1
|
Duymaz D, Kebabci AO, Kizilel S. Harnessing the immunomodulatory potential of chitosan and its derivatives for advanced biomedical applications. Int J Biol Macromol 2025; 307:142055. [PMID: 40090654 DOI: 10.1016/j.ijbiomac.2025.142055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 02/21/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
The success of biomaterial applications in medicine, particularly in tissue engineering, relies on achieving a balance between promoting tissue regeneration and controlling the immune response. Due to its natural origin, high biocompatibility, and versatility, chitosan has emerged as a promising biomaterial especially for immunomodulation purposes. Immunomodulation, refers to the deliberate alteration of the immune system's activity to achieve a desired therapeutic effect either by enhancing or suppressing the function of specific immune cells, signaling pathways, or cytokine production. This modulation opens up the unlimited possibilities for the use of biomaterials, especially about the use of natural polymers such as chitosan. Although numerous chitosan-based immunoregulatory strategies have been demonstrated over the past two decades, the lack of in-depth exploration hinders the full potential of strategies that include chitosan and its derivatives in biomedical applications. Thus, in this review, the possible immunomodulatory effects of chitosan, chitosan derivatives and their potential combined with various agents and therapies are investigated in detail. Moreover, this report includes agents for localized immune response control, chitosan-based strategies with complementary immunomodulatory properties to create synergistic effects that will influence the success of cell therapies for enhanced tissue acceptance and regeneration. Finally, the challenges and outlook of chitosan-based therapies as a powerful tool for improving immunomodulatory applications are discussed for paving the way for further studies.
Collapse
Affiliation(s)
- Doğukan Duymaz
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye
| | - Aybaran O Kebabci
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye
| | - Seda Kizilel
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye.
| |
Collapse
|
2
|
Sinani G, Sessevmez M, Şenel S. Applications of Chitosan in Prevention and Treatment Strategies of Infectious Diseases. Pharmaceutics 2024; 16:1201. [PMID: 39339237 PMCID: PMC11434819 DOI: 10.3390/pharmaceutics16091201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Chitosan is the most commonly investigated functional cationic biopolymer in a wide range of medical applications due to its promising properties such as biocompatibility, biodegradability, and bioadhesivity, as well as its numerous bioactive properties. Within the last three decades, chitosan and its derivatives have been investigated as biomaterials for drug and vaccine delivery systems, besides for their bioactive properties. Due to the functional groups in its structure, it is possible to tailor the delivery systems with desired properties. There has been a great interest in the application of chitosan-based systems also for the prevention and treatment of infectious diseases, specifically due to their antimicrobial, antiviral, and immunostimulatory effects. In this review, recent applications of chitosan in the prevention and treatment of infectious diseases are reviewed, and possibilities and limitations with regards to technical and regulatory aspects are discussed. Finally, the future perspectives on utilization of chitosan as a biomaterial are discussed.
Collapse
Affiliation(s)
- Genada Sinani
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Altinbas University, 34147 Istanbul, Türkiye;
| | - Melike Sessevmez
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, 34116 Istanbul, Türkiye;
| | - Sevda Şenel
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe Univesity, 06100 Ankara, Türkiye
| |
Collapse
|
3
|
Weiss CM, Liu H, Ball EE, Hoover AR, Wong TS, Wong CF, Lam S, Hode T, Keel MK, Levenson RM, Chen WR, Coffey LL. N-dihydrogalactochitosan reduces mortality in a lethal mouse model of SARS-CoV-2. PLoS One 2023; 18:e0289139. [PMID: 37552656 PMCID: PMC10409267 DOI: 10.1371/journal.pone.0289139] [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: 03/08/2023] [Accepted: 07/11/2023] [Indexed: 08/10/2023] Open
Abstract
The rapid emergence and global dissemination of SARS-CoV-2 that causes COVID-19 continues to cause an unprecedented global health burden resulting in nearly 7 million deaths. While multiple vaccine countermeasures have been approved for emergency use, additional treatments are still needed due to sluggish vaccine rollout, vaccine hesitancy, and inefficient vaccine-mediated protection. Immunoadjuvant compounds delivered intranasally can guide non-specific innate immune responses during the critical early stages of viral replication, reducing morbidity and mortality. N-dihydrogalactochitosan (GC) is a novel mucoadhesive immunostimulatory polymer of β-0-4-linked N-acetylglucosamine that is solubilized by the conjugation of galactose glycans with current applications as a cancer immunotherapeutic. We tested GC as a potential countermeasure for COVID-19. GC was well-tolerated and did not produce histopathologic lesions in the mouse lung. GC administered intranasally before and after SARS-CoV-2 exposure diminished morbidity and mortality in humanized ACE2 receptor expressing mice by up to 75% and reduced infectious virus levels in the upper airway. Fluorescent labeling of GC shows that it is confined to the lumen or superficial mucosa of the nasal cavity, without involvement of adjacent or deeper tissues. Our findings demonstrate a new application for soluble immunoadjuvants such as GC for preventing disease associated with SARS-CoV-2 and may be particularly attractive to persons who are needle-averse.
Collapse
Affiliation(s)
- Christopher M. Weiss
- Department of Pathology, Microbiology & Immunology, University of California, Davis, California, United States of America
| | - Hongwei Liu
- Department of Pathology, Microbiology & Immunology, University of California, Davis, California, United States of America
| | - Erin E. Ball
- Department of Pathology, Microbiology & Immunology, University of California, Davis, California, United States of America
| | - Ashley R. Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, United States of America
| | - Talia S. Wong
- Department of Pathology, Microbiology & Immunology, University of California, Davis, California, United States of America
| | - Chun Fung Wong
- Immunophotonics, Inc., Saint Louis, Missouri, United States of America
| | - Samuel Lam
- Immunophotonics, Inc., Saint Louis, Missouri, United States of America
| | - Tomas Hode
- Immunophotonics, Inc., Saint Louis, Missouri, United States of America
| | - M. Kevin Keel
- Department of Pathology, Microbiology & Immunology, University of California, Davis, California, United States of America
| | - Richard M. Levenson
- Department of Pathology and Laboratory Medicine, UC Davis Health, Sacramento, California, United States of America
| | - Wei R. Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, United States of America
| | - Lark L. Coffey
- Department of Pathology, Microbiology & Immunology, University of California, Davis, California, United States of America
| |
Collapse
|
4
|
Kehagia E, Papakyriakopoulou P, Valsami G. Advances in intranasal vaccine delivery: A promising non-invasive route of immunization. Vaccine 2023:S0264-410X(23)00529-7. [PMID: 37179163 PMCID: PMC10173027 DOI: 10.1016/j.vaccine.2023.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 04/25/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023]
Abstract
The importance of vaccination has been proven particularly significant the last three years, as it is revealed to be the most efficient weapon for the prevention of several infections including SARS-COV-2. Parenteral vaccination is the most applicable method of immunization, for the prevention of systematic and respiratory infections, or central nervous system disorders, involving T and B cells to a whole-body immune response. However, the mucosal vaccines, such as nasal vaccines, can additionally activate the immune cells localized on the mucosal tissue of the upper and lower respiratory tract. This dual stimulation of the immune system, along with their needle-free administration favors the development of novel nasal vaccines to produce long-lasting immunity. In recent years, the nanoparticulate systems have been extensively involved in the formulation of nasal vaccines as polymeric, polysaccharide and lipid ones, as well as in the form of proteosomes, lipopeptides and virosomes. Advanced delivery nanosystems have been designed and evaluated as carriers or adjuvants for nasal vaccination. To this end, several nanoparticulate vaccines are undergone clinical trials as promising candidates for nasal immunization, while nasal vaccines against influenza type A and B and hepatitis B have been approved by health authorities. This comprehensive literature review aims to summarize the critical aspects of these formulations and highlight their potential for the future establishment of nasal vaccination. Both preclinical (in vitro and in vivo) and clinical studies are incorporated, summarized, and critically discussed, as well as the limitations of nasal immunization.
Collapse
Affiliation(s)
- Eleni Kehagia
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15784, Greece
| | - Paraskevi Papakyriakopoulou
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15784, Greece.
| | - Georgia Valsami
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15784, Greece
| |
Collapse
|
5
|
He X, Chen X, Wang H, Du G, Sun X. Recent advances in respiratory immunization: A focus on COVID-19 vaccines. J Control Release 2023; 355:655-674. [PMID: 36787821 PMCID: PMC9937028 DOI: 10.1016/j.jconrel.2023.02.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023]
Abstract
The development of vaccines has always been an essential task worldwide since vaccines are regarded as powerful weapons in protecting the global population. Although the vast majority of currently authorized human vaccinations are administered intramuscularly or subcutaneously, exploring novel routes of immunization has been a prominent area of study in recent years. This is particularly relevant in the face of pandemic diseases, such as COVID-19, where respiratory immunization offers distinct advantages, such as inducing systemic and mucosal responses to prevent viral infections in both the upper and lower respiratory tracts and also leading to higher patient compliance. However, the development of respiratory vaccines confronts challenges due to the physiological barriers of the respiratory tract, with most of these vaccines still in the research and development stage. In this review, we detail the structure of the respiratory tract and the mechanisms of mucosal immunity, as well as the obstacles to respiratory vaccination. We also examine the considerations necessary in constructing a COVID-19 respiratory vaccine, including the dosage form of the vaccines, potential excipients and mucosal adjuvants, and delivery systems and devices for respiratory vaccines. Finally, we present a comprehensive overview of the COVID-19 respiratory vaccines currently under clinical investigation. We hope this review can provide valuable insights and inspiration for the future development of respiratory vaccinations.
Collapse
Affiliation(s)
- Xiyue He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaoyan Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hairui Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
6
|
Jung MH, Jung SJ, Kim T. Saponin and chitosan-based oral vaccine against viral haemorrhagic septicaemia virus (VHSV) provides protective immunity in olive flounder (Paralichthys olivaceus). FISH & SHELLFISH IMMUNOLOGY 2022; 126:336-346. [PMID: 35643353 DOI: 10.1016/j.fsi.2022.05.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 06/15/2023]
Abstract
Production losses of olive flounder (Paralichthys olivaceus) have increased owing to viral haemorrhagic septicaemia virus (VHSV) infection. In this study, we determined safe concentrations of orally administered saponin and chitosan by analysing serum enzyme (AST/ALT) levels as biochemical markers of hepatic injury. Furthermore, we demonstrated the efficacy, duration of protection, and safety of saponin and chitosan-based vaccines with inactivated VHSV (IV). Oral administration of saponin, chitosan, and their combination did not induce fish mortality at all tested concentrations (0.29, 1.45, and 2.9 mg/g of fish body weight/day) 10 days after administration. However, AST level was high at a dose >0.29 mg/g of fish body weight/day. Both saponin and chitosan were found to be safe and acceptable for vaccination studies at a dose of 0.29 mg/g of fish body weight/day. Administration of IV alone did not induce protection at 2 and 4 weeks post vaccination (wpv). Olive flounders administered saponin + IV and chitosan + IV vaccines had higher immunity against VHSV with relative percentage survival (RPS) of 12.5-7.5% and 0-20.1%, respectively; however, additional immunisation with combination of saponin + chitosan + IV clearly enhanced the protection with RPS values of 10-15%, 26.7%, 42.9%, and 37.5% at 4, 8, 12, and 20 wpv, respectively. Although the RPS value of oral immunisation was not comparable to that of injectable vaccines, the manufacturing process is simple and oral administration causes less stress to juvenile fish. To investigate the development of a protective immune response, olive flounder were re-challenged with VHSV (107.8 TCID50/fish) at 70 days postinfection; 100% of the previously unexposed fish died, whereas 80-100% of the previously immunised fish survived. Our results showed the possibility of developing preventive measures against VHSV using saponin and chitosan-based oral vaccines with inactivated virus.
Collapse
Affiliation(s)
- Myung-Hwa Jung
- Department of Marine Bio and Medical Sciences, Hanseo University, Republic of Korea
| | - Sung-Ju Jung
- Department of Aqualife Medicine, Chonnam National University, Republic of Korea.
| | - Taeho Kim
- Department of Marine Production Management, Chonnam National University, Republic of Korea
| |
Collapse
|
7
|
Hasanpour Galehban M, Zeynizadeh B, Mousavi H. Ni II NPs entrapped within a matrix of l-glutamic acid cross-linked chitosan supported on magnetic carboxylic acid-functionalized multi-walled carbon nanotube: a new and efficient multi-task catalytic system for the green one-pot synthesis of diverse heterocyclic frameworks. RSC Adv 2022; 12:16454-16478. [PMID: 35754864 PMCID: PMC9171750 DOI: 10.1039/d1ra08454b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/20/2022] [Indexed: 12/19/2022] Open
Abstract
In the present study, a new l-glutamic acid cross-linked chitosan supported on magnetic carboxylic acid-functionalized multi-walled carbon nanotube (Fe3O4/f-MWCNT-CS-Glu) nanocomposite was prepared through a convenient one-pot multi-component sequential strategy. Then, nickelII nanoparticles (NiII NPs) were entrapped within a matrix of the mentioned nanocomposite. Afterward, the structure of the as-prepared Fe3O4/f-MWCNT-CS-Glu/NiII nanosystem was elucidated by various techniques, including FT-IR, PXRD, SEM, TEM, SEM-based EDX and elemental mapping, ICP-OES, TGA/DTA, and VSM. In the next part of this research, the catalytic applications of the mentioned nickelII-containing magnetic nanocomposite were assessed upon green one-pot synthesis of diverse heterocyclic frameworks, including bis-coumarins (3a-n), 2-aryl(or heteroaryl)-2,3-dihydroquinazolin-4(1H)-ones (5a-r), 9-aryl-3,3,6,6-tetramethyl-3,4,5,6,7,9-hexahydro-1H-xanthene-1,8(2H)-diones (7a-n), and 2-amino-4-aryl-7,7-dimethyl-5-oxo-5,6,7,8-tetrahydro-4H-chromene-3-carbonitriles (9a-n). The good-to-excellent yields of the desired products, satisfactory reaction rates, use of water solvent or solvent-free reaction medium, acceptable turnover numbers (TONs) and turnover frequencies (TOFs), along with comfortable recoverability and satisfying reusability of the as-prepared nanocatalyst for at least eight successive runs, and also easy work-up and purification procedures are some of the advantages of the current synthetic protocols.
Collapse
Affiliation(s)
| | - Behzad Zeynizadeh
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University Urmia Iran
| | - Hossein Mousavi
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University Urmia Iran
| |
Collapse
|
8
|
Dong C, Wang BZ. Engineered Nanoparticulate Vaccines to Combat Recurring and Pandemic Influenza Threats. ADVANCED NANOBIOMED RESEARCH 2022; 2:2100122. [PMID: 35754779 PMCID: PMC9231845 DOI: 10.1002/anbr.202100122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Reoccurring seasonal flu epidemics and occasional pandemics are among the most severe threats to public health. Current seasonal influenza vaccines provide limited protection against drifted circulating strains and no protection against influenza pandemics. Next-generation influenza vaccines, designated as universal influenza vaccines, should be safe, affordable, and elicit long-lasting cross-protective influenza immunity. Nanotechnology plays a critical role in the development of such novel vaccines. Engineered nanoparticles can incorporate multiple advantageous properties into the same nanoparticulate platforms to improve vaccine potency and breadth. These immunological properties include virus-like biomimicry, high antigen-load, controlled antigen release, targeted delivery, and induction of innate signaling pathways. Many nanoparticle influenza vaccines have shown promising results in generating potent and broadly protective immune responses. This review will summarize the necessity and characteristics of next-generation influenza vaccines and the immunological correlates of broad influenza immunity and focus on how cutting-edge nanoparticle technology contributes to such vaccine development. The review will give new insights into the rational design of nanoparticle universal vaccines to combat influenza epidemics and pandemics.
Collapse
Affiliation(s)
- Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia 30303, USA
| |
Collapse
|
9
|
Iyer S, Yadav R, Agarwal S, Tripathi S, Agarwal R. Bioengineering Strategies for Developing Vaccines against Respiratory Viral Diseases. Clin Microbiol Rev 2022; 35:e0012321. [PMID: 34788128 PMCID: PMC8597982 DOI: 10.1128/cmr.00123-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Respiratory viral pathogens like influenza and coronaviruses such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have caused outbreaks leading to millions of deaths. Vaccinations are, to date, the best and most economical way to control such outbreaks and have been highly successful for several pathogens. Currently used vaccines for respiratory viral pathogens are primarily live attenuated or inactivated and can risk reversion to virulence or confer inadequate immunity. The recent trend of using potent biomolecules like DNA, RNA, and protein antigenic components to synthesize vaccines for diseases has shown promising results. Still, it remains challenging to translate due to their high susceptibility to degradation during storage and after delivery. Advances in bioengineering technology for vaccine design have made it possible to control the physicochemical properties of the vaccines for rapid synthesis, heightened antigen presentation, safer formulations, and more robust immunogenicity. Bioengineering techniques and materials have been used to synthesize several potent vaccines, approved or in trials, against coronavirus disease 2019 (COVID-19) and are being explored for influenza, SARS, and Middle East respiratory syndrome (MERS) vaccines as well. Here, we review bioengineering strategies such as the use of polymeric particles, liposomes, and virus-like particles in vaccine development against influenza and coronaviruses and the feasibility of adopting these technologies for clinical use.
Collapse
Affiliation(s)
- Shalini Iyer
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Rajesh Yadav
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Smriti Agarwal
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Shashank Tripathi
- Department of Microbiology and Cell Biology, Center for Infectious Disease Research, Indian Institute of Science, Bengaluru, India
| | - Rachit Agarwal
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
10
|
Akache B, Stark FC, Agbayani G, Renner TM, McCluskie MJ. Adjuvants: Engineering Protective Immune Responses in Human and Veterinary Vaccines. Methods Mol Biol 2022; 2412:179-231. [PMID: 34918246 DOI: 10.1007/978-1-0716-1892-9_9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Adjuvants are key components of many vaccines, used to enhance the level and breadth of the immune response to a target antigen, thereby enhancing protection from the associated disease. In recent years, advances in our understanding of the innate and adaptive immune systems have allowed for the development of a number of novel adjuvants with differing mechanisms of action. Herein, we review adjuvants currently approved for human and veterinary use, describing their use and proposed mechanisms of action. In addition, we will discuss additional promising adjuvants currently undergoing preclinical and/or clinical testing.
Collapse
Affiliation(s)
- Bassel Akache
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Felicity C Stark
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Gerard Agbayani
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Tyler M Renner
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Michael J McCluskie
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada.
| |
Collapse
|
11
|
Jaber N, Al‐Remawi M, Al‐Akayleh F, Al‐Muhtaseb N, Al‐Adham ISI, Collier PJ. A review of the antiviral activity of Chitosan, including patented applications and its potential use against COVID-19. J Appl Microbiol 2022; 132:41-58. [PMID: 34218488 PMCID: PMC8447037 DOI: 10.1111/jam.15202] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022]
Abstract
Chitosan is an abundant organic polysaccharide, which can be relatively easily obtained by chemical modification of animal or fungal source materials. Chitosan and its derivatives have been shown to exhibit direct antiviral activity, to be useful vaccine adjuvants and to have potential anti-SARS-CoV-2 activity. This thorough and timely review looks at the recent history of investigations into the role of chitosan and its derivatives as an antiviral agent and proposes a future application in the treatment of endemic SARS-CoV-2.
Collapse
Affiliation(s)
- Nisrein Jaber
- Faculty of PharmacyAl‐Ahliyya Amman UniversityAmmanJordan
| | - Mayyas Al‐Remawi
- Faculty of Pharmacy & Medical SciencesUniversity of PetraAmmanJordan
| | - Faisal Al‐Akayleh
- Faculty of Pharmacy & Medical SciencesUniversity of PetraAmmanJordan
| | - Najah Al‐Muhtaseb
- Faculty of Pharmacy & Medical SciencesUniversity of PetraAmmanJordan
| | | | | |
Collapse
|
12
|
Xu H, Cai L, Hufnagel S, Cui Z. Intranasal vaccine: Factors to consider in research and development. Int J Pharm 2021; 609:121180. [PMID: 34637935 DOI: 10.1016/j.ijpharm.2021.121180] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023]
Abstract
Most existing vaccines for human use are administered by needle-based injection. Administering vaccines needle-free intranasally has numerous advantages over by needle-based injection, but there are only a few intranasal vaccines that are currently approved for human use, and all of them are live attenuated influenza virus vaccines. Clearly, there are immunological as well as non-immunological challenges that prevent vaccine developers from choosing the intranasal route of administration. We reviewed current approved intranasal vaccines and pipelines and described the target of intranasal vaccines, i.e. nose and lymphoid tissues in the nasal cavity. We then analyzed factors unique to intranasal vaccines that need to be considered when researching and developing new intranasal vaccines. We concluded that while the choice of vaccine formulations, mucoadhesives, mucosal and epithelial permeation enhancers, and ligands that target M-cells are important, safe and effective intranasal mucosal vaccine adjuvants are needed to successfully develop an intranasal vaccine that is not based on live-attenuated viruses or bacteria. Moreover, more effective intranasal vaccine application devices that can efficiently target a vaccine to lymphoid tissues in the nasal cavity as well as preclinical animal models that can better predict intranasal vaccine performance in clinical trials are needed to increase the success rate of intranasal vaccines in clinical trials.
Collapse
Affiliation(s)
- Haiyue Xu
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Lucy Cai
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Stephanie Hufnagel
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States.
| |
Collapse
|
13
|
Kumar A, Dhadi SR, Mai N, Taylor C, Roy JW, Barnett DA, Lewis SM, Ghosh A, Ouellette RJ. The polysaccharide chitosan facilitates the isolation of small extracellular vesicles from multiple biofluids. J Extracell Vesicles 2021; 10:e12138. [PMID: 34478244 PMCID: PMC8409086 DOI: 10.1002/jev2.12138] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 07/08/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022] Open
Abstract
Several studies have demonstrated the potential uses of extracellular vesicles (EVs) for liquid biopsy-based diagnostic tests and therapeutic applications; however, clinical use of EVs presents a challenge as many currently-available EV isolation methods have limitations related to efficiency, purity, and complexity of the methods. Moreover, many EV isolation methods do not perform efficiently in all biofluids due to their differential physicochemical properties. Thus, there continues to be a need for novel EV isolation methods that are simple, robust, non-toxic, and/or clinically-amenable. Here we demonstrate a rapid and efficient method for small extracellular vesicle (sEV) isolation that uses chitosan, a linear cationic polyelectrolyte polysaccharide that exhibits biocompatibility, non-immunogenicity, biodegradability, and low toxicity. Chitosan-precipitated material was characterized using Western blotting, nanoparticle tracking analysis (NTA), transmission electron microscopy (TEM), and relevant proteomic-based gene ontology analyses. We find that chitosan facilitates the isolation of sEVs from multiple biofluids, including cell culture-conditioned media, human urine, plasma and saliva. Overall, our data support the potential for chitosan to isolate a population of sEVs from a variety of biofluids and may have the potential to be a clinically amenable sEV isolation method.
Collapse
Affiliation(s)
- Awanit Kumar
- Atlantic Cancer Research InstituteMonctonNew BrunswickCanada
| | | | - Ngoc‐Nu Mai
- Atlantic Cancer Research InstituteMonctonNew BrunswickCanada
| | | | - Jeremy W. Roy
- Atlantic Cancer Research InstituteMonctonNew BrunswickCanada
- Beatrice Hunter Cancer Research InstituteHalifaxNova ScotiaCanada
| | - David A. Barnett
- Atlantic Cancer Research InstituteMonctonNew BrunswickCanada
- Department of Chemistry and BiochemistryMount Allison UniversitySackvilleNew BrunswickCanada
| | - Stephen M. Lewis
- Atlantic Cancer Research InstituteMonctonNew BrunswickCanada
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNew BrunswickCanada
- Beatrice Hunter Cancer Research InstituteHalifaxNova ScotiaCanada
| | - Anirban Ghosh
- Atlantic Cancer Research InstituteMonctonNew BrunswickCanada
| | - Rodney J. Ouellette
- Atlantic Cancer Research InstituteMonctonNew BrunswickCanada
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNew BrunswickCanada
| |
Collapse
|
14
|
Mousavi H. A comprehensive survey upon diverse and prolific applications of chitosan-based catalytic systems in one-pot multi-component synthesis of heterocyclic rings. Int J Biol Macromol 2021; 186:1003-1166. [PMID: 34174311 DOI: 10.1016/j.ijbiomac.2021.06.123] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 05/16/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022]
Abstract
Heterocyclic compounds are among the most prestigious and valuable chemical molecules with diverse and magnificent applications in various sciences. Due to the remarkable and numerous properties of the heterocyclic frameworks, the development of efficient and convenient synthetic methods for the preparation of such outstanding compounds is of great importance. Undoubtedly, catalysis has a conspicuous role in modern chemical synthesis and green chemistry. Therefore, when designing a chemical reaction, choosing and or preparing powerful and environmentally benign simple catalysts or complicated catalytic systems for an acceleration of the chemical reaction is a pivotal part of work for synthetic chemists. Chitosan, as a biocompatible and biodegradable pseudo-natural polysaccharide is one of the excellent choices for the preparation of suitable catalytic systems due to its unique properties. In this review paper, every effort has been made to cover all research articles in the field of one-pot synthesis of heterocyclic frameworks in the presence of chitosan-based catalytic systems, which were published roughly by the first quarter of 2020. It is hoped that this review paper can be a little help to synthetic scientists, methodologists, and catalyst designers, both on the laboratory and industrial scales.
Collapse
Affiliation(s)
- Hossein Mousavi
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia, Iran.
| |
Collapse
|
15
|
Ghosh T, Singh R, Nesamma AA, Jutur PP. Marine Polysaccharides: Properties and Applications. POLYSACCHARIDES 2021. [DOI: 10.1002/9781119711414.ch3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
16
|
Abstract
Influenza viruses cause seasonal epidemics and represent a pandemic risk. With current vaccine methods struggling to protect populations against emerging strains, there is a demand for a next-generation flu vaccine capable of providing broad protection. Recombinant biotechnology, combined with nanomedicine techniques, could address this demand by increasing immunogenicity and directing immune responses toward conserved antigenic targets on the virus. Various nanoparticle candidates have been tested for use in vaccines, including virus-like particles, protein and carbohydrate nanoconstructs, antigen-carrying lipid particles, and synthetic and inorganic particles modified for antigen presentation. These methods have yielded some promising results, including protection in animal models against antigenically distinct influenza strains, production of antibodies with broad reactivity, and activation of potent T cell responses. Based on the evidence of current research, it is feasible that the next generation of influenza vaccines will combine recombinant antigens with nanoparticle carriers.
Collapse
MESH Headings
- Animals
- Antigens, Viral/administration & dosage
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Disease Models, Animal
- Drug Carriers/chemistry
- Humans
- Immunogenicity, Vaccine
- Influenza A virus/genetics
- Influenza A virus/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza Vaccines/pharmacokinetics
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Nanoparticles/chemistry
- Protein Engineering
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Recombinant Proteins/pharmacokinetics
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Proteins/administration & dosage
- Viral Proteins/genetics
- Viral Proteins/immunology
- Viral Proteins/pharmacokinetics
Collapse
Affiliation(s)
- Zachary R Sia
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Matthew S Miller
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Diseases Research, McMaster Immunology Research Centre, McMaster University, Ontario, Canada
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York, USA
| |
Collapse
|
17
|
Intranasal Vaccine Using P10 Peptide Complexed within Chitosan Polymeric Nanoparticles as Experimental Therapy for Paracoccidioidomycosis in Murine Model. J Fungi (Basel) 2020; 6:jof6030160. [PMID: 32887256 PMCID: PMC7560165 DOI: 10.3390/jof6030160] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023] Open
Abstract
Paracoccidioidomycosis (PCM) is a granulomatous fungal disease caused by the dimorphic fungal species of Paracoccidioides, which mainly affects the lungs. Modern strategies for the treatment and/or prevention of PCM are based on a Th1-type immune response, which is important for controlling the disease. One of the most studied candidates for a vaccine is the P10 peptide, derived from the 43 kDa glycoprotein of Paracoccidioides brasiliensis. In order to improve its immune modulatory effect, the P10 peptide was associated with a chitosan-conjugated nanoparticle. The nanoparticles presented 220 nm medium size, poly dispersion index (PDI) below 0.5, zeta potential of +20 mV and encapsulation efficiency around 90%. The nanoparticles' non-toxicity was verified by hemolytic test and cell viability using murine macrophages. The nanoparticles were stable and presented physicochemical characteristics desirable for biological applications, reducing the fungal load and the usual standard concentration of the peptide from 4 to 20 times.
Collapse
|
18
|
Chitosan-based particulate systems for drug and vaccine delivery in the treatment and prevention of neglected tropical diseases. Drug Deliv Transl Res 2020; 10:1644-1674. [PMID: 32588282 DOI: 10.1007/s13346-020-00806-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neglected tropical diseases (NTDs) are a diverse group of infections which are difficult to prevent or control, affecting impoverished communities that are unique to tropical or subtropical regions. In spite of the low number of drugs that are currently used for the treatment of these diseases, progress on new drug discovery and development for NTDs is still very limited. Therefore, strategies on the development of new delivery systems for current drugs have been the main focus of formulators to provide improved efficacy and safety. In recent years, particulate delivery systems at micro- and nanosize, including polymeric micro- and nanoparticles, liposomes, solid lipid nanoparticles, metallic nanoparticles, and nanoemulsions, have been widely investigated in the treatment and control of NTDs. Among these polymers used for the preparation of such systems is chitosan, which is a marine biopolymer obtained from the shells of crustaceans. Chitosan has been investigated as a delivery system due to the versatility of its physicochemical properties as well as bioadhesive and penetration-enhancing properties. Furthermore, chitosan can be also used to improve treatment due to its bioactive properties such as antimicrobial, tissue regeneration, etc. In this review, after giving a brief introduction to neglected diseases and particulate systems developed for the treatment and control of NTDs, the chitosan-based systems will be described in more detail and the recent studies on these systems will be reviewed. Graphical abstract.
Collapse
|
19
|
El-Sayed A, Kamel M. Advanced applications of nanotechnology in veterinary medicine. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:19073-19086. [PMID: 30547342 DOI: 10.1007/s11356-018-3913-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 12/04/2018] [Indexed: 06/09/2023]
Abstract
The invention of new techniques to manipulate materials at their nanoscale had an evolutionary effect on various medical sciences. At the time, there are thousands of nanomaterials which can be divided according to their shape, origin, or their application. The nanotechnology provided new solutions for old problems. In medical sciences, they are used for diagnostic or therapeutic purposes. They can also be applied in the preparation of nanovaccines and nanoadjuvants. Their use in the treatment of cancer and in gene therapy opened the door for a new era in medicine. Recently, various applications of nanotechnology started to find their way in the veterinary sector. They increasingly invade animal therapeutics, diagnostics, production of veterinary vaccines, farm disinfectants, for animal breeding and reproduction, and even the field of animal nutrition. Their replacement of commonly used antibiotics directly reflects on the public health. By so doing, they minimize the problem of drug resistance in both human and veterinary medicine, and the problem of drug residues in milk and meat. In addition, they have a great economic impact, by minimizing the amounts of discarded milk and the number of culled calves in dairy herds. Nanotechnology was also applied to develop pet care products and hygienic articles. The present review discusses the advantage of using nanomaterials compared to their counterparts, the various classes of nanoparticles, and illustrates the applications and the role of nanotechnology in the field of veterinary medicine.
Collapse
Affiliation(s)
- Amr El-Sayed
- Faculty of Veterinary Medicine, Department of Medicine and Infectious Diseases, Cairo University, Giza, Egypt
| | - Mohamed Kamel
- Faculty of Veterinary Medicine, Department of Medicine and Infectious Diseases, Cairo University, Giza, Egypt.
| |
Collapse
|
20
|
Dewangan HK. Rational application of nanoadjuvant for mucosal vaccine delivery system. J Immunol Methods 2020; 481-482:112791. [PMID: 32387695 DOI: 10.1016/j.jim.2020.112791] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/20/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022]
Abstract
The surface of the mucosa is the biggest path through which pathogens enter the human body. We need an understanding of mucosal immune systems to use vaccines that generate protective mucosal and systemic immunity to regulate the outbreak of various infectious diseases. The better impact of the mucosal vaccine over traditional injectable vaccines are that not only do they induce efficient immune reactions to the mucosa but they are also comfortable in physical aspect & psychological aspect. The material of the vaccine includes pathogens antigens and adjuvants, which enable vaccination to be effective. Vaccines are classified into different criteria, including the used vaccine material and method of administration. Vaccines have traditionally been injected through a needle. However, as most of the pathogens first infect the mucosal surfaces, and growing interest is expressed in establishing protective immunity from the mucosa, which is accomplished through mucosal paths through vaccinosis. To improve the existing vaccines further, innovative strategies derived from interdisciplinary scientific research will need to develop new vaccine production, storage, and delivery systems. A distinctive & vast research and development platform has been set up for the growth of the next generation of mucosal vaccinations. The latest science and technological advancement in the areas of molecular biology, bio and chemical engineering, genome and system biology has provided accumulated understanding of the inborn and acquired multi-dimensional immune system. This review summarizes recent developments in the use of mucosal vaccines and their associated nanoadjuvants for the control of infectious diseases.
Collapse
Affiliation(s)
- Hitesh Kumar Dewangan
- Institute of Pharmaceutical Research (IPR), GLA University, Mathura, NH-2, Mathura Delhi Road, Chaumuhan Mathura, Uttar Pradesh 281406, India.
| |
Collapse
|
21
|
|
22
|
Youssef FS, El-Banna HA, Elzorba HY, Galal AM. Application of some nanoparticles in the field of veterinary medicine. Int J Vet Sci Med 2019; 7:78-93. [PMID: 32010725 PMCID: PMC6968591 DOI: 10.1080/23144599.2019.1691379] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/31/2022] Open
Abstract
Nanotechnology is a fast-growing technology that plays an important great impact on various fields of therapeutic applications. It is capable for solving several problems related to animal health and production. There are different nano-systems such as liposomes, metallic nanoparticles, polymeric micelles, polymeric nanospheres, functionalized fullerenes, carbon nanotubes, dendrimers, polymer-coated nanocrystals and nanoshells. In this review, we mentioned different methods for the preparation and characterization of nanoparticles. This review is concerned mainly on nanoparticle systems for antibiotic delivery which suffer from poor bioavailability and many side effects. Nanoparticles are characterized by many features include their minimal size, colossal surface zone to mass extent. The development of antimicrobials in nanoparticle systems is considered an excellent alternative delivery system for antimicrobials for the treatment of microbial diseases by increasing therapeutic effect and overcoming the side effects. In this paper, we reviewed some antimicrobial nanoparticle preparations and we focused on florfenicol and neomycin nanoparticle preparations as well as chitosan and silver nanoparticles preparations to prepare, characterize and compare their different pharmacological effects.
Collapse
Affiliation(s)
- Fady Sayed Youssef
- Pharmacology department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Hossny Awad El-Banna
- Pharmacology department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | | | - Ahmed Mohamed Galal
- Pharmacology department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
23
|
Komati S, Swain S, Rao MEB, Jena BR, Dasi V. Mucoadhesive Multiparticulate Drug Delivery Systems: An Extensive Review of Patents. Adv Pharm Bull 2019; 9:521-538. [PMID: 31857957 PMCID: PMC6912179 DOI: 10.15171/apb.2019.062] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 06/18/2019] [Accepted: 06/24/2019] [Indexed: 11/09/2022] Open
Abstract
Innovations in pharmaceutical research are striving for designing newer drug therapies to eradicate deadly diseases. Strategies for such inventions always flourish with keys and objectives of minimal adverse effects and effective treatment. Recent trends in pharmaceutical technology specify that mucoadhesive drug delivery system is particularly appropriate than oral control release, for getting local systematic delivery of drugs in GIT for an extended interval of time at a predetermined rate. However, it is somehow expensive and unpleasant sensation for some patients, but still it is needful for getting short enzymatic activity, simple administration without pain and evasion of fast pass metabolism. Usually the vehicles employed in drug delivery of mucoadhesive system have a significant impact that draws further attention to potential benefits like improved bioavailability of therapeutic agents, extensive drug residence time at the site of administration and a comparatively faster drug uptake into the systemic circulation. The drug release from mucoadhesive multiparticulates is contingent on several types of factors comprising carrier need to produce the multiparticles and quantity of medication drug contained in them. Mucoadhesion is characterized by selected theories and mechanisms. Various strategies emergent in mucoadhesive multiparticulate drug delivery system (MMDDS) by in-vitro as well as ex-vivo description and characterization are also critically discussed. Apart from these, the primary focus during this review is to highlight current patents, clinical status, and regulatory policy for enhancement of mucoadhesive multi-particulate drug delivery system in the present scenario.
Collapse
Affiliation(s)
- Someshwar Komati
- Department of Pharmaceutics, University College of Pharmaceutical Sciences, Palamuru University, Mahaboobnagar, Telangana-509001, India
| | - Suryakanta Swain
- Southern Institute of Medical Sciences, College of Pharmacy, Mangaldas Nagar, Vijyawada Road, Guntur-522 001, Andhra Pradesh, India
| | - Muddana Eswara Bhanoji Rao
- Department of Pharmaceutics, Roland Institute of Pharmaceutical Sciences, Khodasinghi, Berhampur-760 010, Ganjam, Odisha, India
| | - Bikash Ranjan Jena
- Southern Institute of Medical Sciences, College of Pharmacy, Mangaldas Nagar, Vijyawada Road, Guntur-522 001, Andhra Pradesh, India
| | - Vishali Dasi
- Department of Pharmaceutics, University College of Pharmaceutical Sciences, Palamuru University, Mahaboobnagar, Telangana-509001, India
| |
Collapse
|
24
|
Calzas C, Chevalier C. Innovative Mucosal Vaccine Formulations Against Influenza A Virus Infections. Front Immunol 2019; 10:1605. [PMID: 31379823 PMCID: PMC6650573 DOI: 10.3389/fimmu.2019.01605] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/27/2019] [Indexed: 12/11/2022] Open
Abstract
Despite efforts made to develop efficient preventive strategies, infections with influenza A viruses (IAV) continue to cause serious clinical and economic problems. Current licensed human vaccines are mainly inactivated whole virus particles or split-virion administered via the parenteral route. These vaccines provide incomplete protection against IAV in high-risk groups and are poorly/not effective against the constant antigenic drift/shift occurring in circulating strains. Advances in mucosal vaccinology and in the understanding of the protective anti-influenza immune mechanisms suggest that intranasal immunization is a promising strategy to fight against IAV. To date, human mucosal anti-influenza vaccines consist of live attenuated strains administered intranasally, which elicit higher local humoral and cellular immune responses than conventional parenteral vaccines. However, because of inconsistent protective efficacy and safety concerns regarding the use of live viral strains, new vaccine candidates are urgently needed. To prime and induce potent and long-lived protective immune responses, mucosal vaccine formulations need to ensure the immunoavailability and the immunostimulating capacity of the vaccine antigen(s) at the mucosal surfaces, while being minimally reactogenic/toxic. The purpose of this review is to compile innovative delivery/adjuvant systems tested for intranasal administration of inactivated influenza vaccines, including micro/nanosized particulate carriers such as lipid-based particles, virus-like particles and polymers associated or not with immunopotentiatory molecules including microorganism-derived toxins, Toll-like receptor ligands and cytokines. The capacity of these vaccines to trigger specific mucosal and systemic humoral and cellular responses against IAV and their (cross)-protective potential are considered.
Collapse
Affiliation(s)
- Cynthia Calzas
- VIM, UR892, Equipe Virus Influenza, INRA, University PARIS-SACLAY, Jouy-en-Josas, France
| | - Christophe Chevalier
- VIM, UR892, Equipe Virus Influenza, INRA, University PARIS-SACLAY, Jouy-en-Josas, France
| |
Collapse
|
25
|
Cohen E, Merzendorfer H. Chitin/Chitosan: Versatile Ecological, Industrial, and Biomedical Applications. EXTRACELLULAR SUGAR-BASED BIOPOLYMERS MATRICES 2019; 12. [PMCID: PMC7115017 DOI: 10.1007/978-3-030-12919-4_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chitin is a linear polysaccharide of N-acetylglucosamine, which is highly abundant in nature and mainly produced by marine crustaceans. Chitosan is obtained by hydrolytic deacetylation. Both polysaccharides are renewable resources, simply and cost-effectively extracted from waste material of fish industry, mainly crab and shrimp shells. Research over the past five decades has revealed that chitosan, in particular, possesses unique and useful characteristics such as chemical versatility, polyelectrolyte properties, gel- and film-forming ability, high adsorption capacity, antimicrobial and antioxidative properties, low toxicity, and biocompatibility and biodegradability features. A plethora of chemical chitosan derivatives have been synthesized yielding improved materials with suggested or effective applications in water treatment, biosensor engineering, agriculture, food processing and storage, textile additives, cosmetics fabrication, and in veterinary and human medicine. The number of studies in this research field has exploded particularly during the last two decades. Here, we review recent advances in utilizing chitosan and chitosan derivatives in different technical, agricultural, and biomedical fields.
Collapse
Affiliation(s)
- Ephraim Cohen
- Department of Entomology, The Robert H. Smith Faculty of Agriculture Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Hans Merzendorfer
- School of Science and Technology, Institute of Biology – Molecular Biology, University of Siegen, Siegen, Germany
| |
Collapse
|
26
|
Brown HE, Esher SK, Alspaugh JA. Chitin: A "Hidden Figure" in the Fungal Cell Wall. Curr Top Microbiol Immunol 2019; 425:83-111. [PMID: 31807896 DOI: 10.1007/82_2019_184] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chitin and chitosan are two related polysaccharides that provide important structural stability to fungal cell walls. Often embedded deeply within the cell wall structure, these molecules anchor other components at the cell surface. Chitin-directed organization of the cell wall layers allows the fungal cell to effectively monitor and interact with the external environment. For fungal pathogens, this interaction includes maintaining cellular strategies to avoid excessive detection by the host innate immune system. In turn, mammalian and plant hosts have developed their own strategies to process fungal chitin, resulting in chitin fragments of varying molecular size. The size-dependent differences in the immune activation behaviors of variably sized chitin molecules help to explain how chitin and related chitooligomers can both inhibit and activate host immunity. Moreover, chitin and chitosan have recently been exploited for many biomedical applications, including targeted drug delivery and vaccine development.
Collapse
Affiliation(s)
- Hannah E Brown
- Department of Medicine, Department of Molecular Genetics and Microbiology, Duke University School of Medicine, 303 Sands Research Building, DUMC, 102359, Durham, 27710, NC, USA
| | - Shannon K Esher
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - J Andrew Alspaugh
- Department of Medicine, Department of Molecular Genetics and Microbiology, Duke University School of Medicine, 303 Sands Research Building, DUMC, 102359, Durham, 27710, NC, USA.
| |
Collapse
|
27
|
Schuerer N, Stein E, Inic-Kanada A, Ghasemian E, Stojanovic M, Montanaro J, Bintner N, Hohenadl C, Sachsenhofer R, Barisani-Asenbauer T. Effects of chitosan and chitosan N-acetylcysteine solutions on conjunctival epithelial cells. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.xjec.2018.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
28
|
Corthésy B, Bioley G. Lipid-Based Particles: Versatile Delivery Systems for Mucosal Vaccination against Infection. Front Immunol 2018; 9:431. [PMID: 29563912 PMCID: PMC5845866 DOI: 10.3389/fimmu.2018.00431] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/19/2018] [Indexed: 12/19/2022] Open
Abstract
Vaccination is the process of administering immunogenic formulations in order to induce or harness antigen (Ag)-specific antibody and T cell responses in order to protect against infections. Important successes have been obtained in protecting individuals against many deleterious pathological situations after parenteral vaccination. However, one of the major limitations of the current vaccination strategies is the administration route that may not be optimal for the induction of immunity at the site of pathogen entry, i.e., mucosal surfaces. It is now well documented that immune responses along the genital, respiratory, or gastrointestinal tracts have to be elicited locally to ensure efficient trafficking of effector and memory B and T cells to mucosal tissues. Moreover, needle-free mucosal delivery of vaccines is advantageous in terms of safety, compliance, and ease of administration. However, the quest for mucosal vaccines is challenging due to (1) the fact that Ag sampling has to be performed across the epithelium through a relatively limited number of portals of entry; (2) the deleterious acidic and proteolytic environment of the mucosae that affect the stability, integrity, and retention time of the applied Ags; and (3) the tolerogenic environment of mucosae, which requires the addition of adjuvants to elicit efficient effector immune responses. Until now, only few mucosally applicable vaccine formulations have been developed and successfully tested. In animal models and clinical trials, the use of lipidic structures such as liposomes, virosomes, immune stimulating complexes, gas-filled microbubbles and emulsions has proven efficient for the mucosal delivery of associated Ags and the induction of local and systemic immune reponses. Such particles are suitable for mucosal delivery because they protect the associated payload from degradation and deliver concentrated amounts of Ags via specialized sampling cells (microfold cells) within the mucosal epithelium to underlying antigen-presenting cells. The review aims at summarizing recent development in the field of mucosal vaccination using lipid-based particles. The modularity ensured by tailoring the lipidic design and content of particles, and their known safety as already established in humans, make the continuing appraisal of these vaccine candidates a promising development in the field of targeted mucosal vaccination.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory, Division of Immunology and Allergy, Centre des Laboratoires d'Epalinges, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Gilles Bioley
- R&D Laboratory, Division of Immunology and Allergy, Centre des Laboratoires d'Epalinges, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| |
Collapse
|
29
|
Halder SK, Mondal KC. Microbial Valorization of Chitinous Bioresources for Chitin Extraction and Production of Chito-Oligomers and N-Acetylglucosamine: Trends, Perspectives and Prospects. Microb Biotechnol 2018. [DOI: 10.1007/978-981-10-7140-9_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
|
30
|
|
31
|
Zhu G, Falahat R, Wang K, Mailloux A, Artzi N, Mulé JJ. Tumor-Associated Tertiary Lymphoid Structures: Gene-Expression Profiling and Their Bioengineering. Front Immunol 2017; 8:767. [PMID: 28713385 PMCID: PMC5491937 DOI: 10.3389/fimmu.2017.00767] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/16/2017] [Indexed: 11/15/2022] Open
Abstract
Tertiary lymphoid structures (TLSs) have been identified in the parenchyma and/or in the peripheral margins of human solid tumors. Uncovering the functional nature of these structures is the subject of much intensive investigation. Studies have shown a direct correlation of the presence of human tumor-localized TLS and better patient outcome (e.g., increase in overall survival) in certain solid tumor histologies, but not all. We had identified a tumor-derived immune gene-expression signature, encoding 12 distinct chemokines, which could reliably identify the presence of TLSs, of different degrees, in various human solid tumors. We are focused on understanding the influence of TLSs on the tumor microenvironment and leveraging this understanding to both manipulate the antitumor immune response and potentially enhance immunotherapy applications. Moreover, as not all human solid tumors show the presence of these lymphoid structures, we are embarking on bioengineering approaches to design and build “designer” TLSs to address, and potentially overcome, an unmet medical need in cancer patients whose tumors lack such lymphoid structures.
Collapse
Affiliation(s)
- Genyuan Zhu
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
| | - Rana Falahat
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
| | - Kui Wang
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Adam Mailloux
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
| | - Natalie Artzi
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - James J Mulé
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States.,Cutaneous Oncology Department, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
32
|
Schulze K, Ebensen T, Riese P, Prochnow B, Lehr CM, Guzmán CA. New Horizons in the Development of Novel Needle-Free Immunization Strategies to Increase Vaccination Efficacy. Curr Top Microbiol Immunol 2017; 398:207-234. [PMID: 27370343 DOI: 10.1007/82_2016_495] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The young twenty-first century has already brought several medical advances, such as a functional artificial human liver created from stem cells, improved antiviral (e.g., against HIV) and cancer (e.g., against breast cancer) therapies, interventions controlling cardiovascular diseases, and development of new and optimized vaccines (e.g., HPV vaccine). However, despite this substantial progress and the achievements of the last century, humans still suffer considerably from diseases, especially from infectious diseases. Thus, almost one-fourth of all deaths worldwide are caused directly or indirectly by infectious agents. Although vaccination has led to the control of many diseases, including smallpox, diphtheria, and tetanus, emerging diseases are still not completely contained. Furthermore, pathogens such as Bordetella pertussis undergo alterations making adaptation of the respective vaccine necessary. Moreover, insufficient implementation of vaccination campaigns leads to re-emergence of diseases which were believed to be already under control (e.g., poliomyelitis). Therefore, novel vaccination strategies need to be developed in order to meet the current challenges including lack of compliance, safety issues, and logistic constraints. In this context, mucosal and transdermal approaches constitute promising noninvasive vaccination strategies able to match these demands.
Collapse
Affiliation(s)
- Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.
| | - Peggy Riese
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Blair Prochnow
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery, Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Braunschweig, Germany.,Department of Pharmacy, Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| |
Collapse
|
33
|
Lone NA, Spackman E, Kapczynski D. Immunologic evaluation of 10 different adjuvants for use in vaccines for chickens against highly pathogenic avian influenza virus. Vaccine 2017; 35:3401-3408. [DOI: 10.1016/j.vaccine.2017.05.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/17/2017] [Accepted: 05/03/2017] [Indexed: 12/16/2022]
|
34
|
Park J, Ramanathan R, Pham L, Woodrow KA. Chitosan enhances nanoparticle delivery from the reproductive tract to target draining lymphoid organs. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2015-2025. [PMID: 28435136 DOI: 10.1016/j.nano.2017.04.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/29/2017] [Accepted: 04/11/2017] [Indexed: 12/21/2022]
Abstract
To prime adaptive immune responses from the female reproductive tract (FRT), particulate antigens must be transported to draining lymph nodes (dLNs) since there are no local organized lymphoid structures equivalent to those found in the respiratory or gastrointestinal tracts. However, little is known about how to safely and effectively navigate successive barriers to transport such as crossing the epithelium and gaining access to migratory cells and lymphatic drainage that provide entry into dLNs. Here, we demonstrate that intravaginal pre-treatment with chitosan significantly facilitates translocation of nanoparticles (NPs) across the multilayered vaginal epithelium to target dLNs. In addition, chitosan pre-treatment was found to enhance NP associations with immunogenic antigen presenting cells in the vaginal submucosa. These observations indicate that chitosan may have great potential as an adjuvant for both local and systemic protective immunity against viral infections in the FRT.
Collapse
Affiliation(s)
- Jaehyung Park
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Renuka Ramanathan
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Linhchi Pham
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
35
|
Caetano LA, Figueiredo L, Almeida AJ, Gonçalves LMD. BCG-loaded chitosan microparticles: interaction with macrophages and preliminary in vivo studies. J Microencapsul 2017; 34:203-217. [PMID: 28378596 DOI: 10.1080/02652048.2017.1316325] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The aim of this study was to develop a novel BCG-loaded chitosan vaccine with high association efficiency which can afford efficient interaction with APC and elicit local and Th1-type-specific immune response after intranasal administration. Chitosan-suspended BCG and BCG-loaded chitosan-alginate microparticles were prepared by ionotropic gelation. Interaction with APC was evaluated by fluorescence microscopy using rBCG-GFP. Specific immune responses were evaluated following intranasal immunisation of mice. Cellular uptake was approximately two-fold higher for chitosan-suspended BCG. A single dose of BCG-loaded microparticles or chitosan-suspended BCG by intranasal route improved Th1-type response compared with subcutaneous BCG. Chitosan-suspended BCG originated the highest mucosal response in the lungs by intranasal route. These positive results indicate that the proposed approach of whole live BCG microencapsulation in chitosan-alginate for intranasal immunisation was successful in allowing efficient interaction with APC, while improving the cellular immune response, which is of interest for local immunisation against tuberculosis.
Collapse
Affiliation(s)
- Liliana Aranha Caetano
- a Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy , University of Lisbon , Lisbon , Portugal.,b Department of Ciências e Tecnologias Laboratoriais e Saúde Comunitária, ESTeSL - Escola Superior de Tecnologia da Saúde de Lisboa , Instituto Politécnico de Lisboa , Lisbon , Portugal
| | - Lara Figueiredo
- a Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy , University of Lisbon , Lisbon , Portugal
| | - António J Almeida
- a Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy , University of Lisbon , Lisbon , Portugal
| | - L M D Gonçalves
- a Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy , University of Lisbon , Lisbon , Portugal
| |
Collapse
|
36
|
Molecular Weight-Dependent Immunostimulative Activity of Low Molecular Weight Chitosan via Regulating NF-κB and AP-1 Signaling Pathways in RAW264.7 Macrophages. Mar Drugs 2016; 14:md14090169. [PMID: 27657093 PMCID: PMC5039540 DOI: 10.3390/md14090169] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/12/2016] [Accepted: 09/13/2016] [Indexed: 11/17/2022] Open
Abstract
Chitosan and its derivatives such as low molecular weight chitosans (LMWCs) have been found to possess many important biological properties, such as antioxidant and antitumor effects. In our previous study, LMWCs were found to elicit a strong immunomodulatory response in macrophages dependent on molecular weight. Herein we further investigated the molecular weight-dependent immunostimulative activity of LMWCs and elucidated its mechanism of action on RAW264.7 macrophages. LMWCs (3 kDa and 50 kDa of molecular weight) could significantly enhance the mRNA expression levels of COX-2, IL-10 and MCP-1 in a molecular weight and concentration-dependent manner. The results suggested that LMWCs elicited a significant immunomodulatory response, which was dependent on the dose and the molecular weight. Regarding the possible molecular mechanism of action, LMWCs promoted the expression of the genes of key molecules in NF-κB and AP-1 pathways, including IKKβ, TRAF6 and JNK1, and induced the phosphorylation of protein IKBα in RAW264.7 macrophage. Moreover, LMWCs increased nuclear translocation of p65 and activation of activator protein-1 (AP-1, C-Jun and C-Fos) in a molecular weight-dependent manner. Taken together, our findings suggested that LMWCs exert immunostimulative activity via activation of NF-κB and AP-1 pathways in RAW264.7 macrophages in a molecular weight-dependent manner and that 3 kDa LMWC shows great potential as a novel agent for the treatment of immune suppression diseases and in future vaccines.
Collapse
|
37
|
Kavaliauskis A, Arnemo M, Speth M, Lagos L, Rishovd AL, Estepa A, Griffiths G, Gjøen T. Protective effect of a recombinant VHSV-G vaccine using poly(I:C) loaded nanoparticles as an adjuvant in zebrafish (Danio rerio) infection model. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 61:248-257. [PMID: 27084059 DOI: 10.1016/j.dci.2016.04.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 04/09/2016] [Accepted: 04/09/2016] [Indexed: 06/05/2023]
Abstract
There is a constant need to increase the efficiency of vaccines in the aquaculture industry. Although several nano-based vaccine formulations have been reported, to the best of our knowledge so far only one of them have been implemented in the industry. Here we report on chitosan-poly(I:C) nanoparticles (NPs) that could be used as a non-specific adjuvant in antiviral vaccines in aquaculture. We have characterized the physical parameters of the NPs, studied the in vivo and in vitro bio-distribution of fluorescent NPs and verified NP uptake by zebrafish leucocytes. We used the zebrafish model to test the protective efficiency of the recombinant glycoprotein G (rgpG) of VHSV compared to inactivated whole virus (iV) against VHSV using NPs as an adjuvant in both formulations. In parallel we tested free poly(I:C) and rgpG (pICrgpG), and free chitosan and rgpG (CSrgpG) vaccine formulations. While the iV group (with NP adjuvant) provided the highest overall survival, all vaccine formulations with poly(I:C) provided a significant protection against VHSV; possibly through an early induction of an anti-viral state. Our results suggest that chitosan-poly(I:C) NPs are a promising adjuvant candidate for future vaccine formulations.
Collapse
Affiliation(s)
- Arturas Kavaliauskis
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316 Oslo, Norway
| | - Marianne Arnemo
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316 Oslo, Norway
| | - Martin Speth
- Department of Biosciences, University of Oslo, P.O. Box 1066 Blindern, 0371 Oslo, Norway
| | - Leidy Lagos
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316 Oslo, Norway
| | - Anne-Lise Rishovd
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316 Oslo, Norway
| | | | - Gareth Griffiths
- Department of Biosciences, University of Oslo, P.O. Box 1066 Blindern, 0371 Oslo, Norway
| | - Tor Gjøen
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316 Oslo, Norway.
| |
Collapse
|
38
|
Calcium Carbonate-Based Mucoadhesive Microcontainers for Intranasal Delivery of Drugs Bypassing the Blood–Brain Barrier. BIONANOSCIENCE 2016. [DOI: 10.1007/s12668-016-0212-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
39
|
Zeng Z, Dai S, Jiao Y, Jiang L, Zhao Y, Wang B, Zong L. Mannosylated protamine as a novel DNA vaccine carrier for effective induction of anti-tumor immune responses. Int J Pharm 2016; 506:394-406. [DOI: 10.1016/j.ijpharm.2016.04.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 03/10/2016] [Accepted: 04/15/2016] [Indexed: 01/25/2023]
|
40
|
Amirnasr M, Fallah Tafti T, Sankian M, Rezaei A, Tafaghodi M. Immunization against HTLV-I with chitosan and tri-methylchitosan nanoparticles loaded with recombinant env23 and env13 antigens of envelope protein gp46. Microb Pathog 2016; 97:38-44. [PMID: 27235335 DOI: 10.1016/j.micpath.2016.05.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 05/08/2016] [Accepted: 05/17/2016] [Indexed: 10/21/2022]
Abstract
To prevent the spread of HTLV-I (Human T-lymphotropic virus type 1), a safe and effective vaccine is required. To increase immune responses against the peptide antigens can be potentiated with polymer-based nanoparticles, like chitosan (CHT) and trimethylchitosan (TMC), as delivery system/adjuvant. CHT and TMC nanoparticles loaded with recombinant proteins (env23 & env13) of gp46 were prepared by direct coating of antigens with positively charged polymers. The size of CHT and TMC nanoparticles (NPs) loaded with each antigen was about 400 nm. The physical stability of NPs was followed for 4 weeks. Both formulations showed to be stable for about 15 days. The immunogenicity of NPs loaded with antigens was studied after nasal and subcutaneous immunization in mice. Three immunizations (7.5 μg antigen) were performed with 2 weeks intervals. Two weeks after the last booster dose, sera IgG subtypes were measured. After subcutaneous administration, for both nanoparticulate antigens, serum IgG1 and IgGtotal levels were higher than antigen solution (P < 0.001). After nasal administration, for env23, IgG2a levels and IgG2a/IgG1 ratio was significantly higher than groups with subcutaneous administration (P < 0.001). Both nanoparticles showed good immunoadjuvant potential. Env23 antigen was a better candidate for vaccination against HTLV-I, as it induced higher cellular immune responses, compared with env13.
Collapse
Affiliation(s)
- Maryam Amirnasr
- Student Research Committee, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Tannan Fallah Tafti
- Student Research Committee, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mojtaba Sankian
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Abdorrahim Rezaei
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohsen Tafaghodi
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
41
|
Ahmed TA, Aljaeid BM. Preparation, characterization, and potential application of chitosan, chitosan derivatives, and chitosan metal nanoparticles in pharmaceutical drug delivery. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:483-507. [PMID: 26869768 PMCID: PMC4734734 DOI: 10.2147/dddt.s99651] [Citation(s) in RCA: 366] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Naturally occurring polymers, particularly of the polysaccharide type, have been used pharmaceutically for the delivery of a wide variety of therapeutic agents. Chitosan, the second abundant naturally occurring polysaccharide next to cellulose, is a biocompatible and biodegradable mucoadhesive polymer that has been extensively used in the preparation of micro-as well as nanoparticles. The prepared particles have been exploited as a potential carrier for different therapeutic agents such as peptides, proteins, vaccines, DNA, and drugs for parenteral and nonparenteral administration. Therapeutic agent-loaded chitosan micro- or nanoparticles were found to be more stable, permeable, and bioactive. In this review, we are highlighting the different methods of preparation and characterization of chitosan micro- and nanoparticles, while reviewing the pharmaceutical applications of these particles in drug delivery. Moreover, the roles of chitosan derivatives and chitosan metal nanoparticles in drug delivery have been illustrated.
Collapse
Affiliation(s)
- Tarek A Ahmed
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Bader M Aljaeid
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| |
Collapse
|
42
|
Wu N, Wen ZS, Xiang XW, Huang YN, Gao Y, Qu YL. Immunostimulative Activity of Low Molecular Weight Chitosans in RAW264.7 Macrophages. Mar Drugs 2015; 13:6210-25. [PMID: 26437419 PMCID: PMC4626685 DOI: 10.3390/md13106210] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/09/2015] [Accepted: 09/21/2015] [Indexed: 12/26/2022] Open
Abstract
Chitosan and its derivatives such as low molecular weight chitosans (LMWCs) have been reported to exert many biological activities, such as antioxidant and antitumor effects. However, complex and molecular weight dependent effects of chitosan remain controversial and the mechanisms that mediate these complex effects are still poorly defined. This study was carried out to investigate the immunostimulative effect of different molecular weight chitosan in RAW264.7 macrophages. Our data suggested that two LMWCs (molecular weight of 3 kDa and 50 kDa) both possessed immunostimulative activity, which was dependent on dose and, at the higher doses, also on the molecular weight. LMWCs could significantly enhance the the pinocytic activity, and induce the production of tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), interferon-γ (IFN-γ), nitric oxide (NO) and inducible nitric oxide synthase (iNOS) in a molecular weight and concentration-dependent manner. LMWCs were further showed to promote the expression of the genes including iNOS, TNF-α. Taken together, our findings suggested that LMWCs elicited significantly immunomodulatory response through up-regulating mRNA expression of proinflammatory cytokines and activated RAW264.7 macrophage in a molecular weight- and concentration-dependent manner.
Collapse
Affiliation(s)
- Ning Wu
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Food and Pharmacy College, Zhejiang Ocean University, Zhoushan 316000, China.
| | - Zheng-Shun Wen
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Food and Pharmacy College, Zhejiang Ocean University, Zhoushan 316000, China.
| | - Xing-Wei Xiang
- Zhejiang Marine Development Research Institute, Zhoushan 316000, China.
| | - Yan-Na Huang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China.
| | - Yang Gao
- School of Fishery, Zhejiang Ocean University, Zhoushan, 316000, China.
| | - You-Le Qu
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Food and Pharmacy College, Zhejiang Ocean University, Zhoushan 316000, China.
| |
Collapse
|
43
|
Savelkoul HFJ, Ferro VA, Strioga MM, Schijns VEJC. Choice and Design of Adjuvants for Parenteral and Mucosal Vaccines. Vaccines (Basel) 2015; 3:148-71. [PMID: 26344951 PMCID: PMC4494243 DOI: 10.3390/vaccines3010148] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 10/11/2014] [Accepted: 02/24/2015] [Indexed: 11/16/2022] Open
Abstract
The existence of pathogens that escape recognition by specific vaccines, the need to improve existing vaccines and the increased availability of therapeutic (non-infectious disease) vaccines necessitate the rational development of novel vaccine concepts based on the induction of protective cell-mediated immune responses. For naive T-cell activation, several signals resulting from innate and adaptive interactions need to be integrated, and adjuvants may interfere with some or all of these signals. Adjuvants, for example, are used to promote the immunogenicity of antigens in vaccines, by inducing a pro-inflammatory environment that enables the recruitment and promotion of the infiltration of phagocytic cells, particularly antigen-presenting cells (APC), to the injection site. Adjuvants can enhance antigen presentation, induce cytokine expression, activate APC and modulate more downstream adaptive immune reactions (vaccine delivery systems, facilitating immune Signal 1). In addition, adjuvants can act as immunopotentiators (facilitating Signals 2 and 3) exhibiting immune stimulatory effects during antigen presentation by inducing the expression of co-stimulatory molecules on APC. Together, these signals determine the strength of activation of specific T-cells, thereby also influencing the quality of the downstream T helper cytokine profiles and the differentiation of antigen-specific T helper populations (Signal 3). New adjuvants should also target specific (innate) immune cells in order to facilitate proper activation of downstream adaptive immune responses and homing (Signal 4). It is desirable that these adjuvants should be able to exert such responses in the context of mucosal administered vaccines. This review focuses on the understanding of the potential working mechanisms of the most well-known classes of adjuvants to be used effectively in vaccines.
Collapse
Affiliation(s)
- Huub F J Savelkoul
- Cell Biology and Immunology, Wageningen University, Wageningen, P.O. Box 338, 6700 AH Wageningen, The Netherlands.
| | - Valerie A Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - Marius M Strioga
- Department of Immunology, Center of Oncosurgery, National Cancer Institute, P. Baublio Str. 3b-321, LT-08406 Vilnius, Lithuania.
| | - Virgil E J C Schijns
- Cell Biology and Immunology, Wageningen University, Wageningen, P.O. Box 338, 6700 AH Wageningen, The Netherlands.
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
- ERC-Belgium and ERC-The Netherlands, 5374 RE Schaijk, The Netherlands.
| |
Collapse
|
44
|
Zhai Y, Zhou Y, Li X, Feng G. Immune-enhancing effect of nano-DNA vaccine encoding a gene of the prME protein of Japanese encephalitis virus and BALB/c mouse granulocyte-macrophage colony-stimulating factor. Mol Med Rep 2015; 12:199-209. [PMID: 25738258 PMCID: PMC4438877 DOI: 10.3892/mmr.2015.3419] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 01/02/2015] [Indexed: 11/12/2022] Open
Abstract
Plasmid-encoded granulocyte-macrophage colony-stimulating factor (GM-CSF) is an adjuvant for genetic vaccines; however, how GM-CSF enhances immunogenicity remains to be elucidated. In the present study, it was demonstrated that injection of a plasmid encoding the premembrane (prM) and envelope (E) protein of Japanese encephalitis virus and mouse GM-CSF (pJME/GM-CSF) into mouse muscle recruited large and multifocal conglomerates of macrophages and granulocytes, predominantly neutrophils. During the peak of the infiltration, an appreciable number of immature dendritic cells (DCs) appeared, although no T and B-cells was detected. pJME/GM-CSF increased the number of splenic DCs and the expression of major histocompatibility complex class II (MHCII) on splenic DC, and enhanced the antigenic capture, processing and presentation functions of splenic DCs, and the cell-mediated immunity induced by the vaccine. These findings suggested that the immune-enhancing effect by pJME/GM-CSF was associated with infiltrate size and the appearance of integrin αx (CD11c)+cells. Chitosan-pJME/GM-CSF nanoparticles, prepared by coacervation via intramuscular injection, outperformed standard pJME/GM-CSF administrations in DC recruitment, antigen processing and presentation, and vaccine enhancement. This revealed that muscular injection of chitosan-pJME/GM-CSF nanoparticles may enhance the immunoadjuvant properties of GM-CSF.
Collapse
Affiliation(s)
- Yongzhen Zhai
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yan Zhou
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Ximei Li
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Guohe Feng
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
45
|
InCVAX--a novel strategy for treatment of late-stage, metastatic cancers through photoimmunotherapy induced tumor-specific immunity. Cancer Lett 2015; 359:169-77. [PMID: 25633839 DOI: 10.1016/j.canlet.2015.01.029] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/20/2015] [Accepted: 01/21/2015] [Indexed: 12/31/2022]
Abstract
A novel, promising potential cancer vaccine strategy was proposed to use a two-injection procedure for solid tumors to prompt the immune system to identify and systemically eliminate primary and metastatic cancers. The two-injection procedure consists of local photothermal application on a selected tumor intended to liberate whole cell tumor antigens, followed by a local injection of an immunoadjuvant that consists of a semi-synthetic functionalized glucosamine polymer, N-dihydro-galacto-chitosan (GC), which is intended to activate antigen presenting cells and facilitate an increased uptake of tumor antigens. This strategy is thus proposed as an in situ autologous cancer vaccine (inCVAX) that may activate antigen presenting cells and expose them to tumor antigens in situ, with the intention of inducing a systemic tumor specific T-cell response. Here, the development of inCVAX for the treatment of metastatic cancers in the past decades is systematically reviewed. The antitumor immune responses of local photothermal treatment and immunological stimulation with GC are also discussed. This treatment approach is also commonly referred to as laser immunotherapy (LIT).
Collapse
|
46
|
Neimert-Andersson T, Binnmyr J, Enoksson M, Langebäck J, Zettergren L, Hällgren AC, Franzén H, Lind Enoksson S, Lafolie P, Lindberg A, Al-Tawil N, Andersson M, Singer P, Grönlund H, Gafvelin G. Evaluation of safety and efficacy as an adjuvant for the chitosan-based vaccine delivery vehicle ViscoGel in a single-blind randomised Phase I/IIa clinical trial. Vaccine 2014; 32:5967-74. [DOI: 10.1016/j.vaccine.2014.08.057] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 08/21/2014] [Accepted: 08/22/2014] [Indexed: 11/30/2022]
|
47
|
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
|
48
|
Kumar A, Pandey AN, Jain SK. Nasal-nanotechnology: revolution for efficient therapeutics delivery. Drug Deliv 2014; 23:681-93. [PMID: 24901207 DOI: 10.3109/10717544.2014.920431] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
CONTEXT In recent years, nanotechnology-based delivery systems have gained interest to overcome the problems of restricted absorption of therapeutic agents from the nasal cavity, depending upon the physicochemical properties of the drug and physiological properties of the human nose. OBJECTIVE The well-tolerated and non-invasive nasal drug delivery when combined with the nanotechnology-based novel formulations and carriers, opens the way for the effective systemic and brain targeting delivery of various therapeutic agents. To accomplish competent drug delivery, it is imperative to recognize the interactions among the nanomaterials and the nasal biological environment, targeting cell-surface receptors, drug release, multiple drug administration, stability of therapeutic agents and molecular mechanisms of cell signaling involved in patho-biology of the disease under consideration. METHODS Quite a few systems have been successfully formulated using nanomaterials for intranasal (IN) delivery. Carbon nanotubes (CNTs), chitosan, polylactic-co-glycolic acid (PLGA) and PLGA-based nanosystems have also been studied in vitro and in vivo for the delivery of several therapeutic agents which shown promising concentrations in the brain after nasal administration. RESULTS AND CONCLUSION The use of nanomaterials including peptide-based nanotubes and nanogels (NGs) for vaccine delivery via nasal route is a new approach to control the disease progression. In this review, the recent developments in nanotechnology utilized for nasal drug delivery have been discussed.
Collapse
Affiliation(s)
- Amrish Kumar
- a Department of Pharmaceutics , Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University) , Bilaspur , Chhattisgarh , India
| | - Aditya Nath Pandey
- a Department of Pharmaceutics , Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University) , Bilaspur , Chhattisgarh , India
| | - Sunil Kumar Jain
- a Department of Pharmaceutics , Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University) , Bilaspur , Chhattisgarh , India
| |
Collapse
|
49
|
Bueter CL, Lee CK, Wang JP, Ostroff GR, Specht CA, Levitz SM. Spectrum and mechanisms of inflammasome activation by chitosan. THE JOURNAL OF IMMUNOLOGY 2014; 192:5943-51. [PMID: 24829412 DOI: 10.4049/jimmunol.1301695] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chitosan, the deacetylated derivative of chitin, can be found in the cell wall of some fungi and is used in translational applications. We have shown that highly purified preparations of chitosan, but not chitin, activate the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome in primed mouse bone marrow-derived macrophages (BMMΦ), inducing a robust IL-1β response. In this article, we further define specific cell types that are activated and delineate mechanisms of activation. BMMΦ differentiated to promote a classically activated (M1) phenotype released more IL-1β in response to chitosan than intermediate or alternatively activated macrophages (M2). Chitosan, but not chitin, induced a robust IL-1β response in mouse dendritic cells, peritoneal macrophages, and human PBMCs. Three mechanisms for NLRP3 inflammasome activation may contribute: K(+) efflux, reactive oxygen species, and lysosomal destabilization. The contributions of these mechanisms were tested using a K(+) efflux inhibitor, high extracellular potassium, a mitochondrial reactive oxygen species inhibitor, lysosomal acidification inhibitors, and a cathepsin B inhibitor. These studies revealed that each of these pathways participated in optimal NLRP3 inflammasome activation by chitosan. Finally, neither chitosan nor chitin stimulated significant release from unprimed BMMΦ of any of 22 cytokines and chemokines assayed. This study has the following conclusions: 1) chitosan, but not chitin, stimulates IL-1β release from multiple murine and human cell types; 2) multiple nonredundant mechanisms appear to participate in inflammasome activation by chitosan; and 3) chitin and chitosan are relatively weak stimulators of inflammatory mediators from unprimed BMMΦ. These data have implications for understanding the nature of the immune response to microbes and biomaterials that contain chitin and chitosan.
Collapse
Affiliation(s)
- Chelsea L Bueter
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Chrono K Lee
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Jennifer P Wang
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Gary R Ostroff
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Charles A Specht
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Stuart M Levitz
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605; and
| |
Collapse
|
50
|
Klein K, Mann JFS, Rogers P, Shattock RJ. Polymeric penetration enhancers promote humoral immune responses to mucosal vaccines. J Control Release 2014; 183:43-50. [PMID: 24657807 DOI: 10.1016/j.jconrel.2014.03.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/07/2014] [Accepted: 03/10/2014] [Indexed: 11/26/2022]
Abstract
Protective mucosal immune responses are thought best induced by trans-mucosal vaccination, providing greater potential to generate potent local immune responses than conventional parenteral vaccination. However, poor trans-mucosal permeability of large macromolecular antigens limits bioavailability to local inductive immune cells. This study explores the utility of polymeric penetration enhancers to promote trans-mucosal bioavailability of insulin, as a biomarker of mucosal absorption, and two vaccine candidates: recombinant HIV-1 envelope glycoprotein (CN54gp140) and tetanus toxoid (TT). Responses to vaccinating antigens were assessed by measurement of serum and the vaginal humoral responses. Polyethyleneimine (PEI), Dimethyl-β-cyclodextrin (DM-β-CD) and Chitosan enhanced the bioavailability of insulin following intranasal (IN), sublingual (SL), intravaginal (I.Vag) and intrarectal (IR) administration. The same penetration enhancers also increased antigen-specific IgG and IgA antibody responses to the model vaccine antigens in serum and vaginal secretions following IN and SL application. Co-delivery of both antigens with PEI or Chitosan showed the highest increase in systemic IgG and IgA responses following IN or SL administration. However the highest IgA titres in vaginal secretions were achieved after IN immunisations with PEI and Chitosan. None of the penetration enhancers were able to increase antibody responses to gp140 after I.Vag immunisations, while in contrast PEI and Chitosan were able to induce TT-specific systemic IgG levels following I.Vag administration. In summary, we present supporting data that suggest appropriate co-formulation of vaccine antigens with excipients known to influence mucosal barrier functions can increase the bioavailability of mucosally applied antigens promoting the induction of mucosal and systemic antibody responses.
Collapse
Affiliation(s)
- Katja Klein
- Imperial College London, Department of Infectious Diseases, Division of Medicine, Norfolk Place, London W2 1PG, UK
| | - Jamie F S Mann
- Imperial College London, Department of Infectious Diseases, Division of Medicine, Norfolk Place, London W2 1PG, UK
| | - Paul Rogers
- Imperial College London, Department of Infectious Diseases, Division of Medicine, Norfolk Place, London W2 1PG, UK
| | - Robin J Shattock
- Imperial College London, Department of Infectious Diseases, Division of Medicine, Norfolk Place, London W2 1PG, UK.
| |
Collapse
|