Adjuvant-enhanced CD4 T Cell Responses are Critical to Durable Vaccine Immunity

Karen A O Martins, Christopher L Cooper, Sabrina M Stronsky, Sarah L W Norris, Steven A Kwilas, Jesse T Steffens, Jacqueline G Benko, Sean A van Tongeren, Sina Bavari, Karen A O Martins, Christopher L Cooper, Sabrina M Stronsky, Sarah L W Norris, Steven A Kwilas, Jesse T Steffens, Jacqueline G Benko, Sean A van Tongeren, Sina Bavari

Abstract

Protein-based vaccines offer a safer alternative to live-attenuated or inactivated vaccines but have limited immunogenicity. The identification of adjuvants that augment immunogenicity, specifically in a manner that is durable and antigen-specific, is therefore critical for advanced development. In this study, we use the filovirus virus-like particle (VLP) as a model protein-based vaccine in order to evaluate the impact of four candidate vaccine adjuvants on enhancing long term protection from Ebola virus challenge. Adjuvants tested include poly-ICLC (Hiltonol), MPLA, CpG 2395, and alhydrogel. We compared and contrasted antibody responses, neutralizing antibody responses, effector T cell responses, and T follicular helper (Tfh) cell frequencies with each adjuvant's impact on durable protection. We demonstrate that in this system, the most effective adjuvant elicits a Th1-skewed antibody response and strong CD4 T cell responses, including an increase in Tfh frequency. Using immune-deficient animals and adoptive transfer of serum and cells from vaccinated animals into naïve animals, we further demonstrate that serum and CD4 T cells play a critical role in conferring protection within effective vaccination regimens. These studies inform on the requirements of long term immune protection, which can potentially be used to guide screening of clinical-grade adjuvants for vaccine clinical development.

Keywords: Adjuvant; BME, beta mercaptoethanol; CD, cluster of differentiation; DSCF, Dwass, Steel, Critchlow-Fligner; Durable protection; ELISA, Enzyme linked immunosorbent assay; ELISPOT, enzyme-linked immunospot assay; Ebola virus; FACS, fluorescence activated cell sorting; FBS, fetal bovine serum; GP, glycoprotein; IACUC, Institutional Animal Care and Use Committee; IM, intramuscular; IP, intraperitoneal; IQR, interquartile range; Immune correlates; LN, lymph node; MPLA, monophosphoryl lipid A; NAb, neutralizing antibody; Ns, not significant; PBS, phosphate buffered saline; PRR, pattern recognition receptor; Pfu, plaque forming unit; PsVNA, pseudovirion neutralization assay; TLR, Toll-like receptor; USAMRIID, United States Army Medical Research Institute of Infectious Diseases; VLP, virus-like particle; Vaccine; ma-EBOV, mouse-adapted Ebola virus.

Figures

Fig. 1
Supplemental Fig. 1 Effective adjuvants increase the frequency of whole glycoprotein-specific T cells. IFNγ ELISPOT analysis of splenocytes from C57BL/6 mice vaccinated two times with indicated vaccine and adjuvant combination. Splenocytes were collected on day 4 or day 147 after the second vaccination, which occurred on day 21. Data is pooled from two separate experiments each containing 3–4 mice per group, as described in Fig. 3a and d. Recombinant Ebola GP was used as antigen. Mean and IQR shown. Comparisons between animals receiving VLP with or with adjuvant are shown, where “*” indicates 0.01 

Fig. 1

Adjuvants impact the durability of…

Fig. 1

Adjuvants impact the durability of protection conferred by eVLP vaccination. (a) C57BL/6 mice…

Fig. 1
Adjuvants impact the durability of protection conferred by eVLP vaccination. (a) C57BL/6 mice were vaccinated IM two times with 10 μg of eVLP and challenged four (short-term) weeks or twenty-two (long-term) weeks after the vaccine boost. Data in A are pooled from 8 individual studies with 6–10 animals/group. Fisher's exact test: survival in the short-term group was significantly higher than in the long-term group (p 

Fig. 2

Adjuvants have variable impact on…

Fig. 2

Adjuvants have variable impact on IgG subclasses and antibody neutralization. (a) Serum was…

Fig. 2
Adjuvants have variable impact on IgG subclasses and antibody neutralization. (a) Serum was collected 14 days and 147 days after the vaccine boost (days 35 and 168, respectively) and evaluated for anti-GP IgG, IgG1, IgG2c, and IgG3 levels using an ELISA. Data shown are pooled from at least two separate experiments per group. (b) Pairwise comparison using DSCF multiple pairwise comparison was used and p values greater than 0.05 are shown as “ns”. (c) Summary of results shown in A, B and D. (d) Neutralizing antibody titers were evaluated using the PsVNA, with titers giving 80% neutralization shown; median and IQR shown. Samples within each group were selected randomly from three separate studies for evaluation in the assay. Pairwise comparison using post-hoc Tukey's studentized range test procedure was used to evaluate differences between groups at both days 35 and day 168, where “*” indicates 0.01 

Fig. 3

Effective adjuvants increase the frequency…

Fig. 3

Effective adjuvants increase the frequency of antigen-specific T cells. (a) IFNγ ELISPOT analysis…

Fig. 3
Effective adjuvants increase the frequency of antigen-specific T cells. (a) IFNγ ELISPOT analysis of splenocytes from C57BL/6 mice vaccinated two times with indicated vaccine and adjuvant combination. Splenocytes were collected on day 4 after the second vaccination. Data is pooled from four separate experiments each containing 2–3 mice per group; median with IQR shown. (B&C) Frequency of IFNγ +, IL2 +, or TNFα + cells after vaccination with VLP and polyICLC or VLP and CpG; median shown. Gating is on viable T cells and then CD4 + CD44high T cells (b) or CD8 + CD44high T cells (c). (d) IFNγ ELISPOT analysis of splenocytes from C57BL/6 mice vaccinated with indicated vaccine and adjuvant combination. Animals received the standard two vaccinations and then received a third vaccine boost 22 weeks after the second vaccination. Splenocytes were collected 4 days later. Data is pooled from two separate experiments each containing 3–4 mice per group; median with IQR shown. (e) Four weeks after the second vaccination, splenocytes were collected from animals and T cells were isolated using negative bead selection. T cells were then sorted to collect central memory, effector memory, and CD44int/low cell populations. Gating strategy is shown. (f) Sorted T cells were cultured with peptide-exposed, T cell-depleted, naïve splenocytes and the frequency of cytokine-positive CD4 and CD8 T cells in each sorted population was quantified after 5 days of culture. Gating is on CD4 or CD8 T cells and data shown are the frequency of cells expressing IFNγ, TNFα, or IL2; median is shown. “*” indicates 0.01 

Fig. 4

CD8 T cell deficiency does…

Fig. 4

CD8 T cell deficiency does not impact short term or long term survival…

Fig. 4
CD8 T cell deficiency does not impact short term or long term survival of vaccinated C57BL/6J mice. (a) Mice were treated IM twice with saline, VLP, VLP and polyICLC, or VLP and CpG. Four weeks after the second vaccination, mice were challenged. Closed symbols represent wild type C57BL/6J mice and open symbols indicate CD8-deficient mice. (b) Mice were vaccinated on the same schedule as in A, but challenge occurred 22 weeks after the second vaccination. (c) Two weeks after the second vaccination and 1 week prior to challenge, blood was collected from vaccinated animals and evaluated for anti-GP IgG antibody titers. (D) A subset of vaccinated animals was euthanized 4 days after the vaccine boost to evaluate CD4 + T cell responses. Median response of C57BL/6J mice and CD8-deficient mice is shown. N = 8–10 per treated group for A–C and D presents data pooled from two separate evaluations of 4–8 mice per group each. “*” indicates 0.005 

Fig. 5

Tfh frequencies are increased by…

Fig. 5

Tfh frequencies are increased by adjuvants associated with protection from challenge. C57BL/6 mice…

Fig. 5
Tfh frequencies are increased by adjuvants associated with protection from challenge. C57BL/6 mice were vaccinated a single time IM with VLP, with or without adjuvant. Seven days after vaccination, the draining popliteal LN was isolated. (a) Untouched T cells were isolated from the LN (n = 3 or 4/group). RNA was isolated from the purified T cells and pooled for each vaccination group. Samples were then evaluated using the SABiosciences T and B cell activation PCR array in triplicate. For the volcano plot, the fold difference is the average of triplicate, and the p value was calculated comparing animals vaccinated with VLP to those vaccinated with VLP and polyICLC. (b) Seven days after vaccination, cells from the draining LN were evaluated for Tfh populations. Cells were gated on viable lymphocytes after doublet exclusion, and then on CD4 + T cells expressing CXCR5 and PD1. Median and IQR shown. (c) CD3 + CD4 + CXCR5 + PD1 + cell population in mice vaccinated with VLP and polyICLC. ICOS and Bcl6 expression of this population is shown. Data shown are pooled from three separate vaccination experiments. Comparisons between animals receiving VLP with or with adjuvant are shown, where “*” indicates 0.01 
Similar articles
Cited by
References
    1. Agnandji S.T., Huttner A., Zinser M.E., Njuguna P., Dahlke C., Fernandes J.F., Yerly S., Dayer J.A., Kraehling V., Kasonta R., Adegnika A.A., Altfeld M., Auderset F., Bache E.B., Biedenkopf N., Borregaard S., Brosnahan J.S., Burrow R., Combescure C., Desmeules J., Eickmann M., Fehling S.K., Finckh A., Goncalves A.R., Grobusch M.P., Hooper J., Jambrecina A., Kabwende A.L., Kaya G., Kimani D., Lell B., Lemaitre B., Lohse A.W., Massinga-Loembe M., Matthey A., Mordmuller B., Nolting A., Ogwang C., Ramharter M., Schmidt-Chanasit J., Schmiedel S., Silvera P., Stahl F.R., Staines H.M., Strecker T., Stubbe H.C., Tsofa B., Zaki S., Fast P., Moorthy V., Kaiser L., Krishna S., Becker S., Kieny M.P., Bejon P., Kremsner P.G., Addo M.M., Siegrist C.A. Phase 1 trials of rVSV ebola vaccine in Africa and Europe — preliminary report. N. Engl. J. Med. 2015 (ePub ahead of print) - PMC - PubMed
    1. Alexopoulou L., Holt A.C., Medzhitov R., Flavell R.A. Recognition of double-stranded RNA and activation of NF-kappab by Toll-Like receptor 3. Nature. 2001;413:732–738. - PubMed
    1. Audet J., Wong G., Wang H., Lu G., Gao G.F., Kobinger G., Qiu X. Molecular characterization of the monoclonal antibodies composing zmab: a protective cocktail against Ebola virus. Sci. Rep. 2014;4:6881. - PMC - PubMed
    1. Bale S., Dias J.M., Fusco M.L., Hashiguchi T., Wong A.C., Liu T., Keuhne A.I., Li S., Woods V.L., Jr., Chandran K., Dye J.M., Saphire E.O. Structural basis for differential neutralization of ebolaviruses. Viruses. 2012;4:447–470. - PMC - PubMed
    1. Baumgartner C.K., Malherbe L.P. Regulation of CD4 T-cell receptor diversity by vaccine adjuvants. Immunology. 2010;130:16–22. - PMC - PubMed
Show all 87 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig. 1
Fig. 1
Adjuvants impact the durability of protection conferred by eVLP vaccination. (a) C57BL/6 mice were vaccinated IM two times with 10 μg of eVLP and challenged four (short-term) weeks or twenty-two (long-term) weeks after the vaccine boost. Data in A are pooled from 8 individual studies with 6–10 animals/group. Fisher's exact test: survival in the short-term group was significantly higher than in the long-term group (p 

Fig. 2

Adjuvants have variable impact on…

Fig. 2

Adjuvants have variable impact on IgG subclasses and antibody neutralization. (a) Serum was…

Fig. 2
Adjuvants have variable impact on IgG subclasses and antibody neutralization. (a) Serum was collected 14 days and 147 days after the vaccine boost (days 35 and 168, respectively) and evaluated for anti-GP IgG, IgG1, IgG2c, and IgG3 levels using an ELISA. Data shown are pooled from at least two separate experiments per group. (b) Pairwise comparison using DSCF multiple pairwise comparison was used and p values greater than 0.05 are shown as “ns”. (c) Summary of results shown in A, B and D. (d) Neutralizing antibody titers were evaluated using the PsVNA, with titers giving 80% neutralization shown; median and IQR shown. Samples within each group were selected randomly from three separate studies for evaluation in the assay. Pairwise comparison using post-hoc Tukey's studentized range test procedure was used to evaluate differences between groups at both days 35 and day 168, where “*” indicates 0.01 

Fig. 3

Effective adjuvants increase the frequency…

Fig. 3

Effective adjuvants increase the frequency of antigen-specific T cells. (a) IFNγ ELISPOT analysis…

Fig. 3
Effective adjuvants increase the frequency of antigen-specific T cells. (a) IFNγ ELISPOT analysis of splenocytes from C57BL/6 mice vaccinated two times with indicated vaccine and adjuvant combination. Splenocytes were collected on day 4 after the second vaccination. Data is pooled from four separate experiments each containing 2–3 mice per group; median with IQR shown. (B&C) Frequency of IFNγ +, IL2 +, or TNFα + cells after vaccination with VLP and polyICLC or VLP and CpG; median shown. Gating is on viable T cells and then CD4 + CD44high T cells (b) or CD8 + CD44high T cells (c). (d) IFNγ ELISPOT analysis of splenocytes from C57BL/6 mice vaccinated with indicated vaccine and adjuvant combination. Animals received the standard two vaccinations and then received a third vaccine boost 22 weeks after the second vaccination. Splenocytes were collected 4 days later. Data is pooled from two separate experiments each containing 3–4 mice per group; median with IQR shown. (e) Four weeks after the second vaccination, splenocytes were collected from animals and T cells were isolated using negative bead selection. T cells were then sorted to collect central memory, effector memory, and CD44int/low cell populations. Gating strategy is shown. (f) Sorted T cells were cultured with peptide-exposed, T cell-depleted, naïve splenocytes and the frequency of cytokine-positive CD4 and CD8 T cells in each sorted population was quantified after 5 days of culture. Gating is on CD4 or CD8 T cells and data shown are the frequency of cells expressing IFNγ, TNFα, or IL2; median is shown. “*” indicates 0.01 

Fig. 4

CD8 T cell deficiency does…

Fig. 4

CD8 T cell deficiency does not impact short term or long term survival…

Fig. 4
CD8 T cell deficiency does not impact short term or long term survival of vaccinated C57BL/6J mice. (a) Mice were treated IM twice with saline, VLP, VLP and polyICLC, or VLP and CpG. Four weeks after the second vaccination, mice were challenged. Closed symbols represent wild type C57BL/6J mice and open symbols indicate CD8-deficient mice. (b) Mice were vaccinated on the same schedule as in A, but challenge occurred 22 weeks after the second vaccination. (c) Two weeks after the second vaccination and 1 week prior to challenge, blood was collected from vaccinated animals and evaluated for anti-GP IgG antibody titers. (D) A subset of vaccinated animals was euthanized 4 days after the vaccine boost to evaluate CD4 + T cell responses. Median response of C57BL/6J mice and CD8-deficient mice is shown. N = 8–10 per treated group for A–C and D presents data pooled from two separate evaluations of 4–8 mice per group each. “*” indicates 0.005 

Fig. 5

Tfh frequencies are increased by…

Fig. 5

Tfh frequencies are increased by adjuvants associated with protection from challenge. C57BL/6 mice…

Fig. 5
Tfh frequencies are increased by adjuvants associated with protection from challenge. C57BL/6 mice were vaccinated a single time IM with VLP, with or without adjuvant. Seven days after vaccination, the draining popliteal LN was isolated. (a) Untouched T cells were isolated from the LN (n = 3 or 4/group). RNA was isolated from the purified T cells and pooled for each vaccination group. Samples were then evaluated using the SABiosciences T and B cell activation PCR array in triplicate. For the volcano plot, the fold difference is the average of triplicate, and the p value was calculated comparing animals vaccinated with VLP to those vaccinated with VLP and polyICLC. (b) Seven days after vaccination, cells from the draining LN were evaluated for Tfh populations. Cells were gated on viable lymphocytes after doublet exclusion, and then on CD4 + T cells expressing CXCR5 and PD1. Median and IQR shown. (c) CD3 + CD4 + CXCR5 + PD1 + cell population in mice vaccinated with VLP and polyICLC. ICOS and Bcl6 expression of this population is shown. Data shown are pooled from three separate vaccination experiments. Comparisons between animals receiving VLP with or with adjuvant are shown, where “*” indicates 0.01 
Similar articles
Cited by
References
    1. Agnandji S.T., Huttner A., Zinser M.E., Njuguna P., Dahlke C., Fernandes J.F., Yerly S., Dayer J.A., Kraehling V., Kasonta R., Adegnika A.A., Altfeld M., Auderset F., Bache E.B., Biedenkopf N., Borregaard S., Brosnahan J.S., Burrow R., Combescure C., Desmeules J., Eickmann M., Fehling S.K., Finckh A., Goncalves A.R., Grobusch M.P., Hooper J., Jambrecina A., Kabwende A.L., Kaya G., Kimani D., Lell B., Lemaitre B., Lohse A.W., Massinga-Loembe M., Matthey A., Mordmuller B., Nolting A., Ogwang C., Ramharter M., Schmidt-Chanasit J., Schmiedel S., Silvera P., Stahl F.R., Staines H.M., Strecker T., Stubbe H.C., Tsofa B., Zaki S., Fast P., Moorthy V., Kaiser L., Krishna S., Becker S., Kieny M.P., Bejon P., Kremsner P.G., Addo M.M., Siegrist C.A. Phase 1 trials of rVSV ebola vaccine in Africa and Europe — preliminary report. N. Engl. J. Med. 2015 (ePub ahead of print) - PMC - PubMed
    1. Alexopoulou L., Holt A.C., Medzhitov R., Flavell R.A. Recognition of double-stranded RNA and activation of NF-kappab by Toll-Like receptor 3. Nature. 2001;413:732–738. - PubMed
    1. Audet J., Wong G., Wang H., Lu G., Gao G.F., Kobinger G., Qiu X. Molecular characterization of the monoclonal antibodies composing zmab: a protective cocktail against Ebola virus. Sci. Rep. 2014;4:6881. - PMC - PubMed
    1. Bale S., Dias J.M., Fusco M.L., Hashiguchi T., Wong A.C., Liu T., Keuhne A.I., Li S., Woods V.L., Jr., Chandran K., Dye J.M., Saphire E.O. Structural basis for differential neutralization of ebolaviruses. Viruses. 2012;4:447–470. - PMC - PubMed
    1. Baumgartner C.K., Malherbe L.P. Regulation of CD4 T-cell receptor diversity by vaccine adjuvants. Immunology. 2010;130:16–22. - PMC - PubMed
Show all 87 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig. 2
Fig. 2
Adjuvants have variable impact on IgG subclasses and antibody neutralization. (a) Serum was collected 14 days and 147 days after the vaccine boost (days 35 and 168, respectively) and evaluated for anti-GP IgG, IgG1, IgG2c, and IgG3 levels using an ELISA. Data shown are pooled from at least two separate experiments per group. (b) Pairwise comparison using DSCF multiple pairwise comparison was used and p values greater than 0.05 are shown as “ns”. (c) Summary of results shown in A, B and D. (d) Neutralizing antibody titers were evaluated using the PsVNA, with titers giving 80% neutralization shown; median and IQR shown. Samples within each group were selected randomly from three separate studies for evaluation in the assay. Pairwise comparison using post-hoc Tukey's studentized range test procedure was used to evaluate differences between groups at both days 35 and day 168, where “*” indicates 0.01 

Fig. 3

Effective adjuvants increase the frequency…

Fig. 3

Effective adjuvants increase the frequency of antigen-specific T cells. (a) IFNγ ELISPOT analysis…

Fig. 3
Effective adjuvants increase the frequency of antigen-specific T cells. (a) IFNγ ELISPOT analysis of splenocytes from C57BL/6 mice vaccinated two times with indicated vaccine and adjuvant combination. Splenocytes were collected on day 4 after the second vaccination. Data is pooled from four separate experiments each containing 2–3 mice per group; median with IQR shown. (B&C) Frequency of IFNγ +, IL2 +, or TNFα + cells after vaccination with VLP and polyICLC or VLP and CpG; median shown. Gating is on viable T cells and then CD4 + CD44high T cells (b) or CD8 + CD44high T cells (c). (d) IFNγ ELISPOT analysis of splenocytes from C57BL/6 mice vaccinated with indicated vaccine and adjuvant combination. Animals received the standard two vaccinations and then received a third vaccine boost 22 weeks after the second vaccination. Splenocytes were collected 4 days later. Data is pooled from two separate experiments each containing 3–4 mice per group; median with IQR shown. (e) Four weeks after the second vaccination, splenocytes were collected from animals and T cells were isolated using negative bead selection. T cells were then sorted to collect central memory, effector memory, and CD44int/low cell populations. Gating strategy is shown. (f) Sorted T cells were cultured with peptide-exposed, T cell-depleted, naïve splenocytes and the frequency of cytokine-positive CD4 and CD8 T cells in each sorted population was quantified after 5 days of culture. Gating is on CD4 or CD8 T cells and data shown are the frequency of cells expressing IFNγ, TNFα, or IL2; median is shown. “*” indicates 0.01 

Fig. 4

CD8 T cell deficiency does…

Fig. 4

CD8 T cell deficiency does not impact short term or long term survival…

Fig. 4
CD8 T cell deficiency does not impact short term or long term survival of vaccinated C57BL/6J mice. (a) Mice were treated IM twice with saline, VLP, VLP and polyICLC, or VLP and CpG. Four weeks after the second vaccination, mice were challenged. Closed symbols represent wild type C57BL/6J mice and open symbols indicate CD8-deficient mice. (b) Mice were vaccinated on the same schedule as in A, but challenge occurred 22 weeks after the second vaccination. (c) Two weeks after the second vaccination and 1 week prior to challenge, blood was collected from vaccinated animals and evaluated for anti-GP IgG antibody titers. (D) A subset of vaccinated animals was euthanized 4 days after the vaccine boost to evaluate CD4 + T cell responses. Median response of C57BL/6J mice and CD8-deficient mice is shown. N = 8–10 per treated group for A–C and D presents data pooled from two separate evaluations of 4–8 mice per group each. “*” indicates 0.005 

Fig. 5

Tfh frequencies are increased by…

Fig. 5

Tfh frequencies are increased by adjuvants associated with protection from challenge. C57BL/6 mice…

Fig. 5
Tfh frequencies are increased by adjuvants associated with protection from challenge. C57BL/6 mice were vaccinated a single time IM with VLP, with or without adjuvant. Seven days after vaccination, the draining popliteal LN was isolated. (a) Untouched T cells were isolated from the LN (n = 3 or 4/group). RNA was isolated from the purified T cells and pooled for each vaccination group. Samples were then evaluated using the SABiosciences T and B cell activation PCR array in triplicate. For the volcano plot, the fold difference is the average of triplicate, and the p value was calculated comparing animals vaccinated with VLP to those vaccinated with VLP and polyICLC. (b) Seven days after vaccination, cells from the draining LN were evaluated for Tfh populations. Cells were gated on viable lymphocytes after doublet exclusion, and then on CD4 + T cells expressing CXCR5 and PD1. Median and IQR shown. (c) CD3 + CD4 + CXCR5 + PD1 + cell population in mice vaccinated with VLP and polyICLC. ICOS and Bcl6 expression of this population is shown. Data shown are pooled from three separate vaccination experiments. Comparisons between animals receiving VLP with or with adjuvant are shown, where “*” indicates 0.01 
Similar articles
Cited by
References
    1. Agnandji S.T., Huttner A., Zinser M.E., Njuguna P., Dahlke C., Fernandes J.F., Yerly S., Dayer J.A., Kraehling V., Kasonta R., Adegnika A.A., Altfeld M., Auderset F., Bache E.B., Biedenkopf N., Borregaard S., Brosnahan J.S., Burrow R., Combescure C., Desmeules J., Eickmann M., Fehling S.K., Finckh A., Goncalves A.R., Grobusch M.P., Hooper J., Jambrecina A., Kabwende A.L., Kaya G., Kimani D., Lell B., Lemaitre B., Lohse A.W., Massinga-Loembe M., Matthey A., Mordmuller B., Nolting A., Ogwang C., Ramharter M., Schmidt-Chanasit J., Schmiedel S., Silvera P., Stahl F.R., Staines H.M., Strecker T., Stubbe H.C., Tsofa B., Zaki S., Fast P., Moorthy V., Kaiser L., Krishna S., Becker S., Kieny M.P., Bejon P., Kremsner P.G., Addo M.M., Siegrist C.A. Phase 1 trials of rVSV ebola vaccine in Africa and Europe — preliminary report. N. Engl. J. Med. 2015 (ePub ahead of print) - PMC - PubMed
    1. Alexopoulou L., Holt A.C., Medzhitov R., Flavell R.A. Recognition of double-stranded RNA and activation of NF-kappab by Toll-Like receptor 3. Nature. 2001;413:732–738. - PubMed
    1. Audet J., Wong G., Wang H., Lu G., Gao G.F., Kobinger G., Qiu X. Molecular characterization of the monoclonal antibodies composing zmab: a protective cocktail against Ebola virus. Sci. Rep. 2014;4:6881. - PMC - PubMed
    1. Bale S., Dias J.M., Fusco M.L., Hashiguchi T., Wong A.C., Liu T., Keuhne A.I., Li S., Woods V.L., Jr., Chandran K., Dye J.M., Saphire E.O. Structural basis for differential neutralization of ebolaviruses. Viruses. 2012;4:447–470. - PMC - PubMed
    1. Baumgartner C.K., Malherbe L.P. Regulation of CD4 T-cell receptor diversity by vaccine adjuvants. Immunology. 2010;130:16–22. - PMC - PubMed
Show all 87 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig. 3
Fig. 3
Effective adjuvants increase the frequency of antigen-specific T cells. (a) IFNγ ELISPOT analysis of splenocytes from C57BL/6 mice vaccinated two times with indicated vaccine and adjuvant combination. Splenocytes were collected on day 4 after the second vaccination. Data is pooled from four separate experiments each containing 2–3 mice per group; median with IQR shown. (B&C) Frequency of IFNγ +, IL2 +, or TNFα + cells after vaccination with VLP and polyICLC or VLP and CpG; median shown. Gating is on viable T cells and then CD4 + CD44high T cells (b) or CD8 + CD44high T cells (c). (d) IFNγ ELISPOT analysis of splenocytes from C57BL/6 mice vaccinated with indicated vaccine and adjuvant combination. Animals received the standard two vaccinations and then received a third vaccine boost 22 weeks after the second vaccination. Splenocytes were collected 4 days later. Data is pooled from two separate experiments each containing 3–4 mice per group; median with IQR shown. (e) Four weeks after the second vaccination, splenocytes were collected from animals and T cells were isolated using negative bead selection. T cells were then sorted to collect central memory, effector memory, and CD44int/low cell populations. Gating strategy is shown. (f) Sorted T cells were cultured with peptide-exposed, T cell-depleted, naïve splenocytes and the frequency of cytokine-positive CD4 and CD8 T cells in each sorted population was quantified after 5 days of culture. Gating is on CD4 or CD8 T cells and data shown are the frequency of cells expressing IFNγ, TNFα, or IL2; median is shown. “*” indicates 0.01 

Fig. 4

CD8 T cell deficiency does…

Fig. 4

CD8 T cell deficiency does not impact short term or long term survival…

Fig. 4
CD8 T cell deficiency does not impact short term or long term survival of vaccinated C57BL/6J mice. (a) Mice were treated IM twice with saline, VLP, VLP and polyICLC, or VLP and CpG. Four weeks after the second vaccination, mice were challenged. Closed symbols represent wild type C57BL/6J mice and open symbols indicate CD8-deficient mice. (b) Mice were vaccinated on the same schedule as in A, but challenge occurred 22 weeks after the second vaccination. (c) Two weeks after the second vaccination and 1 week prior to challenge, blood was collected from vaccinated animals and evaluated for anti-GP IgG antibody titers. (D) A subset of vaccinated animals was euthanized 4 days after the vaccine boost to evaluate CD4 + T cell responses. Median response of C57BL/6J mice and CD8-deficient mice is shown. N = 8–10 per treated group for A–C and D presents data pooled from two separate evaluations of 4–8 mice per group each. “*” indicates 0.005 

Fig. 5

Tfh frequencies are increased by…

Fig. 5

Tfh frequencies are increased by adjuvants associated with protection from challenge. C57BL/6 mice…

Fig. 5
Tfh frequencies are increased by adjuvants associated with protection from challenge. C57BL/6 mice were vaccinated a single time IM with VLP, with or without adjuvant. Seven days after vaccination, the draining popliteal LN was isolated. (a) Untouched T cells were isolated from the LN (n = 3 or 4/group). RNA was isolated from the purified T cells and pooled for each vaccination group. Samples were then evaluated using the SABiosciences T and B cell activation PCR array in triplicate. For the volcano plot, the fold difference is the average of triplicate, and the p value was calculated comparing animals vaccinated with VLP to those vaccinated with VLP and polyICLC. (b) Seven days after vaccination, cells from the draining LN were evaluated for Tfh populations. Cells were gated on viable lymphocytes after doublet exclusion, and then on CD4 + T cells expressing CXCR5 and PD1. Median and IQR shown. (c) CD3 + CD4 + CXCR5 + PD1 + cell population in mice vaccinated with VLP and polyICLC. ICOS and Bcl6 expression of this population is shown. Data shown are pooled from three separate vaccination experiments. Comparisons between animals receiving VLP with or with adjuvant are shown, where “*” indicates 0.01 
Similar articles
Cited by
References
    1. Agnandji S.T., Huttner A., Zinser M.E., Njuguna P., Dahlke C., Fernandes J.F., Yerly S., Dayer J.A., Kraehling V., Kasonta R., Adegnika A.A., Altfeld M., Auderset F., Bache E.B., Biedenkopf N., Borregaard S., Brosnahan J.S., Burrow R., Combescure C., Desmeules J., Eickmann M., Fehling S.K., Finckh A., Goncalves A.R., Grobusch M.P., Hooper J., Jambrecina A., Kabwende A.L., Kaya G., Kimani D., Lell B., Lemaitre B., Lohse A.W., Massinga-Loembe M., Matthey A., Mordmuller B., Nolting A., Ogwang C., Ramharter M., Schmidt-Chanasit J., Schmiedel S., Silvera P., Stahl F.R., Staines H.M., Strecker T., Stubbe H.C., Tsofa B., Zaki S., Fast P., Moorthy V., Kaiser L., Krishna S., Becker S., Kieny M.P., Bejon P., Kremsner P.G., Addo M.M., Siegrist C.A. Phase 1 trials of rVSV ebola vaccine in Africa and Europe — preliminary report. N. Engl. J. Med. 2015 (ePub ahead of print) - PMC - PubMed
    1. Alexopoulou L., Holt A.C., Medzhitov R., Flavell R.A. Recognition of double-stranded RNA and activation of NF-kappab by Toll-Like receptor 3. Nature. 2001;413:732–738. - PubMed
    1. Audet J., Wong G., Wang H., Lu G., Gao G.F., Kobinger G., Qiu X. Molecular characterization of the monoclonal antibodies composing zmab: a protective cocktail against Ebola virus. Sci. Rep. 2014;4:6881. - PMC - PubMed
    1. Bale S., Dias J.M., Fusco M.L., Hashiguchi T., Wong A.C., Liu T., Keuhne A.I., Li S., Woods V.L., Jr., Chandran K., Dye J.M., Saphire E.O. Structural basis for differential neutralization of ebolaviruses. Viruses. 2012;4:447–470. - PMC - PubMed
    1. Baumgartner C.K., Malherbe L.P. Regulation of CD4 T-cell receptor diversity by vaccine adjuvants. Immunology. 2010;130:16–22. - PMC - PubMed
Show all 87 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Fig. 4
Fig. 4
CD8 T cell deficiency does not impact short term or long term survival of vaccinated C57BL/6J mice. (a) Mice were treated IM twice with saline, VLP, VLP and polyICLC, or VLP and CpG. Four weeks after the second vaccination, mice were challenged. Closed symbols represent wild type C57BL/6J mice and open symbols indicate CD8-deficient mice. (b) Mice were vaccinated on the same schedule as in A, but challenge occurred 22 weeks after the second vaccination. (c) Two weeks after the second vaccination and 1 week prior to challenge, blood was collected from vaccinated animals and evaluated for anti-GP IgG antibody titers. (D) A subset of vaccinated animals was euthanized 4 days after the vaccine boost to evaluate CD4 + T cell responses. Median response of C57BL/6J mice and CD8-deficient mice is shown. N = 8–10 per treated group for A–C and D presents data pooled from two separate evaluations of 4–8 mice per group each. “*” indicates 0.005 

Fig. 5

Tfh frequencies are increased by…

Fig. 5

Tfh frequencies are increased by adjuvants associated with protection from challenge. C57BL/6 mice…

Fig. 5
Tfh frequencies are increased by adjuvants associated with protection from challenge. C57BL/6 mice were vaccinated a single time IM with VLP, with or without adjuvant. Seven days after vaccination, the draining popliteal LN was isolated. (a) Untouched T cells were isolated from the LN (n = 3 or 4/group). RNA was isolated from the purified T cells and pooled for each vaccination group. Samples were then evaluated using the SABiosciences T and B cell activation PCR array in triplicate. For the volcano plot, the fold difference is the average of triplicate, and the p value was calculated comparing animals vaccinated with VLP to those vaccinated with VLP and polyICLC. (b) Seven days after vaccination, cells from the draining LN were evaluated for Tfh populations. Cells were gated on viable lymphocytes after doublet exclusion, and then on CD4 + T cells expressing CXCR5 and PD1. Median and IQR shown. (c) CD3 + CD4 + CXCR5 + PD1 + cell population in mice vaccinated with VLP and polyICLC. ICOS and Bcl6 expression of this population is shown. Data shown are pooled from three separate vaccination experiments. Comparisons between animals receiving VLP with or with adjuvant are shown, where “*” indicates 0.01 
Similar articles
Cited by
References
    1. Agnandji S.T., Huttner A., Zinser M.E., Njuguna P., Dahlke C., Fernandes J.F., Yerly S., Dayer J.A., Kraehling V., Kasonta R., Adegnika A.A., Altfeld M., Auderset F., Bache E.B., Biedenkopf N., Borregaard S., Brosnahan J.S., Burrow R., Combescure C., Desmeules J., Eickmann M., Fehling S.K., Finckh A., Goncalves A.R., Grobusch M.P., Hooper J., Jambrecina A., Kabwende A.L., Kaya G., Kimani D., Lell B., Lemaitre B., Lohse A.W., Massinga-Loembe M., Matthey A., Mordmuller B., Nolting A., Ogwang C., Ramharter M., Schmidt-Chanasit J., Schmiedel S., Silvera P., Stahl F.R., Staines H.M., Strecker T., Stubbe H.C., Tsofa B., Zaki S., Fast P., Moorthy V., Kaiser L., Krishna S., Becker S., Kieny M.P., Bejon P., Kremsner P.G., Addo M.M., Siegrist C.A. Phase 1 trials of rVSV ebola vaccine in Africa and Europe — preliminary report. N. Engl. J. Med. 2015 (ePub ahead of print) - PMC - PubMed
    1. Alexopoulou L., Holt A.C., Medzhitov R., Flavell R.A. Recognition of double-stranded RNA and activation of NF-kappab by Toll-Like receptor 3. Nature. 2001;413:732–738. - PubMed
    1. Audet J., Wong G., Wang H., Lu G., Gao G.F., Kobinger G., Qiu X. Molecular characterization of the monoclonal antibodies composing zmab: a protective cocktail against Ebola virus. Sci. Rep. 2014;4:6881. - PMC - PubMed
    1. Bale S., Dias J.M., Fusco M.L., Hashiguchi T., Wong A.C., Liu T., Keuhne A.I., Li S., Woods V.L., Jr., Chandran K., Dye J.M., Saphire E.O. Structural basis for differential neutralization of ebolaviruses. Viruses. 2012;4:447–470. - PMC - PubMed
    1. Baumgartner C.K., Malherbe L.P. Regulation of CD4 T-cell receptor diversity by vaccine adjuvants. Immunology. 2010;130:16–22. - PMC - PubMed
Show all 87 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Fig. 5
Fig. 5
Tfh frequencies are increased by adjuvants associated with protection from challenge. C57BL/6 mice were vaccinated a single time IM with VLP, with or without adjuvant. Seven days after vaccination, the draining popliteal LN was isolated. (a) Untouched T cells were isolated from the LN (n = 3 or 4/group). RNA was isolated from the purified T cells and pooled for each vaccination group. Samples were then evaluated using the SABiosciences T and B cell activation PCR array in triplicate. For the volcano plot, the fold difference is the average of triplicate, and the p value was calculated comparing animals vaccinated with VLP to those vaccinated with VLP and polyICLC. (b) Seven days after vaccination, cells from the draining LN were evaluated for Tfh populations. Cells were gated on viable lymphocytes after doublet exclusion, and then on CD4 + T cells expressing CXCR5 and PD1. Median and IQR shown. (c) CD3 + CD4 + CXCR5 + PD1 + cell population in mice vaccinated with VLP and polyICLC. ICOS and Bcl6 expression of this population is shown. Data shown are pooled from three separate vaccination experiments. Comparisons between animals receiving VLP with or with adjuvant are shown, where “*” indicates 0.01 

References

    1. Agnandji S.T., Huttner A., Zinser M.E., Njuguna P., Dahlke C., Fernandes J.F., Yerly S., Dayer J.A., Kraehling V., Kasonta R., Adegnika A.A., Altfeld M., Auderset F., Bache E.B., Biedenkopf N., Borregaard S., Brosnahan J.S., Burrow R., Combescure C., Desmeules J., Eickmann M., Fehling S.K., Finckh A., Goncalves A.R., Grobusch M.P., Hooper J., Jambrecina A., Kabwende A.L., Kaya G., Kimani D., Lell B., Lemaitre B., Lohse A.W., Massinga-Loembe M., Matthey A., Mordmuller B., Nolting A., Ogwang C., Ramharter M., Schmidt-Chanasit J., Schmiedel S., Silvera P., Stahl F.R., Staines H.M., Strecker T., Stubbe H.C., Tsofa B., Zaki S., Fast P., Moorthy V., Kaiser L., Krishna S., Becker S., Kieny M.P., Bejon P., Kremsner P.G., Addo M.M., Siegrist C.A. Phase 1 trials of rVSV ebola vaccine in Africa and Europe — preliminary report. N. Engl. J. Med. 2015 (ePub ahead of print)
    1. Alexopoulou L., Holt A.C., Medzhitov R., Flavell R.A. Recognition of double-stranded RNA and activation of NF-kappab by Toll-Like receptor 3. Nature. 2001;413:732–738.
    1. Audet J., Wong G., Wang H., Lu G., Gao G.F., Kobinger G., Qiu X. Molecular characterization of the monoclonal antibodies composing zmab: a protective cocktail against Ebola virus. Sci. Rep. 2014;4:6881.
    1. Bale S., Dias J.M., Fusco M.L., Hashiguchi T., Wong A.C., Liu T., Keuhne A.I., Li S., Woods V.L., Jr., Chandran K., Dye J.M., Saphire E.O. Structural basis for differential neutralization of ebolaviruses. Viruses. 2012;4:447–470.
    1. Baumgartner C.K., Malherbe L.P. Regulation of CD4 T-cell receptor diversity by vaccine adjuvants. Immunology. 2010;130:16–22.
    1. Beran J. Safety and immunogenicity of a new Hepatitis B vaccine for the protection of patients with renal insufficiency including pre-haemodialysis and haemodialysis patients. Expert. Opin. Biol. Ther. 2008;8:235–247.
    1. Bohannon J.K., Hernandez A., Enkhbaatar P., Adams W.L., Sherwood E.R. The immunobiology of Toll-Like receptor 4 agonists: from endotoxin tolerance to immunoadjuvants. Shock. 2013;40:451–462.
    1. Bray M., Davis K., Geisbert T., Schmaljohn C., Huggins J. A mouse model for evaluation of prophylaxis and therapy of Ebola hemorrhagic fever. J. Infect. Dis. 1998;178:651–661.
    1. Brown D.M., Lee S., Garcia-Hernandez Mde L., Swain S.L. Multifunctional CD4 cells expressing gamma interferon and perforin mediate protection against lethal influenza virus infection. J. Virol. 2012;86:6792–6803.
    1. Brunner R., Jensen-Jarolim E., Pali-Scholl I. The ABC of clinical and experimental adjuvants—a brief overview. Immunol. Lett. 2010;128:29–35.
    1. Caproni E., Tritto E., Cortese M., Muzzi A., Mosca F., Monaci E., Baudner B., Seubert A., De Gregorio E. MF59 and Pam3CSK4 boost adaptive responses to influenza subunit vaccine through an IFN type I-independent mechanism of Action. J. Immunol. 2012;188:3088–3098.
    1. Coffman R.L., Sher A., Seder R.A. Vaccine adjuvants: putting innate immunity to work. Immunity. 2010;33:492–503.
    1. Doll K.L., Harty J.T. Correlates of protective immunity following whole sporozoite vaccination against malaria. Immunol. Res. 2014;59:166–176.
    1. Dye J.M., Herbert A.S., Kuehne A.I., Barth J.F., Muhammad M.A., Zak S.E., Ortiz R.A., Prugar L.I., Pratt W.D. Postexposure antibody prophylaxis protects nonhuman primates from filovirus disease. Proc. Natl. Acad. Sci. U. S. A. 2012;109:5034–5039.
    1. Einstein M.H., Levin M.J., Chatterjee A., Chakhtoura N., Takacs P., Catteau G., Dessy F.J., Moris P., Lin L., Struyf F., Dubin G., Group, H. P. V. S. Comparative humoral and cellular immunogenicity and safety of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine and HPV-6/11/16/18 vaccine in healthy women aged 18–45 years: follow-up through month 48 in a Phase III randomized study. Hum. Vaccines Immunother. 2014;10:3455–3465.
    1. Einstein M.H., Takacs P., Chatterjee A., Sperling R.S., Chakhtoura N., Blatter M.M., Lalezari J., David M.P., Lin L., Struyf F., Dubin G., Group, H. P. V. S. Comparison of long-term immunogenicity and safety of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine and HPV-6/11/16/18 vaccine in healthy women aged 18–45 years: end-of-study analysis of a Phase III randomized trial. Hum. Vaccines Immunother. 2014;10:3435–3445.
    1. Finkelman F.D., Katona I.M., Mosmann T.R., Coffman R.L. IFN-Gamma Regulates the isotypes of Ig secreted during in vivo humoral immune responses. J. Immunol. 1988;140:1022–1027.
    1. Grant-Klein R.J., Altamura L.A., Badger C.V., Bounds C.E., Van Deusen N.M., Kwilas S.A., Vu H.A., Warfield K.L., Hooper J.W., Hannaman D., Dupuy L.C., Schmaljohn C.S. Codon-optimized Filovirus DNA Vaccines Delivered by Intramuscular Electroporation Protect Cynomolgus Macaques from Lethal Ebola and Marburg Virus Challenges. Hum. Vaccines Immunother. 2015;11(8):1991–2004.
    1. Gupta R.K. Aluminum compounds as vaccine adjuvants. Adv. Drug Deliv. Rev. 1998;32:155–172.
    1. Guven E., Duus K., Laursen I., Hojrup P., Houen G. Aluminum hydroxide adjuvant differentially activates the three complement pathways with major involvement of the alternative pathway. PLoS One. 2013;8:e74445.
    1. Hanson M.C., Crespo M.P., Abraham W., Moynihan K.D., Szeto G.L., Chen S.H., Melo M.B., Mueller S., Irvine D.J. Nanoparticulate STING agonists are potent lymph node-targeted vaccine adjuvants. J. Clin. Invest. 2015;125:2532–2546.
    1. Hartmann G., Battiany J., Poeck H., Wagner M., Kerkmann M., Lubenow N., Rothenfusser S., Endres S. Rational design of new CpG oligonucleotides that combine B cell activation with high IFN-alpha induction in plasmacytoid dendritic cells. Eur. J. Immunol. 2003;33:1633–1641.
    1. Henao-Tamayo M.I., Ordway D.J., Irwin S.M., Shang S., Shanley C., Orme I.M. Phenotypic definition of effector and memory T-lymphocyte subsets in mice chronically infected with Mycobacterium tuberculosis. Clin. Vaccine Immunol. 2010;17:618–625.
    1. Hikono H., Kohlmeier J.E., Takamura S., Wittmer S.T., Roberts A.D., Woodland D.L. Activation phenotype, rather than central- or effector-memory phenotype, predicts the recall efficacy of memory CD8 + T cells. J. Exp. Med. 2007;204:1625–1636.
    1. Hogenesch H. Mechanisms of stimulation of the immune response by aluminum adjuvants. Vaccine. 2002;20(Suppl. 3):S34–S39.
    1. Hutchison S., Benson R.A., Gibson V.B., Pollock A.H., Garside P., Brewer J.M. Antigen depot is not required for alum adjuvanticity. FASEB J. 2012;26:1272–1279.
    1. Jain S., O'Hagan D.T., Singh M. The long-term potential of biodegradable poly(lactide-co-glycolide) microparticles as the next-generation vaccine adjuvant. Expert Rev. Vaccines. 2011;10:1731–1742.
    1. Johnson S., Eller M., Teigler J.E., Maloveste S.M., Schultz B.T., Soghoian D.Z., Lu R., Oster A.F., Chenine A.L., Alter G., Dittmer U., Marovich M., Robb M.L., Michael N.L., Bolton D., Streeck H. Cooperativity of HIV-specific cytolytic CD4 + T cells and CD8 + T cells in control of HIV viremia. J. Virol. 2015;89(15):7494–7505.
    1. Kastenmuller K., Espinosa D.A., Trager L., Stoyanov C., Salazar A.M., Pokalwar S., Singh S., Dutta S., Ockenhouse C.F., Zavala F., Seder R.A. Full-length P. falciparum circumsporozoite protein administered with poly-ICLC or GLA/SE elicits potent antibody and CD4 + T cell immunity and protection in Mice. Infect. Immun. 2012;81(3):789–800.
    1. Kok T., Gaeguta A., Finnie J., Gorry P.R., Churchill M., Li P. Designer antigens for elicitation of broadly neutralizing antibodies against HIV. Clin. Transl. Immunol. 2014;3:e24.
    1. Krishnan L., Gurnani K., Dicaire C.J., van Faassen H., Zafer A., Kirschning C.J., Sad S., Sprott G.D. Rapid clonal expansion and prolonged maintenance of memory CD8 + T cells of the effector (CD44highCD62Llow) and central (CD44highCD62Lhigh) phenotype by an archaeosome adjuvant independent of TLR2. J. Immunol. 2007;178:2396–2406.
    1. Lee J.E., Fusco M.L., Hessell A.J., Oswald W.B., Burton D.R., Saphire E.O. Structure of the Ebola virus glycoprotein bound to an antibody from a human survivor. Nature. 2008;454:177–182.
    1. Lefrancois L., Masopust D. T cell immunity in lymphoid and non-lymphoid tissues. Curr. Opin. Immunol. 2002;14:503–508.
    1. Levy H.B., Baer G., Baron S., Buckler C.E., Gibbs C.J., Iadarola M.J., London W.T., Rice J. A modified polyriboinosinic-polyribocytidylic acid complex that induces interferon in primates. J. Infect. Dis. 1975;132:434–439.
    1. Longhi M.P., Trumpfheller C., Idoyaga J., Caskey M., Matos I., Kluger C., Salazar A.M., Colonna M., Steinman R.M. Dendritic cells require a systemic type I interferon response to mature and induce CD4 + Th1 immunity with poly IC as adjuvant. J. Exp. Med. 2009;206:1589–1602.
    1. Marrack P., Mckee A.S., Munks M.W. Towards an understanding of the adjuvant action of aluminium. Nat. Rev. Immunol. 2009;9:287–293.
    1. Marshall J.D., Fearon K., Abbate C., Subramanian S., Yee P., Gregorio J., Coffman R.L., Van Nest G. Identification of a novel cpg DNA class and motif that optimally stimulate B cell and plasmacytoid dendritic cell functions. J. Leukoc. Biol. 2003;73:781–792.
    1. Martins K.A., Steffens J.T., van Tongeren S.A., Wells J.B., Bergeron A.A., Dickson S.P., Dye J.M., Salazar A.M., Bavari S. Toll-like receptor agonist augments virus-like particle-mediated protection from Ebola virus with transient immune activation. PLoS One. 2014;9:e89735.
    1. Martins K., Carra J.H., Cooper C.L., Kwilas S.A., Robinson C.G., Shurtleff A.C., Schokman R.D., Kuehl K.A., Wells J.B., Steffens J.T., van Tongeren S.A., Hooper J.W., Bavari S. Cross-protection conferred by filovirus virus-like particles containing trimeric hybrid glycoprotein. Viral Immunol. 2015;28:62–70.
    1. Martins K.A., Bavari S., Salazar A.M. Vaccine adjuvant uses of poly-ic and derivatives. Expert Rev. Vaccines. 2015;14:447–459.
    1. Marzi A., Engelmann F., Feldmann F., Haberthur K., Shupert W.L., Brining D., Scott D.P., Geisbert T.W., Kawaoka Y., Katze M.G., Feldmann H., Messaoudi I. Antibodies are necessary for rvsv/ZEBOV-GP-mediated protection against lethal Ebola virus challenge in nonhuman primates. Proc. Natl. Acad. Sci. U. S. A. 2013;110:1893–1898.
    1. McAleer J.P., Vella A.T. Educating CD4 T cells with vaccine adjuvants: lessons from lipopolysaccharide. Trends Immunol. 2010;31:429–435.
    1. McKinstry K.K., Strutt T.M., Kuang Y., Brown D.M., Sell S., Dutton R.W., Swain S.L. Memory CD4 + T cells protect against influenza through multiple synergizing mechanisms. J. Clin. Invest. 2012;122:2847–2856.
    1. Mendoza D., Migueles S.A., Rood J.E., Peterson B., Johnson S., Doria-Rose N., Schneider D., Rakasz E., Trivett M.T., Trubey C.M., Coalter V., Hallahan C.W., Watkins D., Franchini G., Lifson J.D., Connors M. Cytotoxic capacity of SIV-specific CD8(+) T cells against primary autologous targets correlates with immune control in SIV-infected rhesus macaques. PLoS Pathog. 2013;9:e1003195.
    1. Monaci E., Mancini F., Lofano G., Bacconi M., Tavarini S., Sammicheli C., Arcidiacono L., Giraldi M., Galletti B., Rossi Paccani S., Torre A., Fontana M.R., Grandi G., De Gregorio E., Bensi G., Chiarot E., Nuti S., Bagnoli F., Soldaini E., Bertholet S. MF59- and Al(OH)3-Adjuvanted Staphylococcus aureus (4C-Staph) Vaccines Induce Sustained Protective Humoral and Cellular Immune Responses, with a Critical Role for Effector CD4 T Cells at Low Antibody Titers. Front. Immunol. 2015;6:439.
    1. Moon J.J., Suh H., Li A.V., Ockenhouse C.F., Yadava A., Irvine D.J. Enhancing humoral responses to a malaria antigen with nanoparticle vaccines that expand Tfh cells and promote germinal center induction. Proc. Natl. Acad. Sci. U. S. A. 2012;109:1080–1085.
    1. Nemes M.M., Tytell A.A., Lampson G.P., Field A.K., Hilleman M.R. Inducers of interferon and host resistance. VI. Antiviral efficacy of poly I:C in animal models. Proc. Soc. Exp. Biol. Med. 1969;132:776–783.
    1. O'Hagan D.T., Fox C.B. New generation adjuvants — From empiricism to rational design. Vaccine. 2015;33(Suppl. 2):B14–B20.
    1. Olinger G.G., Bailey M.A., Dye J.M., Bakken R., Kuehne A., Kondig J., Wilson J., Hogan R.J., Hart M.K. Protective cytotoxic T-cell responses induced by Venezuelan equine encephalitis virus replicons expressing Ebola virus proteins. J. Virol. 2005;79:14189–14196.
    1. Oswald W.B., Geisbert T.W., Davis K.J., Geisbert J.B., Sullivan N.J., Jahrling P.B., Parren P.W., Burton D.R. Neutralizing antibody fails to impact the course of Ebola virus infection in monkeys. PLoS Pathog. 2007;3:e9.
    1. Perret R., Sierro S.R., Botelho N.K., Corgnac S., Donda A., Romero P. Adjuvants that improve the ratio of antigen-specific effector to regulatory T cells enhance tumor immunity. Cancer Res. 2013;73:6597–6608.
    1. Pitoiset F., Vazquez T., Bellier B. Enveloped virus-like particle platforms: Vaccines of the future? Expert Rev. Vaccines. 2015:1–3.
    1. Plotkin S.A. Correlates of protection induced by vaccination. Clin. Vaccine Immunol. 2010;17:1055–1065.
    1. Qiu X., Audet J., Wong G., Pillet S., Bello A., Cabral T., Strong J.E., Plummer F., Corbett C.R., Alimonti J.B., Kobinger G.P. Successful treatment of ebola virus-infected cynomolgus macaques with monoclonal antibodies. Sci. Transl. Med. 2012;4:138ra81.
    1. Quinn K.M., Da Costa A., Yamamoto A., Berry D., Lindsay R.W., Darrah P.A., Wang L., Cheng C., Kong W.P., Gall J.G., Nicosia A., Folgori A., Colloca S., Cortese R., Gostick E., Price D.A., Gomez C.E., Esteban M., Wyatt L.S., Moss B., Morgan C., Roederer M., Bailer R.T., Nabel G.J., Koup R.A., Seder R.A. Comparative analysis of the magnitude, quality, phenotype, and protective capacity of simian immunodeficiency virus gag-specific CD8 + T cells following human-, simian-, and chimpanzee-derived recombinant adenoviral vector immunization. J. Immunol. 2013;190:2720–2735.
    1. Quinn K.M., Yamamoto A., Costa A., Darrah P.A., Lindsay R.W., Hegde S.T., Johnson T.R., Flynn B.J., Lore K., Seder R.A. Coadministration of polyinosinic:polycytidylic acid and immunostimulatory complexes modifies antigen processing in dendritic cell subsets and enhances HIV gag-specific T cell immunity. J. Immunol. 2013;191:5085–5096.
    1. Rao M., Bray M., Alving C.R., Jahrling P., Matyas G.R. Induction of immune responses in mice and monkeys to Ebola virus after immunization with liposome-encapsulated irradiated Ebola virus: protection in mice requires CD4(+) T cells. J. Virol. 2002;76:9176–9185.
    1. Roy C.J., Brey R.N., Mantis N.J., Mapes K., Pop I.V., Pop L.M., Ruback S., Killeen S.Z., Doyle-Meyers L., Vinet-Oliphant H.S., Didier P.J., Vitetta E.S. Thermostable ricin vaccine protects rhesus macaques against aerosolized ricin: Epitope-specific neutralizing antibodies correlate with protection. Proc. Natl. Acad. Sci. U. S. A. 2015;112:3782–3787.
    1. Sant A.J., McMichael A. Revealing the role of CD4(+) T cells in viral immunity. J. Exp. Med. 2012;209:1391–1395.
    1. Seder R., Reed S.G., O'Hagan D., Malyala P., D'Oro U., Laera D., Abrignani S., Cerundolo V., Steinman L., Bertholet S. Gaps in knowledge and prospects for research of adjuvanted vaccines. Vaccine. 2015;33(Suppl. 2):B40–B43.
    1. Shedlock D.J., Aviles J., Talbott K.T., Wong G., Wu S.J., Villarreal D.O., Myles D.J., Croyle M.A., Yan J., Kobinger G.P., Weiner D.B. Induction of broad cytotoxic T cells by protective DNA vaccination against Marburg and Ebola. Mol. Ther. 2013;21:1432–1444.
    1. Snapper C.M., Paul W.E. Interferon-gamma and B cell stimulatory factor-1 reciprocally regulate Ig isotype production. Science. 1987;236:944–947.
    1. Soghoian D.Z., Streeck H. Cytolytic CD4(+) T cells in viral immunity. Expert Rev. Vaccines. 2010;9:1453–1463.
    1. Sokolovska A., Hem S.L., Hogenesch H. Activation of dendritic cells and induction of CD4(+) T cell differentiation by aluminum-containing adjuvants. Vaccine. 2007;25:4575–4585.
    1. Sridhar S., Begom S., Bermingham A., Hoschler K., Adamson W., Carman W., Bean T., Barclay W., Deeks J.J., Lalvani A. Cellular immune correlates of protection against symptomatic pandemic influenza. Nat. Med. 2013;19:1305–1312.
    1. Stahl-Hennig C., Eisenblatter M., Jasny E., Rzehak T., Tenner-Racz K., Trumpfheller C., Salazar A.M., Uberla K., Nieto K., Kleinschmidt J., Schulte R., Gissmann L., Muller M., Sacher A., Racz P., Steinman R.M., Uguccioni M., Ignatius R. Synthetic double-stranded rnas are adjuvants for the induction of T helper 1 and humoral immune responses to human papillomavirus in rhesus macaques. PLoS Pathog. 2009;5:e1000373.
    1. Steinhagen F., Kinjo T., Bode C., Klinman D.M. TLR-based immune adjuvants. Vaccine. 2011;29:3341–3355.
    1. Sullivan N.J., Martin J.E., Graham B.S., Nabel G.J. Correlates of protective immunity for Ebola vaccines: implications for regulatory approval by the animal rule. Nat. Rev. Microbiol. 2009;7:393–400.
    1. Sullivan N.J., Hensley L., Asiedu C., Geisbert T.W., Stanley D., Johnson J., Honko A., Olinger G., Bailey M., Geisbert J.B., Reimann K.A., Bao S., Rao S., Roederer M., Jahrling P.B., Koup R.A., Nabel G.J. CD8 + cellular immunity mediates rAd5 vaccine protection against Ebola virus infection of nonhuman primates. Nat. Med. 2011;17:1128–1131.
    1. Surquin M., Tielemans C., Nortier J., Jadoul M., Peeters P., Ryba M., Roznovsky L., Doman J., Barthelemy X., Crasta P.D., Messier M., Houard S. Anti-HBs antibody persistence following primary vaccination with an investigational AS02(v)-adjuvanted hepatitis B vaccine in patients with renal insufficiency. Hum. Vaccines. 2011;7:913–918.
    1. Swenson D.L., Warfield K.L., Kuehl K., Larsen T., Hevey M.C., Schmaljohn A., Bavari S., Aman M.J. Generation of Marburg virus-like particles by co-expression of glycoprotein and matrix protein. FEMS Immunol. Med. Microbiol. 2004;40:27–31.
    1. Swenson D.L., Warfield K.L., Negley D.L., Schmaljohn A., Aman M.J., Bavari S. Virus-like particles exhibit potential as a pan-filovirus vaccine for both Ebola and Marburg viral infections. Vaccine. 2005;23:3033–3042.
    1. Swenson D.L., Wang D., Luo M., Warfield K.L., Woraratanadharm J., Holman D.H., Dong J.Y., Pratt W.D. Vaccine to confer to nonhuman primates complete protection against multistrain Ebola and Marburg virus infections. Clin. Vaccine Immunol. 2008;15:460–467.
    1. Swenson D.L., Warfield K.L., Larsen T., Alves D.A., Coberley S.S., Bavari S. Monovalent virus-like particle vaccine protects guinea pigs and nonhuman primates against infection with multiple Marburg viruses. Expert Rev. Vaccines. 2008;7:417–429.
    1. Trumpfheller C., Caskey M., Nchinda G., Longhi M.P., Mizenina O., Huang Y., Schlesinger S.J., Colonna M., Steinman R.M. The microbial mimic poly IC induces durable and protective CD4 + T cell immunity together with a dendritic cell targeted vaccine. Proc. Natl. Acad. Sci. U. S. A. 2008;105:2574–2579.
    1. van Gils M.J., Sanders R.W. In vivo protection by broadly neutralizing HIV antibodies. Trends Microbiol. 2014;22:550–551.
    1. Verthelyi D., Ishii K.J., Gursel M., Takeshita F., Klinman D.M. Human peripheral blood cells differentially recognize and respond to two distinct CPG motifs. J. Immunol. 2001;166:2372–2377.
    1. Wang Y., Cella M., Gilfillan S., Colonna M. Cutting edge: polyinosinic:polycytidylic acid boosts the generation of memory CD8 T cells through melanoma differentiation-associated protein 5 expressed in stromal cells. J. Immunol. 2010;184:2751–2755.
    1. Warfield K.L., Bosio C.M., Welcher B.C., Deal E.M., Mohamadzadeh M., Schmaljohn A., Aman M.J., Bavari S. Ebola virus-like particles protect from lethal Ebola virus infection. Proc. Natl. Acad. Sci. U. S. A. 2003;100:15889–15894.
    1. Warfield K.L., Swenson D.L., Negley D.L., Schmaljohn A.L., Aman M.J., Bavari S. Marburg virus-like particles protect guinea pigs from lethal Marburg virus infection. Vaccine. 2004;22:3495–3502.
    1. Warfield K.L., Swenson D.L., Olinger G.G., Kalina W.V., Aman M.J., Bavari S. Ebola virus-like particle-based vaccine protects nonhuman primates against lethal Ebola virus challenge. J. Infect. Dis. 2007;196(Suppl. 2):s430–s437.
    1. Watkins D.I. The hope for an HIV vaccine based on induction of CD8 + T lymphocytes–a review. Mem. Inst. Oswaldo Cruz. 2008;103:119–129.
    1. Wilkinson T.M., Li C.K., Chui C.S., Huang A.K., Perkins M., Liebner J.C., Lambkin-Williams R., Gilbert A., Oxford J., Nicholas B., Staples K.J., Dong T., Douek D.C., Mcmichael A.J., Xu X.N. Preexisting influenza-specific CD4 + T cells correlate with disease protection against influenza challenge in humans. Nat. Med. 2012;18:274–280.
    1. Wong G., Richardson J.S., Pillet S., Patel A., Qiu X., Alimonti J., Hogan J., Zhang Y., Takada A., Feldmann H., Kobinger G.P. Immune parameters correlate with protection against Ebola virus infection in rodents and nonhuman primates. Sci. Transl. Med. 2012;4:158ra146.
    1. Wong G., Audet J., Fernando L., Fausther-Bovendo H., Alimonti J.B., Kobinger G.P., Qiu X. Immunization with vesicular stomatitis virus vaccine expressing the Ebola glycoprotein provides sustained long-term protection in rodents. Vaccine. 2014;32:5722–5729.
    1. Zens K.D., Farber D.L. Memory CD4 T cells in influenza. Curr. Top. Microbiol. Immunol. 2015;386:399–421.
    1. Zhou Y., Callendret B., Xu D., Brasky K.M., Feng Z., Hensley L.L., Guedj J., Perelson A.S., Lemon S.M., Lanford R.E., Walker C.M. Dominance of the CD4(+) T helper cell response during acute resolving hepatitis A virus infection. J. Exp. Med. 2012;209:1481–1492.

Source: PubMed

3
購読する