1
|
Riel AMS, Rungelrath V, Elwaie TA, Rasheed OK, Hicks L, Ettenger G, You DC, Smith M, Buhl C, Abdelwahab W, Miller SM, Smith AJ, Burkhart D, Evans JT, Ryter KT. Systematic Evaluation of Regiochemistry and Lipidation of Aryl Trehalose Mincle Agonists. Int J Mol Sci 2024; 25:10031. [PMID: 39337517 PMCID: PMC11432005 DOI: 10.3390/ijms251810031] [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: 08/08/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
The Macrophage-Inducible C-type Lectin receptor (Mincle) plays a critical role in innate immune recognition and pathology, and therefore represents a promising target for vaccine adjuvants. Innovative trehalose-based Mincle agonists with improved pharmacology and potency may prove useful in the development of Th17-mediated adaptive immune responses. Herein, we report on in vitro and in silico investigations of specific Mincle ligand-receptor interactions required for the effective receptor engagement and activation of Th17-polarizing cytokines. Specifically, we employed a library of trehalose benzoate scaffolds, varying the degree of aryl lipidation and regiochemistry that produce inflammatory cytokines in a Mincle-dependent fashion. In vitro interleukin-6 (IL-6) cytokine production by human peripheral blood mononuclear cells (hPBMCs) indicated that the lipid regiochemistry is key to potency and maximum cytokine output, with the tri-substituted compounds inducing higher levels of IL-6 in hPBMCs than the di-substituted derivatives. Additionally, IL-6 production trended higher after stimulation with compounds that contained lipids ranging from five to eight carbons long, compared to shorter (below five) or longer (above eight) carbon chains, across all the substitution patterns. An analysis of the additional cytokines produced by hPBMCs revealed that compound 4d, tri-substituted and five carbons long, induced significantly greater levels of interleukin-1β (IL-1β), tumor necrosis factor- α (TNF-α), interleukin-23 (IL-23), and interferon- γ (IFN-γ) than the other compounds tested in this study. An in silico assessment of 4d highlighted the capability of this analogue to bind to the human Mincle carbohydrate recognition domain (CRD) efficiently. Together, these data highlight important structure-activity findings regarding Mincle-specific cytokine induction, generating a lead adjuvant candidate for future formulations and immunological evaluations.
Collapse
Affiliation(s)
- Asia Marie S Riel
- Department of Chemistry and Biochemistry, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
| | - Viktoria Rungelrath
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
| | - Tamer A Elwaie
- Department of Chemistry and Biochemistry, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Omer K Rasheed
- Department of Chemistry and Biochemistry, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
- Inimmune Corporation, Missoula, MT 59802, USA
| | - Linda Hicks
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
| | - George Ettenger
- Department of Chemistry and Biochemistry, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
| | - Dai-Chi You
- Department of Chemistry and Biochemistry, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
| | - Mira Smith
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
| | - Cassandra Buhl
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
- Inimmune Corporation, Missoula, MT 59802, USA
| | - Walid Abdelwahab
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
| | - Shannon M Miller
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
- Inimmune Corporation, Missoula, MT 59802, USA
| | - Alyson J Smith
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
- Inimmune Corporation, Missoula, MT 59802, USA
| | - David Burkhart
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
- Inimmune Corporation, Missoula, MT 59802, USA
| | - Jay T Evans
- Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
- Inimmune Corporation, Missoula, MT 59802, USA
| | - Kendal T Ryter
- Department of Chemistry and Biochemistry, Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA
- Inimmune Corporation, Missoula, MT 59802, USA
| |
Collapse
|
2
|
Abdelwahab WM, Le-Vinh B, Riffey A, Hicks L, Buhl C, Ettenger G, Jackson KJ, Weiss AM, Miller S, Ryter K, Evans JT, Burkhart DJ. Promotion of Th17 Polarized Immunity via Co-Delivery of Mincle Agonist and Tuberculosis Antigen Using Silica Nanoparticles. ACS APPLIED BIO MATERIALS 2024; 7:3877-3889. [PMID: 38832760 DOI: 10.1021/acsabm.4c00245] [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: 06/05/2024]
Abstract
Adjuvants and immunomodulators that effectively drive a Th17-skewed immune response are not part of the standard vaccine toolkit. Vaccine adjuvants and delivery technologies that can induce Th17 or Th1/17 immunity and protection against bacterial pathogens, such as tuberculosis (TB), are urgently needed. Th17-polarized immune response can be induced using agonists that bind and activate C-type lectin receptors (CLRs) such as macrophage inducible C-type lectin (Mincle). A simple but effective strategy was developed for codelivering Mincle agonists with the recombinant Mycobacterium tuberculosis fusion antigen, M72, using tunable silica nanoparticles (SNP). Anionic bare SNP, hydrophobic phenyl-functionalized SNP (P-SNP), and cationic amine-functionalized SNP (A-SNP) of different sizes were coated with three synthetic Mincle agonists, UM-1024, UM-1052, and UM-1098, and evaluated for adjuvant activity in vitro and in vivo. The antigen and adjuvant were coadsorbed onto SNP via electrostatic and hydrophobic interactions, facilitating multivalent display and delivery to antigen presenting cells. The cationic A-SNP showed the highest coloading efficiency for the antigen and adjuvant. In addition, the UM-1098-adsorbed A-SNP formulation demonstrated slow-release kinetics in vitro, excellent stability over 12 months of storage, and strong IL-6 induction from human peripheral blood mononuclear cells. Co-adsorption of UM-1098 and M72 on A-SNP significantly improved antigen-specific humoral and Th17-polarized immune responses in vivo in BALB/c mice relative to the controls. Taken together, A-SNP is a promising platform for codelivery and proper presentation of adjuvants and antigens and provides the basis for their further development as a vaccine delivery platform for immunization against TB or other diseases for which Th17 immunity contributes to protection.
Collapse
Affiliation(s)
- Walid M Abdelwahab
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Bao Le-Vinh
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Alexander Riffey
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Linda Hicks
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Cassandra Buhl
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - George Ettenger
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Chemistry, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Konner J Jackson
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| | - Adam M Weiss
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Shannon Miller
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| | - Kendal Ryter
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Chemistry, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| | - Jay T Evans
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| | - David J Burkhart
- Center for Translational Medicine, 32 campus drive, Missoula, Montana 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
- Inimmune Corporation, 1121 East Broadway, Missoula, Montana 59812, United States
| |
Collapse
|
3
|
Foster M, Dangerfield EM, Timmer MSM, Stocker BL, Wilkinson BL. Probing Isosteric Replacement for Immunoadjuvant Design: Bis-Aryl Triazole Trehalolipids are Mincle Agonists. ACS Med Chem Lett 2024; 15:899-905. [PMID: 38894898 PMCID: PMC11181483 DOI: 10.1021/acsmedchemlett.4c00100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
Herein, we report the modular synthesis and immunological activity of seven bis-aryl triazole trehalolipids (1a-1g) as Brartemicin analogs. The compounds comprised one or two octyloxy (C8) alkyl chains and were synthesized using the venerable CuAAc reaction between the respective aryl acetylenes and a trehalose diazide. A Mincle reporter cell assay revealed that all lipidated analogs activated Mincle. Two compounds, 1c and 1d, produced strong Mincle-dependent immune responses in vitro. The activity was dependent on the degree of alkylation and regiochemistry, with 1c and 1d showing significantly increased IL-1β production in vitro compared to monoalkylated compounds and dialkylated compounds lacking ortho substitution. Molecular docking of 1c positioned the triazole in proximity to Arg-183, which may offer additional interactions that could explain the binding affinity for this class of ligand. These findings demonstrate the capability of triazole-linked Brartemicin analogs as Mincle-mediated Th1/Th17 vaccine adjuvants.
Collapse
Affiliation(s)
- Michael
J. Foster
- School
of Science and Technology, University of
New England, Armidale 2351, Australia
| | - Emma M. Dangerfield
- School
of Chemical and Physical Sciences, Victoria
University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| | - Mattie S. M. Timmer
- School
of Chemical and Physical Sciences, Victoria
University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| | - Bridget L. Stocker
- School
of Chemical and Physical Sciences, Victoria
University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| | - Brendan L. Wilkinson
- School
of Science and Technology, University of
New England, Armidale 2351, Australia
| |
Collapse
|
4
|
Rungelrath V, Ahmed M, Hicks L, Miller SM, Ryter KT, Montgomery K, Ettenger G, Riffey A, Abdelwahab WM, Khader SA, Evans JT. Vaccination with Mincle agonist UM-1098 and mycobacterial antigens induces protective Th1 and Th17 responses. NPJ Vaccines 2024; 9:100. [PMID: 38844494 PMCID: PMC11156909 DOI: 10.1038/s41541-024-00897-x] [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] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is one of the top infectious killers in the world. The only licensed vaccine against TB, Bacille Calmette-Guérin (BCG), provides variable protection against pulmonary TB, especially in adults. Hence, novel TB vaccine approaches are urgently needed. Both Th1 and Th17 responses are necessary for protection against TB, yet effective adjuvants and vaccine delivery systems for inducing robust Th1 and Th17 immunity are lacking. Herein we describe a synthetic Mincle agonist, UM-1098, and a silica nanoparticle delivery system that drives Th1/Th17 responses to Mtb antigens. Stimulation of human peripheral blood mononuclear cells (hPBMCs) with UM-1098 induced high levels of Th17 polarizing cytokines IL-6, IL-1β, IL-23 as well as IL-12p70, IL-4 and TNF-α in vitro. PBMCs from both C57BL/6 and BALB/c mice responded with a similar cytokine pattern in vitro and in vivo. Importantly, intramuscular (I.M.) vaccination with UM-1098-adjuvanted TB antigen M72 resulted in significantly higher antigen-specific IFN-γ and IL-17A levels in C57BL/6 wt mice than Mincle KO mice. Vaccination of C57BL/6 wt mice with immunodominant Mtb antigens ESAT6/Ag85B or M72 resulted in predominantly Th1 and Th17 responses and induced antigen-specific serum antibodies. Notably, in a virulent Mtb challenge model, vaccination with UM-1098 adjuvanted ESAT6/Ag85B or M72 significantly reduced lung bacterial burden when compared with unvaccinated mice and protection occurred in the absence of pulmonary inflammation. These data demonstrate that the synthetic Mincle agonist UM-1098 induces strong Th1 and Th17 immunity after vaccination with Mtb antigens and provides protection against Mtb infection in mice.
Collapse
Affiliation(s)
- Viktoria Rungelrath
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Mushtaq Ahmed
- Department of Microbiology, University of Chicago, 920 E. 58th St., Chicago, IL, 60637, USA
| | - Linda Hicks
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Shannon M Miller
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kendal T Ryter
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kyle Montgomery
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - George Ettenger
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Alexander Riffey
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Walid M Abdelwahab
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Shabaana Abdul Khader
- Department of Microbiology, University of Chicago, 920 E. 58th St., Chicago, IL, 60637, USA
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA.
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA.
| |
Collapse
|
5
|
Gyu Choi H, Woong Kwon K, Jae Shin S. Importance of adjuvant selection in tuberculosis vaccine development: Exploring basic mechanisms and clinical implications. Vaccine X 2023; 15:100400. [PMID: 37965276 PMCID: PMC10641539 DOI: 10.1016/j.jvacx.2023.100400] [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: 05/02/2023] [Revised: 09/13/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
The global emergency of unexpected pathogens, exemplified by SARS-CoV-2, has emphasized the importance of vaccines in thwarting infection and curtailing the progression of severe disease. The scourge of tuberculosis (TB), emanating from the Mycobacterium tuberculosis (Mtb) complex, has inflicted a more profound toll in terms of mortality and morbidity than any other infectious agents prior to the SARS-CoV-2 pandemic. Despite the existence of Bacillus Calmette-Guérin (BCG), the only licensed vaccine developed a century ago, its efficacy against TB remains unsatisfactory, particularly in preventing pulmonary Mtb infections in adolescents and adults. However, collaborations between academic and industrial entities have led to a renewed impetus in the development of TB vaccines, with numerous candidates, particularly subunit vaccines with specialized adjuvants, exhibiting promising outcomes in recent clinical studies. Adjuvants are crucial in modulating optimal immunological responses, by endowing immune cells with sufficient antigen and immune signals. As exemplified by the COVID-19 vaccine landscape, the interplay between vaccine efficacy and adverse effects is of paramount importance, particularly for the elderly and individuals with underlying ailments such as diabetes and concurrent infections. In this regard, adjuvants hold the key to optimizing vaccine efficacy and safety. This review accentuates the pivotal roles of adjuvants and their underlying mechanisms in the development of TB vaccines. Furthermore, we expound on the prospects for the development of more efficacious adjuvants and their synergistic combinations for individuals in diverse states, such as aging, HIV co-infection, and diabetes, by examining the immunological alterations that arise with aging and comparing them with those observed in younger cohorts.
Collapse
Affiliation(s)
- Han Gyu Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| |
Collapse
|
6
|
Salunke DB, Lindsley CW. Call for Papers: Medicinal Chemistry of Next Generation Vaccine Adjuvants. J Med Chem 2023; 66:10119-10121. [PMID: 37490392 DOI: 10.1021/acs.jmedchem.3c01248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Affiliation(s)
- Deepak B Salunke
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
- National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials (NICOVIA), Panjab University, Chandigarh 160 014, India
| | - Craig W Lindsley
- Vanderbilt Institute of Chemical Biology Program in Drug Discovery, Department of Pharmacology, Vanderbilt Medical Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
7
|
Hendricksen AT, Ezzatpour S, Pulukuri AJ, Ryan AT, Flanagan TJ, Frantz W, Buchholz DW, Ortega V, Monreal IA, Sahler JM, Nielsen AE, Aguilar HC, Mancini RJ. Thermophobic Trehalose Glycopolymers as Smart C-Type Lectin Receptor Vaccine Adjuvants. Adv Healthc Mater 2023; 12:e2202918. [PMID: 37002787 PMCID: PMC11212414 DOI: 10.1002/adhm.202202918] [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: 11/21/2022] [Revised: 02/06/2023] [Indexed: 04/04/2023]
Abstract
Herein, this work reports the first synthetic vaccine adjuvants that attenuate potency in response to small, 1-2 °C changes in temperature about their lower critical solution temperature (LCST). Adjuvant additives significantly increase vaccine efficacy. However, adjuvants also cause inflammatory side effects, such as pyrexia, which currently limits their use. To address this, a thermophobic vaccine adjuvant engineered to attenuate potency at temperatures correlating to pyrexia is created. Thermophobic adjuvants are synthesized by combining a rationally designed trehalose glycolipid vaccine adjuvant with thermoresponsive poly-N-isoporpylacrylamide (NIPAM) via reversible addition fragmentation chain transfer (RAFT) polymerization. The resulting thermophobic adjuvants exhibit LCSTs near 37 °C, and self-assembled into nanoparticles with temperature-dependent sizes (90-270 nm). Thermophobic adjuvants activate HEK-mMINCLE and other innate immune cell lines as well as primary mouse bone marrow derived dendritic cells (BMDCs) and bone marrow derived macrophages (BMDMs). Inflammatory cytokine production is attenuated under conditions mimicking pyrexia (above the LCST) relative to homeostasis (37 °C) or below the LCST. This thermophobic behavior correlated with decreased adjuvant Rg is observed by DLS, as well as glycolipid-NIPAM shielding interactions are observed by NOESY-NMR. In vivo, thermophobic adjuvants enhance efficacy of a whole inactivated influenza A/California/04/2009 virus vaccine, by increasing neutralizing antibody titers and CD4+ /44+ /62L+ lung and lymph node central memory T cells, as well as providing better protection from morbidity after viral challenge relative to unadjuvanted control vaccine. Together, these results demonstrate the first adjuvants with potency regulated by temperature. This work envisions that with further investigation, this approach can enhance vaccine efficacy while maintaining safety.
Collapse
Affiliation(s)
- Aaron T. Hendricksen
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - Shahrzad Ezzatpour
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Anunay J. Pulukuri
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - Austin T. Ryan
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - Tatum J. Flanagan
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - William Frantz
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
| | - David W. Buchholz
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Victoria Ortega
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Isaac A. Monreal
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Julie M. Sahler
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Amy E. Nielsen
- Astante Therapeutics Inc.120 N Pine Street, Suite 270ASpokaneWA99202USA
| | - Hector C. Aguilar
- Department of Microbiology and ImmunologyCornell University618 Tower RoadIthacaNY14850USA
| | - Rock J. Mancini
- Department of ChemistryWashington State University1470 NE College AvePullmanWA99164USA
- Department of Chemistry and BiochemistryMiami University651 E. High StreetOxfordOH45056USA
| |
Collapse
|
8
|
Lee B, Nanishi E, Levy O, Dowling DJ. Precision Vaccinology Approaches for the Development of Adjuvanted Vaccines Targeted to Distinct Vulnerable Populations. Pharmaceutics 2023; 15:1766. [PMID: 37376214 PMCID: PMC10305121 DOI: 10.3390/pharmaceutics15061766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Infection persists as one of the leading global causes of morbidity and mortality, with particular burden at the extremes of age and in populations who are immunocompromised or suffer chronic co-morbid diseases. By focusing discovery and innovation efforts to better understand the phenotypic and mechanistic differences in the immune systems of diverse vulnerable populations, emerging research in precision vaccine discovery and development has explored how to optimize immunizations across the lifespan. Here, we focus on two key elements of precision vaccinology, as applied to epidemic/pandemic response and preparedness, including (a) selecting robust combinations of adjuvants and antigens, and (b) coupling these platforms with appropriate formulation systems. In this context, several considerations exist, including the intended goals of immunization (e.g., achieving immunogenicity versus lessening transmission), reducing the likelihood of adverse reactogenicity, and optimizing the route of administration. Each of these considerations is accompanied by several key challenges. On-going innovation in precision vaccinology will expand and target the arsenal of vaccine components for protection of vulnerable populations.
Collapse
Affiliation(s)
- Branden Lee
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
| | - Etsuro Nanishi
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David J. Dowling
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115, USA; (B.L.); (E.N.); (O.L.)
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
9
|
Igarashi Y. Development of a drug discovery approach from microbes with a special focus on isolation sources and taxonomy. J Antibiot (Tokyo) 2023:10.1038/s41429-023-00625-y. [PMID: 37188757 DOI: 10.1038/s41429-023-00625-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 05/17/2023]
Abstract
After the successful discoveries of numerous antibiotics from microorganisms, frequent reisolation of known compounds becomes an obstacle in further development of new drugs from natural products. Exploration of biological sources that can provide novel scaffolds is thus an urgent matter in drug lead screening. As an alternative source to the conventionally used soil microorganisms, we selected endophytic actinomycetes, marine actinomycetes, and actinomycetes in tropical areas for investigation and found an array of new bioactive compounds. Furthermore, based on the analysis of the distribution pattern of biosynthetic gene clusters in bacteria together with available genomic data, we speculated that biosynthetic gene clusters for secondary metabolites are specific to each genus. Based on this assumption, we investigated actinomycetal and marine bacterial genera from which no compounds have been reported, which led to the discovery of a variety of skeletally novel bioactive compounds. These findings suggest that consideration of environmental factor and taxonomic position is critically effective in the selection of potential strains producing structurally unique compounds.
Collapse
Affiliation(s)
- Yasuhiro Igarashi
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama, 939-0398, Japan.
| |
Collapse
|
10
|
Kodar K, Dangerfield EM, Foster AJ, Forsythe D, Ishizuka S, McConnell MJ, Yamasaki S, Timmer MSM, Stocker BL. Aryl-functionalised α,α'-Trehalose 6,6'-Glycolipid Induces Mincle-independent Pyroptotic Cell Death. Inflammation 2023:10.1007/s10753-023-01814-5. [PMID: 37140682 PMCID: PMC10359228 DOI: 10.1007/s10753-023-01814-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/16/2023] [Accepted: 03/31/2023] [Indexed: 05/05/2023]
Abstract
α,α'-Trehalose 6,6'-glycolipids have long been known for their immunostimulatory properties. The adjuvanticity of α,α'-trehalose 6,6'-glycolipids is mediated by signalling through the macrophage inducible C-type lectin (Mincle) and the induction of an inflammatory response. Herein, we present an aryl-functionalised trehalose glycolipid, AF-2, that leads to the release of cytokines and chemokines, including IL-6, MIP-2 and TNF-α, in a Mincle-dependent manner. Furthermore, plate-coated AF-2 also leads to the Mincle-independent production of IL-1β, which is unprecedented for this class of glycolipid. Upon investigation into the mode of action of plate-coated AF-2, it was observed that the treatment of WT and Mincle-/- bone marrow derived macrophages (BMDM), murine RAW264.7 cells, and human monocytes with AF-2 led to lytic cell death, as evidenced using Sytox Green and lactate dehydrogenase assays, and confocal and scanning electron microscopy. The requirement for functional Gasdermin D and Caspase-1 for IL-1β production and cell death by AF-2 confirmed pyroptosis as the mode of action of AF-2. The inhibition of NLRP3 and K+ efflux reduced AF-2 mediated IL-1β production and cell death, and allowed us to conclude that AF-2 leads to Capase-1 dependent NLRP3 inflammasome-mediated cell death. The unique mode of action of plate-coated AF-2 was surprising and highlights how the physical presentation of Mincle ligands can lead to dramatically different immunological outcomes.
Collapse
Affiliation(s)
- Kristel Kodar
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Emma M Dangerfield
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Amy J Foster
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Devlin Forsythe
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
- School of Biological Sciences, PO Box 600, Wellington, New Zealand
| | - Shigenari Ishizuka
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Melanie J McConnell
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
- School of Biological Sciences, PO Box 600, Wellington, New Zealand
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Division of Molecular Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Fukuoka, Japan
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Mattie S M Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| | - Bridget L Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| |
Collapse
|
11
|
Thathsaranie P Manthrirathna MA, Kodar K, Ishizuka S, Dangerfield EM, Xiuyuan L, Yamasaki S, Stocker BL, S M Timmer M. 6-C-Linked trehalose glycolipids signal through Mincle and exhibit potent adjuvant activity. Bioorg Chem 2023; 133:106345. [PMID: 36764230 DOI: 10.1016/j.bioorg.2023.106345] [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: 11/15/2022] [Revised: 12/19/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023]
Abstract
Many studies have investigated the Mincle-mediated agonist activity of α,α'-trehalose-6,6́-glycolipids, however, none have considered how the position, or absence, of the ester moiety influences Mincle-mediated agonist activity. We prepared a variety of 6-C-linked α,α'-trehalose glycolipids containing inverted esters, ketone, carboxy or no carbonyl moieties. Modelling studies indicated that these derivatives bind to the CRD of Mincle in a manner similar to that of the prototypical Mincle agonist, trehalose dibehenate (TDB), with NFAT-GFP reporter cell assays confirming that all compounds, apart from derivatives with short alkyl chains, led to robust Mincle signalling. It was also observed that a carbonyl moiety was needed for good Mincle-mediated signalling. The ability of the compounds to induce mIL-1 β and mIL-6 production by bone marrow-derived macrophages (BMDMs) further demonstrated the agonist activity of the compounds, with the presence of a carbonyl moiety and longer lipid chains augmenting cytokine production. Notably, a C20 inverted ester led to levels of mIL-1β that were significantly greater than those induced by TDB. The same C20 inverted ester also led to a significant increase in hIL-1β and hIL-6 by human monocytes, and exhibited no toxicity, as demonstrated using BMDMs in an in vitro Sytox Green assay. The ability of the inverted ester to enhance antigen-mediated immune responses was then determined. In these studies, the inverted ester was found to augment the OVA-specific Th1/Th7 immune response in vitro, and exhibit adjuvanticity that was better than that of TDB in vivo, as evidenced by a significant increase in IgG antibodies for the inverted ester but not TDB when using OVA as a model antigen.
Collapse
Affiliation(s)
| | - Kristel Kodar
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand; Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Shigenari Ishizuka
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Emma M Dangerfield
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand; Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Lu Xiuyuan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Division of Molecular Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Fukuoka, Japan; Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Bridget L Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand; Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Mattie S M Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand; Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| |
Collapse
|
12
|
Rasheed OK, Buhl C, Evans JT, Holley D, Ryter KT. Synthesis and Biological Evaluation of Trehalose-based Bi-aryl Derivatives as C-type Lectin Ligands. Tetrahedron 2023; 132:133241. [PMID: 36874612 PMCID: PMC9979692 DOI: 10.1016/j.tet.2022.133241] [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] [Indexed: 01/11/2023]
Abstract
The identification of Mincle as the C-type lectin receptor on innate immune cells responsible for binding TDM and the realization that this receptor could be key to productive vaccines for mycobacterial infection has raised interest in the development of synthetic Mincle ligands as novel adjuvants. We recently reported on the synthesis and evaluation of Brartemicin analog UM-1024 that demonstrated Mincle agonist activity, exhibiting potent Th1/Th17 adjuvant activity that was greater than that of trehalose dibehenate (TDB). Our pursuit to understand Mincle/ligand relationships and improve the pharmacologic properties of the ligands has expanded and continues to reveal new and exciting structure activity relationships. Herein we report the synthesis of novel bi-aryl trehalose derivatives in good to excellent yields. These compounds were evaluated for their ability to engage the human Mincle receptor and tested for the induction of cytokines from human peripheral blood mononuclear cells. A preliminary structure-activity relationship (SAR) of these novel bi-aryl derivatives revealed that bi-aryl trehalose ligand 3D showed relatively high potency in cytokine production in comparison to trehalose glycolipid adjuvant TDB and the natural ligand TDM and induced dose-dependent, Mincle selective stimulation in hMincle HEK reporter cells. Also, through computational studies, we provide an insight into the potential mode of binding of 6,6'-Biaryl trehalose compounds on human Mincle receptor.
Collapse
Affiliation(s)
- Omer K. Rasheed
- Department of Chemistry and Biochemistry, University of Montana, 32 campus drive, Missoula, MT, 59812, United States
- Current address: Inimmune Corp., 1121 E. Broadway St, Missoula, MT, 59808, United States
| | - Cassandra Buhl
- Center for Translational Medicine, University of Montana, 32 campus drive, Missoula, MT, 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Jay T. Evans
- Center for Translational Medicine, University of Montana, 32 campus drive, Missoula, MT, 59812, United States
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - David Holley
- Center for Bimolecular Structure and Dynamics, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - Kendal T. Ryter
- Department of Chemistry and Biochemistry, University of Montana, 32 campus drive, Missoula, MT, 59812, United States
- Center for Translational Medicine, University of Montana, 32 campus drive, Missoula, MT, 59812, United States
| |
Collapse
|
13
|
Manthrirathna MATP, Dangerfield EM, Ishizuka S, Woods A, Luong BS, Yamasaki S, Timmer MSM, Stocker BL. Water-soluble trehalose glycolipids show superior Mincle binding and signaling but impaired phagocytosis and IL-1β production. Front Mol Biosci 2022; 9:1015210. [PMID: 36504717 PMCID: PMC9729344 DOI: 10.3389/fmolb.2022.1015210] [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: 08/09/2022] [Accepted: 10/25/2022] [Indexed: 11/25/2022] Open
Abstract
The tremendous potential of trehalose glycolipids as vaccine adjuvants has incentivized the study of how the structures of these ligands relate to their Mincle-mediated agonist activities. Despite this, structure-activity work in the field has been largely empirical, and less is known about how Mincle-independent pathways might be affected by different trehalose glycolipids, and whether Mincle binding by itself can serve as a proxy for adjuvanticity. There is also much demand for more water-soluble Mincle ligands. To address this need, we prepared polyethylene glycol modified trehalose glycolipids (PEG-TGLs) with enhanced water solubility and strong murine Mincle (mMincle) binding and signaling. However, only modest cytokine and chemokine responses were observed upon the treatment of GM-CSF treated bone-marrow cells with the PEG-TGLs. Notability, no IL-1β was observed. Using RNA-Seq analysis and a representative PEG-TGL, we determined that the more water-soluble adducts were less able to activate phagocytic pathways, and hence, failed to induce IL-1β production. Taken together, our data suggests that in addition to strong Mincle binding, which is a pre-requisite for Mincle-mediated cellular responses, the physical presentation of trehalose glycolipids in colloidal form is required for inflammasome activation, and hence, a strong inflammatory immune response.
Collapse
Affiliation(s)
| | - Emma M. Dangerfield
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand,Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Shigenari Ishizuka
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan,Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Aodhamair Woods
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Brenda S. Luong
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand,Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan,Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan,Division of Molecular Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan,Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Mattie S. M. Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand,Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand,*Correspondence: Bridget L. Stocker, ; Mattie S. M. Timmer,
| | - Bridget L. Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand,Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand,*Correspondence: Bridget L. Stocker, ; Mattie S. M. Timmer,
| |
Collapse
|
14
|
Matsumaru T. Lipid Conjugates as Ligands for the C-type Lectin Receptor Mincle. TRENDS GLYCOSCI GLYC 2022. [DOI: 10.4052/tigg.2029.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
15
|
Matsumaru T. Lipid Conjugates as Ligands for the C-type Lectin Receptor Mincle. TRENDS GLYCOSCI GLYC 2022. [DOI: 10.4052/tigg.2029.1e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
16
|
Desel C, Murray PJ, Lehmann CHK, Heger L, Christensen D, Andersen P, Mack M, Dudziak D, Lang R. Monocytes Elicit a Neutrophil-Independent Th1/Th17 Response Upon Immunization With a Mincle-Dependent Glycolipid Adjuvant. Front Immunol 2022; 13:880474. [PMID: 35585969 PMCID: PMC9108773 DOI: 10.3389/fimmu.2022.880474] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/01/2022] [Indexed: 12/24/2022] Open
Abstract
Successful subunit vaccination with recombinant proteins requires adjuvants. The glycolipid trehalose-dibehenate (TDB), a synthetic analog of the mycobacterial cord factor, potently induces Th1 and Th17 immune responses and is a candidate adjuvant for human immunization. TDB binds to the C-type lectin receptor Mincle and triggers Syk-Card9-dependent APC activation. In addition, interleukin (IL)-1 receptor/MyD88-dependent signaling is required for TDB adjuvanticity. The role of different innate immune cell types in adjuvant-stimulated Th1/Th17 responses is not well characterized. We investigated cell recruitment to the site of injection (SOI) and to the draining lymph nodes (dLNs) after immunization with the TDB containing adjuvant CAF01 in a protein-based vaccine. Recruitment of monocytes and neutrophils to the SOI and the dramatic increase in lymph node cellularity was partially dependent on both Mincle and MyD88. Despite their large numbers at the SOI, neutrophils were dispensable for the induction of Th1/Th17 responses. In contrast, CCR2-dependent monocyte recruitment was essential for the induction of Th1/Th17 cells. Transport of adjuvant to the dLN did not require Mincle, MyD88, or CCR2. Together, adjuvanticity conferred by monocytes can be separated at the cellular level from potential tissue damage by neutrophils.
Collapse
Affiliation(s)
- Christiane Desel
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- *Correspondence: Christiane Desel, ; Roland Lang,
| | - Peter J. Murray
- Department of Infectious Disease, St. Jude Children’s Research Hospital, Memphis, TN, United States
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Christian H. K. Lehmann
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lukas Heger
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- *Correspondence: Christiane Desel, ; Roland Lang,
| |
Collapse
|
17
|
Matsumaru T, Sakuratani K, Yanaka S, Kato K, Yamasaki S, Fujimoto Y. Fungal β‐mannosyloxymannitol glycolipids and their analogues: synthesis and Mincle‐mediated signaling activity. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Takanori Matsumaru
- Keio University: Keio Gijuku Daigaku Faculty of Science and Technology JAPAN
| | - Kasumi Sakuratani
- Keio University Faculty of Science and Technology Graduate School of Science and Technology: Keio Gijuku Daigaku Rikogakubu Daigakuin Rikogaku Kenkyuka Faculty of Science and Technology JAPAN
| | - Saeko Yanaka
- National institutes of Natural Sciences Exploratory Research Center On Life and Living Systems (ExCELLS) and Institute for Molecular Science (IMS) JAPAN
| | - Koichi Kato
- National Institutes of Natural Sciences Exploratory Research Center On Life and Living Systems (ExCELLS) and Institute for Molecular Science (IMS) JAPAN
| | - Sho Yamasaki
- Osaka University: Osaka Daigaku Department of Molecular Immunology, Research Institute for Microbial Diseases/Laboratory of Molecular Immunology, Immunology Frontier Research Center (WPI-IFReC) JAPAN
| | - Yukari Fujimoto
- Keio University Department of Chemistry, Faculty of Science and Technology 3-14-1 Hiyoshi, Kohoku-ku 223-8522 Yokohama JAPAN
| |
Collapse
|
18
|
Cramer J. Medicinal chemistry of the myeloid C-type lectin receptors Mincle, Langerin, and DC-SIGN. RSC Med Chem 2021; 12:1985-2000. [PMID: 35024612 PMCID: PMC8672822 DOI: 10.1039/d1md00238d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/14/2021] [Indexed: 01/07/2023] Open
Abstract
In their role as pattern-recognition receptors on cells of the innate immune system, myeloid C-type lectin receptors (CLRs) assume important biological functions related to immunity, homeostasis, and cancer. As such, this family of receptors represents an appealing target for therapeutic interventions for modulating the outcome of many pathological processes, in particular related to infectious diseases. This review summarizes the current state of research into glycomimetic or drug-like small molecule ligands for the CLRs Mincle, Langerin, and DC-SIGN, which have potential therapeutic applications in vaccine research and anti-infective therapy.
Collapse
Affiliation(s)
- Jonathan Cramer
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University of Düsseldorf Universitätsstr. 1 40225 Düsseldorf Germany
| |
Collapse
|
19
|
Luo X, Lian Q, Li W, Chen L, Zhang R, Yang D, Gao L, Qi X, Liu Z, Liao G. Fully synthetic Mincle-dependent self-adjuvanting cancer vaccines elicit robust humoral and T cell-dependent immune responses and protect mice from tumor development. Chem Sci 2021; 12:15998-16013. [PMID: 35024123 PMCID: PMC8672726 DOI: 10.1039/d1sc05736g] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/15/2021] [Indexed: 12/11/2022] Open
Abstract
A new strategy based on a macrophage-inducible C-type lectin (Mincle) agonist was established to construct synthetic cancer vaccines. Using sialyl-Tn (STn) as a model antigen, four conjugates with the Mincle agonist as a built-in adjuvant were designed and synthesized through a facile and efficient method. All conjugates could induce BMDMs to produce inflammatory cytokines in a Mincle-dependent manner and were found to elicit robust humoral and T cell-dependent immune responses alone in mice. The corresponding antibodies could recognize, bind and exhibit complement-dependent cytotoxicity to STn-positive cancer cells, leading to tumor cell lysis. Moreover, all conjugates could effectively inhibit tumor growth and prolong the mice survival time in vivo, with therapeutic effects better than STn-CRM197/Al. Notably, compared to conventional glycoprotein conjugate vaccines, these fully synthetic conjugate vaccines do not cause "epitope suppression." Mincle ligands thus hold great potential as a platform for the development of new vaccine carriers with self-adjuvanting properties for cancer treatment. Preliminary structure-activity relationship analysis shows that a vaccine containing one STn antigen carried by vizantin exhibits the best efficacy, providing support for further optimization and additional investigation into Mincle agonists as the carrier of self-adjuvanting cancer vaccines.
Collapse
Affiliation(s)
- 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
| | - Qinghai Lian
- 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
| | - Wenwei 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
| | - Liqing Chen
- 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
| | - Renyu Zhang
- 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
| | - Deying Yang
- 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
| | - 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
| | - Xiaoxiao Qi
- 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
| | - 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
|
20
|
Enriquez AB, Izzo A, Miller SM, Stewart EL, Mahon RN, Frank DJ, Evans JT, Rengarajan J, Triccas JA. Advancing Adjuvants for Mycobacterium tuberculosis Therapeutics. Front Immunol 2021; 12:740117. [PMID: 34759923 PMCID: PMC8572789 DOI: 10.3389/fimmu.2021.740117] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/26/2021] [Indexed: 01/15/2023] Open
Abstract
Tuberculosis (TB) remains one of the leading causes of death worldwide due to a single infectious disease agent. BCG, the only licensed vaccine against TB, offers limited protection against pulmonary disease in children and adults. TB vaccine research has recently been reinvigorated by new data suggesting alternative administration of BCG induces protection and a subunit/adjuvant vaccine that provides close to 50% protection. These results demonstrate the need for generating adjuvants in order to develop the next generation of TB vaccines. However, development of TB-targeted adjuvants is lacking. To help meet this need, NIAID convened a workshop in 2020 titled “Advancing Vaccine Adjuvants for Mycobacterium tuberculosis Therapeutics”. In this review, we present the four areas identified in the workshop as necessary for advancing TB adjuvants: 1) correlates of protective immunity, 2) targeting specific immune cells, 3) immune evasion mechanisms, and 4) animal models. We will discuss each of these four areas in detail and summarize what is known and what we can advance on in order to help develop more efficacious TB vaccines.
Collapse
Affiliation(s)
- Ana B Enriquez
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Angelo Izzo
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Sydney, NSW, Australia
| | - Shannon M Miller
- Center for Translational Medicine, University of Montana, Missoula, MT, United States.,Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, United States
| | - Erica L Stewart
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Institute for Infectious Diseases and Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Robert N Mahon
- Division of AIDS, Columbus Technologies & Services Inc., Contractor to National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Daniel J Frank
- Division of AIDS, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, Missoula, MT, United States.,Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, United States
| | - Jyothi Rengarajan
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States.,Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States
| | - James A Triccas
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Sydney Institute for Infectious Diseases and Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
21
|
Dangerfield EM, Lynch AT, Kodar K, Stocker BL, Timmer MSM. Amide-linked brartemicin glycolipids exhibit Mincle-mediated agonist activity in vitro. Carbohydr Res 2021; 511:108461. [PMID: 34753005 DOI: 10.1016/j.carres.2021.108461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023]
Abstract
Lipidated derivatives of the natural product brartemicin show much promise as vaccine adjuvants due to their ability to signal through the Macrophage Inducible C-type Lectin (Mincle). We synthesised three lipophilic amide-linked brartemicin derivatives and compared their agonist activity to that of their ester-linked counterparts in vitro. We demonstrate that the brartemicin amide derivatives activate bone-marrow-derived macrophages (BMDMs) in a Mincle-dependent manner, as evidenced by the production of the pro-inflammatory cytokine IL-1β in wildtype but not Mincle-/- cells. The amide derivatives showed activity that was as good as, if not better than, their ester counterparts. Two of the amide derivatives, but none of the ester-derivatives, also led to the production of IL-1β by human-derived monocytes. As the production of IL-1β is a good indicator of vaccine adjuvanticity potential, these findings suggest that amide-linked brartemicin derivatives show particular promise as vaccine adjuvants.
Collapse
Affiliation(s)
- Emma M Dangerfield
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand; Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Amy T Lynch
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand; Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Kristel Kodar
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand; Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Bridget L Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand; Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| | - Mattie S M Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand; Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| |
Collapse
|
22
|
Anderluh M, Berti F, Bzducha‐Wróbel A, Chiodo F, Colombo C, Compostella F, Durlik K, Ferhati X, Holmdahl R, Jovanovic D, Kaca W, Lay L, Marinovic‐Cincovic M, Marradi M, Ozil M, Polito L, Reina‐Martin JJ, Reis CA, Sackstein R, Silipo A, Švajger U, Vaněk O, Yamamoto F, Richichi B, van Vliet SJ. Emerging glyco-based strategies to steer immune responses. FEBS J 2021; 288:4746-4772. [PMID: 33752265 PMCID: PMC8453523 DOI: 10.1111/febs.15830] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/12/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023]
Abstract
Glycan structures are common posttranslational modifications of proteins, which serve multiple important structural roles (for instance in protein folding), but also are crucial participants in cell-cell communications and in the regulation of immune responses. Through the interaction with glycan-binding receptors, glycans are able to affect the activation status of antigen-presenting cells, leading either to induction of pro-inflammatory responses or to suppression of immunity and instigation of immune tolerance. This unique feature of glycans has attracted the interest and spurred collaborations of glyco-chemists and glyco-immunologists to develop glycan-based tools as potential therapeutic approaches in the fight against diseases such as cancer and autoimmune conditions. In this review, we highlight emerging advances in this field, and in particular, we discuss on how glycan-modified conjugates or glycoengineered cells can be employed as targeting devices to direct tumor antigens to lectin receptors on antigen-presenting cells, like dendritic cells. In addition, we address how glycan-based nanoparticles can act as delivery platforms to enhance immune responses. Finally, we discuss some of the latest developments in glycan-based therapies, including chimeric antigen receptor (CAR)-T cells to achieve targeting of tumor-associated glycan-specific epitopes, as well as the use of glycan moieties to suppress ongoing immune responses, especially in the context of autoimmunity.
Collapse
Affiliation(s)
- Marko Anderluh
- Chair of Pharmaceutical ChemistryFaculty of PharmacyUniversity of LjubljanaSlovenia
| | | | - Anna Bzducha‐Wróbel
- Department of Biotechnology and Food MicrobiologyWarsaw University of Life Sciences‐SGGWPoland
| | - Fabrizio Chiodo
- Department of Molecular Cell Biology and ImmunologyCancer Center AmsterdamAmsterdam Infection and Immunity InstituteAmsterdam UMCVrije Universiteit AmsterdamNetherlands
| | - Cinzia Colombo
- Department of Chemistry and CRC Materiali Polimerici (LaMPo)University of MilanItaly
| | - Federica Compostella
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanItaly
| | - Katarzyna Durlik
- Department of Microbiology and ParasitologyJan Kochanowski UniversityKielcePoland
| | - Xhenti Ferhati
- Department of Chemistry ‘Ugo Schiff’University of FlorenceFlorenceItaly
| | - Rikard Holmdahl
- Division of Medical Inflammation ResearchDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Dragana Jovanovic
- Vinča Institute of Nuclear Sciences ‐ National Institute of the Republic of SerbiaUniversity of BelgradeSerbia
| | - Wieslaw Kaca
- Department of Microbiology and ParasitologyJan Kochanowski UniversityKielcePoland
| | - Luigi Lay
- Department of Chemistry and CRC Materiali Polimerici (LaMPo)University of MilanItaly
| | - Milena Marinovic‐Cincovic
- Vinča Institute of Nuclear Sciences ‐ National Institute of the Republic of SerbiaUniversity of BelgradeSerbia
| | - Marco Marradi
- Department of Chemistry ‘Ugo Schiff’University of FlorenceFlorenceItaly
| | - Musa Ozil
- Department of ChemistryFaculty of Arts and SciencesRecep Tayyip Erdogan University RizeTurkey
| | | | | | - Celso A. Reis
- I3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortugal
- IPATIMUP‐Institute of Molecular Pathology and ImmunologyInstituto de Ciências Biomédicas Abel SalazarUniversity of PortoPortugal
| | - Robert Sackstein
- Department of Translational Medicinethe Translational Glycobiology InstituteHerbert Wertheim College of MedicineFlorida International UniversityMiamiFLUSA
| | - Alba Silipo
- Department of Chemical SciencesUniversity of Naples Federico IIComplesso Universitario Monte Sant’AngeloNapoliItaly
| | - Urban Švajger
- Blood Transfusion Center of SloveniaLjubljanaSlovenia
| | - Ondřej Vaněk
- Department of BiochemistryFaculty of ScienceCharles UniversityPragueCzech Republic
| | - Fumiichiro Yamamoto
- Immunohematology & Glycobiology LaboratoryJosep Carreras Leukaemia Research InstituteBadalonaSpain
| | - Barbara Richichi
- Department of Chemistry ‘Ugo Schiff’University of FlorenceFlorenceItaly
| | - Sandra J. van Vliet
- Department of Molecular Cell Biology and ImmunologyCancer Center AmsterdamAmsterdam Infection and Immunity InstituteAmsterdam UMCVrije Universiteit AmsterdamNetherlands
| |
Collapse
|
23
|
In-vivo expressed Mycobacterium tuberculosis antigens recognised in three mouse strains after infection and BCG vaccination. NPJ Vaccines 2021; 6:81. [PMID: 34083546 PMCID: PMC8175414 DOI: 10.1038/s41541-021-00343-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 04/23/2021] [Indexed: 01/15/2023] Open
Abstract
Novel tuberculosis (TB)-vaccines preferably should (i) boost host immune responses induced by previous BCG vaccination and (ii) be directed against Mycobacterium tuberculosis (Mtb) proteins expressed throughout the Mtb infection-cycle. Human Mtb antigen-discovery screens identified antigens encoded by Mtb-genes highly expressed during in vivo murine infection (IVE-TB antigens). To translate these findings towards animal models, we determined which IVE-TB-antigens are recognised by T-cells following Mtb challenge or BCG vaccination in three different mouse strains. Eleven Mtb-antigens were recognised across TB-resistant and susceptible mice. Confirming previous human data, several Mtb-antigens induced cytokines other than IFN-γ. Pulmonary cells from susceptible C3HeB/FeJ mice produced less TNF-α, agreeing with the TB-susceptibility phenotype. In addition, responses to several antigens were induced by BCG in C3HeB/FeJ mice, offering potential for boosting. Thus, recognition of promising Mtb-antigens identified in humans validates across multiple mouse TB-infection models with widely differing TB-susceptibilities. This offers translational tools to evaluate IVE-TB-antigens as diagnostic and vaccine antigens.
Collapse
|
24
|
Rasheed OK, Buhl C, Evans JT, Ryter KT. Design of Trehalose-Based Amide/Sulfonamide C-type Lectin Receptor Signaling Compounds. ChemMedChem 2021; 16:1246-1251. [PMID: 33415819 PMCID: PMC8068603 DOI: 10.1002/cmdc.202000775] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/06/2021] [Indexed: 02/06/2023]
Abstract
Mincle agonists have been shown to induce inflammatory cytokine production, such as tumor necrosis factor-alpha (TNF) and promote the development of a Th1/Th17 immune response that might be crucial to development of effective vaccination against pathogens such as Mycobacterium tuberculosis. As an expansion of our previous work, a library of 6,6'-amide and sulfonamide α,α-d-trehalose compounds with various substituents on the aromatic ring was synthesized efficiently in good to excellent yields. These compounds were evaluated for their ability to activate the human C-type lectin receptor Mincle by the induction of cytokines from human peripheral blood mononuclear cells. A preliminary structure-activity relationship (SAR) of these novel trehalose diamides and sulfonamides revealed that aryl amide-linked trehalose compounds demonstrated improved activity and relatively high potency cytokine production compared to the Mincle ligand trehalose dibehenate adjuvant (TDB) and the natural ligand trehalose dimycolate (TDM) inducing dose-dependent and human-Mincle-specific stimulation in a HEK reporter cell line.
Collapse
Affiliation(s)
- Omer K Rasheed
- Department of Chemistry and Biochemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
- Inimmune Corp., 1121 E. Broadway, Suite 121, Missoula, MT 59802, USA
| | - Cassandra Buhl
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
| | - Jay T Evans
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
| | - Kendal T Ryter
- Department of Chemistry and Biochemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA
| |
Collapse
|
25
|
Trehalose diamide glycolipids augment antigen-specific antibody responses in a Mincle-dependent manner. Bioorg Chem 2021; 110:104747. [PMID: 33799177 DOI: 10.1016/j.bioorg.2021.104747] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/15/2021] [Accepted: 02/13/2021] [Indexed: 12/11/2022]
Abstract
Many studies have investigated how trehalose glycolipid structures can be modified to improve their Macrophage inducible C-type lectin (Mincle)-mediated adjuvanticity. However, in all instances, the ester-linkage of α,ά-trehalose to the lipid of choice remained. We investigated how changing this ester-linkage to an amide influences Mincle signalling and agonist activity and demonstrated that Mincle tolerates this functional group change. In in vivo vaccination studies in murine and ovine model systems, using OVA or Mannheimia haemolytica and Mycoplasma ovipneumoniae as vaccine antigens, respectively, it was demonstrated that a representative trehalose diamide glycolipid was able to enhance antibody-specific immune responses. Notably, IgG titres against M. ovipneumoniae were significantly greater when using trehalose dibehenamide (A-TDB) compared to trehalose dibehenate (TDB). This is particularly important as infection with M. ovipneumoniae predisposes sheep to pneumonia.
Collapse
|
26
|
Abdelwahab WM, Riffey A, Buhl C, Johnson C, Ryter K, Evans JT, Burkhart DJ. Co-adsorption of synthetic Mincle agonists and antigen to silica nanoparticles for enhanced vaccine activity: A formulation approach to co-delivery. Int J Pharm 2020; 593:120119. [PMID: 33249249 DOI: 10.1016/j.ijpharm.2020.120119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/12/2020] [Accepted: 11/22/2020] [Indexed: 01/14/2023]
Abstract
To date there is no clinically approved adjuvant to drive a protective T-helper cell 17 (Th17) immune response against Mycobacterium tuberculosis (Mtb). Trehalose Dimycolate (TDM) is a glycolipid molecule found in the cell wall of Mtb and similar species. Our team has discovered novel synthetic TDM derivatives that target Mincle receptors and when presented on the surface of amine functionalized silica nanoparticles (A-SNPs) adopt the requisite supramolecular structure for Mincle receptor agonism. Here we describe the preparation and characterization methods for these critical silica nanoparticles (SNPs) co-loaded with Mincle agonists (MAs) and a model antigen. In this work, A-SNPs with a particle diameter of 246 ± 11 nm were prepared and examined for co-adsorption of two synthetic MAs along with ovalbumin (OVA). Due to the insolubility of the studied MAs in aqueous environment, aggregation of the MAs made separation of the adjuvant-loaded A-SNPs from the free-form MAs via centrifugation very challenging. To facilitate separation, we synthesized modified SNPs with comparable amine surface functionalization to the original A-SNPs, but with a superparamagnetic iron oxide core (M-A-SNPs), to allow for magnetic separation. We also substituted Alexa Fluor 488-labeled ovalbumin (AF 488 OVA) for the un-tagged OVA to improve the sensitivity of our quantitation method. A RP-HPLC method was developed to simultaneously determine the amount of adsorption of both the Mincle adjuvant and the model antigen to the A-SNPs. AF488 OVA demonstrated higher than 90% adsorption, with or without the co-adsorption of MAs. Likewise, MAs exhibited higher than 80% adsorption in the presence or absence of antigen. The developed formulations were tested in vitro using murine RAW cells and human peripheral blood mononuclear cells, exhibiting good cytokine induction in both cell lines. Results from these studies indicate that A-SNPs could be used as a customizable presentation platform to co-deliver antigens along with different MAs of varying structural features and biophysical properties.
Collapse
Affiliation(s)
- Walid M Abdelwahab
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Alexander Riffey
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Cassie Buhl
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Craig Johnson
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Chemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Kendal Ryter
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Chemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - David J Burkhart
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States.
| |
Collapse
|
27
|
O'Hagan DT, Lodaya RN, Lofano G. The continued advance of vaccine adjuvants - 'we can work it out'. Semin Immunol 2020; 50:101426. [PMID: 33257234 DOI: 10.1016/j.smim.2020.101426] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/20/2020] [Accepted: 11/16/2020] [Indexed: 12/19/2022]
Abstract
In the last decade there have been some significant advances in vaccine adjuvants, particularly in relation to their inclusion in licensed products. This was proceeded by several decades in which such advances were very scarce, or entirely absent, but several novel adjuvants have now been included in licensed products, including in the US. These advances have relied upon several key technological insights that have emerged in this time period, which have finally allowed an in depth understanding of how adjuvants work. These advances include developments in systems biology approaches which allow the hypotheses first advanced in pre-clinical studies to be critically evaluated in human studies. This review highlights these recent advances, both in relation to the adjuvants themselves, but also the technologies that have enabled their successes. Moreover, we critically appraise what will come next, both in terms of new adjuvant molecules, and the technologies needed to allow them to succeed. We confidently predict that additional adjuvants will emerge in the coming years that will reach approval in licensed products, but that the components might differ significantly from those which are currently used. Gradually, the natural products that were originally used to build adjuvants, since they were readily available at the time of initial development, will come to be replaced by synthetic or biosynthetic materials, with more appealing attributes, including more reliable and robust supply, along with reduced heterogeneity. The recent advance in vaccine adjuvants is timely, given the need to create novel vaccines to deal with the COVID-19 pandemic. Although, we must ensure that the rigorous safety evaluations that allowed the current adjuvants to advance are not 'short-changed' in the push for new vaccines to meet the global challenge as quickly as possible, we must not jeopardize what we have achieved, by pushing less established technologies too quickly, if the data does not fully support it.
Collapse
Affiliation(s)
- Derek T O'Hagan
- GSK, Slaoui Center for Vaccines Research, Rockville, MD, 20850, USA
| | - Rushit N Lodaya
- GSK, Slaoui Center for Vaccines Research, Rockville, MD, 20850, USA
| | - Giuseppe Lofano
- GSK, Slaoui Center for Vaccines Research, Rockville, MD, 20850, USA.
| |
Collapse
|
28
|
Izumi S, Kobayashi Y, Takemoto Y. Stereoselective Synthesis of 1,1′‐Disaccharides by Organoboron Catalysis. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202004476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Sanae Izumi
- Graduate School of Pharmaceutical Sciences Kyoto University 46-29 Shimoadachi-cho, Yoshida, Sakyo-ku Kyoto 606-8501 Japan
| | - Yusuke Kobayashi
- Graduate School of Pharmaceutical Sciences Kyoto University 46-29 Shimoadachi-cho, Yoshida, Sakyo-ku Kyoto 606-8501 Japan
| | - Yoshiji Takemoto
- Graduate School of Pharmaceutical Sciences Kyoto University 46-29 Shimoadachi-cho, Yoshida, Sakyo-ku Kyoto 606-8501 Japan
| |
Collapse
|
29
|
Lindenwald DL, Lepenies B. C-Type Lectins in Veterinary Species: Recent Advancements and Applications. Int J Mol Sci 2020; 21:ijms21145122. [PMID: 32698416 PMCID: PMC7403975 DOI: 10.3390/ijms21145122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 02/06/2023] Open
Abstract
C-type lectins (CTLs), a superfamily of glycan-binding receptors, play a pivotal role in the host defense against pathogens and the maintenance of immune homeostasis of higher animals and humans. CTLs in innate immunity serve as pattern recognition receptors and often bind to glycan structures in damage- and pathogen-associated molecular patterns. While CTLs are found throughout the whole animal kingdom, their ligand specificities and downstream signaling have mainly been studied in humans and in model organisms such as mice. In this review, recent advancements in CTL research in veterinary species as well as potential applications of CTL targeting in veterinary medicine are outlined.
Collapse
|
30
|
Rasheed OK, Ettenger G, Buhl C, Child R, Miller SM, Evans JT, Ryter KT. 6,6'-Aryl trehalose analogs as potential Mincle ligands. Bioorg Med Chem 2020; 28:115564. [PMID: 32616186 PMCID: PMC7372699 DOI: 10.1016/j.bmc.2020.115564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/27/2020] [Accepted: 05/20/2020] [Indexed: 12/17/2022]
Abstract
6,6'-Aryl trehalose derivatives have been synthesized with a view towards identifying novel Th-17-inducing vaccine adjuvants based on the high affinity Mincle ligand Brartemicin. The initial structure-activity relationships of these novel trehalose-based compounds were investigated. All compounds have been evaluated for their ability to engage the Mincle receptor and induce a potential pro-Th17 cytokine profile from human peripheral blood mononuclear cells based on IL-6 production in human peripheral blood mononuclear cells. The preliminary biological characterization of the designed analogs presented in this paper should aid in the future design and testing of more affine ligands that may foster the discovery of novel adjuvants with improved pharmacological properties.
Collapse
Affiliation(s)
- Omer K Rasheed
- Department of Chemistry and Biochemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - George Ettenger
- Department of Chemistry and Biochemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Cassandra Buhl
- Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Robert Child
- Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Shannon M Miller
- Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Kendal T Ryter
- Department of Chemistry and Biochemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States.
| |
Collapse
|
31
|
Stereoselective Synthesis of 1,1′‐Disaccharides by Organoboron Catalysis. Angew Chem Int Ed Engl 2020; 59:14054-14059. [DOI: 10.1002/anie.202004476] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Indexed: 12/12/2022]
|