitters can be pharmacologically delivered for irradiation of single cancer cells, but cellular lethality could be further enhanced by targeting alpha-emitters directly to the nucleus. PARP-1 is a druggable protein in the nucleus that is overexpressed in neuroblastoma compared with normal tissues and is associated with decreased survival in high-risk patients. To exploit this, we have functionalized a PARP inhibitor (PARPi) with an alpha-emitter astatine-211. This approach offers enhanced cytotoxicity from conventional PARPis by not requiring enzymatic inhibition of PARP-1 to elicit DNA damage; instead, the alpha-particle directly induces multiple double-strand DNA breaks across the particle track. Here, we explored the efficacy of [211At]MM4 in multiple cancers and found neuroblastoma to be highly sensitive in vitro and in vivo Furthermore, alpha-particles delivered to neuroblastoma show antitumor effects and durable responses in a neuroblastoma xenograft model, especially when administered in a fractionated regimen. This work provides the preclinical proof of concept for an alpha-emitting drug conjugate that directly targets cancer chromatin as a therapeutic approach for neuroblastoma and perhaps other cancers.


Asunto(s)
Neuroblastoma/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Neuroblastoma/mortalidad , Neuroblastoma/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Análisis de Supervivencia
12.
Clin Cancer Res ; 25(10): 3063-3073, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30692100

RESUMEN

PURPOSE: Cyclin-dependent kinase 4/6 (CDK4/6) inhibitors in combination with endocrine-therapy have emerged as an important regimen of care for estrogen receptor (ER)-positive metastatic breast cancer, although identifying predictive biomarkers remains a challenge. We assessed the ability of two PET-proliferation tracers, [18F]FLT and [18F]ISO-1, for evaluating response to CDK4/6-inhibitor (palbociclib) and ER-antagonist (fulvestrant). EXPERIMENTAL DESIGN: To determine the effect of CDK4/6 inhibition combined with estrogen-blockade, we assessed cell proliferation in six breast cancer cell lines after 1, 3, and 6 days of treatment with palbociclib and/or fulvestrant. These data were correlated to in vitro radiotracer assays and results were verified by longitudinal [18F]FLT and [18F]ISO-1 micro-PET imaging performed in MCF7 tumor-bearing mice. RESULTS: All palbociclib-sensitive cell lines showed decreased [18F]FLT accumulation and S-phase depletion after treatment, with both measures augmented by combination therapy. In contrast, these cells showed changes in [18F]ISO-1 analogue-binding and G0 arrest only after prolonged treatment. MicroPET imaging of MCF7 xenografts showed a significant decrease in [18F]FLT but no changes in [18F]ISO-1 uptake in all treated mice on day 3. On day 14, however, mice treated with combination therapy showed a significant decrease in [18F]ISO-1, corresponding to G0 arrest, while maintaining reduced [18F]FLT uptake, which corresponded to S-phase depletion. CONCLUSIONS: Our data suggest complementary roles of [18F]FLT and [18F]ISO-1 PET in evaluating tumor-proliferation after combined CDK4/6 inhibitor and endocrine therapy in breast cancer. [18F]FLT is more sensitive to immediate changes in S-phase, whereas [18F]ISO-1 can assess more delayed changes related to cell-cycle arrest and transition to G0 quiescence from combination therapy. These data suggest a potential role for early prediction of long-term response using these imaging biomarkers.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Animales , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Didesoxinucleósidos , Antagonistas del Receptor de Estrógeno/administración & dosificación , Femenino , Radioisótopos de Flúor , Fulvestrant/administración & dosificación , Humanos , Estudios Longitudinales , Células MCF-7 , Ratones , Ratones SCID , Piperazinas/administración & dosificación , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Piridinas/administración & dosificación , Radiofármacos , Distribución Aleatoria , Receptores de Estrógenos/antagonistas & inhibidores , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Bioorg Chem ; 83: 242-249, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30390553

RESUMEN

Poly(ADP-ribose)polymerase-1 inhibitor (PARPi) AZD2461 was designed to be a weak P-glycoprotein (P-gp) analogue of FDA approved olaparib. With this chemical property in mind, we utilized the AZD2461 ligand architecture to develop a CNS penetrant and PARP-1 selective imaging probe, in order to investigate PARP-1 mediated neuroinflammation and neurodegenerative diseases, such as Alzheimer's and Parkinson's. Our work led to the identification of several high-affinity PARPi, including AZD2461 congener 9e (PARP-1 IC50 = 3.9 ±â€¯1.2 nM), which was further evaluated as a potential 18F-PET brain imaging probe. However, despite the similar molecular scaffolds of 9e and AZD2461, our studies revealed non-appreciable brain-uptake of [18F]9e in non-human primates, suggesting AZD2461 to be non-CNS penetrant.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Ftalazinas/farmacología , Piperidinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/agonistas , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Radioisótopos de Flúor/química , Humanos , Macaca mulatta , Masculino , Ratones Endogámicos BALB C , Ftalazinas/síntesis química , Piperidinas/síntesis química
14.
EJNMMI Phys ; 5(1): 21, 2018 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-30294746

RESUMEN

BACKGROUND: [18F]Fluortriopride (FTP) was developed as a dopamine D3-selective radiotracer, thought to be important to neurobiological reward pathways and implicated in drug addiction, Parkinson's disease, and schizophrenia. Preclinical radiation dosimetry studies found the gallbladder wall received the highest dose. A gallbladder dose reduction intervention was simulated using a novel reduction model for healthy adults following fatty-meal consumption. The goals of this study were to assess whole body FTP human dosimetry and determine the feasibility of reducing absorbed dose to the gallbladder wall. RESULTS: Effective dose without a fatty meal was 0.022 ± 0.002 mSv/MBq (± standard deviation) with highest organ dose of 0.436 ± 0.178 mSv/MBq to the gallbladder wall (n = 10). Predicted gallbladder dose reduction with fatty meal consumed was 67.4% (n = 10). Meal consumption by four repeat volunteers decreased average gallbladder dose by 71.3% (n = 4) compared to the original ten volunteers. CONCLUSIONS: Observed effective doses were adequately low to continue studying FTP uptake in humans. Validated dosimetry simulations indicate up to a 71% reduction in gallbladder dose can be achieved by employing intrinsic physiology to contract the gallbladder via fatty meal ingestion. This methodology for predicting gallbladder absorbed dose reduction from fatty meal consumption can be applied to other radiopharmaceuticals and radiotherapies.

15.
J Clin Invest ; 128(5): 2116-2126, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29509546

RESUMEN

BACKGROUND: Poly(ADP-ribose) polymerase (PARP) inhibitors are effective in a broad population of patients with ovarian cancer; however, resistance caused by low enzyme expression of the drug target PARP-1 remains to be clinically evaluated in this context. We hypothesize that PARP-1 expression is variable in ovarian cancer and can be quantified in primary and metastatic disease using a novel PET imaging agent. METHODS: We used a translational approach to describe the significance of PET imaging of PARP-1 in ovarian cancer. First, we produced PARP1-KO ovarian cancer cell lines using CRISPR/Cas9 gene editing to test the loss of PARP-1 as a resistance mechanism to all clinically used PARP inhibitors. Next, we performed preclinical microPET imaging studies using ovarian cancer patient-derived xenografts in mouse models. Finally, in a phase I PET imaging clinical trial we explored PET imaging as a regional marker of PARP-1 expression in primary and metastatic disease through correlative tissue histology. RESULTS: We found that deletion of PARP1 causes resistance to all PARP inhibitors in vitro, and microPET imaging provides proof of concept as an approach to quantify PARP-1 in vivo. Clinically, we observed a spectrum of standard uptake values (SUVs) ranging from 2-12 for PARP-1 in tumors. In addition, we found a positive correlation between PET SUVs and fluorescent immunohistochemistry for PARP-1 (r2 = 0.60). CONCLUSION: This work confirms the translational potential of a PARP-1 PET imaging agent and supports future clinical trials to test PARP-1 expression as a method to stratify patients for PARP inhibitor therapy. TRIAL REGISTRATION: Clinicaltrials.gov NCT02637934. FUNDING: Research reported in this publication was supported by the Department of Defense OC160269, a Basser Center team science grant, NIH National Cancer Institute R01CA174904, a Department of Energy training grant DE-SC0012476, Abramson Cancer Center Radiation Oncology pilot grants, the Marsha Rivkin Foundation, Kaleidoscope of Hope Foundation, and Paul Calabresi K12 Career Development Award 5K12CA076931.


Asunto(s)
Medios de Contraste/administración & dosificación , Resistencia a Antineoplásicos , Neoplasias Ováricas , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas/administración & dosificación , Tomografía Computarizada por Tomografía de Emisión de Positrones , Adulto , Anciano , Animales , Línea Celular Tumoral , Femenino , Xenoinjertos , Humanos , Ratones , Persona de Mediana Edad , Proteínas de Neoplasias , Trasplante de Neoplasias , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo
16.
ACS Chem Neurosci ; 9(3): 578-586, 2018 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-29099578

RESUMEN

Reactive oxygen species (ROS) are believed to play a major role in the proinflammatory, M1-polarized form of neuroinflammation. However, it has been difficult to assess the role of ROS and their role in neuroinflammation in animal models of disease because of the absence of probes capable of measuring their presence with the functional imaging technique positron emission tomography (PET). This study describes the synthesis and in vivo evaluation of [18F]ROStrace, a radiotracer for imaging superoxide in vivo with PET, in an LPS model of neuroinflammation. [18F]ROStrace was found to rapidly cross the blood-brain barrier (BBB) and was trapped in the brain of LPS-treated animals but not the control group. [18F] ox-ROStrace, the oxidized form of [18F]ROStrace, did not cross the BBB. These data suggest that [18F]ROStrace is a suitable radiotracer for imaging superoxide levels in the central nervous system with PET.


Asunto(s)
Radioisótopos de Flúor/metabolismo , Inflamación/metabolismo , Tomografía de Emisión de Positrones , Radiofármacos/farmacología , Superóxidos/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Inflamación/patología , Ratones , Tomografía de Emisión de Positrones/métodos , Especies Reactivas de Oxígeno/metabolismo , Distribución Tisular/fisiología
17.
Proc Natl Acad Sci U S A ; 114(31): 8372-8377, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28716936

RESUMEN

There is often overlap in the diagnostic features of common pathologic processes such as infection, sterile inflammation, and cancer both clinically and using conventional imaging techniques. Here, we report the development of a positron emission tomography probe for live bacterial infection based on the small-molecule antibiotic trimethoprim (TMP). [18F]fluoropropyl-trimethoprim, or [18F]FPTMP, shows a greater than 100-fold increased uptake in vitro in live bacteria (Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa) relative to controls. In a rodent myositis model, [18F]FPTMP identified live bacterial infection without demonstrating confounding increased signal in the same animal from other etiologies including chemical inflammation (turpentine) and cancer (breast carcinoma). Additionally, the biodistribution of [18F]FPTMP in a nonhuman primate shows low background in many important tissues that may be sites of infection such as the lungs and soft tissues. These results suggest that [18F]FPTMP could be a broadly useful agent for the sensitive and specific imaging of bacterial infection with strong translational potential.


Asunto(s)
Antibacterianos/farmacología , Infecciones por Escherichia coli/diagnóstico , Escherichia coli/metabolismo , Tomografía de Emisión de Positrones/métodos , Infecciones por Pseudomonas/diagnóstico , Pseudomonas aeruginosa/metabolismo , Infecciones Estafilocócicas/diagnóstico , Staphylococcus aureus/metabolismo , Trimetoprim/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/microbiología , Radioisótopos de Flúor/química , Células HCT116 , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Infecciones por Pseudomonas/microbiología , Radiofármacos/farmacología , Infecciones Estafilocócicas/microbiología , Trimetoprim/química
18.
Mol Ther ; 25(1): 120-126, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28129108

RESUMEN

There is a need for improved methods to image genetically engineered cells, including immune cells used for cell-based therapy. Given the genetic manipulation inherent to gene therapy, the use of a reporter protein is a logical solution and positron emission tomography (PET) can provide the desired sensitivity and spatial localization. We developed a broadly applicable PET imaging strategy based on the small bacterial protein E. coli dihydrofolate reductase (Ec dhfr) and its highly specific small molecule inhibitor, trimethoprim (TMP). The difference in TMP affinity for bacterial compared to mammalian DHFR suggests that a TMP radioligand would have a low background in unmodified mammalian tissues and high retention in Ec dhfr engineered cells, providing high contrast imaging. Here, we describe the in vitro properties of [11C]TMP and show over 10-fold increased signal in transgenic Ec dhfr cells compared to control. In a mouse xenograft model, [11C]TMP rapidly accumulated in Ec dhfr carrying cells within minutes of intravenous administration. Moreover, [11C]TMP can identify less than a million xenografted cells in a small volume in tissues other than the abdominal compartment. This limit of detection is a clinically relevant number and bodes well for clinical translation especially given that [11C]TMP is an isotopologue of clinically approved antibiotic.


Asunto(s)
Radioisótopos de Carbono , Genes Reporteros , Imagen Molecular , Tomografía de Emisión de Positrones/métodos , Trimetoprim , Animales , Línea Celular , Ratones , Sensibilidad y Especificidad , Microtomografía por Rayos X
19.
Cancer Biother Radiopharm ; 32(1): 9-15, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28118040

RESUMEN

INTRODUCTION: Poly (ADP-ribose) polymerase 1 (PARP-1) is the major target of clinical PARP inhibitors and is a potential predictive biomarker for response to therapy. Due to the limited success of PARP inhibitors as monotherapy, investigators have shifted the clinical role of PARP inhibitors to the adjuvant setting. In this study, we evaluate the radiotracer [18F]FluorThanatrace ([18F]FTT) as a marker of PARP expression in vitro and the associated biological implications of PARP-1 expression in PARP inhibitor treatment adjuvant to radiation therapy. MATERIALS AND METHODS: SNU-251 (BRCA1-mutant) and SKOV3 (BRCA1-WT) cell lines were evaluated in vitro by using the radiotracer [18F]FTT. Pharmacological binding assays were performed at baseline and were correlated with PARP-1 protein expression measured by Western blot protein analysis. Cell viability and clonogenic assays were used to characterize in vitro cytotoxicity for treatments, including: PARP inhibitors alone, radiation alone, and PARP inhibitor adjuvant to radiation. Western blot protein analysis was used to assess response to treatment by using γH2AX to measure DNA damage and PAR to measure the catalytic inhibition of PARP. RESULTS: [18F]FTT was capable of measuring PARP-1 protein expression in vitro and corresponded to Western blot protein analysis at baseline. The addition of a PARP inhibitor enhanced radiation effects in both cell lines; however, a greater synergy was observed in the SNU-251 cell line that expresses a BRCA1 mutation and homologous recombination deficiency. Western blot protein analysis showed that the addition of a PARP inhibitor adjuvant to radiation increases DNA damage in both cell lines and reduces PARP enzymatic activity as measured by PAR. CONCLUSIONS: In this work, we found that PARP-1 expression positively corresponds in vitro to the response of PARP inhibitors in combination with radiation therapy in ovarian cancer.


Asunto(s)
Biomarcadores Farmacológicos/metabolismo , Neoplasias Ováricas/terapia , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Línea Celular Tumoral , Supervivencia Celular , Quimioterapia Adyuvante , Daño del ADN/efectos de los fármacos , Femenino , Radioisótopos de Flúor , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/radioterapia , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico
20.
Nucl Med Biol ; 43(11): 721-731, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27589334

RESUMEN

INTRODUCTION: Nine novel analogues were synthesized including a 6-carbon spacer analogue of ISO-1 (7). They have moderate binding affinity for sigma-2 (σ2) receptors and high selectivity for σ2 receptors relative to sigma-1 (σ1) receptors. METHODS: ([18F]7) was synthesized and evaluated as a candidate ligand for positron emission (PET) imaging of the σ2 receptor in tumors. Radioligand [18F]7 was radiolabeled with 18F via displacement of the corresponding mesylate precursor with [18F]fluoride. Cellular uptake study of [18F]7 was performed in EMT-6 tumor cell, and in vivo biodistribution study of [18F]7 and microPET imaging study of [18F]3 and [18F]7 carried out in female Balb/c mice bearing EMT-6 tumors. RESULTS: [18F]7 had a respectable tumor uptake (1.55%ID/g at 60min post-injection) and high tumor/muscle ratios at 60 and 120min post-injection. MicroPET imaging of [18F]7 in tumor-bearing mice as above showed significant tumor localization and a high tumor/muscle ratio as well. CONCLUSIONS: These results are similar to or better than [18F]ISO-1 ([18F]3), which indicates that [18F]7 has potential for imaging the σ2 receptor status of solid tumors.


Asunto(s)
Benzamidas/química , Carbono/química , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Receptores sigma/metabolismo , Tetrahidroisoquinolinas/química , Animales , Benzamidas/metabolismo , Benzamidas/farmacocinética , Transporte Biológico , Línea Celular Tumoral , Femenino , Ratones , Conformación Molecular , Radioquímica , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...