1
|
Lai Y, Yang X, Wei D, Wang X, Sun R, Li Y, Ji P, Bao Y, Chu T, Zhang C, Liang Q, Xu J, Zhang X, Chen Y, Wang Y. BCG-trained macrophages couple LDLR upregulation to type I IFN responses and antiviral immunity. Cell Rep 2025; 44:115493. [PMID: 40178982 DOI: 10.1016/j.celrep.2025.115493] [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: 09/28/2024] [Revised: 01/08/2025] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
Trained immunity refers to memory-like responses of innate immune cells when they re-encounter pathogenic stimuli. Bacillus Calmette-Guérin (BCG) vaccination implies enhanced antiviral immunity, whereas the underlying mechanisms remain unclear. Herein, we have uncovered elevated expression of low-density lipoprotein receptor (LDLR) on BCG-trained macrophages with robust type I interferon (IFNI) production and antiviral effects both in vivo and in vitro. Consequently, cholesterol is accumulated in BCG-trained macrophages, leading to the augmentation of NFE2L1 expression and the formation of NFE2L1/IRAK1/TRIM25 complex where TRIM25 mediates IRAK1 K63 polyubiquitination to exaggerate IFNI responses in an RIG-I-dependent manner. We have also observed LDLR+ macrophages displaying heightened IFNI responses in BCG-treated human macrophages. To antagonize LDLR degradation by PCSK9 inhibitors increases IFNI responses in the macrophages and accelerated viral clearance. Our study thus couples LDLR upregulation to antiviral activity in BCG-trained macrophages, making commercial PCSK9 inhibitors potential antiviral indications in clinic.
Collapse
Affiliation(s)
- Yangdian Lai
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxu Yang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong Wei
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruiming Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunfei Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Ji
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yujie Bao
- Department of Infectious Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiancheng Chu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenxing Zhang
- Department of Nephrology, Shanghai Children's Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiming Liang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Xu
- Department of Infectious Diseases, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinxin Zhang
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingying Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Scariot DB, Staneviciute A, Machado RRB, Yuk SA, Liu YG, Sharma S, Almunif S, Arona Mbaye EH, Nakamura CV, Engman DM, Scott EA. Efficacy of benznidazole delivery during Chagas disease nanotherapy is dependent on the nanocarrier morphology. Biomaterials 2025; 322:123358. [PMID: 40318604 DOI: 10.1016/j.biomaterials.2025.123358] [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: 10/31/2024] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/07/2025]
Abstract
The causative agent of Chagas disease, the protozoan Trypanosoma cruzi, is an obligate intracellular parasite that is typically treated with daily oral administration of Benznidazole (BNZ), a parasiticidal pro-drug with considerable side effects. Previously, we effectively targeted intracellular parasites using ∼100 nm diameter BNZ-loaded poly(ethylene glycol)-b-poly(propylene sulfide) (PEG-b-PPS) vesicular nanocarriers (a.k.a. polymersomes) in a T. cruzi-infected mouse model, without causing the typical side effects associated with standard BNZ treatment. Here, we exploit the structural versatility of the PEG-b-PPS system to investigate the impact of nanocarrier structure on the efficacy of BNZ nanotherapy. Despite sharing the same surface chemistry and oxidation-sensitive biodegradation, solid core ∼25 nm PEG-b-PPS micelles failed to produce in vivo trypanocidal effects. By applying the Förster Resonance Energy Transfer strategy, we demonstrated that PEG-b-PPS polymersomes promoted sustained intracellular drug release and enhanced tissue accumulation, offering a significant advantage for intracellular drug delivery compared to micelles with the same surface chemistry. Our studies further revealed that the lack of parasiticidal effect in PEG-b-PPS micelles is likely due to their slower rate of accumulation into solid tissues, consistent with the prolonged circulation time of intact micelles. Considering the cardiac damage typically induced by T. cruzi infection, this study also investigated the contributions of cardiac cellular biodistribution and payload release for both nanocarriers to the treatment outcomes of BNZ delivery. Our findings emphasize the crucial role of cardiac macrophages in the parasiticidal effect of BNZ formulations and highlight the critical importance of nanobiomaterial structure during therapeutic delivery.
Collapse
Affiliation(s)
- Debora B Scariot
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA; Department of Biomedical Engineering, NanoSTAR Institute, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Austeja Staneviciute
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Rayanne R B Machado
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA; Department of Biological Sciences, State University of Maringa, Parana, 87020-900, Brazil
| | - Simseok A Yuk
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Yu-Gang Liu
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Swagat Sharma
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Sultan Almunif
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA; Bioengineering Institute, King Abdulaziz City for Science and Technology, Riyadh, 12354, Saudi Arabia
| | - El Hadji Arona Mbaye
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Celso Vataru Nakamura
- Department of Biological Sciences, State University of Maringa, Parana, 87020-900, Brazil
| | - David M Engman
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA; Department of Pathology, Northwestern University, Chicago, IL, 60611, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA; Department of Biomedical Engineering, NanoSTAR Institute, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA.
| |
Collapse
|
3
|
Merriam D, Weinberg A. PBMC and fibroblast cocultures to mimic the in vivo effect of BCG on trained immunity. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001449. [PMID: 39981222 PMCID: PMC11840551 DOI: 10.17912/micropub.biology.001449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/18/2025] [Accepted: 02/03/2025] [Indexed: 02/22/2025]
Abstract
BCG greatly stimulates innate immune cells. Previous studies demonstrated that BCG-stimulated monocytes develop trained immunity whereby they respond to homologous and heterologous antigens. Previous studies used isolated monocytes or animal models to study BCG-induced trained immunity, which have benefits and limitations. To approximate in vivo conditions, we stimulated peripheral blood mononuclear cells (PBMCs) with BCG-treated human fibroblasts. We found that compared with BCG stimulation, the addition of fibroblasts increased the expression of IFN-γ in NK and γδ T cells and of TNF-α and IL-10 in monocytes. We conclude that BCG-treated fibroblasts offer advantages over BCG alone for studying trained immunity.
Collapse
Affiliation(s)
- David Merriam
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Biology, Metropolitan State University of Denver, Denver, Colorado, United States
| | - Adriana Weinberg
- Departments of Pediatrics, Medicine, and Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
4
|
Netea MG, Joosten LAB. Trained innate immunity: Concept, nomenclature, and future perspectives. J Allergy Clin Immunol 2024; 154:1079-1084. [PMID: 39278362 DOI: 10.1016/j.jaci.2024.09.005] [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: 07/08/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
During the past decade, compelling evidence has accumulated demonstrating that innate immune cells can mount adaptive characteristics, leading to long-term changes in their function. This de facto innate immune memory has been termed trained immunity. Trained immunity, which is mediated through extensive metabolic rewiring and epigenetic modifications, has important effects in human diseases. Although the upregulation of trained immunity by certain vaccines provides heterologous protection against infections, the inappropriate activation of trained immunity by endogenous stimuli contributes to the pathogenesis of inflammatory and neurodegenerative disorders. Development of vaccines that can induce both classical adaptive immunity and trained immunity may lead to a new generation of vaccines with increased efficacy. Activation of trained immunity can also lead to novel strategies for the treatment of cancer, whereas modulation of trained immunity can provide new approaches to the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands; Department for Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany.
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
5
|
Singh A, Boggiano C, Yin DE, Polakowski L, Majji SP, Leitner WW, Levy O, De Paris K. Precision adjuvants for pediatric vaccines. Sci Transl Med 2024; 16:eabq7378. [PMID: 39231242 PMCID: PMC11911902 DOI: 10.1126/scitranslmed.abq7378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024]
Abstract
Elucidating optimal vaccine adjuvants for harnessing age-specific immune pathways to enhance magnitude, breadth, and durability of immunogenicity remains a key gap area in pediatric vaccine design. A better understanding of age-specific adjuvants will inform precision discovery and development of safe and effective vaccines for protecting children from preventable infectious diseases.
Collapse
Affiliation(s)
- Anjali Singh
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - César Boggiano
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Dwight E. Yin
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Laura Polakowski
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Sai P. Majji
- Maternal and Pediatric Infectious Disease Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20817, USA
| | - Wolfgang W. Leitner
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Soni D, Borriello F, Scott DA, Feru F, DeLeon M, Brightman SE, Cheng WK, Melhem G, Smith JA, Ramirez JC, Barman S, Cameron M, Kelly A, Walker K, Nanishi E, van Haren SD, Phan T, Qi Y, Kinsey R, Raczy MM, Ozonoff A, Pettengill MA, Hubbell JA, Fox CB, Dowling DJ, Levy O. From hit to vial: Precision discovery and development of an imidazopyrimidine TLR7/8 agonist adjuvant formulation. SCIENCE ADVANCES 2024; 10:eadg3747. [PMID: 38959314 PMCID: PMC11221515 DOI: 10.1126/sciadv.adg3747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/29/2024] [Indexed: 07/05/2024]
Abstract
Vaccination can help prevent infection and can also be used to treat cancer, allergy, and potentially even drug overdose. Adjuvants enhance vaccine responses, but currently, the path to their advancement and development is incremental. We used a phenotypic small-molecule screen using THP-1 cells to identify nuclear factor-κB (NF-κB)-activating molecules followed by counterscreening lead target libraries with a quantitative tumor necrosis factor immunoassay using primary human peripheral blood mononuclear cells. Screening on primary cells identified an imidazopyrimidine, dubbed PVP-037. Moreover, while PVP-037 did not overtly activate THP-1 cells, it demonstrated broad innate immune activation, including NF-κB and cytokine induction from primary human leukocytes in vitro as well as enhancement of influenza and SARS-CoV-2 antigen-specific humoral responses in mice. Several de novo synthesis structural enhancements iteratively improved PVP-037's in vitro efficacy, potency, species-specific activity, and in vivo adjuvanticity. Overall, we identified imidazopyrimidine Toll-like receptor-7/8 adjuvants that act in synergy with oil-in-water emulsion to enhance immune responses.
Collapse
Affiliation(s)
- Dheeraj Soni
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Francesco Borriello
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - David A. Scott
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Frederic Feru
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria DeLeon
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Spencer E. Brightman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Wing Ki Cheng
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Gandolina Melhem
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Juan C. Ramirez
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Soumik Barman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Michael Cameron
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Aisling Kelly
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Kristina Walker
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Etsuro Nanishi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Simon Daniel van Haren
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tony Phan
- Access to Advanced Health Institute (AAHI), Seattle, WA, USA
| | - Yizhi Qi
- Access to Advanced Health Institute (AAHI), Seattle, WA, USA
| | - Robert Kinsey
- Access to Advanced Health Institute (AAHI), Seattle, WA, USA
| | - Michal M. Raczy
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Matthew A. Pettengill
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jeffery A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Christopher B. Fox
- Access to Advanced Health Institute (AAHI), Seattle, WA, USA
- Department of Global Health, University of Washington, 3980 15th Ave NE, Seattle, WA 98195, USA
| | - David J. Dowling
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| |
Collapse
|
7
|
Escher TE, Yuk SA, Qian Y, Stubbs CK, Scott EA, Satchell KJF. Therapeutic expression of RAS Degrader RRSP in Pancreatic Cancer via Nanocarrier-mediated mRNA delivery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598439. [PMID: 38948803 PMCID: PMC11212117 DOI: 10.1101/2024.06.11.598439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
About one-third of all human cancers encode abnormal RAS proteins locked in a constitutively activated state to drive malignant transformation and uncontrolled tumor growth. Despite progress in development of small molecules for treatment of mutant KRAS cancers, there is a need for a pan-RAS inhibitor that is effective against all RAS isoforms and variants and that avoids drug resistance. We have previously shown that the naturally occurring bacterial enzyme RAS/RAP1-specific endopeptidase (RRSP) is a potent RAS degrader that can be re-engineered as a biologic therapy to induce regression of colorectal, breast, and pancreatic tumors. Here, we have developed a strategy for in vivo expression of this RAS degrader via mRNA delivery using a synthetic nonviral gene delivery platform composed of the poly(ethylene glycol)-b-poly(propylene sulfide) (PEG-b-PPS) block copolymer conjugated to a dendritic cationic peptide (PPDP2). Using this strategy, PPDP2 is shown to deliver mRNA to both human and mouse pancreatic cells resulting in RRSP gene expression, activity, and loss of cell proliferation. Further, pancreatic tumors are reduced with residual tumors lacking detectable RAS and phosphorylated ERK. These data support that mRNA-loaded synthetic nanocarrier delivery of a RAS degrader can interrupt the RAS signaling system within pancreatic cancer cells while avoiding side effects during therapy.
Collapse
Affiliation(s)
- Taylor E Escher
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611 United States
| | - Simseok A Yuk
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Yuan Qian
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Caleb K Stubbs
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611 United States
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
- Robert H. Lurie Comprehensive Cancer Research Center, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611 United States
| | - Karla J F Satchell
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611 United States
- Robert H. Lurie Comprehensive Cancer Research Center, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611 United States
| |
Collapse
|
8
|
Li X, Gao J, Wu C, Wang C, Zhang R, He J, Xia ZJ, Joshi N, Karp JM, Kuai R. Precise modulation and use of reactive oxygen species for immunotherapy. SCIENCE ADVANCES 2024; 10:eadl0479. [PMID: 38748805 PMCID: PMC11095489 DOI: 10.1126/sciadv.adl0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/10/2024] [Indexed: 05/19/2024]
Abstract
Reactive oxygen species (ROS) play an important role in regulating the immune system by affecting pathogens, cancer cells, and immune cells. Recent advances in biomaterials have leveraged this mechanism to precisely modulate ROS levels in target tissues for improving the effectiveness of immunotherapies in infectious diseases, cancer, and autoimmune diseases. Moreover, ROS-responsive biomaterials can trigger the release of immunotherapeutics and provide tunable release kinetics, which can further boost their efficacy. This review will discuss the latest biomaterial-based approaches for both precise modulation of ROS levels and using ROS as a stimulus to control the release kinetics of immunotherapeutics. Finally, we will discuss the existing challenges and potential solutions for clinical translation of ROS-modulating and ROS-responsive approaches for immunotherapy, and provide an outlook for future research.
Collapse
Affiliation(s)
- Xinyan Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jingjing Gao
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Biomedical Engineering, Material Science and Engineering Graduate Program and The Center for Bioactive Delivery-Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Chengcheng Wu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Chaoyu Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ruoshi Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jia He
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ziting Judy Xia
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nitin Joshi
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey M. Karp
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rui Kuai
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| |
Collapse
|
9
|
Du F, Yuk SA, Qian Y, Vincent MP, Bobbala S, Abbott TM, Kim H, Li Y, Li H, Yi S, Qiao B, Scott EA. A Biomimetic Multi-Component Subunit Vaccine via Ratiometric Loading of Hierarchical Hydrogels. RESEARCH SQUARE 2024:rs.3.rs-4177821. [PMID: 38746232 PMCID: PMC11092859 DOI: 10.21203/rs.3.rs-4177821/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The development of subunit vaccines that mimic the molecular complexity of attenuated vaccines has been limited by the difficulty of intracellular co-delivery of multiple chemically diverse payloads at controllable concentrations. We report on hierarchical hydrogel depots employing simple poly(propylene sulfone) homopolymers to enable ratiometric loading of a protein antigen and four physicochemically distinct adjuvants in a hierarchical manner. The optimized vaccine consisted of immunostimulants either adsorbed to or encapsulated within nanogels, which were capable of noncovalent anchoring to subcutaneous tissues. These 5-component nanogel vaccines demonstrated enhanced humoral and cell-mediated immune responses compared to formulations with standard single adjuvant and antigen pairing. The use of a single simple homopolymer capable of rapid and stable loading and intracellular delivery of diverse molecular cargoes holds promise for facile development and optimization of scalable subunit vaccines and complex therapeutic formulations for a wide range of biomedical applications.
Collapse
Affiliation(s)
- Fanfan Du
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Simseok A. Yuk
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Yuan Qian
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Michael P. Vincent
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sharan Bobbala
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Tirzah M. Abbott
- Northwestern University Atomic and Nanoscale Characterization Experimental Center, Evanston, IL 60208, USA
| | - Hyeohn Kim
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Yang Li
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60201, USA
| | - Haoyu Li
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sijia Yi
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Baofu Qiao
- Department of Natural Sciences, Baruch Colleg-e, City University of New York, New York, 10010, USA
| | - Evan A. Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL 60208, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
10
|
Hua C, Qiu L. Polymersomes for Therapeutic Protein and Peptide Delivery: Towards Better Loading Properties. Int J Nanomedicine 2024; 19:2317-2340. [PMID: 38476284 PMCID: PMC10929215 DOI: 10.2147/ijn.s444910] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/24/2024] [Indexed: 03/14/2024] Open
Abstract
Therapeutics based on proteins and peptides have profoundly transformed the landscape of treatment for diseases, from diabetes mellitus to cancers, yet the short half-life and low bioavailability of therapeutic proteins and peptides hinder their wide applications. To break through this bottleneck, biomolecules-loaded polymersomes with strong adjustability and versatility have attracted more and more attentions recently. Loading proteins or peptides into polymersomes is the first but extremely important step towards developing high-quality formulation products. However, increasing protein and peptide loading content is quite challenging due to the inherent nature of self-assembled vesicle formation mechanism and physiochemical characteristics of biomacromolecules. This review highlights the potential of polymersomes as the next-generation therapeutic proteins and peptides carrier and emphatically introduces novel approaches and recent progress to achieve satisfactory encapsulation capability of polymersomes for proteins and peptides. On the one hand, with the help of intermolecular interactions, such as electrostatic, lipid-protein, and hydrophobic interactions, the drug loading could be significantly improved. On the other hand, loading improvement could be attained through innovation of preparation methods, ranging from modified traditional film hydration techniques to the novel phase-guided assembly method.
Collapse
Affiliation(s)
- Chengxu Hua
- Ministry of Educational (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, People’s Republic of China
| | - Liyan Qiu
- Ministry of Educational (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, People’s Republic of China
| |
Collapse
|
11
|
Kaur A, Vaccari M. Exploring HIV Vaccine Progress in the Pre-Clinical and Clinical Setting: From History to Future Prospects. Viruses 2024; 16:368. [PMID: 38543734 PMCID: PMC10974975 DOI: 10.3390/v16030368] [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: 01/09/2024] [Revised: 02/08/2024] [Accepted: 02/21/2024] [Indexed: 04/01/2024] Open
Abstract
The human immunodeficiency virus (HIV) continues to pose a significant global health challenge, with millions of people affected and new cases emerging each year. While various treatment and prevention methods exist, including antiretroviral therapy and non-vaccine approaches, developing an effective vaccine remains the most crucial and cost-effective solution to combating the HIV epidemic. Despite significant advancements in HIV research, the HIV vaccine field has faced numerous challenges, and only one clinical trial has demonstrated a modest level of efficacy. This review delves into the history of HIV vaccines and the current efforts in HIV prevention, emphasizing pre-clinical vaccine development using the non-human primate model (NHP) of HIV infection. NHP models offer valuable insights into potential preventive strategies for combating HIV, and they play a vital role in informing and guiding the development of novel vaccine candidates before they can proceed to human clinical trials.
Collapse
Affiliation(s)
- Amitinder Kaur
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA;
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Monica Vaccari
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA;
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
12
|
Klug N, Burke J, Scott E. Rational Engineering of Islet Tolerance via Biomaterial-Mediated Immune Modulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:216-224. [PMID: 38166244 PMCID: PMC10766078 DOI: 10.4049/jimmunol.2300527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/17/2023] [Indexed: 01/04/2024]
Abstract
Type 1 diabetes (T1D) onset is characterized by an autoimmune attack on β islet cells within the pancreas, preventing the insulin secretion required to maintain glucose homeostasis. Targeted modulation of key immunoregulatory cell populations is a promising strategy to restore tolerance to β cells. This strategy can be used to prevent T1D onset or reverse T1D with transplanted islets. To this end, drug delivery systems can be employed to transport immunomodulatory cargo to specific cell populations that inhibit autoreactive T cell-mediated destruction of the β cell mass. The rational engineering of biomaterials into nanoscale and microscale drug carriers can facilitate targeted interactions with immune cells. The physicochemical properties of the biomaterial, the delivered immunomodulatory agent, and the target cell populations are critical variables in the design of these delivery systems. In this review, we discuss recent biomaterials-based drug delivery approaches to induce islet tolerance and the need to consider both immune and metabolic markers of disease progression.
Collapse
Affiliation(s)
- Natalie Klug
- Department of Biomedical Engineering, Robert R. McCormick School of Engineering and Applied Science, Northwestern University, Evanston, IL
| | - Jacqueline Burke
- Department of Biomedical Engineering, Robert R. McCormick School of Engineering and Applied Science, Northwestern University, Evanston, IL
| | - Evan Scott
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
13
|
Morrocchi E, van Haren S, Palma P, Levy O. Modeling human immune responses to vaccination in vitro. Trends Immunol 2024; 45:32-47. [PMID: 38135599 PMCID: PMC11688643 DOI: 10.1016/j.it.2023.11.002] [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: 10/31/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
The human immune system is a complex network of coordinated components that are crucial for health and disease. Animal models, commonly used to study immunomodulatory agents, are limited by species-specific differences, low throughput, and ethical concerns. In contrast, in vitro modeling of human immune responses can enable species- and population-specific mechanistic studies and translational development within the same study participant. Translational accuracy of in vitro models is enhanced by accounting for genetic, epigenetic, and demographic features such as age, sex, and comorbidity. This review explores various human in vitro immune models, considers evidence that they may resemble human in vivo responses, and assesses their potential to accelerate and de-risk vaccine discovery and development.
Collapse
Affiliation(s)
- Elena Morrocchi
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, Rome, Italy; Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | - Simon van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Paolo Palma
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, Rome, Italy; Chair of Pediatrics, Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy.
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
14
|
Pashirova T, Shaihutdinova Z, Tatarinov D, Mansurova M, Kazakova R, Bogdanov A, Chabrière E, Jacquet P, Daudé D, Akhunzianov AA, Miftakhova RR, Masson P. Tuning the Envelope Structure of Enzyme Nanoreactors for In Vivo Detoxification of Organophosphates. Int J Mol Sci 2023; 24:15756. [PMID: 37958742 PMCID: PMC10649860 DOI: 10.3390/ijms242115756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Encapsulated phosphotriesterase nanoreactors show their efficacy in the prophylaxis and post-exposure treatment of poisoning by paraoxon. A new enzyme nanoreactor (E-nRs) containing an evolved multiple mutant (L72C/Y97F/Y99F/W263V/I280T) of Saccharolobus solfataricus phosphotriesterase (PTE) for in vivo detoxification of organophosphorous compounds (OP) was made. A comparison of nanoreactors made of three- and di-block copolymers was carried out. Two types of morphology nanoreactors made of di-block copolymers were prepared and characterized as spherical micelles and polymersomes with sizes of 40 nm and 100 nm, respectively. The polymer concentrations were varied from 0.1 to 0.5% (w/w) and enzyme concentrations were varied from 2.5 to 12.5 μM. In vivo experiments using E-nRs of diameter 106 nm, polydispersity 0.17, zeta-potential -8.3 mV, and loading capacity 15% showed that the detoxification efficacy against paraoxon was improved: the LD50 shift was 23.7xLD50 for prophylaxis and 8xLD50 for post-exposure treatment without behavioral alteration or functional physiological changes up to one month after injection. The pharmacokinetic profiles of i.v.-injected E-nRs made of three- and di-block copolymers were similar to the profiles of the injected free enzyme, suggesting partial enzyme encapsulation. Indeed, ELISA and Western blot analyses showed that animals developed an immune response against the enzyme. However, animals that received several injections did not develop iatrogenic symptoms.
Collapse
Affiliation(s)
- Tatiana Pashirova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, 420088 Kazan, Russia; (Z.S.); (D.T.); (A.B.)
| | - Zukhra Shaihutdinova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, 420088 Kazan, Russia; (Z.S.); (D.T.); (A.B.)
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (M.M.); (R.K.); (A.A.A.); (R.R.M.)
| | - Dmitry Tatarinov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, 420088 Kazan, Russia; (Z.S.); (D.T.); (A.B.)
| | - Milana Mansurova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (M.M.); (R.K.); (A.A.A.); (R.R.M.)
| | - Renata Kazakova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (M.M.); (R.K.); (A.A.A.); (R.R.M.)
| | - Andrei Bogdanov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Arbuzov Str., 8, 420088 Kazan, Russia; (Z.S.); (D.T.); (A.B.)
| | - Eric Chabrière
- Gene&GreenTK, 19–21 Boulevard Jean Moulin, 13005 Marseille, France; (E.C.); (P.J.); (D.D.)
- IRD, APHM, MEPHI, IHU-Méditerranée Infection, Aix Marseille Université, 19–21 Boulevard Jean Moulin, 13005 Marseille, France
| | - Pauline Jacquet
- Gene&GreenTK, 19–21 Boulevard Jean Moulin, 13005 Marseille, France; (E.C.); (P.J.); (D.D.)
| | - David Daudé
- Gene&GreenTK, 19–21 Boulevard Jean Moulin, 13005 Marseille, France; (E.C.); (P.J.); (D.D.)
| | - Almaz A. Akhunzianov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (M.M.); (R.K.); (A.A.A.); (R.R.M.)
| | - Regina R. Miftakhova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (M.M.); (R.K.); (A.A.A.); (R.R.M.)
| | - Patrick Masson
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (M.M.); (R.K.); (A.A.A.); (R.R.M.)
| |
Collapse
|
15
|
Malloy AMW, Lu Z, Kehl M, Pena DaMata J, Lau-Kilby AW, Turfkruyer M. Increased innate immune activation induces protective RSV-specific lung-resident memory T cells in neonatal mice. Mucosal Immunol 2023; 16:593-605. [PMID: 37392972 DOI: 10.1016/j.mucimm.2023.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/24/2023] [Indexed: 07/03/2023]
Abstract
Young infants frequently experience respiratory tract infections, yet vaccines designed to provide mucosal protection are lacking. Localizing pathogen-specific cellular and humoral immune responses to the lung could provide improved immune protection. We used a well-characterized murine model of respiratory syncytial virus (RSV) to study the development of lung-resident memory T cells (TRM) in neonatal compared to adult mice. We demonstrated that priming with RSV during the neonatal period failed to retain RSV-specific clusters of differentiation (CD8) TRM 6 weeks post infection, in contrast to priming during adulthood. The reduced development of RSV-specific TRM was associated with poor acquisition of two key markers of tissue residence: CD69 and CD103. However, by augmenting both innate immune activation and antigen exposure, neonatal RSV-specific CD8 T cells increased expression of tissue-residence markers and were maintained in the lung at memory time points. Establishment of TRM correlated with more rapid control of the virus in the lungs upon reinfection. This is the first strategy to effectively establish RSV-specific TRM in neonates providing new insight into neonatal memory T cell development and vaccine strategies.
Collapse
Affiliation(s)
- Allison M W Malloy
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, USA.
| | - Zhongyan Lu
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, USA
| | - Margaret Kehl
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, USA
| | - Jarina Pena DaMata
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, USA
| | - Annie W Lau-Kilby
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, USA
| | - Mathilde Turfkruyer
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, USA
| |
Collapse
|
16
|
Crofts KF, Page CL, Swedik SM, Holbrook BC, Meyers AK, Zhu X, Parsonage D, Westcott MM, Alexander-Miller MA. An Analysis of Linker-Dependent Effects on the APC Activation and In Vivo Immunogenicity of an R848-Conjugated Influenza Vaccine. Vaccines (Basel) 2023; 11:1261. [PMID: 37515076 PMCID: PMC10383912 DOI: 10.3390/vaccines11071261] [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: 06/06/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Subunit or inactivated vaccines comprise the majority of vaccines used against viral and bacterial pathogens. However, compared to their live/attenuated counterparts, these vaccines often demonstrate reduced immunogenicity, requiring multiple boosters and or adjuvants to elicit protective immune responses. For this reason, studies of adjuvants and the mechanism through which they can improve inactivated vaccine responses are critical for the development of vaccines with increased efficacy. Studies have shown that the direct conjugation of adjuvant to antigen promotes vaccine immunogenicity, with the advantage of both the adjuvant and antigen targeting the same cell. Using this strategy of direct linkage, we developed an inactivated influenza A (IAV) vaccine that is directly conjugated with the Toll-like receptor 7/8 agonist resiquimod (R848) through a heterobifunctional crosslinker. Previously, we showed that this vaccine resulted in improved protection and viral clearance in newborn nonhuman primates compared to a non-adjuvanted vaccine. We subsequently discovered that the choice of linker used to conjugate R848 to the virus alters the stimulatory activity of the vaccine, promoting increased maturation and proinflammatory cytokine production from DC differentiated in vitro. With this knowledge, we explored how the choice of crosslinker impacts the stimulatory activity of these vaccines. We found that the linker choice alters signaling through the NF-κB pathway in human monocyte-derived dendritic cells (moDCs). Further, we extended our analyses to in vivo differentiated APC present in human peripheral blood, replicating the linker-dependent differences found in in vitro differentiated cells. Finally, we demonstrated in a mouse model that the choice of linker impacts the amount of IAV-specific IgG antibody produced in response to vaccination. These data enhance our understanding of conjugation approaches for improving vaccine immunogenicity.
Collapse
Affiliation(s)
- Kali F. Crofts
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (K.F.C.); (C.L.P.); (S.M.S.); (B.C.H.); (A.K.M.); (X.Z.); (M.M.W.)
| | - Courtney L. Page
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (K.F.C.); (C.L.P.); (S.M.S.); (B.C.H.); (A.K.M.); (X.Z.); (M.M.W.)
| | - Stephanie M. Swedik
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (K.F.C.); (C.L.P.); (S.M.S.); (B.C.H.); (A.K.M.); (X.Z.); (M.M.W.)
| | - Beth C. Holbrook
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (K.F.C.); (C.L.P.); (S.M.S.); (B.C.H.); (A.K.M.); (X.Z.); (M.M.W.)
| | - Allison K. Meyers
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (K.F.C.); (C.L.P.); (S.M.S.); (B.C.H.); (A.K.M.); (X.Z.); (M.M.W.)
| | - Xuewei Zhu
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (K.F.C.); (C.L.P.); (S.M.S.); (B.C.H.); (A.K.M.); (X.Z.); (M.M.W.)
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Derek Parsonage
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA;
| | - Marlena M. Westcott
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (K.F.C.); (C.L.P.); (S.M.S.); (B.C.H.); (A.K.M.); (X.Z.); (M.M.W.)
| | - Martha A. Alexander-Miller
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (K.F.C.); (C.L.P.); (S.M.S.); (B.C.H.); (A.K.M.); (X.Z.); (M.M.W.)
| |
Collapse
|
17
|
Sharifi E, Yousefiasl S, Trovato M, Sartorius R, Esmaeili Y, Goodarzi H, Ghomi M, Bigham A, Moghaddam FD, Heidarifard M, Pourmotabed S, Nazarzadeh Zare E, Paiva-Santos AC, Rabiee N, Wang X, Tay FR. Nanostructures for prevention, diagnosis, and treatment of viral respiratory infections: from influenza virus to SARS-CoV-2 variants. J Nanobiotechnology 2023; 21:199. [PMID: 37344894 PMCID: PMC10283343 DOI: 10.1186/s12951-023-01938-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/24/2023] [Indexed: 06/23/2023] Open
Abstract
Viruses are a major cause of mortality and socio-economic downfall despite the plethora of biopharmaceuticals designed for their eradication. Conventional antiviral therapies are often ineffective. Live-attenuated vaccines can pose a safety risk due to the possibility of pathogen reversion, whereas inactivated viral vaccines and subunit vaccines do not generate robust and sustained immune responses. Recent studies have demonstrated the potential of strategies that combine nanotechnology concepts with the diagnosis, prevention, and treatment of viral infectious diseases. The present review provides a comprehensive introduction to the different strains of viruses involved in respiratory diseases and presents an overview of recent advances in the diagnosis and treatment of viral infections based on nanotechnology concepts and applications. Discussions in diagnostic/therapeutic nanotechnology-based approaches will be focused on H1N1 influenza, respiratory syncytial virus, human parainfluenza virus type 3 infections, as well as COVID-19 infections caused by the SARS-CoV-2 virus Delta variant and new emerging Omicron variant.
Collapse
Affiliation(s)
- Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran.
| | - Satar Yousefiasl
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maria Trovato
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), 80131, Naples, Italy
| | - Rossella Sartorius
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), 80131, Naples, Italy
| | - Yasaman Esmaeili
- School of Advanced Technologies in Medicine, Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran
| | - Hamid Goodarzi
- Centre de recherche, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
- Départment d'Ophtalmologie, Université de Montréal, Montreal, QC, Canada
| | - Matineh Ghomi
- School of Chemistry, Damghan University, Damghan, 36716-45667, Iran
| | - Ashkan Bigham
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | - Farnaz Dabbagh Moghaddam
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, 00133, Rome, Italy
| | - Maryam Heidarifard
- Centre de recherche, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
- Départment d'Ophtalmologie, Université de Montréal, Montreal, QC, Canada
| | - Samiramis Pourmotabed
- Department of Emergency Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | | | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Xiangdong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
18
|
Lee B, Nanishi E, Levy O, Dowling DJ. Precision Vaccinology Approaches for the Development of Adjuvanted Vaccines Targeted to Distinct Vulnerable Populations. Pharmaceutics 2023; 15:1766. [PMID: 37376214 PMCID: PMC10305121 DOI: 10.3390/pharmaceutics15061766] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Infection persists as one of the leading global causes of morbidity and mortality, with particular burden at the extremes of age and in populations who are immunocompromised or suffer chronic co-morbid diseases. By focusing discovery and innovation efforts to better understand the phenotypic and mechanistic differences in the immune systems of diverse vulnerable populations, emerging research in precision vaccine discovery and development has explored how to optimize immunizations across the lifespan. Here, we focus on two key elements of precision vaccinology, as applied to epidemic/pandemic response and preparedness, including (a) selecting robust combinations of adjuvants and antigens, and (b) coupling these platforms with appropriate formulation systems. In this context, several considerations exist, including the intended goals of immunization (e.g., achieving immunogenicity versus lessening transmission), reducing the likelihood of adverse reactogenicity, and optimizing the route of administration. Each of these considerations is accompanied by several key challenges. On-going innovation in precision vaccinology will expand and target the arsenal of vaccine components for protection of vulnerable populations.
Collapse
Affiliation(s)
- Branden Lee
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
| | - Etsuro Nanishi
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David J. Dowling
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
19
|
Jin SM, Yoo YJ, Shin HS, Kim S, Lee SN, Lee CH, Kim H, Kim JE, Bae YS, Hong J, Noh YW, Lim YT. A nanoadjuvant that dynamically coordinates innate immune stimuli activation enhances cancer immunotherapy and reduces immune cell exhaustion. NATURE NANOTECHNOLOGY 2023; 18:390-402. [PMID: 36635335 DOI: 10.1038/s41565-022-01296-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/15/2022] [Indexed: 06/17/2023]
Abstract
Although conventional innate immune stimuli contribute to immune activation, they induce exhausted immune cells, resulting in suboptimal cancer immunotherapy. Here we suggest a kinetically activating nanoadjuvant (K-nanoadjuvant) that can dynamically integrate two waves of innate immune stimuli, resulting in effective antitumour immunity without immune cell exhaustion. The combinatorial code of K-nanoadjuvant is optimized in terms of the order, duration and time window between spatiotemporally activating Toll-like receptor 7/8 agonist and other Toll-like receptor agonists. K-nanoadjuvant induces effector/non-exhausted dendritic cells that programme the magnitude and persistence of interleukin-12 secretion, generate effector/non-exhausted CD8+ T cells, and activate natural killer cells. Treatment with K-nanoadjuvant as a monotherapy or in combination therapy with anti-PD-L1 or liposomes (doxorubicin) results in strong antitumour immunity in murine models, with minimal systemic toxicity, providing a strategy for synchronous and dynamic tailoring of innate immunity for enhanced cancer immunotherapy.
Collapse
Affiliation(s)
- Seung Mo Jin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yeon Jeong Yoo
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hong Sik Shin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sohyun Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sang Nam Lee
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Chang Hoon Lee
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hyunji Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jung-Eun Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Department of Biological Science, Sungkyunkwan University, Suwon, Republic of Korea
| | - JungHyub Hong
- Department of Biological Sciences, Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Department of Biological Science, Sungkyunkwan University, Suwon, Republic of Korea
| | - Young-Woock Noh
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
20
|
Zhao L, Shu M, Shi K, Tang S, Li Z. Novel use of graphene oxide quantum dots in a pickering emulsion as a Chlamydia trachomatis vaccine adjuvant. Int Immunopharmacol 2023; 118:110035. [PMID: 36958212 DOI: 10.1016/j.intimp.2023.110035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/01/2023] [Accepted: 03/10/2023] [Indexed: 03/25/2023]
Abstract
Graphene oxide quantum dots (GOQDs), which are graphene-based nanoparticles, are potential surfactant substitutes for stabilizing Pickering emulsions, due to their high surface area, biodegradability, and reasonable biocompatibility. In the present study, GOQDs stabilized Pickering emulsion (GQPE) was prepared by simple sonication and then used as an adjuvant to enhance immune responses to the Chlamydia trachomatis Pgp3 recombinant vaccine. Immunization of mice showed that GQPE robustly activates adaptive immunity by efficiently stimulating IgG, sIgA, IFN-γ, IL-4, and TNF-α production. Controlled release repository of antigens both in vivo and in vitro prolonged the immune response. In addition, GQPE enhanced dendritic cell recruitment at the injection site, ensuring rapid and efficient innate immunity. Safety assessment revealed that GQPE does not cause liver, kidney, and myocardial damage in mice, suggesting its favorable biocompatibility. This study provides evidence for the use of GOPE as a facile, effective, and safe strategy to enhance the immune response to Pgp3 recombinant vaccines.
Collapse
Affiliation(s)
- Lanhua Zhao
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, the School of Nuring, University of South China, Hengyang, 421001 Hunan, PR China
| | - Mingyi Shu
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, the School of Nuring, University of South China, Hengyang, 421001 Hunan, PR China
| | - Keliang Shi
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, the School of Nuring, University of South China, Hengyang, 421001 Hunan, PR China
| | - Shuangyang Tang
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, the School of Nuring, University of South China, Hengyang, 421001 Hunan, PR China
| | - Zhongyu Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, the School of Nuring, University of South China, Hengyang, 421001 Hunan, PR China.
| |
Collapse
|
21
|
Ma J, Wang S, Zhao C, Yan X, Ren Q, Dong Z, Qiu J, Liu Y, Shan Q, Xu M, Yan B, Liu S. Computer-Aided Discovery of Potent Broad-Spectrum Vaccine Adjuvants. Angew Chem Int Ed Engl 2023; 62:e202301059. [PMID: 36815280 DOI: 10.1002/anie.202301059] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 02/24/2023]
Abstract
Adjuvants stimulate the immune system to vigorously respond to a vaccine. While current adjuvants such as aluminum salts and oil-in-water emulsions have been used for decades, they do not generate broad and long-lasting responses in many vaccines. Consequently, more potent adjuvants are needed. Here, using computer-aided molecule design and machine learning, we discovered 2 new, broad-spectrum adjuvants that can boost vaccine responses. Our library containing 46 toll-like receptor (TLR)-targeting agonist ligands were assembled on Au nanoparticles. Comprehensive in vitro, ex vivo and in vivo studies showed both leads promoted dendritic cell activation via multiple TLRs and enhanced antigen presentation to T cells. When used together with tumor-specific antigens to immunize mice against B16-OVA melanoma and 4T1-PD1 breast cancer, both adjuvants unleashed strong immune responses that suppressed tumor growth and lung metastases. Our results show computer-aided design and screening can rapidly uncover potent adjuvants for tackling waning immunity in current vaccines.
Collapse
Affiliation(s)
- Juan Ma
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- School of Environmental Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Shenqing Wang
- Institute of Environmental Research at Greater Bays, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, P. R. China
| | - Chuanfang Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiliang Yan
- Institute of Environmental Research at Greater Bays, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, P. R. China
| | - Quanzhong Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- Department of toxicology and sanitary chemistry, School of public health, Capital Medical University, Beijing, 100069, P. R. China
| | - Zheng Dong
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- School of Environmental Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jiahuang Qiu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- School of Environmental Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yin Liu
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 330106, P. R. China
| | - Qing'e Shan
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P. R. China
| | - Ming Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- School of Environmental Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Bing Yan
- Institute of Environmental Research at Greater Bays, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, P. R. China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, P. R. China
- School of Environmental Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P. R. China
| |
Collapse
|
22
|
Gouveia MG, Wesseler JP, Ramaekers J, Weder C, Scholten PBV, Bruns N. Polymersome-based protein drug delivery - quo vadis? Chem Soc Rev 2023; 52:728-778. [PMID: 36537575 PMCID: PMC9890519 DOI: 10.1039/d2cs00106c] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Indexed: 12/24/2022]
Abstract
Protein-based therapeutics are an attractive alternative to established therapeutic approaches and represent one of the fastest growing families of drugs. While many of these proteins can be delivered using established formulations, the intrinsic sensitivity of proteins to denaturation sometimes calls for a protective carrier to allow administration. Historically, lipid-based self-assembled structures, notably liposomes, have performed this function. After the discovery of polymersome-based targeted drug-delivery systems, which offer manifold advantages over lipid-based structures, the scientific community expected that such systems would take the therapeutic world by storm. However, no polymersome formulations have been commercialised. In this review article, we discuss key obstacles for the sluggish translation of polymersome-based protein nanocarriers into approved pharmaceuticals, which include limitations imparted by the use of non-degradable polymers, the intricacies of polymersome production methods, and the complexity of the in vivo journey of polymersomes across various biological barriers. Considering this complex subject from a polymer chemist's point of view, we highlight key areas that are worthy to explore in order to advance polymersomes to a level at which clinical trials become worthwhile and translation into pharmaceutical and nanomedical applications is realistic.
Collapse
Affiliation(s)
- Micael G Gouveia
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Justus P Wesseler
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Jobbe Ramaekers
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Christoph Weder
- Adolphe Merkle Institute, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Philip B V Scholten
- Adolphe Merkle Institute, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Nico Bruns
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
- Department of Chemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany.
| |
Collapse
|
23
|
Yang H, Hu Y, Kong D, Chen P, Yang L. Intralesional Bacillus Calmette-Guérin injections and hypo-fractionated radiation synergistically induce systemic antitumor immune responses. Int Immunopharmacol 2023; 114:109542. [PMID: 36521291 DOI: 10.1016/j.intimp.2022.109542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022]
Abstract
Radiotherapy, an important treatment for multiple malignancies, produces systemic anti-tumor effects in combination with immunotherapies, especially immune checkpoint inhibitors (ICBs). However, for some patients who do not respond to ICB treatment or show ICB-induced autoimmune symptoms, new alternatives need to be explored. Innovative immunomodulatory strategies, including the administration of immunostimulants, could be used to improve the immunogenicity induced by radiotherapy. In this study, we explored the synergistic effect of Bacillus Calmette-Guérin (BCG) combined with hypo-fractionated radiotherapy (H-RT) in inducing anti-tumor immune responses. We observed the systemic and abscopal effects of this combination in mice with 4 T1 breast cancer. H-RT combined with BCG could remodel the immune microenvironment and alleviate leukocyte-like responses by increasing the infiltration of CD8 + T cells, promoting the maturation of dendritic cells (DCs), decreasing the infiltration of immunosuppressive cells, and downregulating the expression of immunosuppressive cytokines. Therefore, this combination could enhance the systemic anti-tumor response, leading to the regression of untreated synchronous tumors and a decrease in the systemic metastatic burden. These results highlight the potential of BCG in assisting antitumor therapy and the therapeutic potential of this combination treatment.
Collapse
Affiliation(s)
- Hanshan Yang
- Medical Center of Hematology, the Second Affiliated Hospital, Army Medical University, Chongqing 400000, China; Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yuru Hu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Deyi Kong
- Department of Encephalopathy, Jiang 'an Hospital of Traditional Chinese Medicine, Yibin 644000, China
| | - Ping Chen
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Linglin Yang
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
24
|
Dowling DJ, Barman S, Smith AJ, Borriello F, Chaney D, Brightman SE, Melhem G, Brook B, Menon M, Soni D, Schüller S, Siram K, Nanishi E, Bazin HG, Burkhart DJ, Levy O, Evans JT. Development of a TLR7/8 agonist adjuvant formulation to overcome early life hyporesponsiveness to DTaP vaccination. Sci Rep 2022; 12:16860. [PMID: 36258023 PMCID: PMC9579132 DOI: 10.1038/s41598-022-20346-w] [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: 06/06/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022] Open
Abstract
Infection is the most common cause of mortality early in life, yet the broad potential of immunization is not fully realized in this vulnerable population. Most vaccines are administered during infancy and childhood, but in some cases the full benefit of vaccination is not realized in-part. New adjuvants are cardinal to further optimize current immunization approaches for early life. However, only a few classes of adjuvants are presently incorporated in vaccines approved for human use. Recent advances in the discovery and delivery of Toll-like receptor (TLR) agonist adjuvants have provided a new toolbox for vaccinologists. Prominent among these candidate adjuvants are synthetic small molecule TLR7/8 agonists. The development of an effective infant Bordetella pertussis vaccine is urgently required because of the resurgence of pertussis in many countries, contemporaneous to the switch from whole cell to acellular vaccines. In this context, TLR7/8 adjuvant based vaccine formulation strategies may be a promising tool to enhance and accelerate early life immunity by acellular B. pertussis vaccines. In the present study, we optimized (a) the formulation delivery system, (b) structure, and (c) immunologic activity of novel small molecule imidazoquinoline TLR7/8 adjuvants towards human infant leukocytes, including dendritic cells. Upon immunization of neonatal mice, this TLR7/8 adjuvant overcame neonatal hyporesponsiveness to acellular pertussis vaccination by driving a T helper (Th)1/Th17 biased T cell- and IgG2c-skewed humoral response to a licensed acellular vaccine (DTaP). This potent immunization strategy may represent a new paradigm for effective immunization against pertussis and other pathogens in early life.
Collapse
Affiliation(s)
- David J Dowling
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Soumik Barman
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Alyson J Smith
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59802, USA
- Seagen, Bothell, WA, USA
| | - Francesco Borriello
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, 80131, Italy
- WAO Center of Excellence, Naples, 80131, Italy
- Generate Biomedicines, Cambridge, MA, USA
| | - Danielle Chaney
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Spencer E Brightman
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
| | - Gandolina Melhem
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
| | - Byron Brook
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Manisha Menon
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
| | - Dheeraj Soni
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Simone Schüller
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Karthik Siram
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
| | - Etsuro Nanishi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Hélène G Bazin
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59802, USA
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
| | - David J Burkhart
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59802, USA
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Harvard Institutes of Medicine, Room 842, 4 Blackfan Circle, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT & Harvard, Cambridge, MA, USA.
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59802, USA.
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA.
| |
Collapse
|
25
|
Barman S, Borriello F, Brook B, Pietrasanta C, De Leon M, Sweitzer C, Menon M, van Haren SD, Soni D, Saito Y, Nanishi E, Yi S, Bobbala S, Levy O, Scott EA, Dowling DJ. Shaping Neonatal Immunization by Tuning the Delivery of Synergistic Adjuvants via Nanocarriers. ACS Chem Biol 2022; 17:2559-2571. [PMID: 36028220 PMCID: PMC9486804 DOI: 10.1021/acschembio.2c00497] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/15/2022] [Indexed: 01/19/2023]
Abstract
Adjuvanted nanocarrier-based vaccines hold substantial potential for applications in novel early-life immunization strategies. Here, via mouse and human age-specific in vitro modeling, we identified the combination of a small-molecule STING agonist (2'3'-cyclic GMP-AMP, cGAMP) and a TLR7/8 agonist (CL075) to drive the synergistic activation of neonatal dendritic cells and precision CD4 T-helper (Th) cell expansion via the IL-12/IFNγ axis. We further demonstrate that the vaccination of neonatal mice with quadrivalent influenza recombinant hemagglutinin (rHA) and an admixture of two polymersome (PS) nanocarriers separately encapsulating cGAMP (cGAMP-PS) and CL075 (CL075-PS) drove robust Th1 bias, high frequency of T follicular helper (TFH) cells, and germinal center (GC) B cells along with the IgG2c-skewed humoral response in vivo. Dual-loaded cGAMP/CL075-PSs did not outperform admixed cGAMP-PS and CL075-PS in vivo. These data validate an optimally designed adjuvantation system via age-selected small-molecule synergy and a multicomponent nanocarrier formulation as an effective approach to induce type 1 immune responses in early life.
Collapse
Affiliation(s)
- Soumik Barman
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Harvard
Medical School, Boston, Massachusetts 02115, United States
| | - Francesco Borriello
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Harvard
Medical School, Boston, Massachusetts 02115, United States
- Department
of Translational Medical Sciences and Center for Basic and Clinical
Immunology Research (CISI), University of
Naples Federico II, Naples 80131, Italy
- WAO
Center of Excellence, Naples 80131, Italy
| | - Byron Brook
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Harvard
Medical School, Boston, Massachusetts 02115, United States
| | - Carlo Pietrasanta
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Harvard
Medical School, Boston, Massachusetts 02115, United States
- Fondazione
IRCCS Ca’ Granda Ospedale Maggiore Policlinico, NICU, Milan 20122, Italy
- Department
of Clinical Sciences and Community Health, University of Milan, Milan 20122, Italy
| | - Maria De Leon
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
| | - Cali Sweitzer
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
| | - Manisha Menon
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
| | - Simon D. van Haren
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Harvard
Medical School, Boston, Massachusetts 02115, United States
| | - Dheeraj Soni
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Harvard
Medical School, Boston, Massachusetts 02115, United States
| | - Yoshine Saito
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
| | - Etsuro Nanishi
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Harvard
Medical School, Boston, Massachusetts 02115, United States
| | - Sijia Yi
- Department
of Biomedical Engineering, Northwestern
University, Evanston, Chicago, Illinois 60208, United States
| | - Sharan Bobbala
- Department
of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Ofer Levy
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Harvard
Medical School, Boston, Massachusetts 02115, United States
- Broad
Institute of MIT & Harvard, Cambridge, Massachusetts 02142, United States
| | - Evan A. Scott
- Department
of Biomedical Engineering, Northwestern
University, Evanston, Chicago, Illinois 60208, United States
| | - David J. Dowling
- Precision
Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Harvard
Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
26
|
Ko CN, Zang S, Zhou Y, Zhong Z, Yang C. Nanocarriers for effective delivery: modulation of innate immunity for the management of infections and the associated complications. J Nanobiotechnology 2022; 20:380. [PMID: 35986268 PMCID: PMC9388998 DOI: 10.1186/s12951-022-01582-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/01/2022] [Indexed: 12/24/2022] Open
Abstract
Innate immunity is the first line of defense against invading pathogens. Innate immune cells can recognize invading pathogens through recognizing pathogen-associated molecular patterns (PAMPs) via pattern recognition receptors (PRRs). The recognition of PAMPs by PRRs triggers immune defense mechanisms and the secretion of pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6. However, sustained and overwhelming activation of immune system may disrupt immune homeostasis and contribute to inflammatory disorders. Immunomodulators targeting PRRs may be beneficial to treat infectious diseases and their associated complications. However, therapeutic performances of immunomodulators can be negatively affected by (1) high immune-mediated toxicity, (2) poor solubility and (3) bioactivity loss after long circulation. Recently, nanocarriers have emerged as a very promising tool to overcome these obstacles owning to their unique properties such as sustained circulation, desired bio-distribution, and preferred pharmacokinetic and pharmacodynamic profiles. In this review, we aim to provide an up-to-date overview on the strategies and applications of nanocarrier-assisted innate immune modulation for the management of infections and their associated complications. We first summarize examples of important innate immune modulators. The types of nanomaterials available for drug delivery, as well as their applications for the delivery of immunomodulatory drugs and vaccine adjuvants are also discussed.
Collapse
|
27
|
Dowling DJ, Levy O. A Precision Adjuvant Approach to Enhance Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Vaccines Optimized for Immunologically Distinct Vulnerable Populations. Clin Infect Dis 2022; 75:S30-S36. [PMID: 35512145 PMCID: PMC9129145 DOI: 10.1093/cid/ciac342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Indexed: 01/19/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has caused significant mortality, especially among older adults whose distinct immune system reflects immunosenescence. Multiple SARS-CoV-2 vaccines have received emergency use authorization and/or licensure from the US Food and Drug Administration and throughout the world. However, their deployment has heighted significant limitations, such by age-dependent immunogenicity, requirements for multiple vaccine doses, refrigeration infrastructure that is not universally available, as well as waning immunity. Thus, there was, and continues to be a need for continued innovation during the pandemic given the desire for dose-sparing, formulations stable at more readily achievable temperatures, need for robust immunogenicity in vulnerable populations, and development of safe and effective pediatric vaccines. In this context, optimal SARS-CoV-2 vaccines may ultimately rely on inclusion of adjuvants as they can potentially enhance protection of vulnerable populations and provide dose-sparing effects enabling single shot protection.
Collapse
Affiliation(s)
- David J Dowling
- Precision Vaccines Program
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ofer Levy
- Precision Vaccines Program
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
28
|
Nanishi E, Angelidou A, Rotman C, Dowling DJ, Levy O, Ozonoff A. Precision Vaccine Adjuvants for Older Adults: A Scoping Review. Clin Infect Dis 2022; 75:S72-S80. [PMID: 35439286 PMCID: PMC9376277 DOI: 10.1093/cid/ciac302] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Older adults, defined as those ≥60 years of age, are a growing population vulnerable to infections including severe acute respiratory syndrome coronavirus 2. Although immunization is a key to protecting this population, immunosenescence can impair responses to vaccines. Adjuvants can increase the immunogenicity of vaccine antigens but have not been systematically compared in older adults. We conducted a scoping review to assess the comparative effectiveness of adjuvants in aged populations. Adjuvants AS01, MF59, AS03, and CpG-oligodeoxynucleotide, included in licensed vaccines, are effective in older human adults. A growing menu of investigational adjuvants, such as Matrix-M and CpG plus alum, showed promising results in early phase clinical trials and preclinical studies. Most studies assessed only 1 or 2 adjuvants and no study has directly compared >3 adjuvants among older adults. Enhanced preclinical approaches enabling direct comparison of multiple adjuvants including human in vitro modeling and age-specific animal models may derisk and accelerate vaccine development for older adults.
Collapse
Affiliation(s)
- Etsuro Nanishi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital,Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Asimenia Angelidou
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital,Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Chloe Rotman
- Medical Library, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - David J Dowling
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital,Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital,Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA
| | - Al Ozonoff
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital,Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
29
|
Weiss AM, Hossainy S, Rowan SJ, Hubbell JA, Esser-Kahn AP. Immunostimulatory Polymers as Adjuvants, Immunotherapies, and Delivery Systems. Macromolecules 2022; 55:6913-6937. [PMID: 36034324 PMCID: PMC9404695 DOI: 10.1021/acs.macromol.2c00854] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/16/2022] [Indexed: 12/14/2022]
Abstract
![]()
Activating innate immunity in a controlled manner is
necessary
for the development of next-generation therapeutics. Adjuvants, or
molecules that modulate the immune response, are critical components
of vaccines and immunotherapies. While small molecules and biologics
dominate the adjuvant market, emerging evidence supports the use of
immunostimulatory polymers in therapeutics. Such polymers can stabilize
and deliver cargo while stimulating the immune system by functioning
as pattern recognition receptor (PRR) agonists. At the same time,
in designing polymers that engage the immune system, it is important
to consider any unintended initiation of an immune response that results
in adverse immune-related events. Here, we highlight biologically
derived and synthetic polymer scaffolds, as well as polymer–adjuvant
systems and stimuli-responsive polymers loaded with adjuvants, that
can invoke an immune response. We present synthetic considerations
for the design of such immunostimulatory polymers, outline methods
to target their delivery, and discuss their application in therapeutics.
Finally, we conclude with our opinions on the design of next-generation
immunostimulatory polymers, new applications of immunostimulatory
polymers, and the development of improved preclinical immunocompatibility
tests for new polymers.
Collapse
Affiliation(s)
- Adam M. Weiss
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
- Department of Chemistry, University of Chicago 5735 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Samir Hossainy
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| | - Stuart J. Rowan
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
- Department of Chemistry, University of Chicago 5735 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| |
Collapse
|
30
|
Yi S, Kim SY, Vincent MP, Yuk SA, Bobbala S, Du F, Scott EA. Dendritic peptide-conjugated polymeric nanovectors for non-toxic delivery of plasmid DNA and enhanced non-viral transfection of immune cells. iScience 2022; 25:104555. [PMID: 35769884 PMCID: PMC9234717 DOI: 10.1016/j.isci.2022.104555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/29/2022] [Accepted: 06/02/2022] [Indexed: 10/26/2022] Open
Abstract
Plasmid DNA (pDNA) transfection is advantageous for gene therapies requiring larger genetic elements, including "all-in-one" CRISPR/Cas9 plasmids, but is limited by toxicity as well as poor intracellular release and transfection efficiency in immune cell populations. Here, we developed a synthetic non-viral gene delivery platform composed of poly(ethylene glycol)-b-poly(propylene sulfide) copolymers linked to a cationic dendritic peptide (DP) via a reduceable bond, PEG-b-PPS-ss-DP (PPDP). A library of self-assembling PPDP polymers was synthesized and screened to identify optimal constructs capable of transfecting macrophages with small (pCMV-DsRed, 4.6 kb) and large (pL-CRISPR.EFS.tRFP, 11.7 kb) plasmids. The optimized PPDP construct transfected macrophages, fibroblasts, dendritic cells, and T cells more efficiently and with less toxicity than a commercial Lipo2K reagent, regardless of pDNA size and under standard culture conditions in the presence of serum. The PPDP technology described herein is a stimuli-responsive polymeric nanovector that can be leveraged to meet diverse challenges in gene delivery.
Collapse
Affiliation(s)
- Sijia Yi
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sun-Young Kim
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Michael P. Vincent
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Simseok A. Yuk
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26505, USA
| | - Fanfan Du
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Evan Alexander Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
- Department of Microbiology-Immunology, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
31
|
Barman S, Soni D, Brook B, Nanishi E, Dowling DJ. Precision Vaccine Development: Cues From Natural Immunity. Front Immunol 2022; 12:662218. [PMID: 35222350 PMCID: PMC8866702 DOI: 10.3389/fimmu.2021.662218] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 12/21/2021] [Indexed: 12/31/2022] Open
Abstract
Traditional vaccine development against infectious diseases has been guided by the overarching aim to generate efficacious vaccines normally indicated by an antibody and/or cellular response that correlates with protection. However, this approach has been shown to be only a partially effective measure, since vaccine- and pathogen-specific immunity may not perfectly overlap. Thus, some vaccine development strategies, normally focused on targeted generation of both antigen specific antibody and T cell responses, resulting in a long-lived heterogenous and stable pool of memory lymphocytes, may benefit from better mimicking the immune response of a natural infection. However, challenges to achieving this goal remain unattended, due to gaps in our understanding of human immunity and full elucidation of infectious pathogenesis. In this review, we describe recent advances in the development of effective vaccines, focusing on how understanding the differences in the immunizing and non-immunizing immune responses to natural infections and corresponding shifts in immune ontogeny are crucial to inform the next generation of infectious disease vaccines.
Collapse
Affiliation(s)
- Soumik Barman
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Dheeraj Soni
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Byron Brook
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Etsuro Nanishi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - David J Dowling
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
32
|
Czysch C, Medina-Montano C, Dal NJK, Dinh T, Fröder Y, Winterwerber P, Maxeiner K, Räder HJ, Schuppan D, Schild H, Bros M, Biersack B, Feranoli F, Grabbe S, Nuhn L. End Group Dye-labeled Polycarbonate Block Copolymers for Micellar (immuno-)Drug Delivery. Macromol Rapid Commun 2022; 43:e2200095. [PMID: 35339115 DOI: 10.1002/marc.202200095] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Indexed: 11/08/2022]
Abstract
Defined conjugation of functional molecules to block copolymer end groups is a powerful strategy to enhance the scope of micellar carriers for drug delivery. In this study, we have established an approach to access well-defined polycarbonate-based block copolymers by labeling their end groups with single fluorescent dye molecules. Following controlled polymerization conditions, the block copolymers' primary hydroxy end group can be converted into activated pentafluorophenyl ester carbonates and subsequently aminolyzed with fluorescent dyes that are equipped with primary amines. During a solvent evaporation process, the resulting end group dye-labeled block copolymers self-assemble into narrowly dispersed 26 nm sized micelles and simultaneously encapsulate hydrophobic (immuno-)drugs. The covalently attached fluorescent tracer can be used to monitor both uptake into cells and stability under biologically relevant conditions, including incubation with blood plasma or during blood circulation in zebrafish embryos. By encapsulation of the TLR7/8 agonist CL075, immune stimulatory polymeric micelles are generated that get internalized by various antigen presenting dendritic cells and promote their maturation. Generally, such end group dye-labeled polycarbonate block copolymers display ideal features to permit targeted delivery of hydrophobic drugs to key immune cells for vaccination and cancer immunotherapy. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
| | | | | | - Thi Dinh
- Max Planck Institute for Polymer Research Mainz, Germany
| | - Yannick Fröder
- Max Planck Institute for Polymer Research Mainz, Germany
| | | | | | | | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center Mainz, Germany.,Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School Boston, United States
| | - Hansjörg Schild
- Institute of Immunology, University Medical Center Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Germany
| | | | | | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, Germany
| | - Lutz Nuhn
- Max Planck Institute for Polymer Research Mainz, Germany
| |
Collapse
|
33
|
Vincent MP, Navidzadeh JO, Bobbala S, Scott EA. Leveraging self-assembled nanobiomaterials for improved cancer immunotherapy. Cancer Cell 2022; 40:255-276. [PMID: 35148814 PMCID: PMC8930620 DOI: 10.1016/j.ccell.2022.01.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/22/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022]
Abstract
Nanomaterials and targeted drug delivery vehicles improve the therapeutic index of drugs and permit greater control over their pharmacokinetics, biodistribution, and bioavailability. Here, nanotechnologies applied to cancer immunotherapy are discussed with a focus on current and next generation self-assembling drug delivery systems composed of lipids and/or polymers. Topics covered include the fundamental design, suitability, and inherent properties of nanomaterials that induce anti-tumor immune responses and support anti-cancer vaccination. Established active and passive targeting strategies as well as newer "indirect" methods are presented together with insights into how nanocarrier structure and surface chemistry can be leveraged for controlled delivery to the tumor microenvironment while minimizing off-target effects.
Collapse
Affiliation(s)
- Michael P Vincent
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Justin O Navidzadeh
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA; Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
34
|
Burke JA, Zhang X, Bobbala S, Frey MA, Bohorquez Fuentes C, Freire Haddad H, Allen SD, Richardson RAK, Ameer GA, Scott EA. Subcutaneous nanotherapy repurposes the immunosuppressive mechanism of rapamycin to enhance allogeneic islet graft viability. NATURE NANOTECHNOLOGY 2022; 17:319-330. [PMID: 35039683 PMCID: PMC8934301 DOI: 10.1038/s41565-021-01048-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/09/2021] [Indexed: 05/03/2023]
Abstract
Standard oral rapamycin (that is, Rapamune) administration is plagued by poor bioavailability and broad biodistribution. Thus, this pleotropic mammalian target of rapamycin (mTOR) inhibitor has a narrow therapeutic window and numerous side effects and provides inadequate protection to transplanted cells and tissues. Furthermore, the hydrophobicity of rapamycin limits its use in parenteral formulations. Here, we demonstrate that subcutaneous delivery via poly(ethylene glycol)-b-poly(propylene sulfide) polymersome nanocarriers significantly alters rapamycin's cellular biodistribution to repurpose its mechanism of action for tolerance, instead of immunosuppression, and minimize side effects. While oral rapamycin inhibits T cell proliferation directly, subcutaneously administered rapamycin-loaded polymersomes modulate antigen presenting cells in lieu of T cells, significantly improving maintenance of normoglycemia in a clinically relevant, major histocompatibility complex-mismatched, allogeneic, intraportal (liver) islet transplantation model. These results demonstrate the ability of a rationally designed nanocarrier to re-engineer the immunosuppressive mechanism of a drug by controlling cellular biodistribution.
Collapse
Affiliation(s)
- Jacqueline A Burke
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Xiaomin Zhang
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Molly A Frey
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Carolina Bohorquez Fuentes
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Helena Freire Haddad
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Sean D Allen
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Reese A K Richardson
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA
| | - Guillermo A Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA.
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA.
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
35
|
Brennan K, Craven S, Cheung M, Kane D, Noone E, O'Callaghan J, Molloy EJ, Walsh PT, McAuliffe FM, Doyle SL. Cytosolic dsRNA improves neonatal innate immune responses to adjuvants in use in pediatric vaccines. J Leukoc Biol 2022; 112:523-537. [PMID: 35098572 PMCID: PMC9542317 DOI: 10.1002/jlb.5a0521-242r] [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: 05/07/2021] [Revised: 11/17/2021] [Accepted: 01/06/2022] [Indexed: 11/12/2022] Open
Abstract
Pattern recognition receptors (PRRs) of the innate immune system represent the critical front‐line defense against pathogens, and new vaccine formulations target these PRR pathways to boost vaccine responses, through activation of cellular/Th1 immunity. The majority of pediatric vaccines contain aluminum (ALUM) or monophosphoryl lipid A (MPLA) as adjuvants to encourage immune activation. Evidence suggests that elements of the innate immune system, currently being targeted for vaccine adjuvanticity do not fully develop until puberty and it is likely that effective adjuvants for the neonatal and pediatric populations are being overlooked due to modeling of responses in adult systems. We recently reported that the activity of the cytosolic nucleic acid (CNA) sensing family of PRRs is strong in cord blood and peripheral blood of young children. This study investigates the function of CNA sensors in subsets of neonatal innate immune cells and shows that myeloid cells from cord blood can be activated to express T cell costimulatory markers, and also to produce Th1 promoting cytokines. CD80 and CD86 were consistently up‐regulated in response to cytosolic Poly(I:C) stimulation in all cell types examined and CNA activation also induced robust Type I IFN and low levels of TNFα in monocytes, monocyte‐derived macrophages, and monocyte‐derived dendritic cells. We have compared CNA activation to adjuvants currently in use (MPLA or ALUM), either alone or in combination and found that cytosolic Poly(I:C) in combination with MPLA or ALUM can improve expression of activation marker levels above those observed with either adjuvant alone. This may prove particularly promising in the context of improving the efficacy of existing ALUM‐ or MPLA‐containing vaccines, through activation of T cell‐mediated immunity.
Collapse
Affiliation(s)
- Kiva Brennan
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Crumlin Dublin Ireland
- Department of Clinical Medicine, School of Medicine Trinity College Dublin Dublin Ireland
| | - Simon Craven
- UCD Perinatal Research Centre, Obstetrics & Gynaecology School of Medicine, University College Dublin, National Maternity Hospital Dublin Ireland
| | - Maria Cheung
- UCD Perinatal Research Centre, Obstetrics & Gynaecology School of Medicine, University College Dublin, National Maternity Hospital Dublin Ireland
| | - Daniel Kane
- UCD Perinatal Research Centre, Obstetrics & Gynaecology School of Medicine, University College Dublin, National Maternity Hospital Dublin Ireland
| | - Eleanor Noone
- Department of Clinical Medicine, School of Medicine Trinity College Dublin Dublin Ireland
| | - Joseph O'Callaghan
- Department of Clinical Medicine, School of Medicine Trinity College Dublin Dublin Ireland
| | - Eleanor J Molloy
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Crumlin Dublin Ireland
- Department of Paediatrics School of Medicine, Trinity College Dublin Dublin Ireland
- Coombe Women and Infants University Hospital Dublin Ireland
| | - Patrick T Walsh
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Crumlin Dublin Ireland
- Department of Clinical Medicine, School of Medicine Trinity College Dublin Dublin Ireland
| | - Fionnuala M McAuliffe
- UCD Perinatal Research Centre, Obstetrics & Gynaecology School of Medicine, University College Dublin, National Maternity Hospital Dublin Ireland
| | - Sarah L Doyle
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Crumlin Dublin Ireland
- Department of Clinical Medicine, School of Medicine Trinity College Dublin Dublin Ireland
| |
Collapse
|
36
|
Wirsching S, Machtakova M, Borgans F, Pretsch L, Fichter M, Cacicedo ML, Thérien-Aubin H, Landfester K, Gehring S. OVA-PEG-R848 nanocapsules stimulate neonatal conventional and plasmacytoid dendritic cells. Front Pediatr 2022; 10:966113. [PMID: 36177449 PMCID: PMC9513203 DOI: 10.3389/fped.2022.966113] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/17/2022] [Indexed: 12/04/2022] Open
Abstract
Childhood mortality represents a major issue with 5. 3 million worldwide deaths of children under 5 years of age in 2019. Approximately half of those deaths can be attributed to easily preventable, infectious diseases. Currently approved neonatal vaccines are typically effective only after multiple doses leaving infants especially vulnerable during the first 6 months of life. Survival rates could be improved significantly by developing new and more potent vaccines that are capable of overcoming inherently tolerogenic neonatal immune systems. TLR agonists have garnered a great deal of attention in recent years due to their extensive capacities to activate innate immunity. Herein, the superior capacity of the TLR7/8 agonist, resiquimod (R848), to activate adult and neonatal primary peripheral blood dendritic cells is demonstrated. Moreover, R848 can be conjugated to polyethylene glycol and encapsulated in ovalbumin nanocapsules to efficiently co-deliver antigen and adjuvant in vitro. This study is among the first to demonstrate the capacity of encapsulated R848 to activate neonatal dendritic cells. These findings support the potential incorporation of R848 as adjuvant in neonatal vaccines, making them more effective in eliciting a robust immune response.
Collapse
Affiliation(s)
- Sebastian Wirsching
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | - Frauke Borgans
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.,Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt, Germany
| | - Leah Pretsch
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Michael Fichter
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.,Max Planck Institute for Polymer Research, Mainz, Germany.,Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Maximiliano L Cacicedo
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Héloïse Thérien-Aubin
- Max Planck Institute for Polymer Research, Mainz, Germany.,Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | | | - Stephan Gehring
- Children's Hospital, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
37
|
Angelidou A, Pittet LF, Faustman D, Curtis N, Levy O. BCG vaccine's off-target effects on allergic, inflammatory, and autoimmune diseases: Worth another shot? J Allergy Clin Immunol 2022; 149:51-54. [PMID: 34673049 PMCID: PMC11688639 DOI: 10.1016/j.jaci.2021.09.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 01/02/2023]
Affiliation(s)
- Asimenia Angelidou
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, Mass; Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Laure F Pittet
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Royal Children's Hospital Melbourne, Parkville, Australia; Unit of Pediatric Infectious Diseases, Division of General Pediatrics, Department of Pediatrics, Gynecology and Obstetrics, Children's Hospital, University Hospitals of Geneva, Geneva, Switzerland; Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Denise Faustman
- Department of Pediatrics, Harvard Medical School, Boston, Mass; Department of Immunobiology, Massachusetts General Hospital, Boston, Mass
| | - Nigel Curtis
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Royal Children's Hospital Melbourne, Parkville, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass; Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Mass.
| |
Collapse
|
38
|
Li X, Pan C, Sun P, Peng Z, Feng E, Wu J, Wang H, Zhu L. Orthogonal modular biosynthesis of nanoscale conjugate vaccines for vaccination against infection. NANO RESEARCH 2022; 15:1645-1653. [PMID: 34405037 PMCID: PMC8359766 DOI: 10.1007/s12274-021-3713-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/17/2021] [Accepted: 06/24/2021] [Indexed: 05/21/2023]
Abstract
UNLABELLED Conjugate vaccines represent one of the most effective means for controlling the occurrence of bacterial diseases. Although nanotechnology has been greatly applied in the field of vaccines, it is seldom used for conjugate vaccine research because it is very difficult to connect polysaccharides and nanocarriers. In this work, an orthogonal and modular biosynthesis method was used to produce nanoconjugate vaccines using the SpyTag/SpyCatcher system. When SpyTag/SpyCatcher system is combined with protein glycosylation technology, bacterial O-polysaccharide obtained from Shigela flexneri 2a can be conjugated onto the surfaces of different virus-like particles (VLPs) in a biocompatible and controlled manner. After confirming the excellent lymph node targeting and humoral immune activation abilities, these nanoconjugate vaccines further induced efficient prophylactic effects against infection in a mouse model. These results demonstrated that natural polysaccharide antigens can be easily connected to VLPs to prepare highly efficient nanoconjugate vaccines. To the best of the researchers' knowledge, this is the first time VLP-based nanoconjugate vaccines are produced efficiently, and this strategy could be applied to develop various pathogenic nanoconjugate vaccines. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (Figs. S1-S9) is available in the online version of this article at 10.1007/s12274-021-3713-4.
Collapse
Affiliation(s)
- Xin Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Peng Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Zhehui Peng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Erling Feng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Jun Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Hengliang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| |
Collapse
|
39
|
Ariawan AD, van Eersel J, Martin AD, Ke YD, Ittner LM. Recent progress in synthetic self-adjuvanting vaccine development. Biomater Sci 2022; 10:4037-4057. [DOI: 10.1039/d2bm00061j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Vaccination is a proven way to protect individuals against many infectious diseases, as currently highlighted in the global COVID-19 pandemic. Peptides- or small molecule antigen-based vaccination offer advantages over the...
Collapse
|
40
|
Allen ME, Golding A, Rus V, Karabin NB, Li S, Lescott CJ, Bobbala S, Scott EA, Szeto GL. Targeted Delivery of Chloroquine to Antigen-Presenting Cells Enhances Inhibition of the Type I Interferon Response. ACS Biomater Sci Eng 2021; 7:5666-5677. [PMID: 34813288 DOI: 10.1021/acsbiomaterials.1c01047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Systemic lupus erythematosus (SLE) causes damaging inflammation in multiple organs via the accumulation of immune complexes. These complexes activate plasmacytoid dendritic cells (pDCs) via toll-like receptors (TLRs), contributing to disease pathogenesis by driving the secretion of inflammatory type I interferons (IFNs). Antimalarial drugs, such as chloroquine (CQ), are TLR antagonists used to alleviate inflammation in SLE. However, they require ∼3 months of continuous use before achieving therapeutic efficacy and can accumulate in the retinal pigment epithelium with chronic use, resulting in retinopathy. We hypothesized that poly(ethylene glycol)-b-poly(propylene sulfide) filamentous nanocarriers, filomicelles (FMs), could directly deliver CQ to pDCs via passive, morphology-based targeting to concentrate drug delivery to specific immune cells, improve drug activity by increased inhibition of type I IFN, and enhance efficacy per dose. Healthy human peripheral blood mononuclear cells were treated with soluble CQ or CQ-loaded FMs, stimulated with TLR agonists or SLE patient sera, and type I IFN secretion was quantified via multi-subtype IFN-α ELISA and MX1 gene expression using real-time reverse transcription-quantitative polymerase chain reaction. Our results showed that 50 μg CQ/mg FM decreased MX1 expression and IFN-α production after TLR activation with either synthetic nucleic acid agonists or immune complex-rich sera from SLE patients. Cellular uptake and biodistribution studies showed that FMs preferentially accumulate in human pDCs and monocytes in vitro and in tissues frequently damaged in SLE patients (i.e., kidneys), while sparing the eye in vivo. These results showed that nanocarrier morphology enables drug delivery, and CQ-FMs may be equally effective and more targeted than soluble CQ at inhibiting SLE-relevant pathways.
Collapse
Affiliation(s)
- Marilyn E Allen
- Department of Chemical, Biochemical & Environmental Engineering, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, Maryland 21250, United States
| | - Amit Golding
- Department of Medicine, Division of Rheumatology & Clinical Immunology, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, Maryland 21201, United States
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology & Clinical Immunology, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, Maryland 21201, United States
| | - Nicholas B Karabin
- Department of Biomedical Engineering, Northwestern University, 633 Clark Street, Evanston, Illinois 60208, United States
| | - Sophia Li
- Department of Biomedical Engineering, Northwestern University, 633 Clark Street, Evanston, Illinois 60208, United States
| | - Chamille J Lescott
- Department of Biomedical Engineering, Northwestern University, 633 Clark Street, Evanston, Illinois 60208, United States
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, 1 Medical Center Drive, Morgantown, West Virginia 26506, United States
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, 633 Clark Street, Evanston, Illinois 60208, United States
| | - Gregory L Szeto
- Allen Institute for Immunology, 615 Westlake Avenue North, Seattle, Washington 98109, United States
| |
Collapse
|
41
|
Niculescu AG, Grumezescu AM. Polymer-Based Nanosystems-A Versatile Delivery Approach. MATERIALS (BASEL, SWITZERLAND) 2021; 14:6812. [PMID: 34832213 PMCID: PMC8619478 DOI: 10.3390/ma14226812] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 01/10/2023]
Abstract
Polymer-based nanoparticles of tailored size, morphology, and surface properties have attracted increasing attention as carriers for drugs, biomolecules, and genes. By protecting the payload from degradation and maintaining sustained and controlled release of the drug, polymeric nanoparticles can reduce drug clearance, increase their cargo's stability and solubility, prolong its half-life, and ensure optimal concentration at the target site. The inherent immunomodulatory properties of specific polymer nanoparticles, coupled with their drug encapsulation ability, have raised particular interest in vaccine delivery. This paper aims to review current and emerging drug delivery applications of both branched and linear, natural, and synthetic polymer nanostructures, focusing on their role in vaccine development.
Collapse
Affiliation(s)
- Adelina-Gabriela Niculescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania;
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania;
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
- Academy of Romanian Scientists, Ilfov no. 3, 50044 Bucharest, Romania
| |
Collapse
|
42
|
Vincent MP, Karabin NB, Allen SD, Bobbala S, Frey MA, Yi S, Yang Y, Scott EA. The Combination of Morphology and Surface Chemistry Defines the Immunological Identity of Nanocarriers in Human Blood. ADVANCED THERAPEUTICS 2021; 4:2100062. [PMID: 34485684 PMCID: PMC8411909 DOI: 10.1002/adtp.202100062] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Indexed: 12/13/2022]
Abstract
Upon exposure to blood, a corona of proteins adsorbs to nanocarrier surfaces to confer a biological identity that interfaces with the immune system. While the nanocarrier surface chemistry has long been the focus of protein corona formation, the influence of nanostructure has remained unclear despite established influences on biodistribution, clearance, and inflammation. Here, combinations of nanocarrier morphology and surface chemistry are engineered to i) achieve compositionally distinct protein coatings in human blood and ii) control protein-mediated interactions with the immune system. A library of nine PEGylated nanocarriers differing in their combination of morphology (spheres, vesicles, and cylinders) and surface chemistry (methoxy, hydroxyl, and phosphate) are synthesized to represent properties of therapeutic and biomimetic delivery vehicles. Analysis by quantitative label-free proteomic techniques reveal that specific surface chemistry and morphology combinations adsorb unique protein signatures from human blood, resulting in differential complement activation and elicitation of distinct proinflammatory cytokine responses. Furthermore, nanocarrier morphology is shown to primarily influence uptake and clearance by human monocytes, macrophages, and dendritic cells. This comprehensive analysis provides mechanistic insights into rational design choices that impact the immunological identity of nanocarriers in human blood, which can be leveraged to engineer drug delivery vehicles for precision medicine and immunotherapy.
Collapse
Affiliation(s)
- Michael P Vincent
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Nicholas B Karabin
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sean D Allen
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Sharan Bobbala
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Molly A Frey
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Sijia Yi
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Yufan Yang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA; Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA; Simpson Querrey Institute, Robert H. Lurie Medical Research Center, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
43
|
Bhagchandani S, Johnson JA, Irvine DJ. Evolution of Toll-like receptor 7/8 agonist therapeutics and their delivery approaches: From antiviral formulations to vaccine adjuvants. Adv Drug Deliv Rev 2021; 175:113803. [PMID: 34058283 PMCID: PMC9003539 DOI: 10.1016/j.addr.2021.05.013] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/04/2021] [Accepted: 05/15/2021] [Indexed: 02/07/2023]
Abstract
Imidazoquinoline derivatives (IMDs) and related compounds function as synthetic agonists of Toll-like receptors 7 and 8 (TLR7/8) and one is FDA approved for topical antiviral and skin cancer treatments. Nevertheless, these innate immune system-activating drugs have potentially much broader therapeutic utility; they have been pursued as antitumor immunomodulatory agents and more recently as candidate vaccine adjuvants for cancer and infectious disease. The broad expression profiles of TLR7/8, poor pharmacokinetic properties of IMDs, and toxicities associated with systemic administration, however, are formidable barriers to successful clinical translation. Herein, we review IMD formulations that have advanced to the clinic and discuss issues related to biodistribution and toxicity that have hampered the further development of these compounds. Recent strategies aimed at enhancing safety and efficacy, particularly through the use of bioconjugates and nanoparticle formulations that alter pharmacokinetics, biodistribution, and cellular targeting, are described. Finally, key aspects of the biology of TLR7 signaling, such as TLR7 tolerance, that may need to be considered in the development of new IMD therapeutics are discussed.
Collapse
Affiliation(s)
- Sachin Bhagchandani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jeremiah A Johnson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
44
|
Li X, Yi S, Scariot DB, Martinez SJ, Falk BA, Olson CL, Romano PS, Scott EA, Engman DM. Nanocarrier-enhanced intracellular delivery of benznidazole for treatment of Trypanosoma cruzi infection. JCI Insight 2021; 6:145523. [PMID: 33986194 PMCID: PMC8262286 DOI: 10.1172/jci.insight.145523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/31/2021] [Indexed: 11/17/2022] Open
Abstract
Chagas disease is caused by infection with the protozoan parasite Trypanosoma cruzi (T. cruzi), an intracellular pathogen that causes significant morbidity and death among millions in the Americas from Canada to Argentina. Current therapy involves oral administration of the nitroimidazole benznidazole (BNZ), which has serious side effects that often necessitate cessation of treatment. To both avoid off-target side effects and reduce the necessary dosage of BNZ, we packaged the drug within poly(ethylene glycol)-block-poly(propylene sulfide) polymersomes (BNZ-PSs). We show that these vesicular nanocarriers enhanced intracellular delivery to phagocytic cells and tested this formulation in a mouse model of T. cruzi infection. BNZ-PS is not only nontoxic but also significantly more potent than free BNZ, effectively reducing parasitemia, intracellular infection, and tissue parasitosis at a 466-fold lower dose of BNZ. We conclude that BNZ-PS was superior to BNZ for treatment of T. cruzi infection in mice and that further modifications of this nanocarrier formulation could lead to a wide range of custom controlled delivery applications for improved treatment of Chagas disease in humans.
Collapse
Affiliation(s)
- Xiaomo Li
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Pathology, Northwestern University, Chicago, Illinois, USA
| | - Sijia Yi
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, Illinois, USA
| | - Débora B. Scariot
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, Illinois, USA
| | - Santiago J. Martinez
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Institute of Histology and Embryology, “Dr. Mario H. Burgos”, IHEM-CONICET, National University of Cuyo, Mendoza, Argentina
| | - Ben A. Falk
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Cheryl L. Olson
- Department of Pathology, Northwestern University, Chicago, Illinois, USA
| | - Patricia S. Romano
- Institute of Histology and Embryology, “Dr. Mario H. Burgos”, IHEM-CONICET, National University of Cuyo, Mendoza, Argentina
| | - Evan A. Scott
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, Illinois, USA
| | - David M. Engman
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Pathology, Northwestern University, Chicago, Illinois, USA
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
45
|
Stack T, Liu Y, Frey M, Bobbala S, Vincent M, Scott E. Enhancing subcutaneous injection and target tissue accumulation of nanoparticles via co-administration with macropinocytosis inhibitory nanoparticles (MiNP). NANOSCALE HORIZONS 2021; 6:393-400. [PMID: 33884386 PMCID: PMC8127988 DOI: 10.1039/d0nh00679c] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
A significant barrier to the application of nanoparticles for precision medicine is the mononuclear phagocyte system (MPS), a diverse population of phagocytic cells primarily located within the liver, spleen and lymph nodes. The majority of nanoparticles are indiscriminately cleared by the MPS via macropinocytosis before reaching their intended targets, resulting in side effects and decreased efficacy. Here, we demonstrate that the biodistribution and desired tissue accumulation of targeted nanoparticles can be significantly enhanced by co-injection with polymeric micelles containing the actin depolymerizing agent latrunculin A. These macropinocytosis inhibitory nanoparticles (MiNP) were found to selectively inhibit non-specific uptake of a second "effector" nanoparticle in vitro without impeding receptor-mediated endocytosis. In tumor bearing mice, co-injection with MiNP in a single multi-nanoparticle formulation significantly increased the accumulation of folate-receptor targeted nanoparticles within tumors. Furthermore, subcutaneous co-administration with MiNP allowed effector nanoparticles to achieve serum levels that rivaled a standard intravenous injection. This effect was only observed if the effector nanoparticles were injected within 24 h following MiNP administration, indicating a temporary avoidance of MPS cells. Co-injection with MiNP therefore allows reversible evasion of the MPS for targeted nanoparticles and presents a previously unexplored method of modulating and improving nanoparticle biodistribution following subcutaneous administration.
Collapse
Affiliation(s)
- Trevor Stack
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA.
| | - Yugang Liu
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA.
| | - Molly Frey
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA.
| | - Sharan Bobbala
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA.
| | - Michael Vincent
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA.
| | - Evan Scott
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA.
| |
Collapse
|
46
|
England R, Pak J, Liu M, Rao S, Ozonoff A, Levy O, van Haren SD. Human Blood Plasma Shapes Distinct Neonatal TLR-Mediated Dendritic Cell Activation via Expression of the MicroRNA Let-7g. Immunohorizons 2021; 5:246-256. [PMID: 33931496 PMCID: PMC11688640 DOI: 10.4049/immunohorizons.2000081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/19/2021] [Indexed: 11/19/2022] Open
Abstract
The newborn innate immune system is characterized as functionally distinct, resulting in impaired proinflammatory responses to many stimuli and a bias toward Th2 development. Although the magnitude of impairment can be partially overcome, for instance through activation of TLR7/8 in newborn dendritic cells, the newborn innate response remains distinct from that of adults. Using human in vitro modeling of newborn and adult dendritic cells, we investigated the role of extracellular and intracellular regulators in driving age-specific responses to TLR7/8 stimulation. MicroRNA expression profiling and plasma switch experiments identified Let-7g as a novel regulator of newborn innate immunity. Activation-induced expression of Let-7g in monocyte-derived dendritic cells (MoDCs) is driven by newborn plasma and reduces expression of costimulatory receptors CD86, MHC class I, and CCR7 and secretion of IFN-α and sCD40L. Conversely, an increase in secretion of the Th2-polarizing cytokine IL-12p40 is observed. Overexpression of Let-7g in adult MoDCs resulted in the same observations. Small interfering RNA-mediated ablation of Let-7g levels in newborn MoDCs resulted in an adult-like phenotype. In conclusion, this study reveals for the first time (to our knowledge) that age-specific differences in human plasma induce the microRNA Let-7g as a key mediator of the newborn innate immune phenotype. These observations shed new light on the mechanisms of immune ontogeny and may inform approaches to discover age-specific immunomodulators, such as adjuvants.
Collapse
Affiliation(s)
- Ross England
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | - Jensen Pak
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | - Mark Liu
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | - Shun Rao
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | - Al Ozonoff
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Simon D van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA;
- Harvard Medical School, Boston, MA; and
| |
Collapse
|
47
|
An Overview of Nanocarrier-Based Adjuvants for Vaccine Delivery. Pharmaceutics 2021; 13:pharmaceutics13040455. [PMID: 33801614 PMCID: PMC8066039 DOI: 10.3390/pharmaceutics13040455] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 12/12/2022] Open
Abstract
The development of vaccines is one of the most significant medical accomplishments which has helped to eradicate a large number of diseases. It has undergone an evolutionary process from live attenuated pathogen vaccine to killed whole organisms or inactivated toxins (toxoids), each of them having its own advantages and disadvantages. The crucial parameters in vaccination are the generation of memory response and protection against infection, while an important aspect is the effective delivery of antigen in an intelligent manner to evoke a robust immune response. In this regard, nanotechnology is greatly contributing to developing efficient vaccine adjuvants and delivery systems. These can protect the encapsulated antigen from the host’s in-vivo environment and releasing it in a sustained manner to induce a long-lasting immunostimulatory effect. In view of this, the present review article summarizes nanoscale-based adjuvants and delivery vehicles such as viral vectors, virus-like particles and virosomes; non-viral vectors namely nanoemulsions, lipid nanocarriers, biodegradable and non-degradable nanoparticles, calcium phosphate nanoparticles, colloidally stable nanoparticles, proteosomes; and pattern recognition receptors covering c-type lectin receptors and toll-like receptors.
Collapse
|
48
|
Tokuhara D, Hikita N. Cord Blood-Based Approach to Assess Candidate Vaccine Adjuvants Designed for Neonates and Infants. Vaccines (Basel) 2021; 9:vaccines9020095. [PMID: 33514054 PMCID: PMC7911524 DOI: 10.3390/vaccines9020095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Neonates and infants are particularly susceptible to infections, for which outcomes tend to be severe. Vaccination is a key strategy for preventing infectious diseases, but the protective immunity achieved through vaccination typically is weaker in infants than in healthy adults. One possible explanation for the poor acquisition of vaccine-induced immunity in infants is that their innate immune response, represented by toll-like receptors, is immature. The current system for developing pediatric vaccines relies on the confirmation of their safety and effectiveness in studies involving the use of mature animals or adult humans. However, creating vaccines for neonates and infants requires an understanding of their uniquely immature innate immunity. Here we review current knowledge regarding the innate immune system of neonates and infants and challenges in developing vaccine adjuvants for those children through analyses of cord blood.
Collapse
|
49
|
Velluto D, Bojadzic D, De Toni T, Buchwald P, Tomei AA. Drug-Integrating Amphiphilic Nanomaterial Assemblies: 1. Spatiotemporal control of cyclosporine delivery and activity using nanomicelles and nanofibrils. J Control Release 2021; 329:955-970. [PMID: 33086102 PMCID: PMC7904645 DOI: 10.1016/j.jconrel.2020.10.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/12/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022]
Abstract
Immunomodulatory therapies are limited by unavoidable side effects as well as poor solubility, stability, and pharmacokinetic properties. Nanomaterial-based drug delivery may overcome these limitations by increasing drug solubility, site-targeting, and duration of action. Here, we prepared innovative drug-integrating amphiphilic nanomaterial assemblies (DIANA) with tunable hydrophobicity, size, and morphology, and we evaluated their ability to deliver cyclosporine A (CsA) for immunomodulatory applications. We synthesized amphiphilic block copolymers made of poly(ethylene glycol)-poly(propylene sulfide) (PEG-PPS) and poly(ethylene glycol)-oligo(ethylene sulfide) (PEG-OES) that can self-assemble into solid core nanomicelles (nMIC, with ≈20 nm diameter) and nanofibrils (nFIB, with ≈5 nm diameter and > 500 nm length), respectively. nMIC and nFIB displayed good CsA encapsulation efficiency (up to 4.5 and 2 mg/mL, respectively in aqueous solution), superior to many other solubilization methods, and provided sustained release (>14 and > 7 days for the nMIC and nFIB) without compromising CsA's pharmacological activity. Treatment of insulin-secreting cells with unloaded DIANAs did not impair cell viability and functionality. Both CsA-loaded DIANAs inhibited the proliferation and activation of insulin-reactive cytotoxic T cells in vitro. Subcutaneous injections of CsA-loaded DIANAs in mice provided CsA sustained release, decreasing alloantigen-induced immune responses in the draining lymph node at lower doses and reduced administration frequency than unformulated CsA. While nMIC solubilized higher amounts and provided more sustained release of CsA in vitro, nFIB enhanced cellular uptake and promoted local retention due to slower trafficking in vivo. DIANAs provide a versatile platform for a local immune suppression regimen that can be applied to allogeneic cell transplantation.
Collapse
Affiliation(s)
- Diana Velluto
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Damir Bojadzic
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Teresa De Toni
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Alice A Tomei
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Biomedical Engineering, University of Miami, Miami, FL, USA; Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
50
|
Soni D, Bobbala S, Li S, Scott EA, Dowling DJ. The sixth revolution in pediatric vaccinology: immunoengineering and delivery systems. Pediatr Res 2021; 89:1364-1372. [PMID: 32927471 PMCID: PMC7511675 DOI: 10.1038/s41390-020-01112-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 05/08/2020] [Accepted: 05/28/2020] [Indexed: 02/08/2023]
Abstract
Infection is the predominant cause of mortality in early life, and immunization is the most promising biomedical intervention to reduce this burden. However, very young infants fail to respond optimally to most vaccines currently in use, especially neonates. In 2005, Stanley Plotkin proposed that new delivery systems would spur a new revolution in pediatric vaccinology, just as attenuation, inactivation, cell culture of viruses, genetic engineering, and adjuvantation had done in preceding decades. Recent advances in the field of immunoengineering, which is evolving alongside vaccinology, have begun to increasingly influence vaccine formulation design. Historically, the particulate nature of materials used in many vaccine formulations was empiric, often because of the need to stabilize antigens or reduce endotoxin levels. However, present vaccine delivery systems are rationally engineered to mimic the size, shape, and surface chemistry of pathogens, and are therefore often referred to as "pathogen-like particles". More than a decade from his original assessment, we re-assess Plotkin's prediction. In addition, we highlight how immunoengineering and advanced delivery systems may be uniquely capable of enhancing vaccine responses in vulnerable populations, such as infants. IMPACT: Immunoengineering and advanced delivery systems are leading to new developments in pediatric vaccinology. Summarizes delivery systems currently in use and development, and prospects for the future. Broad overview of immunoengineering's impact on vaccinology, catering to Pediatric Clinicians and Immunologists.
Collapse
Affiliation(s)
- Dheeraj Soni
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Sharan Bobbala
- grid.16753.360000 0001 2299 3507Department of Biomedical Engineering, Northwestern University, Evanston, IL USA
| | - Sophia Li
- grid.16753.360000 0001 2299 3507Department of Biomedical Engineering, Northwestern University, Evanston, IL USA
| | - Evan A. Scott
- grid.16753.360000 0001 2299 3507Department of Biomedical Engineering, Northwestern University, Evanston, IL USA
| | - David J. Dowling
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| |
Collapse
|