Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Med Chem ; 33(9): 2421-9, 1990 Sep.
Article in English | MEDLINE | ID: mdl-1975275

ABSTRACT

With an eye toward the development of novel atypical antipsychotic agents, we have studied the structure-affinity relationships of N,N'-di-o-tolylguanidine (DTG, 3) and its congeners at the haloperidol-sensitive sigma receptor. A number of DTG analogues were synthesized and evaluated in in vitro radioligand displacement experiments with guinea pig brain membrane homogenates, using the highly sigma-specific radioligands [3H]-3 and [3H]-(+)-3-(3-hydroxyphenyl)-N-(1-propyl)piperidine and the phencyclidine (PCP) receptor specific compounds [3H]-N-[1-(2-thienyl)-cyclohexyl]piperidine and [3H]-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10- imine. The affinity of N,N'-diarylguanidines for the sigma receptor decreases with increasing steric bulk of ortho substituents larger than C2H5. Hydrophobic substituents are generally preferred over similarly positioned hydrophilic ones. Furthermore, electroneutral substituents are preferred over strongly electron donating or withdrawing groups. Significant binding to the sigma receptor is usually retained as long as at least one side of the guanidine bears a preferred group (e.g. 2-CH3C6H5). Replacement of one or both aryl rings with certain saturated carbocycles (e.g. cyclohexyl, norbornyl, or adamantyl) leads to a significant increase in affinity. By combining the best aromatic and best saturated carbocyclic substituents in the same molecule, we arrived at some of the most potent sigma ligands described to date (e.g. N-exo-2-norbornyl-N'-(2-iodophenyl)guanidine, IC50 = 3 nM vs [3H]-3). All of the compounds tested were several orders of magnitude more potent at the sigma receptor than at the PCP receptor, with a few notable exceptions. This series of disubstituted guanidines may be of value in the development of potential antipsychotics and in the further pharmacological and biochemical characterization of the sigma receptor.


Subject(s)
Antipsychotic Agents/chemical synthesis , Guanidines/chemical synthesis , Animals , Antipsychotic Agents/pharmacology , Chemical Phenomena , Chemistry , Guanidines/pharmacology , Guinea Pigs , Haloperidol/pharmacology , Radioligand Assay , Receptors, Opioid/drug effects , Receptors, sigma , Structure-Activity Relationship
2.
J Med Chem ; 37(2): 260-7, 1994 Jan 21.
Article in English | MEDLINE | ID: mdl-8295213

ABSTRACT

Diarylguanidines, acting as NMDA receptor ion channel site ligands, represent a new class of potential neuroprotective drugs. Several diarylguanidines structurally related to N,N'-di-o-tolylguanidine (DTG), a known selective sigma receptor ligand, were synthesized and evaluated in in vitro radioligand displacement assays, with rat or guinea pig brain membrane homogenates, using the NMDA receptor ion channel site specific radioligand [3H]-(+)-5(S)-methyl-10(R),11-dihydro-5H-dibenzo[a,d]cyclohepten-5 ,10- imine (MK-801, 3), and the sigma receptor-specific radioligand [3H]-di-o-tolylguanidine (DTG, 5). This paper presents the structure-activity relationships leading to novel tri- and tetrasubstituted guanidines, which exhibit high selectivity for NMDA receptor ion channel sites and weak or negligible affinity for sigma receptors. The in vitro binding results from symmetrically substituted diphenylguanidines indicated that compounds having ortho or meta substituents (with respect to the position of the guanidine nitrogen) on the phenyl rings showed greater affinity for the NMDA receptor ion channel site compared with para-substituted derivatives. Among the group of ring substituents studied for symmetrical diarylguanidines, an isopropyl group was preferred at the ortho position and an ethyl group was preferred at the meta position. Several unsymmetrical guanidines containing a naphthalene ring on one nitrogen atom and an ortho- or a meta-substituted phenyl ring on the second nitrogen atom, e.g., N-1-naphthyl-N'-(3-ethylphenyl)guanidine (36), showed a 3-5-fold increase in affinity for the NMDA receptor ion channel site and no change in sigma receptor affinity compared to the respective symmetrical counterparts. Additional small substituents on the guanidine nitrogen atoms bearing the aryl rings resulted in tri- and tetrasubstituted guanidine derivatives which retained affinity for NMDA receptor ion channel sites but exhibited a significant reduction in their affinities for sigma receptors. For example, N-1-naphthyl-N'-(3-ethylphenyl)-N'-methylguanidine (40) showed high affinity for the NMDA receptor ion channel site (IC50 = 36 nM vs [3H]-3) and low affinity for sigma receptors (IC50 = 2540 nM vs [3H]-5). Selectivity for the NMDA receptor ion channel sites over sigma receptors appears to be dependent upon the structure of the additional substituents on the guanidine nitrogen atoms bearing the aryl groups. Methyl and ethyl substituents are most preferred in the tri- and tetrasubstituted diarylguanidines. The trisubstituted guanidine, N-1-naphthyl-N'-(3-ethylphenyl)-N'-methylguanidine (40) and its close analogues showed good in vivo neuroprotection and are potential neuroprotective drug candidates for the treatment of stroke and other neurodegenerative disorders.


Subject(s)
Guanidines/chemical synthesis , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Brain/drug effects , Guanidines/pharmacology , Guinea Pigs , In Vitro Techniques , Radioligand Assay , Rats , Structure-Activity Relationship
3.
J Med Chem ; 41(17): 3298-302, 1998 Aug 13.
Article in English | MEDLINE | ID: mdl-9703475

ABSTRACT

Synthesis and structure-activity relationships (SAR) are described for a series of N,N'-diarylguanidines related to N-acenaphth-5-yl-N'-(4-methoxynaphth-1-yl)guanidine (3) as anticonvulsants through blockade of sodium channels. SAR studies on compound 3 led to several simpler diphenylguanidines with improved in vitro and in vivo activity. Compounds were screened for blockade of sodium channels in a veratridine-induced [14C]guanidinium influx assay (type IIA sodium channels) and for anticonvulsant activity in the audiogenic DBA/2 mouse model. Results indicated that N, N'-diphenylguanidines substituted with flexible and moderate size lipophilic groups were preferred over aryl and/or hydrophilic groups for biological activity. Among the compounds studied, n-butyl- and/or n-butoxy-containing guanidines showed superior biological activity. A possible relationship between in vitro and in vivo activity of this compound series and their measured/calculated lipophilicities was investigated. Compounds of this series showed only weak NMDA ion channel-blocking activity indicating that the anticonvulsant activity of these compounds is unlikely to be mediated by NMDA ion channels but, more likely, by acting at voltage-gated sodium channels.


Subject(s)
Anticonvulsants/chemical synthesis , Guanidines/chemical synthesis , Seizures/prevention & control , Sodium Channel Blockers , Acoustic Stimulation , Animals , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , CHO Cells , Cell Line , Cricetinae , Drug Design , Guanidine/metabolism , Guanidines/chemistry , Guanidines/pharmacology , Mice , Mice, Inbred DBA , Molecular Structure , Piperazines/chemistry , Piperazines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Riluzole/chemistry , Riluzole/pharmacology , Structure-Activity Relationship , Veratridine/pharmacology
4.
J Med Chem ; 36(14): 1938-46, 1993 Jul 09.
Article in English | MEDLINE | ID: mdl-8101572

ABSTRACT

IDDC (3, 10,5-(iminomethano)-10,11-dihydro-5H-dibenzo[a,d]cycloheptene++ +) and a series of substituted derivatives were synthesized and evaluated in vitro for their ability to displace tritiated MK-801 ([3H]-2) from its specific binding site in guinea pig brain homogenate. Substitution at the 3-position of 3 with bromine, chlorine, and fluorine led to increased binding affinity. In contrast, substitution of donor groups at the 3-position gave decreased binding affinities, as did all substitutions at the 7-position and on nitrogen. Where racemic mixtures were resolved, the (+)-optical antipodes were more active than their enantiomers or racemates. The most active ligand found in this study was (+)-13e (IC50 = 15.5 +/- 4.5 nM). The affinity of (+)-13e for the PCP receptor makes it among the most potent ligands known. In vitro neuroprotection was demonstrated by 3, (+)-3, and (+)-6 (N-Me-IDDC) against glutamate-induced cell death in rat hippocampal cells.


Subject(s)
Dibenzocycloheptenes/chemical synthesis , Dibenzocycloheptenes/pharmacology , Receptors, Phencyclidine/drug effects , Animals , Binding Sites , Brain/drug effects , Brain/metabolism , Cells, Cultured , Dibenzocycloheptenes/chemistry , Dizocilpine Maleate/metabolism , Excitatory Amino Acid Antagonists , Glutamates/toxicity , Glutamic Acid , Guinea Pigs , Rats , Rats, Sprague-Dawley , Receptors, Phencyclidine/metabolism , Stereoisomerism , Structure-Activity Relationship
5.
J Neurotrauma ; 9 Suppl 2: S531-43, 1992 May.
Article in English | MEDLINE | ID: mdl-1319500

ABSTRACT

Ischemic insults to the brain in stroke or traumatic brain injury produce excessive release of glutamate from depolarized nerve terminals. This excessive glutamate release in turn stimulates massive calcium entry into nerve cells, activating a biochemical cascade that results in cell death. A major pathway of calcium entry into depolarized nerve cells is through voltage-sensitive, high threshold calcium channels. A large fraction of this calcium entry is mediated through "R-type" calcium channels, channels resistant to blockage by dihydropyridine calcium antagonists such as nimodipine. A newly discovered compound derived from spider venom, CNS 2103, antagonizes both R-type channels and dihydropyridine-sensitive ("L-type") calcium channels. This broad spectrum of action, coupled with selectivity for calcium channels over other classes of voltage-sensitive and ligand-gated ion channels, makes CNS 2103 an interesting lead for development of drugs to treat ischemic brain injury. Activation of presynaptic ("N-type") calcium channels in nerve terminals is a primary cause of excessive neurotransmitter release in brain ischemia. Prevention of glutamate release by blockade of N-type channels in glutamatergic nerve terminals may, at an early stage in the pathophysiological cascade, abort the process leading to nerve cell death. Cambridge NeuroScience has developed a novel rapid kinetic approach for monitoring glutamate release from brain nerve terminals in vitro, and this has led to CNS 1145, a substituted guanidine that selectively blocks a kinetic component of calcium-dependent glutamate release mediated by persistent depolarization. Additional evidence suggests that CNS 1145 antagonizes presynaptic N-type calcium channels, and this may account at least in part for its ability to block glutamate release.


Subject(s)
Brain Injuries/drug therapy , Brain Ischemia/drug therapy , Calcium Channel Blockers/therapeutic use , Calcium Channels/physiology , Calcium/metabolism , Central Nervous System/physiology , Cerebrovascular Disorders/drug therapy , Neurons/physiology , Animals , Brain Injuries/physiopathology , Brain Ischemia/physiopathology , Calcium Channels/drug effects , Cell Death , Cerebrovascular Disorders/physiopathology , Humans , Neurons/cytology , Neurons/pathology , Synapses/drug effects , Synapses/physiology
6.
Ann N Y Acad Sci ; 765: 210-29, 1995 Sep 15.
Article in English | MEDLINE | ID: mdl-7486608

ABSTRACT

We have originated a family of N,N'-disubstituted guanidines that block the voltage-activated Ca2+ and Na+ channels governing glutamate release. These compounds, CNS 1237 (N-acenaphthyl-N'-methoxynaphthyl guanidine) and its analogues, are "use dependent" in their ability to attenaute neurotransmitter release: they block glutamate release with greater efficacy under conditions of persistent or repetitive depolarization, as would be encountered under pathophysiological circumstances, relative to their ability to block glutamate release elicited by brief, transient depolarizations more characteristic of normal physiological release events in nonischemic brain. Using electrophysiological and rapid kinetic methods, we have differentiated the use-dependent block of the relevant Na+ and Ca2+ channels governing neurotransmitter release from the mechanism of channel antagonism exhibited by, respectively, the substituted guanidine Na+ channel blocker tetrodotoxin (TTX) and venom peptide Ca2+ antagonists. To characterize use-dependent Na+ channel block by CNS 1237, we have employed whole-cell voltage-clamp recordings from a Chinese hamster ovary (CHO) cell line expressing cloned mammalian type II Na+ channels. These experiments demonstrated that, in contrast to the actions of TTX under the same conditions, the potency of Na+ channel block by CNS 1237 is greatly enhanced by depolarizing stimuli in a frequency-dependent manner. Ca2+ channel-activated glutamate release from brain nerve terminal preparations was measured with approximately 300 msec time resolution over a 5-second period of high K(+)-depolarization, using a rapid superfusion technique. CNS 1237 and analogues, at 1-3 microM, accelerated the decay of glutamate release by 40-70%, reflecting depolarization-induced enhancement of block. In contrast, blockade of glutamate release by the Ca2+ channel antagonist peptide toxins omega-aga IV-A (from spider venom) and omega-conotoxin M-VII-C (from cone snail venom) exhibited "reverse-use-dependence:" at concentrations of 0.3 microM, which blocked the initial amplitude of glutamate release by 40-60%, the decay time constant for glutamate release was significantly increased, indicating depolarization-induced relief of block. These findings establish that CNS 1237 and other members of this compound series are use-dependent blockers of the voltage-activated ion channels governing glutamate release. Studies of CNS 1237 in the rat middle cerebral artery occlusion (MCAO) focal stroke model have indicated infarct size reduction comparable to that observed by the same investigators for the glutamate release blocker (BW 619C89 (Burroughs-Wellcome, now in clinical development). Maximal infarct size reduction is achieved with a 3-mg/kg bolus followed by a 4-hour infusion of 0.75 mg/kg/hr.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Brain/physiology , Calcium Channel Blockers/pharmacology , Glutamic Acid/metabolism , Guanidines/pharmacology , Ischemic Attack, Transient/prevention & control , Neuroprotective Agents/pharmacology , Receptors, N-Methyl-D-Aspartate/physiology , Sodium Channel Blockers , Animals , Blood Pressure/drug effects , Brain/drug effects , Brain/physiopathology , CHO Cells , Cricetinae , Electrophysiology/methods , Heart Rate/drug effects , Ischemic Attack, Transient/physiopathology , Kinetics , Neurotransmitter Uptake Inhibitors/pharmacology , Piperazines/pharmacology , Pyrimidines/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/drug effects , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Sodium Channels/physiology
8.
J Pharmacol Exp Ther ; 271(2): 1080-5, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7525936

ABSTRACT

Radioligand binding techniques were used to determine the affinity of a series of substituted guanidine derivatives for 1) the binding site within the ion channel of the N-methyl-D-aspartate (NMDA) receptor, as defined by displacement of MK-801 ([3H]dizocilpine) and 2) sigma sites as defined by displacement of [3H]N,N'-di-(o-tolyl)guanidine. The goal was to find ligands with high affinity and selectivity for the NMDA receptor ion-channel site. The neuroprotective activity of these compounds was assessed by their ability to protect cortical neurons from injury caused by a 5-min exposure to 500 microM glutamate in vitro. Release of lactate dehydrogenase into the culture medium by damaged neurons was used as an index of neuronal injury. The 14 compounds tested had IC50 values ranging from 37.3 nM to 12.7 microM for the NMDA receptor ion-channel site and from 8.3 nM to 7.25 microM for sigma sites. Affinity for the ion-channel site was improved by unsymmetrical substitutions on the guanidine moiety. All compounds in the series protected cortical neurons against glutamate toxicity, with EC50 values (concentration affording 50% protection) ranging from 0.38 to 28.25 microM. The neuroprotective effect of each compound was positively correlated with its ion-channel site affinity (r = 0.94); no correlation between neuroprotective efficacy and sigma site binding affinity was found (r V -0.13) establishing clearly that neuroprotection in this assay was linked to NMDA antagonist properties.


Subject(s)
Guanidines/pharmacology , Ion Channels/metabolism , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, sigma/metabolism , Animals , Cells, Cultured , Dizocilpine Maleate/metabolism , Glutamic Acid/toxicity , Guanidines/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Structure-Activity Relationship
9.
Bioconjug Chem ; 4(6): 545-8, 1993.
Article in English | MEDLINE | ID: mdl-8305523

ABSTRACT

The enhanced photolabeling properties of chlorinated phenyl azides are demonstrated by the synthesis and photolysis of methyl 4-azido-2,3,5,6-tetrachlorobenzoate (3) and methyl 4-azido-3,5-dichlorobenzoate (4). Photolysis of azide 3 in 1 M diethylamine/cyclohexane as the trapping medium gave 34% NH-insertion product. Similar photolysis of azide 4 gave 35% NH insertion product. These results demonstrate that chlorinated phenyl azides are significantly better at undergoing NH insertion than nonhalogenated analogs and suggest that improvement of existing aryl azide-based photolabels might be achieved by introduction of chlorine atoms on either side of the azide group. As an application, 3-azido-2,4-dichloro-10,5-(iminomethano)-10,11-dihydro-5H- dibenzo[a,d]cycloheptene (19), an analog of the potent PCP receptor ligand IDDC (14), was synthesized and its affinity for the PCP receptor was determined to be 6.3 +/- 0.7 microM (IC50 against [3H]MK801).


Subject(s)
Affinity Labels/chemical synthesis , Azides/chemical synthesis , Hydrocarbons, Chlorinated/chemical synthesis , Receptors, Phencyclidine/metabolism , Affinity Labels/metabolism , Animals , Central Nervous System/ultrastructure , Ligands
SELECTION OF CITATIONS
SEARCH DETAIL