A Novel Synthetic Dual Agonistic Liposomal TLR4/7 Adjuvant Promotes Broad Immune Responses in an Influenza Vaccine With Minimal Reactogenicity

The limited efficacy of seasonal influenza vaccines is usually attributed to ongoing variation in the major antigenic targets for protective antibody responses including hemagglutinin (HA) and neuraminidase (NA). Hence, vaccine development has largely focused on broadening antigenic epitopes to gene...

Full description

Bibliographic Details
Main Authors: Fumi Sato-Kaneko, Shiyin Yao, Fitzgerald S. Lao, Jonathan Shpigelman, Karen Messer, Minya Pu, Nikunj M. Shukla, Howard B. Cottam, Michael Chan, Paul J. Chu, David Burkhart, Roman Schoener, Takaji Matsutani, Dennis A. Carson, Maripat Corr, Tomoko Hayashi
Format: Article
Language:English
Published: Frontiers Media S.A. 2020-06-01
Series:Frontiers in Immunology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fimmu.2020.01207/full
id doaj-878ad460cb7e47e1886c8e90719228b5
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Fumi Sato-Kaneko
Shiyin Yao
Fitzgerald S. Lao
Jonathan Shpigelman
Karen Messer
Minya Pu
Nikunj M. Shukla
Howard B. Cottam
Michael Chan
Paul J. Chu
David Burkhart
Roman Schoener
Takaji Matsutani
Dennis A. Carson
Maripat Corr
Tomoko Hayashi
spellingShingle Fumi Sato-Kaneko
Shiyin Yao
Fitzgerald S. Lao
Jonathan Shpigelman
Karen Messer
Minya Pu
Nikunj M. Shukla
Howard B. Cottam
Michael Chan
Paul J. Chu
David Burkhart
Roman Schoener
Takaji Matsutani
Dennis A. Carson
Maripat Corr
Tomoko Hayashi
A Novel Synthetic Dual Agonistic Liposomal TLR4/7 Adjuvant Promotes Broad Immune Responses in an Influenza Vaccine With Minimal Reactogenicity
Frontiers in Immunology
vaccine
combination adjuvant
synthetic TLR4 agonist
synthetic TLR7 agonists
small molecule
influenza virus infection
author_facet Fumi Sato-Kaneko
Shiyin Yao
Fitzgerald S. Lao
Jonathan Shpigelman
Karen Messer
Minya Pu
Nikunj M. Shukla
Howard B. Cottam
Michael Chan
Paul J. Chu
David Burkhart
Roman Schoener
Takaji Matsutani
Dennis A. Carson
Maripat Corr
Tomoko Hayashi
author_sort Fumi Sato-Kaneko
title A Novel Synthetic Dual Agonistic Liposomal TLR4/7 Adjuvant Promotes Broad Immune Responses in an Influenza Vaccine With Minimal Reactogenicity
title_short A Novel Synthetic Dual Agonistic Liposomal TLR4/7 Adjuvant Promotes Broad Immune Responses in an Influenza Vaccine With Minimal Reactogenicity
title_full A Novel Synthetic Dual Agonistic Liposomal TLR4/7 Adjuvant Promotes Broad Immune Responses in an Influenza Vaccine With Minimal Reactogenicity
title_fullStr A Novel Synthetic Dual Agonistic Liposomal TLR4/7 Adjuvant Promotes Broad Immune Responses in an Influenza Vaccine With Minimal Reactogenicity
title_full_unstemmed A Novel Synthetic Dual Agonistic Liposomal TLR4/7 Adjuvant Promotes Broad Immune Responses in an Influenza Vaccine With Minimal Reactogenicity
title_sort novel synthetic dual agonistic liposomal tlr4/7 adjuvant promotes broad immune responses in an influenza vaccine with minimal reactogenicity
publisher Frontiers Media S.A.
series Frontiers in Immunology
issn 1664-3224
publishDate 2020-06-01
description The limited efficacy of seasonal influenza vaccines is usually attributed to ongoing variation in the major antigenic targets for protective antibody responses including hemagglutinin (HA) and neuraminidase (NA). Hence, vaccine development has largely focused on broadening antigenic epitopes to generate cross-reactive protection. However, the vaccine adjuvant components which can accelerate, enhance and prolong antigenic immune responses, can also increase the breadth of these responses. We previously demonstrated that the combination of synthetic small-molecule Toll-like receptor 4 (TLR4) and TLR7 ligands is a potent adjuvant for recombinant influenza virus HA, inducing rapid, and sustained antibody responses that are protective against influenza viruses in homologous and heterologous murine challenge models. To further enhance adjuvant efficacy, we performed a structure-activity relationship study for the TLR4 ligand, N-cyclohexyl-2-((5-methyl-4-oxo-3-phenyl-4,5-dihydro-3H-pyrimido[5,4-b]indol-2-yl)thio)acetamide (C25H26N4O2S; 1Z105), and identified the 8-(furan-2-yl) substituted pyrimido[5,4-b]indole analog (C29H28N4O3S; 2B182C) as a derivative with higher potency in activating both human and mouse TLR4-NF-κB reporter cells and primary cells. In a prime-boost immunization model using inactivated influenza A virus [IIAV; A/California/04/2009 (H1N1)pdm09], 2B182C used as adjuvant induced higher serum anti-HA and anti-NA IgG1 levels compared to 1Z105, and also increased the anti-NA IgG2a responses. In combination with a TLR7 ligand, 1V270, 2B182C induced equivalent levels of anti-NA and anti-HA IgG1 to 1V270+1Z105. However, the combination of 1V270+2B182C induced 10-fold higher anti-HA and anti-NA IgG2a levels compared to 1V270+1Z105. A stable liposomal formulation of 1V270+2B182C was developed, which synergistically enhanced anti-HA and anti-NA IgG1 and IgG2a responses without demonstrable reactogenicity after intramuscular injection. Notably, vaccination with IIAV plus the liposomal formulation of 1V270+2B182C protected mice against lethal homologous influenza virus (H1N1)pdm09 challenge and reduced lung viral titers and cytokine levels. The combination adjuvant induced a greater diversity in B cell clonotypes of immunoglobulin heavy chain (IGH) genes in the draining lymph nodes and antibodies against a broad spectrum of HA epitopes encompassing HA head and stalk domains and with cross-reactivity against different subtypes of HA and NA. This novel combination liposomal adjuvant contributes to a more broadly protective vaccine while demonstrating an attractive safety profile.
topic vaccine
combination adjuvant
synthetic TLR4 agonist
synthetic TLR7 agonists
small molecule
influenza virus infection
url https://www.frontiersin.org/article/10.3389/fimmu.2020.01207/full
work_keys_str_mv AT fumisatokaneko anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT shiyinyao anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT fitzgeraldslao anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT jonathanshpigelman anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT karenmesser anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT minyapu anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT nikunjmshukla anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT howardbcottam anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT michaelchan anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT pauljchu anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT davidburkhart anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT romanschoener anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT takajimatsutani anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT dennisacarson anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT maripatcorr anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT tomokohayashi anovelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT fumisatokaneko novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT shiyinyao novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT fitzgeraldslao novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT jonathanshpigelman novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT karenmesser novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT minyapu novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT nikunjmshukla novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT howardbcottam novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT michaelchan novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT pauljchu novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT davidburkhart novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT romanschoener novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT takajimatsutani novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT dennisacarson novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT maripatcorr novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
AT tomokohayashi novelsyntheticdualagonisticliposomaltlr47adjuvantpromotesbroadimmuneresponsesinaninfluenzavaccinewithminimalreactogenicity
_version_ 1724627885868711936
spelling doaj-878ad460cb7e47e1886c8e90719228b52020-11-25T03:18:15ZengFrontiers Media S.A.Frontiers in Immunology1664-32242020-06-011110.3389/fimmu.2020.01207545505A Novel Synthetic Dual Agonistic Liposomal TLR4/7 Adjuvant Promotes Broad Immune Responses in an Influenza Vaccine With Minimal ReactogenicityFumi Sato-Kaneko0Shiyin Yao1Fitzgerald S. Lao2Jonathan Shpigelman3Karen Messer4Minya Pu5Nikunj M. Shukla6Howard B. Cottam7Michael Chan8Paul J. Chu9David Burkhart10Roman Schoener11Takaji Matsutani12Dennis A. Carson13Maripat Corr14Tomoko Hayashi15Moores Cancer Center, University of California, San Diego, La Jolla, CA, United StatesMoores Cancer Center, University of California, San Diego, La Jolla, CA, United StatesMoores Cancer Center, University of California, San Diego, La Jolla, CA, United StatesMoores Cancer Center, University of California, San Diego, La Jolla, CA, United StatesDivision of Biostatistics, University of California, San Diego, La Jolla, CA, United StatesDivision of Biostatistics, University of California, San Diego, La Jolla, CA, United StatesMoores Cancer Center, University of California, San Diego, La Jolla, CA, United StatesMoores Cancer Center, University of California, San Diego, La Jolla, CA, United StatesMoores Cancer Center, University of California, San Diego, La Jolla, CA, United StatesMoores Cancer Center, University of California, San Diego, La Jolla, CA, United StatesInimmune Corp., Missoula, MT, United StatesInimmune Corp., Missoula, MT, United StatesRepertoire Genesis Inc., Saito-Asagai, Osaka, JapanMoores Cancer Center, University of California, San Diego, La Jolla, CA, United StatesDepartment of Medicine, University of California, San Diego, La Jolla, CA, United StatesMoores Cancer Center, University of California, San Diego, La Jolla, CA, United StatesThe limited efficacy of seasonal influenza vaccines is usually attributed to ongoing variation in the major antigenic targets for protective antibody responses including hemagglutinin (HA) and neuraminidase (NA). Hence, vaccine development has largely focused on broadening antigenic epitopes to generate cross-reactive protection. However, the vaccine adjuvant components which can accelerate, enhance and prolong antigenic immune responses, can also increase the breadth of these responses. We previously demonstrated that the combination of synthetic small-molecule Toll-like receptor 4 (TLR4) and TLR7 ligands is a potent adjuvant for recombinant influenza virus HA, inducing rapid, and sustained antibody responses that are protective against influenza viruses in homologous and heterologous murine challenge models. To further enhance adjuvant efficacy, we performed a structure-activity relationship study for the TLR4 ligand, N-cyclohexyl-2-((5-methyl-4-oxo-3-phenyl-4,5-dihydro-3H-pyrimido[5,4-b]indol-2-yl)thio)acetamide (C25H26N4O2S; 1Z105), and identified the 8-(furan-2-yl) substituted pyrimido[5,4-b]indole analog (C29H28N4O3S; 2B182C) as a derivative with higher potency in activating both human and mouse TLR4-NF-κB reporter cells and primary cells. In a prime-boost immunization model using inactivated influenza A virus [IIAV; A/California/04/2009 (H1N1)pdm09], 2B182C used as adjuvant induced higher serum anti-HA and anti-NA IgG1 levels compared to 1Z105, and also increased the anti-NA IgG2a responses. In combination with a TLR7 ligand, 1V270, 2B182C induced equivalent levels of anti-NA and anti-HA IgG1 to 1V270+1Z105. However, the combination of 1V270+2B182C induced 10-fold higher anti-HA and anti-NA IgG2a levels compared to 1V270+1Z105. A stable liposomal formulation of 1V270+2B182C was developed, which synergistically enhanced anti-HA and anti-NA IgG1 and IgG2a responses without demonstrable reactogenicity after intramuscular injection. Notably, vaccination with IIAV plus the liposomal formulation of 1V270+2B182C protected mice against lethal homologous influenza virus (H1N1)pdm09 challenge and reduced lung viral titers and cytokine levels. The combination adjuvant induced a greater diversity in B cell clonotypes of immunoglobulin heavy chain (IGH) genes in the draining lymph nodes and antibodies against a broad spectrum of HA epitopes encompassing HA head and stalk domains and with cross-reactivity against different subtypes of HA and NA. This novel combination liposomal adjuvant contributes to a more broadly protective vaccine while demonstrating an attractive safety profile.https://www.frontiersin.org/article/10.3389/fimmu.2020.01207/fullvaccinecombination adjuvantsynthetic TLR4 agonistsynthetic TLR7 agonistssmall moleculeinfluenza virus infection