15 March 2005

Use of Articles in the Bibliography

The articles in the PC Bibliography may be restricted by copyright laws. These have been made available to you by PC Project for the exclusive use in teaching, scholar- ship or research regarding Pachyonychia Congenita.

To the best of our understanding, in supplying this material to you we have followed the guidelines of Sec 107 regarding fair use of copyright materials. That section reads as follows:

Sec. 107. - Limitations on exclusive rights: Fair use Notwithstanding the provisions of sections 106 and 106A, the fair use of a copyrighted work, including such use by reproduction in copies or phonorecords or by any other means specified by that section, for purposes such as criticism, comment, news reporting, teaching (including multiple copies for classroom use), scholarship, or research, is not an infringement of copyright. In determining whether the use made of a work in any particular case is a fair use the factors to be considered shall include - (1) the purpose and character of the use, including whether such use is of a commercial nature or is for nonprofit educational purposes; (2) the nature of the copyrighted work; (3) the amount and substantiality of the portion used in relation to the copyrighted work as a whole; and (4) the effect of the use upon the potential market for or value of the copyrighted work. The fact that a work is unpublished shall not itself bar a finding of fair use if such finding is made upon consideration of all the above factors.

We hope that making available the relevant information on Pachyonychia Congenita will be a means of furthering research to find effective therapies and a cure for PC.

2386 East Heritage Way, Suite B, Salt Lake City, Utah 84109 USA Phone +1-877-628-7300 • Email—[email protected] www.pachyonychia.org COREL-07857; No of Pages 12 Journal of Controlled Release xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Journal of Controlled Release

journal homepage: www.elsevier.com/locate/jconrel

Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges

Michael Zakrewsky, Sunny Kumar, Samir Mitragotri ⁎

Center for Bioengineering and Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA article info abstract

Article history: Nucleic acids (NAs) hold significant potential for the treatment of several diseases. Topical delivery of NAs for the Received 10 June 2015 treatment of skin diseases is especially advantageous since it bypasses the challenges associated with systemic Received in revised form 7 September 2015 administration which suffers from enzymatic degradation, systemic toxicity and lack of targeting to skin. Howev- Accepted 9 September 2015 er, the skin's protective barrier function limits the delivery of NAs into skin after topical application. Here, we Available online xxxx highlight strategies for enhancing delivery of NAs into skin, and provide evidence that translation of topical NA

Keywords: therapies could have a transformative impact on the treatment of skin diseases. Nucleic acids © 2015 Elsevier B.V. All rights reserved. Dermal drug delivery Skin disease

1. Introduction applied drugs. Nevertheless, significant efforts have been expended over the years to overcome the skin barrier. This review highlights strat- Nucleic acids (NAs) hold great potential for the treatment of various egies to effectively deliver NAs into the skin. Focus is placed on their ap- diseases, and there has been a significant amount of both academic plications for the treatment of dermatological diseases. Further, we as well as commercial interest in a variety of NA-based therapeutics provide evidence of the transformative impact of topical NA therapies including genes, antisense oligodeoxynucleotides (ODNs), siRNA, on the treatment of skin diseases. The efforts here highlight significant aptamers, and CpG oligonucleotides [1–4]. However, translation of advances in topical delivery of NAs over the last decade, and aim to these platforms to the clinic has been significantly limited by challenges guide future technologies and their translation into the clinic. associated with delivering NAs to the diseased site. Enzymatic degrada- tion in the blood, rapid clearance from systemic circulation, poor bio- 2. The skin barrier availability at the target site, and immunological response have yet to be adequately addressed for the implementation of systemically admin- The outermost layer of skin, the SC, is primarily responsible for istered NA therapies [1,4]. its barrier function. The SC is a thin layer only 10–20 μmthickthat Delivery of NAs through the skin offers a potential solution to these is made up of corneocytes. Corneocytes are anucleate cells heavily issues, especially for the treatment of dermatological diseases (Table 1). enriched with intracellular keratin filaments. Corneocytes are held to- The skin is the largest organ of the body, and can provide a pain free and gether in a “brick and mortar” structure by a lipid matrix composed of compliant interface for drug delivery [5,6]. Topical delivery of NAs offers ceramides, free fatty acids, and cholesterol. Materials traversing the several advantages over alternative delivery routes including avoidance skin barrier must, therefore, diffuse through the tortuous lipid channels, of major degradative pathways in the GI tract, avoidance of enzymatic and/or traverse transcellularly through corneocytes, or enter the skin degradation and clearance from the bloodstream, sustained and con- through hair follicles or sweat ducts (Fig. 1). Transport within the lipid trolled delivery, reduction in systemic toxicity, the ability to easily bilayers, however, is the most common mode of passage through the observe and treat sites of adverse reactions, and improved patient com- skin. This results in the exclusion of most foreign materials, and more pliance [6]. Concurrently, it provides a means to directly target the dis- specifically, renders passage of large, hydrophilic molecules (N500 Da eased sites for the treatment of dermatological diseases e.g. skin cancer, and Log Po/w b 1.5) such as NAs (typically NN10,000 Da, Log Po/w b 0) psoriasis, and . to virtually negligible levels without some form of enhancement Topical delivery of NAs, however, is quite challenging. The challenge strategy. originates from the outermost layer of the skin – the The layer underlying the SC, the epidermis, can also serve as (SC) – which serves as a formidable barrier to the entry of topically another transport barrier [7]. The epidermis is the first viable tissue layer of the skin where the pathology of several dermatological disor- – μ ⁎ Corresponding author. ders resides. The epidermis is 50 100 m thick and is composed primar- E-mail address: [email protected] (S. Mitragotri). ily of keratinocytes. As keratinocytes migrate upward from deeper

http://dx.doi.org/10.1016/j.jconrel.2015.09.017 0168-3659/© 2015 Elsevier B.V. All rights reserved.

Please cite this article as: M. Zakrewsky, et al., Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges, J. Control. Release (2015), http://dx.doi.org/10.1016/j.jconrel.2015.09.017 2 M. Zakrewsky et al. / Journal of Controlled Release xxx (2015) xxx–xxx

Table 1 overwhelming. Needle-phobia is a serious concern for a large number Advantages of NA topical delivery for the treatment of skin disease. of children as well as adults leading to significant patient non- Advantages of NA topical delivery compliance [9]. Moreover, intradermal injections are limited only to the site of application, and injection into multiple sites during a single • Targeted delivery into skin • Large surface area for drug application administration is challenging. To avoid many of these drawbacks, • Easily observe and excise sites of adverse reaction microneedle arrays have been developed. Microneedle arrays comprise • Easily monitor disease progress and adjust treatment needles that are only 100–700 μminlength(Fig. 2). When placed on the • Needle-free application skin, their sharp tips allow easy insertion into the stratum corneum, • Sustained and controlled delivery • Avoidance of GI tract while the short length ensures adequate penetration into the skin • Limited to negligible systemic toxicity without disrupting nerves in deeper skin tissue. Microneedles have • Limited to negligible clearance from diseased tissue been used extensively for the delivery of NAs. Mikszta et al. used • User-friendly application microneedle arrays to deliver plasmid DNA encoding a hepatitis B sur- face antigen for immunization [10], and they showed extensive immu- nological response in mice. Antibody titers following application of portions of the epidermis to the SC they gradually begin to keratinize microneedle arrays were significantly higher and less variable than and secrete lipids that eventually form the SC bilayers. This process con- when delivered using either intradermal or intramuscular injection. tinues as keratinocytes terminally differentiate into corneocytes and Chabri et al. [11] used microneedles to deliver cationic lipid-DNA com- serves to rejuvenate the SC from underneath while the outermost plexes (~100 nm diameter) into the skin. Ding et al. [12] demonstrated layer of the SC sloughs away. Within the epidermis, keratinocytes are successful immunization of mice with co-administration of diphtheria held together by cell–cell tight junctions. In the epidermis, claudin-1, toxoid and CpG oligonucleotide delivered by microneedle array, and claudin-4, occludin, and zonula occludens-1 are responsible for inhib- Gonzalez-Gonzalez et al. [13] demonstrated effective delivery of anti- iting paracellular transport [8]. This makes transport of NAs, other luciferase siRNA and gene silencing in luciferase expressing transgenic large macromolecules, and drug carriers such as nanoparticles and mice. NA–lipid complexes difficult both vertically, deeper into the skin, as well as laterally from the site of administration to peripheral areas of 3.1.2. Microporation the skin. For effective treatment, both the SC as well as epidermal trans- Microporation is another technique that employs physical dis- port barriers must be overcome to deliver NAs to all areas of the disease. ruption of the SC for delivery of large therapeutics or therapeutic carriers. An array of resistive elements can be placed on the skin. An 3. Methods of transport enhancement electric current pulsed through the array results in localized abla- tion of corneocytes in contact with the array [14]. Alternatively, Over the years, a large number of strategies have been devised for erbium:yttrium–aluminum–garnet (Er: YAG) laser arrays can be used perturbing the SC to enhance the delivery of drugs into and through for localized ablation of the SC and epidermis [15]. Similar to micro- the skin. These strategies can be generally categorized into three main needle arrays, microporation has gained considerable interest over the groups: physical, active, and passive methods. The advantages and dis- last decade. For example, Lee et al. [16] used laser microporation to de- advantages of each class of perturbation methods are summarized in liver antisense oligonucleotide as well as plasmid DNA into the skin. De- Table 2. Their use for the delivery of NAs is described below. livery of antisense oligonucleotide was enhanced 3–30 fold compared to intact skin in vitro. In addition, expression of GFP in nude mice was en- 3.1. NA delivery using physical methods hanced 160 fold after application of GFP plasmid DNA. The amount of enhancementcorrelatedwithboththelaserfluency as well as the size 3.1.1. Microneedles of oligonucleotide. The same group also showed enhanced delivery of Intradermal injections are the simplest and most direct method siRNA [17]. siRNA delivery into skin was enhanced 3.5 fold and localized for delivering NAs into the skin. Here, the barrier properties of the SC mainly in the dermis. Hessenberger et al. [18] used laser microporation are overcome completely by injecting NAs directly into the viable to deliver CpG oligonucleotides into the skin and successfully protected tissue layers of the skin. Intradermal injections are typically used for against immune response to grass pollen in mice. They used the Precise evaluating efficacy of NAs or other cutaneous therapeutics, or as the Laser Epidermal System (Pantec Biosolutions) which creates well- positive control for evaluating dermal delivery technologies, however, defined arrays of micropores in the skin, and allows precise control the downsides of intradermal injection for treating skin disease are over the number, density, and depth of the micropores giving the user

Fig. 1. Transport pathways into the skin. A: Intercellular pathway through lipid bilayers. B: Transcellular pathway through keratin-rich corneocytes. C: Shunt pathway through hair follicles and sweat ducts.

Please cite this article as: M. Zakrewsky, et al., Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges, J. Control. Release (2015), http://dx.doi.org/10.1016/j.jconrel.2015.09.017 M. Zakrewsky et al. / Journal of Controlled Release xxx (2015) xxx–xxx 3

Table 2 Advantages and disadvantages of physical, active, and passive topical NA delivery methods.

Class Methods Advantages Limitations

Physical Microneedles; microporation • High enhancement • Cost • No therapeutic limitations • Limited application area • Easily incorporate large depot • Slow return of skin barrier function • Delivery not majorly affected by diseased state of skin • Potentially irritating • No direct cell internalization enhancement Active Electroporation; iontophoresis; • Skin penetration and cell internalization • Cost sonophoresis • Controlled delivery • Limited application area • Easily incorporate large depot • Localized penetration zones • Repeated application at the same site • Transient perturbation of skin Passive Polymeric nanoparticles; liposomes; • Low cost • Nanoparticles, liposomes may localize in hair follicles peptides; dendrimers • Application to large skin surface • Peptides, dendrimers may be limited by size of NA • Can protect therapeutic from degradation • Lower enhancement • Multi-functional (SC perturbation, epidermal tight junction • Delivery can vary depending on state of SC and epidermis perturbation, cell membrane perturbation) • Targeting of specific cell-types

unprecedented tuneability over the amount and localization of thera- of SC microstructure creating pores in the skin [14].Ontheotherhand, peutic delivered [19]. electroporation is the application of short duration (b0.5 s) and high in- tensity (b100 V) electric pulses to the skin [20] which result in transient 3.2. NA delivery using active methods permeabilization of the lipid bilayers in the skin and concurrently permeabilize cell membranes of epidermal keratinocytes. Electropora- 3.2.1. Electroporation tion is also expected to create aqueous pores through the skin. However, Electroporation can be used to permeabilize the skin and enhance electroporation, unlike microporation, acts through non-thermal mech- passive diffusion of drug. The mechanism of electroporation is quite dif- anisms as lipid rearrangement, reduced skin resistance, and enhanced ferent from that of electrically-induced microporation. Electrically- transdermal transport are observed in the absence of a significant tem- induced microporation utilizes electric fields to induce thermal ablation perature rise in the pulse medium [20]. Using electroporation, Regnier et al. [21] showed enhanced permeability of the stratum corneum to a phosphorothioate antisense ODN. The permeability enhancement lasted up to 1 hr post-electroporation in rat skin in vitro. Specifically, transport was enhanced N 4-fold into the SC and N 3-fold into viable skin tissue when oligonucleotide solution was applied immediately fol- lowing electroporation. Further, Zhang et al. [22] demonstrated effective gene transfection and expression in the epidermis of human skin.

3.2.2. Iontophoresis Iontophoresis can be used to drive transport of charged drugs like NAs. Applying a continuous low intensity (b10 V) electric field at a con- stant current [20] has been used extensively to deliver a wide range of charged therapeutics including calcitonin, luteinizing hormone- releasing hormone, and dexamethasone [5]. In contrast to electropora- tion which acts primarily on the skin structure, iontophoresis is not be- lieved to cause major changes to the skin. Minor structural effects can be observed and may partially contribute to delivery enhancement through sweat ducts and hair follicles [20,23]. Nevertheless, iontopho- resis is believed to primarily act on the drug itself, driving transport of a charged molecule by means of an applied electric field. Using ionto- phoresis, Kigasawa et al. [24] demonstrated successful delivery of anti- IL-10 siRNA. Specifically, using an atopic dermatitis model in rats they showed a 73% reduction in IL-10 mRNA levels after treatment with anti-IL-10 siRNA and iontophoresis. The same group later extended the technique for vaccination as well as treatment of melanoma tumors [25].Kigasawaet al. reported enhanced delivery of CpG oligonucleotides into the epidermis and dermis using iontophoresis compared to that by free diffusion. Further, for the treatment of melanoma tumors in hairless mice, CpG oligonucleotides delivered using iontophoresis resulted in tumor reduction comparable to subcutaneous injection. Abu Hashim et al. also demonstrated the ability to deliver NF-κB decoy ODN into skin using iontophoresis for atopic dermatitis treatment [26].While passive diffusion of FITC-NF-κB decoy ODN was negligible for all con- centrations tested, ~100 pmol/cm2 was delivered after 6 hr of iontopho- Fig. 2. Microneedle arrays offer controlled length and sharp tips for easy insertion through resis using 0.5 mA current density. In addition, the authors observed a the SC and into the epidermis. (a) Scanning electron micrograph of a 20 × 20 silicon fl microneedle array. (b) Scanning electron micrograph of a microneedle tip. Figure linear dependence between the ux of ODN into the skin with both cur- reproduced with permission from Henry, et al. [104]. rent density and concentration of drug. When the technique was

Please cite this article as: M. Zakrewsky, et al., Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges, J. Control. Release (2015), http://dx.doi.org/10.1016/j.jconrel.2015.09.017 4 M. Zakrewsky et al. / Journal of Controlled Release xxx (2015) xxx–xxx applied for the treatment of atopic dermatitis in mice, the authors noted skin disease in vivo.Tranet al. [29] demonstrated ~30% reduction in significant reduction in both tissue swelling and TNFα expression. In melanoma tumor size when an anti-B-Raf siRNA liposome formulation comparison, treatment with buffer alone, as well as treatment with a was delivered into skin using sonophoresis. The liposome was postulat- scrambledODNsequence,resultedinnosignificant reduction in either ed to protect siRNA from enzymatic degradation in skin, as well as aid ear swelling or TNFα. Iontophoresis has also been used to deliver NA intracellular uptake by melanoma cells. complexes into the skin. Brus et al. [27] achieved enhanced delivery of polyethylenimine (PEI) complexed with ODN. Negatively charged 3.3. NA delivery using passive methods ODN was electrostatically complexed with positively charged PEI at a charge ratio of 1:13.3 to create positively charged complexes. 3.3.1. Nanoparticles Complexing was shown to protect oligonucleotides from enzymatic Nanoparticles are promising due to their high loading capacity, abil- degradation and enhance intracellular delivery in epidermal ity to shield enzymatic degradation, and reduce immunogenicity. Due to keratinocytes. Complexes did not show any measureable passive diffu- their size, nanoparticles are expected to require physical perturbation of sion, however, under constant electrical current the complexes did pas- the SC and underlying epidermis for effective delivery. Recently, howev- sage into the skin primarily via the shunt pathway. er, functionalized nanoparticles have been shown to passively diffuse through the skin and elicit a therapeutic response. For example, Siu et 3.2.3. Sonophoresis al. [30] demonstrated effective delivery of functionalized carbon nano- Low-frequency ultrasound has also been demonstrated to transient- tubes for the treatment of melanoma in vivo. Single-walled carbon ly permeabilize the SC lipid bilayers, facilitating the delivery of a large nanotubes functionalized with PEI were shown to deliver anti-B-Raf number of macromolecules including insulin, bovine serum albumin, siRNA and result in B-Raf silencing and attenuation of tumor growth and heparin [5]. Tezel et al. [28] first reported delivery of therapeutic in a mouse melanoma model. Ozbas-Turan et al. [31] delivered chitosan quantities of NAs into the skin using sonophoresis. Delivery of ODNs complexed with β-galactosidase plasmid DNA into mouse skin resulting into porcine skin in vitro was significantly enhanced by applying in significant expression of β-galactosidase after 7 days of treatment. 2.4 W/cm2 ultrasound for 10 min (Fig. 3). Interestingly, enhancement Similarly, Cui and Mumper [32] showed delivery and expression of lu- seemed to occur in localized regions of the skin (Fig. 3), however, ciferase plasmid DNA when complexed with chitosan. these regions were not observed to be hair follicles as is commonly re- ported for iontophoresis-mediated delivery of NAs. Instead, localized 3.3.2. Liposomes penetration zones most likely correspond to localized areas of acoustic Liposomes have also been studied extensively for nucleic acid deliv- cavitation inherent to this technique, and occupied ~5% of skin surface ery for the treatment of skin disease. For example, Desai et al. [33] used area. In addition, the authors showed no observable effect of ultrasound cationic lipid nanoparticles complexed with anti-TNFα siRNA and cap- on skin histology. NA delivery with ultrasound was also shown to treat saicin to treat psoriasis in vivo. Treatment resulted in significantly

Fig. 3. Sonophoresis enhances delivery of NA into skin through localized perturbation zones. (a) Top view of skin exposed to sulforhodamine B and ultrasound. (b and c) FITC-NA delivery into skin after ultrasound treatment. (b) Cross-section of skin corresponding to a localized perturbation zone. (c) Cross-section of skin corresponding to a non-localized perturbation zone. Figure modified with permission from Tezel et al. [28].

Please cite this article as: M. Zakrewsky, et al., Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges, J. Control. Release (2015), http://dx.doi.org/10.1016/j.jconrel.2015.09.017 M. Zakrewsky et al. / Journal of Controlled Release xxx (2015) xxx–xxx 5 reduced expression of a number of inflammatory cytokines including 3.3.4. Peptides TNFα,IL-17,IL-23,andNF-κB. Bracke et al. [34] used ultraflexible Peptides hold potential as drug delivery vehicles owing to their sim- liposomes loaded with anti-β defensin-2 to treat psoriasis in vivo.Plas- plicity, diversity, biocompatibility, and potential for multi-functionality. mid DNA can also be delivered using liposomes. Li et al. delivered IL-4 In addition, peptides can be easily screened and selected from phage- encoding plasmid DNA into mouse skin in vivo for the treatment of display libraries for various functions (Fig. 5). Phage-display screening psoriasis [35]. Enhanced expression of IL-4 led to suppressed hyper- offers a powerful tool for high-throughput discovery of novel peptide plastic and inflamed vessels in the skin. Similarly, Kim et al. [36] used sequences that can enhance penetration of large cargos into skin. More- liposomes complexed with antisense ODN for IL-13 mRNA to treat over, peptide sequences that localize in unique regions of the skin can atopic dermatitis in vivo. Treatment with liposome formulation resulted also be selected to target specific cell types and minimize side effects. in a significant reduction in skin thickness and inflammatory cell Over the last ten years, several peptides have been identified which pos- infiltration. sess the ability to enhance transport of NAs into the skin and elicit a therapeutic response. The first of these peptides discovered using 3.3.3. Spherical NAs phage-display screening was TD-1 (ACSSSPSKHCG) [42].Linet al. [43] Highly ordered spherical complexes of nucleic acids (spherical showed TD-1 could enhance transport of GAPDH siRNA into viable nucleic acids) have shown potential for treating skin disease due to tissue in the skin, as well as subcutaneous tissue, and silence GAPDH their enhanced delivery into skin, internalization into skin cells, and expression. Co-incubation of GAPDH siRNA with TD-1 resulted in simi- protection of NAs from degradation (Fig. 4) [37]. Further, several differ- lar GAPDH expression levels as intradermal injection, while expression ent sequences of NAs can be incorporated into a single construct for levels for both methods of application were significantly less than siRNA multifactorial diseases. Imaging agents like quantum dots can be used applied on the skin without peptide [43]. Further, application with TD-1 as the core. Alternatively, hollow spherical nucleic acids can be prepared resulted in significantly reduced levels of target mRNA in the skin for for incorporation of additional drug. Zheng et al. successfully delivered up to 3 days and target protein in the skin for up to 7 days. Hsu and anti-EGFR (epidermal growth factor receptor) siRNA into mouse skin Mitragotri [44] identified another peptide using phage-display screen- using gold-core nanoparticles [38].Specifically, gold nanoparticles coat- ing, SPACE peptide (ACTGSTQHQCG), with the ability to not only en- ed with a dense layer of highly-ordered and covalently bound siRNA hance delivery of siRNA across the skin but also enhance intracellular resulted in passive transport through intact mouse SC and localized ex- uptake. In vivo application of SPACE peptide conjugated to IL-10 siRNA clusively in the dermis and epidermis. After 3 weeks of treatment the or GAPDH siRNA resulted in ~30% or ~45% knockdown in protein authors observed nearly complete knockdown of EGFR as well as re- expression, respectively. This result is in contrast to the control formu- pression of downstream phosphorylation, and reduction in epidermal lation containing siRNA alone which showed negligible gene silencing. thickness. Spherical nucleic acids also demonstrated the ability to stifle Further, SPACE peptide was shown to be non-toxic at concentrations nuclease degradation and efficiently internalize into a large variety of as high as 10 mg/mL [45]. Uchida et al. [46] used a dual-peptide sys- cell types to stimulate gene silencing [39]. Further, spherical nucleic tem with both Tat peptide (GRKKRRQRRRCG) and AT1002 (FCIGRLCG) acids have been used to treat psoriasis [40] as well as aid wound healing to enhance gene silencing in the skin. Tat peptide and siRNA were com- [41] in vivo through knockdown of TNFα and ganglioside GM3 synthase, plexed via electrostatic interactions. Complexes were then co-incubated respectively. with AT1002 and applied on the skin. Here, Tat peptide was used as a

Fig. 4. The anatomy of spherical nucleic acid nanostructures. An inorganic core is densely functionalized with oligonucleotides containing three segments: a recognition sequence, a spacer segment, and a chemical-attachment group. Additionally, other functional groups such as dye molecules, quenchers, modified bases, and drugs can be attached along any segment of the oligonucleotide. Figure reproduced with permission from Cutler et al. [37].

Please cite this article as: M. Zakrewsky, et al., Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges, J. Control. Release (2015), http://dx.doi.org/10.1016/j.jconrel.2015.09.017 6 M. Zakrewsky et al. / Journal of Controlled Release xxx (2015) xxx–xxx

Fig. 5. Experimental design to screen for skin-penetrating peptides using phage-display. The same strategy could be used to screen for aptamers. cell-penetrating peptide to enhance intracellular delivery, and AT1002 resulted in significantly enhanced apoptosis and reduction in tumor was used as a tight-junction modulator to enhance permeation into size in mice. the epidermis and dermis. Tat peptide has also been shown to enhance macromolecule transport across the SC [47,48]. The system was later 4. Potential impact applied to treat atopic dermatitis in mice [49]. Application of anti-RelA siRNA complexed with Tat peptide and co-incubated with AT1002 led The burden of skin disease is alarming and growing fast [53]. 1 out to reductions in ear thickness, clinical skin severity, topical cytokine of 3 individuals is estimated to have a skin disease at any given time, levels, and serum IgE production. Yi et al. [50] successfully delivered resulting in direct healthcare costs over $30 billion annually in the US anti-microphthalmia-associated transcription factor (anti-MITF) siRNA alone [54]. Symptoms from the ~3000 different skin diseases range conjugated to TD-1 R8 peptide (ACSSSPSKHCGRRRRRRRR) for the treat- from itching, redness, and irritation to physical disfigurement, or ment of patients with melisma. Treatment with a cream formulation death. Furthermore, skin diseases manifest externally leading to severe containing TD-1 R8 peptide co-administered with anti-MITF siRNA re- and even debilitating emotional distress and social prejudice [54]. sulted in reductions in tyrosinase, tyrosinase-related protein 1, and Dermatological drugs aimed at treating skin disease reap an estimated melanocortin 1 receptor leading to measurable inhibition of melanin $24.4 billion in annual revenue [55]. In addition, many other skin con- production and melanocyte apoptosis. In fact, after 4 weeks application ditions, not considered disease, are of major concern. These include cos- patients demonstrated a significant lightening of facial hypermelanosis metic conditions like wrinkling, cellulite, discoloration, and skin pliancy. lesions and almost completely restored dark lesions to normal skin color Growing demand for more effective treatments of cosmetic conditions after 12 weeks. is evidenced by the emergence and growth of the cosmeceutical market which was estimated to be ~$35 billion globally in 2013 [56].TopicalNA therapies have exciting potential to reduce this burden and be transfor- 3.3.5. Dendrimers mative in the way we deal with and treat skin disease (Table 3). Exam- Although less studied than peptides for delivering NAs into skin, ples are discussed in detail below. dedrimers are similarly advantageous due to their diversity, ease of Targeting orphan disease can help speed translation by allowing synthesis, and functional group density. Bielinska et al. [51] demonstrat- fast-tracked regulatory processing from orphan drug designation. ed the use of dendrimers for topical gene delivery. Dendrimer com- There are several orphan skin diseases with recognized NA targets. plexes resulted in measurable gene expression of chloramphenicol Hidradenitis suppurativa (Hurley disease) is an orphan disease that af- transacetylase that was ~7-fold higher than with naked plasmid. In fects over 100,000 people in the US [57], most commonly women [58]. addition, Venuganti et al. [52] used a similar dendrimer in combination It is a chronic and debilitating disease characterized by painful abscess- with iontophoresis to deliver antisense oligonucleotide. Treatment with es, redness, and scarring, leading to significant physical and psycholog- antisense oligonucleotide targeting the anti-apoptotic protein, Bcl-2, ical impairment and reduced quality of life [59]. Currently, the most

Please cite this article as: M. Zakrewsky, et al., Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges, J. Control. Release (2015), http://dx.doi.org/10.1016/j.jconrel.2015.09.017 M. Zakrewsky et al. / Journal of Controlled Release xxx (2015) xxx–xxx 7

Table 3 Topical NA delivery can have a large impact in the clinic due to the large number of skin diseases with NA therapeutic targets responsible for their etiology and pathogenesis.

Disease/Condition People affected Causative molecule Role in pathogenesis Up/downregulation Ref. and NA target for for therapy therapy

Hurley disease 100,000 in US TNFα, IL-17, IL-23 Promotes inflammation through activation of vascular Down [61,62] endothelial cells and immune cells Epidermolysis 500,000 globally IL-1β, IL-1β receptor Mutant K14 leads to upregulation of IL-1β, which then Down [67] bullosa results in a positive feedback signaling cascade that ends up further activating mutant K14 expression Mutant K5, K14 Mutations result in keratin filament aggregation in Down (mutant) [68,69] keratinocytes leading to dysfunction Collagen VII Mutation results in loss of anchoring-fibril formation Up (wild-type) [70] Pachyonychia congenita 5000–10,000 globally Mutant K6a, K6b, K17 Mutations disrupt keratin filament formation resulting Down (mutant) [72] in skin cell fragility Netherton syndrome 1/50,000 globally Mutation in serine protease inhibitor Kazal type 5 leads Down [79] to serine protease hyperactivity and excessive protein degradation in skin Psoriasis 4.5 million adults in US TNFα, IL-17, IL-23 Same as above Down [64] IL-4 IL-4 producing T helper cell population is repressed Up [84] β defensin-2 Highly upregulated in psoriatic lesions Down [34] Atopic dermatitis 10%–20% of children; RelA Important member of the NF-κBinflammatory pathway Down [49] 1%–3% of adults IL-4, -5, -10, -13 Overexpressed in AD lesions Down [85] Th2-type immune Th2-type dominated immunological response Down [88] response implicated in AD pathogenesis Cellulite 80%–90% post-adolescent ACE ACE has been linked to higher production of angiotensin Down [89] women II, a vasoconstrictive peptide, along with lower production of bradykinin, a vasodilatative peptide Collagenase, lipase, Enzyme hyperactivity leads to breakdown of subcutaneous Down [90] elastase and dermal ECM. Lipase suppression leads to build up of (collagenase, fatty tissue elastase); up (lipase) HIF-1 HIF-1 induces fibrosis and inflammation which is Down [89] implicated in the formation of cellulite Wrinkling Universal with aging Elastase Breakdown of elastic fibers leads to reduced skin Down [91] elasticity and wrinkle formation Melasma 4%–10% of all MITF Inhibition of MITF gene leads to suppression of melanin Down [50] dermatological visits production in dark facial lesions common form of treatment is surgical excision of diseased areas of the with severe EB survive past the age of 30 due to extensive wounds skin [60]. For severe forms of Hurley disease there is no FDA approved and . There is no FDA approved drug indication for EB. Manage- treatment. Studies into the etiology and pathogenesis of Hurley disease ment of EB consists of limiting blister formation from scratching and suggest it is a multifactorial inflammatory disease characterized by friction [66]. For more effective treatment, studies suggest IL-1β and overexpression of TNFα [61] as well as other inflammatory cytokines its receptor may be a valuable target. Indeed, overexpression of IL-1β like IL-17 and IL-23 [62]. Treatment of Hurley disease with anti-TNFα has been detected in patients with EB and treatment of EB in mice monoclonal antibodies or other biologics has been shown to be effective with anti-IL-1β or IL-1β receptor antagonist resulted in improvement [60,63].TNFα promotes inflammation through recruitment of vascular of symptoms [67]. Further, TWi Biotechnology Inc. was recently granted endothelial cells and immune cells. Further, it is overly expressed at orphan drug designation for its anti-IL-1β and anti-IL-1β receptor small sites of inflammation while repression generally results in alleviation molecule drug AC-201 [65]. EB can also be targeted at the genetic level. of inflammation [64]. While there is no FDA approved drug indication One subtype of EB ( simplex), is predominantly for treating moderate to severe Hurley disease, several anti-TNFα bio- caused by dominant-negative mutations in the genes encoding logics are already on the market including etanercept and adalimumab. keratin-5 and keratin-14 (K5 and K14). siRNA therapies that specifically In fact, the anti-TNFα monoclonal antibody, Humira (adalimumab, knockdown mutant keratin without affecting wild-type keratins have Abbvie Inc.), was recently granted orphan drug designation by the shown promise [68,69]. A second subtype of EB (dystrophic epider- FDA to start clinical trials on moderate to severe Hurley disease patients molysis bullosa) is caused by mutations in collagen VII and gene deliv- [65]. On the other hand, these drugs can only be administered by injec- ery of wild-type collagen VII has shown promise as a therapeutic in tion or infusion and systemic side effects limit their long-term use. phase I/II clinical trials [70]. Alarmingly, the most common side effect of systemic delivery is the de- Pachyonychia congenita (PC) is a rare genetic autosomal dominant velopment of other autoimmune diseases as a direct result of treatment skin disorder that affects 5000 to 10,000 people [71]. PC may be caused due to increased expression of IL-17 and IL-23 as well as other inflam- by mutations in either K6a, K6b, K6c, K16, or K17 [66]. The disease typ- matory cytokines [64]. Therefore, topical delivery of anti-TNFα siRNA ically manifests with calluses and blisters on the feet and palms, as well or antisense oligonucleotide is an attractive platform to limit off target as discoloration, cysts, and of the hair follicles [72]. effects, improve patient compliance, and reduce hospital-related costs. Moreover, patients typically live in constant pain. Due to the large num- Even more impactful, a cocktail of topical anti-TNFα, anti-IL-17, and ber of different mutations that can result in PC, topical application of anti-IL-23 NA therapeutics may prove synergistic at alleviating Hurley personalized NA therapies holds the most potential. In fact, treatment disease symptoms while avoiding adverse reactions. of patients with topical siRNA therapies are showing promise both in Epidermolysis bullosa (EB) is another rare skin disease with few ef- mouse models as well as in humans [73–77]. fective treatments. EB affects roughly 30,000 people in the US and Netherton syndrome is an autosomal recessive disorder estimated to ~500,000 people globally [66]. EB symptoms typically include severe affect 1/50,000 individuals [78]. Symptoms present at birth or shortly skin fragility to the point where large segments of skin can be removed after and include painful peeling and scaling of skin. More alarmingly, from simply scratching or rubbing skin [66]. Alarmingly, few patients Netherton syndrome manifests significant impairment of skin barrier

Please cite this article as: M. Zakrewsky, et al., Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges, J. Control. Release (2015), http://dx.doi.org/10.1016/j.jconrel.2015.09.017 8 M. Zakrewsky et al. / Journal of Controlled Release xxx (2015) xxx–xxx function leading to life-threatening dehydration and frequent systemic 80%–90% of post-adolescent women [89]. It is characterized by an “or- . Netherton syndrome is caused by a mutation of the serine ange peel” appearance of the skin primarily in the thighs and buttocks. protease inhibitor Kazal type 5 (SPINK5) gene leading to serine protease While physical burden is not associated with cellulite, significant emo- hyperactivity and excessive protein degradation in skin. Essentially, tional burden is typically associated with cellulite. Cellulite pathology protease hyperactivity results in excessive and premature epidermal is generally not well understood, however, a few NA targets that show [79]. Netherton syndrome may be treated by down- promise are proposed in literature. For example, decreased microcircu- regulating production of serine proteases in the skin, or alternatively, in- lation in subcutaneous adipose tissue is believed to play a role in cellu- ducing expression of wild-type serine protease inhibitor [80]. Due to the lite formation. Specifically, overexpression of angiotensin-converting significant reduction in skin barrier function, initial treatment with top- enzyme has been linked to higher production of angiotensin II, a vaso- ical NAs may not require dermal enhancement. Instead, technologies to constrictive peptide, along with lower production of bradykinin, a localize drug in the skin such as cell-targeting or skin targeting peptides vasodilatative peptide [89]. Breakdown of the extracellular matrix may be required. However, as skin integrity improves more significant (ECM) due to enzyme hyperactivity may also play an important role enhancement may be required. in cellulite [90]. Normal ECM may potentially be restored by silencing Mainstream disease would also benefit from topical NA therapies. enzymes like collagenase, elastase, and lipase. Finally, fibrosis and local- For example, psoriasis is estimated to affect nearly 4.5 million adults ized inflammation may play an important role in cellulite formation in the US alone [81]. Children are also commonly affected by the disease. [89].Specifically, hypoxic inducible factor-1 (HIF-1) mutations that It is a chronic skin disease that presents as severe lesions, redness, and minimize its ability to induce fibrosis and inflammation have been itching and poses a significant burden on patients' quality of life. shown to be associated with individuals who do not typically have cel- About 20%–25% of patients are estimated to be dissatisfied with their lulite, or have reduced severity of cellulite, while individuals with wild- current treatment [81]. Of even more concern, for patients suffering type HIF-1 were shown to have a higher probability of more severe pre- from the most severe form of psoriasis, erythrodermic psoriasis, there sentation of cellulite. is no standard treatment regimen [82]. Due to the multifactorial nature Skin wrinkling is another cosmetic condition that generates signifi- of psoriasis, NA therapies may be advantageous because NA cocktails cant emotional burden. Wrinkling affects everyone as they age, howev- targeting a number of different targets can be formulated and delivered er, treatment is most typically sought by post-adolescent females. leading to down/up regulation as needed. Psoriasis is one of the more Unfortunately, few effective treatments exist for the reduction of wrin- exhaustively studied forms of inflammatory skin disease and numerous kles. Currently, the most common treatment is intradermal injections of therapeutic targets have been proposed; a subset is described here. Sim- botulinum toxin A (Botox). Botox, however, is extremely toxic and re- ilar to Hurley disease, TNFα is perhaps the most established drug target sults in temporary muscular paralysis at the site of injection. Further, for treating severe forms of psoriasis [64]. Topical delivery of anti-TNFα Botox must be administered by a trained physician limiting its use and siRNA as well as a siRNA cocktail including anti-TNFα and anti-STAT3 increasing its cost. Alternatively, topical NA therapies may be beneficial was shown to be effective at alleviating psoriasis-like symptoms in as both a treatment and prevention strategy. For example, elastase up- mice [33,83]. Activated T helper cells have also been shown to play a regulation has been proposed as an important factor for ultraviolet major role in the manifestation of psoriasis, and selective skewing irradiation induced wrinkle formation [91]. Inhibition of elastase with from Th1 phenotype to the IL-4 producing Th2 phenotype can alleviate N-phenethylphosphonyl-L-leucyl-L-tryptophane resulted in reduced psoriasis symptoms [84]. Further, selective differentiation to the Th2 formation of wrinkles in mice exposed to daily doses of ultraviolet irra- phenotype can be simply induced by subcutaneous injection of IL-4 diation. Therefore, knockdown of elastase with topically applied NAs [84]. As an alternative to injection, psoriasis may be alleviated through may be a viable option for the treatment and prevention of skin wrin- topical delivery of plasmid DNA to induce expression of IL-4 in the kling, as well as provide patients with a safer and cheaper alternative skin. Proof-of-concept has been demonstrated in a K14-VEGF transgenic to Botox injections. mouse model of psoriasis [35]. Melasma is a hyperpigmentation disorder that manifests as darken- Atopic dermatitis (AD) affects 10%–20% of children and 1%–3% of ing or browning of the skin, typically on the face [92]. Melasma dispro- adults worldwide [85]. AD typically manifests with severe redness and portionately affects women and disproportionately affects ethnicities itching, skin lesions, and papules resulting in significant impact on qual- with darker skin color, however overall, melasma is generally estimated ity of life. Similar to psoriasis, the inflammatory nature of the disease to account for 4%–10% of all dermatological-related doctor visits [93]. makes topical NA treatment appealing. Although the exact cause of AD Moreover, melasma is commonly reported to induce significant psycho- is not known, upregulation of inflammatory pathways such as the NF- logical burden, social impairment, and reduced quality of life [92,93]. κBinflammatory pathway are typically observed, and inhibition of The most common treatments are skin lightening agents like tyrosinase these pathways have shown promise. For example, delivery of anti- inhibitors and spot removing agents like retinoic acid, kojic acid, and RelA, an important member of the NF-κBinflammatory pathway, result- azelaic acid [50]. However, these are typically either ineffective or result ed in significant improvement in disease symptoms in mice [49].Inad- in unsightly white spots when used in the clinic. Topical NA therapies dition, knockdown of NF-κB with decoy ODNs also showed therapeutic may prove beneficial for treating melasma without adverse side effects promise in mice [26]. Several cytokines are also believed to be associat- associated with current treatments. Specifically, studies have shown si- ed with the pathogenesis of AD including IL-4, -5, -10, and -13. These lencing of the microphthalmia-associated transcription factor (MITF) cytokines are typically expressed in significantly higher quantities in gene could be an effective treatment strategy. MITF encodes for tyrosi- AD lesions compared to healthy skin [85], and knockdown of these cy- nase, tyrosinase-related protein 1, and melanocortin 1 receptor which tokines and others with topical NA therapies has shown promise in are all involved in melanin synthesis. Topical delivery of anti-MITF mouse models of AD [24,36,86,87]. Further, suppressing the Th2-type siRNA has shown promise in human trials [50]. dominated immunological response typical of AD may hold therapeutic promise. Indeed, delivery of CpG oligonucleotides was shown to signif- icantly reduce AD lesions in mice by shifting from Th2-type dominant 5. Remaining challenges immune response to a more balanced Th1/Th2 immune response [88]. Cosmetic conditions would also benefit significantly from topical NA Despite significant efforts to deliver NAs into the skin as well as iden- therapies developed as either pharmaceuticals or cosmeceuticals. How- tifying NA targets for treating skin disease, significant hurdles remain. ever, cosmeceutical development may benefit from faster translation to Three major areas posed serious challenges in the past and still remain the clinic due to reduced FDA regulations provided claims of the cosme- the bottleneck to successful translation of topical NA therapies: (1) skin ceutical are appropriately chosen. For example, cellulite manifests in delivery, (2) cellular internalization, and (3) stability of NAs. These

Please cite this article as: M. Zakrewsky, et al., Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges, J. Control. Release (2015), http://dx.doi.org/10.1016/j.jconrel.2015.09.017 M. Zakrewsky et al. / Journal of Controlled Release xxx (2015) xxx–xxx 9 challenges must be addressed concurrently to develop successful NA microneedle arrays and microporation devices see widespread use in topical delivery systems for use in humans. the clinic. Microneedles and other physical methods are the most effective at Perhaps the most significant drawback of both physical and active enhancing delivery into the skin, and the type and size of therapeutic methods, however, is limited application area. For diseases like Hurley is not restricted. Further, large depots can be easily incorporated for disease, EB, moderate to severe psoriasis, skin wrinkling, and many sustained release. However, significant questions remain in regards to others which can occupy a large percentage of skin surface area, physi- their effectiveness at localized and homogeneous delivery into skin tis- cal and active methods may not be practical. In these cases, passive per- sue. Specifically, physical methods inherently result in localized pene- turbation methods may be most effective. tration zones. They do not address horizontal diffusion of NA which is Passive transport enhancers can be formulated as a cream and retarded by cell–cell tight junctions in the epidermis [7,8].Active applied to all areas of the body, including the face, in a patient friendly methods such as electroporation [94] and sonophoresis [28] may also manner. Further, formulations incorporating synergistic combina- be limited by localized penetration zones (Fig. 3). The effect of size of tions of skin penetrating and cell internalizing transport enhancers penetration area on clinical outcome needs to be determined. Combina- can be easily envisaged. The main limitation of passive methods tion therapies with co-delivery of NAs and, for example, tight junction remains their low transport enhancement relative to physical and active modulators may be required for efficacy in humans. methods. Further, passive transport enhancers like peptides and Also of concern, physical methods do not enhance cellular inter- dendrimers may be limited to use with NAs of a certain size like anti- nalization nor protect NAs from degradation in the skin. NAs need sense oligonucleotides and siRNA as opposed to plasmid DNA. Effort to enter into the cytoplasm or nucleus of cells to elicit a therapeutic re- has been spent over the years to identify and understand passive trans- sponse. Physical methods alone cannot facilitate this enhancement, port enhancers. High-throughput screening for chemical enhancers and therefore, they must inherently be combined with other technolo- is well-documented [105] and has proven useful for identifying gies to enable cell internalization. For example, microneedle arrays inte- synergistic combinations of chemicals that perturb the skin with mini- grating active methods like electroporation [95–97] and sonophoresis mal irritation [106]. Along the same lines, peptides can be screened in [98] have been designed to facilitate NA delivery enhancement into a high-throughput fashion using phage-display (Fig. 5). Phage libraries cells. However, incorporating active methods for cell internalization applied to the skin are selected based on their ability to transport further adds complexity and cost. Passive methods for cell internaliza- through the skin. After only a few rounds, a library of ~109 peptide se- tion, for example, tagging NAs with cell-penetrating peptides, cell- quences can be screened. Particularly advantageous, this technique penetrating dendrimers, or cell-penetrating aptamers have the poten- can be used to identify peptides that localize in particular layers of the tial to provide affordable means of permeation enhancement. In skin e.g. epidermis or dermis. Delivery of NA therapies to specific loca- fact, several cell-penetrating peptides are currently in clinical trials tions in the skin could be beneficial to limit off-target effects. Phage which makes them an attractive option [99]. On the other hand, cell- screening is a powerful technique, and an extensive number of phage li- penetrating peptides combined with physical or active dermal penetra- braries exist. Effort should be place on identifying novel peptides with tion enhancement still neglect the stability issues of NAs. siRNA, ODN, powerful skin-penetrating ability. The same methodology could be and plasmid DNA are susceptible to enzymes in the epidermis and der- used to screen for skin-penetrating aptamers, which to the authors' mis which can lead to significant degradation. To address this concern, knowledge has not been attempted to-date, further broadening the packaging NAs in cationic lipid complexes or liposomes would both pro- space of chemically and structurally distinct passive transporters for de- tect the NA from enzymatic degradation as well as aid cell internaliza- livering NAs. tion. In addition, chemical modifications of NAs like attaching charge As new transporters are identified, we can better understand how to neutral moieties via cleavable or reducible linkers or use of a phospho- overcome the skin barrier to deliver increasingly large cargos. For exam- rothioate backbone have been shown to aid in internalization [100] and ple, the number of skin-penetrating peptides identified thus far have protect from degradation [101], respectively, without compromising afforded a better understanding of their mechanism of transport en- function. hancement [107]. Interestingly, TD-1, Tat, and SPACE peptide all seem The regulatory hurdles for medical devices and drug–device combi- to bind and interact with keratin in the corneocytes in the skin facilitat- nations must also be addressed. Physical methods are sophisticated and ing transport of cyclosporine A through the transcellular pathway. As a will be highly regulated. Fabrication, sterilization, and implementation result of this work, we posit screening of low molecular weight ligands of the device must be demonstrated to result in safe and effective ther- for affinity to keratin may help identify ligands with improved NA deliv- apy. This regulation is in addition to any oversight process for the NA ery. It is also conceivable that the mechanism of enhancement is depen- itself and further complicates translation of topical NA therapies. dent on the cargo delivered, however, similar studies with NA cargos are Along this line, a lot of effort has been spent over the years to address severely lacking. fabrication concerns. For example, methods have been devised to fabri- Concurrently, definitive mechanistic studies to elucidate modes of cate microneedle devices using steel and other strong materials [102]. transport enhancement of nanoparticles, liposomes, and spherical Further, biocompatible and biodegradable microneedles have been pro- nucleic acids are lacking and studies that have been reported do not posed [103]. Many of these methods can be scaled using the same pro- reach adequate consensus. Alvarez-Roman et al. [108] have studied cesses used for integrated circuits [104] which affords cost-effective the distribution of polystyrene particles after topical application. They manufacturing as well as the potential for outsourcing manufacturing. observed almost exclusive uptake into the hair follicles. Follicular up- Lacking, however, are studies assessing effects of sterilization and im- take was dependent on size of the particles with 20 nm particles accu- plementation protocols on patient safety. For example, all physical mulating to a larger extent than 200 nm particles. Similar localization methods result in disruption of the skin barrier. This disruption will re- in the hair follicles has been observed for liposomes [109–111] and main until re-epithelialization at the site of ablation can occur. While ultradeformable liposomes [35], as well as titanium dioxide microparti- barrier function of the skin is diminished, pathogens may potentially cles [112], micro/nanoemulsion droplets [113], lipoplexes [114],and enter the skin and cause infection. Therefore, the site of application solid lipid nanoparticles [112]. However, naked NAs have also been may need to repeatedly be sterilized, or sealed with a sterile bandage, shown to localize in hair follicles when applied without any nanocarrier for days or weeks after application of the NA therapy. Depending on [115–117] which suggests nanocarriers may only hold potential as drug the frequency of application as well as the site of application (e.g. face, depots for controlled release topical formulations of NAs. Still others hands, feet), requiring a permanent or semi-permanent sterile bandage have demonstrated dispersed distributions of nanocarriers into viable may not be tolerated by the patient. Prevalence of adverse events from epidermis and dermis. Verma et al. [118] demonstrated enhanced physical ablation of the SC and epidermis must be determined before drug delivery using liposomes and also concluded that size was the

Please cite this article as: M. Zakrewsky, et al., Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges, J. Control. Release (2015), http://dx.doi.org/10.1016/j.jconrel.2015.09.017 10 M. Zakrewsky et al. / Journal of Controlled Release xxx (2015) xxx–xxx most important parameter. However, it is important to note that 6. Conclusion these studies were performed with fluorescent drug and therefore do not distinguish between penetration of intact liposomes versus Topical application of NAs for the treatment of skin disease is an ad- enhancement in drug delivery through simple fluidization of lipid vantageous route for the translation of NA therapies to the clinic. Topical bilayers. In fact, Kirjavainen et al. concluded that non-fusogenic application offers many advantages over alternative methods of admin- fluorescently-tagged liposomes do not penetrate skin [119]. Instead, istration including avoidance of many challenges that are current road- only liposomes that were fusogenic with SC lipids were able to enhance blocks to systemic NA therapy implementation. Although the skin does drug delivery. Moreover, fusogenic liposomes enhanced penetration pose a significant barrier to drug delivery, especially to large hydrophilic of drug applied in free solution subsequent to application of lipo- macromolecules like NAs, extensive effort has been spent to overcome somes. These data suggest that liposomes may act through incor- this barrier. These efforts have been highlighted here, and include the poration into and fluidization of the lipid bilayers to enhance drug use of microneedles, microporation, electroporation, iontophoresis, delivery. Other factors of liposome design such as charge, amount of sonophoresis, nanoparticles, liposomes, spherical nucleic acids, pep- cholesterol, and acyl chain length did not appear to influence drug tides, and dendrimers. Translation of these technologies to the clinic is delivery, In contrast, Geusens et al. concluded liposomes can penetrate sure to have a transformative impact on the burden of both skin disease intact skin if flexible enough [120]. It is reasonable that liposomes as well as cosmetic conditions. A large number of genetic and multifac- that can penetrate via the multilamellar lipid bilayers in the SC without torial inflammatory diseases result in chronic, physical and emotional breaking apart may aid delivery of cargo into the skin. Others have also burden to patients who have few treatment options other than repeated reached similar conclusions arguing ultraflexible liposomes create dosing by injection or infusion, or invasive surgery. Similarly, cosmetic hydration-driven transport mechanisms through the skin [121].How- conditions result in significant emotional burden with limited effective ever, since studies have shown localization of ultraflexible treatment options other than invasive surgery. Future efforts should liposomes in hair follicles [35], another possibility is that flexibility focus on validating the safety of long-term physical disruption of the does not aid penetration across multilamellar SC but instead enhances skin, better understanding the mechanisms of NA enhancement into transport only across the unilamellar lining of hair follicles. If confirmed, skin via passive methods, and exhaustively screening for novel ligands this may preclude treatment of regions devoid of hair follicles like palms and synergistic combinations of enhancers to realize the full potential and foot pads. Interestingly, spherical nucleic acids, are not be expected of topical NA therapies in the clinic. to fluidize lipid bilayers, nor are they be expected deform to facilitate passive diffusion through tight intercellular lipid channels in the skin; Acknowledgment therefore, we would expect them to localize in hair follicles like the ma- jority of solid nanoparticles N 40 nm studied to date [122].Yet,spherical The authors acknowledge support from Duncan and Suzanne nucleic acids exhibit extensive penetration homogenously into epider- Mellichamp Chair and fellowship as well as from the National Institutes mal and dermal tissue [38,40]. How spherical nucleic acids penetrate of health (1R21CA191133). skin has yet to be reported but should be of immediate interest given the significant potential this platform appears to have for treating skin disease. Although in vitro testing using Franz diffusion cells can be per- References formed easily and quickly to screen large libraries of phage-displayed [1] C.V. Pecot, G.A. Calin, R.L. Coleman, G. Lopez-Berestein, A.K. Sood, RNA interference peptides and aptamers, testing individual formulations of nanocarriers in the clinic: challenges and future directions, Nat. Rev. Cancer 11 (2011) 59–67. remains tedious. Thus, studies that continue to identify mechanisms of [2] R.K.M. Leung, P.A. Whittaker, RNA interference: from gene silencing to gene- nanocarrier delivery and important design parameters for delivery en- specific therapeutics, Pharmacol. Ther. 107 (2005) 222–239. [3] S.I. Pai, Y.Y. Lin, B. Macaes, A. Meneshian, C.F. Hung, T.C. Wu, Prospects of RNA in- hancement are crucial to advance passive delivery methods. terference therapy for cancer, Gene Ther. 13 (2006) 464–477. On the other hand, passive enhancers can be significantly affected [4] E.R. Rayburn, R.W. Zhang, Antisense, RNAi, and gene silencing strategies for thera- by the state of the SC. For example, a liposome formulation may be py: mission possible or impossible? Drug Discov. Today 13 (2008) 513–521. engineered to be non-toxic or non-irritating to intact skin. How- [5] M.R. Prausnitz, S. Mitragotri, R. Langer, Current status and future potential of trans- dermal drug delivery, Nat. Rev. Drug Discov. 3 (2004) 115–124. ever, when applied on diseased skin, because barrier function is dimin- [6] M.B. Brown, G.P. Martin, S.A. Jones, F.K. Akomeah, Dermal and transdermal drug ished, the same formulation may deliver an irritating or cytotoxic dose. delivery systems: current and future prospects, Drug Deliv. 13 (2006) 175–187. In contrast, delivery using physical methods should be minimally affect- [7] N. Kirschner, P. Houdek, M. Fromm, I. Moll, J.M. Brandner, Tight junctions form a barrier in human epidermis, Eur. J. Cell Biol. 89 (2010) 839–842. ed by the diseased state of skin and active methods can be easily tuned [8] N. Kirschner, R. Rosenthal, M. Furuse, I. Moll, M. Fromm, J.M. Brandner, Contribu- to control the amount of dose delivered. Formulations of passive en- tion of tight junction proteins to ion, macromolecule, and water barrier in hancers, especially complex formulations, are not as easily tuned. This keratinocytes, J. Investig. Dermatol. 133 (2013) 1161–1169. [9] S. Mitragotri, Immunization without needles, Nat. Rev. Immunol. 5 (2005) must be considered during the design and implementation of passive 905–916. enhancers. [10] J.A. Mikszta, J.B. Alarcon, J.M. Brittingham, D.E. Sutter, R.J. Pettis, N.G. Harvey, Im- Clearly, much is still unknown; however, translation of NA therapies proved genetic immunization via micromechanical disruption of skin-barrier func- tion and targeted epidermal delivery, Nat. Med. 8 (2002) 415–419. appears to be close. Near-term challenges include validating safety of [11] F. Chabri, K. Bouris, T. Jones, D. Barrow, A. Hann, C. Allender, et al., Microfabricated microneedle and microporation arrays. Establishment of regulatory silicon microneedles for nonviral cutaneous gene delivery, Br. J. Dermatol. 150 guidelines will help on this front. In particular, we posit physical (2004) 869–877. [12] Z. Ding, E. Van Riet, S. Romeijn, G.F. Kersten, W. Jiskoot, J.A. Bouwstra, Immune methods for delivering CpG oligonucleotides or other NA-based vaccine modulation by adjuvants combined with diphtheria toxoid administered topically will be the first realization of topical NA therapy in the clinic. Indeed, the in BALB/c mice after microneedle array pretreatment, Pharm. Res. 26 (2009) benefits of needle-free immunization are well-established [123].How- 1635–1643. ever, despite their superior delivery efficacy, active and physical [13] E. Gonzalez-Gonzalez, T.J. Speaker, R.P. Hickerson, R. Spitler, M.A. Flores, D. Leake, et al., Silencing of reporter gene expression in skin using siRNAs and expression methods are severely limited by application area, and therefore, are of plasmid DNA delivered by a soluble protrusion array device (PAD), Mol. Ther. not ideal for local delivery of NAs to skin tissue where disease symptoms 18 (2010) 1667–1674. manifest. To this end, long-term challenges include high-throughput [14] J. Bramson, K. Dayball, C. Evelegh, Y.H. Wan, D. Page, A. Smith, Enabling topical im- munization via microporation: a novel method for pain-free and needle-free deliv- screening of passive enhancers, followed by systematic studies to eluci- ery of adenovirus-based vaccines, Gene Ther. 10 (2003) 251–260. date mechanisms of NA delivery. A synergistic combination of passive [15] C.-H. Lin, I.A. Aljuffali, J.-Y. Fang, Lasers as an approach for promoting drug delivery SC, tight junction, and cell membrane perturbers may be required to via skin, Expert Opin. Drug Deliv. 11 (2014) 599–614. [16] W.-R. Lee, S.-C. Shen, C.-R. Liu, C.-L. Fang, C.-H. Hu, J.-Y. Fang, Erbium: YAG laser- tackle most forms of skin disease and realize the full transformative po- mediated oligonucleotide and DNA delivery via the skin: an animal study, J. Con- tential of topical NA therapies in the clinic. trol. Release 115 (2006) 344–353.

Please cite this article as: M. Zakrewsky, et al., Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges, J. Control. Release (2015), http://dx.doi.org/10.1016/j.jconrel.2015.09.017 M. Zakrewsky et al. / Journal of Controlled Release xxx (2015) xxx–xxx 11

[17] W.-R. Lee, S.-C. Shen, R.-Z. Zhuo, K.-C. Wang, J.-Y. Fang, Enhancement of topical [46] T. Uchida, T. Kanazawa, Y. Takashima, H. Okada, Development of an efficient trans- small interfering RNA delivery and expression by low-fluence erbium: YAG laser dermal delivery system of small interfering RNA using functional peptides, Tat and pretreatment of skin, Hum. Gene Ther. 20 (2009) 580–588. AT-1002, Chem. Pharm. Bull. 59 (2011) 196–201. [18] M. Hessenberger, R. Weiss, E.E. Weinberger, C. Boehler, J. Thalhamer, S. [47] J. Manosroi, W. Lohcharoenkal, F. Goetz, R.G. Werner, W. Manosroi, A. Manosroi, Scheiblhofer, Transcutaneous delivery of CpG-adjuvanted allergen via laser- Transdermal absorption and stability enhancement of salmon calcitonin by Tat generated micropores, Vaccine 31 (2013) 3427–3434. peptide, Drug Dev. Ind. Pharm. 39 (2013) 520–525. [19] R. Weiss, M. Hessenberger, S. Kitzmuller, D. Bach, E.E. Weinberger, W.D. [48] J.B. Rothbard, S. Garlington, Q. Lin, T. Kirschberg, E. Kreider, P.L. McGrane, et al., Krautgartner, et al., Transcutaneous vaccination via laser microporation, J. Control. Conjugation of arginine oligomers to cyclosporin A facilitates topical delivery and Release 162 (2012) 391–399. inhibition of inflammation, Nat. Med. 6 (2000) 1253–1257. [20] A.K. Banga, S. Bose, T.K. Ghosh, Iontophoresis and electroporation: comparisons [49] T. Uchida, T. Kanazawa, M. Kawai, Y. Takashima, H. Okada, Therapeutic effects on and contrasts, Int. J. Pharm. 179 (1999) 1–19. atopic dermatitis by anti-RelA short interfering RNA combined with functional [21] V. Regnier, N. De Morre, A. Jadoul, V. Preat, Mechanisms of a phosphorothioate ol- peptides Tat and AT1002, J. Pharmacol. Exp. Ther. 338 (2011) 443–450. igonucleotide delivery by skin electroporation, Int. J. Pharm. 184 (1999) 147–156. [50] X. Yi, G. Zhao, H. Zhang, D. Guan, R. Meng, Y. Zhang, et al., MITF-siRNA formulation [22] L. Zhang, E. Nolan, S. Kreitschitz, D.P. Rabussay, Enhanced delivery of naked DNA to is a safe and effective therapy for human melasma, Mol. Ther. 19 (2011) 362–371. the skin by non-invasive in vivo electroporation, Biochim. Biophys. Acta Gen. Subj. [51] A.U. Bielinska, A. Yen, H.L. Wu, K.M. Zahos, R. Sun, N.D. Weiner, et al., Application of 1572 (2002) 1–9. membrane-based dendrimer/DNA complexes for solid phase transfection in vitro [23] A. Jadoul, J. Bouwstra, V. Preat, Effects of iontophoresis and electroporation on the and in vivo, Biomaterials 21 (2000) 877–887. stratum corneum—review of the biophysical studies, Adv. Drug Deliv. Rev. 35 [52] V.V.K. Venuganti, M. Saraswathy, C. Dwivedi, R.S. Kaushik, O.P. Perumal, Topical (1999) 89–105. gene silencing by iontophoretic delivery of an antisense oligonucleotide- [24] K. Kigasawa, K. Kajimoto, S. Hama, A. Saito, K. Kanamura, K. Kogure, Noninvasive dendrimer nanocomplex: the proof of concept in a skin cancer mouse model, delivery of siRNA into the epidermis by iontophoresis using an atopic dermatitis- Nanoscale 7 (2015) 3903–3914. like model rat, Int. J. Pharm. 383 (2010) 157–160. [53] R.J. Hay, N.E. Johns, H.C. Williams, I.W. Bolliger, R.P. Dellavalle, D.J. Margolis, et al., [25] K. Kigasawa, K. Kajimoto, T. Nakamura, S. Hama, K. Kanamura, H. Harashima, et al., The global burden of skin disease in 2010: an analysis of the prevalence and impact Noninvasive and efficient transdermal delivery of CpG-oligodeoxynucleotide for of skin conditions, J. Investig. Dermatol. 134 (2014) 1527–1534. cancer immunotherapy, J. Control. Release 150 (2011) 256–265. [54] D.R. Bickers, H.W. Lim, D. Margolis, M.A. Weinstock, C. Goodman, E. Faulkner, et al., [26] I.I. Abu Hashim, K. Motoyama, A.-E.H. Abd-ElGawad, M.H. El-Shabouri, T.M. Borg, The burden of skin diseases: 2004: a joint project of the American Academy of H. Arima, Potential use of iontophoresis for transdermal delivery of NF-kappa B Dermatology Association and the Society for Investigative Dermatology, J. Am. decoy oligonucleotides, Int. J. Pharm. 393 (2010) 127–134. Acad. Dermatol. 55 (2006) 490–500. [27] C. Brus, P. Santi, P. Colombo, T. Kissel, Distribution and quantification oligodeoxy- [55] Visiongain, Dermatological drugs market forecast 2014–2024: future prospects for nucleotide complexes of polyethylenimine in human skin after iontophoretic leading companies, 2014 272. delivery using confocal scanning laser microscopy, J. Control. Release 84 (2002) [56] Ltd. IR, Global cosmeceuticals market 2015–2019, 2014 67. 171–181. [57] I. Cosmatos, A. Matcho, R. Weinstein, M.O. Montgomery, P. Stang, Analysis of pa- [28] A. Tezel, S. Dokka, S. Kelly, G.E. Hardee, S. Mitragotri, Topical delivery of anti-sense ol- tient claims data to determine the prevalence of hidradenitis suppurativa in the igonucleotides using low-frequency sonophoresis, Pharm. Res. 21 (2004) 2219–2225. United States, J. Am. Acad. Dermatol. 68 (2013) 412–419. [29] M.A. Tran, R. Gowda, E.-J. Park, J. Adair, N. Smith, M. Kester, et al., Noninvasive drug [58] V. Shahi, A. Alikhan, B.G. Vazquez, A.L. Weaver, M.D. Davis, Prevalence of delivery using ultrasound: targeting melanoma using siRNA against mutant hidradenitis suppurativa: a population-based study in Olmsted County, Minnesota, (V600E) B-Raf, in: E.S. Ebbini (Ed.), 8th International Symposium on Therapeutic Dermatology 229 (2014) 154–158. Ultrasound 2009, pp. 423–427. [59] L. Matusiak, A. Bieniek, J.C. Szepietowski, Psychophysical aspects of hidradenitis [30] K.S. Siu, D. Chen, X. Zheng, X. Zhang, N. Johnston, Y. Liu, et al., Non-covalently func- suppurativa, Acta Derm. Venereol. 90 (2010) 264–268. tionalized single-walled carbon nanotube for topical siRNA delivery into melano- [60] S. Yazdanyar, G.B.E. Jemec, Hidradenitis suppurativa: a review of cause and treat- ma, Biomaterials 35 (2014) 3435–3442. ment, Curr. Opin. Infect. Dis. 24 (2011) 118–123. [31] S. Ozbas-Turan, J. Akbuga, Plasmid DNA-loaded chitosan/TPP nanoparticles for top- [61] E. Mozeika, M. Pilmane, B.M. Nurnberg, G.B.E. Jemec, Tumour necrosis factor-alpha ical gene delivery, Drug Deliv. 18 (2011) 215–222. and matrix metalloproteinase-2 are expressed strongly in hidradenitis suppurativa, [32] Z.R. Cui, R.J. Mumper, Chitosan-based nanoparticles for topical genetic immuniza- Acta Derm. Venereol. 93 (2013) 301–304. tion, J. Control. Release 75 (2001) 409–419. [62] C. Schlapbach, T. Haenni, N. Yawalkar, R.E. Hunger, Expression of the IL-23/Th17 path- [33] P.R. Desai, S. Marepally, A.R. Patel, C. Voshavar, A. Chaudhuri, M. Singh, Topical de- way in lesions of hidradenitis suppurativa, J. Am. Acad. Dermatol. 65 (2011) 790–798. livery of anti-TNF alpha siRNA and capsaicin via novel lipid-polymer hybrid nano- [63] V. Harde, U. Mrowietz, Treatment of severe recalcitrant hidradenitis suppurativa particles efficiently inhibits skin inflammation in vivo, J. Control. Release 170 with adalimumab, J. Dtsch. Dermatol. Ges. 7 (2009) 139–141. (2013) 51–63. [64] L. Grine, L. Dejager, C. Libert, R.E. Vandenbroucke, An inflammatory triangle in pso- [34] S. Bracke, M. Carretero, S. Guerrero-Aspizua, E. Desmet, N. Illera, M. Navarro, et al., riasis: TNF, type I IFNs and IL-17, Cytokine Growth Factor Rev. 26 (2015) 25–33. Targeted silencing of DEFB4 in a bioengineered skin-humanized mouse model for [65] FDA, Search Orphan Drug Designations and Approvals, http://wwwaccessdatafdagov/. psoriasis: development of siRNA SECosome-based novel therapies, Exp. Dermatol. [66] NIH, Genetic and Rare Diseases Information Center: Skin Diseases, https:// 23 (2014) 199–201. rarediseasesinfonihgov/gard/diseases-by-category/22/skin-diseases. [35] J. Li, X. Li, Y. Zhang, X.K. Zhou, H.S. Yang, X.C. Chen, et al., Gene therapy for psoriasis [67] H. Sadeghi, A. Lockmann, A.C. Hund, U. Samavedam, E. Pipi, K. Vafia, et al., Caspase- in the K14-VEGF transgenic mouse model by topical transdermal delivery of 1-independent IL-1 release mediates blister formation in autoantibody-induced interleukin-4 using ultradeformable cationic liposome, J. Gene Med. 12 (2010) tissue injury through modulation of endothelial adhesion molecules, J. Immunol. 481–490. 194 (2015) 3656–3663. [36] S.T. Kim, K.-M. Lee, H.-J. Park, S.-E. Jin, W.S. Ahn, C.-K. Kim, Topical delivery of [68] S.D. Atkinson, V.E. McGilligan, H. Liao, I. Szeverenyi, F.J.D. Smith, C.B.T. Moore, et al., interleukin-13 antisense oligonucleotides with cationic elastic liposome for the Development of allele-specific therapeutic siRNA for keratin 5 mutations in treatment of atopic dermatitis, J. Gene Med. 11 (2009) 26–37. epidermolysis bullosa simplex, J. Investig. Dermatol. 131 (2011) 2079–2086. [37] J.I. Cutler, E. Auyeung, C.A. Mirkin, Spherical nucleic acids, J. Am. Chem. Soc. 134 [69] V.E. McGillan, S. Atkinson, H. Liao, T. Moore, I. McLean, Mutation-specific siRNA (2012) 1376–1391. therapy for epidermolysis bullosa simplex, J. Investig. Dermatol. 130 (2010) S91-S. [38] D. Zheng, D.A. Giljohann, D.L. Chen, M.D. Massich, X.Q. Wang, H. Iordanov, et al., [70] B. Geusens, T. Strobbe, S. Bracke, P. Dynoodt, N. Sanders, M. Van Gele, et al., Lipid- Topical delivery of siRNA-based spherical nucleic acid nanoparticle conjugates mediated gene delivery to the skin, Eur. J. Pharm. Sci. 43 (2011) 199–211. for gene regulation, Proc. Natl. Acad. Sci. U. S. A. 109 (2012) 11975–11980. [71] M.J. Eliason, S.A. Leachman, Feng B-j, M.E. Schwartz, C.D. Hansen, A review of the [39] K.L. Young, A.W. Scott, L. Hao, S.E. Mirkin, G. Liu, C.A. Mirkin, Hollow spherical clinical phenotype of 254 patients with genetically confirmed pachyonychia nucleic acids for intracellular gene regulation based upon biocompatible silica congenita, J. Am. Acad. Dermatol. 67 (2012) 680–686. shells, Nano Lett. 12 (2012) 3867–3871. [72] B.R. Liu, J.F. Li, S.W. Lu, H.J. Lee, Y.W. Huang, K.B. Shannon, et al., Cellular internal- [40] K. Lewandowski, W. Daniel, R. Kang, D. Giljohann, C. Mirkin, A.S. Paller, Topically ization of quantum dots noncovalently conjugated with arginine-rich cell- delivered spherical nucleic acid nanoconjugates targeting TNF improve the psori- penetrating peptides, J. Nanosci. Nanotechnol. 10 (2010) 6534–6543. atic phenotype, J. Investig. Dermatol. 135 (2015) S71-S. [73] R.P. Hickerson, M.A. Flores, D. Leake, M.F. Lara, C.H. Contag, S.A. Leachman, et al., [41] P.S. Randeria, M.A. Seeger, X.-Q. Wang, H. Wilson, D. Shipp, C.A. Mirkin, et al., Use of self-delivery siRNAs to inhibit gene expression in an organotypic siRNA-based spherical nucleic acids reverse impaired wound healing in diabetic pachyonychia congenita model, J. Investig. Dermatol. 131 (2011) 1037–1044. mice by ganglioside GM3 synthase knockdown, Proc. Natl. Acad. Sci. U. S. A. 112 [74] R.P. Hickerson, F.J.D. Smith, W.H.I. McLean, M. Landthaler, R.E. Leube, R.L. Kaspar, (2015) 5573–5578. SiRNA-mediated selective inhibition of mutant keratin mRNAs responsible for [42] Y. Chen, Y. Shen, X. Guo, C. Zhang, W. Yang, M. Ma, et al., Transdermal protein de- the skin disorder pachyonychia congenita, in: T. Tuschl, J. Rossi (Eds.), Oligonucle- livery by a coadministered peptide identified via phage display, Nat. Biotechnol. 24 otide Therapeutics 2006, pp. 56–61. (2006) 455–460. [75] S.A. Leachman, R.P. Hickerson, P.R. Hull, F.J.D. Smith, L.M. Milstone, E.B. Lane, et al., [43] C.-M. Lin, K. Huang, Y. Zeng, X.-C. Chen, S. Wang, Y. Li, A simple, noninvasive and Therapeutic siRNAs for dominant genetic skin disorders including pachyonychia efficient method for transdermal delivery of siRNA, Arch. Dermatol. Res. 304 congenita, J. Dermatol. Sci. 51 (2008) 151–157. (2012) 139–144. [76] S.A. Leachman, R.P. Hickerson, M.E. Schwartz, E.E. Bullough, S.L. Hutcherson, K.M. [44] T. Hsu, S. Mitragotri, Delivery of siRNA and other macromolecules into skin and cells Boucher, et al., Mutation-specific siRNA resolves callus in a pachyonychia using a peptide enhancer, Proc. Natl. Acad. Sci. U. S. A. 108 (2011) 15816–15821. congenita patient, J. Investig. Dermatol. 129 (2009) S54-S. [45] M. Chen, M. Zakrewsky, V. Gupta, A.C. Anselmo, D.H. Slee, J.A. Muraski, et al., Top- [77] T. Uchida, W. Fujimoto, Silencing disease-causing mutant K6a gene in a Japanese ical delivery of siRNA into skin using SPACE-peptide carriers, J. Control. Release 179 patient with pachyonychia congenita via short interfering RNA, J. Investig. (2014) 33–41. Dermatol. 133 (2013) S141-S.

Please cite this article as: M. Zakrewsky, et al., Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges, J. Control. Release (2015), http://dx.doi.org/10.1016/j.jconrel.2015.09.017 12 M. Zakrewsky et al. / Journal of Controlled Release xxx (2015) xxx–xxx

[78] E. Sprecher, S. Chavanas, J.J. DiGiovanna, S. Amin, K. Nielsen, J.S. Prendiville, et al., [101] R. Eritja, M. Terrazas, S. Grijalvo, A. Avino, A. Alagia, S. Perez-Rentero, et al., Chal- The spectrum of pathogenic mutations in SPINK5 in 19 families with Netherton lenges and opportunities for oligonucleotide-based therapeutics by antisense and syndrome: implications for mutation detection and first case of prenatal diagnosis, RNA interference mechanisms, Springer-Verlag Berlin, Berlin, 2014. J. Investig. Dermatol. 117 (2001) 179–187. [102] S. Indermun, R. Luttge, Y.E. Choonara, P. Kumar, L.C. du Toit, G. Modi, et al., Current [79] A. Abdul-Wahab, W. Qasim, J.A. McGrath, Gene therapies for inherited skin disor- advances in the fabrication of microneedles for transdermal delivery, J. Control. Re- ders, Semin. Cutan. Med. Surg. 33 (2014) 83–90. lease 185 (2014) 130–138. [80] D. Roedl, V. Oji, J.T.M. Buters, H. Behrendt, M. Braun-Falco, rAAV2-mediated resto- [103] X. Hong, L. Wei, F. Wu, Z. Wu, L. Chen, Z. Liu, et al., Dissolving and biodegradable ration of LEKTI in LEKTI-deficient cells from Netherton patients, J. Dermatol. Sci. 61 microneedle technologies for transdermal sustained delivery of drug and vaccine, (2011) 194–198. Drug Des. Dev. Ther. 7 (2013) 945–952. [81] R.S. Stern, T. Nijsten, S.R. Feldman, D.J. Margolis, T. Rolstad, Psoriasis is common, [104] S. Henry, D.V. McAllister, M.G. Allen, M.R. Prausnitz, Microfabricated microneedles: carries a substantial burden even when not extensive, and is associated with wide- a novel approach to transdermal drug delivery, J. Pharm. Sci. 87 (1998) 922–925. spread treatment dissatisfaction, J. Investig. Dermatol. Symp. Proc. 9 (2004) 136–139. [105] P. Karande, A. Jain, S. Mitragotri, Discovery of transdermal penetration enhancers [82] M. Rosenbach, S. Hsu, N.J. Korman, M.G. Lebwohl, M. Young, B.E. Bebo Jr., et al., by high-throughput screening, Nat. Biotechnol. 22 (2004) 192–197. Treatment of erythrodermic psoriasis: from the medical board of the National Pso- [106] P. Karande, S. Mitragotri, Enhancement of transdermal drug delivery via synergistic riasis Foundation, J. Am. Acad. Dermatol. 62 (2010) 655–662. action of chemicals, Biochim. Biophys. Acta 2009 (1788) 2362–2373. [83] S. Marepally, C.H.A. Boakye, A.R. Patel, C. Godugu, R. Doddapaneni, P.R. Desai, et al., [107] S. Kumar, M. Zakrewsky, M. Chen, S. Menegatti, J.A. Muraski, S. Mitragotri, Peptides Topical administration of dual siRNAs using fusogenic lipid nanoparticles for as skin penetration enhancers: mechanisms of action, J. Control. Release 199 treating psoriatic-like plaques, Nanomedicine 9 (2014) 2157–2174. (2015) 168–178. [84] K. Ghoreschi, P. Thomas, S. Breit, M. Dugas, R. Mailhammer, W. van Eden, et al., [108] R. Alvarez-Roman, A. Naik, Y. Kalia, R.H. Guy, H. Fessi, Skin penetration and distri- Interleukin-4 therapy of psoriasis induces Th2 responses and improves human au- bution of polymeric nanoparticles, J. Control. Release 99 (2004) 53–62. toimmune disease, Nat. Med. 9 (2003) 40–46. [109] S.N. Ciotti, N. Weiner, Follicular liposomal delivery systems, J. Liposome Res. 12 [85] D.Y.M. Leung, T. Bieber, Atopic dermatitis, Lancet 361 (2003) 151–160. (2002) 143–148. [86] M. Azuma, P. Ritprajak, M. Hashiguchi, Topical application of siRNA targeting cuta- [110] R.M. Hoffman, Topical liposome targeting of dyes, melanins, genes, and proteins neous dendritic cells in allergic skin disease, in: W.P. Min, T. Ichim (Eds.),Rna Inter- selectively to hair follicles, J. Drug Target. 5 (1998) 67–74. ference: From Biology to Clinical Applications 2010, pp. 373–381. [111] L.N. Li, R.M. Hoffman, Topical liposome delivery of molecules to hair follicles in [87] P. Ritprajak, M. Hashiguchi, M. Azuma, Topical application of cream-emulsified mice, J. Dermatol. Sci. 14 (1997) 101–108. CD86 siRNA ameliorates allergic skin disease by targeting cutaneous dendritic [112] H. Wosicka, K. Cal, Targeting to the hair follicles: current status and potential, J. cells, Mol. Ther. 16 (2008) 1323–1330. Dermatol. Sci. 57 (2010) 83–89. [88] J. Inoue, Y. Aramaki, Suppression of skin lesions by transdermal application of CpG- [113] H.L. Wu, C. Ramachandran, A.U. Bielinska, K. Kingzett, R. Sun, N.D. Weiner, et al., oligodeoxynucleotides in NC/Nga mice, a model of human atopic dermatitis, J. Topical transfection using plasmid DNA in a water-in-oil nanoemulsion, Int. J. Immunol. 178 (2007) 584–591. Pharm. 221 (2001) 23–34. [89] E. Emanuele, M. Bertona, D. Geroldi, A multilocus candidate approach identifies [114] A. Domashenko, S. Gupta, G. Cotsarelis, Efficient delivery of transgenes to human ACE and HIF1A as susceptibility genes for cellulite, J. Eur. Acad. Dermatol. Venereol. hair follicle progenitor cells using topical lipoplex, Nat. Biotechnol. 18 (2000) 24 (2010) 930–935. 420–423. [90] D. Hexsel, M. Soirefmann, Cosmeceuticals for cellulite, Semin. Cutan. Med. Surg. 30 [115] S. Dokka, S.R. Cooper, S. Kelly, G.E. Hardee, J.G. Karras, Dermal delivery of topically (2011) 167–170. applied oligonucleotides via follicular transport in mouse skin, J. Investig. [91] N. Tsuji, S. Moriwaki, Y. Suzuki, Y. Takema, G. Imokawa, The role of elastases secret- Dermatol. 124 (2005) 971–975. ed by fibroblasts in wrinkle formation: implication through selective inhibition of [116] C. Dou, F. Lay, A.M. Ansari, D.J. Rees, A.K. Ahmed, O. Kovbasnjuk, et al., Strengthen- elastase activity, Photochem. Photobiol. 74 (2001) 283–290. ing the skin with topical delivery of keratinocyte growth factor-1 using a novel [92] F.M. Freitag, T.F. Cestari, L.R. Leopoldo, P. Paludo, J.C. Boza, Effect of melasma on DNA plasmid, Mol. Ther. 22 (2014) 752–761. quality of life in a sample of women living in southern Brazil, J. Eur. Acad. [117] H.R. Fan, Q. Lin, G.R. Morrissey, P.A. Khavari, Immunization via hair follicles by top- Dermatol. Venereol. 22 (2008) 655–662. ical application of naked DNA to normal skin, Nat. Biotechnol. 17 (1999) 870–872. [93] D. KrupaShankar, V. Somani, M. Kohli, J. Sharad, A. Ganjoo, S. Kandhari, et al., A [118] D.D. Verma, S. Verma, G. Blume, A. Fahr, Particle size of liposomes influences der- cross-sectional, multicentric clinico-epidemiological study of melasma in India, mal delivery of substances into skin, Int. J. Pharm. 258 (2003) 141–151. Dermatol. Ther. (Heidelb) 4 (2014) 71–81. [119] M. Kirjavainen, A. Urtti, I. Jaaskelainen, T.M. Suhonen, P. Paronen, R. [94] J.C. Weaver, T.E. Vaughan, Y. Chizmadzhev, Theory of electrical creation of aqueous ValjakkaKoskela, et al., Interaction of liposomes with human skin in vitro—the in- pathways across skin transport barriers, Adv. Drug Deliv. Rev. 35 (1999) 21–39. fluence of lipid composition and structure, Biochim. Biophys. Acta Lipids Lipid [95] S.-O. Choi, Y.C. Kim, J.-H. Park, J. Hutcheson, H.S. Gill, Y.-K. Yoon, et al., An electrical- Metab. 1304 (1996) 179–189. ly active microneedle array for electroporation, Biomed. Microdevices 12 (2010) [120] B. Geusens, M. Van Gele, S. Braat, S.C. De Smedt, M.C.A. Stuart, T.W. Prow, et al., 263–273. Flexible nanosomes (SECosomes) enable efficient siRNA delivery in cultured pri- [96] L. Daugimont, N. Baron, G. Vandermeulen, N. Pavselj, D. Miklavcic, M.-C. Jullien, mary skin cells and in the viable epidermis of ex vivo human skin, Adv. Funct. et al., Hollow microneedle arrays for intradermal drug delivery and DNA electropo- Mater. 20 (2010) 4077–4090. ration, J. Membr. Biol. 236 (2010) 117–125. [121] G. Cevc, D. Gebauer, Hydration-driven transport of deformable lipid vesicles [97] K. Yan, H. Todo, K. Sugibayashi, Transdermal drug delivery by in-skin electropora- through fine pores and the skin barrier, Biophys. J. 84 (2003) 1010–1024. tion using a microneedle array, Int. J. Pharm. 397 (2010) 77–83. [122] J. Lademann, H. Richter, M.C. Meinke, B. Lange-Asschenfeldt, C. Antoniou, W.C. [98] B. Chen, J. Wei, C. Iliescu, Sonophoretic enhanced microneedles array (SEMA)-Im- Mak, et al., Drug delivery with topically applied nanoparticles: science fiction or re- proving the efficiency of transdermal drug delivery, Sensors Actuators B Chem. 145 ality? Skin Pharmacol. Physiol. 26 (2013) 227–233. (2010) 54–60. [123] R.J. Pettis, A.J. Harvey, Microneedle delivery: clinical studies and emerging medical [99] F. Milletti, Cell-penetrating peptides: classes, origin, and current landscape, Drug applications, Ther. Deliv. 3 (2012) 357–371. Discov. Today 17 (2012) 850–860. [100] B.R. Meade, K. Gogoi, A.S. Hamil, C. Palm-Apergi, Berg Avd, J.C. Hagopian, et al., Ef- ficient delivery of RNAi prodrugs containing reversible charge-neutralizing phosphotriester backbone modifications, Nat. Biotechnol. 32 (2014) 1256–1261.

Please cite this article as: M. Zakrewsky, et al., Nucleic acid delivery into skin for the treatment of skin disease: Proofs-of-concept, potential impact, and remaining challenges, J. Control. Release (2015), http://dx.doi.org/10.1016/j.jconrel.2015.09.017