Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Neuropharmacology ; 195: 108653, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34126123

ABSTRACT

Therapeutic vaccines offer a viable strategy to treat opioid use disorders (OUD) complementary to current pharmacotherapies. The candidate Oxy(Gly)4-sKLH vaccine targeting oxycodone displayed pre-clinical proof of efficacy, selectivity and safety, and it is now undergoing clinical evaluation. To further support its implementation in the clinic, this study tested critical in vivo neuropsychopharmacological properties of the Oxy(Gly)4-sKLH vaccine in rats. While repeated immunizations with Oxy(Gly)4-sKLH were necessary to maintain the antibody response overtime, exposure to free oxycodone did not boost oxycodone-specific antibody levels in vaccinated rats, limiting concerns of immune-related side effects. Immunization with Oxy(Gly)4-sKLH achieved sustained antibody titers over a period of five months following initial vaccination, supporting its potential for providing long-lasting protection. In vivo studies of selectivity showed that vaccination prevented oxycodone-induced but not methadone-induced antinociception, while still preserving the opioid antagonist naloxone's pharmacological effects. Vaccination did not interfere with fentanyl-induced antinociception or fentanyl distribution to the brain. These in vivo data confirm the previously reported in vitro selectivity profile of Oxy(Gly)4-sKLH. Vaccination extended oxycodone's half-life up to 25 h compared to control. While vaccination reduced the reinforcing efficacy of oxycodone in an intravenous self-administration model, signs of toxicity were not observed. These rodent studies confirm that active immunization with Oxy(Gly)4-sKLH induces highly specific and long-lasting antibodies which are effective in decreasing the reinforcing effects of oxycodone while preserving the efficacy of medications used to treat OUD and overdose.


Subject(s)
Antibodies , Brain/drug effects , Opioid-Related Disorders/drug therapy , Oxycodone , Vaccination , Vaccines/pharmacology , Animals , Fentanyl/pharmacology , Male , Rats
2.
Mol Pharm ; 16(6): 2364-2375, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31018096

ABSTRACT

This study focused on formulating conjugate vaccines targeting oxycodone and heroin for technology transfer, good manufacturing practice (GMP), and clinical evaluation. Lead vaccines used the highly immunogenic carrier protein keyhole limpet hemocyanin (KLH), which poses formulation problems because of its size. To address this barrier to translation, an oxycodone-based hapten conjugated to GMP-grade subunit KLH (OXY-sKLH) and adsorbed on alum adjuvant was studied with regard to carbodiimide coupling reaction time, buffer composition, purification methods for conjugates, conjugate size, state of aggregation, and protein/alum ratio. Vaccine formulations were screened for post-immunization antibody levels and efficacy in reducing oxycodone distribution to the brain in rats. While larger conjugates were more immunogenic, their size prevented characterization of the haptenation ratio by standard analytical methods and sterilization by filtration. To address this issue, conjugation chemistry and vaccine formulation were optimized for maximal efficacy, and conjugate size was measured by dynamic light scattering prior to adsorption to alum. An analogous heroin vaccine (M-sKLH) was also optimized for conjugation chemistry, formulated in alum, and characterized for potency against heroin in rats. Finally, this study found that the efficacy of OXY-sKLH was preserved when co-administered with M-sKLH, supporting the proof of concept for a bivalent vaccine formulation targeting both heroin and oxycodone. This study suggests methods for addressing the unique formulation and characterization challenges posed by conjugating small molecules to sKLH while preserving vaccine efficacy.


Subject(s)
Opioid-Related Disorders/prevention & control , Vaccines, Conjugate/chemistry , Animals , Hemocyanins/metabolism , Heroin/toxicity , Humans , Oxycodone/toxicity , Rats , Vaccines/chemistry , Vaccines/therapeutic use , Vaccines, Conjugate/therapeutic use
3.
PLoS One ; 12(12): e0184876, 2017.
Article in English | MEDLINE | ID: mdl-29194445

ABSTRACT

Among vaccines aimed at treating substance use disorders, those targeting opioids present several unique medication development challenges. 1) Opioid overdose is a common complication of abuse, so it is desirable for an opioid vaccine to block the toxic as well as the addictive effects of opioids. 2) It is important that an opioid vaccine not interfere with the action of opioid antagonists used to reverse opioid overdose or treat addiction. 3) Some opioids are immunosuppressive and chronic ongoing opioid use could interfere with vaccine immunogenicity. 4) Although antibody-bound oxycodone is unable to enter the brain because of its size, it might still be able to activate peripheral opioid receptors. To assess vaccine impact on opioid toxicity, rats vaccinated with oxycodone conjugated to keyhole limpet hemocyanin subunit dimer (OXY-dKLH) adsorbed to alum or controls vaccinated with dKLH were compared with regard to oxycodone-induced hotplate analgesia and oxycodone-induced respiratory depression and bradycardia. Vaccination shifted the dose-response curves to the right, representing protection, for each of these endpoints. Naloxone was equally effective in both OXY-dKLH and control groups, providing complete and rapid reversal of respiratory depression. The administration of a long-acting naltrexone formulation during vaccination did not impair vaccine immunogenicity in mice. Similarly, serum anti-oxycodone antibody titers were not altered by continuous morphine infusion during vaccination compared to opioid-naïve controls. Competitive ELISA assay showed negligible or low affinity of immune antiserum for endogenous opioids or opioid antagonists. In vitro receptor binding assays showed that antibody-bound oxycodone does not activate mu opioid receptors. These data support further study of OXY-dKLH as a potential treatment for oxycodone abuse and suggest that vaccination might also reduce the severity of oxycodone overdose.


Subject(s)
Opioid-Related Disorders/therapy , Oxycodone/immunology , Vaccines/therapeutic use , Animals , Antidotes/administration & dosage , Male , Mice , Naloxone/administration & dosage , Rats , Vaccines/adverse effects
4.
Drug Alcohol Depend ; 168: 76-88, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27627814

ABSTRACT

BACKGROUND: The popularity of electronic cigarettes (ECs) has increased dramatically despite their unknown health consequences. Because the abuse liability of ECs is one of the leading concerns of the Food and Drug Administration (FDA), models to assess it are urgently needed to inform FDA regulatory decisions regarding these products. The purpose of this study was to assess the relative abuse liability of an EC liquid compared to nicotine alone in rats. Because this EC liquid contains non-nicotine constituents that may enhance its abuse liability, we hypothesized that it would have greater abuse liability than nicotine alone. METHODS: Nicotine alone and nicotine dose-equivalent concentrations of EC liquid were compared in terms of their acute effects on intracranial self-stimulation (ICSS) thresholds, acquisition of self-administration, reinforcing efficacy (i.e., elasticity of demand), blockade of these behavioral effects by mecamylamine, nicotine pharmacokinetics and nicotinic acetylcholine receptor binding and activation. RESULTS: There were no significant differences between formulations on any measure, except that EC liquid produced less of an elevation in ICSS thresholds at high nicotine doses. CONCLUSIONS: Collectively, these findings suggest that the relative abuse liability of this EC liquid is similar to that of nicotine alone in terms of its reinforcing and reinforcement-enhancing effects, but that it may have less aversive/anhedonic effects at high doses. The present methods may be useful for assessing the abuse liability of other ECs to inform potential FDA regulation of those products.


Subject(s)
Electronic Nicotine Delivery Systems , Nicotine/administration & dosage , Reinforcement, Psychology , Self Administration , Self Stimulation/drug effects , Animals , Dose-Response Relationship, Drug , Male , Mecamylamine/pharmacology , Nicotinic Agonists/administration & dosage , Nicotinic Antagonists/pharmacokinetics , Rats , Rats, Sprague-Dawley
5.
Drug Alcohol Depend ; 141: 107-17, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24934691

ABSTRACT

Nicotine dependence plays a critical role in addiction to tobacco products, and thus contributes to a variety of devastating tobacco-related diseases (SGR 2014). Annual costs associated with smoking in the US are estimated to be between $289 and $333 billion. Effective interventions for nicotine dependence, especially in smokers, are a critical barrier to the eradication of tobacco-related diseases. This overview highlights research presented at the Plenary Symposium of Behavior, Biology and Chemistry: Translational Research in Addiction Conference (BBC), hosted by the UT Health Science Center San Antonio, on March 9-10, 2013. The Plenary Symposium focused on tobacco addiction, and covered topics ranging from basic science to national policy. As in previous years, the meeting brought together globally-renowned scientists, graduate student recruits, and young scientists from underrepresented populations in Texas and other states with the goal of fostering interest in drug addiction research in young generations.


Subject(s)
Behavior, Addictive/drug therapy , Smoking Cessation/methods , Smoking/drug therapy , Substance Withdrawal Syndrome/diagnosis , Tobacco Use Disorder/drug therapy , Behavior, Addictive/physiopathology , Humans , Research , Smoking/physiopathology , Substance Withdrawal Syndrome/physiopathology , Tobacco Use Disorder/physiopathology
6.
J Pharmacol Exp Ther ; 344(2): 397-406, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23220743

ABSTRACT

Morphine conjugate vaccines have effectively reduced behavioral effects of heroin in rodents and primates. To better understand how these effects are mediated, heroin and metabolite distribution studies were performed in rats in the presence and absence of vaccination. In non-vaccinated rats 6-monoacetylmorphine (6-MAM) was the predominant opioid in plasma and brain as early as 1 minute after i.v. administration of heroin and for up to 14 minutes. Vaccination with morphine conjugated to keyhole limpet hemocyanin (M-KLH) elicited high titers and concentrations of antibodies with high affinity for heroin, 6-MAM, and morphine. Four minutes after heroin administration vaccinated rats showed substantial retention of all three opioids in plasma compared to controls and reduced 6-MAM and morphine, but not heroin, distribution to brain. Administration of 6-MAM rather than heroin in M-KLH vaccinated rats showed a similar drug distribution pattern. Vaccination reduced heroin-induced analgesia and blocked heroin-induced locomotor activity throughout 2 weeks of repeated testing. Higher serum opioid-specific antibody concentrations were associated with higher plasma opioid concentrations, lower brain 6-MAM and morphine concentrations, and lower heroin-induced locomotor activity. Serum antibody concentrations over 0.2 mg/ml were associated with substantial effects on these measures. These data support a critical role for 6-MAM in mediating the early effects of i.v. heroin and suggest that reducing 6-MAM concentration in brain is essential to the efficacy of morphine conjugate vaccines.


Subject(s)
Behavior, Animal/drug effects , Heroin , Morphine/immunology , Vaccines, Conjugate/immunology , Adjuvants, Immunologic , Animals , Antibodies/blood , Binding, Competitive , Brain/metabolism , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay , Hemocyanins/immunology , Heroin/adverse effects , Heroin/immunology , Heroin/pharmacokinetics , Injections, Intravenous , Male , Morphine/blood , Morphine/pharmacokinetics , Morphine Derivatives/blood , Morphine Derivatives/immunology , Morphine Derivatives/metabolism , Motor Activity/drug effects , Protein Binding , Rats , Rats, Sprague-Dawley , Time Factors , Tissue Distribution , Vaccines, Conjugate/administration & dosage
7.
Vaccine ; 30(31): 4617-24, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22583811

ABSTRACT

Opioid conjugate vaccines have shown promise in animal models as a potential treatment for opioid addiction. Individual vaccines are quite specific and each targets only a limited number of structurally similar opioids. Since opioid users can switch or transition between opioids, we studied a bivalent immunization strategy of combining 2 vaccines that could target several of the most commonly abused opioids; heroin, oxycodone and their active metabolites. Morphine (M) and oxycodone (OXY) haptens were conjugated to keyhole limpet hemocyanin (KLH) through tetraglycine (Gly)(4) linkers at the C6 position. Immunization of rats with M-KLH alone produced high titers of antibodies directed against heroin, 6-monoacetylmorphine (6-MAM) and morphine. Immunization with OXY-KLH produced high titers of antibodies against oxycodone and oxymorphone. Immunization with the bivalent vaccine produced consistently high antibody titers against both immunogens. Bivalent vaccine antibody titers against the individual immunogens were higher than with the monovalent vaccines alone owing, at least in part, to cross-reactivity of the antibodies. Administration of a single concurrent intravenous dose of 6-MAM and oxycodone to rats immunized with the bivalent vaccine increased 6-MAM, morphine and oxycodone retention in serum and reduced the distribution of 6-MAM and oxycodone to brain. Vaccine efficacy correlated with serum antibody titers for both monovalent vaccines, alone or in combination. Efficacy of the individual vaccines was not compromised by their combined use. Consistent with the enhanced titers in the bivalent group, a trend toward enhanced pharmacokinetic efficacy with the bivalent vaccine was observed. These data support the possibility of co-administering two or more opioid vaccines concurrently to target multiple abusable opioids without compromising the immunogenicity or efficacy of the individual components.


Subject(s)
Brain/metabolism , Heroin/pharmacokinetics , Morphine Derivatives/pharmacokinetics , Oxycodone/pharmacokinetics , Vaccines/administration & dosage , Animals , Antibodies/blood , Antibody Specificity , Cross Reactions , Haptens , Heroin/immunology , Male , Mice , Mice, Inbred BALB C , Morphine/blood , Morphine/immunology , Morphine/pharmacokinetics , Morphine Derivatives/blood , Morphine Derivatives/immunology , Oxycodone/blood , Oxycodone/immunology , Rats , Vaccines, Combined/administration & dosage
8.
J Pharmacol Exp Ther ; 341(1): 225-32, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22262924

ABSTRACT

Opioid conjugate vaccines have shown promise in attenuating the behavioral effects of heroin or morphine in animals. The goal of this study was to extend this approach to oxycodone (OXY), a commonly abused prescription opioid. Haptens were generated by adding tetraglycine (Gly)(4) or hemisuccinate (HS) linkers at the 6-position of OXY. Immunization of rats with OXY(Gly)(4) conjugated to the carrier proteins bovine serum albumin (BSA) or keyhole limpet hemocyanin (KLH) produced high-titer antibodies to OXY and its metabolite oxymorphone with substantially lower affinities for other structurally related opioid agonists and antagonists. There was no measurable binding of antibody by the (Gly)(4) linker alone or off-target opioids methadone and buprenorphine. OXY(HS) conjugates were less immunogenic despite achieving protein haptenation ratios comparable to OXY(Gly)(4)-BSA. In rats given a single intravenous dose of OXY, immunization with OXY(Gly)(4)-KLH increased OXY protein binding and retention in serum while decreasing its unbound (free) concentration in plasma and distribution to brain. Vaccine efficacy correlated with serum antibody titers, and it was greatest in rats given the lowest OXY dose (0.05 mg/kg) but was significant even after a larger OXY dose (0.5 mg/kg), equivalent to the high end of the therapeutic range in humans. These effects of OXY(Gly)(4)-KLH on drug disposition were comparable to those of nicotine or cocaine vaccines that are in clinical trials as addiction treatments. Immunization with OXY(Gly)(4)-KLH also reduced OXY analgesia in a thermal nociception test. These data support further study of vaccination with the OXY(Gly)(4)-KLH immunogen as a potential treatment option for OXY abuse or addiction.


Subject(s)
Analgesia/methods , Antibody Formation , Brain/metabolism , Hot Temperature , Oxycodone/administration & dosage , Oxycodone/immunology , Vaccines, Conjugate/immunology , Animals , Antibodies/blood , Brain/immunology , Cattle , Chickens , Dose-Response Relationship, Immunologic , Hot Temperature/adverse effects , Male , Oxycodone/antagonists & inhibitors , Pain/blood , Pain/immunology , Pain/prevention & control , Pain Measurement/methods , Protein Binding/immunology , Rats , Rats, Sprague-Dawley , Vaccines, Conjugate/administration & dosage
9.
Biochem Pharmacol ; 83(4): 543-50, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22100986

ABSTRACT

Nicotine conjugate vaccine efficacy is limited by the concentration of nicotine-specific antibodies that can be reliably generated in serum. Previous studies suggest that the concurrent use of 2 structurally distinct nicotine immunogens in rats can generate additive antibody responses by stimulating distinct B cell populations. In the current study we investigated whether it is possible to identify a third immunologically distinct nicotine immunogen. The new 1'-SNic immunogen (2S)-N,N'-(disulfanediyldiethane-2,1-diyl)bis[4-(2-pyridin-3-ylpyrrolidin-1-yl)butanamide] conjugated to keyhole limpet hemocyanin (KLH) differed from the existing immunogens 3'-AmNic-rEPA and 6-CMUNic-BSA in linker position, linker composition, conjugation chemistry, and carrier protein. Vaccination of rats with 1'-SNic-KLH elicited high concentrations of high affinity nicotine-specific antibodies. The antibodies produced in response to 1'-SNic-KLH did not appreciably cross-react in ELISA with either 3'-AmNic-rEPA or 6-CMUNic-BSA or vice versa, showing that the B cell populations activated by each of these nicotine immunogens were non-overlapping and distinct. Nicotine retention in serum was increased and nicotine distribution to brain substantially reduced in rats vaccinated with 1'-SNic-KLH compared to controls. Effects of 1'-SNic-KLH on nicotine distribution were comparable to those of 3'-AmNic-rEPA which has progressed to late stage clinical trials as an adjunct to smoking cessation. These data show that it is possible to design multiple immunogens from a small molecule such as nicotine which elicit independent immune responses. This approach could be applicable to other addiction vaccines or small molecule targets as well.


Subject(s)
Antibodies/immunology , Antibody Specificity/immunology , Nicotine/immunology , Pyridines/immunology , Pyrrolidines/immunology , Vaccines, Synthetic/immunology , Animals , Antibodies/metabolism , Hemocyanins , Molecular Structure , Nicotine/chemistry , Pyridines/chemistry , Pyrrolidines/chemistry , Rats , Vaccines, Synthetic/chemistry
10.
Biochem Pharmacol ; 81(9): 1164-70, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21333633

ABSTRACT

Preclinical models of nicotine vaccine pharmacology have relied on i.v. or s.c. administration of nicotine. Models using cigarette smoke inhalation might more accurately simulate nicotine exposure in smokers. Nicotine vaccine effects were examined in rats using two cigarette smoke exposure models: a 10 min nose-only exposure (NSE) producing serum nicotine levels equivalent to the nicotine boost from 1 cigarette in a smoker, and a 2h whole-body exposure (WBE) producing serum nicotine levels similar to those associated with regular mid-day smoking. Vaccination prior to 10min smoke NSE reduced nicotine distribution to brain by 90%, comparable to its effect on nicotine administered i.v. Vaccination prior to 2 h smoke WBE reduced nicotine distribution to brain by 35%. The nicotine concentration in broncheoalveolar lavage (BAL) fluid obtained after 2 h WBE was increased by 230% in vaccinated rats but was also increased in rats passively immunized with a nicotine-specific monoclonal antibody, and so was likely due to transfer of antibody from serum rather than local production at the pulmonary mucosa. Nicotine-specific IgA was not detectable in BAL fluid, but titers in serum were appreciable at 21-25% of the IgG titer and could contribute to vaccine efficacy. Both vaccination and passive immunization are effective in reducing nicotine distribution to brain in rats when nicotine is delivered via inhaled cigarette smoke. These data validate results previously obtained in rodents for nicotine vaccines using i.v. or s.c. nicotine dosing and provide a quantitative method for studying aspects of nicotine exposure which are unique to cigarette smoke inhalation.


Subject(s)
Nicotine/immunology , Smoke , Vaccines/administration & dosage , Administration, Inhalation , Animals , Enzyme-Linked Immunosorbent Assay , Male , Nicotine/administration & dosage , Nicotine/pharmacokinetics , Rats , Rats, Sprague-Dawley , Nicotiana
11.
Int Immunopharmacol ; 8(11): 1589-94, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18656557

ABSTRACT

The efficacy of nicotine vaccines for smoking cessation is dependent upon their ability to elicit sufficiently high serum antibody concentrations. This study compared two nicotine immunogens representing different hapten presentations, 3'-aminomethyl nicotine conjugated to recombinant Pseudomonas exoprotein A (3'-AmNic-rEPA) and 6-carboxymethlureido nicotine conjugated to keyhole limpet hemocyanin (6-CMUNic-KLH), and assessed whether their concurrent administration would produce additive serum antibody concentrations in rats. Effects of vaccination on nicotine pharmacokinetics were also studied. Vaccination of rats with these immunogens produced non cross-reacting nicotine-specific antibodies (NicAb). Serum NicAb concentrations elicited by each individual immunogen were not affected by whether the immunogens were administered alone as monovalent vaccines or together as a bivalent vaccine. The total NicAb concentration in the bivalent vaccine group was additive compared to that of the monovalent vaccines alone. Higher serum NicAb concentrations, irrespective of which immunogen elicited the antibodies, were associated with greater binding of nicotine in serum, a lower unbound nicotine concentration in serum, and lower brain nicotine concentration. These results demonstrate that it is possible to design immunogens which provide distinct nicotine epitopes for immune presentation, and which produce additive serum antibody levels. The concurrent administration of these immunogens as a bivalent vaccine may provide a general strategy for enhancing the antibody response to small molecules such as nicotine.


Subject(s)
Bacterial Proteins/immunology , Hemocyanins/immunology , Nicotine/immunology , Smoking Cessation/methods , Smoking/therapy , Vaccination , Animals , Antigens, Bacterial/immunology , Bacterial Proteins/chemistry , Cross Reactions/immunology , Hemocyanins/chemistry , Nicotine/chemistry , Rats , Vaccines, Conjugate/chemistry , Vaccines, Conjugate/immunology
12.
Neurotoxicol Teratol ; 28(5): 589-96, 2006.
Article in English | MEDLINE | ID: mdl-16996246

ABSTRACT

Gestational exposure of rats to nicotine produces long-lasting alterations in brain development. Vaccination of adult female rats against nicotine reduces the distribution of maternally administered nicotine to fetal brain, suggesting that vaccination might protect against these effects. In the current study, the effects of vaccination on nicotine-induced changes in fetal (3)H-epibatidine binding and c-fos mRNA expression were evaluated using tissue from a previous pharmacokinetic study of vaccination. An intermittent nicotine dosing regimen designed to resemble nicotine intake in a smoker was administered from GD1-20. Peak nicotine levels in fetal brain were reduced by vaccination, whereas the chronic accumulation of nicotine in fetal brain was not. Gestational nicotine exposure produced significant increases in (125)I-epibatidine binding to brain and spinal cord on GD20, and decreased c-fos mRNA expression in fetal striatum, adrenal and lung. Vaccination did not significantly alter these effects. These data suggest that nicotine dosing, using a clinically relevant intermittent bolus dose regimen, produces substantial changes in fetal nicotinic receptor and c-fos mRNA expression. The decrease in c-fos mRNA expression contrasts with previously reported increases, and suggests that the nicotine dosing regimen used may influence its effects. The lack of effect of vaccination suggests that the cumulative exposure of fetal tissues to nicotine may influence the measured parameters to a greater extent than peak exposure levels.


Subject(s)
Gene Expression/physiology , Nicotine/immunology , Nicotine/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Nicotinic/metabolism , Vaccination , Analysis of Variance , Animals , Autoradiography/methods , Brain/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Embryo, Mammalian , Enzyme-Linked Immunosorbent Assay/methods , Female , Gene Expression/drug effects , Nicotinic Agonists/pharmacokinetics , Pregnancy , Protein Binding/drug effects , Proto-Oncogene Proteins c-fos/genetics , Pyridines/pharmacokinetics , RNA, Messenger/metabolism , Rats , Receptors, Nicotinic/drug effects , Tritium/pharmacokinetics
13.
Int Immunopharmacol ; 6(11): 1665-72, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16979120

ABSTRACT

Vaccination against nicotine to elicit the production of nicotine-specific antibodies is a potential treatment for tobacco addiction which reduces nicotine distribution from serum to brain. Vaccination of pregnant rats also reduces the distribution of maternally-administered nicotine to the fetal brain. Whether this is due to maternal antibody reducing the transfer of nicotine from mother to fetus, or to fetal antibody altering the distribution of nicotine within the fetus, is not clear. In the current study, passive immunization of rats with the murine monoclonal nicotine-specific antibody Nic311 was used as a surrogate for vaccination because antibody transfer to the fetus was anticipated to be lower than with vaccination. Pregnant rats received nicotine from gestational day (GD) 18-20 as frequent i.v. boluses to simulate nicotine exposure from smoking. Nic311 was administered at doses of 30, 80 or 240 mg/kg on GD 19. Fetal serum Nic311 levels on GD 20 were <3% of concurrent maternal levels, but concentrations of up to 20 ug/ml in fetal serum were obtained owing to the very high levels in maternal serum. Accumulation of the chronically administered nicotine, measured on GD 20, was not changed by Nic311 treatment in either maternal or fetal brain. The early distribution of nicotine to maternal brain, measured 5 min after a dose, was markedly reduced by Nic311, while the early distribution of nicotine to whole fetus and fetal brain was not substantially altered. These data suggest that the limited transfer of Nic311 to the fetus in turn limits the ability of Nic311 to reduce nicotine distribution to the fetal brain.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunization, Passive , Nicotine/immunology , Nicotine/pharmacokinetics , Animals , Antibodies, Monoclonal/pharmacokinetics , Brain/metabolism , Female , Maternal-Fetal Exchange , Mice , Nicotine/blood , Pregnancy , Rats , Rats, Sprague-Dawley
14.
J Pharmacol Exp Ther ; 317(2): 660-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16407464

ABSTRACT

Vaccination against nicotine blocks or attenuates nicotine-related behaviors relevant to addiction in rats. Passive immunization with nicotine-specific antibodies is an alternative to vaccination with the potential advantages of allowing control of antibody dose and affinity. In the current study, the effects of two antibodies on the distribution of nicotine to brain were evaluated during chronic nicotine administration in rats; the monoclonal antibody Nic311 (K(d) = 60 nM) and nicotine-specific antiserum (K(d) = 1.6 nM). Nicotine was administered via repeated i.v. bolus doses over 2 days and antibody was administered during the first day. Neither antibody appreciably reduced the chronic accumulation of nicotine in brain, despite high protein binding of nicotine in serum (98.9%) and a 73% reduction in the unbound serum nicotine concentration with the highest Nic311 dose. However, both antibodies substantially reduced the early distribution of nicotine to brain 5 min after a dose. The higher affinity antibody was no more effective than Nic311. The highest Nic311 dose produced serum antibody levels 10 times higher than those reported with vaccination. The efficacy of Nic311 was dose-related, with the highest dose producing a 76% decrease in the early distribution of nicotine to brain. These findings, along with previous data, suggest that the primary effect of passive immunization is to slow, rather than prevent, the distribution of nicotine to brain. In the setting of chronic nicotine dosing, antibodies with a moderate affinity for nicotine produced substantial effects on the early distribution of nicotine to brain and were as effective as higher affinity antibodies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Brain/metabolism , Immunization, Passive , Nicotine/pharmacokinetics , Tobacco Use Disorder/prevention & control , Animals , Antibodies, Monoclonal/administration & dosage , Antibody Affinity/immunology , Cotinine/blood , Male , Nicotine/blood , Nicotine/immunology , Rats , Rats, Sprague-Dawley , Tissue Distribution , Tobacco Use Disorder/immunology
15.
Drug Metab Dispos ; 33(7): 1056-61, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15843487

ABSTRACT

Vaccination against nicotine is being studied as a potential treatment for nicotine dependence. Some of the limitations of vaccination, such as variability in antibody titer and affinity, might be overcome by instead using passive immunization with nicotine-specific monoclonal antibodies. The effects of antibodies on nicotine distribution to brain were studied using nicotine-specific monoclonal antibodies (NICmAbs) with K(d) values ranging from 60 to 250 nM and a high-affinity polyclonal rabbit antiserum (K(d) = 1.6 nM). Pretreatment with NICmAbs substantially increased the binding of nicotine in serum after a single nicotine dose, reduced the unbound nicotine concentration in serum, and reduced the distribution of nicotine to brain. Efficacy was directly related to antibody affinity for nicotine. Efficacy of the highest affinity NICmAb, NICmAb311, was dose-related, with the highest dose reducing nicotine distribution to brain by 78%. NICmAb311 decreased nicotine clearance by 90% and prolonged the terminal half-life of nicotine by 120%. At equivalent doses, NICmAb311 was less effective than the higher affinity rabbit antiserum but comparable efficacy could be achieved by increasing the NICmAb311 dose. These data suggest that passive immunization with nicotine-specific monoclonal antibodies substantially alters nicotine pharmacokinetics in a manner similar to that previously reported for vaccination against nicotine. Antibody efficacy is a function of both dose and affinity for nicotine.


Subject(s)
Antibodies, Monoclonal/immunology , Brain/metabolism , Nicotine/immunology , Animals , Antibody Affinity , Enzyme-Linked Immunosorbent Assay , Mice , Nicotine/pharmacokinetics , Rabbits , Rats
16.
Int Immunopharmacol ; 3(7): 957-70, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12810353

ABSTRACT

Vaccination of rats against nicotine reduces nicotine distribution to brain even at nicotine doses greatly exceeding the estimated binding capacity of the available antibody. This observation suggests a differential effect by which vaccination reduces nicotine distribution to brain to a greater extent than to other tissues. To test this hypothesis, vaccinated rats received a single intravenous nicotine dose equal to twice the estimated binding capacity of nicotine-specific antibody in vaccinated rats. The total and bound serum nicotine concentrations were higher in the vaccinated rats compared to controls, while the unbound serum nicotine concentration was lower. Distribution of nicotine to brain was reduced in vaccinated rats in a time-dependent manner, with a greater reduction at 1 min (64%) than at 25 min (45%). Vaccination reduced nicotine distribution to muscle, testis, spleen, liver, heart, and kidney, but to a lesser extent than to brain, while nicotine distribution to fat was increased. Chronically infused nicotine showed a similarly altered pattern of tissue distribution in vaccinated rats, but differences were in general smaller than after a single nicotine dose; brain nicotine concentration was 24% lower in vaccinated rats, while lung nicotine concentration was higher. The presence of nicotine-specific antibody in tissues may have contributed to the increased nicotine concentrations in fat and lung. These data suggest that vaccination reduces nicotine distribution to brain not only by sequestering nicotine in serum but also by redirecting tissue distribution disproportionately away from brain, such that nicotine concentrations are reduced to a greater extent in brain than in other tissues.


Subject(s)
Nicotine/immunology , Nicotine/pharmacokinetics , Vaccines, Conjugate/immunology , Animals , Antibodies/blood , Antibodies/metabolism , Brain/metabolism , Immunization , Male , Nicotine/blood , Rats , Rats, Sprague-Dawley , Time Factors , Tissue Distribution , Vaccines, Conjugate/administration & dosage
17.
Nicotine Tob Res ; 5(3): 387-96, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12791522

ABSTRACT

Adverse outcomes from tobacco use may take decades to develop. Biomarkers are measures that can be used in the early stages of tobacco use to assess exposure to tobacco toxins or to predict adverse health outcomes with which they are associated. Examples of biomarkers include specific chemical components of tobacco or their metabolites; early biochemical, histological, or physiological effects; and early health effects. Mechanistically relevant and quantitatively valid biomarkers are essential for assessing the ultimate impact of new products, treatments, preventive measures, and public health policies on tobacco-related disease. The tobacco industry's recent introduction of a variety of new tobacco products or devices with implied claims of reduced health risks highlights the need to develop methods for assessing their potential for benefit or harm. A wide variety of biomarkers for tobacco exposure or harm has been studied. Although many questions about their use remain unanswered, substantial data exist regarding their validity and utility. This conference reviewed both the general issues surrounding biomarker use and the current state of knowledge regarding the most widely studied and promising biomarkers.


Subject(s)
Biomarkers/analysis , Environmental Exposure/analysis , Epidemiologic Studies , Smoking/adverse effects , Tobacco Smoke Pollution/adverse effects , Alkaloids/analysis , Ganglionic Stimulants/analysis , Hemoglobins/analysis , Hemoglobins/chemistry , Humans , Nicotine/analysis , Reproducibility of Results , Risk Assessment , Sensitivity and Specificity
18.
Tob Control ; 12(2): 189-94, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12773730

ABSTRACT

OBJECTIVE: This study assessed renters' preferences for official smoking policies in their buildings and their practices concerning restricting tobacco smoking in their apartments. DESIGN: Renters (n = 301) living in large apartment complexes in a suburb of Minneapolis, Minnesota, completed a mail survey. MAIN OUTCOME MEASURES: The survey asked about the official smoking policies in place in their apartment buildings, their preferences for policies, whether they had smelled tobacco smoke coming into their apartments from without, and, if so, what they had done about it. RESULTS: The majority of non-smokers (79%) preferred that their building be smoke-free. When asked to identify the current smoking policy in their buildings, residents disagreed substantially. Most renters (60%) reported smoke-free policies in their own apartments and another significant proportion (23%) restricted smoking to certain areas or occasions or persons. 75% thought that enforcing a smoke-free policy for guests would not be difficult. 53% of those in non-smoking households had smelled tobacco smoke in their apartments; most of these reported being bothered by it. However, very few complained to the building owner or manager (15.5%) or to the smoker (6.9%). CONCLUSIONS: The majority of non-smokers preferred that their buildings be smoke-free. A failure to report problems to apartment managers might be an impediment to instituting smoke-free policies in apartment buildings. The considerable disagreement among residents within apartment complexes about the current official smoking policy in their buildings suggests that policies are lacking or are not well communicated.


Subject(s)
Consumer Behavior , Health Knowledge, Attitudes, Practice , Housing/legislation & jurisprudence , Smoking/psychology , Adolescent , Adult , Aged , Female , Health Policy , Humans , Male , Middle Aged , Minnesota , Perception , Public Policy , Smoking/legislation & jurisprudence , Tobacco Smoke Pollution/legislation & jurisprudence , Tobacco Smoke Pollution/prevention & control
19.
J Pharmacol Exp Ther ; 305(2): 587-92, 2003 May.
Article in English | MEDLINE | ID: mdl-12606612

ABSTRACT

Cigarette smoking during pregnancy is associated with a variety of adverse fetal outcomes. Nicotine is a likely contributor to these adverse effects, with fetal brain as one target organ. Vaccination of adult male rats against nicotine has been shown to reduce nicotine distribution to the brain. The current study examined whether vaccination of female rats before pregnancy would reduce the distribution to fetal brain of a single nicotine dose administered during gestation. Female rats immunized with a nicotine conjugate vaccine received a single dose of nicotine 0.03 mg/kg i.v. on gestational day 16 to 22. Five minutes later, vaccinated rats had substantially higher bound and lower unbound serum nicotine concentration and lower brain nicotine concentration than controls. Fetal brain nicotine concentration was reduced by 43% in vaccinated rats, comparable to the reduction in the maternal brain nicotine concentration. The whole-fetus nicotine concentration was not altered by vaccination. A similar experiment was performed in which pregnant rats were passively immunized with rabbit nicotine-specific IgG 7 or 21 mg/kg just before nicotine dosing. The effects of passive immunization on nicotine distribution in the mother were IgG dose-related and the higher dose reduced nicotine distribution to fetal brain by 60%. These data suggest that vaccine effects on nicotine distribution to serum and brain are similar in pregnant female rats to those previously reported in adult males. Vaccination of female rats before pregnancy, or passive immunization during pregnancy, can reduce the exposure of fetal brain to a single dose of maternally administered nicotine.


Subject(s)
Brain/metabolism , Nicotine/immunology , Nicotine/pharmacokinetics , Nicotinic Agonists/immunology , Nicotinic Agonists/pharmacokinetics , Vaccination , Animals , Antibodies/chemistry , Brain/embryology , Female , Fetal Weight/drug effects , Haptens/immunology , Immunization, Passive , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Male , Pregnancy , Protein Binding , Rabbits/immunology , Rats , Tissue Distribution
20.
Exp Clin Psychopharmacol ; 9(2): 228-34, 2001 May.
Article in English | MEDLINE | ID: mdl-11518099

ABSTRACT

The ability of a nicotine vaccine to protect against nicotine-induced seizures was studied in rats. Groups of 10 rats were vaccinated with 3 doses of either a nicotine conjugate vaccine over 6 weeks to elicit high titers of nicotine-specific antibodies or with a control vaccine. Rats were then pretreated with a 1-week subcutaneous infusion of either nicotine 1 mg/kg/day or saline and then received a single 2 mg/kg ip dose of nicotine to provoke seizures. Vaccination reduced the incidence of seizures. The combination of vaccination and pretreatment with nicotine infusion was more effective than either treatment alone. These data suggest that vaccination is protective against this toxic effect of nicotine and that combining vaccination and chronic nicotine administration may provide a novel strategy for blocking some effects of nicotine.


Subject(s)
Nicotine/immunology , Nicotine/pharmacology , Nicotinic Agonists/immunology , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Seizures/chemically induced , Seizures/prevention & control , Vaccination , Animals , Brain/metabolism , Carrier Proteins , Enzyme-Linked Immunosorbent Assay , Haptens/immunology , Immunoglobulin E/blood , Infusions, Intravenous , Male , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...