151
|
Bashir SM, Ahmed Rather G, Patrício A, Haq Z, Sheikh AA, Shah MZUH, Singh H, Khan AA, Imtiyaz S, Ahmad SB, Nabi S, Rakhshan R, Hassan S, Fonte P. Chitosan Nanoparticles: A Versatile Platform for Biomedical Applications. MATERIALS (BASEL, SWITZERLAND) 2022; 15:ma15196521. [PMID: 36233864 PMCID: PMC9570720 DOI: 10.3390/ma15196521] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/05/2022] [Accepted: 09/15/2022] [Indexed: 05/10/2023]
Abstract
Chitosan is a biodegradable and biocompatible natural polymer that has been extensively explored in recent decades. The Food and Drug Administration has approved chitosan for wound treatment and nutritional use. Furthermore, chitosan has paved the way for advancements in different biomedical applications including as a nanocarrier and tissue-engineering scaffold. Its antibacterial, antioxidant, and haemostatic properties make it an excellent option for wound dressings. Because of its hydrophilic nature, chitosan is an ideal starting material for biocompatible and biodegradable hydrogels. To suit specific application demands, chitosan can be combined with fillers, such as hydroxyapatite, to modify the mechanical characteristics of pH-sensitive hydrogels. Furthermore, the cationic characteristics of chitosan have made it a popular choice for gene delivery and cancer therapy. Thus, the use of chitosan nanoparticles in developing novel drug delivery systems has received special attention. This review aims to provide an overview of chitosan-based nanoparticles, focusing on their versatile properties and different applications in biomedical sciences and engineering.
Collapse
Affiliation(s)
- Showkeen Muzamil Bashir
- Molecular Biology Laboratory, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Srinagar 190006, India
- Correspondence: (S.M.B.); (G.A.R.); (P.F.)
| | - Gulzar Ahmed Rather
- Department of Biomedical Engineering, Sathyabama Institute of Science & Technology (Deemed to be University), Chennai 600119, India
- Correspondence: (S.M.B.); (G.A.R.); (P.F.)
| | - Ana Patrício
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Zulfiqar Haq
- ICAR-Poultry Seed Project, Division of LPM, Skuast-K 132001, India
| | - Amir Amin Sheikh
- International Institute of Veterinary Education and Research (IIVER), Bahu Akbarpur, Rohtak 124001, India
| | - Mohd Zahoor ul Haq Shah
- Laboratory of Endocrinology, Department of Bioscience, Barkatullah University, Bhopal 462026, India
| | - Hemant Singh
- Department of Polymer and Process Engineering, Indian Institute of Technology, Roorkee 247667, India
| | - Azmat Alam Khan
- ICAR-Poultry Seed Project, Division of LPM, Skuast-K 132001, India
| | - Sofi Imtiyaz
- Molecular Biology Laboratory, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Srinagar 190006, India
| | - Sheikh Bilal Ahmad
- Molecular Biology Laboratory, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Srinagar 190006, India
| | - Showket Nabi
- Large Animal Diagnostic Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Srinagar 190006, India
| | - Rabia Rakhshan
- Molecular Biology Laboratory, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama Alusteng, Srinagar 190006, India
| | - Saqib Hassan
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Pedro Fonte
- iBB—Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Center for Marine Sciences (CCMAR), Gambelas Campus, University of Algarve, 8005-139 Faro, Portugal
- Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, Gambelas Campus, University of Algarve, 8005-139 Faro, Portugal
- Correspondence: (S.M.B.); (G.A.R.); (P.F.)
| |
Collapse
|
152
|
Weiss AM, Hossainy S, Rowan SJ, Hubbell JA, Esser-Kahn AP. Immunostimulatory Polymers as Adjuvants, Immunotherapies, and Delivery Systems. Macromolecules 2022; 55:6913-6937. [PMID: 36034324 PMCID: PMC9404695 DOI: 10.1021/acs.macromol.2c00854] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/16/2022] [Indexed: 12/14/2022]
Abstract
![]()
Activating innate immunity in a controlled manner is
necessary
for the development of next-generation therapeutics. Adjuvants, or
molecules that modulate the immune response, are critical components
of vaccines and immunotherapies. While small molecules and biologics
dominate the adjuvant market, emerging evidence supports the use of
immunostimulatory polymers in therapeutics. Such polymers can stabilize
and deliver cargo while stimulating the immune system by functioning
as pattern recognition receptor (PRR) agonists. At the same time,
in designing polymers that engage the immune system, it is important
to consider any unintended initiation of an immune response that results
in adverse immune-related events. Here, we highlight biologically
derived and synthetic polymer scaffolds, as well as polymer–adjuvant
systems and stimuli-responsive polymers loaded with adjuvants, that
can invoke an immune response. We present synthetic considerations
for the design of such immunostimulatory polymers, outline methods
to target their delivery, and discuss their application in therapeutics.
Finally, we conclude with our opinions on the design of next-generation
immunostimulatory polymers, new applications of immunostimulatory
polymers, and the development of improved preclinical immunocompatibility
tests for new polymers.
Collapse
Affiliation(s)
- Adam M. Weiss
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
- Department of Chemistry, University of Chicago 5735 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Samir Hossainy
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| | - Stuart J. Rowan
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
- Department of Chemistry, University of Chicago 5735 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| |
Collapse
|
153
|
Functionalized chitosan as a promising platform for cancer immunotherapy: A review. Carbohydr Polym 2022; 290:119452. [DOI: 10.1016/j.carbpol.2022.119452] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/20/2022]
|
154
|
V. Lima B, Oliveira MJ, Barbosa MA, Gonçalves RM, Castro F. Harnessing chitosan and poly-(γ-glutamic acid)-based biomaterials towards cancer immunotherapy. MATERIALS TODAY ADVANCES 2022; 15:100252. [DOI: 10.1016/j.mtadv.2022.100252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
155
|
Liao Z, Huang J, Lo PC, Lovell JF, Jin H, Yang K. Self-adjuvanting cancer nanovaccines. J Nanobiotechnology 2022; 20:345. [PMID: 35883176 PMCID: PMC9316869 DOI: 10.1186/s12951-022-01545-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/04/2022] [Indexed: 11/12/2022] Open
Abstract
Nanovaccines, a new generation of vaccines that use nanoparticles as carriers and/or adjuvants, have been widely used in the prevention and treatment of various diseases, including cancer. Nanovaccines have sparked considerable interest in cancer therapy due to a variety of advantages, including improved access to lymph nodes (LN), optimal packing and presentation of antigens, and induction of a persistent anti-tumor immune response. As a delivery system for cancer vaccines, various types of nanoparticles have been designed to facilitate the delivery of antigens and adjuvants to lymphoid organs and antigen-presenting cells (APCs). Particularly, some types of nanoparticles are able to confer an immune-enhancing capability and can themselves be utilized for adjuvant-like effect for vaccines, suggesting a direction for a better use of nanomaterials and the optimization of cancer vaccines. However, this role of nanoparticles in vaccines has not been well studied. To further elucidate the role of self-adjuvanting nanovaccines in cancer therapy, we review the mechanisms of antitumor vaccine adjuvants with respect to nanovaccines with self-adjuvanting properties, including enhancing cross-presentation, targeting signaling pathways, biomimicking of the natural invasion process of pathogens, and further unknown mechanisms. We surveyed self-adjuvanting cancer nanovaccines in clinical research and discussed their advantages and challenges. In this review, we classified self-adjuvanting cancer nanovaccines according to the underlying immunomodulatory mechanism, which may provide mechanistic insights into the design of nanovaccines in the future.
Collapse
Affiliation(s)
- Zhiyun Liao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Huang
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Pui-Chi Lo
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Honglin Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
156
|
Activation of Stimulation of Interferon Genes (STING) Signal and Cancer Immunotherapy. Molecules 2022; 27:molecules27144638. [PMID: 35889509 PMCID: PMC9325158 DOI: 10.3390/molecules27144638] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
Stimulator of interferon gene (STING), an intracellular receptor in the endoplasmic reticulum, could induce the production of cytokines such as type I interferon (IFN) by activating the cGAS-STING signal pathway. In recent years, activation of STING has shown great potential to enhance anti-tumor immunity and reshape the tumor microenvironment, which is expected to be used in tumor immunotherapy. A number of STING agonists have demonstrated promising biological activity and showed excellent synergistic anti-tumor effects in combination with other cancer therapies in preclinical studies and some clinical trials. The combination of STING agonists and ICI also showed a potent effect in improving anti-tumor immunity. In this review, we introduce the cGAS-STING signaling pathway and its effect in tumor immunity and discuss the recent strategies of activation of the STING signaling pathway and its research progress in tumor immunotherapy.
Collapse
|
157
|
Zeng H, Gao Y, Yu W, Liu J, Zhong C, Su X, Wen S, Liang H. Pharmacological Inhibition of STING/TBK1 Signaling Attenuates Myeloid Fibroblast Activation and Macrophage to Myofibroblast Transition in Renal Fibrosis. Front Pharmacol 2022; 13:940716. [PMID: 35924048 PMCID: PMC9340478 DOI: 10.3389/fphar.2022.940716] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022] Open
Abstract
Renal fibrosis is an important pathological biomarker of chronic kidney disease (CKD). Stimulator of interferon genes/TANK binding kinase 1 (STING/TBK1) axis has been identified as the main regulator of innate immune response and closely related to fibrotic disorder. However, the role of STING/TBK1 signaling pathway in kidney fibrosis is still unknown. In this study, we investigated the effect of pharmacological inhibition of STING/TBK1 signaling on renal fibrosis induced by folic acid (FA). In mice, TBK1 was significantly activated in interstitial cells of FA-injured kidneys, which was markedly inhibited by H-151 (a STING inhibitor) treatment. Specifically, pharmacological inhibition of STING impaired bone marrow-derived fibroblasts activation and macrophage to myofibroblast transition in folic acid nephropathy, leading to reduction of extracellular matrix proteins expression, myofibroblasts formation and development of renal fibrosis. Furthermore, pharmacological inhibition of TBK1 by GSK8612 reduced myeloid myofibroblasts accumulation and impeded macrophage to myofibroblast differentiation, resulting in less deposition of extracellular matrix protein and less severe fibrotic lesion in FA-injured kidneys. In cultured mouse bone marrow-derived monocytes, TGF-β1 activated STING/TBK1 signaling. This was abolished by STING or TBK1 inhibitor administration. In addition, GSK8612 treatment decreased levels of α-smooth muscle actin and extracellular matrix proteins and prevents bone marrow-derived macrophages to myofibroblasts transition in vitro. Collectively, our results revealed that STING/TBK1 signaling has a critical role in bone marrow-derived fibroblast activation, macrophages to myofibroblasts transition, and kidney fibrosis progression.
Collapse
Affiliation(s)
- Haimei Zeng
- Department of Anesthesiology, Foshan Women and Children Hospital, Foshan, China
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
- Department of Anesthesiology, Huidong People’s Hospital, Huizhou, China
| | - Ying Gao
- Department of Anesthesiology, The First People’s Hospital of Foshan, Foshan, China
| | - Wenqiang Yu
- Department of Anesthesiology, The First People’s Hospital of Foshan, Foshan, China
| | - Jiping Liu
- Department of Anesthesiology, Foshan Women and Children Hospital, Foshan, China
| | - Chaoqun Zhong
- Department of Anesthesiology, The First People’s Hospital of Foshan, Foshan, China
| | - Xi Su
- Department of Paediatrics, Foshan Women and Children Hospital, Foshan, China
- *Correspondence: Xi Su, ; Hua Liang,
| | - Shihong Wen
- Department of Anesthesiology, The First Affiliated Hospital of SUN YAT-SEN University, Guangzhou, China
| | - Hua Liang
- Department of Anesthesiology, Foshan Women and Children Hospital, Foshan, China
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
- *Correspondence: Xi Su, ; Hua Liang,
| |
Collapse
|
158
|
Lee D, Huntoon K, Kang M, Lu Y, Gallup T, Jiang W, Kim BYS. Harnessing cGAS‐STING Pathway for Cancer Immunotherapy: From Bench to Clinic. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- DaeYong Lee
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Kristin Huntoon
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Minjeong Kang
- Department of radiation oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Yifei Lu
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Thomas Gallup
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Wen Jiang
- Department of radiation oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Betty Y S Kim
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| |
Collapse
|
159
|
Wang D, Zou Y, Wang N, Wu J. Chitosan hydrochloride salt stabilized emulsion as vaccine adjuvant. Carbohydr Polym 2022; 296:119879. [DOI: 10.1016/j.carbpol.2022.119879] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/04/2022] [Accepted: 07/13/2022] [Indexed: 11/02/2022]
|
160
|
Seesen M, Jearanaiwitayakul T, Limthongkul J, Sunintaboon P, Ubol S. Mice immunized with trimethyl chitosan nanoparticles containing DENV-2 envelope domain III elicit neutralizing antibodies with undetectable antibody-dependent enhancement activity. J Gen Virol 2022; 103. [PMID: 35833704 DOI: 10.1099/jgv.0.001768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dengue is a disease that poses a significant global public health concern. Although a tetravalent live-attenuated dengue vaccine has been licensed, its efficacy is still debated due to evidence of vaccine breakthrough infection. To avoid this issue, dengue vaccines should stimulate a high degree of serotype-specific response. Thus, envelope domain III (EDIII), which contains serotype-specific neutralizing epitopes, is an attractive target for dengue vaccine development. In this study, we investigated how EDIII encapsidated in N, N, N-trimethyl chitosan chloride nanoparticles (TMC NPs) stimulates a serotype-specific response and whether this response exerts a potential in vitro breakthrough infection. The immune response to DENV-2 elicited by EDIII TMC NP-immunized mice was monitored. We demonstrated that immunization with EDIII TMC NPs resulted in a high level of anti-EDIII antibody production. These antibodies included IgG, IgG1, and IgG2a subtypes. Importantly, antibodies from the immunized mice exerted efficient neutralizing activity with undetectable antibody dependent enhancement (ADE) activity. We also found that EDIII TMC NPs activated functional EDIII-specific CD4+ and CD8+ T cell responses. In conclusion, EDIII TMC NPs stimulated humoral immunity with a strong neutralizing antibody response, as well as a cellular immune response against DENV-2.
Collapse
Affiliation(s)
- Mathurin Seesen
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | - Jitra Limthongkul
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Panya Sunintaboon
- Department of Chemistry, Faculty of Science, Mahidol University, Salaya, Nakornpatom 73170, Thailand
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
161
|
Turley JL, Lavelle EC. Resolving adjuvant mode of action to enhance vaccine efficacy. Curr Opin Immunol 2022; 77:102229. [PMID: 35779364 DOI: 10.1016/j.coi.2022.102229] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/15/2022] [Accepted: 05/25/2022] [Indexed: 12/11/2022]
Abstract
Adjuvants are a miscellaneous range of molecules and materials that can enhance the magnitude, functionality, breadth and durability of immune responses. Despite the multiplicity of compounds with adjuvant properties, less than a dozen are in clinical use in vaccines against infectious diseases. While many factors have contributed to their slow development, among the major challenges are the high safety and efficacy standards set by current adjuvants in human vaccines and our limited understanding of how adjuvants mediate their effects. This review outlines why it is so difficult to elucidate their mechanism of action, highlights areas that require in-depth research and discusses recent advancements that are revitalising adjuvant development. It is hoped that a fuller understanding of adjuvant sensing, signalling and function will facilitate the design of vaccines that promote sustained protective immunity against challenging bacterial and viral pathogens.
Collapse
Affiliation(s)
- Joanna L Turley
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02R590, Ireland.
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02R590, Ireland.
| |
Collapse
|
162
|
Activation of Cellular Players in Adaptive Immunity via Exogenous Delivery of Tumor Cell Lysates. Pharmaceutics 2022; 14:pharmaceutics14071358. [PMID: 35890254 PMCID: PMC9316852 DOI: 10.3390/pharmaceutics14071358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 12/04/2022] Open
Abstract
Tumor cell lysates (TCLs) are a good immunogenic source of tumor-associated antigens. Since whole necrotic TCLs can enhance the maturation and antigen-presenting ability of dendritic cells (DCs), multiple strategies for the exogenous delivery of TCLs have been investigated as novel cancer immunotherapeutic solutions. The TCL-mediated induction of DC maturation and the subsequent immunological response could be improved by utilizing various material-based carriers. Enhanced antitumor immunity and cancer vaccination efficacy could be eventually achieved through the in vivo administration of TCLs. Therefore, (1) important engineering methodologies to prepare antigen-containing TCLs, (2) current therapeutic approaches using TCL-mediated DC activation, and (3) the significant sequential mechanism of DC-based signaling and stimulation in adaptive immunity are summarized in this review. More importantly, the recently reported developments in biomaterial-based exogenous TCL delivery platforms and co-delivery strategies with adjuvants for effective cancer vaccination and antitumor effects are emphasized.
Collapse
|
163
|
Argenziano M, Occhipinti S, Scomparin A, Angelini C, Novelli F, Soster M, Giovarelli M, Cavalli R. Exploring chitosan-shelled nanobubbles to improve HER2 + immunotherapy via dendritic cell targeting. Drug Deliv Transl Res 2022; 12:2007-2018. [PMID: 35672651 PMCID: PMC9172608 DOI: 10.1007/s13346-022-01185-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 11/29/2022]
Abstract
Immunotherapy is a valuable approach to cancer treatment as it is able to activate the immune system. However, the curative methods currently in clinical practice, including immune checkpoint inhibitors, present some limitations. Dendritic cell vaccination has been investigated as an immunotherapeutic strategy, and nanotechnology-based delivery systems have emerged as powerful tools for improving immunotherapy and vaccine development. A number of nanodelivery systems have therefore been proposed to promote cancer immunotherapy. This work aims to design a novel immunotherapy nanoplatform for the treatment of HER2 + breast cancer, and specially tailored chitosan-shelled nanobubbles (NBs) have been developed for the delivery of a DNA vaccine. The NBs have been functionalized with anti-CD1a antibodies to target dendritic cells (DCs). The NB formulations possess dimensions of approximately 300 nm and positive surface charge, and also show good physical stability up to 6 months under storage at 4 °C. In vitro characterization has confirmed that these NBs are capable of loading DNA with good encapsulation efficiency (82%). The antiCD1a-functionalized NBs are designed to target DCs, and demonstrated the ability to induce DC activation in both human and mouse cell models, and also elicited a specific immune response that was capable of slowing tumor growth in mice in vivo. These findings are the proof of concept that loading a tumor vaccine into DC-targeted chitosan nanobubbles may become an attractive nanotechnology approach for the future immunotherapeutic treatment of cancer.
Collapse
Affiliation(s)
- Monica Argenziano
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125, Turin, Italy
| | - Sergio Occhipinti
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, 10126, Turin, Italy
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125, Turin, Italy
| | - Costanza Angelini
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, 10126, Turin, Italy
| | - Francesco Novelli
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, 10126, Turin, Italy
| | - Marco Soster
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125, Turin, Italy
| | - Mirella Giovarelli
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, 10126, Turin, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125, Turin, Italy.
| |
Collapse
|
164
|
Feng H, Sun R, Song G, Zhu S, Nie Z, Lin L, Yi R, Wu S, Wang G, He Y, Wang S, Wang P, Wu L, Shu J. A Glycolipid α-GalCer Derivative, 7DW8-5 as a Novel Mucosal Adjuvant for the Split Inactivated Influenza Vaccine. Viruses 2022; 14:v14061174. [PMID: 35746644 PMCID: PMC9230830 DOI: 10.3390/v14061174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/15/2022] [Accepted: 05/27/2022] [Indexed: 12/10/2022] Open
Abstract
Influenza virus infects the host and transmits through the respiratory tract (i.e., the mouth and nose); therefore, the development of intranasal influenza vaccines that mimic the natural infection, coupled with an efficient mucosal adjuvant, is an attractive alternative to current parenteral vaccines. However, with the withdrawal of cholera toxin and Escherichia coli heat-labile endotoxin from clinical use due to side effects, there are no approved adjuvants for intranasal vaccines. Therefore, safe and effective mucosal adjuvants are urgently needed. Previously, we reported that one derivative of α-Galactosylceramide (α-GalCer), 7DW8-5, could enhance the protective efficacy of split influenza vaccine by injection administration. However, the mucosal adjuvanticity of 7DW8-5 is still unclear. In this study, we found that 7DW8-5 promotes the production of secret IgA antibodies and IgG antibodies and enhances the protective efficacy of the split influenza vaccine by intranasal administration. Furthermore, co-administration of 7DW8-5 with the split influenza vaccine significantly reduces the virus shedding in the upper and lower respiratory tract after lethal challenge. Our results demonstrate that 7DW8-5 is a novel mucosal adjuvant for the split influenza vaccine.
Collapse
Affiliation(s)
- Huapeng Feng
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
- Correspondence: (H.F.); (L.W.); (J.S.)
| | - Ruolin Sun
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
| | - Guanru Song
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
| | - Shunfan Zhu
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
| | - Zhenyu Nie
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
| | - Liming Lin
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
| | - Ruonan Yi
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
| | - Shixiang Wu
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
| | - Genzhu Wang
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
| | - Yulong He
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
| | - Siquan Wang
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
| | - Pei Wang
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
| | - Li Wu
- Department of Biology, College of Life Sciences, China Jiliang University, Hangzhou 310018, China
- Correspondence: (H.F.); (L.W.); (J.S.)
| | - Jianhong Shu
- Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (R.S.); (G.S.); (S.Z.); (Z.N.); (L.L.); (R.Y.); (S.W.); (G.W.); (Y.H.); (S.W.); (P.W.)
- Correspondence: (H.F.); (L.W.); (J.S.)
| |
Collapse
|
165
|
Intranasal administration of a recombinant RBD vaccine induces long-term immunity against Omicron-included SARS-CoV-2 variants. Signal Transduct Target Ther 2022; 7:159. [PMID: 35581200 PMCID: PMC9112270 DOI: 10.1038/s41392-022-01002-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/23/2022] [Accepted: 04/05/2022] [Indexed: 02/05/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) has posed great threats to global health and economy. Several effective vaccines are available now, but additional booster immunization is required to retain or increase the immune responses owing to waning immunity and the emergency of new variant strains. The deficiency of intramuscularly delivered vaccines to induce mucosal immunity urged the development of mucosal vaccines. Here, we developed an adjuvanted intranasal RBD vaccine and monitored its long-term immunogenicity against both wild-type and mutant strains of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), including Omicron variants, in mice. Three-dose intranasal immunization with this vaccine induced and maintained high levels of neutralizing IgG antibodies in the sera for at least 1 year. Strong mucosal immunity was also provoked, including mucosal secretory IgA and lung-resident memory T cells (TRM). We also demonstrated that the long-term persistence of lung TRM cells is a consequence of local T-cell proliferation, rather than T-cell migration from lymph nodes. Our data suggested that the adjuvanted intranasal RBD vaccine is a promising vaccine candidate to establish robust, long-lasting, and broad protective immunity against SARS-CoV-2 both systemically and locally.
Collapse
|
166
|
Chitooligosaccharides Improve the Efficacy of Checkpoint Inhibitors in a Mouse Model of Lung Cancer. Pharmaceutics 2022; 14:pharmaceutics14051046. [PMID: 35631632 PMCID: PMC9147765 DOI: 10.3390/pharmaceutics14051046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 12/10/2022] Open
Abstract
YKL-40 (also named chitinase 3 like-1 protein [CHI3L1]) is a secreted chitinase-like protein which is upregulated in cancers and suggested to have pro-tumorigenic activity. YKL-40 lacks enzymatic function, but it can bind carbohydrates such as chitin. Chitooligosaccharides (COS) derived from deacetylation and hydrolysis of chitin might be used for the blockade of YKL-40 function. Here, public single-cell RNA sequencing datasets were used to elucidate the cellular source of YKL-40 gene expression in human tumors. Fibroblasts and myeloid cells were the primary sources of YKL-40. Screening of YKL-40 gene expression in syngeneic mouse cancer models showed the highest expression in the Lewis lung carcinoma (LL2) model. LL2 was used to investigate COS monotherapy and combinations with immune checkpoint inhibitors (anti-PD-L1 and anti-CTLA-4) (ICIs) and radiotherapy (8 Gy × 3) (RT). COS tended to reduce plasma YKL-40 levels, but it did not affect tumor growth. LL2 showed minimal responses to ICIs, or to RT alone. Interestingly, ICIs combined with COS led to delayed tumor growth. RT also enhanced the efficacy of ICIs; however, the addition of COS did not further delay the tumor growth. COS may exert their anti-tumorigenic effects through the inhibition of YKL-40, but additional functions of COS should be investigated.
Collapse
|
167
|
Nguyen KG, Mantooth SM, Vrabel MR, Zaharoff DA. Intranasal Delivery of Thermostable Subunit Vaccine for Cross-Reactive Mucosal and Systemic Antibody Responses Against SARS-CoV-2. Front Immunol 2022; 13:858904. [PMID: 35592324 PMCID: PMC9110812 DOI: 10.3389/fimmu.2022.858904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/01/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the remarkable efficacy of currently approved COVID-19 vaccines, there are several opportunities for continued vaccine development against SARS-CoV-2 and future lethal respiratory viruses. In particular, restricted vaccine access and hesitancy have limited immunization rates. In addition, current vaccines are unable to prevent breakthrough infections, leading to prolonged virus circulation. To improve access, a subunit vaccine with enhanced thermostability was designed to eliminate the need for an ultra-cold chain. The exclusion of infectious and genetic materials from this vaccine may also help reduce vaccine hesitancy. In an effort to prevent breakthrough infections, intranasal immunization to induce mucosal immunity was explored. A prototype vaccine comprised of receptor-binding domain (RBD) polypeptides formulated with additional immunoadjuvants in a chitosan (CS) solution induced high levels of RBD-specific antibodies in laboratory mice after 1 or 2 immunizations. Antibody responses were durable with high titers persisting for at least five months following subcutaneous vaccination. Serum anti-RBD antibodies contained both IgG1 and IgG2a isotypes suggesting that the vaccine induced a mixed Th1/Th2 response. RBD vaccination without CS formulation resulted in minimal anti-RBD responses. The addition of CpG oligonucleotides to the CS plus RBD vaccine formulation increased antibody titers more effectively than interleukin-12 (IL-12). Importantly, generated antibodies were cross-reactive against RBD mutants associated with SARS-CoV-2 variants of concern, including alpha, beta and delta variants, and inhibited binding of RBD to its cognate receptor angiotensin converting enzyme 2 (ACE2). With respect to stability, vaccines did not lose activity when stored at either room temperature (21-22°C) or 4°C for at least one month. When delivered intranasally, vaccines induced RBD-specific mucosal IgA antibodies, which may protect against breakthrough infections in the upper respiratory tract. Altogether, data indicate that the designed vaccine platform is versatile, adaptable and capable of overcoming key constraints of current COVID-19 vaccines.
Collapse
Affiliation(s)
- Khue G. Nguyen
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Siena M. Mantooth
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Maura R. Vrabel
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - David A. Zaharoff
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
168
|
Yu Z, He K, Cao W, Aleem MT, Yan R, Xu L, Song X, Li X. Nano vaccines for T. gondii Ribosomal P2 Protein With Nanomaterials as a Promising DNA Vaccine Against Toxoplasmosis. Front Immunol 2022; 13:839489. [PMID: 35265084 PMCID: PMC8899214 DOI: 10.3389/fimmu.2022.839489] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Caused by Toxoplasma gondii, toxoplasmosis has aroused great threats to public health around the world. So far, no effective vaccine or drug is commercially available, and the demands for a safe and effective therapeutic strategy have become more and more urgent. In the current study, we constructed a DNA vaccine encoding T. gondii ribosomal P2 protein (TgP2) and denoted as TgP2-pVAX1 plasmid. To improve the immunoprotection, nanomaterial poly-lactic-co-glycolic acid (PLGA) and chitosan were used as the delivery vehicle to construct TgP2-pVAX1/PLGA and TgP2-pVAX1/CS nanospheres. Before vaccinations in BALB/c mice, TgP2-pVAX1 plasmids were transiently transfected into Human Embryonic Kidney (HEK) 293-T cells, and the expression of the eukaryotic plasmids was detected by laser confocal microscopy and Western blotting. Then the immunoprotection of naked DNA plasmids and their two nano-encapsulations were evaluated in the laboratory animal model. According to the investigations of antibody, cytokine, dendritic cell (DC) maturation, molecule expression, splenocyte proliferation, and T lymphocyte proportion, TgP2-pVAX1 plasmid delivered by two types of nanospheres could elicit a mixed Th1/Th2 immune response and Th1 immunity as the dominant. In addition, TgP2-pVAX1/PLGA and TgP2-pVAX1/CS nanospheres have great advantages in enhancing immunity against a lethal dose of T. gondii RH strain challenge. All these results suggested that TgP2-pVAX1 plasmids delivered by PLGA or chitosan nanomaterial could be promising vaccines in resisting toxoplasmosis and deserve further investigations and applications.
Collapse
Affiliation(s)
- ZhengQing Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ke He
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - WanDi Cao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Muhammad Tahir Aleem
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - RuoFeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - LiXin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - XiaoKai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - XiangRui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
169
|
An all-in-one adjuvanted therapeutic cancer vaccine targeting dendritic cell cytosol induces long-lived tumor suppression through NLRC4 inflammasome activation. Biomaterials 2022; 286:121542. [DOI: 10.1016/j.biomaterials.2022.121542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/29/2022] [Accepted: 04/22/2022] [Indexed: 11/21/2022]
|
170
|
Lee S, Byun S, Lee C, Park SH, Rudra D, Iwakura Y, Lee YJ, Im S, Hwang DS. Resolving the Mutually Exclusive Immune Responses of Chitosan with Nanomechanics and Immunological Assays. Adv Healthc Mater 2022; 11:e2102667. [PMID: 35397156 DOI: 10.1002/adhm.202102667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/18/2022] [Indexed: 12/22/2022]
Abstract
Multifaceted functions displayed by both pro- and anti-inflammatory properties of chitosan hinder its effective development as an immunomodulatory agent. Herein, the contributions of the bending stiffness of chitosan with regard to its immune regulatory properties toward inflammation are investigated. The anti-inflammatory properties of chitosan molecular weight (MW) with a shorter (≈1 kDa) or longer (≈15 kDa) than the persistent length (LP ) are compared using immunological assays and nanomechanics-based experiments on the surface forces apparatus (SFA). Interestingly, 1 kDa chitosan significantly enhances the generation of anti-inflammatory regulatory T cells (Tregs) through the Dectin-1-dependent pattern recognition receptor (PRR) on antigen-presenting cells. SFA analyses also show a similar trend of interaction forces between chitosan and diverse PRRs depending on their MW. The results obtained in the immunological and nanomechanical experiments are consistent and imply that the binding features of PRRs vary depending on the MW of chitosan, which may alter immune activity. In accordance, in vivo administration of only 1 kDa represses inflammatory responses and suppresses the progression of experimental colitis. This study elucidates a previously unexplored bending stiffness-dependent immune regulatory property of chitosan and suggests the applicability of low MW (rod-like) chitosan as a pharmaceutical ingredient to treat diverse inflammatory disorders.
Collapse
Affiliation(s)
- Suyoung Lee
- Division of Integrative Biosciences and Biotechnology Department of Life Science Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro Pohang 37673 Republic of Korea
- Division of Environmental Science and Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro Pohang 37673 Republic of Korea
| | - Seohyun Byun
- Division of Integrative Biosciences and Biotechnology Department of Life Science Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro Pohang 37673 Republic of Korea
| | - Changhon Lee
- Division of Integrative Biosciences and Biotechnology Department of Life Science Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro Pohang 37673 Republic of Korea
| | - Sun Hee Park
- ImmmunoBiome Inc. 77 Cheongam‐Ro, Nam‐Gu Pohang 37673 Republic of Korea
| | - Dipayan Rudra
- ImmmunoBiome Inc. 77 Cheongam‐Ro, Nam‐Gu Pohang 37673 Republic of Korea
- School of Life Sciences and Technology ShanghaiTech University 393 Huaxia Middle Rd Pudong Shanghai 201210 China
| | - Yoichiro Iwakura
- Center for Animal Disease Models Research Institute for Science and Technology Tokyo University of Science 2669 Yamazaki Noda Chiba 278‐0022 Japan
- Center for Experimental Medicine and Systems Biology Institute of Medical Science the University of Tokyo Minato‐ku Tokyo 108‐0071 Japan
| | - You Jeong Lee
- Division of Integrative Biosciences and Biotechnology Department of Life Science Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro Pohang 37673 Republic of Korea
- Department of Pharmacy Seoul National University Gwanak‐ro 38‐gil Seoul 08826 Republic of Korea
| | - Sin‐Hyeog Im
- Division of Integrative Biosciences and Biotechnology Department of Life Science Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro Pohang 37673 Republic of Korea
- ImmmunoBiome Inc. 77 Cheongam‐Ro, Nam‐Gu Pohang 37673 Republic of Korea
| | - Dong Soo Hwang
- Division of Integrative Biosciences and Biotechnology Department of Life Science Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro Pohang 37673 Republic of Korea
- Division of Environmental Science and Engineering Pohang University of Science and Technology (POSTECH) 77 Cheongam‐ro Pohang 37673 Republic of Korea
| |
Collapse
|
171
|
Wang J, Wu CS, Hu YZ, Yang L, Zhang XJ, Zhang YA. Plasmablasts induced by chitosan oligosaccharide secrete natural IgM to enhance the humoral immunity in grass carp. Carbohydr Polym 2022; 281:119073. [PMID: 35074109 DOI: 10.1016/j.carbpol.2021.119073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/13/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023]
Abstract
Chitosan oligosaccharide (COS) is an attractive immunopotentiator capable of driving humoral immunity in vertebrates, but its cellular and molecular mechanisms still require elucidation. In this study, COS induced the proliferation and differentiation of splenic IgM+ B cells into IgMlo and IgMhi B cell subsets in grass carp (Ctenopharyngodon idella). The IgMlo B cells were further identified as short-lived plasmablasts that secreted natural IgM with binding-abilities to lipopolysaccharide (LPS) and peptidoglycan (PGN). Moreover, the mannose receptor (MR) and integrins were discovered and identified as the binding-receptors of COS on IgMlo plasmablasts. The MR synergized with integrins to trigger intracellular signal transduction to boost plasmablast generation and expansion. Notably, IgMlo plasmablasts originally generated in spleen but they migrated into blood to secrete natural IgM, which augmented the serum bactericidal activity. Taken together, this study revealed the cellular and molecular mechanisms of COS-triggered humoral immunity in fish.
Collapse
Affiliation(s)
- Jie Wang
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China
| | - Chang-Song Wu
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China
| | - Ya-Zhen Hu
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China
| | - Lan Yang
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China
| | - Xu-Jie Zhang
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China.
| | - Yong-An Zhang
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Hubei Hongshan Laboratory, Wuhan, China.
| |
Collapse
|
172
|
Garland KM, Sheehy TL, Wilson JT. Chemical and Biomolecular Strategies for STING Pathway Activation in Cancer Immunotherapy. Chem Rev 2022; 122:5977-6039. [PMID: 35107989 PMCID: PMC8994686 DOI: 10.1021/acs.chemrev.1c00750] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The stimulator of interferon genes (STING) cellular signaling pathway is a promising target for cancer immunotherapy. Activation of the intracellular STING protein triggers the production of a multifaceted array of immunostimulatory molecules, which, in the proper context, can drive dendritic cell maturation, antitumor macrophage polarization, T cell priming and activation, natural killer cell activation, vascular reprogramming, and/or cancer cell death, resulting in immune-mediated tumor elimination and generation of antitumor immune memory. Accordingly, there is a significant amount of ongoing preclinical and clinical research toward further understanding the role of the STING pathway in cancer immune surveillance as well as the development of modulators of the pathway as a strategy to stimulate antitumor immunity. Yet, the efficacy of STING pathway agonists is limited by many drug delivery and pharmacological challenges. Depending on the class of STING agonist and the desired administration route, these may include poor drug stability, immunocellular toxicity, immune-related adverse events, limited tumor or lymph node targeting and/or retention, low cellular uptake and intracellular delivery, and a complex dependence on the magnitude and kinetics of STING signaling. This review provides a concise summary of the STING pathway, highlighting recent biological developments, immunological consequences, and implications for drug delivery. This review also offers a critical analysis of an expanding arsenal of chemical strategies that are being employed to enhance the efficacy, safety, and/or clinical utility of STING pathway agonists and lastly draws attention to several opportunities for therapeutic advancements.
Collapse
Affiliation(s)
- Kyle M Garland
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
| | - Taylor L Sheehy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
| |
Collapse
|
173
|
Oliveira NA, Sevim H. Dendritic cell differentiation from human induced pluripotent stem cells: challenges and progress. Stem Cells Dev 2022; 31:207-220. [PMID: 35316109 DOI: 10.1089/scd.2021.0305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Dendritic cells (DCs) are the major antigen-presenting cells of the immune system responsible for initiating and coordinating immune responses. These abilities provide potential for several clinical applications, such as the development of immunogenic vaccines. However, difficulty in obtaining DCs from conventional sources, such as bone marrow (BM), peripheral blood (PBMC), and cord blood (CB), is a significantly hinders routine application. The use of human induced pluripotent stem cells (hiPSCs) is a valuable alternative for generating sufficient numbers of DCs to be used in basic and pre-clinical studies. Despite the many challenges that must be overcome to achieve an efficient protocol for obtaining the major DC types from hiPSCs, recent progress has been made. Here we review the current state of developing DCs from hiPSCs, as well as the key elements required to enable the routine use of hiPSC-derived DCs in pre-clinical and clinical assays.
Collapse
Affiliation(s)
- Nelio Aj Oliveira
- Jackson Laboratory - Farmington, 481263, Cell Engineering , Farmington, Connecticut, United States, 06032-2374;
| | - Handan Sevim
- Hacettepe Universitesi, 37515, Faculty of Science Department of Biology, Ankara, Ankara, Turkey;
| |
Collapse
|
174
|
Safarzadeh M, Mohammadi-Yeganeh S, Ghorbani-Bidkorbeh F, Haji Molla Hoseini M. Chitosan based nanoformulation expressing miR-155 as a promising adjuvant to enhance Th1-biased immune responses. Life Sci 2022; 297:120459. [PMID: 35248524 DOI: 10.1016/j.lfs.2022.120459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/18/2022] [Accepted: 03/01/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND AIM MiR-155 could act as a key modulator of different aspects of immune system including Th1 responses. In this study, we designed chitosan nanoparticles containing miR-155-expressing plasmid and explored their effects as an adjuvant to enhance Th1 responses for potential future application against intracellular pathogens. METHODS Nanoparticles were formulated by complex coacervation method and characterized for physicochemical and functional characteristics. Transfection efficiency in Raw 264.7 cells, effects on miR-155 target genes and NO production were evaluated. The prepared nanoparticles were co-administered as an adjuvant with ovalbumin to immunize mice and finally production of IFN-γ and IL-4 were measured by ELISA in splenocyte recall responses. RESULTS The prepared nanoparticles had the mean size of 244 nm and zeta potential of +17 mV, respectively. Electrophoresis analysis indicated the high capability of nanoparticles to protect the plasmid from DNaseI degradation. Furthermore, nanoparticles showed an appropriate transfection efficiency in Raw 264.7 cells and could downregulate the expression of miR-155 target genes and also upregulate NO production. In vivo immunization examinations revealed successful shift of T cell responses toward Th1. CONCLUSION Our data suggests the high potential of chitosan nanoparticles containing miR-155-expressing plasmid as an adjuvant for significantly enhanced Th1-biased immune responses upon immunization with a given antigen.
Collapse
Affiliation(s)
- Mehrnoush Safarzadeh
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ghorbani-Bidkorbeh
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Haji Molla Hoseini
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
175
|
Dmour I, Islam N. Recent advances on chitosan as an adjuvant for vaccine delivery. Int J Biol Macromol 2022; 200:498-519. [PMID: 34973993 DOI: 10.1016/j.ijbiomac.2021.12.129] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/05/2021] [Accepted: 12/19/2021] [Indexed: 12/21/2022]
Abstract
Chitosan (CS) is a natural polymer derived from chitin that has wide applications in drugs, vaccines, and antigen delivery. The distinctive mucoadhesive, biocompatibility, biodegradable, and less toxic properties of chitosan compared to the currently used vaccine adjuvants made it a promising candidate for use as an adjuvant/carrier in vaccine delivery. In addition, chitosan exhibits intrinsic immunomodulating properties making it a suitable adjuvant in preparing vaccines delivery systems. Nanoparticles (NPs) of chitosan and its derivatives loaded with antigen have been shown to induce cellular and humoral responses. Versatility in the physicochemical properties of chitosan can provide an excellent opportunity to engineer antigen-specific adjuvant/delivery systems. This review discusses the recent advances of chitosan and its derivatives as adjuvants in vaccine deliveryand the published literature in the last fifteen years. The impact of physicochemical properties of chitosan on vaccine formulation has been described in detail. Applications of chitosan and its derivatives, their physicochemical properties, and mechanisms in enhancing immune responses have been discussed. Finally, challenges and future aspects of chitosan use has been pointed out.
Collapse
Affiliation(s)
- Isra Dmour
- Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan.
| | - Nazrul Islam
- Pharmacy Discipline, School of Clinical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; Centre for Immunology and Infection Control (CIIC), Queensland University of Technology (QUT), Brisbane, QLD, Australia
| |
Collapse
|
176
|
Kim NE, Song YJ. Coordinated regulation of interferon and inflammasome signaling pathways by SARS-CoV-2 proteins. J Microbiol 2022; 60:300-307. [PMID: 35089584 PMCID: PMC8795727 DOI: 10.1007/s12275-022-1502-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/06/2021] [Accepted: 12/15/2021] [Indexed: 12/16/2022]
Abstract
Type I and III interferons (IFNs) and the nucleotide-binding domain (NBD) leucine-rich repeat (LRR)-containing receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome play pivotal roles in the pathogenesis of SARS-CoV-2. While optimal IFN and inflammasome responses are essential for limiting SARS-CoV-2 infection, aberrant activation of these innate immune responses is associated with COVID-19 pathogenesis. In this review, we focus our discussion on recent findings on SARS-CoV-2-induced type I and III IFNs and NLRP3 inflammasome responses and the viral proteins regulating these mechanisms.
Collapse
Affiliation(s)
- Na-Eun Kim
- Department of Life Science, Gachon University, Seongnam, 13120, Republic of Korea
| | - Yoon-Jae Song
- Department of Life Science, Gachon University, Seongnam, 13120, Republic of Korea.
| |
Collapse
|
177
|
MUC1 vaccines using β-cyclodextrin grafted chitosan (CS-g-CD) as carrier via host-guest interaction elicit robust immune responses. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.02.072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
178
|
Du Y, Liu Y, Wang D, Bai H, Wang Z, He X, Zhang P, Tian J, Wang J. Peptidic microarchitecture-trapped tumor vaccine combined with immune checkpoint inhibitor or PI3Kγ inhibitor can enhance immunogenicity and eradicate tumors. J Immunother Cancer 2022; 10:jitc-2021-003564. [PMID: 35217574 PMCID: PMC8883272 DOI: 10.1136/jitc-2021-003564] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2022] [Indexed: 12/30/2022] Open
Abstract
Background With the rapid development of immune checkpoint inhibitors and neoantigen (NeoV)-based personalized tumor vaccines, tumor immunotherapy has shown promising therapeutic results. However, the limited efficacy of available tumor vaccines impedes the development of personalized tumor immunotherapy. In this study, we developed a novel tumor vaccine system and proposed combined therapeutic strategies for improving treatment effects. Methods We developed a novel tumor vaccine system comprising a newly synthesized peptidic microarchitecture (PMA) with high assembly efficacy. The PMA-trapped neoantigen vaccine was developed to codeliver tumor neoantigen and the Toll-like receptor 9 agonist CpG (NeoV), abbreviated as PMA-NeoV. A microfluidic chip was used to produce PMA particles in a uniform and precise manner. Vaccine effectiveness was investigated both in vitro and in vivo. The combined immunotherapeutic effect of PMA-NeoV with anti-programmed cell death ligand 1 antibody (aPD-L1) or with the phosphatidylinositol 3‑kinase γ (PI3Kγ) inhibitor IPI-549 was further tested in MC38 mouse tumor model. Results PMA-NeoV not only promoted codelivery of the tumor vaccine but also potentiated vaccine immunogenicity. Moreover, compared with free NeoV, PMA-NeoV significantly increased the number of tumor-infiltrating lymphocytes, promoted the neoantigen-specific systemic immune response, and suppressed murine colon MC38 tumor growth. Furthermore, PMA-NeoV increased the expression of programmed cell death receptor-1 on T lymphocytes, and in combination with aPD-L1 eradicated seven of eight MC38 tumors by rescuing exhausted T lymphocytes. Moreover, we combined the PMA-NeoV with the IPI-549, a molecular switch that controls immune suppression, and found that this combination significantly suppressed tumor growth and eradicated five of eight inoculated tumors, by switching suppressive macrophages to their active state and activating T cells to prime a robust tumor immune microenvironment. Conclusions We developed a tumor vaccine delivery system and presented a promising personalized tumor vaccine-based therapeutic regimen in which a tumor vaccine delivery system is combined with an aPD-L1 or PI3Kγ inhibitor to improve tumor immunotherapy outcomes.
Collapse
Affiliation(s)
- Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,The University of Chinese Academy of Sciences, Beijing, China
| | - Ye Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, China
| | - Di Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hua Bai
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhijie Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiran He
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Pei Zhang
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China .,The University of Chinese Academy of Sciences, Beijing, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China.,School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, China
| | - Jie Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
179
|
Zhuo SH, Wu JJ, Zhao L, Li WH, Zhao YF, Li YM. A chitosan-mediated inhalable nanovaccine against SARS-CoV-2. NANO RESEARCH 2022; 15:4191-4200. [PMID: 35126879 PMCID: PMC8809230 DOI: 10.1007/s12274-021-4012-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 06/14/2023]
Abstract
UNLABELLED Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with several antigenic variants, has grown into a global challenge, and the rapid establishment of an immune barrier is crucial to achieving long-term control of the virus. This has led to a great demand for easy preparation and scalable vaccines, especially in low-income countries. Here, we present an inhalable nanovaccine comprising chitosan and SARS-CoV-2 spike protein. The chitosan-mediated nanovaccine enabled a strong spike-specific antibody immune response and augmented local mucosal immunity in bronchoalveolar lavage and lungs, which might be capable of protecting the host from infection without systemic toxicity. In addition, the enhanced adaptive immunity stimulated by chitosan showed potential protection against SARS-CoV-2. Furthermore, inhalation of the nanovaccine induced a comparable antibody response compared to intramuscular injection. This inhalable nanovaccine against SARS-CoV-2 offers a convenient and compliant strategy to reduce the use of needles and the need for medical staff. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (the immune activation of CS-mediated nanovacccine on BMDCs, cell viability, immune responses in lungs and BALF, serum chemistry and H&E histopathological analysis.) is available in the online version of this article at 10.1007/s12274-021-4012-9.
Collapse
Affiliation(s)
- Shao-Hua Zhuo
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084 China
| | - Jun-Jun Wu
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084 China
| | - Lang Zhao
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084 China
| | - Wen-Hao Li
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084 China
| | - Yu-Fen Zhao
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084 China
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315221 China
| | - Yan-Mei Li
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084 China
- Beijing Institute for Brain Disorders, Beijing, 100069 China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| |
Collapse
|
180
|
Alu A, Chen L, Lei H, Wei Y, Tian X, Wei X. Intranasal COVID-19 vaccines: From bench to bed. EBioMedicine 2022; 76:103841. [PMID: 35085851 PMCID: PMC8785603 DOI: 10.1016/j.ebiom.2022.103841] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 01/09/2022] [Accepted: 01/12/2022] [Indexed: 02/05/2023] Open
Abstract
Currently licensed COVID-19 vaccines are all designed for intramuscular (IM) immunization. However, vaccination today failed to prevent the virus infection through the upper respiratory tract, which is partially due to the absence of mucosal immunity activation. Despite the emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, the next generation of COVID-19 vaccine is in demand and intranasal (IN) vaccination method has been demonstrated to be potent in inducing both mucosal and systemic immune responses. Presently, although not licensed, various IN vaccines against SARS-CoV-2 are under intensive investigation, with 12 candidates reaching clinical trials at different phases. In this review, we give a detailed description about current status of IN COVID-19 vaccines, including virus-vectored vaccines, recombinant subunit vaccines and live attenuated vaccines. The ongoing clinical trials for IN vaccines are highlighted. Additionally, the underlying mechanisms of mucosal immunity and potential mucosal adjuvants and nasal delivery devices are also summarized.
Collapse
Affiliation(s)
- Aqu Alu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hong Lei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
181
|
Second Messenger 2'3'-cyclic GMP-AMP (2'3'-cGAMP):Synthesis, transmission, and degradation. Biochem Pharmacol 2022; 198:114934. [PMID: 35104477 DOI: 10.1016/j.bcp.2022.114934] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 01/07/2023]
Abstract
Cyclic GMP-AMP synthase (cGAS) senses foreign DNA to produce 2'3'-cyclic GMP-AMP (2'3'-cGAMP). 2'3'-cGAMP is a second messenger that binds and activates the adaptor protein STING, which triggers the innate immune response. As a STING agonist, the small molecule 2'3'-cGAMP plays pivotal roles in antiviral defense and has adjuvant applications, and anti-tumor effects. 2'3'-cGAMP and its analogs are thus putative targets for immunotherapy and are currently being testedin clinical trials to treat solid tumors. However, several barriers to further development have emerged from these studies, such as evidence of immune and inflammatory side-effects, poor pharmacokinetics, and undesirable biodistribution. Here, we review the status of 2'3'-cGAMP research and outline the role of 2'3'-cGAMP in immune signaling, adjuvant applications, and cancer immunotherapy, as well as various 2'3'-cGAMP detection methods.
Collapse
|
182
|
Ito S, Hirobe S, Yamashita R, Sugiyama A, Takeuchi H, Eguchi R, Yoshida J, Oyamada T, Tachibana M, Okada N. Analysis of immune response induction mechanisms implicating the dose-sparing effect of transcutaneous immunization using a self-dissolving microneedle patch. Vaccine 2022; 40:862-872. [PMID: 34998604 DOI: 10.1016/j.vaccine.2021.12.055] [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: 09/20/2021] [Revised: 12/19/2021] [Accepted: 12/25/2021] [Indexed: 11/28/2022]
Abstract
Transcutaneous immunization (TCI) is an effective vaccination method that is easier and less painful than the conventional injectable vaccination method. We previously developed self-dissolving microneedle patches (sdMN) and demonstrated that this TCI method has a high vaccination efficacy in mice and humans. To elucidate the mechanism of immune response induction, which is the basis for the efficacy and safety of TCI with sdMN, we examined the local reaction of the skin where sdMN was applied and the kinetics and differentiation status of immune cells in the draining lymph nodes (DLNs). We found that gene expression of the proinflammatory cytokine Il1b and the downstream transcription factor Irf7 was markedly upregulated in skin tissues after sdMN application. Moreover, activation of Langerhans cells and CD207- dermal dendritic cells, which are subsets of antigen-presenting cells (APCs) in the skin, and their migration to the DLNs were promoted. Furthermore, the activated APC subsets promoted CD4+ T cell and B cell differentiation and the formation of germinal centers, which are the sites of high-affinity antibody production. These phenomena associated with sdMN application may contribute to the efficient production of antigen-specific antibodies after TCI using sdMN. These findings provide essential information regarding immune response induction mechanisms for the development and improvement of TCI preparations.
Collapse
Affiliation(s)
- Sayami Ito
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Sachiko Hirobe
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Pharmacy, Osaka University Hospital, 2-15 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryo Yamashita
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Arisa Sugiyama
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Honoka Takeuchi
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryosuke Eguchi
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Junya Yoshida
- FUJIFILM Advanced Research Laboratories, Fujifilm Holdings Corporation, 577-1 Ushijima, Kaisei Town, Ashigarakami-gun, Kanagawa Prefecture 258-8577, Japan
| | - Takayoshi Oyamada
- FUJIFILM Advanced Research Laboratories, Fujifilm Holdings Corporation, 577-1 Ushijima, Kaisei Town, Ashigarakami-gun, Kanagawa Prefecture 258-8577, Japan
| | - Masashi Tachibana
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Vaccine and Immune Regulation (BIKEN), Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Naoki Okada
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Vaccine and Immune Regulation (BIKEN), Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
183
|
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
|
184
|
The Future of Nanomedicine. Nanomedicine (Lond) 2022. [DOI: 10.1007/978-981-13-9374-7_24-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
185
|
Zhao J, Song W, Tang Z, Chen X. Macromolecular Effects in Medicinal Chemistry ※. ACTA CHIMICA SINICA 2022. [DOI: 10.6023/a21120602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
186
|
Ou L, Zhang A, Cheng Y, Chen Y. The cGAS-STING Pathway: A Promising Immunotherapy Target. Front Immunol 2021; 12:795048. [PMID: 34956229 PMCID: PMC8695770 DOI: 10.3389/fimmu.2021.795048] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/22/2021] [Indexed: 01/07/2023] Open
Abstract
With the continuous development of immunotherapy, researchers have paid more attention to the specific immune regulatory mechanisms of various immune responses in different diseases. As a novel and vital innate immune signal pathway, the cGAS-STING signal pathway activated by nucleic acid substances, interplays with other immune responses, by which it participates in regulating cancer, autoimmune and inflammatory diseases, microbial and parasitic infectious diseases, and other diseases. With the exception of its role in innate immunity, the growing list of researches demonstrated expanding roles of the cGAS-STING signal pathway in bridging the innate immunity (macrophage polarization) with the adaptive immunity (T lymphocytes differentiation). Macrophages and T lymphocytes are the most representative cells of innate immunity and adaptive immunity, respectively. Their polarization or differentiation are involved in the pathogenesis and progression of various diseases. Here we mainly summarized recent advanced discoveries of how the cGAS-STING signal pathway regulated macrophages polarization and T lymphocytes differentiation in various diseases and vaccine applications, providing a promising direction for the development and clinical application of immunotherapeutic strategies for related diseases.
Collapse
Affiliation(s)
- Liang Ou
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, China
| | - Ao Zhang
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, China
| | - Yuxing Cheng
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, China
| | - Ying Chen
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
187
|
An oral vaccine based on chitosan/aluminum adjuvant induces both local and systemic immune responses in turbot (Scophthalmus maximus). Vaccine 2021; 39:7477-7484. [PMID: 34844823 DOI: 10.1016/j.vaccine.2021.10.063] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/14/2021] [Accepted: 10/25/2021] [Indexed: 02/03/2023]
Abstract
Oral vaccination is the most convenient method for disease protection in aquaculture due to possibility of adding vaccines as part of fish diet. However, low protective efficiency is the major problem of oral vaccination owing to some reasons, especially for antigens degradation. In this study, we developed an oral inactivated vaccine based on a chitosan/aluminum adjuvant system, and investigated immune response induced by the vaccine. As a result, Th1 and Th2 cells mediated immune responses were observed after the vaccination according to the upregulation of IL-6, IL-12, IFNγ and IgM genes expression that were deemed as the driver cytokines for triggering the responses. Moreover, ratio of IgT+/IgM+ B cells was elevated in intestine after vaccination, while IgM antibodies were also observed in the sera of vaccinated fish, suggested that both local and systemic immune responses were induced by the inactivated vaccine. Totally, this oral vaccine can be used for V. anguillarum protection and this chitosan/aluminum system is an efficient candidate adjuvant for developing more vaccines in the future.
Collapse
|
188
|
Wei Q, Su Y, Xin H, Zhang L, Ding J, Chen X. Immunologically Effective Biomaterials. ACS APPLIED MATERIALS & INTERFACES 2021; 13:56719-56724. [PMID: 34797622 DOI: 10.1021/acsami.1c14781] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Immunoregulation represents a booming field of biomaterial design. The unique physical and chemical properties of biomaterials offer tremendous opportunities for development. Each of their parameters exerts immunogenic effects at the immune system, cellular, and molecular levels. Herein, the perspective summarizes the interaction of biomaterials with immune cells and the underlying mechanisms to control immunoregulation in a top-down manner, providing solid inspiration for biomedical applications of immunologically effective biomaterials.
Collapse
Affiliation(s)
- Qi Wei
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, P. R. China
| | - Yuanzhen Su
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| | - Hua Xin
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, P. R. China
| | - Lening Zhang
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 220 Handan Road, Shanghai 200433, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| |
Collapse
|
189
|
Kimoto T. Development of a safe and effective novel synthetic mucosal adjuvant SF-10 derived from physiological metabolic pathways and function of human pulmonary surfactant. Vaccine 2021; 40:544-553. [PMID: 34887132 DOI: 10.1016/j.vaccine.2021.11.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A safe and effective mucosal adjuvant is required for vaccination against influenza A virus (IAV) infection. Previously, we described that intranasally administration of surfacten®, a medicine derived from bovine pulmonary surfactant (PS), with IAV vaccine can induce IAV-specific IgA in the respiratory tract mucosa and IgG in serum. PS is secreted by alveolar type II cells and Clara cells and serves to reduce lung surface tension. PS finished its rules is incorporated by antigen presenting cells (APCs), such as alveolar macrophages and dendritic cells, and alveolar type II cells and rapidly metabolized. We focused on the metabolic pathways and rapid metabolic turnover of PS and developed a PS-based mucosal adjuvant. First, we determined the essential components of PS adjuvanticity and found that the complex of three PS lipids and surfactant protein-C can enhance to deliver the vaccine antigen and activate APCs. Later, we improved the safety, efficacy and ease of manufacture and finally succeeded in developing SF-10. The use of SF-10 with influenza split vaccine (HAv) (HAv-SF-10) enhances HAv incorporation into APCs both in vitro and in vivo, and intranasal instillation of HAv-SF-10 induced systemic and mucosal HAv-specific immunities in not only mice but also cynomolgus monkeys. The report that PS has physiological effects on the gastrointestinal mucosa prompted us develop a new SF-10-based vaccine that can be administered orally. In this review, We summarize our work on the development of clinically effective PS-based nasal and oral mucosal adjuvants for influenza vaccine.
Collapse
Affiliation(s)
- Takashi Kimoto
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Tokushima, Japan.
| |
Collapse
|
190
|
Rezaei FS, Sharifianjazi F, Esmaeilkhanian A, Salehi E. Chitosan films and scaffolds for regenerative medicine applications: A review. Carbohydr Polym 2021; 273:118631. [PMID: 34561021 DOI: 10.1016/j.carbpol.2021.118631] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 01/01/2023]
Abstract
Over the last years, chitosan has demonstrated unparalleled characteristics for regenerative medicine applications. Beside excellent antimicrobial and wound healing properties, this polysaccharide biopolymer offers favorable characteristics such as biocompatibility, biodegradability, and film and fiber-forming capabilities. Having plentiful active amine groups, chitosan can be also readily modified to provide auxiliary features for growing demands in regenerative medicine, which is constantly confronted with new problems, necessitating the creation of biocompatible, immunogenic and biodegradable film/scaffold composites. A new look at the chitosan composites structure/activity/application tradeoff is the primary focus of the current review, which can help researchers to detect the bottlenecks and overcome the shortcomings that arose from this intersection. In the current review, the most recent advances in chitosan films and scaffolds in terms of preparation techniques and modifying methods for improving their functional properties, in three major biomedical fields i.e., tissue engineering, wound healing, and drug delivery are surveyed and discussed.
Collapse
Affiliation(s)
- Farnoush Sadat Rezaei
- Department of Chemical Engineering, Faculty of Engineering, Amir Kabir University, Tehran, Iran
| | - Fariborz Sharifianjazi
- Department of Mining and Metallurgical Engineering, Faculty of Engineering, Amir Kabir University, Tehran, Iran
| | - Amirhossein Esmaeilkhanian
- Department of Mining and Metallurgical Engineering, Faculty of Engineering, Amir Kabir University, Tehran, Iran
| | - Ehsan Salehi
- Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak 38156-88349, Iran.
| |
Collapse
|
191
|
Enriquez AB, Izzo A, Miller SM, Stewart EL, Mahon RN, Frank DJ, Evans JT, Rengarajan J, Triccas JA. Advancing Adjuvants for Mycobacterium tuberculosis Therapeutics. Front Immunol 2021; 12:740117. [PMID: 34759923 PMCID: PMC8572789 DOI: 10.3389/fimmu.2021.740117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/26/2021] [Indexed: 01/15/2023] Open
Abstract
Tuberculosis (TB) remains one of the leading causes of death worldwide due to a single infectious disease agent. BCG, the only licensed vaccine against TB, offers limited protection against pulmonary disease in children and adults. TB vaccine research has recently been reinvigorated by new data suggesting alternative administration of BCG induces protection and a subunit/adjuvant vaccine that provides close to 50% protection. These results demonstrate the need for generating adjuvants in order to develop the next generation of TB vaccines. However, development of TB-targeted adjuvants is lacking. To help meet this need, NIAID convened a workshop in 2020 titled “Advancing Vaccine Adjuvants for Mycobacterium tuberculosis Therapeutics”. In this review, we present the four areas identified in the workshop as necessary for advancing TB adjuvants: 1) correlates of protective immunity, 2) targeting specific immune cells, 3) immune evasion mechanisms, and 4) animal models. We will discuss each of these four areas in detail and summarize what is known and what we can advance on in order to help develop more efficacious TB vaccines.
Collapse
Affiliation(s)
- Ana B Enriquez
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Angelo Izzo
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Sydney, NSW, Australia
| | - Shannon M Miller
- Center for Translational Medicine, University of Montana, Missoula, MT, United States.,Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, United States
| | - Erica L Stewart
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Institute for Infectious Diseases and Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Robert N Mahon
- Division of AIDS, Columbus Technologies & Services Inc., Contractor to National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Daniel J Frank
- Division of AIDS, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, Missoula, MT, United States.,Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, United States
| | - Jyothi Rengarajan
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States.,Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States
| | - James A Triccas
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Institute for Infectious Diseases and Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
192
|
Wang Z, Yuan Y, Chen C, Zhang C, Huang F, Zhou M, Chen H, Fu ZF, Zhao L. Colloidal Manganese Salt Improves the Efficacy of Rabies Vaccines in Mice, Cats, and Dogs. J Virol 2021; 95:e0141421. [PMID: 34495701 PMCID: PMC8577392 DOI: 10.1128/jvi.01414-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022] Open
Abstract
Rabies, caused by rabies virus (RABV), remains a serious threat to public health in most countries worldwide. At present, the administration of rabies vaccines has been the most effective strategy to control rabies. Herein, we evaluate the effect of colloidal manganese salt (Mn jelly [MnJ]) as an adjuvant of rabies vaccine in mice, cats, and dogs. The results showed that MnJ promoted type I interferon (IFN-I) and cytokine production in vitro and the maturation of dendritic cells (DCs) in vitro and in vivo. Besides, MnJ serving as an adjuvant for rabies vaccines could significantly facilitate the generation of T follicular helper (Tfh) cells, germinal center (GC) B cells, plasma cells (PCs), and RABV-specific antibody-secreting cells (ASCs), consequently improve the immunogenicity of rabies vaccines, and provide better protection against virulent RABV challenge. Similarly, MnJ enhanced the humoral immune response in cats and dogs as well. Collectively, our results suggest that MnJ can facilitate the maturation of DCs during rabies vaccination, which can be a promising adjuvant candidate for rabies vaccines. IMPORTANCE Extending the humoral immune response by using adjuvants is an important strategy for vaccine development. In this study, a novel adjuvant, MnJ, supplemented in rabies vaccines was evaluated in mice, cats, and dogs. Our results in the mouse model revealed that MnJ increased the numbers of mature DCs, Tfh cells, GC B cells, PCs, and RABV-specific ASCs, resulting in enhanced immunogenicity and protection rate of rabies vaccines. We further found that MnJ had the same stimulative effect in cats and dogs. Our study provides the first evidence that MnJ serving as a novel adjuvant of rabies vaccines can boost the immune response in both a mouse and pet model.
Collapse
Affiliation(s)
- Zongmei Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yueming Yuan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chen Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chengguang Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Fei Huang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhen F. Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
193
|
Moine L, Canali MM, Porporatto C, Correa SG. Reviewing the biological activity of chitosan in the mucosa: Focus on intestinal immunity. Int J Biol Macromol 2021; 189:324-334. [PMID: 34419549 DOI: 10.1016/j.ijbiomac.2021.08.098] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/08/2021] [Accepted: 08/12/2021] [Indexed: 12/13/2022]
Abstract
Chitosan is a polymer derived from the partial deacetylation of chitin with particular characteristics, such as mucoadhesiveness, tolerability, biocompatibility and biodegradability. Biomedical uses of chitosan cover a wide spectrum of applications as dietary fiber, immunoadjuvant and regulator of the intestinal microbiota or delivery agent. Chemical modification of chitosan is feasible because its reactive amino and hydroxyl groups can be modified by a diverse array of ligands, functional groups and molecules. This gives rise to numerous derivatives that allow different formulation types influencing their activity. Considering the multiple events resulting from the interaction with mucosal tissues, chitosan is a singular candidate for strategies targeting immune stimulation (i.e., tolerance induction, vaccination). Its role as a prebiotic and probiotic carrier represents an effective option to manage intestinal dysbiosis. In the intestinal scenario where the exposure of the immune system to a wide variety of antigens is permanent, chitosan increases IgA levels and favors a tolerogenic environment, thus becoming a key ally for host homeostasis.
Collapse
Affiliation(s)
- L Moine
- Departamento de Bioquímica Clínica-Facultad de Ciencias Químicas-Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, CP: 5016 Córdoba Capital, Córdoba, Argentina
| | - M M Canali
- Université Côte d'Azur, INSERM, CNRS, IPMC, France
| | - C Porporatto
- Instituto Multidisciplinario de Investigación y Transferencia Agroalimentaria y Biotecnológica (IMITAB-CONICET), Universidad Nacional de Villa María (UNVM), Arturo Jauretche 1555, CP: 5900 Villa María, Córdoba, Argentina
| | - S G Correa
- Departamento de Bioquímica Clínica-Facultad de Ciencias Químicas-Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, CP: 5016 Córdoba Capital, Córdoba, Argentina.
| |
Collapse
|
194
|
Trincado V, Gala RP, Morales JO. Buccal and Sublingual Vaccines: A Review on Oral Mucosal Immunization and Delivery Systems. Vaccines (Basel) 2021; 9:vaccines9101177. [PMID: 34696284 PMCID: PMC8539688 DOI: 10.3390/vaccines9101177] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022] Open
Abstract
Currently, most vaccines available on the market are for parental use; however, this may not be the best option on several occasions. Mucosal routes of administration such as intranasal, sublingual, and buccal generate great interest due to the benefits they offer. These range from increasing patient compliance to inducing a more effective immune response than that achieved through conventional routes. Due to the activation of the common mucosal immune system, it is possible to generate an effective systemic and local immune response, which is not achieved through parenteral administration. Protection against pathogens that use mucosal entry routes is provided by an effective induction of mucosal immunity. Mucosal delivery systems are being developed, such as films and microneedles, which have proven to be effective, safe, and easy to administer. These systems have multiple advantages over commonly used injections, which are simple to manufacture, stable at room temperature, painless for the patient since they do not require puncture. Therefore, these delivery systems do not require to be administered by medical personnel; in fact, they could be self-administered.
Collapse
Affiliation(s)
- Valeria Trincado
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago 8380494, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile
- Center of New Drugs for Hypertension (CENDHY), Santiago 8380494, Chile
| | - Rikhav P. Gala
- Biotechnology Division, Center Mid-Atlantic, Fraunhofer USA, Newark, DE 19702, USA;
| | - Javier O. Morales
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago 8380494, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile
- Center of New Drugs for Hypertension (CENDHY), Santiago 8380494, Chile
- Correspondence:
| |
Collapse
|
195
|
Abstract
Cancer is one of the major causes of death worldwide. Chemotherapeutic drugs have become a popular choice as anticancer agents. Despite the therapeutic benefits of chemotherapeutic drugs, patients often experience side effects and drug resistance. Biopolymers could be used to overcome some of the limitations of chemotherapeutic drugs, as well as be used either as anticancer agents or drug delivery vehicles. Chitosan is a biocompatible polymer derived from chitin. Chitosan, chitosan derivatives, or chitosan nanoparticles have shown their promise as an anticancer agent. Additionally, functionally modified chitosan can be used to deliver nucleic acids, chemotherapeutic drugs, and anticancer agents. More importantly, chitosan-based drug delivery systems improved the efficacy, potency, cytotoxicity, or biocompatibility of these anticancer agents. In this review, we will investigate the properties of chitosan and chemically tuned chitosan derivatives, and their application in cancer therapy.
Collapse
|
196
|
Abstract
Innate immunity is regulated by a broad set of evolutionary conserved receptors to finely probe the local environment and maintain host integrity. Besides pathogen recognition through conserved motifs, several of these receptors also sense aberrant or misplaced self-molecules as a sign of perturbed homeostasis. Among them, self-nucleic acid sensing by the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway alerts on the presence of both exogenous and endogenous DNA in the cytoplasm. We review recent literature demonstrating that self-nucleic acid detection through the STING pathway is central to numerous processes, from cell physiology to sterile injury, auto-immunity and cancer. We address the role of STING in autoimmune diseases linked to dysfunctional DNAse or related to mutations in DNA sensing pathways. We expose the role of the cGAS/STING pathway in inflammatory diseases, neurodegenerative conditions and cancer. Connections between STING in various cell processes including autophagy and cell death are developed. Finally, we review proposed mechanisms to explain the sources of cytoplasmic DNA.
Collapse
Affiliation(s)
| | - Nicolas Riteau
- Experimental and Molecular Immunology and Neurogenetics Laboratory (INEM), Centre National de la Recherche Scientifique (CNRS), UMR7355 and University of Orleans, Orleans, France
| |
Collapse
|
197
|
Loading Effect of Chitosan Derivative Nanoparticles on Different Antigens and Their Immunomodulatory Activity on Dendritic Cells. Mar Drugs 2021; 19:md19100536. [PMID: 34677435 PMCID: PMC8541265 DOI: 10.3390/md19100536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 11/28/2022] Open
Abstract
Drug carrier nanoparticles (NPs) were prepared by the polyelectrolyte method, with chitosan sulfate, with different substituents and quaternary ammonium chitosan, including C236-HACC NPs, C36-HACC NPs, and C6-HACC NPs. To evaluate whether the NPs are suitable for loading different antigens, we chose bovine serum albumin (BSA), ovalbumin (OVA), and myoglobin (Mb) as model antigens to investigate the encapsulation effect of the NPs. The characteristics (size, potential, and encapsulation efficiency) of the NPs were measured. Moreover, the NPs with higher encapsulation efficiency were selected for the immunological activity research. The results showed that chitosan derivative NPs with different substitution sites had different loading effects on the three antigens, and the encapsulation rate of BSA and OVA was significantly better than that of Mb. Moreover, the NPs encapsulated with different antigens have different immune stimulating abilities to DCS cells, the immune effect of OVA-coated NPs was significantly better than that of BSA-coated NPs and blank NPs, especially C236-HACC-OVA NPs. Furthermore, we found that C236-HACC-OVA NPs could increase the phosphorylation level of intracellular proteins to activate cell pathways. Therefore, C236-HACC NPs are more suitable for the loading of antigens similar to the OVA structure.
Collapse
|
198
|
Vo NTN, Huang L, Lemos H, Mellor AL, Novakovic K. Genipin‐crosslinked chitosan hydrogels: Preliminary evaluation of the in vitro biocompatibility and biodegradation. J Appl Polym Sci 2021. [DOI: 10.1002/app.50848] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Nga T. N. Vo
- School of Engineering Newcastle University Newcastle Upon Tyne UK
| | - Lei Huang
- Translational and Clinical Research Institute Newcastle University Newcastle Upon Tyne UK
| | - Henrique Lemos
- Translational and Clinical Research Institute Newcastle University Newcastle Upon Tyne UK
| | - Andrew L. Mellor
- Translational and Clinical Research Institute Newcastle University Newcastle Upon Tyne UK
| | | |
Collapse
|
199
|
Cai SS, Li T, Akinade T, Zhu Y, Leong KW. Drug delivery carriers with therapeutic functions. Adv Drug Deliv Rev 2021; 176:113884. [PMID: 34302897 PMCID: PMC8440421 DOI: 10.1016/j.addr.2021.113884] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 01/07/2023]
Abstract
Design of micro- or nanocarriers for drug delivery has primarily been focused on properties such as hydrophobicity, biodegradability, size, shape, surface charge, and toxicity, so that they can achieve optimal delivery with respect to drug loading, release kinetics, biodistribution, cellular uptake, and biocompatibility. Incorporation of stimulus-sensitive moieties into the carriers would lead to "smart" delivery systems. A further evolution would be to endow the carrier with a therapeutic function such that it no longer serves as a mere passive entity to release the drug at the target tissue but can be viewed as a therapeutic agent in itself. In this review, we will discuss recent and ongoing efforts over the past decade to design therapeutic drug carriers that confer a biological benefit, including ROS scavenging or generating, pro- or anti-inflammatory, and immuno-evasive properties, to enhance the overall therapeutic efficacy of the delivery systems.
Collapse
Affiliation(s)
- Shuting S. Cai
- Department of Biomedical Engineering, Columbia University, New York 10027, New York, United States
| | - Tianyu Li
- Department of Biomedical Engineering, Columbia University, New York 10027, New York, United States
| | - Tolulope Akinade
- Graduate Program in Cellular, Molecular and Biomedical Studies, Vagelos College of Physicians and Surgeons, Columbia University, New York 10027, New York, United States
| | - Yuefei Zhu
- Department of Biomedical Engineering, Columbia University, New York 10027, New York, United States
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York 10027, New York, United States,Department of Systems Biology, Columbia University, New York 10027, New York, United States,Corresponding author , Mailing address: 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY 10027
| |
Collapse
|
200
|
Recent advances in immunotherapy, immunoadjuvant, and nanomaterial-based combination immunotherapy. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|