1
|
Lu T, Li X, Zheng W, Kuang C, Wu B, Liu X, Xue Y, Shi J, Lu L, Han Y. Vaccines to Treat Substance Use Disorders: Current Status and Future Directions. Pharmaceutics 2024; 16:84. [PMID: 38258095 PMCID: PMC10820210 DOI: 10.3390/pharmaceutics16010084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/24/2024] Open
Abstract
Addiction, particularly in relation to psychostimulants and opioids, persists as a global health crisis with profound social and economic ramifications. Traditional interventions, including medications and behavioral therapies, often encounter limited success due to the chronic and relapsing nature of addictive disorders. Consequently, there is significant interest in the development of innovative therapeutics to counteract the effects of abused substances. In recent years, vaccines have emerged as a novel and promising strategy to tackle addiction. Anti-drug vaccines are designed to stimulate the immune system to produce antibodies that bind to addictive compounds, such as nicotine, cocaine, morphine, methamphetamine, and heroin. These antibodies effectively neutralize the target molecules, preventing them from reaching the brain and eliciting their rewarding effects. By obstructing the rewarding sensations associated with substance use, vaccines aim to reduce cravings and the motivation to engage in drug use. Although anti-drug vaccines hold significant potential, challenges remain in their development and implementation. The reversibility of vaccination and the potential for combining vaccines with other addiction treatments offer promise for improving addiction outcomes. This review provides an overview of anti-drug vaccines, their mechanisms of action, and their potential impact on treatment for substance use disorders. Furthermore, this review summarizes recent advancements in vaccine development for each specific drug, offering insights for the development of more effective and personalized treatments capable of addressing the distinct challenges posed by various abused substances.
Collapse
Affiliation(s)
- Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; (T.L.); (X.L.); (Y.X.); (J.S.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xue Li
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; (T.L.); (X.L.); (Y.X.); (J.S.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wei Zheng
- Peking-Tsinghua Centre for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China;
| | - Chenyan Kuang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China;
| | - Bingyi Wu
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China;
| | - Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China;
| | - Yanxue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; (T.L.); (X.L.); (Y.X.); (J.S.)
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; (T.L.); (X.L.); (Y.X.); (J.S.)
| | - Lin Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; (T.L.); (X.L.); (Y.X.); (J.S.)
- Peking-Tsinghua Centre for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China;
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China;
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; (T.L.); (X.L.); (Y.X.); (J.S.)
| |
Collapse
|
2
|
Sabato B, Augusto PSDA, Lima Gonçalves Pereira R, Coutinho Batista Esteves F, Caligiorne SM, Rodrigues Dias Assis B, Apolo Correia Marcelino S, Pires do Espírito Santo L, Dias Dos Reis K, Da Silva Neto L, Goulart G, de Fátima Â, Pierezan F, Toshio Fujiwara R, Castro M, Garcia F. Safety and immunogenicity of the anti-cocaine vaccine UFMG-VAC-V4N2 in a non-human primate model. Vaccine 2023; 41:2127-2136. [PMID: 36822966 DOI: 10.1016/j.vaccine.2023.02.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 02/01/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
A promising strategy for cocaine addiction treatment is the anti-drug vaccine. These vaccines induce the production of anticocaine antibodies, capable of linking to cocaine, and decrease the passage of cocaine throughout the blood-brain barrier, decreasing drug activity in the brain. Our research group developed a new vaccine candidate, the UFMG-V4N2, to treat cocaine use disorders (CUD) using an innovative carrier based on calixarenes. This study assessed the safety and immunogenicity of the anti-cocaine vaccine UFMG-VAC-V4N2 in a non-human primate toxicity study using single and multiple vaccine doses. The UFMG-VAC-V4N2 yielded only mild effects in the injection site and did not influence the general health, feeding behavior, or hematological, renal, hepatic, or metabolic parameters in the vaccinated marmosets. The anti-cocaine vaccine UFMG-VAC-V4N2 presented a favorable safety profile and induced the expected immune response in a non-human primate model of Callithrix penicillata. This preclinical UFMG-VAC-V4N2 study responds to the criteria required by international regulatory agencies contributing to future anticocaine clinical trials of this anti-cocaine vaccine.
Collapse
Affiliation(s)
- Brian Sabato
- Center of research on Health Vulnerability (Núcleo de Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil; Pós-graduação em Neurociências, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte - MG, Brazil.
| | - Paulo Sérgio de Almeida Augusto
- Center of research on Health Vulnerability (Núcleo de Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil; Pós-graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte - MG, Brazil.
| | - Raissa Lima Gonçalves Pereira
- Center of research on Health Vulnerability (Núcleo de Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil; Pós-graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte - MG, Brazil.
| | - Felipe Coutinho Batista Esteves
- Laboratório de Imunologia e Genômica de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte - MG, Brazil
| | - Sordaini M Caligiorne
- Center of research on Health Vulnerability (Núcleo de Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| | - Bruna Rodrigues Dias Assis
- Center of research on Health Vulnerability (Núcleo de Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil; Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| | | | - Larissa Pires do Espírito Santo
- Center of research on Health Vulnerability (Núcleo de Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil; Pós-graduação em Neurociências, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte - MG, Brazil.
| | - Karine Dias Dos Reis
- Center of research on Health Vulnerability (Núcleo de Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Leonardo Da Silva Neto
- Grupo de Estudos em Química Orgânica e Biológica (GEQOB), Departamento de Química, Universidade Federal de Minas Gerias (UFMG), Belo Horizonte, MG, Brazil
| | - Gisele Goulart
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| | - Ângelo de Fátima
- Grupo de Estudos em Química Orgânica e Biológica (GEQOB), Departamento de Química, Universidade Federal de Minas Gerias (UFMG), Belo Horizonte, MG, Brazil.
| | - Felipe Pierezan
- Escola de Veterinária, Universidade Federal de Minas Gerias (UFMG), Belo Horizonte, MG, Brazil.
| | - Ricardo Toshio Fujiwara
- Laboratório de Imunologia e Genômica de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte - MG, Brazil.
| | - Maila Castro
- Center of research on Health Vulnerability (Núcleo de Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil; Pós-graduação em Neurociências, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte - MG, Brazil.
| | - Frederico Garcia
- Center of research on Health Vulnerability (Núcleo de Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil; Pós-graduação em Neurociências, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte - MG, Brazil; Pós-graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte - MG, Brazil.
| |
Collapse
|
3
|
Abstract
The current addiction crisis has destroyed a multitude of lives, leaving millions of fatalities worldwide in its wake. At the same time, various governmental agencies dedicated to solving this seemingly never-ending dilemma have not yet succeeded or delivered on their promises. We understand that addictive behavioral seeking is a multi-faceted neurobiological and spiritually complicated phenomenon. However, although the substitution replacement approach, especially to treat Opioid Use Disorder (OUD), has importance for harm reduction in the short term, it does not bring about a harm-free recovery or prevention. Instead, we propose a promising novel approach that uses genetic risk testing with induction of dopamine homeostasis and an objective Brain Health Check during youth. Our model involves a six-hit approach known as the "Reward Dysregulation Syndrome Solution System," which can identify addiction risk and target the root cause of addiction, dopamine dysregulation. While we applaud all past sophisticated neurogenetic and neuropharmacological research, our opinion is that in the long term, addiction scientists and clinicians might characterize preaddiction using tests; for example, administering the validated RDSQuestionarre29, genetic risk assessment, a modified brain health check, or diagnostic framing of mild to moderate Substance Use Disorder (SUD). The preaddiction concept could incentivize the development of interventions to prevent addiction from developing in the first place and target and treat neurotransmitter imbalances and other early indications of addiction. WC 222.
Collapse
|
4
|
de Almeida Augusto PS, Pereira RLG, Caligiorne SM, Sabato B, Assis BRD, do Espírito Santo LP, Dos Reis KD, Castro Goulart GA, de Fátima Â, de Castro Lourenço das Neves M, Garcia FD. The GNE-KLH anti-cocaine vaccine protects dams and offspring from cocaine-induced effects during the prenatal and lactating periods. Mol Psychiatry 2021; 26:7784-7791. [PMID: 34381172 DOI: 10.1038/s41380-021-01210-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/03/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023]
Abstract
Protecting children from prenatal cocaine exposure is a significant challenge for physicians and childbearing women with cocaine use disorder. Cocaine use is highly prevalent among reproductive-aged women and prenatal cocaine exposure produces obstetric, foetal neurodevelopmental and long-term behavioural impairments. Cocaine crosses the maternal and foetal blood-brain barrier and the placenta by diffusion. The best approach to prevent prenatal cocaine exposure is to stop cocaine use. However, only 25% of cocaine users can discontinue their use during pregnancy. Anti-cocaine vaccination decreases cocaine passage through the blood-brain barrier. This study describes an innovative approach for preventing prenatal cocaine exposure using the GNE-KLH anti-cocaine vaccine, a novel use for the named anti-drug vaccines. Here, we show that anti-cocaine vaccination with GNE-KLH produced and maintained anti-cocaine IgG antibody titres and avidity during pregnancy. These antibodies protected the pregnant rats and their pups against prenatal cocaine damage during pregnancy until weaning. The present work is the first preclinical evidence of the efficacy of an innovative mechanism to prevent prenatal cocaine exposure damage, a worldwide public health care issue. In the future, this mechanism may be useful in pregnant women with cocaine use disorder. Further studies to understand the mechanisms of how anti-cocaine antibodies exert their protective effects in pregnancy are warranted.
Collapse
Affiliation(s)
- Paulo Sérgio de Almeida Augusto
- Centre of Research on Health Vulnerability (Núcleo de Pesquisa em Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, sala 240, Belo Horizonte, MG, Brazil.,Pós-graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, Belo Horizonte, MG, Brazil
| | - Raissa Lima Gonçalves Pereira
- Centre of Research on Health Vulnerability (Núcleo de Pesquisa em Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, sala 240, Belo Horizonte, MG, Brazil.,Pós-graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, Belo Horizonte, MG, Brazil
| | - Sordaini Maria Caligiorne
- Centre of Research on Health Vulnerability (Núcleo de Pesquisa em Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, sala 240, Belo Horizonte, MG, Brazil
| | - Brian Sabato
- Centre of Research on Health Vulnerability (Núcleo de Pesquisa em Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, sala 240, Belo Horizonte, MG, Brazil.,Pós-graduação em Neurociências, Universidade Federal de Minas Gerais (UFMG). Avenida Antônio Carlos, 6627, Belo Horizonte, MG, Brazil
| | - Bruna Rodrigues Dias Assis
- Centre of Research on Health Vulnerability (Núcleo de Pesquisa em Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, sala 240, Belo Horizonte, MG, Brazil.,Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais (UFMG). Avenida Antônio Carlos, 6627, Belo Horizonte, MG, Brazil
| | - Larissa Pires do Espírito Santo
- Centre of Research on Health Vulnerability (Núcleo de Pesquisa em Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, sala 240, Belo Horizonte, MG, Brazil.,Pós-graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, Belo Horizonte, MG, Brazil
| | - Karine Dias Dos Reis
- Centre of Research on Health Vulnerability (Núcleo de Pesquisa em Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, sala 240, Belo Horizonte, MG, Brazil
| | - Gisele Assis Castro Goulart
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais (UFMG). Avenida Antônio Carlos, 6627, Belo Horizonte, MG, Brazil
| | - Ângelo de Fátima
- Grupo de Estudos em Química Orgânica e Biológica (GEQOB), Departament of Chemistry, Universidade Federal de Minas Gerias (UFMG). Avenida Antônio Carlos, 6627, Belo Horizonte, MG, Brazil
| | - Maila de Castro Lourenço das Neves
- Centre of Research on Health Vulnerability (Núcleo de Pesquisa em Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, sala 240, Belo Horizonte, MG, Brazil.,Pós-graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, Belo Horizonte, MG, Brazil.,Pós-graduação em Neurociências, Universidade Federal de Minas Gerais (UFMG). Avenida Antônio Carlos, 6627, Belo Horizonte, MG, Brazil
| | - Frederico Duarte Garcia
- Centre of Research on Health Vulnerability (Núcleo de Pesquisa em Vulnerabilidade e Saúde - NAVES), Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, sala 240, Belo Horizonte, MG, Brazil. .,Pós-graduação em Medicina Molecular, Universidade Federal de Minas Gerais (UFMG). Av. Alfredo Balena, 190, Belo Horizonte, MG, Brazil. .,Pós-graduação em Neurociências, Universidade Federal de Minas Gerais (UFMG). Avenida Antônio Carlos, 6627, Belo Horizonte, MG, Brazil.
| |
Collapse
|
5
|
Blum K, Cadet JL, Gold MS. Psychostimulant use disorder emphasizing methamphetamine and the opioid -dopamine connection: Digging out of a hypodopaminergic ditch. J Neurol Sci 2021; 420:117252. [PMID: 33279726 DOI: 10.1016/j.jns.2020.117252] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Approved food and drug administration (FDA) medications to treat Psychostimulant Use Disorder (PUD) are needed. Both acute and chronic neurological deficits related to the neurophysiological effects of these powerfully addictive drugs can cause stroke and alterations in mood and cognition. OBJECTIVE This article presents a brief review of the psychiatric and neurobiological sequelae of methamphetamine use disorder, some known neurogenetic associations impacted by psychostimulants, and explores treatment modalities and outcomes. HYPOTHESIS The authors propose that gentle D2 receptor stimulation accomplished via some treatment modalities can induce dopamine release, causing alteration of D2-directed mRNA and thus enhanced function of D2 receptors in the human. This proliferation of D2 receptors, in turn, will induce the attenuation of craving behavior, especially in genetically compromised high-risk populations. DISCUSSION A better understanding of the involvement of molecular neurogenetic opioid, mesolimbic dopamine, and psychostimulant connections in "wanting" supports this hypothesis. While both scientific and, clinical professionals search for an FDA approved treatment for PUD the induction of dopamine homeostasis, via activation of the brain reward circuitry, offers treatment for underlying neurotransmitter functional deficits, potential prophylaxis, and support for recovery efforts. CONCLUSION Dopamine regulation may help people dig out of their hypodopaminergia ditch.
Collapse
Affiliation(s)
- Kenneth Blum
- Graduate College, Western University Health Sciences, Pomona, CA, Baltimore, MD, United States of America.
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH National Institute on Drug Abuse, United States of America
| | - Mark S Gold
- Department of Psychiatry, Washington University, St Louis, MO, United States of America.
| |
Collapse
|
6
|
Psychopharmacology: neuroimmune signaling in psychiatric disease-developing vaccines against abused drugs using toll-like receptor agonists. Psychopharmacology (Berl) 2019; 236:2899-2907. [PMID: 30726515 DOI: 10.1007/s00213-019-5176-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 01/16/2019] [Indexed: 01/05/2023]
Abstract
RATIONALE Since substance use disorders have few or no effective pharmacotherapies, researchers have developed vaccines as immune-therapies against nicotine, cocaine, methamphetamine, and opioids including fentanyl. OBJECTIVES We focus on enhancing antibody (AB) production through stimulation of toll-like receptor-5 (TLR5) during active vaccination. The stimulating adjuvant is Entolimod, a novel protein derivative of flagellin. We review the molecular and cellular mechanisms underlying Entolimod's actions on TLR5. RESULTS Entolimod shows excellent efficacy for increasing AB levels to levels well beyond those produced by anti-addiction vaccines alone in animal models and humans. These ABs also significantly block the behavioral effects of the targeted drug of abuse. The TLR5 stimulation involves a wide range of immune cell types such as dendritic, antigen presenting, T and B cells. Entolimod binding to TLR5 initiates an intracellular signaling cascade that stimulates cytokine production of tumor necrosis factor and two interleukins (IL-6 and IL-12). While cytokine release can be catastrophic in cytokine storm, Entolimod produces a modulated release with few side effects even at doses 30 times greater than doses needed in these vaccine studies. Entolimod has markedly increased AB responses to all of our anti-addiction vaccines in rodent models, and in normal humans. CONCLUSIONS Entolimod and TLR5 stimulation has broad application to vaccines and potentially to other psychiatric disorders like depression, which has critical inflammatory contributions that Entolimod could reduce.
Collapse
|
7
|
Hwang CS, Bremer PT, Wenthur CJ, Ho SO, Chiang S, Ellis B, Zhou B, Fujii G, Janda KD. Enhancing Efficacy and Stability of an Antiheroin Vaccine: Examination of Antinociception, Opioid Binding Profile, and Lethality. Mol Pharm 2018; 15:1062-1072. [PMID: 29420901 DOI: 10.1021/acs.molpharmaceut.7b00933] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In recent years, drug conjugate vaccines have shown promise as therapeutics for substance use disorder. As a means to improve the efficacy of a heroin conjugate vaccine, we systematically explored 20 vaccine formulations with varying combinations of carrier proteins and adjuvants. In regard to adjuvants, we explored a Toll-like receptor 9 (TLR9) agonist and a TLR3 agonist in the presence of alum. The TLR9 agonist was cytosine-guanine oligodeoxynucleotide 1826 (CpG ODN 1826), while the TLR3 agonist was virus-derived genomic doubled-stranded RNA (dsRNA). The vaccine formulations containing TLR3 or TLR9 agonist alone elicited strong antiheroin antibody titers and blockade of heroin-induced antinociception when formulated with alum; however, a combination of TLR3 and TLR9 adjuvants did not result in improved efficacy. Investigation of month-long stability of the two lead formulations revealed that the TLR9 but not the TLR3 formulation was stable when stored as a lyophilized solid or as a liquid over 30 days. Furthermore, mice immunized with the TLR9 + alum heroin vaccine gained significant protection from lethal heroin doses, suggesting that this vaccine formulation is suitable for mitigating the harmful effects of heroin, even following month-long storage at room temperature.
Collapse
Affiliation(s)
- Candy S Hwang
- Departments of Chemistry, Immunology, and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Paul T Bremer
- Departments of Chemistry, Immunology, and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Cody J Wenthur
- Departments of Chemistry, Immunology, and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Sam On Ho
- Molecular Express, Inc., Rancho Dominguez , California 90220 , United States
| | - SuMing Chiang
- Molecular Express, Inc., Rancho Dominguez , California 90220 , United States
| | - Beverly Ellis
- Departments of Chemistry, Immunology, and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Bin Zhou
- Departments of Chemistry, Immunology, and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Gary Fujii
- Molecular Express, Inc., Rancho Dominguez , California 90220 , United States
| | - Kim D Janda
- Departments of Chemistry, Immunology, and Microbial Science, Skaggs Institute for Chemical Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| |
Collapse
|
8
|
Raleigh MD, Peterson SJ, Laudenbach M, Baruffaldi F, Carroll FI, Comer SD, Navarro HA, Langston TL, Runyon SP, Winston S, Pravetoni M, Pentel PR. Safety and efficacy of an oxycodone vaccine: Addressing some of the unique considerations posed by opioid abuse. PLoS One 2017; 12:e0184876. [PMID: 29194445 PMCID: PMC5711015 DOI: 10.1371/journal.pone.0184876] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/01/2017] [Indexed: 11/19/2022] Open
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.
Collapse
Affiliation(s)
- M. D. Raleigh
- Minneapolis Medical Research Foundation, Minneapolis, MN, United States of America
- * E-mail:
| | - S. J. Peterson
- Minneapolis Medical Research Foundation, Minneapolis, MN, United States of America
| | - M. Laudenbach
- Minneapolis Medical Research Foundation, Minneapolis, MN, United States of America
| | - F. Baruffaldi
- Minneapolis Medical Research Foundation, Minneapolis, MN, United States of America
| | - F. I. Carroll
- Research Triangle Institute, Research Triangle Park, NC, United States of America
| | - S. D. Comer
- New York State Psychiatric Institute and Department of Psychiatry, College of Physicians and Surgeons of Columbia University, New York, NY, United States of America
| | - H. A. Navarro
- Research Triangle Institute, Research Triangle Park, NC, United States of America
| | - T. L. Langston
- Research Triangle Institute, Research Triangle Park, NC, United States of America
| | - S. P. Runyon
- Research Triangle Institute, Research Triangle Park, NC, United States of America
| | - S. Winston
- Winston Biopharmaceutical Consulting, Boulder, CO, United States of America
| | - M. Pravetoni
- Minneapolis Medical Research Foundation, Minneapolis, MN, United States of America
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, United States of America
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - P. R. Pentel
- Minneapolis Medical Research Foundation, Minneapolis, MN, United States of America
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, United States of America
| |
Collapse
|
9
|
Abstract
INTRODUCTION Substance use disorders are a group of chronic relapsing disorders of the brain, which have massive public health and societal impact. In some disorders (e.g., heroin/prescription opioid addictions) approved medications have a major long-term benefit. For other substances (e.g., cocaine, amphetamines and cannabis) there are no approved medications, and for alcohol there are approved treatments, which are not in wide usage. Approved treatments for tobacco use disorders are available, and novel medications are also under study. Areas covered: Medication-based approaches which are in advanced preclinical stages, or which have reached proof-of concept clinical laboratory studies, as well as clinical trials. Expert opinion: Current challenges involve optimizing translation between preclinical and clinical development, and between clinical laboratory studies to therapeutic clinical trials. Comorbidities including depression or anxiety are challenges for study design and analysis. Improved pharmacogenomics, biomarker and phenotyping approaches are areas of interest. Pharmacological mechanisms currently under investigation include modulation of glutamatergic, GABA, vasopressin and κ-receptor function, as well as inhibition of monoamine re-uptake. Other factors that affect potential market size for emerging medications include stigma, availability of treatment settings, adoption by clinicians, and the prevalence of persons with SUD who are not actively treatment-seeking.
Collapse
Affiliation(s)
- Eduardo R Butelman
- a Laboratory in the Biology of Addictive Diseases , The Rockefeller University , New York , NY , USA
| | - Mary Jeanne Kreek
- a Laboratory in the Biology of Addictive Diseases , The Rockefeller University , New York , NY , USA
| |
Collapse
|
10
|
Brashier DBS, Sharma AK, Akhoon N. Are therapeutic vaccines an answer to the global problem of drug and alcohol abuse? Indian J Pharmacol 2016; 48:487-489. [PMID: 27721531 PMCID: PMC5051239 DOI: 10.4103/0253-7613.190717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Drug Abuse has become a major challenging problem for the society. It effects people of all countries economical strata's and all ages. According. Monetary loss all over the world regarding drug abuse is in million dollars, it not only has an impact on human productivity and healthcare cost but also on cost of crimes conducted by these drugs and alcohol abuse. Therapeutic vaccine has come as new approach to deal with this problem, after failures in search for a pharmaceutical agent to deal with drug of abuse and alcohol. Research in field of nicotine abuse has gone a way ahead with number of vaccines being tried clinically followed by cocaine, opioids, methamphetamine, phencyclidine and alcohol. All of them have a common mechanism of action by antibody production whereas alcohol acts by genetic intervention. None have being approved yet due to poor results in phase II trials, possibly due to not able to trigger an adequate immunological response. But still quest is on for cracking the ice by developing first successful vaccine against drug of abuse, that would follow for other drugs too. It would be great step in field of therapeutic vaccines for drug abuse after similar successful vaccines being approved for other diseases like cancer.
Collapse
Affiliation(s)
- Dick B S Brashier
- Department of Pharmacology, Armed Forces Medical College, Pune, Maharashtra, India
| | - Ashok Kumar Sharma
- Department of Pharmacology, Armed Forces Medical College, Pune, Maharashtra, India
| | - Neha Akhoon
- Department of Pharmacology, Armed Forces Medical College, Pune, Maharashtra, India
| |
Collapse
|
11
|
Pravetoni M. Biologics to treat substance use disorders: Current status and new directions. Hum Vaccin Immunother 2016; 12:3005-3019. [PMID: 27441896 DOI: 10.1080/21645515.2016.1212785] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Biologics (vaccines, monoclonal antibodies (mAb), and genetically modified enzymes) offer a promising class of therapeutics to treat substance use disorders (SUD) involving abuse of opioids and stimulants such as nicotine, cocaine, and methamphetamine. In contrast to small molecule medications targeting brain receptors, biologics for SUD are larger molecules that do not cross the blood-brain barrier (BBB), but target the drug itself, preventing its distribution to the brain and blunting its effects on the central nervous system (CNS). Active and passive immunization approaches rely on antibodies (Ab) that bind drugs of abuse in serum and block their distribution to the brain, preventing the rewarding effects of drugs and addiction-related behaviors. Alternatives to vaccines and anti-drug mAb are genetically engineered human or bacterial enzymes that metabolize drugs of abuse, lowering the concentration of free active drug. Pre-clinical and clinical data support development of effective biologics for SUD.
Collapse
Affiliation(s)
- Marco Pravetoni
- a Minneapolis Medical Research Foundation, and University of Minnesota Medical School, Departments of Medicine and Pharmacology , Center for Immunology , Minneapolis , MN , USA
| |
Collapse
|
12
|
Rudra JS, Ding Y, Neelakantan H, Ding C, Appavu R, Stutz S, Snook JD, Chen H, Cunningham KA, Zhou J. Suppression of Cocaine-Evoked Hyperactivity by Self-Adjuvanting and Multivalent Peptide Nanofiber Vaccines. ACS Chem Neurosci 2016; 7:546-52. [PMID: 26926328 DOI: 10.1021/acschemneuro.5b00345] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The development of anti-cocaine vaccines that counteract the rewarding effects of the drug are currently being investigated as adjunct therapies for prevention of relapse in abstinent users. However, cocaine is weakly immunogenic and requires conjugation to carrier proteins and coadministration with strong adjuvants, which carry the risk of local reactogenicity and systemic toxicity. Here we report synthetic and multivalent self-assembling peptide nanofibers as adjuvant-free carriers for cocaine vaccines. A novel cocaine hapten modified at the P3 site was conjugated to the N-terminus of an amphipathic self-assembling domain KFE8. In aqueous buffers the cocaine-KFE8 conjugate assembled into β-sheet rich nanofibers, which raised anti-cocaine antibodies without the need for added adjuvants in mice. Vaccinated mice were treated with cocaine and a significant negative correlation was observed between antibody levels and cocaine-evoked hyperactivity. These totally synthetic and multivalent nanofibers with well-defined chemical composition represent the first generation of adjuvant-free cocaine vaccines.
Collapse
Affiliation(s)
- Jai S. Rudra
- Department of Pharmacology & Toxicology and ⊥Center for Addiction Research, and ‡Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555, United States
| | - Ye Ding
- Department of Pharmacology & Toxicology and ⊥Center for Addiction Research, and ‡Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555, United States
| | - Harshini Neelakantan
- Department of Pharmacology & Toxicology and ⊥Center for Addiction Research, and ‡Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555, United States
| | - Chunyong Ding
- Department of Pharmacology & Toxicology and ⊥Center for Addiction Research, and ‡Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555, United States
| | - Rajagopal Appavu
- Department of Pharmacology & Toxicology and ⊥Center for Addiction Research, and ‡Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555, United States
| | - Sonja Stutz
- Department of Pharmacology & Toxicology and ⊥Center for Addiction Research, and ‡Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555, United States
| | - Joshua D. Snook
- Department of Pharmacology & Toxicology and ⊥Center for Addiction Research, and ‡Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555, United States
| | - Haiying Chen
- Department of Pharmacology & Toxicology and ⊥Center for Addiction Research, and ‡Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555, United States
| | - Kathryn A. Cunningham
- Department of Pharmacology & Toxicology and ⊥Center for Addiction Research, and ‡Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555, United States
| | - Jia Zhou
- Department of Pharmacology & Toxicology and ⊥Center for Addiction Research, and ‡Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas 77555, United States
| |
Collapse
|
13
|
Blum K, Thanos PK, Badgaiyan RD, Febo M, Oscar-Berman M, Fratantonio J, Demotrovics Z, Gold MS. Neurogenetics and gene therapy for reward deficiency syndrome: are we going to the Promised Land? Expert Opin Biol Ther 2015; 15:973-85. [PMID: 25974314 DOI: 10.1517/14712598.2015.1045871] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Addiction is a substantial health issue with limited treatment options approved by the FDA and as such currently available. The advent of neuroimaging techniques that link neurochemical and neurogenetic mechanisms to the reward circuitry brain function provides a framework for potential genomic-based therapies. AREAS COVERED Through candidate and genome-wide association studies approaches, many gene polymorphisms and clusters have been implicated in drug, food and behavioral dependence linked by the common rubric reward deficiency syndrome (RDS). The results of selective studies that include the role of epigenetics, noncoding micro RNAs in RDS behaviors especially drug abuse involving alcohol, opioids, cocaine, nicotine, pain and feeding are reviewed in this article. New targets for addiction treatment and relapse prevention, treatment alternatives such as gene therapy in animal models, and pharmacogenomics and nutrigenomics methods to manipulate transcription and gene expression are explored. EXPERT OPINION The recognition of the clinical benefit of early genetic testing to determine addiction risk stratification and dopaminergic agonistic, rather than antagonistic therapies are potentially the genomic-based wave of the future. In addition, further development, especially in gene transfer work and viral vector identification, could make gene therapy for RDS a possibility in the future.
Collapse
Affiliation(s)
- Kenneth Blum
- Department of Psychiatry & McKnight Brain Institute, University of Florida College of Medicine , Gainesville, FL , USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
McCreary AC, Müller CP, Filip M. Psychostimulants: Basic and Clinical Pharmacology. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 120:41-83. [PMID: 26070753 DOI: 10.1016/bs.irn.2015.02.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Substance use disorder, and particularly psychostimulant use disorder, has considerable socioeconomic burden globally. The psychostimulants include several chemical classes, being derivatives of benzoylecgonine, phenethylamine, phenylpropanolamine, or aminoaryloxazoline. Psychostimulant drugs activate the brain reward pathways of the mesoaccumbal system, and continued use leads to persistent neuroplastic and dysfunctional changes of a variety of structures involved in learning and memory, habit-forming learning, salience attribution, and inhibitory control. There are a variety of neurochemical and neurobehavioral changes in psychostimulant addiction, for example, dopaminergic, glutamatergic, serotonergic (5-HT-ergic), and γ-amino butyric acid (GABA) changes have all noted. In this chapter, we will review pharmacological changes associated with psychostimulant use and abuse in humans and animals, and on the basis of the best characterized and most widely abused psychostimulants (amphetamines, cocaine) discuss why use transitions into abuse and review basic science and clinical strategies that might assist in treating psychostimulant abuse.
Collapse
Affiliation(s)
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Małgorzata Filip
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland; Department of Toxicology, Faculty of Pharmacy, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
15
|
Lockner JW, Eubanks LM, Choi JL, Lively JM, Schlosburg JE, Collins KC, Globisch D, Rosenfeld-Gunn RJ, Wilson IA, Janda KD. Flagellin as carrier and adjuvant in cocaine vaccine development. Mol Pharm 2015; 12:653-62. [PMID: 25531528 PMCID: PMC4319694 DOI: 10.1021/mp500520r] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cocaine abuse is problematic, directly and indirectly impacting the lives of millions, and yet existing therapies are inadequate and usually ineffective. A cocaine vaccine would be a promising alternative therapeutic option, but efficacy is hampered by variable production of anticocaine antibodies. Thus, new tactics and strategies for boosting cocaine vaccine immunogenicity must be explored. Flagellin is a bacterial protein that stimulates the innate immune response via binding to extracellular Toll-like receptor 5 (TLR5) and also via interaction with intracellular NOD-like receptor C4 (NLRC4), leading to production of pro-inflammatory cytokines. Reasoning that flagellin could serve as both carrier and adjuvant, we modified recombinant flagellin protein to display a cocaine hapten termed GNE. The resulting conjugates exhibited dose-dependent stimulation of anti-GNE antibody production. Moreover, when adjuvanted with alum, but not with liposomal MPLA, GNE-FliC was found to be better than our benchmark GNE-KLH. This work represents a new avenue for exploration in the use of hapten-flagellin conjugates to elicit antihapten immune responses.
Collapse
Affiliation(s)
- Jonathan W Lockner
- Departments of Chemistry, Integrative Structural and Computational Biology, and Immunology and Microbial Science, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|