首页 > 最新文献

CNS drug reviews最新文献

英文 中文
Efficacy of Rimonabant and Other Cannabinoid CB1 Receptor Antagonists in Reducing Food Intake and Body Weight: Preclinical and Clinical Data 利莫那班和其他大麻素CB1受体拮抗剂减少食物摄入和体重的疗效:临床前和临床数据
Pub Date : 2006-08-29 DOI: 10.1111/j.1527-3458.2006.00091.x
Mauro A. M. Carai, Giancarlo Colombo, Paola Maccioni, Gian Luigi Gessa

The present paper focuses on the different lines of evidence indicating that cannabinoid CB1 receptor antagonists, including the prototype rimonabant, reduce food intake and body weight in laboratory animals. Recent clinical surveys demonstrated that rimonabant significantly reduced body weight also in overweight/obese humans. Treatment with rimonabant was associated with a beneficial effect on different metabolic parameters and cardiovascular risk factors linked to overweight. The data reviewed in this paper suggest that cannabinoid CB1 receptor antagonists may constitute a novel class of drugs potentially effective in the treatment of obesity-related disorders.

本论文着重于不同的证据表明大麻素CB1受体拮抗剂,包括原型利莫那班,减少食物摄入量和体重在实验动物。最近的临床调查表明,利莫那班也能显著减轻超重/肥胖人群的体重。利莫那班治疗对不同代谢参数和与超重相关的心血管危险因素有有益影响。本文回顾的数据表明,大麻素CB1受体拮抗剂可能构成一类潜在有效治疗肥胖相关疾病的新型药物。
{"title":"Efficacy of Rimonabant and Other Cannabinoid CB1 Receptor Antagonists in Reducing Food Intake and Body Weight: Preclinical and Clinical Data","authors":"Mauro A. M. Carai,&nbsp;Giancarlo Colombo,&nbsp;Paola Maccioni,&nbsp;Gian Luigi Gessa","doi":"10.1111/j.1527-3458.2006.00091.x","DOIUrl":"10.1111/j.1527-3458.2006.00091.x","url":null,"abstract":"<div>\u0000 \u0000 <p>The present paper focuses on the different lines of evidence indicating that cannabinoid CB<sub>1</sub> receptor antagonists, including the prototype rimonabant, reduce food intake and body weight in laboratory animals. Recent clinical surveys demonstrated that rimonabant significantly reduced body weight also in overweight/obese humans. Treatment with rimonabant was associated with a beneficial effect on different metabolic parameters and cardiovascular risk factors linked to overweight. The data reviewed in this paper suggest that cannabinoid CB<sub>1</sub> receptor antagonists may constitute a novel class of drugs potentially effective in the treatment of obesity-related disorders.</p>\u0000 </div>","PeriodicalId":94307,"journal":{"name":"CNS drug reviews","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2006-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1111/j.1527-3458.2006.00091.x","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"26300003","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 46
Humanin and Colivelin: Neuronal-Death-Suppressing Peptides for Alzheimer's Disease and Amyotrophic Lateral Sclerosis 人蛋白和Colivelin:阿尔茨海默病和肌萎缩侧索硬化症的神经元死亡抑制肽
Pub Date : 2006-08-29 DOI: 10.1111/j.1527-3458.2006.00113.x
Masaaki Matsuoka, Yuichi Hashimoto, Sadakazu Aiso, Ikuo Nishimoto

Humanin (HN), a 24-amino-acid neuroprotective peptide, was originally found in the occipital lobe of an autopsied Alzheimer's disease (AD) patient. HN inhibits neuronal death by binding to its specific receptor on the cell membrane and triggering a Jak2/STAT3 prosurvival pathway. The activation of this pathway may represent a therapeutic approach to AD. HN also exhibits neuroprotective activity against toxicity by familial amyotrophic lateral sclerosis (ALS)-related mutant superoxide dismutase (SOD1). Recent investigations established that AGA-(C8R)-HNG17, a 17-amno-acid derivative of HN, is 105 times more potent as a neuroprotective than HN; at 10-picomolar and higher concentrations in vitro it completely suppresses neuronal death. Moreover, a 26-amino-acid peptide colivelin (CL), composed of activity-dependent neurotrophic factor (ADNF) C-terminally fused to AGA-(C8R)-HNG17, provides complete neuroprotection at 100-femtomolar or higher concentrations in vitro. A series of experiments using mouse AD and ALS models further established the efficacy of HN derivatives, including CL, against these diseases in vivo. HN and CL can be viewed as drug candidates for neuronal death suppression therapy in AD or ALS.

Humanin (HN)是一种由24个氨基酸组成的神经保护肽,最初是在一名阿尔茨海默病(AD)尸检患者的枕叶中发现的。HN通过结合其在细胞膜上的特异性受体并触发Jak2/STAT3促存活通路来抑制神经元死亡。激活这一途径可能是一种治疗AD的方法。HN还表现出抗家族性肌萎缩性侧索硬化症(ALS)相关突变体超氧化物歧化酶(SOD1)毒性的神经保护活性。最近的研究证实,HN的17-氨基酸衍生物AGA-(C8R)- hng17的神经保护作用是HN的105倍;在10皮摩尔及更高的体外浓度下,它完全抑制神经元死亡。此外,一种由活性依赖性神经营养因子(ADNF) c末端融合到AGA-(C8R)- hng17的26氨基酸肽colivelin (CL)在体外提供100飞摩尔或更高浓度的完全神经保护。利用小鼠AD和ALS模型进行的一系列实验进一步证实了HN衍生物(包括CL)在体内对这些疾病的治疗作用。HN和CL可被视为AD或ALS神经元死亡抑制治疗的候选药物。
{"title":"Humanin and Colivelin: Neuronal-Death-Suppressing Peptides for Alzheimer's Disease and Amyotrophic Lateral Sclerosis","authors":"Masaaki Matsuoka,&nbsp;Yuichi Hashimoto,&nbsp;Sadakazu Aiso,&nbsp;Ikuo Nishimoto","doi":"10.1111/j.1527-3458.2006.00113.x","DOIUrl":"10.1111/j.1527-3458.2006.00113.x","url":null,"abstract":"<div>\u0000 \u0000 <p>Humanin (HN), a 24-amino-acid neuroprotective peptide, was originally found in the occipital lobe of an autopsied Alzheimer's disease (AD) patient. HN inhibits neuronal death by binding to its specific receptor on the cell membrane and triggering a Jak2/STAT3 prosurvival pathway. The activation of this pathway may represent a therapeutic approach to AD. HN also exhibits neuroprotective activity against toxicity by familial amyotrophic lateral sclerosis (ALS)-related mutant superoxide dismutase (SOD1). Recent investigations established that AGA-(C8R)-HNG17, a 17-amno-acid derivative of HN, is 10<sup>5</sup> times more potent as a neuroprotective than HN; at 10-picomolar and higher concentrations in vitro it completely suppresses neuronal death. Moreover, a 26-amino-acid peptide colivelin (CL), composed of activity-dependent neurotrophic factor (ADNF) C-terminally fused to AGA-(C8R)-HNG17, provides complete neuroprotection at 100-femtomolar or higher concentrations in vitro. A series of experiments using mouse AD and ALS models further established the efficacy of HN derivatives, including CL, against these diseases in vivo. HN and CL can be viewed as drug candidates for neuronal death suppression therapy in AD or ALS.</p>\u0000 </div>","PeriodicalId":94307,"journal":{"name":"CNS drug reviews","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2006-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1111/j.1527-3458.2006.00113.x","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"26242146","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Metabotropic Glutamate Receptor Subtype 5 Antagonists MPEP and MTEP 代谢性谷氨酸受体亚型5拮抗剂MPEP和MTEP
Pub Date : 2006-08-29 DOI: 10.1111/j.1527-3458.2006.00149.x
Paul M. Lea IV, Alan I. Faden

Glutamate regulates the function of central nervous system (CNS), in part, through the cAMP and/or IP3/DAG second messenger-associated metabotropic glutamate receptors (mGluRs). The mGluR5 antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP) has been extensively used to elucidate potential physiological and pathophysiological functions of mGluR5. Unfortunately, recent evidence indicates significant non-specific actions of MPEP, including inhibition of NMDA receptors. In contrast, in vivo and in vitro characterization of the newer mGluR5 antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine (MTEP) indicates that it is more highly selective for mGluR5 over mGluR1, has no effect on other mGluR subtypes, and has fewer off-target effects than MPEP. This article reviews literature on both of these mGluR5 antagonists, which suggests their possible utility in neurodegeneration, addiction, anxiety and pain management.

谷氨酸调节中枢神经系统(CNS)的功能,部分通过cAMP和/或IP3/DAG第二信使相关代谢谷氨酸受体(mGluRs)。mGluR5拮抗剂2-甲基-6-(苯乙基)吡啶(MPEP)已被广泛用于阐明mGluR5潜在的生理和病理生理功能。不幸的是,最近的证据表明MPEP具有显著的非特异性作用,包括抑制NMDA受体。相比之下,较新的mGluR5拮抗剂3-[(2-甲基-1,3-噻唑-4-基)乙基]吡啶(MTEP)的体内和体外表征表明,它对mGluR5的选择性比mGluR1更高,对其他mGluR亚型没有影响,并且脱靶效应比MPEP更少。本文回顾了这两种mGluR5拮抗剂的文献,提示它们在神经退行性疾病、成瘾、焦虑和疼痛管理方面的可能用途。
{"title":"Metabotropic Glutamate Receptor Subtype 5 Antagonists MPEP and MTEP","authors":"Paul M. Lea IV,&nbsp;Alan I. Faden","doi":"10.1111/j.1527-3458.2006.00149.x","DOIUrl":"10.1111/j.1527-3458.2006.00149.x","url":null,"abstract":"<div>\u0000 \u0000 <p>Glutamate regulates the function of central nervous system (CNS), in part, through the cAMP and/or IP3/DAG second messenger-associated metabotropic glutamate receptors (mGluRs). The mGluR5 antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP) has been extensively used to elucidate potential physiological and pathophysiological functions of mGluR5. Unfortunately, recent evidence indicates significant non-specific actions of MPEP, including inhibition of NMDA receptors. In contrast, in vivo and in vitro characterization of the newer mGluR5 antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine (MTEP) indicates that it is more highly selective for mGluR5 over mGluR1, has no effect on other mGluR subtypes, and has fewer off-target effects than MPEP. This article reviews literature on both of these mGluR5 antagonists, which suggests their possible utility in neurodegeneration, addiction, anxiety and pain management.</p>\u0000 </div>","PeriodicalId":94307,"journal":{"name":"CNS drug reviews","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2006-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1111/j.1527-3458.2006.00149.x","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"26242150","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 153
Preclinical and Clinical Pharmacology of DOV 216,303, a “Triple” Reuptake Inhibitor “三重”再摄取抑制剂DOV 216,303的临床前和临床药理学研究
Pub Date : 2006-08-29 DOI: 10.1111/j.1527-3458.2006.00123.x
Phil Skolnick, Philip Krieter, Joseph Tizzano, Anthony Basile, Piotr Popik, Pal Czobor, Arnold Lippa

DOV 216,303 [(±)-1-(3,4-dichlorophenyl)-3-azabicyclo-[3.1.0]hexane hydrochloride] is the prototype of a class of compounds referred to as “triple” reuptake inhibitors. Such compounds inhibit the reuptake of norepinephrine (NE), serotonin (5-HT), and dopamine (DA), the three neurotransmitters most closely linked to major depressive disorder. DOV 216,303 inhibits [3H]NE, [3H]5-HT, and [3H]DA uptake to the corresponding human recombinant transporters (expressed in HEK 293 cells) with IC50 values of ∼20, 14, and 78 nM, respectively. DOV 216,303 is active in tests predictive of antidepressant activity including the mouse forced swim test and reversal of tetrabenazine-induced ptosis and locomotor depression. The pharmacodynamic, pharmacokinetic, and toxicological profile of DOV 216,303 in animals prompted us to initiate clinical studies. In both single and multiple dose studies using normal volunteers, DOV 216,303 was safe and well-tolerated. Furthermore, both Cmax and AUC values were dose-proportional between 5–150 mg. The plasma concentrations of DOV 216,303 at doses >10 mg were in excess of the IC50 values for inhibition of biogenic amine reuptake. In a Phase II study designed to explore the safety and tolerability of DOV 216,303 in depressed individuals, patients received either 100 mg DOV 216,303 (50 mg b.i.d.) or 40 mg citalopram (20 mg, b.i.d.) for two weeks. A placebo arm was not employed in this study because several institutional review boards required administration of an active control to severely depressed individuals. Time dependent reductions in HAM-D scores (the primary outcome measure) were observed in both the DOV 216,303 and citalopram groups compared to baseline scores (p < 0.0001). The side effect profile was not remarkably different between treatment arms. These findings provide preliminary evidence of a clinically meaningful antidepressant action with a molecule capable of inhibiting the three transmitters most closely linked to major depressive disorder.

DOV 216,303[(±)-1-(3,4-二氯苯基)-3-氮杂环-[3.1.0]盐酸己烷]是一类被称为“三重”再摄取抑制剂的化合物的原型。这些化合物抑制去甲肾上腺素(NE)、血清素(5-HT)和多巴胺(DA)的再摄取,这三种神经递质与重度抑郁症最密切相关。DOV 216,303抑制[3H]NE、[3H]5-HT和[3H]DA对相应的人重组转运体(在HEK 293细胞中表达)的摄取,IC50值分别为~ 20、14和78 nM。DOV 216,303在预测抗抑郁活性的试验中具有活性,包括小鼠强迫游泳试验和逆转tetrabenazine诱导的上下垂和运动抑制。DOV 216303在动物体内的药效学、药代动力学和毒理学特征促使我们开始临床研究。在正常志愿者的单次和多次剂量研究中,DOV 216,303是安全且耐受性良好的。Cmax和AUC值在5 - 150mg之间呈剂量正比关系。DOV 216,303在10 mg剂量下的血浆浓度超过抑制生物胺再摄取的IC50值。在一项旨在探索DOV 216303在抑郁症患者中的安全性和耐受性的II期研究中,患者接受100mg DOV 216303 (50mg,每日一次)或40mg西酞普兰(20mg,每日一次)两周。本研究没有使用安慰剂组,因为几个机构审查委员会要求对严重抑郁症患者进行积极控制。与基线评分相比,DOV 216,303组和西酞普兰组均观察到HAM-D评分(主要预后指标)的时间依赖性降低(p < 0.0001)。治疗组之间的副作用没有显著差异。这些发现为临床有意义的抗抑郁作用提供了初步证据,这种分子能够抑制与重度抑郁症最密切相关的三种递质。
{"title":"Preclinical and Clinical Pharmacology of DOV 216,303, a “Triple” Reuptake Inhibitor","authors":"Phil Skolnick,&nbsp;Philip Krieter,&nbsp;Joseph Tizzano,&nbsp;Anthony Basile,&nbsp;Piotr Popik,&nbsp;Pal Czobor,&nbsp;Arnold Lippa","doi":"10.1111/j.1527-3458.2006.00123.x","DOIUrl":"10.1111/j.1527-3458.2006.00123.x","url":null,"abstract":"<div>\u0000 \u0000 <p>DOV 216,303 [(±)-1-(3,4-dichlorophenyl)-3-azabicyclo-[3.1.0]hexane hydrochloride] is the prototype of a class of compounds referred to as “triple” reuptake inhibitors. Such compounds inhibit the reuptake of norepinephrine (NE), serotonin (5-HT), and dopamine (DA), the three neurotransmitters most closely linked to major depressive disorder. DOV 216,303 inhibits [<sup>3</sup>H]NE, [<sup>3</sup>H]5-HT, and [<sup>3</sup>H]DA uptake to the corresponding human recombinant transporters (expressed in HEK 293 cells) with IC<sub>50</sub> values of ∼20, 14, and 78 nM, respectively. DOV 216,303 is active in tests predictive of antidepressant activity including the mouse forced swim test and reversal of tetrabenazine-induced ptosis and locomotor depression. The pharmacodynamic, pharmacokinetic, and toxicological profile of DOV 216,303 in animals prompted us to initiate clinical studies. In both single and multiple dose studies using normal volunteers, DOV 216,303 was safe and well-tolerated. Furthermore, both C<sub>max</sub> and AUC values were dose-proportional between 5–150 mg. The plasma concentrations of DOV 216,303 at doses &gt;10 mg were in excess of the IC<sub>50</sub> values for inhibition of biogenic amine reuptake. In a Phase II study designed to explore the safety and tolerability of DOV 216,303 in depressed individuals, patients received either 100 mg DOV 216,303 (50 mg b.i.d.) or 40 mg citalopram (20 mg, b.i.d.) for two weeks. A placebo arm was not employed in this study because several institutional review boards required administration of an active control to severely depressed individuals. Time dependent reductions in HAM-D scores (the primary outcome measure) were observed in both the DOV 216,303 and citalopram groups compared to baseline scores (p &lt; 0.0001). The side effect profile was not remarkably different between treatment arms. These findings provide preliminary evidence of a clinically meaningful antidepressant action with a molecule capable of inhibiting the three transmitters most closely linked to major depressive disorder.</p>\u0000 </div>","PeriodicalId":94307,"journal":{"name":"CNS drug reviews","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2006-08-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1111/j.1527-3458.2006.00123.x","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"26242147","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 102
Neuroprotective Activity of 3-Aminopyridine-2-Carboxaldehyde Thiosemicarbazone (PAN-811), a Cancer Therapeutic Agent 肿瘤治疗剂3-氨基吡啶-2-羧基硫代氨基脲(PAN-811)的神经保护作用
Pub Date : 2006-07-10 DOI: 10.1111/j.1527-3458.2006.00077.x
Zhi-Gang Jiang, Michael S. Lebowitz, Hossein A. Ghanbari

3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP) is a highly-hydrophobic small molecule that was originally developed for cancer therapy (Triapine®, Vion Pharmaceuticals) due to its ability to inhibit ribonucleotide reductase, a key enzyme required for DNA synthesis. 3-AP has a high affinity for divalent cations, chelating the Fe2+ at the R2 subunit of the enzyme and inhibiting formation of a tyrosyl radical essential for ribonucleotide reduction. We have demonstrated that 3-AP is also a potent neuroprotectant (as such, it is referred to as “PAN-811”). In vitro it completely blocks ischemic neurotoxicity at a concentration of 0.5 μM (EC50 ≊ 0.35 μM) and hypoxic toxicity at 1.2 μM (EC50 ≊ 0.75 μM). Full protection of primary cortical and striatal neurons can be achieved with 3-AP when it is added to the medium at up to six hours after an ischemic insult. 3-AP also suppresses cell death induced by neurotoxic agents, including staurosporine, veratridine and glutamate, indicating activity against a central target(s) in the neurodegenerative process. 3-AP acts via neutralization of two important intracellular effectors of excitatory neurotoxicity; calcium and free radicals. Its reported ability to elevate anti-apoptotic proteins is likely to be a consequence of the suppression of excessive intracellular free calcium. In a rat model of transient ischemia, a single bolus delivery of 3-AP 1 h after the initiation of ischemic attack reduced infarct volume by 59% when administered i.c.v. (50 μg per rat) and by 35% when administered i.v. (1 mg/kg). In Phase I clinical trials in cancer therapy 3-AP had no cardiovascular, CNS or other major adverse effects. Thus, 3-AP has a high potential for development as a novel, potent neuroprotectant for the treatment of neurodegenerative diseases.

3-氨基吡啶-2-羧基硫代氨基脲(3-AP)是一种高度疏水的小分子,最初被开发用于癌症治疗(Triapine®,Vion Pharmaceuticals),因为它能够抑制核糖核苷酸还原酶,DNA合成所需的关键酶。3-AP对二价阳离子有很高的亲和力,在酶的R2亚基上螯合Fe2+,抑制核糖核苷酸还原所必需的酪氨酸自由基的形成。我们已经证明3-AP也是一种有效的神经保护剂(因此,它被称为“PAN-811”)。体外完全阻断0.5 μM (EC50 0.35 μM)浓度下的缺血神经毒性和1.2 μM (EC50 0.75 μM)浓度下的缺氧毒性。缺血损伤后6小时将3-AP添加到培养基中,可实现对初级皮质和纹状体神经元的充分保护。3-AP还能抑制神经毒性药物(包括staurosporine、veratridine和谷氨酸)诱导的细胞死亡,表明其对神经退行性过程中的一个中心靶点具有活性。3-AP通过中和兴奋性神经毒性的两种重要细胞内效应物起作用;钙和自由基。据报道,其提高抗凋亡蛋白的能力可能是抑制过量细胞内游离钙的结果。在短暂性缺血大鼠模型中,缺血发作开始后1小时单次给药3-AP可使梗死面积减少59%(每只大鼠50 μg),静脉给药(1 mg/kg)可使梗死面积减少35%。在癌症治疗的I期临床试验中,3-AP没有心血管、中枢神经系统或其他主要不良反应。因此,3-AP作为一种治疗神经退行性疾病的新型有效神经保护剂具有很高的发展潜力。
{"title":"Neuroprotective Activity of 3-Aminopyridine-2-Carboxaldehyde Thiosemicarbazone (PAN-811), a Cancer Therapeutic Agent","authors":"Zhi-Gang Jiang,&nbsp;Michael S. Lebowitz,&nbsp;Hossein A. Ghanbari","doi":"10.1111/j.1527-3458.2006.00077.x","DOIUrl":"10.1111/j.1527-3458.2006.00077.x","url":null,"abstract":"<p>3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP) is a highly-hydrophobic small molecule that was originally developed for cancer therapy (Triapine®, Vion Pharmaceuticals) due to its ability to inhibit ribonucleotide reductase, a key enzyme required for DNA synthesis. 3-AP has a high affinity for divalent cations, chelating the Fe<sup>2+</sup> at the R2 subunit of the enzyme and inhibiting formation of a tyrosyl radical essential for ribonucleotide reduction. We have demonstrated that 3-AP is also a potent neuroprotectant (as such, it is referred to as “PAN-811”). In vitro it completely blocks ischemic neurotoxicity at a concentration of 0.5 μM (EC<sub>50</sub> ≊ 0.35 μM) and hypoxic toxicity at 1.2 μM (EC<sub>50</sub> ≊ 0.75 μM). Full protection of primary cortical and striatal neurons can be achieved with 3-AP when it is added to the medium at up to six hours after an ischemic insult. 3-AP also suppresses cell death induced by neurotoxic agents, including staurosporine, veratridine and glutamate, indicating activity against a central target(s) in the neurodegenerative process. 3-AP acts via neutralization of two important intracellular effectors of excitatory neurotoxicity; calcium and free radicals. Its reported ability to elevate anti-apoptotic proteins is likely to be a consequence of the suppression of excessive intracellular free calcium. In a rat model of transient ischemia, a single bolus delivery of 3-AP 1 h after the initiation of ischemic attack reduced infarct volume by 59% when administered i.c.v. (50 μg per rat) and by 35% when administered i.v. (1 mg/kg). In Phase I clinical trials in cancer therapy 3-AP had no cardiovascular, CNS or other major adverse effects. Thus, 3-AP has a high potential for development as a novel, potent neuroprotectant for the treatment of neurodegenerative diseases.</p>","PeriodicalId":94307,"journal":{"name":"CNS drug reviews","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2006-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1111/j.1527-3458.2006.00077.x","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"26140610","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 39
A New Look at the 5α-Reductase Inhibitor Finasteride 5α-还原酶抑制剂非那雄胺的新研究
Pub Date : 2006-07-10 DOI: 10.1111/j.1527-3458.2006.00053.x
Deborah A. Finn, Amy S. Beadles-Bohling, Ethan H. Beckley, Matthew M. Ford, Katherine R. Gililland, Rebecca E. Gorin-Meyer, Kristine M. Wiren

Finasteride is the first 5α-reductase inhibitor that received clinical approval for the treatment of human benign prostatic hyperplasia (BPH) and androgenetic alopecia (male pattern hair loss). These clinical applications are based on the ability of finasteride to inhibit the Type II isoform of the 5α-reductase enzyme, which is the predominant form in human prostate and hair follicles, and the concomitant reduction of testosterone to dihydrotestosterone (DHT). In addition to catalyzing the rate-limiting step in the reduction of testosterone, both isoforms of the 5α-reductase enzyme are responsible for the reduction of progesterone and deoxycorticosterone to dihydroprogesterone (DHP) and dihydrodeoxycorticosterone (DHDOC), respectively. Recent preclinical data indicate that the subsequent 3α-reduction of DHT, DHP and DHDOC produces steroid metabolites with rapid non-genomic effects on brain function and behavior, primarily via an enhancement of γ-aminobutyric acid (GABA)ergic inhibitory neurotransmission. Consistent with their ability to enhance the action of GABA at GABAA receptors, these steroid derivatives (termed neuroactive steroids) possess anticonvulsant, antidepressant and anxiolytic effects in addition to altering aspects of sexual- and alcohol-related behaviors. Thus, finasteride, which inhibits both isoforms of 5α-reductase in rodents, has been used as a tool to manipulate neuroactive steroid levels and determine the impact on behavior. Results of some preclinical studies and clinical observations with finasteride are described in this review article. The data suggest that endogenous neuroactive steroid levels may be inversely related to symptoms of premenstrual and postpartum dysphoric disorder, catamenial epilepsy, depression, and alcohol withdrawal.

非那雄胺是首个获得临床批准用于治疗人类良性前列腺增生(BPH)和雄激素性脱发(男性型脱发)的5α-还原酶抑制剂。这些临床应用是基于非那雄胺抑制5α-还原酶II型异构体的能力,5α-还原酶是人类前列腺和毛囊中的主要形式,并伴随睾酮还原为双氢睾酮(DHT)。5α-还原酶的两种同工型除了催化睾酮还原的限速步骤外,还分别负责将孕酮和脱氧皮质酮还原为二氢孕酮(DHP)和二氢脱氧皮质酮(DHDOC)。最近的临床前数据表明,随后DHT、DHP和DHDOC的3α-还原产生类固醇代谢物,主要通过增强γ-氨基丁酸(GABA)能抑制性神经传递,对脑功能和行为产生快速的非基因组效应。与增强GABA对GABAA受体作用的能力相一致,这些类固醇衍生物(称为神经活性类固醇)除了改变性和酒精相关行为外,还具有抗惊厥、抗抑郁和抗焦虑的作用。因此,非那雄胺抑制啮齿类动物5α-还原酶的两种亚型,已被用作操纵神经活性类固醇水平和确定对行为影响的工具。本文综述了非那雄胺的一些临床前研究和临床观察结果。数据表明,内源性神经活性类固醇水平可能与经前和产后烦躁不安、睡眠性癫痫、抑郁和酒精戒断的症状呈负相关。
{"title":"A New Look at the 5α-Reductase Inhibitor Finasteride","authors":"Deborah A. Finn,&nbsp;Amy S. Beadles-Bohling,&nbsp;Ethan H. Beckley,&nbsp;Matthew M. Ford,&nbsp;Katherine R. Gililland,&nbsp;Rebecca E. Gorin-Meyer,&nbsp;Kristine M. Wiren","doi":"10.1111/j.1527-3458.2006.00053.x","DOIUrl":"10.1111/j.1527-3458.2006.00053.x","url":null,"abstract":"<p>Finasteride is the first 5α-reductase inhibitor that received clinical approval for the treatment of human benign prostatic hyperplasia (BPH) and androgenetic alopecia (male pattern hair loss). These clinical applications are based on the ability of finasteride to inhibit the Type II isoform of the 5α-reductase enzyme, which is the predominant form in human prostate and hair follicles, and the concomitant reduction of testosterone to dihydrotestosterone (DHT). In addition to catalyzing the rate-limiting step in the reduction of testosterone, both isoforms of the 5α-reductase enzyme are responsible for the reduction of progesterone and deoxycorticosterone to dihydroprogesterone (DHP) and dihydrodeoxycorticosterone (DHDOC), respectively. Recent preclinical data indicate that the subsequent 3α-reduction of DHT, DHP and DHDOC produces steroid metabolites with rapid non-genomic effects on brain function and behavior, primarily via an enhancement of γ-aminobutyric acid (GABA)ergic inhibitory neurotransmission. Consistent with their ability to enhance the action of GABA at GABA<sub>A</sub> receptors, these steroid derivatives (termed neuroactive steroids) possess anticonvulsant, antidepressant and anxiolytic effects in addition to altering aspects of sexual- and alcohol-related behaviors. Thus, finasteride, which inhibits both isoforms of 5α-reductase in rodents, has been used as a tool to manipulate neuroactive steroid levels and determine the impact on behavior. Results of some preclinical studies and clinical observations with finasteride are described in this review article. The data suggest that endogenous neuroactive steroid levels may be inversely related to symptoms of premenstrual and postpartum dysphoric disorder, catamenial epilepsy, depression, and alcohol withdrawal.</p>","PeriodicalId":94307,"journal":{"name":"CNS drug reviews","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2006-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1111/j.1527-3458.2006.00053.x","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"26140611","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 140
Neuroprotective Effects of Edaravone: a Novel Free Radical Scavenger in Cerebrovascular Injury 依达拉奉:一种新型自由基清除剂对脑血管损伤的神经保护作用
Pub Date : 2006-07-10 DOI: 10.1111/j.1527-3458.2006.00009.x
Hiroshi Yoshida, Hidekatsu Yanai, Yoshihisa Namiki, Kayoko Fukatsu-Sasaki, Nobuyuki Furutani, Norio Tada

Recanalization and neuroprotection have been mainly targeted for the specific treatment of acute ischemic stroke. Free radicals play a crucial role in brain ischemic injury by exacerbating membrane damage through peroxidation of unsaturated fatty acids of cell membrane, leading to neuronal death and brain edema. Free radicals have been implicated in stroke pathophysiology as pivotal contributors to cell injury. Edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one) is a novel potent free radical scavenger that has been clinically used to reduce the neuronal damage following ischemic stroke. Edaravone exerts neuroprotective effects by inhibiting endothelial injury and by ameliorating neuronal damage in brain ischemia. Edaravone provides the desirable features of NOS: it increases eNOS (beneficial NOS for rescuing ischemic stroke) and decreases nNOS and iNOS (detrimental NOS). Post- reperfusion brain edema and hemorrhagic events induced by thrombolytic therapy may be reduced by edaravone pretreatment. Increased productions of superoxide and NO in the brain after reperfusion and a concomitant surge in oxygen free radicals with increased NO during recirculation lead to formation of peroxynitrite, a superpotent radical. Edaravone, which inhibits oxidation and enhances NO production derived from increased eNOS expression, may improve and conserve cerebral blood flow without peroxynitrite generation during reperfusion. Clinical experience with edaravone suggests that this drug has a wide therapeutic time window. The combination therapy (a thrombolytic plus edaravone) is likely to target brain edema, reduce stroke death and improve the recovery from neurological deficits in stoke patients.

再通和神经保护一直是急性缺血性脑卒中特异性治疗的主要目标。自由基在脑缺血损伤中起着至关重要的作用,自由基通过细胞膜不饱和脂肪酸的过氧化作用加重膜损伤,导致神经元死亡和脑水肿。自由基在脑卒中病理生理学中被认为是细胞损伤的关键因素。依达拉奉(3-甲基-1-苯基-2-吡唑啉-5- 1)是一种新型有效的自由基清除剂,已被临床用于减少缺血性中风后的神经元损伤。依达拉奉通过抑制内皮损伤和改善脑缺血时的神经元损伤发挥神经保护作用。依达拉奉提供了NOS的理想特征:增加eNOS(挽救缺血性卒中的有益NOS),降低nNOS和iNOS(有害NOS)。依达拉奉预处理可减少溶栓治疗引起的再灌注后脑水肿和出血事件。再灌注后大脑中超氧化物和一氧化氮的产生增加,再循环过程中氧自由基的激增与一氧化氮的增加导致过氧亚硝酸盐的形成,这是一种超强自由基。依达拉奉可以抑制氧化并增加eNOS表达引起的NO的产生,可能在再灌注时改善和保存脑血流量而不产生过氧亚硝酸盐。依达拉奉的临床经验表明,该药具有较宽的治疗时间窗。联合治疗(溶栓加依达拉奉)可能针对脑水肿,减少卒中死亡,并改善脑卒中患者神经功能障碍的恢复。
{"title":"Neuroprotective Effects of Edaravone: a Novel Free Radical Scavenger in Cerebrovascular Injury","authors":"Hiroshi Yoshida,&nbsp;Hidekatsu Yanai,&nbsp;Yoshihisa Namiki,&nbsp;Kayoko Fukatsu-Sasaki,&nbsp;Nobuyuki Furutani,&nbsp;Norio Tada","doi":"10.1111/j.1527-3458.2006.00009.x","DOIUrl":"10.1111/j.1527-3458.2006.00009.x","url":null,"abstract":"<p>Recanalization and neuroprotection have been mainly targeted for the specific treatment of acute ischemic stroke. Free radicals play a crucial role in brain ischemic injury by exacerbating membrane damage through peroxidation of unsaturated fatty acids of cell membrane, leading to neuronal death and brain edema. Free radicals have been implicated in stroke pathophysiology as pivotal contributors to cell injury. Edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one) is a novel potent free radical scavenger that has been clinically used to reduce the neuronal damage following ischemic stroke. Edaravone exerts neuroprotective effects by inhibiting endothelial injury and by ameliorating neuronal damage in brain ischemia. Edaravone provides the desirable features of NOS: it increases eNOS (beneficial NOS for rescuing ischemic stroke) and decreases nNOS and iNOS (detrimental NOS). Post- reperfusion brain edema and hemorrhagic events induced by thrombolytic therapy may be reduced by edaravone pretreatment. Increased productions of superoxide and NO in the brain after reperfusion and a concomitant surge in oxygen free radicals with increased NO during recirculation lead to formation of peroxynitrite, a superpotent radical. Edaravone, which inhibits oxidation and enhances NO production derived from increased eNOS expression, may improve and conserve cerebral blood flow without peroxynitrite generation during reperfusion. Clinical experience with edaravone suggests that this drug has a wide therapeutic time window. The combination therapy (a thrombolytic plus edaravone) is likely to target brain edema, reduce stroke death and improve the recovery from neurological deficits in stoke patients.</p>","PeriodicalId":94307,"journal":{"name":"CNS drug reviews","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2006-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1111/j.1527-3458.2006.00009.x","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"26139674","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 335
Bryostatin-1: Pharmacology and Therapeutic Potential as a CNS Drug 苔藓虫素-1:作为中枢神经系统药物的药理学和治疗潜力
Pub Date : 2006-07-10 DOI: 10.1111/j.1527-3458.2006.00001.x
Miao-Kun Sun, Daniel L. Alkon

Bryostatin-1 is a powerful protein kinase C (PKC) agonist, activating PKC isozymes at nanomolar concentrations. Pharmacological studies of bryostatin-1 have mainly been focused on its action in preventing tumor growth. Emerging evidence suggests, however, that bryostatin-1 exhibits additional important pharmacological activities. In preclinical studies bryostatin-1 has been shown at appropriate doses to have cognitive restorative and antidepressant effects. The underlying pharmacological mechanisms may involve an activation of PKC isozymes, induction of synthesis of proteins required for long-term memory, restoration of stress-evoked inhibition of PKC activity, and reduction of neurotoxic amyloid accumulation and tau protein hyperphosphorylation. The therapeutic potential of bryostatin-1 as a CNS drug should be further explored.

苔藓虫素-1是一种强大的蛋白激酶C (PKC)激动剂,在纳摩尔浓度下激活PKC同工酶。苔藓虫素-1的药理研究主要集中在其抑制肿瘤生长的作用上。然而,新出现的证据表明苔藓虫素-1具有其他重要的药理活性。在临床前研究中,苔藓抑素-1在适当剂量下具有认知恢复和抗抑郁作用。潜在的药理学机制可能涉及PKC同工酶的激活,诱导长期记忆所需蛋白质的合成,恢复应激引起的PKC活性抑制,减少神经毒性淀粉样蛋白积累和tau蛋白过度磷酸化。苔藓虫素-1作为中枢神经系统药物的治疗潜力有待进一步挖掘。
{"title":"Bryostatin-1: Pharmacology and Therapeutic Potential as a CNS Drug","authors":"Miao-Kun Sun,&nbsp;Daniel L. Alkon","doi":"10.1111/j.1527-3458.2006.00001.x","DOIUrl":"10.1111/j.1527-3458.2006.00001.x","url":null,"abstract":"<p>Bryostatin-1 is a powerful protein kinase C (PKC) agonist, activating PKC isozymes at nanomolar concentrations. Pharmacological studies of bryostatin-1 have mainly been focused on its action in preventing tumor growth. Emerging evidence suggests, however, that bryostatin-1 exhibits additional important pharmacological activities. In preclinical studies bryostatin-1 has been shown at appropriate doses to have cognitive restorative and antidepressant effects. The underlying pharmacological mechanisms may involve an activation of PKC isozymes, induction of synthesis of proteins required for long-term memory, restoration of stress-evoked inhibition of PKC activity, and reduction of neurotoxic amyloid accumulation and tau protein hyperphosphorylation. The therapeutic potential of bryostatin-1 as a CNS drug should be further explored.</p>","PeriodicalId":94307,"journal":{"name":"CNS drug reviews","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2006-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1111/j.1527-3458.2006.00001.x","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"26139673","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 95
Pharmacological Profile of the Selective FAAH Inhibitor KDS-4103 (URB597) 选择性FAAH抑制剂KDS-4103 (URB597)的药理作用
Pub Date : 2006-07-10 DOI: 10.1111/j.1527-3458.2006.00021.x
Daniele Piomelli, Giorgio Tarzia, Andrea Duranti, Andrea Tontini, Marco Mor, Timothy R. Compton, Olivier Dasse, Edward P. Monaghan, Jeff A. Parrott, David Putman

In the present article, we review the pharmacological properties of KDS-4103 (URB597), a highly potent and selective inhibitor of the enzyme fatty-acid amide hydrolase (FAAH), which catalyzes the intracellular hydrolysis of the endocannabinoid anandamide. In vitro, KDS-4103 inhibits FAAH activity with median inhibitory concentrations (IC50) of 5 nM in rat brain membranes and 3 nM in human liver microsomes. In vivo, KDS-4103 inhibits rat brain FAAH activity after intraperitoneal (i.p.) administration with a median inhibitory dose (ID50) of 0.15 mg/kg. The compound does not significantly interact with other cannabinoid-related targets, including cannabinoid receptors and anandamide transport, or with a broad panel of receptors, ion channels, transporters and enzymes. By i.p. administration to rats and mice KDS-4103 elicits significant, anxiolytic-like, antidepressant-like and analgesic effects, which are prevented by treatment with CB1 receptor antagonists. By contrast, at doses that significantly inhibit FAAH activity and substantially raise brain anandamide levels, KDS-4103 does not evoke classical cannabinoid-like effects (e.g., catalepsy, hypothermia, hyperphagia), does not cause place preference, and does not produce generalization to the discriminative effects of the active ingredient of cannabis, Δ9-tetrahydrocannabinol (Δ9-THC). These findings suggest that KDS-4103 acts by enhancing the tonic actions of anandamide on a subset of CB1 receptors, which may normally be engaged in controlling emotions and pain. KDS-4103 is orally available in rats and cynomolgus monkeys. Sub-chronic repeated dose studies (1500 mg/kg, per os) in these two species have not demonstrated systemic toxicity. Likewise, no toxicity was noted in bacterial cytotoxicity tests in vitro and in the Ames test. Furthermore, no deficits were observed in rats on the rotarod test after acute i.p. treatment with KDS-4103 at doses up to 5 mg/kg or in a functional observation battery after oral doses up to 1500 mg/kg. The results suggest that KDS-4103 will offer a novel approach with a favorable therapeutic window for the treatment of anxiety, depression and pain.

在这篇文章中,我们回顾了KDS-4103 (URB597)的药理学性质,它是一种高效的选择性脂肪酸酰胺水解酶(FAAH)抑制剂,催化细胞内内源性大麻素anandamide的水解。在体外,KDS-4103抑制FAAH活性的中位抑制浓度(IC50)在大鼠脑膜和人肝微粒体中分别为5 nM和3 nM。在体内,KDS-4103腹腔内给药后抑制大鼠脑FAAH活性,中位抑制剂量(ID50)为0.15 mg/kg。该化合物与其他大麻素相关靶点,包括大麻素受体和大麻酰胺运输,或与广泛的受体、离子通道、转运体和酶不显着相互作用。KDS-4103经大鼠和小鼠腹腔注射可引起显著的抗焦虑、抗抑郁和镇痛作用,而CB1受体拮抗剂可阻止这些作用。相比之下,在显著抑制FAAH活性并显著提高脑内anandamide水平的剂量下,KDS-4103不会引起经典的大麻素样效应(例如,嗜睡、体温过低、嗜食),不会引起位置偏好,也不会对大麻活性成分的鉴别作用产生普遍性,Δ9-tetrahydrocannabinol (Δ9-THC)。这些发现表明,KDS-4103通过增强anandamide对CB1受体亚群的强直作用而起作用,而CB1受体通常参与控制情绪和疼痛。KDS-4103可口服给大鼠和食蟹猴。对这两个物种进行的亚慢性重复给药研究(1500mg /kg / s)未显示出全身毒性。同样,体外细菌细胞毒性试验和Ames试验均未发现毒性。此外,大鼠在5mg /kg剂量的KDS-4103急性腹腔治疗后,或口服1500mg /kg剂量的KDS-4103功能观察电池中,在rottarod试验中未观察到缺陷。结果表明,KDS-4103将为焦虑、抑郁和疼痛的治疗提供一种新的途径和良好的治疗窗口。
{"title":"Pharmacological Profile of the Selective FAAH Inhibitor KDS-4103 (URB597)","authors":"Daniele Piomelli,&nbsp;Giorgio Tarzia,&nbsp;Andrea Duranti,&nbsp;Andrea Tontini,&nbsp;Marco Mor,&nbsp;Timothy R. Compton,&nbsp;Olivier Dasse,&nbsp;Edward P. Monaghan,&nbsp;Jeff A. Parrott,&nbsp;David Putman","doi":"10.1111/j.1527-3458.2006.00021.x","DOIUrl":"10.1111/j.1527-3458.2006.00021.x","url":null,"abstract":"<p>In the present article, we review the pharmacological properties of KDS-4103 (URB597), a highly potent and selective inhibitor of the enzyme fatty-acid amide hydrolase (FAAH), which catalyzes the intracellular hydrolysis of the endocannabinoid anandamide. In vitro, KDS-4103 inhibits FAAH activity with median inhibitory concentrations (IC<sub>50</sub>) of 5 nM in rat brain membranes and 3 nM in human liver microsomes. In vivo, KDS-4103 inhibits rat brain FAAH activity after intraperitoneal (i.p.) administration with a median inhibitory dose (ID<sub>50</sub>) of 0.15 mg/kg. The compound does not significantly interact with other cannabinoid-related targets, including cannabinoid receptors and anandamide transport, or with a broad panel of receptors, ion channels, transporters and enzymes. By i.p. administration to rats and mice KDS-4103 elicits significant, anxiolytic-like, antidepressant-like and analgesic effects, which are prevented by treatment with CB1 receptor antagonists. By contrast, at doses that significantly inhibit FAAH activity and substantially raise brain anandamide levels, KDS-4103 does not evoke classical cannabinoid-like effects (e.g., catalepsy, hypothermia, hyperphagia), does not cause place preference, and does not produce generalization to the discriminative effects of the active ingredient of cannabis, Δ9-tetrahydrocannabinol (Δ9-THC). These findings suggest that KDS-4103 acts by enhancing the tonic actions of anandamide on a subset of CB<sub>1</sub> receptors, which may normally be engaged in controlling emotions and pain. KDS-4103 is orally available in rats and cynomolgus monkeys. Sub-chronic repeated dose studies (1500 mg/kg, per os) in these two species have not demonstrated systemic toxicity. Likewise, no toxicity was noted in bacterial cytotoxicity tests in vitro and in the Ames test. Furthermore, no deficits were observed in rats on the rotarod test after acute i.p. treatment with KDS-4103 at doses up to 5 mg/kg or in a functional observation battery after oral doses up to 1500 mg/kg. The results suggest that KDS-4103 will offer a novel approach with a favorable therapeutic window for the treatment of anxiety, depression and pain.</p>","PeriodicalId":94307,"journal":{"name":"CNS drug reviews","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2006-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1111/j.1527-3458.2006.00021.x","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"26140608","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 364
Pharmacological Characterization of DM232 (Unifiram) and DM235 (Sunifiram), New Potent Cognition Enhancers 新型认知增强剂DM232 (Unifiram)和DM235 (Sunifiram)的药理特性
Pub Date : 2006-07-10 DOI: 10.1111/j.1527-3458.2006.00039.x
M. N. Romanelli, N. Galeotti, C. Ghelardini, D. Manetti, E. Martini, F. Gualtieri

DM232 (unifiram) and DM235 (sunifiram) are potent cognition-enhancers, which are four order of magnitude more potent than piracetam. These compounds, although not showing affinity in binding studies for the most important central receptors or channels, are able to prevent amnesia induced by modulation of several neurotransmission systems. These compounds are able to increase the release of acetylcholine from rat cerebral cortex, and, as far as unifiram is concerned, to increase the amplitude of fEPSP in rat hippocampal slices. In vitro experiments, performed on hippocampal slices, also supported the hypothesis of a role of the AMPA receptors for the cognition-enhancing properties of unifiram and sunifiram.

DM232 (unifiram)和DM235 (sunfiram)是有效的认知增强剂,其效力比吡拉西坦强4个数量级。这些化合物,虽然在结合研究中没有显示出对最重要的中枢受体或通道的亲和力,但能够防止由几种神经传递系统的调节引起的健忘症。这些化合物能够增加大鼠大脑皮层乙酰胆碱的释放,并且就unifiram而言,可以增加大鼠海马切片中fEPSP的振幅。在海马体切片上进行的体外实验也支持AMPA受体在unifiram和sununifiram的认知增强特性中发挥作用的假设。
{"title":"Pharmacological Characterization of DM232 (Unifiram) and DM235 (Sunifiram), New Potent Cognition Enhancers","authors":"M. N. Romanelli,&nbsp;N. Galeotti,&nbsp;C. Ghelardini,&nbsp;D. Manetti,&nbsp;E. Martini,&nbsp;F. Gualtieri","doi":"10.1111/j.1527-3458.2006.00039.x","DOIUrl":"10.1111/j.1527-3458.2006.00039.x","url":null,"abstract":"<p>DM232 (unifiram) and DM235 (sunifiram) are potent cognition-enhancers, which are four order of magnitude more potent than piracetam. These compounds, although not showing affinity in binding studies for the most important central receptors or channels, are able to prevent amnesia induced by modulation of several neurotransmission systems. These compounds are able to increase the release of acetylcholine from rat cerebral cortex, and, as far as unifiram is concerned, to increase the amplitude of fEPSP in rat hippocampal slices. In vitro experiments, performed on hippocampal slices, also supported the hypothesis of a role of the AMPA receptors for the cognition-enhancing properties of unifiram and sunifiram.</p>","PeriodicalId":94307,"journal":{"name":"CNS drug reviews","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2006-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1111/j.1527-3458.2006.00039.x","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"26140609","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 17
期刊
CNS drug reviews
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1