Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
BMC Neurol ; 24(1): 116, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38594621

RESUMO

BACKGROUND: The authors sought to examine the impact of the K-variant of butyrylcholinesterase (BCHE-K) carrier status on age-at-diagnosis of Alzheimer disease (AD) in APOE4 carriers. METHODS: Patients aged 50-74 years with cerebrospinal fluid (CSF) biomarker-confirmed AD, were recruited to clinical trial (NCT03186989 since June 14, 2017). Baseline demographics, disease characteristics, and biomarkers were evaluated in 45 patients according to BCHE-K and APOE4 allelic status in this post-hoc study. RESULTS: In APOE4 carriers (N = 33), the mean age-at-diagnosis of AD in BCHE-K carriers (n = 11) was 6.4 years earlier than in BCHE-K noncarriers (n = 22, P < .001, ANOVA). In APOE4 noncarriers (N = 12) there was no observed influence of BCHE-K. APOE4 carriers with BCHE-K also exhibited slightly higher amyloid and tau accumulations compared to BCHE-K noncarriers. A predominantly amyloid, limited tau, and limbic-amnestic phenotype was exemplified by APOE4 homozygotes with BCHE-K. In the overall population, multiple regression analyses demonstrated an association of amyloid accumulation with APOE4 carrier status (P < .029), larger total brain ventricle volume (P < .021), less synaptic injury (Ng, P < .001), and less tau pathophysiology (p-tau181, P < .005). In contrast, tau pathophysiology was associated with more neuroaxonal damage (NfL, P = .002), more synaptic injury (Ng, P < .001), and higher levels of glial activation (YKL-40, P = .01). CONCLUSION: These findings have implications for the genetic architecture of prognosis in early AD, not the genetics of susceptibility to AD. In patients with early AD aged less than 75 years, the mean age-at-diagnosis of AD in APOE4 carriers was reduced by over 6 years in BCHE-K carriers versus noncarriers. The functional status of glia may explain many of the effects of APOE4 and BCHE-K on the early AD phenotype. TRIAL REGISTRATION: NCT03186989 since June 14, 2017.


Assuntos
Doença de Alzheimer , Criança , Humanos , Doença de Alzheimer/líquido cefalorraquidiano , Peptídeos beta-Amiloides , Apolipoproteína E4/genética , Apolipoproteínas E/genética , Butirilcolinesterase/genética , Fenótipo
2.
JAMA Ophthalmol ; 142(4): 301-308, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38421662

RESUMO

Importance: Reliable biomarkers with diagnostic and prognostic values are needed for upcoming gene therapy trials for spinocerebellar ataxias. Objective: To identify ophthalmological biomarkers in a sample of spinocerebellar ataxia type 7 (SCA7) carriers. Design, Setting, and Participants: This article presents baseline data from a cross-sectional natural history study conducted in Paris, France, reference centers for rare diseases from May 2020 to April 2021. Data were analyzed from September to December 2022. Fifteen adult ATXN7 pathogenic expansion carriers (9 with preataxia and 6 with ataxia) were included, all with a Scale for the Assessment and Rating of Ataxia (SARA) score of 15 of 40 or lower. Patients were recruited at the Paris Brain Institute, and all contacted patients accepted to participate in the study. Main Outcomes and Measures: Three visits (baseline, 6 months, and 12 months) were planned, including neurological examination (SARA and Composite Cerebellar Functional Severity Score), ophthalmological examination (best-corrected visual acuity, microperimetry, full-field electroretinogram, optical coherence tomography, and fundus autofluorescence imaging), and neurofilament light chain (NfL) measurements. Here we report the baseline ophthalmic data from the cohort and determine whether there is a correlation between disease scores and ophthalmic results. Results: Among the 15 included SCA7 carriers (median [range] age, 38 [18-60] years; 8 women and 7 men), 12 displayed cone or cone-rod dystrophy, with the number of CAG repeats correlating with disease severity (ρ, 0.73, 95% CI, 0.34 to 0.90; P < .001). Two patients with cone-rod dystrophy exhibited higher repeat numbers and greater ataxia scores (median [range] SARA score, 9 [7-15]) compared to those with only cone dystrophy (median [range] SARA score, 2 [0-5]). A correlation emerged for outer nuclear layer thickness with SARA score (ρ, -0.88; 95% CI, -0.96 to -0.59; P < .001) and NfL levels (ρ, -0.87; 95% CI, -0.86 to 0.96; P < .001). Moreover, ataxia severity was correlated with visual acuity (ρ: 0.89; 95% CI, 0.68 to 0.96; P < .001) and retinal sensitivity (ρ, -0.88; 95% CI, -0.96 to 0.59; P < .001). Conclusions and Relevance: In this cross-sectional study, retinal abnormalities were found at preataxic stages of the disease. Most of the carriers presented with cone dystrophy and preserved rod function. The outer nuclear layer thickness correlated with SARA score and plasma NfL levels suggesting nuclear layer thickness to be a biomarker of disease severity. These findings contribute to understanding the dynamics of SCA7-related retinal dystrophy and may help lay the groundwork for future therapeutic intervention monitoring and clinical trials. Trial Registration: ClinicalTrials.gov Identifier: NCT04288128.


Assuntos
Distrofia de Cones , Distrofias de Cones e Bastonetes , Ataxias Espinocerebelares , Adulto , Masculino , Humanos , Feminino , Estudos Transversais , Ataxias Espinocerebelares/diagnóstico , Cerebelo , Biomarcadores
4.
JAMA Neurol ; 80(12): 1344-1352, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37902726

RESUMO

Importance: Accumulation of hyperphosphorylated, tangled microtubule-associated protein tau (MAPT) is a pathological hallmark of Alzheimer disease (AD) associated with disease progression and cognitive decline. Objective: To evaluate the effect of tau synthesis reduction on tau biomarkers in patients with mild AD. Design, Setting, and Participants: This randomized clinical trial was a double-blind, placebo-controlled 36-week multiple-ascending dose (MAD) phase 1b trial (October 2017 to September 2020), followed by a 64- or 71-week open-label long-term extension (LTE) (October 2019 to May 2022). After being assessed for eligibility at 12 sites in Canada and Europe, participants with mild AD and confirmed amyloid pathology were randomized 3:1 (BIIB080:placebo) in 4 dose cohorts. Intervention: Intrathecal administration of BIIB080, a MAPT-targeting antisense oligonucleotide, or placebo. Active dose arms included 10 mg every 4 weeks, 30 mg every 4 weeks, 60 mg every 4 weeks, and 115 mg every 12 weeks during the MAD period and 60 mg every 12 weeks or 115 mg every 12 weeks during the LTE. Main Outcome and Measures: The original primary end point was safety. Additionally, BIIB080, total tau (t-tau), and phosphorylated tau 181 (p-tau181) cerebrospinal fluid (CSF) concentrations were evaluated. Tau positron emission tomography (PET) was collected in a substudy, and standard uptake value ratios (SUVRs) were calculated in a priori-defined composite regions of interest. Results: Of 102 participants assessed for eligibility, 46 participants with mild AD were enrolled; 23 (50%) were female, and mean (SD) age was 65.8 (5.70) years. BIIB080 was generally well tolerated and was associated with a dose-dependent reduction in CSF t-tau and p-tau181 in the MAD period (56% reduction; 95% CI, 50% to 62%; and 51% reduction; 95% CI, 38% to 63%, of CSF t-tau in the 2 higher-dose cohorts) that continued and/or was maintained through quarterly dosing in the LTE. Tau PET demonstrated reduced accumulation vs placebo at week 25 (n = 13). At week 100, tau PET showed a reduction from baseline across all regions assessed (n = 12), with the largest reductions from baseline observed in the temporal composite (-0.71 SUVR; 95% CI, -1.40 to -0.02). A moderate correlation was observed between model-predicted cumulative CSF drug exposure and tau PET change. Conclusions and Relevance: In this randomized clinical trial, BIIB080 reduced tau biomarkers, including CSF t-tau, CSF p-tau181, and tau PET, which is associated with cognitive decline, in participants with mild AD. Effects of BIIB080 on biomarkers and clinical outcomes are being further evaluated in a phase 2 trial. Trial Registration: ClinicalTrials.gov Identifier: NCT03186989.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Humanos , Feminino , Idoso , Masculino , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/líquido cefalorraquidiano , Proteínas tau/líquido cefalorraquidiano , Biomarcadores/líquido cefalorraquidiano , Tomografia por Emissão de Pósitrons , Método Duplo-Cego , Peptídeos beta-Amiloides/líquido cefalorraquidiano
5.
Nat Med ; 29(6): 1437-1447, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37095250

RESUMO

Tau plays a key role in Alzheimer's disease (AD) pathophysiology, and accumulating evidence suggests that lowering tau may reduce this pathology. We sought to inhibit MAPT expression with a tau-targeting antisense oligonucleotide (MAPTRx) and reduce tau levels in patients with mild AD. A randomized, double-blind, placebo-controlled, multiple-ascending dose phase 1b trial evaluated the safety, pharmacokinetics and target engagement of MAPTRx. Four ascending dose cohorts were enrolled sequentially and randomized 3:1 to intrathecal bolus administrations of MAPTRx or placebo every 4 or 12 weeks during the 13-week treatment period, followed by a 23 week post-treatment period. The primary endpoint was safety. The secondary endpoint was MAPTRx pharmacokinetics in cerebrospinal fluid (CSF). The prespecified key exploratory outcome was CSF total-tau protein concentration. Forty-six patients enrolled in the trial, of whom 34 were randomized to MAPTRx and 12 to placebo. Adverse events were reported in 94% of MAPTRx-treated patients and 75% of placebo-treated patients; all were mild or moderate. No serious adverse events were reported in MAPTRx-treated patients. Dose-dependent reduction in the CSF total-tau concentration was observed with greater than 50% mean reduction from baseline at 24 weeks post-last dose in the 60 mg (four doses) and 115 mg (two doses) MAPTRx groups. Clinicaltrials.gov registration number: NCT03186989 .


Assuntos
Doença de Alzheimer , Proteínas tau , Humanos , Proteínas tau/genética , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/líquido cefalorraquidiano , Oligonucleotídeos Antissenso/uso terapêutico , Resultado do Tratamento , Método Duplo-Cego
6.
N Engl J Med ; 383(2): 109-119, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32640130

RESUMO

BACKGROUND: Tofersen is an antisense oligonucleotide that mediates the degradation of superoxide dismutase 1 (SOD1) messenger RNA to reduce SOD1 protein synthesis. Intrathecal administration of tofersen is being studied for the treatment of amyotrophic lateral sclerosis (ALS) due to SOD1 mutations. METHODS: We conducted a phase 1-2 ascending-dose trial evaluating tofersen in adults with ALS due to SOD1 mutations. In each dose cohort (20, 40, 60, or 100 mg), participants were randomly assigned in a 3:1 ratio to receive five doses of tofersen or placebo, administered intrathecally for 12 weeks. The primary outcomes were safety and pharmacokinetics. The secondary outcome was the change from baseline in the cerebrospinal fluid (CSF) SOD1 concentration at day 85. Clinical function and vital capacity were measured. RESULTS: A total of 50 participants underwent randomization and were included in the analyses; 48 participants received all five planned doses. Lumbar puncture-related adverse events were observed in most participants. Elevations in CSF white-cell count and protein were reported as adverse events in 4 and 5 participants, respectively, who received tofersen. Among participants who received tofersen, one died from pulmonary embolus on day 137, and one from respiratory failure on day 152; one participant in the placebo group died from respiratory failure on day 52. The difference at day 85 in the change from baseline in the CSF SOD1 concentration between the tofersen groups and the placebo group was 2 percentage points (95% confidence interval [CI], -18 to 27) for the 20-mg dose, -25 percentage points (95% CI, -40 to -5) for the 40-mg dose, -19 percentage points (95% CI, -35 to 2) for the 60-mg dose, and -33 percentage points (95% CI, -47 to -16) for the 100-mg dose. CONCLUSIONS: In adults with ALS due to SOD1 mutations, CSF SOD1 concentrations decreased at the highest concentration of tofersen administered intrathecally over a period of 12 weeks. CSF pleocytosis occurred in some participants receiving tofersen. Lumbar puncture-related adverse events were observed in most participants. (Funded by Biogen; ClinicalTrials.gov number, NCT02623699; EudraCT number, 2015-004098-33.).


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos/administração & dosagem , Superóxido Dismutase-1/líquido cefalorraquidiano , Adulto , Esclerose Lateral Amiotrófica/líquido cefalorraquidiano , Esclerose Lateral Amiotrófica/genética , Progressão da Doença , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Cefaleia/induzido quimicamente , Humanos , Injeções Espinhais/efeitos adversos , Filamentos Intermediários , Leucocitose/induzido quimicamente , Masculino , Pessoa de Meia-Idade , Mutação , Oligonucleotídeos/efeitos adversos , Oligonucleotídeos/farmacocinética , Oligonucleotídeos Antissenso/efeitos adversos , Oligonucleotídeos Antissenso/farmacocinética , Superóxido Dismutase-1/genética , Capacidade Vital
7.
N Engl J Med ; 380(24): 2307-2316, 2019 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-31059641

RESUMO

BACKGROUND: Huntington's disease is an autosomal-dominant neurodegenerative disease caused by CAG trinucleotide repeat expansion in HTT, resulting in a mutant huntingtin protein. IONIS-HTTRx (hereafter, HTTRx) is an antisense oligonucleotide designed to inhibit HTT messenger RNA and thereby reduce concentrations of mutant huntingtin. METHODS: We conducted a randomized, double-blind, multiple-ascending-dose, phase 1-2a trial involving adults with early Huntington's disease. Patients were randomly assigned in a 3:1 ratio to receive HTTRx or placebo as a bolus intrathecal administration every 4 weeks for four doses. Dose selection was guided by a preclinical model in mice and nonhuman primates that related dose level to reduction in the concentration of huntingtin. The primary end point was safety. The secondary end point was HTTRx pharmacokinetics in cerebrospinal fluid (CSF). Prespecified exploratory end points included the concentration of mutant huntingtin in CSF. RESULTS: Of the 46 patients who were enrolled in the trial, 34 were randomly assigned to receive HTTRx (at ascending dose levels of 10 to 120 mg) and 12 were randomly assigned to receive placebo. Each patient received all four doses and completed the trial. Adverse events, all of grade 1 or 2, were reported in 98% of the patients. No serious adverse events were seen in HTTRx-treated patients. There were no clinically relevant adverse changes in laboratory variables. Predose (trough) concentrations of HTTRx in CSF showed dose dependence up to doses of 60 mg. HTTRx treatment resulted in a dose-dependent reduction in the concentration of mutant huntingtin in CSF (mean percentage change from baseline, 10% in the placebo group and -20%, -25%, -28%, -42%, and -38% in the HTTRx 10-mg, 30-mg, 60-mg, 90-mg, and 120-mg dose groups, respectively). CONCLUSIONS: Intrathecal administration of HTTRx to patients with early Huntington's disease was not accompanied by serious adverse events. We observed dose-dependent reductions in concentrations of mutant huntingtin. (Funded by Ionis Pharmaceuticals and F. Hoffmann-La Roche; ClinicalTrials.gov number, NCT02519036.).


Assuntos
Proteína Huntingtina/antagonistas & inibidores , Doença de Huntington/tratamento farmacológico , Nucleotídeos/farmacologia , Oligonucleotídeos/uso terapêutico , Adulto , Relação Dose-Resposta a Droga , Feminino , Humanos , Proteína Huntingtina/líquido cefalorraquidiano , Proteína Huntingtina/genética , Injeções Espinhais , Masculino , Pessoa de Meia-Idade , Mutação , Nucleotídeos/síntese química , Oligonucleotídeos/líquido cefalorraquidiano
8.
Methods Mol Biol ; 1780: 497-523, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29856033

RESUMO

Advances in molecular biology and genetics have been used to elucidate the fundamental genetic mechanisms underlying central nervous system (CNS) diseases, yet disease-modifying therapies are currently unavailable for most CNS conditions. Antisense oligonucleotides (ASOs) are synthetic single stranded chains of nucleic acids that bind to a specific sequence on ribonucleic acid (RNA) and regulate posttranscriptional gene expression. Decreased gene expression with ASOs might be able to reduce production of the disease-causing protein underlying dominantly inherited neurodegenerative disorders. Huntington's disease (HD), which is caused by a CAG repeat expansion in exon 1 of the huntingtin (HTT) gene and leads to the pathogenic expansion of a polyglutamine (PolyQ ) tract in the N terminus of the huntingtin protein (Htt), is a prime candidate for ASO therapy.State-of-the art translational science techniques can be applied to the development of an ASO targeting HTT RNA, allowing for a data-driven, stepwise progression through the drug development process. A deep and wide-ranging understanding of the basic, preclinical, clinical, and epidemiologic components of drug development will improve the likelihood of success. This includes characterizing the natural history of the disease, including evolution of biomarkers indexing the underlying pathology; using predictive preclinical models to assess the putative gain-of-function of mutant Htt protein and any loss-of-function of the wild-type protein; characterizing toxicokinetic and pharmacodynamic effects of ASOs in predictive animal models; developing sensitive and reliable biomarkers to monitor target engagement and effects on pathology that translate from animal models to patients with HD; establishing a drug delivery method that ensures reliable distribution to relevant CNS tissue; and designing clinical trials that move expeditiously from proof of concept to proof of efficacy. This review focuses on the translational science techniques that allow for efficient and informed development of an ASO for the treatment of HD.


Assuntos
Proteína Huntingtina/genética , Doença de Huntington/terapia , Oligonucleotídeos Antissenso/uso terapêutico , Reparo Gênico Alvo-Dirigido/métodos , Pesquisa Translacional Biomédica/métodos , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Desenvolvimento de Medicamentos , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Doença de Huntington/patologia , Macaca fascicularis , Camundongos , Mutação , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacologia , Precursores de RNA/genética , Ratos , Resultado do Tratamento
9.
J Clin Psychopharmacol ; 35(4): 454-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25961781

RESUMO

Two phase 2B, randomized, double-blind studies assessed the efficacy and safety of fixed or flexible dose of triple monoamine uptake inhibitor BMS-820836 in patients with treatment-resistant depression to demonstrate whether switching to BMS-820836 was superior to the continuation of standard antidepressant treatment. Patients with a history of inadequate response to 1 to 3 adequate trials of antidepressant therapies were prospectively treated with duloxetine 60 mg/d for 8 weeks (CN162-006) or duloxetine 60 mg/d or escitalopram 20 mg/d for 7 weeks (CN162-007). Inadequate responders were randomized to continue their prospective phase treatment or switch to flexible-dose (0.5-2 mg/d; CN162-006) or fixed-dose (0.25, 0.5, 1, or 2 mg/d; CN162-007) BMS-820836 for 6 weeks. The primary end point in both studies was mean change in Montgomery-Åsberg Depression Rating Scale (MADRS) total score from randomization to study end point. BMS-820836 flexible (0.5-2 mg/d) or fixed dose of 1 mg/d or greater showed efficacy similar to the continuation of antidepressant treatment, with no statistically significant or clinically meaningful differences. In the CN162-006 study, the adjusted mean (SE) change in MADRS total score was -8.7 (0.661) and -8.1 (0.656) for BMS-820836 and duloxetine, respectively (P = 0.526). In the CN162-007 study, the adjusted mean (SE) change in MADRS total score was -7.3 (0.830) and -6.6 (0.842) for BMS-820836 of 1 and 2 mg, respectively, and -6.9 (0.602) for the continuation group (P = 0.910). Thus, BMS-820836 was well tolerated, with no evidence of dose-dependent discontinuations due to adverse events, but it failed to demonstrate superiority to the continuation of an existing antidepressant in patients with treatment-resistant depression.


Assuntos
Transtorno Depressivo Maior/diagnóstico , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Resistente a Tratamento/diagnóstico , Transtorno Depressivo Resistente a Tratamento/tratamento farmacológico , Internacionalidade , Isoquinolinas/uso terapêutico , Piridazinas/uso terapêutico , Adolescente , Adulto , Idoso , Transtorno Depressivo Maior/psicologia , Transtorno Depressivo Resistente a Tratamento/psicologia , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Humanos , Isoquinolinas/efeitos adversos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Piridazinas/efeitos adversos , Resultado do Tratamento , Adulto Jovem
10.
J Alzheimers Dis ; 44(4): 1039-62, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25408210

RESUMO

Cholinesterase enzymes metabolize acetylcholine (ACh). Inhibition of acetylcholinesterase (AChE) in damaged but functional cholinergic synapses in the brains of dementia patients increases intrasynaptic ACh. This enhances cholinergic neurotransmission and improves cognition. There is a window of opportunity for this symptomatic treatment effect that opens and closes during the course of dementia depending on when significant synaptic damage occurs. Cholinesterases also metabolize extrasynaptic ACh with butyrylcholinesterase (BuChE) apparently playing the major dynamic role in extracellular ACh homeostasis. Extracellular ACh plays a key regulatory role in controlling the reactivity and functional states of non-excitable cells, such as neuroglia. Current inhibitors of cholinesterases (ChEIs) have similar effects on intrasynaptic ACh, but differ markedly in abilities to upregulate extracellular AChE, inhibit BuChE, and influence the fibrilization of amyloid-ß peptides. Importantly, ChEIs can have detrimental disease modifying effects in particular individuals characterized by age, gender, and genotype. In contrast, preliminary evidence suggests that the right dose of the right ChEI in the right patient might significantly slow the progression of neurodegenerative processes. For a particular patient, understanding the condition of cholinergic synapses and the reactivity and functional status of neuroglia could allow administration of appropriate ChEI therapy for symptomatic and disease modifying benefits.


Assuntos
Inibidores da Colinesterase/uso terapêutico , Demência/tratamento farmacológico , Indanos/uso terapêutico , Fenilcarbamatos/uso terapêutico , Piperidinas/uso terapêutico , Acetilcolina/metabolismo , Acetilcolinesterase/metabolismo , Peptídeos beta-Amiloides/metabolismo , Apolipoproteínas E/genética , Butirilcolinesterase/metabolismo , Demência/genética , Demência/metabolismo , Demência/patologia , Donepezila , Humanos , Microglia/efeitos dos fármacos , Rivastigmina
11.
J Psychopharmacol ; 29(5): 526-44, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25315829

RESUMO

Many patients with major depressive disorder (MDD) only partially respond, and some have no clinically meaningful response, to current widely used antidepressant drugs. Due to the purported role of dopamine in the pathophysiology of depression, triple-reuptake inhibitors (TRIs) that simultaneously inhibit serotonin (5-HT), norepinephrine (NE) and dopamine reuptake could be a useful addition to the armamentarium of treatments for MDD. A TRI should more effectively activate mesolimbic dopamine-related reward-networks, restore positive mood and reduce potent 5-HT reuptake blockade associated "hypodopaminergic" adverse effects of decreased libido, weight gain and "blunting" of emotions. On the other hand, dopaminergic effects raise concern over abuse liability and TRIs may have many of the cardiovascular effects associated with NET inhibition. Several clinical development programs for potential TRI antidepressants have failed to demonstrate significantly greater efficacy than placebo or standard of care. Successful late-stage clinical development of a TRI is more likely if experimental research studies in the target population of depressed patients have demonstrated target engagement that differentially and dose-dependently improves assessments of reward-network dysfunction relative to existing antidepressants. TRI treatment could be individualized on the basis of predictive markers such as the burden of decreased positive mood symptoms and/or neuroimaging evidence of reward network dysfunction. This review focuses on how the next generation of monoamine-based treatments could be efficiently developed to address unmet medical need in MDD.


Assuntos
Inibidores da Captação Adrenérgica/uso terapêutico , Antidepressivos/uso terapêutico , Transtorno Depressivo Maior/tratamento farmacológico , Inibidores da Captação de Dopamina/uso terapêutico , Desenho de Fármacos , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Combinação de Medicamentos , Humanos
12.
Psychopharmacology (Berl) ; 232(3): 529-40, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25116481

RESUMO

RATIONALE: BMS-820836 is a novel antidepressant that selectively inhibits the reuptake of serotonin, norepinephrine, and dopamine. OBJECTIVE: This Phase I study assessed safety, tolerability, and pharmacokinetics of multiple daily doses of BMS-820836 in healthy subjects. Central serotonin transporter (SERT) and dopamine transporter (DAT) occupancy were assessed using positron emission tomography and [(11)C]MADAM or [(11)C]PE2I, respectively. METHODS: Fifty-seven healthy volunteers were enrolled in this double-blind, placebo-controlled, ascending multiple-dose study (ClincalTrials.gov identifier: NCT00892840). Eight participants in seven dose cohorts received oral doses of BMS-820836 (0.1-4 mg) or placebo for 14 days to assess safety, tolerability, and pharmacokinetics. Additionally, SERT and DAT occupancies were evaluated in 4-8 subjects per cohort at 8 h post-dose on Day 10 and 24 h post-dose on Day 15 at anticipated steady-state conditions. RESULTS: Most adverse events were mild to moderate; there were no serious safety concerns. Median maximum concentrations of BMS-820836 were observed at 4.0-5.5 h post-dose; estimated elimination half-life was 44-74 h. About 80 % striatal SERT occupancy was achieved after multiple doses of 0.5 mg BMS-820836 at both 8 and 24 h post-dose. Striatal DAT occupancy ranged between 14 % and 35 % at 8 h post-dose with a slight decline at 24 h post-dose. CONCLUSIONS: Multiple daily doses of up to 4 mg BMS-820836 appeared to be generally safe and well tolerated in a healthy population. SERT and DAT occupancies were in a range associated with therapeutic efficacy of antidepressants. Together with the pharmacokinetic profile of BMS-820836, the occupancy data support once-daily administration.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Dopamina/efeitos dos fármacos , Isoquinolinas , Neostriado/efeitos dos fármacos , Inibidores da Captação de Neurotransmissores , Piridazinas , Proteínas da Membrana Plasmática de Transporte de Serotonina/efeitos dos fármacos , Adulto , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Humanos , Isoquinolinas/administração & dosagem , Isoquinolinas/efeitos adversos , Isoquinolinas/farmacocinética , Isoquinolinas/farmacologia , Masculino , Pessoa de Meia-Idade , Inibidores da Captação de Neurotransmissores/administração & dosagem , Inibidores da Captação de Neurotransmissores/efeitos adversos , Inibidores da Captação de Neurotransmissores/farmacocinética , Inibidores da Captação de Neurotransmissores/farmacologia , Tomografia por Emissão de Pósitrons , Piridazinas/administração & dosagem , Piridazinas/efeitos adversos , Piridazinas/farmacocinética , Piridazinas/farmacologia , Adulto Jovem
13.
J Psychopharmacol ; 28(6): 527-35, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24811705

RESUMO

Restoration of positive mood, in addition to reducing negative mood, is an important treatment goal in the management of depression. The need to restore positive mood states in depressed patients is not adequately addressed by available treatments for major depressive disorder (MDD), suggesting that this mood dimension could be a useful target for drug development. However, for positive mood restoration to become a valid target for antidepressant drug development certain questions should be answered: are symptoms of decreased positive mood phenomenologically distinct from other symptoms of MDD? Should they be considered a distinct aspect of MDD in the diagnostic nomenclature? Is there evidence for differential responsiveness to treatment? Is the underlying pathophysiology understood and different from that of other MDD symptoms? Is low positive mood specific to depression or does it contribute to psychopathology in other disorders? Beyond these basic questions, this review identifies a number of design issues that need to be considered when conducting studies that target improving positive mood. These design issues include (1) what population to study, (2) what line of treatment to target, (3) the appropriateness and validation of methods and measures to evaluate positive mood and its restoration, (4) the role of functional outcome measures in determining success of the treatment, and (5) optimal designs for add-on therapy versus monotherapy agents.


Assuntos
Afeto/efeitos dos fármacos , Antidepressivos/uso terapêutico , Encéfalo/efeitos dos fármacos , Transtorno Depressivo Maior/tratamento farmacológico , Descoberta de Drogas/métodos , Animais , Encéfalo/fisiopatologia , Transtorno Depressivo Maior/diagnóstico , Transtorno Depressivo Maior/fisiopatologia , Transtorno Depressivo Maior/psicologia , Humanos , Recompensa , Resultado do Tratamento
14.
Alzheimers Dement ; 9(2): e1-73, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22402324

RESUMO

Retrospective data are presented to support a spectrum of early Alzheimer's disease (AD) along a continuum defined by gender and genotype. The putative neurodegenerative mechanisms driving distinct phenotypes at each end of the spectrum are glial hypoactivity associated with early failure of synaptic cholinergic neurotransmission and glial overactivation associated with loss of neural network connectivity due to accelerated age-related breakdown of myelin. In early AD, male butyrylcholinesterase K-variant carriers with one or two apolipoprotein ɛ4 alleles have prominent medial temporal atrophy, synaptic failure, cognitive decline, and accumulation of aggregated beta-amyloid peptide. Increasing synaptic acetylcholine in damaged but still functional cholinergic synapses improves cognitive symptoms, whereas increasing the ability of glia to support synapses and to clear beta-amyloid peptide might be disease-modifying. Conversely, chronic glial overactivation can also drive degenerative processes and in butyrylcholinesterase K-variant negative females generalized glial overactivation may be the main driver from mild cognitive impairment to AD. Females are more likely than males to have accelerated age-related myelin breakdown, more widespread white matter loss, loss of neural network connectivity, whole brain atrophy, and functional decline. Increasing extracellular acetylcholine levels blocks glial activation, reduces myelin loss and damage to neural network connectivity, and is disease-modifying. Between extremes characterized by gender, genotype, and age, pathophysiology may be mixed and this spectrum may explain much of the heterogeneity of amnestic mild cognitive impairment. Preservation of the functional integrity of the neural network may be an important component of strengthening cognitive reserve and significantly delaying the onset and progression of dementia, particularly in females. Prospective confirmation of these hypotheses is required. Implications for future research and therapeutic opportunities are discussed.


Assuntos
Doença de Alzheimer/genética , Butirilcolinesterase/genética , Predisposição Genética para Doença/genética , Doença de Alzheimer/patologia , Feminino , Genótipo , Humanos , Masculino , Caracteres Sexuais
15.
J Alzheimers Dis ; 20(1): 301-11, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20164585

RESUMO

Hallucinations in Alzheimer's disease (AD) may indicate greater cortical cholinergic deficits. Rivastigmine has shown larger treatment benefits versus placebo in dementia with Lewy bodies and Parkinson's disease dementia patients with hallucinations. In this retrospective, hypothesis-generating analysis, we investigated whether hallucinations in AD were associated with greater treatment benefits with rivastigmine. Data were pooled from two randomized, double-blind, 6-month, mild-to-moderate AD trials comparing rivastigmine with placebo. Co-primary efficacy parameters were the Alzheimer Disease Assessment Scale-cognitive subscale (ADAS-cog) and Clinician's Interview-Based Impression of Change plus Caregiver Input (CIBIC-plus). Efficacy data were analyzed for two sub-populations: those with and those without hallucinations at baseline. Of 927 patients, 194 (21%) reported hallucinations at baseline. Hallucinators tended to have greater decline on placebo on all outcome measures. On the ADAS-cog, mean rivastigmine - placebo differences of 3.7 points in hallucinators and 2.2 points in non-hallucinators were reported at 6 months (both p < 0.001). In hallucinators, a significant rivastigmine - placebo difference of -1.0 points (a beneficial effect) was seen on the CIBIC-plus at 6 months (p< 0.001). Non-hallucinators showed a smaller significant treatment difference of -0.3 points (p< 0.05). Interaction testing suggested that differences in treatment effects were significant between hallucinators and non-hallucinators. Hallucinations predicted greater treatment responses to oral rivastigmine.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Alucinações/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Fenilcarbamatos/uso terapêutico , Atividades Cotidianas , Idoso , Doença de Alzheimer/complicações , Análise de Variância , Cognição/efeitos dos fármacos , Relação Dose-Resposta a Droga , Método Duplo-Cego , Eletrocardiografia , Feminino , Alucinações/etiologia , Humanos , Masculino , Entrevista Psiquiátrica Padronizada , Efeito Placebo , Rivastigmina , Índice de Gravidade de Doença , Fatores de Tempo , Resultado do Tratamento
16.
Am J Alzheimers Dis Other Demen ; 25(2): 142-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19359706

RESUMO

Disease-specific assessments are not currently available for patients with Parkinson's disease dementia (PDD). This study evaluated the criterion-related validity and test-retest reliability of the Alzheimer's Disease Assessment scale cognitive subscale (ADAS-cog) in terms of sensitivity for differentiation between mild and moderate severity impairment in PDD. Six other dementia rating scales and cognitive tests were also examined. A total of 113 patients with PDD or Alzheimer disease were recruited into this 4-week, multicenter study, segregated into 2 severity groups based on Mini-Mental State Examination (MMSE) score. Mean ADAS-cog scores showed a statistically significant separation between mild and moderate severity patients in both dementias (P < .001). For the ADAS-cog, test-retest Spearman correlation coefficients were significant for each dementia type and severity. This study demonstrated the criterion-related validity and test-retest reliability for ADAS-cog in patients with PDD and strong correlations with MMSE. This supports the validity of previous results obtained with these measures in studies of patients with PDD.


Assuntos
Doença de Alzheimer/psicologia , Transtornos Cognitivos/psicologia , Cognição , Testes Neuropsicológicos , Doença de Parkinson/diagnóstico , Doença de Parkinson/psicologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/diagnóstico , Transtornos Cognitivos/diagnóstico , Feminino , Humanos , Masculino , Entrevista Psiquiátrica Padronizada , Pessoa de Meia-Idade , Testes Neuropsicológicos/normas , Reprodutibilidade dos Testes , Índice de Gravidade de Doença
17.
J Neurol Neurosurg Psychiatry ; 81(5): 547-51, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19965852

RESUMO

AIM: The authors investigated the prevalence of behavioural and psychological symptoms in vascular dementia (VaD) from baseline data of the VantagE study and compared the severity and relative frequency of symptoms between small-vessel VaD and large-vessel VaD. METHODS: Behavioural and psychological symptoms of 484 VaD patients included in a large multicentre clinical trial (registration number NCT00099216) were determined using the 12-item Neuropsychiatric Inventory (NPI). Symptoms were considered present when the score was > or =1. Based on MRI, patients were classified as having small-vessel VaD (83%) or large-vessel VaD (17%). RESULTS: Behavioural and psychological symptoms were reported in 92% of the VaD patients. The median NPI score of the total study population was 9 (0-76), with a median number of three symptoms per patient. Apathy (65%) was most prevalent, followed by depressive symptoms (45%), irritability (42%) and agitation/aggression (40%). Patients with small-vessel VaD reported more apathy, aberrant motor behaviour and hallucinations than patients with large-vessel VaD (p<0.05). In contrast, patients with large-vessel VaD reported a higher severity of agitation/aggression and euphoria (p<0.05). CONCLUSION: Behavioural and psychological symptoms are common in VaD. Patients with small-vessel and large-vessel VaD demonstrate different profiles of symptoms, with especially more apathy in small-vessel VaD and more agitation/agression in large-vessel VaD.


Assuntos
Circulação Cerebrovascular/fisiologia , Transtornos Cerebrovasculares/patologia , Demência Vascular/patologia , Demência Vascular/psicologia , Idoso , Capilares/patologia , Estudos de Coortes , Método Duplo-Cego , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Masculino , Fármacos Neuroprotetores/uso terapêutico , Fenilcarbamatos/uso terapêutico , Rivastigmina , Fatores Socioeconômicos
18.
Gend Med ; 6(2): 345-55, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19682661

RESUMO

BACKGROUND: Epidemiologic studies have identified several demographic factors, including gender, that may influence the progression from mild cognitive impairment (MCI) to Alzheimer's disease (AD). OBJECTIVE: This analysis aimed to develop a sensitive model for detecting treatment benefits in patients with MCI by controlling for factors that predict progression to AD. The study used this statistical modeling to investigate the effect of gender on treatment response in patients with MCI. METHODS: This post hoc analysis used data from the InDDEx (Investigation in Delay to Diagnosis of Alzheimer's disease with Exelon) study, a long-term (3- to 4-year), multicenter, randomized, double-blind, placebo-controlled trial of rivastigmine 3- to 12-mg/d capsules in 1018 patients with MCI. Baseline variables that were significantly associated with progression to AD within the InDDEx study population were identified. A Cox proportional hazards multivariate regression model that adjusted for these predictive factors was applied to the overall study population and to the gender subgroups. RESULTS: Of 31 baseline variables analyzed, 13 were found to be significantly associated with progression to AD in the overall population. After adjustment using the Cox proportional hazards regression model, rivastigmine was associated with a significantly lower risk for progression to AD compared with placebo in the overall population (1016 patients; hazard ratio [HR] = 0.747; P = 0.045). This effect of rivastigmine was evident in women (530 patients; HR = 0.676; P = 0.046) but not in men. CONCLUSIONS: In these patients with MCI, a significant decrease in the risk for progression to AD was found with rivastigmine over 3 to 4 years. The proportion of women whose disease progressed to AD was significantly lower in the rivastigmine group than the placebo group (P = 0.046). This effect was not found in men. These data suggest that prospectively adjusting for predictive factors could lead to a more accurate estimation of treatment benefits in future studies of MCI.


Assuntos
Doença de Alzheimer/prevenção & controle , Transtornos Cognitivos/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Fenilcarbamatos/uso terapêutico , Idoso , Progressão da Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos de Riscos Proporcionais , Rivastigmina , Fatores Sexuais
19.
Curr Med Res Opin ; 25(10): 2439-46, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19678754

RESUMO

BACKGROUND: Cholinesterase inhibitors are broadly established as first-line symptomatic therapy for Alzheimer's disease (AD). Symptomatic effects are mediated by the inhibition of acetyl- and/or butyryl-cholinesterase (AChE and/or BuChE) - the enzymes that degrade acetylcholine (ACh) in the synapse. However, ACh is also found outside the synapse ('extracellular ACh') where, among other activities, it plays a role in controlling inflammation and might impact on pathological changes. OBJECTIVE/SCOPE: New data and clinical findings are reviewed and discussed to build a preliminary case for possible disease-modifying effects of cholinesterase inhibition. FINDINGS: Trials seeking to demonstrate disease-modifying effects in subjects with mild cognitive impairment failed to reach their primary endpoints, but these failures might relate to aspects of trial methods and analyses. A re-analysis of one of these trials, using a more sensitive model controlling for factors that predict progression to AD, showed a significant delay in progression to AD with dual cholinesterase inhibition over 3 to 4 years. Taken with other evidence, it is plausible that cholinesterase inhibition might contribute to disease modification. The detection of putative disease-modifying effects may be most easily implemented in certain patient subpopulations, and genotyping studies suggest a particular role for BuChE. CONCLUSION: Long-term inhibition of BuChE might be especially important when exploring any disease-modifying effects of cholinesterase inhibitors. Elucidation of the mechanisms involved could provide insights leading to the development of new treatments that modify the development and progression of AD.


Assuntos
Inibidores da Colinesterase/uso terapêutico , Acetilcolinesterase/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Butirilcolinesterase/efeitos dos fármacos , Humanos
20.
Pharmacogenet Genomics ; 19(8): 635-46, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19617863

RESUMO

OBJECTIVE: Evaluate the effect of sex and butyrylcholinesterase (BuChE) genotype on the incidence of Alzheimer's disease (AD), cognitive and functional decline, brain volume changes, and response to rivastigmine treatment in individuals with mild cognitive impairment (MCI). METHODS: This retrospective exploratory analysis from a 3-4 year, randomized, placebo-controlled study of rivastigmine in MCI patients included participants who consented to pharmacogenetic testing. RESULTS: Of a total of 1018 patients, 490 [253 (52%) female] were successfully genotyped for BuChE. In patients receiving placebo, the BuChE wt/wt genotype was associated with a statistically significant higher incidence of progression to AD and functional decline in women, compared with men with the BuChE wt/wt genotype. In patients with a BuChE-K allele receiving placebo, incidence of progression to AD and rate of functional decline were not significantly different by sex; however, cognitive decline was significantly faster in men. Statistically significant benefits of rivastigmine treatment on incident AD, functional decline, ventricular volume expansion, whole-brain atrophy, and white matter loss were evident in female BuChE wt/wt. CONCLUSION: Sex and BuChE genotype seem to differentially influence the type of decline in MCI patients, with more rapid progression of cognitive decline in male BuChE-K, and more incident AD and functional decline in female BuChE wt/wt. Cognitive decline in male BuChE-K and functional decline and incident AD in female BuChE wt/wt were significantly attenuated by rivastigmine. Rivastigmine treatment also significantly reduced ventricular expansion, whole-brain atrophy rate, and white matter loss in female BuChE wt/wt, suggesting a possible disease-modifying effect.


Assuntos
Doença de Alzheimer/diagnóstico , Doença de Alzheimer/genética , Butirilcolinesterase/genética , Transtornos Cognitivos/diagnóstico , Transtornos Cognitivos/genética , Fenilcarbamatos/farmacologia , Biomarcadores/metabolismo , Inibidores da Colinesterase/farmacologia , Cognição , Progressão da Doença , Feminino , Genótipo , Humanos , Masculino , Fármacos Neuroprotetores/farmacologia , Farmacogenética/métodos , Placebos , Estudos Retrospectivos , Rivastigmina , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...