4,741
Views
105
CrossRef citations to date
0
Altmetric
Review

The success of microneedle-mediated vaccine delivery into skin

, &
Pages 2975-2983 | Received 28 Jan 2016, Accepted 23 Mar 2016, Published online: 01 Nov 2016

ABSTRACT

Microneedles (MNs) are designed to specifically target the outermost, skin barrier layer, the stratum corneum, creating transient pathways for minimally invasive transcutaneous delivery. It is reported that MNs can facilitate delivery without stimulating the pain receptors or damaging blood vessels that lie beneath, thus being perceived as painless and associated with reduced bleeding. This immunocompetence of the skin, coupled with its ease of access, makes this organ an attractive vaccination site. The purpose of this review was to collate primary scientific literature pertaining to MN-mediated in vivo vaccination programmes. A total of 62 original research articles are presented, compiling vaccination strategies in 6 different models (mouse, rat, guinea pig, rabbit, pig, macaque and human). Vaccines tested span a wide range of viral, bacterial and protozoan pathogens and includes 7 of the 13 vaccine-preventable diseases, as defined by the WHO. This review highlights the paucity of available clinical trial data. MN-delivered vaccines have demonstrated safety and immunogenicity in pre-clinical models and boast desirable attributes such as painless administration, thermostability, dose-sparing capacity and the potential for self-administration. These advantages should contribute to enhanced global vaccine access.

Introduction

Vaccine delivery

Vaccines are conventionally administered using a hypodermic needle.Citation1 This form of administration provides a rapid and direct method of vaccine delivery. Despite familiarity, widespread use and proven efficacy, the hypodermic needle is associated with accidental needle stick injury, spread of blood-borne infections,Citation2-4 as well as phobias, pain and significant anxiety.Citation5-8 In addition, these needles are not easily self-administered, unless the individual has received specialized training on injection technique and needle disposal.Citation9 Oral vaccination is an attractive alternative Citation10 and a limited number of oral vaccines have been approved for human use.Citation11-15 However, this mode of immunisation can be less effective, as vaccine antigens undergo digestion in the gastrointestinal tract prior to induction of an adequate immune response Citation10 and research on their use has been limited almost exclusively to protection against mucosally transmitted pathogens, with some notable recent exceptions.Citation15 The transdermal route, based on diffusion, has also been investigated. However this route limits delivery only to lipophilic, low molecular weight potent products Citation16 and would prevent a vaccine from crossing the skin due to the presence of the relatively impermeable, outer stratum corneum (SC) layer. Intradermal vaccination is not a novel concept. In 1910, French physician Charles Mantoux published his clinical research on the intradermal injection of tuberculin as a diagnostic test for tuberculosis disease.Citation17 This diagnostic technique formed the basis for intradermal vaccination, a technique still in use for vaccines such as rabiesCitation18 and BCG.Citation19 However, intradermal delivery is technically challenging, requiring significant operator training Citation20 and has been associated with adverse events such as pain, inflammatory changesCitation21 and the development of abscesses.Citation22 Taking into account the limitations of parenteral, oral and traditional transdermal and intradermal vaccination, the concept of the microneedle (MN) emerged as a solution to these issues. MNs can be 1µm in diameter and range from 50µm to 1000µm in length, while mini-needles range from 1000µm to 1500µm.Citation23 They are designed to specifically target the outermost, rate limiting, skin barrier layer, the SC, creating transient pathways for minimally invasive transcutaneous delivery.Citation24 There are 4 different types of MNs: solid, coated, hollow and dissolving. It is reported that MNs can facilitate delivery through SC interruption without stimulating the pain receptors and blood vessels that lie beneath, thus being perceived as painless and associated with a reduction in bleeding.Citation1,25,26 Other advantages of microneedle-mediated delivery include avoidance of first pass metabolism; potential for highly targeted administration to individual cells;Citation26,27 improved patient compliance;Citation28 dose sparing;Citation29,30 thermostability of certain platforms Citation31-33 and potential for self-administration.

The skin: An immune organ and vaccine target

Skin is the largest immune organ in the human body,Citation34 composed of 2 primary layers, the epidermis and dermis.Citation35 These layers provide a protective interface between internal organs and the external environment, encountering a host of toxins, pathogenic organisms and physical stresses.Citation36 The skin functions as more than just a physical barrier. It is capable of mounting a potent immune response due to the residence of specialized antigen presenting cells. Langerhans cells are abundant in the epidermis, comprising 2% to 4% of epithelial cells,Citation36 while more classical dendritic cells are found in the dermis.Citation36-40 Other immune-competent accessory cells residing in the skin include keratinocytes, epidermal cells which play a role in initiating cell-mediated immune responses through the release of cytokines and the expression of cellular adhesion molecules to facilitate movement and coordination with other immune cells; T lymphocytes; melanocytes, epidermal pigment cells which produce a number of cytokines that mediate inflammation and mast cells, leukocytes which modulate host innate immune response through the release of granular and secreted mediators and recruit multiple inflammatory cells through the production of chemotactic factors.Citation36,41-43 The resident professional APC are adept at antigen capture, and upon appropriate activation through intracellular interaction, migrate to proximal lymph nodes to activate B and T lymphocytes and mediate initiation of an adaptive immune response.Citation37,44 This immunocompetence, coupled with its ease of access, makes the skin an attractive vaccination target.

Literature review

The purpose of this review is to collate literature detailing the success of MN-mediated in vivo vaccination programmes. Keywords including ‘microneedle’, ‘solid microneedle’, ‘coated microneedle’, ‘hollow microneedle’ ‘dissolvable microneedle’, ‘dissolving microneedle’, were combined with ‘vaccine’, ‘vaccination’ and ‘immunisation’. Using Google as a search engine, these keywords were combined in various permutations and combinations to search PubMed. This yielded a total of 748 results. Following removal of duplications, 180 results remained. The title and abstract of each result were examined and included or excluded in the final review based on the criteria outlined in . A total of 62 results were included in the final review.Citation29,45-105

Table 1. Inclusion and exclusion criteria.

Solid MNs

The simplest forms of MNs are solid devices. Solid MNs create transient micropores in the SC, thereby increasing permeability of the barrier layer. Vaccine applied onto the treated surface diffuses into the skin (from a loaded patch or semi-solid formulation) through the pores created by MN pre-treatment. The applied vaccine can exert a local effect in the skin and a systemic effect following uptake.Citation1 Solid MNs have been used to deliver vaccines for diphtheria,Citation45,46,48 influenza,Citation46 hepatitis B Citation47,49,50 and malaria Citation51,52 in mice. Microenhancer array devices were developed to cause mild abrasion of the SC. These devices scrape the skin with blunt-tipped microneedles and have been used to increase the delivery of an anthrax vaccine in mice and rabbits,Citation53 a Japanese encephalitis vaccine in cynomolgus monkeys Citation56 and a rabies vaccine in humans.Citation57,58 While these devices were shown to be effective, intradermal injection of the vaccine in each of these studies was significantly more effective, potentially due to inefficient delivery into the skin from the formulation. In spite of immunogenicity, the popularity of solid MNs has reduced in recent years, potentially due to the requirement for a multi-step administration process, the lack of consistency and the increased number of advantages of other MN systems.

Coated microneedles

Advancement on solid MNs was the development of coated devices. Solid MNs are pre-coated with a vaccine in a formulation suitable for coating and dissolution,Citation1 thus resulting in a one-step delivery process. The vaccine coated MNs are inserted into the skin, where dissolution of the vaccine occurs. Vaccine delivery via coated MNs is limited by the dimensions of the MN shaft and tip.Citation106,107 Successful vaccine coated MNs include influenza,Citation54,55,59-77 human papillomavirus,Citation78,79 chikungunya virus,Citation80 West Nile virus,Citation80 rotavirus,Citation81 herpes simplex virus Citation82 and hepatitis C Citation83 in mice, influenza virus Citation84 and bacillus Calmette-Guérin in guinea pigs,Citation85 hepatitis B virus in pigs,Citation86 and measles Citation87 and polio Citation88 viruses in rats. This literature search did not reveal any clinical trials pertaining to vaccine delivery via coated microneedles.

Hollow microneedles

Hollow MNs provide a pre-defined conduit for vaccine delivery into the skin or other tissue. Currently there are 2 hollow MN designs: a single MN or mini-needle, which mimics the conventional hypodermic needle Citation108 or an array of multiple MNs Citation109. The latter permits simultaneous application of a vaccine formulation over a wider area of skin, potentially resulting in higher bioavailability and increasing the likelihood of lymphatic uptake of presented antigens.Citation110 Vaccine may be delivered by passive diffusion through the MN. Conversely, a syringe may be attached to the MN, permitting active vaccine delivery. There are several commercially available hollow MN systems; Soluvia® is licensed for use Citation57,91 and MicronJet® is being clinically tested.Citation29 Soluvia® is a pre-fillable microinjection system with a single 1500µm hollow silicon MN, while MicronJet® is composed of 4 600µm hollow silicon MNs arranged on a plastic adaptor for attachment to a standard syringe barrel.Citation23 Hollow MNs have been successfully developed to immunise human subjects with polio Citation89,90 or influenza Citation29,91,92 vaccines, to immunise mice against plague Citation93 and to administer polio vaccine to rats.Citation94

Dissolving microneedles

The final, most advanced and complex MN is the dissolving MN. Dissolving MNs are polymeric and encapsulate vaccine within their matrix.Citation1,111,112 Insertion of the MNs into the skin catalyzes the degradation of the polymeric compound, subsequently releasing the vaccine.Citation112 Unlike the alternate MN platforms already discussed, dissolving MNs are fully biocompatible and do not generate biohazardous waste, a distinct advantage.Citation113,114 Other advantages include robustness and scalability.Citation115,116 However, unlike hollow MN, a limitation is placed on the amount of vaccine that can be incorporated into the system Citation117 and vaccinees may be obliged to wait for extended periods of time to ensure complete MN degradation.Citation114 Dissolving MNs have been developed to incorporate vaccines for influenza virus,Citation95-100 hepatitis B,Citation101,102 tetanus Citation97, diphtheria,Citation97 malaria Citation97 and HIV Citation103 in mice and measles Citation104 and polio Citation105 in rhesus macaques, with a long term aim to create a thermostable, self-administration platform. Although an attractive platform, dissolvable microneedle (DMN) systems for vaccine delivery have required more time to reach clinical trials compared to hollow or solid microneedles. Hollow and solid MN devices have a traditional medical device classification. In contrast, DMN patches will likely be seen, from a regulatory perspective, as a combination product of a medicinal product (the vaccine) and a device (potential backing layers and/or applicators). However, as a new dosage format, the product specifications, critical quality attributes of each product and regulatory pathway of DMN systems has not yet been defined. Furthermore, to ensure the quality of vaccine-loaded DMN patches that will be clinically used, they must be produced in the appropriate environment that complies with, good manufacturing practice (GMP). These processes, guidelines and regulatory strategies are only recently being defined.Citation118

Discussion

The purpose of this review was to collate primary scientific literature pertaining to MN-mediated in vivo vaccination programs, according to the inclusion and exclusion criteria outlined in . A total of 62 original research articles are presented, compiling vaccination strategies in 6 different models (mouse,Citation45-52,54,55,59-83,93,95-103] rat,Citation87,88,94, guinea pig,Citation84,85 rabbit,Citation53 pig,Citation86 cynomolgus Citation56 or rhesus macaqueCitation104,105) and in human subjects.Citation29,57,58,89-92 The review highlights MN compatibility with live, inactivated, subunit and DNA vaccines. Vaccines tested span a wide range of viral, bacterial and protozoan pathogens; including influenza,Citation29,46,54,55,59-77,84,91,92,95-100 hepatitis B,Citation47,49,50,86,101,102 Japanese encephalitis,Citation56 rabies,Citation57,58 human papillomavirus,Citation78,79 chikungunya virus,Citation80 West Nile virus,Citation80 rotavirus,Citation81 herpes simplex,Citation82 hepatitis C,Citation83 measles,Citation87,104 polio Citation88-90,94,105 and HIV,Citation103 bacterial illnesses including diphtheria,Citation45,46,48,97 anthrax,Citation53 tuberculosis,Citation85 plague Citation93 and tetanus Citation97 and protozoan illnesses including malaria,Citation51,52,97 as summarized in . This list includes 7 of the 13 vaccine-preventable diseases, as defined by the WHO.Citation119 This review highlights the paucity of clinical trial data, with only 11.29% of the 62 trials presented conducted in human subjects.

Table 2. Disease targets for microneedle-mediated vaccine delivery.

Influenza: A popular vaccine target

The influenza virus vaccine is as a popular vaccine target, being the pathogen of interest in 32 of the 62 research articles presented. Influenza is a highly contagious respiratory illness, with influenza A and B viruses causing annual seasonal epidemics and sporadic pandemics of disease, leading to hospitalizations and occasionally death.Citation121-125 In the US, it is estimated that influenza resulted in greater than half a million hospitalizations, 18,491-95,390 intensive care admissions and 4,915-27,174 deaths per year between 2010 and 2013.Citation126 Investment in the development of an influenza vaccine offers significant commercial and technical gain. Unlike other vaccines, which offer life-long immunity with a single dose, influenza immunity requires annual re-vaccination as a result of antigenic variation of the virus.Citation127 The target end-user of a microneedle patch-based influenza vaccine is the adult population and not the pediatric, thus reducing the barrier to clinical use. Vaccination is effective in preventing infection.Citation128 Furthermore, unlike many other vaccines, serological correlates of protection exist and the CHMP criteria are accepted to measure immunogenicity. However, coverage rates in target populations are far below the WHO-recommended 75%.Citation129-131 In addition, there are unmet needs associated with current influenza vaccines.Citation132 This motivates the development of alternate delivery systems such as MNs that may offer enhanced vaccine uptake and acceptance.Citation133 If a MN-based vaccine exhibited enhanced stability and lower vaccine doses could be used,Citation29,30 then this could be attractive to vaccine manufacturers. From a user perspective, the prospect of a painless, potentially self-administered vaccine may lead to improved vaccination coverage.Citation28 However all of these features still remain to be rigorously tested and developed in a clinical context.

Improving vaccine coverage in developing countries

Even though vaccination programs are frequently cited as one of the most low-cost, high-impact public health measures,Citation134 1.5 million children die every year as a result of vaccine preventable illnesses, including some of those presented in this review. Vaccines are temperature sensitive biological products, requiring refrigeration. In many developing world countries, a cold-chain infrastructure is almost prohibitively expensive thus preventing adequate vaccine distribution.Citation135,136 The thermostability of MN vaccines eliminates cold-chain requirements, thus reducing logistic costs and potentially improving distribution.Citation31-33 This thermostability would permit stock-piling in regular drug distribution networks, combatting the frequently encountered issue of supply shortage. In addition to being thermolabile, conventional vaccines often require administration by trained personnel. In LMIC countries, there are shortages of medical personnel at all levels of trainingCitation137: Africa has 2.3 healthcare workers per 1000 population, compared to 24.8 per 1000 in the Americas.Citation138 Therefore the previously discussed potential for self-administration with MN vaccines could further improve vaccine coverage in these countries, in tandem with other public health efforts. However, most pediatric vaccines in the Expanded Program of Immunization are adjuvanted. Pharmaceutical, immunological, safety and efficacy issues of incorporating licensed adjuvants into solid dosage formats of microneedles must be addressed before this technology will be licensed and deployed for these vaccines. Significant research and development effort is being focused in these areas to resolve these concerns.

Translation into clinical use

This review presents a variety of MN vaccines in the pre-clinical development stage, demonstrating safety and immunogenicity in animal models but also highlights the scarcity of clinical trial data. There is a progression from evaluation in small animal models such as mice, to higher animal models such as rhesus macaques, prior to transition to clinical development and evaluation in human subjects.Citation139 While preclinical research answers basic questions, it is not a surrogate for clinical research. It is hoped that the MN vaccines presented in this review, especially those that have undergone assessment in non-human primates, will progress through the developmental stages, ultimately leading to vaccine licensing and introduction into clinical use. An issue that needs to be assessed is the habituality of hypodermic needle-mediated vaccination. Despite the aforementioned disadvantages, traditional immunization has repeatedly demonstrated efficacy and safety. Familiarity breeds acceptance. Therefore a paradigm shift is required to drive the transition of MN-vaccines into clinical use. Increased end-user acceptability of MN-based vaccines will be required for widespread adoption. Positive attributes such as pain-free, bloodless administration must be rigorously tested and defined and acceptance of this technology by the end-user must be assessed, understood and the technology adapted to incorporate end-users' needs. MN fabrication considerations include scalability and dose loading capacity must also be addressed so that the vaccine manufacturer can assimilate the technology into their fill-finish systems. The majority of MN research has been conducted at laboratory scale in small quantities and the development of alternate fabrication approaches has begun to demonstrate scalability.Citation140,141 There is an inherent dose loading capacity associated with some MN technologies, whereby there is a limit to the amount of vaccine that can be coated on or incorporated in the MN.Citation1 The inclusion of adjuvants may reduce the vaccine dose required to elicit an appropriate immune response,Citation86,142 although their inclusion will also necessitate appropriate validation and production in GMP environments. Finally, there is a need for the development of universal acceptance criteria and Good Manufacturing Practice specifications, permitting MN characterization and subsequent commercialization.Citation118

This review presented the research pertaining to in vivo MN vaccines. Vaccines have been delivered via solid, coated, hollow and dissolving MNs. The dissolving MN offers a significant advantage over other MN platforms: the elimination of sharp, biohazardous waste after vaccination. MNs have the potential to improve vaccine access in developing countries. These vaccines have demonstrated safety and immunogenicity in pre-clinical models. The paucity of clinical data presented in this review highlights the need to incentivize vaccine research in human subjects. The technology possesses desirable attributes for the end-user including painless administration and potential for self-application, which may increase compliance and subsequent vaccine coverage, as well as benefits for the manufacturer including thermostability and dose-sparing capacity. All of these advantages demonstrate the high potential for microneedle technologies to have a positive impact on global immunization programs in the future.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

References

  • Kim Y-C, Park J-H, Prausnitz MR. Microneedles for drug and vaccine delivery. Adv Drug Deli Rev 2012; 64(14):1547-68; PMID:22575858; http://dx.doi.org/10.1016/j.addr.2012.04.005
  • Drucker E, Alcabes PG, Marx PA. The injection century: massive unsterile injections and the emergence of human pathogens. Lancet 2001; 358(9297):1989-92; PMID:11747942; http://dx.doi.org/10.1016/S0140-6736(01)06967-7
  • Kermode M. Unsafe injections in low-income country health settings: need for injection safety promotion to prevent the spread of blood-borne viruses. Health Promot Int 2004; 19(1):95-103; PMID:14976177; http://dx.doi.org/10.1093/heapro/dah110
  • Hauri AM, Armstrong GL, Hutin YJ. The global burden of disease attributable to contaminated injections given in health care settings. Int J STD AIDS 2004; 15(1):7-16; PMID:14769164; http://dx.doi.org/10.1258/095646204322637182
  • Nir Y, Paz A, Sabo E, Potasman I. Fear of injections in young adults: prevalence and associations. AmJ Trop Med Hyg 2003; 68(3):341-44; PMID:12685642
  • Hamilton JG. Needle phobia: a neglected diagnosis. J Fam Pract 1995; 41:169-75; PMID:7636457
  • Marks I. Blood-injury phobia: a review. Am J Psychiatry 1988; 145(10):1207-13; PMID:3048117; http://dx.doi.org/10.1176/ajp.145.10.1207
  • Kleinknecht RA. Acquisition of blood, injury, and needle fears and phobias. Behav Res Ther 1994; 32(8):817-23; PMID:7993325; http://dx.doi.org/10.1016/0005-7967(94)90161-9
  • Giudice EL, Campbell JD. Needle-free vaccine delivery. Adv Drug Deliv Rev 2006; 58(1): p. 68-89; PMID:16564111; http://dx.doi.org/10.1016/j.addr.2005.12.003
  • Wang L, Coppel RL. Oral vaccine delivery: can it protect against non-mucosal pathogens? Expert Rev Vaccines 2008; 7:729-38; PMID:18665772
  • Top FH, Buescher EL, Bancroft WH, Russell PK. Immunization with live types 7 and 4 adenovirus vaccines. II. Antibody response and protective effect against acute respiratory disease due to adenovirus type 7. J Infect Dis 1971; 124(2):155-60; PMID:4330998; http://dx.doi.org/10.1093/infdis/124.2.155
  • Bernstein DI. Rotarix: development of a live attenuated monovalent human rotavirus vaccine. Pediatr Ann 2006; 35(1):38; PMID:16466074; http://dx.doi.org/10.3928/0090-4481-20060101-12
  • Ciarlet M, Schödel F. Development of a rotavirus vaccine: clinical safety, immunogenicity, and efficacy of the pentavalent rotavirus vaccine, RotaTeq→. Vaccine 2009; 27:G72-81; PMID:20006144; http://dx.doi.org/10.1016/j.vaccine.2009.09.107
  • Germanier R, Fürer E. Characteristics of the attenuated oral vaccine strain“ S. typhi” Ty 21a. Dev Biol Stand 1982; 53:3-7; PMID:6873472
  • Liebowitz D, Lindbloom JD, Brandl JR, Garg SJ, Tucker SN. High titre neutralising antibodies to influenza after oral tablet immunisation: a phase 1, randomised, placebo-controlled trial. Lancet Infect Dis 2015; 15(9):1041-8; PMID:26333337; http://dx.doi.org/10.1016/S1473-3099(15)00266-2
  • Bora P, Kumar L, Bansal AK. Microneedle technology for advanced drug delivery: Evolving vistas. Review Article, Deaprtment of Pharmaceutical Technology, NIPER, CRIPS 2008; 9(1).
  • Lambert PH, Laurent PE. Intradermal vaccine delivery: will new delivery systems transform vaccine administration? Vaccine 2008; 26(26):3197-208; PMID:18486285; http://dx.doi.org/10.1016/j.vaccine.2008.03.095
  • Briggs DJ, Banzhoff A, Nicolay U, Sirikwin S, Dumavibhat B, Tongswas S, Wasi C. Antibody response of patients after postexposure rabies vaccination with small intradermal doses of purified chick embryo cell vaccine or purified Vero cell rabies vaccine. Bull World Health Organ 2000; 78(5):693-8; PMID:10859864
  • Hawkridge A, Hatherill M, Little F, Goetz MA, Barker L, Mahomed H, Sadoff J, Hanekom W, Geiter L, Hussey G, et al. Efficacy of percutaneous versus intradermal BCG in the prevention of tuberculosis in South African infants: randomised trial. Bmj 2008; 337:a2052; PMID:19008268; http://dx.doi.org/10.1136/bmj.a2052
  • Emerging WHODO, Surveillance OCD. WHO recommendations on rabies post-exposure treatment and the correct technique of intradermal immunization against rabies1997: World Health Organization, Division of Emerging and other Communicable Diseases Surveillance and Control.
  • Al Jarad N, Empey D, Duckworth G. Administration of the BCG vaccination using the multipuncture method in schoolchildren: a comparison with the intradermal method. Thorax 1999; 54(9):762-4; PMID:10456967; http://dx.doi.org/10.1136/thx.54.9.762
  • Ormerod L, Palmer C. Tuberculin reactivity after neonatal percutaneous BCG immunisation. Arch Dis Child 1993; 69(1):155; PMID:8024303; http://dx.doi.org/10.1136/adc.69.1.155
  • Donnelly RF, Singh TRR. Novel Delivery Systems for Transdermal and Intradermal Drug Delivery 2015: John Wiley & Sons.
  • Haq M, Smith E, John DN, Kalavala M, Edwards C, Anstey A, Morrissey A, Birchall JC. Clinical administration of microneedles: skin puncture, pain and sensation. Biomed Microdevices 2009; 11(1):35-47; PMID:18663579; http://dx.doi.org/10.1007/s10544-008-9208-1
  • Birchall JC, Clemo R, Anstey A, John DN. Microneedles in clinical practice–an exploratory study into the opinions of healthcare professionals and the public. Pharm Res 2011; 28(1):95-106; PMID:20238152; http://dx.doi.org/10.1007/s11095-010-0101-2
  • Escobar‐Chávez JJ, Bonilla-Martínez D, Villegas-González MA, Molina-Trinidad E, Casas-Alancaster N, Revilla-Vázquez AL. Microneedles: a valuable physical enhancer to increase transdermal drug delivery. J Clin Pharmacol 2011; 51(7):964-977; PMID:21148047; http://dx.doi.org/10.1177/0091270010378859
  • Birchall JC. Microneedle array technology: the time is right but is the science ready? Expert Rev Med Devices 2006; 3(1):1-4; PMID:16359245; http://dx.doi.org/10.1586/17434440.3.1.1
  • Norman JJ, Prausnitz MR. Improving patient acceptance of insulin therapy by improving needle design. J Diabetes Sci Technol 2012; 6(2):336-8; PMID:22538143; http://dx.doi.org/10.1177/193229681200600217
  • Van Damme P, Oosterhuis-Kafeja F, Van der Wielen M, Almagor Y, Sharon O, Levin Y. Safety and efficacy of a novel microneedle device for dose sparing intradermal influenza vaccination in healthy adults. Vaccine 2009; 27(3):454-9; PMID:19022318; http://dx.doi.org/10.1016/j.vaccine.2008.10.077
  • Al-Zahrani S, Zaric M, McCrudden C, Scott C, Kissenpfennig A, Donnelly RF. Microneedle-mediated vaccine delivery: harnessing cutaneous immunobiology to improve efficacy. Expert Opin Drug Deliv 2012; 9(5):541-50; PMID:22475249; http://dx.doi.org/10.1517/17425247.2012.676038
  • Mistilis MJ, Bommarius AS, Prausnitz MR. Development of a Thermostable Microneedle Patch for Influenza Vaccination. J Pharm Sci 2015; 104(2):740-9; PMID:25448542; http://dx.doi.org/10.1002/jps.24283
  • Choi H-J, Yoo DG, Bondy BJ, Quan FS, Compans RW, Kang SM, Prausnitz MR. Stability of influenza vaccine coated onto microneedles. Biomaterials 2012; 33(14):3756-69; PMID:22361098; http://dx.doi.org/10.1016/j.biomaterials.2012.01.054
  • Chen X, Fernando GJ, Crichton ML, Flaim C, Yukiko SR, Fairmaid EJ, Corbett HJ, Primiero CA, Ansaldo AB, Frazer IH. Improving the reach of vaccines to low-resource regions, with a needle-free vaccine delivery device and long-term thermostabilization. J Control Release 2011; 152(3):349-55; PMID:21371510; http://dx.doi.org/10.1016/j.jconrel.2011.02.026
  • Jepps OG, Dancik Y, Anissimov YG, Roberts MS. Modeling the human skin barrier—Towards a better understanding of dermal absorption. Adv Drug Deliv Rev 2013; 65(2):152-68; PMID:22525516; http://dx.doi.org/10.1016/j.addr.2012.04.003
  • Skountzou I, Quan FS, Jacob J, Compans RW, Kang SM. Transcutaneous immunization with inactivated influenza virus induces protective immune responses. Vaccine 2006; 24(35):6110-19; PMID:16766095; http://dx.doi.org/10.1016/j.vaccine.2006.05.014
  • Salmon J, Armstrong C, Ansel J. The skin as an immune organ. West J Med 1994; 160(2):146; PMID:8160465
  • Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature 1998; 392(6673):245-252; PMID:9521319; http://dx.doi.org/10.1038/32588
  • Steinman RM. Dendritic cells and the control of immunity: enhancing the efficiency of antigen presentation. Mt Sinai J Med 2001; 68(3):160-166; PMID:11373688
  • Schroder JM, Reich K, Kabashima K, Liu FT, Romani N, Metz M, Kerstan A, Lee PH, Loser K, Schon MP, Maurer M, Stoitzner P, Beissert S, Tokura Y, Gallo RL. Who is really in control of skin immunity under physiological circumstances - lymphocytes, dendritic cells or keratinocytes? Exp Dermatol 2006; 15(11):913–6.
  • Lee HK, Iwasaki A. Innate control of adaptive immunity: dendritic cells and beyond. in Seminars in immunology 2007: Elsevier.
  • Zachariae CO, Thestrup-Pedersen K, Matsushima K. Expression and secretion of leukocyte chemotactic cytokines by normal human melanocytes and melanoma cells. J Invest Dermatol 1991; 97(3):593-9; PMID:1875058; http://dx.doi.org/10.1111/1523-1747.ep12481934
  • Urb M, Sheppard DC. The role of mast cells in the defence against pathogens. PLoS Pathog 2012; 8(4):e1002619; PMID:22577358; http://dx.doi.org/10.1371/journal.ppat.1002619
  • Abraham SN, John ALS. Mast cell-orchestrated immunity to pathogens. Nat Rev Immunol 2010; 10(6):440-52; PMID:20498670; http://dx.doi.org/10.1038/nri2782
  • Koutsonanos DG, Esser ES, McMaster SR, Kalluri P, Lee JW, Prausnitz MR, Skountzou I, Denning TL, Kohlmeier JE, Compans RW. Enhanced immune responses by skin vaccination with influenza subunit vaccine in young hosts. Vaccine 2015; 33:4675-82; PMID:25744228
  • Bal SM, Ding Z, Kersten GF, Jiskoot W, Bouwstra JA. Microneedle-based transcutaneous immunisation in mice with N-trimethyl chitosan adjuvanted diphtheria toxoid formulations. Pharm Res 2010; 27(9):1837-47; PMID:20559701; http://dx.doi.org/10.1007/s11095-010-0182-y
  • Ding Z, Verbaan FJ, Bivas-Benita M, Bungener L, Huckriede A, van den Berg DJ, Kersten G, Bouwstra JA. Microneedle arrays for the transcutaneous immunization of diphtheria and influenza in BALB/c mice. J Control Release 2009; 136(1):71-78; PMID:19331846; http://dx.doi.org/10.1016/j.jconrel.2009.01.025
  • Guo L, Qiu Y, Chen J, Zhang S, Xu B, Gao Y. Effective transcutaneous immunization against hepatitis B virus by a combined approach of hydrogel patch formulation and microneedle arrays. Biomed Microdevices 2013; 15(6):1077-85; PMID:23893014; http://dx.doi.org/10.1007/s10544-013-9799-z
  • Ding Z, Van Riet E, Romeijn S, Kersten GF, Jiskoot W, Bouwstra JA. Immune modulation by adjuvants combined with diphtheria toxoid administered topically in BALB/c mice after microneedle array pretreatment. Pharm Res 2009; 26(7):1635-43; PMID:19326190; http://dx.doi.org/10.1007/s11095-009-9874-6
  • Hirschberg H, van Kuijk S, Loch J, Jiskoot W, Bouwstra J, Kersten G, Amorij JP. A combined approach of vesicle formulations and microneedle arrays for transcutaneous immunization against hepatitis B virus. Eur J Pharm Sci 2012; 46(1):1-7; PMID:22330147; http://dx.doi.org/10.1016/j.ejps.2012.01.013
  • Yin D, Liang W, Xing S, Gao Z, Zhang W, Guo Z, Gao S. Hepatitis B DNA vaccine-polycation nano-complexes enhancing immune response by percutaneous administration with microneedle. Biol Pharm Bull 2013; 36(8):1283-91; PMID:23676787; http://dx.doi.org/10.1248/bpb.b13-00050
  • Carey JB, Vrdoljak A, O'Mahony C, Hill AV, Draper SJ, Moore AC. Microneedle-mediated immunization of an adenovirus-based malaria vaccine enhances antigen-specific antibody immunity and reduces anti-vector responses compared to the intradermal route. Sci Rep 2014; 4:6154; PMID:25142082; http://dx.doi.org/10.1038/srep06154
  • Pearson FE, O'Mahony C, Moore AC, Hill AV. Induction of CD8+ T cell responses and protective efficacy following microneedle-mediated delivery of a live adenovirus-vectored malaria vaccine. Vaccine 2015; 33:3248-55 PMID:2583910
  • Mikszta JA, Sullivan VJ, Dean C, Waterston AM, Alarcon JB, Dekker JP 3rd, Brittingham JM, Huang J, Hwang CR, Ferriter M. Protective immunization against inhalational anthrax: a comparison of minimally invasive delivery platforms. J Infect Dis 2005; 191(2):278-88; PMID:15609239; http://dx.doi.org/10.1086/426865
  • Kim Y-C, Quan FS, Compans RW, Kang SM, Prausnitz MR. Formulation and coating of microneedles with inactivated influenza virus to improve vaccine stability and immunogenicity. J Control Release 2010; 142(2):187-95; PMID:19840825; http://dx.doi.org/10.1016/j.jconrel.2009.10.013
  • Kim Y-C, Quan FS, Compans RW, Kang SM, Prausnitz MR. Formulation of microneedles coated with influenza virus-like particle vaccine. AAPS PharmSciTech 2010; 11(3):1193-201; PMID:20676947; http://dx.doi.org/10.1208/s12249-010-9471-3
  • Dean CH, Alarcon JB, Waterston AM, Draper K, Early R, Guirakhoo F, Monath TP, Mikszta JA. Cutaneous delivery of a live, attenuated chimeric flavivirus vaccines against Japanese encephalitis (ChimeriVaxTM-JE) in non-human primates. Hum Vaccin 2005; 1(3):106-11; PMID:17012854; http://dx.doi.org/10.4161/hv.1.3.1797
  • Laurent PE, Bonnet S, Alchas P, Regolini P, Mikszta JA, Pettis R, Harvey NG. Evaluation of the clinical performance of a new intradermal vaccine administration technique and associated delivery system. Vaccine 2007; 25(52):8833-42; PMID:18023942; http://dx.doi.org/10.1016/j.vaccine.2007.10.020
  • Laurent PE, Bourhy H, Fantino M, Alchas P, Mikszta JA. Safety and efficacy of novel dermal and epidermal microneedle delivery systems for rabies vaccination in healthy adults. Vaccine 2010; 28(36):5850-6; PMID:20600481; http://dx.doi.org/10.1016/j.vaccine.2010.06.062
  • Kim Y-C, Quan FS, Yoo DG, Compans RW, Kang SM, Prausnitz MR. Improved influenza vaccination in the skin using vaccine coated microneedles. Vaccine 2009; 27(49):6932-8; PMID:19761836; http://dx.doi.org/10.1016/j.vaccine.2009.08.108
  • Kim Y-C, Quan FS, Yoo DG, Compans RW, Kang SM, Prausnitz MR. Enhanced memory responses to seasonal H1N1 influenza vaccination of the skin with the use of vaccine-coated microneedles. J Infect Dis 2010; 201(2):190-8; PMID:20017632; http://dx.doi.org/10.1086/649228
  • Kim Y-C, Yoo DG, Compans RW, Kang SM, Prausnitz MR. Cross-protection by co-immunization with influenza hemagglutinin DNA and inactivated virus vaccine using coated microneedles. J Control Release 2013; 172(2):579-88; PMID:23643528; http://dx.doi.org/10.1016/j.jconrel.2013.04.016
  • Koutsonanos DG, del Pilar Martin M, Zarnitsyn VG, Sullivan SP, Compans RW, Prausnitz MR, Skountzou I. Transdermal influenza immunization with vaccine-coated microneedle arrays. PloS one 2009; 4(3):e4773; PMID:19274084; http://dx.doi.org/10.1371/journal.pone.0004773
  • Zhu Q, Zarnitsyn VG, Ye L, Wen Z, Gao Y, Pan L, Skountzou I, Gill HS, Prausnitz MR, Yang C, et al. Immunization by vaccine-coated microneedle arrays protects against lethal influenza virus challenge. Proc Natl Acad Sci U S A 2009; 106(19):7968-73; PMID:19416832; http://dx.doi.org/10.1073/pnas.0812652106
  • Weldon WC, Martin MP, Zarnitsyn V, Wang B, Koutsonanos D, Skountzou I, Prausnitz MR, Compans RW. Microneedle vaccination with stabilized recombinant influenza virus hemagglutinin induces improved protective immunity. Clin Vaccine Immunol 2011; 18(4):647-54; PMID:21288996; http://dx.doi.org/10.1128/CVI.00435-10
  • Wang B-Z, Gill HS, He C, Ou C, Wang L, Wang YC, Feng H, Zhang H, Prausnitz MR, Compans RW. Microneedle delivery of an M2e-TLR5 ligand fusion protein to skin confers broadly cross-protective influenza immunity. J Control Release 2014; 178:1-7; PMID:24417966; http://dx.doi.org/10.1016/j.jconrel.2014.01.002
  • Song J-M, Kim YC, Lipatov AS, Pearton M, Davis CT, Yoo DG, Park KM, Chen LM, Quan FS, Birchall JC, et al. Microneedle delivery of H5N1 influenza virus-like particles to the skin induces long-lasting B-and T-cell responses in mice. Clin Vaccine Immunol 2010; 17(9):1381-9; PMID:20631330; http://dx.doi.org/10.1128/CVI.00100-10
  • Song J-M, Kim YC, O E, Compans RW, Prausnitz MR, Kang SM. DNA vaccination in the skin using microneedles improves protection against influenza. Mol Ther 2012; 20(7):1472-80; PMID:22508490; http://dx.doi.org/10.1038/mt.2012.69
  • Song J-M, Kim YC, Barlow PG, Hossain MJ, Park KM, Donis RO, Prausnitz MR, Compans RW, Kang SM. Improved protection against avian influenza H5N1 virus by a single vaccination with virus-like particles in skin using microneedles. Antiviral Res 2010; 88(2):244-7; PMID:20851715; http://dx.doi.org/10.1016/j.antiviral.2010.09.001
  • Shin J-H, Park JK, Lee DH, Quan FS, Song CS, Kim YC. Microneedle Vaccination Elicits Superior Protection and Antibody Response over Intranasal Vaccination against Swine-Origin Influenza A (H1N1) in Mice. PloS one 2015; 10(6):e0130684; PMID:26086590; http://dx.doi.org/10.1371/journal.pone.0130684
  • Quan F-S, Kim YC, Yoo DG, Compans RW, Prausnitz MR, Kang SM. Stabilization of influenza vaccine enhances protection by microneedle delivery in the mouse skin. PloS one 2009; 4(9):e7152; PMID:19779615; http://dx.doi.org/10.1371/journal.pone.0007152
  • Quan F-S, Kim YC, Vunnava A, Yoo DG, Song JM, Prausnitz MR, Compans RW, Kang SM. Intradermal vaccination with influenza virus-like particles by using microneedles induces protection superior to that with intramuscular immunization. J Virol 2010; 84(15):7760-9; PMID:20484519; http://dx.doi.org/10.1128/JVI.01849-09
  • Quan F-S, Kim YC, Song JM, Hwang HS, Compans RW, Prausnitz MR, Kang SM. Long-term protective immunity from an influenza virus-like particle vaccine administered with a microneedle patch. Clin Vaccine Immunol 2013; 20(9):1433-9; PMID:23863506; http://dx.doi.org/10.1128/CVI.00251-13
  • Quan F-S, Kim YC, Compans RW, Prausnitz MR, Kang SM. Dose sparing enabled by skin immunization with influenza virus-like particle vaccine using microneedles. J Control Release 2010; 147(3):326-32; PMID:20692307; http://dx.doi.org/10.1016/j.jconrel.2010.07.125
  • Koutsonanos DG, Vassilieva EV, Stavropoulou A, Zarnitsyn VG, Esser ES, Taherbhai MT, Prausnitz MR, Compans RW, Skountzou I. Delivery of subunit influenza vaccine to skin with microneedles improves immunogenicity and long-lived protection. Sci Rep 2012; 2:357;PMID:22500210; http://dx.doi.org/10.1038/srep00357
  • Kim Y-C, Song JM, Lipatov AS, Choi SO, Lee JW, Donis RO, Compans RW, Kang SM, Prausnitz MR. Increased immunogenicity of avian influenza DNA vaccine delivered to the skin using a microneedle patch. Eur J Pharm Biopharm 2012; 81(2):239-47; PMID:22504442; http://dx.doi.org/10.1016/j.ejpb.2012.03.010
  • Kim MC, Lee JW, Choi HJ, Lee YN, Hwang HS, Lee J, Kim C, Lee JS, Montemango C, Prausnitz MR, Kang SM. Microneedle patch delivery to the skin of virus-like particles containing heterologous M2e extracellular domains of influenza virus induces broad heterosubtypic cross-protection. J Control Release 2015; 210:208–16.
  • del Pilar Martin M, Weldon WC, Zarnitsyn VG, Koutsonanos DG, Akbari H, Skountzou I, Jacob J, Prausnitz MR, Compans RW. Local response to microneedle-based influenza immunization in the skin. MBio 2012; 3(2):e00012-12; PMID:22396479; http://dx.doi.org/10.1128/mBio.00012-12
  • Corbett HJ, Fernando GJ, Chen X, Frazer IH, Kendall MA. Skin vaccination against cervical cancer associated human papillomavirus with a novel micro-projection array in a mouse model. PLoS One 2010; 5(10):e13460; PMID:20976136; http://dx.doi.org/10.1371/journal.pone.0013460
  • Kines RC, Zarnitsyn V, Johnson TR, Pang YY, Corbett KS, Nicewonger JD, Gangopadhyay A, Chen M, Liu J, Prausnitz MR. Vaccination with Human Papillomavirus Pseudovirus-Encapsidated Plasmids Targeted to Skin Using Microneedles. PloS one 2015; 10(3):e0120797; PMID:25785935; http://dx.doi.org/10.1371/journal.pone.0120797
  • Prow TW, Chen X, Prow NA, Fernando GJ, Tan CS, Raphael AP, Chang D, Ruutu MP, Jenkins DW, Pyke A, et al. Nanopatch‐Targeted Skin Vaccination against West Nile Virus and Chikungunya Virus in Mice. Small 2010; 6(16):1776-84; PMID:20665754; http://dx.doi.org/10.1002/smll.201000331
  • Moon S, Wang Y, Edens C, Gentsch JR, Prausnitz MR, Jiang B. Dose sparing and enhanced immunogenicity of inactivated rotavirus vaccine administered by skin vaccination using a microneedle patch. Vaccine 2013; 31(34):3396-402; PMID:23174199; http://dx.doi.org/10.1016/j.vaccine.2012.11.027
  • Chen X, Kask AS, Crichton ML, McNeilly C, Yukiko S, Dong L, Marshak JO, Jarrahian C, Fernando GJ, Chen D, et al. Improved DNA vaccination by skin-targeted delivery using dry-coated densely-packed microprojection arrays. J Control Release 2010; 148(3):327-33; PMID:20850487; http://dx.doi.org/10.1016/j.jconrel.2010.09.001
  • Gill HS, Söderholm J, Prausnitz MR, Sällberg M. Cutaneous vaccination using microneedles coated with hepatitis C DNA vaccine. Gene therapy 2010; 17(6):811-4; PMID:20200562; http://dx.doi.org/10.1038/gt.2010.22
  • Kommareddy S, Baudner BC, Bonificio A, Gallorini S, Palladino G, Determan AS, Dohmeier DM, Kroells KD, Sternjohn JR, Singh M, et al. Influenza subunit vaccine coated microneedle patches elicit comparable immune responses to intramuscular injection in guinea pigs. Vaccine 2013; 31(34):3435-41; PMID:23398932; http://dx.doi.org/10.1016/j.vaccine.2013.01.050
  • Hiraishi Y, Nandakumar S, Choi SO, Lee JW, Kim YC, Posey JE, Sable SB, Prausnitz MR. Bacillus Calmette-Guerin vaccination using a microneedle patch. Vaccine 2011; 29(14):2626-36; PMID:21277407; http://dx.doi.org/10.1016/j.vaccine.2011.01.042
  • Andrianov AK, DeCollibus DP, Gillis HA, Kha HH, Marin A, Prausnitz MR, Babiuk LA, Townsend H, Mutwiri G. Poly [di (carboxylatophenoxy) phosphazene] is a potent adjuvant for intradermal immunization. Proc Natl Acad Sci U S A 2009; 106(45):18936-41; PMID:19864632; http://dx.doi.org/10.1073/pnas.0908842106
  • Edens C, Collins ML, Ayers J, Rota PA, Prausnitz MR. Measles vaccination using a microneedle patch. Vaccine 2013; 31(34):3403-9; PMID:23044406; http://dx.doi.org/10.1016/j.vaccine.2012.09.062
  • van der Maaden K, Sekerdag E, Schipper P, Kersten G, Jiskoot W, Bouwstra J. Layer-by-layer assembly of inactivated poliovirus and N-trimethyl chitosan on pH-sensitive microneedles for dermal vaccination. Langmuir 2015; 31(31):8654-60; PMID:26145437; http://dx.doi.org/10.1021/acs.langmuir.5b01262
  • Troy SB, Kouiavskaia D, Siik J, Kochba E, Beydoun H, Mirochnitchenko O, Levin Y, Khardori N, Chumakov K, Maldonado Y. Comparison of the Immunogenicity of Various Booster Doses of Inactivated Polio Vaccine Delivered Intradermally Versus Intramuscularly to HIV-Infected Adults. J Infect Dis 2015; 211(12):1969-76; PMID:25567841; http://dx.doi.org/10.1093/infdis/jiu841
  • Anand A, Zaman K, Estívariz CF, Yunus M, Gary HE, Weldon WC, Bari TI, Steven Oberste M, Wassilak SG, Luby SP. Early priming with inactivated poliovirus vaccine (IPV) and intradermal fractional dose IPV administered by a microneedle device: A randomized controlled trial. Vaccine 2015; 33:6816-22; PMID:26476367
  • Atmar RL, Patel SM, Keitel WA. Intanza(®): A new intradermal vaccine for seasonal influenza. Expert Rev Vaccines 2010; 9(12):1399–409.
  • Leroux-Roels I, Weber F. Intanza→ 9 µg intradermal seasonal influenza vaccine for adults 18 to 59 years of age. Hum Vaccin & immunotherapeutics 2013; 9(1):115-21; PMID:23442585; http://dx.doi.org/10.4161/hv.22342
  • Huang J, D'Souza AJ, Alarcon JB, Mikszta JA, Ford BM, Ferriter MS, Evans M, Stewart T, Amemiya K, Ulrich RG, et al. Protective immunity in mice achieved with dry powder formulation and alternative delivery of plague F1-V vaccine. Clin Vaccine Immunol 2009; 16(5):719-25; PMID:19261773; http://dx.doi.org/10.1128/CVI.00447-08
  • van der Maaden K, Trietsch SJ, Kraan H, Varypataki EM, Romeijn S, Zwier R, van der Linden HJ, Kersten G, Hankemeier T, Jiskoot W, et al. Novel hollow microneedle technology for depth-controlled microinjection-mediated dermal vaccination: a study with polio vaccine in rats. Pharm Res 2014; 31(7):1846-154; PMID:24469907
  • Hirobe S, Azukizawa H, Hanafusa T, Matsuo K, Quan YS, Kamiyama F, Katayama I, Okada N, Nakagawa S. Clinical study and stability assessment of a novel transcutaneous influenza vaccination using a dissolving microneedle patch. Biomaterials 2015; 57:50-8; PMID:25913250; http://dx.doi.org/10.1016/j.biomaterials.2015.04.007
  • Kommareddy S, Baudner BC, Oh S, Kwon SY, Singh M, O'Hagan DT. Dissolvable microneedle patches for the delivery of cell‐culture‐derived influenza vaccine antigens. J Pharm Sci 2012; 101(3):1021-7; PMID:22190403; http://dx.doi.org/10.1002/jps.23019
  • Matsuo K, Hirobe S, Yokota Y, Ayabe Y, Seto M, Quan YS, Kamiyama F, Tougan T, Horii T, Mukai Y, et al. Transcutaneous immunization using a dissolving microneedle array protects against tetanus, diphtheria, malaria, and influenza. J Control Release 2012; 160(3):495-501; PMID:22516091; http://dx.doi.org/10.1016/j.jconrel.2012.04.001
  • Sullivan SP, Koutsonanos DG, Del Pilar Martin M, Lee JW, Zarnitsyn V, Choi SO, Murthy N, Compans RW, Skountzou I, Prausnitz MR. Dissolving polymer microneedle patches for influenza vaccination. Nature medicine 2010; 16(8):915-20; PMID:20639891; http://dx.doi.org/10.1038/nm.2182
  • Vassilieva EV, Kalluri H, McAllister D, Taherbhai MT, Esser ES, Pewin WP, Pulit-Penaloza JA, Prausnitz MR, Compans RW, Skountzou I. Improved immunogenicity of individual influenza vaccine components delivered with a novel dissolving microneedle patch stable at room temperature. Drug Deliv Transl Res 2015; 5(4):360–71.
  • Wang J, Li B, Wu MX. Effective and lesion-free cutaneous influenza vaccination. Proc Natl Acad Sci U S A 2015; 112(16):5005-10; PMID:25848020; http://dx.doi.org/10.1073/pnas.1500408112
  • Qiu Y, Guo L, Zhang S, Xu B, Gao Y, Hu Y, Hou J, Bai B, Shen H, Mao P. DNA-based vaccination against hepatitis B virus using dissolving microneedle arrays adjuvanted by cationic liposomes and CpG ODN. Drug Deliv 2015:1–8 [Epub ahead of print]
  • Wang T, Zhen Y, Ma X, Wei B, Li S, Wang N. Mannosylated and lipid A-incorporating cationic liposomes constituting microneedle arrays as an effective oral mucosal HBV vaccine applicable in the controlled temperature chain. Colloids Surf Biointerfaces 2015; 126:520-30; PMID:25612819; http://dx.doi.org/10.1016/j.colsurfb.2015.01.005
  • Pattani A, McKay PF, Garland MJ, Curran RM, Migalska K, Cassidy CM, Malcolm RK, Shattock RJ, McCarthy HO, Donnelly RF. Microneedle mediated intradermal delivery of adjuvanted recombinant HIV-1 CN54gp140 effectively primes mucosal boost inoculations. J Control Release 2012; 162(3):529–37.
  • Edens C, Collins ML, Goodson JL, Rota PA, Prausnitz MR. A microneedle patch containing measles vaccine is immunogenic in non-human primates. Vaccine 2015; 33:4712-8; PMID:25770786
  • Edens C, Dybdahl-Sissoko NC, Weldon WC, Oberste MS, Prausnitz MR. Inactivated polio vaccination using a microneedle patch is immunogenic in the rhesus macaque. Vaccine 2015; 33:4683-90; PMID:25749246
  • Gill HS, Prausnitz MR. Coated microneedles for transdermal delivery. J Control Release 2007; 117(2):227-37; PMID:17169459; http://dx.doi.org/10.1016/j.jconrel.2006.10.017
  • Vrdoljak A, McGrath MG, Carey JB, Draper SJ, Hill AV, O'Mahony C, Crean AM, Moore AC. Coated microneedle arrays for transcutaneous delivery of live virus vaccines. J Control Release 2012; 159(1):34-42; PMID:22245683; http://dx.doi.org/10.1016/j.jconrel.2011.12.026
  • Wonglertnirant N, Todo H, Opanasopit P, Ngawhirunpat T, Sugibayashi K. Macromolecular delivery into skin using a hollow microneedle. Biol Pharm Bull 2010; 33(12):1988-93; PMID:21139238; http://dx.doi.org/10.1248/bpb.33.1988
  • Davis SP, Martanto W, Allen MG, Prausnitz MR. Hollow metal microneedles for insulin delivery to diabetic rats. IEEE Trans Biomed Eng 2005; 52(5):909–15.
  • Harvey AJ, Kaestner SA, Sutter DE, Harvey NG, Mikszta JA, Pettis RJ. Microneedle-based intradermal delivery enables rapid lymphatic uptake and distribution of protein drugs. Pharm Res 2011; 28(1):107-16; PMID:20354765; http://dx.doi.org/10.1007/s11095-010-0123-9
  • Sullivan SP, Murthy N, Prausnitz MR. Minimally invasive protein delivery with rapidly dissolving polymer microneedles. Adv Mater 2008; 20(5):933-8; PMID:23239904; http://dx.doi.org/10.1002/adma.200701205
  • Lee JW, Park J-H, Prausnitz MR. Dissolving microneedles for transdermal drug delivery. Biomaterials 2008; 29(13):2113-24; PMID:18261792; http://dx.doi.org/10.1016/j.biomaterials.2007.12.048
  • Prausnitz MR, Langer R. Transdermal drug delivery. Nat Biotechnol 2008; 26(11): p. 1261-1268; PMID:18997767; http://dx.doi.org/10.1038/nbt.1504
  • Lahiji SF, Dangol M, Jung H. A patchless dissolving microneedle delivery system enabling rapid and efficient transdermal drug delivery. Sci Rep2015; 5:7914; PMID:25604728; http://dx.doi.org/10.1038/srep07914
  • Donnelly RF, Singh TRR, Woolfson AD. Microneedle-based drug delivery systems: microfabrication, drug delivery, and safety. Drug Deliv 2010; 17(4):187-207; PMID:20297904; http://dx.doi.org/10.3109/10717541003667798
  • McGrath MG, Vucen S, Vrdoljak A, Kelly A, O'Mahony C, Crean AM, Moore A. Production of dissolvable microneedles using an atomised spray process: Effect of microneedle composition on skin penetration. Eur J Pharm Biopharm 2014; 86(2):200-11; PMID:23727511; http://dx.doi.org/10.1016/j.ejpb.2013.04.023
  • Chu LY, Choi SO, Prausnitz MR. Fabrication of dissolving polymer microneedles for controlled drug encapsulation and delivery: bubble and pedestal microneedle designs. J Pharm Sci 2010; 99(10):4228-38; PMID:20737630; http://dx.doi.org/10.1002/jps.22140
  • Lutton RE, et al. Microneedle characterisation: the need for universal acceptance criteria and GMP specifications when moving towards commercialisation. Drug Deliv Transl Res 2015: p. 1-19.
  • WHO. Vaccine-preventable diseases 2015 [cited 2015 24th November]; Available from: http://apps.who.int/immunization_monitoring/diseases/en/.
  • Mikszta JA, Dekker JP 3rd, Harvey NG, Dean CH, Brittingham JM, Huang J, Sullivan VJ, Dyas B, Roy CJ, Ulrich RG. Microneedle-based intradermal delivery of the anthrax recombinant protective antigen vaccine. Infect Immun 2006; 74(12):6806-10; PMID:17030580; http://dx.doi.org/10.1128/IAI.01210-06
  • Jansen AG, Sanders EA, Hoes AW, van Loon AM, Hak E. Influenza-and respiratory syncytial virus-associated mortality and hospitalisations. Eur Respir J 2007; 30(6):1158-66; PMID:17715167; http://dx.doi.org/10.1183/09031936.00034407
  • Rivetti D, Jefferson T, Thomas R, Rudin M, Rivetti A, Di Pietrantonj C, Demicheli V. Vaccines for preventing influenza in the elderly. Cochrane Database Syst Rev 2006; (3):CD004876.
  • Thompson WW, Shay DK, Weintraub E, Brammer L, Bridges CB, Cox NJ, Fukuda K. Influenza-associated hospitalizations in the United States. Jama 2004; 292(11):1333-40; PMID:15367555; http://dx.doi.org/10.1001/jama.292.11.1333
  • Zhou H, Thompson WW, Viboud CG, Ringholz CM, Cheng PY, Steiner C, Abedi GR, Anderson LJ, Brammer L, Shay DK. Hospitalizations associated with influenza and respiratory syncytial virus in the United States, 1993–2008. Clin Infect Dis 2012; 54(10):1427-36; PMID:22495079; http://dx.doi.org/10.1093/cid/cis211
  • Chaves SS, Lynfield R, Lindegren ML, Bresee J, Finelli L. The US influenza hospitalization surveillance network. Emerg Infect Dis 2015; 9: 1543-50; PMID:26291121; http://dx.doi.org/10.3201/eid2109.141912
  • Reed C, Chaves SS, Daily Kirley P, Emerson R, Aragon D, Hancock EB, Butler L, Baumbach J, Hollick G, Bennett NM, et al. Estimating Influenza Disease Burden from Population-Based Surveillance Data in the United States. PloS one 2015; 10(3):e0118369; PMID:25738736; http://dx.doi.org/10.1371/journal.pone.0118369
  • Treanor J. Influenza vaccine—outmaneuvering antigenic shift and drift. N Engl J Med 2004; 350(3):218-20; PMID:14724300; http://dx.doi.org/10.1056/NEJMp038238
  • Demicheli V, Rivetti D, Deeks JJ, Jefferson TO. Vaccines for preventing influenza in healthy adults. Cochrane Database Syst Rev 2004; (3):CD001269.
  • Executive Board, Prevention and control of influenza pandemics and annual epidemics 2003.
  • Blank PR, Schwenkglenks M, Szucs TD. Vaccination coverage rates in eleven European countries during two consecutive influenza seasons. J Infect 2009; 58(6):446-58; PMID:19446340; http://dx.doi.org/10.1016/j.jinf.2009.04.001
  • CDC: Flu vaccination coverage, United States, 2014 -15 influenza season. 2015. Available from: http://www.cdc.gov/flu/fluvaxview/coverage-1415estimates.htm
  • Noh JY, Kim WJ. Influenza vaccines: unmet needs and recent developments. Infect Chemother 2013; 45(4):375-86; PMID:24475351; http://dx.doi.org/10.3947/ic.2013.45.4.375
  • Norman JJ, Arya JM, McClain MA, Frew PM, Meltzer MI, Prausnitz MR. Microneedle patches: usability and acceptability for self-vaccination against influenza. Vaccine 2014; 32(16):1856-62; PMID:24530146; http://dx.doi.org/10.1016/j.vaccine.2014.01.076
  • Miller MA, Sentz JT. Chapter 12 Vaccine-Preventable Diseases. In Disease and mortality in sub-Saharan Africa. 2nd edition. Washington, DC: The International Bank for Reconstruction and Development/World Bank; 2006.
  • Ren Q, Xiong H, Li Y, Xu R, Zhu C. Evaluation of an outside-the-cold-chain vaccine delivery strategy in remote regions of western China. Public Health Rep 2009; 124(5):745; PMID:19753953
  • Wang L, Li J, Chen H, Li F, Armstrong GL, Nelson C, Ze W, Shapiro CN. Hepatitis B vaccination of newborn infants in rural China: evaluation of a village-based, out-of-cold-chain delivery strategy. Bull World Health Organ 2007; 85(9):688-94; PMID:18026625; http://dx.doi.org/10.2471/BLT.06.037002
  • Dowling JM, Yap CF. Happiness and poverty in developing countries: A global perspective 2012: Palgrave Macmillan.
  • Naicker S, Plange-Rhule J, Tutt RC, Eastwood JB. Shortage of healthcare workers in developing countries–Africa. Ethn Dis 2009; 19(1 Suppl 1):S1-60-4.
  • Wolfe DN, Florence W, Bryant P. Current biodefense vaccine programs and challenges. Hum Vaccin Immunother 2013; 9(7):1591-7; PMID:23428906; http://dx.doi.org/10.4161/hv.24063
  • Moga KA, Bickford LR, Geil RD, Dunn SS, Pandya AA, Wang Y, Fain JH, Archuleta CF, O'Neill AT, Desimone JM. Rapidly–dissolvable microneedle patches via a highly scalable and reproducible soft lithography approach. Adv Mater 2013; 25(36):5060-6; PMID:23893866; http://dx.doi.org/10.1002/adma.201300526
  • Lutton RE, Larrañeta E, Kearney MC, Boyd P, Woolfson AD, Donnelly RF. A novel scalable manufacturing process for the production of hydrogel-forming microneedle arrays. Int J Pharm 2015; 494(1):417-29; PMID:26302858; http://dx.doi.org/10.1016/j.ijpharm.2015.08.049
  • Weldon WC, Zarnitsyn VG, Esser ES, Taherbhai MT, Koutsonanos DG, Vassilieva EV, Skountzou I, Prausnitz MR, Compans RW. Effect of adjuvants on responses to skin immunization by microneedles coated with influenza subunit vaccine. PloS one 2012; 7(7):e41501; PMID:22848514; http://dx.doi.org/10.1371/journal.pone.0041501

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.