Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Neil Vasdev is active.

Publication


Featured researches published by Neil Vasdev.


Annals of Neurology | 2016

Tau positron emission tomographic imaging in aging and early Alzheimer disease

Keith Johnson; Aaron P. Schultz; Rebecca A. Betensky; J. Alex Becker; Jorge Sepulcre; Dorene M. Rentz; Elizabeth C. Mormino; Jasmeer P. Chhatwal; Rebecca Amariglio; Kate V. Papp; Gad A. Marshall; Mark W. Albers; Samantha Mauro; Lesley Pepin; Jonathan Alverio; Kelly Judge; Marlie Philiossaint; Timothy M. Shoup; Daniel Yokell; Bradford C. Dickerson; Teresa Gomez-Isla; Bradley T. Hyman; Neil Vasdev; Reisa A. Sperling

Detection of focal brain tau deposition during life could greatly facilitate accurate diagnosis of Alzheimer disease (AD), staging and monitoring of disease progression, and development of disease‐modifying therapies.


Annals of Neurology | 2015

Tau PET imaging in aging and early Alzheimer's disease

Keith Johnson; Aaron P. Schultz; Rebecca A. Betensky; J. Alex Becker; Jorge Sepulcre; Dorene M. Rentz; Elizabeth C. Mormino; Jasmeer P. Chhatwal; Rebecca Amariglio; Kate V. Papp; Gad A. Marshall; Mark W. Albers; Samantha Mauro; Lesley Pepin; Jonathan Alverio; Kelly Judge; Marlie Philiossaint; Timothy M. Shoup; Daniel Yokell; Bradford C. Dickerson; Teresa Gomez-Isla; Bradley T. Hyman; Neil Vasdev; Reisa A. Sperling

Detection of focal brain tau deposition during life could greatly facilitate accurate diagnosis of Alzheimer disease (AD), staging and monitoring of disease progression, and development of disease‐modifying therapies.


Annals of Neurology | 2015

Validating novel tau positron emission tomography tracer [F-18]-AV-1451 (T807) on postmortem brain tissue

Marta Marquié; Marc D. Normandin; Charles R. Vanderburg; Isabel Costantino; Elizabeth A. Bien; Lisa G. Rycyna; William E. Klunk; Chester A. Mathis; Milos D. Ikonomovic; Manik L. Debnath; Neil Vasdev; Bradford C. Dickerson; Stephen N. Gomperts; John H. Growdon; Keith Johnson; Matthew P. Frosch; Bradley T. Hyman; Teresa Gomez-Isla

To examine region‐ and substrate‐specific autoradiographic and in vitro binding patterns of positron emission tomography tracer [F‐18]‐AV‐1451 (previously known as T807), tailored to allow in vivo detection of paired helical filament‐tau–containing lesions, and to determine whether there is off‐target binding to other amyloid/non‐amyloid proteins.


Nuclear Medicine and Biology | 2008

Radiosynthesis and initial evaluation of [18F]-FEPPA for PET imaging of peripheral benzodiazepine receptors

Alan A. Wilson; Armando Garcia; Jun Parkes; Patrick McCormick; Karin A. Stephenson; Sylvain Houle; Neil Vasdev

INTRODUCTION A novel [18F]-radiolabelled phenoxyanilide, [18F]-FEPPA, has been synthesized and evaluated, in vitro and ex vivo, as a potential positron emission tomography imaging agent for the peripheral benzodiazepine receptor (PBR). METHODS [18F]-FEPPA and two other radiotracers for imaging PBR, namely [11C]-PBR28 and [11C]-PBR28-d3, were synthesised and evaluated in vitro and ex vivo as potential PBR imaging agents. RESULTS [18F]-FEPPA is efficiently prepared in one step from its tosylate precursor and [18F]-fluoride in high radiochemical yields and at high specific activity. FEPPA displayed a Ki of 0.07 nM for PBR in rat mitochondrial membrane preparations and a suitable lipophilicity for brain penetration (log P of 2.99 at pH 7.4). Upon intravenous injection into rats, [18F]-FEPPA showed moderate brain uptake [standard uptake value (SUV) of 0.6 at 5 min] and a slow washout (SUV of 0.35 after 60 min). Highest uptake of radioactivity was seen in the hypothalamus and olfactory bulb, regions previously reported to be enriched in PBR in rat brain. Analysis of plasma and brain extracts demonstrated that [18F]-FEPPA was rapidly metabolized, but no lipophilic metabolites were observed in either preparation and only 5% radioactive metabolites were present in brain tissue extracts. Blocking studies to determine the extent of specific binding of [18F]-FEPPA in rat brain were problematic due to large perturbations in circulating radiotracer and the lack of a reference region. CONCLUSIONS Further evaluation of the potential of [18F]-FEPPA will require the employment of rigorous kinetic models and/or appropriate animal models.


Nature Communications | 2014

Spirocyclic hypervalent iodine(III)-mediated radiofluorination of non-activated and hindered aromatics

Benjamin H. Rotstein; Nickeisha A. Stephenson; Neil Vasdev; Steven H. Liang

Fluorine-18 (t½=109.7 min) is the most commonly used isotope to prepare radiopharmaceuticals for molecular imaging by positron emission tomography (PET). Nucleophilic aromatic substitution reactions of suitably activated (electron-deficient) aromatic substrates with no-carrier-added [(18)F]fluoride ion are routinely carried out in the synthesis of radiotracers in high specific activities. Despite extensive efforts to develop a general (18)F-labelling technique for non-activated arenes there is an urgent and unmet need to achieve this goal. Here we describe an effective solution that relies on the chemistry of spirocyclic hypervalent iodine(III) complexes, which serve as precursors for rapid, one-step regioselective radiofluorination with [(18)F]fluoride. This methodology proves to be efficient for radiolabelling a diverse range of non-activated functionalized arenes and heteroarenes, including arene substrates bearing electron-donating groups, bulky ortho functionalities, benzylic substituents and meta-substituted electron-withdrawing groups. Polyfunctional molecules and a range of previously elusive (18)F-labelled building blocks, compounds and radiopharmaceuticals are synthesized.


JAMA Neurology | 2017

Association of In Vivo [18F]AV-1451 Tau PET Imaging Results With Cortical Atrophy and Symptoms in Typical and Atypical Alzheimer Disease

Chenjie Xia; Sara Makaretz; Christina Caso; Scott M. McGinnis; Stephen N. Gomperts; Jorge Sepulcre; Teresa Gomez-Isla; Bradley T. Hyman; Aaron P. Schultz; Neil Vasdev; Keith Johnson; Bradford C. Dickerson

Importance Previous postmortem studies have long demonstrated that neurofibrillary tangles made of hyperphosphorylated tau proteins are closely associated with Alzheimer disease clinical phenotype and neurodegeneration pattern. Validating these associations in vivo will lead to new diagnostic tools for Alzheimer disease and better understanding of its neurobiology. Objective To examine whether topographical distribution and severity of hyperphosphorylated tau pathologic findings measured by fluorine 18–labeled AV-1451 ([18F]AV-1451) positron emission tomographic (PET) imaging are linked with clinical phenotype and cortical atrophy in patients with Alzheimer disease. Design, Setting, and Participants This observational case series, conducted from July 1, 2012, to July 30, 2015, in an outpatient referral center for patients with neurodegenerative diseases, included 6 patients: 3 with typical amnesic Alzheimer disease and 3 with atypical variants (posterior cortical atrophy, logopenic variant primary progressive aphasia, and corticobasal syndrome). Patients underwent [18F]AV-1451 PET imaging to measure tau burden, carbon 11–labeled Pittsburgh Compound B ([11C]PiB) PET imaging to measure amyloid burden, and structural magnetic resonance imaging to measure cortical thickness. Seventy-seven age-matched controls with normal cognitive function also underwent structural magnetic resonance imaging but not tau or amyloid PET imaging. Main Outcomes and Measures Tau burden, amyloid burden, and cortical thickness. Results In all 6 patients (3 women and 3 men; mean age 61.8 years), the underlying clinical phenotype was associated with the regional distribution of the [18F]AV-1451 signal. Furthermore, within 68 cortical regions of interest measured from each patient, the magnitude of cortical atrophy was strongly correlated with the magnitude of [18F]AV-1451 binding (3 patients with amnesic Alzheimer disease, r = –0.82; P < .001; r = –0.70; P < .001; r = –0.58; P < .001; and 3 patients with nonamnesic Alzheimer disease, r = –0.51; P < .001; r = –0.63; P < .001; r = –0.70; P < .001), but not of [11C]PiB binding. Conclusions and Relevance These findings provide further in vivo evidence that distribution of the [18F]AV-1451 signal as seen on results of PET imaging is a valid marker of clinical symptoms and neurodegeneration. By localizing and quantifying hyperphosphorylated tau in vivo, results of tau PET imaging will likely serve as a key biomarker that links a specific type of molecular Alzheimer disease neuropathologic condition with clinically significant neurodegeneration, which will likely catalyze additional efforts to develop disease-modifying therapeutics.


Nuclear Medicine and Biology | 2011

[11C]CURB: Evaluation of a novel radiotracer for imaging fatty acid amide hydrolase by positron emission tomography

Alan A. Wilson; Armando Garcia; Jun Parkes; Sylvain Houle; Junchao Tong; Neil Vasdev

INTRODUCTION Fatty acid amide hydrolase (FAAH) is the enzyme responsible for metabolising the endogenous cannabinoid, anandamide, and thus represents an important target for molecular imaging. To date, no radiotracer has been shown to be useful for imaging of FAAH using either positron emission tomography (PET) or single photon emission computed tomography (SPECT). We here determine the suitability of a novel carbon-11-labeled inhibitor of FAAH via ex vivo biodistribution studies in rat brain in conjunction with pharmacological challenges. METHODS A potent irreversible inhibitor of FAAH, URB694, radiolabeled with carbon-11 in the carbonyl position ([(11)C]CURB), was administered to male rats via tail-vein injection. Rats were sacrificed at various time points postinjection, and tissue samples were dissected, counted and weighed. Specific binding to FAAH was investigated by pretreatment of animals with URB694 or URB597. For metabolism and mechanism of binding studies, whole brains were excised post-radiotracer injection, homogenised and extracted exhaustively with 80% aq. acetonitrile to determine the time course and fraction of radioactivity that was irreversibly bound to brain parenchyma. RESULTS Upon intravenous injection into rats, [(11)C]CURB showed high brain uptake [standard uptake value (SUV) of 1.6-2.4 at 5 min] with little washout over time, which is characteristic of irreversible binding. Highest uptake of radioactivity was seen in the cortex, intermediate in the cerebellum and lowest in the hypothalamus, reflecting the reported distribution of FAAH. Brain uptake of radioactivity was decreased in a dose-dependent manner by pretreatment with increasing amounts of URB694, demonstrating that binding was saturable. Pretreatment with the well-characterised FAAH inhibitor, URB597, reduced binding in all brain regions by 70-80%. Homogenised brain extraction experiments demonstrated unequivocally that [(11)C]CURB was irreversibly bound to FAAH. CONCLUSIONS The title radiotracer demonstrates favourable properties such as good brain uptake, regional heterogeneity and specificity of binding based on ex vivo biodistribution studies in conscious rat brain. [(11)C]CURB represents a highly promising radiotracer for the imaging of FAAH using PET.


Molecules | 2010

Radiolabeled Small Molecule Protein Kinase Inhibitors for Imaging with PET or SPECT

Justin W. Hicks; Henry F. VanBrocklin; Alan A. Wilson; Sylvain Houle; Neil Vasdev

Imaging protein kinase expression with radiolabeled small molecule inhibitors has been actively pursued to monitor the clinical potential of targeted therapeutics and treatments as well as to determine kinase receptor density changes related to disease progression. The goal of the present review is to provide an overview of the breadth of radiolabeled small molecules that have been synthesized to target intracellular protein kinases, not only for imaging in oncology, but also for other areas of interest, particularly the central nervous system. Considerable radiotracer development has focused on imaging receptor tyrosine kinases of growth factors, protein kinases A, B and C, and glycogen synthase kinase–3β. Design considerations, structural attributes and relevant biological results are summarized.


Chemical Communications | 2013

11CO2 fixation: a renaissance in PET radiochemistry

Benjamin H. Rotstein; Steven H. Liang; Jason P. Holland; Thomas Lee Collier; Jacob M. Hooker; Alan A. Wilson; Neil Vasdev

Carbon-11 labelled carbon dioxide is the cyclotron-generated feedstock reagent for most positron emission tomography (PET) tracers using this radionuclide. Most carbon-11 labels, however, are installed using derivative reagents generated from [(11)C]CO2. In recent years, [(11)C]CO2 has seen a revival in applications for the direct incorporation of carbon-11 into functional groups such as ureas, carbamates, oxazolidinones, carboxylic acids, esters, and amides. This review summarizes classical [(11)C]CO2 fixation strategies using organometallic reagents and then focuses on newly developed methods that employ strong organic bases to reversibly capture [(11)C]CO2 into solution, thereby enabling highly functionalized labelled compounds to be prepared. Labelled compounds and radiopharmaceuticals that have been translated to the clinic are highlighted.


Journal of Labelled Compounds and Radiopharmaceuticals | 2013

A concise radiosynthesis of the tau radiopharmaceutical, [18F]T807

Timothy M. Shoup; Daniel Yokell; Peter A. Rice; Raul N. Jackson; Eli Livni; Keith Johnson; Thomas J. Brady; Neil Vasdev

Fluorine-18 labeled 7-(6-fluoropyridin-3-yl)-5H-pyrido[4,3-b]indole ([(18) F]T807) is a potent and selective agent for imaging paired helical filaments of tau and is among the most promising PET radiopharmaceuticals for this target in early clinical trials. The present study reports a simplified one-step method for the synthesis of [(18) F]T807 that is broadly applicable for routine clinical production using a GE TRACERlab™ FXFN radiosynthesis module. Key facets of our optimized radiosynthesis include development and use of a more soluble protected precursor, tert-butyl 7-(6-nitropyridin-3-yl)-5H-pyrido[4,3-b]indole-5-carboxylate, as well as new HPLC separation conditions that enable a facile one-step synthesis. During the nucleophilic fluorinating reaction with potassium cryptand [(18) F]fluoride (K[(18) F]/K222 ) in DMSO at 130 °C over 10 min the precursor is concurrently deprotected. Formulated [(18) F]T807 was prepared in an uncorrected radiochemical yield of 14 ± 3%, with a specific activity of 216 ± 60 GBq/µmol (5837 ± 1621 mCi/µmol) at the end of synthesis (60 min; n = 3) and validated for human use. This methodology offers the advantage of faster synthesis in fewer steps, with simpler automation that we anticipate will facilitate widespread clinical use of [(18) F]T807.

Collaboration


Dive into the Neil Vasdev's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alan A. Wilson

Centre for Addiction and Mental Health

View shared research outputs
Top Co-Authors

Avatar

Sylvain Houle

Centre for Addiction and Mental Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge