Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Pharm ; 604: 120740, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34062232

ABSTRACT

Oxaliplatin palmitate acetate (OPA), a platinum (IV) oxaliplatin derivative, was previously designed with the aim to improve the platinum-based anti-cancer therapy. In this work, we further explore the potential of OPA in extensive in vitro and in vivo studies. OPA in pancreatic (BxPC3-luc), lung (NCI-H1993) and liver (Hep3B) cancer cell lines showed a higher toxicity in comparison to oxaliplatin. The in vitro release kinetic experiments of OPA from the nanoparticles (NPs) under sink conditions exhibited a very rapid profile. Furthermore, OPA cannot be considered a prodrug of oxaliplatin, based on the OPA intact molecule pharmacokinetic profile study in rats. The formation of oxaliplatin from the biodegradation of OPA ranges only from 5% to 7% and both drugs were rapidly eliminated from the plasma. Pharmacokinetics of OPA PLGA nanoparticles in mice showed that nanoparticles failed to prolong the release of OPA in the plasma and did not add any therapeutic benefit over OPA solution, as suggested by the rapid in vitro release of OPA from nanoparticles. In pancreatic xenograft BxPC3-luc cancer model, both OPA in solution and OPA nanoparticles inhibited the tumor growth, equally and significantly, as compared to oxaliplatin. In liver xenograft Hep3B cancer model, OPA solution and cisplatin demonstrated good and similar antitumor efficacy. In lung xenograft NCI-H1993 cancer model, OPA solution, with a significant antitumor efficacy, was superior to cisplatin, which did not differ from the vehicle. In conclusion, OPA may offer a promising advance in platinum-based chemotherapy against various forms of cancers in an adequate dose and schedule.


Subject(s)
Antineoplastic Agents , Palmitates , Acetates , Animals , Cisplatin , Mice , Oxaliplatin , Rats , Tissue Distribution
2.
Oncotarget ; 6(27): 24560-70, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26009994

ABSTRACT

PURPOSE: The miniature biodegradable implant siG12D-LODER™ was inserted into a tumor and released a siRNA drug against KRAS(G12D) along four months. This novel siRNA based drug was studied, in combination with chemotherapy, as targeted therapy for Locally Advanced Pancreatic Cancer (LAPC). METHODS: An open-label Phase 1/2a study in the first-line setting of patients with non-operable LAPC was initiated. In this study patients were assigned to receive a single dose of siG12D-LODERs, in three escalating dose cohorts (0.025mg, 0.75mg and 3.0mg). Gemcitabine was given on a weekly basis, following the siG12D-LODERTM insertion, until disease progression. The recommended dose was further examined with modified FOLFIRINOX. The follow up period was eight weeks and survival until death. RESULTS: Fifteen patients with LAPC were enrolled. Among the 15 treated patients, the most frequent adverse events observed were grade 1or 2 in severity (89%); five patients experienced serious adverse events (SAEs). In 12 patients analyzed by CT scans, none showed tumor progression, the majority (10/12) demonstrated stable disease and two showed partial response. Decrease in tumor marker CA19-9 was observed in 70% (7/10) of patients. Median overall survival was 15.12 months; 18 month survival was 38.5%. CONCLUSIONS: The combination of siG12D-LODER™ and chemotherapy is well tolerated, safe and demonstrated a potential efficacy in patients with LAPC. NCT01188785.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Drug Implants , Molecular Targeted Therapy , Pancreatic Neoplasms/therapy , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , RNA, Small Interfering , RNAi Therapeutics/methods , Absorbable Implants , Aged , Aged, 80 and over , Antigens, Neoplasm/blood , Antineoplastic Agents/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Combined Modality Therapy , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Disease-Free Survival , Female , Fluorouracil/therapeutic use , Humans , Irinotecan , Leucovorin/therapeutic use , Male , Middle Aged , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins p21(ras)/genetics , Gemcitabine
3.
Epilepsia ; 55(2): 353-61, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24313671

ABSTRACT

OBJECTIVE: Valnoctamide (VCD), a central nervous system (CNS)-active chiral constitutional isomer of valpromide, the corresponding amide of valproic acid (VPA), is currently undergoing phase IIb clinical trials in acute mania. VCD exhibits stereoselective pharmacokinetics (PK) in animals and humans. The current study comparatively evaluated the pharmacodynamics (PD; anticonvulsant activity and teratogenicity) and PK of the four individual stereoisomers of VCD. METHODS: The anticonvulsant activity of VCD individual stereoisomers was evaluated in several rodent anticonvulsant models including maximal electroshock, 6 Hz psychomotor, subcutaneous metrazol, and the pilocarpine-induced and soman-induced status epilepticus (SE). The PK-PD (anticonvulsant activity) relationship of VCD stereoisomers was evaluated following intraperitoneal administration (70 mg/kg) to rats. Induction of neural tube defects (NTDs) by VCD stereoisomers was evaluated in a mouse strain that was highly susceptible to teratogen-induced NTDs. RESULTS: VCD had a stereoselective PK, with (2S,3S)-VCD exhibiting the lowest clearance, and consequently a twice-higher plasma exposure than all other stereoisomers. Nervertheless, there was less stereoselectivity in VCD anticonvulsant activity and each stereoisomer had similar median effective dose (ED)50 values in most models. VCD stereoisomers (258 or 389 mg/kg) did not cause NTDs. These doses are 3-12 times higher than VCD anticonvulsant ED50 values. SIGNIFICANCE: VCD displayed stereoselective PK that did not lead to significant stereoselective activity in various anticonvulsant rodent models. If VCD exerted its broad-spectrum anticonvulsant activity using a single mechanism of action (MOA), it is likely that it would exhibit a stereoselective PD. The fact that there was no significant difference between racemic VCD and its individual stereoisomers suggests that VCD's anticonvulsant activity is due to multiple MOAs.


Subject(s)
Amides/pharmacokinetics , Anticonvulsants/pharmacokinetics , Central Nervous System Stimulants/pharmacokinetics , Teratogens/pharmacokinetics , Valproic Acid/pharmacokinetics , Amides/chemistry , Amides/toxicity , Animals , Anticonvulsants/chemistry , Anticonvulsants/toxicity , Central Nervous System Stimulants/chemistry , Central Nervous System Stimulants/toxicity , Male , Mice , Neural Tube Defects/chemically induced , Random Allocation , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/prevention & control , Stereoisomerism , Teratogens/chemistry , Teratogens/toxicity , Valproic Acid/chemistry , Valproic Acid/toxicity
4.
Birth Defects Res B Dev Reprod Toxicol ; 98(4): 318-27, 2013 Aug.
Article in English | MEDLINE | ID: mdl-24039104

ABSTRACT

BACKGROUND: Valproic acid (VPA), widely used to treat epilepsy, bipolar disorders, and migraine prophylaxis, is known to cause neural tube and skeletal defects in humans and animals. Aminobenzensulfonamide derivatives of VPA with branched aliphatic carboxylic acids, namely 2-methyl-N-(4-sulfamoyl-phenyl)-pentanamide (MSP), 2-ethyl-N-(4-sulfamoyl-phenyl)-butyramide (ESB), 2-ethyl-4-methyl-N-(4-sulfamoyl-phenyl)-pentanamide (EMSP), and 2-ethyl-N-(4-sulfamoyl-benzyl)-butyramide (ESBB), have shown more potent anticonvulsant activity than VPA in preclinical testing. Here, we investigated the teratogenic effects of these analogous compounds of VPA in NMRI mice. METHODS: Pregnant NMRI mice were given a single subcutaneous injection of either VPA at 1.8 or 3.6 mmol/kg, or MSP, ESB, EMSP, or ESBB at 1.8, 3.6, or 4.8 mmol/kg on gestation day (GD) 8. Cesarean section was performed on GD 18, and the live fetuses were examined for external and skeletal malformations. RESULTS: Compared with VPA, which induced neural tube defects (NTDs) in fetuses at 1.8 and 3.6 mmol/kg, the analog derivatives induced no NTDs at dose levels up to 4.8 mmol/kg (except for a single case of exencephaly at 4.8 mmol/kg MSP). Skeletal examination showed several abnormalities mainly at the axial skeletal level with VPA at 1.8 mmol/kg. Fused vertebrae and/or fused ribs were also observed with MSP, ESB, EMSP, and ESBB, they were less severe and seen at a lower incidence that those induced by VPA at the same dose level. CONCLUSIONS: In addition to exerting more potent preclinical antiepileptic activity, teratology comparison indicates that aminobenzensulfonamide analogs are generally more weakly teratogenic than VPA.


Subject(s)
Carboxylic Acids/toxicity , Congenital Abnormalities/pathology , Fatty Acids/toxicity , Sulfanilamides/toxicity , Sulfonamides/toxicity , Animals , Body Weight/drug effects , Bone and Bones/abnormalities , Bone and Bones/drug effects , Bone and Bones/pathology , Carboxylic Acids/chemistry , Congenital Abnormalities/embryology , Embryo, Mammalian/abnormalities , Embryo, Mammalian/drug effects , Fatty Acids/chemistry , Female , Mice , Neural Tube Defects/chemically induced , Neural Tube Defects/embryology , Neural Tube Defects/pathology , Pregnancy , Sulfanilamide , Sulfanilamides/chemistry , Sulfonamides/chemistry , Teratology , Valproic Acid/analogs & derivatives , Valproic Acid/chemistry , Valproic Acid/toxicity
5.
Epilepsia ; 54 Suppl 6: 99-102, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24001086

ABSTRACT

sec-Butyl-propylacetamide (SPD) is a one-carbon homolog of valnoctamide (VCD), a chiral constitutional isomer of valproic acid's (VPA) corresponding amide valpromide. VCD has potential as a therapy in epilepsy including status epilepticus (SE) and neuropathic pain, and is currently being developed for the treatment of bipolar disorder. Both VCD and SPD possess two stereogenic carbons in their chemical structure. SPD possesses a unique and broad-spectrum antiseizure profile superior to that of valproic acid (VPA) and better than that of VCD. In addition SPD blocked behavioral- and electrographic-SE induced by pilocarpine and soman (organophosphate nerve gas) and afforded in vivo neuroprotection that was associated with cognitive sparing. VCD has activity similar to that of SPD in pilocarpine-induced status epilepticus (SE), although at higher doses. The activity of SPD and VCD against SE is superior to that of diazepam in terms of rapid onset, potency, and ability to block SE when given 20-60 min after seizure onset. When administered 20 and 40 min after SE onset, SPD (100-174 mg/kg) produced long-lasting efficacy (e.g., 4-8 h) against soman-induced convulsive and electrographic SE in both rats and guinea pigs. SPD activity in the pilocarpine and soman-induced SE models when administered 20-60 min after seizure onset, differentiates SPD from benzodiazepines and all other antiepileptic drugs .


Subject(s)
Amides/therapeutic use , Anticonvulsants/therapeutic use , Seizures/drug therapy , Status Epilepticus/drug therapy , Valproic Acid/analogs & derivatives , Acute Disease , Amides/chemistry , Animals , Disease Models, Animal , Guinea Pigs , Humans , Rats , Seizures/chemically induced , Status Epilepticus/chemically induced , Treatment Outcome , Valproic Acid/chemistry , Valproic Acid/therapeutic use
6.
J Med Chem ; 56(16): 6467-77, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23879329

ABSTRACT

sec-Butylpropylacetamide (racemic-SPD) is a chiral CNS-active amide derivative of valproic acid (VPA). This study describes synthesis and stereospecific comparative pharmacodynamics (PD, anticonvulsant activity and teratogenicity) and pharmacokinetic (PK) analysis of four individual SPD stereoisomers. SPD stereoisomers' anticonvulsant activity was comparatively evaluated in several anticonvulsant animal models including the benzodiazepine-resistant status epilepticus (SE). SPD stereoisomers' PK-PD relationship was evaluated in rats. Teratogenicity of SPD stereoisomers was evaluated in SWV mice strain, susceptible to VPA-induced neural tube defect (NTD). SPD stereoisomers (141 or 283 mg/kg) did not cause NTD. SPD has stereoselective PK and PD. (2R,3S)-SPD and (2S,3R)-SPD higher clearance led to a 50% lower plasma exposure that may contribute to their relative lower activity in the pilocarpine-induced SE model. (2S,3S)-SPD, (2R,3R)-SPD, and racemic-SPD have similar anticonvulsant activity and a PK profile that are better than those of (2R,3S)-SPD and (2S,3R)-SPD, making them good candidates for development as new, potent antiepileptics with a potential in benzodiazepine-resistant SE.


Subject(s)
Acetamides/therapeutic use , Anticonvulsants/therapeutic use , Status Epilepticus/drug therapy , Valproic Acid/therapeutic use , Acetamides/pharmacokinetics , Acetamides/pharmacology , Amides , Animals , Anticonvulsants/pharmacokinetics , Anticonvulsants/pharmacology , Disease Models, Animal , Rats , Soman/antagonists & inhibitors , Stereoisomerism , Teratogens/toxicity , Valproic Acid/pharmacokinetics , Valproic Acid/pharmacology
7.
J Med Chem ; 55(6): 2835-45, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22339381

ABSTRACT

A novel class of 19 carbamates was synthesized, and their anticonvulsant activity was comparatively evaluated in the rat maximal electroshock (MES) and subcutaneous metrazol (scMet) seizure tests and pilocarpine-induced status epilepticus (SE) model. In spite of the alkyl-carbamates' close structural features, only compounds 34, 38, and 40 were active at the MES test. The analogues 2-ethyl-3-methyl-butyl-carbamate (34) and 2-ethyl-3-methyl-pentyl-carbamate (38) also exhibited potent activity in the pilocarpine-SE model 30 min postseizure onset. Extending the aliphatic side chains of homologous carbamates from 7 to 8 (34 to 35) and from 8 to 9 carbons in the homologues 38 and 43 decreased the activity in the pilocarpine-SE model from ED(50) = 81 mg/kg (34) to 94 mg/kg (35) and from 96 mg/kg (38) to 114 mg/kg (43), respectively. The most potent carbamate, phenyl-ethyl-carbamate (47) (MES ED(50) = 16 mg/kg) contains an aromatic moiety in its structure. Compounds 34, 38, 40, and 47 offer the optimal efficacy-safety profile and, consequently, are promising candidates for development as new antiepileptics.


Subject(s)
Anticonvulsants/chemical synthesis , Carbamates/chemical synthesis , Animals , Anticonvulsants/pharmacology , Anticonvulsants/toxicity , Carbamates/pharmacology , Carbamates/toxicity , Male , Mice , Neurotoxicity Syndromes/etiology , Rats , Rats, Sprague-Dawley , Seizures/drug therapy , Status Epilepticus/drug therapy , Structure-Activity Relationship
8.
Epilepsia ; 53(1): 134-46, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22150444

ABSTRACT

PURPOSE: sec-Butyl-propylacetamide (SPD) is a one-carbon homolog of valnoctamide (VCD), a central nervous system (CNS)-active amide derivative of valproic acid (VPA) currently in phase II clinical trials. The study reported herein evaluated the anticonvulsant activity of SPD in a battery of rodent seizure and epilepsy models and assessed its efficacy in rat and guinea pig models of status epilepticus (SE) and neuroprotection in an organotypic hippocampal slice model of excitotoxic cell death. METHODS: The anticonvulsant activity of SPD was evaluated in several rodent seizure and epilepsy models, including maximal electroshock (MES), 6-Hz psychomotor; subcutaneous (s.c.) metrazol-, s.c. picrotoxin, s.c. bicuculline, and audiogenic, corneal, and hippocampal kindled seizures following intraperitoneal administration. Results obtained with SPD are discussed in relationship to those obtained with VPA and VCD. SPD was also evaluated for its ability to block benzodiazepine-resistant SE induced by pilocarpine (rats) and soman (rats and guinea pigs) following intraperitoneal administration. SPD was tested for its ability to block excitotoxic cell death induced by the glutamate agonists N-methyl-D-aspartate (NMDA) and kainic acid (KA) using organotypic hippocampal slices and SE-induced hippocampal cell death using FluoroJade B staining. The cognitive function of SPD-treated rats that were protected against pilocarpine-induced convulsive SE was examined 10-14 days post-SE using the Morris water maze (MWM). The relationship between the pharmacokinetic profile of SPD and its efficacy against soman-induced SE was evaluated in two parallel studies following SPD (60 mg/kg, i.p.) administration in the soman SE rat model. KEY FINDINGS: SPD was highly effective and displayed a wide protective index (PI = median neurotoxic dose/median effective dose [TD(50)/ED(50)]) in the standardized seizure and epilepsy models employed. The wide PI values of SPD demonstrate that it is effective at doses well below those that produce behavioral impairment. Unlike VCD, SPD also displayed anticonvulsant activity in the rat pilocarpine model of SE. Thirty minutes after the induction of SE, the calculated rat ED(50) for SPD against convulsive SE in this model was 84 mg/kg. SPD was not neuroprotective in the organotypic hippocampal slice preparation; however, it did display hippocampal neuroprotection in both SE models and cognitive sparing in the MWM, which was associated with its antiseizure effect against pilocarpine-induced SE. When administered 20 and 40 min after SE onset, SPD (100-174 mg/kg) produced long-lasting efficacy (e.g., 4-8 h) against soman-induced convulsive and electrographic SE in both rats and guinea pigs. SPD ED(50) values in guinea pigs were 67 and 92 mg/kg when administered at SE onset or 40 min after SE onset, respectively. Assuming linear pharmacokinetics (PK), the PK-PD (pharmacodynamic) results (rats) suggests that effective SPD plasma levels ranged between 8 and 40 mg/L (20 min after the onset of soman-induced seizures) and 12-50 mg/L (40 min after the onset of soman-induced seizures). The time to peak (t(max)) pharmacodynamic effect (PD-t(max)) occurred after the PK-t(max), suggesting that SPD undergoes slow distribution to extraplasmatic sites, which is likely responsible for antiseizure activity of SPD. SIGNIFICANCE: The results demonstrate that SPD is a broad-spectrum antiseizure compound that blocks SE induced by pilocarpine and soman and affords in vivo neuroprotection that is associated with cognitive sparing. Its activity against SE is superior to that of diazepam in terms of rapid onset, potency, and its effect on animal mortality and functional improvement.


Subject(s)
Anticonvulsants/pharmacology , Anticonvulsants/pharmacokinetics , Seizures/drug therapy , Status Epilepticus/drug therapy , Valproic Acid/chemistry , Amides/chemistry , Amides/pharmacokinetics , Amides/pharmacology , Animals , Anticonvulsants/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Guinea Pigs , Hippocampus/drug effects , Male , Mice , Neurons/drug effects , Neuroprotective Agents , Rats , Rats, Sprague-Dawley , Time Factors , Treatment Outcome , Valproic Acid/analogs & derivatives , Valproic Acid/pharmacokinetics , Valproic Acid/pharmacology
9.
Epilepsy Behav ; 22(3): 461-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21959082

ABSTRACT

A series of glycinamide conjugates and N-methoxy amide derivatives of valproic acid (VPA) analogs and constitutional isomers were synthesized and evaluated for anticonvulsant activity. Of all compounds synthesized and tested, only N-methoxy-valnoctamide (N-methoxy-VCD) possessed better activity than VPA in the following anticonvulsant tests: maximal electroshock, subcutaneous metrazol, and 6-Hz (32-mA) seizure tests. In mice, the ED(50) values of N-methoxy-VCD were 142 mg/kg (maximal electroshock test), 70 mg/kg (subcutaneous metrazol test), and 35 mg/kg (6-Hz test), and its neurotoxicity TD(50) was 118 mg/kg. In rats, the ED(50) of N-methoxy-VCD in the subcutaneous metrazol test was 36 mg/kg and its protective index (PI=TD(50)/ED(50)) was >5.5. In the rat pilocarpine-induced status epilepticus model, N-methoxy-VCD demonstrated full protection at 200mg/kg, without any neurotoxicity. N-Methoxy-VCD was tested for its ability to induce teratogenicity in a mouse strain susceptible to VPA-induced teratogenicity and was found to be nonteratogenic, although it caused some resorptions. Nevertheless, a safety margin was still maintained between the ED(50) values of N-methoxy-VCD in the mouse subcutaneous metrazol test and the doses that caused the resorptions. On the basis of these results, N-methoxy-VCD is a good candidate for further evaluation as a new anticonvulsant and central nervous system drug.


Subject(s)
Amides , Anticonvulsants/chemistry , Anticonvulsants/therapeutic use , Seizures/drug therapy , Valproic Acid , Amides/chemistry , Amides/therapeutic use , Animals , Convulsants/toxicity , Disease Models, Animal , Electroshock/adverse effects , Female , Isomerism , Male , Mice , Neural Tube Defects/chemically induced , Pentylenetetrazole/toxicity , Rats , Rats, Sprague-Dawley , Seizures/etiology , Structure-Activity Relationship , Valproic Acid/analogs & derivatives , Valproic Acid/chemistry , Valproic Acid/therapeutic use
10.
J Med Chem ; 54(11): 3977-81, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21506569

ABSTRACT

Aromatic amides comprising branched aliphatic carboxylic acids and 4-aminobenzenesulfonamide were evaluated for their inhibition of carbonic anhydrase (CA) isoforms. Of the most anticonvulsant-active compounds (2, 4, 13, 16, and 17), only 13, 16, and 17 were potent inhibitors of CAs VII and XIV. Compounds 9, 14, and 19 inhibited CA II, while 10 and 12 inhibited all isoforms. Structural studies suggest that differences in the active sites' hydrophobicity modulate the affinity of the inhibitors.


Subject(s)
Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Anticonvulsants/chemical synthesis , Binding Sites , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrases/metabolism , Crystallography, X-Ray , Epilepsy/drug therapy , Humans , Hydrophobic and Hydrophilic Interactions , Protein Isoforms/chemical synthesis , Protein Isoforms/chemistry , Protein Isoforms/pharmacology , Structure-Activity Relationship , Sulfonamides/chemical synthesis
11.
Epilepsia ; 51(10): 1944-53, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20738383

ABSTRACT

PURPOSE: α-Fluoro-2,2,3,3-tetramethylcyclopropanecarboxamide (α-F-TMCD) and α-Cl-TMCD, are α-halo derivatives of TMCD, the corresponding amide of a cyclopropane analog of valproic acid (VPA). This study aimed to comparatively evaluate the pharmacodynamics and pharmacokinetics of α-F-TMCD and α-Cl-TMCD in rodent models of epilepsy and for antiepileptic drug (AED)-induced teratogenicity. The potential of α-F-TMCD as an antiallodynic and antinociceptive compound was also evaluated. METHODS: α-F-TMCD and α-Cl-TMCD were synthesized. α-Cl-TMCD anticonvulsant activity was evaluated in comparison to VPA in the mouse maximal-electroshock-seizure (MES), Metrazol (scMet), and 6-Hz psychomotor-seizure tests. Neurotoxicity was assessed by the Rotorod-ataxia test. Induction of neural tube defects (NTDs) by α-Cl-TMCD and α-F-TMCD was evaluated after intraperitoneal administration to a mouse strain highly susceptible to VPA-induced teratogenicity. The ability of α-F-TMCD to reduce pain was evaluated in the rat spinal nerve ligation (SNL) model for neuropathic pain and in the formalin test. α-F-TMCD and α-Cl-TMCD pharmacokinetics was evaluated following intraperitoneal (40 mg/kg) and oral (60 mg/kg) administration to rats. RESULTS: α-F-TMCD and α-Cl-TMCD had similar potencies in the 6-Hz test and were more potent than VPA in this model and in the scMet test. Neither induced NTDs, and both exhibited wide safety margins. α-F-TMCD was active in the two pain models, and was found to be equipotent to gabapentin in the SNL model (ED(50) = 37 and 32 mg/kg, respectively). Comparative pharmacokinetic analysis showed that α-Cl-TMCD is less susceptible to liver first-pass effect than α-F-TMCD because of lower total (metabolic) clearance and liver extraction ratio. CONCLUSIONS: Based on their potent anticonvulsant activity and lack of teratogenicity, α-F-TMCD and α-Cl-TMCD have the potential for development as new antiepileptics and central nervous system (CNS) drugs.


Subject(s)
Amides/pharmacology , Amides/pharmacokinetics , Anticonvulsants/pharmacology , Anticonvulsants/pharmacokinetics , Cyclopropanes/pharmacology , Cyclopropanes/pharmacokinetics , Epilepsy/prevention & control , Valproic Acid/analogs & derivatives , Abnormalities, Drug-Induced/etiology , Abnormalities, Drug-Induced/prevention & control , Amides/adverse effects , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Cyclopropanes/adverse effects , Disease Models, Animal , Humans , Mice , Neural Tube Defects/chemically induced , Neural Tube Defects/prevention & control , Pain/prevention & control , Pentylenetetrazole/pharmacology , Rats , Valproic Acid/pharmacokinetics , Valproic Acid/pharmacology
12.
J Med Chem ; 53(10): 4177-86, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20420384

ABSTRACT

Despite the availability of 14 new antiepileptic drugs (AEDs), about 30% of epileptic patients are not seizure-free. Consequently there is substantial need to develop new effective AEDs. A novel class of aromatic amides composed of phenylacetic acid or branched aliphatic carboxylic acids, with five to nine carbons in their carboxylic moiety, and aminobenzenesulfonamide were synthesized and evaluated in the anticonvulsant rat-maximal electroshock (MES) and subcutaneous metrazol seizure (scMet) tests. Fourteen of the synthesized amides had an anticonvulsant ED(50) of <50 mg/kg in the rat-MES test. The amides 2-methyl-N-(4-sulfamoylphenyl)butyramide (10), 2-ethyl-N-(4-sulfamoylphenyl)butyramide (11), and 3,3-dimethyl-N-(4-sulfamoylphenyl)butyramide (15) were the most potent compounds possessing MES-ED(50) values of 7.6, 9.9, and 9.4 mg/kg and remarkable protective index (PI = TD(50)/ED(50)) values of 65.7, 50.5, and 53.2, respectively. These potent sulfanylamides caused neural tube defects only at doses markedly exceeding their effective dose. The anticonvulsant properties of these compounds make them potential candidates for further development as new, potent, and safe AEDs.


Subject(s)
Anilides/chemical synthesis , Anticonvulsants/chemical synthesis , Neural Tube Defects/chemically induced , Seizures/drug therapy , Sulfonamides/chemical synthesis , Anilides/pharmacology , Anilides/toxicity , Animals , Anticonvulsants/pharmacology , Anticonvulsants/toxicity , Convulsants , Electroshock , Mice , Pentylenetetrazole , Rats , Rats, Sprague-Dawley , Seizures/etiology , Structure-Activity Relationship , Sulfonamides/pharmacology , Sulfonamides/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...