1
|
Gupta A, Rudra A, Reed K, Langer R, Anderson DG. Advanced technologies for the development of infectious disease vaccines. Nat Rev Drug Discov 2024; 23:914-938. [PMID: 39433939 DOI: 10.1038/s41573-024-01041-z] [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] [Accepted: 08/28/2024] [Indexed: 10/23/2024]
Abstract
Vaccines play a critical role in the prevention of life-threatening infectious disease. However, the development of effective vaccines against many immune-evading pathogens such as HIV has proven challenging, and existing vaccines against some diseases such as tuberculosis and malaria have limited efficacy. The historically slow rate of vaccine development and limited pan-variant immune responses also limit existing vaccine utility against rapidly emerging and mutating pathogens such as influenza and SARS-CoV-2. Additionally, reactogenic effects can contribute to vaccine hesitancy, further undermining the ability of vaccination campaigns to generate herd immunity. These limitations are fuelling the development of novel vaccine technologies to more effectively combat infectious diseases. Towards this end, advances in vaccine delivery systems, adjuvants, antigens and other technologies are paving the way for the next generation of vaccines. This Review focuses on recent advances in synthetic vaccine systems and their associated challenges, highlighting innovation in the field of nano- and nucleic acid-based vaccines.
Collapse
Affiliation(s)
- Akash Gupta
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnab Rudra
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kaelan Reed
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
2
|
Zhang RY, Wen Y, He CB, Zhou SH, Wu YH, Wang EY, Feng RR, Ding D, Du JJ, Gao XF, Guo J. Conjugation of TLR7 and TLR7/8 agonists onto weak protein antigen via versatile oxime ligation for enhanced vaccine efficacy. Int J Biol Macromol 2024; 278:134620. [PMID: 39127274 DOI: 10.1016/j.ijbiomac.2024.134620] [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: 04/17/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Protein-based subunit vaccines are weakly immunogenic, and developing self-adjuvanting vaccines with adjuvant conjugated to antigen is a promising approach for generating optimal immune responses. Here, we report a novel adjuvant-protein conjugate vaccine based on versatile oxime ligation technique. Firstly, the adjuvant properties of a series of TLR7 and TLR7/8 small molecule agonists in self-adjuvanting vaccines were systematically compared by coupling them to proteins in consistent ratio via p-carboxybenzaldehyde (p-CBA) for the first time. All conjugate vaccines induced cytokine secretion in murine and human macrophages in vitro, and promoted specific antibody production in vivo. Notably, a conjugate containing imidazoquinoline TLR7/8 agonist (TLR7/8a1) showed the greatest enhancement in Th1/2 balanced antibody response. To minimize the interference with the protein antigenic integrity, we further developed a systematic glycoconjugation strategy to conjugate this TLR7/8a1 onto the glycan chains of SARS-CoV-2 S1 glycoprotein via oxime ligation, in which S1 containing different numbers of aldehyde groups were obtained by differential periodate oxidation. The resulting TLR7/8a1-S1 conjugate triggered a potent humoral and cellular immunity in vivo. Together these data demonstrate the promise of these TLR7 and TLR7/8 agonists as effective built-in adjuvants, and the versatile oxime ligation strategy might broaden potential applications in designing different conjugate vaccines.
Collapse
Affiliation(s)
- Ru-Yan Zhang
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China; Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, College of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Yu Wen
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Chen-Bin He
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Shi-Hao Zhou
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Ye-Hui Wu
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - En-Yang Wang
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Ran-Ran Feng
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Dong Ding
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Jing-Jing Du
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, College of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Xiao-Fei Gao
- Jiangxi Key Laboratory for Mass Spectrometry and Instrumentation, School of Chemistry and Materials Science, East China University of Technology, Nanchang 330013, China
| | - Jun Guo
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China.
| |
Collapse
|
3
|
Kumar K, Sihag B, Patil MT, Singh R, Sakala IG, Honda-Okubo Y, Singh KN, Petrovsky N, Salunke DB. Design and Synthesis of Polyphenolic Imidazo[4,5- c]quinoline Derivatives to Modulate Toll Like Receptor-7 Agonistic Activity and Adjuvanticity. ACS Pharmacol Transl Sci 2024; 7:2063-2079. [PMID: 39022355 PMCID: PMC11249636 DOI: 10.1021/acsptsci.4c00163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/16/2024] [Accepted: 05/28/2024] [Indexed: 07/20/2024]
Abstract
TLR-7/8 agonists are a well-known class of vaccine adjuvants, with a leading example now included in Covaxin, a licensed human COVID-19 vaccine. This thereby provides the opportunity to develop newer, more potent adjuvants based on structure-function studies of these classes of compounds. Imidazoquinoline-based TLR7/8 agonists are the most potent, but when used as a vaccine adjuvant side effects can arise due to diffusion from the injection site into a systemic circulation. In this work, we sought to address this issue through structural modifications in the agonists to enhance their adsorption capacity to the classic adjuvant alum. We selected a potent TLR7-selective agonist, BBIQ (EC50 = 0.85 μM), and synthesized polyphenolic derivatives to assess their TLR7 agonistic activity and adjuvant potential alone or in combination with alum. Most of the phenolic derivatives were more active than BBIQ and, except for 12b, all were TLR7 specific. Although the synthesized compounds were less active than resiquimod, the immunization data on combination with alum, specifically the IgG1, IgG2b and IgG2c responses, were superior in comparison to BBIQ as well as the reference standard resiquimod. Compound 12b was 5-fold more potent (EC50 = 0.15 μM in TLR7) than BBIQ and induced double the IgG response to SARS-CoV-2 and hepatitis antigens. Similarly, compound 12c (EC50 = 0.31 μM in TLR7) was about 3-fold more potent than BBIQ and doubled the IgG levels. Even though compound 12d exhibited low TLR7 activity (EC50 = 5.13 μM in TLR7), it demonstrated superior adjuvant results, which may be attributed to its enhanced alum adsorption capability as compared with BBIQ and resiquimod. Alum-adsorbed polyphenolic TLR7 agonists thereby represent promising combination adjuvants resulting in a balanced Th1/Th2 immune response.
Collapse
Affiliation(s)
- Kushvinder Kumar
- Department
of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Binita Sihag
- Department
of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Madhuri T. Patil
- Mehr
Chand Mahajan DAV College for Women, Sector 36A, Chandigarh 160 036, India
| | - Rahul Singh
- Department
of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Isaac G. Sakala
- Vaxine
Pty Ltd, 11 Walkley Avenue, Warradale, South Australia 5046, Australia
- College
of Medicine and Public Health, Flinders
University, Bedford Park, South Australia 5042, Australia
| | - Yoshikazu Honda-Okubo
- Vaxine
Pty Ltd, 11 Walkley Avenue, Warradale, South Australia 5046, Australia
- College
of Medicine and Public Health, Flinders
University, Bedford Park, South Australia 5042, Australia
| | - Kamal Nain Singh
- Department
of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Nikolai Petrovsky
- Vaxine
Pty Ltd, 11 Walkley Avenue, Warradale, South Australia 5046, Australia
- College
of Medicine and Public Health, Flinders
University, Bedford Park, South Australia 5042, Australia
| | - Deepak B. Salunke
- Department
of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
- National
Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials
(NICOVIA), Panjab University, Chandigarh 160 014, India
| |
Collapse
|
4
|
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
|
5
|
Mayer DP, Nelson ME, Andriyanova D, Filler RB, Ökten A, Antao OQ, Chen JS, Scumpia PO, Weaver WM, Wilen CB, Deshayes S, Weinstein JS. A novel microporous biomaterial vaccine platform for long-lasting antibody mediated immunity against viral infection. J Control Release 2024; 370:570-582. [PMID: 38734312 PMCID: PMC11665867 DOI: 10.1016/j.jconrel.2024.05.008] [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] [Received: 02/07/2024] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Current antigen delivery platforms, such as alum and nanoparticles, are not readily tunable, thus may not generate optimal adaptive immune responses. We created an antigen delivery platform by loading lyophilized Microporous Annealed Particle (MAP) with aqueous solution containing target antigens. Upon administration of antigen loaded MAP (VaxMAP), the biomaterial reconstitution forms an instant antigen-loaded porous scaffold area with a sustained release profile to maximize humoral immunity. VaxMAP induced CD4+ T follicular helper (Tfh) cells and germinal center (GC) B cell responses in the lymph nodes similar to Alum. VaxMAP loaded with SARS-CoV-2 spike protein improved the magnitude, neutralization, and duration of anti-receptor binding domain antibodies compared to Alum vaccinated mice. A single injection of Influenza specific HA1-loaded-VaxMAP enhanced neutralizing antibodies and elicited greater protection against influenza virus challenge than HA1-loaded-Alum. Thus, VaxMAP is a platform that can be used to promote adaptive immune cell responses to generate more robust neutralizing antibodies, and better protection upon pathogen challenge.
Collapse
Affiliation(s)
- Daniel P. Mayer
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, United States of America
| | - Mariah E. Nelson
- Tempo Therapeutics, 3030 Bunker Hill st., suite 104, San Diego, CA 92109, United States of America
| | - Daria Andriyanova
- Tempo Therapeutics, 3030 Bunker Hill st., suite 104, San Diego, CA 92109, United States of America
| | - Renata B. Filler
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06510, United States of America
| | - Arya Ökten
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06510, United States of America
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, United States of America
| | - Olivia Q. Antao
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, United States of America
| | - Jennifer S. Chen
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06510, United States of America
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, United States of America
| | - Philip O. Scumpia
- Department of Medicine, Division of Dermatology, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Dermatology, West Los Angeles Veteran Affairs Medical Center, Los Angeles, California, United States of America
| | - Westbrook M. Weaver
- Tempo Therapeutics, 3030 Bunker Hill st., suite 104, San Diego, CA 92109, United States of America
| | - Craig B. Wilen
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06510, United States of America
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, United States of America
| | - Stephanie Deshayes
- Tempo Therapeutics, 3030 Bunker Hill st., suite 104, San Diego, CA 92109, United States of America
| | - Jason S. Weinstein
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, United States of America
| |
Collapse
|
6
|
Shen K, Zhang J, Zhao Z, Ma H, Wang Y, Zheng W, Xu J, Li Y, Wang B, Zhang Z, Wu S, Hou L, Chen W. Microparticulated Polygonatum sibiricum polysaccharide shows potent vaccine adjuvant effect. Int J Pharm 2024; 652:123802. [PMID: 38218508 DOI: 10.1016/j.ijpharm.2024.123802] [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: 09/11/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Adjuvants are necessary for protein vaccines and have been used for nearly 100 years. However, developing safe and effective adjuvants is still urgently needed. Polysaccharides isolated from traditional Chinese medicine are considered novel vaccine adjuvant sources. This study aimed to investigate the adjuvant activity and immune-enhancing mechanisms of the microparticulated Polygonatum sibiricum polysaccharide (MP-PSP) modified by calcium carbonate. PSP demonstrated adjuvant activity, and MP-PSP further showed a higher humoral response compared to PSP. Subsequently, MP-PSP was elucidated to improving the immunity by slowing the rate of antigen release and activating dendritic cells along with interleukin-6 secretion through toll-like receptor 4 signaling, followed by T follicular helper cell and B cell interactions. Moreover, MP-PSP had a good safety profile in vaccinated mice. Thus, MP-PSP may be a promising vaccine adjuvant and warrants further investigation.
Collapse
Affiliation(s)
- Kai Shen
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China; Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China; Department of Pharmacy, Affiliated Hospital of Nantong University, 20 West Temple Road, Nantong 226001, China
| | - Jinlong Zhang
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Zhenghao Zhao
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Hao Ma
- Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China
| | - Yudong Wang
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Wanru Zheng
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Jinghan Xu
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Yao Li
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Busen Wang
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Zhe Zhang
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China.
| | - Wei Chen
- Beijing Institute of Biotechnology, 20 Dongdajie Street, Beijing 100071, China.
| |
Collapse
|
7
|
Slezak A, Chang K, Hossainy S, Mansurov A, Rowan SJ, Hubbell JA, Guler MO. Therapeutic synthetic and natural materials for immunoengineering. Chem Soc Rev 2024; 53:1789-1822. [PMID: 38170619 PMCID: PMC11557218 DOI: 10.1039/d3cs00805c] [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: 01/05/2024]
Abstract
Immunoengineering is a rapidly evolving field that has been driving innovations in manipulating immune system for new treatment tools and methods. The need for materials for immunoengineering applications has gained significant attention in recent years due to the growing demand for effective therapies that can target and regulate the immune system. Biologics and biomaterials are emerging as promising tools for controlling immune responses, and a wide variety of materials, including proteins, polymers, nanoparticles, and hydrogels, are being developed for this purpose. In this review article, we explore the different types of materials used in immunoengineering applications, their properties and design principles, and highlight the latest therapeutic materials advancements. Recent works in adjuvants, vaccines, immune tolerance, immunotherapy, and tissue models for immunoengineering studies are discussed.
Collapse
Affiliation(s)
- Anna Slezak
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Kevin Chang
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Samir Hossainy
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Aslan Mansurov
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Stuart J Rowan
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Jeffrey A Hubbell
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Mustafa O Guler
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
8
|
Mayer DP, Neslon ME, Andriyanova D, Antao OQ, Chen JS, Scumpia PO, Weaver WM, Deshayes S, Weinstein JS. A novel microporous biomaterial vaccine platform for long-lasting antibody mediated immunity against viral infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.578038. [PMID: 38352398 PMCID: PMC10862793 DOI: 10.1101/2024.01.30.578038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
Current antigen delivery platforms, such as alum and nanoparticles, are not readily tunable, thus may not generate optimal adaptive immune responses. We created an antigen delivery platform by loading lyophilized Microporous Annealed Particle (MAP) with aqueous solution containing target antigens. Upon administration of antigen loaded MAP (VaxMAP), the biomaterial reconstitution forms an instant antigen-loaded porous scaffold area with a sustained release profile to maximize humoral immunity. VaxMAP induced CD4+ T follicular helper (Tfh) cells and germinal center (GC) B cell responses in the lymph nodes similar to Alum. VaxMAP loaded with SARS-CoV-2 spike protein improved the magnitude and duration of anti-receptor binding domain antibodies compared to Alum and mRNA-vaccinated mice. A single injection of Influenza specific HA1-loaded-VaxMAP enhanced neutralizing antibodies and elicited greater protection against influenza virus challenge than HA1-loaded-Alum. Thus, VaxMAP is a platform that can be used to promote adaptive immune cell responses to generate more robust neutralizing antibodies, and better protection upon pathogen challenge.
Collapse
|
9
|
Wallace RP, Refvik KC, Antane JT, Brünggel K, Tremain AC, Raczy MR, Alpar AT, Nguyen M, Solanki A, Slezak AJ, Watkins EA, Lauterbach AL, Cao S, Wilson DS, Hubbell JA. Synthetically mannosylated antigens induce antigen-specific humoral tolerance and reduce anti-drug antibody responses to immunogenic biologics. Cell Rep Med 2024; 5:101345. [PMID: 38128533 PMCID: PMC10829756 DOI: 10.1016/j.xcrm.2023.101345] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/21/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
Immunogenic biologics trigger an anti-drug antibody (ADA) response in patients that reduces efficacy and increases adverse reactions. Our laboratory has shown that targeting protein antigen to the liver microenvironment can reduce antigen-specific T cell responses; herein, we present a strategy to increase delivery of otherwise immunogenic biologics to the liver via conjugation to a synthetic mannose polymer, p(Man). This delivery leads to reduced antigen-specific T follicular helper cell and B cell responses resulting in diminished ADA production, which is maintained throughout subsequent administrations of the native biologic. We find that p(Man)-antigen treatment impairs the ADA response against recombinant uricase, a highly immunogenic biologic, without a dependence on hapten immunodominance or control by T regulatory cells. We identify increased T cell receptor signaling and increased apoptosis and exhaustion in T cells as effects of p(Man)-antigen treatment via transcriptomic analyses. This modular platform may enhance tolerance to biologics, enabling long-term solutions for an ever-increasing healthcare problem.
Collapse
Affiliation(s)
- Rachel P Wallace
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Kirsten C Refvik
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Jennifer T Antane
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Kym Brünggel
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Andrew C Tremain
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Michal R Raczy
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Aaron T Alpar
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Mindy Nguyen
- Animal Resources Center, University of Chicago, Chicago, IL 60637, USA
| | - Ani Solanki
- Animal Resources Center, University of Chicago, Chicago, IL 60637, USA
| | - Anna J Slezak
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Elyse A Watkins
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Abigail L Lauterbach
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Shijie Cao
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - D Scott Wilson
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Biomedical Engineering Department, Johns Hopkins University, Baltimore, MD 21211, USA.
| | - Jeffrey A Hubbell
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Committee on Immunology, University of Chicago, Chicago, IL 60637, USA; Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
10
|
Miwa H, Antao OQ, Kelly‐Scumpia KM, Baghdasarian S, Mayer DP, Shang L, Sanchez GM, Archang MM, Scumpia PO, Weinstein JS, Di Carlo D. Improved Humoral Immunity and Protection against Influenza Virus Infection with a 3d Porous Biomaterial Vaccine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302248. [PMID: 37750461 PMCID: PMC10625058 DOI: 10.1002/advs.202302248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/03/2023] [Indexed: 09/27/2023]
Abstract
New vaccine platforms that activate humoral immunity and generate neutralizing antibodies are required to combat emerging pathogens, including influenza virus. A slurry of antigen-loaded hydrogel microparticles that anneal to form a porous scaffold with high surface area for antigen uptake by infiltrating immune cells as the biomaterial degrades is demonstrated to enhance humoral immunity. Antigen-loaded-microgels elicited a robust cellular humoral immune response, with increased CD4+ T follicular helper (Tfh) cells and prolonged germinal center (GC) B cells comparable to the commonly used adjuvant, aluminum hydroxide (Alum). Increasing the weight fraction of polymer material led to increased material stiffness and antigen-specific antibody titers superior to Alum. Vaccinating mice with inactivated influenza virus loaded into this more highly cross-linked formulation elicited a strong antibody response and provided protection against a high dose viral challenge. By tuning physical and chemical properties, adjuvanticity can be enhanced leading to humoral immunity and protection against a pathogen, leveraging two different types of antigenic material: individual protein antigen and inactivated virus. The flexibility of the platform may enable design of new vaccines to enhance innate and adaptive immune cell programming to generate and tune high affinity antibodies, a promising approach to generate long-lasting immunity.
Collapse
Affiliation(s)
- Hiromi Miwa
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Olivia Q. Antao
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Kindra M. Kelly‐Scumpia
- Division of CardiologyDepartment of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Sevana Baghdasarian
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Daniel P. Mayer
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Lily Shang
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Gina M. Sanchez
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Maani M. Archang
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
- MSTP ProgramDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Philip O. Scumpia
- Division of DermatologyDepartment of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
- Department of DermatologyVA Greater Los Angeles Healthcare SystemLos AngelesCA90073USA
- Jonsson Comprehensive Cancer CenterUniversity of California Los AngelesLos AngelesCA90095USA
| | - Jason S Weinstein
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Dino Di Carlo
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
- Jonsson Comprehensive Cancer CenterUniversity of California Los AngelesLos AngelesCA90095USA
- Department of Mechanical and Aerospace EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
- California Nano Systems Institute (CNSI)University of California Los AngelesLos AngelesCA90095USA
| |
Collapse
|
11
|
Naushad SM, Mandadapu G, Ramaiah MJ, Almajhdi FN, Hussain T. The role of TLR7 agonists in modulating COVID-19 severity in subjects with loss-of-function TLR7 variants. Sci Rep 2023; 13:13078. [PMID: 37567916 PMCID: PMC10421879 DOI: 10.1038/s41598-023-40114-8] [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] [Received: 02/01/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
We investigate the mechanism associated with the severity of COVID-19 in men with TLR7 mutation. Men with loss-of-function (LOF) mutations in TLR7 had severe COVID-19. LOF mutations in TLR7 increased the risk of critical COVID by 16.00-fold (95% confidence interval 2.40-106.73). The deleterious mutations affect the binding of SARS-CoV2 RNA (- 328.66 ± 26.03 vs. - 354.08 ± 27.70, p = 0.03) and MYD88 (β: 40.279, p = 0.003) to TLR7 resulting in the disruption of TLR7-MyD88-TIRAP complex. In certain hypofunctional variants and all neutral/benign variants, there is no disruption of TLR7-MyD88-TIRAP complex and four TLR7 agonists showed binding affinity comparable to that of wild protein. N-acetylcysteine (NAC) also showed a higher binding affinity for the LOF variants (p = 0.03). To conclude, TLR7 LOF mutations increase the risk of critical COVID-19 due to loss of viral RNA sensing ability and disrupted MyD88 signaling. Majority of hypofunctional and neutral variants of TLR7 are capable of carrying MyD88 signaling by binding to different TLR7 agonists and NAC.
Collapse
Affiliation(s)
- Shaik Mohammad Naushad
- Yoda LifeLine Diagnostics Pvt Ltd, 6-3-862/A, Lal Bungalow Add on, Ameerpet, Hyderabad, 500016, India.
| | | | | | - Fahad N Almajhdi
- COVID-19 Virus Research Chair, Department of Botany and Microbiology, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Tajamul Hussain
- Center of Excellence in Biotechnology Research, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| |
Collapse
|
12
|
Ren H, Jia W, Xie Y, Yu M, Chen Y. Adjuvant physiochemistry and advanced nanotechnology for vaccine development. Chem Soc Rev 2023; 52:5172-5254. [PMID: 37462107 DOI: 10.1039/d2cs00848c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Vaccines comprising innovative adjuvants are rapidly reaching advanced translational stages, such as the authorized nanotechnology adjuvants in mRNA vaccines against COVID-19 worldwide, offering new strategies to effectively combat diseases threatening human health. Adjuvants are vital ingredients in vaccines, which can augment the degree, extensiveness, and longevity of antigen specific immune response. The advances in the modulation of physicochemical properties of nanoplatforms elevate the capability of adjuvants in initiating the innate immune system and adaptive immunity, offering immense potential for developing vaccines against hard-to-target infectious diseases and cancer. In this review, we provide an essential introduction of the basic principles of prophylactic and therapeutic vaccination, key roles of adjuvants in augmenting and shaping immunity to achieve desired outcomes and effectiveness, and the physiochemical properties and action mechanisms of clinically approved adjuvants for humans. We particularly focus on the preclinical and clinical progress of highly immunogenic emerging nanotechnology adjuvants formulated in vaccines for cancer treatment or infectious disease prevention. We deliberate on how the immune system can sense and respond to the physicochemical cues (e.g., chirality, deformability, solubility, topology, and chemical structures) of nanotechnology adjuvants incorporated in the vaccines. Finally, we propose possible strategies to accelerate the clinical implementation of nanotechnology adjuvanted vaccines, such as in-depth elucidation of nano-immuno interactions, antigen identification and optimization by the deployment of high-dimensional multiomics analysis approaches, encouraging close collaborations among scientists from different scientific disciplines and aggressive exploration of novel nanotechnologies.
Collapse
Affiliation(s)
- Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Yujie Xie
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
13
|
Zhang X, Zhou Z. The Mechanism of bnAb Production and Its Application in Mutable Virus Broad-Spectrum Vaccines: Inspiration from HIV-1 Broad Neutralization Research. Vaccines (Basel) 2023; 11:1143. [PMID: 37514959 PMCID: PMC10384589 DOI: 10.3390/vaccines11071143] [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: 05/18/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Elite controllers among HIV-1-infected individuals have demonstrated a stronger ability to control the viral load in their bodies. Scientists have isolated antibodies with strong neutralizing ability from these individuals, which can neutralize HIV-1 variations; these are known as broadly neutralizing antibodies. The nucleic acid of some viruses will constantly mutate during replication (such as SARS-CoV-2), which will reduce the protective ability of the corresponding vaccines. The immune escape caused by this mutation is the most severe challenge faced by humans in the battle against the virus. Therefore, developing broad-spectrum vaccines that can induce broadly neutralizing antibodies against various viruses and their mutated strains is the best way to combat virus mutations. Exploring the mechanism by which the human immune system produces broadly neutralizing antibodies and its induction strategies is crucial in the design process of broad-spectrum vaccines.
Collapse
Affiliation(s)
- Xinyu Zhang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, No. 31 Huatuo Street, Daxing District, Beijing 102629, China
- College of Life Science, Jilin University, Changchun 130012, China
| | - Zehua Zhou
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
14
|
Wen Y, Zhang RY, Wang J, Zhou SH, Peng XQ, Ding D, Zhang ZM, Wei HW, Guo J. Novel sialoglycan linkage for constructing adjuvant-protein conjugate as potent vaccine for COVID-19. J Control Release 2023; 355:238-247. [PMID: 36716860 PMCID: PMC9907060 DOI: 10.1016/j.jconrel.2023.01.062] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 02/01/2023]
Abstract
Self-adjuvanting protein vaccines have been proved to be highly immunogenic with efficient codelivery of adjuvant and antigen. Current protein vaccines with built-in adjuvants are all modified at the peptide backbone of antigen protein, which could not achieve minor epitope interference and adjuvant multivalency at the same time. Herein, we developed a new conjugate strategy to construct effective adjuvant-protein vaccine with adjuvant cluster effect and minimal epitope interference. The toll-like receptor 7 agonist (TLR7a) is covalently conjugated on the terminal sialoglycans of SARS-CoV-2-S1 protein, leading to intracellular release of the small-molecule stimulators with greatly reduced risks of systemic toxicity. The resulting TLR7a-S1 conjugate elicited strong activation of immune cells in vitro, and potent antibody and cellular responses with a significantly enhanced Th1-bias in vivo. TLR7a-S1-induced antibody also effectively cross-neutralized all variants of concern. This sialoglycoconjugation approach to construct protein conjugate vaccines will have more applications to combat SARS-CoV-2 and other diseases.
Collapse
Affiliation(s)
- Yu Wen
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Ru-Yan Zhang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Jian Wang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Shi-Hao Zhou
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Xiao-Qian Peng
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Dong Ding
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Zhi-Ming Zhang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Hua-Wei Wei
- Jiangsu East-Mab Biomedical Technology Co. Ltd, Nantong 226499, China
| | - Jun Guo
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, College of Chemistry, Central China Normal University, Wuhan 430079, China.
| |
Collapse
|
15
|
Wei Q, Liu S, Huang X, Xin H, Ding J. Immunologically effective biomaterials-enhanced vaccines against infection of pathogenic microorganisms. BIOSAFETY AND HEALTH 2023; 5:45-61. [PMID: 40078604 PMCID: PMC11894984 DOI: 10.1016/j.bsheal.2022.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 11/27/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
Infectious diseases are severe public health events that threaten global health. Prophylactic vaccines have been considered as the most effective strategy to train the immune system to recognize and clear pathogenic infections. However, the existing vaccines against infectious diseases have several limitations, such as difficulties in mass manufacturing and storage, weak immunogenicity, and low efficiency of available adjuvants. Biomaterials, especially functional polymers, are expected to break through these bottlenecks based on the advantages of biocompatibility, degradability, controlled synthesis, easy modification, precise targeting, and immune modulation, which are excellent carriers and adjuvants of vaccines. This review mainly summarizes the application of immunologically effective polymers-enhanced vaccines against viruses- and bacteria-related infectious diseases and predicted their potential improvements.
Collapse
Affiliation(s)
- Qi Wei
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, China
| | - Shixian Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China
| | - Xu Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Hua Xin
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| |
Collapse
|
16
|
Slezak AJ, Mansurov A, Raczy MM, Chang K, Alpar AT, Lauterbach AL, Wallace RP, Weathered RK, Medellin JE, Battistella C, Gray LT, Marchell TM, Gomes S, Swartz MA, Hubbell JA. Tumor Cell-Surface Binding of Immune Stimulating Polymeric Glyco-Adjuvant via Cysteine-Reactive Pyridyl Disulfide Promotes Antitumor Immunity. ACS CENTRAL SCIENCE 2022; 8:1435-1446. [PMID: 36313164 PMCID: PMC9615125 DOI: 10.1021/acscentsci.2c00704] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Indexed: 06/10/2023]
Abstract
Immune stimulating agents like Toll-like receptor 7 (TLR7) agonists induce potent antitumor immunity but are limited in their therapeutic window due to off-target immune activation. Here, we developed a polymeric delivery platform that binds excess unpaired cysteines on tumor cell surfaces and debris to adjuvant tumor neoantigens as an in situ vaccine. The metabolic and enzymatic dysregulation in the tumor microenvironment produces these exofacial free thiols, which can undergo efficient disulfide exchange with thiol-reactive pyridyl disulfide moieties upon intratumoral injection. These functional monomers are incorporated into a copolymer with pendant mannose groups and TLR7 agonists to target both antigen and adjuvant to antigen presenting cells. When tethered in the tumor, the polymeric glyco-adjuvant induces a robust antitumor response and prolongs survival of tumor-bearing mice, including in checkpoint-resistant B16F10 melanoma. The construct additionally reduces systemic toxicity associated with clinically relevant small molecule TLR7 agonists.
Collapse
Affiliation(s)
- Anna J. Slezak
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Aslan Mansurov
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Michal M. Raczy
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Kevin Chang
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Aaron T. Alpar
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Abigail L. Lauterbach
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Rachel P. Wallace
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Rachel K. Weathered
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Jorge E.G. Medellin
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Claudia Battistella
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Laura T. Gray
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Tiffany M. Marchell
- Committee
on Immunology, University of Chicago, Chicago, Illinois 60637, United States
| | - Suzana Gomes
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Melody A. Swartz
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
- Committee
on Immunology, University of Chicago, Chicago, Illinois 60637, United States
- Ben
May Department for Cancer Research, University
of Chicago, Chicago, Illinois 60637, United
States
- Committee
on Cancer Biology, University of Chicago, Chicago, Illinois 60637, United States
| | - Jeffrey A. Hubbell
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
- Committee
on Immunology, University of Chicago, Chicago, Illinois 60637, United States
- Committee
on Cancer Biology, University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
17
|
Young A. T cells in SARS-CoV-2 infection and vaccination. Ther Adv Vaccines Immunother 2022; 10:25151355221115011. [PMID: 36051003 PMCID: PMC9425900 DOI: 10.1177/25151355221115011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 07/04/2022] [Indexed: 12/24/2022] Open
Abstract
While antibodies garner the lion’s share of attention in SARS-CoV-2 immunity, cellular immunity (T cells) may be equally, if not more important, in controlling infection. Both CD8+ and CD4+ T cells are elicited earlier and are associated with milder disease, than antibodies, and T-cell activation appears to be necessary for control of infection. Variants of concern (VOCs) such as Omicron have escaped the neutralizing antibody responses after two mRNA vaccine doses, but T-cell immunity is largely intact. The breadth and patient-specific nature of the latter offers a formidable line of defense that can limit the severity of illness, and are likely to be responsible for most of the protection from natural infection or vaccination against VOCs which have evaded the antibody response. Comprehensive searches for T-cell epitopes, T-cell activation from infection and vaccination of specific patient groups, and elicitation of cellular immunity by various alternative vaccine modalities are here reviewed. Development of vaccines that specifically target T cells is called for, to meet the needs of patient groups for whom cellular immunity is weaker, such as the elderly and the immunosuppressed. While VOCs have not yet fully escaped T-cell immunity elicited by natural infection and vaccines, some early reports of partial escape suggest that future VOCs may achieve the dreaded result, dislodging a substantial proportion of cellular immunity, enough to cause a grave public health burden. A proactive, rather than reactive, solution which identifies and targets immutable sequences in SARS-CoV-2, not just those which are conserved, may be the only recourse humankind has to disarm these future VOCs before they disarm us.
Collapse
Affiliation(s)
- Arthur Young
- InvVax, 2265 E. Foohill Blvd., Pasadena, CA 91107, USA
| |
Collapse
|
18
|
Zhang RY, Zhou SH, He CB, Wang J, Wen Y, Feng RR, Yin XG, Yang GF, Guo J. Adjuvant-Protein Conjugate Vaccine with Built-In TLR7 Agonist on S1 Induces Potent Immunity against SARS-CoV-2 and Variants of Concern. ACS Infect Dis 2022; 8:1367-1375. [PMID: 35748575 PMCID: PMC9260725 DOI: 10.1021/acsinfecdis.2c00259] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Indexed: 11/29/2022]
Abstract
With the global pandemic of the new coronavirus disease (COVID-19), a safe, effective, and affordable mass-produced vaccine remains the current focus of research. Herein, we designed an adjuvant-protein conjugate vaccine candidate, in which the TLR7 agonist (TLR7a) was conjugated to S1 subunit of SARS-CoV-2 spike protein, and systematically compared the effect of different numbers of built-in TLR7a on the immune activity for the first time. As the number of built-in TLR7a increased, a bell-shaped reaction was observed in three TLR7a-S1 conjugates, with TLR7a(10)-S1 (with around 10 built-in adjuvant molecules on one S1 protein) eliciting a more potent immune response than TLR7a(2)-S1 and TLR7a(18)-S1. This adjuvant-protein conjugate strategy allows the built-in adjuvant to provide cluster effects and prevents systemic toxicity and facilitates the co-delivery of adjuvant and antigen. Vaccination of mice with TLR7a(10)-S1 triggered a potent humoral and cellular immunity and a balanced Th1/Th2 immune response. Meanwhile, the vaccine induces effective neutralizing antibodies against SARS-CoV-2 and all variants of concern (B.1.1.7/alpha, B.1.351/beta, P.1/gamma, B.1.617.2/delta, and B.1.1.529/omicron). It is expected that the adjuvant-protein conjugate strategy has great potential to construct a potent recombinant protein vaccine candidate against various types of diseases.
Collapse
Affiliation(s)
| | | | - Chen-Bin He
- Key Laboratory of Pesticide & Chemical
Biology of Ministry of Education, International Joint Research Center
for Intelligent Biosensing Technology and Health, Hubei International
Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central
China Normal University, Wuhan 430079, China
| | - Jian Wang
- Key Laboratory of Pesticide & Chemical
Biology of Ministry of Education, International Joint Research Center
for Intelligent Biosensing Technology and Health, Hubei International
Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central
China Normal University, Wuhan 430079, China
| | - Yu Wen
- Key Laboratory of Pesticide & Chemical
Biology of Ministry of Education, International Joint Research Center
for Intelligent Biosensing Technology and Health, Hubei International
Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central
China Normal University, Wuhan 430079, China
| | - Ran-Ran Feng
- Key Laboratory of Pesticide & Chemical
Biology of Ministry of Education, International Joint Research Center
for Intelligent Biosensing Technology and Health, Hubei International
Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central
China Normal University, Wuhan 430079, China
| | - Xu-Guang Yin
- Key Laboratory of Pesticide & Chemical
Biology of Ministry of Education, International Joint Research Center
for Intelligent Biosensing Technology and Health, Hubei International
Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central
China Normal University, Wuhan 430079, China
| | - Guang-Fu Yang
- Key Laboratory of Pesticide & Chemical
Biology of Ministry of Education, International Joint Research Center
for Intelligent Biosensing Technology and Health, Hubei International
Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central
China Normal University, Wuhan 430079, China
| | - Jun Guo
- Key Laboratory of Pesticide & Chemical
Biology of Ministry of Education, International Joint Research Center
for Intelligent Biosensing Technology and Health, Hubei International
Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central
China Normal University, Wuhan 430079, China
| |
Collapse
|
19
|
Kaur A, Baldwin J, Brar D, Salunke DB, Petrovsky N. Toll-like receptor (TLR) agonists as a driving force behind next-generation vaccine adjuvants and cancer therapeutics. Curr Opin Chem Biol 2022; 70:102172. [PMID: 35785601 DOI: 10.1016/j.cbpa.2022.102172] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/06/2022] [Accepted: 05/18/2022] [Indexed: 01/06/2023]
Abstract
Until recently, the development of new human adjuvants was held back by a poor understanding of their mechanisms of action. The field was revolutionized by the discovery of the toll-like receptors (TLRs), innate immune receptors that directly or indirectly are responsible for detecting pathogen-associated molecular patterns (PAMPs) and respond to them by activating innate and adaptive immune pathways. Hundreds of ligands targeting various TLRs have since been identified and characterized as vaccine adjuvants. This work has important implications not only for the development of vaccines against infectious diseases but also for immuno-therapies against cancer, allergy, Alzheimer's disease, drug addiction and other diseases. Each TLR has its own specific tissue localization and downstream gene signalling pathways, providing researchers the opportunity to precisely tailor adjuvants with specific immune effects. TLR agonists can be combined with other TLR or alternative adjuvants to create combination adjuvants with synergistic or modulatory effects. This review provides an introduction to the various classes of TLR adjuvants and their respective signalling pathways. It provides an overview of recent advancements in the TLR field in the past 2-3 years and discusses criteria for selecting specific TLR adjuvants based on considerations, such as disease mechanisms and correlates of protection, TLR immune biasing capabilities, route of administration, antigen compatibility, new vaccine technology platforms, and age- and species-specific effects.
Collapse
Affiliation(s)
- Arshpreet Kaur
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | | | - Deshkanwar Brar
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | - Deepak B Salunke
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Bedford Park, Adelaide 5042, Australia; College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia.
| |
Collapse
|
20
|
Zhou SH, Li YT, Zhang RY, Liu YL, You ZW, Bian MM, Wen Y, Wang J, Du JJ, Guo J. Alum Adjuvant and Built-in TLR7 Agonist Synergistically Enhance Anti-MUC1 Immune Responses for Cancer Vaccine. Front Immunol 2022; 13:857779. [PMID: 35371101 PMCID: PMC8965739 DOI: 10.3389/fimmu.2022.857779] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/17/2022] [Indexed: 12/16/2022] Open
Abstract
The tumor-associated antigen mucin 1 (MUC1) is an attractive target of antitumor vaccine, but its weak immunogenicity is a big challenge for the development of vaccine. In order to enhance immune responses against MUC1, herein, we conjugated small molecular toll-like receptor 7 agonist (TLR7a) to carrier protein BSA via MUC1 glycopeptide to form a three-component conjugate (BSA-MUC1-TLR7a). Furthermore, we combined the three-component conjugate with Alum adjuvant to explore their synergistic effects. The immunological studies indicated that Alum adjuvant and built-in TLR7a synergistically enhanced anti-MUC1 antibody responses and showed Th1-biased immune responses. Meanwhile, antibodies elicited by the vaccine candidate effectively recognized tumor cells and induced complement-dependent cytotoxicity. In addition, Alum adjuvant and built-in TLR7a synergistically enhanced MUC1 glycopeptide-specific memory CD8+ T-cell immune responses. More importantly, the vaccine with the binary adjuvant can significantly inhibit tumor growth and prolong the survival time of mice in the tumor challenge experiment. This novel vaccine construct provides an effective strategy to develop antitumor vaccines.
Collapse
Affiliation(s)
- Shi-Hao Zhou
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Bio-sensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Yu-Ting Li
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Bio-sensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Ru-Yan Zhang
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Bio-sensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Yan-Ling Liu
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Bio-sensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Zi-Wei You
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Bio-sensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Miao-Miao Bian
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Bio-sensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Yu Wen
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Bio-sensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Jian Wang
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Bio-sensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Jing-Jing Du
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, School of Medicine, Hubei Polytechnic University, Huangshi, China
| | - Jun Guo
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Bio-sensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| |
Collapse
|
21
|
Cano-Vicent A, Hashimoto R, Takayama K, Serrano-Aroca Á. Biocompatible Films of Calcium Alginate Inactivate Enveloped Viruses Such as SARS-CoV-2. Polymers (Basel) 2022; 14:polym14071483. [PMID: 35406356 PMCID: PMC9002394 DOI: 10.3390/polym14071483] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023] Open
Abstract
The current pandemic is urgently demanding the development of alternative materials capable of inactivating the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes the coronavirus 2019 (COVID-19) disease. Calcium alginate is a crosslinked hydrophilic biopolymer with an immense range of biomedical applications due to its excellent chemical, physical, and biological properties. In this study, the cytotoxicity and antiviral activity of calcium alginate in the form of films were studied. The results showed that these films, prepared by solvent casting and subsequent crosslinking with calcium cations, are biocompatible in human keratinocytes and are capable of inactivating enveloped viruses such as bacteriophage phi 6 with a 1.43-log reduction (94.92% viral inactivation) and SARS-CoV-2 Delta variant with a 1.64-log reduction (96.94% viral inactivation) in virus titers. The antiviral activity of these calcium alginate films can be attributed to its compacted negative charges that may bind to viral envelopes inactivating membrane receptors.
Collapse
Affiliation(s)
- Alba Cano-Vicent
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain;
| | - Rina Hashimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan;
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan;
- Correspondence: (K.T.); (Á.S.-A.)
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain;
- Correspondence: (K.T.); (Á.S.-A.)
| |
Collapse
|
22
|
Zhang N, Li K, Liu Z, Nandakumar KS, Jiang S. A Perspective on the Roles of Adjuvants in Developing Highly Potent COVID-19 Vaccines. Viruses 2022; 14:v14020387. [PMID: 35215980 PMCID: PMC8875727 DOI: 10.3390/v14020387] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
Several countries have made unremitting efforts to develop an optimal vaccine in the fight against coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). With the increasing occurrence of SARS-CoV-2 variants, current vaccines show decreased neutralizing activities, especially towards the Omicron variant. In this context, adding appropriate adjuvants to COVID-19 vaccines can substantially reduce the number of required doses and improve efficacy or cross-neutralizing protection. We mainly focus on research progress and achievements associated with adjuvanted COVID-19 subunit and inactivated vaccines. We further compare the advantages and disadvantages of different adjuvant formulations in order to provide a scientific reference for designing an effective strategy for future vaccine development.
Collapse
Affiliation(s)
- Naru Zhang
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou 310015, China;
- Correspondence: (N.Z.); (S.J.)
| | - Kangchen Li
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou 310015, China;
| | - Zezhong Liu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China;
| | - Kutty Selva Nandakumar
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden;
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China;
- Correspondence: (N.Z.); (S.J.)
| |
Collapse
|