1
|
Bonyek-Silva Í, Bastos R, Nunes S, Tibúrcio R, Lago A, Silva J, Carvalho LP, Khouri R, Arruda SM, Barral A, Boaventura V, Serezani HC, Carvalho EM, Brodskyn CI, Tavares NM. High glucose heightens vulnerability to Leishmania braziliensis infection in human macrophages by hampering the production of reactive oxygen species through TLR2 and TLR4. Emerg Microbes Infect 2025; 14:2475824. [PMID: 40052633 PMCID: PMC11948364 DOI: 10.1080/22221751.2025.2475824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/26/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025]
Abstract
Diabetes increases susceptibility to infections, including Leishmania braziliensis (Lb). Our group previously demonstrated that diabetic patients with cutaneous leishmaniasis (CL) take longer to heal lesions compared to non-diabetics. Since macrophages play a critical role in CL pathogenesis, we investigated how high glucose levels impact their response during Lb infection. Macrophages cultured in high glucose conditions showed increased parasite load than those in normal glucose conditions. The production of inflammatory mediators was similar between glucose conditions, but basal reactive oxygen species (ROS) production was elevated under high glucose conditions and remained unchanged after Lb infection, indicating glucose-induced oxidative stress does not control the parasite. In contrast, macrophages in normal glucose conditions, exhibited increased ROS production only after infection. Additionally, high glucose reduced TLR2 and TLR4 expression, which was also observed after Lb infection. TLR2/4 inhibition increased Lb infection in normal glucose conditions, mediated by TLR-dependent ROS production. However, this mechanism was absent under high glucose conditions, where elevated basal ROS production appeared TLR-independent. Biopsies from diabetic CL patients corroborated these findings, showing decreased TLR2 and TLR4 expression compared to non-diabetics. These findings suggest that high glucose levels induce oxidative stress and reduces TLR expression, impairing macrophage functions and rendering them less effective at controlling Lb infection.
Collapse
Affiliation(s)
- Ícaro Bonyek-Silva
- Federal Institute of Education, Science and Technology Baiano (IFBA), Xique-Xique, Brazil
- Nursing School, Irecê College, Irecê, Brazil
| | - Rana Bastos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- School of Medicine, Federal University of Bahia (UFBA), Salvador, Brazil
| | - Sara Nunes
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
| | - Rafael Tibúrcio
- Division of Experimental Medicine, Department of Medicine, School of Medicine, University of California San Francisco, San Francisco, United States
| | - Alexsandro Lago
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
| | - Juliana Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
| | - Lucas P. Carvalho
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- School of Medicine, Federal University of Bahia (UFBA), Salvador, Brazil
| | - Ricardo Khouri
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- School of Medicine, Federal University of Bahia (UFBA), Salvador, Brazil
| | - Sergio M. Arruda
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
| | - Aldina Barral
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- InCor (Heart Institute), University of São Paulo, Institute of Investigation in Immunology (iii), São Paulo, Brazil
| | - Viviane Boaventura
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- School of Medicine, Federal University of Bahia (UFBA), Salvador, Brazil
- InCor (Heart Institute), University of São Paulo, Institute of Investigation in Immunology (iii), São Paulo, Brazil
| | - Henrique C. Serezani
- Department of Medicine, Division of Infectious Diseases, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, USA
| | - Edgar M. Carvalho
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- School of Medicine, Federal University of Bahia (UFBA), Salvador, Brazil
| | - Cláudia Ida Brodskyn
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- InCor (Heart Institute), University of São Paulo, Institute of Investigation in Immunology (iii), São Paulo, Brazil
| | - Natalia Machado Tavares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- InCor (Heart Institute), University of São Paulo, Institute of Investigation in Immunology (iii), São Paulo, Brazil
| |
Collapse
|
2
|
Wei H, Liu Y, Huang C, Wang C, Jiang H, Wang L, Wang Y, Wang Z. Ginsenoside Rg1 targets TLR2 to inhibit the NF-κB signaling pathway and ameliorate hematopoietic support of mesenchymal stromal cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119917. [PMID: 40348305 DOI: 10.1016/j.jep.2025.119917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/19/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginsenoside Rg1 is a key bioactive compound in ginseng, a traditional herbal medicine known for tonifying qi and nourishing blood, traditionally used to replenish "qi" by regulating hematopoietic function. But its underlying mechanism remains to be elucidated. AIM OF THE STUDY This study aims to delve into the role of Rg1 on senescent hematopoiesis and its mechanisms. MATERIALS AND METHODS A model of D-galactose-induced hematopoietic injury was established with ginsenoside Rg1. The hematopoietic supportive effect of Rg1 was assessed by quantifying the levels of hematopoietic supportive factors VCAM1, CXCL12 and SCF, CFU-Mix formation and cellular senescence; and the levels of inflammatory factors and oxidative stress were measured by ELISA in the serum and cellular supernatant of mice. Co-culture technique was used to examine the ability of Rg1 to restore impaired hematopoiesis by improving the inflammatory hematopoietic microenvironment. For mechanism exploration, RNA-Seq was used to detect differential genes in Rg1-treated MSCs, GO- and KEGG-based enrichment analyses were used to screen the key pathways in which Rg1 exerts its effects, and molecular docking was used to demonstrate the feasibility of molecular interconnections between Rg1 and TLR2. To further explore the mechanism, pathway activators were further used and the expression levels of target proteins downstream of the TLR2 pathway were quantified using Western blotting. RESULTS Rg1 decreased the levels of inflammatory factors IL-1β, IL-6 and TNFα, while enhancing the expression of hematopoietic support factors in senescent MSCs, thereby improving the self-renewal and differentiation of aged HSPCs. Additionally, Rg1 also delayed HSPC senescence and reduced the level of oxidative stress. KEGG and GO were enriched for the Toll/NF-κB signaling pathway, based on differential genes obtained by transcriptional sequencing. Rg1 could inhibit the elevated levels of MyD88, NF-κB-p65 and IκBα proteins, and their phosphorylation levels by binding to TLR2 protein and inhibiting them. In conclusion, Rg1 ameliorates the inflammatory hematopoietic microenvironment induced by MSCs senescence via the TLR2/NF-κB-p65 signaling pathway, alleviating HSPCs senescence. CONCLUSIONS Our results reveal the mechanism by which Rg1 regulates HSPCs function and represent a potential therapeutic strategy for hematopoietic dysfunction, highlighting the potential value of traditional Chinese medicine extracts in clinical applications.
Collapse
Affiliation(s)
- Han Wei
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, PR China; Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yonggang Liu
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, PR China; Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, PR China
| | - Caihong Huang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, PR China; Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, PR China
| | - Cheng Wang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, PR China; Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, PR China
| | - Honghui Jiang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, PR China; Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, PR China
| | - Lu Wang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, PR China; Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yaping Wang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, PR China; Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Ziling Wang
- Department of Pathology, Basic Medicine College, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, PR China; Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
3
|
Allela OQB, Al-Hussainy AF, Sanghvi G, Roopashree R, Kashyap A, Anand DA, Panigrahi R, Garifulina LM, Taher SG, Alwan M, Jawad M, Mushtaq H. Tumor immune evasion and the Let-7 family: insights into mechanisms and therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04283-9. [PMID: 40423803 DOI: 10.1007/s00210-025-04283-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 05/09/2025] [Indexed: 05/28/2025]
Abstract
Tumor immune evasion is a complex and adaptive mechanism that allows cancer cells to escape immune detection and destruction, contributing to malignancy progression and poor therapeutic outcomes. This review article explores the integral role of the let-7 family of microRNAs (miRNAs) in mediating tumor immune evasion, particularly how these regulators influence the tumor microenvironment (TME) and immune cell functionality. The let-7 family, known for its tumor-suppressive roles, modulates key immune checkpoints, including PD-L1, and pathways linked to immune response regulation, such as the STAT3/SOCS axis, impacts macrophage polarization and modulates immune cell function. Dysregulation of let-7 miRNAs can enhance tumor immune evasion through mechanisms such as downregulating major histocompatibility complex (MHC) expressions, promoting immunosuppressive cell populations, and manipulating metabolic pathways, which together establish an immunosuppressive TME. Conversely, specific let-7 members show potential in restoring anti-tumor immunity by reversing immune suppression and improving T cell responses. By synthesizing current research, this article underscores the dual role of let-7 in both promoting and inhibiting tumor immune evasion, suggesting their potential as therapeutic targets and biomarkers in cancer immunotherapy. Future studies on the context-dependent roles and advanced delivery systems for let-7-targeting therapies are crucial for enhancing immunotherapeutic efficacy and improving patient outcomes across malignancies.
Collapse
Affiliation(s)
| | | | - Gaurav Sanghvi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Aditya Kashyap
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - D Alex Anand
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Rajashree Panigrahi
- Department of Microbiology, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | | | - Sada Ghalib Taher
- College of Health and Medical Technology, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Mariem Alwan
- Pharmacy college, Al-Farahidi University, Baghdad, Iraq
| | - Mahmood Jawad
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | |
Collapse
|
4
|
Zhang J, Huang Q, Yang H, Shi X, Su Y, Xia J, Li Y, Liu X. Lipid Coating of Mesoporous Silica Nanoparticles Leads to Efficient Antigen Delivery to Lymph Nodes for Cancer Vaccination. ACS APPLIED BIO MATERIALS 2025; 8:4294-4302. [PMID: 40310259 DOI: 10.1021/acsabm.5c00403] [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] [Indexed: 05/02/2025]
Abstract
The enrichment of antigens in lymph nodes and the ensuing antigen presentation constitute crucial steps in determining the efficacy of tumor vaccines. However, antigen delivery is restricted by enzyme degradation, immune system clearance, and the difficulty of crossing biological barriers. In this study, mesoporous silica nanoparticles (MSNPs) were prepared for antigen loading and were further coated with a phospholipid bilayer membrane (named silicasomes) to improve the delivery efficiency to lymph nodes. Our results showed that silicasomes exhibited superior lymph node enrichment compared to the bare MSNPs following subcutaneous injection. Moreover, their efficacy as a tumor vaccine was validated in the B16-OVA tumor model by loading the ovalbumin antigens (OVA257-264). Besides, the toll-like receptor 4 (TLR4) agonist monophosphoryl lipid A (MPLA), a component of bacterial lipopolysaccharides, was incorporated into the phospholipid membrane on the surface of silicasomes as an adjuvant. The silicasome-OVA257-264 with the addition of MPLA exhibited a more potent antitumor effect, triggering the infiltration of specific T cells into the tumor. These results demonstrated that lipid coating on MSNPs significantly enhanced their delivery efficiency to lymph nodes and enabled synergistic immune activation of tumor antigens and adjuvants, highlighting their potential as effective vehicles for cancer immunotherapy.
Collapse
Affiliation(s)
- Jia Zhang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
| | - Qiang Huang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
- Department of Urology, The First Hospital of China Medical University, Institute of Urology, China Medical University, Shenyang 110001, China
| | - Honghong Yang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
| | - Xiayu Shi
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
| | - Yang Su
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
- Hangzhou Institute for Advanced Study (UCAS), Hangzhou, Zhejiang 310000, China
| | - Junlong Xia
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yuting Li
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
| | - Xiangsheng Liu
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
| |
Collapse
|
5
|
Konopka EN, Edgerton AO, Kutzler MA. Nucleic acid vaccines: innovations, efficacy, and applications in at-risk populations. Front Immunol 2025; 16:1584876. [PMID: 40438110 PMCID: PMC12116436 DOI: 10.3389/fimmu.2025.1584876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/09/2025] [Indexed: 06/01/2025] Open
Abstract
For more than two centuries, the field of vaccine development has progressed through the adaptation of novel platforms in parallel with technological developments. Building off the advantages and shortcomings of first and second-generation vaccine platforms, the advent of third-generation nucleic acid vaccines has enabled new approaches to tackle emerging infectious diseases, cancers, and pathogens where vaccines remain unavailable. Unlike traditional vaccine platforms, nucleic acid vaccines offer several new advantages, including their lower cost and rapid production, which was widely demonstrated during the COVID-19 pandemic. Beyond production, DNA and mRNA vaccines can elicit unique and targeted responses through specialized design and delivery approaches. Considering the growth of nucleic acid vaccine research over the past two decades, the evaluation of their efficacy in at-risk populations is paramount for refining and improving vaccine design. Importantly, the aging population represents a significant portion of individuals highly susceptible to infection and disease. This review seeks to outline the major impairments in vaccine-induced responses due to aging that may be targeted for improvement with design and delivery components encompassing mRNA and DNA vaccine formulations. Results of pre-clinical and clinical applications of these vaccines in aged animal models and humans will also be evaluated to outline current successes and limitations observed in these platforms.
Collapse
Affiliation(s)
- Emily N. Konopka
- Drexel University College of Medicine, Department of Microbiology and Immunology, Philadelphia, PA, United States
- Drexel University College of Medicine, Department of Medicine, Division of Infectious Diseases and HIV Medicine, Philadelphia, PA, United States
| | - Arden O. Edgerton
- Drexel University College of Medicine, Department of Microbiology and Immunology, Philadelphia, PA, United States
- Drexel University College of Medicine, Department of Medicine, Division of Infectious Diseases and HIV Medicine, Philadelphia, PA, United States
| | - Michele A. Kutzler
- Drexel University College of Medicine, Department of Microbiology and Immunology, Philadelphia, PA, United States
- Drexel University College of Medicine, Department of Medicine, Division of Infectious Diseases and HIV Medicine, Philadelphia, PA, United States
| |
Collapse
|
6
|
Xing J, Zhao X, Li X, Fang R, Sun M, Zhang Y, Song N. The recent advances in vaccine adjuvants. Front Immunol 2025; 16:1557415. [PMID: 40433383 PMCID: PMC12106398 DOI: 10.3389/fimmu.2025.1557415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Vaccine adjuvants, as key components in enhancing vaccine immunogenicity, play a vital role in modern vaccinology. This review systematically examines the historical evolution and mechanisms of vaccine adjuvants, with particular emphasis on innovative advancements in aluminum-based adjuvants, emulsion-based adjuvants, and nucleic acid adjuvants (e.g., CpG oligonucleotides). Specifically, aluminum adjuvants enhance immune responses through particle formation/antigen adsorption, inflammatory cascade activation, and T-cell stimulation. Emulsion adjuvants amplify immunogenicity via antigen depot effects and localized inflammation, while nucleic acid adjuvants like CpG oligonucleotides directly activate B cells and dendritic cells to promote Th1-type immune responses and memory T-cell generation. The article further explores the prospective applications of these novel adjuvants in combating emerging pathogens (including influenza and SARS-CoV-2), particularly highlighting their significance in improving vaccine potency and durability. Moreover, this review underscores the critical importance of adjuvant development in next-generation vaccine design and provides theoretical foundations for creating safer, effective adjuvant.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ningning Song
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, School of Life Science and Technology, Shandong Second Medical University, Weifang, China
| |
Collapse
|
7
|
Fu Y, Kim H, Lee DS, Han AR, Heine H, Zamyatina A, Kim HM. Structural insight into TLR4/MD-2 activation by synthetic LPS mimetics with distinct binding modes. Nat Commun 2025; 16:4164. [PMID: 40325026 PMCID: PMC12053604 DOI: 10.1038/s41467-025-59550-3] [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: 10/23/2024] [Accepted: 04/28/2025] [Indexed: 05/07/2025] Open
Abstract
The mammalian pattern-recognition receptor TLR4/MD-2 (Toll-like receptor 4/myeloid differentiation factor-2) can be activated by a wide variety of pathogen-associated and endogenous molecules, with Gram-negative bacterial lipopolysaccharide (LPS) being the primary natural TLR4 agonist. Activation of TLR4 triggers cellular signaling that enables the beneficial innate immune responses and enhances adaptive immunity, thereby emphasizing the potential of TLR4 agonists for the management of diseases with an immunopathological background and for use as vaccine adjuvants. Given the challenges associated with LPS-derived products, including structural complexity, heterogeneity, toxicity, and species specificity, synthetic molecules targeting TLR4/MD-2 offer a promising alternative. Here, we elucidate the structural basis for the recognition of synthetic LPS-mimicking glycolipids, Disaccharide Lipid A Mimetics (DLAMs), by human and mouse TLR4/MD-2 through cryo-EM structures of six dimeric [TLR4/MD-2/ligand]2 complexes resolved at 2.2-3.1 Å. We reveal that the specific binding modes of DLAMs, distinct from those of LPS, are essential for the species-independent TLR4 agonistic activity. DLAMs function as a molecular bridge, effectively induce the dimerization of TLR4/MD-2 complexes through specific carbohydrate structure-relevant ligand-protein interactions. Our findings reveal the distinct molecular modes of TLR4 activation, and provide a structural basis for the rationale design and development of innovative, highly potent TLR4-targeting immunotherapeutics and adjuvants.
Collapse
Affiliation(s)
- Yaoyao Fu
- Department of Biological Science, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- Center for Biomolecular & Cellular Structure, Institute for Basic Science, Daejeon, Republic of Korea
| | - Hyojin Kim
- Center for Biomolecular & Cellular Structure, Institute for Basic Science, Daejeon, Republic of Korea
| | - Dong Sun Lee
- Department of Biological Science, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- Center for Biomolecular & Cellular Structure, Institute for Basic Science, Daejeon, Republic of Korea
| | - Ah-Reum Han
- Center for Biomolecular & Cellular Structure, Institute for Basic Science, Daejeon, Republic of Korea
| | - Holger Heine
- Research Group Innate Immunity, Research Center Borstel - Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Alla Zamyatina
- Department of Natural Sciences and Sustainable Resources, Institute of Organic Chemistry, BOKU University, Vienna, Austria.
| | - Ho Min Kim
- Department of Biological Science, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
- Center for Biomolecular & Cellular Structure, Institute for Basic Science, Daejeon, Republic of Korea.
| |
Collapse
|
8
|
Sala E, Nelli M, Laura C, Di Lucia P, Beccaria CG, Bono EB, Mangione M, Marotta D, Sperto V, Grillo M, Giustini L, Tosi F, Nie J, Kim D, Furiato G, Malpighi C, Consolo E, Becher B, David E, Cohen M, Giladi A, Amit I, Bosselut R, Guidotti LG, Iannacone M, Kuka M. T-cell-derived IFN-γ suppresses T follicular helper cell differentiation and antibody responses. EMBO J 2025; 44:2400-2423. [PMID: 40169810 PMCID: PMC12048687 DOI: 10.1038/s44318-025-00414-3] [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: 09/17/2024] [Revised: 02/23/2025] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Abstract
CD4+ T cells play a critical role in antiviral humoral and cellular immune responses. We have previously reported that subcutaneous lymphocytic choriomeningitis virus (s.c. LCMV) infection is characterized by a stark compartmentalization of CD4+ T cells, leading to strong TH1 cell polarization but virtually absent T follicular helper (TFH) cells, key drivers of humoral immunity. Here, we investigate the mechanisms responsible for this impaired TFH differentiation. We show that T-bet+ cells induced by LCMV infection encompass a TH1 cell subset expressing granzyme B (GzmB), and a Tcf-1+ cell subset that retains the potential for TFH differentiation without expressing mature TFH markers. Notably, IFN-γ blockade enables full differentiation of Tcf-1+ cells into TFH cells, formation of germinal centers, and increased antibody production. Suppression of TFH cells by IFN-γ is not directly mediated by CD4+ T cells but rather involves another cell type, likely dendritic cells (DCs). Our study provides novel insights into the mechanisms underlying early CD4+ T-cell polarization and humoral responses to viruses, with the potential to facilitate the development of effective vaccine strategies.
Collapse
Affiliation(s)
- Eleonora Sala
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Nelli
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Laura
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pietro Di Lucia
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristian Gabriel Beccaria
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa B Bono
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Mangione
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Davide Marotta
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Sperto
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Grillo
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Leonardo Giustini
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Tosi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Jia Nie
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daehong Kim
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Giuliana Furiato
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Malpighi
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eleonora Consolo
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Merav Cohen
- Department of Clinical Microbiology and Immunology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Amir Giladi
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Remy Bosselut
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Luca G Guidotti
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Iannacone
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Mirela Kuka
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
9
|
Coats SR, Su TH, Luderman Miller Z, King AJ, Ortiz J, Reddy A, Alaei SR, Jain S. Porphyromonas gingivalis outer membrane vesicles divert host innate immunity and promote inflammation via C4' monophosphorylated lipid A. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:1008-1021. [PMID: 40131356 PMCID: PMC12123218 DOI: 10.1093/jimmun/vkae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/05/2024] [Indexed: 03/27/2025]
Abstract
Porphyromonas gingivalis (Pg) is a prevalent pathogen that promotes human periodontal disease (PD) and exacerbates systemic comorbidities such as atherosclerosis, rheumatoid arthritis, and Alzheimer's disease. Pg produces nonphosphorylated tetra-acylated lipid A (NPLA) in its outer membrane (OM) that evades host Toll-like receptor 4 (TLR4), inflammasome pathways, and cationic peptides, enhancing bacterial survival. Here, we show that Pg also releases outer membrane vesicles (OMVs) that engage and divert host cell TLR4, inflammasome, and LL-37 responses away from the microbe. We determined that Pg OMVs are enriched for C4' monophosphoryl lipid A (C4'-MPLA), an established agonist for TLR4-TRIF-IFNβ and inflammasome-IL-1β responses. Comparisons of Pg 381 and Pg 33277 stationary phase cultures revealed higher OMV production by Pg 381, which correlates with its higher proinflammatory pathogenicity. The cationic peptide, polymyxin B (PMB), which selectively binds lipid A C4'-phosphate, reduces OMV-stimulated HEK cell TLR4 activation and THP-1 cell IL-1β production, confirming the proinflammatory role for OMV-C4'-MPLA. Similar to PMB, the host defense peptide, LL-37, inhibits OMV-C4'-MPLA-dependent HEK cell TLR4 activation. PMB and LL-37 also blocked OMV-C4'-MPLA-driven TLR4 activation in human umbilical vein endothelial cells. Finally, wild-type Pg-containing OM-NPLA is highly resistant to LL-37 antimicrobial activity, whereas the ΔlpxF mutant bacterium, retaining OM-C4'-MPLA, is killed by the peptide. In summary, Pg escapes host TLR4 signaling, inflammasome activation, and LL-37 interaction by retaining immunoevasive OM-NPLA. Moreover, Pg dispenses proinflammatory OMV-C4'-MPLA, which engages and redirects those host defenses. We suggest that OMV-C4'-MPLA triggers elevated IFNβ and IL-1β cytokines, which typify PD comorbidities, and drive PD-related alveolar bone loss.
Collapse
Affiliation(s)
- Stephen R Coats
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA, United States
| | - Thet Hnin Su
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA, United States
| | - Zoe Luderman Miller
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA, United States
| | - Alisa J King
- Sciences and Mathematics Division, School of Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, United States
| | - Joshua Ortiz
- Sciences and Mathematics Division, School of Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, United States
| | - Angel Reddy
- Sciences and Mathematics Division, School of Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, United States
| | - Sarah R Alaei
- Sciences and Mathematics Division, School of Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, United States
| | - Sumita Jain
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA, United States
| |
Collapse
|
10
|
Sainz-Mejías M, Ma C, Hou Y, Jurado-Martin I, Romerio A, Franco AR, Shaik MM, Tomás-Cortázar J, Peri F, McClean S. Monosaccharide-Based Synthetic TLR4 Agonist Enhances Vaccine Efficacy against Pseudomonas aeruginosa Challenge. ACS Infect Dis 2025; 11:894-904. [PMID: 40129118 PMCID: PMC11998000 DOI: 10.1021/acsinfecdis.4c00932] [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: 11/19/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025]
Abstract
Vaccine adjuvants are critical to improve the immunogenicity, efficacy, and durability of vaccines; however, their development has lagged behind that of vaccine antigens. Monophosphoryl lipid A (MPLA), a clinically approved adjuvant that stimulates Toll-like receptor 4 (TLR4), faces manufacturing challenges due to its complex and long synthesis. With the aim of simplifying the structure of MPLA while retaining its biological activity, we developed monosaccharide-based molecules FP18 and FP20Rha that activate TLR4 signaling. Both TLR4 agonists induced robust antibody activity against the model antigen, ovalbumin. Here, we report the potential of these TLR4 agonists to enhance the protective efficacy of the well-characterized OprF antigen against P. aeruginosa infection. OprF adjuvanted with FP18 showed reduced bacterial loads in lungs and spleens, relative to antigen alone in an acute P. aeruginosa pneumonia model. FP18-adjuvanted OprF also enhanced the production of anti-OprF antibodies and stimulated IFNγ and TNF in CD4+ T cells, suggesting a Th1-skewed cellular immune response. These adjuvants have promise for accelerating the development of effective vaccines against P. aeruginosa and other infectious diseases.
Collapse
Affiliation(s)
- Maite Sainz-Mejías
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Chaoying Ma
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Yueran Hou
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Irene Jurado-Martin
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Alessio Romerio
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, Milano 20126, Italy
| | - Ana Rita Franco
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, Milano 20126, Italy
| | - Mohammed Monsoor Shaik
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, Milano 20126, Italy
| | - Julen Tomás-Cortázar
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Francesco Peri
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, Milano 20126, Italy
| | - Siobhán McClean
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| |
Collapse
|
11
|
Li D, Li Y, Cang J, Yan X, Wu F, Sun X, Zhang W. Synergistic chemo-immunotherapy for osteosarcoma via a pH-responsive multi-component nanoparticle system. Front Pharmacol 2025; 16:1584245. [PMID: 40264674 PMCID: PMC12011790 DOI: 10.3389/fphar.2025.1584245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 03/26/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction Osteosarcoma (OS) is the most common primary malignant bone tumor in pediatric populations. Its treatment is complicated by chemotherapy-induced toxicity and limited induction of immunogenic cell death (ICD). Methods To address these challenges, we developed a pH-responsive, multi-component nanoparticle system designed to co-deliver doxorubicin (DOX), monophosphoryl lipid A (MPLA), and a PD-1/PD-L1-targeting peptide, integrated with the immune-modulating polymer PEG-PC7A. The system was optimized using both one-factor-at-a-time (OFAT) and Box-Behnken design (BBD). Results The optimized nanoparticles had a hydrodynamic size of 110 nm, high encapsulation efficiency (97.15%), and pH-sensitive drug release (91% at pH 6.5). In vitro studies showed enhanced ICD markers, including calreticulin exposure and ATP/HMGB1 release, aswell as synergistic dendritic cell maturation via dual STING/TLR4 pathway activation. In an orthotopic LM8 osteosarcoma model, the nanoparticles significantly suppressed tumor growth, promoted cytotoxic T lymphocyte infiltration, reduced regulatory T cells, and established long-term immune memory. Discussion The combination of ICD induction, innate immune activation, and checkpoint blockade reprogrammed the tumor microenvironment, amplifying anti-tumor immune responses. These results demonstrate the potential of this multifunctional nanoparticle platform as an effective immunochemotherapeutic strategy for osteosarcoma, offering enhanced therapeutic efficacy and reduced systemic toxicity.
Collapse
Affiliation(s)
- Dapeng Li
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yuanfan Li
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jie Cang
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xianwen Yan
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Feipeng Wu
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xuan Sun
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wenchao Zhang
- Department of Orthopedics, Affiliated Jintan Hospital of Jiangsu University, Jintan, Jiangsu, China
| |
Collapse
|
12
|
Chen Y, Feng D, Cheng Y, Jiang X, Qiu L, Zhang L, Shi D, Wang J. Research progress of metal-CpG composite nanoadjuvants in tumor immunotherapy. Biomater Sci 2025; 13:1605-1623. [PMID: 39998438 DOI: 10.1039/d4bm01399a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
The practical benefits and therapeutic potential of tumor vaccines in immunotherapy have drawn significant attention in the field of cancer treatment. Among the available vaccines, nanovaccines that utilize nanoparticles as carriers or adjuvants have demonstrated considerable effectiveness in combating cancer. Cytosine-phosphate-guanine oligodeoxynucleotide (CpG ODN), a common adjuvant in tumor nanovaccines, activates both humoral and cellular immunity by recognizing toll-like receptor 9 (TLR9), thereby aiding in the prevention and treatment of cancer. Metal nanoparticles hold great promise in tumor immunotherapy due to their adjustable size, surface functionalization, ability to regulate innate immunity, and capacity for controlled delivery of antigens or immunomodulators. Consequently, composite nanoadjuvants, formed by combining metal nanoparticles with CpG ODNs, can be customized to meet the specific performance requirements of different application scenarios, effectively overcoming the limitations of conventional immunotherapy approaches. This review provides a comprehensive analysis of the critical role of metal-CpG composite nanoadjuvants in advancing vaccine adjuvants for cancer therapy and prevention, highlighting their efficacy in preclinical settings.
Collapse
Affiliation(s)
- Yifan Chen
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Danna Feng
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Yilin Cheng
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Xianmeng Jiang
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| | - Li Zhang
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Dongjian Shi
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| |
Collapse
|
13
|
Tang Y, Tang T, Li Y, Wu J, Liu X, Xiang D, Hu X. Biomimetic Self-Oxygenated Immunoliposome for Cancer-Targeted Photodynamic Immunotherapy. Int J Nanomedicine 2025; 20:2743-2759. [PMID: 40066322 PMCID: PMC11892501 DOI: 10.2147/ijn.s508696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
Objective Photodynamic therapy (PDT) is a promising strategy with significant clinical application potential for tumor treatment. However, the tumor hypoxia and limited efficacy against tumor metastasis present significant limitations in the clinical application of PDT. To alleviate tumor hypoxia for PDT against tumor growth and metastasis, we developed a self-oxygenated immunoliposome by encapsulating the catalase (CAT) within the liposome cavity and loading the photosensitizer chlorin e6 (Ce6) and immunoadjuvant MPLA in the lipid bilayer of the immunoliposome (CAT@LP-Ce6-A). Subsequently, we fused it with the cancer cell membrane (CCM) to create the hybrid immunoliposome (CAT@LP-CCM-Ce6-A). The in vitro and in vivo anti-cancer efficacy of CAT@LP-CCM-Ce6-A-based photodynamic immunotherapy (PDIT) was evaluated. Methods CAT@LP-CCM-Ce6-A were characterized by size, zeta potential, transmission electron microscopy (TEM), Coomassie bright blue staining, UV spectrophotometer, and standard Goth's method. Cellular uptake, cell viability, reactive oxygen species (1O2) generation, calreticulin exposure, and ability to promote BMDCs maturation of CAT@LP-CCM-Ce6-A were evaluated in vitro. Biodistribution, anti-cancer therapeutic efficacy, and in vivo safety of CAT@LP-CCM-Ce6-A were investigated in orthotopic triple-negative breast cancer (TNBC) lung metastasis mouse models. Results CAT@LP-CCM-Ce6-A was successfully developed via the thin film hydration and co-extrusion method. The loading capacity of Ce6 and CAT was 4.7 ± 0.9% and 8.5 ± 0.9% respectively. CAT@LP-CCM-Ce6-A exhibited improved cellular uptake efficiency and cytotoxicity under laser irradiation against TNBC. Furthermore, CAT@LP-CCM-Ce6-A possessed enhanced anti-enzymatic degradation ability and promotion of DC maturation. In TNBC-bearing mice, CAT@LP-CCM-Ce6-A-based PDIT demonstrated remarkable therapeutic effect and antitumor immunity while maintaining minimal systemic toxicity. Conclusion CAT@LP-CCM-Ce6-A could be employed as an innovative approach for self-oxygenated photodynamic immunotherapy against cancer.
Collapse
Affiliation(s)
- Yucheng Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, People’s Republic of China
- Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Changsha, 410011, People’s Republic of China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, People’s Republic of China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, People’s Republic of China
- Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Changsha, 410011, People’s Republic of China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, People’s Republic of China
| | - Yongjiang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, People’s Republic of China
- Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Changsha, 410011, People’s Republic of China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, People’s Republic of China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, People’s Republic of China
- Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Changsha, 410011, People’s Republic of China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, People’s Republic of China
| | - Xinyi Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, People’s Republic of China
- Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Changsha, 410011, People’s Republic of China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, People’s Republic of China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, People’s Republic of China
- Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Changsha, 410011, People’s Republic of China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, People’s Republic of China
| | - Xiongbin Hu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, People’s Republic of China
- Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Changsha, 410011, People’s Republic of China
- Institute of Clinical Pharmacy, Central South University, Changsha, 410011, People’s Republic of China
| |
Collapse
|
14
|
Jorda A, Pracher L, Eberl S, Nussbaumer-Pröll A, Sarhan M, Weber M, Wahrmann M, Al Jalali V, Bergmann F, Prager M, Leutzendorff A, Sanz-Codina M, Tegrovsky L, Pecho T, Jilma B, Müller L, Spittler A, Rocha-Hasler M, Eckl-Dorna J, Kusienicka A, Farlik M, Zeitlinger M. Stability of cytokine, cellular and clinical response to the intravenous LPS challenge repeated after one year: a healthy volunteer trial. Med Microbiol Immunol 2025; 214:14. [PMID: 40047923 PMCID: PMC11885351 DOI: 10.1007/s00430-025-00823-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025]
Abstract
Whether the magnitude of individual cytokine, cellular, and clinical responses to the intravenous lipopolysaccharide (LPS) challenge is constant in individuals over extended time periods is unknown. Nine healthy volunteers received an intravenous LPS injection of 2 ng/kg bodyweight twice at intervals of at least one year. Circulating cytokines and leukocyte subsets were quantified using a multiplex immunoassay and cytometry by time-of-flight, respectively. Self-reported symptoms and vital signs were also assessed. We observed moderate to strong intra-individual correlations in the responsiveness of most cytokines (IL-6 [AUC0 - 10]: R = 0.93, p < 0.001; CRP [mg/dL]: R = 0.88, p = 0.004; IL-8 [AUC0 - 10]: R = 0.71, p = 0.031; TNF-alpha [AUC0 - 10]: R = 0.67, p = 0.047; IL-10 [AUC0 - 10]: R = 0.42, p = 0.26) and cellular subsets (CD8 T lymphocytes: R = 0.9, p = 0.002; B lymphocytes [G/L]: R = 0.89, p = 0.003; CD4 T lymphocytes: R = 0.84, p = 0.001; neutrophils: R = 0.80, p = 0.017; monocytes: R = 0.16, p = 0.710) between the 1st and 2nd LPS challenges. Vital signs and symptoms were not reproducible. While the average cellular and clinical response was similar between the two LPS challenges, we found a significantly attenuated AUC0 - 10 of IL-6 (percent difference, -41.9% [95% CI -73.0 - -10.7]) and TNF-alpha (percent difference, -35.7% [95% CI -70.0 - -1.6]) at the 2nd LPS challenge. Individual cytokine and cellular responses to intravenous LPS showed a significant degree of correlation when measured more than one year apart. These correlations did not translate to the reproducibility of clinical symptoms and vital signs, which showed greater variability and were not constant over time. The partly reduced cytokine release in the 2nd LPS challenge might be interpreted as an indicator of a long-lasting tolerance to endotoxin.
Collapse
Affiliation(s)
- Anselm Jorda
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Lena Pracher
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Sabine Eberl
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Alina Nussbaumer-Pröll
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Maysa Sarhan
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Vienna, Austria
| | - Maria Weber
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Markus Wahrmann
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Vienna, Austria
| | - Valentin Al Jalali
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Felix Bergmann
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Marlene Prager
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Amelie Leutzendorff
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine III, Vienna, Austria
| | - Maria Sanz-Codina
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Lara Tegrovsky
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Theresa Pecho
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Lena Müller
- Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Andreas Spittler
- Research Laboratories, Core Facility Flow Cytometry & Department of Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Julia Eckl-Dorna
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Anna Kusienicka
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria.
| |
Collapse
|
15
|
Chang J, Shin K, Lewis JM, Suh HW, Lee J, Damsky W, Xu S, Bosenberg M, Saltzman WM, Girardi M. Enhanced Intratumoral Delivery of Immunomodulator Monophosphoryl Lipid A through Hyperbranched Polyglycerol-Coated Biodegradable Nanoparticles. J Invest Dermatol 2025; 145:593-604. [PMID: 39122142 DOI: 10.1016/j.jid.2024.07.019] [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: 03/12/2024] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
Immunomodulatory agents have significant potential to enhance cancer treatment but have demonstrated limited efficacy beyond the preclinical setting owing to poor pharmacokinetics and toxicity associated with systemic administration. Conversely, when locally delivered, immunomodulatory agents require repeated administration to optimize immune stimulation. To overcome these challenges, we encapsulated the toll-like receptor 4 agonist monophosphoryl lipid A (MPLA) within hyperbranched polyglycerol-coated biodegradable nanoparticles (NPs) engineered for gradual drug release from the NP core, resulting in a more persistent stimulation of antitumor immune responses while minimizing systemic side effects. In a model of malignant melanoma, we demonstrate that hyperbranched polyglycerol-NP encapsulation significantly improves the antitumor efficacy of MPLA by enhancing its ability to remodel the tumor microenvironment. Relative to free MPLA, hyperbranched polyglycerol-coated NP-encapsulated MPLA significantly increased the NK cell- and cytotoxic T-cell-mediated antitumor immune response and tuned the tumor-draining lymph nodes toward a T helper 1 response. Furthermore, when combined with local delivery of a chemotherapeutic agent, hyperbranched polyglycerol-NP-MPLA induces the conversion of an immunosuppressive tumor microenvironment to immunogenic tumor microenvironment and significantly improves survival.
Collapse
Affiliation(s)
- Jungsoo Chang
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Kwangsoo Shin
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Julia M Lewis
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Hee Won Suh
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA
| | - Joohyung Lee
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - William Damsky
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Suzanne Xu
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Marcus Bosenberg
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, Connecticut, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA; Yale Center for Immuno-Oncology, Yale School of Medicine, New Haven, Connecticut, USA
| | - W Mark Saltzman
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA; Department of Chemical & Environmental Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michael Girardi
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
16
|
Islam EA, Fegan JE, Zeppa JJ, Ahn SK, Ng D, Currie EG, Lam J, Moraes TF, Gray-Owen SD. Adjuvant-dependent impacts on vaccine-induced humoral responses and protection in preclinical models of nasal and genital colonization by pathogenic Neisseria. Vaccine 2025; 48:126709. [PMID: 39817984 DOI: 10.1016/j.vaccine.2025.126709] [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: 09/07/2024] [Revised: 11/05/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
Neisseria gonorrhoeae, which causes the sexually transmitted infection gonorrhea and Neisseria meningitidis, a leading cause of bacterial meningitis and septicemia, are closely related human-restricted pathogens that inhabit distinct primary mucosal niches. While successful vaccines against invasive meningococcal disease have been available for decades, the rapid rise in antibiotic resistance has led to an urgent need to develop an effective gonococcal vaccine. Several surface antigens are shared among these two pathogens, making cross-species protection an exciting prospect. However, the type of vaccine-mediated immune response required to achieve protection against respiratory versus genital infection remains ill defined. In this study, we utilize well established mouse models of female lower genital tract colonization by N. gonorrhoeae and upper respiratory tract colonization by N. meningitidis to examine the performance of transferrin binding protein B (TbpB) vaccines formulated with immunologically distinct vaccine adjuvants. We demonstrate that vaccine-mediated protection is influenced by the choice of adjuvant, with Th1/2-balanced adjuvants performing optimally against N. gonorrhoeae, and both Th1/2-balanced and Th2-skewing adjuvants leading to a significant reduction in N. meningitidis burden. We further establish a lack of correlation between protection status and the humoral response or bactericidal titre. Combined, this work supports the feasibility for a single vaccine formulation to achieve pan-neisserial coverage.
Collapse
Affiliation(s)
- Epshita A Islam
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Jamie E Fegan
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Joseph J Zeppa
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Sang Kyun Ahn
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Dixon Ng
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Elissa G Currie
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Jessica Lam
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Trevor F Moraes
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Scott D Gray-Owen
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Canada.
| |
Collapse
|
17
|
Xia W, Tan Y, Liu Y, Xie N, Zhu H. Prospect of extracellular vesicles in tumor immunotherapy. Front Immunol 2025; 16:1525052. [PMID: 40078996 PMCID: PMC11897508 DOI: 10.3389/fimmu.2025.1525052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/28/2025] [Indexed: 03/14/2025] Open
Abstract
Extracellular vesicles (EVs), as cell-derived small vesicles, facilitate intercellular communication within the tumor microenvironment (TME) by transporting biomolecules. EVs from different sources have varied contents, demonstrating differentiated functions that can either promote or inhibit cancer progression. Thus, regulating the formation, secretion, and intake of EVs becomes a new strategy for cancer intervention. Advancements in EV isolation techniques have spurred interest in EV-based therapies, particularly for tumor immunotherapy. This review explores the multifaceted functions of EVs from various sources in tumor immunotherapy, highlighting their potential in cancer vaccines and adoptive cell therapy. Furthermore, we explore the potential of EVs as nanoparticle delivery systems in tumor immunotherapy. Finally, we discuss the current state of EVs in clinical settings and future directions, aiming to provide crucial information to advance the development and clinical application of EVs for cancer treatment.
Collapse
Affiliation(s)
- Wenbo Xia
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yunhan Tan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yongen Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Huili Zhu
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Manivannan K, Fathy Mohamed Y, Fernandez RC. Determining the Bordetella LPS structural features that influence TLR4 downstream signaling. Front Microbiol 2025; 16:1540534. [PMID: 40071202 PMCID: PMC11895810 DOI: 10.3389/fmicb.2025.1540534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
Upon recognizing bacterial lipopolysaccharide (LPS), human TLR4 initiates two distinct signaling pathways: the MyD88 pathway from the cell surface or the TRIF pathway following endocytosis. While the first is associated with strong pro-inflammatory responses, the latter is linked to dendritic cell maturation and T cell priming. Changes in LPS structure can influence the activation of either or both pathways. This study investigates the influence of specific structural features of Bordetella LPS on these pathways: the O antigen, the number of acyl chains in lipid A and the glucosamine modification of the phosphates of the lipid A diglucosamine backbone. Systematically engineered Bordetella LPS differing in one or more of these features were studied by quantifying NFκB and IRF3 activation-indicators of MyD88 and TRIF pathway activation, respectively. The findings reveal that the glucosamine modification of lipid A plays a dominant role in TLR4-mediated signaling, overriding the influence of the O antigen and lipid A acylation. The absence of glucosamine modification significantly reduced the activation of both MyD88 and TRIF pathways, underscoring its importance in promoting TLR4 dimerization. Furthermore, under-acylation of LPS (with 4 or 5 acyl chains) partially reduced NFκB activation, while completely abrogating TRIF pathway activation. In contrast, hexa-and hepta-acylated LPS equally and robustly activated both pathways. Lastly, the Bordetella O antigen selectively biased signaling towards the TRIF pathway without affecting the MyD88 pathway. This study provides valuable insights into how specific LPS structural modifications can be leveraged to tailor TLR4-mediated signaling.
Collapse
Affiliation(s)
- Kiruthika Manivannan
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Yasmine Fathy Mohamed
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Rachel C. Fernandez
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
19
|
Franco AR, Aladailleh Z, Romerio A, Italia A, Lami F, Shaik MM, Skupinska N, Artusa V, Pirianov G, Peri F. Towards more efficient synthetic immunomodulators: biological characterization and mechanism of action of monosaccharide-derived TLR4 agonists. RSC Med Chem 2025:d4md00950a. [PMID: 40124897 PMCID: PMC11926738 DOI: 10.1039/d4md00950a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/15/2025] [Indexed: 03/25/2025] Open
Abstract
Toll-like receptors (TLRs), including TLR4, play a crucial role in innate immunity activation, and small molecular TLR4 activators (agonists) are in the preclinical and clinical phases of development as vaccine adjuvants or tumor immunotherapeutics. Recently, we generated novel glucosamine-derived compounds, FP molecules, that are active as TLR4 agonists. Despite their chemical structure differing from LPS, some of these compounds, including compound FP18, mimicked the biological activity of LPS and its capacity to activate TLR4 and its downstream pathways. In contrast to FP18, compound FP20 showed immunostimulant activity that was only partially due to TLR4 agonism. This activity was mainly associated with NLRP3 inflammasome activation. We generated a panel of glycosylated FP20 derivatives (glyco-FP20) bearing different monosaccharides linked to C6 of the glucosamine. The biological activity of glyco-FP20 was related to TLR4 activation, as assessed from preliminary experiments in HEK-Blue cells. We presented a comprehensive study of the mechanism of action of glyco-FP20 derivatives and their effect on TLR4 signalling, leading to macrophage M1 polarisation and pyroptosis in THP-1 derived macrophages. Results revealed that, similarly to LPS and differently from FP20, glyco-FP20 derivatives were potent TLR4 agonists inducing TLR4/MyD88 signalling pathways that led to M1 macrophage polarisation, associated with NF-kB activation/translocation and release of a number of proinflammatory mediators in THP-1-derived macrophages. In particular, compound FP20 Rha activated TLR4/TRIF signalling, associated with phosphorylation of STAT1/IRF3, leading to the production of IFN-β in THP-1-derived macrophages. Furthermore, using a specific GSD inhibitor (U73), we demonstrated the ability of FP20 and glyco-FP20 to induce GSD-dependent pyroptosis, which was associated with IL-1β/IL-18 and LDH release in THP-1-derived macrophages. These results show that the optimization of FP20 glycosylation can increase the biological potency of the parent molecule and can be used in preclinical development as vaccine adjuvants or macrophage-based cancer immunotherapy.
Collapse
Affiliation(s)
- Ana Rita Franco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Piazza della Scienza, 2 20154 Milano (MI) Italy
| | - Zaineh Aladailleh
- School of Life Sciences, Anglia Ruskin University East Road Cambridge CB1 1PT UK
| | - Alessio Romerio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Piazza della Scienza, 2 20154 Milano (MI) Italy
| | - Alice Italia
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Piazza della Scienza, 2 20154 Milano (MI) Italy
| | - Federico Lami
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Piazza della Scienza, 2 20154 Milano (MI) Italy
| | - Mohammed Monsoor Shaik
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Piazza della Scienza, 2 20154 Milano (MI) Italy
| | - Natalia Skupinska
- School of Life Sciences, Anglia Ruskin University East Road Cambridge CB1 1PT UK
| | - Valentina Artusa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Piazza della Scienza, 2 20154 Milano (MI) Italy
| | - Grisha Pirianov
- School of Life Sciences, Anglia Ruskin University East Road Cambridge CB1 1PT UK
| | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Piazza della Scienza, 2 20154 Milano (MI) Italy
| |
Collapse
|
20
|
Li N, Qin H, Zhu F, Ding H, Chen Y, Lin Y, Deng R, Ma T, Lv Y, Xiong C, Li R, Wei Y, Shi J, Chen H, Zhao Y, Zhou G, Guo H, Lv M, Lin Y, Han B, Nie G, Zhao R. Potent prophylactic cancer vaccines harnessing surface antigens shared by tumour cells and induced pluripotent stem cells. Nat Biomed Eng 2025; 9:215-233. [PMID: 39730914 DOI: 10.1038/s41551-024-01309-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 11/01/2024] [Indexed: 12/29/2024]
Abstract
The development of prophylactic cancer vaccines typically involves the selection of combinations of tumour-associated antigens, tumour-specific antigens and neoantigens. Here we show that membranes from induced pluripotent stem cells can serve as a tumour-antigen pool, and that a nanoparticle vaccine consisting of self-assembled commercial adjuvants wrapped by such membranes robustly stimulated innate immunity, evaded antigen-specific tolerance and activated B-cell and T-cell responses, which were mediated by epitopes from the abundant number of antigens shared between the membranes of tumour cells and pluripotent stem cells. In mice, the vaccine elicited systemic antitumour memory T-cell and B-cell responses as well as tumour-specific immune responses after a tumour challenge, and inhibited the progression of melanoma, colon cancer, breast cancer and post-operative lung metastases. Harnessing antigens shared by pluripotent stem cell membranes and tumour membranes may facilitate the development of universal cancer vaccines.
Collapse
Affiliation(s)
- Nan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Hao Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Fei Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Hao Ding
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Yang Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Yixuan Lin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Ronghui Deng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Tianyu Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Yuanyuan Lv
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Changhao Xiong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Rong Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Yaohua Wei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Hanqing Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing, P. R. China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, P. R. China
| | - Guangbiao Zhou
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Hua Guo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Mengyao Lv
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Yongfang Lin
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Bing Han
- Department of Orthodontics, Cranial-Facial Growth and Development Center, Peking University School and Hospital of Stomatology, Beijing, P. R. China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China.
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China.
| |
Collapse
|
21
|
Matthews RL, Khan N, Beckman B, Sharma S, Dietz Z, Picking WD, Izmirlian G, Sanders C, Stocks SM, Difilippantonio S, Kirnbauer R, Roden RB, Pinto LA, Shoemaker RH, Ernst RK, Marshall JD. Immune profile diversity is achieved with synthetic TLR4 agonists combined with the RG1-VLP vaccine in mice. Vaccine 2025; 44:126577. [PMID: 39632208 DOI: 10.1016/j.vaccine.2024.126577] [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: 03/25/2024] [Revised: 11/04/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
The TLR4 (Toll-like receptor 4)-activating agonist MPLA (monophosphoryl lipid A) is a key component of the adjuvant systems AS01 and AS04, utilized in marketed preventive vaccines for several infectious pathogens. As MPLA is a biologically-derived product containing a mixture of several lipid A congeners with a 4' phosphoryl group and varying numbers of acyl chains with distinct activities, extensive efforts to refine its production and immunogenicity are ongoing; notably, the development of the BECC (Bacterial Enzymatic Combinatorial Chemistry) system in which bacteria express lipid A-modifying enzymes to produce a panoply of lipid A congeners. In an effort to characterize the adjuvant activity of these lipid A congeners, we compared biologically-derived and synthetic versions of BECC470 and BECC438 for adjuvant activity in BALB/c mice vaccinated with the HPV (Human papilloma virus) VLP-based vaccine, RG1-VLP. Synthetic BECC compounds compared favorably to biological versions and, in the case of synthetic BECC470, were routinely superior to their biologically-derived BECC counterpart. Synthetic BECC470-adjuvanted vaccines achieved broad spectrum immune activity characterized by elevated levels of total IgG and IgG2a subtype specific to HPV16 L1 VLPs and the HPV16 L2 peptide, as well as robust HPV16-neutralizing antibody titers. In addition, synthetic BECC470 promoted strong T cell responses to HPV16 L1, increased memory B cell frequency, and increased the T follicular helper cell (Tfh) population in draining lymph nodes. In contrast, the biologically-derived form of BECC470 induced an immune profile specific for highest levels of HPV16 L2-specific IgG2a as well as antibodies cross-neutralizing to HPV18 and HPV39. These data confirm that a synthetically-derived BECC compound can be combined with Alhydrogel to adjuvant the RG1-VLP vaccine as can biologically-derived BECC compounds and MPLA, albeit with subtly distinct immune responses. The distinctions in immune profiles triggered by these BECC compounds warrant further exploration for their capacity to activate TLR4 and modulate immune responses to vaccines.
Collapse
Affiliation(s)
- Rebecca L Matthews
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Nazneen Khan
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Bradley Beckman
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Simran Sharma
- Department of Veterinary Pathobiology and Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Zackary Dietz
- Department of Veterinary Pathobiology and Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - William D Picking
- Department of Veterinary Pathobiology and Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Grant Izmirlian
- Biometry Research Group, Division of Cancer Prevention, NCI, Bethesda, MD, USA
| | - Chelsea Sanders
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Stacy M Stocks
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Reinhard Kirnbauer
- Laboratory of Viral Oncology (LVO), Department of Dermatology, Medical University of Vienna, Austria
| | - Richard B Roden
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Ligia A Pinto
- Vaccine, Immunity, and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Robert H Shoemaker
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, NCI, Bethesda, MD, USA
| | - Robert K Ernst
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Jason D Marshall
- Cancer ImmunoPrevention Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| |
Collapse
|
22
|
Zúquete S, Ferreira M, Delgado IL, Gazalle P, Andaluz S, Rosa MT, Mendes AC, Santos D, Nolasco S, Graça L, Leitão A, Basto AP. Combined TLR2/TLR4 activation equip non-mucosal dendritic cells to prime Th1 cells with gut tropism. iScience 2024; 27:111232. [PMID: 39759015 PMCID: PMC11700634 DOI: 10.1016/j.isci.2024.111232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/03/2024] [Accepted: 10/21/2024] [Indexed: 01/07/2025] Open
Abstract
Activated CD4+ T cells located at mucosal surfaces orchestrate local effector immune mechanisms. When properly polarized, these cells contribute to block infections at early stages and may be essential to restrain the local growth of mucosal tumors, playing a critical role in host protection. How CD4+ T cells simultaneously integrate gut-homing instructions and Th polarization signals transmitted by TLR activated dendritic cells (DCs) is unknown. Here, we show that the combined activation through TLR2, which alone does not induce a clear Th polarization, and TLR4, which alone does not imprint mucosal tropism, equip non-mucosal DCs to prime gut-homing CD4+ T cells with reinforced Th1 polarization. These results show that targeting DCs with combined innate stimuli with distinct properties is a rational strategy to program the outcome of T cell polarization and simultaneously control their tissue tropism. Exploring this strategy could enhance the efficacy of vaccines and immune cell therapies.
Collapse
Affiliation(s)
- Sara Zúquete
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Mariana Ferreira
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Inês L.S. Delgado
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), 1300-477 Lisboa, Portugal
- Faculdade de Medicina Veterinária, Universidade Lusófona – Centro Universitário de Lisboa, 1749-024 Lisboa, Portugal
| | - Paula Gazalle
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), 1300-477 Lisboa, Portugal
- Faculdade de Veterinária, Universidade Federal de Pelotas, Capão do Leão CEP 96160-000, Brazil
| | - Stephanie Andaluz
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal
- Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Maria Teresa Rosa
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal
- Faculdade de Ciência e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Ana Catarina Mendes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto Gulbenkian da Ciência, 2780-156 Oeiras, Portugal
| | - Dulce Santos
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal
| | - Sofia Nolasco
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), 1300-477 Lisboa, Portugal
- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal
| | - Luís Graça
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Instituto Gulbenkian da Ciência, 2780-156 Oeiras, Portugal
| | - Alexandre Leitão
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Afonso P. Basto
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), 1300-477 Lisboa, Portugal
| |
Collapse
|
23
|
Luna E, Ruiz S, Garinot M, Chavagnac C, Agrawal P, Escobar J, Revet L, Asensio MJ, Piras F, Fang FG, Drake DR, Rokbi B, Larocque D, Haensler J. SPA14 liposomes combining saponin with fully synthetic TLR4 agonist provide adjuvanticity to hCMV vaccine candidate. NPJ Vaccines 2024; 9:253. [PMID: 39702373 PMCID: PMC11659416 DOI: 10.1038/s41541-024-01046-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024] Open
Abstract
In the aim of designing and developing a novel saponin-based adjuvant system, we combined the QS21 saponin with low microgram amounts of the fully synthetic TLR4 agonist, E6020, in cholesterol-containing liposomes. The resulting adjuvant system, termed SPA14, appeared as a long-term stable and homogeneous suspension of mostly unilamellar and a few multilamellar vesicles, with an average hydrodynamic diameter of 93 nm, when formulated in citrate buffer at pH 6.0-6.5. When compared in an in vitro human innate immunity construct to AS01B, the QS21/MPL® liposomal adjuvant system of GSK, and with QS21-Liposomes used as benchmarks, SPA14 displayed the strongest immunostimulatory potential based on antigen-presenting cell (APC) activation and cytokine secretion, which was essentially driven by the highly active E6020 agonist in this assay. When tested as an adjuvant in vivo with human cytomegalovirus glycoprotein B (gB) and pentamer complex (PC) as test antigens, SPA14 was generally well tolerated and as active as AS01B for the induction of long-lasting CMV-neutralizing antibodies in mice and non-human primates (NHPs). Both adjuvants promoted the induction of Th-1 responses based on IgG2c production in mice and IFN-γ production in mice and NHPs, but in mice, a higher level of Th-2 cytokines (IL-5) and higher IgG1 over IgG2c secreting cells ratios were obtained with SPA14 indicating that the adjuvant profile of SPA14 could be less Th-1 biased than that of AS01B. From a developability standpoint, SPA14 could be manufactured by a simple and scalable ethanol injection method, owing to the high solubility in ethanol of all its lipidic components, including E6020. Furthermore, E6020 is a single molecule, well-characterized fully synthetic TLR4 agonist constructed in eight synthetic steps from entirely crystalline starting materials and intermediates via an optimized high-yield synthetic route. Overall, our data suggest that SPA14 is a viable, easy-to-manufacture, potent novel adjuvant system that could be broadly applicable as a ready-to-mix adjuvant in the form of a long-term stable liquid formulation.
Collapse
Affiliation(s)
- Ernesto Luna
- Sanofi Vaccines business unit, R&D, Orlando, FL, USA
| | - Sophie Ruiz
- Sanofi Vaccines business unit, R&D, Marcy L'Etoile, France
| | - Marie Garinot
- Sanofi Vaccines business unit, R&D, Marcy L'Etoile, France
| | | | | | - John Escobar
- Sanofi Vaccines business unit, R&D, Orlando, FL, USA
| | - Laurent Revet
- Sanofi Vaccines business unit, R&D, Marcy L'Etoile, France
| | | | - Fabienne Piras
- Sanofi Vaccines business unit, R&D, Marcy L'Etoile, France
| | | | | | - Bachra Rokbi
- Sanofi Vaccines business unit, R&D, Marcy L'Etoile, France
| | | | - Jean Haensler
- Sanofi Vaccines business unit, R&D, Marcy L'Etoile, France.
| |
Collapse
|
24
|
Zhao Z, Jia H, Sun Z, Li Y, Liu L. A new perspective on macrophage-targeted drug research: the potential of KDELR2 in bladder cancer immunotherapy. Front Immunol 2024; 15:1485109. [PMID: 39691708 PMCID: PMC11649672 DOI: 10.3389/fimmu.2024.1485109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/04/2024] [Indexed: 12/19/2024] Open
Abstract
Introduction Bladder cancer was recognized as one of the most common malignant tumors in the urinary system, and treatment options remained largely limited to conventional surgery, radiotherapy, and chemotherapy, which limited patient benefits. Methods Researchers constructed an RNA transcriptome map of bladder cancer by integrating single-cell RNA sequencing and clinical data, identifying potential molecular targets for diagnosis and treatment. We also verified the antitumor activity of the target through in vitro experiment. Results A distinct tumor cell subpopulation characterized by elevated S100A8 expression exhibited high copy number variation, high stemness, and low differentiation. It interacted with myeloid cells via the MIF-(CD74+CD44) and MIF-(CD74+CXCR4) signaling pathways. This study underscored KDELR2's role in promoting cell proliferation, invasion, and migration, providing new therapeutic insights. Prognostic analysis revealed that KDELR2 correlated with poor survival, higher immune scores, and increased macrophage infiltration. Discussion The findings suggested that patients with high KDELR2 expression might benefit from immune checkpoint therapy. KDELR2 was also shown to enhance bladder cancer cell proliferation, invasion, and migration, highlighting it as a promising target for macrophage-focused drug development.
Collapse
Affiliation(s)
- Zhiyi Zhao
- Department of Andrology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongling Jia
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhou Sun
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Yumeng Li
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lingyun Liu
- Department of Andrology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
25
|
Zhang W, Park HB, Yadav D, An EK, Kim SJ, Ryu D, Agrawal R, Ryu JH, Kwak M, Lee PCW, Jin JO. P-type pilus PapG protein elicits toll-like receptor 2-mediated immune activation during cancer immunotherapy. Int J Biol Macromol 2024; 282:137061. [PMID: 39481736 DOI: 10.1016/j.ijbiomac.2024.137061] [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: 06/12/2024] [Revised: 10/14/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
The immune activation ability of FimH, an adhesion protein in pili of Escherichia coli (E. coli), has been recently reported. However, studies on the immune activity of PapG, another major pili terminal protein, have not been well explored. In this study, the immune stimulatory effect of purified recombinant PapG was evaluated. PapG treatment promoted dramatic changes in dendritic morphology of the bone marrow-derived dendritic cells (BMDCs) and induced upregulation of co-stimulatory molecule levels, major histocompatibility complex (MHC) I and II expression, and pro-inflammatory cytokine production in BMDCs. To identify the stimulatory receptor of PapG, an in silico study was performed. PapG exhibited strong binding affinity with murine toll-like receptor 2 (TLR2). In addition, PapG-induced activation of splenic DC and its subsets was unsuccessful in TLR2-knock out mice. Combination of PapG and ovalbumin (OVA) elicited OVA-specific T cell proliferation and cytokine production and cytotoxicity that consequently promoted anti-cancer immune responses against OVA-expressing B16 melanoma. Furthermore, PapG treatment induced activation of peripheral blood DCs and its subsets in humans in a TLR2 dependent manner. PapG-stimulated human conventional DC2 promoted syngeneic T cell proliferation and activation. The findings of this study demonstrated that PapG could be a useful immune stimulator for immunotherapy against cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Hae-Bin Park
- Department of Microbiology, Brain Korea 21 project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Dhananjay Yadav
- Department of Life Science, Yeungnam University, Gyeongsan 38541, South Korea; USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, Florida 33613, USA
| | - Eun-Koung An
- Department of Microbiology, Brain Korea 21 project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - So-Jung Kim
- Department of Microbiology, Brain Korea 21 project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Dayoung Ryu
- Department of Microbiology, Brain Korea 21 project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea
| | - Richa Agrawal
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, The University of Chicago, 929 E 57(th) street, Chicago, IL 60637, USA
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
| | - Minseok Kwak
- Department of Chemistry, Pukyong National University, Busan 48513, South Korea
| | - Peter C W Lee
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea.
| | - Jun-O Jin
- Department of Microbiology, Brain Korea 21 project, University of Ulsan College of Medicine, ASAN Medical Center, Seoul 05505, South Korea.
| |
Collapse
|
26
|
Holze J, Lauber F, Soler S, Kostenis E, Weindl G. Label-free biosensor assay decodes the dynamics of Toll-like receptor signaling. Nat Commun 2024; 15:9554. [PMID: 39532846 PMCID: PMC11558003 DOI: 10.1038/s41467-024-53770-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The discovery of Toll-like receptors (TLRs) represented a significant breakthrough that paved the way for the study of host-pathogen interactions in innate immunity. However, there are still major gaps in understanding TLR function, especially regarding the early dynamics of downstream TLR pathways. Here, we present a label-free optical biosensor-based assay as a method for detecting TLR activation in a native and label-free environment and defining the dynamics of TLR pathway activation. This technology is sufficiently sensitive to detect TLR signaling and readily discriminates between different TLR signaling pathways. We define pharmacological modulators of cell surface and endosomal TLRs and downstream signaling molecules and uncover TLR signaling signatures, including potential biased receptor signaling. These findings highlight that optical biosensor assays complement traditional assays that use a single endpoint and have the potential to facilitate the future design of selective drugs targeting TLRs and their downstream effector cascades.
Collapse
Affiliation(s)
- Janine Holze
- Pharmaceutical Institute, Section Pharmacology and Toxicology, University of Bonn, Bonn, Germany
| | - Felicitas Lauber
- Pharmaceutical Institute, Section Pharmacology and Toxicology, University of Bonn, Bonn, Germany
| | - Sofía Soler
- Institute of Experimental Haematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Evi Kostenis
- Institute for Pharmaceutical Biology, Molecular, Cellular and Pharmacobiology Section, University of Bonn, Bonn, Germany
| | - Günther Weindl
- Pharmaceutical Institute, Section Pharmacology and Toxicology, University of Bonn, Bonn, Germany.
| |
Collapse
|
27
|
Strobl S, Zucchetta D, Vašíček T, Monti A, Ruda A, Widmalm G, Heine H, Zamyatina A. Nonreducing Sugar Scaffold Enables the Development of Immunomodulatory TLR4-specific LPS Mimetics with Picomolar Potency. Angew Chem Int Ed Engl 2024; 63:e202408421. [PMID: 38870340 DOI: 10.1002/anie.202408421] [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: 05/03/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/15/2024]
Abstract
Innate immune defense mechanisms against infection and cancer encompass the modulation of pattern recognition receptor (PRR)-mediated inflammation, including upregulation of various transcription factors and the activation of pro-inflammatory pathways important for immune surveillance. Dysfunction of PRRs-mediated signaling has been implicated in cancer and autoimmune diseases, while the overactivation of PRRs-driven responses during infection can lead to devastating consequences such as acute lung injury or sepsis. We used crystal structure-based design to develop immunomodulatory lipopolysaccharide (LPS) mimetics targeting one of the ubiquitous PRRs, Toll-like Receptor 4 (TLR4). Taking advantage of an exo-anomeric conformation and specific molecular shape of synthetic nonreducing β,β-diglucosamine, which was investigated by NMR, we developed two sets of lipid A mimicking glycolipids capable of either potently activating innate immune responses or inhibiting pro-inflammatory signaling. Stereoselective 1,1'-glycosylation towards fully orthogonally protected nonreducing GlcNβ(1↔1')βGlcN followed by stepwise assembly of differently functionalised phosphorylated glycolipids provided biologically active molecules that were evaluated for their ability to trigger or to inhibit cellular innate immune responses. Two LPS mimetics, identified as potent TLR4-specific inducers of the intracellular signaling pathways, serve as vaccine adjuvant- and immunotherapy candidates, while anionic glycolipids with TLR4-inhibitory potential hold therapeutic promise for the management of acute or chronic inflammation.
Collapse
Affiliation(s)
- Sebastian Strobl
- Department of Chemistry, BOKU University, Muthgasse 18, Vienna, A-1190, Austria
| | - Daniele Zucchetta
- Department of Chemistry, BOKU University, Muthgasse 18, Vienna, A-1190, Austria
| | - Tomáš Vašíček
- Department of Chemistry, BOKU University, Muthgasse 18, Vienna, A-1190, Austria
| | - Alessandro Monti
- Department of Chemistry, BOKU University, Muthgasse 18, Vienna, A-1190, Austria
| | - Alessandro Ruda
- Department of Organic Chemistry, Stockholm University, S-106 91, Stockholm, Sweden
| | - Göran Widmalm
- Department of Organic Chemistry, Stockholm University, S-106 91, Stockholm, Sweden
| | - Holger Heine
- Research Group Innate Immunity, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Parkallee 22, Borstel, 23845, Germany
| | - Alla Zamyatina
- Department of Chemistry, BOKU University, Muthgasse 18, Vienna, A-1190, Austria
| |
Collapse
|
28
|
Fathy Mohamed Y, Fernandez RC. Programming Bordetella pertussis lipid A to promote adjuvanticity. Microb Cell Fact 2024; 23:250. [PMID: 39272136 PMCID: PMC11401268 DOI: 10.1186/s12934-024-02518-7] [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: 06/18/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Bordetella pertussis is the causative agent of whooping cough or pertussis. Although both acellular (aP) and whole-cell pertussis (wP) vaccines protect against disease, the wP vaccine, which is highly reactogenic, is better at preventing colonization and transmission. Reactogenicity is mainly attributed to the lipid A moiety of B. pertussis lipooligosaccharide (LOS). Within LOS, lipid A acts as a hydrophobic anchor, engaging with TLR4-MD2 on host immune cells to initiate both MyD88-dependent and TRIF-dependent pathways, thereby influencing adaptive immune responses. Lipid A variants, such as monophosphoryl lipid A (MPLA) can also act as adjuvants. Adjuvants may overcome the shortcomings of aP vaccines. RESULTS This work used lipid A modifying enzymes from other bacteria to produce an MPLA-like adjuvant strain in B. pertussis. We created B. pertussis strains with distinct lipid A modifications, which were validated using MALDI-TOF. We engineered a hexa-acylated monophosphorylated lipid A that markedly decreased human TLR4 activation and activated the TRIF pathway. The modified lipooligosaccharide (LOS) promoted IRF3 phosphorylation and type I interferon production, similar to MPLA responses. We generated three other variants with increased adjuvanticity properties and reduced endotoxicity. Pyrogenicity studies using the Monocyte Activation Test (MAT) revealed that these four lipid A variants significantly decreased the IL-6, a marker for fever, response in peripheral blood mononuclear cells (PBMCs). CONCLUSION These findings pave the way for developing wP vaccines that are possibly less reactogenic and designing adaptable adjuvants for current vaccine formulations, advancing more effective immunization strategies against pertussis.
Collapse
Affiliation(s)
- Yasmine Fathy Mohamed
- Department of Microbiology & Immunology, The University of British Columbia, Vancouver, British Columbia, V6T1Z3, Canada
- Department of Microbiology & Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Rachel C Fernandez
- Department of Microbiology & Immunology, The University of British Columbia, Vancouver, British Columbia, V6T1Z3, Canada.
| |
Collapse
|
29
|
Zhang S, Yu B, Sheng C, Yao C, Liu Y, Wang J, Zeng Q, Mao Y, Bei J, Zhu B, Chen S. SHISA3 Reprograms Tumor-Associated Macrophages Toward an Antitumoral Phenotype and Enhances Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403019. [PMID: 39054639 PMCID: PMC11423144 DOI: 10.1002/advs.202403019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/22/2024] [Indexed: 07/27/2024]
Abstract
The main challenge for immune checkpoint blockade (ICB) therapy lies in immunosuppressive tumor microenvironment (TME). Repolarizing M2-like tumor-associated macrophages (TAMs) into inflammatory M1 phenotype is a promising strategy for cancer immunotherapy. Here, this study shows that the tumor suppressive protein SHISA3 regulates the antitumor functions of TAMs. Local delivery of mRNA encoding Shisa3 enables cancer immunotherapy by reprogramming TAMs toward an antitumoral phenotype, thus enhancing the efficacy of programmed cell death 1 (PD-1) antibody. Enforced expression of Shisa3 in TAMs increases their phagocytosis and antigen presentation abilities and promotes CD8+ T cell-mediated antitumor immunity. The expression of SHISA3 is induced by damage/pathogen-associated molecular patterns (DAMPs/PAMPs) in macrophages via nuclear factor-κB (NF-κB) transcription factors. Reciprocally, SHISA3 forms a complex with heat shock protein family A member 8 (HSPA8) to activate NF-κB signaling thus maintaining M1 polarization of macrophages. Knockout Shisa3 largely abolishes the antitumor efficacy of combination immunotherapy with Toll-like receptor 4 (TLR4) agonist monophosphoryl lipid A (MPLA) and PD-1 antibody. It further found that higher expression of SHISA3 in antitumoral TAMs is associated with better overall survival in lung cancer patients. Taken together, the findings describe the role of SHISA3 in reprogramming TAMs that ameliorate cancer immunotherapy.
Collapse
Affiliation(s)
- Shimeng Zhang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Bingbing Yu
- Key Laboratory of Molecular Biophysicsthe Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanHubei430074P. R. China
| | - Chunjie Sheng
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Chen Yao
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Yang Liu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Jing Wang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Qi Zeng
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Yizhi Mao
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Jinxin Bei
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Bin Zhu
- Key Laboratory of Molecular Biophysicsthe Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanHubei430074P. R. China
- Shenzhen Huazhong University of Science and Technology Research InstituteShenzhen518063P. R. China
| | - Shuai Chen
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| |
Collapse
|
30
|
Wang Z, Sha T, Li J, Luo H, Liu A, Liang H, Qiang J, Li L, Whittaker AK, Yang B, Sun H, Shi C, Lin Q. Turning foes to friends: Advanced " in situ nanovaccine" with dual immunoregulation for enhanced immunotherapy of metastatic triple-negative breast cancer. Bioact Mater 2024; 39:612-629. [PMID: 38883315 PMCID: PMC11179173 DOI: 10.1016/j.bioactmat.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/21/2024] [Accepted: 04/21/2024] [Indexed: 06/18/2024] Open
Abstract
As a "cold tumor", triple-negative breast cancer (TNBC) exhibits limited responsiveness to current immunotherapy. How to enhance the immunogenicity and reverse the immunosuppressive microenvironment of TNBC remain a formidable challenge. Herein, an "in situ nanovaccine" Au/CuNDs-R848 was designed for imaging-guided photothermal therapy (PTT)/chemodynamic therapy (CDT) synergistic therapy to trigger dual immunoregulatory effects on TNBC. On the one hand, Au/CuNDs-R848 served as a promising photothermal agent and nanozyme, achieving PTT and photothermal-enhanced CDT against the primary tumor of TNBC. Meanwhile, the released antigens and damage-associated molecular patterns (DAMPs) promoted the maturation of dendritic cells (DCs) and facilitated the infiltration of T lymphocytes. Thus, Au/CuNDs-R848 played a role as an "in situ nanovaccine" to enhance the immunogenicity of TNBC by inducing immunogenic cell death (ICD). On the other hand, the nanovaccine suppressed the myeloid-derived suppressor cells (MDSCs), thereby reversing the immunosuppressive microenvironment. Through the dual immunoregulation, "cold tumor" was transformed into a "hot tumor", not only implementing a "turning foes to friends" therapeutic strategy but also enhancing immunotherapy against metastatic TNBC. Furthermore, Au/CuNDs-R848 acted as an excellent nanoprobe, enabling high-resolution near-infrared fluorescence and computed tomography imaging for precise visualization of TNBC. This feature offers potential applications in clinical cancer detection and surgical guidance. Collectively, this work provides an effective strategy for enhancing immune response and offers novel insights into the potential clinical applications for tumor immunotherapy.
Collapse
Affiliation(s)
- Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Tong Sha
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Jinwei Li
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Huanyu Luo
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Annan Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Hao Liang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Jinbiao Qiang
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Lei Li
- Department of Endocrinology, Lequn Branch, The First Hospital of Jilin University, 130021, PR China
| | - Andrew K Whittaker
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Hongchen Sun
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Ce Shi
- Department of Oral Pathology, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
- Jilin Provincial Key Laboratory of Science and Technology for Stomatology Nanoengineering, Changchun, 130021, PR China
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| |
Collapse
|
31
|
Chen Y, Wang Y, Li Z, Jiang H, Pan W, Liu M, Jiang W, Zhang X, Wang F. Preparation and immunological activity evaluation of an intranasal protein subunit vaccine against ancestral and mutant SARS-CoV-2 with curdlan sulfate/O-linked quaternized chitosan nanoparticles as carrier and adjuvant. Int J Biol Macromol 2024; 276:133733. [PMID: 39002905 DOI: 10.1016/j.ijbiomac.2024.133733] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 06/07/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Chitosan and its derivatives are ideal nasal vaccine adjuvant to deliver antigens to immune cells. Previously, we successfully used a chitosan derivative, O-(2-Hydroxyl) propyl-3-trimethyl ammonium chitosan chloride (O-HTCC), and a β-glucan derivative, curdlan sulfate (CS), to prepare a nanoparticle adjuvant CS/O-HTCC which could deliver ovalbumin to antigen presenting cells (APCs) through nasal inhalation. In this article, we used SARS-CoV-2 spike receptor binding domain (S-RBD) as the antigen and CS/O-HTCC nanoparticles as the adjuvant to develop a nasal mucosal protein subunit vaccine, CS/S-RBD/O-HTCC. The humoral immunity, cell-mediated immunity and mucosal immunity induced by vaccines were evaluated. The results showed that CS/S-RBD/O-HTCC could induce desirable immunization with single or bivalent antigen through nasal inoculation, giving one booster vaccination with mutated S-RBD (beta) could bring about a broad cross reaction with ancestral and different mutated S-RBD, and vaccination of the BALB/c mice with CS/S-RBD/O-HTCC containing S-RBD mix antigens (ancestral and omicron) could induce the production of binding and neutralizing antibodies against both of the two antigens. Our results indicate that CS/O-HTCC is a promising nasal mucosal adjuvant to prepare protein subunit vaccine for both primary and booster immunization, and the adjuvant is suitable for loading more than one antigen for preparing multivalent vaccines.
Collapse
MESH Headings
- Chitosan/chemistry
- Animals
- Nanoparticles/chemistry
- beta-Glucans/chemistry
- beta-Glucans/immunology
- SARS-CoV-2/immunology
- Vaccines, Subunit/immunology
- Mice
- Administration, Intranasal
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Adjuvants, Immunologic/pharmacology
- Mice, Inbred BALB C
- COVID-19/prevention & control
- COVID-19/immunology
- Female
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/chemistry
- Antibodies, Viral/immunology
- Immunity, Mucosal/drug effects
- Mutation
- Antibodies, Neutralizing/immunology
- Drug Carriers/chemistry
- Adjuvants, Vaccine/chemistry
- Humans
Collapse
Affiliation(s)
- Yipan Chen
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Yan Wang
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Zuyi Li
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Honglei Jiang
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Wei Pan
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Minghui Liu
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Wenjie Jiang
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China.
| | - Xinke Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Fengshan Wang
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China; NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, National Glycoengineering Research Center, Shandong University, Jinan 250012, Shandong, China.
| |
Collapse
|
32
|
Chibaya L, DeMarco KD, Lusi CF, Kane GI, Brassil ML, Parikh CN, Murphy KC, Chowdury SR, Li J, Ma B, Naylor TE, Cerrutti J, Mori H, Diaz-Infante M, Peura J, Pitarresi JR, Zhu LJ, Fitzgerald KA, Atukorale PU, Ruscetti M. Nanoparticle delivery of innate immune agonists combined with senescence-inducing agents promotes T cell control of pancreatic cancer. Sci Transl Med 2024; 16:eadj9366. [PMID: 39196958 PMCID: PMC11811823 DOI: 10.1126/scitranslmed.adj9366] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 03/18/2024] [Accepted: 06/11/2024] [Indexed: 08/30/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has quickly risen to become the third leading cause of cancer-related death in the United States. This is in part because of its fibrotic tumor microenvironment (TME) that contributes to poor vascularization and immune infiltration and subsequent chemo- and immunotherapy failure. Here, we investigated an immunotherapy approach combining delivery of stimulator of interferon genes (STING) and Toll-like receptor 4 (TLR4) innate immune agonists by lipid-based nanoparticle (NP) coencapsulation with senescence-inducing RAS-targeted therapies, which can remodel the immune suppressive PDAC TME through the senescence-associated secretory phenotype. Treatment of transplanted and autochthonous PDAC mouse models with these regimens led to enhanced uptake of NPs by multiple cell types in the PDAC TME, induction of type I interferon and other proinflammatory signaling pathways, increased antigen presentation by tumor cells and antigen-presenting cells, and subsequent activation of both innate and adaptive immune responses. This two-pronged approach produced potent T cell-driven and type I interferon-mediated tumor regression and long-term survival in preclinical PDAC models dependent on both tumor and host STING activation. STING and TLR4-mediated type I interferon signaling was also associated with enhanced natural killer and CD8+ T cell immunity in human PDAC samples. Thus, combining localized immune agonist delivery with systemic tumor-targeted therapy can orchestrate a coordinated type I interferon-driven innate and adaptive immune response with durable antitumor efficacy against PDAC.
Collapse
Affiliation(s)
- Loretah Chibaya
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Kelly D. DeMarco
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Christina F. Lusi
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Griffin I. Kane
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Meghan L. Brassil
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Chaitanya N. Parikh
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Katherine C. Murphy
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Shreya R. Chowdury
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Junhui Li
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Boyang Ma
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Tiana E. Naylor
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Julia Cerrutti
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Haruka Mori
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Miranda Diaz-Infante
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Jessica Peura
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Jason R. Pitarresi
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Katherine A. Fitzgerald
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Prabhani U. Atukorale
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Marcus Ruscetti
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Immunology and Microbiology Program, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| |
Collapse
|
33
|
Shen X, Yang YB, Gao Y, Wang S, Wang H, Sun M, Meng F, Tang YD, Tu Y, Kong Q, An TQ, Cai XH. Lipid A-modified Escherichia coli can produce porcine parvovirus virus-like particles with high immunogenicity and minimal endotoxin activity. Microb Cell Fact 2024; 23:222. [PMID: 39118114 PMCID: PMC11308658 DOI: 10.1186/s12934-024-02497-9] [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: 06/17/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND A cost-effective Escherichia coli expression system has gained popularity for producing virus-like particle (VLP) vaccines. However, the challenge lies in balancing the endotoxin residue and removal costs, as residual endotoxins can cause inflammatory reactions in the body. RESULTS In this study, porcine parvovirus virus-like particles (PPV-VLPs) were successfully assembled from Decreased Endotoxic BL21 (BL21-DeE), and the effect of structural changes in the lipid A of BL21 on endotoxin activity, immunogenicity, and safety was investigated. The lipopolysaccharide purified from BL21-DeE produced lower IL-6 and TNF-α than that from wild-type BL21 (BL21-W) in both RAW264.7 cells and BALB/c mice. Additionally, mice immunized with PPV-VLP derived form BL21-DeE (BL21-DeE-VLP) showed significantly lower production of inflammatory factors and a smaller increase in body temperature within 3 h than those immunized with VLP from BL21-W (BL21-W-VLP) and endotoxin-removed VLP (ReE-VLP). Moreover, mice in the BL21-DeE-VLP immunized group had similar levels of serum antibodies as those in the BL21-W-VLP group but significantly higher levels than those in the ReE-VLP group. Furthermore, the liver, lungs, and kidneys showed no pathological damage compared with the BL21-W-VLP group. CONCLUSION Overall, this study proposes a method for producing VLP with high immunogenicity and minimal endotoxin activity without chemical or physical endotoxin removal methods. This method could address the issue of endotoxin residues in the VLP and provide production benefits.
Collapse
Affiliation(s)
- Xuegang Shen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Yong-Bo Yang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
- Heilongjiang Veterinary Biopharmaceutical Engineering Technology Research Center, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yanfei Gao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Shujie Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Haiwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Mingxia Sun
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Fandan Meng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Yan-Dong Tang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Yabin Tu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China
| | - Qingke Kong
- College of Veterinary Medicine, Southwest University, No. 2 Tiansheng Road, Beibei District, Chongqing, 400715, China.
| | - Tong-Qing An
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China.
- Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| | - Xue-Hui Cai
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 678 Haping Road, Xiangfang District, Harbin, 150069, China.
- Heilongjiang Veterinary Biopharmaceutical Engineering Technology Research Center, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| |
Collapse
|
34
|
Alugupalli KR. Characterization of Turbo, a TLR Ligand-based Adjuvant for Glycoconjugate Vaccines. Immunohorizons 2024; 8:527-537. [PMID: 39093309 PMCID: PMC11374753 DOI: 10.4049/immunohorizons.2400040] [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: 04/17/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024] Open
Abstract
Many bacterial polysaccharide vaccines, including the typhoid Vi polysaccharide (ViPS) and tetravalent meningococcal polysaccharide conjugate (MCV4) vaccines, do not incorporate adjuvants and are not highly immunogenic, particularly in infants. I found that endotoxin, a TLR4 ligand in ViPS, contributes to the immunogenicity of typhoid vaccines. Because endotoxin is pyrogenic, and its levels are highly variable in vaccines, I developed monophosphoryl lipid A, a nontoxic TLR4 ligand-based adjuvant named Turbo. Admixing Turbo with ViPS and MCV4 vaccines improved their immunogenicity across all ages and eliminated booster requirement. To understand the characteristics of this adjuvanticity, I compared Turbo with alum. Unlike alum, which polarizes the response toward the IgG1 isotype, Turbo promoted Ab class switching to all IgG isotypes with affinity maturation; the magnitude of this IgG response is durable and accompanied by the presence of long-lived plasma cells in the mouse bone marrow. In striking contrast with the pathways employed by alum, Turbo adjuvanticity is independent of NLPR3, pyroptotic cell death effector Gasdermin D, and canonical and noncanonical inflammasome activation mediated by Caspase-1 and Caspase-11, respectively. Turbo adjuvanticity is primarily dependent on the MyD88 axis and is lost in mice deficient in costimulatory molecules CD86 and CD40, indicating that Turbo adjuvanticity includes activation of these pathways. Because Turbo formulations containing either monophosphoryl lipid A or TLR2 ligands, Pam2CysSerLys4, and Pam3CysSerLys4 help generate Ab response of all IgG isotypes, as an adjuvant Turbo can improve the immunogenicity of glycoconjugate vaccines against a wide range of bacterial pathogens whose elimination requires appropriate IgG isotypes.
Collapse
Affiliation(s)
- Kishore R. Alugupalli
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
- TurboVax Inc., Philadelphia, PA
| |
Collapse
|
35
|
Chen Y, Tang S, Cai F, Wan Y. Strategies for Small Extracellular Vesicle-Based Cancer Immunotherapy. RESEARCH (WASHINGTON, D.C.) 2024; 7:0421. [PMID: 39040921 PMCID: PMC11260559 DOI: 10.34133/research.0421] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/10/2024] [Indexed: 07/24/2024]
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed vesicles released by cells. EVs encapsulate proteins and nucleic acids of their parental cell and efficiently deliver the cargo to recipient cells. These vesicles act as mediators of intercellular communication and thus play a crucial role in various physiological and pathological processes. Moreover, EVs hold promise for clinical use. They have been explored as drug delivery vehicles, therapeutic agents, and targets for disease diagnosis. In the landscape of cancer research, while strides have been made in EV-focused cancer physiopathology, liquid biopsy, and drug delivery, the exploration of EVs as immunotherapeutic agents may not have seen substantial progress to date. Despite promising findings reported in cell and animal studies, the clinical translation of EV-based cancer immunotherapeutics encounters challenges. Here, we review the existing strategies used in EV-based cancer immunotherapy, aiming to propel the development of this emerging yet crucial field.
Collapse
Affiliation(s)
- Yundi Chen
- Department of Breast Surgery, Tongji Hospital, School of Medicine,
Tongji University, Shanghai, China
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering,
Binghamton University, Binghamton, NY, USA
| | - Shasha Tang
- Department of Breast Surgery, Tongji Hospital, School of Medicine,
Tongji University, Shanghai, China
| | - Fengfeng Cai
- Department of Breast Surgery, Tongji Hospital, School of Medicine,
Tongji University, Shanghai, China
| | - Yuan Wan
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering,
Binghamton University, Binghamton, NY, USA
| |
Collapse
|
36
|
Goetz M, Thotathil N, Zhao Z, Mitragotri S. Vaccine adjuvants for infectious disease in the clinic. Bioeng Transl Med 2024; 9:e10663. [PMID: 39036089 PMCID: PMC11256182 DOI: 10.1002/btm2.10663] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 07/23/2024] Open
Abstract
Adjuvants, materials added to vaccines to enhance the resulting immune response, are important components of vaccination that are many times overlooked. While vaccines always include an antigen to tell the body what to vaccinate to, of equal importance the adjuvant provides the how, a significant factor in producing a complete response. The adjuvant space has been slow to develop with the first use of an adjuvant in a licensed vaccine occurring in the 1930s, and remaining the only adjuvant in licensed vaccines for the next 80 years. However, with vaccination at the forefront of protection against new and complex pathogens, it is important to consider all components when designing an effective vaccine. Here we summarize the adjuvant space in licensed vaccines as well as the novel adjuvant space in clinical trials with a specific focus on the materials utilized and their resulting impact on the immune response. We discuss five major categories of adjuvant materials: aluminum salts, nanoparticles, viral vectors, TLR agonists, and emulsions. For each category, we delve into the current clinical trials space, the impact of these materials on vaccination, as well as some of the ways in which they could be improved. Adjuvants present an exciting opportunity to improve vaccine responses and stability, this review will help inform about the current progress of this space. Translational impact statement In the aftermath of the COVID-19 pandemic, vaccines for infectious diseases have come into the spotlight. While antigens have always been an important focus of vaccine design, the adjuvant is a significant tool for enhancing the immune response to the vaccine that has been largely underdeveloped. This article provides a broad review of the history of adjuvants and, the current vaccine adjuvant space, and the progress seen in adjuvants in clinical trials. There is specific emphasis on the material landscape for adjuvants and their resulting mechanism of action. Looking ahead, while the novel vaccine adjuvant space features exciting new technologies and materials, there is still a need for more to meet the protective needs of new and complex pathogens.
Collapse
Affiliation(s)
- Morgan Goetz
- John A Paulson School of Engineering & Applied SciencesHarvard UniversityAllstonMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Naaz Thotathil
- University of Massachusetts AmherstAmherstMassachusettsUSA
| | - Zongmin Zhao
- Department of Pharmaceutical SciencesCollege of Pharmacy, University of Illinois ChicagoChicagoIllinoisUSA
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied SciencesHarvard UniversityAllstonMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| |
Collapse
|
37
|
Gao L, Li G, Qiu C, Ye Y, Li X, Liao P, Ming W, Liu Z, Luo X, Liao G. Design, Synthesis, and Bioactivity Evaluation of a TF-Based Cancer Vaccine Candidate Using Lipid A Mimetics As a Built-In Adjuvant. J Med Chem 2024; 67:9976-9990. [PMID: 38886162 DOI: 10.1021/acs.jmedchem.4c00042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
This study describes the design and synthesis of five TF-based cancer vaccine candidates using a lipid A mimetic as the carrier and a built-in adjuvant. All synthesized conjugates elicited robust and consistent TF-specific immune responses in mice without external adjuvants. Immunological studies subsequently conducted in wild-type and TLR4 knockout C57BL/6 mice demonstrated that the activation of TLR4 was the main reason that the synthesized lipid A mimetics increased the TF-specific immune responses. All antisera induced by these conjugates can specifically recognize, bind to, and induce the lysis of TF-positive cancer cells. Moreover, representative conjugates 2 and 3 could effectively reduce the growth of tumors and prolong the survival time of mice in vivo, and the efficacies were better than glycoprotein TF-CRM197 with alum adjuvant. Lipid A mimetics could therefore be a promising platform for the development of new carbohydrate-based vaccine carriers with self-adjuvanting properties for the treatment of cancer.
Collapse
Affiliation(s)
- Lingqiang Gao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Guiqi Li
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Cuiping Qiu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yifan Ye
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaohui Li
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Pan Liao
- Guangzhou Yuemei Pharmaceutical Technology Co., Ltd, Guangzhou 510535, China
| | - Wenbo Ming
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhongqiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiang Luo
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Guochao Liao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
38
|
Hjálmsdóttir Á, Hasler F, Waeckerle-Men Y, Duda A, López-Deber MP, Pihlgren M, Vukicevic M, Kündig TM, Johansen P. T cell independent antibody responses with class switch and memory using peptides anchored on liposomes. NPJ Vaccines 2024; 9:115. [PMID: 38909055 PMCID: PMC11193769 DOI: 10.1038/s41541-024-00902-3] [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: 06/05/2023] [Accepted: 05/23/2024] [Indexed: 06/24/2024] Open
Abstract
Vaccines generally require T lymphocytes for B-cell activation and immunoglobulin class switching in response to peptide or protein antigens. In the absence of T cells, limited IgG class switch takes place, germinal centers are short-lived, and the B cells lack memory. Here, immunization of mice with liposomes containing 15mer peptides and monophosphoryl lipid A (MPLA) as adjuvant, induced T-cell independent (TI) IgG class switch within three days, as well as germinal center formation. The antibody responses were long-lived, strictly dependent on Toll-like receptor 4 (TLR4) signaling, partly dependent on Bruton's tyrosine kinase (BTK) signal transmission, and independent of signaling through T-cell receptors, MHC class II and inflammasome. The antibody response showed characteristics of both TI type 1 and TI type 2. All IgG subclasses could be boosted months after primary immunization, and the biological function of the secreted antibodies was demonstrated in murine models of allergic anaphylaxis and of bacterial infection. Moreover, antibody responses after immunization with peptide- and MPLA-loaded liposomes could be triggered in neonatal mice and in mice receiving immune-suppressants. This study demonstrates T-cell independent endogenous B-cell memory and recall responses in vivo using a peptide antigen. The stimulation of these antibody responses required a correct and dense assembly and administration of peptide and adjuvant on the surface of liposomes. In the future, TI vaccines may prove beneficial in pathological conditions in which T-cell immunity is compromised through disease or medicines or when rapid, antibody-mediated immune protection is needed.
Collapse
Affiliation(s)
| | - Fabio Hasler
- Department of Dermatology, University of Zurich, Zurich, Switzerland
| | | | - Agathe Duda
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | | | - Maria Pihlgren
- AC Immune SA, EPFL Innovation Park EPFL, Lausanne, Switzerland
| | | | - Thomas M Kündig
- Department of Dermatology, University of Zurich, Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Pål Johansen
- Department of Dermatology, University of Zurich, Zurich, Switzerland.
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
39
|
Adams CS, Kim H, Burtner AE, Lee DS, Dobbins C, Criswell C, Coventry B, Kim HM, King NP. De novo design of protein minibinder agonists of TLR3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.17.589973. [PMID: 38659926 PMCID: PMC11042314 DOI: 10.1101/2024.04.17.589973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Toll-like Receptor 3 (TLR3) is a pattern recognition receptor that initiates antiviral immune responses upon binding double-stranded RNA (dsRNA). Several nucleic acid-based TLR3 agonists have been explored clinically as vaccine adjuvants in cancer and infectious disease, but present substantial manufacturing and formulation challenges. Here, we use computational protein design to create novel miniproteins that bind to human TLR3 with nanomolar affinities. Cryo-EM structures of two minibinders in complex with TLR3 reveal that they bind the target as designed, although one partially unfolds due to steric competition with a nearby N-linked glycan. Multimeric forms of both minibinders induce NF-κB signaling in TLR3-expressing cell lines, demonstrating that they may have therapeutically relevant biological activity. Our work provides a foundation for the development of specific, stable, and easy-to-formulate protein-based agonists of TLRs and other pattern recognition receptors.
Collapse
Affiliation(s)
- Chloe S. Adams
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Hyojin Kim
- Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon 34126, South Korea
| | - Abigail E. Burtner
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Dong Sun Lee
- Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon 34126, South Korea
| | - Craig Dobbins
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Cameron Criswell
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Brian Coventry
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| | - Ho Min Kim
- Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon 34126, South Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, South Korea
| | - Neil P. King
- Institute for Protein Design, University of Washington, Seattle, WA, 98195 USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98195 USA
| |
Collapse
|
40
|
Verpalen ECJM, Brouwer AJ, Wolfert MA, Boons GJ. Structure-Based Design and Synthesis of Lipid A Derivatives to Modulate Cytokine Responses. Chemistry 2024:e202400429. [PMID: 38587187 DOI: 10.1002/chem.202400429] [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: 01/31/2024] [Revised: 03/28/2024] [Accepted: 04/07/2024] [Indexed: 04/09/2024]
Abstract
Agonists of Toll like receptors (TLRs) have attracted interest as adjuvants and immune modulators. A crystal structure of TLR4/MD2 with E. coli LPS indicates that the fatty acid at C-2 of the lipid A component of LPS induces dimerization of two TLR4-MD2 complexes, which in turn initiates cell signaling leading to the production of (pro)inflammatory cytokines. To probe the importance of the (R)-3-hydroxymyristate at C-2 of lipid A, a range of bis- and mono-phosphoryl lipid A derivatives with different modifications at C-2 were prepared by a strategy in which 2-methylnaphthyl ethers were employed as permanent protecting group that could be readily removed by catalytic hydrogenation. The C-2 amine was protected as 9-fluorenylmethyloxycarbamate, which at a later stage could be removed to give a free amine that was modified by different fatty acids. LPS and the synthetic lipid As induced the same cytokines, however, large differences in activity were observed. A compound having a hexanoyl moiety at C-2 still showed agonistic properties, but further shortening to a butanoyl abolished activity. The modifications had a larger influence on monophosphoryl lipid As. The lipid As having a butanoyl moiety at C-2 could selectively antagonize TRIF associated cytokines induced by LPS or lipid A.
Collapse
Affiliation(s)
- Enrico C J M Verpalen
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Arwin J Brouwer
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Margreet A Wolfert
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Geert-Jan Boons
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
- Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
- Chemistry Department, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
41
|
Manabe Y, Gárate-Reyes B, Ito K, Hurtado-Guerrero R, Kabayama K, Fukase K. Synthesis and immunological evaluation of TLR1/2 ligand-conjugated RBDs as self-adjuvanting vaccine candidates against SARS-CoV-2. Chem Commun (Camb) 2024; 60:3946-3949. [PMID: 38497901 DOI: 10.1039/d4cc00462k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
We synthesized and evaluated Pam3CSK4-conjugated receptor binding domain (RBD)/deglycosylated RBD as potential anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine candidates. Our investigation revealed the critical importance of limiting the number of introduced Pam3CSK4 molecules to the RBD in order to preserve its antigenicity. We also confirmed the harmonious integration of the adjuvant-conjugation strategy with the glycan-shield removal strategy.
Collapse
Affiliation(s)
- Yoshiyuki Manabe
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan.
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Brandon Gárate-Reyes
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan.
| | - Keita Ito
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan.
| | - Ramón Hurtado-Guerrero
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I + D, Zaragoza, Spain
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Laboratorio de Microscopías Avanzada (LMA), University of Zaragoza, Mariano Esquillor s/n, Campus Rio Ebro, Edificio I + D, Zaragoza, Spain
- Fundación ARAID, Zaragoza, Spain
| | - Kazuya Kabayama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan.
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Koichi Fukase
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan.
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Center for Advanced Modalities and DDS, Osaka University, 11 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
42
|
Chen Y, Song Y, Zhu X, Dong CM, Chen M. Design and Update of Multifunctional Polypeptides and Their Applications for the Prevention of Viral Infections and Cancer Immunotherapies. POLYM REV 2024; 64:528-574. [DOI: 10.1080/15583724.2023.2281462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/07/2023] [Accepted: 11/04/2023] [Indexed: 01/06/2025]
Affiliation(s)
- Yanzheng Chen
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Yingying Song
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Chang-Ming Dong
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Mingsheng Chen
- Shanghai Public Health Clinic Center, Fudan University, Shanghai, P. R. China
| |
Collapse
|
43
|
Pal S, Chaudhari R, Baurceanu I, Hill BJ, Nagy BA, Wolf MT. Extracellular Matrix Scaffold-Assisted Tumor Vaccines Induce Tumor Regression and Long-Term Immune Memory. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309843. [PMID: 38302823 PMCID: PMC11009079 DOI: 10.1002/adma.202309843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/24/2024] [Indexed: 02/03/2024]
Abstract
Injectable scaffold delivery is a strategy to enhance the efficacy of cancer vaccine immunotherapy. The choice of scaffold biomaterial is crucial, impacting both vaccine release kinetics and immune stimulation via the host response. Extracellular matrix (ECM) scaffolds prepared from decellularized tissues facilitate a pro-healing inflammatory response that promotes local cancer immune surveillance. Here, an ECM scaffold-assisted therapeutic cancer vaccine that maintains an immune microenvironment consistent with tissue reconstruction is engineered. Several immune-stimulating adjuvants are screened to develop a cancer vaccine formulated with decellularized small intestinal submucosa (SIS) ECM scaffold co-delivery. It is found that the STING pathway agonist cyclic di-AMP most effectively induces cytotoxic immunity in an ECM scaffold vaccine, without compromising key interleukin 4 (IL-4) mediated immune pathways associated with healing. ECM scaffold delivery enhances therapeutic vaccine efficacy, curing 50-75% of established E.G-7OVA lymphoma tumors in mice, while none are cured with soluble vaccine. SIS-ECM scaffold-assisted vaccination prolonged antigen exposure is dependent on CD8+ cytotoxic T cells and generates long-term antigen-specific immune memory for at least 10 months post-vaccination. This study shows that an ECM scaffold is a promising delivery vehicle to enhance cancer vaccine efficacy while being orthogonal to characteristics of pro-healing immune hallmarks.
Collapse
Affiliation(s)
- Sanjay Pal
- Cancer Biomaterial Engineering Section, Cancer Innovation
Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD
21702
| | - Rohan Chaudhari
- Cancer Biomaterial Engineering Section, Cancer Innovation
Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD
21702
- OHSU School of Medicine, Oregon Health & Science
University, Portland, OR 97239
| | - Iris Baurceanu
- Cancer Biomaterial Engineering Section, Cancer Innovation
Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD
21702
| | - Brenna J. Hill
- AIDS and Cancer Virus Program, Frederick National
Laboratory for Cancer Research, Frederick, MD 21702
| | - Bethany A. Nagy
- Laboratory Animal Sciences Program (LASP), National Cancer
Institute, Frederick, MD 21702
| | - Matthew T. Wolf
- Cancer Biomaterial Engineering Section, Cancer Innovation
Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD
21702
| |
Collapse
|
44
|
Lin YJ, Zimmermann J, Schülke S. Novel adjuvants in allergen-specific immunotherapy: where do we stand? Front Immunol 2024; 15:1348305. [PMID: 38464539 PMCID: PMC10920236 DOI: 10.3389/fimmu.2024.1348305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Type I hypersensitivity, or so-called type I allergy, is caused by Th2-mediated immune responses directed against otherwise harmless environmental antigens. Currently, allergen-specific immunotherapy (AIT) is the only disease-modifying treatment with the potential to re-establish clinical tolerance towards the corresponding allergen(s). However, conventional AIT has certain drawbacks, including long treatment durations, the risk of inducing allergic side effects, and the fact that allergens by themselves have a rather low immunogenicity. To improve AIT, adjuvants can be a powerful tool not only to increase the immunogenicity of co-applied allergens but also to induce the desired immune activation, such as promoting allergen-specific Th1- or regulatory responses. This review summarizes the knowledge on adjuvants currently approved for use in human AIT: aluminum hydroxide, calcium phosphate, microcrystalline tyrosine, and MPLA, as well as novel adjuvants that have been studied in recent years: oil-in-water emulsions, virus-like particles, viral components, carbohydrate-based adjuvants (QS-21, glucans, and mannan) and TLR-ligands (flagellin and CpG-ODN). The investigated adjuvants show distinct properties, such as prolonging allergen release at the injection site, inducing allergen-specific IgG production while also reducing IgE levels, as well as promoting differentiation and activation of different immune cells. In the future, better understanding of the immunological mechanisms underlying the effects of these adjuvants in clinical settings may help us to improve AIT.
Collapse
Affiliation(s)
- Yen-Ju Lin
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Stefan Schülke
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
- Section Research Allergology (ALG 5), Division of Allergology, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
45
|
Shafieichaharberoud F, Lang S, Whalen C, Rivera Quiles C, Purcell L, Talbot C, Wang P, Norton EB, Mazei-Robison M, Sulima A, Jacobson AE, Rice KC, Matyas GR, Huang X. Enhancing Protective Antibodies against Opioids through Antigen Display on Virus-like Particles. Bioconjug Chem 2024; 35:164-173. [PMID: 38113481 PMCID: PMC11259974 DOI: 10.1021/acs.bioconjchem.3c00415] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Opioid use disorder (OUD) has become a public health crisis, with recent significant increases in the number of deaths due to overdose. Vaccination can provide an attractive complementary strategy to combat OUD. A key for high vaccine efficacy is the induction of high levels of antibodies specific to the drug of abuse. Herein, a powerful immunogenic carrier, virus-like particle mutant bacteriophage Qβ (mQβ), has been investigated as a carrier of a small molecule hapten 6-AmHap mimicking heroin. The mQβ-6-AmHap conjugate was able to induce significantly higher levels of IgG antibodies against 6-AmHap than mice immunized with the corresponding tetanus toxoid-6-AmHap conjugate in head-to-head comparison studies in multiple strains of mice. The IgG antibody responses were persistent with high anti-6-AmHap titers 600 days after being immunized with mQβ-6-AmHap. The antibodies induced exhibited strong binding toward multiple heroin/morphine derivatives that have the potential to be abused, while binding weakly to medications used for OUD treatment and pain relief. Furthermore, vaccination effectively reduced the impacts of morphine on mice in both ambulation and antinociception assays, highlighting the translational potential of the mQβ-6-AmHap conjugate to mitigate the harmful effects of drugs of abuse.
Collapse
Affiliation(s)
- Fatemeh Shafieichaharberoud
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, Michigan 48824, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Shuyao Lang
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, Michigan 48824, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Connor Whalen
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Cristina Rivera Quiles
- Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, Michigan 48824, United States
| | - Lillie Purcell
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, Michigan 48824, United States
| | - Cameron Talbot
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, Michigan 48824, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Pengfei Wang
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Elizabeth B Norton
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Michelle Mazei-Robison
- Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, Michigan 48824, United States
| | - Agnieszka Sulima
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Arthur E Jacobson
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kenner C Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Gary R Matyas
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, Michigan 48824, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
46
|
Liu D, Fu L, Gong L, Li S, Li K, Liu K, Yang D. Proton-Gradient-Driven Porphyrin-Based Liposome Remote-Loaded with Imiquimod as In Situ Nanoadjuvants for Synergistically Augmented Tumor Photoimmunotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:8403-8416. [PMID: 38334116 DOI: 10.1021/acsami.3c17133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Cancer immunotherapy is expected to achieve tumor treatment mainly by stimulating the patient's own immune system to kill tumor cells. However, the low immunogenicity of the tumor and the poor efficiency of tumor antigen presentation result in a variety of solid tumors that do not respond to immunotherapy. Herein, we designed a proton-gradient-driven porphyrin-based liposome (PBL) with highly efficient Toll-like receptor 7 (TLR7) agonist (imiquimod, R837) encapsulation (R837@PBL). R837@PBL rapidly released R837 in the acid microenvironment to activate the TLR in the endosome inner membrane to promote bone-marrow-derived dendritic cell maturation and enhance antigen presentation. R837@PBL upon laser irradiation triggered immunogenic cell death of tumor cells and tumor-associated antigen release after subcutaneous injection, activated TLR7, formed in situ tumor nanoadjuvants, and enhanced the antigen presentation efficiency. Photoimmunotherapy promoted the infiltration of cytotoxic T lymphocytes into tumor tissues, inhibited the growth of the treated and abscopal tumors, and exerted highly effective photoimmunotherapeutic effects. Hence, our designed in situ tumor nanoadjuvants are expected to be an effective treatment for treated and abscopal tumors, providing a novel approach for synergistic photoimmunotherapy of tumors.
Collapse
Affiliation(s)
- Dechun Liu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, Shaanxi, China
| | - Luyao Fu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, Shaanxi, China
| | - Linlin Gong
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, Shaanxi, China
| | - Shasha Li
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, Shaanxi, China
| | - Kunwei Li
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, Shaanxi, China
| | - Kunhong Liu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, Shaanxi, China
| | - Dan Yang
- Department of Pharmaceutical Sciences, School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Weiyang University Park, Xi'an 710021, China
| |
Collapse
|
47
|
Wang G, Wang Y, Ma F. Exploiting bacterial-origin immunostimulants for improved vaccination and immunotherapy: current insights and future directions. Cell Biosci 2024; 14:24. [PMID: 38368397 PMCID: PMC10874560 DOI: 10.1186/s13578-024-01207-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/06/2024] [Indexed: 02/19/2024] Open
Abstract
Vaccination is a valid strategy to prevent and control newly emerging and reemerging infectious diseases in humans and animals. However, synthetic and recombinant antigens are poor immunogenic to stimulate efficient and protective host immune response. Immunostimulants are indispensable factors of vaccines, which can promote to trigger fast, robust, and long-lasting immune responses. Importantly, immunotherapy with immunostimulants is increasing proved to be an effective and promising treatment of cancer, which could enhance the function of the immune system against tumor cells. Pattern recognition receptors (PRRs) play vital roles in inflammation and are central to innate and adaptive immune responses. Toll-like receptors (TLRs)-targeting immunostimulants have become one of the hotspots in adjuvant research and cancer therapy. Bacterial-origin immunoreactive molecules are usually the ligands of PRRs, which could be fast recognized by PRRs and activate immune response to eliminate pathogens. Varieties of bacterial immunoreactive molecules and bacterial component-mimicking molecules have been successfully used in vaccines and clinical therapy so far. This work provides a comprehensive review of the development, current state, mechanisms, and applications of bacterial-origin immunostimulants. The exploration of bacterial immunoreactive molecules, along with their corresponding mechanisms, holds immense significance in deepening our understanding of bacterial pathogenicity and in the development of promising immunostimulants.
Collapse
Affiliation(s)
- Guangyu Wang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, Jiangsu, 210023, China
| | - Yongkang Wang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, Jiangsu, 210023, China
| | - Fang Ma
- Institute of Veterinary Immunology & Engineering, National Research Center of Engineering and Technology for Veterinary Biologicals, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China.
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, 212013, China.
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, 225300, China.
| |
Collapse
|
48
|
Yamaguchi M, Mtali YS, Sonokawa H, Takashima K, Fukushima Y, Kouwaki T, Oshiumi H. HPV vaccines induce trained immunity and modulate pro-inflammatory cytokine expression in response to secondary Toll-like receptor stimulations. Microbiol Immunol 2024; 68:65-74. [PMID: 38105559 DOI: 10.1111/1348-0421.13108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/25/2023] [Accepted: 12/03/2023] [Indexed: 12/19/2023]
Abstract
Cervical cancer is caused mostly by human papillomavirus (HPV), and several HPV vaccines have been developed to prevent its onset. Vaccines include antigens as well as adjuvants, with adjuvants playing an important role in activating the innate immune responses necessary for inducing adaptive immunological responses. Recent research has shown the presence of trained immunity inside the innate immune system. However, trained immunity conferred by HPV vaccinations is not well understood. In this work, we explored the innate immune responses and trained immunity caused by two HPV vaccines, Cervarix and Gardasil. Cervarix includes monophosphoryl lipid A and an aluminum adjuvant, and it significantly increased the expression of IL-6 and IFN-β mRNAs in RAW264.7 cells. On the contrary, Gardasil, which only includes an aluminum adjuvant, exhibited little cytokine expression but increased the expression of TLRs. Furthermore, Cervarix significantly increased IL-1β secretion from mouse macrophages, while Gardasil only mildly induced IL-1β secretion. Interestingly, initial stimulation with Gardasil enhanced the expression of IL-6 and TNF-α mRNAs upon secondary stimulation with TLR ligands, indicating that Gardasil induced trained immunity in macrophages. Moreover, Gardasil injection into mice resulted in enhanced TNF-α production in sera following secondary TLR stimulation. Our findings suggest that HPV vaccinations have the ability to induce trained immunity that modulate TLR ligand responses.
Collapse
Affiliation(s)
- Mako Yamaguchi
- School of Medicine, Kumamoto University, Kumamoto, Japan
| | - Yohana S Mtali
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Ken Takashima
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshimi Fukushima
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takahisa Kouwaki
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
49
|
Roman F, Burny W, Ceregido MA, Laupèze B, Temmerman ST, Warter L, Coccia M. Adjuvant system AS01: from mode of action to effective vaccines. Expert Rev Vaccines 2024; 23:715-729. [PMID: 39042099 DOI: 10.1080/14760584.2024.2382725] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION The use of novel adjuvants in human vaccines continues to expand as their contribution to preventing disease in challenging populations and caused by complex pathogens is increasingly understood. AS01 is a family of liposome-based vaccine Adjuvant Systems containing two immunostimulants: 3-O-desacyl-4'-monophosphoryl lipid A and the saponin QS-21. AS01-containing vaccines have been approved and administered to millions of individuals worldwide. AREAS COVERED Here, we report advances in our understanding of the mode of action of AS01 that contributed to the development of efficacious vaccines preventing disease due to malaria, herpes zoster, and respiratory syncytial virus. AS01 induces early innate immune activation that induces T cell-mediated and antibody-mediated responses with optimized functional characteristics and induction of immune memory. AS01-containing vaccines appear relatively impervious to baseline immune status translating into high efficacy across populations. Currently licensed AS01-containing vaccines have shown acceptable safety profiles in clinical trials and post-marketing settings. EXPERT OPINION Initial expectations that adjuvantation with AS01 could support effective vaccine responses and contribute to disease control have been realized. Investigation of the utility of AS01 in vaccines to prevent other challenging diseases, such as tuberculosis, is ongoing, together with efforts to fully define its mechanisms of action in different vaccine settings.
Collapse
|
50
|
He S, Liu S. Zwitterionic materials for nucleic acid delivery and therapeutic applications. J Control Release 2024; 365:919-935. [PMID: 38103789 DOI: 10.1016/j.jconrel.2023.12.017] [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: 11/28/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Nucleic acid therapeutics have demonstrated substantial potential in combating various diseases. However, challenges persist, particularly in the delivery of multifunctional nucleic acids. To address this issue, numerous gene delivery vectors have been developed to fully unlock the potential of gene therapy. The advancement of innovative materials with exceptional delivery properties is critical to propel the clinical translation of nucleic acid drugs. Cationic vector materials have received extensive attention, while zwitterionic materials remain relatively underappreciated in delivery. In this review, we outline a diverse range of zwitterionic material nucleic acid carriers, predominantly encompassing zwitterionic lipids, polymers and peptides. Their respective chemical structures, synthesis approaches, properties, advantages, and therapeutic applications are summarized and discussed. Furthermore, we highlight the challenges and future opportunities associated with the development of zwitterionic vector materials. This review will aid to understand the zwitterionic materials in aiding gene delivery, contributing to the continual progress of nucleic acid therapeutics.
Collapse
Affiliation(s)
- Shun He
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shuai Liu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|