1
|
Berger S, Zeyn Y, Wagner E, Bros M. New insights for the development of efficient DNA vaccines. Microb Biotechnol 2024; 17:e70053. [PMID: 39545748 PMCID: PMC11565620 DOI: 10.1111/1751-7915.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Despite the great potential of DNA vaccines for a broad range of applications, ranging from prevention of infections, over treatment of autoimmune and allergic diseases to cancer immunotherapies, the implementation of such therapies for clinical treatment is far behind the expectations up to now. The main reason is the poor immunogenicity of DNA vaccines in humans. Consequently, the improvement of the performance of DNA vaccines in vivo is required. This mini-review provides an overview of the current state of DNA vaccines and the various strategies to enhance the immunogenic potential of DNA vaccines, including (i) the optimization of the DNA construct itself regarding size, nuclear transfer and transcriptional regulation; (ii) the use of appropriate adjuvants; and (iii) improved delivery, for example, by careful choice of the administration route, physical methods such as electroporation and nanomaterials that may allow cell type-specific targeting. Moreover, combining nanoformulated DNA vaccines with other immunotherapies and prime-boost strategies may help to enhance success of treatment.
Collapse
Affiliation(s)
- Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Yanira Zeyn
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Matthias Bros
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| |
Collapse
|
2
|
Gong L, Zhang Y, Wang L, Zhao X, Wang L, Qiu X, Yang X, Zhu W, Lv L, Kang Y, Wu Y, Zhang A, Du Y, Wang X, Zhang G, Sun A, Zhuang G. Advancing vaccine development: Evaluation of a mannose-modified lipid nanoparticle-based candidate for African swine fever p30 mRNA vaccine eliciting robust immune response in mice. Int J Biol Macromol 2024; 270:132432. [PMID: 38761609 DOI: 10.1016/j.ijbiomac.2024.132432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024]
Abstract
The African swine fever virus (ASFV) continues to pose significant economic and pandemic risks. Consequently, discovering new, efficient vaccines is crucial. Messenger RNA (mRNA) vaccines have emerged as promising candidates, providing minimal risk of insertional mutagenesis, high safety profiles, effectiveness, rapid scalability in production, and cost-effectiveness. In this study, we have developed an ASF p30 mRNA vaccine candidate (mRNA/Man-LNP) employing mannose-modified lipid nanoparticles (LNPs). The mRNA/Man-LNP exhibited effective antigen presentation and facilitated dendritic cells (DCs) maturation. Notably, it elicited strong IgG titers and activated CD4+ and CD8+ T-cells in immunized mice, all while adhering to stringent biosafety standards. This investigation demonstrates that mRNA/Man-LNP can trigger both humoral and cellular immune responses, suggesting its potential as a potent and promising vaccine candidate for controlling African swine fever (ASF).
Collapse
Affiliation(s)
- Lele Gong
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Yuanyuan Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Lele Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Xuyang Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Lucai Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Xiangqi Qiu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Xilong Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Wenhui Zhu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Lijie Lv
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Yunzhe Kang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Yanan Wu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Angke Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Yongkun Du
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Xuannian Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Gaiping Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Longhu Laboratory of Advanced Immunology, Zhengzhou 450046, China; School of Advanced Agriculture Sciences, Peking University, Beijing 100871, China
| | - Aijun Sun
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China.
| | - Guoqing Zhuang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China.
| |
Collapse
|
3
|
Steffens RC, Folda P, Fendler NL, Höhn M, Bücher-Schossau K, Kempter S, Snyder NL, Hartmann L, Wagner E, Berger S. GalNAc- or Mannose-PEG-Functionalized Polyplexes Enable Effective Lectin-Mediated DNA Delivery. Bioconjug Chem 2024; 35:351-370. [PMID: 38440876 DOI: 10.1021/acs.bioconjchem.3c00546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
A cationic, dendrimer-like oligo(aminoamide) carrier with four-arm topology based on succinoyl tetraethylene pentamine and histidines, cysteines, and N-terminal azido-lysines was screened for plasmid DNA delivery on various cell lines. The incorporated azides allow modification with various shielding agents of different polyethylene glycol (PEG) lengths and/or different ligands by copper-free click reaction, either before or after polyplex formation. Prefunctionalization was found to be advantageous over postfunctionalization in terms of nanoparticle formation, stability, and efficacy. A length of 24 ethylene oxide repetition units and prefunctionalization of ≥50% of azides per carrier promoted optimal polyplex shielding. PEG shielding resulted in drastically reduced DNA transfer, which could be successfully restored by active lectin targeting via novel GalNAc or mannose ligands, enabling enhanced receptor-mediated endocytosis of the carrier system. The involvement of the asialoglycoprotein receptor (ASGPR) in the uptake of GalNAc-functionalized polyplexes was confirmed in the ASGPR-positive hepatocarcinoma cell lines HepG2 and Huh7. Mannose-modified polyplexes showed superior cellular uptake and transfection efficacy compared to unmodified and shielded polyplexes in mannose-receptor-expressing dendritic cell-like DC2.4 cells.
Collapse
Affiliation(s)
- Ricarda C Steffens
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Paul Folda
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
| | - Nikole L Fendler
- Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
| | - Katharina Bücher-Schossau
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Susanne Kempter
- Faculty of Physics, LMU Munich, 80539 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Nicole L Snyder
- Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Laura Hartmann
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
- Institute for Macromolecular Chemistry, University Freiburg, Stefan-Meier-Str. 31, 79104 Freiburg im Breisgau, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| |
Collapse
|
4
|
Tang Y, Li L. The Application of Nanovaccines in Autoimmune Diseases. Int J Nanomedicine 2024; 19:367-388. [PMID: 38229706 PMCID: PMC10790641 DOI: 10.2147/ijn.s440612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/05/2024] [Indexed: 01/18/2024] Open
Abstract
Autoimmune diseases are diseases caused by the body's chronic immune responses to self-antigens and attacks on the host's own cells, tissues and organs. The dysfunction of innate immunity and adaptive immunity leads to the destruction of autoimmune tolerance, which is the most basic factor leading to pathogenesis. The optimal strategy for autoimmune diseases is to modify the host immune system to restore tolerance. The ideal effect of therapeutic autoimmune diseases is to eliminate the autoantigen-specific spontaneous immune response without interfering with the immune response against other antigens. Therapeutic nanovaccines that produce immune tolerance conform to this principle. Nanomaterials provide a platform for antigen loading and modification due to their unique physical and chemical properties. Nanovaccines based on nanomaterial technology can simultaneously enable antigens and adjuvants to be absorbed by immune cells and induce rapid and durable immunity. Nanovaccines have the advantages of being able to be designed and loaded and of better protecting antigens from premature degradation. Nanovaccines also have the ability to target specific tissues or cells through optimized design. We review the latest research progress of nanovaccines for autoimmune diseases and the design strategies of nanovaccines to promote the development of more effective nanovaccines for autoimmune diseases.
Collapse
Affiliation(s)
- Yuhong Tang
- Department of Dermatology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Lili Li
- Department of Dermatology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| |
Collapse
|
5
|
Ghafelehbashi R, Salehi M, Kouhi M, AlizadehNaini A, Sajadi-Javan ZS, Nejatidanesh F. Recent progress in cancer immunotherapy: Application of nano-therapeutic systems. J Drug Deliv Sci Technol 2024; 91:105184. [DOI: 10.1016/j.jddst.2023.105184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
6
|
Zeyn Y, Hobernik D, Wilk U, Pöhmerer J, Hieber C, Medina-Montano C, Röhrig N, Strähle CF, Thoma-Kress AK, Wagner E, Bros M, Berger S. Transcriptional Targeting of Dendritic Cells Using an Optimized Human Fascin1 Gene Promoter. Int J Mol Sci 2023; 24:16938. [PMID: 38069260 PMCID: PMC10706967 DOI: 10.3390/ijms242316938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Deeper knowledge about the role of the tumor microenvironment (TME) in cancer development and progression has resulted in new strategies such as gene-based cancer immunotherapy. Whereas some approaches focus on the expression of tumoricidal genes within the TME, DNA-based vaccines are intended to be expressed in antigen-presenting cells (e.g., dendritic cells, DCs) in secondary lymphoid organs, which in turn induce anti-tumor T cell responses. Besides effective delivery systems and the requirement of appropriate adjuvants, DNA vaccines themselves need to be optimized regarding efficacy and selectivity. In this work, the concept of DC-focused transcriptional targeting was tested by applying a plasmid encoding for the luciferase reporter gene under the control of a derivative of the human fascin1 gene promoter (pFscnLuc), comprising the proximal core promoter fused to the normally more distantly located DC enhancer region. DC-focused activity of this reporter construct was confirmed in cell culture in comparison to a standard reporter vector encoding for luciferase under the control of the strong ubiquitously active cytomegalovirus promoter and enhancer (pCMVLuc). Both plasmids were also compared upon intravenous administration in mice. The organ- and cell type-specific expression profile of pFscnLuc versus pCMVLuc demonstrated favorable activity especially in the spleen as a central immune organ and within the spleen in DCs.
Collapse
Affiliation(s)
- Yanira Zeyn
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Dominika Hobernik
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Ulrich Wilk
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (U.W.); (J.P.); (E.W.)
| | - Jana Pöhmerer
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (U.W.); (J.P.); (E.W.)
| | - Christoph Hieber
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Carolina Medina-Montano
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Nadine Röhrig
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Caroline F. Strähle
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.F.S.); (A.K.T.-K.)
| | - Andrea K. Thoma-Kress
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.F.S.); (A.K.T.-K.)
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (U.W.); (J.P.); (E.W.)
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University (JGU) Mainz, 55131 Mainz, Germany; (Y.Z.); (D.H.); (C.H.); (C.M.-M.); (N.R.)
| | - Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScience, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany; (U.W.); (J.P.); (E.W.)
| |
Collapse
|
7
|
Clemente B, Denis M, Silveira CP, Schiavetti F, Brazzoli M, Stranges D. Straight to the point: targeted mRNA-delivery to immune cells for improved vaccine design. Front Immunol 2023; 14:1294929. [PMID: 38090568 PMCID: PMC10711611 DOI: 10.3389/fimmu.2023.1294929] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
With the deepening of our understanding of adaptive immunity at the cellular and molecular level, targeting antigens directly to immune cells has proven to be a successful strategy to develop innovative and potent vaccines. Indeed, it offers the potential to increase vaccine potency and/or modulate immune response quality while reducing off-target effects. With mRNA-vaccines establishing themselves as a versatile technology for future applications, in the last years several approaches have been explored to target nanoparticles-enabled mRNA-delivery systems to immune cells, with a focus on dendritic cells. Dendritic cells (DCs) are the most potent antigen presenting cells and key mediators of B- and T-cell immunity, and therefore considered as an ideal target for cell-specific antigen delivery. Indeed, improved potency of DC-targeted vaccines has been proved in vitro and in vivo. This review discusses the potential specific targets for immune system-directed mRNA delivery, as well as the different targeting ligand classes and delivery systems used for this purpose.
Collapse
|
8
|
Dong H, Li Q, Zhang Y, Ding M, Teng Z, Mou Y. Biomaterials Facilitating Dendritic Cell-Mediated Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301339. [PMID: 37088780 PMCID: PMC10288267 DOI: 10.1002/advs.202301339] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/22/2023] [Indexed: 05/03/2023]
Abstract
Dendritic cell (DC)-based cancer immunotherapy has exhibited remarkable clinical prospects because DCs play a central role in initiating and regulating adaptive immune responses. However, the application of traditional DC-mediated immunotherapy is limited due to insufficient antigen delivery, inadequate antigen presentation, and high levels of immunosuppression. To address these challenges, engineered biomaterials have been exploited to enhance DC-mediated immunotherapeutic effects. In this review, vital principal components that can enhance DC-mediated immunotherapeutic effects are first introduced. The parameters considered in the rational design of biomaterials, including targeting modifications, size, shape, surface, and mechanical properties, which can affect biomaterial optimization of DC functions, are further summarized. Moreover, recent applications of various engineered biomaterials in the field of DC-mediated immunotherapy are reviewed, including those serve as immune component delivery platforms, remodel the tumor microenvironment, and synergistically enhance the effects of other antitumor therapies. Overall, the present review comprehensively and systematically summarizes biomaterials related to the promotion of DC functions; and specifically focuses on the recent advances in biomaterial designs for DC activation to eradicate tumors. The challenges and opportunities of treatment strategies designed to amplify DCs via the application of biomaterials are discussed with the aim of inspiring the clinical translation of future DC-mediated cancer immunotherapies.
Collapse
Affiliation(s)
- Heng Dong
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Qiang Li
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Yu Zhang
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Meng Ding
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023P. R. China
| | - Yongbin Mou
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| |
Collapse
|
9
|
Winkeljann B, Keul DC, Merkel OM. Engineering poly- and micelleplexes for nucleic acid delivery - A reflection on their endosomal escape. J Control Release 2023; 353:518-534. [PMID: 36496051 PMCID: PMC9900387 DOI: 10.1016/j.jconrel.2022.12.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022]
Abstract
For the longest time, the field of nucleic acid delivery has remained skeptical whether or not polycationic drug carrier systems would ever make it into clinical practice. Yet, with the disclosure of patents on polyethyleneimine-based RNA carriers through leading companies in the field of nucleic acid therapeutics such as BioNTech SE and the progress in clinical studies beyond phase I trials, this aloofness seems to regress. As one of the most striking characteristics of polymer-based vectors, the extraordinary tunability can be both a blessing and a curse. Yet, knowing about the adjustment screws and how they impact the performance of the drug carrier provides the formulation scientist committed to its development with a head start. Here, we equip the reader with a toolbox - a toolbox that should advise and support the developer to conceptualize a cutting-edge poly- or micelleplex system for the delivery of therapeutic nucleic acids; to be specific, to engineer the vector towards maximum endosomal escape performance at minimum toxicity. Therefore, after briefly sketching the boundary conditions of polymeric vector design, we will dive into the topic of endosomal trafficking. We will not only discuss the most recent knowledge of the endo-lysosomal compartment but further depict different hypotheses and mechanisms that facilitate the endosomal escape of polyplex systems. Finally, we will combine the different facets introduced in the previous chapters with the fundamental building blocks of polymer vector design and evaluate the advantages and drawbacks. Throughout the article, a particular focus will be placed on cellular peculiarities, not only as an additional barrier, but also to give inspiration to how such cell-specific traits might be capitalized on.
Collapse
Affiliation(s)
- Benjamin Winkeljann
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Butenandtstrasse 5-13, Haus B, 81377 Munich, Germany,Center for NanoScience (CeNS), Ludwig-Maximilians-University Munich, 80799 Munich, Germany
| | - David C. Keul
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Butenandtstrasse 5-13, Haus B, 81377 Munich, Germany
| | - Olivia M. Merkel
- Department of Pharmacy, Ludwig-Maximilians-University Munich, Butenandtstrasse 5-13, Haus B, 81377 Munich, Germany,Center for NanoScience (CeNS), Ludwig-Maximilians-University Munich, 80799 Munich, Germany,Corresponding author at: Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Haus B, 81377 München, Germany
| |
Collapse
|
10
|
Hussain Z, Thu HE, Khan S, Sohail M, Sarfraz RM, Mahmood A, Abourehab MA. Phytonanomedicines, a state-of-the-art strategy for targeted delivery of anti-inflammatory phytochemicals: A review of improved pharmacokinetic profile and therapeutic efficacy. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
|
11
|
Abstract
RNA-based therapeutics have shown great promise in treating a broad spectrum of diseases through various mechanisms including knockdown of pathological genes, expression of therapeutic proteins, and programmed gene editing. Due to the inherent instability and negative-charges of RNA molecules, RNA-based therapeutics can make the most use of delivery systems to overcome biological barriers and to release the RNA payload into the cytosol. Among different types of delivery systems, lipid-based RNA delivery systems, particularly lipid nanoparticles (LNPs), have been extensively studied due to their unique properties, such as simple chemical synthesis of lipid components, scalable manufacturing processes of LNPs, and wide packaging capability. LNPs represent the most widely used delivery systems for RNA-based therapeutics, as evidenced by the clinical approvals of three LNP-RNA formulations, patisiran, BNT162b2, and mRNA-1273. This review covers recent advances of lipids, lipid derivatives, and lipid-derived macromolecules used in RNA delivery over the past several decades. We focus mainly on their chemical structures, synthetic routes, characterization, formulation methods, and structure-activity relationships. We also briefly describe the current status of representative preclinical studies and clinical trials and highlight future opportunities and challenges.
Collapse
Affiliation(s)
- Yuebao Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Changzhen Sun
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Chang Wang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Katarina E Jankovic
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
- Department of Biomedical Engineering, The Center for Clinical and Translational Science, The Comprehensive Cancer Center, Dorothy M. Davis Heart & Lung Research Institute, Department of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
12
|
Importance of particle size of oligomannose-coated liposomes for induction of Th1 immunity. Int Immunopharmacol 2021; 99:108068. [PMID: 34426114 DOI: 10.1016/j.intimp.2021.108068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/09/2021] [Accepted: 08/09/2021] [Indexed: 12/21/2022]
Abstract
Oligomannose-coated liposomes (OMLs) comprised of dipalmitoylphosphatidylcholine, cholesterol and Man3-DPPE at a molar ratio of 1:1:0.1 and particle diameters of about 1000 nm can induce liposome-encased antigen-specific strong Th1 immunity. In this study, we evaluated the effect of particle sizes of OMLs on induction of Th1 immune responses in mice. Spleen cells obtained from mice immunized with antigen-encapsulating OMLs with 1000- and 800-nm diameters secreted remarkably high levels of IFN-γ upon in vitro stimulation. In addition, sera of mice that received these OMLs had significantly higher titers of antigen-specific IgG2a than those of IgG1, which are commonly associated with Th1 responses. In contrast, treatment with antigen-encapsulating OMLs with 400- and 200-nm diameters failed to induce IFN-γ secretion from spleen cells, although these OMLs did elicit elevation of antigen-specific IgGs. In addition, the titers of serum antigen-specific IgG2a were the same as those of IgG1 in mice that received 400-nm OMLs. Resident peritoneal mononuclear phagocytes (MNPs) treated with OMLs of diameter ≥ 600 nm secreted IL-12, which is essential for induction of Th1 immune responses, while those treated with OMLs of ≤ 400 nm failed to produce this cytokine. However, 400-nm OMLs did induce enhanced expression of MHC class II and costimulatory molecules on MNPs, similarly to OMLs of ≥ 600 nm. Taken together, these results strongly indicate that OMLs of diameter ≥ 600 nm are required to induce Th1 immune responses against OML-encased antigens, although OMLs of diameter ≤ 400 nm can activate MNPs.
Collapse
|
13
|
Mannosylated polylactic-co-glycolic acid (MN-PLGA) nanoparticles induce potent anti-tumor immunity in murine model of breast cancer. Biomed Pharmacother 2021; 142:111962. [PMID: 34358752 DOI: 10.1016/j.biopha.2021.111962] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 11/20/2022] Open
Abstract
Nanoparticle-based cancer immunotherapy is considered a novel and promising therapeutic strategy aimed at stimulating host immune responses against tumors. To this end, in the present study, mannan-decorated polylactic-co-glycolic acid (PLGA) nanoparticles containing tumor cell lysate (TCL) and poly riboinosinic polycytidylic acid (poly I:C) were used as antigen delivery systems to immunize breast tumor-bearing Balb/c mice. PLGA nanoparticles were fabricated employing a double emulsion solvent evaporation method. The formation of spherical and uniform nanoparticles (NPs) ranging 150-250 nm was detected by field emission scanning electron microscopy (FESEM) and dynamic light scattering (DLS). Four nanoformulation were used to treat mice and vaccination-induced immunological responses. Tumor regression and overall survival rate were evaluated in four experimental groups. Tumor cell lysate and poly I:C loaded mannan-decorated nanoparticles (TCL-Poly I:C) NP-MN caused a significant decrease in tumor growth and 2- to 3-fold improvement in survival times of the treated mice. The NPs with or without mannan decoration elicited stronger responses in terms of lymphocyte proliferation, delayed-type hypersensitivity and CD107a expression. Moreover, our data indicated that the production of IFN-γ and IL-2 increased while the production of IL-4 and IL-10 decreased in splenocytes culture supernatants. In the pathological evaluations, we found that necrosis and immune cells infiltration rate in the tumor tissue of the treated mice was elevated, while tumor cellularity and lung metastases significantly decreased in particular in the group that received (TCL-Poly I:C) NP-MN. Altogether, our findings suggested that the mannan-decorated PLGA NPs antigen delivery system had significant anti-tumor effects against the murine model of breast cancer and it could be considered as a step forward to human breast cancer immunotherapy.
Collapse
|
14
|
Pappalardo JS, Salmaso S, Levchenko TS, Mastrotto F, Bersani S, Langellotti CA, Vermeulen M, Ghersa F, Quattrocchi V, Zamorano PI, Hartner WC, Toniutti M, Musacchio T, Torchilin VP. Characterization of a Nanovaccine Platform Based on an α1,2-Mannobiose Derivative Shows Species-non-specific Targeting to Human, Bovine, Mouse, and Teleost Fish Dendritic Cells. Mol Pharm 2021; 18:2540-2555. [PMID: 34106726 DOI: 10.1021/acs.molpharmaceut.1c00048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Dendritic cells serve as the main immune cells that trigger the immune response. We developed a simple and cost-effective nanovaccine platform based on the α1',2-mannobiose derivative for dendritic cell targeting. In previous work, we have formulated the α1,2-mannobiose-based nanovaccine platform with plasmid DNA and tested it in cattle against BoHV-1 infection. There, we have shown that the dendritic cell targeting using this nanovaccine platform in vivo can boost the immunogenicity, resulting in a long-lasting immunity. In this work, we aim to characterize the α1',2-mannobiose derivative, which is key in the nanovaccine platform. This DC-targeting strategy takes advantage of the specific receptor known as DC-SIGN and exploits its capacity to bind α1,2-mannobiose that is present at terminal ends of oligosaccharides in certain viruses, bacteria, and other pathogens. The oxidative conjugation of α1',2-mannobiose to NH2-PEG2kDa-DSPE allowed us to preserve the chemical structure of the non-reducing mannose of the disaccharide and the OH groups and the stereochemistry of all carbons of the reducing mannose involved in the binding to DC-SIGN. Here, we show specific targeting to DC-SIGN of decorated micelles incubated with the Raji/DC-SIGN cell line and uptake of targeted liposomes that took place in human, bovine, mouse, and teleost fish DCs in vitro, by flow cytometry. Specific targeting was found in all cultures, demonstrating a species-non-specific avidity for this ligand, which opens up the possibility of using this nanoplatform to develop new vaccines for various species, including humans.
Collapse
Affiliation(s)
- Juan Sebastian Pappalardo
- Veterinary Nanomedicine Group, Instituto de Investigaciones Forestales y Agropecuarias Bariloche (IFAB, INTA-CONICET), EEA Bariloche, Instituto Nacional de Tecnología Agropecuaria, Bote Modesta Victoria 4450, San Carlos de Bariloche, Río Negro R8403DVZ, Argentina.,Immunology and Immunomodulators Group, Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), IV, Instituto Nacional de Tecnología Agropecuaria, Nicolás Repetto 2799, William Morris, Buenos Aires B1681FUU, Argentina.,Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padova, Via F. Marzolo, 5, Padova 35121, Padova, Italy
| | - Tatyana S Levchenko
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padova, Via F. Marzolo, 5, Padova 35121, Padova, Italy
| | - Sara Bersani
- Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padova, Via F. Marzolo, 5, Padova 35121, Padova, Italy
| | - Cecilia A Langellotti
- Immunology and Immunomodulators Group, Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), IV, Instituto Nacional de Tecnología Agropecuaria, Nicolás Repetto 2799, William Morris, Buenos Aires B1681FUU, Argentina.,National Council of Scientific and Technical Research (CONICET), Avenida Rivadavia 1917, Ciudad de Buenos Aires C1033AAJ, Argentina
| | - Monica Vermeulen
- National Council of Scientific and Technical Research (CONICET), Avenida Rivadavia 1917, Ciudad de Buenos Aires C1033AAJ, Argentina.,Institute of Experimental Medicine (IMEX, ANM-CONICET), Academia Nacional de Medicina, Pacheco de Melo 3081, Ciudad de Buenos Aires C1425AUM, Argentina
| | - Federica Ghersa
- Veterinary Nanomedicine Group, Instituto de Investigaciones Forestales y Agropecuarias Bariloche (IFAB, INTA-CONICET), EEA Bariloche, Instituto Nacional de Tecnología Agropecuaria, Bote Modesta Victoria 4450, San Carlos de Bariloche, Río Negro R8403DVZ, Argentina.,Parasitology Laboratory, Instituto de Investigaciones en Biodiversidad y Medioambiente (INIBIOMA, UNCo-CONICET) Universidad Nacional del Comahue, Quintral 1250, San Carlos de Bariloche, Río Negro R8400FRF, Argentina
| | - Valeria Quattrocchi
- Immunology and Immunomodulators Group, Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), IV, Instituto Nacional de Tecnología Agropecuaria, Nicolás Repetto 2799, William Morris, Buenos Aires B1681FUU, Argentina
| | - Patricia I Zamorano
- Immunology and Immunomodulators Group, Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), IV, Instituto Nacional de Tecnología Agropecuaria, Nicolás Repetto 2799, William Morris, Buenos Aires B1681FUU, Argentina.,National Council of Scientific and Technical Research (CONICET), Avenida Rivadavia 1917, Ciudad de Buenos Aires C1033AAJ, Argentina
| | - William C Hartner
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Micaela Toniutti
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Tiziana Musacchio
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
15
|
Shi W, Qiu Q, Feng Z, Tong Z, Guo W, Zou F, Yue N, Huang W, Qian H. Design, synthesis and immunological evaluation of self-assembled antigenic peptides from dual-antigen targets: a broad-spectrum candidate for an effective antibreast cancer therapy. J Immunother Cancer 2021; 9:jitc-2021-002523. [PMID: 34083420 PMCID: PMC8183215 DOI: 10.1136/jitc-2021-002523] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2021] [Indexed: 12/11/2022] Open
Abstract
Background Considering the narrow immune response spectrum of a single epitope, and the nanoparticles (NPs) as a novel adjuvant can achieve efficient delivery of antigenic peptides safely, a nano-system (denoted as DSPE-PEG-Man@EM-NPs) based on cathepsin B-responsive antigenic peptides was designed and synthesized. Methods Highly affinitive antigenic peptides were delivered by self-assembled NPs, and targeted erythrocyte membranes acted as a peptide carrier to improve antigenic peptides presentation and to strengthen cytotoxic T-cells reaction. Cathepsin B coupling could release antigenic peptides rapidly in dendritic cells. Results Evaluations showed that DSPE-PEG-Man@EM-NPs had obvious inhibitory effects towards both MCF-7 and MDA-MB-231 human breast cancer cell lines. Conclusion Overall, this strategy provides a novel strategy for boosting cytotoxic T lymphocytes response, thereby expanding the adaptation range of tumor antigenic peptides and improving the therapeutic effect of tumor immunotherapy with nanomedicine.
Collapse
Affiliation(s)
- Wei Shi
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Qianqian Qiu
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.,School of Pharmacy, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng Teachers' University, Yancheng 224002, China
| | - Ziying Feng
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Zhenzhen Tong
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Weiwei Guo
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Feng Zou
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Na Yue
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Wenlong Huang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, Jiangsu, China
| | - Hai Qian
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China .,Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, Jiangsu, China
| |
Collapse
|
16
|
Goswami R, O’Hagan DT, Adamo R, Baudner BC. Conjugation of Mannans to Enhance the Potency of Liposome Nanoparticles for the Delivery of RNA Vaccines. Pharmaceutics 2021; 13:pharmaceutics13020240. [PMID: 33572332 PMCID: PMC7916126 DOI: 10.3390/pharmaceutics13020240] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/28/2022] Open
Abstract
Recent approval of mRNA vaccines to combat COVID-19 have highlighted the potential of this platform. Lipid nanoparticles (LNP) is the delivery vehicle of choice for mRNA as they prevent its enzymatic degradation by encapsulation. We have recently shown that surface exposition of mannose, incorporated in LNPs as stable cholesterol-amine conjugate, enhances the potency of self-amplifying RNA (SAM) replicon vaccines through augmented uptake by antigen presenting cells (APCs). Here, we generated a new set of LNPs whose surface was modified with mannans of different length (from mono to tetrasaccharide), in order to study the effect on antibody response of model SAM replicon encoding for the respiratory syncytial virus fusion F protein. Furthermore, the impact of the mannosylated liposomal delivery through intradermal as well as intramuscular routes was investigated. The vaccine priming response showed to improve consistently with increase in the chain length of mannoses; however, the booster dose response plateaued above the length of disaccharide. An increase in levels of IgG1 and IgG2a was observed for mannnosylated lipid nanoparticles (MLNPs) as compared to LNPs. This work confirms the potential of mannosylated SAM LNPs for both intramuscular and intradermal delivery, and highlights a disaccharide length as sufficient to ensure improved immunogenicity compared to the un-glycosylated delivery system.
Collapse
Affiliation(s)
- Roshan Goswami
- mAbxience, Julia Morros s/n, Armunia, 24009 León, Spain;
- GSK, Via Fiorentina 1, 53100 Siena, Italy
| | | | - Roberto Adamo
- GSK, Via Fiorentina 1, 53100 Siena, Italy
- Correspondence: (R.A.); (B.C.B.)
| | - Barbara C. Baudner
- GSK, Via Fiorentina 1, 53100 Siena, Italy
- Correspondence: (R.A.); (B.C.B.)
| |
Collapse
|
17
|
Cifuentes-Rius A, Desai A, Yuen D, Johnston APR, Voelcker NH. Inducing immune tolerance with dendritic cell-targeting nanomedicines. NATURE NANOTECHNOLOGY 2021; 16:37-46. [PMID: 33349685 DOI: 10.1038/s41565-020-00810-2] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/29/2020] [Indexed: 04/14/2023]
Abstract
Induced tolerogenic dendritic cells are a powerful immunotherapy for autoimmune disease that have shown promise in laboratory models of disease and early clinical trials. In contrast to conventional immunosuppressive treatments, tolerogenic immunotherapy leverages the cells and function of the immune system to quell the autoreactive lymphocytes responsible for damage and disease. The principle techniques of isolating and reprogramming dendritic cells (DCs), central to this approach, are well characterized. However, the broader application of this technology is limited by its high cost and bespoke nature. Nanomedicine offers an alternative route by performing this reprogramming process in situ. Here, we review the challenges and opportunities in using nanoparticles as a delivery mechanism to target DCs and induce immunomodulation, emphasizing their versatility. We then highlight their potential to solve critical problems in organ transplantation and increasingly prevalent autoimmune disorders such as type 1 diabetes mellitus and multiple sclerosis, where new immunotherapy approaches have begun to show promise.
Collapse
Affiliation(s)
- Anna Cifuentes-Rius
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, Victoria, Australia.
| | - Anal Desai
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, Victoria, Australia
| | - Daniel Yuen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, Victoria, Australia
| | - Angus P R Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, Victoria, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, Parkville, Victoria, Australia.
- CSIRO Manufacturing, Bayview Avenue, Clayton, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia.
| |
Collapse
|
18
|
Manček-Keber M, Ribić R, Chain F, Sinnaeve D, Martins JC, Jerala R, Tomić S, Fehér K. Adamantane Containing Peptidoglycan Fragments Enhance RANTES and IL-6 Production in Lipopolysaccharide-Induced Macrophages. Molecules 2020; 25:molecules25163707. [PMID: 32823878 PMCID: PMC7465286 DOI: 10.3390/molecules25163707] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 07/08/2020] [Accepted: 08/06/2020] [Indexed: 11/24/2022] Open
Abstract
We report the enhancement of the lipopolysaccharide-induced immune response by adamantane containing peptidoglycan fragments in vitro. The immune stimulation was detected by Il-6 (interleukine 6) and RANTES (regulated on activation, normal T cell expressed and secreted) chemokine expression using cell assays on immortalized mouse bone-marrow derived macrophages. The most active compound was a α-D-mannosyl derivative of an adamantylated tripeptide with L-chirality at the adamantyl group attachment, whereby the mannose moiety assumed to target mannose receptors expressed on macrophage cell surfaces. The immune co-stimulatory effect was also influenced by the configuration of the adamantyl center, revealing the importance of specific molecular recognition event taking place with its receptor. The immunostimulating activities of these compounds were further enhanced upon their incorporation into lipid bilayers, which is likely related to the presence of the adamantyl group that helps anchor the peptidoglycan fragment into lipid nanoparticles. We concluded that the proposed adamantane containing peptidoglycan fragments act as co-stimulatory agents and are also suitable for the preparation of lipid nanoparticle-based delivery of peptidoglycan fragments.
Collapse
Affiliation(s)
- Mateja Manček-Keber
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, P.O. Box 660, SI-1001 Ljubljana, Slovenia; (M.M.-K.); (R.J.)
- Centre of Excelence EN-FIST, SI-1000 Ljubljana, Slovenia
| | - Rosana Ribić
- University Center Varaždin, University North, Jurja Križanića 31b, HR-42 000 Varaždin, Croatia;
| | - Fernando Chain
- Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, 9000 Ghent, Belgium; (F.C.); (D.S.); (J.C.M.)
| | - Davy Sinnaeve
- Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, 9000 Ghent, Belgium; (F.C.); (D.S.); (J.C.M.)
- Univ. Lille, Inserm, Institut Pasteur de Lille, CHU Lille, U1167—Labex DISTALZ—RID-AGE—Risk Factors and Molecular Determinants of Aging-Related Diseases, F-59000 Lille, France
- CNRS, ERL9002—Integrative Structural Biology, F-59000 Lille, France
| | - José C. Martins
- Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, 9000 Ghent, Belgium; (F.C.); (D.S.); (J.C.M.)
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, P.O. Box 660, SI-1001 Ljubljana, Slovenia; (M.M.-K.); (R.J.)
- Centre of Excelence EN-FIST, SI-1000 Ljubljana, Slovenia
| | - Srđanka Tomić
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102A, HR-10 000 Zagreb, Croatia;
| | - Krisztina Fehér
- Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
- Molecular Recognition and Interaction Research Group, Hungarian Academy of Sciences, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- Correspondence: or ; Tel.: +36-52-512-900; Fax: +36-52-518-660
| |
Collapse
|
19
|
Ribić R, Manček-Keber M, Chain F, Sinnaeve D, Martins JC, Jerala R, Tomić S, Fehér K. Targeted Delivery of Adamantylated Peptidoglycan Immunomodulators in Lipid Nanocarriers: NMR Shows That Cargo Fragments Are Available on the Surface. J Phys Chem B 2020; 124:4132-4145. [PMID: 32283934 DOI: 10.1021/acs.jpcb.0c00029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We present an in-depth investigation of the membrane interactions of peptidoglycan (PGN)-based immune adjuvants designed for lipid-based delivery systems using NMR spectroscopy. The derivatives contain a cargo peptidoglycan (PGN) dipeptide fragment and an adamantyl group, which serves as an anchor to the lipid bilayer. Furthermore, derivatives with a mannose group that can actively target cell surface receptors on immune cells are also studied. We showed that the targeting mannose group and the cargo PGN fragment are both available on the lipid bilayer surface, thereby enabling interactions with cognate receptors. We found that the nonmannosylated compounds are incorporated stronger into the lipid assemblies than the mannosylated ones, but the latter compounds penetrate deeper in the bilayer. This might be explained by stronger electrostatic interactions available for zwitterionic nonmannosylated derivatives as opposed to the compounds in which the charged N-terminus is capped by mannose groups. The higher incorporation efficiency of the nonmannosylated compounds correlated with a larger relative enhancement in immune stimulation activities upon lipid incorporation compared to that of the derivatives with the mannose group. The chirality of the adamantyl group also influenced the incorporation efficiency, which in turn correlated with membrane-associated conformations that affect possible intermolecular interactions with lipid molecules. These findings will help in improving the development of PGN-based immune adjuvants suitable for delivery in lipid nanoparticles.
Collapse
Affiliation(s)
- Rosana Ribić
- University Center Varaždin, University North, Jurja Križanića 31b, HR-42 000 Varaždin, Croatia.,Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
| | - Mateja Manček-Keber
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, P.O. Box 660, SI-1001 Ljubljana, Slovenia.,Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
| | - Fernando Chain
- Department of Organic and Macromolecular Chemistry, Ghent University, Campus Sterre S4, Krijgslaan 281, 9000 Ghent, Belgium
| | - Davy Sinnaeve
- Department of Organic and Macromolecular Chemistry, Ghent University, Campus Sterre S4, Krijgslaan 281, 9000 Ghent, Belgium.,Univ. Lille, Inserm, Institut Pasteur de Lille, CHU Lille, U1167 - Labex DISTALZ - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, F-59000 Lille, France.,CNRS, ERL9002 - Integrative Structural Biology, F-59000 Lille, France
| | - José C Martins
- Department of Organic and Macromolecular Chemistry, Ghent University, Campus Sterre S4, Krijgslaan 281, 9000 Ghent, Belgium
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, P.O. Box 660, SI-1001 Ljubljana, Slovenia
| | - Srđanka Tomić
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102A, HR-10 000 Zagreb, Croatia
| | - Krisztina Fehér
- Department of Organic and Macromolecular Chemistry, Ghent University, Campus Sterre S4, Krijgslaan 281, 9000 Ghent, Belgium.,Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany.,Molecular Recognition and Interaction Research Group, Hungarian Academy of Sciences, Egyetem tér 1, H-4032 Debrecen, Hungary
| |
Collapse
|
20
|
Hogervorst TP, Li RJE, Marino L, Bruijns SCM, Meeuwenoord NJ, Filippov DV, Overkleeft HS, van der Marel GA, van Vliet SJ, van Kooyk Y, Codée JDC. C-Mannosyl Lysine for Solid Phase Assembly of Mannosylated Peptide Conjugate Cancer Vaccines. ACS Chem Biol 2020; 15:728-739. [PMID: 32045202 PMCID: PMC7091534 DOI: 10.1021/acschembio.9b00987] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Dendritic
cells (DCs) are armed with a multitude of Pattern Recognition
Receptors (PRRs) to recognize pathogens and initiate pathogen-tailored
T cell responses. In these responses, the maturation of DCs is key,
as well as the production of cytokines that help to accomplish T cell
responses. DC-SIGN is a frequently exploited PRR that can effectively
be targeted with mannosylated antigens to enhance the induction of
antigen-specific T cells. The natural O-mannosidic
linkage is susceptible to enzymatic degradation, and its chemical
sensitivity complicates the synthesis of mannosylated antigens. For
this reason, (oligo)mannosides are generally introduced in a late
stage of the antigen synthesis, requiring orthogonal conjugation handles
for their attachment. To increase the stability of the mannosides
and streamline the synthesis of mannosylated peptide antigens, we
here describe the development of an acid-stable C-mannosyl lysine, which allows for the inline introduction of mannosides
during solid-phase peptide synthesis (SPPS). The developed amino acid
has been successfully used for the assembly of both small ligands
and peptide antigen conjugates comprising an epitope of the gp100
melanoma-associated antigen and a TLR7 agonist for DC activation.
The ligands showed similar internalization capacities and binding
affinities as the O-mannosyl analogs. Moreover, the
antigen conjugates were capable of inducing maturation, stimulating
the secretion of pro-inflammatory cytokines, and providing enhanced
gp100 presentation to CD8+ and CD4+ T cells,
similar to their O-mannosyl counterparts. Our results
demonstrate that the C-mannose lysine is a valuable
building block for the generation of anticancer peptide-conjugate
vaccine modalities.
Collapse
Affiliation(s)
- Tim P. Hogervorst
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - R. J. Eveline Li
- Amsterdam UMC-Location Vrije Universiteit Amsterdam, Deptartment of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Laura Marino
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Sven C. M. Bruijns
- Amsterdam UMC-Location Vrije Universiteit Amsterdam, Deptartment of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Nico J. Meeuwenoord
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Dmitri V. Filippov
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Herman S. Overkleeft
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Gijsbert A. van der Marel
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Sandra J. van Vliet
- Amsterdam UMC-Location Vrije Universiteit Amsterdam, Deptartment of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Amsterdam UMC-Location Vrije Universiteit Amsterdam, Deptartment of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Jeroen D. C. Codée
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| |
Collapse
|
21
|
Wagener K, Bros M, Krumb M, Langhanki J, Pektor S, Worm M, Schinnerer M, Montermann E, Miederer M, Frey H, Opatz T, Rösch F. Targeting of Immune Cells with Trimannosylated Liposomes. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900185] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Karolin Wagener
- Institute of Nuclear Chemistry Johannes Gutenberg University Fritz‐Strassmann‐Weg 2 Mainz 55128 Germany
| | - Matthias Bros
- Department of DermatologyUniversity Medical Center Langenbeckstraße 1 Mainz 55101 Germany
| | - Matthias Krumb
- Department of ChemistryJohannes Gutenberg University Duesbergweg 10–14 Mainz 55128 Germany
| | - Jens Langhanki
- Department of ChemistryJohannes Gutenberg University Duesbergweg 10–14 Mainz 55128 Germany
| | - Stefanie Pektor
- Clinic and Polyclinic of Nuclear MedicineUniversity Medical Center Langenbeckstraße 1 Mainz 55101 Germany
| | - Matthias Worm
- Department of ChemistryJohannes Gutenberg University Duesbergweg 10–14 Mainz 55128 Germany
| | - Meike Schinnerer
- Department of ChemistryJohannes Gutenberg University Duesbergweg 10–14 Mainz 55128 Germany
- Institute of Physical ChemistryJohannes Gutenberg University Jakob‐Welder‐Weg 11 Mainz 55128 Germany
| | - Evelyn Montermann
- Department of DermatologyUniversity Medical Center Langenbeckstraße 1 Mainz 55101 Germany
| | - Matthias Miederer
- Clinic and Polyclinic of Nuclear MedicineUniversity Medical Center Langenbeckstraße 1 Mainz 55101 Germany
| | - Holger Frey
- Department of ChemistryJohannes Gutenberg University Duesbergweg 10–14 Mainz 55128 Germany
| | - Till Opatz
- Department of ChemistryJohannes Gutenberg University Duesbergweg 10–14 Mainz 55128 Germany
| | - Frank Rösch
- Institute of Nuclear Chemistry Johannes Gutenberg University Fritz‐Strassmann‐Weg 2 Mainz 55128 Germany
| |
Collapse
|
22
|
Busold S, Nagy NA, Tas SW, van Ree R, de Jong EC, Geijtenbeek TBH. Various Tastes of Sugar: The Potential of Glycosylation in Targeting and Modulating Human Immunity via C-Type Lectin Receptors. Front Immunol 2020; 11:134. [PMID: 32117281 PMCID: PMC7019010 DOI: 10.3389/fimmu.2020.00134] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/20/2020] [Indexed: 12/31/2022] Open
Abstract
C-type lectin receptors (CLRs) are important in several immune regulatory processes. These receptors recognize glycans expressed by host cells or by pathogens. Whereas pathogens are recognized through their glycans, which leads to protective immunity, aberrant cellular glycans are now increasingly recognized as disease-driving factors in cancer, auto-immunity, and allergy. The vast variety of glycan structures translates into a wide spectrum of effects on the immune system ranging from immune suppression to hyper-inflammatory responses. CLRs have distinct expression patterns on antigen presenting cells (APCs) controlling their role in immunity. CLRs can also be exploited to selectively target specific APCs, modulate immune responses and enhance antigen presentation. Here we will discuss the role of glycans and their receptors in immunity as well as potential strategies for immune modulation. A special focus will be given to different dendritic cell subsets as these APCs are crucial orchestrators of immune responses in infections, cancer, auto-immunity and allergies. Furthermore, we will highlight the potential use of nanoscale lipid bi-layer structures (liposomes) in targeted immunotherapy.
Collapse
Affiliation(s)
- Stefanie Busold
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Noémi A Nagy
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W Tas
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Centers, Amsterdam Rheumatology and Immunology Center, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald van Ree
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Department of Otorhinolaryngology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Esther C de Jong
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
23
|
Shahzad KA, Naeem M, Zhang L, Wan X, Song S, Pei W, Zhao C, Jin X, Shen C. Design and Optimization of PLGA Particles to Deliver Immunomodulatory Drugs for the Prevention of Skin Allograft Rejection. Immunol Invest 2019; 49:840-857. [PMID: 31809611 DOI: 10.1080/08820139.2019.1695134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Background: Recent advancements in therapeutic strategies have attracted considerable attention to control the acute organs and tissues rejection, which is the main cause of mortality in transplant recipients. The long-term usage of immunosuppressive drugs compromises the body immunity against simple infections and decrease the patients' quality of life. Tolerance of allograft in recipients without harming the rest of host immune system is the basic idea to develop the therapeutic approaches after induction of donor-specific transplant. Methods: Controlled and targeted delivery system by using biomimetic micro and nanoparticles as carriers is an effective strategy to deplete the immune cells in response to allograft in an antigen-specific manner. Polylactic-co-glycolic acid (PLGA) is a biocompatible and biodegradable polymer, which has frequently being used as drug delivery vehicle. Results: This review focuses on the biomedical applications of PLGA based biomimetic micro and nano-sized particles in drug delivery systems to prolong the survival of alloskin graft. Conclusion: We will discuss the mediating factors for rejection of alloskin graft, selective depletion of immune cells, controlled release mechanism, physiochemical properties, size-based body distribution of PLGA particles and their effect on overall host immune system.
Collapse
Affiliation(s)
- Khawar Ali Shahzad
- Department of Microbiology and Immunology, Medical School, Southeast University , Nanjing, Jiangsu, China.,School of Pharmacy, Taizhou Polytechnic College , Taizhou, Jiangsu, China
| | - Muhammad Naeem
- Institute of Pure and Applied Biology, Zoology Division, Bahauddin Zakariya University , Multan, Pakistan
| | - Lei Zhang
- Department of Microbiology and Immunology, Medical School, Southeast University , Nanjing, Jiangsu, China.,Department of Clinical Laboratory, Lishui District People's Hospital of Nanjing , Nanjing, Jiangsu, China
| | - Xin Wan
- Department of Microbiology and Immunology, Medical School, Southeast University , Nanjing, Jiangsu, China
| | - Shilong Song
- Department of Microbiology and Immunology, Medical School, Southeast University , Nanjing, Jiangsu, China
| | - Weiya Pei
- Department of Microbiology and Immunology, Medical School, Southeast University , Nanjing, Jiangsu, China
| | - Chen Zhao
- Department of Microbiology and Immunology, Medical School, Southeast University , Nanjing, Jiangsu, China
| | - Xiaoxiao Jin
- Department of Microbiology and Immunology, Medical School, Southeast University , Nanjing, Jiangsu, China
| | - Chuanlai Shen
- Department of Microbiology and Immunology, Medical School, Southeast University , Nanjing, Jiangsu, China
| |
Collapse
|
24
|
Goswami R, Chatzikleanthous D, Lou G, Giusti F, Bonci A, Taccone M, Brazzoli M, Gallorini S, Ferlenghi I, Berti F, O’Hagan DT, Pergola C, Baudner BC, Adamo R. Mannosylation of LNP Results in Improved Potency for Self-Amplifying RNA (SAM) Vaccines. ACS Infect Dis 2019; 5:1546-1558. [PMID: 31290323 DOI: 10.1021/acsinfecdis.9b00084] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Mannosylation of Lipid Nanoparticles (LNP) can potentially enhance uptake by Antigen Presenting Cells, which are highly abundant in dermal tissues, to improve the potency of Self Amplifying mRNA (SAM) vaccines in comparison to the established unmodified LNP delivery system. In the current studies, we evaluated mannosylated LNP (MLNP), which were obtained by incorporation of a stable Mannose-cholesterol amine conjugate, for the delivery of an influenza (hemagglutinin) encoded SAM vaccine in mice, by both intramuscular and intradermal routes of administration. SAM MLNP exhibited in vitro enhanced uptake in comparison to unglycosylated LNP from bone marrow-derived dendritic cells, and in vivo more rapid onset of the antibody response, independent of the route. The increased binding antibody levels also translated into higher functional hemagglutinin inhibition titers, particularly following intradermal administration. T cell assay on splenocytes from immunized mice also showed an increase in antigen specific CD8+ T responses, following intradermal administration of MLNP SAM vaccines. Induction of enhanced antigen specific CD4+ T cells, correlating with higher IgG2a antibody responses, was also observed. Hence, the present work illustrates the benefit of mannosylation of LNPs to achieve a faster immune response with SAM vaccines and these observations could contribute to the development of novel skin delivery systems for SAM vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Derek T. O’Hagan
- GSK, 14200 Shady Grove Road, Rockville, Maryland 20850, United States
| | | | | | | |
Collapse
|
25
|
Ribić R, Stojković R, Milković L, Antica M, Cigler M, Tomić S. Design, synthesis and biological evaluation of immunostimulating mannosylated desmuramyl peptides. Beilstein J Org Chem 2019; 15:1805-1814. [PMID: 31467600 PMCID: PMC6693374 DOI: 10.3762/bjoc.15.174] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/11/2019] [Indexed: 12/28/2022] Open
Abstract
Muramyl dipeptide is the minimal structure of peptidoglycan with adjuvant properties. Replacement of the N-acetylmuramyl moiety and increase of lipophilicity are important approaches in the preparation of muramyl dipeptide analogues with improved pharmacological properties. Mannose receptors present on immunocompetent cells are pattern-recognition receptors and by mannose ligands binding they affect the immune system. Here we present the design, synthesis and biological evaluation of novel mannosylated desmuramyl peptide derivatives. Mannose was coupled to dipeptides containing a lipophilic adamantane on N- or C-terminus through a glycolyl or hydroxyisobutyryl linker. Adjuvant activities of synthesized compounds were investigated in the mouse model using ovalbumin as an antigen. Their activities were compared to the previously described mannosylated adamantane-containing desmuramyl peptide and peptidoglycan monomer. Tested compounds exhibited adjuvant activity and the strongest enhancement of IgG production was stimulated by compound 21 (Man-OCH2-ᴅ-(1-Ad)Gly-ʟ-Ala-ᴅ-isoGln).
Collapse
Affiliation(s)
- Rosana Ribić
- University Center Varaždin, University North, Jurja Križanića 31b, HR-42000 Varaždin, Croatia
| | - Ranko Stojković
- Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia
| | - Lidija Milković
- Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia
| | | | - Marko Cigler
- Department of Chemistry, Technical University Munich, Lichtenbergstraße 4, D-85748 Garching, Germany
| | - Srđanka Tomić
- Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia
| |
Collapse
|
26
|
Bartheldyová E, Turánek Knotigová P, Zachová K, Mašek J, Kulich P, Effenberg R, Zyka D, Hubatka F, Kotouček J, Čelechovská H, Héžová R, Tomečková A, Mašková E, Fojtíková M, Macaulay S, Bystrický P, Paulovičová L, Paulovičová E, Drož L, Ledvina M, Raška M, Turánek J. N-Oxy lipid-based click chemistry for orthogonal coupling of mannan onto nanoliposomes prepared by microfluidic mixing: Synthesis of lipids, characterisation of mannan-coated nanoliposomes and in vitro stimulation of dendritic cells. Carbohydr Polym 2018; 207:521-532. [PMID: 30600036 DOI: 10.1016/j.carbpol.2018.10.121] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/18/2018] [Accepted: 10/25/2018] [Indexed: 11/26/2022]
Abstract
New synthetic aminooxy lipid was designed and synthesized as a building block for the formulation of functionalised nanoliposomes (presenting onto the outer surface of aminooxy groups) by microfluidic mixing. Orthogonal binding of cellular mannan (Candida glabrata (CCY 26-20-1) onto the outer surface of functionalised nanoliposomes was modified by orthogonal binding of reducing termini of mannans to oxime lipids via a click chemistry reaction based on aminooxy coupling (oxime ligation). The aminooxy lipid was proved as a suitable active component for preparation of functionalised nanoliposomes by the microfluidic mixing method performed with the instrument NanoAssemblr™. This "on-chip technology" can be easily scaled-up. The structure of mannan-liposomes was visualized by transmission and scanning electron microscopy, including immunogold staining of recombinant mannan receptor bound onto mannosylated-liposomes. The observed structures are in a good correlation with data obtained by DLS, NTA, and TPRS methods. In vitro experiments on human and mouse dendritic cells demonstrate selective internalisation of fluorochrome-labelled mannan-liposomes and their ability to stimulate DC comparable to lipopolysaccharide. We describe a potentially new drug delivery platform for mannan receptor-targeted antimicrobial drugs as well as for immunotherapeutics. Furthermore, the platform based on mannans bound orthogonally onto the surface of nanoliposomes represents a self-adjuvanted carrier for construction of liposome-based recombinant vaccines for both systemic and mucosal routes of administration.
Collapse
Affiliation(s)
- Eliška Bartheldyová
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Pavlína Turánek Knotigová
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Kateřina Zachová
- Department of Immunology and Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hněvotínská 3, 775 15 Olomouc, Czech Republic
| | - Josef Mašek
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Pavel Kulich
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Roman Effenberg
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Technická 5, 166, 28 Prague 6, Czech Republic
| | - Daniel Zyka
- APIGENEX s.r.o., Poděbradská 173/5, Prague 9, 190 00, Czech Republic
| | - František Hubatka
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Jan Kotouček
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Hana Čelechovská
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Renata Héžová
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Andrea Tomečková
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Eliška Mašková
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | - Martina Fojtíková
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic
| | | | - Peter Bystrický
- Division of Neurosciences, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University, Malá Hora 10701/4A, 036 01 Martin, Slovakia
| | - Lucia Paulovičová
- Department of Immunochemistry of Glycoconjugates, Immunology & Cell Culture Laboratory, Institute of Chemistry, Center for Glycomics Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia
| | - Ema Paulovičová
- Department of Immunochemistry of Glycoconjugates, Immunology & Cell Culture Laboratory, Institute of Chemistry, Center for Glycomics Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia.
| | - Ladislav Drož
- APIGENEX s.r.o., Poděbradská 173/5, Prague 9, 190 00, Czech Republic
| | - Miroslav Ledvina
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Technická 5, 166, 28 Prague 6, Czech Republic.
| | - Milan Raška
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; Department of Immunology and Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hněvotínská 3, 775 15 Olomouc, Czech Republic.
| | - Jaroslav Turánek
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic.
| |
Collapse
|
27
|
Mukwaya V, Wang C, Dou H. Saccharide-based nanocarriers for targeted therapeutic and diagnostic applications. POLYM INT 2018. [DOI: 10.1002/pi.5702] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Vincent Mukwaya
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering; Shanghai Jiao Tong University; Shanghai PR China
| | - Chenglong Wang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering; Shanghai Jiao Tong University; Shanghai PR China
| | - Hongjing Dou
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering; Shanghai Jiao Tong University; Shanghai PR China
| |
Collapse
|
28
|
Ochando J, Braza MS. Nanoparticle-Based Modulation and Monitoring of Antigen-Presenting Cells in Organ Transplantation. Front Immunol 2017; 8:1888. [PMID: 29312352 PMCID: PMC5743935 DOI: 10.3389/fimmu.2017.01888] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/11/2017] [Indexed: 11/13/2022] Open
Abstract
Donor-specific unresponsiveness while preserving an intact immune function remains difficult to achieve in organ transplantation. Induction of tolerance requires a fine modulation of the interconnected innate and adaptive immune systems. Antigen-presenting cells (APCs) predominate during allograft rejection and create a highly inflammatory context where allospecific T cells are primed. Currently, the available protocols to prevent allograft rejection include a cocktail of drugs that are efficient in the short-term, but with severe long-term side effects and considerable toxicity. Consequently, better and less burdensome strategies are needed to promote indefinite allograft survival. Targeted delivery of immunosuppressive drugs that prevent the alloimmune response may address some of these problems. Nanoparticle-based approaches represent a promising strategy to negatively modulate the alloresponse by specifically delivering small compounds to APCs in vivo. Nanoparticles are also used as integrating imaging moieties to monitor inflammation for diagnostic purposes. Therefore, nanotechnology approaches represent an attractive strategy to deliver and monitor the efficacy of immunosuppressive therapy in organ transplantation with the potential to improve the clinical treatment of transplant patients.
Collapse
Affiliation(s)
- Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, Immunology Institute, New York, NY, United States
| | - Mounia S Braza
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, Immunology Institute, New York, NY, United States
| |
Collapse
|
29
|
De Serrano LO, Burkhart DJ. Liposomal vaccine formulations as prophylactic agents: design considerations for modern vaccines. J Nanobiotechnology 2017; 15:83. [PMID: 29149896 PMCID: PMC5693489 DOI: 10.1186/s12951-017-0319-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/09/2017] [Indexed: 01/04/2023] Open
Abstract
Vaccinology is one of the most important cornerstones in modern medicine, providing better quality of life. The human immune system is composed of innate and adaptive immune processes that interplay when infection occurs. Innate immunity relies on pathogen-associated molecular patterns which are recognized by pathogen recognition receptors localized in antigen presenting cells. After antigen processing and presentation, CD4+ T cell polarization occurs, further leading to B cell and CD8+ activation and humoral and cell-mediated adaptive immune responses. Liposomes are being employed as vaccine technologies and their design is of importance to ensure proper immune responses. Physicochemical parameters like liposome size, charge, lamellarity and bilayer fluidity must be completely understood to ensure optimal vaccine stability and efficacy. Liposomal vaccines can be developed to target specific immune cell types for the induction of certain immune responses. In this review, we will present promising liposomal vaccine approaches for the treatment of important viral, bacterial, fungal and parasitic infections (including tuberculosis, TB). Cationic liposomes are the most studied liposome types due to their enhanced interaction with the negatively charged immune cells. Thus, a special section on the cationic lipid dimethyldioctadecylammonium and TB is also presented.
Collapse
Affiliation(s)
- Luis O. De Serrano
- Department of Biomedical & Pharmaceutical Sciences and Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812 USA
| | - David J. Burkhart
- Department of Biomedical & Pharmaceutical Sciences and Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812 USA
| |
Collapse
|
30
|
Askarizadeh A, Jaafari MR, Khamesipour A, Badiee A. Liposomal adjuvant development for leishmaniasis vaccines. THERAPEUTIC ADVANCES IN VACCINES 2017; 5:85-101. [PMID: 29201374 PMCID: PMC5697592 DOI: 10.1177/2051013617741578] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/18/2017] [Indexed: 08/29/2023]
Abstract
Leishmaniasis is a parasitic disease that ranges in severity from skin lesions to fatality. Since long-lasting protection is induced upon recovery from cutaneous leishmaniasis, development of an effective vaccine is promising. However, there is no vaccine for use in humans yet. It seems limited efficacy in leishmaniasis vaccines is due to lack of an appropriate adjuvant or delivery system. Hence, the use of particulate adjuvants such as liposomes for effective delivery to the antigen presenting cells (APCs) is a valuable strategy to enhance leishmaniasis vaccine efficacy. The extraordinary versatility of liposomes because of their unique amphiphilic and biphasic nature allows for using antigens or immunostimulators within the core, on the surface or within the bilayer, and modulates both the magnitude and the T-helper bias of the immune response. In this review article, we attempt to summarize the role of liposomal adjuvants in the development of Leishmania vaccines and describe the main physicochemical properties of liposomes like phospholipid composition, surface charge, and particle size during formulation design. We also suggest potentially useful formulation strategies in order for future experiments to have a chance to succeed as liposomal vaccines against leishmaniasis.
Collapse
Affiliation(s)
- Anis Askarizadeh
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran Department of Pharmaceutical Nanotechnology, School of Pharmacy Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Khamesipour
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
31
|
Fagan V, Hussein WM, Su M, Giddam AK, Batzloff MR, Good MF, Toth I, Simerska P. Synthesis, Characterization and Immunological Evaluation of Self‐Adjuvanting Group A Streptococcal Vaccine Candidates Bearing Various Lipidic Adjuvanting Moieties. Chembiochem 2017; 18:545-553. [DOI: 10.1002/cbic.201600639] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Vincent Fagan
- School of Chemistry and Molecular Biosciences The University of Queensland Cooper Road St. Lucia QLD 4072 Australia
| | - Waleed M. Hussein
- School of Chemistry and Molecular Biosciences The University of Queensland Cooper Road St. Lucia QLD 4072 Australia
| | - Mei Su
- School of Chemistry and Molecular Biosciences The University of Queensland Cooper Road St. Lucia QLD 4072 Australia
| | - Ashwini K. Giddam
- School of Chemistry and Molecular Biosciences The University of Queensland Cooper Road St. Lucia QLD 4072 Australia
| | | | - Michael F. Good
- Institute for Glycomics Griffith University Gold Coast 4215 Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences The University of Queensland Cooper Road St. Lucia QLD 4072 Australia
- School of Pharmacy Pharmacy Australia Centre of Excellence The University of Queensland Cornwall Street Woolloongabba QLD 4072 Australia
- Institute for Molecular Bioscience The University of Queensland St. Lucia QLD 4072 Australia
| | - Pavla Simerska
- School of Chemistry and Molecular Biosciences The University of Queensland Cooper Road St. Lucia QLD 4072 Australia
| |
Collapse
|
32
|
Giddam AK, Reiman JM, Zaman M, Skwarczynski M, Toth I, Good MF. A semi-synthetic whole parasite vaccine designed to protect against blood stage malaria. Acta Biomater 2016; 44:295-303. [PMID: 27544810 DOI: 10.1016/j.actbio.2016.08.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/12/2016] [Accepted: 08/16/2016] [Indexed: 12/14/2022]
Abstract
UNLABELLED Although attenuated malaria parasitized red blood cells (pRBCs) are promising vaccine candidates, their application in humans may be restricted for ethical and regulatory reasons. Therefore, we developed an organic microparticle-based delivery platform as a whole parasite malaria-antigen carrier to mimic pRBCs. Killed blood stage parasites were encapsulated within liposomes that are targeted to antigen presenting cells (APCs). Mannosylated lipid core peptides (MLCPs) were used as targeting ligands for the liposome-encapsulated parasite antigens. MLCP-liposomes, but not unmannosylated liposomes, were taken-up efficiently by APCs which then significantly upregulated expression of MHC-ll and costimulatory molecules, CD80 and CD86. Two such vaccines using rodent model systems were constructed - one with Plasmodium chabaudi and the other with P. yoelii. MLCP-liposome vaccines were able to control the parasite burden and extended the survival of mice. Thus, we have demonstrated an alternative delivery system to attenuated pRBCs with similar vaccine efficacy and added clinical advantages. Such liposomes are promising candidates for a human malaria vaccine. STATEMENT OF SIGNIFICANCE Attenuated whole parasite-based vaccines, by incorporating all parasite antigens, are very promising candidates, but issues relating to production, storage and safety concerns are significantly slowing their development. We therefore developed a semi-synthetic whole parasite malaria vaccine that is easily manufactured and stored. Two such prototype vaccines (a P. chabaudi and a P. yoelii vaccine) have been constructed. They are non-infectious, highly immunogenic and give good protection profiles. This semi-synthetic delivery platform is an exciting strategy to accelerate the development of a licensed malaria vaccine. Moreover, this strategy can be potentially applied to a wide range of pathogens.
Collapse
|
33
|
Sedaghat B, Stephenson RJ, Giddam AK, Eskandari S, Apte SH, Pattinson DJ, Doolan DL, Toth I. Synthesis of Mannosylated Lipopeptides with Receptor Targeting Properties. Bioconjug Chem 2016; 27:533-48. [PMID: 26735314 DOI: 10.1021/acs.bioconjchem.5b00547] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Present on the surface of antigen presenting cells (APCs), the mannose receptor (MR) has long been recognized as a front-line receptor in pathogen recognition. During the past decade many attempts have been made to target this receptor for applications including vaccine and drug development. In the present study, a library of vaccine constructs comprising fluorescently labeled mannosylated lipid-dendrimers that contained the ovalbumin CD4(+) epitope, OVA(323-339), as the model peptide antigen were synthesized using fluorenylmethyloxycarbonyl (Fmoc) solid phase peptide synthesis (SPPS). The vaccine constructs were designed with an alanine spacer between the O-linked mannose moieties to investigate the impact of distance between the mannose units on receptor-mediated uptake and/or binding in APCs. Uptake studies performed on F4/80(+) and CD11c(+) cells showed significant uptake and/or binding for lipopeptides containing mannose, and also the lipopeptide without mannose when compared to the control peptides (peptide with no lipid and peptide with no mannose and no lipid). Furthermore, mannan inhibition assays demonstrated that uptake of the mannosylated and lipidated peptides was receptor mediated. To address the specificity of receptor uptake, surface plasmon resonance studies were performed using biacore technology and confirmed high affinity of the mannosylated and lipidated vaccine constructs toward the MR. These studies confirm that both mannose and lipid moieties play significant roles in receptor-mediated uptake on APCs, potentially facilitating vaccine development.
Collapse
Affiliation(s)
| | | | | | | | - Simon H Apte
- Infectious Diseases Program, QIMR Berghofer Medical Research Institute , Brisbane, Queensland 4029, Australia
| | - David J Pattinson
- Infectious Diseases Program, QIMR Berghofer Medical Research Institute , Brisbane, Queensland 4029, Australia
| | - Denise L Doolan
- Infectious Diseases Program, QIMR Berghofer Medical Research Institute , Brisbane, Queensland 4029, Australia
| | - Istvan Toth
- School of Pharmacy, The University of Queensland , Woolloongabba, Queensland 4012, Australia
| |
Collapse
|
34
|
Nguyen H, Katavic P, Bashah NAH, Ferro V. Synthesis of Mannose-Cholesterol Conjugates for Targeted Liposomal Drug Delivery. ChemistrySelect 2016. [DOI: 10.1002/slct.201600007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Huong Nguyen
- The University of Queensland; School of Chemistry and Molecular Biosciences; Brisbane QLD 4072 Australia
| | - Peter Katavic
- The University of Queensland; School of Chemistry and Molecular Biosciences; Brisbane QLD 4072 Australia
| | - Nur Atikah Halim Bashah
- The University of Queensland; School of Chemistry and Molecular Biosciences; Brisbane QLD 4072 Australia
| | - Vito Ferro
- The University of Queensland; School of Chemistry and Molecular Biosciences; Brisbane QLD 4072 Australia
| |
Collapse
|
35
|
Apte SH, Stephenson RJ, Simerska P, Groves PL, Aljohani S, Eskandari S, Toth I, Doolan DL. Systematic evaluation of self-adjuvanting lipopeptide nano-vaccine platforms for the induction of potent CD8(+) T-cell responses. Nanomedicine (Lond) 2015; 11:137-52. [PMID: 26653407 DOI: 10.2217/nnm.15.184] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIM Systematically evaluate lipid core peptide vaccine delivery platforms to identify core features promoting strong CD8(+) T-cell responses. MATERIALS & METHODS Three different self-adjuvanting lipid core peptide nanovaccines each comprising four copies of the dominant ovalbumin CD8(+) T-cell epitope and varying in the utilization of a polylysine or glucose core with 2-amino-hexadecanoic acid (C16) or 2-amino-dodecanoic acid (C12) lipids were synthesized. Vaccines were tested for ability to induce CD8(+) T-cell responses and inhibit tumor growth in vivo. RESULTS The construct utilizing C12 lipids and polylysine core induced very robust effector T cells shown to have in vivo effector capability as demonstrated by in vivo cytotoxicity and ability to inhibit tumor growth as well as modulation of dendritic cell activation. CONCLUSION The C12 polylysine platform was an effective configuration for induction of potent CD8(+) T-cell responses.
Collapse
Affiliation(s)
- Simon H Apte
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Rachel J Stephenson
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Pavla Simerska
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Penny L Groves
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Salwa Aljohani
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Sharareh Eskandari
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Istvan Toth
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia.,School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4012, Australia.,Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Denise L Doolan
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| |
Collapse
|
36
|
Markov OV, Mironova NL, Shmendel EV, Serikov RN, Morozova NG, Maslov MA, Vlassov VV, Zenkova MA. Multicomponent mannose-containing liposomes efficiently deliver RNA in murine immature dendritic cells and provide productive anti-tumour response in murine melanoma model. J Control Release 2015; 213:45-56. [DOI: 10.1016/j.jconrel.2015.06.028] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/15/2015] [Accepted: 06/21/2015] [Indexed: 12/21/2022]
|
37
|
Baio JE, Schach D, Fuchs AV, Schmüser L, Billecke N, Bubeck C, Landfester K, Bonn M, Bruns M, Weiss CK, Weidner T. Reversible activation of pH-sensitive cell penetrating peptides attached to gold surfaces. Chem Commun (Camb) 2015; 51:273-275. [PMID: 25329926 DOI: 10.1039/c4cc07278b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
pH-sensitive viral fusion protein mimics are widely touted as a promising route towards site-specific delivery of therapeutic compounds across lipid membranes. Here, we demonstrate that a fusion protein mimic, designed to achieve a reversible, pH-driven helix-coil transition mechanism, retains its functionality when covalently bound to a surface.
Collapse
Affiliation(s)
- Joe E Baio
- Max Planck Institute for Polymer Research, 55270 Mainz, Germany
| | - Denise Schach
- Max Planck Institute for Polymer Research, 55270 Mainz, Germany
| | - Adrian V Fuchs
- Max Planck Institute for Polymer Research, 55270 Mainz, Germany
| | - Lars Schmüser
- Max Planck Institute for Polymer Research, 55270 Mainz, Germany
| | - Nils Billecke
- Max Planck Institute for Polymer Research, 55270 Mainz, Germany
| | | | | | - Mischa Bonn
- Max Planck Institute for Polymer Research, 55270 Mainz, Germany
| | - Michael Bruns
- Karlsruhe Institute of Technology, Institute for Applied Materials and Karlsruhe Nano Micro Facility, 76344 Eggenstein-Leopoldshafen, Germany
| | - Clemens K Weiss
- Max Planck Institute for Polymer Research, 55270 Mainz, Germany.,University of Applies Sciences Bingen, 55411 Bingen, Germany
| | - Tobias Weidner
- Max Planck Institute for Polymer Research, 55270 Mainz, Germany
| |
Collapse
|
38
|
Hoang MD, Jung SH, Lee HJ, Lee YK, Nguyen-Pham TN, Choi NR, Vo MC, Lee SS, Ahn JS, Yang DH, Kim YK, Kim HJ, Lee JJ. Dendritic Cell-Based Cancer Immunotherapy against Multiple Myeloma: From Bench to Clinic. Chonnam Med J 2015; 51:1-7. [PMID: 25914874 PMCID: PMC4406989 DOI: 10.4068/cmj.2015.51.1.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 03/19/2015] [Accepted: 03/23/2015] [Indexed: 01/27/2023] Open
Abstract
Although the introduction of stem cell transplantation and novel agents has improved survival, multiple myeloma (MM) is still difficult to cure. Alternative approaches are clearly needed to prolong the survival of patients with MM. Dendritic cell (DC) therapy is a very promising tool immunologically in MM. We developed a method to generate potent DCs with increased Th1 polarization and migration ability for inducing strong myeloma-specific cytotoxic T lymphocytes. In this review, we discuss how the efficacy of cancer immunotherapy using DCs can be improved in MM.
Collapse
Affiliation(s)
- My-Dung Hoang
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Sung-Hoon Jung
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea. ; Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Hyun-Ju Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | | | - Thanh-Nhan Nguyen-Pham
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Nu-Ri Choi
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Manh-Cuong Vo
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Seung-Shin Lee
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Jae-Sook Ahn
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Deok-Hwan Yang
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Yeo-Kyeoung Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Korea. ; Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea. ; Vaxcell-Bio Therapeutics, Hwasun, Korea
| |
Collapse
|
39
|
Cellular and molecular targeting for nanotherapeutics in transplantation tolerance. Clin Immunol 2015; 160:14-23. [PMID: 25805659 DOI: 10.1016/j.clim.2015.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 02/23/2015] [Accepted: 03/03/2015] [Indexed: 11/21/2022]
Abstract
The induction of donor-specific tolerance to transplanted cells and organs, while preserving immune function as a whole, remains a highly sought after and elusive strategy for overcoming transplant rejection. Tolerance necessitates modulating a diverse array of cell types that recognize and respond to alloantigens, including antigen presenting cells and T lymphocytes. Nanotherapeutic strategies that employ cellular and biomaterial engineering represent an emerging technology geared towards the goal of inducing transplant tolerance. Nanocarriers offer a platform for delivering antigens of interest to specific cell types in order to achieve tolerogenic antigen presentation. Furthermore, the technologies also provide an opportunity for local immunomodulation at the graft site. Nanocarriers delivering a combination of antigens and immunomodulating agents, such as rapamycin, provide a unique technology platform with the potential to enhance outcomes for the induction of transplant tolerance.
Collapse
|
40
|
Liu TY, Hussein WM, Giddam AK, Jia Z, Reiman JM, Zaman M, McMillan NAJ, Good MF, Monteiro MJ, Toth I, Skwarczynski M. Polyacrylate-based delivery system for self-adjuvanting anticancer peptide vaccine. J Med Chem 2014; 58:888-96. [PMID: 25489968 DOI: 10.1021/jm501514h] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vaccination can provide a safe alternative to chemotherapy by using the body's natural defense mechanisms to create a potent immune response against tumor cells. Peptide-based therapeutic vaccines against human papillomavirus (HPV)-related cancers are usually designed to elicit cytotoxic T cell responses by targeting the HPV-16 E7 oncoprotein. However, peptides alone lack immunogenicity, and an additional adjuvant or external delivery system is required. In this study, we developed new polymer-peptide conjugates to create an efficient self-adjuvanting system for peptide-based therapeutic vaccines. These conjugates reduced tumor growth and eradicated E7-positive TC-1 tumors in mice after a "single shot" immunization, without the help from an external adjuvant. The new conjugates had a significantly higher anticancer efficacy than the antigen formulated with a commercial adjuvant. Furthermore, the polymer-peptide conjugates were promptly taken up by antigen presenting cells, including dendritic cells and macrophages, and efficiently activated CD4(+) T-helper cells and CD8(+) cytotoxic T lymphocyte cells.
Collapse
Affiliation(s)
- Tzu-Yu Liu
- School of Chemistry and Molecular Biosciences, ‡Australian Institute for Bioengineering and Nanotechnology, and §School of Pharmacy, The University of Queensland , Brisbane, QLD 4072, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Haddadi A, Hamdy S, Ghotbi Z, Samuel J, Lavasanifar A. Immunoadjuvant activity of the nanoparticles' surface modified with mannan. NANOTECHNOLOGY 2014; 25:355101. [PMID: 25119543 DOI: 10.1088/0957-4484/25/35/355101] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Mannan (MN) is the natural ligand for mannose receptors, which are widely expressed on dendritic cells (DCs). The purpose of this study was to assess the effect of formulation parameters on the immunogenicity of MN-decorated poly (D, L-lactide-co-glycolide) (PLGA) nanoparticles (NPs) in terms of their ability to stimulate DC phenotypic as well as functional maturation. For this purpose, NPs were formulated from either ester-terminated or COOH-terminated PLGA. Incorporation of MN in NPs was achieved through encapsulation, physical adsorption or chemical conjugation. Murine bone marrow derived DCs (BMDCs) were treated with various NP formulations and assessed for their ability to up-regulate DC cell surface markers, secrete immunostimulatory cytokines and to activate allogenic T cell responses. DCs treated with COOH-terminated PLGA-NPs containing chemically conjugated MN (MN-Cov-COOH) have shown superior performance in improving DC biological functions, compared to the rest of the formulations tested. This may be attributed to the higher level of MN incorporation in the former formulation. Incorporation of MN in PLGA NPs through chemical conjugation can lead to enhanced DC maturation and stimulatory function. This strategy may be used to develop more effective PLGA-based vaccine formulations.
Collapse
Affiliation(s)
- Azita Haddadi
- Division of Pharmacy, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | | | | | | | | |
Collapse
|
42
|
Glycosylation-mediated targeting of carriers. J Control Release 2014; 190:542-55. [DOI: 10.1016/j.jconrel.2014.06.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 05/29/2014] [Accepted: 06/02/2014] [Indexed: 12/24/2022]
|
43
|
Al-Barwani F, Young SL, Baird MA, Larsen DS, Ward VK. Mannosylation of virus-like particles enhances internalization by antigen presenting cells. PLoS One 2014; 9:e104523. [PMID: 25122183 PMCID: PMC4133192 DOI: 10.1371/journal.pone.0104523] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 07/11/2014] [Indexed: 12/05/2022] Open
Abstract
Internalization of peptides by antigen presenting cells is crucial for the initiation of the adaptive immune response. Mannosylation has been demonstrated to enhance antigen uptake through mannose receptors, leading to improved immune responses. In this study we test the effect of surface mannosylation of protein-based virus-like particles (VLP) derived from Rabbit hemorrhagic disease virus (RHDV) on uptake by murine and human antigen presenting cells. A monomannoside and a novel dimannoside were synthesized and successfully conjugated to RHDV VLP capsid protein, providing approximately 270 mannose groups on the surface of each virus particle. VLP conjugated to the mannoside or dimannoside exhibited significantly enhanced binding and internalization by murine dendritic cells, macrophages and B cells as well as human dendritic cells and macrophages. This uptake was inhibited by the inclusion of mannan as a specific inhibitor of mannose specific uptake, demonstrating that mannosylation of VLP targets mannose receptor-based uptake. Consistent with mannose receptor-based uptake, partial retargeting of the intracellular processing of RHDV VLP was observed, confirming that mannosylation of VLP provides both enhanced uptake and modified processing of associated antigens.
Collapse
Affiliation(s)
- Farah Al-Barwani
- Department of Microbiology and Immunology, Otago School of Medical Science, University of Otago, Dunedin, New Zealand
| | - Sarah L. Young
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Margaret A. Baird
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - David S. Larsen
- Department of Chemistry, Division of Sciences, University of Otago, Dunedin, New Zealand
| | - Vernon K. Ward
- Department of Microbiology and Immunology, Otago School of Medical Science, University of Otago, Dunedin, New Zealand
- * E-mail:
| |
Collapse
|
44
|
Hotaling NA, Cummings RD, Ratner DM, Babensee JE. Molecular factors in dendritic cell responses to adsorbed glycoconjugates. Biomaterials 2014; 35:5862-74. [PMID: 24746228 PMCID: PMC4127877 DOI: 10.1016/j.biomaterials.2014.03.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/18/2014] [Indexed: 11/23/2022]
Abstract
Carbohydrates and glycoconjugates have been shown to exert pro-inflammatory effects on the dendritic cells (DCs), supporting pathogen-induced innate immunity and antigen processing, as well as immunosuppressive effects in the tolerance to self-proteins. Additionally, the innate inflammatory response to implanted biomaterials has been hypothesized to be mediated by inflammatory cells interacting with adsorbed proteins, many of which are glycosylated. However, the molecular factors relevant for surface displayed glycoconjugate modulation of dendritic cell (DC) phenotype are unknown. Thus, in this study, a model system was developed to establish the role of glycan composition, density, and carrier cationization state on DC response. Thiol modified glycans were covalently bound to a model protein carrier, maleimide functionalized bovine serum albumin (BSA), and the number of glycans per BSA modulated. Additionally, the carrier isoelectric point was scaled from a pI of ∼4.0 to ∼10.0 using ethylenediamine (EDA). The DC response to the neoglycoconjugates adsorbed to wells of a 384-well plate was determined via a high throughput assay. The underlying trends in DC phenotype in relation to conjugate properties were elucidated via multivariate general linear models. It was found that glycoconjugates with more than 20 glycans per carrier had the greatest impact on the pro-inflammatory response from DCs, followed by conjugates having an isoelectric point above 9.5. Surfaces displaying terminal α1-2 linked mannose structures were able to increase the inflammatory DC response to a greater extent than did any other terminal glycan structure. The results herein can be applied to inform the design of the next generation of combination products and biomaterials for use in future vaccines and implanted materials.
Collapse
Affiliation(s)
- Nathan A Hotaling
- Wallace H. Coulter Dept. of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | | - Daniel M Ratner
- Dept. of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Julia E Babensee
- Wallace H. Coulter Dept. of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
45
|
Hubert M, Larsen DS, Hayman CM, Rades T, Hook S. Physical Characterization of Synthetic Phosphatidylinositol Dimannosides and Analogues in Binary Systems with Phosphatidylcholine. Mol Pharm 2014; 11:913-21. [DOI: 10.1021/mp400588y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | | | - Colin M. Hayman
- Carbohydrate
Chemistry Team, Callaghan Innovation, P.O. Box 31-310, Lower Hutt, New Zealand
| | - Thomas Rades
- Department
of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | | |
Collapse
|
46
|
Oligomannose-coated liposome as a novel adjuvant for the induction of cellular immune responses to control disease status. BIOMED RESEARCH INTERNATIONAL 2013; 2013:562924. [PMID: 24224170 PMCID: PMC3810488 DOI: 10.1155/2013/562924] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/03/2013] [Indexed: 01/28/2023]
Abstract
Professional phagocytic cells, such as dendritic cells, are mainly responsible for phagocytosis, antigen presentation, and cytokine secretion, which induce subsequent activation of T cell-mediated immunity. Thus, strategies that deliver antigens and stimulatory signals to the cells have significant implications for vaccine design. In this paper, we summarize the potential for liposomes coated with the neoglycolipids containing oligomannose residues (OMLs) as a novel adjuvant for induction of Th1 immune responses and CTLs specific for the encased antigen. OMLs preferentially take up peripheral phagocytic cells. In response to OML uptake, the cells secrete IL-12 selectively, enhance the expression of costimulatory molecules, and migrate into lymphoid tissues from peripheral tissues. OMLs also have the ability to deliver encapsulated protein antigens to the MHC class I and class II pathways to generate antigen-specific CTLs and Th1 cells, respectively, and lipid antigen to CD1d to activate NKT cells. Since administration of OML-based vaccines can eliminate an established tumor, inhibit elevation of the serum IgE level, and prevent progression of protozoan infections in several murine, human, and bovine models, OML-based vaccines have revealed their potential for clinical use in vaccination for a variety of diseases in which CTLs and/or Th1 cells act as effector cells.
Collapse
|
47
|
Hirsjärvi S, Dufort S, Bastiat G, Saulnier P, Passirani C, Coll JL, Benoît JP. Surface modification of lipid nanocapsules with polysaccharides: from physicochemical characteristics to in vivo aspects. Acta Biomater 2013; 9:6686-93. [PMID: 23395817 DOI: 10.1016/j.actbio.2013.01.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 01/27/2013] [Accepted: 01/30/2013] [Indexed: 11/26/2022]
Abstract
Attaching polysaccharides to the surface of nanoparticles offers the possibility of modifying the physicochemical and biological properties of the core particles. The surface of lipid nanocapsules (LNCs) was modified by post-insertion of amphiphilic lipochitosan (LC) or lipodextran (LD). Modelling of these LNCs by the drop tensiometer technique revealed that the positively charged LC made the LNC surface more rigid, whereas the neutral, higher M(W) LD had no effect on the surface elasticity. Both LNC-LC and LNC-LD activated the complement system more than the blank LNC, thus suggesting increased capture by the mononuclear phagocyte system. In vitro, the positively charged LNC-LC were more efficiently bound by the model HEK293(β3) cells compared to LNC and LNC-LD. Finally, it was observed that neither LC nor LD changed the in vivo biodistribution properties of LNCs in mice. These polysaccharide-coated LNCs, especially LNC-LC, are promising templates for targeting ligands (e.g. peptides, proteins) or therapeutic molecules (e.g. siRNA).
Collapse
|
48
|
Thomann-Harwood L, Kaeuper P, Rossi N, Milona P, Herrmann B, McCullough K. Nanogel vaccines targeting dendritic cells: Contributions of the surface decoration and vaccine cargo on cell targeting and activation. J Control Release 2013; 166:95-105. [DOI: 10.1016/j.jconrel.2012.11.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 11/23/2012] [Accepted: 11/25/2012] [Indexed: 10/27/2022]
|
49
|
Nicolas J, Mura S, Brambilla D, Mackiewicz N, Couvreur P. Design, functionalization strategies and biomedical applications of targeted biodegradable/biocompatible polymer-based nanocarriers for drug delivery. Chem Soc Rev 2013; 42:1147-235. [DOI: 10.1039/c2cs35265f] [Citation(s) in RCA: 977] [Impact Index Per Article: 81.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
50
|
Rizwan S, McBurney W, Young K, Hanley T, Boyd B, Rades T, Hook S. Cubosomes containing the adjuvants imiquimod and monophosphoryl lipid A stimulate robust cellular and humoral immune responses. J Control Release 2013; 165:16-21. [DOI: 10.1016/j.jconrel.2012.10.020] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 10/20/2012] [Accepted: 10/29/2012] [Indexed: 01/28/2023]
|