Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
NPJ Vaccines ; 8(1): 107, 2023 Jul 24.
Article in English | MEDLINE | ID: mdl-37488109

ABSTRACT

Opioid use disorders (OUD) and overdose are public health threats worldwide. Widespread access to highly potent illicit synthetic opioids such as fentanyl is driving the recent rise in fatal overdoses. Vaccines containing fentanyl-based haptens conjugated to immunogenic carrier proteins offer a long-lasting, safe, and cost-effective strategy to protect individuals from overdose upon accidental or deliberate exposure to fentanyl and its analogs. Prophylactic or therapeutic active immunization with an anti-fentanyl vaccine induces the production of fentanyl-specific antibodies that bind the drug in the blood and prevent its distribution to the brain, which reduces its reinforcing effects and attenuates respiratory depression and bradycardia. To increase the efficacy of a lead anti-fentanyl vaccine, this study tested whether the incorporation of synthetic toll-like receptor (TLR) 4 and TLR7/8 agonists as vaccine adjuvants would increase vaccine efficacy against fentanyl challenge, overdose, and self-administration in either rats or Hanford miniature pigs. Formulation of the vaccine with a nucleolipid TLR7/8 agonist enhanced its immunogenicity and efficacy in preventing fentanyl-induced respiratory depression, analgesia, bradycardia, and self-administration in either rats or mini-pigs. These studies support the use of TLR7/8 adjuvants in vaccine formulations to improve their clinical efficacy against OUD and potentially other substance use disorders (SUD).

2.
Vaccine ; 40(23): 3244-3252, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35469698

ABSTRACT

Opioid use disorders (OUD) and overdose represent a public health threat, resulting in thousands of deaths annually worldwide. Vaccines offer a promising treatment for OUD and potentially the prevention of fatal overdoses. The Oxy(Gly)4-sKLH Conjugate Vaccine, Adsorbed (Oxy(Gly)4-sKLH) has shown promising pre-clinical efficacy at reducing the behavioral and pharmacological effects of oxycodone. To support its clinical evaluation, a GLP toxicology study was performed to address the safety of Oxy(Gly)4-sKLH. Sprague Dawley rats were vaccinated with either aluminum adjuvant (alum) or vaccine adsorbed on alum. Low and high doses of Oxy(Gly)4-sKLH, equivalent to a 1X or 47X human dose, respectively, were administered every two weeks for a total of four vaccinations. Both vaccine doses induced high antibody titers. Vaccine-related toxicity was assessed postmortem in one experimental group after receiving the fourth immunization of the vaccine's high dose. For the remaining experimental groups, rats were challenged with 1.5 mg/kg/day s.c. oxycodone for 7 days after the fourth vaccination to assess whether concurrent exposure to oxycodone in vaccinated animals resulted in toxicity. All rats, except a subset of the aluminum control and the high dose vaccine groups, were sacrificed following oxycodone exposure. These subsets were allowed a four weeks recovery period prior to euthanasia. In this study, no Oxy(Gly)4-sKLH-related hematology, clinical chemistry, urinalysis, body weight, organ weight, or anatomic pathology toxicological findings were observed. These results demonstrate that the Oxy(Gly)4-sKLH vaccine is well tolerated, is immunogenic even at low doses, and does not produce undesired side effects in rats.


Subject(s)
Drug Overdose , Opioid-Related Disorders , Aluminum , Animals , Opioid-Related Disorders/drug therapy , Opioid-Related Disorders/prevention & control , Oxycodone/therapeutic use , Rats , Rats, Sprague-Dawley , Vaccines, Conjugate
3.
PLoS One ; 16(7): e0254247, 2021.
Article in English | MEDLINE | ID: mdl-34329335

ABSTRACT

Use of nicotine-specific monoclonal antibodies (mAbs) to sequester and reduce nicotine distribution to brain has been proposed as a therapeutic approach to treat nicotine addiction (the basis of tobacco use disorder). A series of monoclonal antibodies with high affinity for nicotine (nic•mAbs) was isolated from B-cells of vaccinated smokers. Genes encoding 32 unique nicotine binding antibodies were cloned, and the mAbs expressed and tested by surface plasmon resonance to determine their affinity for S-(-)-nicotine. The highest affinity nic•mAbs had binding affinity constants (KD) between 5 and 67 nM. The 4 highest affinity nic•mAbs were selected to undergo additional secondary screening for antigen-specificity, protein properties (including aggregation and stability), and functional in vivo studies to evaluate their capacity for reducing nicotine distribution to brain in rats. The 2 most potent nic•mAbs in single-dose nicotine pharmacokinetic experiments were further tested in a dose-response in vivo study. The most potent lead, ATI-1013, was selected as the lead candidate based on the results of these studies. Pretreatment with 40 and 80 mg/kg ATI-1013 reduced brain nicotine levels by 56 and 95%, respectively, in a repeated nicotine dosing experiment simulating very heavy smoking. Nicotine self-administration was also significantly reduced in rats treated with ATI-1013. A pilot rat 30-day repeat-dose toxicology study (4x200mg/kg ATI-1013) in the presence of nicotine indicated no drug-related safety concerns. These data provide evidence that ATI-1013 could be a potential therapy for the treatment of nicotine addiction.


Subject(s)
Antibodies, Monoclonal , Antibody Affinity , Brain/metabolism , Nicotine , Tobacco Use Disorder , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antigen-Antibody Complex/chemistry , Humans , Nicotine/chemistry , Nicotine/pharmacokinetics , Rats , Rats, Sprague-Dawley , Tobacco Use Disorder/drug therapy , Tobacco Use Disorder/metabolism
4.
Drug Alcohol Depend ; 219: 108433, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33310485

ABSTRACT

BACKGROUND: The Food and Drug Administration (FDA) is considering setting a nicotine standard for tobacco products to reduce their addictiveness. Such a standard should account for the apparent greater vulnerability to nicotine addiction in some subpopulations, such as adolescents with depression. The present study examined whether the reinforcement threshold and elasticity of demand (i.e., reinforcing efficacy) for nicotine in a genetic inbred rat model of depression (Flinders Sensitive Line [FSL]) differs from an outbred control strain. METHODS: Acquisition of nicotine self-administration (NSA) across a wide range of nicotine doses was measured in both FSL and Sprague-Dawley (SD) control adolescent rats. At the highest dose, elasticity of demand was also measured. Nicotine pharmacokinetics was examined to determine whether it might modulate NSA, as it does smoking in humans. RESULTS: FSL rats acquired self-administration quicker and showed more inelastic demand (greater reinforcing efficacy) than SDs at the highest unit dose. However, there was no strain difference in the reinforcement threshold of nicotine. FSL rats exhibited faster nicotine clearance, larger volume of distribution, and lower plasma and brain nicotine concentrations. However, these differences were not consistently related to strain differences in NSA measures. CONCLUSION: These findings are consistent with studies showing greater dependence and reinforcing efficacy of cigarettes in smokers with depression and those with relatively fast nicotine metabolism. However, these findings also suggest that a nicotine standard to reduce initiation of tobacco use should be similarly effective in both the general adolescent population and those with depression.


Subject(s)
Depression/physiopathology , Nicotine/pharmacology , Tobacco Use Disorder/physiopathology , Animals , Disease Models, Animal , Elasticity , Humans , Rats , Rats, Sprague-Dawley , Reinforcement, Psychology , Self Administration , Smokers , Smoking
5.
Nanomedicine ; 20: 102023, 2019 08.
Article in English | MEDLINE | ID: mdl-31181264

ABSTRACT

The treatment efficacy of a nicotine vaccine largely relies on its ability to induce high titers of nicotine-specific antibodies. Due to its strong immune-potentiating effects, aluminum salt (Alum) has been commonly used as an adjuvant in various nicotine vaccine formulations. In this study, we attempted to improve the immunological performance of a hybrid nanoparticle-based nicotine vaccine (NanoNicVac) by co-administering it with Alum. It was found that Alum severely restricted the release of NanoNicVac at the site of injection. Moreover, Alum damaged the hybrid structure of the vaccine. In the animal trial, mice immunized with NanoNicVac alone achieved an anti-nicotine IgG titer of 3.5 ±â€¯0.2 × 104 after three injections. Unexpectedly, Alum with quantities of 125, 250, 500, and 1000 µg did not enhance the immunogenicity of NanoNicVac. In addition, Alum did not improve the ability of the vaccine to reduce the entry of nicotine into the brain.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/chemistry , Nanoparticles/chemistry , Nicotine/immunology , Vaccines/immunology , Animals , Dendritic Cells/metabolism , Endocytosis , Female , Liposomes , Mice, Inbred BALB C , Nanoparticles/ultrastructure , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Time Factors
6.
Mol Pharm ; 16(6): 2766-2775, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31075204

ABSTRACT

Adjuvants are a critical component for vaccines, especially for a poorly immunogenic antigen, such as nicotine. However, the impact of adjuvant release rate from a vaccine formulation on its immunogenicity has not been well illustrated. In this study, we fabricated a series of hybrid-nanoparticle-based nicotine vaccines to study the impact of adjuvant release rate on their immunological efficacy. It was found that the nanovaccine with a medium or slow adjuvant release rate induced a significantly higher anti-nicotine antibody titer than that with a fast release rate. Furthermore, the medium and slow adjuvant release rates resulted in a significantly lower brain nicotine concentration than the fast release rate after nicotine challenge. All findings suggest that adjuvant release rate affects the immunological efficacy of nanoparticle-based nicotine vaccines, providing a potential strategy to rationally designing vaccine formulations against psychoactive drugs or even other antigens. The hybrid-nanoparticle-based nicotine vaccine with an optimized adjuvant release rate can be a promising next-generation immunotherapeutic candidate against nicotine.


Subject(s)
Nanoparticles/chemistry , Nicotine/chemistry , Vaccines/chemistry , Adjuvants, Immunologic , Animals , Female , Kinetics , Mice , Mice, Inbred BALB C
7.
J Pharmacol Exp Ther ; 368(2): 282-291, 2019 02.
Article in English | MEDLINE | ID: mdl-30409833

ABSTRACT

Fentanyl is an extremely potent synthetic opioid that has been increasingly used to adulterate heroin, cocaine, and counterfeit prescription pills, leading to an increase in opioid-induced fatal overdoses in the United States, Canada, and Europe. A vaccine targeting fentanyl could offer protection against the toxic effects of fentanyl in both recreational drug users and others in professions at risk of accidental exposure. This study focuses on the development of a vaccine consisting of a fentanyl-based hapten (F) conjugated to keyhole limpet hemocyanin (KLH) carrier protein or to GMP-grade subunit KLH (sKLH). Immunization with F-KLH in mice and rats reduced fentanyl-induced hotplate antinociception, and in rats reduced fentanyl distribution to the brain compared with controls. F-KLH did not reduce the antinociceptive effects of equianalgesic doses of heroin or oxycodone in rats. To assess the vaccine effect on fentanyl toxicity, rats immunized with F-sKLH or unconjugated sKLH were exposed to increasing subcutaneous doses of fentanyl. Vaccination with F-sKLH shifted the dose-response curves to the right for both fentanyl-induced antinociception and respiratory depression. Naloxone reversed fentanyl effects in both groups, showing that its ability to reverse respiratory depression was preserved. These data demonstrate preclinical selectivity and efficacy of a fentanyl vaccine and suggest that vaccines may offer a therapeutic option in reducing fentanyl-induced side effects.


Subject(s)
Analgesics, Opioid/antagonists & inhibitors , Analgesics, Opioid/metabolism , Fentanyl/antagonists & inhibitors , Fentanyl/metabolism , Vaccines/pharmacology , Analgesics, Opioid/toxicity , Animals , Bradycardia/blood , Bradycardia/chemically induced , Bradycardia/prevention & control , Fentanyl/toxicity , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley , Respiratory Insufficiency/blood , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/prevention & control , Vaccines/therapeutic use
8.
Biomaterials ; 182: 72-81, 2018 11.
Article in English | MEDLINE | ID: mdl-30107271

ABSTRACT

Polyethylene glycol (PEG) has long been used in nanoparticle-based drug or vaccine delivery platforms. In this study, nano-nicotine vaccines (NanoNicVac) were PEGylated to different degrees to investigate the impact of PEG on the immunological efficacy of the vaccine. Hybrid nanoparticles with various degrees of PEGylation (2.5%-30%) were assembled. It was found that 30% PEGylation resulted in a hybrid nanoparticle of a compromised core-shell structure. A higher concentration of PEG also led to a slower cellular uptake of hybrid nanoparticles by dendritic cells. However, increasing the quantity of the PEG could effectively reduce nanoparticle aggregation during storage and improve the stability of the hybrid nanoparticles. Subsequently, nicotine vaccines were synthesized by conjugating nicotine haptens to the differently PEGylated hybrid nanoparticles. In both in vitro and in vivo studies, it was found that a nicotine vaccine with 20% PEGylation (NanoNicVac 20.0) was significantly more stable than the vaccines with lower PEGylation. In addition, NanoNicVac 20.0 induced a significantly higher anti-nicotine antibody titer of 3.7 ±â€¯0.6 × 104 in mice than the other NanoNicVacs with lower concentrations of PEG. In a subsequent pharmacokinetic study, the lowest brain nicotine concentration of 34 ±â€¯11 ng/g was detected in mice that were immunized with NanoNicVac 20.0. In addition, no apparent adverse events were observed in mice immunized with NanoNicVac. In summary, 20% PEGylation confers NanoNicVac with desirable safety, the highest stability, and the best immunological efficacy in mice.


Subject(s)
Drug Delivery Systems/methods , Haptens/administration & dosage , Nanoparticles/chemistry , Nicotine/immunology , Polyethylene Glycols/chemistry , Tobacco Use Disorder/prevention & control , Vaccines/administration & dosage , Animals , Female , Haptens/immunology , Humans , Liposomes/chemistry , Mice , Mice, Inbred BALB C , Smoking Cessation/methods , Tobacco Use Disorder/immunology , Vaccines/immunology
9.
BMC Biotechnol ; 18(1): 46, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30041697

ABSTRACT

BACKGROUND: The bacterial nicotine-degrading enzyme NicA2 isolated from P. putida was studied to assess its potential use in the treatment of tobacco dependence. RESULTS: Rats were pretreated with varying i.v. doses of NicA2, followed by i.v. administration of nicotine at 0.03 mg/kg. NicA2 had a rapid onset of action reducing blood and brain nicotine concentrations in a dose-related manner, with a rapid onset of action. A 5 mg/kg NicA2 dose reduced the nicotine concentration in blood by > 90% at 1 min after the nicotine dose, compared to controls. Brain nicotine concentrations were reduced by 55% at 1 min and 92% at 5 min post nicotine dose. To evaluate enzyme effects at a nicotine dosing rate equivalent to heavy smoking, rats pretreated with NicA2 at 10 mg/kg were administered 5 doses of nicotine 0.03 mg/kg i.v. over 40 min. Nicotine levels in blood were below the assay detection limit 3 min after either the first or fifth nicotine dose, and nicotine levels in brain were reduced by 82 and 84%, respectively, compared to controls. A 20 mg/kg NicA2 dose attenuated nicotine discrimination and produced extinction of nicotine self-administration (NSA) in most rats, or a compensatory increase in other rats, when administered prior to each daily NSA session. In rats showing compensation, increasing the NicA2 dose to 70 mg/kg resulted in extinction of NSA. An enzyme construct with a longer duration of action, via fusion with an albumin-binding domain, similarly reduced NSA in a 23 h nicotine access model at a dose of 70 mg/kg. CONCLUSIONS: These data extend knowledge of NicA2's effects on nicotine distribution to brain and its ability to attenuate addiction-relevant behaviors in rats and support its further investigation as a treatment for tobacco use disorder.


Subject(s)
Monoamine Oxidase/administration & dosage , Nicotine/metabolism , Animals , Brain/metabolism , Discrimination, Psychological , Dose-Response Relationship, Drug , Monoamine Oxidase/metabolism , Monoamine Oxidase/pharmacokinetics , Nicotine/blood , Nicotine/cerebrospinal fluid , Pseudomonas putida , Rats , Rats, Sprague-Dawley , Reinforcement, Psychology , Self Administration
10.
Nanomedicine ; 14(5): 1655-1665, 2018 07.
Article in English | MEDLINE | ID: mdl-29719216

ABSTRACT

A series of hybrid nanoparticle-based nicotine nanovaccines (NanoNicVac) were engineered in this work by conjugating potent carrier protein candidates (Keyhole limpet hemocyanin (KLH) multimer, KLH subunit, cross-reactive material 197 (CRM197), or tetanus toxoid (TT)) for enhanced immunological efficacy. NanoNicVac with CRM197 or TT were processed by dendritic cells more efficiently than that with KLH multimer or subunit. NanoNicVac carrying CRM197 or TT exhibited a significantly higher immunogenicity against nicotine and a considerably lower immunogenicity against carrier proteins than NanoNicVac carrying KLH multimer or subunit in mice. The in vivo results revealed that NanoNicVac with CRM197 or TT resulted in lower levels of nicotine in the brain of mice after nicotine challenge. All findings suggest that an enhanced immunological efficacy of NanoNicVac can be achieved by using CRM197 or TT instead of KLH or KLH subunit as carrier proteins, making NanoNicVac a promising next-generation immunotherapeutic candidate against nicotine addiction.


Subject(s)
Bacterial Proteins/immunology , Nanoparticles/administration & dosage , Nicotine/immunology , Tetanus Toxoid/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Bacterial Proteins/chemistry , Brain/immunology , Brain/metabolism , Female , Hemocyanins/chemistry , Hemocyanins/immunology , Immunotherapy , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Tetanus Toxoid/chemistry , Tobacco Use Disorder/immunology , Tobacco Use Disorder/prevention & control , Vaccines, Synthetic/chemistry
11.
J Pharmacol Exp Ther ; 365(2): 346-353, 2018 05.
Article in English | MEDLINE | ID: mdl-29535156

ABSTRACT

Heroin and oxycodone abuse occurs over a wide range of drug doses and by various routes of administration characterized by differing rates of drug absorption. The current study addressed the efficacy of a heroin vaccine [morphine hapten conjugated to keyhole limpet hemocyanin (M-KLH)] or oxycodone vaccine [oxycodone hapten conjugated to keyhole limpet hemocyanin (OXY-KLH)] for reducing drug distribution to brain after intravenous heroin or oxycodone, or subcutaneous oxycodone. Rats immunized with M-KLH or keyhole limpet hemocyanin (KLH) control received an intravenous bolus dose of 0.26 or 2.6 mg/kg heroin. Vaccination with M-KLH increased retention of heroin and its active metabolites 6-acetylmorphine (6-AM) and morphine in plasma compared with KLH controls, and reduced total opioid (heroin + 6-AM + morphine) distribution to brain but only at the lower heroin dose. Immunization also protected against respiratory depression at the lower heroin dose. Rats immunized with OXY-KLH or KLH control received 0.22 or 2.2 mg/kg oxycodone intravenously, the molar equivalent of the heroin doses. Immunization with OXY-KLH significantly reduced oxycodone distribution to brain after either oxycodone dose, although the magnitude of effect of immunization at the higher oxycodone dose was small (12%). By contrast, vaccination with OXY-KLH was more effective when oxycodone was administered subcutaneously rather than intravenously, reducing oxycodone distribution to brain by 44% after an oxycodone dose of 2.3 mg/kg. Vaccination also reduced oxycodone-induced antinociception. These data suggest that the efficacy of OXY-KLH and M-KLH opioid vaccines is highly dependent upon opioid dose and route of administration.


Subject(s)
Heroin/administration & dosage , Opioid-Related Disorders/prevention & control , Oxycodone/administration & dosage , Vaccines/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Drug Administration Routes , Heroin/metabolism , Heroin/pharmacology , Male , Oxycodone/metabolism , Oxycodone/pharmacology , Rats
12.
Biomaterials ; 155: 165-175, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29179132

ABSTRACT

Current clinically-tested nicotine vaccines have yet shown enhanced smoking cessation efficacy due to their low immunogenicity. Achieving a sufficiently high immunogenicity is a necessity for establishing a clinically-viable nicotine vaccine. This study aims to facilitate the immunogenicity of a hybrid nanoparticle-based nicotine vaccine by rationally incorporating toll-like receptor (TLR)-based adjuvants, including monophosphoryl lipid A (MPLA), Resiquimod (R848), CpG oligodeoxynucleotide 1826 (CpG ODN 1826), and their combinations. The nanoparticle-delivered model adjuvant was found to be taken up more efficiently by dendritic cells than the free counterpart. Nanovaccine particles were transported to endosomal compartments upon cellular internalization. The incorporation of single or dual TLR adjuvants not only considerably increased total anti-nicotine IgG titers but also significantly affected IgG subtype distribution in mice. Particularly, the nanovaccines carrying MPLA+R848 or MPLA+ODN 1826 generated a much higher anti-nicotine antibody titer than those carrying none or one adjuvant. Meanwhile, the anti-nicotine antibody elicited by the nanovaccine adjuvanted with MPLA+R848 had a significantly higher affinity than that elicited by the nanovaccine carrying MPLA+ODN 1826. Moreover, the incorporation of all the selected TLR adjuvants (except MPLA) reduced the brain nicotine levels in mice after nicotine challenge. Particularly, the nanovaccine with MPLA+R848 exhibited the best ability to reduce the level of nicotine entering the brain. Collectively, rational incorporation of TLR adjuvants could enhance the immunological efficacy of the hybrid nanoparticle-based nicotine vaccine, making it a promising next-generation immunotherapeutic candidate for treating nicotine addiction.


Subject(s)
Nanoparticles/chemistry , Nicotine/chemistry , Tobacco Use Disorder/prevention & control , Vaccines/blood , Adjuvants, Immunologic/chemistry , Animals , Imidazoles/chemistry , Immunotherapy , Lipid A/analogs & derivatives , Lipid A/chemistry , Mice , Oligodeoxyribonucleotides/chemistry
13.
Biomaterials ; 138: 46-56, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28551462

ABSTRACT

A lipid-polymeric hybrid nanoparticle-based next-generation nicotine nanovaccine was rationalized in this study to combat nicotine addiction. A series of nanovaccines, which had nicotine-haptens localized on carrier protein (LPKN), nanoparticle surface (LPNK), or both (LPNKN), were designed to study the impact of hapten localization on their immunological efficacy. All three nanovaccines were efficiently taken up and processed by dendritic cells. LPNKN induced a significantly higher immunogenicity against nicotine and a significantly lower anti-carrier protein antibody level compared to LPKN and LPNK. Meanwhile, it was found that the anti-nicotine antibodies elicited by LPKN and LPNKN bind nicotine stronger than those elicited by LPKN, and LPNK and LPNKN resulted in a more balanced Th1-Th2 immunity than LPKN. Moreover, LPNKN exhibited the best ability to block nicotine from entering the brain of mice. Collectively, the results demonstrated that the immunological efficacy of the hybrid nanoparticle-based nicotine vaccine could be enhanced by modulating hapten localization, providing a promising strategy to combatting nicotine addiction.


Subject(s)
Immunogenicity, Vaccine , Nicotine/immunology , Tobacco Use Disorder/therapy , Vaccines/immunology , Analysis of Variance , Animals , Brain/metabolism , Carrier Proteins/immunology , Female , Haptens/blood , Haptens/immunology , Haptens/metabolism , Lactic Acid/chemistry , Lipids/chemistry , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Nicotine/antagonists & inhibitors , Nicotine/blood , Nicotine/metabolism , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Th1-Th2 Balance , Vaccines/administration & dosage , Vaccines/adverse effects , Vaccines/pharmacokinetics
14.
ACS Comb Sci ; 19(5): 286-298, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28383252

ABSTRACT

Since the demonstration of nicotine vaccines as a possible therapeutic intervention for the effects of tobacco smoke, extensive effort has been made to enhance nicotine specific immunity. Linker modifications of nicotine haptens have been a focal point for improving the immunogenicity of nicotine, in which the evaluation of these modifications usually relies on in vivo animal models, such as mice, rats or nonhuman primates. Here, we present two in vitro screening strategies to estimate and predict the immunogenic potential of our newly designed nicotine haptens. One utilizes a competition enzyme-linked immunoabsorbent assay (ELISA) to profile the interactions of nicotine haptens or hapten-protein conjugates with nicotine specific antibodies, both polyclonal and monoclonal. Another relies on computational modeling of the interactions between haptens and amino acid residues near the conjugation site of the carrier protein to infer linker-carrier protein conjugation effect on antinicotine antibody response. Using these two in vitro methods, we ranked the haptens with different linkers for their potential as viable vaccine candidates. The ELISA-based hapten ranking was in an agreement with the results obtained by in vivo nicotine pharmacokinetic analysis. A correlation was found between the average binding affinity (IC50) of the haptens to an anti-Nic monoclonal antibody and the average brain nicotine concentration in the immunized mice. The computational modeling of hapten and carrier protein interactions helps exclude conjugates with strong linker-carrier conjugation effects and low in vivo efficacy. The simplicity of these in vitro screening strategies should facilitate the selection and development of more effective nicotine conjugate vaccines. In addition, these data highlight a previously under-appreciated contribution of linkers and hapten-protein conjugations to conjugate vaccine immunogenicity by virtue of their inclusion in the epitope that binds and activates B cells.


Subject(s)
Antibodies/chemistry , Haptens/chemistry , Nicotine/chemistry , Animals , Antibodies/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , B-Lymphocytes/metabolism , Computer Simulation , Enzyme-Linked Immunosorbent Assay , Haptens/immunology , Humans , Mice, Inbred BALB C , Nicotine/immunology , Nicotine/pharmacokinetics , Streptavidin/chemistry , Vaccines, Conjugate/chemistry , Vaccines, Conjugate/immunology
15.
Biomaterials ; 123: 107-117, 2017 04.
Article in English | MEDLINE | ID: mdl-28167389

ABSTRACT

Although vaccination is a promising way to combat nicotine addiction, most traditional hapten-protein conjugate nicotine vaccines only show limited efficacy due to their poor recognition and uptake by immune cells. This study aimed to develop a hybrid nanoparticle-based nicotine vaccine with improved efficacy. The focus was to study the impact of hapten density on the immunological efficacy of the proposed hybrid nanovaccine. It was shown that the nanovaccine nanoparticles were taken up by the dendritic cells more efficiently than the conjugate vaccine, regardless of the hapten density on the nanoparticles. At a similar hapten density, the nanovaccine induced a significantly stronger immune response against nicotine than the conjugate vaccine in mice. Moreover, the high- and medium-density nanovaccines resulted in significantly higher anti-nicotine antibody titers than their low-density counterpart. Specifically, the high-density nanovaccine exhibited better immunogenic efficacy, resulting in higher anti-nicotine antibody titers and lower anti-carrier protein antibody titers than the medium- and low-density versions. The high-density nanovaccine also had the best ability to retain nicotine in serum and to block nicotine from entering the brain. These results suggest that the hybrid nanoparticle-based nicotine vaccine can elicit strong immunogenicity by modulating the hapten density, thereby providing a promising next-generation immunotherapeutic strategy against nicotine addiction.


Subject(s)
Brain/immunology , Haptens/immunology , Nanoconjugates/chemistry , Nicotine/immunology , Tobacco Use Disorder/immunology , Tobacco Use Disorder/prevention & control , Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacokinetics , Adjuvants, Immunologic/pharmacology , Animals , Brain/drug effects , Female , Haptens/administration & dosage , Mice , Mice, Inbred BALB C , Nanocapsules/chemistry , Nanocapsules/ultrastructure , Nanoconjugates/ultrastructure , Tobacco Use Cessation Devices , Vaccination/methods , Vaccines/administration & dosage
16.
Nanomedicine ; 13(2): 443-454, 2017 02.
Article in English | MEDLINE | ID: mdl-27520729

ABSTRACT

Traditional hapten-protein conjugate nicotine vaccines have shown less than desired immunological efficacy due to their poor recognition and internalization by immune cells. We developed a novel lipid-polymeric hybrid nanoparticle-based nicotine vaccine to enhance the immunogenicity of the conjugate vaccine, and studied the influence of particle size on its immunogenicity and pharmacokinetic efficacy. The results demonstrated that the nanovaccines, regardless of size, could induce a significantly stronger immune response against nicotine compared to the conjugate vaccine. Particularly, a significantly higher anti-nicotine antibody titer was achieved by the 100 compared to the 500nm nanovaccine. In addition, both the 100 and 500nm nanovaccines reduced the distribution of nicotine into the brain significantly. The 100nm nanovaccine exhibited better pharmacokinetic efficacy than the 500nm nanovaccine in the presence of alum adjuvant. These results suggest that a lipid-polymeric nanoparticle-based nicotine vaccine is a promising candidate to treat nicotine dependence.


Subject(s)
Nanoparticles , Nicotine/pharmacology , Vaccines , Adjuvants, Immunologic , Animals , Female , Mice, Inbred BALB C , Particle Size , Tobacco Use Disorder/therapy
17.
J Biomed Nanotechnol ; 11(12): 2197-210, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26510313

ABSTRACT

Tobacco addiction is the second-leading cause of death in the world. Due to the nature of nicotine (a small molecule), finding ways to combat nicotine's deleterious effects has been a constant challenge to the society and the medical field. In the present work, a novel anti-nicotine vaccine based on nanohorn supported liposome nanoparticles (NsL NPs) was developed. The nano-vaccine was constructed by using negatively charged carbon nanohorns as a scaffold for the assembly of cationic liposomes, which allow the conjugation of hapten conjugated carrier proteins. The assembled bio-nanoparticles are stable. Mice were immunized subcutaneously with the nano-vaccine, which induced high titer and high affinity of nicotine specific antibodies in mice. Furthermore, no evidence of clinical signs or systemic toxicity followed multiple administrations of NsL-based anti-nicotine vaccine. These results suggest that NsL-based anti-nicotine vaccine is a promising candidate in treating nicotine dependence and could have potential to significantly contribute to smoking cessation.


Subject(s)
Carbon/chemistry , Nanoparticles/chemistry , Nicotine/immunology , Vaccines/administration & dosage , Vaccines/immunology , Animals , Antibody Affinity , Antibody Specificity , Body Weight/immunology , Drinking/immunology , Female , Immunoglobulin G/immunology , Liposomes , Mice , Mice, Inbred BALB C , Nicotine/chemistry , Particle Size , Th1 Cells/immunology , Th2 Cells/immunology , Tobacco Use Disorder/prevention & control , Vaccination
19.
Drug Alcohol Depend ; 151: 181-93, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25891231

ABSTRACT

BACKGROUND: The FDA is considering reducing the nicotine content in tobacco products as a population-based strategy to reduce tobacco addiction. Research is needed to determine the threshold level of nicotine needed to maintain smoking and the extent of compensatory smoking that could occur during nicotine reduction. Sources of variability in these measures across sub-populations also need to be identified so that policies can take into account the risks and benefits of nicotine reduction in vulnerable populations. METHODS: The present study examined these issues in a rodent nicotine self-administration model of nicotine reduction policy to characterize individual differences in nicotine reinforcement thresholds, degree of compensation, and elasticity of demand during progressive reduction of the unit nicotine dose. The ability of individual differences in baseline nicotine intake and nicotine pharmacokinetics to predict responses to dose reduction was also examined. RESULTS: Considerable variability in the reinforcement threshold, compensation, and elasticity of demand was evident. High baseline nicotine intake was not correlated with the reinforcement threshold, but predicted less compensation and less elastic demand. Higher nicotine clearance predicted low reinforcement thresholds, greater compensation, and less elastic demand. Less elastic demand also predicted lower reinforcement thresholds. CONCLUSIONS: These findings suggest that baseline nicotine intake, nicotine clearance, and the essential value of nicotine (i.e. elasticity of demand) moderate the effects of progressive nicotine reduction in rats and warrant further study in humans. They also suggest that smokers with fast nicotine metabolism may be more vulnerable to the risks of nicotine reduction.


Subject(s)
Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Tobacco Use Disorder/prevention & control , Tobacco Use Disorder/psychology , Animals , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Extinction, Psychological/drug effects , Female , Individuality , Male , Nicotine/pharmacokinetics , Nicotinic Agonists/pharmacokinetics , Predictive Value of Tests , Rats , Rats, Sprague-Dawley , Reinforcement, Psychology , Self Administration
20.
PLoS One ; 10(3): e0118787, 2015.
Article in English | MEDLINE | ID: mdl-25742165

ABSTRACT

Passive immunization with monoclonal antibodies (mAbs) against (+)-methamphetamine (METH) is being evaluated for the treatment of METH addiction. A human/mouse chimeric form of the murine anti-METH mAb7F9 has entered clinical trials. This study examined the effects of murine mAb7F9 on certain addiction-related behavioral effects of METH in rats as measured using intracranial self-stimulation (ICSS). Initial studies indicated that acute METH (0.1-0.56 mg/kg, s.c.) lowered the minimal (threshold) stimulation intensity that maintained ICSS. METH (0.3 mg/kg, s.c.) also blocked elevations in ICSS thresholds (anhedonia-like behavior) during spontaneous withdrawal from a chronic METH infusion (10 mg/kg/day x 7 days). In studies examining effects of i.v. pretreatment with mAb7F9 (at 30, 100, or 200 mg/kg), 200 mg/kg blocked the ability of an initial injection of METH (0.3 mg/kg, s.c.) to reduce baseline ICSS thresholds, but was less capable of attenuating the effect of subsequent daily injections of METH. MAb7F9 (200 mg/kg) also produced a small but significant reduction in the ability of METH (0.3 mg/kg, s.c.) to reverse METH withdrawal-induced elevations in ICSS thresholds. These studies demonstrate that mAb7F9 can partially attenuate some addiction-related effects of acute METH in an ICSS model, and provide some support for the therapeutic potential of mAb7F9 for the treatment of METH addiction.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Brain/drug effects , Methamphetamine/administration & dosage , Methamphetamine/immunology , Self Stimulation , Animals , Behavior, Addictive/immunology , Brain/immunology , Immunization, Passive , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...