Microcrystalline Tyrosine and Aluminum as Adjuvants in Allergen-Specific Immunotherapy Protect from IgE-Mediated Reactivity in Mouse Models and Act Independently of Inflammasome and TLR Signaling

Deborah S Leuthard, Agathe Duda, Sandra N Freiberger, Sina Weiss, Isabella Dommann, Gabriele Fenini, Emmanuel Contassot, Matthias F Kramer, Murray A Skinner, Thomas M Kündig, Matthew D Heath, Pål Johansen, Deborah S Leuthard, Agathe Duda, Sandra N Freiberger, Sina Weiss, Isabella Dommann, Gabriele Fenini, Emmanuel Contassot, Matthias F Kramer, Murray A Skinner, Thomas M Kündig, Matthew D Heath, Pål Johansen

Abstract

Allergen immunotherapy (AIT) is the only modality that can modify immune responses to allergen exposure, but therapeutic coverage is low. One strategy to improve AIT safety and efficacy is the use of new or improved adjuvants. This study investigates immune responses produced by microcrystalline tyrosine (MCT)-based vaccines as compared with conventional aluminum hydroxide (alum). Wild-type, immune-signaling-deficient, and TCR-transgenic mice were treated with different Ags (e.g., OVA and cat dander Fel d 1), plus MCT or alum as depot adjuvants. Specific Ab responses in serum were measured by ELISA, whereas cytokine secretion was measured both in culture supernatants by ELISA or by flow cytometry of spleen cells. Upon initiation of AIT in allergic mice, body temperature and further clinical signs were used as indicators for anaphylaxis. Overall, MCT and alum induced comparable B and T cell responses, which were independent of TLR signaling. Alum induced stronger IgE and IL-4 secretion than MCT. MCT and alum induced caspase-dependent IL-1β secretion in human monocytes in vitro, but inflammasome activation had no functional effect on inflammatory and Ab responses measured in vivo. In sensitized mice, AIT with MCT-adjuvanted allergens caused fewer anaphylactic reactions compared with alum-adjuvanted allergens. As depot adjuvants, MCT and alum are comparably effective in strength and mechanism of Ag-specific IgG induction and induction of T cell responses. The biocompatible and biodegradable MCT seems therefore a suitable alternative adjuvant to alum-based vaccines and AIT.

Copyright © 2018 The Authors.

Figures

FIGURE 1.
FIGURE 1.
Immunogenicity testing of tyrosine- and alum-based vaccines in BALB/c mice. (A) Mice (n = 3–4) were injected with 0.01 or 0.1 μg OVA on alum (black symbols), MCT (open symbols), or in PBS (dashed lines) on days 0 and 28, and Ab titers were measured on days 28 and 42. Titration curves for individual mice are illustrated. (B) After a third injection with OVA 0.1 μg on day 56, splenocytes were harvested on day 62 and restimulated with OVA for analysis of cytokine secretion (mean + SD). (C) Mice (n = 5) were immunized with 1 (left) or 100 (right) μg OVA on days, 0, 14, and 28; total IgG, IgG1, IgG2a, IgG2b, IgG3, and IgE Abs were determined (mean ± SEM). The latter experiment is representative of three independent experiments with comparable results.
FIGURE 2.
FIGURE 2.
T cell response after MCT- or alum-adjuvanted immunization. C57BL/6 mice (n = 5) were adoptively transferred with OT-I and OT-II cells and immunized with 100 μg OVA on MCT (gray bars) or alum (black bars) 1 and 8 d later (individual mice are indicated). On day 15, spleen cells were analyzed by flow cytometry. (A) Dot blots and histograms show SIINFEKL-specific CD8 T cell activation (CD44) and proliferation (H2Kb pentamer). Frequencies of IFN-γ–producing and IFN-γ– and TNF-α–double-producing CD8 T cells (B) and CD4 T cells (C). (D) Splenocytes were also restimulated in vitro with MHC class I–restricted OVA aa 257–264 (SIINFEKL), MHC class II–restricted OVA aa 323–339, or OVA protein, and IFN-γ in culture supernatants was measured by ELISA. Mean ± SD of means are illustrated. The experiment is representative of two independent experiments with comparable results. *p < 0.05, **p < 0.01.
FIGURE 3.
FIGURE 3.
Immunotherapy of cat dander allergy. BALB/C mice (n = 5) were sensitized by weekly injections of cat fur allergen extract and received AIT with recombinant Fel d 1 allergen on alum (red) or MCT (blue) thrice s.c., as indicated (A). Finally, the mice were challenged with cat fur allergen extract to test tolerance to anaphylaxis. (B) Cat fur allergen-specific IgE (left) and Fel d 1–specific IgG2a (right) Ab titers were measured in sera before and after sensitization and AIT. (C) After challenge, changes in the body temperature were measured as an indicator for anaphylaxis. Left, Rectal body temperature as function of time after challenge. Right, Integrated AUC with baseline 38°C for the body temperature curves. (D) Splenocytes were restimulated with rFel d 1 for 20 h (IL-2) or 96 h (IFN-γ and IL-10), and cytokines were measured in the supernatants by ELISA. Abs are illustrated as mean ± SEM, and other results are illustrated as mean ± SD.
FIGURE 4.
FIGURE 4.
Safety testing of MCT- and alum-based vaccines in mice. BALB/C mice (n = 5) were sensitized by four weekly i.p. injections of OVA adsorbed on alum. Four weeks later, the mice received a single s.c. AIT with OVA on MCT (open circles and bars) or on alum (closed circles and bars). Body temperature changes are illustrated as a function of time after AIT (A) and integrated as AUC (B). The experiment is representative of two independent experiments with comparable results.
FIGURE 5.
FIGURE 5.
Immunogenicity testing in mice deficient in TLR signaling. (A) C57BL/6 wild-type mice as well as syngeneic MyD88- and TLR4-deficient mice were immunized with 40 μg OVA-MCT (gray circles) or 40 μg OVA-alum (black circles) on days 0 and 14. Abs in blood were measured by ELISA. (B) C57BL/6 mice as well as syngeneic TRIF-deficient mice were immunized with 10 μg OVA-MCT on days 0 and 14. Abs in blood were measured by ELISA. The experiment is representative of two independent experiments with comparable results and with four to six mice per group.
FIGURE 6.
FIGURE 6.
Assessment of inflammasome activation and other inflammatory reactions in vivo and in vitro. (AC) THP-1 human monocytes were incubated with various doses of MCT in the presence or absence of zVAD. IL-1β secretion (A) and viability (B) were measured by cytokine and lactate dehydrogenase cytotoxicity ELISA, respectively, in supernatants. (C) Pro–IL-1β and cleaved IL-1β were separated by Western blot using nigericin as a positive control. For the sake of presentation, the blot was cropped and spliced as indicated with the vertical white line, excess sample replicates being omitted. (D) MCT, alum, or PBS were injected i.p. in mice, and cell populations in the peritoneal lavage were analyzed by flow cytometry as indicated. Infiltration of CD11b– and Ly6G–double-positive neutrophils (E), CD11b– and SiglecF–double-positive eosinophils (F), F4/80-positive macrophages (G), MHC class II– and CD11b–double-positive cells (H), and CD11c–CD11b double-positive DCs (I) were measured 4 h (open bars) and 24 h (closed bars) postinjection. The results with wild-type mice are representative of three independent experiments. (J) Percentage of neutrophils, macrophages, and DCs in wild-type versus NALP3 inflammasome knockout mice.
FIGURE 7.
FIGURE 7.
Analysis of the role of inflammasome activation on adjuvant mechanism of action of MCT and alum with regard to stimulation of B and T cell responses. Wild-type C57BL/6 and ASC knockout mice (n = 5) were immunized on days 0, 14, and 26 with 0.1 μg OVA on MCT or alum. (A) Mice were bled on day 42, and OVA-specific IgG1, IgG2b, IgG2c, IgG3, and IgE Abs were measured by ELISA. The Ab results (mean + SD) are illustrated as OD measured at a serum dilution of 1:3000. (B) Mice were euthanized on day 48, and splenocytes were restimulated with OVA (+) or were not restimulated (−) for analysis of cytokine secretion by ELISA (mean + SD). The experiment was not repeated.

References

    1. Platts-Mills T. A. 2015. The allergy epidemics: 1870-2010. J. Allergy Clin. Immunol. 136: 3–13.
    1. Pawankar R. 2014. Allergic diseases and asthma: a global public health concern and a call to action. World Allergy Organ. J. 7: 12.
    1. Calderón M. A., Casale T. B., Togias A., Bousquet J., Durham S. R., Demoly P. 2011. Allergen-specific immunotherapy for respiratory allergies: from meta-analysis to registration and beyond. J. Allergy Clin. Immunol. 127: 30–38.
    1. Musa F., Al-Ahmad M., Arifhodzic N., Al-Herz W. 2017. Compliance with allergen immunotherapy and factors affecting compliance among patients with respiratory allergies. Hum. Vaccin. Immunother. 13: 514–517.
    1. Jutel M., Agache I., Bonini S., Burks A. W., Calderon M., Canonica W., Cox L., Demoly P., Frew A. J., O’Hehir R., et al. 2015. International consensus on allergy immunotherapy. J. Allergy Clin. Immunol. 136: 556–568.
    1. Reed S. G., Orr M. T., Fox C. B. 2013. Key roles of adjuvants in modern vaccines. Nat. Med. 19: 1597–1608.
    1. Hogenesch H. 2013. Mechanism of immunopotentiation and safety of aluminum adjuvants. Front. Immunol. 3: 406.
    1. Sun H., Pollock K. G., Brewer J. M. 2003. Analysis of the role of vaccine adjuvants in modulating dendritic cell activation and antigen presentation in vitro. Vaccine 21: 849–855.
    1. Eisenbarth S. C., Colegio O. R., O’Connor W., Sutterwala F. S., Flavell R. A. 2008. Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nature 453: 1122–1126.
    1. Kool M., Pétrilli V., De Smedt T., Rolaz A., Hammad H., van Nimwegen M., Bergen I. M., Castillo R., Lambrecht B. N., Tschopp J. 2008. Cutting edge: alum adjuvant stimulates inflammatory dendritic cells through activation of the NALP3 inflammasome. J. Immunol. 181: 3755–3759.
    1. Li H., Willingham S. B., Ting J. P., Re F. 2008. Cutting edge: inflammasome activation by alum and alum’s adjuvant effect are mediated by NLRP3. J. Immunol. 181: 17–21.
    1. McKee A. S., Munks M. W., MacLeod M. K., Fleenor C. J., Van Rooijen N., Kappler J. W., Marrack P. 2009. Alum induces innate immune responses through macrophage and mast cell sensors, but these sensors are not required for alum to act as an adjuvant for specific immunity. J. Immunol. 183: 4403–4414.
    1. Awate S., Babiuk L. A., Mutwiri G. 2013. Mechanisms of action of adjuvants. Front. Immunol. 4: 114.
    1. Jensen-Jarolim E. 2015. Aluminium in allergies and allergen immunotherapy. World Allergy Organ. J. 8: 7.
    1. Rosewich M., Lee D., Zielen S. 2013. Pollinex Quattro: an innovative four injections immunotherapy in allergic rhinitis. Hum. Vaccin. Immunother. 9: 1523–1531.
    1. Roger A., Depreux N., Jurgens Y., Serra A. T., Heath M. D., Garcia G., Skinner M. A. 2016. A novel microcrystalline tyrosine-adsorbed, mite-allergoid subcutaneous immunotherapy: 1-year follow-up report. Immunotherapy 8: 1169–1174.
    1. Cabral-Miranda G., Heath M. D., Gomes A. C., Mohsen M. O., Montoya-Diaz E., Salman A. M., Atcheson E., Skinner M. A., Kramer M. F., Reyes-Sandoval A., Bachmann M. F. 2017. Microcrystalline tyrosine (MCT®): a depot adjuvant in licensed allergy immunotherapy offers new opportunities in malaria. Vaccines (Basel) 5: E32.
    1. Cabral-Miranda G., Heath M. D., Mohsen M. O., Gomes A. C., Engeroff P., Flaxman A., Leoratti F. M. S., El-Turabi A., Reyes-Sandoval A., Skinner M. A., et al. 2017. Virus-like particle (VLP) plus microcrystalline tyrosine (MCT) adjuvants enhance vaccine efficacy improving T and B cell immunogenicity and protection against Plasmodium berghei/vivax. Vaccines (Basel) 5: E10.
    1. Heath M. D., Swan N. J., Marriott A. C., Silman N. J., Hallis B., Prevosto C., Gooch K. E., Skinner M. A. 2017. Comparison of a novel microcrystalline tyrosine adjuvant with aluminium hydroxide for enhancing vaccination against seasonal influenza. BMC Infect. Dis. 17: 232.
    1. Martinon F., Pétrilli V., Mayor A., Tardivel A., Tschopp J. 2006. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 440: 237–241.
    1. Li P., Allen H., Banerjee S., Franklin S., Herzog L., Johnston C., McDowell J., Paskind M., Rodman L., Salfeld J., et al. 1995. Mice deficient in IL-1 beta-converting enzyme are defective in production of mature IL-1 beta and resistant to endotoxic shock. Cell 80: 401–411.
    1. Hjálmsdóttir Á., Bühler C., Vonwil V., Roveri M., Håkerud M., Wäckerle-Men Y., Gander B., Johansen P. 2016. Cytosolic delivery of liposomal vaccines by means of the concomitant photosensitization of phagosomes. Mol. Pharm. 13: 320–329.
    1. Johansen P., Senti G., Martinez Gomez J. M., Storni T., von Beust B. R., Wüthrich B., Bot A., Kündig T. M. 2005. Toll-like receptor ligands as adjuvants in allergen-specific immunotherapy. Clin. Exp. Allergy 35: 1591–1598.
    1. Johansen P., Senti G., Martínez Gómez J. M., Wüthrich B., Bot A., Kündig T. M. 2005. Heat denaturation, a simple method to improve the immunotherapeutic potential of allergens. Eur. J. Immunol. 35: 3591–3598.
    1. Martínez-Gómez J. M., Johansen P., Erdmann I., Senti G., Crameri R., Kündig T. M. 2009. Intralymphatic injections as a new administration route for allergen-specific immunotherapy. Int. Arch. Allergy Immunol. 150: 59–65.
    1. Martínez-Gómez J. M., Johansen P., Rose H., Steiner M., Senti G., Rhyner C., Crameri R., Kündig T. M. 2009. Targeting the MHC class II pathway of antigen presentation enhances immunogenicity and safety of allergen immunotherapy. Allergy 64: 172–178.
    1. Oleszycka E., Moran H. B., Tynan G. A., Hearnden C. H., Coutts G., Campbell M., Allan S. M., Scott C. J., Lavelle E. C. 2016. IL-1α and inflammasome-independent IL-1β promote neutrophil infiltration following alum vaccination. FEBS J. 283: 9–24.
    1. Klimek L., Schmidt-Weber C. B., Kramer M. F., Skinner M. A., Heath M. D. 2017. Clinical use of adjuvants in allergen-immunotherapy. Expert Rev. Clin. Immunol. 13: 599–610.
    1. Pfaar O., Cazan D., Klimek L., Larenas-Linnemann D., Calderon M. A. 2012. Adjuvants for immunotherapy. Curr. Opin. Allergy Clin. Immunol. 12: 648–657.
    1. Moingeon P. 2012. Adjuvants for allergy vaccines. Hum. Vaccin. Immunother. 8: 1492–1498.
    1. Parmley S. 2014. Boosting adjuvants. Science-Business eXchange 7: 1–3.
    1. Casale T. B., Stokes J. R. 2014. Immunotherapy: what lies beyond. J. Allergy Clin. Immunol. 133: 612–619, quiz 620.
    1. Wheeler A. W., Moran D. M., Robins B. E., Driscoll A. 1982. l-Tyrosine as an immunological adjuvant. Int. Arch. Allergy Appl. Immunol. 69: 113–119.
    1. Shamji M. H., Kappen J. H., Akdis M., Jensen-Jarolim E., Knol E. F., Kleine-Tebbe J., Bohle B., Chaker A. M., Till S. J., Valenta R., et al. 2017. Biomarkers for monitoring clinical efficacy of allergen immunotherapy for allergic rhinoconjunctivitis and allergic asthma: an EAACI position paper. Allergy 72: 1156–1173.
    1. Magnan A., Mély L., Prato S., Vervloet D., Romagné F., Camilla C., Necker A., Casano B., Montero-Jullian F., Fert V., et al. 2000. Relationships between natural T cells, atopy, IgE levels, and IL-4 production. Allergy 55: 286–290.
    1. Poulsen L. K., Hummelshoj L. 2007. Triggers of IgE class switching and allergy development. Ann. Med. 39: 440–456.
    1. Akdis C. A., Akdis M. 2015. Mechanisms of allergen-specific immunotherapy and immune tolerance to allergens. World Allergy Organ. J. 8: 17.
    1. Shamji M. H., Ljørring C., Francis J. N., Calderon M. A., Larché M., Kimber I., Frew A. J., Ipsen H., Lund K., Würtzen P. A., Durham S. R. 2012. Functional rather than immunoreactive levels of IgG4 correlate closely with clinical response to grass pollen immunotherapy. Allergy 67: 217–226.
    1. Vasou A., Sultanoglu N., Goodbourn S., Randall R. E., Kostrikis L. G. 2017. Targeting pattern recognition receptors (PRR) for vaccine adjuvantation: from synthetic PRR agonists to the potential of defective interfering particles of viruses. Viruses 9: 186.
    1. Gavin A. L., Hoebe K., Duong B., Ota T., Martin C., Beutler B., Nemazee D. 2006. Adjuvant-enhanced antibody responses in the absence of toll-like receptor signaling. Science 314: 1936–1938.
    1. Bell A. J., Heath M. D., Hewings S. J., Skinner M. A. 2015. The adsorption of allergoids and 3-O-desacyl-4′-monophosphoryl lipid A (MPL®) to microcrystalline tyrosine (MCT) in formulations for use in allergy immunotherapy. J. Inorg. Biochem. 152: 147–153.
    1. McDougall S. A., Heath M. D., Kramer M. F., Skinner M. A. 2016. Analysis of aluminium in rat following administration of allergen immunotherapy using either aluminium or microcrystalline-tyrosine-based adjuvants. Bioanalysis 8: 547–556.
    1. Baldrick P., Richardson D., Wheeler A. W. 2002. Review of L-tyrosine confirming its safe human use as an adjuvant. J. Appl. Toxicol. 22: 333–344.
    1. Bortolatto J., Mirotti L., Rodriguez D., Gomes E., Russo M. 2015. Adsorption of Toll-like receptor 4 agonist to alum-based tetanus toxoid vaccine dampens pro-T helper 2 activities and enhances antibody responses. J. Immunol. Res. 2015: 280238.
    1. Wheeler A. W., Marshall J. S., Ulrich J. T. 2001. A Th1-inducing adjuvant, MPL, enhances antibody profiles in experimental animals suggesting it has the potential to improve the efficacy of allergy vaccines. Int. Arch. Allergy Immunol. 126: 135–139.
    1. Drachenberg K. J., Wheeler A. W., Stuebner P., Horak F. 2001. A well-tolerated grass pollen-specific allergy vaccine containing a novel adjuvant, monophosphoryl lipid A, reduces allergic symptoms after only four preseasonal injections. Allergy 56: 498–505.
    1. Mothes N., Heinzkill M., Drachenberg K. J., Sperr W. R., Krauth M. T., Majlesi Y., Semper H., Valent P., Niederberger V., Kraft D., Valenta R. 2003. Allergen-specific immunotherapy with a monophosphoryl lipid A-adjuvanted vaccine: reduced seasonally boosted immunoglobulin E production and inhibition of basophil histamine release by therapy-induced blocking antibodies. Clin. Exp. Allergy 33: 1198–1208.
    1. Creticos P. S., Schroeder J. T., Hamilton R. G., Balcer-Whaley S. L., Khattignavong A. P., Lindblad R., Li H., Coffman R., Seyfert V., Eiden J. J., Broide D., Immune Tolerance Network Group 2006. Immunotherapy with a ragweed-toll-like receptor 9 agonist vaccine for allergic rhinitis. N. Engl. J. Med. 355: 1445–1455.
    1. DuBuske L. M., Frew A. J., Horak F., Keith P. K., Corrigan C. J., Aberer W., Holdich T., von Weikersthal-Drachenberg K. J. 2011. Ultrashort-specific immunotherapy successfully treats seasonal allergic rhinoconjunctivitis to grass pollen. Allergy Asthma Proc. 32: 239–247.
    1. Jensen-Jarolim E., Pali-Schöll I., Roth-Walter F. 2017. Outstanding animal studies in allergy II. From atopic barrier and microbiome to allergen-specific immunotherapy. Curr. Opin. Allergy Clin. Immunol. 17: 180–187.
    1. Jensen-Jarolim E., Pali-Schöll I., Roth-Walter F. 2017. Outstanding animal studies in allergy I. From asthma to food allergy and anaphylaxis. Curr. Opin. Allergy Clin. Immunol. 17: 169–179.

Source: PubMed

3
Abonner