51
|
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: 152] [Impact Index Per Article: 21.7] [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
|
52
|
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
|
53
|
Liang R, Xie J, Li J, Wang K, Liu L, Gao Y, Hussain M, Shen G, Zhu J, Tao J. Liposomes-coated gold nanocages with antigens and adjuvants targeted delivery to dendritic cells for enhancing antitumor immune response. Biomaterials 2017; 149:41-50. [DOI: 10.1016/j.biomaterials.2017.09.029] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 09/11/2017] [Accepted: 09/24/2017] [Indexed: 01/07/2023]
|
54
|
Co-delivery of nucleoside-modified mRNA and TLR agonists for cancer immunotherapy: Restoring the immunogenicity of immunosilent mRNA. J Control Release 2017; 266:287-300. [DOI: 10.1016/j.jconrel.2017.09.041] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/21/2017] [Accepted: 09/29/2017] [Indexed: 12/23/2022]
|
55
|
Liu Z, Guo J. NKT-cell glycolipid agonist as adjuvant in synthetic vaccine. Carbohydr Res 2017; 452:78-90. [DOI: 10.1016/j.carres.2017.10.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 10/14/2017] [Accepted: 10/14/2017] [Indexed: 01/07/2023]
|
56
|
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
|
57
|
Xie J, Yang C, Liu Q, Li J, Liang R, Shen C, Zhang Y, Wang K, Liu L, Shezad K, Sullivan M, Xu Y, Shen G, Tao J, Zhu J, Zhang Z. Encapsulation of Hydrophilic and Hydrophobic Peptides into Hollow Mesoporous Silica Nanoparticles for Enhancement of Antitumor Immune Response. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1701741. [PMID: 28861951 DOI: 10.1002/smll.201701741] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/18/2017] [Indexed: 06/07/2023]
Abstract
Codelivery of combinational antigenic peptides and adjuvant to antigen presenting cells is expected to amplify tumor specific T lymphocytes immune responses while minimizing the possibility of tumor escaping and reducing immune tolerance to single antigenic peptide. However, the varied hydrophobicities of these multivariant derived short antigenic peptides limit their codelivery efficiency in conventional delivery systems. Here, a facile yet effective route is presented to generate monodisperse and stable hollow mesoporous silica nanoparticles (HMSNs) for codelivering of HGP10025-33 and TRP2180-188 , two melanoma-derived peptides with varied hydrophobicities. The HMSNs with large pore size can improve the encapsulation efficiency of both HGP100 and TRP2 after NH2 modification on the inner hollow core and COOH modification in the porous channels. HGP100 and TRP2 loaded HMSNs (HT@HMSNs) are further enveloped within monophosphoryl lipid A adjuvant entrapped lipid bilayer (HTM@HMLBs), for improved stability/biocompatibility and codelivery efficiency of multiple peptides, adjuvant, and enhanced antitumor immune responses. HTM@HMLBs increase uptake by dendritic cells (DCs) and stimulate DCs maturation efficiently, which further induce the activation of both tumor specific CD8+ and CD4+ T lymphocytes. Moreover, HTM@HMLBs can significantly inhibit tumor growth and lung metastasis in murine melanoma models with good safety profiles. HMSNs enveloped with lipid bilayers (HMLBs) are believed to be a promising platform for codelivery of multiple peptides, adjuvant, and enhancement of antitumor efficacy of conventional vaccinations.
Collapse
Affiliation(s)
- Jun Xie
- Tongji School of Pharmacy and National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology (HUST), Wuhan, 430030, China
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Chaohua Yang
- Tongji School of Pharmacy and National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology (HUST), Wuhan, 430030, China
| | - Qianqian Liu
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Jun Li
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Ruijing Liang
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Chen Shen
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Yi Zhang
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Ke Wang
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Liping Liu
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Khurram Shezad
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Martin Sullivan
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Yong Xu
- Department of Immunology, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Guanxin Shen
- Department of Immunology, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Juan Tao
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Jintao Zhu
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Zhiping Zhang
- Tongji School of Pharmacy and National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology (HUST), Wuhan, 430030, China
| |
Collapse
|
58
|
El Bissati K, Zhou Y, Paulillo SM, Raman SK, Karch CP, Roberts CW, Lanar DE, Reed S, Fox C, Carter D, Alexander J, Sette A, Sidney J, Lorenzi H, Begeman IJ, Burkhard P, McLeod R. Protein nanovaccine confers robust immunity against Toxoplasma. NPJ Vaccines 2017; 2:24. [PMID: 29263879 PMCID: PMC5627305 DOI: 10.1038/s41541-017-0024-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 11/08/2022] Open
Abstract
We designed and produced a self-assembling protein nanoparticle. This self-assembling protein nanoparticle contains five CD8+ HLA-A03-11 supertypes-restricted epitopes from antigens expressed during Toxoplasma gondii's lifecycle, the universal CD4+ T cell epitope PADRE, and flagellin as a scaffold and TLR5 agonist. These CD8+ T cell epitopes were separated by N/KAAA spacers and optimized for proteasomal cleavage. Self-assembling protein nanoparticle adjuvanted with TLR4 ligand-emulsion GLA-SE were evaluated for their efficacy in inducing IFN-γ responses and protection of HLA-A*1101 transgenic mice against T. gondii. Immunization, using self-assembling protein nanoparticle-GLA-SE, activated CD8+ T cells to produce IFN-γ. Self-assembling protein nanoparticle-GLA-SE also protected HLA-A*1101 transgenic mice against subsequent challenge with Type II parasites. Hence, combining CD8+ T cell-eliciting peptides and PADRE into a multi-epitope protein that forms a nanoparticle, administered with GLA-SE, leads to efficient presentation by major histocompatibility complex Class I and II molecules. Furthermore, these results suggest that activation of TLR4 and TLR5 could be useful for development of vaccines that elicit T cells to prevent toxoplasmosis in humans.
Collapse
Affiliation(s)
- Kamal El Bissati
- Departments of OVS, The University of Chicago, 5841S Maryland Ave, Chicago, IL 60637 USA
| | - Ying Zhou
- Departments of OVS, The University of Chicago, 5841S Maryland Ave, Chicago, IL 60637 USA
| | | | | | - Christopher P. Karch
- Institute of Materials Science and Department of Molecular and Cell Biology, University of Connecticut, 97 North Eagleville Road, Storrs, CT 06269 USA
| | - Craig W. Roberts
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE UK
| | - David E. Lanar
- Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD 20910 USA
| | - Steve Reed
- Infectious Diseases Research Institute, 1616 Eastlake Ave E #400, Seattle, WA 98102 USA
| | - Chris Fox
- Infectious Diseases Research Institute, 1616 Eastlake Ave E #400, Seattle, WA 98102 USA
| | - Darrick Carter
- Infectious Diseases Research Institute, 1616 Eastlake Ave E #400, Seattle, WA 98102 USA
| | - Jeff Alexander
- PaxVax, 3985-A Sorrento Valley Blvd, San Diego, CA 92121 USA
| | - Alessandro Sette
- La Jolla Institute of Allergy and Immunology, 9420 Athena Cir, La Jolla, CA 92037 USA
| | - John Sidney
- La Jolla Institute of Allergy and Immunology, 9420 Athena Cir, La Jolla, CA 92037 USA
| | - Hernan Lorenzi
- J. Craig Venter Institute, 9714 Medical Center Drive, Rockville, MD 20850 USA
| | - Ian J. Begeman
- Departments of OVS, The University of Chicago, 5841S Maryland Ave, Chicago, IL 60637 USA
| | - Peter Burkhard
- Alpha-O Peptides AG, Lörracherstrasse 50, 4125 Riehen, Switzerland
- Institute of Materials Science and Department of Molecular and Cell Biology, University of Connecticut, 97 North Eagleville Road, Storrs, CT 06269 USA
| | - Rima McLeod
- Departments of OVS, The University of Chicago, 5841S Maryland Ave, Chicago, IL 60637 USA
- Pediatrics (Infectious Diseases), The University of Chicago, 5841S Maryland Ave, Chicago, IL 60637 USA
| |
Collapse
|
59
|
Torres OB, Matyas GR, Rao M, Peachman KK, Jalah R, Beck Z, Michael NL, Rice KC, Jacobson AE, Alving CR. Heroin-HIV-1 (H2) vaccine: induction of dual immunologic effects with a heroin hapten-conjugate and an HIV-1 envelope V2 peptide with liposomal lipid A as an adjuvant. NPJ Vaccines 2017; 2:13. [PMID: 29263870 PMCID: PMC5604742 DOI: 10.1038/s41541-017-0013-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 03/15/2017] [Accepted: 03/21/2017] [Indexed: 12/20/2022] Open
Abstract
A synthetic heroin analog (MorHap) and a synthetic 42 amino acid V2 loop peptide from A/E strain of HIV-1 gp120 envelope protein that was previously used in a successful phase III vaccine trial were constructed as antigens together with liposomes containing monophosphoryl lipid A as an adjuvant, to explore the feasibility of producing a dual use vaccine both for treatment of heroin addiction and prevention of HIV-1 infection among injection drug users. The V2 peptide was tethered by a palmitoyl fatty acyl tail embedded in the liposomal lipid bilayer, and the heroin analog was conjugated to tetanus toxoid as a carrier protein that was mixed with the adjuvant. Upon comparison of a linear V2 peptide with a cyclic peptide, differences were found in the secondary configurations by circular dichroism, with the tethered cyclic peptide (palm-cyclic peptide) entirely in a random coil, and the tethered linear V2 peptide (palm-linear V2 peptide) entirely in a beta-sheet. Upon immunization of mice, palm-cyclic peptide induced anti-cyclic peptide endpoint titers >106 and was considered to be a better immunogen overall than palm-linear V2 peptide for inducing antibodies to gp120 and gp70-V1V2. The antibodies also inhibited the binding of V2 peptide to the HIV-1 α4β7 integrin receptor. Antibody titers to MorHap, even with the presence of injected cyclic peptide, were very high, and resulted in inhibition of the hyper-locomotion and antinociception effects of injected heroin. From these initial experiments, we conclude that with a potent adjuvant and mostly synthetic constituents, a vaccine directed to heroin and HIV-1 (H2 vaccine) could be a feasible objective. A vaccine designed to treat heroin addiction while at the same time preventing HIV infection elicited strong immune responses in mice. Scientists from the US government led by Carl Alving from the Walter Reed Army Institute of Research in Bethesda, Maryland, created a dual vaccine formulated with three main components: a segment of a protein expressed on the surface of HIV; synthetic molecules that resemble heroin and its degradation products; and a potent adjuvant to stimulate the immune system. Mice immunized with this vaccine had high antibody titers against the HIV surface protein as well as heroin and its derivatives. These mice also showed dulled responses to injected heroin. The findings suggest this vaccine strategy could help fight heroin abuse and the high risk of HIV infection among intravenous drug users.
Collapse
Affiliation(s)
- Oscar B Torres
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, 20817 MD USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Gary R Matyas
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Mangala Rao
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Kristina K Peachman
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, 20817 MD USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Rashmi Jalah
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, 20817 MD USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Zoltan Beck
- US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, 20817 MD USA.,U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Nelson L Michael
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| | - Kenner C Rice
- Department of Health and Human Services, Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, 9800 Medical Drive, Bethesda, 20892 MD USA.,National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 9800 Medical Drive, Bethesda, 20892 MD USA
| | - Arthur E Jacobson
- Department of Health and Human Services, Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, 9800 Medical Drive, Bethesda, 20892 MD USA.,National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 9800 Medical Drive, Bethesda, 20892 MD USA
| | - Carl R Alving
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, 20910 MD USA
| |
Collapse
|
60
|
Yuba E, Yamaguchi A, Yoshizaki Y, Harada A, Kono K. Bioactive polysaccharide-based pH-sensitive polymers for cytoplasmic delivery of antigen and activation of antigen-specific immunity. Biomaterials 2017; 120:32-45. [DOI: 10.1016/j.biomaterials.2016.12.021] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 12/03/2016] [Accepted: 12/16/2016] [Indexed: 11/17/2022]
|
61
|
Song P, He S, Zhou A, Lv G, Guo J, Zhou J, Han Y, Zhou H, Hao Z, Cong H. Vaccination with toxofilin DNA in combination with an alum-monophosphoryl lipid A mixed adjuvant induces significant protective immunity against Toxoplasma gondii. BMC Infect Dis 2017; 17:19. [PMID: 28056837 PMCID: PMC5217305 DOI: 10.1186/s12879-016-2147-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 12/21/2016] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND A widely prevalent disease, toxoplasmosis poses serious health threats to both humans and animals; therefore, development of an ideal DNA vaccine against Toxoplasma gondii is needed eagerly. The purpose of the present study is to assess the protective efficacy of a DNA vaccine encoding the T. gondii toxofilin gene (pEGFP-toxofilin). In addition, toxofilin DNA vaccine combined with the individual adjuvants, alum or monophosphoryl lipid A (MPLA), or a mixture of alum-MPLA adjuvant were screened for their ability to enhance antibody responses. METHODS Using bioinformatics, we analyzed the gene and amino acid sequences of the toxofilin protein, recognizing and identifying several potential linear B and T helper (Th)-1 cell epitopes. BALB/c mice were immunized three times with either toxofilin DNA vaccine alone or in combination with the adjuvants such as alum, MPLA or an alum-MPLA mixture. The systemic immune response was evaluated by cytokine, the percentage of CD4 (+) and CD8 (+) T cells and specific antibody measurement. Two weeks after the last immunization, protective efficacy was evaluated by challenging intraperitoneally with 1 × 104 tachyzoites of T. gondii or intragastrically with 20 cysts of T. gondii PRU strain. RESULTS All experimentally immunized mice developed strong humoral and cellular immune responses compared with the control groups. Moreover, by comparison with the non-adjuvant toxofilin DNA vaccine group, adding alum adjuvant to toxofilin DNA vaccine resulted in an increase in humoral response and a skewed Th2 response. However, the MPLA adjuvant with toxofilin DNA vaccine induced significantly enhanced humoral and Th1-biased immune responses. Importantly, the co-administration of alum-MPLA adjuvant in combination with the toxofilin DNA vaccine shifted the Th2 immune response to a Th1 response compared with the alum-toxofilin group, and elicited the strongest humoral and Th1 responses among all the groups. At the same time, a longer survival time and less cyst amounts against T. gondii infection were also observed in the alum-MPLA-toxofilin group in comparison with single or no adjuvant groups. CONCLUSIONS Toxoplasma gondii toxofilin is a promising vaccine candidate that warrants further development. Co-administration of the alum-MPLA adjuvant mixture with DNA vaccine could effectively enhance immunogenicity and strongly skew the cellular immune response towards a Th1 phenotype.
Collapse
Affiliation(s)
- Pengxia Song
- Department of Parasitology, Shandong University School of Medicine, Jinan, Shandong Province, 250012, People's Republic of China
| | - Shenyi He
- Department of Parasitology, Shandong University School of Medicine, Jinan, Shandong Province, 250012, People's Republic of China.
| | - Aihua Zhou
- Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, Shandong University School of Medicine, 250021, Jinan, Shandong Province, People's Republic of China
| | - Gang Lv
- Department of Parasitology, Shandong University School of Medicine, Jinan, Shandong Province, 250012, People's Republic of China
| | - Jingjing Guo
- Department of Parasitology, Shandong University School of Medicine, Jinan, Shandong Province, 250012, People's Republic of China
| | - Jian Zhou
- Shandong University School of Medicine, 250021, Jinan, Shandong Province, People's Republic of China
| | - Yali Han
- Department of Parasitology, Shandong University School of Medicine, Jinan, Shandong Province, 250012, People's Republic of China
| | - Huaiyu Zhou
- Department of Parasitology, Shandong University School of Medicine, Jinan, Shandong Province, 250012, People's Republic of China
| | - Zhen Hao
- Department of Parasitology, Shandong University School of Medicine, Jinan, Shandong Province, 250012, People's Republic of China
| | - Hua Cong
- Department of Parasitology, Shandong University School of Medicine, Jinan, Shandong Province, 250012, People's Republic of China
| |
Collapse
|
62
|
Preparation of Multifunctional Liposomes as a Stable Vaccine Delivery-Adjuvant System by Procedure of Emulsification-Lyophilization. Methods Mol Biol 2016; 1404:635-649. [PMID: 27076327 DOI: 10.1007/978-1-4939-3389-1_41] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Liposomes have been proven to be useful carriers for vaccine antigens and can be modified as a versatile vaccine adjuvant-delivery system (VADS). To fulfill efficiently both functions of adjuvant and delivery, the liposomes are often modified with different functional molecules, such as lipoidal immunopotentiators, APC (antigen-presenting cell) targeting ligands, steric stabilization polymers, and charged lipids. Also, to overcome the weakness of instability, vaccines are often lyophilized as a dry product. In this chapter the procedure of emulsification-lyophilization (PEL) is introduced as an efficient method for preparing a stable anhydrous precursor to the multifunctional liposomes which bear dual modifications with APC targeting molecule of the mannosylated cholesterol and the adjuvant material of monophosphoryl lipid A. The techniques and procedures for synthesis of APC targeting molecule, i.e., the mannosylated cholesterol, and for characterization of the multifunctional liposomes are also described.
Collapse
|
63
|
Wang N, Zhen Y, Jin Y, Wang X, Li N, Jiang S, Wang T. Combining different types of multifunctional liposomes loaded with ammonium bicarbonate to fabricate microneedle arrays as a vaginal mucosal vaccine adjuvant-dual delivery system (VADDS). J Control Release 2016; 246:12-29. [PMID: 27986552 DOI: 10.1016/j.jconrel.2016.12.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 12/01/2016] [Accepted: 12/09/2016] [Indexed: 01/09/2023]
Abstract
To develop effective mucosal vaccines, two types of multifunctional liposomes, the mannosylated lipid A-liposomes (MLLs) with a size of 200nm and the stealth lipid A-liposomes (SLLs) of 50nm, both loaded with a model antigen and NH4HCO3, were fabricated together into microneedles, forming the proSLL/MLL-constituted microneedle array (proSMMA), which upon rehydration dissolved rapidly recovering the initial MLLs and SLLs. Mice vaccinated with proSMMAs by vaginal mucosa patching other than conventional intradermal administration established robust antigen-specific humoral and cellular immunity at both systemic and mucosal levels, especially, in the reproductive and intestinal ducts. Further exploration demonstrated that the MLLs reconstituted from the administered proSMMAs were mostly taken up by vaginal mucosal dendritic cells, whereas the recovered SLLs trafficked directly to draining lymph nodes wherein to be picked up by macrophages. Moreover, the antigens delivered by either liposomes were also cross-presented for MHC-I displaying by APCs thanks to lysosome escape and ROS (reactive oxygen species) stimulation, both of which occurred when lysosomal acidifying the liposome-released NH4HCO3 into CO2 and NH4+/NH3 to rupture lysosomes by gas expansion and to cause ROS production by excessive ammonia induction, resulting in a mixed Th1/Th2 type response which was also promoted by liposomal lipid A via activation of TLR4. In addition, vaginal vaccination of the engineered HSV2 antigen gD-loaded proSMMAs successfully protected mice from the virus challenge. Thus, the proSMMAs are in fact a vaccine adjuvant-dual delivery system capable of eliciting robust humoral and cellular immunity against the invading pathogens, especially, the sexually transmitted ones.
Collapse
Affiliation(s)
- Ning Wang
- School of Biological and Medical Engineering, Hefei University of Technology, 193 Tun Brook Road, Hefei, Anhui Province 230009, China
| | - Yuanyuan Zhen
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Xueting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ning Li
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Shaohong Jiang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China.
| |
Collapse
|
64
|
Abstract
Urinary tract infections (UTI) are among the most common bacterial infections in humans, affecting millions of people every year. UTI cause significant morbidity in women throughout their lifespan, in infant boys, in older men, in individuals with underlying urinary tract abnormalities, and in those that require long-term urethral catheterization, such as patients with spinal cord injuries or incapacitated individuals living in nursing homes. Serious sequelae include frequent recurrences, pyelonephritis with sepsis, renal damage in young children, pre-term birth, and complications of frequent antimicrobial use including high-level antibiotic resistance and Clostridium difficile colitis. Uropathogenic E. coli (UPEC) cause the vast majority of UTI, but less common pathogens such as Enterococcus faecalis and other enterococci frequently take advantage of an abnormal or catheterized urinary tract to cause opportunistic infections. While antibiotic therapy has historically been very successful in controlling UTI, the high rate of recurrence remains a major problem, and many individuals suffer from chronically recurring UTI, requiring long-term prophylactic antibiotic regimens to prevent recurrent UTI. Furthermore, the global emergence of multi-drug resistant UPEC in the past ten years spotlights the need for alternative therapeutic and preventative strategies to combat UTI, including anti-infective drug therapies and vaccines. In this chapter, we review recent advances in the field of UTI pathogenesis, with an emphasis on the identification of promising drug and vaccine targets. We then discuss the development of new UTI drugs and vaccines, highlighting the challenges these approaches face and the need for a greater understanding of urinary tract mucosal immunity.
Collapse
|
65
|
Őrfi E, Szebeni J. The immune system of the gut and potential adverse effects of oral nanocarriers on its function. Adv Drug Deliv Rev 2016; 106:402-409. [PMID: 27693367 DOI: 10.1016/j.addr.2016.09.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is substantial effort in modern pharmacotherapy to use nanoparticle-based drug delivery systems (nDDS) for improving the oral absorption of drugs. An often neglected circumstance regarding this approach is that the gut is a major part of the immune system that may be vulnerable for immune-cell toxicity, or mediate humoral immune response against various components of nDDS, recognized as foreign. This review recapitulates the structure and function of gut-associated lymphoid tissue (GALT), i.e., the enteral section of mucosa-associated lymphoid tissue (MALT) and reminds how virus-like nDDS may potentially induce immunogenicity just as attenuated or killed viruses do in oral vaccines. Furthermore, we present examples for immune toxicities of emulsifiers and polymer-containing micelles, manifested in complement activation-related pseudoallergy (CARPA). A major message of the review is that early testing of immunogenicity or other adverse immune effects of nDDS in appropriate test systems or models may be prudent to recognize the risk of rare immune problems that may surface in late-stage clinical trials or after marketing of nDDS.
Collapse
Affiliation(s)
- Erik Őrfi
- Nanomedicine Research and Education Center, Department of Pathophysiology, Semmelweis University, and SeroScience Ltd, Budapest, Hungary
| | - János Szebeni
- Nanomedicine Research and Education Center, Department of Pathophysiology, Semmelweis University, and SeroScience Ltd, Budapest, Hungary; Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health, Miskolc University, Miskolc, Hungary; SeroScience Ltd., Budapest, Hungary.
| |
Collapse
|
66
|
|
67
|
Liu Y, Chen C. Role of nanotechnology in HIV/AIDS vaccine development. Adv Drug Deliv Rev 2016; 103:76-89. [PMID: 26952542 DOI: 10.1016/j.addr.2016.02.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 12/25/2022]
Abstract
HIV/AIDS is one of the worst crises affecting global health and influencing economic development and social stability. Preventing and treating HIV infection is a crucial task. However, there is still no effective HIV vaccine for clinical application. Nanotechnology has the potential to solve the problems associated with traditional HIV vaccines. At present, various nano-architectures and nanomaterials can function as potential HIV vaccine carriers or adjuvants, including inorganic nanomaterials, liposomes, micelles and polymer nanomaterials. In this review, we summarize the current progress in the use of nanotechnology for the development of an HIV/AIDS vaccine and discuss its potential to greatly improve the solubility, permeability, stability and pharmacokinetics of HIV vaccines. Although nanotechnology holds great promise for applications in HIV/AIDS vaccines, there are still many inadequacies that result in a variety of risks and challenges. The potential hazards to the human body and environment associated with some nano-carriers, and their underlying mechanisms require in-depth study. Non-toxic or low-toxic nanomaterials with adjuvant activity have been identified. However, studying the confluence of factors that affect the adjuvant activity of nanomaterials may be more important for the optimization of the dosage and immunization strategy and investigations into the exact mechanism of action. Moreover, there are no uniform standards for investigations of nanomaterials as potential vaccine adjuvants. These limitations make it harder to analyze and deduce rules from the existing data. Developing vaccine nano-carriers or adjuvants with high benefit-cost ratios is important to ensure their broad usage. Despite some shortcomings, nanomaterials have great potential and application prospects in the fields of AIDS treatment and prevention.
Collapse
Affiliation(s)
- Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| |
Collapse
|
68
|
Wang X, Wang N, Li N, Zhen Y, Wang T. Multifunctional particle-constituted microneedle arrays as cutaneous or mucosal vaccine adjuvant-delivery systems. Hum Vaccin Immunother 2016; 12:2075-2089. [PMID: 27159879 DOI: 10.1080/21645515.2016.1158368] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
To overcome drawbacks of current injection vaccines, such as causing needle phobia, needing health professionals for inoculation, and generating dangerous sharps wastes, researchers have designed novel vaccines that are combined with various microneedle arrays (MAs), in particular, with the multifunctional particle-constructed MAs (MPMAs). MPMAs prove able to enhance vaccine stability through incorporating vaccine ingredients in the carrier, and can be painlessly inoculated by minimally trained workers or by self-administration, leaving behind no metal needle pollution while eliciting robust systemic and mucosal immunity to antigens, thanks to delivering vaccines to cutaneous or mucosal compartments enriched in professional antigen-presenting cells (APCs). Especially, MPMAs can be easily integrated with functional molecules fulfilling targeting vaccine delivery or controlling immune response toward a Th1 or Th2 pathway to generate desired immunity against pathogens. Herein, we introduce the latest research and development of various MPMAs which are a novel but promising vaccine adjuvant delivery system (VADS).
Collapse
Affiliation(s)
- Xueting Wang
- a School of Pharmacy, Anhui Medical University , Hefei , China
| | - Ning Wang
- b School of Medical Engineering, Hefei University of Technology , Hefei , China
| | - Ning Li
- a School of Pharmacy, Anhui Medical University , Hefei , China
| | - Yuanyuan Zhen
- a School of Pharmacy, Anhui Medical University , Hefei , China
| | - Ting Wang
- a School of Pharmacy, Anhui Medical University , Hefei , China
| |
Collapse
|
69
|
Design of pH-sensitive polymer-modified liposomes for antigen delivery and their application in cancer immunotherapy. Polym J 2016. [DOI: 10.1038/pj.2016.31] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
70
|
Abstract
INTRODUCTION Liposomes are well-known as drug carriers, and are now critical components of two of six types of adjuvants present in licensed vaccines. The liposomal vaccine adjuvant field has long been dynamic and innovative, and research in this area is further examined as new commercial products appear in parallel with new vaccines. In an arena where successful products exist the potential for new types of vaccines with liposomal adjuvants, and alternative liposomal adjuvants that could emerge for new types of vaccines, are discussed. AREAS COVERED Major areas include: virosomes, constructed from phospholipids and proteins from influenza virus particles; liposomes containing natural and synthetic neutral or anionic phospholipids, cholesterol, natural or synthetic monophosphoryl lipid A, and QS21 saponin; non-phospholipid cationic liposomes; and combinations and mixtures of liposomes and immunostimulating ingredients as adjuvants for experimental vaccines. EXPERT OPINION Liposomes containing monophosphoryl lipid A and QS21 have considerable momentum that will result soon in emergence of prophylactic vaccines to malaria and shingles, and possible novel cancer vaccines. The licensed virosome vaccines to influenza and hepatitis A will be replaced with virosome vaccines to other infectious diseases. Alternative liposomal formulations are likely to emerge for difficult diseases such as tuberculosis or HIV-1 infection.
Collapse
Affiliation(s)
- Carl R Alving
- a Laboratory of Adjuvant and Antigen Research , US Military HIV Research Program, Walter Reed Army Institute of Research , Silver Spring , MD , USA
| | - Zoltan Beck
- a Laboratory of Adjuvant and Antigen Research , US Military HIV Research Program, Walter Reed Army Institute of Research , Silver Spring , MD , USA.,b US Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine , Bethesda , MD , USA
| | - Gary R Matyas
- a Laboratory of Adjuvant and Antigen Research , US Military HIV Research Program, Walter Reed Army Institute of Research , Silver Spring , MD , USA
| | - Mangala Rao
- a Laboratory of Adjuvant and Antigen Research , US Military HIV Research Program, Walter Reed Army Institute of Research , Silver Spring , MD , USA
| |
Collapse
|
71
|
Akkina R, Allam A, Balazs AB, Blankson JN, Burnett JC, Casares S, Garcia JV, Hasenkrug KJ, Kashanchi F, Kitchen SG, Klein F, Kumar P, Luster AD, Poluektova LY, Rao M, Sanders-Beer BE, Shultz LD, Zack JA. Improvements and Limitations of Humanized Mouse Models for HIV Research: NIH/NIAID "Meet the Experts" 2015 Workshop Summary. AIDS Res Hum Retroviruses 2016; 32:109-19. [PMID: 26670361 DOI: 10.1089/aid.2015.0258] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The number of humanized mouse models for the human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome (AIDS) and other infectious diseases has expanded rapidly over the past 8 years. Highly immunodeficient mouse strains, such as NOD/SCID/gamma chain(null) (NSG, NOG), support better human hematopoietic cell engraftment. Another improvement is the derivation of highly immunodeficient mice, transgenic with human leukocyte antigens (HLAs) and cytokines that supported development of HLA-restricted human T cells and heightened human myeloid cell engraftment. Humanized mice are also used to study the HIV reservoir using new imaging techniques. Despite these advances, there are still limitations in HIV immune responses and deficits in lymphoid structures in these models in addition to xenogeneic graft-versus-host responses. To understand and disseminate the improvements and limitations of humanized mouse models to the scientific community, the NIH sponsored and convened a meeting on April 15, 2015 to discuss the state of knowledge concerning these questions and best practices for selecting a humanized mouse model for a particular scientific investigation. This report summarizes the findings of the NIH meeting.
Collapse
Affiliation(s)
- Ramesh Akkina
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado
| | - Atef Allam
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Silver Spring, Maryland
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | | - Joel N. Blankson
- Department of Medicine, Center for AIDS Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John C. Burnett
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, California
| | - Sofia Casares
- U.S. Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, Maryland
| | - J. Victor Garcia
- Division of Infectious Diseases, Department of Medicine, UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kim J. Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana
| | - Fatah Kashanchi
- School of Systems Biology, National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia
| | - Scott G. Kitchen
- Departments of Medicine and Microbiology, Immunology and Molecular Genetics, UCLA AIDS Institute, Los Angeles, California
| | - Florian Klein
- First Department of Internal Medicine, University Hospital of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Priti Kumar
- School of Medicine, Infectious Diseases/Internal Medicine, Yale University, New Haven, Connecticut
| | - Andrew D. Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Larisa Y. Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Mangala Rao
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Brigitte E. Sanders-Beer
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | | | - Jerome A. Zack
- Departments of Medicine and Microbiology, Immunology and Molecular Genetics, UCLA AIDS Institute, Los Angeles, California
| |
Collapse
|
72
|
Stassijns J, Bollaerts K, Baay M, Verstraeten T. A systematic review and meta-analysis on the safety of newly adjuvanted vaccines among children. Vaccine 2015; 34:714-22. [PMID: 26740250 DOI: 10.1016/j.vaccine.2015.12.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/08/2015] [Accepted: 12/10/2015] [Indexed: 12/15/2022]
Abstract
INTRODUCTION New adjuvants such as the AS- or the MF59-adjuvants improve vaccine efficacy and facilitate dose-sparing. Their use in influenza and malaria vaccines has resulted in a large body of evidence on their clinical safety in children. METHODS We carried out a systematic search for safety data from published clinical trials on newly adjuvanted vaccines in children ≤10 years of age. Serious adverse events (SAEs), solicited AEs, unsolicited AEs and AEs of special interest were evaluated for four new adjuvants: the immuno-stimulants containing adjuvant systems AS01 and AS02, and the squalene containing oil-in-water emulsions AS03 and MF59. Relative risks (RR) were calculated, comparing children receiving newly adjuvanted vaccines to children receiving other vaccines with a variety of antigens, both adjuvanted and unadjuvanted. RESULTS Twenty-nine trials were included in the meta-analysis, encompassing 25,056 children who received at least one dose of the newly adjuvanted vaccines. SAEs did not occur more frequently in adjuvanted groups (RR 0.85, 95%CI 0.75-0.96). Our meta-analyses showed higher reactogenicity following administration of newly adjuvanted vaccines, however, no consistent pattern of solicited AEs was observed across adjuvant systems. Pain was the most prevalent AE, but often mild and of short duration. No increased risks were found for unsolicited AEs, febrile convulsions, potential immune mediated diseases and new onset of chronic diseases. CONCLUSIONS Our meta-analysis did not show any safety concerns in clinical trials of the newly adjuvanted vaccines in children ≤10 years of age. An unexplained increase of meningitis in one Phase III AS01-adjuvanted malaria trial and the link between narcolepsy and the AS03-adjuvanted pandemic vaccine illustrate that continued safety monitoring is warranted.
Collapse
Affiliation(s)
- Jorgen Stassijns
- P-95, Epidemiology and Pharmacovigilance Consulting and Services, Koning Leopold III Laan 1, 3001 Heverlee, Belgium
| | - Kaatje Bollaerts
- P-95, Epidemiology and Pharmacovigilance Consulting and Services, Koning Leopold III Laan 1, 3001 Heverlee, Belgium
| | - Marc Baay
- P-95, Epidemiology and Pharmacovigilance Consulting and Services, Koning Leopold III Laan 1, 3001 Heverlee, Belgium
| | - Thomas Verstraeten
- P-95, Epidemiology and Pharmacovigilance Consulting and Services, Koning Leopold III Laan 1, 3001 Heverlee, Belgium.
| |
Collapse
|
73
|
Differential immune responses to HIV-1 envelope protein induced by liposomal adjuvant formulations containing monophosphoryl lipid A with or without QS21. Vaccine 2015; 33:5578-5587. [PMID: 26372857 DOI: 10.1016/j.vaccine.2015.09.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 08/28/2015] [Accepted: 09/04/2015] [Indexed: 11/22/2022]
Abstract
Liposomes have shown promise as constituents of adjuvant formulations in vaccines to parasitic and viral diseases. A particular type of liposomal construct, referred to as Army Liposome Formulation (ALF), containing neutral and anionic saturated phospholipids, cholesterol, and monophosphoryl lipid A (MPLA), has been used as an adjuvant for many years. Here we investigated the effects of physical and chemical changes of ALF liposomes on adjuvanted immune responses to CN54 gp140, a recombinant HIV-1 envelope protein. While holding the total amounts of liposomal MPLA and the gp140 antigen constant, different liposome sizes and liposomal MPLA:phospholipid molar ratios, and the effect of adding QS21 to the liposomes were compared for inducing immune responses to the gp140. For liposomes lacking QS21, higher titers of IgG binding antibodies to gp140 were induced by small unilamellar vesicle (SUV) rather than by large multilamellar vesicle (MLV) liposomes, and the highest titers were obtained with SUV having the MPLA:phospholipid ratio of 1:5.6. ALF plus QS21 (ALFQ) liposomes induced the same maximal binding antibody titers regardless of the MPLA:phospholipid ratio. ALF MLV liposomes induced mainly IgG1 and very low IgG2a antibodies, while ALF SUV liposomes induced IgG1≥IgG2a>IgG2b antibodies. Liposomes containing QS21 induced IgG1>IgG2a>IgG2b>IgG3 antibodies. ELISPOT analysis of splenocytes from immunized mice revealed that ALF liposomes induced low levels of IFN-γ, but ALFQ induced high levels. ALF and ALFQ liposomes each induced approximately equivalent high levels of IL-4. Based on antibody subtypes and cytokine secretion, we conclude that ALF liposomes predominantly stimulate Th2, while ALFQ strongly induces both Th1 and Th2 immunity. When CN54 gp140 was adjuvanted with either ALF or ALFQ liposomes, antibodies were induced that neutralized two HIV-1 tier 1 clade C strain pseudoviruses.
Collapse
|
74
|
Fan Y, Sahdev P, Ochyl LJ, Akerberg J, Moon JJ. Cationic liposome-hyaluronic acid hybrid nanoparticles for intranasal vaccination with subunit antigens. J Control Release 2015; 208:121-129. [PMID: 25869965 PMCID: PMC4430437 DOI: 10.1016/j.jconrel.2015.04.010] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 04/05/2015] [Accepted: 04/09/2015] [Indexed: 11/20/2022]
Abstract
Here we report the development of a new cationic liposome-hyaluronic acid (HA) hybrid nanoparticle (NP) system and present our characterization of these NPs as an intranasal vaccine platform using a model antigen and F1-V, a candidate recombinant antigen for Yersinia pestis, the causative agent of plague. Incubation of cationic liposomes composed of DOTAP and DOPE with anionic HA biopolymer led to efficient ionic complexation and formation of homogenous liposome-polymer hybrid NPs, as evidenced by fluorescence resonance energy transfer, dynamic light scattering, and nanoparticle tracking analyses. Incorporation of cationic liposomes with thiolated HA allowed for facile surface decoration of NPs with thiol-PEG, resulting in the formation of DOTAP/HA core-PEG shell nanostructures. These NPs, termed DOTAP-HA NPs, exhibited improved colloidal stability and prolonged antigen release. In addition, cytotoxicity associated with DOTAP liposomes (LC50~0.2mg/ml) was significantly reduced by at least 20-fold with DOTAP-HA NPs (LC50>4mg/ml), as measured with bone marrow derived dendritic cells (BMDCs). Furthermore, NPs co-loaded with ovalbumin (OVA) and a molecular adjuvant, monophosphoryl lipid A (MPLA) promoted BMDC maturation and upregulation of co-stimulatory markers, including CD40, CD86, and MHC-II, and C57BL/6 mice vaccinated with NPs via intranasal route generated robust OVA-specific CD8(+) T cell and antibody responses. Importantly, intranasal vaccination with NPs co-loaded with F1-V and MPLA induced potent humoral immune responses with 11-, 23-, and 15-fold increases in F1-V-specific total IgG, IgG1, and IgG2c titers in immune sera by day 77, respectively, and induced balanced Th1/Th2 humoral immune responses, whereas mice immunized with the equivalent doses of soluble F1-V vaccine failed to achieve sero-conversion. Overall, these results suggest that liposome-polymer hybrid NPs may serve as a promising vaccine delivery platform for intranasal vaccination against Y. pestis and other infectious pathogens.
Collapse
Affiliation(s)
- Yuchen Fan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Preety Sahdev
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lukasz J Ochyl
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jonathan Akerberg
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
75
|
Multifunctional liposomes constituting microneedles induced robust systemic and mucosal immunoresponses against the loaded antigens via oral mucosal vaccination. Vaccine 2015; 33:4330-40. [PMID: 25858854 DOI: 10.1016/j.vaccine.2015.03.081] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 03/12/2015] [Accepted: 03/24/2015] [Indexed: 11/20/2022]
Abstract
To develop effective, convenient and stable mucosal vaccines, mannose-PEG-cholesterol (MPC)/lipid A-liposomes (MLLs) entrapping model antigen bovine serum albumin (BSA) were prepared by the procedure of emulsification-lyophilization and used to constitute microneedles, forming the proMLL-filled microneedle arrays (proMMAs). The proMMAs were rather stable and hard enough to pierce porcine skin and, upon rehydration, dissolved rapidly recovering the MLLs without size and entrapment change. The proMMAs given to mice via oral mucosal (o.m.) route, rather than routine intradermal administration, elicited robust systemic and mucosal immunoresponses against the loaded antigens as evidenced by high levels of BSA-specific IgG in the sera and IgA in the salivary, intestinal and vaginal secretions of mice. Enhanced levels of IgG2a and IFN-γ in treated mice revealed that proMMAs induced a mixed Th1/Th2 immunoresponse. Moreover, a significant increase in CD8(+) T cells confirmed that strong cellular immunity had also been established by the immunization of the proMMAs. Thus, the proMMAs can be immunized via o.m. route to set up an effective multiple defense against pathogen invasion and may be an effective vaccine adjuvant-delivery system (VADS) applicable in the controlled temperature chain.
Collapse
|
76
|
Apellániz B, Rujas E, Serrano S, Morante K, Tsumoto K, Caaveiro JMM, Jiménez MÁ, Nieva JL. The Atomic Structure of the HIV-1 gp41 Transmembrane Domain and Its Connection to the Immunogenic Membrane-proximal External Region. J Biol Chem 2015; 290:12999-3015. [PMID: 25787074 DOI: 10.1074/jbc.m115.644351] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Indexed: 11/06/2022] Open
Abstract
The membrane-proximal external region (MPER) C-terminal segment and the transmembrane domain (TMD) of gp41 are involved in HIV-1 envelope glycoprotein-mediated fusion and modulation of immune responses during viral infection. However, the atomic structure of this functional region remains unsolved. Here, based on the high resolution NMR data obtained for peptides spanning the C-terminal segment of MPER and the TMD, we report two main findings: (i) the conformational variability of the TMD helix at a membrane-buried position; and (ii) the existence of an uninterrupted α-helix spanning MPER and the N-terminal region of the TMD. Thus, our structural data provide evidence for the bipartite organization of TMD predicted by previous molecular dynamics simulations and functional studies, but they do not support the breaking of the helix at Lys-683, as was suggested by some models to mark the initiation of the TMD anchor. Antibody binding energetics examined with isothermal titration calorimetry and humoral responses elicited in rabbits by peptide-based vaccines further support the relevance of a continuous MPER-TMD helix for immune recognition. We conclude that the transmembrane anchor of HIV-1 envelope is composed of two distinct subdomains: 1) an immunogenic helix at the N terminus also involved in promoting membrane fusion; and 2) an immunosuppressive helix at the C terminus, which might also contribute to the late stages of the fusion process. The unprecedented high resolution structural data reported here may guide future vaccine and inhibitor developments.
Collapse
Affiliation(s)
- Beatriz Apellániz
- From the Biophysics Unit (Consejo Superior de Investigaciones Científicas, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P. O. Box 644, 48080 Bilbao, Spain
| | - Edurne Rujas
- From the Biophysics Unit (Consejo Superior de Investigaciones Científicas, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P. O. Box 644, 48080 Bilbao, Spain, the Department of Bioengineering, Graduate School of Engineering, University of Tokyo, Bunkyo-ku, 113-8656 Tokyo, Japan, and
| | - Soraya Serrano
- the Institute of Physical Chemistry "Rocasolano" (Consejo Superior de Investigaciones Científicas), Serrano 119, E-28006 Madrid, Spain
| | - Koldo Morante
- the Department of Bioengineering, Graduate School of Engineering, University of Tokyo, Bunkyo-ku, 113-8656 Tokyo, Japan, and
| | - Kouhei Tsumoto
- the Department of Bioengineering, Graduate School of Engineering, University of Tokyo, Bunkyo-ku, 113-8656 Tokyo, Japan, and
| | - Jose M M Caaveiro
- the Department of Bioengineering, Graduate School of Engineering, University of Tokyo, Bunkyo-ku, 113-8656 Tokyo, Japan, and
| | - M Ángeles Jiménez
- the Institute of Physical Chemistry "Rocasolano" (Consejo Superior de Investigaciones Científicas), Serrano 119, E-28006 Madrid, Spain
| | - José L Nieva
- From the Biophysics Unit (Consejo Superior de Investigaciones Científicas, UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P. O. Box 644, 48080 Bilbao, Spain,
| |
Collapse
|
77
|
Beck Z, Matyas GR, Alving CR. Detection of liposomal cholesterol and monophosphoryl lipid A by QS-21 saponin and Limulus polyphemus amebocyte lysate. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:775-80. [DOI: 10.1016/j.bbamem.2014.12.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/26/2014] [Accepted: 12/05/2014] [Indexed: 11/16/2022]
|
78
|
Hanson MC, Abraham W, Crespo MP, Chen SH, Liu H, Szeto GL, Kim M, Reinherz EL, Irvine DJ. Liposomal vaccines incorporating molecular adjuvants and intrastructural T-cell help promote the immunogenicity of HIV membrane-proximal external region peptides. Vaccine 2015; 33:861-8. [PMID: 25559188 DOI: 10.1016/j.vaccine.2014.12.045] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/01/2014] [Accepted: 12/18/2014] [Indexed: 12/11/2022]
Abstract
An HIV vaccine capable of inducing high and durable levels of broadly neutralizing antibodies has thus far proven elusive. A promising antigen is the membrane-proximal external region (MPER) from gp41, a segment of the viral envelope recognized by a number of broadly neutralizing antibodies. Though an attractive vaccine target due to the linear nature of the epitope and its highly conserved sequence, MPER peptides are poorly immunogenic and may require display on membranes to achieve a physiological conformation matching the native virus. Here we systematically explored how the structure and composition of liposomes displaying MPER peptides impacts the strength and durability of humoral responses to this antigen as well as helper T-cell responses in mice. Administration of MPER peptides anchored to the surface of liposomes induced MPER-specific antibodies whereas MPER administered in oil-based emulsion adjuvants or alum did not, even when combined with Toll-like receptor agonists. High-titer IgG responses to liposomal MPER required the inclusion of molecular adjuvants such as monophosphoryl lipid A. Anti-MPER humoral responses were further enhanced by incorporating high-Tm lipids in the vesicle bilayer and optimizing the MPER density to a mean distance of ∼10-15 nm between peptides on the liposomes' surfaces. Encapsulation of helper epitopes within the vesicles allowed efficient "intrastructural" T-cell help, which promoted IgG responses to MPER while minimizing competing B-cell responses against the helper sequence. These results define several key properties of liposome formulations that promote durable, high-titer antibody responses against MPER peptides, which will be a prerequisite for a successful MPER-targeting vaccine.
Collapse
Affiliation(s)
- Melissa C Hanson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wuhbet Abraham
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Monica P Crespo
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stephanie H Chen
- Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Haipeng Liu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Greg Lee Szeto
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; The Ragon Institute of MGH, MIT, and Harvard, 400 Technology Square, Cambridge, MA 02139, USA
| | - Mikyung Kim
- Laboratory of Immunobiology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Ellis L Reinherz
- Laboratory of Immunobiology and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Darrell J Irvine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; The Ragon Institute of MGH, MIT, and Harvard, 400 Technology Square, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
79
|
Apellániz B, Nieva JL. The Use of Liposomes to Shape Epitope Structure and Modulate Immunogenic Responses of Peptide Vaccines Against HIV MPER. PEPTIDE AND PROTEIN VACCINES 2015; 99:15-54. [DOI: 10.1016/bs.apcsb.2015.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
80
|
Schwendener RA. Liposomes as vaccine delivery systems: a review of the recent advances. THERAPEUTIC ADVANCES IN VACCINES 2014; 2:159-82. [PMID: 25364509 DOI: 10.1177/2051013614541440] [Citation(s) in RCA: 329] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Liposomes and liposome-derived nanovesicles such as archaeosomes and virosomes have become important carrier systems in vaccine development and the interest for liposome-based vaccines has markedly increased. A key advantage of liposomes, archaeosomes and virosomes in general, and liposome-based vaccine delivery systems in particular, is their versatility and plasticity. Liposome composition and preparation can be chosen to achieve desired features such as selection of lipid, charge, size, size distribution, entrapment and location of antigens or adjuvants. Depending on the chemical properties, water-soluble antigens (proteins, peptides, nucleic acids, carbohydrates, haptens) are entrapped within the aqueous inner space of liposomes, whereas lipophilic compounds (lipopeptides, antigens, adjuvants, linker molecules) are intercalated into the lipid bilayer and antigens or adjuvants can be attached to the liposome surface either by adsorption or stable chemical linking. Coformulations containing different types of antigens or adjuvants can be combined with the parameters mentioned to tailor liposomal vaccines for individual applications. Special emphasis is given in this review to cationic adjuvant liposome vaccine formulations. Examples of vaccines made with CAF01, an adjuvant composed of the synthetic immune-stimulating mycobacterial cordfactor glycolipid trehalose dibehenate as immunomodulator and the cationic membrane forming molecule dimethyl dioctadecylammonium are presented. Other vaccines such as cationic liposome-DNA complexes (CLDCs) and other adjuvants like muramyl dipeptide, monophosphoryl lipid A and listeriolysin O are mentioned as well. The field of liposomes and liposome-based vaccines is vast. Therefore, this review concentrates on recent and relevant studies emphasizing current reports dealing with the most studied antigens and adjuvants, and pertinent examples of vaccines. Studies on liposome-based veterinary vaccines and experimental therapeutic cancer vaccines are also summarized.
Collapse
Affiliation(s)
- Reto A Schwendener
- Institute of Molecular Cancer Research, Laboratory of Liposome Research, University of Zurich, Winterthurerstrasse 190, Zurich, 8057, Switzerland
| |
Collapse
|
81
|
Sims KH, Tytler EM, Tipton J, Hill KL, Burgess SW, Shaw WA. Avanti lipid tools: connecting lipids, technology, and cell biology. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:1038-48. [PMID: 24954118 DOI: 10.1016/j.bbalip.2014.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 05/21/2014] [Accepted: 05/23/2014] [Indexed: 11/15/2022]
Abstract
Lipid research is challenging owing to the complexity and diversity of the lipidome. Here we review a set of experimental tools developed for the seasoned lipid researcher, as well as, those who are new to the field of lipid research. Novel tools for probing protein-lipid interactions, applications for lipid binding antibodies, enhanced systems for the cellular delivery of lipids, improved visualization of lipid membranes using gold-labeled lipids, and advances in mass spectrometric analysis techniques will be discussed. Because lipid mediators are known to participate in a host of signal transduction and trafficking pathways within the cell, a comprehensive lipid toolbox that aids the science of lipidomics research is essential to better understand the molecular mechanisms of interactions between cellular components. This article is part of a Special Issue entitled Tools to study lipid functions.
Collapse
Affiliation(s)
- Kacee H Sims
- Avanti Polar Lipids, Inc., 700 Industrial Park Drive, Alabaster, Al 35007, USA.
| | - Ewan M Tytler
- Avanti Polar Lipids, Inc., 700 Industrial Park Drive, Alabaster, Al 35007, USA.
| | - John Tipton
- Avanti Polar Lipids, Inc., 700 Industrial Park Drive, Alabaster, Al 35007, USA.
| | - Kasey L Hill
- Avanti Polar Lipids, Inc., 700 Industrial Park Drive, Alabaster, Al 35007, USA.
| | - Stephen W Burgess
- Avanti Polar Lipids, Inc., 700 Industrial Park Drive, Alabaster, Al 35007, USA.
| | - Walter A Shaw
- Avanti Polar Lipids, Inc., 700 Industrial Park Drive, Alabaster, Al 35007, USA.
| |
Collapse
|
82
|
Alving CR, Matyas GR, Torres O, Jalah R, Beck Z. Adjuvants for vaccines to drugs of abuse and addiction. Vaccine 2014; 32:5382-9. [PMID: 25111169 DOI: 10.1016/j.vaccine.2014.07.085] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/15/2014] [Accepted: 07/29/2014] [Indexed: 12/19/2022]
Abstract
Immunotherapeutic vaccines to drugs of abuse, including nicotine, cocaine, heroin, oxycodone, methamphetamine, and others are being developed. The theoretical basis of such vaccines is to induce antibodies that sequester the drug in the blood in the form of antibody-bound drug that cannot cross the blood brain barrier, thereby preventing psychoactive effects. Because the drugs are haptens a successful vaccine relies on development of appropriate hapten-protein carrier conjugates. However, because induction of high and prolonged levels of antibodies is required for an effective vaccine, and because injection of T-independent haptenic drugs of abuse does not induce memory recall responses, the role of adjuvants during immunization plays a critical role. As reviewed herein, preclinical studies often use strong adjuvants such as complete and incomplete Freund's adjuvant and others that cannot be, or in the case of many newer adjuvants, have never been, employed in humans. Balanced against this, the only adjuvant that has been included in candidate vaccines in human clinical trials to nicotine and cocaine has been aluminum hydroxide gel. While aluminum salts have been widely utilized worldwide in numerous licensed vaccines, the experience with human responses to aluminum salt-adjuvanted vaccines to haptenic drugs of abuse has suggested that the immune responses are too weak to allow development of a successful vaccine. What is needed is an adjuvant or combination of adjuvants that are safe, potent, widely available, easily manufactured, and cost-effective. Based on our review of the field we recommend the following adjuvant combinations either for research or for product development for human use: aluminum salt with adsorbed monophosphoryl lipid A (MPLA); liposomes containing MPLA [L(MPLA)]; L(MPLA) adsorbed to aluminum salt; oil-in-water emulsion; or oil-in-water emulsion containing MPLA.
Collapse
Affiliation(s)
- Carl R Alving
- 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.
| | - 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
| | - Oscar 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; U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| | - Rashmi Jalah
- 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; 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; U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| |
Collapse
|
83
|
Wang N, Wang T, Zhang M, Chen R, Niu R, Deng Y. Mannose derivative and lipid A dually decorated cationic liposomes as an effective cold chain free oral mucosal vaccine adjuvant-delivery system. Eur J Pharm Biopharm 2014; 88:194-206. [PMID: 24769065 DOI: 10.1016/j.ejpb.2014.04.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 04/14/2014] [Accepted: 04/16/2014] [Indexed: 01/24/2023]
Abstract
To develop convenient, effective cold chain-free subunit vaccines, a mannose-PEG-cholesterol conjugate (MPC) was synthesized as a lectin binding molecule and anchored onto liposomes which entrapped lipid A and model antigen to form a vaccine adjuvant-delivery system targeting antigen presenting cells. With MPC, soy phosphatidylcholine, stearylamine and monophosphoryl lipid A as emulsifiers dissolved in oil phase (O), and sucrose and BSA in water phase (W), the O/W emulsions were prepared and subsequently lyophilized. The lyophilized product was stable enough to be stored at room temperature and, upon rehydration, formed MPC-/lipid A-liposomes (MLLs) with a size under 300 nm and antigen association rates of around 36%. The MLLs given to mice via oral mucosal (o.m.) administration showed no side effects but induced potent immune responses as evidenced by the high levels of IgG in the sera and IgA in the salivary, intestinal and vaginal secretions of mice. High levels of IgG2a and IFN-γ in treated mice revealed that MLLs via o.m. vaccination induced a mixed Th1/Th2 response against antigens, establishing both humoral and cellular immunity. Thus, the MLLs may be a potent cold chain-free oral mucosal vaccine adjuvant-delivery system.
Collapse
Affiliation(s)
- Ning Wang
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, China
| | - Ting Wang
- Department of Pharmacy, Anhui Medical University, Hefei, China; Department of Pharmacy, Jining Medical College, Sunshine City, China.
| | - Meiling Zhang
- Department of Pharmacy, Anhui Medical University, Hefei, China
| | - Ruonan Chen
- Department of Pharmacy, Anhui Medical University, Hefei, China
| | - Ruowen Niu
- Department of Pharmacy, Anhui Medical University, Hefei, China
| | - Yihui Deng
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
84
|
Matyas GR, Rice KC, Cheng K, Li F, Antoline JFG, Iyer MR, Jacobson AE, Mayorov AV, Beck Z, Torres OB, Alving CR. Facial recognition of heroin vaccine opiates: type 1 cross-reactivities of antibodies induced by hydrolytically stable haptenic surrogates of heroin, 6-acetylmorphine, and morphine. Vaccine 2014; 32:1473-9. [PMID: 24486371 PMCID: PMC4090106 DOI: 10.1016/j.vaccine.2014.01.028] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/21/2013] [Accepted: 01/14/2014] [Indexed: 02/01/2023]
Abstract
Novel synthetic compounds similar to heroin and its major active metabolites, 6-acetylmorphine and morphine, were examined as potential surrogate haptens for the ability to interface with the immune system for a heroin vaccine. Recent studies have suggested that heroin-like haptens must degrade hydrolytically to induce independent immune responses both to heroin and to the metabolites, resulting in antisera containing mixtures of antibodies (type 2 cross-reactivity). To test this concept, two unique hydrolytically stable haptens were created based on presumed structural facial similarities to heroin or to its active metabolites. After conjugation of a heroin-like hapten (DiAmHap) to tetanus toxoid and mixing with liposomes containing monophosphoryl lipid A, high titers of antibodies after two injections in mice had complementary binding sites that exhibited strong type 1 ("true") specific cross-reactivity with heroin and with both of its physiologically active metabolites. Mice immunized with each surrogate hapten exhibited reduced antinociceptive effects caused by injection of heroin. This approach obviates the need to create hydrolytically unstable synthetic heroin-like compounds to induce independent immune responses to heroin and its active metabolites for vaccine development. Facial recognition of hydrolytically stable surrogate haptens by antibodies together with type 1 cross-reactivities with heroin and its metabolites can help to guide synthetic chemical strategies for efficient development of a heroin vaccine.
Collapse
Affiliation(s)
- 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
| | - Kenner C Rice
- Drug Design and Synthesis Section, Chemical Biology Research Branch, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 5625 Fishers Lane, Bethesda, MD 20892-9415, USA; National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 4N03, Bethesda, MD 20892-9415, USA
| | - Kejun Cheng
- Drug Design and Synthesis Section, Chemical Biology Research Branch, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 5625 Fishers Lane, Bethesda, MD 20892-9415, USA; National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 4N03, Bethesda, MD 20892-9415, USA
| | - Fuying Li
- Drug Design and Synthesis Section, Chemical Biology Research Branch, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 5625 Fishers Lane, Bethesda, MD 20892-9415, USA; National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 4N03, Bethesda, MD 20892-9415, USA
| | - Joshua F G Antoline
- Drug Design and Synthesis Section, Chemical Biology Research Branch, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 5625 Fishers Lane, Bethesda, MD 20892-9415, USA; National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 4N03, Bethesda, MD 20892-9415, USA
| | - Malliga R Iyer
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 4N03, Bethesda, MD 20892-9415, USA
| | - Arthur E Jacobson
- Drug Design and Synthesis Section, Chemical Biology Research Branch, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, 5625 Fishers Lane, Bethesda, MD 20892-9415, USA; National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 4N03, Bethesda, MD 20892-9415, USA
| | - Alexander V Mayorov
- 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; 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; 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; U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817 USA
| | - Carl R Alving
- 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.
| |
Collapse
|
85
|
Serrano S, Araujo A, Apellániz B, Bryson S, Carravilla P, de la Arada I, Huarte N, Rujas E, Pai EF, Arrondo JLR, Domene C, Jiménez MA, Nieva JL. Structure and immunogenicity of a peptide vaccine, including the complete HIV-1 gp41 2F5 epitope: implications for antibody recognition mechanism and immunogen design. J Biol Chem 2014; 289:6565-6580. [PMID: 24429284 DOI: 10.1074/jbc.m113.527747] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The membrane-proximal external region (MPER) of gp41 harbors the epitope recognized by the broadly neutralizing anti-HIV 2F5 antibody, a research focus in HIV-1 vaccine development. In this work, we analyze the structure and immunogenic properties of MPERp, a peptide vaccine that includes the following: (i) the complete sequence protected from proteolysis by the 2F5 paratope; (ii) downstream residues postulated to establish weak contacts with the CDR-H3 loop of the antibody, which are believed to be crucial for neutralization; and (iii) an aromatic rich anchor to the membrane interface. MPERp structures solved in dodecylphosphocholine micelles and 25% 1,1,1,3,3,3-hexafluoro-2-propanol (v/v) confirmed folding of the complete 2F5 epitope within continuous kinked helices. Infrared spectroscopy (IR) measurements demonstrated the retention of main helical conformations in immunogenic formulations based on alum, Freund's adjuvant, or two different types of liposomes. Binding to membrane-inserted MPERp, IR, molecular dynamics simulations, and characterization of the immune responses further suggested that packed helical bundles partially inserted into the lipid bilayer, rather than monomeric helices adsorbed to the membrane interface, could encompass effective MPER peptide vaccines. Together, our data constitute a proof-of-concept to support MPER-based peptides in combination with liposomes as stand-alone immunogens and suggest new approaches for structure-aided MPER vaccine development.
Collapse
Affiliation(s)
- Soraya Serrano
- Institute of Physical Chemistry "Rocasolano," Consejo Superior de Investigaciones Científicas (IQFR-CSIC), Serrano 119, E-28006 Madrid, Spain
| | - Aitziber Araujo
- Biophysics Unit, Consejo Superior de Investigaciones Científicas and University of the Basque Country (CSIC-UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P. O. Box 644, 48080 Bilbao, Spain
| | - Beatriz Apellániz
- Biophysics Unit, Consejo Superior de Investigaciones Científicas and University of the Basque Country (CSIC-UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P. O. Box 644, 48080 Bilbao, Spain
| | - Steve Bryson
- Departments of Biochemistry, Medical Biophysics, and Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada; The Campbell Family Institute for Cancer Research, Ontario Cancer Institute/University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Pablo Carravilla
- Biophysics Unit, Consejo Superior de Investigaciones Científicas and University of the Basque Country (CSIC-UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P. O. Box 644, 48080 Bilbao, Spain
| | - Igor de la Arada
- Biophysics Unit, Consejo Superior de Investigaciones Científicas and University of the Basque Country (CSIC-UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P. O. Box 644, 48080 Bilbao, Spain
| | - Nerea Huarte
- Biophysics Unit, Consejo Superior de Investigaciones Científicas and University of the Basque Country (CSIC-UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P. O. Box 644, 48080 Bilbao, Spain
| | - Edurne Rujas
- Biophysics Unit, Consejo Superior de Investigaciones Científicas and University of the Basque Country (CSIC-UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P. O. Box 644, 48080 Bilbao, Spain
| | - Emil F Pai
- Departments of Biochemistry, Medical Biophysics, and Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada; The Campbell Family Institute for Cancer Research, Ontario Cancer Institute/University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - José L R Arrondo
- Biophysics Unit, Consejo Superior de Investigaciones Científicas and University of the Basque Country (CSIC-UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P. O. Box 644, 48080 Bilbao, Spain
| | - Carmen Domene
- Chemistry Research Laboratory, Mansfield Road, University of Oxford, Oxford OX1 3TA, United Kingdom; Department of Chemistry, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - María Angeles Jiménez
- Institute of Physical Chemistry "Rocasolano," Consejo Superior de Investigaciones Científicas (IQFR-CSIC), Serrano 119, E-28006 Madrid, Spain.
| | - José L Nieva
- Biophysics Unit, Consejo Superior de Investigaciones Científicas and University of the Basque Country (CSIC-UPV/EHU) and Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), P. O. Box 644, 48080 Bilbao, Spain.
| |
Collapse
|
86
|
Functionalised Nanoliposomes for Construction of Recombinant Vaccines: Lyme Disease as an Example. MOLECULAR VACCINES 2014. [PMCID: PMC7120364 DOI: 10.1007/978-3-319-00978-0_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Liposomes (phospholipid bilayer vesicles) represent an almost ideal carrier system for the preparation of synthetic vaccines due to their biodegradability and capacity to protect and transport molecules of different physicochemical properties (including size, hydrophilicity, hydrophobicity, and charge). Liposomal carriers can be applied by invasive (e.g. i.m., s.c., i.d.) as well as non-invasive (transdermal and mucosal) routes. In the last 15 years, liposome vaccine technology has matured and several vaccines containing liposome-based adjuvants have been approved for human and veterinary use or have reached late stages of clinical evaluation. Given the intensifying interest in liposome-based vaccines, it is important to understand precisely how liposomes interact with the immune system and how they stimulate immunity. It has become clear that the physicochemical properties of liposomal vaccines – method of antigen attachment, lipid composition, bilayer fluidity, particle charge, and other properties – exert strong effects on the resulting immune response. In this chapter we will discuss some aspects of liposomal vaccines including the effect of novel and emerging immunomodulator incorporation. The application of metallochelating nanoliposomes for development of recombinant vaccine against Lyme disease will be presented as a suitable example.
Collapse
|
87
|
Evaluation of monophosphoryl lipid A as adjuvant for pulmonary delivered influenza vaccine. J Control Release 2013; 174:51-62. [PMID: 24269505 DOI: 10.1016/j.jconrel.2013.11.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 11/12/2013] [Accepted: 11/12/2013] [Indexed: 01/22/2023]
Abstract
Prophylaxis against influenza could be improved by the development of a stable, easy to deliver, potent mucosal vaccine. In this study, we spray-freeze-dried (SFD) whole inactivated virus influenza vaccine (WIV) alone or supplemented with monophosphoryl lipid A (MPLA) using inulin as a lyoprotectant. Physical characterization revealed that the SFD powder consisted of highly porous particles with a size distribution suitable for pulmonary administration. The receptor-binding properties of WIV and the immunostimulatory properties of MPLA were preserved after spray-freeze-drying as indicated by unchanged hemagglutination titers and a retained ability of the vaccine to activate NFkB after incubation with a reporter cell line, respectively. Pulmonary vaccination of mice with MPLA-adjuvanted liquid or powder WIV resulted in induction of higher mucosal and systemic antibody concentrations than vaccination with non-adjuvanted formulations. When exposed to influenza virus, mice immunized with MPLA-adjuvanted pulmonary vaccine showed similar protection in terms of reduction in lung virus titers and prevention of weight loss as mice immunized intramuscularly with subunit vaccine. Characterization of the antibody response revealed a balanced IgG2a-to-IgG1 profile along with induction of both memory IgA- and IgG-producing B cells in mice immunized with MPLA-adjuvanted vaccine. These studies suggest that the mucosal and systemic immune responses to pulmonary delivered influenza vaccines can be significantly enhanced by using MPLA as adjuvant. MPLA-adjuvanted SFD vaccine was particularly effective implying that delivery of adjuvanted vaccine powder to the lungs can be an attractive way of immunization against influenza.
Collapse
|
88
|
Haughney SL, Petersen LK, Schoofs AD, Ramer-Tait AE, King JD, Briles DE, Wannemuehler MJ, Narasimhan B. Retention of structure, antigenicity, and biological function of pneumococcal surface protein A (PspA) released from polyanhydride nanoparticles. Acta Biomater 2013; 9:8262-71. [PMID: 23774257 DOI: 10.1016/j.actbio.2013.06.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 05/29/2013] [Accepted: 06/03/2013] [Indexed: 02/03/2023]
Abstract
Pneumococcal surface protein A (PspA) is a choline-binding protein which is a virulence factor found on the surface of all Streptococcus pneumoniae strains. Vaccination with PspA has been shown to be protective against a lethal challenge with S. pneumoniae, making it a promising immunogen for use in vaccines. Herein the design of a PspA-based subunit vaccine using polyanhydride nanoparticles as a delivery platform is described. Nanoparticles based on sebacic acid (SA), 1,6-bis-(p-carboxyphenoxy)hexane (CPH) and 1,8-bis-(p-carboxyphenoxy)-3,6-dioxaoctane (CPTEG), specifically 50:50 CPTEG:CPH and 20:80 CPH:SA, were used to encapsulate and release PspA. The protein released from the nanoparticle formulations retained its primary and secondary structure as well as its antigenicity. The released PspA was also biologically functional based on its ability to bind to apolactoferrin and prevent its bactericidal activity against Escherichia coli. When the PspA nanoparticle formulations were administered subcutaneously to mice they elicited a high titer and high avidity anti-PspA antibody response. Together these studies provide a framework for the rational design of a vaccine against S. pneumoniae based on polyanhydride nanoparticles.
Collapse
|
89
|
Orr MT, Fox CB, Baldwin SL, Sivananthan SJ, Lucas E, Lin S, Phan T, Moon JJ, Vedvick TS, Reed SG, Coler RN. Adjuvant formulation structure and composition are critical for the development of an effective vaccine against tuberculosis. J Control Release 2013; 172:190-200. [PMID: 23933525 DOI: 10.1016/j.jconrel.2013.07.030] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Revised: 07/12/2013] [Accepted: 07/25/2013] [Indexed: 12/26/2022]
Abstract
One third of the world is infected with Mycobacterium tuberculosis (Mtb) with eight million new cases of active tuberculosis (TB) each year. Development of a new vaccine to augment or replace the only approved TB vaccine, BCG, is needed to control this disease. Mtb infection is primarily controlled by TH1 cells through the production of IFN-γ and TNF which activate infected macrophages to kill the bacterium. Here we examine an array of adjuvant formulations containing the TLR4 agonist GLA to identify candidate adjuvants to pair with ID93, a lead TB vaccine antigen, to elicit protective TH1 responses. We evaluate a variety of adjuvant formulations including alum, liposomes, and oil-in-water emulsions to determine how changes in formulation composition alter adjuvant activity. We find that alum and an aqueous nanosuspension of GLA synergize to enhance generation of ID93-specific TH1 responses, whereas neither on their own are effective adjuvants for generation of ID93-specific TH1 responses. For GLA containing oil-in-water emulsions, the selection of the oil component is critical for adjuvant activity, whereas a variety of lipid components may be used in liposomal formulations of GLA. The composition of the liposome formulation of ID93/GLA does alter the magnitude of the TH1 response. These results demonstrate that there are multiple solutions for an effective formulation of a novel TB vaccine candidate that enhances both TH1 generation and protective efficacy.
Collapse
Affiliation(s)
- Mark T Orr
- Infectious Disease Research Institute, Seattle 98104, USA
| | | | | | | | - Elyse Lucas
- Infectious Disease Research Institute, Seattle 98104, USA
| | - Susan Lin
- Infectious Disease Research Institute, Seattle 98104, USA
| | - Tony Phan
- Infectious Disease Research Institute, Seattle 98104, USA
| | - James J Moon
- Center for Immunology and Inflammatory Diseases and Pulmonary and Critical Care Unit, Massachusetts General Hospital and Harvard Medical School, Charlestown 02129, USA
| | | | - Steven G Reed
- Infectious Disease Research Institute, Seattle 98104, USA
| | - Rhea N Coler
- Infectious Disease Research Institute, Seattle 98104, USA.
| |
Collapse
|
90
|
Matyas GR, Mayorov AV, Rice KC, Jacobson AE, Cheng K, Iyer MR, Li F, Beck Z, Janda KD, Alving CR. Liposomes containing monophosphoryl lipid A: a potent adjuvant system for inducing antibodies to heroin hapten analogs. Vaccine 2013; 31:2804-10. [PMID: 23624097 PMCID: PMC4120113 DOI: 10.1016/j.vaccine.2013.04.027] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Revised: 03/08/2013] [Accepted: 04/09/2013] [Indexed: 12/21/2022]
Abstract
In order to create an effective immunization approach for a potential vaccine to heroin, liposomes containing monophosphoryl lipid A [L(MPLA)] were tested as an adjuvant system to induce antibodies to heroin hapten analogs. Four synthetic haptens and two immunization strategies were employed. In the first strategy, a hydrophobic 23 amino acid immunogenic peptide derived from the membrane proximal external region of gp41 from HIV-1 envelope protein was embedded as a carrier in the outer surface of L(MPLA), to which was conjugated a 15 amino acid universal T cell epitope and a terminal heroin hapten analog. In the second strategy, tetanus toxoid (TT) carrier protein was decorated with haptens by conjugation, and the hapten-conjugated protein was mixed with L(MPLA). After immunization of mice, each of the immunization strategies was effective for induction of IgG anti-hapten antibodies. The first immunization strategy induced a mean end-point IgG titer against one of two haptens tested of approximately 12,800; however, no detectable antibodies were induced against the liposome-associated HIV-1 carrier peptide. In the second immunization strategy, depending on the hapten used for decorating the TT, end-point IgG titers ranged from 100,000 to 6,500,000. In this strategy, in which hapten was conjugated to the TT, end-point IgG titers of 400,000 to the TT carrier were observed with each conjugate. However, upon mixing unconjugated TT with L(MPLA), anti-TT titers of 6,500,000 were observed. We conclude that L(MPLA) serves as a potent adjuvant for inducing antibodies to candidate heroin haptens. However, antibodies to the carrier peptide or protein were partly or completed inhibited by the presence of conjugated hapten.
Collapse
Affiliation(s)
- 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
| | - Alexander V. Mayorov
- 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
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817 USA
| | - Kenner C. Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse, National Institutes of Health, 5625 Fishers Lane, Rockville 20852 MD USA
| | - Arthur E. Jacobson
- Chemical Biology Research Branch, National Institute on Drug Abuse, National Institutes of Health, 5625 Fishers Lane, Rockville 20852 MD USA
| | - Kejun Cheng
- Chemical Biology Research Branch, National Institute on Drug Abuse, National Institutes of Health, 5625 Fishers Lane, Rockville 20852 MD USA
| | - Malliga R. Iyer
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville 20852 MD USA
| | - Fuying Li
- Chemical Biology Research Branch, National Institute on Drug Abuse, National Institutes of Health, 5625 Fishers Lane, Rockville 20852 MD 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
- U.S. Military HIV Research Program, Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817 USA
| | - Kim D. Janda
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology and Worm Institute of Research and Medicine, The Scripps Research Institute, La Jolla, California 92037
| | - Carl R. Alving
- 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
| |
Collapse
|
91
|
Chopra S, Venkatesan N, Betageri GV. Liposomes as nanocarriers for anti-HIV therapy. Drug Deliv Transl Res 2013; 3:471-8. [DOI: 10.1007/s13346-013-0134-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
92
|
Lockner JW, Ho SO, McCague KC, Chiang SM, Do TQ, Fujii G, Janda KD. Enhancing nicotine vaccine immunogenicity with liposomes. Bioorg Med Chem Lett 2012; 23:975-8. [PMID: 23313243 DOI: 10.1016/j.bmcl.2012.12.048] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/10/2012] [Accepted: 12/13/2012] [Indexed: 11/30/2022]
Abstract
A major liability of existing nicotine vaccine candidates is the wide variation in anti-nicotine immune responses among clinical trial participants. In order to address this liability, significant emphasis has been directed at evaluating adjuvants and delivery systems that confer more robust potentiation of the anti-nicotine immune response. Toward that end, we have initiated work that seeks to exploit the adjuvant effect of liposomes, with or without Toll-like receptor agonist(s). The results of the murine immunization study described herein support the hypothesis that a liposomal nicotine vaccine formulation may provide a means for addressing the immunogenicity challenge.
Collapse
Affiliation(s)
- Jonathan W Lockner
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|