1
|
Wijfjes Z, Ramos Tomillero I, Le Gall CM, van Dinther EAW, Turlings F, Classens R, Manna S, van Dalen D, Peters RJRW, Schouren K, Fennemann FL, Hagemans IM, van Dalen FJ, van der Schoot JMS, Figdor CG, Esser-Kahn A, Scheeren FA, Verdoes M. Co-delivery of antigen and adjuvant by site-specific conjugation to dendritic cell-targeted Fab fragments potentiates T cell responses. RSC Chem Biol 2025:d5cb00014a. [PMID: 40343174 PMCID: PMC12057635 DOI: 10.1039/d5cb00014a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
The aim of therapeutic cancer vaccines is to induce tumor-specific cellular immune responses. This requires tumor antigens to be efficiently processed and presented by antigen-presenting cells, in particular dendritic cells (DCs). In addition, DCs require maturation to upregulate the surface expression and secretion of T cell costimulatory molecules, which is achieved by co-administration of adjuvants in vaccines. Peptide-based antigen vaccination is an attractive strategy due to the established biocompatibility of peptides as well as the dosing control. To enhance the efficacy of peptide-based vaccines, antigens can be targeted to DCs. Antigen-adjuvant conjugates are known to enhance T cell activation by ensuring DC maturation upon antigen delivery. In this study, we aim to combine these two approaches in a single molecule, and present a DC-targeted antibody fragment-antigen-adjuvant (AAA)-conjugate. We generate the AAA-conjugate through a combination of site-specific sortase-mediated chemoenzymatic ligation and click chemistry. Ex vivo T cell activation assays show enhanced efficacy of the AAA-conjugate compared to non-adjuvanted control conjugates. The in vivo performance of the AAA-conjugate was suboptimal, which we hypothesize to be a consequence of the hydrophobic character of the conjugate. In vivo efficacy was rescued by co-administration of antibody fragment-antigen conjugates and antibody fragment-adjuvant conjugates, in which the antigen and adjuvant were separatedly delivered using two different DC-targeting molecules. In conclusion, this study provides a proof-of-concept for effective in vivo antigen-specific T cell activation by targeted delivery of both antigen and adjuvant to DCs in a single or separate molecule using site-specific protein engineering.
Collapse
Affiliation(s)
- Zacharias Wijfjes
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
- Institute for Chemical Immunology Nijmegen The Netherlands
| | - Iván Ramos Tomillero
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Camille M Le Gall
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Eric A W van Dinther
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Frederique Turlings
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
- IMAGINE! Consortium Nijmegen The Netherlands
| | - René Classens
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Saikat Manna
- Pritzker School of Molecular Engineering, University of Chicago Chicago USA
| | - Duco van Dalen
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Ruud J R W Peters
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Kayleigh Schouren
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Felix L Fennemann
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Iris M Hagemans
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Floris J van Dalen
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
- Institute for Chemical Immunology Nijmegen The Netherlands
| | | | - Carl G Figdor
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
- Institute for Chemical Immunology Nijmegen The Netherlands
| | - Aaron Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago Chicago USA
| | - Ferenc A Scheeren
- Department of Dermatology, Leiden University Medical Center Leiden The Netherlands
| | - Martijn Verdoes
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
- Institute for Chemical Immunology Nijmegen The Netherlands
- IMAGINE! Consortium Nijmegen The Netherlands
| |
Collapse
|
2
|
Sun Y, Huang S, Li F, Huang S, Li P, Zhao Q, Wang T, Bao H, Fu Y, Sun P, Bai X, Yuan H, Ma X, Zhao Z, Zhang J, Wang J, Li D, Zhang Q, Cao Y, Li K, Lu Z, Fan H. Porcine antibodies reveal novel non-neutralizing universal epitopes on FMDV and their overlaps with neutralization sites. Vet Microbiol 2025; 303:110440. [PMID: 40037011 DOI: 10.1016/j.vetmic.2025.110440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/18/2025] [Accepted: 02/22/2025] [Indexed: 03/06/2025]
Abstract
Foot-and-mouth disease virus (FMDV) is highly infectious and lacks cross-protection among serotypes, with antibodies playing a key role in antiviral immunity. To map conserved epitopes on the FMDV surface that exhibit cross-serotype reactivity, we constructed a pig-specific B-cell receptor (BCR) library through single B-cell sorting and high-throughput sequencing. This led to the identification of 16 broadly reactive, non-neutralizing monoclonal antibodies (mAbs), with 10 targeting VP2 (pOTB-1, pOTB-10, pOTB-13, pOTB-33, pOTB-37, pONY-14, pONY-17, pONY-23, pONY-30, pONY-60) and 6 targeting VP3 (pOTB-6, pOTB-11, pOTB-22, pOTB-23, pONY-3, pONY-59). Among these, a novel free linear epitope was identified at the C-terminus of VP2, recognized by pOTB-1, with the minimal recognition motif "KE." Key residues, T53 and W101, within the complementarity-determining region (CDR) of the pOTB-1 heavy chain, interact with the carboxyl group of the C-terminal glutamate through hydrogen bonding, contributing to the free-form nature of the epitope. Competitive enzyme-linked immunosorbent assays (cELISA) showed that most non-neutralizing antibodies (nNAbs) interfered with the binding of neutralizing antibodies B82 (site 2) and C4 (site 4), confirming the overlap between non-neutralizing and neutralizing epitopes. It has been confirmed that nNAbs mediate antiviral activity in vivo through various mechanisms, such as the formation of immune complexes. These findings reveal new epitopes on VP2 and VP3 and their spatial overlap with neutralizing sites, enhancing our understanding of FMDV immunogenicity and providing novel targets for vaccine and therapeutic development.
Collapse
Affiliation(s)
- Ying Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China; Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China
| | - Shenglin Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Fengjuan Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Shulun Huang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Pinghua Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Qiongqiong Zhao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Tao Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Huifang Bao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Yuanfang Fu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Pu Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Xingwen Bai
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Hong Yuan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Xueqing Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Zhixun Zhao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Jing Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Jian Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Dong Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Qiang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
| | - Yimei Cao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China.
| | - Kun Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China.
| | - Zengjun Lu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China.
| | - Huiying Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China; Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou 510642, China.
| |
Collapse
|
3
|
Roshdy E, Taniguchi H, Nakamura Y, Takahashi H, Kikuchi Y, Celik I, Mohammed ESI, Ishihara Y, Morioka N, Abe M. Design, Synthesis, and Biological Evaluation of BODIPY-Caged Resiquimod as a Dual-Acting Phototherapeutic. J Med Chem 2025; 68:4561-4581. [PMID: 39960426 PMCID: PMC11873906 DOI: 10.1021/acs.jmedchem.4c02606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/28/2025]
Abstract
Resiquimod, an imidazoquinoline scaffold, exhibits potent immunotherapeutic activity but is associated with off-target effects, limiting its clinical utility. To address this limitation, we developed a novel BODIPY-caged resiquimod that is responsive to red light, combining photocaging and photodynamic therapy functionalities. Molecular docking studies guided identification of the optimal caging site for resiquimod, effectively masking its immune activity. BODIPY-caged resiquimod remained inactive under dark conditions, effectively masking resiquimod's immunostimulatory effects. However, red light irradiation precisely uncaged resiquimod, inducing robust immune activation, even in the presence of N-acetyl cysteine as an antioxidant. Notably, the attachment of resiquimod to BODIPY reduced the dark toxicity typically associated with BODIPY as a photosensitizer. In 3D spheroid models of HeLa and A549 cells, BODIPY-caged resiquimod demonstrated spatiotemporal control over cytotoxicity, significantly enhancing cell death only upon irradiation. This dual-function therapeutic approach highlights a "win-win" strategy: precise, red-light-mediated control of immune activation and photodynamic efficacy with reduced collateral toxicity.
Collapse
Affiliation(s)
- Eslam Roshdy
- Department
of Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, Higashi-Hiroshima City, Hiroshima 739-8526, Japan
- Medicinal
Chemistry Department, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Haruto Taniguchi
- Department
of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan
| | - Yoki Nakamura
- Department
of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan
| | - Haruko Takahashi
- Graduate
School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| | - Yutaka Kikuchi
- Graduate
School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| | - Ismail Celik
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, 38039 Kayseri, Turkey
| | - Elsayed S. I. Mohammed
- Avian
Research
Center, King Faisal University, Al Hofuf, Al-Ahsa 31982, Saudi Arabia
- Department
of Histology, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt
| | - Yasuhiro Ishihara
- Program
of Biomedical Science, Graduate School of Integrated Sciences for
Life, Hiroshima University, 1-7-1, Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8521, Japan
| | - Norimitsu Morioka
- Department
of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima 734-8553, Japan
| | - Manabu Abe
- Department
of Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, Higashi-Hiroshima City, Hiroshima 739-8526, Japan
| |
Collapse
|
4
|
Chen Y, Qin H, Li N, Wei Y, Lin Y, Deng R, Ding H, Lv Y, Ma T, Li R, Xiong C, Zheng G, Chen H, Shi J, Zhao Y, Zhao R, Nie G. Neoadjuvant chemotherapy by liposomal doxorubicin boosts immune protection of tumor membrane antigens-based nanovaccine. Cell Rep Med 2025; 6:101877. [PMID: 39708812 PMCID: PMC11866438 DOI: 10.1016/j.xcrm.2024.101877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/26/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024]
Abstract
Autologous tumor cell membrane antigen-based vaccines (TMVs) have garnered extensive attention as personalized immunotherapy. However, patients who take TMVs therapy usually undergo various treatments prior to surgery, and these processes modulate the immunogenicity of the tumor membrane and the tumor immune microenvironment. Herein, we investigate the impact of preoperative chemotherapy on the efficacy of TMVs. Liposomal doxorubicin ameliorates the immunosuppressive tumor microenvironment and enhances immunological molecule expression on the tumor membrane. This has driven TMVs to elicit a more robust immune response than doxorubicin, resulting in more effective immune protection. The TMVs formulated from liposomal doxorubicin-treated tumors induce superior dendritic cell maturation and T cell activation compared to doxorubicin, thus demonstrating better efficacy in preventing recurrence and metastasis in the postoperative murine model. Collectively, our study suggests that chemotherapy can induce immunomodulatory changes that augment the therapeutic potential of immunotherapy and provides valuable insights into the clinical utilization of TMVs.
Collapse
Affiliation(s)
- Yang Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Hao Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Nan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Yaohua Wei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Yixuan Lin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Ronghui Deng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Hao Ding
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Yuanyuan Lv
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Tianyu Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Rong Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Changhao Xiong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Guyu Zheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Hanqing Chen
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P.R. China
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P.R. China
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P.R. China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China.
| |
Collapse
|
5
|
Ye T, Zhong Z, Cappellesso F, Deswarte K, Chen Y, Lauwers H, De Lombaerde E, Gontsarik M, Lienenklaus S, Van Lysebetten D, Sanders NN, Lambrecht BN, De Koker S, Laoui D, De Geest BG. CO-DELIVERY of glutamic acid-extended peptide antigen and imidazoquinoline TLR7/8 agonist via ionizable lipid nanoparticles induces protective anti-tumor immunity. Biomaterials 2024; 311:122693. [PMID: 38996672 DOI: 10.1016/j.biomaterials.2024.122693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/30/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024]
Abstract
Cancer vaccines aim at generating cytotoxic CD8+ T cells that kill cancer cells and confer durable tumor regression. Hereto, CD8+ peptide epitopes should be presented by antigen presenting cells to CD8+ T cells in lymphoid tissue. Unfortunately, in unformulated soluble form, peptide antigens are poorly taken up by antigen presenting cells and do not efficiently reach lymph nodes. Hence, the lack of efficient delivery remains a major limitation for successful clinical translation of cancer vaccination using peptide antigens. Here we propose a generic peptide nanoformulation strategy by extending the amino acid sequence of the peptide antigen epitope with 10 glutamic acid residues. The resulting overall anionic charge of the peptide allows encapsulation into lipid nanoparticles (peptide-LNP) by electrostatic interaction with an ionizable cationic lipid. We demonstrate that intravenous injection of peptide-LNP efficiently delivers the peptide to immune cells in the spleen. Peptide-LNP that co-encapsulate an imidazoquinoline TLR7/8 agonist (IMDQ) induce robust innate immune activation in a broad range of immune cell subsets in the spleen. Peptide-LNP containing the minimal CD8+ T cell epitope of the HPV type 16 E7 oncoprotein and IMDQ induces high levels of antigen-specific CD8+ T cells in the blood, and can confer protective immunity against E7-expressing tumors in both prophylactic and therapeutic settings.
Collapse
Affiliation(s)
- Tingting Ye
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Zifu Zhong
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Federica Cappellesso
- Lab of Cellular and Molecular Immunology, Brussel Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
| | - Kim Deswarte
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Yong Chen
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Heleen Lauwers
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | | | - Mark Gontsarik
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Stefan Lienenklaus
- Institute of Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | | | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Ghent University, Merelbeke, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | | - Damya Laoui
- Lab of Cellular and Molecular Immunology, Brussel Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium.
| | | |
Collapse
|
6
|
Wang J, Zhao Z, Wang Q, Shi J, Wong DWC, Cheung JCW. Advancements in Nanoparticle-Based Adjuvants for Enhanced Tuberculosis Vaccination: A Review. Vaccines (Basel) 2024; 12:1335. [PMID: 39771997 PMCID: PMC11680411 DOI: 10.3390/vaccines12121335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Tuberculosis (TB) remains a leading cause of morbidity and mortality worldwide, necessitating the development of more effective vaccines. Nanoparticle-based adjuvants represent a promising approach to enhancing tuberculosis vaccine efficacy. This review focuses on the advantages of nanoparticulate-loaded vaccines, emphasizing their ability to improve antigen delivery, safety, and immunogenicity. We discuss the various types of nanoparticles and their unique physicochemical properties that contribute to improved antigen delivery and sustained immune activation. Additionally, we highlight the advantages of nanoparticle-based adjuvants in inducing strong cellular and humoral immunity, enhancing vaccine stability, and reducing adverse effects. Finally, we address current challenges and future perspectives in the application of these novel adjuvants, emphasizing their potential to transform TB vaccine strategies and ultimately contribute to better global health outcomes.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
- Department of Clinical Laboratory, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430073, China
| | - Zian Zhao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Quan Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Jingyu Shi
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Duo Wai-Chi Wong
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - James Chung-Wai Cheung
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| |
Collapse
|
7
|
Katebi A, Riazi-Rad F, Varshochian R, Ajdary S. PLGA nanoparticle-delivered Leishmania antigen and TLR agonists as a therapeutic vaccine against cutaneous leishmaniasis in BALB/c mice. Int Immunopharmacol 2024; 138:112538. [PMID: 38924865 DOI: 10.1016/j.intimp.2024.112538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/15/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
Leishmaniasis, caused by Leishmania (L.) species, remains a neglected infection. Therapeutic vaccination presents a promising strategy for its treatment. In this study, we aimed to develop a therapeutic vaccine candidate using Leishmaniaantigens (SLA) and Toll-like receptor (TLR) 7/8 agonist (R848) encapsulated into the poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NPs). Moreover, TLR1/2 agonist (Pam3CSK4) was loaded onto the NPs. The therapeutic effects of these NPs were evaluated in L. major-infected BALB/c mice. Footpad swelling, parasite load, cellular and humoral immune responses, and nitric oxide (NO) production were analyzed. The results demonstrated that the PLGA NPs (SLA-R848-Pam3CSK4) therapeutic vaccine effectively stimulated Th1 cell responses, induced humoral responses, promoted NO production, and restricted parasite burden and lesion size.Our findings suggest that vaccination with SLA combined with TLR1/2 and TLR7/8 agonists in PLGA NPs as a therapeutic vaccine confers strong protection againstL. majorinfection. These results represent a novel particulate therapeutic vaccine against Old World cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Asal Katebi
- Department of Immunology, Pasteur Institute of Iran, Tehran, IR, Iran.
| | - Farhad Riazi-Rad
- Department of Immunology, Pasteur Institute of Iran, Tehran, IR, Iran.
| | - Reyhaneh Varshochian
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, IR, Iran.
| | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, IR, Iran.
| |
Collapse
|
8
|
Chao PH, Chan V, Wu J, Andrew LJ, Li SD. Resiquimod-loaded cationic liposomes cure mice with peritoneal carcinomatosis and induce specific anti-tumor immunity. J Control Release 2024; 372:362-371. [PMID: 38909698 DOI: 10.1016/j.jconrel.2024.06.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Peritoneal carcinomatosis (PC) is characterized by a high recurrence rate and mortality following cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC), primarily due to incomplete cancer elimination. To enhance the standard of care for PC, we developed two cationic liposomal formulations aimed at localizing a toll-like receptor agonist, resiquimod (R848), in the peritoneal cavity to activate the immune system locally to specifically eradicate residual tumor cells. These formulations effectively extended R848 retention in the peritoneum by >10-fold, resulting in up to a 2-fold increase in interferon α (IFN-α) induction in the peritoneal fluid, without increasing the plasma levels. In a CT26 colon cancer model with peritoneal metastases, these liposomal R848 formulations, when combined with oxaliplatin (OXA)-an agent used in HIPEC that induces immunogenic cell death-increased tumor infiltration of effector immune cells, including DCs, CD4, and CD8 T cells. This led to the complete elimination of PC in 60-70% of the mice, while the control mice reached humane endpoints by 30 days. The cured mice developed specific antitumor immunity, as re-challenging them with the same tumor cells did not result in tumor establishment. However, inoculation with a different tumor line led to tumor development. Additionally, exposing CT26 tumor antigens to the splenocytes isolated from the cured mice induced the expansion of CD4 and CD8 T cells and the release of IFN-γ, demonstrating long-term immune memory to the specific tumor. The anti-tumor efficacy of these liposomal R848 formulations was mediated via CD8 T cells with different levels of involvement of CD4 and B cells, and the combination with an anti-PD-1 antibody achieved a cure rate of 90%.
Collapse
Affiliation(s)
- Po-Han Chao
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Vanessa Chan
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Jiamin Wu
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Lucas J Andrew
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.
| |
Collapse
|
9
|
Lou Z, Shi Y, Guo X, Jin Z, Huang J, Hu Y, Liu X, Zhu J, Kuang R, You J. Chronological Management of Adjuvant Effect for Optimized mRNA Vaccine Inspired by Natural Virus Infection. ACS NANO 2024. [PMID: 39011561 DOI: 10.1021/acsnano.4c04953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
The efficacy and safety of mRNA vaccines both rely on a fine-tuning of specific humoral and cellular immune responses. Instead of adjustments in vaccine component, we proposed a concept of chronological management of adjuvant effect to modulate the adaptive immune potency and preference inspired by natural virus infection. By simulating type I interferon expression dynamics during viral infection, three vaccine strategies employing distinct exposure sequences of adjuvant and mRNA have been developed, namely Precede, Coincide, and Follow. Follow, the strategy of adjuvant administration following mRNA, effectively suppressed tumor progression, which was attributed to enhanced mRNA translation, augmented p-MHC I expression, and elevated CD8+ T cell response. Meanwhile, Follow exhibited improved biosafety, characterized by reduced incidences of cardiac and liver toxicity, owing to its alteration to the vaccination microenvironment between successive injections. Our strategy highlights the importance of fine-tuning adjuvant effect dynamics in optimizing mRNA vaccines for clinical application.
Collapse
Affiliation(s)
- Zeliang Lou
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Xuemeng Guo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Zhaolei Jin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Jiaxin Huang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Yilong Hu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Xu Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Jiang Zhu
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P. R. China
| | - Rong Kuang
- Zhejiang Institute for Food and Drug Control, 325 Pingle Street, Hangzhou, Zhejiang 310004, P. R. China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang 310006, P. R. China
- The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310006, P. R. China
- Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang 321299, P. R. China
| |
Collapse
|
10
|
Choi YS, Felgner J, Jan S, Hernandez-Davies JE, Davies DH, Kwon YJ. Administration sequence- and formation-dependent vaccination using acid-degradable polymeric nanoparticles with high antigen encapsulation capability. J Mater Chem B 2024; 12:6577-6586. [PMID: 38872501 DOI: 10.1039/d3tb02834h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Vaccines aim to efficiently and specifically activate the immune system via a cascade of antigen uptake, processing, and presentation by antigen-presenting cells (APCs) to CD4 and CD8 T cells, which in turn drive humoral and cellular immune responses. The specific formulation of vaccine carriers can not only shield the antigens from premature sequestering before reaching APCs but also favorably promote intracellular antigen presentation and processing. This study compares two different acid-degradable polymeric nanoparticles that are capable of encapsulating a moderately immunogenic antigen, GFP, at nearly full efficacy via electrostatic interactions or molecular affinity between His tag and Ni-NTA-conjugated monomners. This resulted in GFP-encapsulating NPs composed of ketal monomers and crosslinkers (KMX/GFP NPs) and NTA-conjugated ketal monomers and crosslinkers (NKMX/GFP NPs), respectively. Encapsulated GFP was found to be released more rapidly from NKMX/GFP NPs (electrostatic encapsulation) than from KMX/GFP NPs (affinity-driven encapsulation). In vivo vaccination studies demonstrated that while repeated injections of either NP formulation resulted in poorer generation of anti-GFP antibodies than injections of the GFP antigen itself, sequential injections of NPs and GFP as prime and booster vaccines, respectively, restored the humoral response. We proposed that NPs primarily assist APCs in antigen presentation by T cells, and B cells need to be further stimulated by free protein antigens to produce antibodies. The findings of this study suggest that the immune response can be modulated by varying the chemistry of vaccine carriers and the sequences of vaccination with free antigens and antigen-encapsulating NPs.
Collapse
Affiliation(s)
- Yeon Su Choi
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA.
| | - Jiin Felgner
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA.
| | - Sharon Jan
- Vaccine Research and Development Center, University of California, Irvine, CA 92697, USA.
| | | | - D Huw Davies
- Vaccine Research and Development Center, University of California, Irvine, CA 92697, USA.
| | - Young Jik Kwon
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA.
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| |
Collapse
|
11
|
Rattan A, Malemnganba T, Sagar, Prajapati VK. Exploring structural engineering approach to formulate and characterize next-generation adjuvants. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:59-90. [PMID: 38762280 DOI: 10.1016/bs.apcsb.2023.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
It is critical to emphasize the importance of vaccination as it protects us against harmful pathogens. Despite significant progress in vaccine development, there is an ongoing need to develop vaccines that are not only safe but also highly effective in protecting against severe infections. Subunit vaccines are generally safe, but they frequently fail to elicit strong immune responses. As a result, there is a need to improve vaccine effectiveness by combining them with adjuvants, which have the potential to boost the immune system many folds. The process of developing these adjuvants requires searching for molecules capable of activating the immune system, combining these promising compounds with an antigen, and then testing this combination using animal models before approving it for clinical use. Liposomal adjuvants work as delivery adjuvants and its activity depends on certain parameters such as surface charge, vesicle size, surface modification and route of administration. Self-assembly property of peptide adjuvants and discovery of hybrid peptides have widened the scope of peptides in vaccine formulations. Since most pathogenic molecules are not peptide based, phage display technique allows for screening peptide mimics for such pathogens that have potential as adjuvants. This chapter discusses about peptide and liposome-based adjuvants focusing on their properties imparting adjuvanticity along with the methods of formulating them. Methods of adjuvant characterization important for an adjuvant to be approved for clinical trials are also discussed. These include assays for cytotoxicity, T-lymphocyte proliferation, dendritic cell maturation, cytokine and antibody production, toll-like receptor dependent signaling and adjuvant half-life.
Collapse
Affiliation(s)
- Aditi Rattan
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Takhellambam Malemnganba
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sagar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
12
|
Shin HS, Kim S, Jin SM, Yoo YJ, Heo JH, Lim YT. Molecular Masking of Synthetic Immunomodulator Evokes Antitumor Immunity With Reduced Immune Tolerance and Systemic Toxicity by Temporal Activity Recovery and Sustained Stimulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309039. [PMID: 37903320 DOI: 10.1002/adma.202309039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/17/2023] [Indexed: 11/01/2023]
Abstract
Activation of the innate immune system counteracts tumor-induced immunosuppression. Hence, small molecule-based toll-like receptor 7/8 agonists (TLR7/8a), which can modulate immunosuppression in the tumor microenvironment along with the activation of innate immunity, are emerging as essential components of cancer immunotherapy. However, the clinical application of synthetic TLR7/8a therapies is limited by systemic immune-associated toxicity and immune tolerance induced by uncontrolled stimulatory activities and repeated treatments. To address these limitations, a dynamic immunomodulation strategy incorporating masking and temporal recovery of the activity of TLR7/8a through prodrug-like TLR7/8a (pro-TLR7/8a) at the molecular level and a sustained and controlled release of active TLR7/8a from nanoliposome (pro-TLR7/8a) (NL(pro-TLR7/8)) in a macroscale depot are designed. Immunization with cationic NL(pro-TLR7/8) and anionic antigens triggers robust activation of innate immune cells as well as antigen-specific T cell responses, eliciting reprogramming of immunosuppressive cells into tumor-suppressive cells, with decreased systemic adverse effects and immune tolerance. Combination treatment with NL(pro-TLR7/8a) and immune checkpoint inhibitors (anti-CTLA-4 plus anti-PD-L1) or nanoliposomes (Doxorubicin) has synergistic effects on antitumor immunity in various tumor models. The concept of pro-TLR7/8a suggested herein may facilitate the advancement of small-molecule-based immunomodulators for clinical translation and safe and effective cancer immunotherapy.
Collapse
Affiliation(s)
- Hong Sik Shin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Sohyun Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Seung Mo Jin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Yeon Jeong Yoo
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Jang Hun Heo
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| |
Collapse
|
13
|
Davis MA, Cho E, Teplensky MH. Harnessing biomaterial architecture to drive anticancer innate immunity. J Mater Chem B 2023; 11:10982-11005. [PMID: 37955201 DOI: 10.1039/d3tb01677c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Immunomodulation is a powerful therapeutic approach that harnesses the body's own immune system and reprograms it to treat diseases, such as cancer. Innate immunity is key in mobilizing the rest of the immune system to respond to disease and is thus an attractive target for immunomodulation. Biomaterials have widely been employed as vehicles to deliver immunomodulatory therapeutic cargo to immune cells and raise robust antitumor immunity. However, it is key to consider the design of biomaterial chemical and physical structure, as it has direct impacts on innate immune activation and antigen presentation to stimulate downstream adaptive immunity. Herein, we highlight the widespread importance of structure-driven biomaterial design for the delivery of immunomodulatory cargo to innate immune cells. The incorporation of precise structural elements can be harnessed to improve delivery kinetics, uptake, and the targeting of biomaterials into innate immune cells, and enhance immune activation against cancer through temporal and spatial processing of cargo to overcome the immunosuppressive tumor microenvironment. Structural design of immunomodulatory biomaterials will profoundly improve the efficacy of current cancer immunotherapies by maximizing the impact of the innate immune system and thus has far-reaching translational potential against other diseases.
Collapse
Affiliation(s)
- Meredith A Davis
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
| | - Ezra Cho
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
| | - Michelle H Teplensky
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
- Department of Materials Science and Engineering, Boston University, Boston, Massachusetts, 02215, USA
| |
Collapse
|
14
|
Tambunlertchai S, Geary SM, Naguib YW, Salem AK. Investigating silver nanoparticles and resiquimod as a local melanoma treatment. Eur J Pharm Biopharm 2023; 183:1-12. [PMID: 36549400 PMCID: PMC10158852 DOI: 10.1016/j.ejpb.2022.12.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/20/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Over the last decade, the potential for silver nanoparticles (AgNP) to be used as an anti-melanoma agent has been supported by both in vitro and in vivo evidence. However, an undesirably high concentration of AgNP is often required to achieve an antitumor effect. Therefore a combination treatment that can maintain or improve antitumor efficacy (with lower amounts of AgNP) while also reducing off-target effects is sought. In this study, the combination of AgNP and resiquimod (RSQ: a Toll-like receptor agonist) was investigated and shown to significantly prolong the survival of melanoma-challenged mice when added sequentially. Results from toxicity studies showed that the treatment was non-toxic in mice. Immune cell depletion studies suggested the possible involvement of CD8+ T cells in the antitumor response observed in the AgNP + RSQ (sequential) treatment. NanoString was also employed to further understand the mechanism underlying the increase in the treatment efficacy of AgNP + RSQ (sequential); showing significant changes, compared to the naive group, in gene expression in pathways involved in apoptosis and immune stimulation. In conclusion, the combination of AgNP and RSQ is a new combination worthy of further investigation in the context of melanoma treatment.
Collapse
Affiliation(s)
- Supreeda Tambunlertchai
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Sean M Geary
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Youssef W Naguib
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
15
|
Aljabali AA, Obeid MA, Bashatwah RM, Serrano-Aroca Á, Mishra V, Mishra Y, El-Tanani M, Hromić-Jahjefendić A, Kapoor DN, Goyal R, Naikoo GA, Tambuwala MM. Nanomaterials and Their Impact on the Immune System. Int J Mol Sci 2023; 24:2008. [PMID: 36768330 PMCID: PMC9917130 DOI: 10.3390/ijms24032008] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/05/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
Nanomaterials have been the focus of intensive development and research in the medical and industrial sectors over the past several decades. Some studies have found that these compounds can have a detrimental impact on living organisms, including their cellular components. Despite the obvious advantages of using nanomaterials in a wide range of applications, there is sometimes skepticism caused by the lack of substantial proof that evaluates potential toxicities. The interactions of nanoparticles (NPs) with cells of the immune system and their biomolecule pathways are an area of interest for researchers. It is possible to modify NPs so that they are not recognized by the immune system or so that they suppress or stimulate the immune system in a targeted manner. In this review, we look at the literature on nanomaterials for immunostimulation and immunosuppression and their impact on how changing the physicochemical features of the particles could alter their interactions with immune cells for the better or for the worse (immunotoxicity). We also look into whether the NPs have a unique or unexpected (but desired) effect on the immune system, and whether the surface grafting of polymers or surface coatings makes stealth nanomaterials that the immune system cannot find and get rid of.
Collapse
Affiliation(s)
- Alaa A. Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, P.O. Box 566, Irbid 21163, Jordan
| | - Mohammad A. Obeid
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, P.O. Box 566, Irbid 21163, Jordan
| | - Rasha M. Bashatwah
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, P.O. Box 566, Irbid 21163, Jordan
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab., Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia, San Vicente Mártir, 46001 Valencia, Spain
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Yachana Mishra
- Department of Zoology, School of Bioengineering and Bioscience, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Mohamed El-Tanani
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina
| | - Deepak N. Kapoor
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India
| | - Rohit Goyal
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India
| | - Gowhar A. Naikoo
- Department of Mathematics and Sciences, College of Arts and Applied Sciences, Dhofar University, Salalah PC 211, Oman
| | - Murtaza M. Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK
| |
Collapse
|
16
|
Butkovich N, Tucker JA, Ramirez A, Li E, Meli VS, Nelson EL, Wang SW. Nanoparticle vaccines can be designed to induce pDC support of mDCs for increased antigen display. Biomater Sci 2023; 11:596-610. [PMID: 36476811 PMCID: PMC10775882 DOI: 10.1039/d2bm01132h] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cancer vaccine immunotherapy facilitates the immune system's recognition of tumor-associated antigens, and the biomolecular design of these vaccines using nanoparticles is one important approach towards obtaining strong anti-tumor responses. Following activation of dendritic cells (DCs), a robust CD8+ T cell-mediated adaptive immune response is critical for tumor elimination. While the role of efficient antigen-presenting myeloid DCs (mDCs) is conventionally attributed towards vaccine efficacy, participation by highly cytokine-producing plasmacytoid DCs (pDCs) is less understood and is often overlooked. We examined vaccines based on the E2 protein nanoparticle platform that delivered encapsulated TLR9 agonist bacterial-like DNA (CpG1826 or CpG1018) or TLR7 agonist viral ssRNA to determine their efficacy over free agonists in activating both mDCs and pDCs for antigen presentation. Although mDCs were only activated by nanoparticle-encapsulated TLR9 agonists, pDCs were activated by all the individually tested constructs, and CpG1826 was shown to induce pDC cytokine production. Transfer of secreted factors from pDCs that were stimulated with a vaccine formulation comprising peptide antigen and CpG1826 enhanced mDC display of the antigen, particularly when delivered in nanoparticles. Only when treated with nanoparticle-conjugated vaccine could pDCs secrete factors to induce antigen display on naïve mDCs. These results reveal that pDCs can aid mDCs, highlighting the importance of activating both pDCs and mDCs in designing effective cancer vaccines, and demonstrate the advantage of using nanoparticle-based vaccine delivery.
Collapse
Affiliation(s)
- Nina Butkovich
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA.
| | - Jo Anne Tucker
- Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Aaron Ramirez
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA.
| | - Enya Li
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA.
| | - Vijaykumar S Meli
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Edward L Nelson
- Department of Medicine, University of California, Irvine, CA 92697, USA
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA
- Institute for Immunology, University of California, Irvine, CA 92697, USA
| | - Szu-Wen Wang
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697, USA.
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA
- Institute for Immunology, University of California, Irvine, CA 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| |
Collapse
|
17
|
Shi T, Sun M, Lu C, Meng F. Self-assembled nanoparticles: A new platform for revolutionizing therapeutic cancer vaccines. Front Immunol 2023; 14:1125253. [PMID: 36895553 PMCID: PMC9988954 DOI: 10.3389/fimmu.2023.1125253] [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: 12/16/2022] [Accepted: 02/07/2023] [Indexed: 02/23/2023] Open
Abstract
Cancer vaccines have had some success in the past decade. Based on in-depth analysis of tumor antigen genomics, many therapeutic vaccines have already entered clinical trials for multiple cancers, including melanoma, lung cancer, and head and neck squamous cell carcinoma, which have demonstrated impressive tumor immunogenicity and antitumor activity. Recently, vaccines based on self-assembled nanoparticles are being actively developed as cancer treatment, and their feasibility has been confirmed in both mice and humans. In this review, we summarize recent therapeutic cancer vaccines based on self-assembled nanoparticles. We describe the basic ingredients for self-assembled nanoparticles, and how they enhance vaccine immunogenicity. We also discuss the novel design method for self-assembled nanoparticles that pose as a promising delivery platform for cancer vaccines, and the potential in combination with multiple therapeutic approaches.
Collapse
Affiliation(s)
- Tianyu Shi
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Mengna Sun
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Changchang Lu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Fanyan Meng
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
18
|
De Mel J, Hossain M, Shofolawe-Bakare O, Mohammad SA, Rasmussen E, Milloy K, Shields M, Roth EW, Arora K, Cueto R, Tang SC, Wilson JT, Smith AE, Werfel TA. Dual-Responsive Glycopolymers for Intracellular Codelivery of Antigen and Lipophilic Adjuvants. Mol Pharm 2022; 19:4705-4716. [PMID: 36374992 PMCID: PMC10013197 DOI: 10.1021/acs.molpharmaceut.2c00750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Traditional approaches to vaccines use whole organisms to trigger an immune response, but they do not typically generate robust cellular-mediated immunity and have various safety risks. Subunit vaccines composed of proteins and/or peptides represent an attractive and safe alternative to whole organism vaccines, but they are poorly immunogenic. Though there are biological reasons for the poor immunogenicity of proteins and peptides, one other key to their relative lack of immunogenicity could be attributed to the poor pharmacokinetic properties of exogenously delivered proteins and peptides. For instance, peptides often aggregate at the site of injection and are not stable in biological fluids, proteins and peptides are rapidly cleared from circulation, and both have poor cellular internalization and endosomal escape. Herein, we developed a delivery system to address the lack of protein immunogenicity by overcoming delivery barriers as well as codelivering immune-stimulating adjuvants. The glycopolymeric nanoparticles (glycoNPs) are composed of a dual-stimuli-responsive block glycopolymer, poly[2-(diisopropylamino)ethyl methacrylate]-b-poly[(pyridyl disulfide ethyl methacrylate)-co-(methacrylamidoglucopyranose)] (p[DPA-b-(PDSMA-co-MAG)]). This polymer facilitates protein conjugation and cytosolic release, the pH-responsive release of lipophilic adjuvants, and pH-dependent membrane disruption to ensure cytosolic delivery of antigens. We synthesized p[DPA-b-(PDSMA-co-MAG)] by reversible addition-fragmentation chain transfer (RAFT) polymerization, followed by the formation and physicochemical characterization of glycoNPs using the p[DPA-b-(PDSMA-co-MAG)] building blocks. These glycoNPs conjugated the model antigen ovalbumin (OVA) and released OVA in response to elevated glutathione levels. Moreover, the glycoNPs displayed pH-dependent drug release of the model hydrophobic drug Nile Red while also exhibiting pH-responsive endosomolytic behavior as indicated by a red blood cell hemolysis assay. GlycoNPs coloaded with OVA and the toll-like receptor 7/8 (TLR-7/8) agonist Resiquimod (R848) activated DC 2.4 dendritic cells (DCs) significantly more than free OVA and R848 and led to robust antigen presentation of the OVA epitope SIINFEKL on major histocompatibility complex I (MHC-I). In sum, the dual-stimuli-responsive glycopolymer introduced here overcomes major protein and peptide delivery barriers and could vastly improve the immunogenicity of protein-based vaccines.
Collapse
Affiliation(s)
- Judith De Mel
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Mehjabeen Hossain
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
| | - Oluwaseyi Shofolawe-Bakare
- Department of Chemical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Sk Arif Mohammad
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Emily Rasmussen
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
| | - Khadeeja Milloy
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Micaela Shields
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Eric W Roth
- Northwestern University Atomic and Nanoscale Characterization Experimental Center, Evanston, Illinois, 60208, United States
| | - Karan Arora
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Rafael Cueto
- Department of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Shou-Ching Tang
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi 39216, United States
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Adam E Smith
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
- Department of Chemical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Thomas A Werfel
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
- Department of Chemical Engineering, University of Mississippi, University, Mississippi 38677, United States
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi 39216, United States
| |
Collapse
|
19
|
Taylor D, Meyer CT, Graves D, Sen R, Fu J, Tran E, Mirza B, Rodriguez G, Lang C, Feng H, Quaranta V, Wilson JT, Kim YJ, Korrer MJ. MuSyC dosing of adjuvanted cancer vaccines optimizes antitumor responses. Front Immunol 2022; 13:936129. [PMID: 36059502 PMCID: PMC9437625 DOI: 10.3389/fimmu.2022.936129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
With the clinical approval of T-cell-dependent immune checkpoint inhibitors for many cancers, therapeutic cancer vaccines have re-emerged as a promising immunotherapy. Cancer vaccines require the addition of immunostimulatory adjuvants to increase vaccine immunogenicity, and increasingly multiple adjuvants are used in combination to bolster further and shape cellular immunity to tumor antigens. However, rigorous quantification of adjuvants' synergistic interactions is challenging due to partial redundancy in costimulatory molecules and cytokine production, leading to the common assumption that combining both adjuvants at the maximum tolerated dose results in optimal efficacy. Herein, we examine this maximum dose assumption and find combinations of these doses are suboptimal. Instead, we optimized dendritic cell activation by extending the Multidimensional Synergy of Combinations (MuSyC) framework that measures the synergy of efficacy and potency between two vaccine adjuvants. Initially, we performed a preliminary in vitro screening of clinically translatable adjuvant receptor targets (TLR, STING, NLL, and RIG-I). We determined that STING agonist (CDN) plus TLR4 agonist (MPL-A) or TLR7/8 agonist (R848) as the best pairwise combinations for dendritic cell activation. In addition, we found that the combination of R848 and CDN is synergistically efficacious and potent in activating both murine and human antigen-presenting cells (APCs) in vitro. These two selected adjuvants were then used to estimate a MuSyC-dose optimized for in vivo T-cell priming using ovalbumin-based peptide vaccines. Finally, using B16 melanoma and MOC1 head and neck cancer models, MuSyC-dose-based adjuvating of cancer vaccines improved the antitumor response, increased tumor-infiltrating lymphocytes, and induced novel myeloid tumor infiltration changes. Further, the MuSyC-dose-based adjuvants approach did not cause additional weight changes or increased plasma cytokine levels compared to CDN alone. Collectively, our findings offer a proof of principle that our MuSyC-extended approach can be used to optimize cancer vaccine formulations for immunotherapy.
Collapse
Affiliation(s)
- David Taylor
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Christian T. Meyer
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, United States
| | - Diana Graves
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rupashree Sen
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Juan Fu
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Emily Tran
- College Arts and Sciences, Vanderbilt University, Nashville, TN, United States
| | - Bilal Mirza
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Gabriel Rodriguez
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Cara Lang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Hanwen Feng
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Vito Quaranta
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
| | - John T. Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Young J. Kim
- Oncology Chair, Global Development, Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Michael J. Korrer
- Department of Otolaryngology Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
20
|
Chakraborty N, Jha D, Roy I, Kumar P, Gaurav SS, Marimuthu K, Ng OT, Lakshminarayanan R, Verma NK, Gautam HK. Nanobiotics against antimicrobial resistance: harnessing the power of nanoscale materials and technologies. J Nanobiotechnology 2022; 20:375. [PMID: 35953826 PMCID: PMC9371964 DOI: 10.1186/s12951-022-01573-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Given the spasmodic increment in antimicrobial resistance (AMR), world is on the verge of “post-antibiotic era”. It is anticipated that current SARS-CoV2 pandemic would worsen the situation in future, mainly due to the lack of new/next generation of antimicrobials. In this context, nanoscale materials with antimicrobial potential have a great promise to treat deadly pathogens. These functional materials are uniquely positioned to effectively interfere with the bacterial systems and augment biofilm penetration. Most importantly, the core substance, surface chemistry, shape, and size of nanomaterials define their efficacy while avoiding the development of AMR. Here, we review the mechanisms of AMR and emerging applications of nanoscale functional materials as an excellent substitute for conventional antibiotics. We discuss the potential, promises, challenges and prospects of nanobiotics to combat AMR.
Collapse
Affiliation(s)
- Nayanika Chakraborty
- Department of Chemistry, University of Delhi, New Delhi, 110007, India.,Department of Immunology and Infectious Disease Biology, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, New Delhi, 110025, India
| | - Diksha Jha
- Department of Immunology and Infectious Disease Biology, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, New Delhi, 110025, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Indrajit Roy
- Department of Chemistry, University of Delhi, New Delhi, 110007, India
| | - Pradeep Kumar
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi University Campus, 110007, New Delhi, India
| | - Shailendra Singh Gaurav
- Department of Genetics and Plant Breeding, Faculty of Agriculture, Chaudhary Charan Singh University, Meerut, 250004, India
| | - Kalisvar Marimuthu
- National Centre for Infectious Diseases (NCID), Singapore, 308442, Singapore.,Tan Tock Seng Hospital (TTSH), 308433, Singapore, Singapore
| | - Oon-Tek Ng
- National Centre for Infectious Diseases (NCID), Singapore, 308442, Singapore.,Tan Tock Seng Hospital (TTSH), 308433, Singapore, Singapore
| | - Rajamani Lakshminarayanan
- Ocular Infections and Anti-Microbials Research Group, Singapore Eye Research Institute, The Academia, 20 College Road, Singapore, 169856, Singapore. .,Department of Pharmacy, National University of Singapore, Singapore, 117543, Singapore. .,Academic Clinical Program in Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore, 169857, Singapore.
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore. .,National Skin Centre, Singapore, 308205, Singapore.
| | - Hemant K Gautam
- Department of Immunology and Infectious Disease Biology, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, New Delhi, 110025, India.
| |
Collapse
|
21
|
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: 33] [Impact Index Per Article: 11.0] [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
|
22
|
Sharma BK, Ramakrishan S, Kaliappan A, Singh M, Kumar A, Dandapat S, Dey S, Chellappa MM. Evaluation of a Lipopolysaccharide and Resiquimod Combination as an Adjuvant with Inactivated Newcastle Disease Virus Vaccine in Chickens. Vaccines (Basel) 2022; 10:vaccines10060894. [PMID: 35746503 PMCID: PMC9229813 DOI: 10.3390/vaccines10060894] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 11/16/2022] Open
Abstract
Various toll-like receptor (TLR) agonists have shown potential as adjuvants with different vaccines in both human and livestock species, including chickens. Our previous studies on combination of lipopolysaccharide (LPS; TLR4 agonist) and resiquimod (R-848; TLR7 agonist) showed the synergistic up-regulation of pro-inflammatory Th1 and Th2 cytokines in chicken peripheral blood mononuclear cells (PMBCs). Hence, the present study aimed to explore the combined adjuvant effect of LPS and R-848 with inactivated Newcastle disease virus (NDV) vaccine in chickens. Two weeks-old SPF chickens were immunized with inactivated NDV vaccine along with a combination of LPS and R-848 as an adjuvant with suitable control groups. A booster dose was given two weeks later. Antibody responses were assessed by enzyme linked immunosorbent assay (ELISA) and hemagglutination inhibition (HI) test, while cell-mediated immune responses were analyzed by a lymphocyte transformation test (LTT) and flow cytometry following vaccination. Two weeks post-booster, the birds were challenged with a velogenic strain of NDV, and protection against clinical signs, mortality and virus shedding was analyzed. The results indicated that inactivated NDV vaccine with R-848 induced significantly higher humoral and cellular immune responses with 100% protection against mortality and viral shedding following a virulent NDV challenge. However, the combination of LPS and R-848 along with inactivated NDV vaccine produced poor humoral and cellular immune responses and could not afford protection against challenge infection and virus shedding when compared to the vaccine-alone group, indicating the deleterious effects of the combination on antigen-specific immune responses. In conclusion, the combination of LPS and R-848 showed the inhibitory effects on antigen-specific humoral, cellular and protective immune responses when used as an adjuvant with inactivated NDV vaccines in chickens. This inhibitory effect might have occurred due to systemic cytokine storm. A nanoparticle-based delivery of the combination of LPS and R-848 for slow and sustained release could be tried as an alternative method to explore the synergistic effect of the combination as an adjuvant in chickens.
Collapse
Affiliation(s)
- Bal Krishnan Sharma
- Immunology Section, Indian Veterinary Research Institute, Bareilly 243122, India; (B.K.S.); (A.K.); (M.S.); (S.D.)
| | - Saravanan Ramakrishan
- Immunology Section, Indian Veterinary Research Institute, Bareilly 243122, India; (B.K.S.); (A.K.); (M.S.); (S.D.)
- Correspondence: ; Tel.: +91-941-246-3498
| | - Abinaya Kaliappan
- Immunology Section, Indian Veterinary Research Institute, Bareilly 243122, India; (B.K.S.); (A.K.); (M.S.); (S.D.)
| | - Mithilesh Singh
- Immunology Section, Indian Veterinary Research Institute, Bareilly 243122, India; (B.K.S.); (A.K.); (M.S.); (S.D.)
| | - Ajay Kumar
- Division of Animal Biochemistry, Indian Veterinary Research Institute, Bareilly 243122, India;
| | - Satyabrata Dandapat
- Immunology Section, Indian Veterinary Research Institute, Bareilly 243122, India; (B.K.S.); (A.K.); (M.S.); (S.D.)
| | - Sohini Dey
- Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Bareilly 243122, India; (S.D.); (M.M.C.)
| | - Madhan Mohan Chellappa
- Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Bareilly 243122, India; (S.D.); (M.M.C.)
| |
Collapse
|
23
|
Systemic Administration of the TLR7/8 Agonist Resiquimod (R848) to Mice Is Associated with Transient, In Vivo-Detectable Brain Swelling. BIOLOGY 2022; 11:biology11020274. [PMID: 35205140 PMCID: PMC8869423 DOI: 10.3390/biology11020274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/17/2022]
Abstract
Peripheral administration of the E. coli endotoxin lipopolysaccharide (LPS) to rats promotes secretion of pro-inflammatory cytokines and in previous studies was associated with transient enlargement of cortical volumes. Here, resiquimod (R848) was administered to mice to stimulate peripheral immune activation, and the effects on brain volumes and neurometabolites determined. After baseline scans, 24 male, wild-type C57BL mice were triaged into three groups including R848 at low (50 μg) and high (100 μg) doses and saline controls. Animals were scanned again at 3 h and 24 h following treatment. Sickness indices of elevated temperature and body weight loss were observed in all R848 animals. Animals that received 50 μg R848 exhibited decreases in hippocampal N-acetylaspartate and phosphocreatine at the 3 h time point that returned to baseline levels at 24 h. Animals that received the 100 μg R848 dose demonstrated transient, localized, volume expansion (~5%) detectable at 3 h in motor, somatosensory, and olfactory cortices; and pons. A metabolic response evident at the lower dose and a volumetric change at the higher dose suggests a temporal evolution of the effect wherein the neurochemical change is demonstrable earlier than neurostructural change. Transient volume expansion in response to peripheral immune stimulation corresponds with previous results and is consistent with brain swelling that may reflect CNS edema.
Collapse
|
24
|
Van Herck S, Feng B, Tang L. Delivery of STING agonists for adjuvanting subunit vaccines. Adv Drug Deliv Rev 2021; 179:114020. [PMID: 34756942 DOI: 10.1016/j.addr.2021.114020] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/16/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023]
Abstract
Adjuvant is an essential component in subunit vaccines. Many agonists of pathogen recognition receptors have been developed as potent adjuvants to optimize the immunogenicity and efficacy of vaccines. Recently discovered cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway has attracted much attention as it is a key mediator for modulating immune responses. Vaccines adjuvanted with STING agonists are found to mediate a robust immune defense against infections and cancer. In this review, we first discuss the mechanisms of STING agonists in the context of vaccination. Next, we present recent progress in novel STING agonist discovery and the delivery strategies. We next highlight recent work in optimizing the efficacy while minimizing toxicity of STING agonist-assisted subunit vaccines for protection against infectious diseases or treatment of cancer. Finally, we share our perspectives of current issues and future directions in further developing STING agonists for adjuvanting subunit vaccines.
Collapse
Affiliation(s)
- Simon Van Herck
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Department of Pharmaceutics, Ghent University, 9000 Ghent, Belgium
| | - Bing Feng
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Institute of Materials Science & Engineering, EPFL, 1015 Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Institute of Materials Science & Engineering, EPFL, 1015 Lausanne, Switzerland.
| |
Collapse
|
25
|
Allahyari M, Golkar M, Fard-Esfahani P, Dimier-Poisson I, Mévélec MN. Co-delivery of PLGA nanoparticles loaded with rSAG1 antigen and TLR ligands: An efficient vaccine against chronic toxoplasmosis. Microb Pathog 2021; 162:105312. [PMID: 34826553 DOI: 10.1016/j.micpath.2021.105312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/19/2022]
Abstract
Although vaccination is a promising approach for the control of toxoplasmosis, there is currently no commercially available human vaccine. Adjuvants such as delivery vehicles and immunomodulators are critical components of vaccine formulations. In this study, Poly (D, l-lactide-co-glycolide) (PLGA) nanoparticles were applied to serve as delivery system for both surface antigen-1 (SAG1), a candidate vaccine against toxoplasmosis and two TLR ligands, monophosphoryl lipid A (MPL) and imiquimod (IMQ), respectively. Compared to rSAG1 alone, CBA/J mice immunized with rSAG1-PLGA produced higher anti-SAG1 IgG antibodies titers. This response was increased by the co-administration of IMQ-PLGA (p < 0.01). Compared to IMQ-PLGA co-administration, MPL-PLGA co-administration further increased the humoral response (p < 0.01) and potentiated the Th1 humoral response. Compared to rSAG1 alone, rSAG1-PLGA, or rSAG1-PLGA mixed with IMQ-PLGA or MPL-PLGA similarly enhanced the cellular response characterized by the production of IFN-γ, IL-2, TNF-α and low levels of IL-5, indicating a Th1-biased immunity. The induced immune responses, led to significant brain cyst reductions (p < 0.01) after oral challenge with T. gondii cysts in mice immunized with either rSAG1-PLGA, rSAG1-PLGA + IMQ-PLGA, rSAG1-PLGA + MPL-PLGA formulations. Taken together the results indicated that PLGA nanoparticles could serve as a platform for dual-delivery of antigens and immunomodulators to provide efficacious vaccines against toxoplasmosis.
Collapse
Affiliation(s)
- Mojgan Allahyari
- Recombinant Protein Production Department, Research and Production Complex, Pasteur Institute of Iran, Karaj, Iran.
| | - Majid Golkar
- Molecular Parasitology Laboratory, Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran.
| | | | | | | |
Collapse
|
26
|
Sartorius R, Trovato M, Manco R, D'Apice L, De Berardinis P. Exploiting viral sensing mediated by Toll-like receptors to design innovative vaccines. NPJ Vaccines 2021; 6:127. [PMID: 34711839 PMCID: PMC8553822 DOI: 10.1038/s41541-021-00391-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
Toll-like receptors (TLRs) are transmembrane proteins belonging to the family of pattern-recognition receptors. They function as sensors of invading pathogens through recognition of pathogen-associated molecular patterns. After their engagement by microbial ligands, TLRs trigger downstream signaling pathways that culminate into transcriptional upregulation of genes involved in immune defense. Here we provide an updated overview on members of the TLR family and we focus on their role in antiviral response. Understanding of innate sensing and signaling of viruses triggered by these receptors would provide useful knowledge to prompt the development of vaccines able to elicit effective and long-lasting immune responses. We describe the mechanisms developed by viral pathogens to escape from immune surveillance mediated by TLRs and finally discuss how TLR/virus interplay might be exploited to guide the design of innovative vaccine platforms.
Collapse
Affiliation(s)
- Rossella Sartorius
- Institute of Biochemistry and Cell Biology, C.N.R., Via Pietro Castellino 111, 80131, Naples, Italy.
| | - Maria Trovato
- Institute of Biochemistry and Cell Biology, C.N.R., Via Pietro Castellino 111, 80131, Naples, Italy
| | - Roberta Manco
- Institute of Biochemistry and Cell Biology, C.N.R., Via Pietro Castellino 111, 80131, Naples, Italy
| | - Luciana D'Apice
- Institute of Biochemistry and Cell Biology, C.N.R., Via Pietro Castellino 111, 80131, Naples, Italy.
| | | |
Collapse
|
27
|
Johnson L, Aglas L, Soh WT, Geppert M, Hofer S, Hofstätter N, Briza P, Ferreira F, Weiss R, Brandstetter H, Duschl A, Himly M. Structural Alterations of Antigens at the Material Interface: An Early Decision Toolbox Facilitating Safe-by-Design Nanovaccine Development. Int J Mol Sci 2021; 22:10895. [PMID: 34639235 PMCID: PMC8509464 DOI: 10.3390/ijms221910895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022] Open
Abstract
Nanomaterials have found extensive interest in the development of novel vaccines, as adjuvants and/or carriers in vaccination platforms. Conjugation of protein antigens at the particle surface by non-covalent adsorption is the most widely used approach in licensed particulate vaccines. Hence, it is essential to understand proteins' structural integrity at the material interface in order to develop safe-by-design nanovaccines. In this study, we utilized two model proteins, the wild-type allergen Bet v 1 and its hypoallergenic fold variant (BM4), to compare SiO2 nanoparticles with Alhydrogel® as particulate systems. A set of biophysical and functional assays including circular dichroism spectroscopy and proteolytic degradation was used to examine the antigens' structural integrity at the material interface. Conjugation of both biomolecules to the particulate systems decreased their proteolytic stability. However, we observed qualitative and quantitative differences in antigen processing concomitant with differences in their fold stability. These changes further led to an alteration in IgE epitope recognition. Here, we propose a toolbox of biophysical and functional in vitro assays for the suitability assessment of nanomaterials in the early stages of vaccine development. These tools will aid in safe-by-design innovations and allow fine-tuning the properties of nanoparticle candidates to shape a specific immune response.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Martin Himly
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; (L.J.); (L.A.); (W.T.S.); (M.G.); (S.H.); (N.H.); (P.B.); (F.F.); (R.W.); (H.B.); (A.D.)
| |
Collapse
|
28
|
Pusch L, Brox R, Scheuer K, Yokosawa T, Wu M, Zubiri BA, Spiecker E, Jandt KD, Fischer D, Hackstein H. Distinct endocytosis and immune activation of poly(lactic-co-glycolic) acid nanoparticles prepared by single- and double-emulsion evaporation. Nanomedicine (Lond) 2021; 16:2075-2094. [PMID: 34523349 DOI: 10.2217/nnm-2021-0022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background: Poly(lactic-co-glycolic) acid (PLGA) nanoparticles can be prepared by emulsion-solvent-evaporation from o/w and w1/o/w2 emulsions. Aims: To elaborate similarities and differences regarding mechanical, morphological and physicochemical properties, as well as endocytosis and dose-dependent immune responses by primary human leukocytes between nanoparticles prepared by these two methods. Methods: Fluorescently labeled as well as TLR agonist (R848)-loaded PLGA nanoparticles were prepared via both single- and double-emulsion solvent evaporation. Results: Particles prepared by both methods were similar in chemical composition and surface charge but exhibited slight differences in size and morphology. Pronounced differences were found for loading, dissolution and mechanical properties. The particles were differently endocytosed by monocytes and induced qualitatively and quantitatively different immune responses. Conclusions: Variations in nanoparticle preparation can affect particle-derived immunological characteristics.
Collapse
Affiliation(s)
- Lennart Pusch
- Department of Transfusion Medicine & Hemostaseology, University Hospital Erlangen, Krankenhausstraße 12, Erlangen, 91054, Germany
| | - Regine Brox
- Department of Transfusion Medicine & Hemostaseology, University Hospital Erlangen, Krankenhausstraße 12, Erlangen, 91054, Germany
| | - Karl Scheuer
- Department of Materials Science & Technology, Otto Schott Institute of Materials Research, Friedrich Schiller University Jena, Löbdergraben 32, Jena, 07743, Germany
| | - Tadahiro Yokosawa
- Institute of Micro- & Nanostructure Research (IMN) & Center for Nanoanalysis & Electron Microscopy (CENEM), Interdisciplinary Center for Nanostructured Films (IZNF), Friedrich-Alexander-University Erlangen-Nürnberg, Cauerstraße 3, Erlangen, 91058, Germany
| | - Mingjian Wu
- Institute of Micro- & Nanostructure Research (IMN) & Center for Nanoanalysis & Electron Microscopy (CENEM), Interdisciplinary Center for Nanostructured Films (IZNF), Friedrich-Alexander-University Erlangen-Nürnberg, Cauerstraße 3, Erlangen, 91058, Germany
| | - Benjamin Apeleo Zubiri
- Institute of Micro- & Nanostructure Research (IMN) & Center for Nanoanalysis & Electron Microscopy (CENEM), Interdisciplinary Center for Nanostructured Films (IZNF), Friedrich-Alexander-University Erlangen-Nürnberg, Cauerstraße 3, Erlangen, 91058, Germany
| | - Erdmann Spiecker
- Institute of Micro- & Nanostructure Research (IMN) & Center for Nanoanalysis & Electron Microscopy (CENEM), Interdisciplinary Center for Nanostructured Films (IZNF), Friedrich-Alexander-University Erlangen-Nürnberg, Cauerstraße 3, Erlangen, 91058, Germany
| | - Klaus D Jandt
- Department of Materials Science & Technology, Otto Schott Institute of Materials Research, Friedrich Schiller University Jena, Löbdergraben 32, Jena, 07743, Germany.,Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, Jena, 07743, Germany
| | - Dagmar Fischer
- Friedrich-Alexander-University Erlangen-Nürnberg, Cauerstraße 4 (Haus 6), Erlangen, 91058, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine & Hemostaseology, University Hospital Erlangen, Krankenhausstraße 12, Erlangen, 91054, Germany
| |
Collapse
|
29
|
Li K, Zhu G, Zhou S, Sun P, Wang H, Bao H, Fu Y, Li P, Bai X, Ma X, Zhang J, Li D, Chen Y, Liu Z, Cao Y, Lu Z. Isolation and characterization of porcine monoclonal antibodies revealed two distinct serotype-independent epitopes on VP2 of foot-and-mouth disease virus. J Gen Virol 2021; 102. [PMID: 34280085 DOI: 10.1099/jgv.0.001608] [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
Pigs are susceptible to foot-and-mouth disease virus (FMDV), and the humoral immune response plays an essential role in protection against FMDV infection. However, little information is available about FMDV-specific mAbs derived from single B cells of pigs. This study aimed to determine the antigenic features of FMDV that are recognized by antibodies from pigs. Therefore, a panel of pig-derived mAbs against FMDV were developed using fluorescence-based single B cell antibody technology. Western blotting revealed that three of the antibodies (1C6, P2-7E and P2-8G) recognized conserved antigen epitopes on capsid protein VP2, and exhibited broad reactivity against both FMDV serotypes A and O. An alanine-substitution scanning assay and sequence conservation analysis elucidated that these porcine mAbs recognized two conserved epitopes on VP2: a linear epitope (2KKTEETTLL10) in the N terminus and a conformational epitope involving residues K63, H65, L66, F67, D68 and L81 on two β-sheets (B-sheet and C-sheet) that depended on the integrity of VP2. Random parings of heavy and light chains of the IgGs confirmed that the heavy chain is predominantly involved in binding to antigen. The light chain of porcine IgG contributes to the binding affinity toward an antigen and may function as a support platform for antibody stability. In summary, this study is the first to reveal the conserved antigenic profile of FMDV recognized by porcine B cells and provides a novel method for analysing the antibody response against FMDV in its natural hosts (i.e. pigs) at the clonal level.
Collapse
Affiliation(s)
- Kun Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Guoqiang Zhu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Shasha Zhou
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Pu Sun
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Hengmei Wang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Huifang Bao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Yuanfang Fu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Pinghua Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Xingwen Bai
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Xueqing Ma
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Jing Zhang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Dong Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Yingli Chen
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Zaixin Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Yimei Cao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| | - Zengjun Lu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Lanzhou 730046, PR China
| |
Collapse
|
30
|
Bhagchandani S, Johnson JA, Irvine DJ. Evolution of Toll-like receptor 7/8 agonist therapeutics and their delivery approaches: From antiviral formulations to vaccine adjuvants. Adv Drug Deliv Rev 2021; 175:113803. [PMID: 34058283 PMCID: PMC9003539 DOI: 10.1016/j.addr.2021.05.013] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/04/2021] [Accepted: 05/15/2021] [Indexed: 02/07/2023]
Abstract
Imidazoquinoline derivatives (IMDs) and related compounds function as synthetic agonists of Toll-like receptors 7 and 8 (TLR7/8) and one is FDA approved for topical antiviral and skin cancer treatments. Nevertheless, these innate immune system-activating drugs have potentially much broader therapeutic utility; they have been pursued as antitumor immunomodulatory agents and more recently as candidate vaccine adjuvants for cancer and infectious disease. The broad expression profiles of TLR7/8, poor pharmacokinetic properties of IMDs, and toxicities associated with systemic administration, however, are formidable barriers to successful clinical translation. Herein, we review IMD formulations that have advanced to the clinic and discuss issues related to biodistribution and toxicity that have hampered the further development of these compounds. Recent strategies aimed at enhancing safety and efficacy, particularly through the use of bioconjugates and nanoparticle formulations that alter pharmacokinetics, biodistribution, and cellular targeting, are described. Finally, key aspects of the biology of TLR7 signaling, such as TLR7 tolerance, that may need to be considered in the development of new IMD therapeutics are discussed.
Collapse
Affiliation(s)
- Sachin Bhagchandani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jeremiah A Johnson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
31
|
Qin L, Zhang H, Zhou Y, Umeshappa CS, Gao H. Nanovaccine-Based Strategies to Overcome Challenges in the Whole Vaccination Cascade for Tumor Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006000. [PMID: 33768693 DOI: 10.1002/smll.202006000] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/26/2020] [Indexed: 06/12/2023]
Abstract
Nanovaccine-based immunotherapy (NBI) has received greater attention recently for its potential to prime tumor-specific immunity and establish a long-term immune memory that prevents tumor recurrence. Despite encouraging results in the recent studies, there are still numerous challenges to be tackled for eliciting potent antitumor immunity using NBI strategies. Based on the principles that govern immune response, here it is proposed that these challenges need to be addressed at the five critical cascading events: Loading tumor-specific antigens by nanoscale drug delivery systems (L); Draining tumor antigens to lymph nodes (D); Internalization by dendritic cells (DCs) (I); Maturation of DCs by costimulatory signaling (M); and Presenting tumor-peptide-major histocompatibility complexes to T cells (P) (LDIMP cascade in short). This review provides a detailed and objective overview of emerging NBI strategies to improve the efficacy of nanovaccines in each step of the LDIMP cascade. It is concluded that the balance between each step must be optimized by delicate designing and modification of nanovaccines and by combining with complementary approaches to provide a synergistic immunity in the fight against cancer.
Collapse
Affiliation(s)
- Lin Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- Chongqing Vocational College of Transportation, Chongqing, 400715, China
| | - Huilin Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yang Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Channakeshava Sokke Umeshappa
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
32
|
Banete A, Gee K, Basta S. Sustained IL-4 priming of macrophages enhances the inflammatory response to TLR7/8 ligand R848. J Leukoc Biol 2021; 111:401-413. [PMID: 34013552 DOI: 10.1002/jlb.3a0520-293rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Macrophages (Mϕ) are highly plastic, and can acquire a variety of functional phenotypes depending on the presence of different stimuli in their local environment. Mφ stimulated by interleukin (IL)-4 induce an alternative activation state and function as anti-inflammatory cells and promote tissue repair. However, there is overwhelming evidence that IL-4 can play a role in promoting inflammation. In asthma and allergic inflammation, IL-4 mediates proinflammatory responses that lead to tissue damage. Thus the effect of IL-4 on the outcome of the immune responses is greatly influenced by other cofactors and cytokines present in the microenvironment. R848 (resiquimod), a TLR7/8 agonist is a novel vaccine adjuvant, triggering a strong Th1-skewed response but its efficacy as a vaccine adjuvant shows variable results. It is not currently known whether the presence of IL-4 can dampen or enhance immunity in response to TLR7 agonists. In the present study, we sought to investigate the impact of IL-4-induced Mφ polarization on the outcome of R848 stimulation. The activation marker expression and production of cytokines were measured in murine spleen-derived Mφ. Protein expression levels of innate recognition molecules and transcription factors involved, including retinoic-acid inducible gene I, mitochondrial antiviral signaling protein, stimulator of interferon genes (STING), and IFN regulatory factors were evaluated in activated Mφ. These play a crucial role in the control of viral replication and optimal CD8+ T cell priming. We report that sustained priming with IL-4 alone promotes an antiviral response in Mφ, and enhances proinflammatory responses to R848 treatment. This highlights the need for better understanding of IL-4 proinflammatory functions and its potential use as a broad-acting antiviral in combination with R848 may be used in combination with other therapies to target the innate arm of immunity against emerging infections.
Collapse
Affiliation(s)
- Andra Banete
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Sameh Basta
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
33
|
Kakwere H, Zhang H, Ingham ES, Nura-Raie M, Tumbale SK, Allen R, Tam SM, Wu B, Liu C, Kheirolomoom A, Fite BZ, Ilovitsh A, Lewis JS, Ferrara KW. Systemic Immunotherapy with Micellar Resiquimod-Polymer Conjugates Triggers a Robust Antitumor Response in a Breast Cancer Model. Adv Healthc Mater 2021; 10:e2100008. [PMID: 33646600 PMCID: PMC8153207 DOI: 10.1002/adhm.202100008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/31/2021] [Indexed: 12/16/2022]
Abstract
Resiquimod is an immunopotent toll-like receptor 7/8 agonist with antitumor activity. Despite being potent against skin cancers, it is poorly tolerated systemically due to toxicity. Integrating resiquimod into nanoparticles presents an avenue to circumvent the toxicity problem. Herein, the preparation of degradable nanoparticles with covalently bound resiquimod and their systemic application in cancer immunotherapy is reported. Dispersion in water of amphiphilic constructs integrating resiquimod covalently bound via degradable amide or ester linkages yields immune-activating nanoparticles. The degradable agonist-nanoparticle bonds allow the release of resiquimod from the carrier nanoparticles. In vitro assays with antigen presenting cells demonstrate that the nanoparticles retain the immunostimulatory activity of resiquimod. Systemic administration of the nanoparticles and checkpoint blockade (aPD-1) to a breast cancer mouse model with multiple established tumors triggers antitumor activity evidenced by suppressed tumor growth and enhanced CD8+ T-cell infiltration. Nanoparticles with ester links, which hydrolyze more readily, yield a stronger immune response with 75% of tumors eliminated when combined with aPD-1. The reduced tumor growth and the presence of activated CD8+ T-cells across multiple tumors suggest the potential for treating metastatic cancer.
Collapse
Affiliation(s)
- Hamilton Kakwere
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Hua Zhang
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California (Davis), Davis, CA, 95616, USA
| | - Marina Nura-Raie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Spencer K Tumbale
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Riley Allen
- Department of Biomedical Engineering, University of California (Davis), Davis, CA, 95616, USA
| | - Sarah M Tam
- Department of Biomedical Engineering, University of California (Davis), Davis, CA, 95616, USA
| | - Bo Wu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Cheng Liu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Azadeh Kheirolomoom
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Brett Z Fite
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Asaf Ilovitsh
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Jamal S Lewis
- Department of Biomedical Engineering, University of California (Davis), Davis, CA, 95616, USA
| | - Katherine W Ferrara
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| |
Collapse
|
34
|
Chin AL, Wang X, Tong R. Aliphatic Polyester-Based Materials for Enhanced Cancer Immunotherapy. Macromol Biosci 2021; 21:e2100087. [PMID: 33909344 DOI: 10.1002/mabi.202100087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/07/2021] [Indexed: 12/19/2022]
Abstract
Poly(lactic acid) (PLA) and its copolymer, poly(lactic-co-glycolic acid) (PLGA), based aliphatic polyesters have been extensively used for biomedical applications, such as drug delivery system and tissue engineering, thanks to their biodegradability, benign toxicity, renewability, and adjustable mechanical properties. A rapidly growing field of cancer research, the development of therapeutic cancer vaccines or treatment modalities is aimed to deliver immunomodulatory signals that control the quality of immune responses against tumors. Herein, the progress and applications of PLA and PLGA are reviewed in delivering immunotherapeutics to treat cancers.
Collapse
Affiliation(s)
- Ai Lin Chin
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| | - Xiaoqian Wang
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| | - Rong Tong
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| |
Collapse
|
35
|
Chen PM, Pan WY, Luo PK, Phung HN, Liu YM, Chiang MC, Chang WA, Tien TL, Huang CY, Wu WW, Chia WT, Sung HW. Pollen-Mimetic Metal-Organic Frameworks with Tunable Spike-Like Nanostructures That Promote Cell Interactions to Improve Antigen-Specific Humoral Immunity. ACS NANO 2021; 15:7596-7607. [PMID: 33760607 DOI: 10.1021/acsnano.1c01129] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The exine capsules of pollen particles exhibit a variety of characteristic surface morphologies that promote their cell interactions; their use as antigen carriers for vaccination has been proposed. However, the allergy-causing substances in pollen particles may not all be removed, even by vigorous chemical treatments. To resolve this issue, this work develops systemic approaches for synthesizing pollen-mimetic metal-organic frameworks (MOFs), which comprise aluminum (Al) ions and an organic linker (2-aminoterephthalic acid), with tunable spike-like nanostructures on their surfaces. The as-synthesized MOFs act not only as a delivery vehicle that carries a model antigen (ovalbumin, OVA) but also as an adjuvant (Al). Scanning and transmission electron microscopies images reveal that the aspect ratio of the nanospikes that are grown on the MOFs can be controlled. A higher aspect ratio of the nanospikes on the MOFs is associated with greater cell attachment and faster and more efficient phagocytosis in cells, which results in greater expressions of pro-inflammatory cytokines. Consequently, a more robust immune response against the antigen of interest is elicited. These findings have broad implications for the rational design of the future antigen/adjuvant-presenting particles for vaccination.
Collapse
Affiliation(s)
- Po-Ming Chen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan, ROC
| | - Wen-Yu Pan
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan, ROC
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan, ROC
| | - Po-Kai Luo
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan, ROC
| | - Hieu Nghia Phung
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan, ROC
| | - Yu-Miao Liu
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan, ROC
| | - Min-Chun Chiang
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan, ROC
| | - Wan-An Chang
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan, ROC
| | - Ting-Lun Tien
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan, ROC
| | - Chih-Yang Huang
- Department of Materials Science and Engineering and Center for Intelligent Semiconductor Nano-System Technology Research, National Chiao Tung University, Hsinchu 300, Taiwan, ROC
| | - Wen-Wei Wu
- Department of Materials Science and Engineering and Center for Intelligent Semiconductor Nano-System Technology Research, National Chiao Tung University, Hsinchu 300, Taiwan, ROC
| | - Wei-Tso Chia
- Department of Orthopedics, National Taiwan University Hospital, Hsinchu Branch, Hsinchu 300, Taiwan, ROC
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan, ROC
| |
Collapse
|
36
|
Lin W, Li C, Xu N, Watanabe M, Xue R, Xu A, Araki M, Sun R, Liu C, Nasu Y, Huang P. Dual-Functional PLGA Nanoparticles Co-Loaded with Indocyanine Green and Resiquimod for Prostate Cancer Treatment. Int J Nanomedicine 2021; 16:2775-2787. [PMID: 33880023 PMCID: PMC8052122 DOI: 10.2147/ijn.s301552] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/24/2021] [Indexed: 12/28/2022] Open
Abstract
PURPOSE With the advance of screening techniques, there is a growing number of low-risk or intermediate-risk prostate cancer (PCa) cases, remaining a serious threat to men's health. To obtain better efficacy, a growing interest has been attracted to develop such emerging treatments as immunotherapy and focal therapy. However, few studies offer guidance on whether and how to combine these modalities against PCa. This study was designed to develop dual-functional nanoparticles (NPs) which combined photothermal therapy (PTT) with immunotherapy and determine the anti-tumor efficacy for PCa treatment. METHODS By a double emulsion technique, the drug nanocarrier, poly(lactic-co-glycolic acid) or PLGA, was applied for co-loading of a fluorescent dye, indocyanine green (ICG) and a toll-like receptor 7/8 (TLR7/8) agonist resiquimod (R848) to synthesize PLGA-ICG-R848 NPs. Next, we determined their characteristic features and evaluated whether they inhibited the cell viability in multiple PCa cell lines. After treatment with PLGA-ICG-R848, the maturation markers of bone marrow-derived dendritic cells (BMDCs) were detected by flow cytometry. By establishing a subcutaneous xenograft model of mouse PCa, we explored both the anti-tumor effect and immune response following the NPs-based laser ablation. RESULTS With a mean diameter of 157.7 nm, PLGA-ICG-R848 exhibited no cytotoxic effect in PCa cells, but they significantly decreased RM9 cell viability to (3.9±1.0)% after laser irradiation. Moreover, PLGA-ICG-R848 promoted BMDCs maturation with the significantly elevated proportions of CD11c+CD86+ and CD11c+CD80+ cells. Following PLGA-ICG-R848-based laser ablation in vivo, the decreased bioluminescent signals indicated a significant inhibition of PCa growth, while the ratio of splenic natural killer (NK) cells in PLGA-ICG-R848 was (3.96±1.88)% compared with (0.99±0.10)% in PBS group, revealing the enhanced immune response against PCa. CONCLUSION The dual-functional PLGA-ICG-R848 NPs under laser irradiation exhibit the anti-tumor efficacy for PCa treatment by combining PTT with immunotherapy.
Collapse
Affiliation(s)
- Wenfeng Lin
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Chaoming Li
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Naijin Xu
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Masami Watanabe
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Ruizhi Xue
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Abai Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Motoo Araki
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ruifen Sun
- Center for Scientific Research, Yunnan University of Chinese Traditional Medicine, Kunming, People’s Republic of China
| | - Chunxiao Liu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yasutomo Nasu
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Peng Huang
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- Okayama Medical Innovation Center, Okayama University, Okayama, Japan
| |
Collapse
|
37
|
Fries CN, Curvino EJ, Chen JL, Permar SR, Fouda GG, Collier JH. Advances in nanomaterial vaccine strategies to address infectious diseases impacting global health. NATURE NANOTECHNOLOGY 2021; 16:1-14. [PMID: 32807876 DOI: 10.1038/s41565-020-0739-9] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 06/23/2020] [Indexed: 05/18/2023]
Abstract
Despite the overwhelming success of vaccines in preventing infectious diseases, there remain numerous globally devastating diseases without fully protective vaccines, particularly human immunodeficiency virus (HIV), malaria and tuberculosis. Nanotechnology approaches are being developed both to design new vaccines against these diseases as well as to facilitate their global implementation. The reasons why a given pathogen may present difficulties for vaccine design are unique and tied to the co-evolutionary history of the pathogen and humans, but there are common challenges that nanotechnology is beginning to help address. In each case, a successful vaccine will need to raise immune responses that differ from the immune responses raised by normal infection. Nanomaterials, with their defined compositions, commonly modular construction, and length scales allowing the engagement of key immune pathways, collectively facilitate the iterative design process necessary to identify such protective immune responses and achieve them reliably. Nanomaterials also provide strategies for engineering the trafficking and delivery of vaccine components to key immune cells and lymphoid tissues, and they can be highly multivalent, improving their engagement with the immune system. This Review will discuss these aspects along with recent nanomaterial advances towards vaccines against infectious disease, with a particular emphasis on HIV/AIDS, malaria and tuberculosis.
Collapse
Affiliation(s)
- Chelsea N Fries
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Jui-Lin Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Sallie R Permar
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Genevieve G Fouda
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
38
|
Wagner J, Gößl D, Ustyanovska N, Xiong M, Hauser D, Zhuzhgova O, Hočevar S, Taskoparan B, Poller L, Datz S, Engelke H, Daali Y, Bein T, Bourquin C. Mesoporous Silica Nanoparticles as pH-Responsive Carrier for the Immune-Activating Drug Resiquimod Enhance the Local Immune Response in Mice. ACS NANO 2021; 15:4450-4466. [PMID: 33648336 DOI: 10.1021/acsnano.0c08384] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nanoparticle-based delivery systems for cancer immunotherapies aim to improve the safety and efficacy of these treatments through local delivery to specialized antigen-presenting cells (APCs). Multifunctional mesoporous silica nanoparticles (MSNs), with their large surface areas, their tunable particle and pore sizes, and their spatially controlled functionalization, represent a safe and versatile carrier system. In this study, we demonstrate the potential of MSNs as a pH-responsive drug carrier system for the anticancer immune-stimulant R848 (resiquimod), a synthetic Toll-like receptor 7 and 8 agonist. Equipped with a biotin-avidin cap, the tailor-made nanoparticles showed efficient stimuli-responsive release of their R848 cargo in an environmental pH of 5.5 or below. We showed that the MSNs loaded with R848 were rapidly taken up by APCs into the acidic environment of the lysosome and that they potently activated the immune cells. Upon subcutaneous injection into mice, the particles accumulated in migratory dendritic cells (DCs) in the draining lymph nodes, where they strongly enhanced the activation of the DCs. Furthermore, simultaneous delivery of the model antigen OVA and the adjuvant R848 by MSNs resulted in an augmented antigen-specific T-cell response. The MSNs significantly improved the pharmacokinetic profile of R848 in mice, as the half-life of the drug was increased 6-fold, and at the same time, the systemic exposure was reduced. In summary, we demonstrate that MSNs represent a promising tool for targeted delivery of the immune modulator R848 to APCs and hold considerable potential as a carrier for cancer vaccines.
Collapse
Affiliation(s)
- Julia Wagner
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Dorothée Gößl
- Department of Chemistry and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Natasha Ustyanovska
- Department of Chemistry and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Mengyao Xiong
- Department of Chemistry and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Daniel Hauser
- Adolphe Merkle Institute, University of Fribourg, 1700 Fribourg, Switzerland
| | - Olga Zhuzhgova
- Department of Chemistry and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Sandra Hočevar
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Betül Taskoparan
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Laura Poller
- Department of Chemistry and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Stefan Datz
- Department of Chemistry and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Hanna Engelke
- Department of Chemistry and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Youssef Daali
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Thomas Bein
- Department of Chemistry and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Carole Bourquin
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
- Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
39
|
Gebre MS, Brito LA, Tostanoski LH, Edwards DK, Carfi A, Barouch DH. Novel approaches for vaccine development. Cell 2021; 184:1589-1603. [PMID: 33740454 PMCID: PMC8049514 DOI: 10.1016/j.cell.2021.02.030] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023]
Abstract
Vaccines are critical tools for maintaining global health. Traditional vaccine technologies have been used across a wide range of bacterial and viral pathogens, yet there are a number of examples where they have not been successful, such as for persistent infections, rapidly evolving pathogens with high sequence variability, complex viral antigens, and emerging pathogens. Novel technologies such as nucleic acid and viral vector vaccines offer the potential to revolutionize vaccine development as they are well-suited to address existing technology limitations. In this review, we discuss the current state of RNA vaccines, recombinant adenovirus vector-based vaccines, and advances from biomaterials and engineering that address these important public health challenges.
Collapse
Affiliation(s)
- Makda S. Gebre
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- co-first authors
| | | | - Lisa H. Tostanoski
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- co-first authors
| | | | - Andrea Carfi
- Moderna, Inc., Cambridge, MA, USA
- Massachusetts Consortium on Pathogen Readiness, Boston, MA, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Massachusetts Consortium on Pathogen Readiness, Boston, MA, USA
| |
Collapse
|
40
|
Genito CJ, Batty CJ, Bachelder EM, Ainslie KM. Considerations for Size, Surface Charge, Polymer Degradation, Co-Delivery, and Manufacturability in the Development of Polymeric Particle Vaccines for Infectious Diseases. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000041. [PMID: 33681864 PMCID: PMC7917382 DOI: 10.1002/anbr.202000041] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/04/2020] [Indexed: 01/15/2023] Open
Abstract
Vaccines have advanced human health for centuries. To improve upon the efficacy of subunit vaccines they have been formulated into nano/microparticles for infectious diseases. Much progress in the field of polymeric particles for vaccine formulation has been made since the push for a tetanus vaccine in the 1990s. Modulation of particle properties such as size, surface charge, degradation rate, and the co-delivery of antigen and adjuvant has been used. This review focuses on advances in the understanding of how these properties influence immune responses to injectable polymeric particle vaccines. Consideration is also given to how endotoxin, route of administration, and other factors influence conclusions that can be made. Current manufacturing techniques involved in preserving vaccine efficacy and scale-up are discussed, as well as those for progressing polymeric particle vaccines toward commercialization. Consideration of all these factors should aid the continued development of efficacious and marketable polymeric particle vaccines.
Collapse
Affiliation(s)
- Christopher J. Genito
- Department of Microbiology and ImmunologyUniversity of North Carolina at Chapel Hill4211 Marsico Hall, 125 Mason Farm RoadChapel HillNC27599USA
| | - Cole J. Batty
- Division of Pharma Engineering & Molecular PharmaceuticsEshelman School of PharmacyUniversity of North Carolina at Chapel Hill4211 Marsico Hall, 125 Mason Farm RoadChapel HillNC27599USA
| | - Eric M. Bachelder
- Division of Pharma Engineering & Molecular PharmaceuticsEshelman School of PharmacyUniversity of North Carolina at Chapel Hill4211 Marsico Hall, 125 Mason Farm RoadChapel HillNC27599USA
| | - Kristy M. Ainslie
- Division of Pharma Engineering & Molecular PharmaceuticsEshelman School of PharmacyUniversity of North Carolina at Chapel Hill4211 Marsico Hall, 125 Mason Farm RoadChapel HillNC27599USA
| |
Collapse
|
41
|
Andrianov AK, Langer R. Polyphosphazene immunoadjuvants: Historical perspective and recent advances. J Control Release 2021; 329:299-315. [PMID: 33285104 PMCID: PMC7904599 DOI: 10.1016/j.jconrel.2020.12.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
The development of successful vaccines has been increasingly reliant on the use of immunoadjuvants - additives, which can enhance and modulate immune responses to vaccine antigens. Immunoadjuvants of the polyphosphazene family encompass synthetic biodegradable macromolecules, which attain in vivo activity via antigen delivery and immunostimulation mechanisms. Over the last decades, the technology has witnessed evolvement of next generation members, expansion to include various antigens and routes of administration, and progression to clinical phase. This was accompanied by gaining important insights into the mechanism of action and the development of a novel class of virus-mimicking nano-assemblies for antigen delivery. The present review evaluates in vitro and in vivo data generated to date in the context of latest advances in understanding the primary function and biophysical behavior of these macromolecules. It also provides an overview of relevant synthetic and characterization methods, macromolecular biodegradation pathways, and polyphosphazene-based multi-component, nanoparticulate, and microfabricated formulations.
Collapse
Affiliation(s)
- Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA.
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
42
|
Islam MA, Rice J, Reesor E, Zope H, Tao W, Lim M, Ding J, Chen Y, Aduluso D, Zetter BR, Farokhzad OC, Shi J. Adjuvant-pulsed mRNA vaccine nanoparticle for immunoprophylactic and therapeutic tumor suppression in mice. Biomaterials 2021; 266:120431. [PMID: 33099060 PMCID: PMC7528902 DOI: 10.1016/j.biomaterials.2020.120431] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/15/2020] [Accepted: 09/30/2020] [Indexed: 12/15/2022]
Abstract
Synthetic mRNA represents an exciting cancer vaccine technology for the implementation of effective cancer immunotherapy. However, inefficient in vivo mRNA delivery along with a requirement for immune co-stimulation present major hurdles to achieving anti-tumor therapeutic efficacy. Here, we demonstrate a proof-of-concept adjuvant-pulsed mRNA vaccine nanoparticle (NP) that is composed of an ovalbumin-coded mRNA and a palmitic acid-modified TLR7/8 agonist R848 (C16-R848), coated with a lipid-polyethylene glycol (lipid-PEG) shell. This mRNA vaccine NP formulation retained the adjuvant activity of encapsulated C16-R848 and markedly improved the transfection efficacy of the mRNA (>95%) and subsequent MHC class I presentation of OVA mRNA derived antigen in antigen-presenting cells. The C16-R848 adjuvant-pulsed mRNA vaccine NP approach induced an effective adaptive immune response by significantly improving the expansion of OVA-specific CD8+ T cells and infiltration of these cells into the tumor bed in vivo, relative to the mRNA vaccine NP without adjuvant. The approach led to an effective anti-tumor immunity against OVA expressing syngeneic allograft mouse models of lymphoma and prostate cancer, resulting in a significant prevention of tumor growth when the vaccine was given before tumor engraftment (84% reduction vs. control) and suppression of tumor growth when given post engraftment (60% reduction vs. control). Our findings indicate that C16-R848 adjuvant pulsation to mRNA vaccine NP is a rational design strategy to increase the effectiveness of synthetic mRNA vaccines for cancer immunotherapy.
Collapse
Affiliation(s)
- Mohammad Ariful Islam
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jamie Rice
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emma Reesor
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Harshal Zope
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Lim
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jianxun Ding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yunhan Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dike Aduluso
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bruce R Zetter
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
43
|
Kaur A, Kaushik D, Piplani S, Mehta SK, Petrovsky N, Salunke DB. TLR2 Agonistic Small Molecules: Detailed Structure-Activity Relationship, Applications, and Future Prospects. J Med Chem 2020; 64:233-278. [PMID: 33346636 DOI: 10.1021/acs.jmedchem.0c01627] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLRs) are the pattern recognition receptors (PRRs) that recognize pathogen-associated molecular patterns (PAMPs) in microbial species. Among the various TLRs, TLR2 has a special place due to its ability to sense the widest repertoire of PAMPs owing to its heterodimerization with either TLR1 or TLR6, broadening its ligand diversity against pathogens. Various scaffolds are reported to activate TLR2, which include naturally occurring lipoproteins, synthetic lipopeptides, and small heterocyclic molecules. We described a detailed SAR in TLR2 agonistic scaffolds and also covered the design and chemistry for the conjugation of TLR2 agonists to antigens, carbohydrates, polymers, and fluorophores. The approaches involved in delivery of TLR2 agonists such as lipidation of antigen, conjugation to polymers, phosphonic acids, and other linkers to achieve surface adsorption, liposomal formulation, and encapsulating nanoparticles are elaborated. The crystal structure analysis and computational modeling are also included with the structural features that facilitate TLR2 activation.
Collapse
Affiliation(s)
- Arshpreet Kaur
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Deepender Kaushik
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Sakshi Piplani
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale, Australia 5046.,College of Medicine and Public Health, Flinders University, Bedford Park, Australia, 5042
| | - Surinder K Mehta
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale, Australia 5046.,College of Medicine and Public Health, Flinders University, Bedford Park, Australia, 5042
| | - Deepak B Salunke
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India.,National Interdisciplinary Centre of Vaccine, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh 160014, India
| |
Collapse
|
44
|
Lang S, Tan Z, Wu X, Huang X. Synthesis of Carboxy-Dimethylmaleic Amide Linked Polymer Conjugate Based Ultra-pH-sensitive Nanoparticles for Enhanced Antitumor Immunotherapy. ACS Macro Lett 2020; 9:1693-1699. [PMID: 33224624 DOI: 10.1021/acsmacrolett.0c00755] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) are an important tool for anticancer immunotherapy. To elicit powerful CTL activities, ultra-pH-sensitive nanoparticles (NPs) based on methoxy poly(ethylene glycol)-b-[poly(diisopropylamino)ethyl methacrylate] have been synthesized as a vaccine delivery platform. A representative CTL epitope, ovalbumin (OVA) peptide antigen, was covalently conjugated to the polymer backbone through an acid responsive carboxy-dimethylmaleic amide linker (CDM) resulting in polymer P-CDM-OVA. Interestingly, while the P-CDM-OVA released OVA peptide slowly in a pH 6.4 buffer, the addition of bovine serum albumin (BSA) mimicking proteins encountered in a cellular and/or in vivo environment significantly accelerated the release process. Successful cell surface presentation of OVA was observed when P-CDM-OVA based ultra-pH-sensitive particles were incubated with antigen presenting cells. These P-CDM-OVA NPs greatly enhanced CTL responses in vivo compared to the free peptide or the previously reported acetalated dextran particles encapsulating OVA. The P-CDM was also investigated for adjuvant conjugation, and the coadministration of P-CDM-OVA and the P-CDM-adjuvant conjugate NPs further improved CTL responses in vivo and effectively reduced tumor growth in mice. Thus, the CDM linked polymer presents a promising platform for anticancer immunotherapy.
Collapse
|
45
|
Auderset F, Belnoue E, Mastelic-Gavillet B, Lambert PH, Siegrist CA. A TLR7/8 Agonist-Including DOEPC-Based Cationic Liposome Formulation Mediates Its Adjuvanticity Through the Sustained Recruitment of Highly Activated Monocytes in a Type I IFN-Independent but NF-κB-Dependent Manner. Front Immunol 2020; 11:580974. [PMID: 33262759 PMCID: PMC7686571 DOI: 10.3389/fimmu.2020.580974] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/15/2020] [Indexed: 12/30/2022] Open
Abstract
Novel adjuvants, such as Toll-like receptors (TLRs) agonists, are needed for the development of new formulations able to circumvent limitations of current vaccines. Among TLRs, TLR7/8 agonists represent promising candidates, as they are well described to enhance antigen-specific antibody responses and skew immunity toward T helper (TH) 1 responses. We find here that the incorporation of the synthetic TLR7/8 ligand 3M-052 in a cationic DOEPC-based liposome formulation shifts immunity toward TH1 responses and elicits strong and long-lasting germinal center and follicular T helper cell responses in adult mice. This reflects the prolonged recruitment of innate cells toward the site of immunization and homing of activated antigen-loaded monocytes and monocyte-derived dendritic cells toward draining lymph nodes. We further show that this adjuvanticity is independent of type I IFN but NF-κB-dependent. Overall, our data identify TLR7/8 agonists incorporated in liposomes as promising and effective adjuvants to enhance TH1 and germinal center responses.
Collapse
Affiliation(s)
- Floriane Auderset
- World Health Organization Collaborating Center for Vaccine Immunology, Departments of Pathology-Immunology and Pediatrics, University of Geneva, Geneva, Switzerland
| | - Elodie Belnoue
- World Health Organization Collaborating Center for Vaccine Immunology, Departments of Pathology-Immunology and Pediatrics, University of Geneva, Geneva, Switzerland
| | - Beatris Mastelic-Gavillet
- World Health Organization Collaborating Center for Vaccine Immunology, Departments of Pathology-Immunology and Pediatrics, University of Geneva, Geneva, Switzerland
| | - Paul-Henri Lambert
- World Health Organization Collaborating Center for Vaccine Immunology, Departments of Pathology-Immunology and Pediatrics, University of Geneva, Geneva, Switzerland
| | - Claire-Anne Siegrist
- World Health Organization Collaborating Center for Vaccine Immunology, Departments of Pathology-Immunology and Pediatrics, University of Geneva, Geneva, Switzerland
| |
Collapse
|
46
|
GenÇ R, YakuboĞullari N, Nalbantsoy A, ÇÖven F, Bedİr E. Adjuvant potency of Astragaloside VII embedded cholesterol nanoparticles for H3N2 influenza vaccine. ACTA ACUST UNITED AC 2020; 44:304-314. [PMID: 33110368 PMCID: PMC7585159 DOI: 10.3906/biy-2003-49] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/23/2020] [Indexed: 11/12/2022]
Abstract
Adjuvants are substances that increase the immune response to a given antigen. In the development of novel vaccine adjuvants/systems, saponins are one of the most attractive molecules due to their altered immunomodulatory activities. In this study, we tried to develop PEG (polyethylene glycol)/cholesterol-based lipid nanoparticles (LNPs) to deliver the Astragaloside VII (AST-VII) and potentiate adjuvant properties of AST-VII for the influenza vaccine. In the formation of PEG/cholesterol/AST-VII-based LNPs (PEG300: Chol-AST-VII LNPs), 3 different primary solvents (acetone, ethanol, and chloroform) were evaluated, employing their effects on hydrodynamic particle size, distribution, surface chemistry, and colloidal stability. Prepared nanoparticles were simply admixtured with inactivated influenza antigen (H3N2) and applied to PMA (phorbol 12-myristate 13-acetate)-ionomycin treated human whole blood to evaluate their cytokine release profile. PEG300: Chol-AST-VII LNPs (80.2 ± 7.7 nm) were obtained using chloroform as a desolvation agent. Co-treatment of PMA-ionomycin with AST-VII and PEG300: Chol-AST-VII LNPs significantly increased the levels of IL-2 and IFN-g, compared to PMA-ionomycin alone. In the presence of H3N2, AST-VII was able to augment IL-17A, while PEG300: Chol-AST-VII LNPs stimulated the production of IFN-g. Hemolysis was only observed in PEG300: Chol-AST-VII LNPs (250 μg/mL) treatment. AST-VII and AST-VII-integrated LNPs could be used as efficacious adjuvants for an inactivated H3N2 vaccine in vitro, and cytokine response through Th1/Th17 route was reported.
Collapse
Affiliation(s)
- Rükan GenÇ
- Department of Chemical Engineering, Faculty of Engineering, Mersin Turkey
| | - Nilgün YakuboĞullari
- Department of Bioengineering, Faculty of Engineering, İzmir Institute of Technology, İzmir Turkey
| | - Ayşe Nalbantsoy
- Department of Bioengineering, Faculty of Engineering, Ege University, İzmir Turkey
| | - Fethiye ÇÖven
- Veterinary Control and Research Institute, İzmir Turkey
| | - Erdal Bedİr
- Department of Bioengineering, Faculty of Engineering, İzmir Institute of Technology, İzmir Turkey
| |
Collapse
|
47
|
Kim Y, Kang S, Shin H, Kim T, Yu B, Kim J, Yoo D, Jon S. Sequential and Timely Combination of a Cancer Nanovaccine with Immune Checkpoint Blockade Effectively Inhibits Tumor Growth and Relapse. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202006117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Yujin Kim
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Sukmo Kang
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Hocheol Shin
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Taewoo Kim
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Byeongjun Yu
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Jinjoo Kim
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Dohyun Yoo
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| | - Sangyong Jon
- Department of Biological Sciences Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- Center for Precision Bio-Nanomedicine Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
- KAIST Institute for the BioCentury Korea Advanced Institute of Science and Technology (KAIST) 291 Daehak-ro Daejeon 34141 Republic of Korea
| |
Collapse
|
48
|
Vaccines based on virus-like nano-particles for use against Middle East Respiratory Syndrome (MERS) coronavirus. Vaccine 2020; 38:5742-5746. [PMID: 32684497 PMCID: PMC7837099 DOI: 10.1016/j.vaccine.2020.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022]
Abstract
Recent advances in virus-like nanoparticles against Middle East respiratory syndrome-related coronavirus (MERS-CoV) can initiate vaccine production faster for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), while ensuring the safety, easy administration, and long-term effects. Patients with this viral pathogen suffer from excess mortality. MERS-CoV can spread through bioaerosol transmission from animal or human sources. The appearance of an outbreak in South Korea sparked off a strong urge to design strategies for developing an effective vaccine since the emergence of MERS-CoV in 2012. Well unfortunately, this is an important fact in virus risk management. The studies showed that virus-like nanoparticles (VLPs) could be effective in its goal of stopping the symptoms of MERS-CoV infection. Besides, due to the genetic similarities in the DNA sequencing of SARS-CoV-2 with MERS-CoV and the first identified severe acute respiratory syndrome (SARS-CoV) in China since 2002/2003, strategic approaches could be used to manage SARS-CoV 2. Gathering the vital piece of information obtained so far could lead to a breakthrough in the development of an effective vaccine against SARS-CoV-2, which is prioritized and focussed by the World Health Organization (WHO). This review focuses on the virus-like nanoparticle that got successful results in animal models of MERS-CoV.
Collapse
|
49
|
Wang J, Zope H, Islam MA, Rice J, Dodman S, Lipert K, Chen Y, Zetter BR, Shi J. Lipidation Approaches Potentiate Adjuvant-Pulsed Immune Surveillance: A Design Rationale for Cancer Nanovaccine. Front Bioeng Biotechnol 2020; 8:787. [PMID: 32850698 PMCID: PMC7399020 DOI: 10.3389/fbioe.2020.00787] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/22/2020] [Indexed: 12/20/2022] Open
Abstract
Adjuvant-pulsed peptide vaccines hold great promise for the prevention and treatment of different diseases including cancer. However, it has been difficult to maximize vaccine efficacy due to numerous obstacles including the unfavorable tolerability profile of adjuvants, instability of peptide antigens, limited cellular uptake, and fast diffusion from the injection site, as well as systemic adverse effects. Here we describe a robust lipidation approach for effective nanoparticle co-delivery of low-molecular weight immunomodulators (TLR7/8 agonists) and peptides (SIINFEKL) with a potent in vivo prophylactic effect. The lipidation approaches (C16-R848 and C16-SIINFEKL) increased their hydrophobicity that is intended not only to improve drug encapsulation efficiency but also to facilitate the membrane association, intracellular trafficking, and subcellular localization. The polymer-lipid hybrid nanoparticles (PLNs) are designed to sustain antigen/adjuvant levels with less systemic exposure. Our results demonstrated that a lipidated nanovaccine can induce effective immunity by enhancing the expansion and activation of antigen-specific CD8+ T cells. This adaptive immune response led to substantial tumor suppression with improved overall survival in a prophylactic setting. Our new methodology enhances the potential of nanovaccines for anti-tumor therapy.
Collapse
Affiliation(s)
- Junqing Wang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Harshal Zope
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Mohammad Ariful Islam
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Immuno-Oncology Group, Immunomic Therapeutics, Inc., Rockville, MD, United States
| | - Jamie Rice
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States.,Silicon Therapeutics, Boston, MA, United States
| | - Sage Dodman
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kevin Lipert
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Yunhan Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Bruce R Zetter
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
50
|
Kim Y, Kang S, Shin H, Kim T, Yu B, Kim J, Yoo D, Jon S. Sequential and Timely Combination of a Cancer Nanovaccine with Immune Checkpoint Blockade Effectively Inhibits Tumor Growth and Relapse. Angew Chem Int Ed Engl 2020; 59:14628-14638. [PMID: 32430981 DOI: 10.1002/anie.202006117] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Indexed: 12/12/2022]
Abstract
We describe a small lipid nanoparticle (SLNP)-based nanovaccine platform and a new combination treatment regimen. Tumor antigen-displaying, CpG adjuvant-embedded SLNPs (OVAPEP -SLNP@CpG) were prepared from biocompatible phospholipids and a cationic cholesterol derivative. The resulting nanovaccine showed highly potent antitumor efficacy in both prophylactic and therapeutic E.G7 tumor models. However, this vaccine induced T cell exhaustion by elevating PD-L1 expression, leading to tumor recurrence. Thus, the nanovaccine was combined with simultaneous anti-PD-1 antibody treatment, but the therapeutic efficacy of this regimen was comparable to that of the nanovaccine alone. Finally, mice that showed a good therapeutic response after the first cycle of immunization with the nanovaccine underwent a second cycle together with anti-PD-1 therapy, resulting in suppression of tumor relapse. This suggests that the antitumor efficacy of combinations of nanovaccines with immune checkpoint blockade therapy is dependent on treatment sequence and the timing of each modality.
Collapse
Affiliation(s)
- Yujin Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Sukmo Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Hocheol Shin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Taewoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Byeongjun Yu
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Jinjoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Dohyun Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| | - Sangyong Jon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.,KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea
| |
Collapse
|