Next Article in Journal
Questionnaire Survey of Possible Association of Allergic Diseases with Adverse Reactions to SARS-CoV-2 Vaccination
Next Article in Special Issue
A Global Study on the Correlates of Gross Domestic Product (GDP) and COVID-19 Vaccine Distribution
Previous Article in Journal
Comirnaty-Elicited and Convalescent Sera Recognize Different Spike Epitopes
Previous Article in Special Issue
Human Papilloma Virus Vaccination: Focus on the Italian Situation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Importance of Nanocarrier Design and Composition for an Efficient Nanoparticle-Mediated Transdermal Vaccination

1
Departamento de Farmacia, Escuela de Química y Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
2
Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
3
Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins No. 340, Santiago 7810000, Chile
4
Departamento de Endocrinología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
5
Millennium Institute on Immunology and Immunotherapy, Departamento de Farmacia, Escuela de Química y Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
6
Centro de Investigación en Nanotecnología y Materiales Avanzados “CIEN-UC”, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
*
Authors to whom correspondence should be addressed.
Vaccines 2021, 9(12), 1420; https://doi.org/10.3390/vaccines9121420
Submission received: 1 October 2021 / Revised: 22 November 2021 / Accepted: 23 November 2021 / Published: 1 December 2021
(This article belongs to the Special Issue Vaccines against Infectious Diseases and Cancer)

Abstract

:
The World Health Organization estimates that the pandemic caused by the SARS-CoV-2 virus claimed more than 3 million lives in 2020 alone. This situation has highlighted the importance of vaccination programs and the urgency of working on new technologies that allow an efficient, safe, and effective immunization. From this perspective, nanomedicine has provided novel tools for the design of the new generation of vaccines. Among the challenges of the new vaccine generations is the search for alternative routes of antigen delivery due to costs, risks, need for trained personnel, and low acceptance in the population associated with the parenteral route. Along these lines, transdermal immunization has been raised as a promising alternative for antigen delivery and vaccination based on a large absorption surface and an abundance of immune system cells. These features contribute to a high barrier capacity and high immunological efficiency for transdermal immunization. However, the stratum corneum barrier constitutes a significant challenge for generating new pharmaceutical forms for transdermal antigen delivery. This review addresses the biological bases for transdermal immunomodulation and the technological advances in the field of nanomedicine, from the passage of antigens facilitated by devices to cross the stratum corneum, to the design of nanosystems, with an emphasis on the importance of design and composition towards the new generation of needle-free nanometric transdermal systems.

1. Introduction

The recent pandemic caused by the SARS-CoV2 infection has shown the great importance of vaccines and their impact on preventing and controlling infectious diseases [1]. As a result, the attention to developing safe and effective vaccines has increased. However, the main route of antigen delivery remains parenteral, reducing the possibility of universal coverage since it can be considered traumatic for some individuals, requires qualified health professionals for application, and, in many cases, efficient cold chain management [2,3]. These factors make it challenging to develop vaccination programs in developing countries or remote areas [4,5]. In addition, the World Health Organization (WHO) estimates that at least 19.4 million infants in the world have not received basic vaccines, a scenario that has worsened due to the pandemic. Therefore, the need to find alternative routes for efficient, safe, and effective antigen delivery to induce protective immunity [6].
The transdermal route of administration provides multiple advantages for achieving this goal because it reduces both the first-pass metabolism and adverse effects, is non-traumatic, and allows self-administration by the patient making it an attractive delivery route for needle-free immunization [7,8]. However, the protective barrier function of the skin can restrain the step of macromolecule and antigen absorption. Those that manage to overcome the stratum corneum (SC) may be available to exert their pharmacological effect [9,10].
The skin is an easily accessible and highly immunocompetent organ [7,11], which can have up to 20 billion cells of various subtypes, such as keratinocytes, Langerhans cells, dendritic cells, T cells, and mast cells that contribute to the immunocompetence of skin [12]. To overcome the barrier that the stratum corneum imposes and favor transdermal permeability, various technologies have been developed, including iontophoresis [13], sonophoresis [14], magnetophoresis [15], electroporation [16], and laser microporation [17]. Unfortunately, these methods have shown significant economic limitations [18]. Therefore, microneedles are the most widely studied method to administer micro and macromolecules through the skin [19]. However, many researchers do not consider this delivery method as a “needle-free” approach. Thus, the development of highly efficient and optimized nanosystems is one of the strategies to cross the skin barrier and benefit from the immunocompetence of this tissue, with design being one of the factors to consider when penetrating the skin without invasive techniques. A solution to this problem could be developing nanosystems capable of transporting the antigen and bypassing this barrier without microneedles. Fulfilling this last objective largely depends on the design composition of the nanoparticles [20]. This article will focus on the advances that have been achieved in the area, with a strong focus on the design techniques of nanoparticle-based transdermal antigen delivery systems and the role of the configuration and use of excipients that favor the crossing of the skin barrier.

2. Mechanisms Involved in the Skin Immune Response

The skin is an extensive and complex organ that accomplishes a fundamental barrier function and comprises various layers that develop in different stages of gestation [21]. The epidermis development is a complex but coordinated process involving cell proliferation, differentiation, and adhesion steps [22]. This process begins in the first weeks of embryonic development, and stratification extends until the end of the first trimester of embryonic development, which ends with the differentiation of spiny cells into granular and cornified cells [22]. The dermis is organized in more advanced stages and continues its maturation weeks after birth [23,24]. Although cells of the immune system in the skin are not usually so abundant, a great density and diversity of immune cells are achieved after a complex development process, which constitutes the skin as a specialized barrier organ [25].
The properties of this barrier are granted mainly by the presence of the stratum corneum, which consists of the outermost layer of the skin, located on the viable epidermis with a thickness of around 15 to 20 layers composed mainly of dead tissue, assuming a barrier almost impenetrable for the vast majority of molecules with therapeutic activity [9]. But not only the intrinsic properties of SC can influence the transdermal passage, but also the physicochemical properties of the compounds can define entry efficiency. The physicochemical properties include molecular weight, solubility, and lipophilicity, which define their ability to be absorbed. Molecules with low molecular weight (less than 500 Da) [26] and log P between 1–4 are expected to diffuse easily [27]. On the other hand, larger molecules but with sufficient lipophilicity could enter through the annexed pathways that we will explain later. All these skin characteristics pose challenging work in administering assets via the transdermal route [8].
The passage of bioactive compounds through the stratum corneum is the first critical point of interest for delivering drugs through the skin. As mentioned above, the specific physicochemical characteristics are restricted and, if it is sufficiently lipophilic, it can enter through the lipophilic stratum corneum [26]. The passage of compounds through the skin can occur through the transepidermal pathway, either through the transcellular pathway that involves the passing through SC cells or the intercellular pathway (also known as paracellular) through the spaces between corneocytes (approximately 75 nm) [28]. On the other hand, the transpedicular pathway consists of skin attachments, such as sweat glands, sebaceous glands, and hair follicles. Normally, this route is not relevant for drug administration as it constitutes only 0.1% of the human skin [29]. However, it plays an important role in highly lipophilic drugs that could form reservoirs in the sebaceous glands or high molecular weight compounds such as nanoparticles, facilitating their entry through hair follicles. Figure 1 shows the various access routes of the active ingredients through the skin [30,31].
Once the molecule enters through the stratum corneum, it will face the interface between the viable epidermis and the dermis with hydrophilic characteristics; given the above, only compounds capable of ionizing will cross, facing the enzymatic activity of the skin [32]. As we can see, the passage of molecules through the skin poses significant challenges. In this regard, various approaches have been developed to facilitate the passage of active compounds. Specifically, in this review, we will address advances in immunization since it is in this field where we find advantageous characteristics for administration.
Drug or antigen delivery via the transdermal route has several advantages (Figure 2). First, pre-systemic metabolism can be avoided, being a candidate for compounds with extensive hepatic metabolism. Second, the transdermal route offers a minimally invasive approach. Third, transdermal delivery avoids infections associated with the manipulation of conventional needles. Fourth, the transdermal route allows dose reduction due to the existence of minimal metabolism. Finally, it has the potential for self-administration and effective induction of the immune system, making it an attractive route for non-invasive immunization [30,31].

Innate and Adaptive Immune Response of the Skin

The potential of transdermal immunization is supported by the abundant presence of cells of the immune system in the skin, which can trigger an effective antigen-specific immune response [33]. Langerhans cells were initially described in 1868 by Paul Langerhans [34] and correspond to a subtype of dendritic cells with a stellate shape located mainly at the base of the epidermis [35]. Langerin expression characterizes these cells; this protein plays a fundamental role in presenting antigens to T25 cells and corresponds to a type C lectin, which is localized in cytoplasmic organelles with a striated appearance inside Langerhans cells called Birbeck granules [36]. It has been recently shown that the expression of the kinases activated by serine/threonine p21 (PAK1) in Langerhans cells contributes significantly to the maintenance of epidermal stem cells, which in turn can be related to autoimmune pathologies and skin cancer, underscoring the importance of this cell type for skin immunomodulation [37]. An essential link between innate and adaptive immunity is the dendritic cell, which can activate naïve T cells and contributes to the initiation of both cellular and adaptive humoral immunity [38], Specifically, in the skin, we can find dermal dendritic cells, which correspond to a subtype of dendritic cells; the evidence shows that they present a greater activation than blood dendritic cells, promoting a strong proliferation of T cells, two populations of dermal dendritic cells; CD1c + DC and CD141 + CD, the latter being responsible for the cross-presentation of CD8 + T antigens [39]. On the other hand, the recent discovery of various subtypes of innate lymphoid cells (ILC), such as ILC1/2/and 3, has contributed to the complexity of the immunomodulation mechanisms in the skin [40,41]. Despite coming from a common lymphoid progenitor, ILCs lack the specific rearranged antigen receptors expressed by T cells and the three ILC subtypes located at different skin layers [42].
Keratinocytes (KCs) are cells that produce keratin in the skin and constitute a high percentage of the cells of the epidermis response [39], forming an efficient barrier, which works as the first line of defense against skin pathogens and exogenous substances [43]. KCs express Toll-Like receptors (TLR) and secrete several types of chemokines and proinflammatory cytokines in response to TLR stimulation by PAMPs. Thus, high expression of interleukin 33 (IL-33), a member of the IL-1 family, has been shown to activate helper T cells, macrophages and induce the ILC innate lymphoid cell family [44]. The recent discovery of various subtypes of innate lymphoid cells (ILC), such as ILC1/2/and 3, has added to the complexity of the immunomodulation mechanisms in the skin [40,41]. Despite coming from a common lymphoid progenitor, ILCs lack the specific rearranged antigen receptors expressed by T cells and the three ILC subtypes located at different skin layers [42]. Dermal fibroblasts are another cell type that makes up the skin and express TLR-type receptors even at higher levels than keratinocytes. One of the functions of these cells is to secrete components of the extracellular matrix [45].

3. Transdermal Immunization Based on Physical Methods to Go across the Stratum Corneum

As discussed above, a significant challenge for administering transdermal vaccines is to overcome the stratum corneum [46]. In this regard, various approaches have been explored, based either on devices disrupting the skin barrier or on vehicles that facilitate antigen passage through the skin. Figure 3 summarizes the most commonly used approaches to achieve this transdermal penetration. These approaches have been used alone or in combination with nanosystems, described in the following sections.

3.1. Transdermal Administration Based on Microneedles

One of the strategies that have been successfully applied for transdermal administration and that causes minor disruption of the stratum corneum is based on microneedles [47]. This strategy manages to overcome the limitations of the conventional parenteral route, as needles with sizes of micrometers can penetrate the layers of the skin, avoiding the typical discomfort in the patient due to pain, bleeding, and risk of infections [48]. Furthermore, microneedles do not suffer from limitations associated with the particle size of bioactive compounds since these devices usually have a larger size. Because of these features, there are already several products marketed for therapies, such as the intradermal influenza vaccine Intanza® [49] or the acne treatment Dermaroller [50]. Furthermore, microneedles are usually classified according to the material with which they have been produced or the active release profile. Thus, we can find dissolving-, solid-, coated- and hollow-microneedles [51].
In the field of transdermal immunization, microneedles associated with free antigen or combined with nanosystems have been used successfully. Table 1 shows some nanosystems designs for diphtheria and a new DNA vaccine against SARS-CoV-2. Microneedles are undoubtedly the most developed devices to date in transdermal immunization [52]. Although recently a very low-cost microneedle-anchored electroporator device for immunization against SARS-CoV-2 was shown, there are still challenges to overcome [53]. Current studies aim to overcome the remaining challenges in this area [54]. New technologies have been developed to manufacture microneedles to obtain improved compatibility when entering the stratum corneum. Examples are soluble microneedles, made with totally biocompatible water-soluble materials that penetrate the skin barrier and then solubilize with the active principle [55]. The oxidation of the material and the shortening of the microneedle length to avoid skin irritation has been another focus of study, leading, for example, to the development of microporous polymeric microneedles [56]. The biocompatibility of the devices has also been an essential subject of study, due to the presence of possible unwanted effects in the application site, such as irritation, inflammation, pore enlargement, and modification of the skin barrier [57,58]. It should be noted that these challenges to be overcome are common to other administration methods; this review will not focus on microneedles mainly; however, we do recommend to the reader some excellent reviews to delve into the advances of this technology [59].

3.2. Transdermal Administration Based on Electrical Techniques

Iontophoresis consists of the application of electrical current through the skin to favor the penetration of specific molecules. This method is an effective and non-invasive route of penetration [67]. Among the limitations that this administration technique faces for transdermal vaccination is the difficulty of administering the antigen in a focused way, avoiding permeation towards the muscle, and efficiently achieving its accumulation and subsequent stimulation of Langerhans cells. Combining this technique with nanoencapsulated bioactive compounds is a promising approach to overcoming this challenge [68,69].
Other techniques that use this basis for the delivery of bioactive compounds include sonophoresis, in which ultrasonic energy is used to induce the entry of assets through the skin [70]. Another technique is magnetophoresis, in which electric charges induce a magnetic field and with this occurs the vectorization of the drug [71]. We also have electroporation; in this technique, an aqueous pore is created in the skin by exposing it to high voltages for short periods, allowing the entry of bioactive compounds [72]. Finally, we find microporation, which, like electroporation, is based on creating a pore that will enable the passage of bioactive compounds. Still, this time the energy is transmitted through a metallic element by conduction, which produces a non-transitory pore in the skin due to the increase in temperature at the level of the stratum corneum [73]. Figure 3 shows a schematic representation of the active diffusion techniques and passive diffusion through barriers. It is important to note that once the stratum corneum has been crossed, the particles can release their content to produce the immune response.

3.3. Transdermal Administration Based on Other Approaches

Star-shaped particles have been developed, made of aluminum oxide or stainless steel, which can generate pores in the skin and thus overcome the stratum corneum barrier. The authors achieved surprising results in improving the survival of mice with cutaneous melanoma treated with 5-fluorouracil and in vaccination against tetanus toxin [74]. However, it is still unclear whether these pores are harmful to the skin in the long term. That is why in the next section, we will review the approaches that have been made to achieve a needle-free vaccination, focusing on the design and composition.
Another relatively novel device used is the PharmaJet Needle-Free Jet injector device, which has been shown to successfully administer the influenza virus vaccine in a randomized trial that compared it with intramuscular administration. The results were not inferior to those obtained with conventional administration, offering an alternative to traditional needle syringes since the device does not have a needle [75]. However, the costs associated with this type of device remain a disadvantage in this field. Table 1 shows another example of the successful use of this device to administer the ovalbumin antigen. Additionally, it incorporates the recent advances in nanosystem technologies used for immunization in in vivo tests for different antigens.

4. Nanosystem-Based Antigen Delivery Systems Noninvasive; Needle-Free Administration

Until now, we have reviewed the characteristics that make the skin and the transdermal pathway promising for immunization and the administration techniques used to cross the stratum corneum and their limitations, highlighting as the main challenge the achievement of immunization without damaging the stratum corneum.
One of the factors that make needle-free immunization desirable is the fact that the rupture methods used to allow the passage of particles such as microneedles, microporation, abrasion, among others, not only produce the response of the innate immune system, generating skin reactions adverse effects in patients, but also interrupt the skin barrier, which implies a greater risk of infections. This does not mean a greater compromise in healthy patients; however, in immunocompromised people, patients with difficult healing, children, and the elderly pose a greater risk that limits extensive use, without also considering the use of these devices complicates administration [76].
In this race to obtain adequate and safe transdermal vaccines, the incorporation of nanomedicine has made a significant contribution [77]. This has allowed, for example, to improve the revised administration techniques with the incorporation of controlled antigen release systems that have allowed to overcome some limitations such as thermostability [78], to enhance permeability by having a small particle size and increasing the contact surface [79], to reduce doses avoiding the manifestation of adverse reactions and to improve pharmacokinetic profiles [80]. The main advantages of nanovaccines are summarized in Figure 4. Next, this review will address the advances in administering vaccines through the skin using nanosystems, their types, designs, approaches, and challenges for the design and composition of nanosystems.
The most widely used nanosystems for antigen delivery are nanoparticles, liposomes, polymeric nanoparticles, niosomes, cubosomes, ethosomes, gold nanoparticles, and nanoemulsions. The choice of the type of nanometric particle depends mainly on the bioactive characteristics of the compound and the chosen route of administration; therefore, it is crucial to consider its use either combined with some administration method of those already reviewed or by itself. The design orientation is based on the delivery of passive or active bioactive compounds. Table 2 summarizes the various types of nanosystems used for antigen administration, their properties, their specific application in the immunization area, and the main challenges that remain to be faced with making their use in the field of immunization. Next, we will address some nanosystems that have been more widely used.

4.1. Liposomes

Liposomes correspond to a double layer commonly formed by phospholipids or other derivatives and cholesterol [95]. These systems have been widely studied to transport antigens and active molecules due to their possible adjuvant effect, triggering an efficient immune response [96,97]. On the other hand, their composition gives them high biocompatibility and the possibility of directing immunological therapies to the different targets in a controlled way [88,98]. The properties that affect this process are the physicochemical characteristics such as particle size, Z potential, polydispersity, and lipid composition [99,100,101]. To date, multiple approaches have been developed to combat infectious diseases by associating antigens with this type of nanocomposite [102,103,104]. Its efficacy for administering antigens against SARS-CoV-2 by different routes of administration is still being evaluated [105]. In the area of transdermal immunization, they have been used in conjunction with dissolvable microneedles to develop vaccines against leishmaniasis [106] and for non-invasive delivery of vaccines against tetanus toxoid [107], among others (see Table 2).

4.2. Nanocomposites Derived to Liposomes

In the field of needle-free transdermal immunization, it has been suggested that the rigid structure of liposomes makes it challenging to pass through the skin barrier [89], which is why multiple modifications have been incorporated to create liposome-derived nanosystems that can circumvent such limitations.

4.2.1. Transferosomes

Transferosomes are elastic liposomes composed of phospholipids, which form deformable vesicles and increase transdermal permeability in the presence of a hydration gradient in the stratum corneum [108]. Their composition, based mainly on edge activating surface surfactants such as sodium cholate, polysorbates, and Sorbites, allows a modulation in the flexibility of the sheath, allowing them to pass through the pores of the skin, thus opening the way to needle-free vaccination [109]. Among the advantages of using these designs are their high flexibility, their ability to encapsulate hydrophilic and hydrophobic compounds, and their ability to incorporate molecules of peptide origin [110]. Due to their composition, they lack biocompatibility problems; like other nanosystems and can be used for topical and systemic treatments.
The mechanism of transdermal entry of transferosomes is based on the presence of border activators, in a first step; They allow the nanosystem to pass through the stratum corneum through channels with diameters of less than 50 nm. In a second step, the component derived from phospholipids is capable of sealing the vesicle and transporting it through the pore, the gradient produced by the difference in water content between the surface of the skin and the epidermis produces what is known as “transdermal gradient”, which allows the passage of the nanosystem [111]. This is how they are capable of crossing the skin barrier by the transcellular or intercellular route [112].
Despite its beneficial properties for transdermal immunization, there is currently a small number of studies in this field, we can mention tetanus vaccine designs [113] and against the virus responsible for hepatitis B [86]. The above is a motivation to overcome the challenges that limit its use, mainly derived from its oxidative stability [114], high cost associated with the constituent lipids, and difficulty of reproducibility of the preparations [115]. So, there is still a long way to understand the interaction between the compounds, components, and stabilization of these nanosystems to develop transdermal vaccines.

4.2.2. Ethosomes

Ethosomes correspond to a type of liposome that contains in its composition between 20 to 40% ethanol. Their properties include a great drug encapsulation capacity and stability in comparison with the classic liposomes, negative Z potential, size smaller than 200 nm, which decrease as the ethanol concentration increases, highly deformable, non-toxic and highly biocompatible [116], skin permeability facilitated by ethanolic content [117,118]. The passage mechanism through the stratum corneum of ethosomes involves two steps: the contact of SC lipids with ethanol produces a composition alteration, known as the “ethanol effect”. In a second phase, the breakdown of the superficial lipids generates a decrease in the skin barrier compaction that allows the flexible structure of ethosomes to enter through the skin and interact with the polar component of lipids, known as the “ethosome effect” [119]. By using ethosomes marked with fluorescent probes, the molecular mechanism of entry of ethosomes was alucidated. Thus, the passage through the SC by intercellular pathways can take place without damaging the structure of the SC and distributing mainly in the cell membrane.
Ethosomes have been developed for various applications, such as treatment of inflammation, analgesia, skin conditions, among others [120]. Although few studies have been carried out in the field of transdermal immunization, it has been shown enhanced transdermal penetration by ethosomes marked with rhodamine [121]. Additionally, ethosomes in conjunction with biopolymers for the administration of ovalbumin, effectively stimulating the response of the immune system and they have been incorporated into carbomer gels to achieve vehicles that can be administered needle-free [122]. It was also shown that ethosomes that include hyaluronic acid or chitosan formulated by layer-by-layer self-assembly promote the stimulation of IL-2 and IL-6 and cytokines associated with dendritic cell maturation when loaded with an antigen [89]. The challenges associated with the formulation of this type of nanosystem are summarized in Table 2.

4.2.3. Niosomes

Niosomes are liposome-type nanometric structures formed by the assembly of non-ionic surfactants, which are mainly derivatives of alkyl or dialkyl polyglycerol ether and cholesterol that are subsequently hydrated. Since they were discovered in the cosmetic industry in the 1980s, these structures have been widely formulated. Depending on their manufacturing method, Niosomes can be unilamellar or multilamellar (obtained by thin layer evaporation, addition of molten lipids, addition of hot water, ether injection, microfluidics, among others) [123]. Advantages of using niosomes include easy large-scale production, high stability, low toxicity, and high transdermal penetration [124].
A successful example of the use of niosomes for transdermal vaccination is a 60% increase in the immune response by encapsulating the antigen to prevent Newcastle disease [125]. However, in recent years, despite the great apogee of niosomes, no significant advances have been reported in the field of transdermal immunization. This scenario could be due to the use of organic solvents, phase heating (non-appropriate for thermolabile compounds), or a large number of existing patents for these vehicles.

4.2.4. Cubosomes

Cubosomes are composed of two aqueous channels, separated by a lipid bilayer arranged three-dimensionally either in a lipid form, water, double diamond, rotated, or primitive [126,127]. Among the lipids most used to formulate them is the [128], while surfactants derived from poloxamers have been the most used to stabilize the cubic structures of these nanosystems [129]. Among the ways of obtaining it, the bottom-up approach and top-down approach stand out. The latter is the most used and consists of the formation of the viscous primary structure formed by lipids and its subsequent dispersion in water by applying high-energy methods [130,131]. The designs for transdermal immunization of niosomes and cubosomes are still small (Table 2) and are not without challenges when formulating them. It is expected that with the progress of research and incorporation of suitable stabilizers, these limitations can be overcome by the encapsulation of assets and transdermal penetration, preventing them from losing their conformation as they pass through the various layers of the skin [132].

4.3. Nanoparticles

Nanoparticles correspond to colloidal structures of nanometric size, for nanomedicine approaches, usually less than 500 nm. Depending on their formulation, we can find polymeric nanoparticles associated with polymers by interacting electrostatic charges or nanocapsules, which generally have a lipid core and a polymer shell. Both designs can incorporate compounds of therapeutic interest either by encapsulation or association by adsorption on the surface [80,133,134].

4.3.1. Polymeric Nanoparticles

New approaches based on incorporating biodegradable polymers into their composition have shown great potential in the biomedical area and the field of immunization [135]. These nanosystems are capable of containing not only antigens of interest but also various adjuvants. Being biodegradable, they can maintain the release of compounds from days to several weeks generating biocompatible waste [81,82]. Table 2 shows recent examples of formulations for transdermal immunization using these nanocarriers.
The pandemic caused by the SARS-CoV-2 virus has not only triggered a broad race to find effective vaccines against this virus. Still, it has also allowed novel vaccines based on nanoparticles loaded with messenger RNA to reach the market, positioning itself as the first of such designs to be approved by regulatory agencies such as the EMA and the FDA [136]. This is how vaccines such as Pfizer and BioNTech RNA: BNT162b2 and modern mRNA-1273 based on purified messenger RNA are used today [137,138]. Although these designs are administered parenterally, without a doubt, they open the door to the development of new technologies, aiming at needle-free vaccination and is not only limited to the transport of antigens but also undoubtedly an open door towards the development of new therapies for the treatment of COVID-19 [139].

4.3.2. Nanocapsules

Systems with nanometric size and core-shell structure and coated mainly with polymeric compounds are called nanocapsules. Among their characteristics, we find the ability to induce long-lasting immune responses, dose reduction, and reduction of adverse effects [140]. Our research group has worked on the development of nanocapsules capable of carrying antigens for needle-free transdermal immunization and shown that polymeric nanocapsules with chitosan shell, loaded with Ovalbumin (OVA) are stable, with a high association of the protein capable of interacting with the immune system and being, in an ex vivo model in pigskin, better retained than OVA in solution [33]. This property has been reinforced by associating hyaluronic acid as a biopolymer, showing promising systems for needle-free transdermal administration [141].

4.4. Nanoemulsions

Nanoemulsions are heterogeneous mixtures that can be formed by drops of oil in an aqueous medium (O/W) or drops of water dispersed in oil (W/O), stabilized by incorporating surfactants and being of nanometric size [60]. They have shown great potential in transdermal vaccination; however, it is still necessary to study the mechanisms by which they could cross the skin barrier. Until now, there is a history that this is dependent on size, which is mainly called the note is that in this case, smaller sizes do not necessarily imply a greater transdermal passage [142].
In the case of transdermal immunization, they have been designed in conjunction with Imiquimod to induce enhanced responses in T lymphocytes [143]. In the case of the incorporation of biopolymers, the influence of the polymeric coating on transdermal penetration has been studied [144]. Recently, the nanoemulsion MF59, an authorized and approved preparation with commercial use for the administration of parenteral vaccines against influenza, has been associated with microneedles, demonstrating a painless administration and maturation of dendritic cells [145]. Although only a few studies for the use of nanoemulsions for transdermal vaccination have been published, their versatility, stability, and a large number of studies associated with their excellent safety profile make them promising vehicles for the fight against COVID-19 disease, not only as antigen carriers but also as carriers of various active molecules against the SARS-CoV-2 virus [146].

5. Novel Approaches to Design Nanoparticles for Needle-Free Transdermal Delivery Based on Their Composition

One of the most recent approaches to address the problem of needle-free vaccination is to focus on the composition of the design without losing sight of the nanometric size. The first approaches in this field were not very far from what is known today; it is necessary to have a small particle size to bypass the stratum corneum [102], and this was the predominant approach in designs for a long time. Large amounts of surfactants were used to achieve this objective. However, today it is known that it is not enough to have a small particle size and adequate lipophilicity; it is also necessary to have excipients that will allow the stratum corneum to be reversibly and non-aggressively opened and thus will enable the passage of nanosystems. Among the components that can be highlighted recently, the use of Compritol 888 ATO has been described as a promoter of transdermal penetration in polymeric nanoparticles that encapsulate ovalbumin and as adjuvant Imiquimod [7].

5.1. Azones

Azones and their derivatives are among the main transdermal permeability enhancers that can be used to manufacture nanosystems. These molecules are composed of a polar and a hydrophobic chain and can enhance transdermal penetration at low concentrations. It is most extensively used alongside laurocapram; however, it should be noted that this or its derivatives have not been yet incorporated for the development of nanosystems for needle-free immunization purposes [117,147].

5.2. Fatty Acids

Fatty acids such as oleic acid, stearic acid, and ethyl oleate are approved by the U.S. Food and Drug Administration (FDA) and can provide various advantages when incorporated into the designs. These excipients allow increasing the transdermal penetration of format dependent on its structure and chain length. The mechanism of action for the transdermal penetration increase is based on the increase in the diffusion coefficient of the skin due to the improvement of the interaction between the preparations with the lipid layer of the skin [30,124].

5.3. Alcohols

Alcohols are excipients that can be incorporated into nanosystems both in their internal and external phases. These can improve the solubility of encapsulated compounds and their ability to enhance the permeability of the skin. Ethanol and isopropyl alcohol have been the most used in topical preparations [148]. However, the use of alcohols represents an excellent design challenge since it is important to optimize the quantity due to their high toxicity when used in high concentrations [149,150]. Otherwise, the encapsulated compounds may be released from the nanosystems, their equilibrium may be broken, and their nanometric structure may be lost. The quantity, length of chain, and phase in which they are added suppose a significant challenge in the design [126].

5.4. Polymers

Polymers are excipients composed of simple subunits joined by covalent bonds called monomers and have been used in nanoparticle designs for various purposes. Among them, we can highlight the increase in the stability of the particles when they are located in external areas due to the particle-particle steric hindrance or as surfactants [108]. On the other hand, these compounds have been described in the field of nanovaccines as thermal stabilizers, and it has been shown that even at concentrations lower than those normally used as surfactants, they are capable of protecting antigens from degradation by keeping them at room temperature, without loss of efficiency [151].

5.5. Polysaccharides

Most polysaccharides are generally recognized as safe (GRAS), and they are widely used for various purposes. In the field of immunization, their use in formulations has been recognized for their ability to activate cells of the immune system. In the field of nanocarrier design of antigens, they stand out for their property of increasing transdermal permeability since they are capable of promoting increased permeability in the stratum corneum, which is, as has been mentioned with much emphasis, one of the main barriers to overcome [152]. These excipients have not only the property already described to enhance permeability; moreover, the incorporation of these excipients in nanoparticle-based vaccine formulations significantly increases the thermostability of antigens. An example of this has been reported for a DNA vaccine for immunization against the Ebola virus, in which the antigen in its non-encapsulated form required −70 °C for storage. In its nano encapsulated state, it no longer requires refrigeration. It is important to note that this strategy was combined with microneedles and has not been applied in needle-free systems, so there is a potential benefit with incorporation into the designs, largely because they are inexpensive excipients but with the limitation that a significant amount must be used to obtain the thermostability enhancer effect [153].

6. Projections

The transdermal route is a promising route of administration for immunization. Among its advantages, we find the great diversity of cells of the immune system, it is not traumatic, its great acceptance by patients, and the potential to improve the deficient characteristics of bioactive compounds. However, although multiple studies have been conducted, we do not yet have commercially available transdermal vaccines. Much progress has been made, especially in the field of microneedles, but there are still many challenges to overcome. When facing these challenges, the safety and efficacy of administering nanocomposites play a very relevant role [137]. The recent approval of parenteral vaccines that use this technology, such as the BNT162b2 vaccine (BioNTech/Pfizer) and the modern design, have shown high efficacy and safety in clinical, double-blind, multicenter, and randomized studies [154,155,156,157]. On the other hand, there is a tremendous challenge in scaling these designs to applications that can be massively developed for the population. Along these lines, it was recently shown that incorporating engineering strategies for production A batch scale of polymeric nanoparticles and even a continuous manufacturing line using microfluidics is feasible and has much potential for these applications [158].
Another relevant characteristic is the manufacturing reproducibility of the nanosystems to comply with the regulatory requirements. Therefore, the improvement of the animal study models used to evaluate the biogenicity of the materials used to constitute nanosystems needs improvement [159]. The costs associated with the production of nanocomposites will continue to be a focus of development. Undoubtedly, immunization of the population continues to be the most cost-effective way to prevent diseases [160]. Efforts continue to focus on low-cost excipients; however, only interdisciplinary work in conjunction with the production laboratories will improve production costs [161]. Needle-free administration must focus on the size and stability of the nanosystems and their composition and rational design. For years, the design and stabilization of nano-sized vehicles have relied on practical strategies and pseudo-ternary phase diagrams [134]. The most elaborate approaches have tackled the problem using Box-Behnken [162] or staggered-level designs in which various combinations of the chosen excipients are tested [163]. However, it is essential to have strategies that optimize designs in less time and consider limited resources [164]. In this context, incorporating computational techniques arises to tackle the design process with a multidisciplinary approach. The use of mathematical models and machine learning as a nanosystem optimization technique is incipient [164]. However, it is a valuable resource capable of reducing design times and the number of experiments required. Experimental data are needed to “fit” or “train” models of the effect of the composition, and they allow to explore the impact of the formulation composition in a range defined for each component [165,166].
One of the projections that this review article seeks to deliver is to motivate researchers to incorporate these types of tools that have been successfully included for the design of new and optimized promising nanosystems that not only limit their use to nanovaccines [167]. Unfortunately, the need to use elaborate programming interfaces, often away from the medical personnel who direct these investigations, has limited their use. Therefore, mathematical modeling and the use of artificial inteligence are an open door, which could allow rapid advances in the field of nanovaccines. Time and the training of interdisciplinary professionals will be able to take advantage of their potential.

Author Contributions

Conceptualization: R.Y.V.-O., J.V.G.-A. and A.M.K., writing—original draft preparation: R.Y.V.-O., writing-review, supervision, and editing: R.Y.V.-O., M.R.-F., M.J.Á.-F., J.V.G.-A. and A.M.K., funding acquisition J.V.G.-A. and A.M.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by FONDECYT grants Nº 1201482 and Nº 1190830, Interdisciplinary Grant 2020 by Pontificia Universidad Católica de Chile and The Millennium Institute on Immunology and Immunotherapy ICN09_016.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

R. Y. Valdivia-Olivares appreciates her scholarships from ANID, Chile (Nº 21212052).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rey-Jurado, E.; Tapia, F.; Muñoz-Durango, N.; Lay, M.K.; Carreño, L.J.; Riedel, C.A.; Bueno, S.M.; Genzel, Y.; Kalergis, A.M. Assessing the importance of domestic vaccine manufacturing centers: An overview of immunization programs, vaccine manufacture, and distribution. Front. Immunol. 2018, 9, 26. [Google Scholar] [CrossRef]
  2. Kim, K.S.; Kim, H.; Park, Y.; Kong, W.H.; Lee, S.W.; Kwok, S.J.J.; Hahn, S.K.; Yun, S.H. Noninvasive Transdermal Vaccination Using Hyaluronan Nanocarriers and Laser Adjuvant. Adv. Funct. Mater. 2016, 26, 2512–2522. [Google Scholar] [CrossRef]
  3. Joyce, J.C.; Carroll, T.D.; Collins, M.L.; Chen, M.H.; Fritts, L.; Dutra, J.C.; Rourke, T.L.; Goodson, J.L.; McChesney, M.B.; Prausnitz, M.R.; et al. A Microneedle Patch for Measles and Rubella Vaccination Is Immunogenic and Protective in Infant Rhesus Macaques. J. Infect. Dis. 2018, 218, 124–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Choi, Y.H.; Perez-Cuevas, M.B.; Kodani, M.; Zhang, X.; Prausnitz, M.R.; Kamili, S.; O’Connor, S.M. Feasibility of Hepatitis B vaccination by microneedle patch: Cellular and humoral immunity studies in rhesus macaques. J. Infect. Dis. 2019, 220, 1926–1934. [Google Scholar] [CrossRef] [PubMed]
  5. Lee, B.Y.; Haidari, L.A. The importance of vaccine supply chains to everyone in the vaccine world. Vaccine 2017, 35, 4475–4479. [Google Scholar] [CrossRef] [PubMed]
  6. Chopra, M.; Bhutta, Z.; Chang Blanc, D.; Checchi, F.; Gupta, A.; Lemango, E.T.; Levine, O.S.; Lyimo, D.; Nandy, R.; O’Brien, K.L.; et al. Addressing the persistent inequities in immunization coverage. Bull. World Health Organ. 2020, 98, 146–148. [Google Scholar] [CrossRef] [PubMed]
  7. Alvarez-Figueroa, M.J.; Narváez-Araya, D.; Armijo-Escalona, N.; Carrasco-Flores, E.A.; González-Aramundiz, J.V. Design of Chitosan Nanocapsules with Compritol 888 ATO® for Imiquimod Transdermal Administration. Evaluation of Their Skin Absorption by Raman Microscopy. Pharm. Res. 2020, 37, 1–10. [Google Scholar] [CrossRef] [PubMed]
  8. Prausnitz, M.R.; Langer, R. Transdermal drug delivery. Nat. Biotechnol. 2008, 26, 1261–1268. [Google Scholar] [CrossRef] [PubMed]
  9. Chen, Z.; Lv, Y.; Qi, J.; Zhu, Q.; Lu, Y.; Wu, W. Overcoming or circumventing the stratum corneum barrier for efficient transcutaneous immunization. Drug Discov. Today 2018, 23, 181–186. [Google Scholar] [CrossRef] [PubMed]
  10. Koutsonanos, D.G.; Martin, M.d.P.; Zarnitsyn, V.G.; Sullivan, S.P.; Compans, R.W.; Prausnitz, M.R.; Skountzou, I. Transdermal influenza immunization with vaccine-coated microneedle arrays. PLoS ONE 2009, 4, e4773. [Google Scholar] [CrossRef]
  11. Gupta, R.; Rai, B. Effect of Size and Surface Charge of Gold Nanoparticles on their Skin Permeability: A Molecular Dynamics Study. Sci. Rep. 2017, 7, 45292. [Google Scholar] [CrossRef] [Green Version]
  12. Ita, K. Transdermal delivery of vaccines–Recent progress and critical issues. Biomed. Pharmacother. 2016, 83, 1080–1088. [Google Scholar] [CrossRef] [PubMed]
  13. Bernardi, D.S.; Bitencourt, C.; da Silveira, D.S.; da Cruz, E.L.; Pereira-da-Silva, M.A.; Faccioli, L.H.; Lopez, R.F. Effective transcutaneous immunization using a combination of iontophoresis and nanoparticles. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 2439–2448. [Google Scholar] [CrossRef] [PubMed]
  14. Ita, K. Recent progress in transdermal sonophoresis. Pharm. Dev. Technol. 2017, 22, 458–466. [Google Scholar] [CrossRef] [PubMed]
  15. Maurya, A.; Lili, C.; Murthy, S.N. Magnetophoresis and Electret-Mediated Transdermal Delivery of Drugs. Nov. Deliv. Syst. Transdermal Intradermal Drug Deliv. 2015, 1, 147. [Google Scholar]
  16. Bernelin-Cottet, C.; Urien, C.; McCaffrey, J.; Collins, D.; Donadei, A.; McDaid, D.; Jakob, V.; Barnier-Quer, C.; Collin, N.; Bouguyon, E. Electroporation of a nanoparticle-associated DNA vaccine induces higher inflammation and immunity compared to its delivery with microneedle patches in pigs. J. Control. Release 2019, 308, 14–28. [Google Scholar] [CrossRef] [PubMed]
  17. Engelke, L.; Winter, G.; Engert, J. Application of water-soluble polyvinyl alcohol-based film patches on laser microporated skin facilitates intradermal macromolecule and nanoparticle delivery. Eur. J. Pharm. Biopharm. 2018, 128, 119–130. [Google Scholar] [CrossRef] [PubMed]
  18. Bhowmik, D.; Duraivel, S.; Kumar, K.S. Recent trends in challenges and opportunities in transdermal drug delivery system. Pharma Innov. 2012, 1, 9–23. [Google Scholar]
  19. Kang, N.-W.; Kim, S.; Lee, J.-Y.; Kim, K.-T.; Choi, Y.; Oh, Y.; Kim, J.; Kim, D.-D.; Park, J.-H. Microneedles for drug delivery: Recent advances in materials and geometry for preclinical and clinical studies. Expert Opin. Drug Deliv. 2021, accepted. [Google Scholar] [CrossRef]
  20. Souto, E.B.; Baldim, I.; Oliveira, W.P.; Rao, R.; Yadav, N.; Gama, F.M.; Mahant, S. SLN and NLC for topical, dermal, and transdermal drug delivery. Expert Opin. Drug Deliv. 2020, 17, 357–377. [Google Scholar] [CrossRef]
  21. Coolen, N.A.; Schouten, K.C.; Middelkoop, E.; Ulrich, M.M. Comparison between human fetal and adult skin. Arch. Dermatol. Res. 2010, 302, 47–55. [Google Scholar] [CrossRef] [Green Version]
  22. Du, H.; Wang, Y.; Haensel, D.; Lee, B.; Dai, X.; Nie, Q. Multiscale modeling of layer formation in epidermis. PLoS Comput. Biol. 2018, 14, e1006006. [Google Scholar] [CrossRef]
  23. Banks-Schlegel, S.P. Keratin alterations during embryonic epidermal differentiation: A presage of adult epidermal maturation. J. Cell Biol. 1982, 93, 551–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Leung, A.; Crombleholme, T.M.; Keswani, S.G. Fetal wound healing: Implications for minimal scar formation. Curr. Opin. Pediatrics 2012, 24, 371. [Google Scholar] [CrossRef] [Green Version]
  25. Botting, R.A.; Haniffa, M. The developing immune network in human prenatal skin. Immunology 2020, 160, 149–156. [Google Scholar] [CrossRef] [PubMed]
  26. He, N.; Warner, K.S.; Chantasart, D.; Shaker, D.S.; Higuchi, W.I.; Li, S.K. Mechanistic study of chemical skin permeation enhancers with different polar and lipophilic functional groups. J. Pharm. Sci. 2004, 93, 1415–1430. [Google Scholar] [CrossRef] [PubMed]
  27. Kang, L.; Yap, C.; Lim, P.; Chen, Y.; Ho, P.; Chan, Y.; Wong, G.; Chan, S. Formulation development of transdermal dosage forms: Quantitative structure-activity relationship model for predicting activities of terpenes that enhance drug penetration through human skin. J. Control. Release 2007, 120, 211–219. [Google Scholar] [CrossRef] [PubMed]
  28. Baroli, B.; Ennas, M.G.; Loffredo, F.; Isola, M.; Pinna, R.; López-Quintela, M.A. Penetration of metallic nanoparticles in human full-thickness skin. J. Investig. Dermatol. 2007, 127, 1701–1712. [Google Scholar] [CrossRef] [PubMed]
  29. Lane, M.E. Skin penetration enhancers. Int. J. Pharm. 2013, 447, 12–21. [Google Scholar] [CrossRef]
  30. Kitaoka, M.; Wakabayashi, R.; Kamiya, N.; Goto, M. Solid-in-oil nanodispersions for transdermal drug delivery systems. Biotechnol. J. 2016, 11, 1375–1385. [Google Scholar] [CrossRef]
  31. He, Y.; Hong, C.; Li, J.; Howard, M.T.; Li, Y.; Turvey, M.E.; Uppu, D.S.S.M.; Martin, J.R.; Zhang, K.; Irvine, D.J.; et al. Synthetic Charge-Invertible Polymer for Rapid and Complete Implantation of Layer-by-Layer Microneedle Drug Films for Enhanced Transdermal Vaccination. ACS Nano 2018, 12, 10272–10280. [Google Scholar] [CrossRef]
  32. Liu, C.; Quan, P.; Fang, L. Effect of drug physicochemical properties on drug release and their relationship with drug skin permeation behaviors in hydroxyl pressure sensitive adhesive. Eur. J. Pharm. Sci. 2016, 93, 437–446. [Google Scholar] [CrossRef]
  33. Bussio, J.I.; Molina-Perea, C.; González-Aramundiz, J.V. Lower-sized chitosan nanocapsules for transcutaneous antigen delivery. Nanomaterials 2018, 8, 659. [Google Scholar] [CrossRef]
  34. De Panfilis, G. Paul Langerhans’ death centennial, July 20, 1988. J. Investig. Derm. 1988, 91, 283. [Google Scholar] [CrossRef] [Green Version]
  35. Merad, M.; Ginhoux, F.; Collin, M. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat. Rev. Immunol. 2008, 8, 935–947. [Google Scholar] [CrossRef] [PubMed]
  36. Yan, B.; Liu, N.; Li, J.; Li, J.; Zhu, W.; Kuang, Y.; Chen, X.; Peng, C. The role of Langerhans cells in epidermal homeostasis and pathogenesis of psoriasis. J. Cell. Mol. Med. 2020, 24, 11646–11655. [Google Scholar] [CrossRef]
  37. Okumura, K.; Saito, M.; Yoshizawa, Y.; Ito, Y.; Isogai, E.; Araki, K.; Wakabayashi, Y. Pak1 maintains epidermal stem cells by regulating Langerhans cells and is required for skin carcinogenesis. Oncogene 2020, 39, 4756–4769. [Google Scholar] [CrossRef]
  38. Banchereau, J.; Steinman, R.M. Dendritic cells and the control of immunity. Nature 1998, 392, 245–252. [Google Scholar] [CrossRef]
  39. Chambers, E.S.; Vukmanovic-Stejic, M. Skin barrier immunity and ageing. Immunology 2020, 160, 116–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Kim, B.S.; Wang, K.; Siracusa, M.C.; Saenz, S.A.; Brestoff, J.R.; Monticelli, L.A.; Noti, M.; Wojno, E.D.T.; Fung, T.C.; Kubo, M.; et al. Basophils Promote Innate Lymphoid Cell Responses in Inflamed Skin. J. Immunol. 2014, 193, 3717–3725. [Google Scholar] [CrossRef] [Green Version]
  41. Yang, J.; Hu, S.M.; Zhao, L.M.; Kaplan, D.H.; Perdew, G.H.; Xiong, N. Selective programming of CCR10(+) innate lymphoid cells in skin-draining lymph nodes for cutaneous homeostatic regulation. Nat. Immunol. 2016, 17, 48–56. [Google Scholar] [CrossRef] [Green Version]
  42. Chen, Y.-L.; Hardman, C.S.; Yadava, K.; Ogg, G. Innate lymphocyte mechanisms in skin diseases. Annu. Rev. Immunol. 2020, 38, 171–202. [Google Scholar] [CrossRef]
  43. Oshio, T.; Komine, M.; Tsuda, H.; Tominaga, S.I.; Saito, H.; Nakae, S.; Ohtsuki, M. Nuclear expression of IL-33 in epidermal keratinocytes promotes wound healing in mice. J. Dermatol. Sci. 2017, 85, 106–114. [Google Scholar] [CrossRef]
  44. Hesse-Macabata, J.; Morgner, B.; Morgenstern, S.; Grimm, M.; Elsner, P.; Hipler, U.; Wiegand, C. Innate immune response of human epidermal keratinocytes and dermal fibroblasts to in vitro incubation of Trichophyton benhamiae DSM 6916. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 1177–1188. [Google Scholar] [CrossRef] [PubMed]
  45. Medzhitov, R. Toll-like receptors and innate immunity. Nat. Rev. Immunol. 2001, 1, 135–145. [Google Scholar] [CrossRef]
  46. Alkilani, A.Z.; McCrudden, M.T.; Donnelly, R.F. Transdermal drug delivery: Innovative pharmaceutical developments based on disruption of the barrier properties of the stratum corneum. Pharmaceutics 2015, 7, 438–470. [Google Scholar] [CrossRef] [Green Version]
  47. Liu, S.; Jin, M.N.; Uuan, Y.S.Q.; Kamiyama, F.; Kusamori, K.; Katsumi, H.; Sakane, T.; Yamamoto, A. Transdermal delivery of relatively high molecular weight drugs using novel self-dissolving microneedle arrays fabricated from hyaluronic acid and their characteristics and safety after application to the skin. Eur. J. Pharm. Biopharm. 2014, 86, 267–276. [Google Scholar] [CrossRef] [PubMed]
  48. Brown, S.; Zambrana, P.N.; Ge, X.; Bagdure, D.; Stinchcomb, A.L.; Rao, G.; Tolosa, L. Minimally invasive technique for measuring transdermal glucose with a fluorescent biosensor. Anal. Bioanal. Chem. 2018, 410, 7249–7260. [Google Scholar] [CrossRef] [PubMed]
  49. Atmar, R.L.; Patel, S.M.; Keitel, W.A. Intanza®: A new intradermal vaccine for seasonal influenza. Expert Rev. Vaccines 2010, 9, 1399–1409. [Google Scholar] [CrossRef]
  50. Gadkari, R.; Nayak, C. A split-face comparative study to evaluate efficacy of combined subcision and dermaroller against combined subcision and cryoroller in treatment of acne scars. J. Cosmet. Dermatol. 2014, 13, 38–43. [Google Scholar] [CrossRef]
  51. Nagarkar, R.; Singh, M.; Nguyen, H.X.; Jonnalagadda, S. A review of recent advances in microneedle technology for transdermal drug delivery. J. Drug Deliv. Sci. Technol. 2020, 59, 101923. [Google Scholar] [CrossRef]
  52. O’Shea, J.; Prausnitz, M.R.; Rouphael, N. Dissolvable microneedle patches to enable increased access to vaccines against SARS-CoV-2 and future pandemic outbreaks. Vaccines 2021, 9, 320. [Google Scholar] [CrossRef] [PubMed]
  53. Xia, D.; Jin, R.; Byagathvalli, G.; Yu, H.; Ye, L.; Lu, C.-Y.; Bhamla, M.S.; Yang, C.; Prausnitz, M.R. An ultra-low-cost electroporator with microneedle electrodes (ePatch) for SARS-CoV-2 vaccination. Proc. Natl. Acad. Sci. USA 2021, 118, e2110817118. [Google Scholar] [CrossRef] [PubMed]
  54. Kolli, C.S. Microneedles: Bench to bedside. Ther. Deliv. 2015, 6, 1081–1088. [Google Scholar] [CrossRef]
  55. Lee, J.W.; Park, J.-H.; Prausnitz, M.R. Dissolving microneedles for transdermal drug delivery. Biomaterials 2008, 29, 2113–2124. [Google Scholar] [CrossRef] [Green Version]
  56. Bao, L.; Park, J.; Bonfante, G.; Kim, B. Recent advances in porous microneedles: Materials, fabrication, and transdermal applications. Drug Deliv. Transl. Res. 2021. Available online: https://link.springer.com/article/10.1007/s13346-021-01045-x (accessed on 20 November 2021). [CrossRef]
  57. Bal, S.M.; Caussin, J.; Pavel, S.; Bouwstra, J.A. In vivo assessment of safety of microneedle arrays in human skin. Eur. J. Pharm. Sci. 2008, 35, 193–202. [Google Scholar] [CrossRef]
  58. Waghule, T.; Singhvi, G.; Dubey, S.K.; Pandey, M.M.; Gupta, G.; Singh, M.; Dua, K. Microneedles: A smart approach and increasing potential for transdermal drug delivery system. Biomed. Pharmacother. 2019, 109, 1249–1258. [Google Scholar] [CrossRef]
  59. Guillot, A.J.; Cordeiro, A.S.; Donnelly, R.F.; Montesinos, M.C.; Garrigues, T.M.; Melero, A. Microneedle-based delivery: An overview of current applications and trends. Pharmaceutics 2020, 12, 569. [Google Scholar] [CrossRef]
  60. Zhang, Y.; Ng, W.; Feng, X.; Cao, F.; Xu, H. Lipid vesicular nanocarrier: Quick encapsulation efficiency determination and transcutaneous application. Int. J. Pharm. 2017, 516, 225–230. [Google Scholar] [CrossRef]
  61. Hernandez-Franco, J.F.; Mosley, Y.-Y.C.; Franco, J.; Ragland, D.; Yao, Y.; HogenEsch, H. Effective and Safe Stimulation of Humoral and Cell-Mediated Immunity by Intradermal Immunization with a Cyclic Dinucleotide/Nanoparticle Combination Adjuvant. J. Immunol. 2021, 206, 700–711. [Google Scholar] [CrossRef]
  62. Fotoran, W.L.; Kleiber, N.; Glitz, C.; Wunderlich, G. A DNA Vaccine Encoding Plasmodium falciparum PfRH5 in Cationic Liposomes for Dermal Tattooing Immunization. Vaccines 2020, 8, 619. [Google Scholar] [CrossRef] [PubMed]
  63. Du, G.; Woythe, L.; van der Maaden, K.; Leone, M.; Romeijn, S.; Kros, A.; Kersten, G.; Jiskoot, W.; Bouwstra, J.A. Coated and Hollow Microneedle-Mediated Intradermal Immunization in Mice with Diphtheria Toxoid Loaded Mesoporous Silica Nanoparticles. Pharm. Res. 2018, 35, 189. [Google Scholar] [CrossRef] [Green Version]
  64. Rostami, H.; Ebtekar, M.; Ardestani, M.S.; Yazdi, M.H.; Mahdavi, M. Co-utilization of a TLR5 agonist and nano-formulation of HIV-1 vaccine candidate leads to increased vaccine immunogenicity and decreased immunogenic dose: A preliminary study. Immunol. Lett. 2017, 187, 19–26. [Google Scholar] [CrossRef] [PubMed]
  65. Yin, Y.; Su, W.; Zhang, J.; Huang, W.; Li, X.; Ma, H.; Tan, M.; Song, H.; Cao, G.; Yu, S.; et al. Separable Microneedle Patch to Protect and Deliver DNA Nanovaccines Against COVID-19. ACS Nano 2021, 15, 14347–14359. [Google Scholar] [CrossRef]
  66. Wang, Y.; Li, S.; Dong, C.; Ma, Y.; Song, Y.; Zhu, W.; Kim, J.; Deng, L.; Denning, T.L.; Kang, S.-M.; et al. Skin Vaccination with Dissolvable Microneedle Patches Incorporating Influenza Neuraminidase and Flagellin Protein Nanoparticles Induces Broad Immune Protection against Multiple Influenza Viruses. ACS Appl. Bio Mater. 2021, 4, 4953–4961. [Google Scholar] [CrossRef] [PubMed]
  67. Wong, T.W. Electrical, magnetic, photomechanical and cavitational waves to overcome skin barrier for transdermal drug delivery. J. Control. Release 2014, 193, 257–269. [Google Scholar] [CrossRef]
  68. Hasan, M.; Khatun, A.; Fukuta, T.; Kogure, K. Noninvasive transdermal delivery of liposomes by weak electric current. Adv. Drug Deliv. Rev. 2020, 154, 227–235. [Google Scholar] [CrossRef]
  69. Toyoda, M.; Hama, S.; Ikeda, Y.; Nagasaki, Y.; Kogure, K. Anti-cancer vaccination by transdermal delivery of antigen peptide-loaded nanogels via iontophoresis. Int. J. Pharm. 2015, 483, 110–114. [Google Scholar] [CrossRef] [PubMed]
  70. Subongkot, T. Combined effect of sonophoresis and a microemulsion on the dermal delivery of celecoxib. Drug Deliv. 2020, 27, 1087–1093. [Google Scholar] [CrossRef]
  71. Akhtar, N.; Singh, V.; Yusuf, M.; Khan, R.A. Non-invasive drug delivery technology: Development and current status of transdermal drug delivery devices, techniques and biomedical applications. Biomed. Eng./Biomed. Tech. 2020, 65, 243–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Chen, X.; Zhu, L.; Li, R.; Pang, L.; Zhu, S.; Ma, J.; Du, L.; Jin, Y. Electroporation-enhanced transdermal drug delivery: Effects of logP, pKa, solubility and penetration time. Eur. J. Pharm. Sci. 2020, 151, 105410. [Google Scholar] [CrossRef] [PubMed]
  73. Bhattaccharjee, S.; Beck-Broichsitter, M.; Banga, A.K. In Situ Gel Formation in Microporated Skin for Enhanced Topical Delivery of Niacinamide. Pharmaceutics 2020, 12, 472. [Google Scholar] [CrossRef]
  74. Tadros, A.R.; Romanyuk, A.; Miller, I.C.; Santiago, A.; Noel, R.K.; O’Farrell, L.; Kwong, G.A.; Prausnitz, M.R. STAR particles for enhanced topical drug and vaccine delivery. Nat. Med. 2020, 26, 341–347. [Google Scholar] [CrossRef] [PubMed]
  75. McAllister, L.; Anderson, J.; Werth, K.; Cho, I.; Copeland, K.; Le Cam Bouveret, N.; Plant, D.; Mendelman, P.M.; Cobb, D.K. Needle-free jet injection for administration of influenza vaccine: A randomised non-inferiority trial. Lancet 2014, 384, 674–681. [Google Scholar] [CrossRef]
  76. Mittal, A.; Schulze, K.; Ebensen, T.; Weißmann, S.; Hansen, S.; Lehr, C.M.; Guzmán, C.A. Efficient nanoparticle-mediated needle-free transcutaneous vaccination via hair follicles requires adjuvantation. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 147–154. [Google Scholar] [CrossRef] [Green Version]
  77. Hashemi, V.; Farhadi, S.; Chaleshtari, M.G.; Seashore-Ludlow, B.; Masjedi, A.; Hojjat-Farsangi, M.; Namdar, A.; Ajjoolabady, A.; Mohammadi, H.; Ghalamfarsa, G. Nanomedicine for improvement of dendritic cell-based cancer immunotherapy. Int. Immunopharmacol. 2020, 83, 106446. [Google Scholar] [CrossRef]
  78. Crecente-Campo, J.; Lorenzo-Abalde, S.; Mora, A.; Marzoa, J.; Csaba, N.; Blanco, J.; González-Fernández, Á.; Alonso, M.J. Bilayer polymeric nanocapsules: A formulation approach for a thermostable and adjuvanted E. coli antigen vaccine. J. Control. Release 2018, 286, 20–32. [Google Scholar] [CrossRef]
  79. Jasim, A.; Abdelghany, S.; Greish, K. Current update on the role of enhanced permeability and retention effect in cancer nanomedicine. In Nanotechnology-Based Approaches for Targeting and Delivery of Drugs and Genes; Elsevier: Amsterdam, The Netherlands, 2017; pp. 62–109. [Google Scholar]
  80. Carabineiro, S.A.C. Applications of gold nanoparticles in nanomedicine: Recent advances in vaccines. Molecules 2017, 22, 857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Ahmed, K.S.; Shan, X.; Mao, J.; Qiu, L.; Chen, J. Derma roller® microneedles-mediated transdermal delivery of doxorubicin and celecoxib co-loaded liposomes for enhancing the anticancer effect. Mater. Sci. Eng. C 2019, 99, 1448–1458. [Google Scholar] [CrossRef]
  82. 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. 2016, 23, 2391–2398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Tyagi, R.K.; Garg, N.K.; Jadon, R.; Sahu, T.; Katare, O.P.; Dalai, S.K.; Awasthi, A.; Marepally, S.K. Elastic liposome-mediated transdermal immunization enhanced the immunogenicity of P-falciparum surface antigen, MSP-1(19). Vaccine 2015, 33, 4630–4638. [Google Scholar] [CrossRef] [PubMed]
  84. Tyagi, R.K.; Garg, N.K.; Dalai, S.K.; Awasthi, A. Transdermal immunization of P-falciparum surface antigen (MSP-1(19)) via elastic liposomes confers robust immunogenicity. Hum. Vaccines Immunother. 2016, 12, 990–992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Chen, Y.C.; Chen, S.J.; Cheng, H.F.; Yeh, M.K. Development of Yersinia pestis F1 antigen-loaded liposome vaccine against plague using microneedles as a delivery system. J. Drug Deliv. Sci. Technol. 2020, 55, 101443. [Google Scholar] [CrossRef]
  86. Mahor, S.; Rawat, A.; Dubey, P.K.; Gupta, P.N.; Khatri, K.; Goyal, A.K.; Vyas, S. Cationic transfersomes based topical genetic vaccine against hepatitis B. Int. J. Pharm. 2007, 340, 13–19. [Google Scholar] [CrossRef]
  87. Yang, X.X.; Wang, X.Y.; Hong, H.Y.; Elfawal, G.; Lin, S.; Wu, J.L.; Jiang, Y.X.; He, C.L.; Mo, X.M.; Kai, G.Y.; et al. Galactosylated chitosan-modified ethosomes combined with silk fibroin nanofibers is useful in transcutaneous immunization. J. Control. Release 2020, 327, 88–99. [Google Scholar] [CrossRef]
  88. Pamornpathomkul, B.; Niyomtham, N.; Yingyongnarongkul, B.-E.; Prasitpuriprecha, C.; Rojanarata, T.; Ngawhirunpat, T.; Opanasopit, P. Cationic niosomes for enhanced skin immunization of plasmid DNA-encoding ovalbumin via hollow microneedles. AAPS PharmSciTech 2018, 19, 481–488. [Google Scholar] [CrossRef] [PubMed]
  89. Rattanapak, T.; Young, K.; Rades, T.; Hook, S. Comparative study of liposomes, transfersomes, ethosomes and cubosomes for transcutaneous immunisation: Characterisation and in vitro skin penetration. J. Pharm. Pharmacol. 2012, 64, 1560–1569. [Google Scholar] [CrossRef] [PubMed]
  90. Rattanapak, T.; Birchall, J.; Young, K.; Ishii, M.; Meglinski, I.; Rades, T.; Hook, S. Transcutaneous immunization using microneedles and cubosomes: Mechanistic investigations using Optical Coherence Tomography and Two-Photon Microscopy. J. Control. Release 2013, 172, 894–903. [Google Scholar] [CrossRef]
  91. Peleteiro, M.; Presas, E.; González-Aramundiz, J.V.; Sánchez-Correa, B.; Simón-Vázquez, R.; Csaba, N.; Alonso, M.J.; González-Fernández, Á. Polymeric nanocapsules for vaccine delivery: Influence of the polymeric shell on the interaction with the immune system. Front. Immunol. 2018, 9, 791. [Google Scholar] [CrossRef]
  92. Boone, C.E.; Wang, C.; Lopez-Ramirez, M.A.; Beiss, V.; Shukla, S.; Chariou, P.L.; Kupor, D.; Rueda, R.; Wang, J.; Steinmetz, N.F. Active microneedle administration of plant virus nanoparticles for cancer in situ vaccination improves immunotherapeutic efficacy. ACS Appl. Nano Mater. 2020, 3, 8037–8051. [Google Scholar] [CrossRef]
  93. Mittal, A.; Schulze, K.; Ebensen, T.; Weissmann, S.; Hansen, S.; Guzmán, C.A.; Lehr, C.-M. Inverse micellar sugar glass (IMSG) nanoparticles for transfollicular vaccination. J. Control. Release 2015, 206, 140–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Takeuchi, I.; Suzuki, T.; Makino, K. Iontophoretic transdermal delivery using chitosan-coated PLGA nanoparticles for transcutaneous immunization. Colloids Surf. A Physicochem. Eng. Asp. 2021, 608, 125607. [Google Scholar] [CrossRef]
  95. Varypataki, E.M.; van der Maaden, K.; Bouwstra, J.; Ossendorp, F.; Jiskoot, W. Cationic liposomes loaded with a synthetic long peptide and poly (I: C): A defined adjuvanted vaccine for induction of antigen-specific T cell cytotoxicity. AAPS J. 2015, 17, 216–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Baldwin, S.L.; Reese, V.A.; Larsen, S.E.; Beebe, E.; Guderian, J.; Orr, M.T.; Fox, C.B.; Reed, S.G.; Coler, R.N. Prophylactic efficacy against Mycobacterium tuberculosis using ID93 and lipid-based adjuvant formulations in the mouse model. PLoS ONE 2021, 16, e0247990. [Google Scholar] [CrossRef]
  97. Coccia, M.; Collignon, C.; Hervé, C.; Chalon, A.; Welsby, I.; Detienne, S.; van Helden, M.J.; Dutta, S.; Genito, C.J.; Waters, N.C.; et al. Cellular and molecular synergy in AS01-adjuvanted vaccines results in an early IFNγ response promoting vaccine immunogenicity. NPJ Vaccines 2017, 2, 25. [Google Scholar] [CrossRef]
  98. Bhardwaj, P.; Tripathi, P.; Gupta, R.; Pandey, S. Niosomes: A review on niosomal research in the last decade. J. Drug Deliv. Sci. Technol. 2020, 56, 101581. [Google Scholar] [CrossRef]
  99. Perrie, Y.; Frederik, P.M.; Gregoriadis, G. Liposome-mediated DNA vaccination: The effect of vesicle composition. Vaccine 2001, 19, 3301–3310. [Google Scholar] [CrossRef]
  100. Abhyankar, M.M.; Orr, M.T.; Lin, S.; Suraju, M.O.; Simpson, A.; Blust, M.; Pham, T.; Guderian, J.A.; Tomai, M.A.; Elvecrog, J.; et al. Adjuvant composition and delivery route shape immune response quality and protective efficacy of a recombinant vaccine for Entamoeba histolytica. NPJ Vaccines 2018, 3, 22. [Google Scholar] [CrossRef] [Green Version]
  101. Saremi, S.S.; Shahryari, M.; Ghoorchian, R.; Eshaghian, H.; Jalali, S.A.; Nikpoor, A.R.; Jafari, M.R.; Badiee, A. The role of nanoliposome bilayer composition containing soluble leishmania antigen on maturation and activation of dendritic cells. Iran. J. Basic Med. Sci. 2018, 21, 536–545. [Google Scholar] [CrossRef] [PubMed]
  102. Yusuf, H.; Ali, A.A.; Orr, N.; Tunney, M.M.; McCarthy, H.O.; Kett, V.L. Novel freeze-dried DDA and TPGS liposomes are suitable for nasal delivery of vaccine. Int. J. Pharm. 2017, 533, 179–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Mansury, D.; Ghazvini, K.; Jamehdar, S.A.; Badiee, A.; Tafaghodi, M.; Nikpoor, A.R.; Amini, Y.; Jaafari, M.R. Enhancement of the effect of BCG vaccine against tuberculosis using DDA/TDB liposomes containing a fusion protein of HspX, PPE44, and EsxV. Artif. Cells Nanomed. Biotechnol. 2019, 47, 370–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Inoh, Y.; Nagai, M.; Matsushita, K.; Nakanishi, M.; Furuno, T. Gene transfection efficiency into dendritic cells is influenced by the size of cationic liposomes/DNA complexes. Eur. J. Pharm. Sci. 2017, 102, 230–236. [Google Scholar] [CrossRef] [PubMed]
  105. Huang, H.; Zhang, C.L.; Yang, S.P.; Xiao, W.; Zheng, Q.; Song, X.R. The investigation of mRNA vaccines formulated in liposomes administrated in multiple routes against SARS-CoV-2. J. Control. Release 2021, 335, 449–456. [Google Scholar] [CrossRef]
  106. Lanza, J.S.; Vucen, S.; Flynn, O.; Donadei, A.; Cojean, S.; Loiseau, P.M.; Fernandes, A.; Frezard, F.; Moore, A.C. A TLR9-adjuvanted vaccine formulated into dissolvable microneedle patches or cationic liposomes protects against leishmaniasis after skin or subcutaneous immunization. Int. J. Pharm. 2020, 586, 119390. [Google Scholar] [CrossRef]
  107. Gupta, P.N.; Mishra, V.; Rawat, A.; Dubey, P.; Mahor, S.; Jain, S.; Chatterji, D.P.; Vyas, S.P. Non-invasive vaccine delivery in transfersomes, niosomes and liposomes: A comparative study. Int. J. Pharm. 2005, 293, 73–82. [Google Scholar] [CrossRef]
  108. Mann, J.L.; Maikawa, C.L.; Smith, A.A.; Grosskopf, A.K.; Baker, S.W.; Roth, G.A.; Meis, C.M.; Gale, E.C.; Liong, C.S.; Correa, S. An ultrafast insulin formulation enabled by high-throughput screening of engineered polymeric excipients. Sci. Transl. Med. 2020, 12, 550. [Google Scholar] [CrossRef]
  109. Rai, S.; Pandey, V.; Rai, G. Transfersomes as versatile and flexible nano-vesicular carriers in skin cancer therapy: The state of the art. Nano Rev. Exp. 2017, 8, 1325708. [Google Scholar] [CrossRef]
  110. Opatha, S.A.T.; Titapiwatanakun, V.; Chutoprapat, R. Transfersomes: A promising nanoencapsulation technique for transdermal drug delivery. Pharmaceutics 2020, 12, 855. [Google Scholar] [CrossRef]
  111. El Zaafarany, G.M.; Awad, G.A.; Holayel, S.M.; Mortada, N.D. Role of edge activators and surface charge in developing ultradeformable vesicles with enhanced skin delivery. Int. J. Pharm. 2010, 397, 164–172. [Google Scholar] [CrossRef] [PubMed]
  112. Schätzlein, A.; Cevc, G. Non-uniform cellular packing of the stratum corneum and permeability barrier function of intact skin: A high-resolution confocal laser scanning microscopy study using highly deformable vesicles (Transfersomes). Br. J. Dermatol. 1998, 138, 583–592. [Google Scholar] [CrossRef]
  113. Gupta, P.N.; Mishra, V.; Singh, P.; Rawat, A.; Dubey, P.; Mahor, S.; Vyas, S.P. Tetanus toxoid-loaded transfersomes for topical immunization. J. Pharm. Pharmacol. 2005, 57, 295–301. [Google Scholar] [CrossRef]
  114. Grit, M.; Crommelin, D.J. Chemical stability of liposomes: Implications for their physical stability. Chem. Phys. Lipids 1993, 64, 3–18. [Google Scholar] [CrossRef]
  115. Van Hoogevest, P.; Wendel, A. The use of natural and synthetic phospholipids as pharmaceutical excipients. Eur. J. Lipid Sci. Technol. 2014, 116, 1088–1107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Zhang, Y.; Xia, Q.; Li, Y.; He, Z.; Li, Z.; Guo, T.; Wu, Z.; Feng, N. CD44 assists the topical anti-psoriatic efficacy of curcumin-loaded hyaluronan-modified ethosomes: A new strategy for clustering drug in inflammatory skin. Theranostics 2019, 9, 48. [Google Scholar] [CrossRef]
  117. Dhawan, S.; Sharma, P.; Nanda, S. Nanosized Labile and Particulate Ingredients in Topical Formulations: A Strategic Approach Against Photoageing and Photocarcinogenesis. In Nanoformulations in Human Health; Springer: Berlin/Heidelberg, Germany, 2020; pp. 287–308. [Google Scholar]
  118. Goyal, R.; Macri, L.K.; Kaplan, H.M.; Kohn, J. Nanoparticles and nanofibers for topical drug delivery. J. Control. Release 2016, 240, 77–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Yang, L.; Wu, L.; Wu, D.; Shi, D.; Wang, T.; Zhu, X. Mechanism of transdermal permeation promotion of lipophilic drugs by ethosomes. Int. J. Nanomed. 2017, 12, 3357–3364. [Google Scholar] [CrossRef] [Green Version]
  120. Paiva-Santos, A.C.; Silva, A.L.; Guerra, C.; Peixoto, D.; Pereira-Silva, M.; Zeinali, M.; Mascarenhas-Melo, F.; Castro, R.; Veiga, F. Ethosomes as Nanocarriers for the Development of Skin Delivery Formulations. Pharm. Res. 2021, 38, 947–970. [Google Scholar] [CrossRef] [PubMed]
  121. Xie, J.; Ji, Y.; Xue, W.; Ma, D.; Hu, Y.F. Hyaluronic acid-containing ethosomes as a potential carrier for transdermal drug delivery. Colloids Surf. B Biointerfaces 2018, 172, 323–329. [Google Scholar] [CrossRef]
  122. Zhang, Y.; Ng, W.; Hu, J.; Mussa, S.S.; Ge, Y.; Xu, H. Formulation and in vitro stability evaluation of ethosomal carbomer hydrogel for transdermal vaccine delivery. Colloids Surf. B Biointerfaces 2018, 163, 184–191. [Google Scholar] [CrossRef]
  123. Marianecci, C.; Di Marzio, L.; Rinaldi, F.; Celia, C.; Paolino, D.; Alhaique, F.; Esposito, S.; Carafa, M. Niosomes from 80s to present: The state of the art. Adv. Colloid Interface Sci. 2014, 205, 187–206. [Google Scholar] [CrossRef] [PubMed]
  124. Ibrahim, S.A.; Li, S.K. Efficiency of fatty acids as chemical penetration enhancers: Mechanisms and structure enhancement relationship. Pharm. Res. 2010, 27, 115–125. [Google Scholar] [CrossRef] [Green Version]
  125. Okore, V.C.; Attama, A.A.; Ofokansi, K.C.; Esimone, C.O.; Onuigbo, E.B. Formulation and evaluation of niosomes. Indian J. Pharm. Sci. 2011, 73, 323–328. [Google Scholar] [CrossRef] [PubMed]
  126. Kopečná, M.; Macháček, M.; Nováčková, A.; Paraskevopoulos, G.; Roh, J.; Vávrová, K. Esters of terpene alcohols as highly potent, reversible, and low toxic skin penetration enhancers. Sci. Rep. 2019, 9, 14617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Patrick, T.S. Cubosomes: Bicontinuous Liquid Crystalline Nanoparticles; CRC Press: Boca Raton, FL, USA, 2009. [Google Scholar]
  128. Montis, C.; Castroflorio, B.; Mendozza, M.; Salvatore, A.; Berti, D.; Baglioni, P. Magnetocubosomes for the delivery and controlled release of therapeutics. J. Colloid Interface Sci. 2015, 449, 317–326. [Google Scholar] [CrossRef]
  129. Kojarunchitt, T.; Hook, S.; Rizwan, S.; Rades, T.; Baldursdottir, S. Development and characterisation of modified poloxamer 407 thermoresponsive depot systems containing cubosomes. Int. J. Pharm. 2011, 408, 20–26. [Google Scholar] [CrossRef] [PubMed]
  130. Um, J.Y.; Chung, H.; Kim, K.S.; Kwon, I.C.; Jeong, S.Y. In vitro cellular interaction and absorption of dispersed cubic particles. Int. J. Pharm. 2003, 253, 71–80. [Google Scholar] [CrossRef]
  131. Gupta, A.; de Campo, L.; Rehmanjan, B.; Willis, S.A.; Waddington, L.J.; Stait-Gardner, T.; Kirby, N.; Price, W.S.; Moghaddam, M.J. Evaluation of Gd-DTPA-monophytanyl and phytantriol nanoassemblies as potential MRI contrast agents. Langmuir 2015, 31, 1556–1563. [Google Scholar] [CrossRef]
  132. Karami, Z.; Hamidi, M. Cubosomes: Remarkable drug delivery potential. Drug Discov. Today 2016, 21, 789–801. [Google Scholar] [CrossRef]
  133. Bolhassani, A.; Javanzad, S.; Saleh, T.; Hashemi, M.; Aghasadeghi, M.R.; Sadat, S.M. Polymeric nanoparticles: Potent vectors for vaccine delivery targeting cancer and infectious diseases. Hum. Vaccines Immunother. 2014, 10, 321–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. González-Aramundiz, J.V.; Olmedo, M.P.; González-Fernández, Á.; Fernández, M.J.A.; Csaba, N.S. Protamine-based nanoparticles as new antigen delivery systems. Eur. J. Pharm. Biopharm. 2015, 97, 51–59. [Google Scholar] [CrossRef]
  135. Alfagih, I.M.; Aldosari, B.; AlQuadeib, B.; Almurshedi, A.; Alfagih, M.M. Nanoparticles as Adjuvants and Nanodelivery Systems for mRNA-Based Vaccines. Pharmaceutics 2020, 13, 45. [Google Scholar] [CrossRef] [PubMed]
  136. Verbeke, R.; Lentacker, I.; De Smedt, S.C.; Dewitte, H. The dawn of mRNA vaccines: The COVID-19 case. J. Control. Release 2021, 333, 511–520. [Google Scholar] [CrossRef] [PubMed]
  137. Chagla, Z. The BNT162b2 (BioNTech/Pfizer) vaccine had 95% efficacy against COVID-19 ≥7 days after the 2nd dose. Ann. Intern. Med. 2021, 174, JC15. [Google Scholar] [CrossRef]
  138. Mahase, E. COVID-19: Moderna applies for US and EU approval as vaccine trial reports 94.1% efficacy. BMJ Br. Med. J. (Online) 2020, 371, m4709. [Google Scholar] [CrossRef]
  139. Tammam, S.N.; El Safy, S.; Ramadan, S.; Arjune, S.; Krakor, E.; Mathur, S. Repurpose but also (nano)-reformulate! The potential role of nanomedicine in the battle against SARS-CoV2. J. Control Release 2021, 337, 258–284. [Google Scholar] [CrossRef]
  140. Vicente, S.; Peleteiro, M.; Gonzalez-Aramundiz, J.V.; Díaz-Freitas, B.; Martínez-Pulgarín, S.; Neissa, J.I.; Escribano, J.M.; Sanchez, A.; González-Fernández, Á.; Alonso, M.J. Highly versatile immunostimulating nanocapsules for specific immune potentiation. Nanomedicine 2014, 9, 2273–2289. [Google Scholar] [CrossRef] [PubMed]
  141. Bussio, J.I.; Molina-Perea, C.; González-Aramundiz, J.V. Hyaluronic acid nanocapsules as a platform for needle-free vaccination. Pharmaceutics 2019, 11, 246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Su, R.; Fan, W.; Yu, Q.; Dong, X.; Qi, J.; Zhu, Q.; Zhao, W.; Wu, W.; Chen, Z.; Li, Y. Size-dependent penetration of nanoemulsions into epidermis and hair follicles: Implications for transdermal delivery and immunization. Oncotarget 2017, 8, 38214. [Google Scholar] [CrossRef] [Green Version]
  143. Lopez, P.A.; Denny, M.; Hartmann, A.-K.; Alflen, A.; Probst, H.C.; von Stebut, E.; Tenzer, S.; Schild, H.; Stassen, M.; Langguth, P.; et al. Transcutaneous immunization with a novel imiquimod nanoemulsion induces superior T cell responses and virus protection. J. Dermatol. Sci. 2017, 87, 252–259. [Google Scholar] [CrossRef] [Green Version]
  144. Alvarez-Figueroa, M.J.; Abarca-Riquelme, J.M.; González-Aramundiz, J.V. Influence of protamine shell on nanoemulsions as a carrier for cyclosporine-A skin delivery. Pharm. Dev. Technol. 2019, 24, 630–638. [Google Scholar] [CrossRef]
  145. Yu, Q.; Huang, Y.; Zhu, C.; Wu, X.; Tai, Z.; Xie, X.; Qi, J.; Wu, W.; Chen, Z.; Lu, Y. Combination of Microneedles and MF59 Adjuvant as a Simple Approach to Enhance Transcutaneous Immunization. J. Biomed. Nanotechnol. 2020, 16, 1776–1786. [Google Scholar] [CrossRef] [PubMed]
  146. Tayeb, H.H.; Felimban, R.; Almaghrabi, S.; Hasaballah, N. Nanoemulsions: Formulation, characterization, biological fate, and potential role against COVID-19 and other viral outbreaks. Colloid Interface Sci. Commun. 2021, 45, 100533. [Google Scholar] [CrossRef]
  147. Quan, P.; Jiao, B.; Shang, R.; Liu, C.; Fang, L. Alternative therapy of rheumatoid arthritis with a novel transdermal patch containing Siegesbeckiae Herba extract. J. Ethnopharmacol. 2021, 265, 113294. [Google Scholar] [CrossRef]
  148. Liu, P.; Cettina, M.; Wong, J. Effects of isopropanol–isopropyl myristate binary enhancers on in vitro transport of estradiol in human epidermis: A mechanistic evaluation. J. Pharm. Sci. 2009, 98, 565–572. [Google Scholar] [CrossRef] [PubMed]
  149. Bommannan, D.; Potts, R.O.; Guy, R.H. Examination of the effect of ethanol on human stratum corneum in vivo using infrared spectroscopy. J. Control. Release 1991, 16, 299–304. [Google Scholar] [CrossRef]
  150. Natsheh, H.; Touitou, E. Phospholipid Vesicles for Dermal/Transdermal and Nasal Administration of Active Molecules: The Effect of Surfactants and Alcohols on the Fluidity of Their Lipid Bilayers and Penetration Enhancement Properties. Molecules 2020, 25, 2959. [Google Scholar] [CrossRef] [PubMed]
  151. Pelliccia, M.; Andreozzi, P.; Paulose, J.; D’Alicarnasso, M.; Cagno, V.; Donalisio, M.; Civra, A.; Broeckel, R.M.; Haese, N.; Silva, P.J. Additives for vaccine storage to improve thermal stability of adenoviruses from hours to months. Nat. Commun. 2016, 7, 13520. [Google Scholar] [CrossRef]
  152. Cordeiro, A.S.; Alonso, M.J.; de la Fuente, M. Nanoengineering of vaccines using natural polysaccharides. Biotechnol. Adv. 2015, 33, 1279–1293. [Google Scholar] [CrossRef] [PubMed]
  153. Yang, H.W.; Ye, L.; Guo, X.D.; Yang, C.; Compans, R.W.; Prausnitz, M.R. Ebola Vaccination Using a DNA Vaccine Coated on PLGA-PLL/γPGA Nanoparticles Administered Using a Microneedle Patch. Adv. Healthc. Mater. 2017, 6, 1600750. [Google Scholar] [CrossRef] [PubMed]
  154. Mahase, E. COVID-19: Pfizer vaccine efficacy was 52% after first dose and 95% after second dose, paper shows. BMJ 2020, 371, m4826. [Google Scholar] [CrossRef]
  155. Brosh-Nissimov, T.; Orenbuch-Harroch, E.; Chowers, M.; Elbaz, M.; Nesher, L.; Stein, M.; Maor, Y.; Cohen, R.; Hussein, K.; Weinberger, M. BNT162b2 vaccine breakthrough: Clinical characteristics of 152 fully vaccinated hospitalized COVID-19 patients in Israel. Clin. Microbiol. Infect. 2021, 27, 1652–1657. [Google Scholar] [CrossRef]
  156. Grupper, A.; Sharon, N.; Finn, T.; Cohen, R.; Israel, M.; Agbaria, A.; Rechavi, Y.; Schwartz, I.F.; Schwartz, D.; Lellouch, Y. Humoral response to the Pfizer BNT162b2 vaccine in patients undergoing maintenance hemodialysis. Clin. J. Am. Soc. Nephrol. 2021, 16, 1037–1042. [Google Scholar] [CrossRef] [PubMed]
  157. Pilishvili, T.; Fleming-Dutra, K.E.; Farrar, J.L.; Gierke, R.; Mohr, N.M.; Talan, D.A.; Krishnadasan, A.; Harland, K.K.; Smithline, H.A.; Hou, P.C. Interim Estimates of Vaccine Effectiveness of Pfizer-BioNTech and Moderna COVID-19 Vaccines Among Health Care Personnel—33 US Sites, January–March 2021. Morb. Mortal. Wkly. Rep. 2021, 70, 753. [Google Scholar] [CrossRef] [PubMed]
  158. Crecente-Campo, J.; Alonso, M.J. Engineering, on-demand manufacturing, and scaling-up of polymeric nanocapsules. Bioeng. Transl. Med. 2019, 4, 38–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Page, A.; Fusil, F.; Cosset, F.-L. Towards physiologically and tightly regulated vectored antibody therapies. Cancers 2020, 12, 962. [Google Scholar] [CrossRef] [Green Version]
  160. Pippa, N.; Gazouli, M.; Pispas, S. Recent Advances and Future Perspectives in Polymer-Based Nanovaccines. Vaccines 2021, 9, 558. [Google Scholar] [CrossRef]
  161. Uskoković, V. Nanomedicine for the poor: A lost cause or an idea whose time has yet to come? Nanomedicine 2021, 16, 1203–1218. [Google Scholar] [CrossRef]
  162. Ferreira, S.C.; Bruns, R.; Ferreira, H.; Matos, G.; David, J.; Brandão, G.; da Silva, E.P.; Portugal, L.; Dos Reis, P.; Souza, A. Box-Behnken design: An alternative for the optimization of analytical methods. Anal. Chim. Acta 2007, 597, 179–186. [Google Scholar] [CrossRef] [PubMed]
  163. Arnouts, H.; Goos, P. Staggered-level designs for experiments with more than one hard-to-change factor. Technometrics 2012, 54, 355–366. [Google Scholar] [CrossRef]
  164. Baghaei, B.; Saeb, M.R.; Jafari, S.H.; Khonakdar, H.A.; Rezaee, B.; Goodarzi, V.; Mohammadi, Y. Modeling and closed-loop control of particle size and initial burst of PLGA biodegradable nanoparticles for targeted drug delivery. J. Appl. Polym. Sci. 2017, 134, 45145. [Google Scholar] [CrossRef]
  165. Dorigatti, I.; Donnelly, C.; Laydon, D.; Small, R.; Jackson, N.; Coudeville, L.; Ferguson, N. Refined efficacy estimates of the Sanofi Pasteur dengue vaccine CYD-TDV using machine learning. Nat. Commun. 2018, 9, 3644. [Google Scholar] [CrossRef] [PubMed]
  166. Chaudhury, S.; Duncan, E.H.; Atre, T.; Storme, C.K.; Beck, K.; Kaba, S.A.; Lanar, D.E.; Bergmann-Leitner, E.S. Identification of immune signatures of novel adjuvant formulations using machine learning. Sci. Rep. 2018, 8, 17508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Cortés-Ríos, J.; Valdivia-Olivares, R.; Álvarez-Figueroa, M.; Rodriguez-Fernandez, M.; González-Aramundiz, J. Optimization of physicochemical properties of novel multiple nanoemulsion for complex food matrices through iterative mathematical modelling. J. Food Eng. 2020, 276, 109883. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the routes of skin penetration of active compounds. On the left, a transpedicular route consists of a. entry through hair follicle, b. entry through sweat glands, c. entry through sebaceous glands. On the right, transepidermal route. d. Transcellular pathway, e. Intercellular pathway.
Figure 1. Schematic representation of the routes of skin penetration of active compounds. On the left, a transpedicular route consists of a. entry through hair follicle, b. entry through sweat glands, c. entry through sebaceous glands. On the right, transepidermal route. d. Transcellular pathway, e. Intercellular pathway.
Vaccines 09 01420 g001
Figure 2. Advantages of transdermal vaccine delivery. Created with BioRender.com (accessed on 27 October 2021).
Figure 2. Advantages of transdermal vaccine delivery. Created with BioRender.com (accessed on 27 October 2021).
Vaccines 09 01420 g002
Figure 3. Schematic representation of the mechanisms involved in immunization based on nanoparticles, either using combined techniques or design of nanoparticles by passive diffusion. Once the stratum corneum has been crossed, the antigens can interact with cells of the immune system already described.
Figure 3. Schematic representation of the mechanisms involved in immunization based on nanoparticles, either using combined techniques or design of nanoparticles by passive diffusion. Once the stratum corneum has been crossed, the antigens can interact with cells of the immune system already described.
Vaccines 09 01420 g003
Figure 4. Main advantages of the use of nanocomposites for the delivery of antigens and the design of nanovaccines. Created with BioRender.com (accessed on 27 October 2021).
Figure 4. Main advantages of the use of nanocomposites for the delivery of antigens and the design of nanovaccines. Created with BioRender.com (accessed on 27 October 2021).
Vaccines 09 01420 g004
Table 1. Transdermal nanovaccine designs for various antigens using novel nanosystems and their immune response from in vivo assays.
Table 1. Transdermal nanovaccine designs for various antigens using novel nanosystems and their immune response from in vivo assays.
AntigenNanosystem DesignImmune Response In Vivo Assays
OvalbuminLiposomes, transferosomes and etosomes formulated using the reverse phase evaporation method.In female BALB/c mice using colloidal Al (OH) 3 as adjuvant, an antiova antibody titer was obtained higher than the other nanosystems, compared to the non-encapsulated control [60].
Phytoglycogen (PG) nanoparticles conjugated to form Nano-11 adjuvant particles with and without cyclic di-AMP, administered with Pharmajet.The compound combining both adjuvants demonstrated a synergistic immune response that resulted in increased production of Abs IgG1 and IgG2a, as well as CD8 T lymphocytes expressing Th1, Th17 and IFN-γ in mice and pigs [61].
Homolog 5 of Plasmodium falciparum reticulocyte-binding protein (PfRH5) and coding sequence of small hepatitis B virus envelope (HBs) antigenTattoo Cationic liposomes fused with VHP antigen, expressing on their surface (PfRH5) formulated from dimethyldioctadecylammonium bromide) and DC-cholesterol by solvent evaporation.A strong humoral response against PfRH5 in malaria vaccines was demonstrated in mice in those with fused tattoo, superior to the non-fused control PfRH5 and to intraperitoneal administration [62].
Diphtheria toxoid (DT)Mesoporous Silica Nanoparticles (MSN) Embedded in Coated, Hollow MicroneedlesThe DT encapsulated in MSN induced a stronger antibody response than the antigen solution when administered by hollow microneedles in BALB/c mice, it is shown that the type of encapsulation and microneedle affect the response [63].
HIV-1P24-Nef peptidePGLA nanoparticles with the sequence of the flagellin molecule from Pseudomonas aeruginosa as TLR5 activating adjuvant.The formulation is shown in mice to improve immunogenicity and reduce the dose [64].
Antigen DNA, based on protein N or S from SARS-CoV-2 virusesLipidoid nanoparticles composed of low molecular weight polyethyleneimines conjugated with deoxycholic acid loaded with the adjuvant Resiquimod in separable microneedles.The authors show in female C57BL/6 mice that the intradermal vaccine is capable of inducing an enhanced and lasting immune response compared to the intramuscular route, the formulation can be kept at room temperature for at least 30 days [65].
Influenza Neuraminidase and Flagellin ProteinInfluenza 2 matrix protein ectodomain (M2e) nanoparticles (M2) by ethanol desolvation and double-layered protein nanoparticles, incorporated in soluble microneedles.The nanovaccine was able to significantly increase the levels of specific antibodies and protect the mice from infection [66].
Table 2. Main Nanocarriers used in transdermal immunization.
Table 2. Main Nanocarriers used in transdermal immunization.
NanosystemApplication in ImmunizationChallengesReferences
LiposomesMicroneedles combined with liposomes co-loaded with doxorubicin HCl (DOX) and celecoxib (CEL)/cationic liposomes encapsulated with hepatitis B DNA vaccine and adjuvant CpG ODN.Conducting clinical trials, limitations associated with the coupled use of microneedles.[81,82]
Liposomes loaded with the surface antigen of P-falciparum MSP-1[83,84]
Yersinia pestis F-1 antigen-loaded liposomes using microneedles[85]
TransferosomesCationic transferosomes composed of cationic lipid DOTMA and sodium deoxycholate.Deficiency of consistent results that validate increased transdermal permeability.[60,86]
EthosomesHyaluronic acid (HA) and galactosylated chitosan (GC) modified ethosome (Eth-HA-GC) loaded ovalbumin.Evaluation of safety and efficacy using other antigens, application suggested by authors in oncology[87]
High ethanolic content can be a “double-edged sword”, producing high drug entrapment, but also large leakage.
NiosomesCationic niosomes loaded with ovalbumin combined with hollow microneedle.Dependence of association with microneedles.[88]
CubosomesCubosomes that encapsulate adjuvants Quil A and monophosphoryl lipid.Ability to cross the stratum corneum by passive diffusion, compatibility to encapsulate antigens and adjuvants in sets still under study.[89]
Cubosomes to transport antigens combined with microneedles[90]
Polimeric NanocapsulesProtamine and polyarginine nanocapsules in association with the recombinant hepatitis B surface antigen.Incorporation of adjuvant molecules to obtain an improved immune response.
Nanocapsules of a vitamin E oily core, surrounded by two layers: a first layer of chitosan and a second of dextran sulphate, antigen, IutA protein from Escherichia coli[91]
Autonomous active microneedle for the direct intratumoral delivery of an immunoadjuvant, cowpea mosaic virus nanoparticles (CPMV).[92,93,94]
Chitosan-coated PLGA nanoparticles
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Valdivia-Olivares, R.Y.; Rodriguez-Fernandez, M.; Álvarez-Figueroa, M.J.; Kalergis, A.M.; González-Aramundiz, J.V. The Importance of Nanocarrier Design and Composition for an Efficient Nanoparticle-Mediated Transdermal Vaccination. Vaccines 2021, 9, 1420. https://doi.org/10.3390/vaccines9121420

AMA Style

Valdivia-Olivares RY, Rodriguez-Fernandez M, Álvarez-Figueroa MJ, Kalergis AM, González-Aramundiz JV. The Importance of Nanocarrier Design and Composition for an Efficient Nanoparticle-Mediated Transdermal Vaccination. Vaccines. 2021; 9(12):1420. https://doi.org/10.3390/vaccines9121420

Chicago/Turabian Style

Valdivia-Olivares, Rayen Yanara, Maria Rodriguez-Fernandez, María Javiera Álvarez-Figueroa, Alexis M. Kalergis, and José Vicente González-Aramundiz. 2021. "The Importance of Nanocarrier Design and Composition for an Efficient Nanoparticle-Mediated Transdermal Vaccination" Vaccines 9, no. 12: 1420. https://doi.org/10.3390/vaccines9121420

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop