Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Dec 1.
Published in final edited form as: Ther Deliv. 2015 Feb;6(2):217–229. doi: 10.4155/tde.14.110

Current and emerging formulation strategies for the effective transdermal delivery of HIV inhibitors

Anthony S Ham 1,*, Robert W Buckheit Jr 1
PMCID: PMC4384645  NIHMSID: NIHMS667136  PMID: 25690088

Abstract

Current and emerging formulation strategies for skin permeation are poised to open the transdermal drug delivery to a broader range of small molecule compounds that do not fit the traditional requirements for successful transdermal drug delivery, allowing the development of new patch technologies to deliver antiretroviral drugs that were previously incapable of being delivered through transdermal means. Transdermal drug delivery offers several distinct advantages over traditional dosage forms. Current antiretroviral drugs used for the treatment of HIV infection include a variety of highly active small molecule compounds with significantly limited skin permeability, and thus new and novel means of enhancing transport through the skin are needed. Current and emerging formulation strategies are poised to open the transdermal drug delivery to a broader range of compounds that do not fit the traditional requirements for successful transdermal drug delivery, allowing the development of new patch technologies to deliver antiretroviral drugs that were previously incapable of being delivered through transdermal means. Thus, with continuing research into skin permeability and patch formulation strategies, there is a large potential for antiretroviral transdermal drug delivery.

The challenges of transdermal drug delivery

The expanding transdermal drug delivery market is expected to reach $32 billion in 2015 from a market worth $12.7 billion in 2005 [1]. Yet, only 12 drugs comprise this entire commercial transdermal product market [2]. Since 1979, when scopolamine was approved by the US Food and Drug Administration (FDA) for the treatment of motion sickness [3], clonidine, estradiol, levonorgestrel, fentanyl, granisetron, methylpheni-date, nicotine, nitroglycerin, oxybutynin, rivastigmine, selegiline and testosterone have been approved for use in humans [2,46]. The small number of approved products constituting this group of approved drugs is primarily related to the selective nature of the skin barrier. The skin barrier limits the systemic delivery of drugs at therapeutically relevant rates to those that have high potency, moderate lipophilicity and low-molecular weight [7]. However, a large number of drugs do not conform to these selective physico-chemical and bioactivity criteria. Despite this, advances in delivery technologies have resulted in an increasing number of drugs being administered transdermally including hydrophobic small molecules, hydrophilic molecules and macromolecules. Recently, patents have been issued in transdermal delivery systems for antiemetic medication, transdermal buprenorphine to treat pain resulting from sickle cell crisis, transdermal delivery of nonsteroidal anti-inflammatory drugs and a transdermal delivery system for water insoluble drugs [6]. Transdermal delivery is also currently being investigated as the formulation vector for topical pain relief, hormone therapy, cardiovascular disease, anti-inflammatory and anti-HIV therapy [811]. Despite the significant development challenges of drug delivery through the skin, transdermal drug delivery remains a significant and attractive area of research to the pharmaceutical industry for a variety of reasons. Transdermal products are attractive because they offer numerous advantages over other routes of drug delivery. One of the largest qualitative advantages of transdermal delivery is the convenient administration of drug. Transdermal delivery eliminates frequent dosing administration and pharmacokinetic ‘peaks and valleys’ in plasma drug concentrations associated with oral and intravenous injectable products. Transdermal delivery provides a constant and sustained release of drug through the skin to maintain a stable drug concentration in the blood. Thus, even drugs with a short plasma half-life can be easily delivered via a transdermal route. Additionally drugs with limited plasma half-life could be administered transdermally at a more frequent regimen to compensate for the limited bioactivity of drug delivered from the dosage form. Additionally, formulations such as transdermal patches allow for the simultaneous dosing of multiple drug products, on-demand or variable-rate drug delivery, and are compatible with most drug regimens. Another advantage to transdermal delivery is the avoidance of hepatic first-pass metabolism and gastrointestinal tract exposure. The avoidance of first-pass metabolism allows for a lower amount of drug to be administered resulting in reduced potential for adverse side effects. All these advantages of transdermal administration of drugs importantly contribute to an overall increase in patient adherence to transdermal drug delivery over other forms of drug delivery [1215].

The challenges of transdermal delivery of HIV inhibitors

Over thirty antiviral drugs have been approved for use in HIV-1-infected patients [16] with vast majority of drugs being small molecules (MW ≤ 750 Da). The FDA approved list includes (Table 1): nucleoside reverse transcriptase inhibitors (NRTIs) (abacavir, didano-sine, emtricitabine, lamivudine, stavudine, tenofovir disoproxil fumarate and zidovudine), non-nucleoside reverse transcriptase inhibitors (NNRTIs) (delavirdine, efavirenz, etravirine, nevirapine, dapiverine and rilpivirine), protease inhibitors (PIs) (atazanavir, darunavir, fosamprenavir, nelfinavir, ritonavir, saquinavir and tipranavir), fusion inhibitors (FIs) (enfuvirtide), entry inhibitors (EIs) (maraviroc) and integrase inhibitors (IIs) (dolutegravir and raltegravir). In addition, a wide variety of small molecule compounds targeting both well-defined as well as novel viral and cellular targets continue in preclinical and clinical development (Table 1) [1719].

Table 1.

HIV inhibitor transdermally relevant physicochemical properties.

Name ABV MW (Da) LogP MP (C)
NRTI Abacavir ABC 286.33 0.9 165
Didanosine ddI 236.23 -1.24 160
Emtricitabine FTC 247.25 -0.6 136
Lamivudine 3TC 229.26 -0.9 160
Tenofovir disoproxil fumarate TDF 287.21 1.25 276
Zidovudine AZT 267.24 0.05 106

NNRTI Delavirdine DLV 456.56 2.34 226
Dfavirenz EFV 315.67 4.7 139
Etravirine ETR 434.05 4.5 265
Nevirapine NVP 266.3 2.0 247
Dapivirine TMC-120 329.4 4.8 219
Rilpivirine RPV 366.42 4.5 242

PI Atazanavir ATV 704.86 5.6 195
Darunavir DRV 547.66 2.9 74
Fosamprenavir IDV 585.61 1.8 282
Nelfinavir NFV 567.78 5.7 350
Ritonavir RTV 720.94 6 120
Sasquinavir SQV 670.84 4.2 350
Tipranavir TPV 602.66 7 86

FI Enfuvirtide T-20 4491.88 -14.7 N/A

EI Maraviroc MVC 513.66 5.1 197

II Dolutegravir DTG 419.38 2.4 188
Raltegravir RAL 444.42 1.1 155

Under development IQP-0528 - 340.42 4.1 216
UC-781 - 335.85 4.9 157
ISIS 5320 - 2584.09 -4.6

To design a transdermal drug delivery system for HIV inhibition, a thorough understanding of the skin and the barrier properties of the stratum corneum is critical [20]. Transport across the skin is primarily driven by diffusion, which is governed by the physicochemical properties of the drug and the barrier through which the drug diffuses. The drug diffuses from the dosage form to the surface of the stratum corneum boundary layer and partitions into the lipid and tissue domains of the lower epidermis and dermis layers. The diffusion of drug is controlled by its skin permeability or the rate at which drug concentration transports into the epidermal tissue. The drug then diffuses through the dermal lipid layers into the microcirculation provided by the local capillary network, which provides and infinite sink for drug absorption. This repeated diffusion-partition process drives the transport of drug into and out of the dermal tissue. Drugs which have been successfully delivered through the skin share similar physicochemical properties, which include low molecular weight, possession of both lipophilic and hydrophilic phases and a low melting point. Since drug delivery through the skin is diffusion driven, the molecular weight of the drug administered has a significant impact on delivery; with smaller molecules having a greater rate of delivery over large molecules. With approved and experimental HIV inhibitors, the majority of the drugs are below 1000 Da (with the exception of Fuzeon (Table 1). Optimal drugs for transdermal drug delivery are ones that contain both lipophilic and hydrophilic phase in order to pass though both the stratum corneum and lipid-rich dermal layer to the underlying capillary system. This transport and partition is defined by the partition coefficient (Log P) and the solubility (melting point). It is well recognized that the lower the melting point of a material, the greater its solubility in a given solvent – including solubility in skin lipids. In the case of HIV inhibitors, with the exception of DRV, TPV and T-20, the melting points of all the drugs exceeds 100°C. However, the one factor that has the greatest influence on drug transport through the skin is the ability of the drug to partition into the tissue. Typically, drugs with Log P values in the range of 2–3 show optimal permeability across the stratum corneum as well as moderate partitioning out of the stratum corneum. Conversely, drugs with a Log P > 3 are expected to exhibit high partitioning into the stratum corneum but poor partition into the systemic circulation [21]. Lipo-philic drugs have a higher residence time and thus a lower systemic exposure rate. Most of the current HIV inhibitors have Log P values exceeding 4.0, and coupled with their high melting points are insoluble small molecules that show high partitioning into the stratum corneum (Table 1). As a direct result of these physicochemical properties, currently there are no transdermal products available for the treatment of infectious diseases such as HIV. The clinical experience with all HIV agents has clearly demonstrated the ability of HIV to evade the anti-viral effects of any monotherapeutic drug administration strategy through the rapid accumulation of amino acid changes in the targeted proteins [22]. The highly error prone HIV reverse transcriptase, with its lack of proofreading capability, generates significant heterogeneity within the highly related but nonidentical populations quasispace of viruses circulating in a patient [23]. It is widely accepted that most drug resistant viruses pre-exist within the population of viruses and are selected from within this heterogeneous environment upon application of selective drug pressure. In addition to the high levels of resistance possible to single agents, each of the anti-HIV agents employed to date have had significant dose limiting and long-term toxicities which render successful long-term therapy for HIV disease difficult to achieve [22]. It is clear that the continued development of new agents with enhanced potency, reduced toxicity and a greater genetic barrier to resistance, as well as targeting other HIV replication steps, is an important need for the continued effectiveness of HIV therapy. In addition, simplification of patient dosing regimens and enhancements in adherence to drug regimens would provide significant long-term advantages to populations of infected individuals. Transdermal drug delivery would be a suitable strategy to avoid many of these problems by eliminating pharmacokinetic peaks and troughs in drug levels which enable resistant viruses to escape drug selective pressure when concentrations fall below effective levels [24]. Furthermore, transdermal administration of multiple drugs is possible using patch technologies, with the ability to control the rate of delivery of individual drug components and overall plasma drug concentrations. Resistance and toxicity is further controlled by sustained drug concentrations for prolonged periods of time while the patch effectively delivers drug in the absence of the peaks of drug concentration associated with adverse effects.

In administering small molecules as therapeutic agents, one of the biggest obstacles to effective treatment is drug bioavailability. In studies with Zidovu-dine (AZT), the first anti-HIV compound approved for clinical use, the therapeutic effectiveness of the drug was significantly limited due to its dose-dependent hematological toxicity, low therapeutic index, short biological half-life and relatively poor bioavail-ability [25]. Additionally, due to first-pass metabolism, oral bioavailability was low and the dosage required to maintain therapeutic levels often resulted in toxic levels of AZT in the blood and other side effects [26]. Therefore, transdermal drug deliveries were investigated to potentially deliver anti-HIV drugs such as AZT through the skin. A variety of strategies to transdermally deliver drugs has been and continues to be evaluated for HIV specific inhibitors.

Current transdermal delivery strategies

Until recently, the transdermal delivery field was primary focused on passive patch delivery technologies that relied upon simple diffusion of a pharmaceutical product across the skin barrier. Emerging active technology patches that ‘drive’ a drug through the skin barrier have opened transdermal drug delivery to a broader range of compounds that do not fit the traditional requirements for successful transdermal drug delivery, allowing the development of new active patches to deliver antiretroviral drugs that were previously incapable of being delivered through transdermal means. In addition, new strategies are also being developed with current passive patch technology to increase the utility and application of transdermal drug delivery.

Successful dermal administration must ensure the delivery of a drug through the stratum corneum, which is an organ designed to selectively act as a barrier to keep things out of the body. Transport through the epidermis is primarily diffusion driven, governed by the physico-chemical properties of the drug and the barrier itself [27]. The drug released from the patches diffuses through the boundary layer (stratum corneum) to the underlying dermal layer and then into the blood vessels (Figure 1). The rate and ultimate success of a drug to diffuse through the skin is largely dependent upon its molecular weight, partition coefficient (Log P), solubility and polarity. Therefore, for dermal administration to be successful for any potential drug, the drug product ideally should be a small molecule with a molecular weight of 500 Da or less with high lipophilicity to permeate through the skin [7]. Depending on the eventual target of the transdermal delivery, which may be either local to the skin or to the systemic circulation, the melting point and partition coefficient of the drug is important in determining the drug compound's suitability for transdermal delivery by defining whether the drug will be partitioned within the dermis or will transit through the stratum corneum and dermis and thus partition out of the tissue and be absorbed into the systemic circulation [21].

Figure 1.

Figure 1

Illustration of transdermal delivery through intact skin.

In considering transdermal delivery as a possible means of drug dosing, it is important to understand the various components of delivery from the patch. First, the drug must be released from the dosage form, which is controlled by the specific formulation utilized, and then the drug permeates through the skin, which is controlled by the skin and physicochemical properties of the drug. As such, there exist several types of transdermal dosage forms which may be used to control drug release from the patch (Figure 2):

  • Single-layer drug-in-adhesive is the most basic formulation where the rate of drug release from the patch is dependent on the diffusion of the drug product across the skin;

  • Multilayer drug-in-adhesive formulations encompasses products with either the addition of a membrane between two distinct drug-in-adhesive layers or the addition of multiple drug-in-adhesive layers under a single backing film;

  • Drug reservoir formulations are comprised of an inclusion of a liquid compartment containing a drug solution or suspension separated from the release liner by a semipermeable membrane and adhesive;

  • Drug matrix formulations include a semi-solid matrix containing a drug solution or suspension [2830].

Figure 2. Types of transdermal patch formulations.

Figure 2

(A) Single-layer drug-in-adhesive: the rate of release is dependent on the diffusion across the skin. (B) Multi-layer drug-in-adhesive: encompasses either the addition of a membrane between two distinct drug-in-adhesive layers or the addition of multiple drug-in-adhesive layers under a single backing film. (C) Drug reservoir: the inclusion of a liquid compartment containing a drug solution or suspension separated from the release liner by a semipermeable membrane and adhesive. (D) Drug matrix: inclusion of a semisolid matrix containing a drug solution or suspension.

Current research in transdermal drug delivery of HIV antiretroviral products has been primarily focused on the addition of permeability enhancers to the formulation. This strategy allows for the use of existing dosage forms as described above while addressing the second stage of transdermal drug delivery, namely the transport of the drug product through the skin barrier after its release from the formulated patch. In the development of HIV transdermal delivery technologies, chemical and physical permeability enhancers have been utilized in the patch to investigate the relative transport of the antiretroviral agent through the skin once released from a patch dosage form.

The use of passive permeability enhancers in transdermal delivery

Chemical permeability enhancers facilitate drug transport through the skin by increasing drug partitioning into the stratum corneum, increasing drug diffusion through the stratum corneum, or a combination of both physicochemical effects [31]. There are several categories of permeability enhancers [32] that were tested in conjunction with the HIV inhibitor AZT, which is the most common HIV drug thus far evaluated in the investigation of transdermal patch anti-HIV drug delivery. Aprotic solvents such as N-methyl-2-pyrrolidone have been shown to enhance plasma concentrations of AZT for 10 h [33]. Carvacrol, t-anethole, thymol, linalool, t-menthol have been shown to enhance AZT transport through the stratum corneum at 5% drug concentrations. Solvents as permeability enhancers increase skin penetration by utilizing the polar pathways and/or fluidizing lipids. Other permeability enhancers added to AZT-containing transdermal patches include propylene glycol, oleic acid, sodium dodecyl [34], ethanol/water [3537], various binary solutions [38], carbonate prodrugs [39] and liposomes [40,41]. Other than solvents, the primary permeability enhancers are surfactants which are classified according to the nature of their polar head group. Surfactants absorb to the skin interface and serve to bind to and denature skin surface proteins. Surfactants then solubilize the intercellular lipids of the skin to permit enhanced penetration of the transported drug through pathways between the cells. The primary concern with the use of surfactants as transdermal permeability enhancers include skin toxicity and irritation [42].

Other than AZT, relatively few HIV inhibitors have been investigated using transdermal delivery [43]. Zal-citabine (ddC) and Didanosine (ddI) have been investigated for transdermal delivery using the same permeability enhancers as utilized with AZT [3537,44,45]. For these compounds, the permeability enhancer Pheroid was shown to effectively transport the nucleoside RT inhibitors across the skin barrier [46]. With Lamivudine (3TC), MPEG carbamates were used as a permeability enhancer for transport through the stratum corneum, and the formulation was shown to increase aqueous solubility while still maintaining constant lipophilicity. Polymeric ethylene glycol carbonates also showed similar permeability enhancement with AZT with an increase of 2–4-fold in drug flux through the skin [39]. These permeability enhancers have also been integrated into current transdermal patch formulations [34,47] and gel formulations [48,49].

The use of active permeability enhancers in transdermal delivery

Systematic innovations such as iontophoresis have been used to actively drive the transport of small drug molecules across the skin barrier [47,50]. Iontophoresis is a strategy of permeability enhancement using a repulsive electromotive force through the skin via an electric charge [51]. With AZT, iontophoresis was shown to enhance in vitro permeation by 5- to 40-fold over that of passive diffusion [47].

Emerging transdermal delivery strategies

While no current investigations of antiretroviral inhibitors are being conducted with emerging transdermal strategies, there are several strategies that show potential in providing enhanced delivery of antiretroviral agents. Dissolvable microneedles are an emerging strategy that is minimally invasive and which serves to bypass the stratum corneum barrier to permit the drug's direct access to the viable epidermal layer of the skin. These novel microdevices have been developed to puncture the skin to facilitate the transdermal delivery of hydrophilic drugs and macromolecules, including peptides, DNA and other molecules, that would otherwise have difficulty passing the outermost layer of the skin, the stratum corneum [5255].

Dissolving microemulsion hydrogels have also been investigated as a method of delivering poorly permeable and low bioavailability compounds [56,57]. Hydrogels are solid dosage forms that completely dissolve over time when applied to the skin. Hydrogels offer advantages over other semisolid dermal formulations such as creams and gels by acting as a drug depot for sustained drug delivery [58]. Microemulsion hydrogels also represent an alternative to traditional transdermal patch formulations by eliminating the persistence of the patch backing when applied to the skin.

HIV inhibitors & transdermal drug delivery

Despite promising data suggesting the successful use of these permeability enhancers with HIV inhibitors, one of the primary problems with anti-HIV compounds and transdermal delivery has been that research has been performed with relatively low potency inhibitors such as AZT, ddc, ddI and 3TC as opposed to compounds that are effective in picomolar to low nanomolar concentration ranges, such as the highly active nonnucleoside reverse transcriptase inhibitors (NNRTIs).

We have worked extensively with a family of NNRTIs being investigated as potential therapeutic and prevention products based on their bioactivity and safety profile. The pyrimidinedione class of inhibitors are highly potent (sub- to low-nanomolar active concentration ranges) small molecule inhibitors that have a dual mechanism of action against HIV infection. The pyrimidinediones primarily act as steric inhibitors of viral reverse transcriptase by binding to the hydrophobic nonnucleoside binding pocket on the RT, but possess a secondary undefined effect on HIV entry [59]. The pyrimidinedione IQP-0410 was identified as a highly potent (TI > 1 million) antiretroviral therapeutic agent, which was being developed as a product for the treatment of HIV-infected individuals [59,60]. Transdermal delivery of IQP-0410 was considered in light of the rapid first-pass metabolism and short plasma half-life (~5 h) of the drug product. Thus, bypass of hepatic clearance of IQP-0410 was determined to be essential for continued development of IQP-0410 as an effective HIV inhibitor. In considering a transdermal drug delivery strategy for IQP-0410 (and other antiretroviral products), the appropriateness of the compound for dermal transport was carefully evaluated. Successful delivery of any antiretroviral agent through the skin is largely dependent upon having a compound of an appropriate molecular weight, partition coefficient (Log P), solubility and polarity. IQP-0410 is practically insoluble and has a molecular weight of 352.43 g/mol. With a calculated Log P of 3–4, IQP-0410 is nonpolar and thus lipophilic [21]. In vitro antiviral evaluations demonstrated that IQP-0410 was efficacious at subnanomolar concentrations against HIV-1 (0.28 nM) with a resulting therapeutic index of greater than 500,000 [60]. How-ever, in vivo PK and bioavailability studies in mice have shown that IQP-0410 only has a 24% oral bioavailability with a half-life of 5.37 h and an intravenous half-life of 30 minutes. This short systemic residence time can be attributed by extensive first-pass metabolism by the liver. Therefore, bypassing first-pass metabolism via dermal delivery will highly improve the pharmacokinetics of IQP-0410 and allow maintenance of effective concentrations of the active product for prolonged periods of time.

The initial studies of the formulation of the preclinical drug candidate IQP-0410 for transdermal patch delivery were performed by incorporating permeability enhancers and IQP-0410 into the polymer matrix of the transdermal patch. In vitro release studies of IQP-0410 from the transdermal patches were performed through synthetic 0.45 micron hydrophilic polyvinylidene fluoride (PVDF) membranes in Franz cell diffusion cells [61]. The transdermal product was applied flush atop the membrane in the donor cell. The receptor cell was filled with a 1:1 Isopropanol (IPA)/Phosphate Buffered Saline (PBS) solution while the donor cell was wetted with PBS to maintain humidity [62]. For 72 h at 37°C, a linear release of IQP-0410 across the membrane barrier into the receptor cells was measured (Figure 3A). While the flux of IQP-0410 across the membrane is not a true measurement of drug delivery and permeability, the drug transport of IQP-0410 from the transdermal film across the membrane does correspond to a zero-order release kinetic profile [27]. The ex vivo permeability of the IQP-0410 transdermal film was evaluated through full thickness human epidermal tissue model that is comprised of normal, human-derived epidermal keratinocytes (NHEK) which have been cultured to form a multilayered, highly differentiated model of the human epidermis [63]. The transdermal films were placed flush with the apical surface of the epidermal tissue and kept dry. The tissues were maintained in DMEM from the receptor (basal) compartment. When the patches were applied to epidermal tissue, ex vivo studies similarly resulted in a linear zero-order release rate (0.94 ± 0.06 μg/cm2/h) into the basal media over a 3 day application (Figure 3B) [64]. IQP-0410 was successfully released and permeated through the full thickness epidermal tissue and collected in the basal media suggesting the potential viability of controlled zero-order delivery of IQP-0410 through the skin. The in vitro dissolution of the transdermal films and release of IQP-0410 from dosage form were conducted in a USP 4 flow-through cell dissolution system [6567] under sink conditions in a dissolution medium of 10:90 ethanol/DI water at a flow rate of 10 ml/min and 37°C. While the in vitro release of IQP-0410 into dissolution media suggested a 22.2% increased rate from films stored under accelerated storage conditions, the cumulative recovered IQP-0410 was unaffected. As noted above, transdermal drug delivery has two functional components which are essential for overall drug delivery: release from the formulation to the epidermis, and permeation through the skin to the underlying blood vessels. Therefore, any increase in drug release rate from the film formulation will be mitigated by the diffusion of the drug through the tissue. The passive diffusion across the skin, the basis of transdermal drug delivery, will be the limiting factor in the drug delivery and permeability [68]. In the ex vivo studies, 45.9 ± 20.8 μM, 94.5 ± 12.7μM and 134 ± 14.7 μM of IQP-0410 was collected each successive day, respectively, from the basal media resulting in an average anti-HIV 50% efficacious concentration (EC50) value of 2.09 ± 0.43 nM in CEM-SS cells and 1.02 ± 0.56 nM in PBMCs over a 3-day application [64]. Ex vivo studies performed over 3 days determined that the concentration of delivered IQP-0410 after 24 h (46 μM) was 5000-fold greater than in vitro defined EC95 values [60]. The results of the transdermal delivery studies with the NNRTI IQP-0410 demonstrated the delivery of sufficient IQP-0410 through the ex vivo tissue model to yield successful in vitro reduction of HIV-1 replication for over 72 h. The physicochemical properties of IQP-0410 thus provide a more feasible compound for transdermal anti-HIV drug delivery when compared to the drug delivery results obtained with AZT, ddC, ddI and 3TC. Continuing studies with the IQP-0410 patch technology involve evaluation of other pyrimidinedione analogs with greater stability for use in the patches as well as the development of patches that might provide even longer periods of effective drug delivery through the skin and into systemic circulation. Patches are being developed specifically for their ability to lower HIV plasma virus loads in infected individuals and will thus require the eventual release of multiple drug products, as well as the development of strategies to prevent the infection of uninfected individuals through pre-exposure prophylaxis.

Figure 3. Illustration of transdermal delivery through intact skin.

Figure 3

In vitro/ex vivo permeability of the IQP-0410 transdermal film through barrier. The in vitro release and subsequent transport of IQP-0410 from the transdermal film (defined here as a transdermal patch without the associated adhesive backing) through a polyvinylidene fluoride membrane over 72 h (A) (n = 3 ± SD). The ex vivo release and permeability of IQP-0410 from the transdermal films through full thickness epidermal tissue over 72 h (B); (black triangles) (n = 3 ± SD). The transdermal films were packaged into air-tight foil pouches and stored under standard (30°C / 65% RH) and accelerated (40°C / 75% RH) environmental conditions for 3 months. At 1, 2 and 3 months, the transdermal films were removed from storage and tested for drug release and permeability through full thickness epidermal tissue. The combined average (n = 6 ± SD) of the transdermal films stored under standard environmental conditions for 3 months (B); (blue diamonds). The combined average (n = 6 ± SD) of the transdermal films stored under accelerated environmental conditions for 3 months (B); (red squares).

Reproduced from [64] © PLoS ONE (2013).

Transdermal delivery for pre-exposure prophylaxis (PrEP) & HIV prevention

While there is significant development in sustained antiretroviral delivery for the therapeutic treatment of HIV, prevention of HIV infection represents a more effective method of confronting the HIV epidemic. This is especially true in the absence of any effective vaccine in the development pipeline to provide immunity and protect against HIV infection. Therefore, an important strategy under development is to prophylactically inhibit the transmission of STIs, including HIV [69,70], through the use of chemical barriers of sexual transmission. For the most part, the application of such topical microbicides in the form of gels is directly associated with coitus and the gels are designed to be applied as a single dose before and after intercourse to provide effective concentrations of the microbicide product at the time of virus transmission.

As such, these prevention products are directly linked to coitus and the timing of exposure to infectious HIV and, therefore, require highly specific compliance and adherence to the dosing strategy to be effective. It is well understood in the microbicide development field that coital dependency is not optimal and thus coitally associated products may not be the most optimal dosage form for women in all parts of the world. Therefore, sustained/controlled release drug delivery is a dosing mechanism that aims to dissociate drug administration from coitus which will hopefully simplify dosing of antiviral products and increase compliance and subsequent rates of prevention. Currently, the field of sustained release microbicide PrEP is still in the early developmental stages. There exist several vaginal rings under development [7174] along with vaginal tablets [75] that confer sustained drug release. However, all of these sustained release drug delivery systems are applied within the vagina or rectum which may limit their use in geographic and cultural regions where specific dosage forms have low acceptability among potential users of the product. The development of transdermal formulations to deliver antiretroviral drugs for PreP will dissociate prevention from both the reproductive tract and coitus. The results of PrEP clinical studies in populations of men who have sex with men (MSM) confirm the potential utility of PrEP with transdermal delivered products. Males were provided oral dosing of two antiretroviral agents (Tenofovir and FTC) and were found to have a significantly reduced rate of HIV transmission as the oral dosing conferred protection of the men through effective concentrations of drug product in susceptible tissue in the reproductive tract [76]. It would be expected that transdermal delivery of anti-HIV compounds would allow even more effective PrEP in this context since dosing would be sustained over longer periods of time using a simpler regimen, providing even greater adherence to PrEP regimens. The transdermal and oral delivery of antiretroviral drugs also has one additional advantage which cannot be overlooked and which greatly adds to prevention strategies. The effective delivery of drugs to infected individuals with simplified regimens and sustained delivery will yield lower rates of HIV transmission of infectious virus to uninfected individuals. This ‘treatment as prevention’ strategy has been evaluated in clinical studies and it has been clearly shown that the reduced viral loads in treated individuals are directly correlated with reduced rates of HIV transmission [77]. Thus, the use of transdermal patches to simplify drug dosing and provide uniform sustained drug release over longer periods of time would have a direct effect on the transmission of infectious virus from infected individuals.

Future perspective

There have been significant advancements in understanding the mechanism of action for HIV and appropriate preventative/treatment strategies; however, an optimum treatment still remains a major challenge. Combinations of nucleotide and nonnucleoside reverse transcriptase inhibitors and protease inhibitors have been effectively used in highly active antiretroviral therapies (HAART) to significantly reduce HIV viral load in infected individuals for prolonged periods of time. The utilization of HAART has dramatically changed the therapeutic landscape of HIV treatment and the application of cocktails of antiretroviral agents is now the standard of care for HIV patients [78]. The dramatic reduction in viral load and clinical improvements achieved with HAART is a rigorous validation of the ability of anti-HIV drugs to contain and manage HIV-disease, and demonstrates that combination of three or more anti-HIV agents – even when directed against a few of the putative ten viral targets – are superior to single or dual drug therapy. Thus, the prevailing belief is that the addition of new anti-HIV agents to HAART regimens will provide additional clinical benefit. However, HAART still suffers from complications with the emergence of multidrug resistant virus strains, toxicity, drug–drug interactions, difficult treatment regimens and inadequate pharmacology (bioavailability and tissue distribution) [7981]. Clearly there is an urgent need for the continued development of new anti-HIV strategies and therapies. One of these strategies for the evolution of effective HIV therapy may include the use of transdermal patches to deliver drug cocktails using stable sustained release technology.

Conclusion

While alternative formulations such as transdermal drug delivery have been evaluated for HIV inhibitors, the design and development of these drug delivery systems are currently lacking. In addition, emerging transdermal formulation strategies have not yet considered the application of antiretroviral agents. Although several studies have reported the in vitro efficacy of the transdermally delivered compound, there remains a significant lack of data on the clinical applicability and toxicity of transdermal antiretroviral product delivery. Due the complex nature of HIV infection, there are several limitations to currently employed HAART drug therapy. Transdermal drug delivery represents an established alternative dosage form that avoids many of the limitations of existing oral drug delivery. While lower potency approved ARV drugs have been extensively investigated for transdermal delivery, recent evaluations of highly potent ARV drugs such as IQP-0410 and new emerging technologies in transdermal formulation must be pursued to truly understand the potential of transdermal delivery for the prevention and treatment of HIV infection.

Key term.

Skin permeability: Rate of drug concentration transport from the formulated dosage form into the epidermal tissue over time and volume of tissue.

Key term.

Drug partitioning: The measure of the difference in solubility of a compound between two immiscible (hydrophobic / hydrophilic) phases. Drug partitioning or partition coefficient (LogP) is determined by the following:

logPoctanolwater=log([solute]octanol[solute]un-ionised water)

The LogP value is known as a measure of lipophilicity.

Key terms.

Pre-exposure prophylaxis: Prevention strategy where people who do not have HIV but who are at substantial risk of getting it take a drug. When someone is exposed to HIV through sex or injection drug use, PreP can prevent the virus from establishing a permanent infection.

Highly active antiretroviral therapies: Anti-HIV treatments that consist of a combination of at least three drugs that suppress HIV replication. Three drugs are used in order to reduce the likelihood of the virus developing resistance and to maintain virus loads in patients below detectable levels.

Executive summary.

Introduction

  • A large number of drugs do not conform to the selective physicochemical and bioactivity criteria for successful transdermal drug delivery. Despite this, advances in delivery technologies have resulted in an increasing number of drugs being administered transdermally.

The challenges of transdermal delivery of HIV inhibitors

  • Currently there are no transdermal products available for the treatment of infectious diseases such as HIV. Existing research into delivering FDA approved small molecule HIV inhibitors has shown some promise; however, significant work in this field is required.

Current transdermal delivery strategies

  • Until recently, the transdermal delivery field was primary focused on passive patch delivery technologies that relied upon simple diffusion of a pharmaceutical product across the skin barrier. Emerging active technology patches that ‘drive’ a drug through the skin barrier have opened transdermal drug delivery to a broader range of compounds that do not fit the traditional requirements for successful transdermal drug delivery, allowing the development of new active patches to deliver antiretroviral drugs that were previously incapable of being delivered through transdermal means.

The use of passive permeability enhancers in transdermal delivery

  • Chemical permeability enhancers facilitate drug transport through the skin by increasing drug partitioning into the stratum corneum, increasing drug diffusion through the stratum corneum, or a combination of both physicochemical effects. Such passive permeability enhancers have been shown to increase permeability of several FDA approved HIV inhibitors through the skin.

The use of active permeability enhancers in transdermal delivery

  • Active permeability enhancers have been used to actively drive the transport of small drug molecules across the skin barrier. While less convenient in regards to patient uses, active permeability enhancers such as iontophoresis have been show to significant improve transdermal permeability.

Emerging transdermal delivery strategies

  • There is still a large area of transdermal drug delivery that has yet to be applied to antiretrovirals. No current investigations of antiretroviral inhibitors are being conducted with emerging transdermal strategies; however, there are several strategies such as microneedles and microemulsions that show potential in providing enhanced delivery of antiretroviral agents.

  • Despite the challenges in transdermal drug delivery, there are several examples of potentially success formulations to deliver anti-HIV compounds. Continuing studies with the IQP-0410 patch technology involve evaluation of other pyrimidinedione analogs with greater stability for use in the patches as well as the development of patches that might provide even longer periods of effective drug delivery through the skin and into systemic circulation. Patches are being developed specifically for their ability to lower HIV plasma virus loads in infected individuals and will thus require the eventual release of multiple drug products, as well as the development of strategies to prevent the infection of uninfected individuals through pre-exposure prophylaxis.

Conclusion

  • While there is significant development in sustained antiretroviral delivery for the therapeutic treatment of HIV, prevention of HIV infection represents a more effective method of confronting the HIV epidemic. Sustained release microbicide PrEP is still in the early developmental stages; however, several formulations under development have shown promise in preventing infection. This ‘treatment as prevention’ strategy has been evaluated in clinical studies and it has been clearly shown that the reduced viral loads in treated individuals are directly correlated with reduced rates of HIV transmission.

Future perspective

  • There have been significant advancements in understanding the mechanism of action for HIV and appropriate preventative/treatment strategies; however, an optimum treatment still remains a major challenge. While alternative formulations such as transdermal drug delivery have been evaluated for HIV inhibitors, the design and development of these drug delivery systems are currently lacking. In addition, emerging transdermal formulation strategies have not yet considered the application of antiretroviral agents which must be pursued to truly understand the potential of transdermal delivery for the prevention and treatment of HIV infection.

Acknowledgments

The authors also acknowledge financial support for the development of the IQP-0410 transdermal patch described in this manuscript from grant R43 AI089331 from the National Institute of Allergy and Infectious Diseases, National Institutes of Health. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Footnotes

Financial & competing interests disclosure

The authors disclose financial involvement for transdermal patch product development activities as founder and stockholder (RWB) and employment and stock option ownership in ImQuest Life Sciences, the parent company of ImQuest Bio-Sciences, Inc. (ASH). The authors also acknowledge financial Sciences, Inc. (ASH).

No writing assistance was utilized in the production of this manuscript.

References

Papers of special note have been highlighted as:

• of interest

•• of considerable interest

  • 1.Transdermal drug delivery-technologies, markets and companies. Jain PharmaBiotech Report. http://pharmabiotech.ch/reports/tdd.
  • 2.Paudel KS, Milewski M, Swadley CL, Brogden NK, Ghosh P, Stinchcomb AL. Challenges and opportunities in dermal/transdermal delivery. Ther. Deliv. 2010;1(11):109–131. doi: 10.4155/tde.10.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Segal M. Patches, pumps and timed release: new ways to deliver drugs. US FDA Consumer. 2011;25(8):13–15. [Google Scholar]
  • 4.Cadth Rotigotine transdermal patched (Neupro) for the treatment of Parkinson’s disease. Issues Emerg. Health Technol. 2008;(112):1–6. [PubMed] [Google Scholar]
  • 5.Kumar R, Philip A. Modified transdermal technologies: breaking the barriers of drug permeation via the skin. Trop. J. Pharm. Res. 2007;6(1):633–644. [Google Scholar]
  • 6.Shah S. Transdermal drug delivery technology revisited: recent advances. www.Pharmainfo.net.
  • 7.Bos JD, Meinardi MMHM. The 500 Dalton rule for the skin penetration of chemical compounds and drugs. Exp. Dermatol. 2000;9(3):165–169. doi: 10.1034/j.1600-0625.2000.009003165.x. [DOI] [PubMed] [Google Scholar]
  • 8••.Takmaz EA, Inal Ö , Baykara T. Studies on transdermal delivery enhancement of zidovudine AAPS PharmSciTech. 2009;10:88–97. doi: 10.1208/s12249-008-9179-9. [Purpose of this study was to investigate physicochemical characteristics and in vitro release of zidovudine from monolithic film of Eudragit RL 100 and ethyl cellulose.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Patel NA, Patel NJ, Patel RP. Design and evaluation of transdermal drug delivery system for curcumin as an anti-inflammatory drug. Drug Dev. Ind. Parm. 2009;35(2):234–242. doi: 10.1080/03639040802266782. [DOI] [PubMed] [Google Scholar]
  • 10.Nath A, Sitruk-Ware R. Parenteral administration of progestins for hormonal replacement therapy. Eur. J. Contracept. Reprod. Health Care. 2009;14(2):88–96. doi: 10.1080/13625180902747425. [DOI] [PubMed] [Google Scholar]
  • 11.Aqil M, Ali A, Sultana Y, Najmi A. Fabrication and evaluation of polymeric films for transdermal delivery of pinacidil. Pharmazie. 2004;59(8):631–635. [PubMed] [Google Scholar]
  • 12.Cleary G. In: Medical applications of controlled release. In: Transdermal controlled release systems. Langer D, editor. CRC Press; FL, USA: 1984. pp. 203–251. [Google Scholar]
  • 13.Kydonieus A. Controlled Release Technologies. CRC Press; FL, USA: 1987. [Google Scholar]
  • 14.Chien Y. In: Transdermal therapeutic system. In: Controlled Drug delivery fundamentals and applications. Robinson J, Lee V, editors. Marcel Dekker, Inc.; New York, USA: 1987. pp. 524–552. [Google Scholar]
  • 15.Keith A. Polymer matrix consideration for transdermal devices. Drug Dev. Ind. Parm. 1983;9:605–621. [Google Scholar]
  • 16.Lane T, Pettifor A, Pascoe S, Fiamma A, Rees H. Heterosexual anal intercourse increases risk of HIV infection among young South African men. AIDS. 2006;20(1):123–125. doi: 10.1097/01.aids.0000198083.55078.02. [DOI] [PubMed] [Google Scholar]
  • 17.Mahalingam A, Simmons AP, Ugaonkar SR, et al. Vaginal microbicide gel for the delivery of IQP-0528, a pyrimidinedione analog with a dual mechanism of action against HIV-1. Antimicrob Agents Chemother. 2011;55(4):1650–1660. doi: 10.1128/AAC.01368-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Buckheit RW, Jr, Snow MJ, Fliakas-Boltz V, et al. Highly potent oxathiin carboxanilide derivatives with efficacy against nonnucleoside reverse transcriptase inhibitor-resistant human immunodeficiency virus isolates. Antimicrob Agents Chemother. 1997;41(4):831–837. doi: 10.1128/aac.41.4.831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Buckheit RW, Jr., Roberson JL, Lackman-Smith C, Wyatt JR, Vickers TA, Ecker DJ. Potent and specific inhibition of HIV envelope-mediated cell fusion and virus binding by G quartet-forming oligonucleotide (ISIS 5320). AIDS Res. Hum. Retroviruses. 1994;10(11):1497–1506. doi: 10.1089/aid.1994.10.1497. [DOI] [PubMed] [Google Scholar]
  • 20.Hadgraft J, Lane ME. Skin permeation: the years of enlightenment. Int. J. Pharm. 2005;305(1-2):2–12. doi: 10.1016/j.ijpharm.2005.07.014. [DOI] [PubMed] [Google Scholar]
  • 21.Lewi P, Arnold E, Andries K, et al. Correlations between factors determining the pharmacokinetics and antiviral activity of HIV-1 non-nucleoside reverse transcriptase inhibitors of the diaryltriazine and diarylpyrimidine classes of compounds. Drugs R D. 2004;5(5):245–257. doi: 10.2165/00126839-200405050-00001. [DOI] [PubMed] [Google Scholar]
  • 22.Buckheit RW., Jr Understanding HIV resistance, fitness, replication capacity and compensation: targeting viral fitness as a therapeutic strategy. Expert Opin. Investig. Drugs. 2004;13(8):933–958. doi: 10.1517/13543784.13.8.933. [DOI] [PubMed] [Google Scholar]
  • 23.Mansky LM, Temin HM. Lower in vivo mutation rate of human immunodeficiency virus type 1 than that predicted from the fidelity of purified reverse transcriptase. J. Virol. 1995;69(8):5087–5094. doi: 10.1128/jvi.69.8.5087-5094.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24•.Ojewole E, Mackraj I, Naidoo P, Govender T. Exploring the use of novel drug delivery systems for antiretroviral drugsExploring the use of novel drug delivery systems for antiretroviral drugs. Eur. J. Pharm. Biopharm. 2008;70:697–710. doi: 10.1016/j.ejpb.2008.06.020. [Review that highlights the significant potential that novel drug delivery systems, including but not limited to nanoparticles, transdermal patches, rectal delivery, and buccal delivery have for the future effective treatment of HIV/AIDS patients on ARV drug therapy.] [DOI] [PubMed] [Google Scholar]
  • 25.Kuksal A, Tiwary A, Jain NK, Jain S. Formulation and in vitro, in vivo evaluation of extended-release matrix tablet of zidovudine: influence of combination of hydrophilic and hydrophobic matrix formers. AAPS PharmSciTech. 2006;7(1):e1–e9. doi: 10.1208/pt070101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Oh S, Jeong S, Park T, Lee J. Enhanced transdermal delivery of AZT (Zidovudine) using iontophoresis and penetration enhancer. J. Control. Release. 1998;51:161–168. doi: 10.1016/s0168-3659(97)00162-4. [DOI] [PubMed] [Google Scholar]
  • 27.Costa P, Sausa Lobo JM. Evaluation of mathematical models describing drug release from estradiol transdermal systems. Drug Dev. Ind. Pharm. 2003;29(1):89–97. doi: 10.1081/ddc-120016687. [DOI] [PubMed] [Google Scholar]
  • 28.Zorbist R, Quan D, Thomas H, Stanworth S. Pharmacokinetics and metabolism of transdermal oxybutynin: in vitro and in vivo performance of a novel delivery system. Pharm. Res. 2003;20:103. doi: 10.1023/a:1022259011052. [DOI] [PubMed] [Google Scholar]
  • 29.Mukherjee B, Mahaoatra S, Gupta R, Patra B, Tiwari A, Arora A. A comparison of povidone-ethylcellulose and povidone-eudragit transdermal dexamethasone matrix patches based on in vitro skin permeation. Eur. J. Pharm. Biopharm. 2005;59:475–483. doi: 10.1016/j.ejpb.2004.09.009. [DOI] [PubMed] [Google Scholar]
  • 30•.Jain A, Jain P, Kurmi J, Jain D, Jain R, et al. Novel strategies for effective transdermal drug delivery: a review. Crit. Rev. Ther. Drug Carrier Syst. 2014;31:219–272. doi: 10.1615/critrevtherdrugcarriersyst.2014008126. [A review on the nanosized aggregates and micro-needle technology for the advanced delivery of vaccines, protein, peptides, nucleic acid and hormones across the skin.] [DOI] [PubMed] [Google Scholar]
  • 31.Thong HY, Zhai H, Maibach HI. Percutaneous penetration enhancers: an overview. Skin Pharm. Physiol. 2007;20(6):272–282. doi: 10.1159/000107575. [DOI] [PubMed] [Google Scholar]
  • 32.Karande P, Mitragotri S. Enhancement of transdermal drug delivery via synergistic action of chemicals. Biochim. Biophys. Acta (BBA) - Biomembranes. 2009;1788(11):2362–2373. doi: 10.1016/j.bbamem.2009.08.015. [DOI] [PubMed] [Google Scholar]
  • 33.Seki T, Kawaguchi T, Juni K, Sugibayashi K, Morimoto Y. Sustained transdermal delivery of zidovudine via controlled release of penetration enhancer. J. Control. Release. 1991;17(1):41–47. [Google Scholar]
  • 34.Jeong S, Lee J, Yuk S, Lee H. Transdermal delivery system of anti-AIDS virus agent using a biopolymer. Polymer (Korea) 1996;20:347–354. [Google Scholar]
  • 35.Kim DD, Chien YW. Transdermal delivery of dideoxynucleoside-type anti-HIV drugs. 1. Stability studies for hairless rat skin permeation. J. Pharm. Sci. 1995;84(9):1061–1066. doi: 10.1002/jps.2600840906. [DOI] [PubMed] [Google Scholar]
  • 36.Kim D-D, Kim JL, Chien YW. Mutual hairless rat skin permeation-enhancing effect of ethanol/water system and oleic acid. J. Pharm. Sci. 1996;85(11):1191–1195. doi: 10.1021/js9601041. [DOI] [PubMed] [Google Scholar]
  • 37••.Kim D-D, Chien YW. Transdermal delivery of dideoxynucleoside-type anti-HIV drugs. 2. The effect of vehicle and enhancer on skin permeation. J. Pharm. Sci. 1996;85:214–219. doi: 10.1021/js950141c. [Effects of vehicles and enhancers on the skin permeation of the anti-HIV drugs Zalcitabine (ddC), Didanosine (ddI) and Zidovudine (AZT) were studied using hairless rat skin. The results suggest that the mutual enhancement effect of ethanol and OA may make trasnsdermal devliery of anti-HIV drug possible.] [DOI] [PubMed] [Google Scholar]
  • 38.Suwanpidokkul N, Thongnopnua P, Umprayn K. Transdermal delivery of zidovudine (AZT): the effects of vehicles, enhancers, and polymer membranes on permeation across cadaver pig skin. AAPS PharmSciTech. 2004;5(3):e48. doi: 10.1208/pt050348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.N'da DD, Breytenbach JC. Synthesis of methoxypoly(ethylene glycol) carbonate prodrugs of zidovudine and penetration through human skin in vitro. J. Pharm. Pharmacol. 2009;61(6):721–731. doi: 10.1211/jpp.61.06.0004. [DOI] [PubMed] [Google Scholar]
  • 40.Jain S, Tiwary AK, Jain NK. PEGylated elastic liposomal formulation for lymphatic targeting of zidovudine. Curr. Drug. Deliv. 2008;5(4):275–281. doi: 10.2174/156720108785915078. [DOI] [PubMed] [Google Scholar]
  • 41.Dubey V, Mishra D, Nahar M, Jain V, Jain NK. Enhanced transdermal delivery of an anti-HIV agent via ethanolic liposomes. Nanomedicine. 2010;6(4):590–596. doi: 10.1016/j.nano.2010.01.002. [DOI] [PubMed] [Google Scholar]
  • 42.Som I, Bhatia K, Yasir M. Status of surfactants as penetration enhancers in transdermal drug delivery. J. Pharm. Bioallied. Sci. 2012;4(1):2–9. doi: 10.4103/0975-7406.92724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Van Heerden J, Breytenbach JC, N'da DD, Breytenbach JW, Du Preez JL. Synthesis and in vitro transdermal penetration of methoxypoly(ethylene glycol) carbonate and carbamate derivatives of lamivudine (3TC). Med. Chem. 2010;6(2):91–99. doi: 10.2174/157340610791321433. [DOI] [PubMed] [Google Scholar]
  • 44.Mukherji E, Millenbaugh NJ, Au JL. Percutaneous absorption of 2’,3’-dideoxyinosine in rats. Pharm. Res. 1994;11(6):809–815. doi: 10.1023/a:1018965305234. [DOI] [PubMed] [Google Scholar]
  • 45.Kim DD, Chien YW. Transdermal delivery of zalcitabine: in vitro skin permeation study. AIDS. 1995;9(12):1331–1336. [PubMed] [Google Scholar]
  • 46.Saunders J, Davis H, Coetzee L, Botha S, Kruger A, Grobler A. A novel skin penetration enhancer: evaluation by membrane diffusion and confocal microscopy. J. Pharm. Pharm. Sci. 1999;2(3):99–107. [PubMed] [Google Scholar]
  • 47.Oh SY, Jeong SY, Park TG, Lee JH. Enhanced transdermal delivery of AZT (Zidovudine) using iontophoresis and penetration enhancer. J. Control. Release. 1998;51(2-3):161–168. doi: 10.1016/s0168-3659(97)00162-4. [DOI] [PubMed] [Google Scholar]
  • 48.Narishetty STK, Panchagnula R. Effect of l-menthol and 1,8-cineole on phase behavior and molecular organization of SC lipids and skin permeation of zidovudine. J. Control. Release. 2005;102(1):59–70. doi: 10.1016/j.jconrel.2004.09.016. [DOI] [PubMed] [Google Scholar]
  • 49.Kararli TT, Kirchhoff CF, Penzotti Jr SC. Enhancement of transdermal transport of azidothymidine (AZT) with novel terpene and terpene-like enhancers: in vivo-in vitro correlations. J. Control. Release. 1995;34(1):43–51. [Google Scholar]
  • 50.Riviere J, Heit M. Electrically-assisted transdermal drug delivery. Pharm. Res. 1997;14(6):687–697. doi: 10.1023/a:1012129801406. [DOI] [PubMed] [Google Scholar]
  • 51.Dhote V, Bhatnagar P, Mishra PK, Mahajan SC, Mishra DK. Iontophoresis: a potential emergence of a transdermal drug delivery system. Sci. Pharm. 2012;80(1):1–28. doi: 10.3797/scipharm.1108-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Indermun S, Luttge R, Choonara YE, et al. Current advances in the fabrication of microneedles for transdermal delivery. J. Control. Release. 2014;185C:130–138. doi: 10.1016/j.jconrel.2014.04.052. [DOI] [PubMed] [Google Scholar]
  • 53.Zhu Z, Luo H, Lu W, et al. Rapidly dissolvable microneedle patches for transdermal delivery of exenatide. Pharm. Res. 2014;31(12):3348–3360. doi: 10.1007/s11095-014-1424-1. [DOI] [PubMed] [Google Scholar]
  • 54.Pierre MB, Rossetti FC. Microneedle-based drug delivery systems for transdermal route. Curr. Drug Targets. 2014;15(3):281–291. doi: 10.2174/13894501113146660232. [DOI] [PubMed] [Google Scholar]
  • 55.Donnelly RF, Morrow DI, Mccrudden MT, et al. Hydrogel-forming and dissolving microneedles for enhanced delivery of photosensitizers and precursors. Photochem. Photobiol. 2014;90(3):641–647. doi: 10.1111/php.12209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Zhang X, Wu Y, Hong Y, Zhu X, Lin L, Lin Q. Preparation and evaluation of dl-praeruptorin A microemulsion based hydrogel for dermal delivery. Drug. Deliv. 2014 doi: 10.3109/10717544.2014.898713. [DOI] [PubMed] [Google Scholar]
  • 57.Elnaggar YS, El-Refaie WM, El-Massik MA, Abdallah OY. Lecithin-based nanostructured gels for skin delivery: an update on state of art and recent applications. J. Control. Release. 2014;180:10–24. doi: 10.1016/j.jconrel.2014.02.004. [DOI] [PubMed] [Google Scholar]
  • 58.Dragicevic-Curic N, Winter S, Stupar M, et al. Temoporfin-loaded liposomal gels: viscoelastic properties and in vitro skin penetration. Int. J. Pharm. 2009;373(1-2):77–84. doi: 10.1016/j.ijpharm.2009.02.010. [DOI] [PubMed] [Google Scholar]
  • 59.Watson KM, Yang L, Buckheit Jr RW. Development of dual-acting pyrimidinediones as novel and highly potent topical anti-HIV microbicides. Antimicrob. Agents Chemother. 2011;55(11):5243–5254. doi: 10.1128/AAC.05237-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Buckheit Jr RW, Watson K, Fliakas-Boltz V, et al. SJ-3366, a unique and highly potent nonnucleoside reverse transcriptase inhibitor of human immunodeficiency virus Type 1 (HIV-1) that also inhibits HIV-2. Antimicrob. Agents Chemother. 2001;45(2):393–400. doi: 10.1128/AAC.45.2.393-400.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Sassi AB, Mccullough KD, Cost MR, Hillier SL, Rohan LC. Permeability of tritiated water through human cervical and vaginal tissue. J. Pharm. Sci. 2004;93(8):2009–2016. doi: 10.1002/jps.20107. [DOI] [PubMed] [Google Scholar]
  • 62.Padula C, Nicoli S, Colombo P, Santi P. Single-layer transdermal film containing lidocaine: modulation of drug release. Eur. J. Pharm. Biopharm. 2007;66(3):422–428. doi: 10.1016/j.ejpb.2006.11.014. [DOI] [PubMed] [Google Scholar]
  • 63.Cannon CL, Neal PJ, Southee JA, Kubilus J, Klausner M. New epidermal model for dermal irritancy testing. Toxicol. In Vitro. 1994;8(4):889–891. doi: 10.1016/0887-2333(94)90095-7. [DOI] [PubMed] [Google Scholar]
  • 64••.Ham AS, Lustig W, Yang L, Boczar A, Buckheit KW, et al. In vitro and ex vivo evaluations on transdermal delivery of the HIV inhibitor IQP-0410. PLoS ONE. 2013;8:e75306. doi: 10.1371/journal.pone.0075306. [An in vitro and ex vivo study into delivering the NNRTI IQP-0410 transdermally via a transdermal patch. With a permeability enhance, IQP-0410 was shown to have zero-order permeability while maintaining anti-HIV effiacy.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Budhian A, Siegel SJ, Winey KI. Controlling the in vitro release profiles for a system of haloperidol-loaded PLGA nanoparticles. Int. J. Pharm. 2008;346(1-2):151. doi: 10.1016/j.ijpharm.2007.06.011. [DOI] [PubMed] [Google Scholar]
  • 66.Li Y-P, Pei Y-Y, Zhang X-Y, et al. PEGylated PLGA nanoparticles as protein carriers: synthesis, preparation and biodistribution in rats. J. Control. Release. 2001;71(2):203. doi: 10.1016/s0168-3659(01)00218-8. [DOI] [PubMed] [Google Scholar]
  • 67.Singh A, Aboul-Enein H. Advantages of USP apparatus IV (flow-through cell apparatus) in dissolution studies. Iranian Chemical Society. 2006;3(3):220–222. [Google Scholar]
  • 68.Scheindlin S. Transdermal drug delivery: PAST, PRESENT, FUTURE. Mol. Interv. 2004;4(6):308–312. doi: 10.1124/mi.4.6.1. [DOI] [PubMed] [Google Scholar]
  • 69.Turpin J. Considerations and development of topical microbicides to inhibit the sexual transmission of HIV. Expert Opin. Investig. Drugs. 2002;11(8):1007–1097. doi: 10.1517/13543784.11.8.1077. [DOI] [PubMed] [Google Scholar]
  • 70.Harrison P, Rosenberg Z, Bowcut J. Topical microbicides for disease prevention: status and challenges. Clin. Infect. Dis. 2003;36(10):1290–1294. doi: 10.1086/374834. [DOI] [PubMed] [Google Scholar]
  • 71.Woolfson AD, Malcolm RK, Morrow RJ, Toner CF, Mccullagh SD. Intravaginal ring delivery of the reverse transcriptase inhibitor TMC 120 as an HIV microbicide Int. J. Pharm. 2006;325(1-2):82–89. doi: 10.1016/j.ijpharm.2006.06.026. [DOI] [PubMed] [Google Scholar]
  • 72.Gupta KM, Pearce SM, Poursaid AE, et al. Polyurethane intravaginal ring for controlled delivery of dapivirine, a nonnucleoside reverse transcriptase inhibitor of HIV-1. J. Pharm. Sci. 2008;97(10):4228–4239. doi: 10.1002/jps.21331. [DOI] [PubMed] [Google Scholar]
  • 73.Johnson TJ, Clark MR, Albright TH, et al. A 90-day tenofovir reservoir intravaginal ring for mucosal HIV prophylaxis. Antimicrob. Agents Chemother. 2012;56(12):6272–6283. doi: 10.1128/AAC.01431-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Singer R, Mawson P, Derby N, et al. An intravaginal ring that releases the NNRTI MIV-150 reduces SHIV transmission in macaques. Sci. Transl. Med. 2012;4(150):1–8. doi: 10.1126/scitranslmed.3003936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mcconville C, Friend DR, Clark MR, Malcolm K. Preformulation and development of a once-daily sustained-release tenofovir vaginal tablet tablet containing a single excipient. J. Pharm. Sci. 2013;102(6):1859–1868. doi: 10.1002/jps.23528. [DOI] [PubMed] [Google Scholar]
  • 76.Marcus JL, Glidden DV, Mcmahan V, et al. Daily oral emtricitabine/tenofovir preexposure prophylaxis and herpes simplex virus type 2 among men who have sex with men. PLoS ONE. 2014;9(3):e91513. doi: 10.1371/journal.pone.0091513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cohen MS, Chen YQ, Mccauley M, et al. Prevention of HIV-1 Infection with early antiretroviral therapy. N. Engl. J. Med. 2011;365(6):493–505. doi: 10.1056/NEJMoa1105243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Yeni PG, Hammer SM, Hirsch MS, et al. Treatment for adult HIV infection: 2004 recommendations of the International AIDS Society-USA Panel. Top HIV Med. 2004;292(2):251–265. doi: 10.1001/jama.292.2.251. [DOI] [PubMed] [Google Scholar]
  • 79.Carpenter CC, Cooper DA, Fischl MA, et al. Antiretroviral therapy in adults: updated recommendations of the International AIDS Society-USA Panel. JAMA. 2000;283(3):381–390. doi: 10.1001/jama.283.3.381. [DOI] [PubMed] [Google Scholar]
  • 80.Richman DD. HIV therapeutics. Science (New York, NY) 1996;272(5270):1886–1888. doi: 10.1126/science.272.5270.1886. [DOI] [PubMed] [Google Scholar]
  • 81.Trabattoni D, Lo Caputo S, Biasin M, et al. Modulation of human immunodeficiency virus (HIV)-specific immune response by using efavirenz, nelfinavir, and stavudine in a rescue therapy regimen for HIV-infected, drug-experienced patients. Clin. Diagn. Lab. Immunol. 2002;9(5):1114–1118. doi: 10.1128/CDLI.9.5.1114-1118.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES