1
|
Jia C, Fu J, Wang Z, Li Z, Yu J, Liu Q, Zhang J, Wang X. Immunogenicity of a novel HMT13 adjuvant-based inactivated vaccine against Avian pathogenic Escherichia coli serogroups O36, O78, and O109. Poult Sci 2025; 104:105097. [PMID: 40209468 PMCID: PMC12008629 DOI: 10.1016/j.psj.2025.105097] [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: 01/06/2025] [Revised: 03/02/2025] [Accepted: 03/24/2025] [Indexed: 04/12/2025] Open
Abstract
Avian colibacillosis, mainly caused by avian pathogenic Escherichia coli (APEC), is one of the most prominent diseases in the poultry industry. Inactivated vaccines are crucial for preventing and controlling APEC infection, with adjuvants playing a key role in ensuring their effectiveness. However, traditional oil-emulsion inactivated vaccines often cause severe adverse reactions in animals. Based on the serogroups of 343 clinically isolated APEC strains from many provinces of China, O36, O78 and O109 were identified as the predominant serogroups currently circulating in these regions, with O36 and O109 being rarely noticed before. Consequently, we further screened these three dominant serogroups for their promising immunogenicity to develop effective inactivated vaccines. The immune side effects of 7 adjuvants including water-in-oil emulsions (Marc-52, ISA 71 VG, and ISA 78 VG), water-in-oil-in-water emulsions (ADJ 501 and HMT 13), oil-in-water emulsion (ADJ 2052), and aluminum hydroxide gels were evaluated and compared. The results showed that the vaccine formulated with HMT 13 as an adjuvant exhibited sufficient protective efficacy against homotypic APEC infection while effectively stimulating humoral and cellular immune responses. Moreover, inactivated vaccines containing HMT 13 could be easily absorbed at the inoculation site without interfering with chicken growth; they also possessed favorable emulsification properties facilitating storage. Therefore, HMT 13 represents an excellent adjuvant for developing an inactivated vaccine against chicken colibacillosis. This study presents a novel multivalent inactivated vaccine against chicken colibacillosis based on the use of HMT13 as an adjuvant, which offers a new approach for the prevention and control of APEC currently prevalent in most of China.
Collapse
Affiliation(s)
- Chaoying Jia
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, PR China
| | - Jiyang Fu
- Wuhan Keqian Biology Co., Ltd., Wuhan 430000, PR China
| | - Zesong Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, PR China
| | - Zugang Li
- Wuhan Keqian Biology Co., Ltd., Wuhan 430000, PR China
| | - Jiangxu Yu
- Wuhan Keqian Biology Co., Ltd., Wuhan 430000, PR China
| | - Qingyun Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, PR China; Engineering Research Center of Animal Biopharmaceuticals, The Ministry of Education of the People's Republic of China (MOE), Wuhan 430070, PR China
| | - Jinqiu Zhang
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, PR China
| | - Xiangru Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, PR China; Engineering Research Center of Animal Biopharmaceuticals, The Ministry of Education of the People's Republic of China (MOE), Wuhan 430070, PR China.
| |
Collapse
|
2
|
Gordy JT, Bates RE, Glass E, Meza J, Li Y, Schill C, Taylor AD, Wang T, Chen F, Plunkett K, Karanika S, Karakousis PC, Markham RB. MIP-3α-antigen fusion DNA vaccine enhances sex differences in tuberculosis model and alters dendritic cell activity early post vaccination. RESEARCH SQUARE 2025:rs.3.rs-5663995. [PMID: 39877094 PMCID: PMC11774437 DOI: 10.21203/rs.3.rs-5663995/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Background Tuberculosis (TB) remains a major cause of global morbidity and mortality. Efforts to control TB are hampered by the lengthy and cumbersome treatment required to eradicate the infection. Bacterial persistence during exposure to bactericidal antibiotics is at least partially mediated by the bacterial stringent response enzyme, RelMtb. A therapeutic DNA vaccine targeting RelMtb has been shown to increase the efficacy of antitubercular drugs, and fusing macrophage-inflammatory protein 3α (MIP-3α), which interacts with CCR6 on immature dendritic cells (iDCs), to RelMtb further increases the vaccine's therapeutic efficacy. A secondary analysis of these prior studies elucidated prominent sex-based differences in vaccine therapeutic efficacy, with female mice showing improved microbial outcomes compared to males as a result of the Rel and MIP-3α-Rel vaccine constructs, with a greater sex-associated difference in the MIP-3α-Rel group. In the current study, we addressed the hypothesis that these sex-related differences are due to differential DC activation/function soon after vaccination. Methods A EαGFP reporter vaccine model was used to track vaccine antigen presentation by an antibody Y-Ae which binds the Eα peptide tag in complex with I-Ab MHC-II molecules. Results MIP-3α-EαGFP groups had more DCs presenting vaccine antigen infiltrating from the periphery, with more abundant Langerhans cells in males and greater CD8 + CD103 + cross-presenting dermal DCs in females. This model also shows there is greater DC activation, as measured by CD80 and MHC II MFI, by MIP-3α compared to EαGFP alone, especially in female mice. Conclusions Our findings are consistent with the sex- and MIP-3α-related differences seen in the therapeutic model and supports the hypothesis that in both sexes MIP-3α enhances vaccine uptake and cell activation by peripheral iDCs. Additionally, Female mice showed greater levels of antigen presentation, especially in DCs able to cross-present antigen, explaining why they had the best outcomes. Further studies are required to understand underlying mechanisms and to link APC results directly to T-cell responses.
Collapse
|
3
|
Sabzehei F, Taromchi AH, Ramazani A, Nedaei K, Feizi A, Arsang-Jang S, Danafar H. Cationic micelle delivery of a multi-epitope vaccine candidate derived from tumor-associated antigens, causing regression in established CT26 colorectal tumors in mice. J Biomed Mater Res A 2024; 112:733-742. [PMID: 38088136 DOI: 10.1002/jbm.a.37654] [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: 08/24/2023] [Revised: 11/18/2023] [Accepted: 11/27/2023] [Indexed: 03/20/2024]
Abstract
Among all the cancers, colorectal cancer (CRC) has the third mortality rank in both genders. Cancer vaccines have shown promising results in boosting patients' immune systems to fight cancer. Using the IEDB database, we predicted mouse MHC-I (H2-Ld) binding epitopes from four tumor-associated antigens (APC, KRAS, TP53, and PIK3CA) and designed a multi-epitope vaccine. We expressed the candidate vaccine and encapsulated it into the cationic micelle with polyethyleneimine conjugated to oleic acid as its building blocks. We studied tumor inhibition effect, cytokine production, and lymphocyte proliferation in the mouse CRC model after vaccination. Our finding illustrated significant tumor growth inhibition in mouse models treated with the candidate nanovaccine. Besides the significant release of IFN-γ and IL-4 by immunized mouse spleen T-lymphocytes, T-cell proliferation assay results confirmed effective immune response after the vaccination. These results demonstrate the potential therapeutic effects of nanovaccines and could be a possible approach to CRC immunotherapy.
Collapse
Affiliation(s)
- Faezeh Sabzehei
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Amir Hossein Taromchi
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Ramazani
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Keivan Nedaei
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolamir Feizi
- Department of Pathology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Shahram Arsang-Jang
- Department of Biostatistics and Epidemiology, School of Medicine, Znjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Danafar
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
4
|
Abbasi S, Matsui-Masai M, Yasui F, Hayashi A, Tockary TA, Mochida Y, Akinaga S, Kohara M, Kataoka K, Uchida S. Carrier-free mRNA vaccine induces robust immunity against SARS-CoV-2 in mice and non-human primates without systemic reactogenicity. Mol Ther 2024; 32:1266-1283. [PMID: 38569556 PMCID: PMC11081875 DOI: 10.1016/j.ymthe.2024.03.022] [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: 10/29/2023] [Revised: 02/21/2024] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
Carrier-free naked mRNA vaccines may reduce the reactogenicity associated with delivery carriers; however, their effectiveness against infectious diseases has been suboptimal. To boost efficacy, we targeted the skin layer rich in antigen-presenting cells (APCs) and utilized a jet injector. The jet injection efficiently introduced naked mRNA into skin cells, including APCs in mice. Further analyses indicated that APCs, after taking up antigen mRNA in the skin, migrated to the lymph nodes (LNs) for antigen presentation. Additionally, the jet injection provoked localized lymphocyte infiltration in the skin, serving as a physical adjuvant for vaccination. Without a delivery carrier, our approach confined mRNA distribution to the injection site, preventing systemic mRNA leakage and associated systemic proinflammatory reactions. In mouse vaccination, the naked mRNA jet injection elicited robust antigen-specific antibody production over 6 months, along with germinal center formation in LNs and the induction of both CD4- and CD8-positive T cells. By targeting the SARS-CoV-2 spike protein, this approach provided protection against viral challenge. Furthermore, our approach generated neutralizing antibodies against SARS-CoV-2 in non-human primates at levels comparable to those observed in mice. In conclusion, our approach offers a safe and effective option for mRNA vaccines targeting infectious diseases.
Collapse
Affiliation(s)
- Saed Abbasi
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Miki Matsui-Masai
- Department of Research, NANO MRNA Co., Ltd., 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Fumihiko Yasui
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, 2-1-6, Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Akimasa Hayashi
- Department of Pathology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Theofilus A Tockary
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Yuki Mochida
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Shiro Akinaga
- Department of Research, NANO MRNA Co., Ltd., 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, 2-1-6, Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan.
| | - Satoshi Uchida
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan; Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
5
|
Sanchez PL, Andre G, Antipov A, Petrovsky N, Ross TM. Advax-SM™-Adjuvanted COBRA (H1/H3) Hemagglutinin Influenza Vaccines. Vaccines (Basel) 2024; 12:455. [PMID: 38793706 PMCID: PMC11125990 DOI: 10.3390/vaccines12050455] [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: 02/08/2024] [Revised: 03/25/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Adjuvants enhance immune responses stimulated by vaccines. To date, many seasonal influenza vaccines are not formulated with an adjuvant. In the present study, the adjuvant Advax-SM™ was combined with next generation, broadly reactive influenza hemagglutinin (HA) vaccines that were designed using a computationally optimized broadly reactive antigen (COBRA) methodology. Advax-SM™ is a novel adjuvant comprising inulin polysaccharide and CpG55.2, a TLR9 agonist. COBRA HA vaccines were combined with Advax-SM™ or a comparator squalene emulsion (SE) adjuvant and administered to mice intramuscularly. Mice vaccinated with Advax-SM™ adjuvanted COBRA HA vaccines had increased serum levels of anti-influenza IgG and IgA, high hemagglutination inhibition activity against a panel of H1N1 and H3N2 influenza viruses, and increased anti-influenza antibody secreting cells isolated from spleens. COBRA HA plus Advax-SM™ immunized mice were protected against both morbidity and mortality following viral challenge and, at postmortem, had no detectable lung viral titers or lung inflammation. Overall, the Advax-SM™-adjuvanted COBRA HA formulation provided effective protection against drifted H1N1 and H3N2 influenza viruses.
Collapse
Affiliation(s)
- Pedro L. Sanchez
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
| | - Greiciely Andre
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Anna Antipov
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Adelaide, SA 5046, Australia; (G.A.); (A.A.); (N.P.)
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA;
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
6
|
Luo R, Wan Y, Liu G, Chen J, Luo X, Li Z, Su D, Lu N, Luo Z. Engineering Self-Assembling Peptide Hydrogel to Enhance the Capacity of Dendritic Cells to Activate In Vivo T-Cell Immunity. Biomacromolecules 2024; 25:1408-1428. [PMID: 38236703 DOI: 10.1021/acs.biomac.3c00511] [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: 03/12/2024]
Abstract
The efficacy of the dendritic cell (DC) has failed to meet expectations thus far, and crucial problems such as the immature state of DCs, low targeting efficiency, insufficient number of dendritic cells, and microenvironment are still the current focus. To address these problems, we developed two self-assembling peptides, RLDI and RQDT, that mimic extracellular matrix (ECM). These peptides can be self-assembled into highly ordered three-dimensional nanofiber scaffold structures, where RLDI can form gelation immediately. In addition, we found that RLDI and RQDT enhance the biological function of DCs, including releasing antigens sustainably, adhering to DCs, promoting the maturation of DCs, and increasing the ability of DC antigen presentation. Moreover, peptide hydrogel-based DC treatment significantly achieved prophylactic and treatment effects on colon cancer. These results have certain implications for the design of new broad-spectrum vaccines in the future.
Collapse
Affiliation(s)
- Ruyue Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Yuan Wan
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa 52242, United States
| | - Guicen Liu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Jialei Chen
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Xinyi Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Zhaoxu Li
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697, United States
| | - Di Su
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Na Lu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
7
|
Edwards C, Shah SA, Gebhardt T, Jewell CM. Exploiting Unique Features of Microneedles to Modulate Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302410. [PMID: 37380199 PMCID: PMC10753036 DOI: 10.1002/adma.202302410] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/01/2023] [Indexed: 06/30/2023]
Abstract
Microneedle arrays (MNAs) are small patches containing hundreds of short projections that deliver signals directly to dermal layers without causing pain. These technologies are of special interest for immunotherapy and vaccine delivery because they directly target immune cells concentrated in the skin. The targeting abilities of MNAs result in efficient immune responses-often more protective or therapeutic-compared to conventional needle delivery. MNAs also offer logistical benefits, such as self-administration and transportation without refrigeration. Thus, numerous preclinical and clinical studies are exploring these technologies. Here the unique advantages of MNA, as well as critical challenges-such as manufacturing and sterility issues-the field faces to enable widespread deployment are discussed. How MNA design parameters can be exploited for controlled release of vaccines and immunotherapies, and the application to preclinical models of infection, cancer, autoimmunity, and allergies are explained. Specific strategies are also discussed to reduce off-target effects compared to conventional vaccine delivery routes, and novel chemical and manufacturing controls that enable cargo stability in MNAs across flexible intervals and temperatures. Clinical research using MNAs is then examined. Drawbacks of MNAs and the implications, and emerging opportunities to exploit MNAs for immune engineering and clinical use are concluded.
Collapse
Affiliation(s)
- Camilla Edwards
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Shrey A Shah
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Thomas Gebhardt
- Department of Microbiology & Immunology, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, VIC, 3000, Australia
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- US Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, 21201, USA
- Robert E. Fischell Institute for Biomedical Devices, College Park, MD, 20742, USA
- Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, 21201, USA
| |
Collapse
|
8
|
Sabzehei F, Taromchi AH, Danafar H, Rashidzadeh H, Ramazani A. In vitro Characterization of Polyethyleneimine-Oleic Acid Cationic Micelle as a Novel Protein Carrier. Adv Biomed Res 2023; 12:126. [PMID: 37434917 PMCID: PMC10331558 DOI: 10.4103/abr.abr_303_22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/13/2022] [Accepted: 10/30/2022] [Indexed: 07/13/2023] Open
Abstract
Background Nanotechnology has introduced valuable carriers for vaccine delivery. The success of vaccination depends on many factors, such as the intact and safe presentation of vaccine candidates to immune cells. We have conjugated branched PEI-2k and oleic acid (OL) as the building block of the cationic micelle. We aimed to introduce a novel carrier for vaccine candidates. Materials and Methods We conjugated polyethyleneimine and OL (POA) to synthesize the building blocks of cationic micelles. The critical micelle concentration (CMC), size and zeta potential of micelles, and their stability in 60 days were determined. Loading, encapsulation efficiency, and in vitro release study were assessed using bovine serum albumin (BSA) as a protein model. Furthermore, the cytotoxicity and hemocompatibility of developed nanosized micelles were evaluated to ascertain the biocompatibility of fabricated micelles. Cell uptake of cationic micelles in the macrophage cell line was also followed up. Results The conjugation of two polymer parts was confirmed by Fourier transform infrared spectroscopy and 1H nuclear magnetic resonance techniques. The CMC of the developed micelles was around 5.62 × 10-8 mg/ ml, whereas the loading and encapsulation efficiencies were 16.5% and 70%, respectively. The size and zeta potential of the cationic micelles were 96.53 ± 18.53 nm and 68.3 mV, respectively. The release of BSA from POA micelles after 8 and 72 hours was 8.5% and 82%, respectively. Finally, fluorescence microscopy showed that the prepared micelles were successfully and effectively taken up by RAW264.7 cells. Conclusion These results may provide a cutting-edge vaccine delivery solution and open up a new avenue for future vaccine research.
Collapse
Affiliation(s)
- Faezeh Sabzehei
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Amir Hossein Taromchi
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Danafar
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamid Rashidzadeh
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Ramazani
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
9
|
Amaya L, Grigoryan L, Li Z, Lee A, Wender PA, Pulendran B, Chang HY. Circular RNA vaccine induces potent T cell responses. Proc Natl Acad Sci U S A 2023; 120:e2302191120. [PMID: 37155869 PMCID: PMC10193964 DOI: 10.1073/pnas.2302191120] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/14/2023] [Indexed: 05/10/2023] Open
Abstract
Circular RNAs (circRNAs) are a class of RNAs commonly found across eukaryotes and viruses, characterized by their resistance to exonuclease-mediated degradation. Their superior stability compared to linear RNAs, combined with previous work showing that engineered circRNAs serve as efficient protein translation templates, make circRNA a promising candidate for RNA medicine. Here, we systematically examine the adjuvant activity, route of administration, and antigen-specific immunity of circRNA vaccination in mice. Potent circRNA adjuvant activity is associated with RNA uptake and activation of myeloid cells in the draining lymph nodes and transient cytokine release. Immunization of mice with engineered circRNA encoding a protein antigen delivered by a charge-altering releasable transporter induced innate activation of dendritic cells, robust antigen-specific CD8 T cell responses in lymph nodes and tissues, and strong antitumor efficacy as a therapeutic cancer vaccine. These results highlight the potential utility of circRNA vaccines for stimulating potent innate and T cell responses in tissues.
Collapse
Affiliation(s)
- Laura Amaya
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA94305
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Lilit Grigoryan
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA94305
| | - Zhijian Li
- Department of Chemistry, Stanford University, Stanford, CA94305
| | - Audrey Lee
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA94305
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, CA94305
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA94305
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA94305
| | - Howard Y. Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| |
Collapse
|
10
|
Dithranol as novel co-adjuvant for non-invasive dermal vaccination. NPJ Vaccines 2022; 7:112. [PMID: 36153349 PMCID: PMC9509335 DOI: 10.1038/s41541-022-00530-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Transcutaneous immunization (TCI) utilizing the TLR7 agonist imiquimod (IMQ-TCI) induces T cell-driven protective immunity upon application onto intact skin. In our present work, we combine the anti-psoriatic agent dithranol with IMQ-TCI to boost vaccination efficacy (Dithranol/IMQ-based transcutaneous vaccination (DIVA)). Using ovalbumin-derived peptides as model antigens in mice, DIVA induced superior cytolytic CD8+ T cells and CD4+ T cells with a TH1 cytokine profile in the priming as well as in the memory phase. Regarding the underlying mechanisms, dithranol induced an oxidant-dependent, monocyte-attracting inflammatory milieu in the skin boosting TLR7-dependent activation of dendritic cells and macrophages leading to superior T cell priming and protective immunity in vaccinia virus infection. In conclusion, we introduce the non-invasive vaccination method DIVA to induce strong primary and memory T cell responses upon a single local treatment. This work provides relevant insights in cutaneous vaccination approaches, paving the way for clinical development in humans.
Collapse
|
11
|
Dar SA, Kole S, Shin SM, Jeong HJ, Jung SJ. Comparative study on antigen persistence and immunoprotective efficacy of intramuscular and intraperitoneal injections of squalene - aluminium hydroxide (Sq + Al) adjuvanted viral hemorrhagic septicaemia virus vaccine in olive flounder (Paralichthys olivaceus). Vaccine 2021; 39:6866-6875. [PMID: 34696933 DOI: 10.1016/j.vaccine.2021.10.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/07/2021] [Accepted: 10/12/2021] [Indexed: 01/23/2023]
Abstract
The profitability of the olive flounder (Paralichthys olivaceus) aquaculture industry in Korea depends on high production and maintenance of flesh quality, as consumers prefer to eat raw flounders from aquaria and relish the raw muscles as 'sashimi'. For sustaining high production, easy-to-deliver and efficient vaccination strategies against serious pathogens, such as viral hemorrhagic septicemia virus (VHSV), is very important as it cause considerable losses to the industry. Whereas, a safe and non-invasive vaccine formulation that is free from unacceptable side-effects and does not devalue the fish is needed to maintain flesh quality. We previously developed a squalene-aluminium hydroxide (Sq + Al) adjuvanted VHSV vaccine that conferred moderate to high protection in flounder, without causing any side effects when administered through the intraperitoneal (IP) injection route. However, farmers often demand intramuscular (IM) injection vaccines as they are relatively easy to administer in small fishes. Therefore, we administered the developed vaccine via IP and IM routes and investigated the safety and persistency of the vaccine at the injection site. In addition, we conducted a comparative analysis of vaccine efficacy and serum antibody response. The clinical and histological observation of the IM and IP groups showed that our vaccine remained persistence at the injection sites for 10-17 weeks post vaccination (wpv), without causing any adverse effects to the fish. The relative percentage of survival were 100% and 71.4% for the IP group and 88.9% and 92.3% for the IM group at 3 and 17 wpv, respectively. Thus, considering the persistency period (24 wpv) and both short and long-term efficacy of our vaccine, the present study offers an option to flounder farmers in selecting either IM or IP delivery strategy according to their cultured fish size and harvesting schedule - IM vaccination for small-sized fish and IP vaccination for table-sized fish.
Collapse
Affiliation(s)
- Showkat Ahmad Dar
- Department of Aqualife Medicine, Chonnam National University, Republic of Korea
| | - Sajal Kole
- Department of Aqualife Medicine, Chonnam National University, Republic of Korea
| | - Su-Mi Shin
- Department of Aqualife Medicine, Chonnam National University, Republic of Korea
| | - Hyeon-Jong Jeong
- Department of Aqualife Medicine, Chonnam National University, Republic of Korea
| | - Sung-Ju Jung
- Department of Aqualife Medicine, Chonnam National University, Republic of Korea.
| |
Collapse
|
12
|
Gabitzsch E, Safrit JT, Verma M, Rice A, Sieling P, Zakin L, Shin A, Morimoto B, Adisetiyo H, Wong R, Bezawada A, Dinkins K, Balint J, Peykov V, Garban H, Liu P, Bacon A, Bone P, Drew J, Sanford DC, Spilman P, Sender L, Rabizadeh S, Niazi K, Soon-Shiong P. Dual-Antigen COVID-19 Vaccine Subcutaneous Prime Delivery With Oral Boosts Protects NHP Against SARS-CoV-2 Challenge. Front Immunol 2021; 12:729837. [PMID: 34603305 PMCID: PMC8481919 DOI: 10.3389/fimmu.2021.729837] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/20/2021] [Indexed: 12/25/2022] Open
Abstract
We have developed a dual-antigen COVID-19 vaccine incorporating genes for a modified SARS-CoV-2 spike protein (S-Fusion) and the viral nucleocapsid (N) protein with an Enhanced T-cell Stimulation Domain (N-ETSD) to increase the potential for MHC class II responses. The vaccine antigens are delivered by a human adenovirus serotype 5 platform, hAd5 [E1-, E2b-, E3-], previously demonstrated to be effective in the presence of Ad immunity. Vaccination of rhesus macaques with the hAd5 S-Fusion + N-ETSD vaccine by subcutaneous prime injection followed by two oral boosts elicited neutralizing anti-S IgG and T helper cell 1-biased T-cell responses to both S and N that protected the upper and lower respiratory tracts from high titer (1 x 106 TCID50) SARS-CoV-2 challenge. Notably, viral replication was inhibited within 24 hours of challenge in both lung and nasal passages, becoming undetectable within 7 days post-challenge.
Collapse
Affiliation(s)
| | | | - Mohit Verma
- ImmunityBio, Inc., Culver City, CA, United States
| | - Adrian Rice
- ImmunityBio, Inc., Culver City, CA, United States
| | | | - Lise Zakin
- ImmunityBio, Inc., Culver City, CA, United States
| | - Annie Shin
- ImmunityBio, Inc., Culver City, CA, United States
| | | | | | - Raymond Wong
- ImmunityBio, Inc., Culver City, CA, United States
| | | | - Kyle Dinkins
- ImmunityBio, Inc., Culver City, CA, United States
| | | | | | | | - Philip Liu
- ImmunityBio, Inc., Culver City, CA, United States
| | | | - Pete Bone
- IosBio, Burgess Hill, United Kingdom
| | - Jeff Drew
- IosBio, Burgess Hill, United Kingdom
| | | | | | | | | | - Kayvan Niazi
- ImmunityBio, Inc., Culver City, CA, United States
| | | |
Collapse
|
13
|
Adjuvant oncolytic virotherapy for personalized anti-cancer vaccination. Nat Commun 2021; 12:2626. [PMID: 33976179 PMCID: PMC8113265 DOI: 10.1038/s41467-021-22929-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 04/07/2021] [Indexed: 01/17/2023] Open
Abstract
By conferring systemic protection and durable benefits, cancer immunotherapies are emerging as long-term solutions for cancer treatment. One such approach that is currently undergoing clinical testing is a therapeutic anti-cancer vaccine that uses two different viruses expressing the same tumor antigen to prime and boost anti-tumor immunity. By providing the additional advantage of directly killing cancer cells, oncolytic viruses (OVs) constitute ideal platforms for such treatment strategy. However, given that the targeted tumor antigen is encoded into the viral genomes, its production requires robust infection and therefore, the vaccination efficiency partially depends on the unpredictable and highly variable intrinsic sensitivity of each tumor to OV infection. In this study, we demonstrate that anti-cancer vaccination using OVs (Adenovirus (Ad), Maraba virus (MRB), Vesicular stomatitis virus (VSV) and Vaccinia virus (VV)) co-administered with antigenic peptides is as efficient as antigen-engineered OVs and does not depend on viral replication. Our strategy is particularly attractive for personalized anti-cancer vaccines targeting patient-specific mutations. We suggest that the use of OVs as adjuvant platforms for therapeutic anti-cancer vaccination warrants testing for cancer treatment. Viruses expressing tumour antigens can prime and boost anti-tumour immunity but the efficiency of this approach depends on the capacity of the virus to infect the host. Here, the authors show that vaccination with oncolytic viruses co-administered with tumour antigenic peptides is as efficient as antigen-engineered oncolytic viruses.
Collapse
|
14
|
Palmitoylated antigens for the induction of anti-tumor CD8 + T cells and enhanced tumor recognition. MOLECULAR THERAPY-ONCOLYTICS 2021; 21:315-328. [PMID: 34141869 PMCID: PMC8170356 DOI: 10.1016/j.omto.2021.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/24/2021] [Indexed: 12/30/2022]
Abstract
Induction of tumor-specific cytotoxic CD8+ T cells (CTLs) via immunization relies on the presentation of tumor-associated peptides in major histocompatibility complex (MHC) class I molecules by dendritic cells (DCs). To achieve presentation of exogenous peptides into MHC class I, cytosolic processing and cross-presentation are required. Vaccination strategies aiming to induce tumor-specific CD8+ T cells via this exogenous route therefore pose a challenge. In this study, we describe improved CD8+ T cell induction and in vivo tumor suppression of mono-palmitic acid-modified (C16:0) antigenic peptides, which can be attributed to their unique processing route, efficient receptor-independent integration within lipid bilayers, and continuous intracellular accumulation and presentation through MHC class I. We propose that this membrane-integrating feature of palmitoylated peptides can be exploited as a tool for quick and efficient antigen enrichment and MHC class I loading. Importantly, both DCs and non-professional antigen-presenting cells (APCs), similar to tumor cells, facilitate anti-tumor immunity by efficient CTL priming via DCs and effective recognition of tumors through enhanced presentation of antigens.
Collapse
|
15
|
Shibuya M, Tamiya S, Kawai A, Yoshioka Y. Nasal-subcutaneous prime-boost regimen for inactivated whole-virus influenza vaccine efficiently protects mice against both upper and lower respiratory tract infections. Biochem Biophys Res Commun 2021; 554:166-172. [PMID: 33798943 DOI: 10.1016/j.bbrc.2021.03.099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/18/2021] [Indexed: 10/21/2022]
Abstract
Although influenza vaccines are effective for reducing viral transmission and the severity of clinical symptoms, influenza viruses still induce considerable morbidity and mortality worldwide. Seasonal influenza viruses infect the upper respiratory tract initially but then often induce severe pulmonary complications in the lower respiratory tract. Therefore, influenza vaccines that prevent viral infection at both the upper and lower respiratory tracts are highly anticipated. Here, we examined whether using different vaccination routes for priming and boosting achieved protection in both regions of the respiratory tract. To this end, we used inactivated whole-virion influenza vaccines to immunize mice either subcutaneously or intranasally for both priming and boosting. Regardless of the route used for boosting, the levels of virus-specific IgG in plasma were higher in mice primed subcutaneously than those in control mice, which received PBS only. In addition, intranasal priming followed by subcutaneous boosting induced higher levels of virus-specific IgG in plasma than those in control mice. The levels of virus-specific nasal IgA were higher in mice that were primed intranasally than in control mice or in mice primed subcutaneously. Furthermore, intranasal priming but not subcutaneous priming provided protection against viral challenge in the upper respiratory tract. In addition, when coupled with subcutaneous boosting, both subcutaneous and intranasal priming protected against viral challenge in the lower respiratory tract. These results indicate that intranasal priming followed by subcutaneous boosting induces both virus-specific IgG in plasma and IgA in nasal washes and protects against virus challenge in both the upper and lower respiratory tracts. Our results will help to develop novel vaccines against influenza viruses and other respiratory viruses.
Collapse
Affiliation(s)
- Meito Shibuya
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shigeyuki Tamiya
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Atsushi Kawai
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasuo Yoshioka
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; The Research Foundation for Microbial Diseases of Osaka University (BIKEN), 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
16
|
Schetters STT, Li RJE, Kruijssen LJW, Engels S, Ambrosini M, Garcia-Vallejo JJ, Kalay H, Unger WWJ, van Kooyk Y. Adaptable antigen matrix platforms for peptide vaccination strategies and T cell-mediated anti-tumor immunity. Biomaterials 2020; 262:120342. [PMID: 32905903 DOI: 10.1016/j.biomaterials.2020.120342] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/17/2020] [Accepted: 08/21/2020] [Indexed: 12/30/2022]
Abstract
Injection of antigenic peptides has been widely used as a vaccine strategy to boost T cell immunity. However, the poor immunogenicity of single peptides can potentially be strengthened through modification of the tertiary structure and the selection of the accompanying adjuvant. Here, we generated antigenic peptides into non-linear trimers by solid phase peptide synthesis, thereby enhancing antigen presentation by dendritic cells to CD8+ T cells in vitro and in vivo. CD8+ T cells from mice vaccinated with trimers showed an KLRG1+ effector phenotype and were able to recognize and kill antigen-expressing tumor cells ex vivo. Importantly, trimers outperformed synthetic long peptide in terms of T cell response even when equal number of epitopes were used for immunization. To improve the synthesis of trimers containing difficult peptide sequences, we developed a novel small molecule that functions as conjugation platform for synthetic long peptides. This platform , termed Antigen MAtriX (AMAX) improved yield, purity and solubility of trimers over conventional solid phase synthesis strategies. AMAX outperformed synthetic long peptides in terms of both CD8+ and CD4+ T cell responses and allowed functionalization with DC-SIGN-binding carbohydrates for in vivo dendritic cell targeting strategies, boosting T cell responses even further. Moreover, we show that agonistic CD40 antibody combined with MF59 (AddaVax) emulsion synergistically improves the antigen-specific T cell response of the AMAX in vivo. Also, tumor-associated antigens and neo-antigens could be incorporated in AMAX for tumor-specific CD8+ T cell responses. Importantly, immunization with a mix of neoantigen AMAX could reduce tumor growth in a pre-clinical syngeneic mouse model. Hence, we provide pre-clinical support for the induction of effector CD8+ T cells through the adaptable AMAX platform as easy implementable peptidic vaccination strategy against any antigen of choice, including neoantigens for anti-tumor immunity.
Collapse
Affiliation(s)
- Sjoerd T T Schetters
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands.
| | - R J Eveline Li
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands
| | - Laura J W Kruijssen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands
| | - Steef Engels
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands
| | - Martino Ambrosini
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands
| | - Juan J Garcia-Vallejo
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands
| | - Hakan Kalay
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands
| | - Wendy W J Unger
- Laboratory of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location VUmc, Amsterdam, Netherlands.
| |
Collapse
|