1
|
Démoulins T, Techakriengkrai N, Ebensen T, Schulze K, Liniger M, Gerber M, Nedumpun T, McCullough KC, Guzmán CA, Suradhat S, Ruggli N. New Generation Self-Replicating RNA Vaccines Derived from Pestivirus Genome. Methods Mol Biol 2024; 2786:89-133. [PMID: 38814391 DOI: 10.1007/978-1-0716-3770-8_4] [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] [Indexed: 05/31/2024]
Abstract
While mRNA vaccines have shown their worth, they have the same failing as inactivated vaccines, namely they have limited half-life, are non-replicating, and therefore limited to the size of the vaccine payload for the amount of material translated. New advances averting these problems are combining replicon RNA (RepRNA) technology with nanotechnology. RepRNA are large self-replicating RNA molecules (typically 12-15 kb) derived from viral genomes defective in at least one essential structural protein gene. They provide sustained antigen production, effectively increasing vaccine antigen payloads over time, without the risk of producing infectious progeny. The major limitations with RepRNA are RNase-sensitivity and inefficient uptake by dendritic cells (DCs), which need to be overcome for efficacious RNA-based vaccine design. We employed biodegradable delivery vehicles to protect the RepRNA and promote DC delivery. Condensing RepRNA with polyethylenimine (PEI) and encapsulating RepRNA into novel Coatsome-replicon vehicles are two approaches that have proven effective for delivery to DCs and induction of immune responses in vivo.
Collapse
Affiliation(s)
- Thomas Démoulins
- The Institute of Virology and Immunology IVI, Bern & Mittelhäusern, Switzerland.
- Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, Bern, Switzerland.
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.
- Center of Excellence in Emerging Infectious Diseases in Animals, Chulalongkorn University (CU-EIDAs), Bangkok, Thailand.
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| | - Navapon Techakriengkrai
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Emerging Infectious Diseases in Animals, Chulalongkorn University (CU-EIDAs), Bangkok, Thailand
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Matthias Liniger
- The Institute of Virology and Immunology IVI, Bern & Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Markus Gerber
- The Institute of Virology and Immunology IVI, Bern & Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Teerawut Nedumpun
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Emerging Infectious Diseases in Animals, Chulalongkorn University (CU-EIDAs), Bangkok, Thailand
| | - Kenneth C McCullough
- The Institute of Virology and Immunology IVI, Bern & Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Sanipa Suradhat
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Emerging Infectious Diseases in Animals, Chulalongkorn University (CU-EIDAs), Bangkok, Thailand
| | - Nicolas Ruggli
- The Institute of Virology and Immunology IVI, Bern & Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
2
|
Seaberg J, Clegg JR, Bhattacharya R, Mukherjee P. Self-Therapeutic Nanomaterials: Applications in Biology and Medicine. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2023; 62:190-224. [PMID: 36938366 PMCID: PMC10022599 DOI: 10.1016/j.mattod.2022.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Over past decades, nanotechnology has contributed to the biomedical field in areas including detection, diagnosis, and drug delivery via opto-electronic properties or enhancement of biological effects. Though generally considered inert delivery vehicles, a plethora of past and present evidence demonstrates that nanomaterials also exude unique intrinsic biological activity based on composition, shape, and surface functionalization. These intrinsic biological activities, termed self-therapeutic properties, take several forms, including mediation of cell-cell interactions, modulation of interactions between biomolecules, catalytic amplification of biochemical reactions, and alteration of biological signal transduction events. Moreover, study of biomolecule-nanomaterial interactions offers a promising avenue for uncovering the molecular mechanisms of biology and the evolution of disease. In this review, we observe the historical development, synthesis, and characterization of self-therapeutic nanomaterials. Next, we discuss nanomaterial interactions with biological systems, starting with administration and concluding with elimination. Finally, we apply this materials perspective to advances in intrinsic nanotherapies across the biomedical field, from cancer therapy to treatment of microbial infections and tissue regeneration. We conclude with a description of self-therapeutic nanomaterials in clinical trials and share our perspective on the direction of the field in upcoming years.
Collapse
Affiliation(s)
- Joshua Seaberg
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
- M.D./Ph.D. Program, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - John R. Clegg
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
3
|
Song C, Xu J, Gao C, Zhang W, Fang X, Shang Y. Nanomaterials targeting macrophages in sepsis: A promising approach for sepsis management. Front Immunol 2022; 13:1026173. [PMID: 36569932 PMCID: PMC9780679 DOI: 10.3389/fimmu.2022.1026173] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction resulting from dysregulated host responses to infection. Macrophages play significant roles in host against pathogens and the immunopathogenesis of sepsis, such as phagocytosis of pathogens, secretion of cytokines, and phenotype reprogramming. However, the rapid progression of sepsis impairs macrophage function, and conventional antimicrobial and supportive treatment are not sufficient to restore dysregulated macrophages roles. Nanoparticles own unique physicochemical properties, surface functions, localized surface plasmon resonance phenomenon, passive targeting in vivo, good biocompatibility and biodegradability, are accessible for biomedical applications. Once into the body, NPs are recognized by host immune system. Macrophages are phagocytes in innate immunity dedicated to the recognition of foreign substances, including nanoparticles, with which an immune response subsequently occurs. Various design strategies, such as surface functionalization, have been implemented to manipulate the recognition of nanoparticles by monocytes/macrophages, and engulfed by them to regulate their function in sepsis, compensating for the shortcomings of sepsis traditional methods. The review summarizes the mechanism of nanomaterials targeting macrophages and recent advances in nanomedicine targeting macrophages in sepsis, which provides good insight for exploring macrophage-based nano-management in sepsis.
Collapse
|
4
|
Saberpour M, Najar-Peeraye S, Shams S, Bakhshi B. Effects of chitosan nanoparticles loaded with mesenchymal stem cell conditioned media on gene expression in Vibrio cholerae and Caco-2 cells. Sci Rep 2022; 12:9781. [PMID: 35697926 PMCID: PMC9192724 DOI: 10.1038/s41598-022-14057-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/31/2022] [Indexed: 11/09/2022] Open
Abstract
Vibrio (V.) cholerae forms a pellicle for self-defense in the pathological conditions in the intestine, which protects it against antibiotics and adverse conditions. Targeting biofilm genes and Toll-like receptors (TLRs) is one of the new strategies to combat multidrug-resistant bacteria. The objective of this study was to evaluate the effect of mesenchymal stem cell conditioned media (MSC CM; 1000 µg), chitosan nanoparticles incorporated with mesenchymal stem cell conditioned media (MSC CM-CS NPs; 1000 µg + 0.05%), and chitosan nanoparticles (CS NPs; 0.05%) on the expression of bap1 and rbmC biofilm genes in V. cholerae and TLR2 and TLR4 genes in Caco-2 cells. The bacteria were inoculated in the presence or absence of MSC CM, MSC CM-CS NPs, and CS NPs for 24 h at 37 °C to evaluate the expression of biofilm genes. The Caco-2 cells were also exposed to V. cholerae for 1 h and then MSC CM, MSC CM-CS NPs, and CS NPs for 18 h at 37 °C. After these times, RNA was extracted from Caco-2 cells and bacteria exposed to the compounds, and the expression of target genes was evaluated using real-time PCR. Caco-2 cell viability was also assessed by MTT assay. After adding MSC CM, MSC CM-CS NPs, and CS NPs to V. cholerae medium, the percentage reduction in gene expression of bap1 was 96, 91, and 39%, and rbmC was 93, 92, and 32%, respectively. After adding MSC CM, MSC CM-CS NPs, and CS NPs to the Caco-2 cell medium, the percentage reduction in the gene expression of TLR4 was 89, 90, and 82%, and TLR2 was 41, 43, and 32%, respectively. MTT showed that Caco-2 cell viability was high and the compounds had little toxicity on these cells. Finally, it suggests that MSC CM-CS NPs designed may be a therapeutic agent to combat inflammation and biofilm formation in multidrug-resistant V. cholerae. However, further studies in vivo are also recommended.
Collapse
Affiliation(s)
- Masoumeh Saberpour
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shahin Najar-Peeraye
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Shams
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| | - Bita Bakhshi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
5
|
Khan A, Alamry KA. Recent advances of emerging green chitosan-based biomaterials with potential biomedical applications: A review. Carbohydr Res 2021; 506:108368. [PMID: 34111686 DOI: 10.1016/j.carres.2021.108368] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/13/2022]
Abstract
Chitosan is the most abundant natural biopolymer, after cellulose. It is mainly derived from the fungi, shrimp's shells, and exoskeleton of crustaceans, through the deacetylation of chitin. The ecological sustainability associated with its exercise and the flexibility of chitosan owing to its active functional hydroxyl and amino groups makes it a promising candidate for a wide range of applications through a variety of modifications. The biodegradability and biocompatibility of chitosan and its derivatives along with their various chemical functionalities make them promising carriers for pharmaceutical, nutritional, medicinal, environmental, agriculture, drug delivery, and biotechnology applications. The present work aims to provide a detailed and organized description of modified chitosan and its derivatives-based nanomaterials for biomedical applications. We addressed the biological and physicochemical benefits of nanocomposite materials made up of chitosan and its derivatives in various formulations, including improved physicochemical stability and cells/tissue interaction, controlled drug release, and increased bioavailability and efficacy in clinical practice. Moreover, several modification techniques and their effective utilization are also reviewed and collected in this review.
Collapse
Affiliation(s)
- Ajahar Khan
- Faculty of Science, Department of Chemistry, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Khalid A Alamry
- Faculty of Science, Department of Chemistry, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
6
|
Pippa N, Gazouli M, Pispas S. Recent Advances and Future Perspectives in Polymer-Based Nanovaccines. Vaccines (Basel) 2021; 9:558. [PMID: 34073648 PMCID: PMC8226647 DOI: 10.3390/vaccines9060558] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/24/2021] [Accepted: 05/24/2021] [Indexed: 01/15/2023] Open
Abstract
Vaccination is the most valuable and cost-effective health measure to prevent and control the spread of infectious diseases. A significant number of infectious diseases and chronic disorders are still not preventable by existing vaccination schemes; therefore, new-generation vaccines are needed. Novel technologies such as nanoparticulate systems and adjuvants can enable safe and effective vaccines for difficult target populations such as newborns, elderly, and the immune-compromised. More recently, polymer-based particles have found application as vaccine platforms and vaccine adjuvants due to their ability to prevent antigen degradation and clearance, coupled with enhanced uptake by professional antigen-presenting cells (APCs). Polymeric nanoparticles have been applied in vaccine delivery, showing significant adjuvant effects as they can easily be taken up by APCs. In other words, polymer-based systems offer a lot of advantages, including versatility and flexibility in the design process, the ability to incorporate a range of immunomodulators/antigens, mimicking infection in different ways, and acting as a depot, thereby persisting long enough to generate adaptive immune responses. The aim of this review is to summarize the properties, the characteristics, the added value, and the limitations of the polymer-based nanovaccines, as well as the process of their development by the pharmaceutical industry.
Collapse
Affiliation(s)
- Natassa Pippa
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece;
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Panepistimioupolis Zografou, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Maria Gazouli
- Department of Basic Medical Science, Laboratory of Biology, School of Medicine National, Kapodistrian University of Athens, 11527 Athens, Greece;
- Department of Radiology, University General Hospital “Attikon”, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Stergios Pispas
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece;
| |
Collapse
|
7
|
Démoulins T, Ruggli N, Gerber M, Thomann-Harwood LJ, Ebensen T, Schulze K, Guzmán CA, McCullough KC. Self-Amplifying Pestivirus Replicon RNA Encoding Influenza Virus Nucleoprotein and Hemagglutinin Promote Humoral and Cellular Immune Responses in Pigs. Front Immunol 2021; 11:622385. [PMID: 33584723 PMCID: PMC7877248 DOI: 10.3389/fimmu.2020.622385] [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] [Received: 10/28/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022] Open
Abstract
Self-amplifying replicon RNA (RepRNA) promotes expansion of mRNA templates encoding genes of interest through their replicative nature, thus providing increased antigen payloads. RepRNA derived from the non-cytopathogenic classical swine fever virus (CSFV) targets monocytes and dendritic cells (DCs), potentially promoting prolonged antigen expression in the DCs, contrasting with cytopathogenic RepRNA. We engineered pestivirus RepRNA constructs encoding influenza virus H5N1 (A/chicken/Yamaguchi/7/2004) nucleoprotein (Rep-NP) or hemagglutinin (Rep-HA). The inherent RNase-sensitivity of RepRNA had to be circumvented to ensure efficient delivery to DCs for intracellular release and RepRNA translation; we have reported how only particular synthetic delivery vehicle formulations are appropriate. The question remained concerning RepRNA packaged in virus replicon particles (VRPs); we have now compared an efficient polyethylenimine (PEI)-based formulation (polyplex) with VRP-delivery as well as naked RepRNA co-administered with the potent bis-(3’,5’)-cyclic dimeric adenosine monophosphate (c-di-AMP) adjuvant. All formulations contained a Rep-HA/Rep-NP mix, to assess the breadth of both humoral and cell-mediated defences against the influenza virus antigens. Assessment employed pigs for their close immunological relationship to humans, and as natural hosts for influenza virus. Animals receiving the VRPs, as well as PEI-delivered RepRNA, displayed strong humoral and cellular responses against both HA and NP, but with VRPs proving to be more efficacious. In contrast, naked RepRNA plus c-di-AMP could induce only low-level immune responses, in one out of five pigs. In conclusion, RepRNA encoding different influenza virus antigens are efficacious for inducing both humoral and cellular immune defences in pigs. Comparisons showed that packaging within VRP remains the most efficacious for delivery leading to induction of immune defences; however, this technology necessitates employment of expensive complementing cell cultures, and VRPs do not target human cells. Therefore, choosing the appropriate synthetic delivery vehicle still offers potential for rapid vaccine design, particularly in the context of the current coronavirus pandemic.
Collapse
Affiliation(s)
- Thomas Démoulins
- The Institute of Virology and Immunology IVI, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Nicolas Ruggli
- The Institute of Virology and Immunology IVI, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Markus Gerber
- The Institute of Virology and Immunology IVI, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Lisa J Thomann-Harwood
- The Institute of Virology and Immunology IVI, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Kenneth C McCullough
- The Institute of Virology and Immunology IVI, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Abstract
Introduction: The development of more efficacious vaccines, especially subunit vaccines administered via non-invasive routes, is a priority in vaccinology. Nanogels are materials that can meet the requirements to serve as efficient vaccine delivery vehicles (in terms of thermo-sensitivity, biocompatibility, and pH-responsiveness; among others); thus there is a growing interest in exploring the potential of nanogels for vaccine development. Areas covered: Herein, a critical analysis of nanogel synthesis methodologies is presented and nanogel-based vaccines under development are summarized and placed in perspective. Promising vaccine candidates based on nanogels have been reported for cancer, obesity, and infectious diseases (mainly respiratory diseases). Some of the candidates were administered by mucosal routes which are highly attractive in terms of simple administration and induction of protective responses at both mucosal and systemic levels. Expert opinion: The most advanced models of nanogel-based vaccines comprise candidates against cancer, based on cholesteryl pullulan nanogels evaluated in clinical trials with promising findings; as well as some vaccines against respiratory pathogens tested in mice thus far. Nonetheless, the challenge for this field is advancing in clinical trials and proving the protective potential in test animals for many other candidates. Implementing green synthesis approaches for nanogels is also required.
Collapse
|
9
|
Qi N, Zhao H, Wang Q, Qin Y, Yuan H, Li Y. Preparing CdS QDs in sodium alginate gel: realizing water solubility and stimuli responsiveness of QDs in an integrative way. SOFT MATTER 2019; 15:2319-2327. [PMID: 30747942 DOI: 10.1039/c8sm02483a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Quantum dots (QDs) are of great interest due to their excellent fluorescence properties and thus, they have been widely studied. Compared with the typical organometallic synthetic routes and hydrothermal methods usually carried out under high temperatures, methods employing colloidal templates can be used for preparing QDs in mild conditions and have gained increasing attention. In this prospect, a hydrogel is an ideal colloidal template for the preparation of QDs in an aqueous medium, while the related study for in situ preparation of QDs in a gel and the consequent functionalization of QDs are in demand. In this paper, we proposed a two-step method to prepare CdS QDs in a sodium alginate (SA) gel, which showed effective constraint in the uniform size distribution of QDs. Without the introduction of additional ligands, the prepared CdS-SA QDs exhibited responsiveness to pH and detection of Fe3+, thus providing a simplified way for the functionalization of QDs. CdS-SA QDs showed good biocompatibility and stability in a certain concentration, which indicated the prospective applications of CdS-SA QDs in the fields of biological labeling and environmental sensing.
Collapse
Affiliation(s)
- Na Qi
- Key Laboratory of Colloid and Interface Chemistry of State Education Ministry, Shandong University, 27 South Road of ShanDa, Jinan, Shandong 250100, P. R. China.
| | | | | | | | | | | |
Collapse
|
10
|
Yao Y, Liao W, Yu R, Du Y, Zhang T, Peng Q. Potentials of combining nanomaterials and stem cell therapy in myocardial repair. Nanomedicine (Lond) 2018; 13:1623-1638. [PMID: 30028249 DOI: 10.2217/nnm-2018-0013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cardiac diseases have become the leading cause of death worldwide. Developing efficient strategies to treat such diseases is of great urgency. Stem cell-based regeneration medicine offers a novel approach for heart repair. However, low retention and poor survival rate of engrafted cells limit its applications. Nanomaterials have shown great potentials in addressing above issues due to nanoparticles-bio interactions. Therefore, combining nanomaterials and stem cell therapy is of great interest and significance for heart repair. Herein, we provide a comprehensive understanding of the applications of four types of nanomaterials (nanogels, polymeric nanomaterials, inorganic nanomaterials and exosomes) in stem cell therapy for myocardial repair. In addition, we launch an initial discussion on current problems and more importantly, possible solutions for myocardial repair.
Collapse
Affiliation(s)
- Yang Yao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.,Department of Oral Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Wen Liao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ruichao Yu
- Department of Pathophysiology & Molecular Pharmacology, Joslin Diabetes Center, Harvard Medical School, 1 Joslin Place, Boston, MA 02215, USA
| | - Yu Du
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ting Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Daneshmandi S, Pourfathollah AA, Forouzandeh-Moghaddam M. Enhanced CD40 and ICOSL expression on dendritic cells surface improve anti-tumor immune responses; effectiveness of mRNA/chitosan nanoparticles. Immunopharmacol Immunotoxicol 2018; 40:375-386. [PMID: 30265161 DOI: 10.1080/08923973.2018.1510959] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Objective: To improve dendritic cells (DCs) function, we targeted DCs to over express CD40 and inducible costimulator ligand (ICOSL) costimulatory molecules along with total messenger RNA (mRNA) of tumor cells to achieve a safe and effective system for treatment of tumor. Materials and methods: We generated CD40 and ICOSL mRNA in vitro and manipulated DCs using chitosan nanoparticles and also lipofectamine transfection system then examined in vitro and in vivo. Results: Mice bone marrow derived DCs pulsed with total tumor mRNA/CD40 mRNA or ICOSL mRNA showed higher expression of DCs maturation markers (CD40, ICOSL, CD86, and MHC-II) and accelerated secretion of pro-inflammatory cytokines. Co-culture of DCs with T cells enhanced proliferation of T cells and shift toward stronger Th1 cytokine responses especially in presence of CD40 over expressed DCs. Intra-tumor administration of manipulated DCs to 4T1 tumor mice model showed delay in growth of tumor volume, trend to increase in mice survival, and stronger anti-tumor cytokines production in splenocytes of mice model (with higher efficacy of mRNA/chitosan nanoparticle system). Conclusions: Hence, we suggest that targeting intra-tumor DCs to elicit expression of CD40 and ICOSL and present broad range of tumor antigens could yield effective anti-tumor responses. In this regard, CD40 molecule manipulation trigger stronger functions, while mRNA/chitosan nanoparticles system could provide a high potent tool for targeting strategies.
Collapse
Affiliation(s)
- Saeed Daneshmandi
- a Department of Immunology, Faculty of Medical Sciences , Tarbiat Modares University , Tehran , Iran
| | - Ali Akbar Pourfathollah
- a Department of Immunology, Faculty of Medical Sciences , Tarbiat Modares University , Tehran , Iran
| | | |
Collapse
|
12
|
Wolf-Grosse S, Mollnes TE, Ali S, Stenvik J, Nilsen AM. Iron oxide nanoparticles enhance Toll-like receptor-induced cytokines in a particle size- and actin-dependent manner in human blood. Nanomedicine (Lond) 2018; 13:1773-1785. [PMID: 30084726 DOI: 10.2217/nnm-2017-0362] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Aim: To assess the effects of different-sized iron oxide nanoparticles (IONPs) on inflammatory responses in human whole blood. Materials & methods: Human whole blood with and without 10 and 30 nm IONPs was incubated with Toll-like receptor (TLR) ligands. Cytokine levels, complement activation, reactive oxygen species and viability were determined. Results: The 10 nm IONPs enhanced the TLR2/6, TLR4 and partly TLR8-mediated cytokine production, whereas the 30 nm IONPs partly enhanced TLR2/6 and decreased TLR8-mediated cytokine production. Particle-mediated enhancement of TLR4-induced cytokines could not be explained by complement activation, but was dependent on TLR4/MD2 and CD14, as well as actin polymerization. Conclusion: The IONPs differentially affected the TLR ligand-induced cytokines, which has important implications for biomedical applications of IONPs.
Collapse
Affiliation(s)
- Susann Wolf-Grosse
- Department of Clinical & Molecular Medicine, Norwegian University of Science & Technology, 7491 Trondheim, Norway
| | - Tom E Mollnes
- Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science & Technology, 7491 Trondheim, Norway.,Department of Immunology, Oslo University Hospital, Rikshospitalet, 0424 Oslo, Norway.,Research Laboratory, Nordland Hospital, 8092 Bodø, Norway.,Faculty of Health Sciences, K.G. Jebsen Thrombosis Research & Expertise Center, University of Tromsø, 9037 Tromsø, Norway
| | - Syed Ali
- Division of Neurotoxicology, US FDA/National Center for Toxicological Research, Jefferson, AR 501, USA
| | - Jørgen Stenvik
- Department of Clinical & Molecular Medicine, Norwegian University of Science & Technology, 7491 Trondheim, Norway.,Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science & Technology, 7491 Trondheim, Norway
| | - Asbjørn M Nilsen
- Department of Clinical & Molecular Medicine, Norwegian University of Science & Technology, 7491 Trondheim, Norway
| |
Collapse
|
13
|
Toma CC, Aloisi A, Bordoni V, Di Corato R, Rauner M, Cuniberti G, Delogu LG, Rinaldi R. Immune Profiling of Polysaccharide Submicron Vesicles. Biomacromolecules 2018; 19:3560-3571. [DOI: 10.1021/acs.biomac.8b00832] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Chiara C. Toma
- CNR-NANO (ECMT) Institute of Nanoscience,
Via Arnesano, 73100 Lecce, Italy
| | - Alessandra Aloisi
- CNR-IMM Institute for Microelectronics and Microsystems, Via Monteroni, University Campus, A3 Building, 73100 Lecce, Italy
- CNR-NANO (ECMT) Institute of Nanoscience,
Via Arnesano, 73100 Lecce, Italy
| | - Valentina Bordoni
- Department of Chemistry and Pharmacy, University of Sassari, via Muroni 23, 07100 Sassari, Italy
| | | | | | | | - Lucia G. Delogu
- Department of Chemistry and Pharmacy, University of Sassari, via Muroni 23, 07100 Sassari, Italy
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, via Licio Giorgeri 1, 34134 Trieste, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, corso Stati Uniti 4 35127 Padova, Italy
| | - Rosaria Rinaldi
- CNR-IMM Institute for Microelectronics and Microsystems, Via Monteroni, University Campus, A3 Building, 73100 Lecce, Italy
| |
Collapse
|
14
|
Mosafer J, Sabbaghi AH, Badiee A, Dehghan S, Tafaghodi M. Preparation, characterization and in vivo evaluation of alginate-coated chitosan and trimethylchitosan nanoparticles loaded with PR8 influenza virus for nasal immunization. Asian J Pharm Sci 2018; 14:216-221. [PMID: 32104453 PMCID: PMC7032123 DOI: 10.1016/j.ajps.2018.04.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 03/14/2018] [Accepted: 04/06/2018] [Indexed: 11/18/2022] Open
Abstract
For efficient mucosal vaccine delivery, nanoparticulate antigens are better taken by microfold cells in the nasal associated lymphoid tissue and also dendritic cells. Nanoparticles based on polymers such as chitosan (CHT) and its water soluble derivative, trimethylchitosan (TMC), could be successfully used as carrier/adjuvant for this purpose. Sodium alginate, a negatively charged biopolymer, could modify the immunostimulatory properties of CHT and TMC NPs and increase their stability. Sodium alginate (ALG)-coated chitosan (CHT) and trimethylchitosan (TMC) nanoparticles (NPs) loaded with inactivated PR8 influenza virus were successfully prepared by direct coating of the virus with CHT or TMC polymers to evaluate their immunoadjuvant potential after nasal immunization. After nasal immunizations in BALB/c mice, PR8-CHT formulation elicited higher IgG2a and IgG1 antibody titers compared with PR8-TMC. ALG coating of this formulation (PR8-CHT-ALG) significantly decreased the antibody titers and a less immune response was induced than PR8-TMC-ALG formulation. PR8-TMC-ALG formulation showed significantly higher IgG2a/IgG1 ratio, as criteria for Th1-type immune response, compared with PR8-CHT-ALG and PR8 virus alone. Altogether, the PR8-TMC-ALG formulation could be considered as an efficient intranasal antigen delivery system for nasal vaccines.
Collapse
Affiliation(s)
- Jafar Mosafer
- Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ali Badiee
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Solmaz Dehghan
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Tafaghodi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Corresponding author. Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, P.O. Box 9196773117, Mashhad, Iran. Tel.: +98 51 31801337.
| |
Collapse
|
15
|
Shen Y, Hao T, Ou S, Hu C, Chen L. Applications and perspectives of nanomaterials in novel vaccine development. MEDCHEMCOMM 2018; 9:226-238. [PMID: 30108916 PMCID: PMC6083789 DOI: 10.1039/c7md00158d] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 10/17/2017] [Indexed: 01/22/2023]
Abstract
Vaccines show great potential for both prophylactic and therapeutic use in infections, cancer, and other diseases. With the rapid development of bio-technologies and materials sciences, nanomaterials are playing essential roles in novel vaccine formulations and can boost antigen effectiveness by operating as delivery systems to enhance antigen processing and/or as immune-potentiating adjuvants to induce or potentiate immune responses. The effect of nanoparticles in vaccinology showed enhanced antigen stability and immunogenicity as well as targeted delivery and slow release. However, obstacles remain due to the lack of fundamental knowledge on the detailed molecular working mechanism and in vivo bio-effects of nanoparticles. This review provides a broad overview of the current improvements in nanoparticles in vaccinology. Modern nanoparticle vaccines are classified by the nanoparticles' action based on either delivery system or immune potentiator approaches. The mechanisms of interaction of nanoparticles with the antigens and the immune system are discussed. Nanoparticle vaccines approved for use are also listed. A fundamental understanding of the in vivo bio-distribution and the fate of nanoparticles will accelerate the rational design of new nanoparticles comprising vaccines in the future.
Collapse
Affiliation(s)
- Yingbin Shen
- Department of Food Science and Engineering , School of Science and Engineering , Jinan University , Guangzhou 510632 , Guangdong , China . ; ; ; ; ; ; Tel: +86 138 801 32918
| | - Tianyao Hao
- Department of Food Science and Engineering , School of Science and Engineering , Jinan University , Guangzhou 510632 , Guangdong , China . ; ; ; ; ; ; Tel: +86 138 801 32918
| | - Shiyi Ou
- Department of Food Science and Engineering , School of Science and Engineering , Jinan University , Guangzhou 510632 , Guangdong , China . ; ; ; ; ; ; Tel: +86 138 801 32918
| | - Churan Hu
- Department of Food Science and Engineering , School of Science and Engineering , Jinan University , Guangzhou 510632 , Guangdong , China . ; ; ; ; ; ; Tel: +86 138 801 32918
| | - Long Chen
- Department of Food Science and Engineering , School of Science and Engineering , Jinan University , Guangzhou 510632 , Guangdong , China . ; ; ; ; ; ; Tel: +86 138 801 32918
| |
Collapse
|
16
|
Démoulins T, Ebensen T, Schulze K, Englezou PC, Pelliccia M, Guzmán CA, Ruggli N, McCullough KC. Self-replicating RNA vaccine functionality modulated by fine-tuning of polyplex delivery vehicle structure. J Control Release 2017; 266:256-271. [PMID: 28935594 DOI: 10.1016/j.jconrel.2017.09.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 12/17/2022]
Abstract
The major limitations with large and complex self-amplifying RNA vaccines (RepRNA) are RNase-sensitivity and inefficient translation in dendritic cells (DCs). Condensing RepRNA with polyethylenimine (PEI) gave positive in vitro readouts, but was largely inferior to virus-like replicon particles (VRP) or direct electroporation. In the present study, we improved such polyplex formulation and determined that fine-tuning of the polyplex structure is essential for ensuring efficacious translation. Thereby, three parameters dominate: (i) PEI molecular weight (MW); (ii) RepRNA:PEI (weight:weight) ratio; and (iii) inclusion of cell penetrating peptides (CPPs). Seven commercially available linear PEIs (MW 2,500-250,000) were classified as strong, intermediate or low for their aptitude at complexing and protecting RepRNA for delivery into porcine blood DCs. Inclusion of (Arg)9 or TAT(57-57) CPPs further modified the translation readouts, but varied for different gene expressions. Dependent on the formulation, translation of the gene of interest (GOI) inserted into the RepRNA (luciferase, or influenza virus hemagglutinin or nucleoprotein) could decrease, while the RepRNA structural gene (E2) translation increased. This was noted in the porcine SK6 cell line, as well as both porcine and, for the first time, human DCs. Two formulations - [Rep/PEI-4,000 (1:3)] and [Rep/PEI-40,000 (1:2)/(Arg)9] were efficacious in vivo in mice and pigs, where specific CD8+ T and CD4+ T-cell responses against the GOI-encoded antigen were observed for the first time. The results demonstrate that different polyplex formulations differ in their interaction with the RepRNA such that only certain genes can be translated. Thus, delivery of these large self-replicating RNA molecules require definition with respect to translation of different genes, rather than just the GOI as is the norm, for identifying optimal delivery for the desired immune activation in vivo.
Collapse
Affiliation(s)
- Thomas Démoulins
- Institute of Virology and Immunology (IVI), Sensemattstrasse 293, CH-3147 Mittelhäusern, Switzerland.
| | - Thomas Ebensen
- Helmholtz Centre for Infection Research Braunschweig (HZI), Department of Vaccinology and Applied Microbiology, Braunschweig, Germany
| | - Kai Schulze
- Helmholtz Centre for Infection Research Braunschweig (HZI), Department of Vaccinology and Applied Microbiology, Braunschweig, Germany
| | - Pavlos C Englezou
- Institute of Virology and Immunology (IVI), Sensemattstrasse 293, CH-3147 Mittelhäusern, Switzerland
| | - Maria Pelliccia
- NorthWest Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Carlos A Guzmán
- Helmholtz Centre for Infection Research Braunschweig (HZI), Department of Vaccinology and Applied Microbiology, Braunschweig, Germany
| | - Nicolas Ruggli
- Institute of Virology and Immunology (IVI), Sensemattstrasse 293, CH-3147 Mittelhäusern, Switzerland
| | - Kenneth C McCullough
- Institute of Virology and Immunology (IVI), Sensemattstrasse 293, CH-3147 Mittelhäusern, Switzerland
| |
Collapse
|
17
|
Bazzi S, Modjtahedi H, Mudan S, Achkar M, Akle C, Bahr GM. Immunomodulatory effects of heat-killed Mycobacterium obuense on human blood dendritic cells. Innate Immun 2017; 23:592-605. [PMID: 28853313 DOI: 10.1177/1753425917727838] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Heat-killed (HK) Mycobacterium obuense is a novel immunomodulator, currently undergoing clinical evaluation as an immunotherapeutic agent in the treatment of cancer. Here, we examined the effect of in vitro exposure to HK M. obuense on the expression of different categories of surface receptors on human blood myeloid (m) and plasmacytoid (p) DCs. Moreover, we have characterized the cytokine and chemokine secretion patterns of purified total blood DCs stimulated with HK M. obuense. HK M. obuense significantly up-regulated the expression of CD11c, CD80, CD83, CD86, CD274 and MHC class II in whole-blood mDCs and CD80, CD123 and MHC class II in whole-blood pDCs. Down-regulation of CD195 expression in both DC subpopulations was also noted. Further analysis showed that HK M. obuense up-regulated the expression of CD80, CD83 and MHC class II on purified blood DC subpopulations. TLR2 and TLR1 were also identified to be engaged in mediating the HK M. obuense-induced up-regulation of surface receptor expression on whole blood mDCs. In addition, our data demonstrated that HK M. obuense augmented the secretion of CCL4, CCL5, CCL22, CXCL8, IL-6, IL-12p40 and TNF-α by purified total blood DCs. Taken together, our data suggest that HK M. obuense exerts potent differential immunomodulatory effects on human DC subpopulations.
Collapse
Affiliation(s)
- Samer Bazzi
- 1 School of Life Sciences, Faculty of Science, Engineering and Computing, 4264 Kingston University , Kingston upon Thames, UK.,2 Biology Department, Faculty of Sciences, 54686 University of Balamand , Al Kurah, Lebanon
| | - Helmout Modjtahedi
- 1 School of Life Sciences, Faculty of Science, Engineering and Computing, 4264 Kingston University , Kingston upon Thames, UK
| | - Satvinder Mudan
- 3 St George's University of London, Imperial College, London and The Royal Marsden Hospital, London, UK
| | - Marcel Achkar
- 4 Clinical Laboratory Department, Nini Hospital, Tripoli, Lebanon
| | | | - Georges M Bahr
- 6 Faculty of Medicine and Medical Sciences, 54686 University of Balamand , Al Kurah, Lebanon
| |
Collapse
|
18
|
Chauhan N, Tiwari S, Iype T, Jain U. An overview of adjuvants utilized in prophylactic vaccine formulation as immunomodulators. Expert Rev Vaccines 2017; 16:491-502. [DOI: 10.1080/14760584.2017.1306440] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Nidhi Chauhan
- Amity Institute of Nanotechnology, Amity University, Noida, India
| | - Sukirti Tiwari
- Amity Institute of Nanotechnology, Amity University, Noida, India
| | - Tessy Iype
- R & D Division, MagGenome Technologies Pvt. Ltd., Kochi, India
| | - Utkarsh Jain
- Amity Institute of Nanotechnology, Amity University, Noida, India
| |
Collapse
|
19
|
Kouskoura T, Katsaros C, von Gunten S. The Potential Use of Pharmacological Agents to Modulate Orthodontic Tooth Movement (OTM). Front Physiol 2017; 8:67. [PMID: 28228735 PMCID: PMC5296343 DOI: 10.3389/fphys.2017.00067] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/24/2017] [Indexed: 11/13/2022] Open
Abstract
The biological processes that come into play during orthodontic tooth movement (OTM) have been shown to be influenced by a variety of pharmacological agents. The effects of such agents are of particular relevance to the clinician as the rate of tooth movement can be accelerated or reduced as a result. This review aims to provide an overview of recent insights into drug-mediated effects and the potential use of drugs to influence the rate of tooth movement during orthodontic treatment. The limitations of current experimental models and the need for well-designed clinical and pre-clinical studies are also discussed.
Collapse
Affiliation(s)
- Thaleia Kouskoura
- Department of Orthodontics and Dentofacial Orthopedics, School of Dental Medicine, University of Bern Bern, Switzerland
| | - Christos Katsaros
- Department of Orthodontics and Dentofacial Orthopedics, School of Dental Medicine, University of Bern Bern, Switzerland
| | | |
Collapse
|
20
|
Démoulins T, Englezou PC, Milona P, Ruggli N, Tirelli N, Pichon C, Sapet C, Ebensen T, Guzmán CA, McCullough KC. Self-Replicating RNA Vaccine Delivery to Dendritic Cells. Methods Mol Biol 2017; 1499:37-75. [PMID: 27987142 DOI: 10.1007/978-1-4939-6481-9_3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Most current vaccines are either inactivated pathogen-derived or protein/peptide-based, although attenuated and vector vaccines have also been developed. The former induce at best moderate protection, even as multimeric antigen, due to limitations in antigen loads and therefore capacity for inducing robust immune defense. While attenuated and vector vaccines offer advantages through their replicative nature, drawbacks and risks remain with potential reversion to virulence and interference from preexisting immunity. New advances averting these problems are combining self-amplifying replicon RNA (RepRNA) technology with nanotechnology. RepRNA are large self-replicating RNA molecules (12-15 kb) derived from viral genomes defective in at least one structural protein gene. They provide sustained antigen production, effectively increasing vaccine antigen payloads over time, without the risk of producing infectious progeny. The major limitation with RepRNA is RNase-sensitivity and inefficient uptake by dendritic cells (DCs)-absolute requirements for efficacious vaccine design. We employed biodegradable delivery vehicles to protect the RepRNA and promote DC delivery. Encapsulating RepRNA into chitosan nanoparticles, as well as condensing RepRNA with polyethylenimine (PEI), cationic lipids, or chitosans, has proven effective for delivery to DCs and induction of immune responses in vivo.
Collapse
Affiliation(s)
- Thomas Démoulins
- Institute of Virology and Immunology (IVI), Sensemattstrasse 293, CH-3147, Mittelhäusern, Switzerland.
| | - Pavlos C Englezou
- Institute of Virology and Immunology (IVI), Sensemattstrasse 293, CH-3147, Mittelhäusern, Switzerland
| | - Panagiota Milona
- Institute of Virology and Immunology (IVI), Sensemattstrasse 293, CH-3147, Mittelhäusern, Switzerland
| | - Nicolas Ruggli
- Institute of Virology and Immunology (IVI), Sensemattstrasse 293, CH-3147, Mittelhäusern, Switzerland
| | - Nicola Tirelli
- Centre of Regenerative Medicine, University of Manchester, Manchester, M13 9PT, UK
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS UPR4301, 45071, Orléans cedex 2, France
| | - Cédric Sapet
- OzBiosciences, Parc scientifique de Luminy, Marseille, France
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Kenneth C McCullough
- Institute of Virology and Immunology (IVI), Sensemattstrasse 293, CH-3147, Mittelhäusern, Switzerland
| |
Collapse
|
21
|
Wang H, Qian J, Ding F. Recent advances in engineered chitosan-based nanogels for biomedical applications. J Mater Chem B 2017; 5:6986-7007. [DOI: 10.1039/c7tb01624g] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent progress in the preparation and biomedical applications of engineered chitosan-based nanogels has been comprehensively reviewed.
Collapse
Affiliation(s)
- Hongxia Wang
- School of Printing and Packaging, Wuhan University
- Wuhan 430072
- P. R. China
| | - Jun Qian
- School of Printing and Packaging, Wuhan University
- Wuhan 430072
- P. R. China
| | - Fuyuan Ding
- School of Printing and Packaging, Wuhan University
- Wuhan 430072
- P. R. China
| |
Collapse
|
22
|
Tsintou M, Wang C, Dalamagkas K, Weng D, Zhang YN, Niu W. Nanogels for biomedical applications. NANOBIOMATERIALS SCIENCE, DEVELOPMENT AND EVALUATION 2017:87-124. [DOI: 10.1016/b978-0-08-100963-5.00005-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
23
|
Huang Y, Qin T, Huang Y, Liu Z, Bo R, Hu Y, Liu J, Wu Y, Wang D. Rehmannia glutinosa polysaccharide liposome as a novel strategy for stimulating an efficient immune response and their effects on dendritic cells. Int J Nanomedicine 2016; 11:6795-6808. [PMID: 28008254 PMCID: PMC5167497 DOI: 10.2147/ijn.s119108] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Nanomedicine, the medical application of nanotechnology, promises a seemingly limitless range of applications from drug delivery to adjuvants and therapeutics. Our current research is focused on natural polymer-based liposome adjuvants. With the aim of inducing protective and long-lasting immunity, the immunological adjuvant activity of Rehmannia glutinosa polysaccharide liposome (RGPL) was investigated. In vivo, the splenic lymphocyte proliferation ratios and ovalbumin-specific immunoglobulin G titers of ovalbumin-RGPL-vaccinated mice were significantly upregulated. In draining lymph nodes, the expression of MHC II+CD11c+ and CD86+CD11c+ was increased by RGPL; in addition, the percentages of central memory cells (TCM) and effector memory cells (TEM) were also elevated. RGPL could effectively provide adequate antigen exposure in lymph nodes. In vitro, RGPL could promote dendritic cell maturation and enhance dendritic cell functions, such as the mixed lymphocyte reaction and antigen presentation. Overall, the results demonstrated that RGPL has the potential to act as an effective controlled release vaccine adjuvant.
Collapse
Affiliation(s)
- Yee Huang
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing
| | - Tao Qin
- College of Animal Science and Veterinary Medicine, Fujian Agriculture and Forestry University, Fuzhou, People's Republic of China
| | - Yifan Huang
- College of Animal Science and Veterinary Medicine, Fujian Agriculture and Forestry University, Fuzhou, People's Republic of China
| | - Zhenguang Liu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing
| | - Ruonan Bo
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing
| | - Yuanliang Hu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing
| | - Jiaguo Liu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing
| | - Yi Wu
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing
| | - Deyun Wang
- College of Veterinary Medicine, Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing; College of Animal Science and Veterinary Medicine, Fujian Agriculture and Forestry University, Fuzhou, People's Republic of China
| |
Collapse
|
24
|
Polyethylenimine-based polyplex delivery of self-replicating RNA vaccines. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 12:711-722. [PMID: 26592962 DOI: 10.1016/j.nano.2015.11.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/20/2015] [Accepted: 11/04/2015] [Indexed: 01/13/2023]
Abstract
UNLABELLED Self-amplifying replicon RNA (RepRNA) are large molecules (12-14 kb); their self-replication amplifies mRNA template numbers, affording several rounds of antigen production, effectively increasing vaccine antigen payloads. Their sensitivity to RNase-sensitivity and inefficient uptake by dendritic cells (DCs) - absolute requirements for vaccine design - were tackled by condensing RepRNA into synthetic, nanoparticulate, polyethylenimine (PEI)-polyplex delivery vehicles. Polyplex-delivery formulations for small RNA molecules cannot be transferred to RepRNA due to its greater size and complexity; the N:P charge ratio and impact of RepRNA folding would influence polyplex condensation, post-delivery decompaction and the cytosolic release essential for RepRNA translation. Polyplex-formulations proved successful for delivery of RepRNA encoding influenza virus hemagglutinin and nucleocapsid to DCs. Cytosolic translocation was facilitated, leading to RepRNA translation. This efficacy was confirmed in vivo, inducing both humoral and cellular immune responses. Accordingly, this paper describes the first PEI-polyplexes providing efficient delivery of the complex and large, self-amplifying RepRNA vaccines. FROM THE CLINICAL EDITOR The use of self-amplifying replicon RNA (RepRNA) to increase vaccine antigen payloads can potentially be useful in effective vaccine design. Nonetheless, its use is limited by the degradation during the uptake process. Here, the authors attempted to solve this problem by packaging RepRNA using polyethylenimine (PEI)-polyplex delivery vehicles. The efficacy was confirmed in vivo by the appropriate humoral and cellular immune responses. This novel delivery method may prove to be very useful for future vaccine design.
Collapse
|
25
|
Tahara Y, Akiyoshi K. Current advances in self-assembled nanogel delivery systems for immunotherapy. Adv Drug Deliv Rev 2015; 95:65-76. [PMID: 26482187 DOI: 10.1016/j.addr.2015.10.004] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/17/2015] [Accepted: 10/09/2015] [Indexed: 10/24/2022]
Abstract
Since nanogels (nanometer-sized gels) were developed two decades ago, they were utilized as carriers of innovative drug delivery systems. In particular, immunological drug delivery via self-assembled nanogels (self-nanogels) owing to their nanometer size and molecular chaperon-like ability to encapsulate large biomolecules is one of the most well studied and successful applications of nanogels. In the present review, we focus on self-nanogel applications as immunological drug delivery systems for cancer vaccines, cytokine delivery, nasal vaccines, and nucleic acid delivery, including several clinical trials. Cancer vaccines were the first practical application of self-nanogels as vehicles for drug delivery. After successful pre-clinical studies, phase I clinical trials were conducted, and it was found that vaccines consisting of self-nanogels could be administered repeatedly to humans without serious adverse effects, and self-nanogel vaccines induced antigen-specific cellular and humoral immunity. Cytokine delivery via self-nanogels led to the sustained release of IL-12, suppressed tumor growth, and increased Th1-type immune responses. Cationic self-nanogels were effective in penetrating the nasal mucosa and resulted in successful nasal vaccines in mice and nonhuman primates. Cationic self-nanogels were also used for the intracellular delivery of proteins and nucleic acids, and were successfully used to knockdown tumor growth factor expression using short interfering RNA with the immunological effect. These studies suggest that self-nanogels are currently one of the most unique and attractive immunological drug delivery systems and are edging closer to practical use.
Collapse
|
26
|
Correia-Pinto JF, Csaba N, Schiller JT, Alonso MJ. Chitosan-Poly (I:C)-PADRE Based Nanoparticles as Delivery Vehicles for Synthetic Peptide Vaccines. Vaccines (Basel) 2015; 3:730-50. [PMID: 26378586 PMCID: PMC4586475 DOI: 10.3390/vaccines3030730] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/14/2015] [Accepted: 08/31/2015] [Indexed: 11/25/2022] Open
Abstract
The safety and precision of peptide antigens has prompted the search for adjuvants capable of increasing the immune response against these intrinsically poorly immunogenic antigens. The integration of both immunostimulants and peptide antigens within nanometric delivery systems for their co-delivery to immune cells is a promising vaccination strategy. With this in mind, the potential synergistic effect of the immunostimulant poly (I:C) (pIC) and a T-Helper peptide (PADRE), integrated into a chitosan (CS) based nanostructure, was explored. The value of this nanostructured combination of materials was assessed for a peptide antigen (1338aa) derived from the HPV-16 L2 protein. These nanoparticles, produced by ionic gelation technique, exhibited a nanometric size (<300 nm), a high positive surface charge (>40 mV) and high pIC association efficiency (>96%). They also showed capacity for the association of both the 1338aa and PADRE peptides. The influence of the presence of pIC and PADRE in the nanocomposition, as well as that of the peptide presentation form (encapsulated versus surface adsorbed) on the antibody induction was evaluated in a preliminary in vivo study. The data obtained highlights the possibility to engineer nanoparticles through the rational combination of a number of adjuvant molecules together with the antigen.
Collapse
Affiliation(s)
- Jorge F Correia-Pinto
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Av. Barcelona s/n, Campus Vida, University of Santiago de Compostela, 15707 Santiago de Compostela, Spain.
| | - Noemi Csaba
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Av. Barcelona s/n, Campus Vida, University of Santiago de Compostela, 15707 Santiago de Compostela, Spain.
| | - John T Schiller
- Laboratory of Cellular Oncology, National Cancer Institute, NIH, Bethesda, MD, USA.
| | - Maria J Alonso
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Av. Barcelona s/n, Campus Vida, University of Santiago de Compostela, 15707 Santiago de Compostela, Spain.
| |
Collapse
|
27
|
Li Y, Maciel D, Rodrigues J, Shi X, Tomás H. Biodegradable Polymer Nanogels for Drug/Nucleic Acid Delivery. Chem Rev 2015; 115:8564-8608. [PMID: 26259712 DOI: 10.1021/cr500131f] [Citation(s) in RCA: 397] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yulin Li
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
- The State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, East China University of Science and Technology , Shanghai 200237, People's Republic of China
| | - Dina Maciel
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
| | - João Rodrigues
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
| | - Xiangyang Shi
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University , Shanghai 201620, People's Republic of China
| | - Helena Tomás
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira , Campus da Penteada 9000-390, Funchal, Portugal
| |
Collapse
|
28
|
Vasiliev YM. Chitosan-based vaccine adjuvants: incomplete characterization complicates preclinical and clinical evaluation. Expert Rev Vaccines 2014; 14:37-53. [PMID: 25262982 DOI: 10.1586/14760584.2015.956729] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
A number of preclinical and clinical studies with chitosan-adjuvanted antigen- and DNA-based vaccines have been carried out. Various chitosans and their modifications, in different forms (solutions, powders, gels and particles), have been evaluated with various antigens administered via different routes. Chitosan is a generic name for a wide array of glucosamine-based substances derived from biological sources, and standardization is necessary. However, in most of the studies published to date, molecular weight, viscosity, deacetylation degree and/or purity level (especially endotoxins) are not provided for the initial chitosan substance and/or final formulation and the preparation procedure is not detailed. Evaluation of adjuvant properties is challenging, given that the only available data are insufficient to demonstrate immunogenicity for chitosans with characteristics within certain intervals to elucidate mechanisms of action or to exclude impurities as the active substance. These and other issues of chitosan-based vaccine adjuvants are summarized and a step-by-step evaluation approach for chitosan-based vaccine adjuvants is outlined.
Collapse
Affiliation(s)
- Yuri M Vasiliev
- Mechnikov Research Institute of Vaccines and Sera, M. Kazeny lane, 5a, Moscow, 105064, Russian Federation
| |
Collapse
|
29
|
Abstract
Most infectious diseases are caused by pathogenic infiltrations from the mucosal tract. Therefore, vaccines delivered to the mucosal tissues can mimic natural infections and provide protection at the first site of infection. Thus, mucosal, especially, oral delivery is becoming the most preferred mode of vaccination. However, oral vaccines have to overcome several barriers such as the extremely low pH of the stomach, the presence of proteolytic enzymes and bile salts as well as low permeability in the intestine. Several formulations based on nanoparticle strategies are currently being explored to prepare stable oral vaccine formulations. This review briefly discusses several molecular mechanisms involved in intestinal immune cell activation and various aspects of oral nanoparticle-based vaccine design that should be considered for improved mucosal and systemic immune responses.
Collapse
Affiliation(s)
- Nirmal Marasini
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | | | | |
Collapse
|
30
|
Démoulins T, Milona P, McCullough KC. Alginate-coated chitosan nanogels differentially modulate class-A and class-B CpG-ODN targeting of dendritic cells and intracellular delivery. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:1739-49. [PMID: 24941461 DOI: 10.1016/j.nano.2014.06.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 05/20/2014] [Accepted: 06/05/2014] [Indexed: 12/26/2022]
Abstract
UNLABELLED CpG-oligodeoxynucleotides (CpG-ODNs) interact with dendritic cells (DCs), but evidence is less clear for CpG-ODN admixed with or incorporated into vaccine delivery vehicles. We loaded alginate-coated chitosan-nanogels (Ng) with class-A or class-B CpG-ODN, and compared with the same CpG-ODNs free or admixed with empty Ng. Experiments were performed on both porcine and human blood DC subpopulations. Encapsulation of class-A CpG-ODN (loading into Ng) strongly reduced the CpG-ODN uptake and intracellular trafficking in the cytosol; this was associated with a marked deficiency in IFN-α induction. In contrast, encapsulation of class-B CpG-ODN increased its uptake and did not influence consistently intracellular trafficking into the nucleus. The choice of CpG-ODN class as adjuvant is thus critical in terms of how it will behave with nanoparticulate vaccine delivery vehicles. The latter can have distinctive modulatory influences on the CpG-ODN, which would require definition for different CpG-ODN and delivery vehicles prior to vaccine formulation. FROM THE CLINICAL EDITOR This basic science study investigates the role of class-A and class-B CpG-oligodeoxynucleotides loaded into alginate-coated chitosan nanogels, demonstrating differential effects between the two classes as related to the use of these nanoformulations as vaccine delivery vehicles.
Collapse
Affiliation(s)
- Thomas Démoulins
- Institute of Virology and Immunology (IVI), Mittelhäusern, Switzerland.
| | - Panagiota Milona
- Institute of Virology and Immunology (IVI), Mittelhäusern, Switzerland
| | | |
Collapse
|
31
|
Sahdev P, Ochyl LJ, Moon JJ. Biomaterials for nanoparticle vaccine delivery systems. Pharm Res 2014; 31:2563-82. [PMID: 24848341 DOI: 10.1007/s11095-014-1419-y] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/12/2014] [Indexed: 01/11/2023]
Abstract
Subunit vaccination benefits from improved safety over attenuated or inactivated vaccines, but their limited capability to elicit long-lasting, concerted cellular and humoral immune responses is a major challenge. Recent studies have demonstrated that antigen delivery via nanoparticle formulations can significantly improve immunogenicity of vaccines due to either intrinsic immunostimulatory properties of the materials or by co-entrapment of molecular adjuvants such as Toll-like receptor agonists. These studies have collectively shown that nanoparticles designed to mimic biophysical and biochemical cues of pathogens offer new exciting opportunities to enhance activation of innate immunity and elicit potent cellular and humoral immune responses with minimal cytotoxicity. In this review, we present key research advances that were made within the last 5 years in the field of nanoparticle vaccine delivery systems. In particular, we focus on the impact of biomaterials composition, size, and surface charge of nanoparticles on modulation of particle biodistribution, delivery of antigens and immunostimulatory molecules, trafficking and targeting of antigen presenting cells, and overall immune responses in systemic and mucosal tissues. This review describes recent progresses in the design of nanoparticle vaccine delivery carriers, including liposomes, lipid-based particles, micelles and nanostructures composed of natural or synthetic polymers, and lipid-polymer hybrid nanoparticles.
Collapse
Affiliation(s)
- Preety Sahdev
- Department of Pharmaceutical Sciences, College of Pharmacy University of Michigan, 2800 Plymouth Road NCRC, Ann Arbor, Michigan, 48109, USA
| | | | | |
Collapse
|
32
|
|
33
|
|
34
|
Shirota H, Klinman DM. Recent progress concerning CpG DNA and its use as a vaccine adjuvant. Expert Rev Vaccines 2013; 13:299-312. [PMID: 24308579 DOI: 10.1586/14760584.2014.863715] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
CpG Oligonucleotides (ODN) are immunomodulatory synthetic oligonucleotides designed to specifically agonize Toll-like receptor 9. Here, we review recent progress in understanding the mechanism of action of CpG ODN and provide an overview of human clinical trial results using CpG ODN to improve the vaccines for cancer, allergy and infectious disease.
Collapse
|