Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Bioconjug Chem ; 33(7): 1340-1349, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35767675

ABSTRACT

It is particularly challenging to develop a truly effective pharmacotherapy for cocaine use disorder (CUD) treatment. Accelerating cocaine metabolism via hydrolysis at cocaine benzoyl ester using an efficient cocaine hydrolase (CocH) is known as a promising pharmacotherapeutic approach to CUD treatment. Preclinical and clinical studies on our first CocH (CocH1), in its human serum albumin-fused form known as TV-1380, have demonstrated the promise of a general concept of CocH-based pharmacotherapy for CUD treatment. However, the biological half-life of TV-1380 (t1/2 = 8 h in rats, associated with t1/2 = 43-77 h in humans) is not long enough for practical treatment of cocaine dependence, which requires enzyme injection for no more than once weekly. Through protein fusion of a human butyrylcholinesterase mutant (denoted as CocH5) with a mutant (denoted as Fc(M6)) of Fc from human IgG1, we have designed, prepared, and tested a new fusion protein (denoted as CocH5-Fc(M6)) for its pharmacokinetic profile and in vivo catalytic activity against (-)-cocaine. CocH5-Fc(M6) represents the currently most efficient long-acting cocaine hydrolase with both the highest catalytic activity against (-)-cocaine and the longest elimination half-life (t1/2 = 229 ± 5 h) in rats. As a result, even at a single modest dose of 3 mg/kg, CocH5-Fc(M6) can significantly and effectively accelerate the metabolism of cocaine in rats for at least 60 days. In addition, ∼70 nM CocH5-Fc(M6) in plasma was able to completely block the toxicity and physiological effects induced by intraperitoneal injection of a lethal dose of cocaine (60 mg/kg).


Subject(s)
Cocaine-Related Disorders , Cocaine , Animals , Butyrylcholinesterase/genetics , Butyrylcholinesterase/pharmacokinetics , Carboxylic Ester Hydrolases/genetics , Cocaine/metabolism , Cocaine/therapeutic use , Cocaine-Related Disorders/drug therapy , Humans , Rats , Recombinant Proteins
2.
Biotechnol Bioeng ; 117(1): 157-166, 2020 01.
Article in English | MEDLINE | ID: mdl-31544955

ABSTRACT

Chinese hamster ovary (CHO) cells typically produce glycoproteins with N-glycans terminating in α-2,3 sialylation. Human cells produce glycoproteins that include α-2,3 and α-2,6 sialic acids. To examine the impact of altering protein sialylation on pharmacokinetic properties, recombinant human butyrylcholinesterase (BChE) was produced in CHO cells by knocking out the α-2,3 sialyltransferase genes followed by overexpression of the α-2,6 sialyltransferase (26BChE) enzyme. The N-glycan composition of 26BChE was compared to BChE with α-2,3 sialylation (23BChE) derived from wild-type CHO cells. Both 23BChE and 26BChE exhibited comparable antennarity distributions with bi-antennary di-sialylated glycans representing the most abundant glycoform. CD-1 mice were intravenously injected with the 23BChE or 26BChE, and residual BChE activities from blood collected at various time points for pharmacokinetic analyses. Although 23BChE contained a slightly lower initial sialylation level compared to 26BChE, the molecule exhibited higher residual activity between 5 and 24 hr postinjection. Pharmacokinetic analyses indicated that 23BChE exhibited an increase in area under the curve and a lower volume of distribution at steady state than that of 26BChE. These findings suggest that the type of sialylation linkage may play a significant role in the pharmacokinetic behavior of a biotherapeutic when tested in in vivo animal models.


Subject(s)
Butyrylcholinesterase/chemistry , Butyrylcholinesterase/pharmacokinetics , N-Acetylneuraminic Acid/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Animals , Butyrylcholinesterase/blood , Butyrylcholinesterase/genetics , CHO Cells , Cricetinae , Cricetulus , Humans , Mice , Recombinant Proteins/blood , Recombinant Proteins/genetics
3.
Proc Natl Acad Sci U S A ; 110(4): 1243-8, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23297221

ABSTRACT

The creation of effective bioscavengers as a pretreatment for exposure to nerve agents is a challenging medical objective. We report a recombinant method using chemical polysialylation to generate bioscavengers stable in the bloodstream. Development of a CHO-based expression system using genes encoding human butyrylcholinesterase and a proline-rich peptide under elongation factor promoter control resulted in self-assembling, active enzyme multimers. Polysialylation gives bioscavengers with enhanced pharmacokinetics which protect mice against 4.2 LD(50) of S-(2-(diethylamino)ethyl) O-isobutyl methanephosphonothioate without perturbation of long-term behavior.


Subject(s)
Butyrylcholinesterase/chemistry , Butyrylcholinesterase/pharmacokinetics , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Amino Acid Sequence , Animals , Butyrylcholinesterase/administration & dosage , Butyrylcholinesterase/genetics , CHO Cells , Chemical Warfare Agents/toxicity , Cricetinae , Cricetulus , Humans , Lethal Dose 50 , Male , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Neuroprotective Agents/administration & dosage , Organothiophosphorus Compounds/antagonists & inhibitors , Organothiophosphorus Compounds/toxicity , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacokinetics , Sialic Acids/chemistry
4.
Addict Biol ; 18(1): 30-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22264200

ABSTRACT

Although substantial research effort has focused on developing pharmacological treatments for cocaine abuse, no effective medications have been developed. Recent studies show that enzymes that metabolize cocaine in the periphery, forestalling its entry into the brain, can prevent cocaine toxicity and its behavioral effects in rodents. Here we report on effects of one such enzyme (Albu-CocH) on the pharmacokinetic and behavioral effects of cocaine in squirrel monkeys. Albu-CocH was developed from successive mutations of human butyrylcholinesterase (BChE) and has 1000-fold greater catalytic activity against cocaine than naturally occurring BChE. Pharmacokinetic studies showed that Albu-CocH (5 mg/kg) had a half-life of 56.6 hours in squirrel monkeys. In these studies, plasma levels of cocaine following i.v. 1 mg/kg cocaine were reduced 2 hours after administration of Albu-CocH, whereas plasma levels of the cocaine metabolite ecgonine methyl ester were increased. These effects were still evident 72 hours following Albu-CocH administration. In behavioral experiments in monkeys, pre-treatment with 5 mg/kg Albu-CocH dramatically decreased self-administration of a reinforcing dose of i.v. cocaine (30 µg/kg/injection) for over 24 hours. Pre-treatment with 5 mg/kg Albu-CocH also attenuated the reinstatement of extinguished cocaine self-administration by an i.v. priming injection of cocaine (0.1 or 0.3 mg/kg) and, in separate studies, attenuated the discriminative-stimulus effects of cocaine. The ability of Albu-CocH to attenuate the abuse-related effects of cocaine in squirrel monkeys indicates that further investigation of BChE mutants as potential treatment for cocaine abuse and toxicity is warranted.


Subject(s)
Albumins/pharmacology , Butyrylcholinesterase/pharmacology , Cocaine-Related Disorders/drug therapy , Cocaine/pharmacokinetics , Dopamine Uptake Inhibitors/pharmacokinetics , Albumins/pharmacokinetics , Analysis of Variance , Animals , Antibody Formation/drug effects , Biocatalysis , Butyrylcholinesterase/pharmacokinetics , Cocaine/administration & dosage , Cocaine/antagonists & inhibitors , Discrimination Learning/drug effects , Dopamine Uptake Inhibitors/administration & dosage , Dopamine Uptake Inhibitors/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions/prevention & control , Drug-Seeking Behavior/drug effects , Half-Life , Humans , Male , Reinforcement, Psychology , Saimiri , Self Administration
5.
Proc Natl Acad Sci U S A ; 107(47): 20251-6, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21059932

ABSTRACT

The concept of using cholinesterase bioscavengers for prophylaxis against organophosphorous nerve agents and pesticides has progressed from the bench to clinical trial. However, the supply of the native human proteins is either limited (e.g., plasma-derived butyrylcholinesterase and erythrocytic acetylcholinesterase) or nonexisting (synaptic acetylcholinesterase). Here we identify a unique form of recombinant human butyrylcholinesterase that mimics the native enzyme assembly into tetramers; this form provides extended effective pharmacokinetics that is significantly enhanced by polyethylene glycol conjugation. We further demonstrate that this enzyme (but not a G117H/E197Q organophosphorus acid anhydride hydrolase catalytic variant) can prevent morbidity and mortality associated with organophosphorous nerve agent and pesticide exposure of animal subjects of two model species.


Subject(s)
Butyrylcholinesterase/pharmacology , Chemical Warfare Agents/toxicity , Neuroprotective Agents/pharmacology , Nicotiana/metabolism , Organophosphorus Compounds/toxicity , Pesticides/toxicity , Animals , Butyrylcholinesterase/metabolism , Butyrylcholinesterase/pharmacokinetics , Chemical Warfare Agents/metabolism , Chromatography, High Pressure Liquid , Guinea Pigs , Humans , Immunoblotting , Kinetics , Mice , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Organophosphorus Compounds/metabolism , Pesticides/metabolism , Polyethylene Glycols/metabolism , Protein Engineering
6.
Vox Sang ; 100(3): 285-97, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20946535

ABSTRACT

BACKGROUND AND OBJECTIVES: A human plasma-derived butyrylcholinesterase preparation manufactured on the industrial scale is described. MATERIAL AND METHODS: The human butyrylcholinesterase (hBChE) product was extensively investigated for its purity using immunological and electrophoretic methods and characterized by thorough glycoproteomic approaches. A comprehensive preclinical testing programme addressing safety and pharmacokinetic parameters supplemented the biochemical characterization. RESULTS: The high-purity hBChE preparation is tetrameric and has high specific activity and molecular integrity of the protein backbone. Acute toxicity studies and in vivo thrombogenicity studies provided evidence of a sufficient safety margin for use in humans. CONCLUSION: Extensive preclinical safety and pharmacokinetic testing confirmed that this hBChE preparation can be used for further efficacy testing as a bioscavenger for toxic organophosphate compounds in appropriate animal models and ultimately in humans.


Subject(s)
Butyrylcholinesterase/isolation & purification , Drug Industry/methods , Butyrylcholinesterase/pharmacokinetics , Butyrylcholinesterase/toxicity , Humans , Materials Testing , Organophosphates , Pharmacokinetics , Quality Control , Viruses
7.
Chem Biol Interact ; 344: 109499, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-33961835

ABSTRACT

Exogenously administered human serum butyrylcholinesterase (Hu BChE) affords protection by binding to organophosphorus (OP) nerve agents and pesticides in circulation. The resulting Hu BChE-OP conjugate undergoes 'aging' and the conjugate circulates until cleared from the body. Thus, we evaluated the effects of Hu BChE-OP conjugates on the general health and operant behavior of macaques. Rhesus macaques trained to perform a six-item serial probe recognition (SPR) task were administered 30 mg/kg of Hu BChE-soman conjugate (n = 4) or Hu BChE-VX conjugate (n = 4) by intramuscular injection. Performance on the SPR task was evaluated at 60-90 min after conjugate administration and daily thereafter for the next 4 weeks. Diazepam (3.2 mg/kg), a positive control, was administered 5 weeks after conjugate administration and performance on the SPR task was evaluated as before. Blood collected throughout the study was analyzed for acetylcholinesterase (AChE) and BChE activities. Residual BChE activity of conjugates displayed a similar pharmacokinetic profile as free Hu BChE. Neither of the Hu BChE-OP conjugates produced clear or pronounced degradations in performance on the SPR task. In contrast, diazepam clearly impaired performance on the SPR task on the day of administration in 7 of 8 macaques (and sometimes longer). Taken together, these results suggest that Hu BChE-OP conjugates are safe and provide further support for the development of Hu BChE as a bioscavenger for use in humans.


Subject(s)
Butyrylcholinesterase/toxicity , Nerve Agents/toxicity , Organothiophosphorus Compounds/toxicity , Soman/toxicity , Animals , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/pharmacokinetics , Diazepam/pharmacology , Female , Humans , Macaca mulatta , Male , Memory/drug effects , Nerve Agents/chemistry , Nerve Agents/pharmacokinetics , Organothiophosphorus Compounds/chemistry , Organothiophosphorus Compounds/pharmacokinetics , Soman/chemistry , Soman/pharmacokinetics
8.
AAPS J ; 22(1): 5, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31754920

ABSTRACT

Therapeutic treatment of cocaine toxicity or addiction is a grand medical challenge. As a promising therapeutic strategy for treatment of cocaine toxicity and addiction to develop a highly efficient cocaine hydrolase (CocH) capable of accelerating cocaine metabolism to produce physiologically/biologically inactive metabolites, our previously designed A199S/S287G/A328W/Y332G mutant of human butyrylcholinesterase (BChE), known as cocaine hydrolase-1 (CocH1), possesses the desirably high catalytic activity against cocaine. The C-terminus of CocH1, truncated after amino acid #529, was fused to human serum albumin (HSA) to extend the biological half-life. The C-terminal HSA-fused CocH1 (CocH1-HSA), known as Albu-CocH1, Albu-CocH, AlbuBChE, Albu-BChE, or TV-1380 in literature, has shown favorable preclinical and clinical profiles. However, the actual therapeutic value of TV-1380 for cocaine addiction treatment is still limited by the short half-life. In this study, we designed and tested a new type of HSA-fused CocH1 proteins, i.e., N-terminal HSA-fused CocH1, with or without a linker between the HSA and CocH1 domains. It has been demonstrated that the catalytic activity of these new fusion proteins against cocaine is similar to that of TV-1380. However, HSA-CocH1 (without a linker) has a significantly longer biological half-life (t1/2 = 14 ± 2 h) compared to the corresponding C-terminal HSA-fused CocH1, i.e., CocH1-HSA (TV-1380 with t1/2 = 5-8 h), in rats. Further, the N-terminal HSA-fused CocH1 proteins with a linker have further prolonged biological half-lives: t1/2 = 17 ± 2 h for both HSA-EAAAK-CocH1 and HSA-PAPAP-CocH1, and t1/2 = 18 ± 3 h for HSA-(PAPAP)2-CocH1. These N-terminal HSA-fused CocH1 proteins may serve as more promising protein drug candidates for cocaine addiction treatment.


Subject(s)
Albumins/pharmacokinetics , Butyrylcholinesterase/pharmacokinetics , Carboxylic Ester Hydrolases/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Proteins/pharmacokinetics , Albumins/chemistry , Animals , Butyrylcholinesterase/chemistry , Carboxylic Ester Hydrolases/chemistry , Half-Life , Mice , Models, Molecular , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Proteins/chemistry
9.
Chem Biol Interact ; 308: 170-178, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31129133

ABSTRACT

Organophosphorus compounds (OP) pose a significant threat. Administration of human butyrylcholinesterase (HuBChE) may reduce or prevent OP toxicity. Thus, we evaluated the safety and efficacy of HuBChE in monkeys using sensitive neurobehavioral tests while concurrently characterizing absorption and elimination in the presence and absence of high-dose soman exposure to predict time course and degree of protection. Eight young adult male cynomolgus macaques were trained on two distinct automated tests of neurobehavioral functioning. HuBChE purified under current Good Manufacturing Practices (CGMP) was injected intramuscularly at 13.1 mg/kg, producing an average peak plasma value (Cmax) of over 27 Units/ml. The apparent time to maximum concentration (Tmax) approximated 7 h, the elimination half-life approximated 102 h, and plasma levels returned to pre-administration (baseline) levels by 14 days. No behavioral disruptions following HuBChE administration were observed on either neurobehavioral test, even in monkeys injected 24 h later with an otherwise lethal dose of soman. Thus, HuBChE provided complete neurobehavioral protection from soman challenge. The present data replicate and extend previous results from our laboratory that had used a different route of administration (intravenous), a different species (rhesus macaque), and a different BChE product (non-CGMP material). The addition of two sensitive neurobehavioral tests coupled with the PK/PD results convincingly demonstrates the neurobehavioral safety of plasma-derived HuBChE at therapeutic levels. Protection against an otherwise-lethal dose of soman by a pre-exposure treatment dose that is devoid of side effects establishes a foundation for additional testing using other exposure routes and treatment times, other challenge agents/routes, or other classes of organophosphate scavengers.


Subject(s)
Behavior, Animal/drug effects , Butyrylcholinesterase/administration & dosage , Chemical Warfare Agents/metabolism , Soman/metabolism , Animals , Butyrylcholinesterase/blood , Butyrylcholinesterase/pharmacokinetics , Chemical Warfare Agents/toxicity , Half-Life , Humans , Injections, Intramuscular , Macaca fascicularis , Male , Soman/toxicity
10.
BMC Biotechnol ; 8: 50, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18485214

ABSTRACT

BACKGROUND: Human butyrylcholinesterase (huBChE) has been shown to be an effective antidote against multiple LD50 of organophosphorus compounds. A prerequisite for such use of huBChE is a prolonged circulatory half-life. This study was undertaken to produce recombinant huBChE fused to human serum albumin (hSA) and characterize the fusion protein. RESULTS: Secretion level of the fusion protein produced in vitro in BHK cells was approximately 30 mg/liter. Transgenic mice and goats generated with the fusion constructs expressed in their milk a bioactive protein at concentrations of 0.04-1.1 g/liter. BChE activity gel staining and a size exclusion chromatography (SEC)-HPLC revealed that the fusion protein consisted of predominant dimers and some monomers. The protein was confirmed to have expected molecular mass of approximately 150 kDa by Western blot. The purified fusion protein produced in vitro was injected intravenously into juvenile pigs for pharmacokinetic study. Analysis of a series of blood samples using the Ellman assay revealed a substantial enhancement of the plasma half-life of the fusion protein (approximately 32 h) when compared with a transgenically produced huBChE preparation containing >70% tetramer (approximately 3 h). In vitro nerve agent binding and inhibition experiments indicated that the fusion protein in the milk of transgenic mice had similar inhibition characteristics compared to human plasma BChE against the nerve agents tested. CONCLUSION: Both the pharmacokinetic study and the in vitro nerve agent binding and inhibition assay suggested that a fusion protein retaining both properties of huBChE and hSA is produced in vitro and in vivo. The production of the fusion protein in the milk of transgenic goats provided further evidence that sufficient quantities of BChE/hSA can be produced to serve as a cost-effective and reliable source of BChE for prophylaxis and post-exposure treatment.


Subject(s)
Butyrylcholinesterase/pharmacokinetics , Kidney/enzymology , Protein Engineering/methods , Recombinant Fusion Proteins/pharmacokinetics , Serum Albumin/pharmacokinetics , Animals , Butyrylcholinesterase/blood , Butyrylcholinesterase/genetics , Cell Line , Cricetinae , Goats , Humans , Metabolic Clearance Rate , Mice , Mice, Transgenic , Serum Albumin/genetics , Swine
11.
Chem Biol Interact ; 175(1-3): 267-72, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18597747

ABSTRACT

Human serum butyrylcholinesterase (Hu BChE) is currently under advanced development as a pretreatment drug for organophosphate (OP) poisoning in humans. It was shown to protect mice, rats, guinea pigs, and monkeys against multiple LD(50) challenges of OP nerve agents by i.v. or s.c. bolus injections. Since inhalation is the most likely route of exposure to OP nerve agents on the battlefield or in public places, the aim of this study was to evaluate the efficacy of Hu BChE against whole-body inhalation exposure to sarin (GB) vapor. Male Göttingen minipigs were subjected to one of the following treatments: (1) air exposure; (2) GB vapor exposure; (3) pretreatment with 3 mg/kg of Hu BChE followed by GB vapor exposure; (4) pretreatment with 6.5 mg/kg of Hu BChE followed by GB vapor exposure; (5) pretreatment with 7.5 mg/kg of Hu BChE followed by GB vapor exposure. Hu BChE was administered by i.m. injection, 24h prior to whole-body exposure to GB vapor at a concentration of 4.1 mg/m(3) for 60 min, a dose lethal to 99% of untreated exposed pigs (LCt99). EEG, ECG, and pupil size were monitored throughout exposure, and blood drawn from a surgically implanted jugular catheter before and throughout the exposure period, was analyzed for acetylcholinesterase (AChE) and BChE activities, and the amount of GB present in plasma. All animals exposed to GB vapor alone or pretreated with 3 or 6.5 mg/kg of Hu BChE, died following exposure to GB vapor. All five animals pretreated with 7.5 mg/kg of Hu BChE survived the GB exposure. The amount of GB bound in plasma was 200-fold higher compared to that from plasma of pigs that did not receive Hu BChE, suggesting that Hu BChE was effective in scavenging GB in blood. Additionally, pretreatment with 7.5 mg/kg of Hu BChE prevented cardiac abnormalities and seizure activity observed in untreated animals and those treated with lower doses of Hu BChE.


Subject(s)
Antidotes/therapeutic use , Butyrylcholinesterase/blood , Butyrylcholinesterase/therapeutic use , Chemical Warfare Agents/toxicity , Sarin/toxicity , Animals , Biological Availability , Butyrylcholinesterase/pharmacokinetics , Humans , Male , Swine , Swine, Miniature
12.
Chem Biol Interact ; 175(1-3): 83-7, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18514640

ABSTRACT

Butyrylcholinesterase (BChE, EC 3.1.1.8) is important in human cocaine metabolism despite its limited ability to hydrolyze this drug. Efforts to improve the catalytic efficiency of this enzyme have led to a quadruple mutant cocaine hydrolase, "CocH", that in animal models of addiction appears promising for treatment of overdose and relapse. We incorporated the CocH mutations into a BChE-albumin fusion protein, "Albu-CocH", and evaluated the pharmacokinetics of the enzyme after i.v. injection in rats. As assessed from the time course of cocaine hydrolyzing activity in plasma, Albu-CocH redistributed into extracellular fluid (16% of estimated total body water) with a t(1/2) of 0.66h and it underwent elimination with a t(1/2) of 8h. These results indicate that the enzyme has ample stability for short-term applications and may be suitable for longer-term treatment as well. Present data also confirm the markedly enhanced power of Albu-CocH for cocaine hydrolysis and they support the view that Albu-CocH might prove valuable in treating phenomena associated with cocaine abuse.


Subject(s)
Albumins/metabolism , Butyrylcholinesterase/metabolism , Cocaine-Related Disorders/drug therapy , Cocaine/toxicity , Albumins/pharmacokinetics , Animals , Biocatalysis , Butyrylcholinesterase/pharmacokinetics , Female , Male , Rats , Rats, Wistar
13.
Drug Alcohol Depend ; 188: 295-303, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29807217

ABSTRACT

BACKGROUND: TV-1380 is a rationally mutated, human BChE fused to human serum albumin that has high hydrolytic enzymatic activity against cocaine and as well as an extended elimination half-life. OBJECTIVE: The present studies examined the safety of TV-1380 and its protective effect when given to monkeys alone or concomitantly with cocaine and ethanol. METHODS: A set of studies was conducted in monkeys with TV-1380. The parameters tested included telemetric assessment of cardiovascular parameters, clinical pathology, plasma analysis of cardiac troponin I, ex-vivo analyses of cocaethylene and PK analysis of serum concentrations of TV-1380, cocaine and its metabolites, and histopathological examinations. RESULTS: TV-1380 treatment in monkeys was well tolerated. TV-1380 pretreatment prior to cocaine significantly attenuated the cardiac effects of cocaine and reduced cocaine-induced elevations in serum cardiac troponin I. TV-1380 changed the metabolic fate of cocaine resulting in decreased exposure to benzoylecgonine, while increasing the exposure to ecgonine methyl ester in plasma.TV-1380 reduced the plasma levels of the toxic metabolite cocaethylene formed after co-administration of ethanol and cocaine. CONCLUSION: The results of this study demonstrate that TV-1380 not only accelerates the elimination of cocaine, but also protects the treated animal from the cardiac effects of cocaine, and inhibits the formation of the toxic cocaethylene metabolite when cocaine is given together with ethanol, supporting further clinical development of modified BChE products as possible treatments for cocaine abuse.


Subject(s)
Albumins/adverse effects , Albumins/pharmacology , Albumins/pharmacokinetics , Butyrylcholinesterase/adverse effects , Butyrylcholinesterase/pharmacology , Butyrylcholinesterase/pharmacokinetics , Cocaine/analogs & derivatives , Cocaine/antagonists & inhibitors , Ethanol/antagonists & inhibitors , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/pharmacokinetics , Animals , Butyrylcholinesterase/blood , Cocaine/blood , Cocaine/metabolism , Cocaine/pharmacokinetics , Cocaine/pharmacology , Drug Interactions , Electrocardiography/drug effects , Ethanol/blood , Ethanol/pharmacokinetics , Ethanol/pharmacology , Female , Heart Rate/drug effects , Humans , Macaca fascicularis , Male , Recombinant Fusion Proteins/blood , Respiration/drug effects , Troponin I/blood
14.
J Control Release ; 230: 73-8, 2016 05 28.
Article in English | MEDLINE | ID: mdl-27063423

ABSTRACT

Butyrylcholinesterase (BChE) is the most promising bioscavenger candidate to treat or prevent organophosphate (OP) poisoning. However, the clinical application of BChE is limited by two obstacles: an inadequate circulation half-life and limited sources for production. Although several modification technologies including glycosylation and PEGylation have been developed to improve its pharmacokinetics, none of them have been able to outperform blood-derived native BChE. In this work, we designed a long-circulating bioscavenger nanogel by coating equine serum-derived BChE with a zwitterionic polymer gel layer. This zwitterionic gel coating protected BChE from denaturation and degradation under harsh conditions. Notably, the nanocapsule exhibited a long circulation half-life of ~45h, a three-fold increase from the unmodified native version, enabling both therapeutic and prophylactic applications. In addition, the gel coating reduced the immunogenicity of equine BChE, unlocking the possibility to use non-human derived BChE as an OP bioscavenger in humans.


Subject(s)
Betaine/analogs & derivatives , Betaine/chemistry , Butyrylcholinesterase/administration & dosage , Nanocapsules/administration & dosage , Animals , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/pharmacokinetics , Cell Survival/drug effects , Cholinesterase Inhibitors/chemistry , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Gels , Horses , Male , Mice , NIH 3T3 Cells , Nanocapsules/chemistry , Paraoxon/analogs & derivatives , Paraoxon/chemistry , Rats, Sprague-Dawley
15.
Chem Biol Interact ; 157-158: 428-9, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16429577

ABSTRACT

Human serum butyrylcholinesterase (Hu BChE) has been demonstrated to be a highly effective detoxifying enzyme for counteracting the acute toxicity of organophosphorus (OP) nerve agents. In order to initiate an investigational new drug (IND) application for human use, the safety and pharmacokinetic properties of the enzyme were assessed in guinea pigs. Sixty milligrams per kilogram of Hu BChE was administered to guinea pigs by either i.p. or i.m. injection. Blood was drawn at various time points for up to 2 weeks following enzyme injection for the measurement of blood BChE activity. Hu BChE displayed a mean residence time of 110 h, regardless of the route of administration and the enzyme activity remained almost 10-fold above baseline level even after 2 weeks post enzyme injection. Fourteen days post Hu BChE administration, all animals were subjected to 20 panel serum chemistry, hematology, and complete gross/histopathology examination. Results showed no toxic effects as measured by general observation, serum chemistry, hematology, and gross and histological tissue changes. In conclusion, Hu BChE displays a long-lasting stability in the circulation of guinea pigs, and is devoid of any toxic side effects. These results provide convincing data for the safe and effective use of Hu BChE as a bioscavenger to protect humans against all OP nerve agents.


Subject(s)
Butyrylcholinesterase/adverse effects , Butyrylcholinesterase/pharmacokinetics , Animals , Butyrylcholinesterase/administration & dosage , Butyrylcholinesterase/blood , Guinea Pigs , Humans
16.
Chem Biol Interact ; 157-158: 199-203, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16263104

ABSTRACT

The use of exogenously administered cholinesterases (ChEs) as bioscavengers of highly toxic organophosphate (OP) nerve agents is now sufficiently well documented to make them a highly viable prophylactic treatment against this potential threat. Of the ChEs evaluated so far, human serum butyrylcholinesterase (HuBChE) is most suitable for human use. A dose of 200 mg (3 mg/kg) of HuBChE is envisioned as a prophylactic treatment in humans that can protect from an exposure of up to 2 x LD50 of soman. In addition to its use as a prophylactic for a variety of wartime scenarios, including covert actions, it also has potential use for first responders (civilians) reacting to terrorist nerve gas release. We recently, developed a procedure for the large-scale purification of HuBChE, which yielded approximately 6 g of highly purified enzyme from 120 kg of Cohn fraction IV-4. The enzyme had a specific activity of 700-750 U/mg and migrated as a single band on SDS-PAGE. To provide data for initiating an investigational new drug (IND) application for the use of this enzyme as a bioscavenger in humans, we established its pharmacokinetic properties, examined its safety in mice, and evaluated its shelf life at various temperatures. In mice administered various doses up to 90 mg/kg, enzyme activity reached peak levels in circulation at 10 and 24 h following i.p. and i.m. injections, respectively. The enzyme displayed a mean residence time (MRT) of 40-50 h, regardless of the route of administration or dose of injected enzyme. Mice were euthanized 2 weeks following enzyme administration and tissues were examined grossly or microscopically for possible toxic effects. Results suggest that HuBChE does not exhibit any toxicity in mice as measured by general observation, serum chemistry, hematology, gross or histologic tissue changes. The shelf life of this enzyme stored at 4, 25, 37, and 45 degrees C was determined in lyophilized form. The enzyme was found to be stable when stored in lyophilized form at -20, 4, 25, or 37 degrees C to date (2 years), as measured by specific activity and SDS polyacrylamide gel electrophoresis. The effect of storage on circulatory stability was determined by measuring MRT in mice; there was no change in the MRT of lyophilized enzyme stored at -20 degrees C to date (2 years). These results provide convincing data that HuBChE is a safe bioscavenger that can provide protection against all OP nerve agents. Efforts are now underway to prepare the required documentation for submission of an IND application to the United States Food and Drug Administration (USFDA).


Subject(s)
Butyrylcholinesterase/adverse effects , Butyrylcholinesterase/pharmacokinetics , Animals , Antidotes , Butyrylcholinesterase/administration & dosage , Enzyme Stability , Humans , Mice , Soman/antagonists & inhibitors , Soman/toxicity , Temperature
17.
Chem Biol Interact ; 157-158: 205-10, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16289064

ABSTRACT

Human butyrylcholinesterase (HuBuChE), purified from outdated human plasma, is being evaluated for efficacy against nerve agents in guinea pigs and cynomolgus monkeys. Previous studies in rodents and nonhuman primates demonstrated that pretreatment of animals with enzymes that can scavenge nerve agents could provide significant protection against behavioral and lethal effects of nerve agent intoxication. In preparation for evaluation of efficacy of HuBuChE prior to initiating an investigational new drug (IND) application, the pharmacokinetics of HuBuChE were evaluated in guinea pigs and in cynomolgus monkeys. HuBuChE was injected intramuscularly (i.m.) at two doses, and blood samples were taken to follow the time-course of HuBuChE in blood for up to 168 h after administration. In guinea pigs, the two doses of HuBuChE, 19.9 and 32.5 mg/kg, produced similar times of maximal blood concentration (T(max) of 26.0 and 26.8 h, respectively) and similar elimination half-times (t(1/2) of 64.6 and 75.5 h, respectively). Enzyme levels were still 10-fold over baseline at 72 h. Based on these data, guinea pigs were administered 150 mg/kg of enzyme i.m. and challenged at T(max). Soman or VX doses were approximately 1.5, 2.0 and 2.0 x LD50 administered subcutaneously (s.c.) in sequence at 90-120 min apart. None of the animals displayed signs of organophosphorus (OP) anticholinesterase intoxication at any of the challenge levels, and all survived for the 14-day duration of the experiment. Similar experiments were carried out with cynomolgus monkeys to determine the pharmacokinetics of HuBuChE and its efficacy against soman. The complete survival of nearly all animals tested to date, coupled with the maximal blood concentration and half-life elimination profile obtained for HuBuChE after i.m. injection, provides strong support for the continued development of HuBuChE as a product to protect against nerve agents.


Subject(s)
Butyrylcholinesterase/pharmacology , Macaca fascicularis/metabolism , Organothiophosphorus Compounds/antagonists & inhibitors , Organothiophosphorus Compounds/poisoning , Animals , Butyrylcholinesterase/administration & dosage , Butyrylcholinesterase/pharmacokinetics , Guinea Pigs , Humans , Lethal Dose 50 , Male , Nervous System Diseases/prevention & control
18.
Chem Biol Interact ; 157-158: 115-21, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16253215

ABSTRACT

Previous studies in rodents and non-human primates have demonstrated that pretreatment of animals with cholinesterases could provide significant protection against organophosphate (OP) nerve agent toxicity. Gene delivery/therapy is emerging as an approach to achieve high-level expression of proteins in vivo that are very similar to their native counterparts. Recently, adenoviral (Ad) vectors have proven to be excellent vehicles for delivering genes to cells in vitro and in vivo. In this study, we explored the use of the newly designed AdenoVATOR system for the expression of recombinant human butyrylcholinesterase (rHu BChE) in human embryonic kidney 293A (HEK-293A) cells. In these cells, rHu BChE was expressed as mostly tetrameric form by the simultaneous expression of proline-rich attachment domain. By optimizing the culture conditions, 1.5-2.0 U/ml of rHu BChE could be expressed in HEK-293A cells. Recombinant Hu BChE was purified to homogeneity by ammonium sulfate fractionation followed by affinity column chromatography using procainamide Sepharose and cobalt Sepharose gels. The enzymatic and physico-chemical properties of purified rHu BChE were similar to those of native serum-derived Hu BChE. To determine the suitability of this preparation for use as an antidote against highly toxic nerve agents, its pharmacokinetics were evaluated in mice. Recombinant Hu BChE exhibited a mean residence time of 18.3 h which was 2.5-fold shorter than that observed for native Hu BChE in mice. However, rHu BChE chemically modified with polyethyleneglycol (PEG) displayed a mean residence time of 36.2 h suggesting that PEG-modification can prolong the circulatory stability of rHu BChE. The efficacy of Ad-Hu BChE to induce the production of therapeutic levels of bioscavenger in vivo is under evaluation.


Subject(s)
Butyrylcholinesterase/chemistry , Butyrylcholinesterase/pharmacokinetics , Polyethylene Glycols/chemistry , Animals , Butyrylcholinesterase/blood , Butyrylcholinesterase/isolation & purification , Cell Line , Female , Gene Expression , Glycosylation , Humans , Kinetics , Male , Mice , Mice, Inbred BALB C , Peptide Hydrolases/metabolism , Recombinant Proteins/blood , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacokinetics , Time Factors
19.
Biochimie ; 118: 51-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26239905

ABSTRACT

Butyrylcholinesterase is a stoichiometric bioscavenger against poisoning by organophosphorus pesticides and nerve agents. The low level of expression and extremely rapid clearance of monomeric recombinant human butyrylcholinesterase (rhBChE) from bloodstream (t½≈2 min) limits its pharmaceutical application. Recently (Ilyushin at al., PNAS, 2013) we described a long-acting polysialylated recombinant butyrylcholinesterase (rhBChE-CAO), stable in the bloodstream, that protects mice against 4.2 LD50 of VR. Here we report a set of modifications of the initial rhBChE expression vector to improve stability of the enzyme in the bloodstream and increase its production in CHO cells by introducing in the expression cassette: (i) the sequence of the natural human PRAD-peptide in frame with rhBChE gene via "self-processing" viral F2A peptide under control of an hEF/HTLV promoter, and (ii) previously predicted in silico MAR 1-68 and MAR X-29 sequences. This provides fully tetrameric rhBChE (4rhBChE) at 70 mg/l, that displays improved pharmacokinetics (t½ = 32 ± 1.2 h, MRT = 43 ± 2 h). 3D Fluorescent visualization and distribution of (125)I-labeled enzyme reveals similar low level 4rhBChE and rhBChE-CAO accumulation in muscle, fat, and brain. Administered 4rhBChE was mainly catabolized in the liver and breakdown products were excreted in kidney. Injection of 1.2 LD50 and 1.1 LD50 of paraoxon to BALB/c and knockout BChE-/- mice pre-treated with 4rhBChE (50 mg/kg) resulted in 100% and 78% survival, respectively, without perturbation of long-term behavior. In contrast, 100% mortality of non-pre-treated mice was observed. The high expression level of 4rhBChE in CHO cells permits consideration of this new expression system for manufacturing BChE as a biopharmaceutical.


Subject(s)
Butyrylcholinesterase/chemical synthesis , Butyrylcholinesterase/pharmacology , Organophosphate Poisoning/prevention & control , Recombinant Proteins/chemical synthesis , Recombinant Proteins/pharmacology , Animals , Butyrylcholinesterase/pharmacokinetics , CHO Cells , Cloning, Molecular , Cricetinae , Cricetulus , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Polymerase Chain Reaction , Recombinant Proteins/pharmacokinetics , Transfection
20.
Chem Biol Interact ; 242: 219-26, 2015 Dec 05.
Article in English | MEDLINE | ID: mdl-26415620

ABSTRACT

Recombinant (r) and native butyrylcholinesterse (BChE) are potent bioscavengers of organophosphates (OPs) such as nerve agents and pesticides and are undergoing development as antidotal treatments for OP-induced toxicity. Because of the lethal properties of such agents, regulatory approval will require extensive testing under the Animal Rule. However, human (Hu) glycoprotein biologicals, such as BChE, present a challenge for assessing immunogenicity and efficacy in heterologous animal models since any immune responses to the small species differences in amino acids or glycans between the host and biologic may alter pharmacodynamics and preclude accurate efficacy testing; possibly underestimating their potential protective value in humans. To establish accurate pharmacokinetic and efficacy data, an homologous animal model has been developed in which native and PEGylated forms of CHO-derived rMaBChE were multiply injected into homologous macaques with no induction of antibody. These now serve as controls for assessing the pharmacokinetics and immunogenicity in macaques of multiple administrations of PEGylated and unmodified human rBChE (rHuBChE) by both intravenous (IV) and pulmonary routes. The results indicate that, except for maximal concentration (Cmax), the pharmacokinetic parameters following IV injection with heterologous PEG-rHuBChE were greatly reduced even after the first injection compared with homologous PEG-rMaBChE. Anti-HuBChE antibody responses were induced in all monkeys after the second and third administrations regardless of the route of delivery; impacting rates of clearance and usually resulting in reduced endogenous MaBChE activity. These data highlight the difficulties inherent in assessing pharmacokinetics and immunogenicity in animal models, but bode well for the efficacy and safety of rHuBChE pretreatments in homologous humans.


Subject(s)
Butyrylcholinesterase/immunology , Butyrylcholinesterase/pharmacokinetics , Lung , Recombinant Proteins/immunology , Recombinant Proteins/pharmacokinetics , Administration, Intravenous , Animals , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/pharmacology , Humans , Macaca , Organophosphorus Compounds/antagonists & inhibitors , Polyethylene Glycols/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL