Research paper
Preparation of H1N1 microneedles by a low-temperature process without a stabilizer

https://doi.org/10.1016/j.ejpb.2019.08.005Get rights and content

Abstract

During the manufacture of H1N1 microneedles, a stabilizer is usually added to maintain the antigenicity of the vaccine. However, finding a suitable stabilizer is difficult, and the addition of a stabilizer can limit the antigen dose and the addition of an adjuvant because of the limited volume of the microneedles. In this study, the authors evaluated whether H1N1 microneedles could be fabricated without a stabilizer by keeping the production environment at a low temperature.

H1N1 microneedle patches without a stabilizer were prepared in a process that involved maintaining a low temperature of 10 °C. The protective immune response to this method of drug application was investigated by comparing it with traditional intramuscular (IM) immunization and with the use of H1N1 microneedles with a stabilizer.

A process-sensitive antigen, H1N1, was stabilized without the use of a stabilizer in a process that maintained a low temperature of 10 °C. The preparation process consisted of coating and drying processes. In animal experiments, mice were immunized using an array of low-temperature H1N1 microneedles without a stabilizer (LT-MN), and they showed strong antibody responses. Compared to three other application methods of traditional IM immunization, low-temperature H1N1 microneedles with a stabilizer (LT-MN-T), and room-temperature H1N1 microneedles with a stabilizer (RT-MN-T), LT-MN produced comparable results in inducing protective immunity. A plaque reduction neutralization test found that LT-MN and LT-MN-T provided greater immunity compared with IM and RT-MN-T.

A process in which the temperature is maintained at 10 °C can provide successful vaccination with H1N1 microneedles without the addition of a stabilizer. This process can be applied to various temperature-sensitive biologics.

Graphical abstract

(1) Plaque reduction neutralization test (PRNT) for virus neutralization. IM (H1N1): intramuscular administration, RT-MN-T: room-temperature H1N1 microneedles with stabilizer, LT-MN-T: low-temperature H1N1 microneedles with stabilizer, LT-MN: low-temperature H1N1 microneedles without stabilizer. (2) The IM administration group had a lower neutralizing antibody than the microneedle group. (3) The microneedle group prepared at low temperature showed a similar antibody titer regardless of the addition of a stabilizer. (4) When the whole microneedle production process is performed at a low temperature, the influence of the stabilizer and the denaturation of the antigen can both be minimized, and as a result, immunogenicity can be induced effectively.

  1. Download : Download high-res image (79KB)
  2. Download : Download full-size image

Introduction

The microneedle system is designed to overcome the limitations of the existing transdermal drug delivery system [1], [2], [3]. Microneedles can deliver active ingredients into the skin through the stratum corneum regardless of the molecular weight and polarity of the ingredients [4], [5]. The use of microneedles for vaccinations has been studied and developed because microneedles can deliver the antigen into the skin layer, where the cells that need to receive the antigen are primarily located [6], [7], [8], [9]. Microneedles also provide the advantages of convenience and user compliance by avoiding the pain and fear of needles that patients often experience [10], [11]. Vaccine microneedles have shown efficacy equivalent to intramuscular (IM) injection [12], [13], [14]. Vaccine microneedles have also shown improved storage stability compared to the conventional formulation [15], [16]. However, in all previous studies, vaccine microneedles have required a stabilizer because the antigen was denatured during the process of preparation and storage [17], [18], [19]. An excessive amount of stabilizer was added to stabilize the antigen during the manufacturing process as well as during subsequent long-term storage [16], [20]. When the antigen was formulated for microneedles, great effort was necessary to screen the optimal stabilizer [21], [22]. There are a number of limitations related to the addition of the stabilizer: (1) the limitation of the appropriate co-solvent of the stabilizer and the antigen, (2) the limitation of the solubility of the stabilizer in the solvent, (3) the limitation of the amount of antigen that can be loaded into the formulation with the stabilizer, (4) the decrease in the mechanical strength of the dissolving microneedles, and (5) the phase separation of the formulation during the drying process [15], [23]. In particular, in the case of influenza antigens, the amount of stabilizer was 20 times more than that of the antigen, and the addition of an excessive amount of stabilizer could cause the above-mentioned limitations [12], [24].

The decrease in the stability of the antigen occurs rapidly at the stage where the liquid formulation is made into a microneedle shape and dried [17], [25]. If antigenicity is maintained during the manufacturing process, the addition of a stabilizer is less constrained because the change in antigenicity was mainly caused during the manufacturing process when the vaccine microneedles were stored at room temperature and below room temperature [20], [26]. Hemagglutinin (HA) activity, including H1N1, was stabilized mainly by adding a stabilizer during manufacturing, but HA activity did not change during long-term storage without a stabilizer at room temperature [16]. If HA activity can be maintained during the manufacturing process, the amount of antigen per unit area of microneedle array can be increased by minimization or elimination of the stabilizer.

The low-temperature process of producing vaccine microneedles was introduced for the first time to improve the stability of the antigen in the manufacturing process, resulting in the removal of the stabilizer during the manufacturing process. H1N1 vaccine microneedles were prepared by a low-temperature microneedle manufacturing system. To investigate the improvement in the stability of the antigen as a result of using the low-temperature process, we compared the antigenicity of low-temperature H1N1 microneedles without a stabilizer (LT-MN) to the antigenicity of IM immunization, room-temperature H1N1 microneedles with a stabilizer (RT-MN-T), and low-temperature H1N1 microneedles with a stabilizer (LT-MN-T). We then conducted animal experiments to confirm the improvement in stability by the low-temperature process.

Section snippets

Materials

Phosphate-buffered saline (PBS) and trehalose-dihydrate were obtained from Sigma-Aldrich (St. Louis, MO). Polylactic acid (PLA) was purchased from Lactel (Birmingham, AL). Carboxymethyl cellulose (CMC) was obtained from Whawon (Gyeonggi-do, South Korea). H1N1 antigen of seasonal influenza vaccine was provided by Il-Yang Pharmaceutical Co. (Yongin, Republic of Korea). The H1N1 antigen of seasonal influenza vaccine that we received from the manufacturer was composed of NaCl, NaH2PO4·nH2O, KH2PO4,

Fabrication of H1N1-coated microneedles

Each microneedle group was coated with each component in the ratio of the components shown in Table 1. The targeted amount, 3 μg of H1N1, was loaded on the surface of the microneedle structure using the dip-coating method at 25 °C for RT-MN-T and at 10 °C for LT-MN-T and LT-MN. The coating and drying process for LT-MN-T and LT-MN was performed in the 10 °C chamber of the open system. If the temperature inside the chamber and the reservoir was as low as 4 °C, condensation of water occurred on

Conclusions

In the case of the temperature-sensitive H1N1 antigen, denaturation of the antigen occurred during the manufacturing process, and the previous solution for inhibiting denaturation was the addition of stabilizers to the formulation. In this study, conducting the manufacturing process at a low temperature without a stabilizer was suggested. According to the results of the in vitro test, the antigenicity of H1N1 could be preserved by maintaining the temperature of the process at 10 °C without

Acknowledgement

This work was funded by grants from Korea Ministry of Trade, Industry & Energy, South Korea (MOTIE, 10067809 (Industrial Strategic Technology Development Program)) and Korea Ministry of Health and Welfare, South Korea (MoHW, HI15C297 (Technology Development Program of Responding to Infectious Disease)). We appreciate IL-yang Phar­ma­ceu­ti­cal Co. to provide anti­gen of sea­sonal in­fluenza vac­cine.

References (44)

  • M.J. Mistilis et al.

    Development of a thermostable microneedle patch for influenza vaccination

    J. Pharm. Sci.

    (2015)
  • N.K. Jain et al.

    Formulation and stabilization of recombinant protein based virus-like particle vaccines

    Adv. Drug Deliv. Rev.

    (2015)
  • S.-J. Kim et al.

    Stability evaluation of H3N2 influenza split vaccine in drying process for solidification

    J. Pharm. Investig.

    (2019)
  • A.P. Raphael et al.

    Formulations for microprojection/microneedle vaccine delivery: Structure, strength and release profiles

    J. Control. Release.

    (2016)
  • Y.-C. Kim et al.

    Formulation and coating of microneedles with inactivated influenza virus to improve vaccine stability and immunogenicity

    J. Control. Release.

    (2010)
  • J.C. Kapteyn et al.

    Haemagglutinin quantification and identification of influenza A&B strains propagated in PER. C6® cells: a novel RP-HPLC method

    Vaccine

    (2006)
  • J.W. Lee et al.

    Dissolving microneedles for transdermal drug delivery

    Biomaterials

    (2008)
  • A. Vrdoljak et al.

    Coated microneedle arrays for transcutaneous delivery of live virus vaccines

    J. Control. Release.

    (2012)
  • C. Edens et al.

    Measles vaccination using a microneedle patch

    Vaccine

    (2013)
  • M.-C. Chen et al.

    Enhancing immunogenicity of antigens through sustained intradermal delivery using chitosan microneedles with a patch-dissolvable design

    Acta Biomater.

    (2018)
  • A. Vrdoljak et al.

    Induction of broad immunity by thermostabilised vaccines incorporated in dissolvable microneedles using novel fabrication methods

    J. Control. Release.

    (2016)
  • Y. Hiraishi et al.

    Performance and characteristics evaluation of a sodium hyaluronate-based microneedle patch for a transcutaneous drug delivery system

    Int. J. Pharm.

    (2013)
  • Cited by (19)

    • Preparation of particle-attached microneedles using a dry coating process

      2022, Journal of Controlled Release
      Citation Excerpt :

      Microneedles provide a transdermal drug delivery system, that can deliver active pharmaceutical ingredients (API) into the skin regardless of the ingredients' molecular weight [1,2].

    • Microneedles

      2022, Microneedles
    • Microarray patches enable the development of skin-targeted vaccines against COVID-19

      2021, Advanced Drug Delivery Reviews
      Citation Excerpt :

      Although these promising results demonstrated the potential of MAP-directed skin vaccination, researchers sought alternative MAP concepts due to the aforementioned limitations of the ‘poke and patch’ approach, especially to enable more consistent and convenient delivery of vaccines to targeted skin microenvironments. Coated MAPs have been evaluated with several different vaccine types (e.g., live attenuated, viral vector, inactivated virus, subunit, DNA, and VLPs) for numerous viral infections, such as influenza, hepatitis B, rotavirus, RSV, Ebola, dengue, measles, polio, HIV, HPV, West Nile virus, chikungunya, HSV, and hepatitis C [270,275–303]. Vaccines delivered by coated MAPs have been mainly tested in mice, but there also have been a limited number of studies in rat, guinea pig, porcine, and non-human primate models [221,291,294,296,304,305].

    View all citing articles on Scopus
    1

    These authors equally contributed to this work.

    View full text