Elsevier

Colloids and Surfaces B: Biointerfaces

Volume 174, 1 February 2019, Pages 536-543
Colloids and Surfaces B: Biointerfaces

Novel imiquimod nanovesicles for topical vaccination

https://doi.org/10.1016/j.colsurfb.2018.11.031Get rights and content

Highlights

  • High accumulation of imiquimod in human skin.

  • Induction of pro-inflammatory cytokines TNF-α and IL-6.

  • Topical induction of cell-biased systemic response against seasonal influenza vaccine.

Abstract

Development of needle and pain free noninvasive immunization procedures is a top priority for public health agencies. In this work the topical adjuvant activity of the immunomodulator imiquimod (IMQ) carried by ultradeformable archaeosomes (UDA2) (nanovesicles containing sn-2,3 ether linked phytanyl saturated archaeolipids) was surveyed and compared with that of ultradeformable liposomes lacking archaeolipids (UDL2) and free IMQ, using the model antigen ovalbumin and a seasonal influenza vaccine in Balb/c mice. UDA2 (250 ± 94 nm, -26 ± 4 mV Z potential) induced higher IMQ accumulation in human skin and higher production of TNF-α and IL-6 by macrophages and keratinocytes than free IMQ and UDL2. Mixed with ovalbumin, UDA2 was more efficient at generating cellular response, as measured by an increase in serum IgG2a and INF-γ production by splenocytes, compared with free IMQ and UDL2. Moreover, mixed with a seasonal influenza vaccine UDA2 produced same IgG titers and IgG2a/IgG1 isotypes ratio (≈1) than the subcutaneously administered influenza vaccine. Topical UDA2 however, induced highest stimulation index and INF-γ levels by splenocytes. UDA2 might be a promising adjuvant for topical immunization, since it produced cell-biased systemic response with ≈ 13-fold lower IMQ dose than the delivered as the commercial IMQ cream, Aldara.

Introduction

The development of needle and pain free noninvasive immunization procedures is a top priority for public health agencies [1]. Topical vaccination however, is challenged by the stratum corneum (SC) barrier interposed between antigens-adjuvants and the skin-associated lymphoid tissue (SALT), lying few hundred micrometers depth from skin surface [2,3]. Topical vaccination with soft matter is a new attractive way to induce immune protection without disrupting the SC. In this scenario, we previously showed that topically applied ultradeformable archaeosomes (UDA: made of total polar archaeolipids (TPA) extracted from the archaea Halorubrum tebenquichense plus soybean phosphatidylcholine and sodium cholate (3:3:1 w/w), can penetrate the SC to induce antigen specific IgG serum responses [[4], [5], [6]]. TPA, key components of UDA, are a mixture of sn-2,3 glycerol ether linked fully saturated polyisoprenoid chains phospholipids. TPA extracted from H. tebenquichense are ligands for scavenger receptor class A expressed on phagocytic cells and immature antigen presenting cells (APC) [7]. Vesicles fully or partly made of TPA such as archaeosomes or UDA, are much more pronouncedly captured by APC than TPA-lacking vesicles. On the other hand, TPA confer UDA stability to heat sterilization and storage under cold-free conditions [8]. The immune response elicited by topical UDA against ovalbumin and leishmania antigens, however, is lower than the one obtained after parenteral administration using the universal adjuvant alum or vesicles fully made of TPA [4,5]. Moreover, UDA elicit an IgG2a/IgG1 ratio ≈ 0.5, suggesting an humoral (Th2) biased response [4,8].

Imiquimod (IMQ), on the other hand, is a synthetic ligand of the endosomal Toll Like Receptor 7 (TLR7), that induces the production of cytokines such as IFN-α, TNF-α, IL-1a, IL-6, IL-8 and IL-12 in immune cells like monocytes/macrophages, dendritic cells (DC) (dermal DC and plasmacytoid DC), B cells and granulocytes [9,10]. IMQ induces cellular memory via the activation of effector T cells, Langerhans cells (LC), lymphocytes, and macrophages, which circulate lymphatically and activate the adaptive immune response [11]. Topically administered IMQ induces functional maturation of epidermal LC cells and bone marrow epidermal DC and stimulates migration of these APC to regional lymph nodes where they promote a specific T cell response [9]. A 5% IMQ cream called Aldara is approved to treat dermatologic skin pathologies including external genital and perianal warts, small superficial basal cell carcinoma and actinic keratoses on the face and scalp [12]. Aldara is an oil-in-water emulsion consisting of isostearic acid as the oil phase in which IMQ is solubilized at 5%. In clinical trials, topical application of Aldara to patients with melanoma and prostate cancer vaccinated with peptides has shown to increase cytotoxic responses [13,14]. However, responses induced by Aldara rapidly fade away, resulting in poor memory formation and only partial tumor protection [15,16]. Moreover, isostearic acid cause frequent cutaneous adverse reactions in 75% of the patients and has been recently found that it contributes to inflammatory responses via inflammasome activation [17]. Besides, this emulsion is not stable, Aldara cream is marketed in single-use packaging and unused packets are recommended to be discarded. On the other hand, IMQ shows very poor skin permeability [18]. For topical vaccination, IMQ needs to cross the SC, enter the skin and be taken up by skin-resident LC and DC for activation and migration to draining lymph nodes. At the same time, to avoid any dangerous systemic adverse effect, IMQ should remain at the administration site, not entering the bloodstream. All these data suggest that a new IMQ formulation for topical vaccination is needed.

IMQ has been formulated in different types of particulate carriers such as nanogels [19], solid nanoemulsions [20] and emulsions [21] for topical immunization. All these approaches, however are based on the release of IMQ from the skin surface and not on the target of IMQ loaded nanoparticles to APC. Recently we have showed that aqueous suspension of vesicles fully made by TPA containing IMQ are capable of inducing cell-biased antigen-specific systemic response upon subcutaneous application [22].

The hypothesis of this work is that the immune response elicited by UDA or IMQ alone, may be improved by the inclusion of IMQ in UDA to gain skin penetration and APC and endosomal targeting. IMQ was loaded into and externally associated with nanovesicles (UDA2 and UDL2 - ultradeformable nanovesicles lacking TPA-). The in vitro penetration on human skin, cytotoxicity, and the release of proinflammatory cytokines by keratinocytes and macrophages upon incubation with nanovesicles were determined. Then, the ability of topical nanovesicles to stimulate immune responses toward the model antigen ovalbumin and a seasonal influenza vaccine were screened. Finally, storage stability was determined.

Section snippets

Materials

Soybean phosphatidylcholine (SPC, purity >90%) was a gift from Lipoid, Ludwigshafen, Germany. Imiquimod (purity >98%) was a gift from Laboratorio Lazar (Buenos Aires, Argentina). Sodium cholate (NaChol), Ovalbumin grade V (OVA), 2,2-azino-bis (3-ethylbenzthiazoline-6-sulphonic acid) (ABTS), lipopolysaccharides from Escherichia coli 0111:B4 (LPS), Laurdan and 3-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyl tetrazolium bromide (MTT) were from Sigma-Aldrich (MO, USA). Roswell Park Memorial Institute

Nanovesicles preparation and characterization

IMQ is a small molecule (MW 240.3 Da) of the imidazoquinoline amines family poorly soluble in either hydrophilic or lipophilic solvents [30]. IMQ is practically insoluble in water at physiological pH, in common organic solvents and in pharmaceutical excipients. As a weak base of pKa 7.3, IMQ is soluble in 0.1 M lactic acid and in fatty acids such as stearic acid, oleic acid and linoleic acid [30]. These solubility issues hinder the incorporation of large amounts of IMQ into ultradeformable

Conclusions

Overall, here we report novel IMQ-ultradeformable archaeosomes for topical vaccination. These nanovesicles induced the secretion of proinflammatory cytokines by macrophages and keratinocytes in vitro. More important after topical application UDA2 elicited high levels of serum IgG2a antibodies and INF-γ production meaning that UDA2 effectively induced a more Th1-mediated immune response, compared with UDA and IMQ alone. UDA2 would be an alternative adjuvant for needle and pain free vaccination

Acknowledgments

The authors would like to thank to LME/LNNano for the use of electron microscopy facility and technical support. This work was supported by the ANPCYT under Grant PICT 2011-2402 and Secretaria de Investigaciones Universidad Nacional de Quilmes under Grant Nanomedicinas-2. ATC and MJA have fellowships from National Council for Scientific and Technological Research (CONICET). ELR, MJM, APP are members of the Research Career Program from CONICET.

References (46)

  • S.M. Bal et al.

    J. Control. Release

    (2010)
  • L. Higa et al.

    Nanomedicine

    (2012)
  • D. Carrer et al.

    Colloids Surf. B Biointerfaces

    (2014)
  • A.T. Caimi et al.

    Colloids Surf. B Biointerfaces

    (2017)
  • P. Stein et al.

    J. Invest. Dermatol.

    (2011)
  • I. Telò et al.

    Int. J. Pharm.

    (2016)
  • K. Gogoll et al.

    Cell. Immunol.

    (2016)
  • P.A. Lopez et al.

    J. Dermatol. Sci.

    (2017)
  • B. Van den Bergh et al.

    Int. J. Pharm.

    (2001)
  • N. Wang et al.

    Eur. J. Pharm. Biopharm.

    (2014)
  • G. Cevc

    Adv. Drug Deliv. Rev.

    (2004)
  • M.M. Elsayed et al.

    Int. J. Pharm.

    (2006)
  • J. Brewer et al.

    J. Invest. Dermatol.

    (2013)
  • M.P. Schön et al.

    J. Invest. Dermatol.

    (2006)
  • A.S. Goebel et al.

    Eur. J. Pharm. Biopharm.

    (2010)
  • J. Hickling et al.
    (2011)
  • N. Kirschner et al.

    Acad. Sci.

    (2012)
  • L. Higa et al.

    PLoS One

    (2016)
  • M.J. Altube et al.

    J. Mater. Chem. B

    (2017)
  • M. Stanley

    Clin. Exp. Dermatol.

    (2002)
  • M. Schön et al.

    Br. J. Dermatol. Syph.

    (2007)
  • G. Rechtsteiner et al.

    J. Immunol.

    (2005)
  • D. Vidal et al.

    Expert Rev. Dermatol.

    (2018)
  • Cited by (8)

    • Ether lipids from archaeas in nano-drug delivery and vaccination

      2023, International Journal of Pharmaceutics
    • Improving Vaccine-Induced Immunity: Can Baseline Predict Outcome?

      2020, Trends in Immunology
      Citation Excerpt :

      Furthermore, oral administration of the immunosuppressant rapamycin has been shown to increase the immune response to influenza virus vaccination (higher antibody response) in older adults relative to those not having received rapamycin [55]. In addition to rapamycin, other small molecules and biologics with immunomodulatory function, such as imiquimod, are currently approved for clinical use [56–58]. Thus, the tools to modulate immune baseline status to potentially improve the outcome of vaccination are already available.

    • The anti MRSA biofilm activity of Thymus vulgaris essential oil in nanovesicles

      2019, Phytomedicine
      Citation Excerpt :

      Opposite to PL, the diphytanylglycerol diethers display sn2,3 stereoisomerism and fully saturated polyprenol chains are linked by ether instead of esters bond to the glycerol backbone. Different types of archaeolipids-containing soft colloidal nanostructures have repeatedly shown increased resistance to multiple insults such as mechanical stress, chemoenzymatic hydrolysis and oxidation, improving the protection to the carried drugs (Caimi et al., 2017 and 2019; Altube et al., 2016 and 2017). We hypothesize that T formulated in archaeolipid-containing nanovesicles would display improved structural protection during handling and storage, a fact that may help to increase its antimicrobial activity.

    View all citing articles on Scopus
    View full text