1
|
Chang J, Shin K, Lewis JM, Suh HW, Lee J, Damsky W, Xu S, Bosenberg M, Saltzman WM, Girardi M. Enhanced Intratumoral Delivery of Immunomodulator Monophosphoryl Lipid A through Hyperbranched Polyglycerol-Coated Biodegradable Nanoparticles. J Invest Dermatol 2025; 145:593-604. [PMID: 39122142 DOI: 10.1016/j.jid.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
Immunomodulatory agents have significant potential to enhance cancer treatment but have demonstrated limited efficacy beyond the preclinical setting owing to poor pharmacokinetics and toxicity associated with systemic administration. Conversely, when locally delivered, immunomodulatory agents require repeated administration to optimize immune stimulation. To overcome these challenges, we encapsulated the toll-like receptor 4 agonist monophosphoryl lipid A (MPLA) within hyperbranched polyglycerol-coated biodegradable nanoparticles (NPs) engineered for gradual drug release from the NP core, resulting in a more persistent stimulation of antitumor immune responses while minimizing systemic side effects. In a model of malignant melanoma, we demonstrate that hyperbranched polyglycerol-NP encapsulation significantly improves the antitumor efficacy of MPLA by enhancing its ability to remodel the tumor microenvironment. Relative to free MPLA, hyperbranched polyglycerol-coated NP-encapsulated MPLA significantly increased the NK cell- and cytotoxic T-cell-mediated antitumor immune response and tuned the tumor-draining lymph nodes toward a T helper 1 response. Furthermore, when combined with local delivery of a chemotherapeutic agent, hyperbranched polyglycerol-NP-MPLA induces the conversion of an immunosuppressive tumor microenvironment to immunogenic tumor microenvironment and significantly improves survival.
Collapse
Affiliation(s)
- Jungsoo Chang
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Kwangsoo Shin
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Julia M Lewis
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Hee Won Suh
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Joohyung Lee
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - William Damsky
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Suzanne Xu
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Marcus Bosenberg
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, Connecticut, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA; Yale Center for Immuno-Oncology, Yale School of Medicine, New Haven, Connecticut, USA
| | - W Mark Saltzman
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA; Department of Chemical & Environmental Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michael Girardi
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
2
|
Tuncturk M, Kushwaha S, Heider RM, Oesterle T, Weinshilboum R, Ho MF. The development of opioid vaccines as a novel strategy for the treatment of opioid use disorder and overdose prevention. Int J Neuropsychopharmacol 2025; 28:pyaf005. [PMID: 39831679 PMCID: PMC11792077 DOI: 10.1093/ijnp/pyaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/17/2025] [Indexed: 01/22/2025] Open
Abstract
Opioid use disorder (OUD) affects over 40 million people worldwide, creating significant social and economic burdens. Medication for opioid use disorder (MOUD) is often considered the primary treatment approach for OUD. MOUD, including methadone, buprenorphine, and naltrexone, is effective for some, but its benefits may be limited by poor adherence to treatment recommendations. Immunopharmacotherapy offers an innovative approach by using vaccines to generate antibodies that neutralize opioids, blocking them from crossing the blood-brain barrier and reducing their psychoactive effects. To date, only 3 clinical trials for opioid vaccines have been published. While these studies demonstrated the potential of opioid vaccines for relapse prevention, there is currently no standardized protocol for evaluating their effectiveness. We have reviewed recent preclinical studies that demonstrated the efficacy of vaccines targeting opioids, including heroin, morphine, oxycodone, hydrocodone, and fentanyl. These studies showed that vaccines against opioids reduced drug reinforcement, decreased opioid-induced antinociception, and increased survival rates against lethal opioid doses. These studies also demonstrated the importance of vaccine formulation and the use of adjuvants in enhancing antibody production and specificity. Finally, we highlighted the strengths and concerns associated with the opioid vaccine treatment, including ethical considerations.
Collapse
Affiliation(s)
- Mustafa Tuncturk
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Shikha Kushwaha
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Robin M Heider
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Tyler Oesterle
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Ming-Fen Ho
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
3
|
Hamid FA, Le NMN, Song D, Amin H, Hicks L, Bird S, Siram K, Hoppe B, Demeler B, Evans JT, Burkhart D, Pravetoni M. A cationic liposome-formulated Toll Like Receptor (TLR)7/8 agonist enhances the efficacy of a vaccine against fentanyl toxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631964. [PMID: 39868149 PMCID: PMC11761771 DOI: 10.1101/2025.01.08.631964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The U.S. opioid epidemic is an extraordinary public health crisis that started in 1990 and significantly accelerated in the last decade. Since 2020, over 100,000 fatal drug overdoses have been reported annually, and 75% of those involved fentanyl and its analogs (F/FA). Accelerating the translation of innovative, effective, and safe treatments is needed to augment existing measures to counteract such a crisis. Active immunization against F/FA and other opioids represents a promising therapeutic and prophylactic strategy for opioid use disorder (OUD) and opioid-induced overdose toxicity. Previously we demonstrated that the anti-fentanyl vaccine comprising a fentanyl-based hapten (F) conjugated to the diphtheria cross-reactive material (CRM), admixed with the novel lipidated toll-like receptor 7/8 (TLR7/8) agonist INI-4001 adsorbed on Alhydrogel ® (alum) induced high-affinity fentanyl-specific polyclonal antibodies that protected against fentanyl-induced pharmacological effects in mice, rats, and mini-pigs. Here, INI-4001 was formulated into liposomes with different surface charges, and their impact on F-CRM adsorption, INI-4001 adjuvanticity, and vaccine efficacy were explored. Additionally, as the role of innate immunity in mediating the efficacy of addiction vaccines is largely unknown, we tested these formulations on the activation of innate immunity in vitro . Cationic INI-4001 liposomes surpassed other liposomal and aluminum-based formulations of INI-4001 by enhancing the efficacy of fentanyl vaccines and protecting rats against bradycardia and respiratory depression by blocking the distribution of fentanyl to the brain. Fentanyl vaccines adjuvanted with either cationic INI-4001 liposomes or the aqueous INI-4001 adsorbed to alum induced significant surface expression of co-stimulatory molecules and maturation markers in a murine dendritic cell line (JAWS II), while the former was superior in enhancing the macrophages surface expression of CD40, CD86 and inducible nitric oxide synthase (iNOS), indicative of maturation and activation. These results warrant further investigation of liposome-based formulations of TLR7/8 agonists for improving the efficacy of vaccines targeting F/FA and other opioids of public health interest. Graphical abstract
Collapse
|
4
|
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
|
5
|
Abstract
Substance use disorders (SUD) present a worldwide challenge with few effective therapies except for the relative efficacy of opioid pharmacotherapies, despite limited treatment access. However, the proliferation of illicit fentanyl use initiated a dramatic and cascading epidemic of lethal overdoses. This rise in fentanyl overdoses regenerated an interest in vaccine immunotherapy, which, despite an optimistic start in animal models over the past 50 years, yielded disappointing results in human clinical trials of vaccines against nicotine, stimulants (cocaine and methamphetamine), and opioids. After a brief review of clinical and selected preclinical vaccine studies, the "lessons learned" from the previous vaccine clinical trials are summarized, and then the newest challenge of a vaccine against fentanyl and its analogs is explored. Animal studies have made significant advances in vaccine technology for SUD treatment over the past 50 years, and the resulting anti-fentanyl vaccines show remarkable promise for ending this epidemic of fentanyl deaths.
Collapse
Affiliation(s)
- Thomas R Kosten
- Waggoner Professor of Psychiatry, Pharmacology, Neuroscience, Immunology, Baylor College of Medicine, Houston
| |
Collapse
|
6
|
Powers N, Massena C, Crouse B, Smith M, Hicks L, Evans JT, Miller S, Pravetoni M, Burkhart D. Self-Adjuvanting TLR7/8 Agonist and Fentanyl Hapten Co-Conjugate Achieves Enhanced Protection against Fentanyl Challenge. Bioconjug Chem 2023; 34:1811-1821. [PMID: 37758302 PMCID: PMC10587865 DOI: 10.1021/acs.bioconjchem.3c00347] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/12/2023] [Indexed: 10/03/2023]
Abstract
Currently approved pharmacotherapies for opioid use disorders (OUDs) and overdose reversal agents are insufficient to slow the spread of OUDs due to the proliferation of fentanyl. This is evident in the 31% rise in drug overdose deaths from 2019 to 2022, with rates increasing from 21.6 to 28.3 overdoses per 100,000 deaths. Vaccines are a potential alternative or adjunct therapy for the treatment of several substance use disorders (nicotine, cocaine) but have shown limited clinical success due to suboptimal antibody titers. In this study, we demonstrate that coconjugation of a Toll-like receptor 7/8 (TLR7/8) agonist (UM-3006) alongside a fentanyl-based hapten (F1) on the surface of the carrier protein cross-reactive material 197 (CRM) significantly increased generation of high-affinity fentanyl-specific antibodies. This demonstrated enhanced protection against fentanyl challenges relative to an unconjugated (admix) adjuvant control in mice. Inclusion of aluminum hydroxide (alum) adjuvant further increased titers and enhanced protection, as determined by analysis of fentanyl concentration in serum and brain tissue. Collectively, our findings present a promising approach to enhance the efficacy of antiopioid vaccines, underscoring the need for extensive exploration of TLR7/8 agonist conjugates as a compelling strategy to combat opioid use disorders.
Collapse
Affiliation(s)
- Noah Powers
- Center
for Translational Medicine, Department of Biomedical and Pharmaceutical
Sciences, University of Montana, 32 Campus Drive, Skaggs Building, Missoula, Montana 59801, United States
| | - Casey Massena
- Center
for Translational Medicine, Department of Biomedical and Pharmaceutical
Sciences, University of Montana, 32 Campus Drive, Skaggs Building, Missoula, Montana 59801, United States
| | - Bethany Crouse
- Department
of Pharmacology, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mira Smith
- Center
for Translational Medicine, Department of Biomedical and Pharmaceutical
Sciences, University of Montana, 32 Campus Drive, Skaggs Building, Missoula, Montana 59801, United States
| | - Linda Hicks
- Center
for Translational Medicine, Department of Biomedical and Pharmaceutical
Sciences, University of Montana, 32 Campus Drive, Skaggs Building, Missoula, Montana 59801, United States
| | - Jay T. Evans
- Center
for Translational Medicine, Department of Biomedical and Pharmaceutical
Sciences, University of Montana, 32 Campus Drive, Skaggs Building, Missoula, Montana 59801, United States
| | - Shannon Miller
- Center
for Translational Medicine, Department of Biomedical and Pharmaceutical
Sciences, University of Montana, 32 Campus Drive, Skaggs Building, Missoula, Montana 59801, United States
| | - Marco Pravetoni
- Department
of Psychiatry and Behavioral Sciences, University
of Washington School of Medicine, Seattle, Washington 98195, United States
| | - David Burkhart
- Center
for Translational Medicine, Department of Biomedical and Pharmaceutical
Sciences, University of Montana, 32 Campus Drive, Skaggs Building, Missoula, Montana 59801, United States
| |
Collapse
|
7
|
Miller SM, Crouse B, Hicks L, Amin H, Cole S, Bazin HG, Burkhart DJ, Pravetoni M, Evans JT. A lipidated TLR7/8 adjuvant enhances the efficacy of a vaccine against fentanyl in mice. NPJ Vaccines 2023; 8:97. [PMID: 37429853 PMCID: PMC10333387 DOI: 10.1038/s41541-023-00694-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/22/2023] [Indexed: 07/12/2023] Open
Abstract
Opioid use disorders (OUD) and opioid-related fatal overdoses are a public health concern in the United States. Approximately 100,000 fatal opioid-related overdoses occurred annually from mid-2020 to the present, the majority of which involved fentanyl or fentanyl analogs. Vaccines have been proposed as a therapeutic and prophylactic strategy to offer selective and long-lasting protection against accidental or deliberate exposure to fentanyl and closely related analogs. To support the development of a clinically viable anti-opioid vaccine suitable for human use, the incorporation of adjuvants will be required to elicit high titers of high-affinity circulating antibodies specific to the target opioid. Here we demonstrate that the addition of a synthetic TLR7/8 agonist, INI-4001, but not a synthetic TLR4 agonist, INI-2002, to a candidate conjugate vaccine consisting of a fentanyl-based hapten, F1, conjugated to the diphtheria cross-reactive material (CRM), significantly increased generation of high-affinity F1-specific antibody concentrations, and reduced drug distribution to the brain after fentanyl administration in mice.
Collapse
Affiliation(s)
- Shannon M Miller
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT, USA
- Inimmune Corporation, Missoula, MT, USA
| | - Bethany Crouse
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, USA
| | - Linda Hicks
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT, USA
| | - Hardik Amin
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT, USA
| | - Shelby Cole
- Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Helene G Bazin
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT, USA
- Inimmune Corporation, Missoula, MT, USA
| | - David J Burkhart
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT, USA
- Inimmune Corporation, Missoula, MT, USA
| | - Marco Pravetoni
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Jay T Evans
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT, USA.
- Inimmune Corporation, Missoula, MT, USA.
| |
Collapse
|
8
|
Lee J, Eubanks LM, Zhou B, Janda KD. Development of an Effective Monoclonal Antibody against Heroin and Its Metabolites Reveals Therapies Have Mistargeted 6-Monoacetylmorphine and Morphine over Heroin. ACS CENTRAL SCIENCE 2022; 8:1464-1470. [PMID: 36313156 PMCID: PMC9615117 DOI: 10.1021/acscentsci.2c00977] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Indexed: 05/29/2023]
Abstract
The opioid epidemic is a global public health crisis that has failed to abate with current pharmaceutical treatments. Moreover, these FDA-approved drugs possess numerous problems such as adverse side effects, short half-lives, abuse potential, and recidivism after discontinued use. An alternative treatment model for opioid use disorders is immunopharmacotherapy, where antibodies are produced to inhibit illicit substances by sequestering the drug in the periphery. Immunopharmacotherapeutics against heroin have engaged both active and passive vaccines targeting heroin's metabolites, 6-monoacetylmorphine (6-AM) and morphine, since decades of research have stated that heroin's psychoactive and lethal effects are mainly attributed to these compounds. However, concerted efforts to develop effective immunopharmacotherapies against heroin abuse have faced little clinical advancement, suggesting a need for reassessing drug target selection. To address this issue, four unique monoclonal antibodies were procured with distinct affinity to either heroin, 6-AM, or morphine. Examination of these antibodies through in vitro and in vivo tests revealed monoclonal antibody 11D12 as the optimal therapeutic and provided crucial insights into the key chemical species to target for blunting heroin's psychoactive and lethal effects. These findings offer clarification into the problematic attempts of therapeutics targeting heroin's metabolites and provide a path forward for future heroin immunopharmacotherapy development.
Collapse
|
9
|
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: 5] [Impact Index Per Article: 1.7] [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
|
10
|
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: 20] [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
|
11
|
The M3-TT Vaccine Decreases the Antinociceptive Effects of Morphine and Heroin in Mice. Int J Ment Health Addict 2021. [DOI: 10.1007/s11469-021-00621-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
12
|
Hosseini SA, Zahedipour F, Mirzaei H, Kazemi Oskuee R. Potential SARS-CoV-2 vaccines: Concept, progress, and challenges. Int Immunopharmacol 2021; 97:107622. [PMID: 33895475 PMCID: PMC8006194 DOI: 10.1016/j.intimp.2021.107622] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Since September 2020, the world has had more than 28 million cases of coronavirus disease 2019 (COVID-19). Many countries are facing a second wave of the COVID-19 outbreak. A pressing need is evident for the development of a potent vaccine to control the SARS-CoV-2. Institutions and companies in many countries have announced their vaccine research programs and progress against the COVID-19. While most vaccines go through the designation and preparation stages, some of them are under evaluation for efficacy among animal models and clinical trials, and three approved vaccine candidates have been introduced for limited exploitation in Russia and China. An effective vaccine must induce a protective response of both cell-mediated and humoral immunity and should meet the safety and efficacy criteria. Although the emergence of new technologies has accelerated the development of vaccines, there are several challenges on the way, such as limited knowledge about the pathophysiology of the virus, inducing humoral or cellular immunity, immune enhancement with animal coronavirus vaccines, and lack of an appropriate animal model. In this review, we firstly discuss the immune responses against SARS-CoV-2 disease, subsequently, give an overview of several vaccine platforms for SARS-CoV-2 under clinical trials and challenges in vaccine development against this virus.
Collapse
Affiliation(s)
- Seyede Atefe Hosseini
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran,Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran,Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran,Corresponding authors
| | - Reza Kazemi Oskuee
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran,Corresponding authors
| |
Collapse
|
13
|
Barbosa-Méndez S, Matus-Ortega M, Hernández-Miramontes R, Salazar-Juárez A. The morphine/heroin vaccine decreased the heroin-induced antinociceptive and reinforcing effects in three inbred strains mouse. Int Immunopharmacol 2021; 98:107887. [PMID: 34186279 DOI: 10.1016/j.intimp.2021.107887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 11/25/2022]
Abstract
Clinical trials have indicated that a vaccine must be immunogenic in genetically diverse human populations and that immunogenicity and protective efficacy in animal models are two key indices required for the approval of a new vaccine. Additionally, the immune response (immunogenicity) and immunoprotection are dependent on the mouse strain. Therefore, the objective of the present study was to determine the immune response (immunogenicity) and the protective efficacy (behavioral response) in three inbred mouse strains immunized with the M6TT vaccine. Female BALB/c, C57Bl/6, and DBA/2 inbred mice were immunized with the M6-TT vaccine. A solid-phase antibody-capture ELISA was used to monitor antibody titer responses after each booster dose in vaccinated animals. The study used tail-flick testing to evaluate the antinociceptive effects induced by heroin. Additionally, heroin-induced locomotor activity and place preference were evaluated. The M6-TT vaccine was able to generate a specific antibody titer in the three inbred mouse strains evaluated. The antibodies reduced the antinociceptive effect of different doses of heroin. In addition, they decreased the heroin-induced locomotor activity and place preference. These findings suggest that the M6-TT vaccine generates a powerful immunogenic response capable of reducing the antinociceptive and reinforcing effects of heroin in different inbred mouse strains, which supports its possible future use in clinical trials in genetically diverse human populations.
Collapse
Affiliation(s)
- Susana Barbosa-Méndez
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Instituto Nacional de Psiquiatría, México DF 14370, Mexico
| | - Maura Matus-Ortega
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Instituto Nacional de Psiquiatría, México DF 14370, Mexico
| | - Ricardo Hernández-Miramontes
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Instituto Nacional de Psiquiatría, México DF 14370, Mexico
| | - Alberto Salazar-Juárez
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Instituto Nacional de Psiquiatría, México DF 14370, Mexico.
| |
Collapse
|
14
|
Sei CJ, Rao M, Schuman RF, Daum LT, Matyas GR, Rikhi N, Muema K, Anderson A, Jobe O, Kroscher KA, Alving CR, Fischer GW. Conserved Influenza Hemagglutinin, Neuraminidase and Matrix Peptides Adjuvanted with ALFQ Induce Broadly Neutralizing Antibodies. Vaccines (Basel) 2021; 9:vaccines9070698. [PMID: 34202178 PMCID: PMC8310080 DOI: 10.3390/vaccines9070698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/15/2021] [Accepted: 06/21/2021] [Indexed: 01/07/2023] Open
Abstract
A universal influenza candidate vaccine that targets multiple conserved influenza virus epitopes from hemagglutinin (HA), neuraminidase (NA) and matrix (M2e) proteins was combined with the potent Army liposomal adjuvant (ALFQ) to promote induction of broad immunity to seasonal and pandemic influenza strains. The unconjugated and CRM-conjugated composite peptides formulated with ALFQ were highly immunogenic and induced both humoral and cellular immune responses in mice. Broadly reactive serum antibodies were induced across various IgG isotypes. Mice immunized with the unconjugated composite peptide developed antibody responses earlier than mice immunized with conjugated peptides, and the IgG antibodies were broadly reactive and neutralizing across Groups 1 and 2 influenza viruses. Multi-epitope unconjugated influenza composite peptides formulated with ALFQ provide a novel strategy for the development of a universal influenza vaccine. These synthetic peptide vaccines avoid the pitfalls of egg-produced influenza vaccines and production can be scaled up rapidly and economically.
Collapse
Affiliation(s)
- Clara J. Sei
- Longhorn Vaccines and Diagnostics, Gaithersburg, MD 20878, USA; (N.R.); (K.M.); (K.A.K.); (G.W.F.)
- Correspondence: ; Tel.: +1-240-848-4293
| | - Mangala Rao
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (M.R.); (G.R.M.); (A.A.); (O.J.); (C.R.A.)
| | | | - Luke T. Daum
- Longhorn Vaccines and Diagnostics, San Antonio, TX 78209, USA;
| | - Gary R. Matyas
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (M.R.); (G.R.M.); (A.A.); (O.J.); (C.R.A.)
| | - Nimisha Rikhi
- Longhorn Vaccines and Diagnostics, Gaithersburg, MD 20878, USA; (N.R.); (K.M.); (K.A.K.); (G.W.F.)
| | - Kevin Muema
- Longhorn Vaccines and Diagnostics, Gaithersburg, MD 20878, USA; (N.R.); (K.M.); (K.A.K.); (G.W.F.)
| | - Alexander Anderson
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (M.R.); (G.R.M.); (A.A.); (O.J.); (C.R.A.)
- Oak Ridge Institute of Science and Education, Oak Ridge, TN 37831, USA
| | - Ousman Jobe
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (M.R.); (G.R.M.); (A.A.); (O.J.); (C.R.A.)
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Kellie A. Kroscher
- Longhorn Vaccines and Diagnostics, Gaithersburg, MD 20878, USA; (N.R.); (K.M.); (K.A.K.); (G.W.F.)
| | - Carl R. Alving
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (M.R.); (G.R.M.); (A.A.); (O.J.); (C.R.A.)
| | - Gerald W. Fischer
- Longhorn Vaccines and Diagnostics, Gaithersburg, MD 20878, USA; (N.R.); (K.M.); (K.A.K.); (G.W.F.)
| |
Collapse
|
15
|
Köteles I, Mazák K, Tóth G, Horváth P, Kiss E, Tűz B, Hosztafi S. Synthesis of 3-O-Carboxyalkyl Morphine Derivatives and Characterization of Their Acid-Base Properties. Chem Biodivers 2021; 18:e2100135. [PMID: 34018677 DOI: 10.1002/cbdv.202100135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/18/2021] [Indexed: 11/11/2022]
Abstract
The C-3 phenolic hydroxy group containing morphine derivatives (morphine, oxymorphone, naloxone, naltrexone) are excellent candidates for the synthesis of 3-O-functionalized molecules. Achieving free carboxylic group containing derivatives gives the opportunity for further modification and conjugation that could be used for immunization and immunoassays. For this purpose ethyl bromo- and chloroacetate can be used as O-alkylating agents. Hydrolyzing the products affords the appropriate free carboxylic group containing 3-O-carboxyalkyl derivatives. As these molecules contain an acidic and a basic functional group the protonation macro- and microconstants were determined too, using pH-potentiometry and NMR-pH titration, beside fully characterizing their structure using IR, CD, NMR and HR-MS measurements.
Collapse
Affiliation(s)
- István Köteles
- Department of Pharmaceutical Chemistry, Semmelweis University, Hőgyes Endre u. 9., H-1092, Budapest, Hungary
| | - Károly Mazák
- Department of Pharmaceutical Chemistry, Semmelweis University, Hőgyes Endre u. 9., H-1092, Budapest, Hungary
| | - Gergő Tóth
- Department of Pharmaceutical Chemistry, Semmelweis University, Hőgyes Endre u. 9., H-1092, Budapest, Hungary
| | - Péter Horváth
- Department of Pharmaceutical Chemistry, Semmelweis University, Hőgyes Endre u. 9., H-1092, Budapest, Hungary
| | - Eszter Kiss
- Department of Pharmaceutical Chemistry, Semmelweis University, Hőgyes Endre u. 9., H-1092, Budapest, Hungary
| | - Boglárka Tűz
- Department of Pharmaceutical Chemistry, Semmelweis University, Hőgyes Endre u. 9., H-1092, Budapest, Hungary
| | - Sándor Hosztafi
- Department of Pharmaceutical Chemistry, Semmelweis University, Hőgyes Endre u. 9., H-1092, Budapest, Hungary
| |
Collapse
|
16
|
Alving CR. Walter Reed Army Institute of Research (WRAIR): Fifty Years of Achievements That Impact Science and Society. Mil Med 2021; 186:72-77. [PMID: 33313776 PMCID: PMC7909454 DOI: 10.1093/milmed/usaa468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
Thirty-four historical achievements since 1970 that emanated from scientific research at the Walter Army Institute of Research are identified and documented. Impact areas include vaccines, drug development, and clinical assays to prevent or treat infectious diseases; neuropsychiatric management of warrior performance and combat casualty; blood delivery management; and radiation protection.
Collapse
Affiliation(s)
- Carl R Alving
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Silver Spring, MD 20910, USA
| |
Collapse
|
17
|
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: 1.0] [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
|
18
|
Blake S, Bremer PT, Zhou B, Petrovsky N, Smith LC, Hwang CS, Janda KD. Developing Translational Vaccines against Heroin and Fentanyl through Investigation of Adjuvants and Stability. Mol Pharm 2021; 18:228-235. [PMID: 33301675 PMCID: PMC9946458 DOI: 10.1021/acs.molpharmaceut.0c00837] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The nearly insurmountable adversity that accompanies opioid use disorder (OUD) creates life-altering complications for opioid users. To worsen matters, existing small-molecule drugs continue to inadequately address OUD due to their engagement of the opioid receptor, which can leave the user to deal with side effects and financial hardships from their repeated use. An alternative therapeutic approach utilizes endogenously generated antibodies through active vaccination to reduce the effect of opioids without modulating the opioid receptor. Here, we explore different adjuvants and storage conditions to improve opioid vaccine efficacy and shelf life. Our results revealed that inulin-based formulations (Advax) containing a CpG oligodeoxynucleotide (ODN) acted as effective adjuvants when combined with a heroin conjugate: immunized mice showed excellent recovery from heroin-induced antinociception accompanied by high titer, high opioid affinity serum antibodies similar to the immunopotentiating properties of traditional alum-based adjuvants. Moreover, nonhuman primates vaccinated with a heroin/fentanyl combination vaccine demonstrated potent antibody responses against opioids when formulated with both inulin and alum adjuvants. Finally, storing a freeze-dried opioid vaccine formulation maintained efficacy for up 1 year at room temperature. The results from our studies represent an advance toward a clinically feasible opioid vaccine.
Collapse
Affiliation(s)
- Steven Blake
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Paul T. Bremer
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States,Cessation Therapeutics, LLC, 3031 Tisch Way, San Jose, California 95128, United States
| | - Bin Zhou
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Nikolai Petrovsky
- Flinders Medical Centre, Flinders University, GPO Box 2100, Adelaide 5001, South Australia, Australia,Vaxine Pty Ltd, 11 Walkley Avenue, Warradale 5046, South Australia, Australia
| | - Lauren C. Smith
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Candy S. Hwang
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, 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, 10550 North Torrey Pines Road, La Jolla, California 92037, United States,Corresponding Author: Kim D. Janda - The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States. Phone: (858), 785-2515. Fax: (858) 784-2595. .
| |
Collapse
|
19
|
Cheng R, Fontana F, Xiao J, Liu Z, Figueiredo P, Shahbazi MA, Wang S, Jin J, Torrieri G, Hirvonen JT, Zhang H, Chen T, Cui W, Lu Y, Santos HA. Recombination Monophosphoryl Lipid A-Derived Vacosome for the Development of Preventive Cancer Vaccines. ACS APPLIED MATERIALS & INTERFACES 2020; 12:44554-44562. [PMID: 32960566 PMCID: PMC7549091 DOI: 10.1021/acsami.0c15057] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/09/2020] [Indexed: 05/09/2023]
Abstract
Recently, there has been an increasing interest for utilizing the host immune system to fight against cancer. Moreover, cancer vaccines, which can stimulate the host immune system to respond to cancer in the long term, are being investigated as a promising approach to induce tumor-specific immunity. In this work, we prepared an effective cancer vaccine (denoted as "vacosome") by reconstructing the cancer cell membrane, monophosphoryl lipid A as a toll-like receptor 4 agonist, and egg phosphatidylcholine. The vacosome triggered and enhanced bone marrow dendritic cell maturation as well as stimulated the antitumor response against breast cancer 4T1 cells in vitro. Furthermore, an immune memory was established in BALB/c mice after three-time preimmunization with the vacosome. After that, the immunized mice showed inhibited tumor growth and prolonged survival period (longer than 50 days). Overall, our results demonstrate that the vacosome can be a potential candidate for clinical translation as a cancer vaccine.
Collapse
Affiliation(s)
- Ruoyu Cheng
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Flavia Fontana
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Junyuan Xiao
- Shanghai Key Laboratory for Prevention and Treatment
of Bone and Joint Diseases, Shanghai Institute of Traumatology and
Orthopaedics, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, 197 Ruijin Second Road, 200025 Shanghai, PR China
| | - Zehua Liu
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Patrícia Figueiredo
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Mohammad-Ali Shahbazi
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran
| | - Shiqi Wang
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jing Jin
- Shanghai Key Laboratory for Prevention and Treatment
of Bone and Joint Diseases, Shanghai Institute of Traumatology and
Orthopaedics, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, 197 Ruijin Second Road, 200025 Shanghai, PR China
| | - Giulia Torrieri
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jouni T. Hirvonen
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Hongbo Zhang
- Shanghai Key Laboratory for Prevention and Treatment
of Bone and Joint Diseases, Shanghai Institute of Traumatology and
Orthopaedics, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, 197 Ruijin Second Road, 200025 Shanghai, PR China
- Department of Pharmaceutical Sciences Laboratory and
Turku Center for Biotechnology, Åbo
Akademi University, FI-20520 Turku, Finland
| | - Tongtong Chen
- Radiology Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025 Shanghai, PR China
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment
of Bone and Joint Diseases, Shanghai Institute of Traumatology and
Orthopaedics, Ruijin Hospital, Shanghai
Jiao Tong University School of Medicine, 197 Ruijin Second Road, 200025 Shanghai, PR China
| | - Yong Lu
- Radiology Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025 Shanghai, PR China
| | - Hélder A. Santos
- Drug Research Program,
Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
- Helsinki Insititute of Life Science, HiLIFE, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
20
|
Synthesis of Potential Haptens with Morphine Skeleton and Determination of Protonation Constants. Molecules 2020; 25:molecules25174009. [PMID: 32887468 PMCID: PMC7504778 DOI: 10.3390/molecules25174009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/24/2020] [Accepted: 09/01/2020] [Indexed: 11/16/2022] Open
Abstract
Vaccination could be a promising alternative warfare against drug addiction and abuse. For this purpose, so-called haptens can be used. These molecules alone do not induce the activation of the immune system, this occurs only when they are attached to an immunogenic carrier protein. Hence obtaining a free amino or carboxylic group during the structural transformation is an important part of the synthesis. Namely, these groups can be used to form the requisite peptide bond between the hapten and the carrier protein. Focusing on this basic principle, six nor-morphine compounds were treated with ethyl acrylate and ethyl bromoacetate, while the prepared esters were hydrolyzed to obtain the N-carboxymethyl- and N-carboxyethyl-normorphine derivatives which are considered as potential haptens. The next step was the coupling phase with glycine ethyl ester, but the reactions did not work or the work-up process was not accomplishable. As an alternative route, the normorphine-compounds were N-alkylated with N-(chloroacetyl)glycine ethyl ester. These products were hydrolyzed in alkaline media and after the work-up process all of the derivatives contained the free carboxylic group of the glycine side chain. The acid-base properties of these molecules are characterized in detail. In the N-carboxyalkyl derivatives, the basicity of the amino and phenolate site is within an order of magnitude. In the glycine derivatives the basicity of the amino group is significantly decreased compared to the parent compounds (i.e., morphine, oxymorphone) because of the electron withdrawing amide group. The protonation state of the carboxylate group significantly influences the basicity of the amino group. All of the glycine ester and the glycine carboxylic acid derivatives are currently under biological tests.
Collapse
|
21
|
Wallis J, Katti P, Martin AM, Hills T, Seymour LW, Shenton DP, Carlisle RC. A liposome-based cancer vaccine for a rapid and high-titre anti-ErbB-2 antibody response. Eur J Pharm Sci 2020; 152:105456. [PMID: 32653563 DOI: 10.1016/j.ejps.2020.105456] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/10/2020] [Accepted: 07/02/2020] [Indexed: 11/26/2022]
Abstract
Vaccines are arguably the most important medical technology developed to date. However, effective treatment of diseases such as breast cancer have so far evaded standard vaccination strategies. One popular target for cancer treatment is the cell surface membrane protein, ErbB-2, also known as Her-2 or neu. It is localised to the cell surface and has raised expression in 15-30% of all breast cancers, as well as in ovarian, colon and lung cancer. Here, a liposomal system comprised of spatially separated ErbB-2 peptide, to activate B cells, and ovalbumin peptide OVA323-339, to provide non-cognate T cell support, was used to generate antibodies against the epitope of the ErbB-2 protein targeted by Pertuzumab, a monoclonal antibody licensed for the treatment of ErbB-2 expressing cancers. After just 7 days a raised (7.3-fold, p<0.01), isotype-switched, humoral immune response specific for the ErbB-2 peptide was achieved in mice with pre-existing immunity to OVA which were exposed to liposomes with external ErbB-2 and internal OVA323-339. The absence of pre-existing OVA immunity in the mice or OVA323-339 peptide in the liposomes removed the effect. The effect of this anti-ErbB-2 antibody response was characterised against an ErbB-2 overexpressing tumour cell line both in vitro and in vivo. Notably, antibody responses were demonstrated to induce cell death in vitro, resulting in 96% reduction in viable cells. This study, therefore, demonstrates the feasibility of this approach to generate a rapid, high-titre, isotype-switched, antibody response that specifically targets ErbB-2 overexpression on tumour cells and is capable of inducing cell death in vitro in the absence of complement or immune cells.
Collapse
Affiliation(s)
- Jamie Wallis
- Institute of Biomedical Engineering, University of Oxford, UK
| | - Prateek Katti
- Institute of Biomedical Engineering, University of Oxford, UK
| | | | - Tom Hills
- Department of Oncology, University of Oxford, UK
| | | | | | | |
Collapse
|
22
|
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: 29] [Impact Index Per Article: 5.8] [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
|
23
|
Mella-Raipán J, Romero-Parra J, Recabarren-Gajardo G. DARK Classics in Chemical Neuroscience: Heroin and Desomorphine. ACS Chem Neurosci 2020; 11:3905-3927. [PMID: 32568519 DOI: 10.1021/acschemneuro.0c00262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Opioids are arguably one of the most important pharmacologic classes, mainly due to their rich history, their useful and potent analgesic effects, and also, just as importantly, their "Dark Side", constituted by their reinforcing properties that have led countless of users to a spiral of addiction, biological dependence, tolerance, withdrawal syndromes, and death. Among the most significant abused and addictive known opioids are heroin and desomorphine, both synthetic derivatives of morphine that belong to the 4,5-epoxymorphinan structural chemical group of the opioid family drugs. These agents share not only structural, pharmacological, and epidemiological features but also a common geographical distribution. A drop in Afghan heroin production and its "exports" to Russia gave rise to widespread consumption of desomorphine in ex-Soviet republics during the first decade of the 21st century, representing an economical and accessible alternative for misusers to this sort of derivative. Herein we review the state of the art of history, chemistry and synthesis, pharmacology, and impact on society of these "cursed cousins".
Collapse
Affiliation(s)
- Jaime Mella-Raipán
- Instituto de Quı́mica y Bioquı́mica, Facultad de Ciencias, Universidad de Valparaı́so, Av. Gran Bretaña 1111, Valparaı́so 2360102, Chile
- Facultad de Farmacia, Centro de Investigación Farmacopea Chilena, Universidad de Valparaı́so,, Av. Gran Bretaña 1093, Valparaı́so 2360102, Chile
| | - Javier Romero-Parra
- Departamento de Quı́mica Orgánica y Fisicoquı́mica, Facultad de Ciencias Quı́micas y Farmacéuticas, Universidad de Chile, Sergio Livingstone 1007, Casilla
233, 8380492 Santiago, Chile
| | - Gonzalo Recabarren-Gajardo
- Bioactive Heterocycles Synthesis Laboratory, BHSL, Departamento de Farmacia, Facultad de Quı́mica y de Farmacia, Pontificia Universidad Católica de Chile, Casilla 306, Avda. Vicuña Mackenna 4860, Macul, 7820436 Santiago, Chile
- Centro Interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile,, Marcoleta 391, 8330024 Santiago, Chile
| |
Collapse
|
24
|
Gradinati V, Baruffaldi F, Abbaraju S, Laudenbach M, Amin R, Gilger B, Velagaleti P, Pravetoni M. Polymer-mediated delivery of vaccines to treat opioid use disorders and to reduce opioid-induced toxicity. Vaccine 2020; 38:4704-4712. [PMID: 32439214 DOI: 10.1016/j.vaccine.2020.05.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/05/2020] [Accepted: 05/09/2020] [Indexed: 11/15/2022]
Abstract
Vaccines offer a potential strategy to treat opioid use disorders (OUD) and to reduce the incidence of opioid-related overdoses. Vaccines induce opioid-specific polyclonal antibodies that selectively and effectively bind the target opioid and prevent its distribution across the blood-brain barrier. Because antibody-mediated reduction of drug distribution to the brain reduces drug-induced behavior and toxicity, vaccine efficacy depends on the quantity and quality of the antibody response. This study tested whether polymer-mediated delivery could improve vaccine efficacy against opioids as well as eliminate the need for booster injections normally required for a successful immunization. A series of novel biodegradable biocompatible thermogelling pentablock co-polymers were used to formulate a candidate vaccine against oxycodone in mice and rats. Polymer-based delivery of the anti-oxycodone vaccine was equally or more effective than administration in aluminum adjuvant in generating oxycodone-specific antibodies and in reducing oxycodone-induced effects and oxycodone distribution to the brain in mice and rats. The composition and release kinetics of the polymer formulations determined vaccine efficacy. Specifically, a formulation consisting of three simultaneous injections of the anti-oxycodone vaccine formulated in three different polymers with slow, intermediate, and fast release kinetics was more effective than an immunization regimen consisting of three sequential injections with the vaccine adsorbed on aluminum. The novel three-phased polymer vaccine formulation was effective in blocking oxycodone-induced antinociception, respiratory depression and bradycardia in rats.
Collapse
Affiliation(s)
- Valeria Gradinati
- Hennepin Healthcare Research Institute, Minneapolis, MN, United States; University of Minnesota Medical School, Department of Pharmacology, Minneapolis, MN, United States
| | | | | | - Megan Laudenbach
- Hennepin Healthcare Research Institute, Minneapolis, MN, United States
| | - Rasidul Amin
- Symmetry Biosciences, Raleigh, NC, United States
| | - Brian Gilger
- North Carolina State University, NC, United States
| | | | - Marco Pravetoni
- Hennepin Healthcare Research Institute, Minneapolis, MN, United States; University of Minnesota Medical School, Department of Pharmacology, Minneapolis, MN, United States; University of Minnesota, Center for Immunology, Minneapolis, MN, United States.
| |
Collapse
|
25
|
Xiaoshan T, Junjie Y, Wenqing W, Yunong Z, Jiaping L, Shanshan L, Kutty Selva N, Kui C. Immunotherapy for treating methamphetamine, heroin and cocaine use disorders. Drug Discov Today 2020; 25:610-619. [DOI: 10.1016/j.drudis.2019.07.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/17/2019] [Accepted: 07/29/2019] [Indexed: 01/05/2023]
|
26
|
Alving CR, Peachman KK, Matyas GR, Rao M, Beck Z. Army Liposome Formulation (ALF) family of vaccine adjuvants. Expert Rev Vaccines 2020; 19:279-292. [PMID: 32228108 PMCID: PMC7412170 DOI: 10.1080/14760584.2020.1745636] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/17/2020] [Indexed: 12/19/2022]
Abstract
Introduction: From its earliest days, the US. military has embraced the use of vaccines to fight infectious diseases. The Army Liposome Formulation (ALF) has been a pivotal innovation as a vaccine adjuvant that provides excellent safety and potency and could lead to dual-use military and civilian benefits. For protection of personnel against difficult disease threats found in many areas of the world, Army vaccine scientists have created novel liposome-based vaccine adjuvants.Areas covered: ALF consists of liposomes containing saturated phospholipids, cholesterol, and monophosphoryl lipid A (MPLA) as an immunostimulant. ALF exhibited safety and strong potency in many vaccine clinical trials. Improvements based on ALF include: ALF adsorbed to aluminum hydroxide (ALFA); ALF containing QS21 saponin (ALFQ); and ALFQ adsorbed to aluminum hydroxide (ALFQA). Preclinical safety and efficacy studies with ALF, LFA, ALFQ, and ALFQA are discussed in preparation for upcoming vaccine trials targeting malaria, HIV-1, bacterial diarrhea, and opioid addiction.Expert opinion: The introduction of ALF in the 1980s stimulated commercial interest in vaccines to infectious diseases, and therapeutic vaccines to cancer, and Alzheimer's disease. It is likely that ALF, ALFA, and ALFQ, will provide momentum for new types of modern vaccines with improved efficacy and safety.
Collapse
Affiliation(s)
- Carl R. Alving
- Laboratory of Adjuvant & Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, USA
| | - Kristina K. Peachman
- Laboratory of Adjuvant & Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| | - Gary R. Matyas
- Laboratory of Adjuvant & Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, USA
| | - Mangala Rao
- Laboratory of Adjuvant & Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, USA
| | - Zoltan Beck
- Laboratory of Adjuvant & Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| |
Collapse
|
27
|
Verma A, Schmidt BA, Elizaldi SR, Nguyen NK, Walter KA, Beck Z, Trinh HV, Dinasarapu AR, Lakshmanappa YS, Rane NN, Matyas GR, Rao M, Shen X, Tomaras GD, LaBranche CC, Reimann KA, Foehl DH, Gach JS, Forthal DN, Kozlowski PA, Amara RR, Iyer SS. Impact of T h1 CD4 Follicular Helper T Cell Skewing on Antibody Responses to an HIV-1 Vaccine in Rhesus Macaques. J Virol 2020; 94:e01737-19. [PMID: 31827000 PMCID: PMC7158739 DOI: 10.1128/jvi.01737-19] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Generating durable humoral immunity through vaccination depends upon effective interactions of follicular helper T (Tfh) cells with germinal center (GC) B cells. Th1 polarization of Tfh cells is an important process shaping the success of Tfh-GC B cell interactions by influencing costimulatory and cytokine-dependent Tfh help to B cells. However, the question remains as to whether adjuvant-dependent modulation of Tfh cells enhances HIV-1 vaccine-induced antienvelope (anti-Env) antibody responses. We investigated whether an HIV-1 vaccine platform designed to increase the number of Th1-polarized Tfh cells enhances the magnitude and quality of anti-Env antibodies. Utilizing a novel interferon-induced protein 10 (IP-10)-adjuvanted HIV-1 DNA prime followed by a monophosphoryl lipid A and QS-21 (MPLA+QS-21)-adjuvanted Env protein boost (DIP-10 PALFQ) in macaques, we observed higher anti-Env serum IgG titers with greater cross-clade reactivity, specificity for V1V2, and effector functions than in macaques primed with DNA lacking IP-10 and boosted with MPLA-plus-alum-adjuvanted Env protein (DPALFA) The DIP-10 PALFQ vaccine regimen elicited higher anti-Env IgG1 and lower IgG4 antibody levels in serum, showing for the first time that adjuvants can dramatically impact the IgG subclass profile in macaques. The DIP-10 PALFQ regimen also increased vaginal and rectal IgA antibodies to a greater extent. Within lymph nodes, we observed augmented GC B cell responses and the promotion of Th1 gene expression profiles in GC Tfh cells. The frequency of GC Tfh cells correlated with both the magnitude and avidity of anti-Env serum IgG. Together, these data suggest that adjuvant-induced stimulation of Th1-Tfh cells is an effective strategy for enhancing the magnitude and quality of anti-Env antibody responses.IMPORTANCE The results of the RV144 trial demonstrated that vaccination could prevent HIV transmission in humans and that longevity of anti-Env antibodies may be key to this protection. Efforts to improve upon the prime-boost vaccine regimen used in RV144 have indicated that booster immunizations can increase serum anti-Env antibody titers but only transiently. Poor antibody durability hampers efforts to develop an effective HIV-1 vaccine. This study was designed to identify the specific elements involved in the immunological mechanism necessary to produce robust HIV-1-specific antibodies in rhesus macaques. By clearly defining immune-mediated pathways that improve the magnitude and functionality of the anti-HIV-1 antibody response, we will have the foundation necessary for the rational development of an HIV-1 vaccine.
Collapse
Affiliation(s)
- Anil Verma
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Brian A Schmidt
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Sonny R Elizaldi
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
- Graduate Group in Immunology, UC Davis, Davis, California, USA
| | - Nancy K Nguyen
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Korey A Walter
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Zoltan Beck
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Hung V Trinh
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Ashok R Dinasarapu
- Emory Department of Human Genetics, Emory University, Atlanta, Georgia, USA
| | | | - Niharika N Rane
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
| | - Gary R Matyas
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Mangala Rao
- U.S. Military HIV Research Program, Laboratory of Adjuvant and Antigen Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Celia C LaBranche
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Keith A Reimann
- Nonhuman Primate Reagent Resource, MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - David H Foehl
- Nonhuman Primate Reagent Resource, MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Johannes S Gach
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, School of Medicine, UC Irvine, Irvine, California, USA
| | - Donald N Forthal
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, School of Medicine, UC Irvine, Irvine, California, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, School of Medicine, UC Irvine, Irvine, California, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Rama R Amara
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Smita S Iyer
- The Center for Immunology and Infectious Diseases, UC Davis, Davis, California, USA
- California National Primate Research Center, School of Veterinary Medicine, UC Davis, Davis, California, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, Davis, California, USA
| |
Collapse
|
28
|
Navashenaq JG, Zamani P, Nikpoor AR, Tavakkol-Afshari J, Jaafari MR. Doxil chemotherapy plus liposomal P5 immunotherapy decreased myeloid-derived suppressor cells in murine model of breast cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 24:102150. [PMID: 31931230 DOI: 10.1016/j.nano.2020.102150] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) play a pivotal role in cancer. To overcome the problem of the MDSCs in the tumor microenvironment in this study, a combination of immunotherapy and chemotherapy was used. For this purpose, a liposomal formulation of P5 peptide and PEGylated liposomal doxorubicin (Doxil®) was utilized to treat mice bearing HER2+ tumor model. The results revealed that Doxil® administration before immunotherapy had not only reduced the population and functions of the MDSCs in the spleen (P < 0.001) and the tumor microenvironment (P < 0.05) but had also supported further immunotherapy including enhanced CD4+ (P < 0.01) and CD8+ lymphocyte (P < 0.001) population and IFN-γ production (P < 0.001). This effect was also more pronounced with a liposomal P5 and Doxil® compared with free peptide and doxorubicin. In conclusion, the results demonstrated that Doxil® plus liposomal P5 could have a decreasing effect on MDSCs and tumor growth, and it could be beneficial in breast cancer treatment.
Collapse
Affiliation(s)
- Jamshid Gholizadeh Navashenaq
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Reza Nikpoor
- Department of Immunology, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Mahmoud Reza Jaafari
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
29
|
Combadière B, Beaujean M, Chaudesaigues C, Vieillard V. Peptide-Based Vaccination for Antibody Responses Against HIV. Vaccines (Basel) 2019; 7:vaccines7030105. [PMID: 31480779 PMCID: PMC6789779 DOI: 10.3390/vaccines7030105] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/29/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
HIV-1 is responsible for a global pandemic of 35 million people and continues to spread at a rate of >2 million new infections/year. It is widely acknowledged that a protective vaccine would be the most effective means to reduce HIV-1 spread and ultimately eliminate the pandemic, whereas a therapeutic vaccine might help to mitigate the clinical course of the disease and to contribute to virus eradication strategies. However, despite more than 30 years of research, we do not have a vaccine capable of protecting against HIV-1 infection or impacting on disease progression. This, in part, denotes the challenge of identifying immunogens and vaccine modalities with a reduced risk of failure in late stage development. However, progress has been made in epitope identification for the induction of broadly neutralizing antibodies. Thus, peptide-based vaccination has become one of the challenges of this decade. While some researchers reconstitute envelope protein conformation and stabilization to conserve the epitope targeted by neutralizing antibodies, others have developed strategies based on peptide-carrier vaccines with a similar goal. Here, we will review the major peptide-carrier based approaches in the vaccine field and their application and recent development in the HIV-1 field.
Collapse
Affiliation(s)
- Behazine Combadière
- Sorbonne University, UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Center of Immunology and Infectious Diseases (CIMI-Paris), 91 Boulevard de l'Hôpital, F-75013 Paris, France.
| | - Manon Beaujean
- Sorbonne University, UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Center of Immunology and Infectious Diseases (CIMI-Paris), 91 Boulevard de l'Hôpital, F-75013 Paris, France
| | - Chloé Chaudesaigues
- Sorbonne University, UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Center of Immunology and Infectious Diseases (CIMI-Paris), 91 Boulevard de l'Hôpital, F-75013 Paris, France
| | - Vincent Vieillard
- Sorbonne University, UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Center of Immunology and Infectious Diseases (CIMI-Paris), 91 Boulevard de l'Hôpital, F-75013 Paris, France
| |
Collapse
|
30
|
Hwang CS, Smith LC, Wenthur CJ, Ellis B, Zhou B, Janda KD. Heroin vaccine: Using titer, affinity, and antinociception as metrics when examining sex and strain differences. Vaccine 2019; 37:4155-4163. [PMID: 31176539 DOI: 10.1016/j.vaccine.2019.05.061] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 01/03/2023]
Abstract
Anti-drug vaccines have potential as new interventions against substance use disorder (SUD). However, given the challenges seen with inter-individual variability in SUD vaccine trials to date, new interventions should ensure a robust immune response and safety profile among a diverse population. This requires accounting for sex and heritable genetic differences in response to both abused substances as well as the vaccination itself. To test response variability to our heroin-tetanus toxoid (Her-TT) immunoconjugate vaccine, we vaccinated male and female mice from several mouse strains including Swiss Webster (SW), BALB/c, and Jackson diversity mice (J:DO). Previous studies with vaccinated male SW mice demonstrated a rare hypersensitivity resulting in mice rapidly expiring with exposure to a low dose of heroin. Our results indicate that this response is limited to only male SW mice, and not to any other strain or female SW mice. Our data suggest that this hypersensitivity is not the result of an overactive cytokine or IgE response. Vaccination was similarly effective among the sexes for each strain and against repeated heroin challenge. Inbred BALB/c and J:DO mice were found to have the best vaccine response against heroin in antinociception behavioral assay. These results highlight the importance of incorporating both male and female subjects, along with different strains to mimic diverse human populations, as new SUD vaccines are being tested.
Collapse
Affiliation(s)
- Candy S Hwang
- Department of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Lauren C Smith
- Department of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Cody J Wenthur
- Department of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Beverly Ellis
- Department of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Bin Zhou
- Department of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Kim D Janda
- Department of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
31
|
Pravetoni M, Comer SD. Development of vaccines to treat opioid use disorders and reduce incidence of overdose. Neuropharmacology 2019; 158:107662. [PMID: 31173759 DOI: 10.1016/j.neuropharm.2019.06.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/01/2019] [Accepted: 06/02/2019] [Indexed: 02/06/2023]
Abstract
Vaccines offer a promising therapeutic strategy to treat substance use disorders (SUD). Vaccines have shown extensive preclinical proof of selectivity, safety, and efficacy against opioids, nicotine, cocaine, methamphetamine, and designer drugs. Despite clinical evaluation of vaccines targeting nicotine and cocaine showing proof of concept for this approach, no vaccine for SUD has yet reached the market. This review first discusses how vaccines for treatment of opioid use disorders (OUD) and reduction of opioid-induced fatal overdoses fit within the current medication assisted treatment (MAT) portfolio, and then summarizes ongoing efforts toward translation of vaccines targeting heroin, oxycodone, fentanyl, and other opioids. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.
Collapse
Affiliation(s)
- Marco Pravetoni
- University of Minnesota Medical School, Departments of Pharmacology and Medicine, Minneapolis, MN, USA; Hennepin Healthcare Research Institute, Minneapolis, MN, USA.
| | - Sandra D Comer
- Columbia University Irving Medical Center, Department of Psychiatry, The New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
32
|
MPL nano-liposomal vaccine containing P5 HER2/neu-derived peptide pulsed PADRE as an effective vaccine in a mice TUBO model of breast cancer. J Control Release 2019; 303:223-236. [DOI: 10.1016/j.jconrel.2019.04.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/11/2019] [Accepted: 04/13/2019] [Indexed: 11/21/2022]
|
33
|
Chemical and Immunological Characteristics of Aluminum-Based, Oil-Water Emulsion, and Bacterial-Origin Adjuvants. J Immunol Res 2019; 2019:3974127. [PMID: 31205956 PMCID: PMC6530223 DOI: 10.1155/2019/3974127] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/02/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022] Open
Abstract
Adjuvants are a diverse family of substances whose main objective is to increase the strength, quality, and duration of the immune response caused by vaccines. The most commonly used adjuvants are aluminum-based, oil-water emulsion, and bacterial-origin adjuvants. In this paper, we will discuss how the election of adjuvants is important for the adjuvant-mediated induction of immunity for different types of vaccines. Aluminum-based adjuvants are the most commonly used, the safest, and have the best efficacy, due to the triggering of a strong humoral response, albeit generating a weak induction of cell-mediated immune response. Freund's adjuvant is the most widely used oil-water emulsion adjuvant in animal trials; it stimulates inflammation and causes aggregation and precipitation of soluble protein antigens that facilitate the uptake by antigen-presenting cells (APCs). Adjuvants of bacterial origin, such as flagellin, E. coli membranes, and monophosphoryl lipid A (MLA), are known to potentiate immune responses, but their safety and risks are the main concern of their clinical use. This minireview summarizes the mechanisms that classic and novel adjuvants produce to stimulate immune responses.
Collapse
|
34
|
Natori Y, Hwang CS, Lin L, Smith LC, Zhou B, Janda KD. A chemically contiguous hapten approach for a heroin-fentanyl vaccine. Beilstein J Org Chem 2019; 15:1020-1031. [PMID: 31164940 DOI: 10.3762/bjoc.15.100] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/23/2019] [Indexed: 12/22/2022] Open
Abstract
Background: Increased death due to the opioid epidemic in the United States has necessitated the development of new strategies to treat addiction. Monoclonal antibodies and antidrug vaccines provide a tool that both aids addiction management and reduces the potential for overdose. Dual drug vaccines formulated by successive conjugation or by mixture have certain drawbacks. The current study examines an approach for combatting the dangers of fentanyl-laced heroin, by using a hapten with one epitope that has domains for both fentanyl and heroin. Results: We evaluated a series of nine vaccines developed from chemically contiguous haptens composed of both heroin- and fentanyl-like domains. Analysis of the results obtained by SPR and ELISA revealed trends in antibody affinity and titers for heroin and fentanyl based on epitope size and linker location. In antinociception studies, the best performing vaccines offered comparable protection against heroin as our benchmark heroin vaccine, but exhibited attenuated protection against fentanyl compared to our fentanyl vaccine. Conclusion: After thorough investigation of this strategy, we have identified key considerations for the development of a chemically contiguous heroin-fentanyl vaccine. Importantly, this is the first report of such a strategy in the opioid-drug-vaccine field.
Collapse
Affiliation(s)
- Yoshihiro Natori
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology; The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA, 92037, USA.,Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Komatsushima 4-4-1, Aoba-ku, Sendai, 981-8558, Japan
| | - Candy S Hwang
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology; The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA, 92037, USA.,Department of Chemistry, Southern Connecticut State University, 501 Crescent St, New Haven, CT, 06515, USA
| | - Lucy Lin
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology; The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Lauren C Smith
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology; The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Bin Zhou
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology; The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Kim D Janda
- Departments of Chemistry, Immunology and Microbial Science, Skaggs Institute for Chemical Biology; The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA, 92037, USA
| |
Collapse
|
35
|
Wallis J, Shenton DP, Carlisle RC. Novel approaches for the design, delivery and administration of vaccine technologies. Clin Exp Immunol 2019; 196:189-204. [PMID: 30963549 PMCID: PMC6468175 DOI: 10.1111/cei.13287] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2019] [Indexed: 12/20/2022] Open
Abstract
It is easy to argue that vaccine development represents humankind's most important and successful endeavour, such is the impact that vaccination has had on human morbidity and mortality over the last 200 years. During this time the original method of Jenner and Pasteur, i.e. that of injecting live-attenuated or inactivated pathogens, has been developed and supplemented with a wide range of alternative approaches which are now in clinical use or under development. These next-generation technologies have been designed to produce a vaccine that has the effectiveness of the original live-attenuated and inactivated vaccines, but without the associated risks and limitations. Indeed, the method of development has undoubtedly moved away from Pasteur's three Is paradigm (isolate, inactivate, inject) towards an approach of rational design, made possible by improved knowledge of the pathogen-host interaction and the mechanisms of the immune system. These novel vaccines have explored methods for targeted delivery of antigenic material, as well as for the control of release profiles, so that dosing regimens can be matched to the time-lines of immune system stimulation and the realities of health-care delivery in dispersed populations. The methods by which vaccines are administered are also the subject of intense research in the hope that needle and syringe dosing, with all its associated issues regarding risk of injury, cross-infection and patient compliance, can be replaced. This review provides a detailed overview of new vaccine vectors as well as information pertaining to the novel delivery platforms under development.
Collapse
Affiliation(s)
- J. Wallis
- Institute of Biomedical EngineeringUniversity of OxfordOxfordUK
| | - D. P. Shenton
- Defence Science and Technology LaboratoryPorton DownUK
| | - R. C. Carlisle
- Institute of Biomedical EngineeringUniversity of OxfordOxfordUK
| |
Collapse
|
36
|
Ramakrishnan A, Schumack NM, Gariepy CL, Eggleston H, Nunez G, Espinoza N, Nieto M, Castillo R, Rojas J, McCoy AJ, Beck Z, Matyas GR, Alving CR, Guerry P, Poly F, Laird RM. Enhanced Immunogenicity and Protective Efficacy of a Campylobacter jejuni Conjugate Vaccine Coadministered with Liposomes Containing Monophosphoryl Lipid A and QS-21. mSphere 2019; 4:e00101-19. [PMID: 31043512 PMCID: PMC6495334 DOI: 10.1128/msphere.00101-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/15/2019] [Indexed: 12/20/2022] Open
Abstract
Campylobacter jejuni is among the most common causes of diarrheal disease worldwide and efforts to develop protective measures against the pathogen are ongoing. One of the few defined virulence factors targeted for vaccine development is the capsule polysaccharide (CPS). We have developed a capsule conjugate vaccine against C. jejuni strain 81-176 (CPS-CRM) that is immunogenic in mice and nonhuman primates (NHPs) but only moderately immunogenic in humans when delivered alone or with aluminum hydroxide. To enhance immunogenicity, two novel liposome-based adjuvant systems, the Army Liposome Formulation (ALF), containing synthetic monophosphoryl lipid A, and ALF plus QS-21 (ALFQ), were evaluated with CPS-CRM in this study. In mice, ALF and ALFQ induced similar amounts of CPS-specific IgG that was significantly higher than levels induced by CPS-CRM alone. Qualitative differences in antibody responses were observed where CPS-CRM alone induced Th2-biased IgG1, whereas ALF and ALFQ enhanced Th1-mediated anti-CPS IgG2b and IgG2c and generated functional bactericidal antibody titers. CPS-CRM + ALFQ was superior to vaccine alone or CPS-CRM + ALF in augmenting antigen-specific Th1, Th2, and Th17 cytokine responses and a significantly higher proportion of CD4+ IFN-γ+ IL-2+ TNF-α+ and CD4+ IL-4+ IL-10+ T cells. ALFQ also significantly enhanced anti-CPS responses in NHPs when delivered with CPS-CRM compared to alum- or ALF-adjuvanted groups and showed the highest protective efficacy against diarrhea following orogastric challenge with C. jejuni This study provides evidence that the ALF adjuvants may provide enhanced immunogenicity of this and other novel C. jejuni capsule conjugate vaccines in humans.IMPORTANCECampylobacter jejuni is a leading cause of diarrheal disease worldwide, and currently no preventative interventions are available. C. jejuni is an invasive mucosal pathogen that has a variety of polysaccharide structures on its surface, including a capsule. In phase 1 studies, a C. jejuni capsule conjugate vaccine was safe but poorly immunogenic when delivered alone or with aluminum hydroxide. Here, we report enhanced immunogenicity of the conjugate vaccine delivered with liposome adjuvants containing monophosphoryl lipid A without or with QS-21, known as ALF and ALFQ, respectively, in preclinical studies. Both liposome adjuvants significantly enhanced immunity in mice and nonhuman primates and improved protective efficacy of the vaccine compared to alum in a nonhuman primate C. jejuni diarrhea model, providing promising evidence that these potent adjuvant formulations may enhance immunogenicity in upcoming human studies with this C. jejuni conjugate and other malaria and HIV vaccine platforms.
Collapse
Affiliation(s)
| | - Nina M Schumack
- Henry M. Jackson Foundation for Military Medicine, Bethesda, Maryland, USA
- Department of Enteric Diseases, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Christina L Gariepy
- Henry M. Jackson Foundation for Military Medicine, Bethesda, Maryland, USA
- Department of Enteric Diseases, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Heather Eggleston
- Henry M. Jackson Foundation for Military Medicine, Bethesda, Maryland, USA
- Department of Enteric Diseases, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Gladys Nunez
- Bacteriology Department, U.S. Naval Medical Research Unit No. 6, Callao, Peru
| | - Nereyda Espinoza
- Bacteriology Department, U.S. Naval Medical Research Unit No. 6, Callao, Peru
| | - Monica Nieto
- Bacteriology Department, U.S. Naval Medical Research Unit No. 6, Callao, Peru
| | - Rosa Castillo
- Bacteriology Department, U.S. Naval Medical Research Unit No. 6, Callao, Peru
| | - Jesus Rojas
- Bacteriology Department, U.S. Naval Medical Research Unit No. 6, Callao, Peru
| | - Andrea J McCoy
- Bacteriology Department, U.S. Naval Medical Research Unit No. 6, Callao, Peru
| | - Zoltan Beck
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for Military Medicine, Bethesda, Maryland, USA
| | - Gary R Matyas
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Carl R Alving
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Patricia Guerry
- Department of Enteric Diseases, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Frédéric Poly
- Department of Enteric Diseases, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Renee M Laird
- Henry M. Jackson Foundation for Military Medicine, Bethesda, Maryland, USA
- Department of Enteric Diseases, Naval Medical Research Center, Silver Spring, Maryland, USA
| |
Collapse
|
37
|
Farzad N, Barati N, Momtazi-Borojeni AA, Yazdani M, Arab A, Razazan A, Shariat S, Mansourian M, Abbasi A, Saberi Z, Badiee A, Jalali SA, Jaafari MR. P435 HER2/neu-derived peptide conjugated to liposomes containing DOPE as an effective prophylactic vaccine formulation for breast cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:665-673. [DOI: 10.1080/21691401.2019.1576702] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Niloofar Farzad
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Science, Mashhad, Iran
| | - Nastaran Barati
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Science, Mashhad, Iran
| | - Amir Abbas Momtazi-Borojeni
- Nanotechnology Research Center, Department of Medical Biotechnology, Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Yazdani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atefeh Arab
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Science, Mashhad, Iran
| | - Atefeh Razazan
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sheida Shariat
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mercedeh Mansourian
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Science, Mashhad, Iran
| | - Azam Abbasi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Saberi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Science, Mashhad, Iran
| | - Seyed Amir Jalali
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Science, Mashhad, Iran
| |
Collapse
|
38
|
Raleigh MD, Baruffaldi F, Peterson SJ, Le Naour M, Harmon TM, Vigliaturo JR, Pentel PR, Pravetoni M. A Fentanyl Vaccine Alters Fentanyl Distribution and Protects against Fentanyl-Induced Effects in Mice and Rats. J Pharmacol Exp Ther 2019; 368:282-291. [PMID: 30409833 PMCID: PMC6346379 DOI: 10.1124/jpet.118.253674] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/07/2018] [Indexed: 11/22/2022] Open
Abstract
Fentanyl is an extremely potent synthetic opioid that has been increasingly used to adulterate heroin, cocaine, and counterfeit prescription pills, leading to an increase in opioid-induced fatal overdoses in the United States, Canada, and Europe. A vaccine targeting fentanyl could offer protection against the toxic effects of fentanyl in both recreational drug users and others in professions at risk of accidental exposure. This study focuses on the development of a vaccine consisting of a fentanyl-based hapten (F) conjugated to keyhole limpet hemocyanin (KLH) carrier protein or to GMP-grade subunit KLH (sKLH). Immunization with F-KLH in mice and rats reduced fentanyl-induced hotplate antinociception, and in rats reduced fentanyl distribution to the brain compared with controls. F-KLH did not reduce the antinociceptive effects of equianalgesic doses of heroin or oxycodone in rats. To assess the vaccine effect on fentanyl toxicity, rats immunized with F-sKLH or unconjugated sKLH were exposed to increasing subcutaneous doses of fentanyl. Vaccination with F-sKLH shifted the dose-response curves to the right for both fentanyl-induced antinociception and respiratory depression. Naloxone reversed fentanyl effects in both groups, showing that its ability to reverse respiratory depression was preserved. These data demonstrate preclinical selectivity and efficacy of a fentanyl vaccine and suggest that vaccines may offer a therapeutic option in reducing fentanyl-induced side effects.
Collapse
Affiliation(s)
- Michael D Raleigh
- Hennepin Healthcare Research Institute (formerly Minneapolis Medical Research Foundation), Minneapolis, Minnesota (M.D.R., F.B., S.J.P., T.M.H., J.R.V., P.R.P., M.P.); Department of Medicinal Chemistry, University of Minnesota College of Pharmacy Minneapolis, Minnesota (M.L.N.); MLN BioPharma Consulting LLC (M.L.N.) Minneapolis, Minnesota; and Departments of Pharmacology (M.P.) and Medicine (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota Medical School, Minneapolis, Minnesota
| | - Federico Baruffaldi
- Hennepin Healthcare Research Institute (formerly Minneapolis Medical Research Foundation), Minneapolis, Minnesota (M.D.R., F.B., S.J.P., T.M.H., J.R.V., P.R.P., M.P.); Department of Medicinal Chemistry, University of Minnesota College of Pharmacy Minneapolis, Minnesota (M.L.N.); MLN BioPharma Consulting LLC (M.L.N.) Minneapolis, Minnesota; and Departments of Pharmacology (M.P.) and Medicine (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota Medical School, Minneapolis, Minnesota
| | - Samantha J Peterson
- Hennepin Healthcare Research Institute (formerly Minneapolis Medical Research Foundation), Minneapolis, Minnesota (M.D.R., F.B., S.J.P., T.M.H., J.R.V., P.R.P., M.P.); Department of Medicinal Chemistry, University of Minnesota College of Pharmacy Minneapolis, Minnesota (M.L.N.); MLN BioPharma Consulting LLC (M.L.N.) Minneapolis, Minnesota; and Departments of Pharmacology (M.P.) and Medicine (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota Medical School, Minneapolis, Minnesota
| | - Morgan Le Naour
- Hennepin Healthcare Research Institute (formerly Minneapolis Medical Research Foundation), Minneapolis, Minnesota (M.D.R., F.B., S.J.P., T.M.H., J.R.V., P.R.P., M.P.); Department of Medicinal Chemistry, University of Minnesota College of Pharmacy Minneapolis, Minnesota (M.L.N.); MLN BioPharma Consulting LLC (M.L.N.) Minneapolis, Minnesota; and Departments of Pharmacology (M.P.) and Medicine (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota Medical School, Minneapolis, Minnesota
| | - Theresa M Harmon
- Hennepin Healthcare Research Institute (formerly Minneapolis Medical Research Foundation), Minneapolis, Minnesota (M.D.R., F.B., S.J.P., T.M.H., J.R.V., P.R.P., M.P.); Department of Medicinal Chemistry, University of Minnesota College of Pharmacy Minneapolis, Minnesota (M.L.N.); MLN BioPharma Consulting LLC (M.L.N.) Minneapolis, Minnesota; and Departments of Pharmacology (M.P.) and Medicine (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota Medical School, Minneapolis, Minnesota
| | - Jennifer R Vigliaturo
- Hennepin Healthcare Research Institute (formerly Minneapolis Medical Research Foundation), Minneapolis, Minnesota (M.D.R., F.B., S.J.P., T.M.H., J.R.V., P.R.P., M.P.); Department of Medicinal Chemistry, University of Minnesota College of Pharmacy Minneapolis, Minnesota (M.L.N.); MLN BioPharma Consulting LLC (M.L.N.) Minneapolis, Minnesota; and Departments of Pharmacology (M.P.) and Medicine (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota Medical School, Minneapolis, Minnesota
| | - Paul R Pentel
- Hennepin Healthcare Research Institute (formerly Minneapolis Medical Research Foundation), Minneapolis, Minnesota (M.D.R., F.B., S.J.P., T.M.H., J.R.V., P.R.P., M.P.); Department of Medicinal Chemistry, University of Minnesota College of Pharmacy Minneapolis, Minnesota (M.L.N.); MLN BioPharma Consulting LLC (M.L.N.) Minneapolis, Minnesota; and Departments of Pharmacology (M.P.) and Medicine (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota Medical School, Minneapolis, Minnesota
| | - Marco Pravetoni
- Hennepin Healthcare Research Institute (formerly Minneapolis Medical Research Foundation), Minneapolis, Minnesota (M.D.R., F.B., S.J.P., T.M.H., J.R.V., P.R.P., M.P.); Department of Medicinal Chemistry, University of Minnesota College of Pharmacy Minneapolis, Minnesota (M.L.N.); MLN BioPharma Consulting LLC (M.L.N.) Minneapolis, Minnesota; and Departments of Pharmacology (M.P.) and Medicine (P.R.P., M.P.), and Center for Immunology (M.P.), University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
39
|
Arora R, Haile CN, Kosten TA, Wu Y, Ramakrishnan M, Hawkins LD, Orson FM, Kosten TR. Preclinical efficacy of an anti-methamphetamine vaccine using E6020 adjuvant. Am J Addict 2019; 28:119-126. [PMID: 30701618 DOI: 10.1111/ajad.12867] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/02/2019] [Accepted: 01/12/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Methamphetamine (MA) substance use disorder (SUD) does not have an efficacious pharmacotherapy. We developed a MA vaccine and investigated its potential to attenuate MA induced responses. METHODS We examined a novel adjuvant, E6020, a Toll-like receptor-4 (TLR-4) agonist combined with tetanus-toxoid conjugated to succinyl-methamphetamine (TT-SMA) adsorbed on aluminum hydroxide (alum). Adult BALB/c female mice received the vaccine and booster injections at weeks 0, 3, and 6. The efficacy of the vaccine was assessed by the level and affinity of anti-MA antibodies elicited, its ability to attenuate MA induced locomotor activation and its reduction in the amount of MA entering the brains of vaccinated mice. RESULTS The TT-SMA vaccine containing alum and E6020 adjuvant produced anti-MA antibodies with nanomolar affinities and showed threefold greater peak titer levels than without E6020 (700 vs 250 μg/ml). These antibodies significantly decreased MA-induced locomotor activation (p < .05), and reduced the brain (p < .005) MA levels following MA administration in actively immunized mice. CONCLUSIONS Thus, this anti-MA vaccine formulated with E6020 demonstrated effective functional protection against behavioral disruptions induced by MA. SCIENTIFIC SIGNIFICANCE Together, anti-MA vaccine showing a promising improvement in the efficacy of the vaccine that could be an effective candidate vaccine for methamphetamine use disorder (MUD). Furthermore, combinations of adjuvants may be a tool to design vaccines for MA dependence in humans. (Am J Addict 2019;XX:1-8).
Collapse
Affiliation(s)
- Reetakshi Arora
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas
| | - Colin N Haile
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas.,Department of Psychology, University of Houston, Houston, Texas
| | - Therese A Kosten
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas.,Department of Psychology, University of Houston, Houston, Texas
| | - Yan Wu
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas
| | - Muthu Ramakrishnan
- The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas.,Immunology Allergy & Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | | | - Frank M Orson
- The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas.,Immunology Allergy & Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Thomas R Kosten
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas
| |
Collapse
|
40
|
Li Z, Ding S, Li Y. Study on the immunity protection of 14-3-3–MPLA–liposome vaccine against cystic echinococcosis in mice. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.flm.2019.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
41
|
Myagkova MA, Morozova VS. Vaccines for substance abuse treatment: new approaches in the immunotherapy of addictions. Russ Chem Bull 2018. [DOI: 10.1007/s11172-018-2290-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
42
|
Baruffaldi F, Kelcher AH, Laudenbach M, Gradinati V, Limkar A, Roslawski M, Birnbaum A, Lees A, Hassler C, Runyon S, Pravetoni M. Preclinical Efficacy and Characterization of Candidate Vaccines for Treatment of Opioid Use Disorders Using Clinically Viable Carrier Proteins. Mol Pharm 2018; 15:4947-4962. [PMID: 30240216 DOI: 10.1021/acs.molpharmaceut.8b00592] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Vaccines may offer a new treatment strategy for opioid use disorders and opioid-related overdoses. To speed translation, this study evaluates opioid conjugate vaccines containing components suitable for pharmaceutical manufacturing and compares analytical assays for conjugate characterization. Three oxycodone-based haptens (OXY) containing either PEGylated or tetraglycine [(Gly)4] linkers were conjugated to a keyhole limpet hemocyanin (KLH) carrier protein via carbodiimide (EDAC) or maleimide chemistry. The EDAC-conjugated OXY(Gly)4-KLH was most effective in reducing oxycodone distribution to the brain in mice. Vaccine efficacy was T cell-dependent. The lead OXY hapten was conjugated to the KLH, tetanus toxoid, diphtheria cross-reactive material (CRM), as well as the E. coli-expressed CRM (EcoCRM) and nontoxic tetanus toxin heavy chain fragment C (rTTHc) carrier proteins. All vaccines induced early hapten-specific B cell expansion and showed equivalent efficacy against oxycodone in mice. However, some hapten-protein conjugates were easier to characterize for molecular weight and size. Finally, heroin vaccines formulated with either EcoCRM or KLH were equally effective in reducing heroin-induced antinociception and distribution to the brain of heroin and its metabolites in mice. This study identifies vaccine candidates and vaccine components for further development.
Collapse
Affiliation(s)
- Federico Baruffaldi
- Hennepin Healthcare Research Institute (HHRI, formerly Minneapolis Medical Research Foundation or MMRF) , 701 Park Avenue , Minneapolis , Minnesota 55415 , United States
| | - April Huseby Kelcher
- Hennepin Healthcare Research Institute (HHRI, formerly Minneapolis Medical Research Foundation or MMRF) , 701 Park Avenue , Minneapolis , Minnesota 55415 , United States
| | - Megan Laudenbach
- Hennepin Healthcare Research Institute (HHRI, formerly Minneapolis Medical Research Foundation or MMRF) , 701 Park Avenue , Minneapolis , Minnesota 55415 , United States
| | - Valeria Gradinati
- Hennepin Healthcare Research Institute (HHRI, formerly Minneapolis Medical Research Foundation or MMRF) , 701 Park Avenue , Minneapolis , Minnesota 55415 , United States.,Dipartimento di Chimica e Tecnologie Farmaceutiche, Socrates Program , Universitá degli Studi di Milano , Milan 20122 , Italy
| | - Ajinkya Limkar
- University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | | | - Angela Birnbaum
- University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - Andrew Lees
- Fina Biosolutions, LLC , Rockville , Maryland 20850 , United States
| | - Carla Hassler
- RTI International , Research Triangle Park , North Carolina 27709-2194 , United States
| | - Scott Runyon
- RTI International , Research Triangle Park , North Carolina 27709-2194 , United States
| | - Marco Pravetoni
- Hennepin Healthcare Research Institute (HHRI, formerly Minneapolis Medical Research Foundation or MMRF) , 701 Park Avenue , Minneapolis , Minnesota 55415 , United States.,Departments of Medicine and Pharmacology, Center for Immunology , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| |
Collapse
|
43
|
Moulahoum H, Zihnioglu F, Timur S, Coskunol H. Novel technologies in detection, treatment and prevention of substance use disorders. J Food Drug Anal 2018; 27:22-31. [PMID: 30648574 PMCID: PMC9298618 DOI: 10.1016/j.jfda.2018.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 08/18/2018] [Accepted: 09/10/2018] [Indexed: 11/03/2022] Open
Abstract
Substance use disorders are a widely recognized problem, which affects various levels of communities and influenced the world socioeconomically. Its source is deeply embedded in the global population. In order to fight against such an adversary, governments have spared no efforts in implementing substance abuse treatment centers and funding research to develop treatments and prevention procedures. In this review, we will discuss the use of immunological-based treatments and detection kit technologies. We will be detailing the steps followed to produce performant antibodies (antigens, carriers, and adjuvants) focusing on cocaine and methamphetamine as examples. Furthermore, part of this review is dedicated to substance use detection. Owing to novel technologies such as bio-functional polymeric surfaces and biosensors manufacturing, detection has become a more convenient method with the fast and on-site developed devices. Commercially available devices are able to test substance use disorders in urine, saliva, hair, and sweat. This improvement has had a tremendous impact on the prevention of driving under influence and other illicit behaviors. Lastly, substance abuse became a major issue involving the cooperation of experts on all levels to devise better treatment programs and prevent abuse-based accidents, injury and death.
Collapse
Affiliation(s)
- Hichem Moulahoum
- Biochemistry Department, Faculty of Science, Ege University, Bornova, Izmir 35100, Turkey.
| | - Figen Zihnioglu
- Biochemistry Department, Faculty of Science, Ege University, Bornova, Izmir 35100, Turkey
| | - Suna Timur
- Biochemistry Department, Faculty of Science, Ege University, Bornova, Izmir 35100, Turkey; Central Research Testing and Analysis Laboratory Research and Application Center, Ege University, Bornova, Izmir 35100, Turkey
| | - Hakan Coskunol
- Addiction Treatment Center, Faculty of Medicine, Ege University, Bornova, Izmir 35100, Turkey.
| |
Collapse
|
44
|
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: 1.1] [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
|
45
|
Ignacio BJ, Albin TJ, Esser-Kahn AP, Verdoes M. Toll-like Receptor Agonist Conjugation: A Chemical Perspective. Bioconjug Chem 2018; 29:587-603. [PMID: 29378134 PMCID: PMC10642707 DOI: 10.1021/acs.bioconjchem.7b00808] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) are vital elements of the mammalian immune system that function by recognizing pathogen-associated molecular patterns (PAMPs), bridging innate and adaptive immunity. They have become a prominent therapeutic target for the treatment of infectious diseases, cancer, and allergies, with many TLR agonists currently in clinical trials or approved as immunostimulants. Numerous studies have shown that conjugation of TLR agonists to other molecules can beneficially influence their potency, toxicity, pharmacokinetics, or function. The functional properties of TLR agonist conjugates, however, are highly dependent on the ligation strategy employed. Here, we review the chemical structural requirements for effective functional TLR agonist conjugation. In addition, we provide similar analysis for those that have yet to be conjugated. Moreover, we discuss applications of covalent TLR agonist conjugation and their implications for clinical use.
Collapse
Affiliation(s)
- Bob J. Ignacio
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Tyler J. Albin
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Aaron P. Esser-Kahn
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
- Institute for Molecular Engineering, University of Chicago, Chicago, Illinois 60637, United States
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
46
|
Nisini R, Poerio N, Mariotti S, De Santis F, Fraziano M. The Multirole of Liposomes in Therapy and Prevention of Infectious Diseases. Front Immunol 2018; 9:155. [PMID: 29459867 PMCID: PMC5807682 DOI: 10.3389/fimmu.2018.00155] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/17/2018] [Indexed: 12/17/2022] Open
Abstract
Liposomes are closed bilayer structures spontaneously formed by hydrated phospholipids that are widely used as efficient delivery systems for drugs or antigens, due to their capability to encapsulate bioactive hydrophilic, amphipathic, and lipophilic molecules into inner water phase or within lipid leaflets. The efficacy of liposomes as drug or antigen carriers has been improved in the last years to ameliorate pharmacokinetics and capacity to release their cargo in selected target organs or cells. Moreover, different formulations and variations in liposome composition have been often proposed to include immunostimulatory molecules, ligands for specific receptors, or stimuli responsive compounds. Intriguingly, independent research has unveiled the capacity of several phospholipids to play critical roles as intracellular messengers in modulating both innate and adaptive immune responses through various mechanisms, including (i) activation of different antimicrobial enzymatic pathways, (ii) driving the fusion–fission events between endosomes with direct consequences to phagosome maturation and/or to antigen presentation pathway, and (iii) modulation of the inflammatory response. These features can be exploited by including selected bioactive phospholipids in the bilayer scaffold of liposomes. This would represent an important step forward since drug or antigen carrying liposomes could be engineered to simultaneously activate different signal transduction pathways and target specific cells or tissues to induce antigen-specific T and/or B cell response. This lipid-based host-directed strategy can provide a focused antimicrobial innate and adaptive immune response against specific pathogens and offer a novel prophylactic or therapeutic option against chronic, recurrent, or drug-resistant infections.
Collapse
Affiliation(s)
- Roberto Nisini
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | - Noemi Poerio
- Dipartimento di Biologia, Università degli Studi di Roma "Tor Vergata", Rome, Italy
| | - Sabrina Mariotti
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | - Federica De Santis
- Dipartimento di Biologia, Università degli Studi di Roma "Tor Vergata", Rome, Italy
| | - Maurizio Fraziano
- Dipartimento di Biologia, Università degli Studi di Roma "Tor Vergata", Rome, Italy
| |
Collapse
|
47
|
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: 6.5] [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
|
48
|
Razazan A, Behravan J, Arab A, Barati N, Arabi L, Gholizadeh Z, Hatamipour M, Reza Nikpoor A, Momtazi-Borojeni AA, Mosaffa F, Ghahremani MH, Jaafari MR. Conjugated nanoliposome with the HER2/neu-derived peptide GP2 as an effective vaccine against breast cancer in mice xenograft model. PLoS One 2017; 12:e0185099. [PMID: 29045460 PMCID: PMC5646774 DOI: 10.1371/journal.pone.0185099] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 09/06/2017] [Indexed: 01/07/2023] Open
Abstract
One of the challenging issues in vaccine development is peptide and adjuvant delivery into target cells. In this study, we developed a vaccine and therapeutic delivery system to increase cytotoxic T lymphocyte (CTL) response against a breast cancer model overexpressing HER2/neu. Gp2, a HER2/neu-derived peptide, was conjugated to Maleimide-mPEG2000-DSPE micelles and post inserted into liposomes composed of DMPC, DMPG phospholipids, and fusogenic lipid dioleoylphosphatidylethanolamine (DOPE) containing monophosphoryl lipid A (MPL) adjuvant (DMPC-DMPG-DOPE-MPL-Gp2). BALB/c mice were immunized with different formulations and the immune response was evaluated in vitro and in vivo. ELISpot and intracellular cytokine analysis by flow cytometry showed that the mice vaccinated with Lip-DOPE-MPL-GP2 incited the highest number of IFN-γ+ in CD8+ cells and CTL response. The immunization led to lower tumor sizes and longer survival time compared to the other groups of mice immunized and treated with the Lip-DOPE-MPL-GP2 formulation in both prophylactic and therapeutic experiments. These results showed that co-formulation of DOPE and MPL conjugated with GP2 peptide not only induces high antitumor immunity but also enhances therapeutic efficacy in TUBO mice model. Lip-DOPE-MPL-GP2 formulation could be a promising vaccine and a therapeutic delivery system against HER2 positive cancers and merits further investigation.
Collapse
Affiliation(s)
- Atefeh Razazan
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran Iran
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Behravan
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atefeh Arab
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nastaran Barati
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Gholizadeh
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdi Hatamipour
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Reza Nikpoor
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Abbas Momtazi-Borojeni
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohamad Hosein Ghahremani
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
49
|
Pouyanfard S, Müller M. Human papillomavirus first and second generation vaccines-current status and future directions. Biol Chem 2017; 398:871-889. [PMID: 28328521 DOI: 10.1515/hsz-2017-0105] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/16/2017] [Indexed: 02/06/2023]
Abstract
It has been more than 10 years that the first prophylactic papillomavirus vaccine became available, although distribution has been mainly limited to the more affluent countries. The first two vaccines have been a great success, hundreds of millions of women and a much smaller number of men have been vaccinated ever since. In a few countries with high vaccination coverage, in particular Australia but also parts of Great Britain and others, clinical impact of vaccination programs is already visible and there are indications for herd immunity as well. Vaccine efficacy is higher than originally estimated and the vaccines have an excellent safety profile. Gardasil9 is a second generation HPV virus-like particle vaccine that was licensed in 2015 and there are more to come in the near future. Currently, burning questions in respect to HPV vaccination are the duration of protection - especially in regard to cross-protection - reduction of the three-dose regimen and its impact on cross-protection; and duration of response, as well as protection against oropharyngeal HPV infections. Furthermore, researchers are seeking to overcome limitations of the VLP vaccines, namely low thermal stability, cost, invasive administration, limited coverage of non-vaccine HPV types, and lack of therapeutic efficacy. In this review we summarize the current status of licensed VLP vaccines and address questions related to second and third generation HPV vaccines.
Collapse
|
50
|
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
|