1
|
Hosztafi S, Galambos AR, Köteles I, Karádi DÁ, Fürst S, Al-Khrasani M. Opioid-Based Haptens: Development of Immunotherapy. Int J Mol Sci 2024; 25:7781. [PMID: 39063024 PMCID: PMC11277321 DOI: 10.3390/ijms25147781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Over the past decades, extensive preclinical research has been conducted to develop vaccinations to protect against substance use disorder caused by opioids, nicotine, cocaine, and designer drugs. Morphine or fentanyl derivatives are small molecules, and these compounds are not immunogenic, but when conjugated as haptens to a carrier protein will elicit the production of antibodies capable of reacting specifically with the unconjugated hapten or its parent compound. The position of the attachment in opioid haptens to the carrier protein will influence the specificity of the antiserum produced in immunized animals with the hapten-carrier conjugate. Immunoassays for the determination of opioid drugs are based on the ability of drugs to inhibit the reaction between drug-specific antibodies and the corresponding drug-carrier conjugate or the corresponding labelled hapten. Pharmacological studies of the hapten-carrier conjugates resulted in the development of vaccines for treating opioid use disorders (OUDs). Immunotherapy for opioid addiction includes the induction of anti-drug vaccines which are composed of a hapten, a carrier protein, and adjuvants. In this review we survey the design of opioid haptens, the development of the opioid radioimmunoassay, and the results of immunotherapy for OUDs.
Collapse
Affiliation(s)
- Sándor Hosztafi
- Department of Pharmaceutical Chemistry, Semmelweis University, Hogyes Endre u. 9., H-1092 Budapest, Hungary;
| | - Anna Rita Galambos
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvá-rad tér 4., H-1445 Budapest, Hungary; (A.R.G.); (D.Á.K.); (S.F.)
| | - István Köteles
- Department of Pharmaceutical Chemistry, Semmelweis University, Hogyes Endre u. 9., H-1092 Budapest, Hungary;
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-412 96 Gothenburg, Sweden
| | - Dávid Á Karádi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvá-rad tér 4., H-1445 Budapest, Hungary; (A.R.G.); (D.Á.K.); (S.F.)
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Üllői út 78., H-1082 Budapest, Hungary
| | - Susanna Fürst
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvá-rad tér 4., H-1445 Budapest, Hungary; (A.R.G.); (D.Á.K.); (S.F.)
| | - Mahmoud Al-Khrasani
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvá-rad tér 4., H-1445 Budapest, Hungary; (A.R.G.); (D.Á.K.); (S.F.)
| |
Collapse
|
2
|
Celik M, Fuehrlein B. A Review of Immunotherapeutic Approaches for Substance Use Disorders: Current Status and Future Prospects. Immunotargets Ther 2022; 11:55-66. [PMID: 36199734 PMCID: PMC9528911 DOI: 10.2147/itt.s370435] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/23/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Muhammet Celik
- Research Division, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Brian Fuehrlein
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Mental Health Service Line, VA Connecticut Healthcare System, West Haven, CT, USA
- Correspondence: Brian Fuehrlein, Mental Health Service Line, VA Connecticut Healthcare System, 950 Campbell Ave, West Haven, CT, 06516, Tel +1-203-932-5711 x4471, Fax +1-203-937-4904, Email
| |
Collapse
|
3
|
Lin M, Marin A, Ellis B, Eubanks LM, Andrianov AK, Janda KD. Polyphosphazene: A New Adjuvant Platform for Cocaine Vaccine Development. Mol Pharm 2022; 19:3358-3366. [PMID: 35984034 DOI: 10.1021/acs.molpharmaceut.2c00489] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cocaine is a highly addictive drug that has seen a steady uptrend causing severe health problems worldwide. Currently, there are no approved therapeutics for treating cocaine use disorder; hence, there is an urgent need to identify new medications. Immunopharmacotherapeutics is a promising approach utilizing endogenous antibodies generated through active vaccination, and if properly programmed, can blunt a drug's psychoactive and addictive effects. However, drug vaccine efficacy has largely been limited by the modest levels of antibodies induced. Herein, we explored an adjuvant system consisting of a polyphosphazene macromolecule, specifically poly[di(carboxylatoethylphenoxy)-phosphazene] (PCEP), a biocompatible synthetic polymer that was solicited for improved cocaine conjugate vaccine delivery performance. Our results demonstrated PCEP's superior assembling efficiency with a cocaine hapten as well as with the combined adjuvant CpG oligodeoxynucleotide (ODN). Importantly, this combination led to a higher titer response, balanced immunity, successful sequestering of cocaine in the blood, and a reduction in the drug in the brain. Moreover, a PCEP-cocaine conjugate vaccine was also found to function well via intranasal administration, where its efficacy was demonstrated through the antibody titer, affinity, mucosal IgA production, and a reduction in cocaine's locomotor activity. Overall, a comprehensive evaluation of PCEP integrated within a cocaine vaccine established an advance in the use of synthetic adjuvants in the drugs of abuse vaccine field.
Collapse
Affiliation(s)
- Mingliang Lin
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, California 92037, United States
| | - Alexander Marin
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland 20850, United States
| | - Beverly Ellis
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, California 92037, United States
| | - Lisa M Eubanks
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, California 92037, United States
| | - Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland 20850, United States
| | - Kim D Janda
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, La Jolla, California 92037, United States
| |
Collapse
|
4
|
Abucayon E, Whalen C, Torres OB, Duval AJ, Sulima A, Antoline JFG, Oertel T, Barrientos RC, Jacobson AE, Rice KC, Matyas GR. A Rapid Method for Direct Quantification of Antibody Binding-Site Concentration in Serum. ACS OMEGA 2022; 7:26812-26823. [PMID: 35936462 PMCID: PMC9352236 DOI: 10.1021/acsomega.2c03237] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/04/2022] [Indexed: 06/15/2023]
Abstract
The quantitation of the available antibody binding-site concentration of polyclonal antibodies in serum is critical in defining the efficacy of vaccines against substances of abuse. We have conceptualized an equilibrium dialysis (ED)-based approach coupled with fluorimetry (ED-fluorimetry) to measure the antibody binding-site concentration to the ligand in an aqueous environment. The measured binding-site concentrations in monoclonal antibody (mAb) and sera samples from TT-6-AmHap-immunized rats by ED-fluorimetry are in agreement with those determined by a more established equilibrium dialysis coupled with ultraperformance liquid chromatography tandem mass spectrometry (ED-UPLC-MS/MS). Importantly, we have shown that the measured antibody binding-site concentrations to the ligand by ED-fluorimetry were not influenced by the sample serum matrix; thus, this method is valid for determining the binding-site concentration of polyclonal antibodies in sera samples. Further, we have demonstrated that under appropriate analytical conditions, this method resolved the total binding-site concentrations on a nanomolar scale with good accuracy and repeatability within the microliter sample volumes. This simple, rapid, and sample preparation-free approach has the potential to reliably perform quantitative antibody binding-site screening in serum and other more complex biological fluids.
Collapse
Affiliation(s)
- Erwin
G. Abucayon
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program,
Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Henry
M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Connor Whalen
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program,
Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Oak
Ridge Institute for Science and Education, Oak Ridge, Tennessee 37831, United States
| | - Oscar B. Torres
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program,
Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Henry
M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Alexander J. Duval
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program,
Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Henry
M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Agnieszka Sulima
- Department
of Health and Human Services, Drug Design
and Synthesis Section, Molecular Targets and Medications Discovery
Branch, Intramural Research Program, National Institute on Drug Abuse
and the National Institute on Alcohol Abuse and Alcoholism, National
Institutes of Health, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Joshua F. G. Antoline
- Department
of Health and Human Services, Drug Design
and Synthesis Section, Molecular Targets and Medications Discovery
Branch, Intramural Research Program, National Institute on Drug Abuse
and the National Institute on Alcohol Abuse and Alcoholism, National
Institutes of Health, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Therese Oertel
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program,
Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Oak
Ridge Institute for Science and Education, Oak Ridge, Tennessee 37831, United States
| | - Rodell C. Barrientos
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program,
Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Henry
M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Arthur E. Jacobson
- Department
of Health and Human Services, Drug Design
and Synthesis Section, Molecular Targets and Medications Discovery
Branch, Intramural Research Program, National Institute on Drug Abuse
and the National Institute on Alcohol Abuse and Alcoholism, National
Institutes of Health, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Kenner C. Rice
- Department
of Health and Human Services, Drug Design
and Synthesis Section, Molecular Targets and Medications Discovery
Branch, Intramural Research Program, National Institute on Drug Abuse
and the National Institute on Alcohol Abuse and Alcoholism, National
Institutes of Health, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Gary R. Matyas
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program,
Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| |
Collapse
|
5
|
Sulima A, Li F, Morgan JB, Truong P, Antoline JFG, Oertel T, Barrientos RC, Torres OB, Beck Z, Imler GH, Deschamps JR, Matyas GR, Jacobson AE, Rice KC. Design, Synthesis, and In Vivo Evaluation of C1-Linked 4,5-Epoxymorphinan Haptens for Heroin Vaccines. Molecules 2022; 27:1553. [PMID: 35268659 PMCID: PMC8911913 DOI: 10.3390/molecules27051553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/16/2022] [Accepted: 02/21/2022] [Indexed: 11/16/2022] Open
Abstract
In our continuing effort to develop effective anti-heroin vaccines as potential medications for the treatment of opioid use disorder, herein we present the design and synthesis of the haptens: 1-AmidoMorHap (1), 1-AmidoMorHap epimer (2), 1 Amido-DihydroMorHap (3), and 1 Amido-DihydroMorHap epimer (4). This is the first report of hydrolytically stable haptenic surrogates of heroin with the attachment site at the C1 position in the 4,5-epoxymorophinan nucleus. We prepared respective tetanus toxoid (TT)-hapten conjugates as heroin vaccine immunogens and evaluated their efficacy in vivo. We showed that all TT-hapten conjugates induced high antibody endpoint titers against the targets but only haptens 2 and 3 can induce protective effects against heroin in vivo. The epimeric analogues of these haptens, 1 and 4, failed to protect mice from the effects of heroin. We also showed that the in vivo efficacy is consistent with the results of the in vitro drug sequestration assay. Attachment of the linker at the C1 position induced antibodies with weak binding to the target drugs. Only TT-2 and TT-3 yielded antibodies that bound heroin and 6-acetyl morphine. None of the TT-hapten conjugates induced antibodies that cross-reacted with morphine, methadone, naloxone, or naltrexone, and only TT-3 interacted weakly with buprenorphine, and that subtle structural difference, especially at the C6 position, can vastly alter the specificity of the induced antibodies. This study is an important contribution in the field of vaccine development against small-molecule targets, providing proof that the chirality at C6 in these epoxymorphinans is a vital key to their effectiveness.
Collapse
Affiliation(s)
- Agnieszka Sulima
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| | - Fuying Li
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| | - Jeffrey Brian Morgan
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| | - Phong Truong
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| | - Joshua F. G. Antoline
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| | - Therese Oertel
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (T.O.); (R.C.B.); (O.B.T.); (Z.B.); (G.R.M.)
| | - Rodell C. Barrientos
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (T.O.); (R.C.B.); (O.B.T.); (Z.B.); (G.R.M.)
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| | - Oscar B. Torres
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (T.O.); (R.C.B.); (O.B.T.); (Z.B.); (G.R.M.)
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| | - Zoltan Beck
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (T.O.); (R.C.B.); (O.B.T.); (Z.B.); (G.R.M.)
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| | - Gregory H. Imler
- Center for Biomolecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA; (G.H.I.); (J.R.D.)
| | - Jeffrey R. Deschamps
- Center for Biomolecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA; (G.H.I.); (J.R.D.)
| | - Gary R. Matyas
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (T.O.); (R.C.B.); (O.B.T.); (Z.B.); (G.R.M.)
| | - Arthur E. Jacobson
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| | - Kenner C. Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA; (A.S.); (F.L.); (J.B.M.); (P.T.); (J.F.G.A.)
| |
Collapse
|
6
|
Barrientos R, Whalen C, Torres OB, Sulima A, Bow EW, Komla E, Beck Z, Jacobson AE, Rice KC, Matyas GR. Bivalent Conjugate Vaccine Induces Dual Immunogenic Response That Attenuates Heroin and Fentanyl Effects in Mice. Bioconjug Chem 2021; 32:2295-2306. [PMID: 34076427 PMCID: PMC8603354 DOI: 10.1021/acs.bioconjchem.1c00179] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/17/2021] [Indexed: 11/29/2022]
Abstract
Opioid use disorders and fatal overdose due to consumption of fentanyl-laced heroin remain a major public health menace in the United States. Vaccination may serve as a promising potential remedy to combat accidental overdose and to mitigate the abuse potential of opioids. We previously reported the heroin and fentanyl monovalent vaccines carrying, respectively, a heroin hapten, 6-AmHap, and a fentanyl hapten, para-AmFenHap, conjugated to tetanus toxoid (TT). Herein, we describe the mixing of these antigens to formulate a bivalent vaccine adjuvanted with liposomes containing monophosphoryl lipid A (MPLA) adsorbed on aluminum hydroxide. Immunization of mice with the bivalent vaccine resulted in IgG titers of >105 against both haptens. The polyclonal sera bound heroin, 6-acetylmorphine, morphine, and fentanyl with dissociation constants (Kd) of 0.25 to 0.50 nM. Mice were protected from the anti-nociceptive effects of heroin, fentanyl, and heroin +9% (w/w) fentanyl. No cross-reactivity to methadone and buprenorphine was observed in vivo. Naloxone remained efficacious in immunized mice. These results highlighted the potential of combining TT-6-AmHap and TT-para-AmFenHap to yield an efficacious bivalent vaccine that could ablate heroin and fentanyl effects. This vaccine warrants further testing to establish its potential translatability to humans.
Collapse
Affiliation(s)
- Rodell
C. Barrientos
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Henry
M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Connor Whalen
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Oscar B. Torres
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Henry
M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Agnieszka Sulima
- Drug
Design and Synthesis Section, Molecular Targets and Medications Discovery
Branch, Intramural Research Program, National
Institute on Drug Abuse and the National Institute on Alcohol Abuse
and Alcoholism, National Institutes of Health, Department of Health
and Human Services, 9800 Medical Center Drive, Bethesda, Maryland 20892, United States
| | - Eric W. Bow
- Drug
Design and Synthesis Section, Molecular Targets and Medications Discovery
Branch, Intramural Research Program, National
Institute on Drug Abuse and the National Institute on Alcohol Abuse
and Alcoholism, National Institutes of Health, Department of Health
and Human Services, 9800 Medical Center Drive, Bethesda, Maryland 20892, United States
| | - Essie Komla
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Henry
M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Zoltan Beck
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
- Henry
M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Arthur E. Jacobson
- Drug
Design and Synthesis Section, Molecular Targets and Medications Discovery
Branch, Intramural Research Program, National
Institute on Drug Abuse and the National Institute on Alcohol Abuse
and Alcoholism, National Institutes of Health, Department of Health
and Human Services, 9800 Medical Center Drive, Bethesda, Maryland 20892, United States
| | - Kenner C. Rice
- Drug
Design and Synthesis Section, Molecular Targets and Medications Discovery
Branch, Intramural Research Program, National
Institute on Drug Abuse and the National Institute on Alcohol Abuse
and Alcoholism, National Institutes of Health, Department of Health
and Human Services, 9800 Medical Center Drive, Bethesda, Maryland 20892, United States
| | - Gary R. Matyas
- Laboratory
of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| |
Collapse
|
7
|
Ban B, Barrientos RC, Oertel T, Komla E, Whalen C, Sopko M, You Y, Banerjee P, Sulima A, Jacobson AE, Rice KC, Matyas GR, Yusibov V. Novel chimeric monoclonal antibodies that block fentanyl effects and alter fentanyl biodistribution in mice. MAbs 2021; 13:1991552. [PMID: 34693882 PMCID: PMC8547829 DOI: 10.1080/19420862.2021.1991552] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The prevalence and societal impact of opioid use disorder (OUD) is an acknowledged public health crisis that is further aggravated by the current pandemic. One of the devastating consequences of OUD is opioid overdose deaths. While multiple medications are now available to treat OUD, given the prevalence and societal burden, additional well-tolerated and effective therapies are still needed. To this point, we have developed chimeric monoclonal antibodies (mAb) that will specifically complex with fentanyl and its analogs in the periphery, thereby preventing them from reaching the central nervous system. Additionally, mAb-based passive immunotherapy offers a high degree of specificity to drugs of abuse and does not interfere with an individual’s ability to use any of the medications used to treat OUD. We hypothesized that sequestering fentanyl and its analogs in the periphery will mitigate their negative effects on the brain and peripheral organs. This study is the first report of chimeric mAb against fentanyl and its analogs. We have discovered, engineered the chimeric versions, and identified the selectivity of these antibodies, through in vitro characterization and in vivo animal challenge studies. Two mAb candidates with very high (0.1–1.3 nM) binding affinities to fentanyl and its analogs were found to be effective in engaging fentanyl in the periphery and blocking its effects in challenged animals. Results presented in this work constitute a major contribution in the field of novel therapeutics targeting OUD.
Collapse
Affiliation(s)
- Bhupal Ban
- Pharmaceutical Center, Indiana Biosciences Research Institute, Indianapolis, Indiana, United States
| | - Rodell C Barrientos
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States.,U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States
| | - Therese Oertel
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States
| | - Essie Komla
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States.,U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States
| | - Connor Whalen
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States
| | - Megan Sopko
- Pharmaceutical Center, Indiana Biosciences Research Institute, Indianapolis, Indiana, United States
| | - Yingjian You
- Pharmaceutical Center, Indiana Biosciences Research Institute, Indianapolis, Indiana, United States
| | - Partha Banerjee
- Pharmaceutical Center, Indiana Biosciences Research Institute, Indianapolis, Indiana, United States
| | - Agnieszka Sulima
- Department of Health and Human Services, Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Arthur E Jacobson
- Department of Health and Human Services, Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Kenner C Rice
- Department of Health and Human Services, Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Gary R Matyas
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States
| | - Vidadi Yusibov
- Pharmaceutical Center, Indiana Biosciences Research Institute, Indianapolis, Indiana, United States
| |
Collapse
|
8
|
Immunochemical method for penthiopyrad detection through thermodynamic and kinetic characterization of monoclonal antibodies. Talanta 2021; 226:122123. [PMID: 33676678 DOI: 10.1016/j.talanta.2021.122123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 10/22/2022]
Abstract
Immunoassays are nowadays being employed for rapid contaminant analysis in clinical, environmental, and agrochemical samples. A thorough characterization of the antibody‒antigen interaction can bring light to the immunoreagent selection process in order to develop sensitive and robust tests. Thus, determination of equilibrium and reaction rate constants is usually recommendable. However, this can be quite tricky for low molecular weight compounds, and competitive strategies are commonly followed to estimate apparent affinity values. In the present study, a collection of monoclonal antibodies to penthiopyrad was raised for the first time, and apparent equilibrium constants were assessed by the Langmuir model using three different competitive enzyme-linked immunosorbent assay formats. The obtained KD values from antibody-coated assays were quite close to the corresponding KD values calculated from surface plasmon resonance (SPR) evaluation. These studies were employed to select a pair of immunoreagents for immunoassay development. The KD value for penthiopyrad of the selected antibody obtained by SPR was 0.28 nM. The optimized direct assay showed an IC50 value for penthiopyrad of 0.42 nM (0.15 ng mL-1) in buffer. The limit of quantification for grape, must, and wine samples was 10 ng mL-1. An excellent correlation was found when immunochemical results were compared with those from LC-MS/MS. As an application case, it was determined that 58% of penthiopyrad was still found in wine after fermentation.
Collapse
|
9
|
Gutman ES, Irvin TC, Morgan JB, Barrientos RC, Torres OB, Beck Z, Matyas GR, Jacobson AE, Rice KC. Synthesis and immunological effects of C14-linked 4,5-epoxymorphinan analogues as novel heroin vaccine haptens. RSC Chem Biol 2021; 2:835-842. [PMID: 34179783 PMCID: PMC8190897 DOI: 10.1039/d1cb00029b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Active immunization is being explored as a potential therapeutic to combat accidental overdose and to mitigate the abuse potential of opioids. Hapten design is one of the crucial factors that determines the efficacy of a candidate vaccine to substance abuse and remains one of the most active areas of research in vaccine development. Herein we report for the first time the synthesis of three novel opiate surrogates with the linker attachment site at C14, 1 (6,14-AmidoHap), 2 (14-AmidoMorHap), and 3 (14-AmidoHerHap) as novel heroin haptens. The compounds 1, 2, and 3 are analogues with different substituents at C6: an acetamide, a hydroxyl moiety, and an acetate, respectively. All three haptens had a phenolic hydroxyl group at C3. The haptens were conjugated to the tetanus toxoid carrier protein, adjuvanted with liposomal monophosphoryl lipid A/aluminum hydroxide and were tested in mice in terms of immunogenicity and efficacy. Immunization of mice resulted in antibody endpoint titers of >105 against all the haptens. Neither of the conjugates of 1, 2, and 3 had induced antibodies with selectivity broad enough to recognize and bind heroin, 6-AM, and morphine resulting in little to no protection against the antinociceptive effects of heroin in vivo. Only the mice immunized with conjugate 3 were partially protected against heroin-induced antinociception. These results contribute to the growing body of knowledge that the linker position and the subtle structural differences in the hapten scaffold impact the selectivity of the induced antibodies. Together, these highlight the importance of rational hapten design for heroin vaccine development. Three novel opiate surrogates with the linker at C14, 1 (6,14-AmidoHap), 2 (14-AmidoMorHap), and 3 (14-AmidoHerHap) were conjugated to tetanus toxoid (TT) and tested as heroin vaccines. The C3 and C6 moieties are crucial in antibody selectivity.![]()
Collapse
Affiliation(s)
- Eugene S Gutman
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and The National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services 9800 Medical Center Drive Bethesda MD 20892-3373 USA +1 301-451-4799 +1 301-451-5028
| | - Thomas C Irvin
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and The National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services 9800 Medical Center Drive Bethesda MD 20892-3373 USA +1 301-451-4799 +1 301-451-5028
| | - J Brian Morgan
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and The National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services 9800 Medical Center Drive Bethesda MD 20892-3373 USA +1 301-451-4799 +1 301-451-5028
| | - Rodell C Barrientos
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research 503 Robert Grant Avenue Silver Spring MD 20910 USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine 6720A Rockledge Drive Bethesda MD 20817 USA
| | - Oscar B Torres
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research 503 Robert Grant Avenue Silver Spring MD 20910 USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine 6720A Rockledge Drive Bethesda MD 20817 USA
| | - Zoltan Beck
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research 503 Robert Grant Avenue Silver Spring MD 20910 USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine 6720A Rockledge Drive Bethesda MD 20817 USA
| | - Gary R Matyas
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research 503 Robert Grant Avenue Silver Spring MD 20910 USA
| | - Arthur E Jacobson
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and The National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services 9800 Medical Center Drive Bethesda MD 20892-3373 USA +1 301-451-4799 +1 301-451-5028
| | - Kenner C Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and The National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services 9800 Medical Center Drive Bethesda MD 20892-3373 USA +1 301-451-4799 +1 301-451-5028
| |
Collapse
|
10
|
Lin M, Lee JC, Blake S, Ellis B, Eubanks LM, Janda KD. Broadly Neutralizing Synthetic Cannabinoid Vaccines. JACS AU 2021; 1:31-40. [PMID: 34467269 PMCID: PMC8395583 DOI: 10.1021/jacsau.0c00057] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Indexed: 05/11/2023]
Abstract
Synthetic cannabinoids (SCs) constitute a significant portion of psychoactive substances forming a major public health risk. Due to the wide variety of SCs, broadly neutralizing antibodies generated by active immunization present an intriguing pathway to combat cannabinoid use disorder. Here, we probed hapten design for antibody affinity and cross reactivity against two classes of SCs. Of the 10 haptens screened, 3 vaccine groups revealed submicromolar IC50, each targeting 5-6 compounds in our panel of 22 drugs. Moreover, SCs were successfully sequestered when administered by vaping or intraperitoneal injection, which was confirmed within animal models by observing locomotion, body temperature, and pharmacokinetics. We also discovered synergistic effects to simultaneously blunt two drug classes through an admixture vaccine approach. Collectively, our study provides a comprehensive foundation for the development of vaccines against SCs.
Collapse
|
11
|
Barrientos RC, Bow EW, Whalen C, Torres OB, Sulima A, Beck Z, Jacobson AE, Rice KC, Matyas GR. Novel Vaccine That Blunts Fentanyl Effects and Sequesters Ultrapotent Fentanyl Analogues. Mol Pharm 2020; 17:3447-3460. [PMID: 32787282 PMCID: PMC7482402 DOI: 10.1021/acs.molpharmaceut.0c00497] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
![]()
Active
immunization is an emerging potential modality to combat
fatal overdose amid the opioid epidemic. In this study, we described
the design, synthesis, formulation, and animal testing of an efficacious
vaccine against fentanyl. The vaccine formulation is composed of a
novel fentanyl hapten conjugated to tetanus toxoid (TT) and adjuvanted
with liposomes containing monophosphoryl lipid A adsorbed on aluminum
hydroxide. The linker and hapten N-phenyl-N-(1-(4-(3-(tritylthio)propanamido)phenethyl)piperidin-4-yl)propionamide
were conjugated sequentially to TT using amine-N-hydroxysuccinimide-ester
and thiol–maleimide reaction chemistries, respectively. Conjugation
was facile, efficient, and reproducible with a protein recovery of
>98% and a hapten density of 30–35 per carrier protein molecule.
In mice, immunization induced high and robust antibody endpoint titers
in the order of >106 against the hapten. The antisera
bound
fentanyl, carfentanil, cyclopropyl fentanyl, para-fluorofentanyl, and furanyl fentanyl in vitro with
antibody-drug dissociation constants in the range of 0.36–4.66
nM. No cross-reactivity to naloxone, naltrexone, methadone, or buprenorphine
was observed. In vivo, immunization shifted the antinociceptive
dose–response curve of fentanyl to higher doses. Collectively,
these preclinical results showcased the desired traits of a potential
vaccine against fentanyl and demonstrated the feasibility of immunization
to combat fentanyl-induced effects.
Collapse
Affiliation(s)
- Rodell C Barrientos
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States.,U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Eric W Bow
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, Department of Health and Human Services, National Institutes of Health, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Connor Whalen
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Oscar B Torres
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States.,U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Agnieszka Sulima
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, Department of Health and Human Services, National Institutes of Health, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Zoltan Beck
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States.,U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, Maryland 20817, United States
| | - Arthur E Jacobson
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, Department of Health and Human Services, National Institutes of Health, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Kenner C Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, Department of Health and Human Services, National Institutes of Health, 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Gary R Matyas
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| |
Collapse
|
12
|
A rapid solution-based method for determining the affinity of heroin hapten-induced antibodies to heroin, its metabolites, and other opioids. Anal Bioanal Chem 2018; 410:3885-3903. [PMID: 29675707 PMCID: PMC5956019 DOI: 10.1007/s00216-018-1060-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/27/2018] [Accepted: 04/03/2018] [Indexed: 11/15/2022]
Abstract
We describe for the first time a method that utilizes microscale thermophoresis (MST) technology to determine polyclonal antibody affinities to small molecules. Using a novel type of heterologous MST, we have accurately measured a solution-based binding affinity of serum antibodies to heroin which was previously impossible with other currently available methods. Moreover, this mismatch approach (i.e., using a cross-reactive hapten tracer) has never been reported in the literature. When compared with equilibrium dialysis combined with ultra-performance liquid chromatography/tandem mass spectrometry (ED-UPLC/MS/MS), this novel MST method yields similar binding affinity values for polyclonal antibodies to the major heroin metabolites 6-AM and morphine. Additionally, we herein report the method of synthesis of this novel cross-reactive hapten, MorHap-acetamide—a useful analog for the study of heroin hapten–antibody interactions. Using heterologous MST, we were able to determine the affinities, down to nanomolar accuracies, of polyclonal antibodies to various abused opioids. While optimizing this method, we further discovered that heroin is protected from serum esterase degradation by the presence of these antibodies in a concentration-dependent manner. Lastly, using affinity data for a number of structurally different opioids, we were able to dissect the moieties that are crucial to antibody binding. The novel MST method that is presented herein can be extended to the analysis of any ligand that is prone to degradation and can be applied not only to the development of vaccines to substances of abuse but also to the analysis of small molecule/protein interactions in the presence of serum. Strategy for the determination of hapten-induced antibody affinities using Microscale thermophoresis ![]()
Collapse
|
13
|
Raleigh MD, Laudenbach M, Baruffaldi F, Peterson SJ, Roslawski MJ, Birnbaum AK, Carroll FI, Runyon SP, Winston S, Pentel PR, Pravetoni M. Opioid Dose- and Route-Dependent Efficacy of Oxycodone and Heroin Vaccines in Rats. J Pharmacol Exp Ther 2018. [PMID: 29535156 DOI: 10.1124/jpet.117.247049] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
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.
Collapse
Affiliation(s)
- Michael D Raleigh
- Minneapolis Medical Research Foundation, Minneapolis, Minnesota (M.D.R., M.L., F.B., S.J.P., P.R.P., M.P.); University of Minnesota College of Pharmacy, Minneapolis, Minnesota (M.J.R., A.K.B.); Research Triangle Institute, Research Triangle Park, North Carolina (F.I.C., S.P.R.); Winston Biopharmaceutical Consulting, Boulder, Colorado (S.W.); and University of Minnesota Medical School (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota, Minneapolis, Minnesota
| | - Megan Laudenbach
- Minneapolis Medical Research Foundation, Minneapolis, Minnesota (M.D.R., M.L., F.B., S.J.P., P.R.P., M.P.); University of Minnesota College of Pharmacy, Minneapolis, Minnesota (M.J.R., A.K.B.); Research Triangle Institute, Research Triangle Park, North Carolina (F.I.C., S.P.R.); Winston Biopharmaceutical Consulting, Boulder, Colorado (S.W.); and University of Minnesota Medical School (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota, Minneapolis, Minnesota
| | - Federico Baruffaldi
- Minneapolis Medical Research Foundation, Minneapolis, Minnesota (M.D.R., M.L., F.B., S.J.P., P.R.P., M.P.); University of Minnesota College of Pharmacy, Minneapolis, Minnesota (M.J.R., A.K.B.); Research Triangle Institute, Research Triangle Park, North Carolina (F.I.C., S.P.R.); Winston Biopharmaceutical Consulting, Boulder, Colorado (S.W.); and University of Minnesota Medical School (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota, Minneapolis, Minnesota
| | - Samantha J Peterson
- Minneapolis Medical Research Foundation, Minneapolis, Minnesota (M.D.R., M.L., F.B., S.J.P., P.R.P., M.P.); University of Minnesota College of Pharmacy, Minneapolis, Minnesota (M.J.R., A.K.B.); Research Triangle Institute, Research Triangle Park, North Carolina (F.I.C., S.P.R.); Winston Biopharmaceutical Consulting, Boulder, Colorado (S.W.); and University of Minnesota Medical School (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota, Minneapolis, Minnesota
| | - Michaela J Roslawski
- Minneapolis Medical Research Foundation, Minneapolis, Minnesota (M.D.R., M.L., F.B., S.J.P., P.R.P., M.P.); University of Minnesota College of Pharmacy, Minneapolis, Minnesota (M.J.R., A.K.B.); Research Triangle Institute, Research Triangle Park, North Carolina (F.I.C., S.P.R.); Winston Biopharmaceutical Consulting, Boulder, Colorado (S.W.); and University of Minnesota Medical School (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota, Minneapolis, Minnesota
| | - Angela K Birnbaum
- Minneapolis Medical Research Foundation, Minneapolis, Minnesota (M.D.R., M.L., F.B., S.J.P., P.R.P., M.P.); University of Minnesota College of Pharmacy, Minneapolis, Minnesota (M.J.R., A.K.B.); Research Triangle Institute, Research Triangle Park, North Carolina (F.I.C., S.P.R.); Winston Biopharmaceutical Consulting, Boulder, Colorado (S.W.); and University of Minnesota Medical School (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota, Minneapolis, Minnesota
| | - F Ivy Carroll
- Minneapolis Medical Research Foundation, Minneapolis, Minnesota (M.D.R., M.L., F.B., S.J.P., P.R.P., M.P.); University of Minnesota College of Pharmacy, Minneapolis, Minnesota (M.J.R., A.K.B.); Research Triangle Institute, Research Triangle Park, North Carolina (F.I.C., S.P.R.); Winston Biopharmaceutical Consulting, Boulder, Colorado (S.W.); and University of Minnesota Medical School (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota, Minneapolis, Minnesota
| | - Scott P Runyon
- Minneapolis Medical Research Foundation, Minneapolis, Minnesota (M.D.R., M.L., F.B., S.J.P., P.R.P., M.P.); University of Minnesota College of Pharmacy, Minneapolis, Minnesota (M.J.R., A.K.B.); Research Triangle Institute, Research Triangle Park, North Carolina (F.I.C., S.P.R.); Winston Biopharmaceutical Consulting, Boulder, Colorado (S.W.); and University of Minnesota Medical School (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota, Minneapolis, Minnesota
| | - Scott Winston
- Minneapolis Medical Research Foundation, Minneapolis, Minnesota (M.D.R., M.L., F.B., S.J.P., P.R.P., M.P.); University of Minnesota College of Pharmacy, Minneapolis, Minnesota (M.J.R., A.K.B.); Research Triangle Institute, Research Triangle Park, North Carolina (F.I.C., S.P.R.); Winston Biopharmaceutical Consulting, Boulder, Colorado (S.W.); and University of Minnesota Medical School (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota, Minneapolis, Minnesota
| | - Paul R Pentel
- Minneapolis Medical Research Foundation, Minneapolis, Minnesota (M.D.R., M.L., F.B., S.J.P., P.R.P., M.P.); University of Minnesota College of Pharmacy, Minneapolis, Minnesota (M.J.R., A.K.B.); Research Triangle Institute, Research Triangle Park, North Carolina (F.I.C., S.P.R.); Winston Biopharmaceutical Consulting, Boulder, Colorado (S.W.); and University of Minnesota Medical School (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota, Minneapolis, Minnesota
| | - Marco Pravetoni
- Minneapolis Medical Research Foundation, Minneapolis, Minnesota (M.D.R., M.L., F.B., S.J.P., P.R.P., M.P.); University of Minnesota College of Pharmacy, Minneapolis, Minnesota (M.J.R., A.K.B.); Research Triangle Institute, Research Triangle Park, North Carolina (F.I.C., S.P.R.); Winston Biopharmaceutical Consulting, Boulder, Colorado (S.W.); and University of Minnesota Medical School (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
14
|
Sulima A, Jalah R, Antoline JFG, Torres OB, Imler GH, Deschamps JR, Beck Z, Alving CR, Jacobson AE, Rice KC, Matyas GR. A Stable Heroin Analogue That Can Serve as a Vaccine Hapten to Induce Antibodies That Block the Effects of Heroin and Its Metabolites in Rodents and That Cross-React Immunologically with Related Drugs of Abuse. J Med Chem 2017; 61:329-343. [PMID: 29236495 PMCID: PMC5767880 DOI: 10.1021/acs.jmedchem.7b01427] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
![]()
An
improved synthesis of a haptenic heroin surrogate 1 (6-AmHap)
is reported. The intermediate needed for the preparation
of 1 was described in the route in the synthesis of 2 (DiAmHap). A scalable procedure was developed to install
the C-3 amido group. Using the Boc protectng group in 18 allowed preparation of 1 in an overall yield of 53%
from 4 and eliminated the necessity of preparing the
diamide 13. Hapten 1 was conjugated to tetanus
toxoid and mixed with liposomes containing monophosphoryl lipid A
as an adjuvant. The 1 vaccine induced high anti-1 IgG levels that reduced heroin-induced antinociception and
locomotive behavioral changes following repeated subcutaneous and
intravenous heroin challenges in mice and rats. Vaccinated mice had
reduced heroin-induced hyperlocomotion following a 50 mg/kg heroin
challenge. The 1 vaccine-induced antibodies bound to
heroin and other abused opioids, including hydrocodone, oxycodone,
hydromorphone, oxymorphone, and codeine.
Collapse
Affiliation(s)
- Agnieszka Sulima
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services , 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Rashmi Jalah
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine , 6720A Rockledge Drive, Bethesda, Maryland 20817, United States.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research , 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Joshua F G Antoline
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services , 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Oscar B Torres
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine , 6720A Rockledge Drive, Bethesda, Maryland 20817, United States.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research , 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Gregory H Imler
- Center for Biomolecular Science and Engineering, Naval Research Laboratory , Washington D.C. 20375, United States
| | - Jeffrey R Deschamps
- Center for Biomolecular Science and Engineering, Naval Research Laboratory , Washington D.C. 20375, United States
| | - Zoltan Beck
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine , 6720A Rockledge Drive, Bethesda, Maryland 20817, United States.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research , 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Carl R Alving
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research , 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| | - Arthur E Jacobson
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services , 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Kenner C Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services , 9800 Medical Center Drive, Bethesda, Maryland 20892-3373, United States
| | - Gary R Matyas
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research , 503 Robert Grant Avenue, Silver Spring, Maryland 20910, United States
| |
Collapse
|
15
|
Abstract
Substance use disorder, especially in relation to opioids such as heroin and fentanyl, is a significant public health issue and has intensified in recent years. As a result, substantial interest exists in developing therapeutics to counteract the effects of abused drugs. A promising universal strategy for antagonizing the pharmacology of virtually any drug involves the development of a conjugate vaccine, wherein a hapten structurally similar to the target drug is conjugated to an immunogenic carrier protein. When formulated with adjuvants and immunized, the immunoconjugate should elicit serum IgG antibodies with the ability to sequester the target drug to prevent its entry to the brain, thereby acting as an immunoantagonist. Despite the failures of first-generation conjugate vaccines against cocaine and nicotine in clinical trials, second-generation vaccines have shown dramatically improved performance in preclinical models, thus renewing the potential clinical utility of conjugate vaccines in curbing substance use disorder. This review explores the critical design elements of drug conjugate vaccines such as hapten structure, adjuvant formulation, bioconjugate chemistry, and carrier protein selection. Methods for evaluating these vaccines are discussed, and recent progress in vaccine development for each drug is summarized.
Collapse
Affiliation(s)
- Paul T Bremer
- Departments of Chemistry and Immunology, The Scripps Research Institute, La Jolla, California
| | - Kim D Janda
- Departments of Chemistry and Immunology, The Scripps Research Institute, La Jolla, California
| |
Collapse
|
16
|
Torres OB, Matyas GR, Rao M, Peachman KK, Jalah R, Beck Z, Michael NL, Rice KC, Jacobson AE, Alving CR. Heroin-HIV-1 (H2) vaccine: induction of dual immunologic effects with a heroin hapten-conjugate and an HIV-1 envelope V2 peptide with liposomal lipid A as an adjuvant. NPJ Vaccines 2017; 2:13. [PMID: 29263870 PMCID: PMC5604742 DOI: 10.1038/s41541-017-0013-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 03/15/2017] [Accepted: 03/21/2017] [Indexed: 12/20/2022] Open
Abstract
A synthetic heroin analog (MorHap) and a synthetic 42 amino acid V2 loop peptide from A/E strain of HIV-1 gp120 envelope protein that was previously used in a successful phase III vaccine trial were constructed as antigens together with liposomes containing monophosphoryl lipid A as an adjuvant, to explore the feasibility of producing a dual use vaccine both for treatment of heroin addiction and prevention of HIV-1 infection among injection drug users. The V2 peptide was tethered by a palmitoyl fatty acyl tail embedded in the liposomal lipid bilayer, and the heroin analog was conjugated to tetanus toxoid as a carrier protein that was mixed with the adjuvant. Upon comparison of a linear V2 peptide with a cyclic peptide, differences were found in the secondary configurations by circular dichroism, with the tethered cyclic peptide (palm-cyclic peptide) entirely in a random coil, and the tethered linear V2 peptide (palm-linear V2 peptide) entirely in a beta-sheet. Upon immunization of mice, palm-cyclic peptide induced anti-cyclic peptide endpoint titers >106 and was considered to be a better immunogen overall than palm-linear V2 peptide for inducing antibodies to gp120 and gp70-V1V2. The antibodies also inhibited the binding of V2 peptide to the HIV-1 α4β7 integrin receptor. Antibody titers to MorHap, even with the presence of injected cyclic peptide, were very high, and resulted in inhibition of the hyper-locomotion and antinociception effects of injected heroin. From these initial experiments, we conclude that with a potent adjuvant and mostly synthetic constituents, a vaccine directed to heroin and HIV-1 (H2 vaccine) could be a feasible objective. A vaccine designed to treat heroin addiction while at the same time preventing HIV infection elicited strong immune responses in mice. Scientists from the US government led by Carl Alving from the Walter Reed Army Institute of Research in Bethesda, Maryland, created a dual vaccine formulated with three main components: a segment of a protein expressed on the surface of HIV; synthetic molecules that resemble heroin and its degradation products; and a potent adjuvant to stimulate the immune system. Mice immunized with this vaccine had high antibody titers against the HIV surface protein as well as heroin and its derivatives. These mice also showed dulled responses to injected heroin. The findings suggest this vaccine strategy could help fight heroin abuse and the high risk of HIV infection among intravenous drug users.
Collapse
Affiliation(s)
- Oscar B Torres
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, 20817 MD USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Gary R Matyas
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Mangala Rao
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Kristina K Peachman
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, 20817 MD USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Rashmi Jalah
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, 20817 MD USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Zoltan Beck
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, 20817 MD USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Nelson L Michael
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Kenner C Rice
- Department of Health and Human Services, Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, 9800 Medical Drive, Bethesda, 20892 MD USA.,National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 9800 Medical Drive, Bethesda, 20892 MD USA
| | - Arthur E Jacobson
- Department of Health and Human Services, Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, 9800 Medical Drive, Bethesda, 20892 MD USA.,National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 9800 Medical Drive, Bethesda, 20892 MD USA
| | - Carl R Alving
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| |
Collapse
|
17
|
Zhang Y, Zeng K, Wang J, Gao H, Nan Y, Zheng X. Identifying the antiasthmatic target of doxofylline using immobilized β2 -adrenoceptor based high-performance affinity chromatography and site-directed molecular docking. J Mol Recognit 2016; 29:492-8. [PMID: 27173639 DOI: 10.1002/jmr.2549] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 02/28/2016] [Accepted: 04/06/2016] [Indexed: 01/16/2023]
Abstract
As a xanthine derivative, doxofylline is believed to be dominant for fighting against asthma in practice. Unlike other xanthines, the antiasthmatic effects of doxofylline lack any definite proof of target and mediating mechanism according to previous reports. In this work, the interaction between doxofylline and β2 -AR was investigated by high performance affinity chromatography using frontal analysis and nonlinear model. The methodology involved the immobilization of β2 -AR on the silica gel by a random linking method, the determination of the binding parameters by frontal analysis and nonlinear chromatography and the exploration of the binding mechanism by site-directed molecular docking. The association constant for doxofylline binding to immobilized β2 -AR was determined to be 7.70 × 10(4) M(-1) by nonlinear chromatography and 5.91 × 10(4) M(-1) by frontal analysis. Ser(169) and Ser(173) were the binding sites for the receptor-drug interaction on which hydrogen bond was believed to be the main driven force during the interaction. These results indicated that the antiasthmatic effects of doxofylline may be behind the mediating mechanism of β2 -AR. High performance affinity chromatography based on immobilized receptor has potential to become an alternative for drug target confirmation and drug-receptor interaction analysis. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yajun Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China.
| | - Kaizhu Zeng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Jing Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Haiyang Gao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Yefei Nan
- College of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| |
Collapse
|