Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Nat Med ; 30(5): 1406-1415, 2024 May.
Article in English | MEDLINE | ID: mdl-38745011

ABSTRACT

GRN mutations cause progranulin haploinsufficiency, which eventually leads to frontotemporal dementia (FTD-GRN). PR006 is an investigational gene therapy delivering the granulin gene (GRN) using an adeno-associated virus serotype 9 (AAV9) vector. In non-clinical studies, PR006 transduced neurons derived from induced pluripotent stem cells of patients with FTD-GRN, resulted in progranulin expression and improvement of lipofuscin, lysosomal and neuroinflammation pathologies in Grn-knockout mice, and was well tolerated except for minimal, asymptomatic dorsal root ganglionopathy in non-human primates. We initiated a first-in-human phase 1/2 open-label trial. Here we report results of a pre-specified interim analysis triggered with the last treated patient of the low-dose cohort (n = 6) reaching the 12-month follow-up timepoint. We also include preliminary data from the mid-dose cohort (n = 7). Primary endpoints were safety, immunogenicity and change in progranulin levels in cerebrospinal fluid (CSF) and blood. Secondary endpoints were Clinical Dementia Rating (CDR) plus National Alzheimer's Disease Coordinating Center (NACC) Frontotemporal Lobar Degeneration (FTLD) rating scale and levels of neurofilament light chain (NfL). One-time administration of PR006 into the cisterna magna was generally safe and well tolerated. All patients developed treatment-emergent anti-AAV9 antibodies in the CSF, but none developed anti-progranulin antibodies. CSF pleocytosis was the most common PR006-related adverse event. Twelve serious adverse events occurred, mostly unrelated to PR006. Deep vein thrombosis developed in three patients. There was one death (unrelated) occurring 18 months after treatment. CSF progranulin increased after PR006 treatment in all patients; blood progranulin increased in most patients but only transiently. NfL levels transiently increased after PR006 treatment, likely reflecting dorsal root ganglia toxicity. Progression rates, based on the CDR scale, were within the broad ranges reported for patients with FTD. These data provide preliminary insights into the safety and bioactivity of PR006. Longer follow-up and additional studies are needed to confirm the safety and potential efficacy of PR006. ClinicalTrials.gov identifier: NCT04408625 .


Subject(s)
Dependovirus , Frontotemporal Dementia , Genetic Therapy , Progranulins , Humans , Frontotemporal Dementia/genetics , Frontotemporal Dementia/therapy , Frontotemporal Dementia/cerebrospinal fluid , Progranulins/genetics , Genetic Therapy/adverse effects , Genetic Therapy/methods , Dependovirus/genetics , Middle Aged , Female , Male , Aged , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/cerebrospinal fluid , Genetic Vectors , Animals , Treatment Outcome , Translational Research, Biomedical , Mice , Neurofilament Proteins/genetics , Neurofilament Proteins/cerebrospinal fluid , Neurofilament Proteins/blood
2.
J Parkinsons Dis ; 11(s2): S183-S188, 2021.
Article in English | MEDLINE | ID: mdl-34151863

ABSTRACT

Human genetic studies as well as studies in animal models indicate that lysosomal dysfunction plays a key role in the pathogenesis of Parkinson's disease. Among the lysosomal genes involved, GBA1 has the largest impact on Parkinson's disease risk. Deficiency in the GBA1 encoded enzyme glucocerebrosidase (GCase) leads to the accumulation of the GCase glycolipid substrates glucosylceramide and glucosylsphingosine and ultimately results in toxicity and inflammation and negatively affect many clinical aspects of Parkinson's disease, including disease risk, the severity of presentation, age of onset, and likelihood of progression to dementia. These findings support the view that re-establishing normal levels of GCase enzyme activity may reduce the progression of Parkinson's disease in patients carrying GBA1 mutations. Studies in mouse models indicate that PR001, a AAV9 vector-based gene therapy designed to deliver a functional GBA1 gene to the brain, suggest that this therapeutic approach may slow or stop disease progression. PR001 is currently being evaluated in clinical trials with Parkinson's disease patients carrying GBA1 mutations.


Subject(s)
Gaucher Disease , Parkinson Disease , Animals , Genetic Therapy , Glucosylceramidase/genetics , Humans , Lysosomes , Mice , Mutation , Parkinson Disease/genetics , Parkinson Disease/therapy , alpha-Synuclein
3.
Pharmacol Res ; 154: 104240, 2020 04.
Article in English | MEDLINE | ID: mdl-31026504

ABSTRACT

The pharmacology of Nerve Growth Factor (NGF) and the discovery and development of tanezumab, a monoclonal anti-NGF antibody for the treatment of pain illustrate the complex and unpredictable nature of modern drug development. Initial efforts attempted to use NGF agonistically for Alzheimer's disease and neuropathies. Most unexpectedly, clinical studies unmasked hyperalgesic effects. These observations together with new data emerging from molecular and animal model studies stimulated the idea of using an NGF antagonist for chronic pain. These events also reflect the advances of neuropharmacology from classical small molecule efforts directed at neurotransmitter receptors to modern biotechnology with significant integration in molecular biology, biochemistry, and protein engineering.


Subject(s)
Analgesics/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Nerve Growth Factor/antagonists & inhibitors , Pain/drug therapy , Animals , Biotechnology , Humans , Protein Engineering
4.
Pain Rep ; 3(3): e653, 2018 May.
Article in English | MEDLINE | ID: mdl-29922745

ABSTRACT

INTRODUCTION: The neurotrophin nerve growth factor has a demonstrated role in pain transduction and pathophysiology. OBJECTIVES: Two randomized, double-blind, placebo-controlled, phase 1 studies were conducted to evaluate safety, tolerability, and analgesic efficacy of single doses of tanezumab, a humanized anti-nerve growth factor monoclonal antibody, in chronic or acute pain. METHODS: In the first study (CL001), patients with moderate to severe pain from osteoarthritis (OA) of the knee received a single intravenous infusion of tanezumab (3-1000 µg/kg) or placebo in a dose-escalation (part 1; N = 42) or parallel-arm (part 2; N = 79) study design. The second study (CL002) was a placebo-controlled dose-escalation (tanezumab 10-1000 µg/kg; N = 50) study in patients undergoing bunionectomy surgery. RESULTS: Adverse event rates were generally similar across treatments. Most adverse events were generally mild to moderate in severity and no patients discontinued as a result of adverse events. Adverse events of abnormal peripheral sensation were more common with higher doses of tanezumab (≥100 µg/kg) than with placebo. These were generally mild to moderate in severity. Tanezumab provided up to 12 weeks of effective analgesia for OA knee pain, with statistically significant improvements at doses ≥100 µg/kg (P < 0.05). By contrast, no trend for analgesic activity was found when tanezumab was administered 8 to 16 hours before bunionectomy. CONCLUSIONS: The demonstration of a favorable safety profile and clinical efficacy in OA pain supports clinical development of tanezumab as a potential treatment for chronic pain conditions.

5.
Sci Rep ; 8(1): 4634, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29545579

ABSTRACT

Soluble amyloid ß oligomers (AßOs) are widely recognized neurotoxins that trigger aberrant signaling in specific subsets of neurons, leading to accumulated neuronal damage and memory disorders in Alzheimer's disease (AD). One of the profound downstream consequences of AßO-triggered events is dysregulation of cytosolic calcium concentration ([Ca2+]i), which has been implicated in synaptic failure, cytoskeletal abnormalities, and eventually neuronal death. We have developed an in vitro/in vivo drug screening assay to evaluate putative AßO-blocking candidates by measuring AßO-induced real-time changes in [Ca2+]i. Our screening assay demonstrated that the anti-AßO monoclonal antibody ACU3B3 exhibits potent blocking capability against a broad size range of AßOs. We showed that picomolar concentrations of AßOs were capable of increasing [Ca2+]i in primary neuronal cultures, an effect prevented by ACU3B3. Topical application of 5 nM AßOs onto exposed cortical surfaces also elicited significant calcium elevations in vivo, which was completely abolished by pre-treatment of the brain with 1 ng/mL (6.67 pM) ACU3B3. Our results provide strong support for the utility of this functional screening assay in identifying and confirming the efficacy of AßO-blocking drug candidates such as the human homolog of ACU3B3, which may emerge as the first experimental AD therapeutic to validate the amyloid oligomer hypothesis.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Brain/drug effects , Calcium/metabolism , Image Processing, Computer-Assisted/methods , Aged, 80 and over , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Case-Control Studies , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL
6.
Curr Alzheimer Res ; 14(4): 393-402, 2017.
Article in English | MEDLINE | ID: mdl-28093972

ABSTRACT

Therapeutic strategies that target pathways of protein misfolding and the toxicity of intermediates along these pathways are mainly at discovery and early development stages, with the exception of monoclonal antibodies that have mainly failed to produce convincing clinical benefits in late stage trials. The clinical failures represent potentially critical lessons for future neurodegenerative disease drug development. More effective drugs may be achieved by pursuing the following two strategies. First, conformational targeting of aggregates of misfolded proteins, rather than less specific binding that includes monomer subunits, which vastly outnumber the toxic targets. Second, since neurodegenerative diseases frequently include more than one potential protein pathology, generic targeting of aggregates by shape might also be a crucial feature of a drug candidate. Incorporating both of these critical features into a viable drug candidate along with high affinity binding has not been achieved with small molecule approaches or with antibody fragments. Monoclonal antibodies developed so far are not broadly acting through conformational recognition. Using GAIM (General Amyloid Interaction Motif) represents a novel approach that incorporates high affinity conformational recognition for multiple protein assemblies, as well as recognition of an array of assemblies along the misfolding pathway between oligomers and fibers. A GAIM-Ig fusion, NPT088, is nearing clinical testing.


Subject(s)
Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/pharmacology , Animals , Humans , Protein Conformation , Protein Folding
7.
Alzheimers Res Ther ; 6(4): 42, 2014.
Article in English | MEDLINE | ID: mdl-25045405

ABSTRACT

Levels of amyloid-beta monomer and deposited amyloid-beta in the Alzheimer's disease brain are orders of magnitude greater than soluble amyloid-beta oligomer levels. Monomeric amyloid-beta has no known direct toxicity. Insoluble fibrillar amyloid-beta has been proposed to be an in vivo mechanism for removal of soluble amyloid-beta and exhibits relatively low toxicity. In contrast, soluble amyloid-beta oligomers are widely reported to be the most toxic amyloid-beta form, both causing acute synaptotoxicity and inducing neurodegenerative processes. None of the amyloid-beta immunotherapies currently in clinical development selectively target soluble amyloid-beta oligomers, and their lack of efficacy is not unexpected considering their selectivity for monomeric or fibrillar amyloid-beta (or both) rather than soluble amyloid-beta oligomers. Because they exhibit acute, memory-compromising synaptic toxicity and induce chronic neurodegenerative toxicity and because they exist at very low in vivo levels in the Alzheimer's disease brain, soluble amyloid-beta oligomers constitute an optimal immunotherapeutic target that should be pursued more aggressively.

8.
Trends Pharmacol Sci ; 34(5): 261-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23582316

ABSTRACT

Soluble Aß oligomers are now widely recognized as key pathogenic structures in Alzheimer's disease. They inhibit synaptic function, leading to early memory deficits and synaptic degeneration, and they trigger the downstream neuronal signaling responsible for phospho-tau Alzheimer's pathology. The marginal effects observed in recent clinical studies of solanezumab, targeting monomeric Aß, and bapineuzumab, targeting amyloid plaques, prompted expert comments that drug discovery efforts in Alzheimer's disease should focus on soluble forms of Aß rather than fibrillar Aß deposits found in amyloid plaques. Accumulating scientific data suggest that soluble Aß oligomers represent the optimal intervention target within the amyloid manifold. Active drug discovery approaches include antibodies that selectively capture soluble Aß oligomers, selective modifiers of oligomer assembly, and receptor antagonists. The onset of symptomatic clinical benefit is expected to be rapid for such agents, because neuronal memory signaling should normalize on blockage of soluble Aß oligomers. This key feature is not shared by amyloid-lowering therapeutics, and it should translate into streamlined clinical development for oligomer-targeting drugs. Oligomer-targeting drugs should also confer long-term disease modification and slowing of disease progression, because they prevent the downstream signaling responsible for phospho-tau mediated cytoskeletal degeneration.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Humans , Molecular Targeted Therapy/methods
9.
Alzheimers Dement ; 8(5): 445-52, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22959699

ABSTRACT

To address the pending public health crisis due to Alzheimer's disease (AD) and related neurodegenerative disorders, the Marian S. Ware Alzheimer Program at the University of Pennsylvania held a meeting entitled "State of the Science Conference on the Advancement of Alzheimer's Diagnosis, Treatment and Care," on June 21-22, 2012. The meeting comprised four workgroups focusing on Biomarkers; Clinical Care and Health Services Research; Drug Development; and Health Economics, Policy, and Ethics. The workgroups shared, discussed, and compiled an integrated set of priorities, recommendations, and action plans, which are presented in this article.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/therapy , Health Policy , Health Services Research , Alzheimer Disease/epidemiology , Cost-Benefit Analysis/statistics & numerical data , Female , Humans , Male , United States
10.
Alzheimer Dis Assoc Disord ; 26(1): 8-16, 2012.
Article in English | MEDLINE | ID: mdl-22354138

ABSTRACT

BACKGROUND: Florbetapir F 18 (F-AV-45) is a positron emission tomography imaging ligand for the detection of amyloid aggregation associated with Alzheimer disease. Earlier data showed that florbetapir F 18 binds with high affinity to ß-amyloid (Aß) plaques in human brain homogenates (Kd=3.7 nM) and has favorable imaging pharmacokinetic properties, including rapid brain penetration and washout. This study used human autopsy brain tissue to evaluate the correlation between in vitro florbetapir F 18 binding and Aß density measured by established neuropathologic methods. METHODS: The localization and density of florbetapir F 18 binding in frozen and formalin-fixed paraffin-embedded sections of postmortem brain tissue from 40 patients with a varying degree of neurodegenerative pathology was assessed by standard florbetapir F 18 autoradiography and correlated with the localization and density of Aß identified by silver staining, thioflavin S staining, and immunohistochemistry. RESULTS: There were strong quantitative correlations between florbetapir F 18 tissue binding and both Aß plaques identified by light microscopy (Silver staining and thioflavin S fluorescence) and by immunohistochemical measurements of Aß using 3 antibodies recognizing different epitopes of the Aß peptide. Florbetapir F 18 did not bind to neurofibrillary tangles. CONCLUSIONS: Florbetapir F 18 selectively binds Aß in human brain tissue. The binding intensity was quantitatively correlated with the density of Aß plaques identified by standard neuropathologic techniques and correlated with the density of Aß measured by immunohistochemistry. As Aß plaques are a defining neuropathologic feature for Alzheimer disease, these results support the use of florbetapir F 18 as an amyloid positron emission tomography ligand to identify the presence of Alzheimer disease pathology in patients with signs and symptoms of progressive late-life cognitive impairment.


Subject(s)
Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/metabolism , Aniline Compounds/analysis , Brain/diagnostic imaging , Ethylene Glycols/analysis , Plaque, Amyloid/diagnostic imaging , Radiopharmaceuticals/analysis , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/metabolism , Amyloid beta-Peptides/analysis , Aniline Compounds/chemistry , Autopsy , Autoradiography , Brain/metabolism , Brain/pathology , Ethylene Glycols/chemistry , Humans , Neurofibrillary Tangles/diagnostic imaging , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , Positron-Emission Tomography
11.
J Psychopharmacol ; 26(7): 973-81, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22236504

ABSTRACT

Although cocaine dependence affects an estimated 1.6 million people in the USA, there are currently no medications approved for the treatment of this disorder. Experiments performed in animal models have demonstrated that inhibitors of the stress response effectively reduce intravenous cocaine self-administration. This exploratory, double-blind, placebo-controlled study was designed to assess the safety and efficacy of combinations of the cortisol synthesis inhibitor metyrapone, and the benzodiazepine oxazepam, in 45 cocaine-dependent individuals. The subjects were randomized to a total daily dose of 500 mg metyrapone/20 mg oxazepam (low dose), a total daily dose of 1500 mg metyrapone/20 mg oxazepam (high dose), or placebo for 6 weeks of treatment. The outcome measures were a reduction in cocaine craving and associated cocaine use as determined by quantitative measurements of the cocaine metabolite benzoylecgonine (BE) in urine at all visits. Of the randomized subjects, 49% completed the study. The combination of metyrapone and oxazepam was well tolerated and tended to reduce cocaine craving and cocaine use, with significant reductions at several time points when controlling for baseline scores. These data suggest that further assessments of the ability of the metyrapone and oxazepam combination to support cocaine abstinence in cocaine-dependent subjects are warranted.


Subject(s)
Cocaine-Related Disorders/drug therapy , Enzyme Inhibitors/therapeutic use , GABA Agonists/therapeutic use , Metyrapone/therapeutic use , Oxazepam/therapeutic use , Steroid 11-beta-Hydroxylase/antagonists & inhibitors , Substance Withdrawal Syndrome/prevention & control , Adult , Cocaine/analogs & derivatives , Cocaine/urine , Cocaine-Related Disorders/prevention & control , Cocaine-Related Disorders/urine , Diagnostic and Statistical Manual of Mental Disorders , Dose-Response Relationship, Drug , Double-Blind Method , Drug Therapy, Combination/adverse effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Female , GABA Agonists/administration & dosage , GABA Agonists/adverse effects , Humans , Louisiana , Male , Metyrapone/administration & dosage , Metyrapone/adverse effects , Middle Aged , Oxazepam/administration & dosage , Oxazepam/adverse effects , Patient Compliance , Patient Dropouts , Pilot Projects , Secondary Prevention , Steroid 11-beta-Hydroxylase/administration & dosage , Steroid 11-beta-Hydroxylase/adverse effects
12.
Proc Natl Acad Sci U S A ; 108(51): 20719-24, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22143775

ABSTRACT

We combined multimodal imaging (bioluminescence, X-ray computed tomography, and PET), tomographic reconstruction of bioluminescent sources, and two unique, complementary models to evaluate three previously synthesized PET radiotracers thought to target pancreatic beta cells. The three radiotracers {[(18)F]fluoropropyl-(+)-dihydrotetrabenazine ([(18)F]FP-DTBZ), [(18)F](+)-2-oxiranyl-3-isobutyl-9-(3-fluoropropoxy)-10-methoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinoline ((18)F-AV-266), and (2S,3R,11bR)-9-(3-fluoropropoxy)-2-(hydroxymethyl)-3-isobutyl-10-methoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinolin-2-ol ((18)F-AV-300)} bind vesicular monoamine transporter 2. Tomographic reconstruction of the bioluminescent signal in mice expressing luciferase only in pancreatic beta cells was used to delineate the pancreas and was coregistered with PET and X-ray computed tomography images. This strategy enabled unambiguous identification of the pancreas on PET images, permitting accurate quantification of the pancreatic PET signal. We show here that, after conditional, specific, and rapid mouse beta-cell ablation, beta-cell loss was detected by bioluminescence imaging but not by PET imaging, given that the pancreatic signal provided by three PET radiotracers was not altered. To determine whether these ligands bound human beta cells in vivo, we imaged mice transplanted with luciferase-expressing human islets. The human islets were imaged by bioluminescence but not with the PET ligands, indicating that these vesicular monoamine transporter 2-directed ligands did not specifically bind beta cells. These data demonstrate the utility of coregistered multimodal imaging as a platform for evaluation and validation of candidate ligands for imaging islets.


Subject(s)
Positron-Emission Tomography/methods , Tomography, X-Ray Computed/methods , Animals , Diabetes Mellitus/metabolism , Diagnostic Imaging/methods , Female , Humans , Insulin/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/metabolism , Ligands , Luminescence , Male , Mice , Mice, Inbred NOD , Rats , Tissue Distribution
13.
J Med Chem ; 54(23): 8085-98, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-22011144

ABSTRACT

ß-Amyloid plaques (Aß plaques) in the brain are associated with cerebral amyloid angiopathy (CAA). Imaging agents that could target the Aß plaques in the living human brain would be potentially valuable as biomarkers in patients with CAA. A new series of (18)F styrylpyridine derivatives with high molecular weights for selectively targeting Aß plaques in the blood vessels of the brain but excluded from the brain parenchyma is reported. The styrylpyridine derivatives, 8a-c, display high binding affinities and specificity to Aß plaques (K(i) = 2.87, 3.24, and 7.71 nM, respectively). In vitro autoradiography of [(18)F]8a shows labeling of ß-amyloid plaques associated with blood vessel walls in human brain sections of subjects with CAA and also in the tissue of AD brain sections. The results suggest that [(18)F]8a may be a useful PET imaging agent for selectively detecting Aß plaques associated with cerebral vessels in the living human brain.


Subject(s)
Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/diagnostic imaging , Plaque, Amyloid/diagnostic imaging , Pyridines/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Styrenes/chemical synthesis , Animals , Autoradiography , Brain/blood supply , Cerebral Amyloid Angiopathy/pathology , Fluorine Radioisotopes , Humans , Mice , Mice, Inbred ICR , Plaque, Amyloid/pathology , Positron-Emission Tomography , Pyridines/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Stereoisomerism , Structure-Activity Relationship , Styrenes/pharmacokinetics , Tissue Distribution
14.
Semin Nucl Med ; 41(4): 300-4, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21624563

ABSTRACT

Florbetapir F-18 is a molecular imaging agent combining high affinity for ß-amyloid, pharmacokinetic properties that allow positron emission tomography (PET) imaging within a convenient time after dose administration, and the wide availability of the radionuclide fluorine-18. Florbetapir F-18 is prepared by nucleophilic radiofluorination in approximately 60 minutes with a decay-corrected yield of 20%-40% and with a specific activity typically exceeding 100 Ci/mmol. The florbetapir F-18 dissociation constant (K(d)) for binding to ß-amyloid in brain tissue from Alzheimer's disease (AD) patients was 3.7 ± 0.3 nmol/L, and the maximum binding capacity (B(max)) was 8800 ± 1600 fmol/mg protein. Autoradiography studies have shown that florbetapir F-18 selectively binds to ß-amyloid aggregates in AD patient brain tissue, and the binding intensity is correlated with the density of ß-amyloid quantified by standard neuropathologic techniques. Studies in animals revealed no safety concerns and rapid and transient normal brain uptake (6.8% injected dose/g at 2 minutes and 1.9% injected dose/g at 60 minutes in the mouse). Florbetapir F-18 has been well-tolerated in studies of more than 2000 human subjects. Biodistribution studies in humans revealed predominantly hepatobiliary excretion. The whole body effective dose was 7 mSv from a dose of 370 MBq. The pharmacokinetic of florbetapir F-18 make it possible to obtain a PET image with a brief (10 minutes) acquisition time within a convenient time window of 30-90 minutes after dose administration. Clinical studies have demonstrated a clear correlation between in vivo PET imaging with florbetapir F-18 and postmortem histopathologic quantitation of ß-amyloid in the brain.


Subject(s)
Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/metabolism , Aniline Compounds/pharmacokinetics , Cerebral Cortex/diagnostic imaging , Ethylene Glycols/pharmacokinetics , Positron-Emission Tomography/methods , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Aniline Compounds/chemistry , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dogs , Ethylene Glycols/chemistry , Humans , Mice , Molecular Imaging , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Tissue Distribution
15.
JAMA ; 305(3): 275-83, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21245183

ABSTRACT

CONTEXT: The ability to identify and quantify brain ß-amyloid could increase the accuracy of a clinical diagnosis of Alzheimer disease. OBJECTIVE: To determine if florbetapir F 18 positron emission tomographic (PET) imaging performed during life accurately predicts the presence of ß-amyloid in the brain at autopsy. DESIGN, SETTING, AND PARTICIPANTS: Prospective clinical evaluation conducted February 2009 through March 2010 of florbetapir-PET imaging performed on 35 patients from hospice, long-term care, and community health care facilities near the end of their lives (6 patients to establish the protocol and 29 to validate) compared with immunohistochemistry and silver stain measures of brain ß-amyloid after their death used as the reference standard. PET images were also obtained in 74 young individuals (18-50 years) presumed free of brain amyloid to better understand the frequency of a false-positive interpretation of a florbetapir-PET image. MAIN OUTCOME MEASURES: Correlation of florbetapir-PET image interpretation (based on the median of 3 nuclear medicine physicians' ratings) and semiautomated quantification of cortical retention with postmortem ß-amyloid burden, neuritic amyloid plaque density, and neuropathological diagnosis of Alzheimer disease in the first 35 participants autopsied (out of 152 individuals enrolled in the PET pathological correlation study). RESULTS: Florbetapir-PET imaging was performed a mean of 99 days (range, 1-377 days) before death for the 29 individuals in the primary analysis cohort. Fifteen of the 29 individuals (51.7%) met pathological criteria for Alzheimer disease. Both visual interpretation of the florbetapir-PET images and mean quantitative estimates of cortical uptake were correlated with presence and quantity of ß-amyloid pathology at autopsy as measured by immunohistochemistry (Bonferroni ρ, 0.78 [95% confidence interval, 0.58-0.89]; P <.001]) and silver stain neuritic plaque score (Bonferroni ρ, 0.71 [95% confidence interval, 0.47-0.86]; P <.001). Florbetapir-PET images and postmortem results rated as positive or negative for ß-amyloid agreed in 96% of the 29 individuals in the primary analysis cohort. The florbetapir-PET image was rated as amyloid negative in the 74 younger individuals in the nonautopsy cohort. CONCLUSIONS: Florbetapir-PET imaging was correlated with the presence and density of ß-amyloid. These data provide evidence that a molecular imaging procedure can identify ß-amyloid pathology in the brains of individuals during life. Additional studies are required to understand the appropriate use of florbetapir-PET imaging in the clinical diagnosis of Alzheimer disease and for the prediction of progression to dementia.


Subject(s)
Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/analysis , Aniline Compounds , Brain Chemistry , Ethylene Glycols , Positron-Emission Tomography/methods , Radiopharmaceuticals , Aged , Autopsy , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , False Positive Reactions , Female , Humans , Immunohistochemistry , Male , Middle Aged , Prospective Studies
16.
PET Clin ; 5(1): 75-82, 2010 Jan.
Article in English | MEDLINE | ID: mdl-27157484

ABSTRACT

The early detection and monitoring of neurodegenerative diseases, including Parkinson disease, Alzheimer disease, dementia with Lewy bodies and other dementias, and movement disorders, represent a significant unmet medical need. Tools for accurate and early differential diagnosis are necessary to determine the appropriate treatment for patients and to minimize inappropriate use of potentially harmful treatments. Such diagnostic imaging tools are expected to permit monitoring of disease progression and will thus accelerate testing and development of disease-modifying drugs. The new imaging tests may be useful as prognostic tools by identifying humans with neurodegenerative diseases before the clinical manifestations become evident.

17.
J Nucl Med ; 50(11): 1887-94, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19837759

ABSTRACT

UNLABELLED: beta-amyloid plaques (Abeta plaques) in the brain, containing predominantly fibrillary Abeta peptide aggregates, represent a defining pathologic feature of Alzheimer disease (AD). Imaging agents targeting the Abeta plaques in the living human brain are potentially valuable as biomarkers of pathogenesis processes in AD. (E)-4-(2-(6-(2-(2-(2-(18)F-fluoroethoxy)ethoxy)ethoxy)pyridin-3-yl)vinyl)-N-methyl benzenamine ((18)F-AV-45) is such as an agent currently in phase III clinical studies for PET of Abeta plaques in the brain. METHODS: In vitro binding of (18)F-AV-45 to Abeta plaques in the postmortem AD brain tissue was evaluated by in vitro binding assay and autoradiography. In vivo biodistribution of (18)F-AV-45 in mice and ex vivo autoradiography of AD transgenic mice (APPswe/PSEN1) with Abeta aggregates in the brain were performed. Small-animal PET of a monkey brain after an intravenous injection of (18)F-AV-45 was evaluated. RESULTS: (18)F-AV-45 displayed a high binding affinity and specificity to Abeta plaques (K(d), 3.72 +/- 0.30 nM). In vitro autoradiography of postmortem human brain sections showed substantial plaque labeling in AD brains and not in the control brains. Initial high brain uptake and rapid washout from the brain of healthy mice and monkey were observed. Metabolites produced in the blood of healthy mice after an intravenous injection were identified. (18)F-AV-45 displayed excellent binding affinity to Abeta plaques in the AD brain by ex vivo autoradiography in transgenic AD model mice. The results lend support that (18)F-AV-45 may be a useful PET agent for detecting Abeta plaques in the living human brain.


Subject(s)
Amyloid beta-Peptides/metabolism , Aniline Compounds/metabolism , Benzene/metabolism , Brain/diagnostic imaging , Brain/pathology , Ethylene Glycols/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Aniline Compounds/pharmacokinetics , Animals , Autoradiography , Benzene/pharmacokinetics , Brain/metabolism , Ethylene Glycols/pharmacokinetics , Female , Haplorhini , Humans , Injections, Intravenous , Male , Mice , Mice, Transgenic , Positron-Emission Tomography , Tissue Distribution
18.
BMC Neurosci ; 9 Suppl 3: S7, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-19091004

ABSTRACT

A drug candidate suitable for clinical testing is expected to bind selectively to the receptor site on the target, to elicit the desired functional response of the target molecule, and to have adequate bioavailability and biodistribution to elicit the desired responses in animals and humans; it must also pass formal toxicity evaluation in animals. The path from lead to clinical drug candidate represents the most idiosyncratic segment of drug discovery and development. Each program is unique and setbacks are common, making it difficult to predict accurately the duration or costs of this segment. Because of incidents of unpredicted human toxicity seen in recent years, the regulatory agencies and public demands for safety of new drug candidates have become very strict, and safety issues are dominant when identifying a clinical drug candidate.


Subject(s)
Drug Design , Drug Discovery/methods , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Animals , Biological Availability , Drug Discovery/economics , Drug Evaluation, Preclinical/economics , Drug Evaluation, Preclinical/methods , Drug Industry/economics , Drug Industry/methods , Drug-Related Side Effects and Adverse Reactions , Humans , Pharmaceutical Preparations/administration & dosage , Tissue Distribution
19.
Aging Cell ; 5(1): 3-8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16441837

ABSTRACT

Many current drugs increase the average lifespan by preventing fatal diseases or by slowing down the progressive degenerative diseases that increase mortality. The existing strategies and guidelines for the development and regulatory approval of new drugs are designed for such compounds. Rapid advances in understanding molecular mechanisms of aging make it possible to envisage future drugs that extend the lifespan by regulating aging mechanism outside of disease pathways. Strategies for development and regulatory approval of such drugs remain to be defined. Since the drug candidates will be given to healthy, elderly subjects, safety requirements will be extremely high. Clinical studies of many years' duration will be necessary to prove changes in longevity. These time intervals may exceed those of patent protection and thus minimize commercial incentives. Despite these challenges, two broadly defined pathways are feasible. First, it may be possible to obtain public funding for studies with voluntary participation of humans consuming existing drugs or natural compounds in the 'expected to be safe' category. Second, the development of novel drugs may proceed on the basis of well-defined biomarkers of aging that can serve as surrogate end points in clinical studies. The emerging approaches will prompt the regulatory agencies into taking the first steps towards regulatory guidance.


Subject(s)
Aging/drug effects , Drug Approval , Longevity/drug effects , Pharmacology, Clinical/legislation & jurisprudence , Aging/physiology , Biomarkers/metabolism , Geriatrics/trends , Humans , Longevity/physiology , Pharmacology, Clinical/trends
20.
Trends Pharmacol Sci ; 27(2): 85-91, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16376998

ABSTRACT

Nerve growth factor (NGF) was identified originally as a survival factor for sensory and sympathetic neurons in the developing nervous system. In adults, NGF is not required for survival but it has a crucial role in the generation of pain and hyperalgesia in several acute and chronic pain states. The expression of NGF is high in injured and inflamed tissues, and activation of the NGF receptor tyrosine kinase trkA on nociceptive neurons triggers and potentiates pain signalling by multiple mechanisms. Inhibition of NGF function and signalling blocks pain sensation as effectively as cyclooxygenase inhibitors and opiates in rodent models of pain. Several pharmaceutical companies have active drug-discovery and development programs that are based on a variety of approaches to antagonise NGF, including NGF 'capture', blocking the binding of NGF to trkA and inhibiting trkA signalling. NGF antagonism is expected to be a highly effective therapeutic approach in many pain states, and to be free of the adverse effects of traditional analgesic drugs.


Subject(s)
Analgesics/pharmacology , Nerve Growth Factors/antagonists & inhibitors , Pain/physiopathology , Animals , Humans , Hyperalgesia/physiopathology , Pain/drug therapy , Receptor, trkA/antagonists & inhibitors , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...