Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 May 1.
Published in final edited form as: Ther Deliv. 2010 Jul;1(1):109–131. doi: 10.4155/tde.10.16

Challenges and opportunities in dermal/transdermal delivery

Kalpana S Paudel 1, Mikolaj Milewski 1, Courtney L Swadley 1, Nicole K Brogden 1, Priyanka Ghosh 1, Audra L Stinchcomb 1,
PMCID: PMC2995530  NIHMSID: NIHMS226982  PMID: 21132122

Abstract

Transdermal drug delivery is an exciting and challenging area. There are numerous transdermal delivery systems currently available on the market. However, the transdermal market still remains limited to a narrow range of drugs. Further advances in transdermal delivery depend on the ability to overcome the challenges faced regarding the permeation and skin irritation of the drug molecules. Emergence of novel techniques for skin permeation enhancement and development of methods to lessen skin irritation would widen the transdermal market for hydrophilic compounds, macromolecules and conventional drugs for new therapeutic indications. As evident from the ongoing clinical trials of a wide variety of drugs for various clinical conditions, there is a great future for transdermal delivery of drugs.


Delivery of drugs through the skin has been an attractive as well as a challenging area for research. Advances in modern technologies are resulting in a larger number of drugs being delivered transdermally including conventional hydrophobic small molecule drugs, hydrophilic drugs and macromolecules. Transdermal systems are a desirable form of drug delivery because of the obvious advantages over other routes of delivery. Transdermal delivery provides convenient and pain-free self-administration for patients. It eliminates frequent dosing administration and plasma level peaks and valleys associated with oral dosing and injections to maintain a constant drug concentration, and a drug with a short half-life can be delivered easily. All this leads to enhanced patient compliance, especially when long-term treatment is required, as in chronic pain treatment and smoking cessation therapy. Avoidance of hepatic first-pass metabolism and the GI tract for poorly bioavailable drugs is another advantage of transdermal delivery. Elimination of this first-pass effect allows the amount of drug administered to be lower, and hence safer in hepato-compromised patients, resulting in the reduction of adverse effects. Transdermal systems are generally inexpensive when compared with other therapies on a monthly cost basis, as patches are designed to deliver drugs from 1 to 7 days. The other advantage of transdermal delivery is that multiple dosing, on-demand or variable-rate delivery of drugs, is possible with the latest programmable systems, adding more benefits to the conventional patch dosage forms. The general acceptability of transdermal products by patients is very high, which is also evident from the increasing market for transdermal products. The transdermal drug delivery market, worth $12.7 billion dollars in 2005, is expected to reach $32 billion in 2015 [1].

Transdermal delivery systems (TDS) were introduced onto the US market in the late 1970s [2], but transdermal delivery of drugs had been around for a very long time. There have been previous reports about the use of mustard plasters to alleviate chest congestion and belladonna plasters used as analgesics. The mustard plasters were homemade as well as available commercially where mustard seeds were ground and mixed with water to form a paste, which was in turn used to form a dispersion type of delivery system. Once applied to the skin, enzymes activated by body heat led to the formation of an active ingredient (allyl isothiocyanate). Transport of the active drug component took place by passive diffusion across the skin – the very basis of transdermal drug delivery [3,4,201].

Since then a long path has been traversed in the field where we have seen the development of numerous transdermal patches ranging from nicotine to methylphenidate and testosterone to lidocaine [4]. The TDSs that have been developed over the years have been classified into different generations by Prausnitz et al. [2]. According to the classification, the first generation dealt mostly with small, lipophilic and uncharged molecules that can be delivered in the therapeutic range by passive diffusion alone. Most of the TDS that are currently on the market belong to this generation. But with the advancement of science and engineering we have seen the use of chemical enhancers and techniques such as ultrasound and iontophoresis for the delivery of drug molecules that cannot undergo passive diffusion. These belong to the second generation of transdermal products that target reversible disruption of the skin’s outer layer, the stratum corneum or use an additional driving force for drug delivery. One of the best examples is the delivery of lidocaine (a charged molecule), for which an iontophoretic delivery system was developed and marketed. A third generation of delivery systems is currently under development employing techniques such as microneedles and electroporation for delivery of macromolecules. This third generation of systems targets its effect towards the stratum corneum, rather than modification of the drug molecule itself.

A number of recently published reviews deal with various aspects of transdermal delivery, for example, classification of transdermals into generations and the transdermal market [2], and nanotechnology in transdermal delivery [5]. Patent reviews on formulation aspects of transdermal delivery and enhanced transdermal delivery techniques [68] have also been published. The first aim of this article is to provide an updated overview of the transdermal products currently on the US market and in clinical trials. The second aim is to focus on the challenges and solutions for overcoming skin permeation and skin irritation.

Transdermal products currently on the US market

A previously published review by Prausnitz and Langer described the various US FDA-approved transdermal patches and delivery systems [2]. This article contains an updated listing of transdermal products currently on the US market, as well as a separate section on transdermal products meant for local dermal delivery, as compared with systemic delivery. Table 1 lists all the transdermal products available in the USA. The first patch to be introduced onto the US market in 1979 was the scopolamine patch. It was a small reservoir type patch where a liquid dispersion of the drug was prepared and sealed between an impermeable backing layer and a rate-controlling membrane, which stays in contact with the skin and is responsible for the delivery of the drug. The nitroglycerin patch was introduced next in 1981 for angina and was much bigger in size. The clonidine patch (Catapres TTS®) was introduced in 1984 for high blood pressure as the first 7-day patch system. These were followed by the estradiol and fentanyl patches in 1986 and 1990, respectively. The original estradiol delivery system was eventually developed as a matrix type patch where the drug was dispersed in the adhesive matrix itself (Climara®), rather than in a separate formulation. But the TDS that revolutionized the transdermal market was the nicotine patch, which was first introduced in 1991 as a treatment for smoking cessation. Since then there has been development of a number of different patches, including a testosterone patch for hypogonadism in males and combination patches of estradiol and norethindrone or levonorgestrel for menopausal symptoms. TDS using ultrasound and iontophoresis were marketed in 1995 for delivery of small charged molecules such as lidocaine and fentanyl HCl. Although some of these prescription drug systems have been removed from the market for various reasons, ultrasound and iontophoresis are still used to deliver drugs for pain treatment in physical therapy clinics. The field of transdermal delivery now seems to be expanding further into the chronic treatment of neurological disorders, which has been showcased by the introduction of TDS-containing drugs such as methylphenidate for attention-deficit hyperactivity disorder (introduced in 2006), rotigotine for Parkinson’s disease (2007) and rivastigmine for dementia (2007) [2,9,201,202]. This expansion into neuroscience-related drugs is quite possibly due to the fact that small, potent molecules that can cross the blood–brain barrier also have good characteristics for crossing the skin, in addition to the fact that chronic disease states are ideal for multiple-day controlled-release delivery.

Table 1.

List of transdermal products currently on the US market.

Active ingredients Type of delivery system Name Company Type of patch Dose and application Regulatory status Uses
Clonidine Transdermal patch extended release Catapres TTS® Boehringer Ingelheim Drug in reservoir and in adhesive formulation 3.5–10.5-cm2 patches deliver
0.1–0.3 mg/day for 7 days
Applied to hairless skin on the upper outer arm or chest
Rx Essential hypertension
Clonidine Par Pharm Reservoir type 0.1–0.3 mg/24 h for 7 days
Estradiol Transdermal patch extended release Alora® Watson Laboratories Adhesive matrix drug reservoir 9–36-cm2 patches deliver
0.025–0.1 mg/day and continuous delivery for twice weekly dosing
Applied to lower abdomen
Rx Menopause, postmenopausal and osteoporosis, in case of lowered estrogen levels
Climara® Bayer Healthcare Adhesive matrix containing drug 6.5–25-cm2 patches deliver
0.025–0.1 mg/day for 7 days
Applied to lower abdomen or upper quadrant of buttock
Estraderm® Novartis Reservoir type 0.05 or 0.1-mg/day and continuous delivery for twice weekly application
Estradiol Mylan Technologies Adhesive matrix containing drug 0.025–0.1 mg/day continuous delivery once weekly patch
Menostar® Bayer Healthcare Adhesive matrix containing drug 3.25-cm2 delivers 14 μg/day for 7 days
Applied near lower abdomen
Vivelle®/Vivelle-Dot Novartis/Novogyne Adhesive formulation contains drug Patches having active surface area of 2.5–10 cm2 deliver 0.025–0.1 mg/day and twice weekly application
Applied to the abdomen
Transdermal gel Divigel® Upsher-Smith Laboratories 0.1% gel 0.25–1 g dose available
Applied to a small area (200 cm2) of the thigh in a thin, quick-drying layer
Elestrin® Azur Pharma 0.06% gel supplied in a non-aerosol, metered-dose pump container Applied once daily to the upper arm using a metered-dose pump that delivers 0.87 g of Elestrin® gel per actuation
Estrogel® Ascend Therapeutics 0.06% estradiol in an absorptive hydroalcoholic gel 1.25 g in a single dose and applied to
750-cm2 area
Applied to arm between wrist and shoulder
Transdermal spray Evamist KV Pharm/Ther-Rx Topical application to the skin of a rapidly drying homogeneous solution of 1.7% drug from a metered-dose pump One, two or three sprays/day (90 μl/spray) to adjacent nonoverlapping 20-cm2 areas on the inner surface of the arm between the elbow and the wrist and allowed to dry Rx Menopause, postmenopausal and osteoporosis, in case of lowered estrogen levels
Estradiol and levonorgestrel Transdermal patch extended release Climara Pro Bayer Healthcare Pharmaceuticals Drug in adhesive layer 22-cm2 Climara Pro system contains 4.4 mg estradiol and 1.39 mg levonorgestrel and delivers 0.045 mg estradiol and 0.015 mg levonorgestrel/day for 7 days applied to lower abdomen Rx Menopausal symptoms
Estradiol and norethindrone acetate Transdermal patch extended release Combipatch® Novartis Adhesive layer contains both drugs 9–16-cm2 patches deliver 0.05/0.14 or 0.05/0.25 mg estradiol/norethindrone acetate per day and applied twice weekly to lower abdomen Rx Menopausal symptoms
Ethinyl estradiol and norelgestromin Transdermal patch extended release Ortho Evra® Ortho McNeil Janssen Adhesive matrix containing drug 6.00 mg norelgestromin and 0.75 mg ethinyl estradiol in each 20-cm2 patch and delivers for 7 days
Applied to buttock, abdomen, upper outer arm or upper torso
Rx Contraception
Fentanyl Transdermal patch extended release Fentanyl transdermal system Actavis, Mylan Technologies, Lavipharm Labs, Noven, Watson Laboratories and Teva Pharms Matrix type (Mylan technologies and Teva Pharms) and reservoir (Actavis and Watson laboratories) 10–40-cm2 patches deliver 25–100 μg/h Schedule II Chronic pain (opioid tolerant) that cannot be managed by any other means
Duragesic® Ortho McNeil Janssen Drug in reservoir and in adhesive formulation 5–40-cm2 patches deliver 12.5–100 μg/h continuous systemic delivery for 72 h applied to flat surface such as the chest, back, flank or upper arm
Granisetron Transdermal patch extended release Sancuso® Prostraken Adhesive matrix containing drug 52-cm2 patch containing 34.3 mg of granisetron. The patch releases 3.1 mg of granisetron per 24 h for up to 7 days
Applied to upper outer arm
Rx Chemotherapy-induced nausea and vomiting
Methylphenidate Transdermal patch extended release Daytrana® Shire Adhesive-based matrix type patch 12.5–37.5-cm2 patches deliver 10–30 mg/9 h per patch applied to the hip area 2 h before an effect is needed and should be removed 9 h after application Schedule II Attention-deficit hyperactivity disorder
Nicotine Transdermal patch extended release Nicoderm® CQ Aventis Matrix type patch 7–21 mg over 24 h at different stages of the treatment OTC Smoking cessation
Nicotine transdermal system Novartis Consumer, Watson Laboratories, Cardinal Health and Aveva Matrix type patch 7–21 mg/24 h at different stages of treatment
Habitrol® Novartis and Novartis Consumer Reservoir type 17.5–52.5 mg that delivers 7–21 mg/day for the duration of treatment
Nitroglycerin Transdermal patch extended release Nitro Dur® Key Pharmaceuticals Drug in adhesive 5–40-cm2 patch delivers 0.1–0.8 mg/h for 12–14 h Rx Angina prophylaxis
Nitroglycerin Noven, Hercon Laboratories, Kremers Urban and Mylan Technologies Drug in adhesive Delivers nitroglycerin at 0.2 mg/h
Minitran Transdermal System Graceway Pharmaceuticals Drug in adhesive Delivers 0.1–0.6 mg/h
Transdermal ointment Nitroglycerin Fougera 2% 7.5–30 mg applied in the morning and again 6 h later to a 36-inch2 area of truncal skin
Oxybutynin Transdermal patch extended release Oxytrol® Watson Laboratories Adhesive matrix containing drug 39 cm2 system containing 36 mg and has a nominal in vivo delivery rate of 3.9 mg oxybutynin per day consistently for 3–4 days
Applied to abdomen, hip or buttock
Rx Bladder muscle dysfunction
Oxybutynin chloride Transdermal gel Gelnique® Watson Labs 10% gel 100 mg applied once daily to dry, intact skin on the abdomen, upper arms/shoulders or thighs (area of application rotated) Rx Bladder muscle dysfunction
Rivastigmine Transdermal patch extended release Exelon® Novartis Matrix reservoir containing drug 4.6–9.5 mg/24 h from 5–10-cm2 patches
Preferable application to upper or lower back
Rx Dementia associated with Alzheimer’s disease and Parkinson’s disease
Scopolamine Transdermal patch extended release Transderm Scop® Novartis Matrix reservoir containing drug 2.5-cm2 patch delivers 1.0 mg for 3 days
Applied to the hairless area behind one ear
Rx Motion sickness, Postoperative nausea and vomiting (prophylaxis).
Selegiline Transdermal patch extended release Emsam® Somerset Drug in adhesive 6–12 mg/24 h from 20–40-cm2 patch
Applied to the upper torso, upper thigh or the outer surface of the upper arm
Rx Major depressive disorder
Testosterone Transdermal patch extended release Androderm® Watson Laboratories and Watson Pharma Reservoir type 2.5 or 5 mg/day from 37–44-cm2 patch Schedule III Rx Hypogonadism (testosterone deficiency)
Transdermal gel Androgel® Unimed Pharma and Solvay/Abbott 1% gel 5–10 g contains 50–100 mg, 10% of the applied testosterone dose is absorbed across skin of average permeability during a 24-h period
Applied 5 g once daily to shoulders and upper arms and/or abdomen

OTC: Over-the-counter drug; Rx: Prescription drug.

Data retrieved from [201,202,211,212].

Most of the focus on transdermal systems has been on the patch dosage form, but a variation in the patch system of the transdermal era is a metered spray, for example Evamist®, which delivers estradiol or the gel formulations such as Androgel®, which delivers testosterone. The newer transdermal gels are technically just a return to the early approved nitroglycerin ointment technology; however, the new formulations have better cosmetic feel and consumer appeal. These systems are highly effective and cost efficient, and provide a good alternative to patch manufacturing for very potent drug molecules. One of the primary cost and safety issues associated with all patches currently on the market is that they usually contain a large amount of residual drug after use in order to maintain a saturated concentration of drug throughout the entire delivery period. For example, only 3% of the total drug content is absorbed from the lidocaine patch (Lidoderm®) [202], so it is very important to remove the patches carefully and discard according to provided directions. The residual drug in the patch can pose a danger to children who may pull the patches from the trash, and this residual drug is also a highly desired source of controlled substance for addicts. Therefore, the development of metered-dose pumps or active diffusion systems might thus be very beneficial to make transdermal products with more efficient drug use and improved safety/abuse liability profiles.

The stability of the transdermal patches is also important for delivering drugs effectively. Proper packaging of the individual patches is critical since the patches may contain a volatile solvent or hygroscopic substance. Each individual patch thus needs to be heat sealed into a multilaminate packaging consisting of foil, paper and heat sealable polyethylene portions. This requires an extra step in the manufacturing process, and adds some extra expense, as compared with most tablets and capsules that can be packaged easily in a multientry bottle.

Table 2 shows the topical gel and patch products currently available in the USA for the transdermal treatment of local tissue sites. These products are mainly indicated for the treatment of pain associated with arthritis, neuropathy, and muscle sprain and strain, apart from their use in local anesthesia. One FDA-approved lidocaine/tetracaine product for local anesthesia (Synera®) utilizes CHADD® technology. CHADD utilizes controlled heat to enhance transdermal drug permeation. CHADD units may either be incorporated directly into the drug-containing patch (as with Synera) or placed on top of a transdermal patch to temporarily increase the local temperature [203]. The FDA approved the first topical diclofenac product for osteoarthritis joint pain in 2007. Topical drugs penetrate slowly below the site of application leading to a relatively high drug concentration in the dermis, subcutaneous tissue and/or in the joint synovium, but delivering small amounts into the systemic circulation. New drug molecules for topical treatment of pain need to be developed given that some topical products such as capsaicin and diclofenac have side effects and are not effective for all patients.

Table 2.

List of transdermal products for local tissue treatment currently on the US market.

Active ingredients Type of delivery system Name Company Concentration Dose and application Regulatory status Uses
Capsaicin Topical patch (drug in adhesive) Qutenza® NeurogesX, Inc. 8% Four patches (maximum) can be applied every 3 months for a 60-min application
Applied by healthcare professional and local topical anesthetic may be required
Rx Neuropathic pain associated with postherpetic neuralgia
Diclofenac epolamine Topical patch (drug in adhesive) Flector® Inst Biochem 1.3% Each 10 × 14-cm patch contains 180 mg
Applied twice daily
Rx Pain relief
Diclofenac sodium Topical gel/jelly Voltaren® gel Novartis Consumer Health 1% 2–4 g applied maximum of four-times daily Rx Osteoarthritis
Solaraze® Nycomed US 3% 0.5 g gel used on each 5 cm2 lesion site Topical treatment of actinic keratoses
Topical solution Pennsaid® Nuvo Research Inc. 1.5% Each 1 ml solution contains 16.05 mg of drug
Applied ten drops at a time, four times for each application
Rx Osteoarthritis of the knee
Lidocaine Topical patch (drug in adhesive) Lidoderm® Teikoku Pharm 5% 10 × 14-cm patch, 70 mg/patch, three patches can be applied at one time for 12 h in a 24-h period Rx Postherpetic neuralgia
Lidocaine Topical cream utilizes SonoPrep® or laser-assisted drug delivery technology Echo Therapeutics and Norwood Abbey 4% Treatment with the device prior to application of 4% topical cream Rx Local dermal anesthesia
Lidocaine and prilocaine Topical cream Emla® App Pharms 2.5% of each drug 1–10 g/10 cm2 of skin surface area is applied for varying lengths of time as required Rx Local anesthetic
Lidocaine and tetracaine Topical patch utilizes CHADD® technology Synera® Zars Pharma 70 mg of each drug/patch Typically applied 20–30 min before the procedure Rx Local anesthetic for superficial dermatological procedures/venous access
Menthol and capsaicin Topical liquid Absorbine Jr® arthritis strength WF Young 4% menthol, 0.025% capsaicin Thin film three- to four-times daily OTC Arthritis pain, musculoskeletal pain and neuropathic pain
Menthol and methyl salicylate Topical patch Salonpas® arthritis pain Hisamitsu America 31.5 mg menthol, 105 mg methyl salicylate Patch can be used up to four-times daily OTC Arthritis pain
Salicylic acid Topical patch Trans-Ver-Sal® Doak 15% 6–20-mm patch OTC Acne vulgaris, hyperkeratotic psoriasis and removal of wart
Trolamine salicylate Topical cream Joint and muscle pain relief Flex-Power 10% cream Massaged three- to four-times daily OTC Muscle and joint pain

OTC: Over-the-counter drug; Rx: Prescription drug.

Data retrieved from [201,202,211,212].

Transdermal products currently in clinical trials

There are currently a large number of ongoing FDA-approved active clinical trials related to transdermal products (Table 3). These trials range from Phase I to IV studies, and most involve previously FDA-approved transdermal systems, including such compounds as fentanyl, nicotine and hormone therapies (to name just a few). These studies target a wide variety of disease states, and many are investigating the use of currently available transdermal products for new indications or in various combinations with other drugs. This article provides a description of the presently ongoing FDA-approved clinical trials related to TDS.

Table 3.

Ongoing FDA-approved active clinical trials involving transdermal delivery systems.

Drug Sponsor (collaborators) Transdermal product/intervention Phase Disease state/other comments
Buprenorphine Mundipharma Research Limited Buprenorphine transdermal patch extended release I and II Chemotherapy-induced mucositis and mouth pain in children
Fentanyl Cristália Produtos Químicos Farmacêuticos Ltda. Transdermal fentanyl III Pain and palliative care
Janssen Korea, Ltd., Korea Transdermal fentanyl IV Pain
Hormone therapy Washington University School of Medicine (American College of Obstetricians and Gynecologists) Transdermal estradiol versus oral naproxen or placebo - Contraception bleeding
Massachusetts General Hospital Transdermal 17β-estradiol versus oral estrogen versus placebo III Amenorrhea and female athlete triad syndrome
Bayer Ethinylestradiol/gestodene (BAY86–5016) II Contraception and ovulation inhibition
Bayer Ethinylestradiol/gestogene (BAY86–5016) II Contraception
Bayer Ethinylestradiol/gestodene (BAY86–5016) III Contraception
Stanford University Transdermal estrogen versus oral estrogen - Ovarian failure, premature
University of Chicago (Novo Nordisk, University of Michigan, Johns Hopkins University, Massachusetts General Hospital, University of Oklahoma, University of South Florida and Thomas Jefferson University) Transdermal estradiol plus growth hormone - Turner’s syndrome
National Institute on Aging Gonadotropin-releasing hormone plus transdermal estradiol III Obesity
Imperial College London (National Cancer Institute) Transdermal estrogen versus luteinizing hormone-releasing hormone analog II Anemia, cardiovascular complications, hot flashes, osteoporosis and prostate cancer
University of Pittsburgh Transdermal estrogen patch versus oral sertraline or placebo IV Postpartum depression
Robert H Lurie Cancer Center (National Cancer Institute) Transdermal 4-hydroxytamoxifen or placebo plus oral tamoxifen citrate or placebo II Breast cancer
Weill Medical College of Cornell University Oral contraceptive pills/microdose lupron versus transdermal estradiol patch/gonadotropin-releasing hormone antagonist IV Infertility
Population Council Transdermal NES/E2 gel II Ovulation
Procter and Gamble Transdermal testosterone versus placebo II Heart failure
Steven Lamm (Solvay Pharmaceuticals) AndroGel® (transdermal testosterone gel) I Hypogonadism
M et P Pharma Transdermal testosterone versus intranasal testosterone I Hypogonadism
Karolinska Institutet (Karolinska University Hospital, UroHealth Skövde, Universitätsklinikum MünsterInstitut für Reproduktionsmedizin, Krankenanstalt der Stadt Wien Rudolfstiftung, Medical University of Graz, Krankenhaus der Stadt Wien Lainz, Medizinische Universität Wien, Endokrinologikum and Charite University) Transdermal testosterone versus placebo III Metabolic syndrome
National Institute on Aging Ganirelix acetate plus transdermal estradiol or placebo - Arterial stiffening, aging and menopause
The Alfred (Stanley Medical Research Institute) Transdermal estradiol versus placebo - Schizophrenia, schizoaffective disorder and schizophreniform disorder (not in manic phase)
Massachusetts General Hospital Transdermal estrogen plus gonadotropin-releasing hormone plus gonadotropin-releasing hormone antagonist II and III Healthy postmenopausal women
University Hospital Transdermal versus oral estradiol IV Postmenopausal
Center for Epidemiology and Health Research (Procter and Gamble) Transdermal testosterone patch plus estrogen IV Ovariectomy, hysterectomy and hypoactive sexual desire disorder
Boston University (Eunice Kennedy Shriver National Institute of Child Health and Human Development) Transdermal testosterone (active or placebo) plus open-label sildenafil citrate IV Erectile dysfunction, testosterone deficiency and diabetes
BioSante Pharmaceuticals Transdermal testosterone versus placebo III Hypoactive sexual desire disorder
Eunice Kennedy Shriver National Institute of Child Health and Human Development (Population Council) Transdermal testosterone plus active or placebo Nestorone® gel I Contraception
Methylphenidate Cox Health Systems Methylphenidate transdermal system (Daytrana®) - Attention-deficit hyperactivity disorder and insomnia
University of Virginia Methylphenidate transdermal system (Daytrana) versus placebo - Attention-deficit hyperactivity disorder
University of Utah (Shire Pharmaceutical Development) Methylphenidate transdermal system versus placebo III Attention-deficit hyperactivity disorder
Nicotine Assistance Publique – Hôpitaux de Paris Transdermal nicotine versus usual treatment of Parkinson’s disease II Idiopathic Parkinson’s disease
Ohio State University (American Thoracic Society) Transdermal nicotine versus no intervention IV Pulmonary sarcoidosis
National Institute on Alcohol Abuse and Alcoholism Smoking cessation (with transdermal nicotine) plus behavioral counseling II Tobacco use cessation and alcohol-related disorders
National Institute on Drug Abuse (NIDA) (National Institutes of Health Clinical Center) Transdermal nicotine or placebo - Nicotine dependence and schizophrenia
University of Pennsylvania Transdermal nicotine ± placebo II Nicotine dependence
Massachusetts General Hospital (Stanley Medical Research Institute, North Suffolk Mental Health Association) Transdermal nicotine versus placebo patch IV Schizophrenia
University of Ottawa Heart Institute (Heart and Stroke Foundation of Ontario) Transdermal nicotine versus oral varenicline IV Coronary heart disease
Ochsner Health System Transdermal nicotine versus placebo - Postoperative nausea and vomiting
National Institute on Drug Abuse (NIDA) Transdermal nicotine IV Nicotine dependence
Yale University Transdermal nicotine versus intranasal nicotine versus placebo II Alcohol drinking
Centre for Addiction and Mental Health Transdermal nicotine plus transcranial magnetic stimulation - Cigarette smoking and schizophrenia
Duke University (Shire Pharmaceutical Development) Transdermal nicotine patch plus lisdexamphetamine or placebo - Attention-deficit hyperactivity disorder and nicotine dependence
Medical University of South Carolina Transdermal nicotine patch versus varenicline IV Nicotine dependence
Butler Hospital (National Institute on Drug Abuse [NIDA]) Transdermal nicotine plus counseling - Nicotine dependence
University of Texas at Austin Transdermal nicotine versus no intervention I Smoking cessation
Stony Brook University Transdermal nicotine versus placebo - Postoperative pain
University of Pennsylvania (NIH) Transdermal nicotine - Nicotine dependence
University of California, San Francisco (National Cancer Institute) Transdermal nicotine - Breast cancer, chemotherapeutic agent toxicity and Palmar–Plantar erythrodysesthesia
Medical University of South Carolina Transdermal nicotine - Nicotine dependence
Butler Hospital (National Institute on Drug Abuse) Behavioral modification plus transdermal nicotine II Nicotine dependence
Yu-Li Hospital (National Health Research Institutes, Taiwan Department of Health, Taiwan) Transdermal nicotine plus bupropion IV Tobacco dependence and schizophrenia
Centre for Addiction and Mental Health (Pfizer, Ontario Ministry of Health and Long Term Care) Transdermal nicotine plus behavioral interventions - Smoking
Department of Veterans Affairs Transdermal nicotine plus behavioral therapy plus bupropion SR 0 Stress disorders and post-traumatic tobacco use disorder
National Institute of Cancerología Transdermal nitroglycerin II Locally advanced non-small-cell lung cancer
University Hospital, Basel, Switzerland Experimental treatment combination that includes transdermal nitrates - Acute heart failure
University Hospital, Basel, Switzerland Targeted therapy with nitrates, including transdermal nitroglycerin - Acute heart failure
Rivastigmine Novartis Pharmaceuticals Transdermal rivastigmine IV Alzheimer’s disease
University Hospital, Lille Transdermal rivastigmine versus placebo III Parkinson’s disease and apathy
Novartis Pharmaceuticals Transdermal rivastigmine versus placebo IV Multiple sclerosis and cognitive impairment
Rotigotine UCB, Inc. Transdermal rotigotine versus placebo III Parkinson’s disease
UCB, Inc. Transdermal rotigotine I Healthy volunteers
University College, London (Medical Research Council) Transdermal rotigotine II Right hemisphere stroke, hemispatial neglect and motor deficits
Scopolamine Drexel University College of Medicine (Merck) Transdermal scopolamine plus oral aprepitant or placebo - Nausea and vomiting
Drexel University College of Medicine (Merck) Transdermal scopolamine or placebo plus oral aprepitant - Nausea and vomiting
Miscellaneous or investigational agents Banner Health (Phoenix Children’s Hospital) Transdermal DMPS (metal chelator approved for use in Europe) I Intoxication
Somerset Pharmaceuticals Transdermal selegiline versus placebo IV Mental health and major depressive disorder
Eli Lilly and Company (TransPharma Medical) Transdermal versus subcutaneous teriparatide II Osteoporosis
Labtec GmbH Transdermal sufentanil versus oral morphine sulfate II Chronic pain
University of Nottingham Transdermal glyceryl trinitrate patch II and III Stroke
Lytix Biopharma AS (Oslo University Hospital, Karolinska University Hospital) LTX-315 (Oncopore), dose escalation study I Cancer with transdermal accessible tumor
Otsuka Pharmaceutical Co., Ltd SPM 962 versus placebo II and III Early Parkinson’s disease
Butler Hospital Oral fluoxetine or placebo plus transdermal nicotine III Major depressive disorder and nicotine dependence depression
McMaster University (The Physicians’ Services Incorporated Foundation and Canadian Network and Centre for Trials Internationally) Transdermal clonidine or placebo; active (oral) aspirin or placebo IV Cardiovascular disease
Fundación Cardiovascular de Colombia (Fundación Santandereana de Diabetes y Obesidad (FUSANDE), The University of Akron, University of Santander and Instituto de Salud de Bucaramanga (ISABU)) Controlled nitric oxide releasing patch versus placebo III Diabetic foot
MedtronicNeuro Standard therapy (including transdermal oxybutynin) versus InterStim® therapy IV Urinary incontinence and urge overactive bladder
Axxonis Pharma AG Transdermal lisuride versus oral ropinorole or placebo III Restless legs syndrome
NuPathe Inc. NP101 (sumatriptan iontophoretic transdermal patch) III Migraine disorders

Data taken from [206].

Besides the trials utilizing both investigational agents and currently approved transdermal products (described in Table 3), there is also extensive work taking place in the development of microporation technologies and devices, most of which are targeted to disrupt or bypass the stratum corneum. Such microporation techniques that are currently in development (and are in various stages of the FDA approval process) include laser, radiofrequency, thermal microporation technologies and microneedle systems. In addition to the large number of technologies in development, these products are being investigated with a wide variety of drug compounds, including (but not limited to) parathyroid hormone, lidocaine, insulin, fentanyl and diclofenac sodium. Several recently published reviews provide excellent overviews of the technologies typically used to enhance transdermal delivery, as well as the technologies currently in development, and the reader is referred to these reviews for more details [911]. Table 4 describes all the currently ongoing FDA-approved active clinical trials involving microporation techniques for transdermal delivery.

Table 4.

Ongoing clinical trials involving microporation techniques for transdermal delivery.

Microporation technique Sponsor (Collaborator) Intervention Disease state/other comments
Electroporation Karolinska University Hospital (Karolinska Institutet, Swedish Institute for Infectious Disease and Control Cyto Pulse Sciences, Inc.) DNA vaccine delivered by intradermal electroporation (device: Derma Vax) Colorectal cancer
Uppsala University (Karolinska Institutet and Cyto Pulse Sciences, Inc.) DNA vaccine delivered by intradermal electroporation (device: Derma Vax) Prostate cancer
Microneedles Emory University Microneedles versus subcutaneous catheter for insulin delivery Type 1 diabetes
The University of Hong Kong (Hospital Authority, Hong Kong) MicronJet versus intramuscular injection of H1N1 vaccine Vaccination and influenza
NanoPass Technologies Ltd Conventional subcutaneous injection versus MicronJet for insulin delivery Intradermal injections and healthy volunteers
Hadassah Medical Organization Intradermal unadjuvanted Pandemrix® via a microneedle device, compared with intramuscular adjuvanted Pandemrix Healthy subjects
NanoPass Technologies Ltd Delivery of influenza vaccine with microneedles versus intramuscular injection Influenza vaccine
Becton, Dickinson and Company Extended microneedle delivery of insulin Types 1 and 2 diabetes mellitus
Radiofrequency ablation Eli Lilly and Company (TransPharma Medical) Transdermal versus subcutaneous teriparatide (device: ViaDerm drug delivery system) Osteoporosis

Data taken from [206].

In addition to the active trials that are ongoing with TDS, there are several products in development that have completed Phase III trials but are not yet available on the US market. These products include a buprenorphine TDS for various pain conditions (Purdue Pharma LP) and a sumatriptan iontophoretic transdermal patch (Zelrix) for migraine disorders (NuPathe Inc., NDA filing expected in 2011) [204206]. There are also products that have completed some Phase II trials and are expected to continue to progress through further trials, such as the Altea thermal ablation device for insulin delivery, transdermal bupivacaine patch (Eladur, Durect and AlPharma Inc.), transdermal sufentanil (TRANSDUR-sufentanil, Durect and AlPharma Inc.) and ViaDerm-human parathyroid hormone (TransPharma Medical) [207,208].

Challenges & solutions for overcoming the skin permeation barrier

Owing to the selective nature of the skin barrier, only a small pool of drugs can be delivered systemically at therapeutically relevant rates. Approximately 15 drugs constitute the whole segment of the transdermal drug market (Table 1). Besides great potency, the physicochemical drug characteristics often evoked as favorable for percutaneous delivery include moderate lipophilicity and low-molecular-weight [12]. However, a large number of pharmaceutical agents do not fulfill these criteria. This is especially true for macromolecules, such as insulin, human growth hormone or cyclosporine, which are very challenging from the drug delivery point of view. Overcoming low skin permeability to xenobiotics can be achieved by a variety of approaches, and is an active field of research. Their effectiveness and applicability will vary from drug to drug depending on the physicochemical nature of the compound. Here, a brief overview of the enhancement methods including chemical methods, physical methods and a prodrug approach will be provided with special emphasis on the recent developments.

Chemical methods

Chemical permeation enhancers facilitate drug permeation across the skin by increasing drug partitioning into the barrier domain of the stratum corneum, increasing drug diffusivity in the barrier domain of the stratum corneum or the combination of both [13]. The heterogeneous stratum corneum is composed of keratin ‘bricks’ and intercellular continuous lipid ‘mortar’ organized in multilamellar strata [1416]. Depending on the nature of the drug either of these two environments may be the rate-limiting milieu (barrier domain) for the percutaneous transport. As a consequence it is anticipated that the magnitude of permeation improvement obtained with a given permeation enhancer will vary between lipophilic and hydrophilic drugs. Several mechanisms of action are known: increasing fluidity of stratum corneum lipid bilayers, extraction of intercellular lipids, increase of drug’s thermodynamic activity, increase in stratum corneum hydration, alteration of proteinaceous corneocyte components and others. More detailed discussion of the modes of action has been reported elsewhere [17,18]. Permeation enhancers are conventionally divided into several groups based on their chemical structure rather than the mechanism of action. This is partially due to the difficulty determining a primary or mixed mode of action for many of them. Furthermore, compounds from the same group can exert their effect through different mechanisms. More than 300 substances have been shown to have skin permeabilization potential and this number is still growing. Most known enhancers fall into the following categories: alcohols (ethanol, pentanol, benzyl alcohol, lauryl alcohol, propylene glycols and glycerol), fatty acids (oleic acid, linoleic acid, valeric acid and lauric acid), amines (diethanolamine and triethanolamine), esters (isopropyl palmitate, isopropyl myristate and ethyl acetate), amides (1-dodecylazacycloheptane-2-one [Azone®], urea, dimethylacetamide, dimethylformamide and pyrrolidone derivatives), hydrocarbons (alkanes and squalene), surfactants (sodium laureate, cetyltrimethylammonium bromide, Brij®, Tween® and sodium cholate), terpenes (D-limonene, carvone and anise oil), sulfoxides (dimethyl sulfoxide) and phospholipids (lecithine). The importance of water, or hydration of the stratum corneum, is not to be underestimated. A fully hydrated stratum corneum (under occlusion) presents lesser diffusional resistance to xenobiotics than its dehydrated counterpart. However, a common drawback of permeation enhancers is that their efficacy is often closely mimicked by skin irritation. In general, the same mechanisms that are responsible for enhanced drug transport such as disrupting ordered stratum corneum lipid bilayers or corneocyte structural organization are also responsible for skin irritation [19]. One possibility to address this concern is to identify mixtures of permeation enhancers that exhibit synergistic effects [20]. Karande et al. successfully used a screening approach to test 5000 binary mixtures of chemicals [21,22]. The details are described in the next section of this article. Most of the products on the transdermal market use the effect of occlusion, which can be classified as an enhancement technique acting through the hydration of the stratum corneum. Additionally, drugs such as nitroglycerin (Nitro-Dur®, Nitro Disc® and Transderm-Nitro®) use fatty acid esters and lidocaine (Lidoderm®) use urea and propylene glycol as chemical enhancers. Traditionally, chemical enhancers have been used to increase the delivery of small molecules and showed only limited success in permeation enhancement of macromolecules. Overall, chemical methods, although effective, cannot compete with physical enhancement methods that provide a greater magnitude of skin permeabilization.

Physical methods

The oldest and by far the most popular way of overcoming the skin barrier physically is the use of hypodermic needles. In many cases it is the only viable method of delivery for poorly absorbable and highly unstable compounds. Typically, drug solution is forced under piston pressure directly into the bloodstream or tissue (skin and muscle). Such drug administration results in quick delivery of large amounts of drug. If controlled drug delivery over longer periods of time is desired, indwelling catheters are used. However, both require mechanical perforation of skin with a needle, which causes pain and trauma. According to Hamilton, needle phobia is a medical condition that affects at least 10% of the population [23]. This condition is a serious problem in the healthcare system in the sense that people with needle phobia tend to avoid medical care. To address these drawbacks, several alternative physical skin enhancement methods such as jet injections, dermabrasion, thermal ablation, laser, microneedles, iontophoresis, electroporation, ultrasound and combinations of the above have been investigated. These methods aim at developing more user-friendly and flexible delivery systems, and are able to produce bolus type as well as sustained drug delivery profiles.

Jet injections involve the delivery of liquid or solid particles driven by high-pressure accelerators across the stratum corneum [24]. The concept of drug delivering jets is not new as first reports date back to the 1940s; however, first attempts of using this technology translated into limited success. Technology advances in the later decades enabled the construction of more reliable and effective devices based on compressed helium [25]. It proved its potential in the delivery of insulin to diabetic patients [26]. This method is similar to the hypodermic injection in that it enables rapid delivery of large amounts of drug, but it has resulted in improved patient compliance [27]. Shortcomings of jet injections include inability to deliver drugs over longer periods of time; however, they seem to be well suited for vaccination delivery [28].

Dermabrasion has been traditionally used to treat acne, scars and other dermatologic and cosmetic conditions [29]. Recently, the potential of this technique for transdermal drug delivery has been recognized. Mechanistically, crystal microdermabrasion uses microcrystals blown onto the skin surface to perform the exfoliating process. Several-fold improvement in flux for low-molecular-weight drugs was shown by various researchers with the use of this technique [30,31]. A recent study by Gill et al. investigated the effect of microdermabrasion on stratum corneum removal in rhesus macaques and human volunteers in vivo. The effectiveness of the skin barrier removal correlated with the number of passes of the mobile dermabrasion device and selective removal without damage to the viable epidermis was possible [32].

Thermal ablation takes advantage of the external source of thermal energy, which propagates into the stratum corneum to create microchannels. Heating of the skin surface to hundreds of degrees for a very short period of time allows the thermal damage to be limited to the stratum corneum alone without further heat propagation to live epidermal layers. An interesting publication by Park et al. discussed the effect of heat on skin permeability [33]. The authors pointed out skin permeability changes to a model hydrophilic compound at different temperature ranges. While an intermittent increase in the skin temperature to 100–150°C causes a moderate increase in the flux of a hydrophilic compound, 150–250°C translates into one to two orders of magnitude increase, and a temperature of over 300°C adds yet another tenfold augmentation in the transdermal flux. Different mechanisms were postulated to be responsible for such change at each temperature range. Altea Therapeutics developed a PassPort system that comprises a single-use disposable patch and a re-useable handheld applicator. Phase I and II clinical trials have been completed for the delivery of insulin via this enhancement system.

A laser can also be used to thermally ablate parts of the stratum corneum creating pores. These pores allow drug molecules applied to the surface of the skin to bypass the diffusional barrier and gain easy access to the vascularized deeper layers of the skin. This method has been able to extract interstitial fluid to measure glucose levels in diabetic patients as well as being effective in augmenting transdermal flux of hydrophilic and hydrophobic molecules [3436]. A transdermal laser system P.L.E.A.S.E.® (Pantec Biosolutions) has been developed and granted a European patent in 2009. Furthermore, laser-assisted drug delivery (Norwood Abbey) was developed and the device has been cleared for marketing by the FDA for the delivery of a topically applied local anesthetic. The pitfall of the laser approach is that it is a relatively complicated technology associated with high costs.

Microneedles became one of the major enhancement techniques in the transdermal drug delivery field after advancements in microfabrication techniques were made in recent years. Long enough to perforate the topmost layers of the epidermis but short enough not to excite nerve endings in the skin, microneedles offer painless and powerful dermal permeabilization [37]. A variety of different microneedle designs have been used in in vitro testing. Solid microneedles are used to pierce microchannels through the skin followed by application of a transdermal patch, and microneedles themselves can be coated with a drug or contain a drug dissolved in their matrix. Yet another option involves hollow microneedles through which a convective flux of drug is delivered into deeper skin layers. Henry et al. showed an increase in the transdermal flux of calcein by up to four orders of magnitude [38]. A multitude of other studies proved that microneedles effectively permeabilize skin to drugs [3942]. A first-inhuman microneedle study showed that therapeutically relevant plasma levels of naltrexone, an opioid antagonist, were achieved after skin pretreatment with microneedles and subsequent application of transdermal patches [43]. Currently, several systems based on microneedles are being investigated, such as Microstructured Transdermal Systems (3M), Macroflux® (Zosano) or MicroCor (Corium), and have shown efficacy in preclinical trials.

There are several methods that use electric current as a means to permeabilize the skin. Iontophoresis relies on the continuous electric current to drive charged drug molecules across the skin via electrophoresis and electroosmosis [44]. It is commonly stated that iontophoresis mainly provides additional electrochemical driving force for drug transport across skin, rather than increasing the permeability of the skin. The first in vivo animal investigation showing the effectiveness of this approach was carried out at the beginning of the 20th century. Since then many drug delivery systems were tested for small molecules such as pilocarpine [45], lidocaine [46] or zolmitriptan [47] and large molecules such as calcitonin, insulin, parathyroid hormone or vasopressin [4852]. Clinical studies, however, have been limited to small agents such as lidocaine, cortisone or fentanyl [5355]. Although there have been reports indicating the presence of erythema under the electrode application site [56], the irritation correlates well with the current and as long as the current is low, adverse effects are not substantial. Both continuous and intermittent delivery of drugs is feasible. A couple of drug delivery systems using iontophoretic devices were approved by the FDA in the past, for example, lidocaine HCl with epinephrine (Iontocaine® and Lidosite® topical system) and fentanyl HCl (IONSYS) [209]. IONSYS, however, was never marketed in the USA. These products have now been listed as discontinued by the FDA (Iontocaine since 2005, Lidosite topical system since 2006 and IONSYS since 2008). The discontinuation of IONSYS in the USA was affected primarily by the European Medicines Agency’s [210] recall of the product and suspension of marketing because of corrosion of a component in the system, which can lead to release of fentanyl without patient activation. The limitations of this delivery method include the relatively high cost of the iontophoretic device.

Another method – electroporation – is based on the use of short (micro- to milli-second), high-voltage electrical pulses to create transient disruptions in the structure of the stratum corneum [57]. Such disruptions, or pores, facilitate the transport of small and large molecules otherwise unable to permeate at all. Drug molecules are believed to be transported through skin by electrophoretic movement and diffusion. Examples proving in vitro effectiveness of electroporation include small molecules such as timolol [58], as well as larger molecules such as oligonucleotides, heparin and IgG antibodies [59].

Ultrasound skin enhancement (sonophoresis) is achieved by the use of high (MHz) or low (kHz) frequency devices, the latter showing greater percutaneous drug flux improvement. At low frequencies the ultrasound applied to the skin results in the production of cavitational bubbles, which by oscillation disrupt the stratum corneum structure. In vitro experiments have shown substantial increase in the skin permeability to small molecules such as estradiol, aldosterone or lidocaine [60], as well as macromolecules such as insulin, erythropoietin or γ-interferon [6165]. In 2004, the FDA approved an ultrasound system (SonoPrep®) to accelerate the action of lidocaine for local dermal anesthesia. The same system is intended to be used for delivering vaccines, insulin and antibiotics, and blood glucose monitoring in the future. An interesting feature of the device is real-time skin impedance feedback that stops the sonication procedure when the desired level of conductance has been achieved. This method does not cause pain or irritation when used within certain limitations. A ViaDerm system (TransPharma) has been developed and preclinical trials demonstrated effectiveness of this method of skin enhancement. Moreover, clinical trials involving the ViaDerm system coupled with human parathyroid hormone (1–34) and human growth hormone are underway.

Combinations of enhancement techniques have the potential of showing synergistic effects [66]. Some combinations that have been studied include electroporation–ultrasound [67], electroporation–iontophoresis [68,69] and microneedle–iontophoresis [70,71]. The observed synergistic effects may be rationalized by different mechanisms by which enhancement methods work, for example electroporation disrupts stratum corneum barrier structure increasing its permeability to drugs, while iontophoresis provides additional driving force for diffusion of charged particles above that of just passive diffusion.

Prodrugs

The prodrug approach in percutaneous drug delivery includes several different areas of interest: permeation across skin, chemical and enzymatic stability, as well as skin irritation potential. While all of these things need to be considered for design of a successful prodrug, usually the main reason to use this approach is to modify the physicochemical properties of a parent drug such that the flux of the prodrug is increased over that of the parent molecule. A chemical modification of the compound of interest presents great opportunity to alter its ability to cross the stratum corneum barrier. In the most conventional case, when the diffusional barrier is represented by the lipid domain of the stratum corneum and parent drug is relatively hydrophilic, increasing its lipophilicity can result in improved percutaneous flux. A penalty, however, is paid for with the increase in the size of the permeant. By contrast, it has also been possible to improve the flux of extremely hydrophobic compounds (which undergo viable tissue controlled diffusion, in comparison to stratum corneum controlled diffusion) by creating more hydrophilic prodrugs. A good discussion of the prodrug approach in percutaneous drug delivery can be found elsewhere [72]. More recent reports include the delivery of ketorolac piperazinylalkyl prodrugs [73], a pilot clinical trial involving prodrugs of levodopa [74], effects of PEGylation on transdermal flux of acetaminophen [75], naltrex-one prodrugs [76,77] and bupropion prodrugs [78]. Currently, there are no transdermal prodrugs available on the market.

Challenges & solutions for overcoming skin irritation

A major advantage of topical and transdermal drug delivery is the reduction of systemic side effects. On the other hand, delivery of drugs by these routes adds the potential for side effects in the form of skin irritation at the delivery site. Skin irritation reactions include irritant contact dermatitis (ICD), an inflammatory response caused by repeated or direct exposure of the skin to weak irritants, and allergic contact dermatitis (ACD), delayed, T-cell-mediated inflammatory response to a specific allergen. ICD reactions range from erythema and scaling to necrotic burns while ACD reactions include erythema, edema and occasionally vesiculation. Additionally, the onset of ACD reactions is highly variable, and is dependent on the irritant that initiated the reaction and the individual who expresses the allergic response.

Factors that contribute to skin irritation include changes in the physiological pH of the skin, disruption of the stratum corneum barrier (i.e., delipidization, hydration and disruption of stratum corneum lipid packing), immunological and physiological reactions, bacteria proliferation at the delivery site, and chemical/pharmacological features of the drug or vehicle. Many features of topical and transdermal systems contribute to skin irritation including the active pharmaceutical ingredient (API), formulation (including skin permeation enhancers and excipients), occlusion of the skin, duration of device application and the type of delivery device used. Additionally, patch adhesives and membranes, solvents, enhancers and the active drug have all been demonstrated to contribute to the development of ACD reactions. The risk of causing cutaneous irritation can be minimized through management of the device and formulation components during the device design process. Additional means to reduce adverse skin responses have been reported; pretreatment of the skin with a corticosteroid before application of the system, inclusion of corticosteroids in the formulation and structural alterations of chemical permeation enhancer are just a few of these. Factors identified as contributing to skin irritation as well as methods to prevent or remedy these reactions are discussed in greater detail later.

The role of drug features in skin irritation

The API included in the topical or transdermal pharmaceutical preparation has been indicated as the major cause of ACD in patients [79]. Drugs that exhibit moderate-to-severe irritation in animal studies are often ruled out for further use in topical formulations. Unfortunately, it is difficult to predict the irritation potential of a drug without first testing it in an animal or cell model. Some studies have been performed to identify characteristics of drugs that may be potential irritants. For instance, it has been reported that the dissociation constant (pKa) of an API can affect the physiological pH of the skin and by this cause cutaneous irritation. Berner et al. have shown that benzoic acid derivatives with pKa values of 4 or less caused irritation after 24 h of exposure to the skin in humans [80,81]. Furthermore, drugs that possess pKa values greater than 8 have been shown to irritate the skin as well. A trend of increasing irritation with increasing pKa was reported by a number of groups [82,83]. Based on these studies, it is predicted that drugs with pKa values between 4 and 8 should provide minimal skin irritation. Additionally, instead of excluding potentially irritating drugs, simply minimizing their concentration included in a topical or transdermal formulation may reduce their resulting irritation to safe and tolerable levels.

Pharmacological responses of the skin to a drug can also present irritation and should be considered. For example, capsaicin is marketed as a topical preparation indicated for relief of local pain and irritation. This drug induces its action through interaction with the epidermal nervous system. After initial application, capsaicin causes pain and a burning sensation at the site of action. To reduce these effects capsaicin is applied multiple times a day at a low dose to induce resistance in the skin to irritation while also building local therapeutic levels. Here, the dose and dosing regimen of capsaicin is controlled to bring about the therapeutic response without causing significant, long-term irritation [84]. It has also been reported that subsequent exposure to an irritant can induce resistance of the skin to the irritant, a process called accommodation. Both downregulation of the inflammatory response and skin hardening by upregulation of ceramide 1 synthesis have been attributed to the development of resistance by the skin [85,86].

Substitution of equally or more potent derivatives of an irritating drug for the parent in a topically applied delivery system may also reduce skin irritation. For example, synthetic derivatives of capsaicin have shown decreased irritation over the parent drug. In a study where capsaicin, nonivamide and sodium nonivamide acetate were applied to the forearms of male human volunteers, sodium nonivamide was found to produce the least erythema and painful sensation. Although its transdermal flux was determined to be the least of the three drugs tested, sodium nonivamide is more potent than capsaicin in its pharmacological action. Nonivamide was also found to be less irritating than capsaicin in this study while displaying similar flux to capsaicin as well [87].

The role of vehicle & devices in skin irritation

The pH of the skin’s surface has been reported to be in the range of 5.4 to 5.9 and is important in the maintenance of skin barrier function and defense against infection and disease [88]. The skin also has an excellent buffering capacity against large changes in pH. Albeit, external factors such as washing and applying solutions, drugs and cosmetics to the surface of the skin can raise its surface pH and can likewise increase or induce skin irritation. For example, alkaline solutions of pH 9 and above applied to the skin have been reported to cause skin irritation. In the same study, aqueous solutions of pH 5 and 7 did not cause irritation when applied to the skin [89]. In another study, Ananthapadmanabhan et al. showed that a solution at pH 10 when applied to the skin, compared with a pH of 4 or 6.5, increased the transition temperature of stratum corneum lipids [90]. Observed adverse effects were swelling of the stratum corneum and disruption of the skin barrier function, as indicated by an increase in transepidermal water loss. Therefore, to avoid skin irritation it is very important to buffer formulations applied to the skin as close to the skin’s surface pH as possible. Some universal pharmaceutical solvents are irritating to the skin and therefore cannot be used in topical preparations. Prior to in vivo investigation, these solvents should be replaced by alternative solvents with acceptable irritation and safety profiles. Topical solvents deemed to be safe for use include isopropyl alcohol, propylene glycol, isopropyl myristate and polyethylene glycols to a certain percentage (up to 60% used in marketed products) [91].

Most topical systems and transdermal systems require prolonged complete occlusion of the skin in the form of a patch, cream, lotion or other pharmaceutical preparation. Water-impermeable occlusion of the skin has been found to increase the pH and temperature of the skin’s surface, as well as trapping moisture and sweat to create a humid environment. These conditions are favorable for bacterial and yeast overgrowth on the surface of the skin. Sweat itself has been shown to cause occlusion-induced irritation [92]. ICD reactions are commonly seen after skin occlusion and its incidence has been reported to increase with the duration of occlusion [93,94]. Additionally, occlusion may increase the irritation potential of the topically administered API. For instance, Van der Valk and Maibach have shown that in ten healthy subjects the irritation response to repeated short-term exposure of sodium lauryl sulfate was enhanced with postexposure occlusion [95]. Functional damage to the skin barrier by occlusion, detected by transepidermal water loss, has also been reported [96].

Finally, the choice of a matrix type versus a reservoir-type patch for drug delivery can affect the incidence of skin irritation. Components of each system have been reported to contribute to skin reactions upon use. In the case of transdermal delivery of estradiol, skin reactions have been described in response to the adhesive, hydroxypropyl cellulose and ethanol in the original TDS [79]. In addition, reactions to methacrylate in the transdermal nicotine patch have been reported [97]. Since most reactions to patches tend to be ACD responses, it is difficult to predict the irritation potential of the individual patch components. Furthermore, studies discerning irritation caused by the application of matrix type versus reservoir type patches are inconclusive. However, one source has reported that drug-loaded matrix patches tend to cause fewer cutaneous reactions than reservoir release patches [98]. By contrast, similar skin irritation was observed after application of either a matrix or reservoir patch containing fentanyl to the skin of healthy human subjects [99].

Impact of formulation on skin irritation

It has been shown that the formulation used to deliver a drug can influence the appearance and degree of skin irritation. Hydrogels, for instance, have been reported to reduce skin irritation by absorbing moisture from the skin’s surface [94]. A study comparing the safety and efficacy of a lotion containing benzoyl peroxide entrapped in polymeric microspheres to a lotion containing free benzoyl peroxide showed that controlled release of benzoyl peroxide from the microspheres reduced skin irritation in humans, without sacrificing efficacy. Based on these results, it was concluded that controlled-release systems might be useful in reducing irritation induced by topically applied drugs [100]. Liposomes have also been shown to reduce skin irritation. The postulated mechanisms for reduction of skin irritation by liposomes include hydration of the epidermis and the sustained release of drugs, hence avoiding the buildup of toxic drug concentrations in the skin [101]. For example, tretinoin formulated in liposomes demonstrated decreased skin irritation with respect to the equipotent gel formulation when administered to patients [102]. It is possible that using a hydrogel or a cream containing liposome- or microsphere-entrapped drug may decrease the skin irritation potential of the formulation applied to the skin’s surface.

Reinforcing the protective barrier of the skin by addition of an emollient to topical formulations has been shown to reduce or prevent skin irritation as well. For example, Zhai et al. showed that applying a cream containing paraffin wax in cetyl alcohol to the skin prior to treatment with sodium lauryl sulfate or ammonium hydroxide prevented irritation caused by these known irritants in the absence of the applied cream [103]. Additionally, in a clinical study conducted by Wigger-Albeti et al. [104], it was shown that the irritant effect of known irritants sodium hydroxide, toluene and lactic acid on human skin was prevented by applying petrolatum to the skin before the irritant. Petrolatum was found to be less effective against toluene-induced irritation.

Chemical permeation enhancers

The stratum corneum is a formidable barrier to exogenous agents including drugs. Therefore, it is often necessary to add permeation-enhancing chemicals to aid drugs in passing through the stratum corneum. Permeation-enhancing chemicals include fatty acids, organic solvents (i.e., acetone and ethanol), alcohols, esters and surfactants, among many others described earlier in this article. It is generally understood that for enhancers, increased potency is directly correlated with increased skin irritation. Difficulty in reducing the irritation of these agents has been expressed since the same mechanisms responsible for increasing permeation cause irritation. While potent enhancers are effective at transiently compromising the integrity of the stratum corneum barrier, their action is not entirely limited to the stratum corneum and the interaction with viable epidermis can cause cytotoxicity and irritation. However, studies have been performed with the objective of reducing the irritancy potential of known permeation enhancers without decreasing their potency with some success. Published methods for reducing the skin irritation of permeation enhancers include combining permeation enhancers (synergistic mixtures) and manipulation of their chemical structures. Ben-Shabat, Baruch and Sintov prepared propylene glycol mono- and di-ester derivatives of saturated and unsaturated fatty acids to improve their permeation enhancement ability without increasing their irritation [105]. None of the derivatives tested showed greater permeation enhancement than the corresponding fatty acid, although the propylene glycol conjugates of oleic and linoleic acid diminished skin irritation and maintained equivalent enhancement capability when compared with the free fatty acid. Sintov and Ben-Shabat have also reported on the formation of fatty acid–drug conjugates to increase permeation without jeopardizing their safety [106]. Finally, Karande and Mitragotri have summarized numerous permeation enhancer combinations, their safety and the synergistic enhancement of these combinations in increasing drug permeability [107]. One example given was a cyclodextrin–enhancer complex that reduced side effects of the enhancer while maintaining its permeabilization ability. In summary, it appears that combinations of permeation enhancers, solvents and drugs may be able to increase potency without causing excess irritation.

Anti-irritants & corticosteroids in reducing skin irritation

Pre-application of steroids to the skin and incorporation of steroid or anti-irritants into irritant formulations have been demonstrated to reduce skin reactions. Topical pretreatment with a 0.1%-triamcinolone acetonide cream was shown to decrease the incidence and/or severity of skin reactions associated with TDS exposure in humans. This was determined based on the comparison of cumulative irritation scores obtained in patients that were pretreated with steroid versus those to whom pretreatment was not applied [108]. Lastly, incorporation of anti-irritants into irritant formulations has been shown to decrease adverse skin reactions [109]. Some anti-irritants reported to decrease side effects caused by irritants are glycerol [110,111], triamcinolone acetonide, [111], lobetasol and diphen-hydramine [112]. These systems were shown to reduce sodium lauryl sulfate-, nonanoic acid- and captopril gel-induced skin irritation, respectively.

Future perspective

Expanding the use of novel permeation enhancement techniques with macromolecules and other conventional molecules for a wider range of indications is highly desirable for the transdermal industry. Physical enhancement methods afford substantial improvement in the rate of delivery of therapeutic agents across skin. Currently, a variety of them are undergoing extensive investigation and new device-based TDS can be expected in the near future. One can also expect the first transdermal prodrug product to emerge on the market in the near future. Novel prodrugs would not only help to reach the therapeutic levels for some drugs, but may also help alleviate skin irritation.

The incidence and significance of skin irritation reactions will decrease with the increasing availability of physical permeation enhancement methods and new breakthroughs in topical drug formulations, such as liposomes, microemulsions, nanoparticles and evaporating gels. Breakthroughs in chemical permeation enhancer analogs showing significant improvements in limiting cutaneous irritation show promise for the development of safe chemical enhancers and should be further examined in the future.

Executive summary.

  • Approximately 20 transdermal and 10 topical/dermal products make up the current US transdermal market.

  • Most transdermal products on the US market today are in the conventional patch form, but a few variations such as a programmable patch, metered spray or the gel formulations have also been approved by the US FDA. These transdermal products are indicated for a wide range of therapies.

  • Although reservoir and matrix patches share an almost equal ratio among the approved patch products, matrix patches are the most desired and the ideal goal for future products. The newer patches are much smaller in size compared with the older conventional systems.

  • Transdermal products for local treatment currently available on the US market come in gel and patch forms. These products are mainly indicated for local pain treatment of a variety of conditions.

  • Transdermal products that are currently in FDA-approved clinical trials are investigational agents, as well as approved transdermal products for new indications or in various combinations with other drugs.

  • Microporation techniques have gained momentum in the drug approval process and products may be on the market soon for a wide range of molecules.

  • Increase in skin permeability is a prerequisite for successful delivery of new macromolecular drugs and improved delivery of conventional drugs.

  • Although many individual chemical enhancers often fail to provide the desired enhancement level without exhibiting safety concerns, some combinations of chemical enhancers act synergistically to overcome these limitations.

  • Physical enhancement methods afford substantial improvement in the rate of delivery of therapeutic agents across skin. Currently, a variety of them are undergoing extensive investigation.

  • Prodrugs allow the physicochemical properties of drug molecules to be changed, which facilitates crossing of the stratum corneum barrier.

  • It is difficult to predict the incidence of cutaneous irritation caused by drugs, patch components and formulation components prior to in vivo testing.

  • Drug and permeation enhancer analogs have shown superior safety over their corresponding parent forms.

  • Reported ways to prevent or reduce skin irritation of topical and transdermal systems include:

    • Maintaining the pH of the topically applied formulation near the skin pH

    • Avoiding drugs that possess a dissociation constant (pKa) of less than 4 or greater than 8

    • Formulating the active pharmaceutical ingredient in a hydrogel or liposomal system

    • Adding corticosteroids or anti-irritants to the drug formulation

Key Term

Chemical permeation enhancer

A substance that temporarily changes the nature of the skin barrier, stratum corneum, so that the passage of the drug molecules across it is facilitated

Prodrugs

A pharmacologically inactive substance that undergoes in vivo activation to form the active metabolite (parent molecule)

Cutaneous irritation

A complex biological reaction to exogenous stimuli applied to the skin or caused by damage to the skin barrier, identified by superficial pH, transepidermal water loss, skin hydration, skin color, blood flow and barrier resistance

Barrier function

Intactness of the stratum corneum barrier of the skin and its ability to deter chemicals, microbes and loss of water from the viable skin layers

Transepidermal water loss

Evaporation of water from the skin’s surface as well as a physical tool for assessing perturbations of the skin barrier

Footnotes

For reprint orders, please contact reprints@future-science.com

Financial & competing interests disclosure

Audra L Stinchcomb is funded by NIH grants R01DA13425, R01DA18822, R43DA26266 and RC2DA28984. Courtney L Swadley is funded by NIH Grant F31DA025424. Nicole K Brogden is funded by NIH grant F31DA029374. Audra L Stinchcomb is a significant shareholder in and Chief Scientific Officer of AllTranz Inc., a transdermal company. 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.

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

Bibliography

Papers of special note have been highlighted as:

▪ of interest

▪ ▪ of considerable interest

  • 1.Jain PharmaBiotech report. Transdermal drug delivery-technologies, markets and companies. 2005. [Google Scholar]
  • 2.Prausnitz MR, Langer R. Transdermal drug delivery. Nat Biotech. 2008;26(11):1261–1268. doi: 10.1038/nbt.1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Martin E, editor. Remington’s Practice of Pharmacy. Mack Publishing Co; Easton, PA, USA: 2003. [Google Scholar]
  • 4.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]
  • 5.Cevc G, Vierl U. Nanotechnology and the transdermal route. A state of the art review and critical appraisal. J Control Release. 2010;141(11):277–299. doi: 10.1016/j.jconrel.2009.10.016. [DOI] [PubMed] [Google Scholar]
  • 6.Tiwary AK, Sapra B, Jain S. Innovations in transdermal drug delivery: formulations and techniques. Recent Pat Drug Deliv Formul. 2007;1:23–36. doi: 10.2174/187221107779814087. [DOI] [PubMed] [Google Scholar]
  • 7.Samad A, Ullah Z, Alam MI, Wais M, Shams MS. Transdermal drug delivery system: patent reviews. Recent Pat Drug Deliv Formul. 2009;3(2):143–52. doi: 10.2174/187221109788452294. [DOI] [PubMed] [Google Scholar]
  • 8.Rizwan M, Aqil M, Talegaonkar S, Azeem A, Sultana Y, Ali A. Enhanced transdermal drug delivery techniques: an extensive review of patents. Recent Pat Drug Deliv Formul. 2009;3(2):105–124. doi: 10.2174/187221109788452285. [DOI] [PubMed] [Google Scholar]
  • 9.Prausnitz MR, Mitragotri S, Langer R. Current status and future potential of transdermal drug delivery. Nat Rev Drug Discov. 2004;3(2):115–124. doi: 10.1038/nrd1304. [DOI] [PubMed] [Google Scholar]
  • 10.Banga AK. Microporation applications for enhancing drug delivery. Expert Opin Drug Deliv. 2009;6(4):343–54. doi: 10.1517/17425240902841935. [DOI] [PubMed] [Google Scholar]
  • 11.Kalluri H, Banga AK. Microneedles and transdermal drug delivery. J Drug Del Sci Tech. 2009;19(5):303–310. [Google Scholar]
  • 12.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]
  • 13▪.Thong HY, Zhai H, Maibach HI. Percutaneous penetration enhancers: an overview. Skin Pharmacol Physiol. 2007;20(6):272–282. doi: 10.1159/000107575. Recent review containing extensive list of popular chemical permeation enhancers. [DOI] [PubMed] [Google Scholar]
  • 14.Michaels AS, Chandrasekaran SK, Shaw JE. Drug permeation through human skin. Theory and in vitro experimental measurement. AIChE J. 1975;21(5):985–996. [Google Scholar]
  • 15.Albery WJ, Hadgraft J. Percutaneous absorption: in vivo experiments. J Pharm Pharmacol. 1979;31(3):140–147. doi: 10.1111/j.2042-7158.1979.tb13456.x. [DOI] [PubMed] [Google Scholar]
  • 16.Tojo K. Random brick model for drug transport across stratum corneum. J Pharm Sci. 1987;76(12):889–891. doi: 10.1002/jps.2600761209. [DOI] [PubMed] [Google Scholar]
  • 17.Goffin V, Henry F, Pierard-Franchimont C, Pierard GE. Penetration enhancers assessed by corneoxenometry. Skin Pharmacol Appl Skin Physiol. 2000;13(5):280–284. doi: 10.1159/000029934. [DOI] [PubMed] [Google Scholar]
  • 18.Barry BW. Lipid–protein-partitioning theory of skin penetration enhancement. J Control Release. 1991;15(3):237–248. doi: 10.1023/a:1015813803205. [DOI] [PubMed] [Google Scholar]
  • 19.Karande P, Jain A, Ergun K, Kispersky V, Mitragotri S. Design principles of chemical penetration enhancers for transdermal drug delivery. Proc Natl Acad Sci USA. 2005;102(13):4688–4693. doi: 10.1073/pnas.0501176102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kogan A, Garti N. Microemulsions as transdermal drug delivery vehicles. Adv Colloid Interface Sci. 2006;123–126:369–385. doi: 10.1016/j.cis.2006.05.014. [DOI] [PubMed] [Google Scholar]
  • 21▪ ▪.Karande P, Jain A, Mitragotri S. Discovery of transdermal penetration enhancers by high-throughput screening. Nat Biotechnol. 2004;22(2):192–197. doi: 10.1038/nbt928. Identification of synergistic chemical permeation enhancer combinations through high-throughput screening. [DOI] [PubMed] [Google Scholar]
  • 22.Karande P, Jain A, Mitragotri S. Insights into synergistic interactions in binary mixtures of chemical permeation enhancers for transdermal drug delivery. J Control Release. 2006;115(1):85–93. doi: 10.1016/j.jconrel.2006.07.001. [DOI] [PubMed] [Google Scholar]
  • 23.Hamilton JG. Needle phobia: a neglected diagnosis. J Fam Practice. 1995;41(2):169–175. [PubMed] [Google Scholar]
  • 24.Burkoth TL, Bellhouse BJ, Hewson G, Longridge DJ, Muddle AG, Sarphie DF. Transdermal and transmucosal powdered drug delivery. Crit Rev Ther Drug Carrier Syst. 1999;16(4):331–384. doi: 10.1615/critrevtherdrugcarriersyst.v16.i4.10. [DOI] [PubMed] [Google Scholar]
  • 25.Schramm J, Mitragotri S. Transdermal drug delivery by jet injectors: energetics of jet formation and penetration. Pharm Res. 2002;19(11):1673–1679. doi: 10.1023/a:1020753329492. [DOI] [PubMed] [Google Scholar]
  • 26.Bremseth DL, Pass F. Delivery of insulin by jet injection: recent observations. Diabetes Technol Ther. 2001;3(2):225–232. doi: 10.1089/152091501300209598. [DOI] [PubMed] [Google Scholar]
  • 27.Denne JR, Andrews KL, Lees DV, Mook W. A survey of patient preference for insulin jet injectors versus needle and syringe. Diabetes Educ. 1992;18(3):223–227. doi: 10.1177/014572179201800309. [DOI] [PubMed] [Google Scholar]
  • 28.Dean Hansi J, Fuller D, Osorio Jorge E. Powder and particle-mediated approaches for delivery of DNA and protein vaccines into the epidermis. Comp Immunol Microbiol Infect Dis. 2003;26(5–6):373–388. doi: 10.1016/S0147-9571(03)00021-3. [DOI] [PubMed] [Google Scholar]
  • 29.Spencer JM. Microdermabrasion. Am J Clin Dermatol. 2005;6(2):89–92. doi: 10.2165/00128071-200506020-00003. [DOI] [PubMed] [Google Scholar]
  • 30.Fang JY, Lee WR, Shen SC, Fang YP, Hu CH. Enhancement of topical 5-aminolaevulinic acid delivery by erbium:YAG laser and microdermabrasion: a comparison with iontophoresis and electroporation. Br J Dermatol. 2004;151(1):132–140. doi: 10.1111/j.1365-2133.2004.06051.x. [DOI] [PubMed] [Google Scholar]
  • 31.Fujimoto T, Shirakami K, Tojo K. Effect of microdermabrasion on barrier capacity of stratum corneum. Chem Pharm Bull. 2005;53(8):1014–1016. doi: 10.1248/cpb.53.1014. [DOI] [PubMed] [Google Scholar]
  • 32.Gill HS, Andrews SN, Sakthivel SK, et al. Selective removal of stratum corneum by microdermabrasion to increase skin permeability. Eur J Pharm Sci. 2009;38(2):95–103. doi: 10.1016/j.ejps.2009.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33▪ ▪.Park JH, Lee JW, Kim YC, Prausnitz MR. The effect of heat on skin permeability. Int J Pharm. 2008;359(1–2):94–103. doi: 10.1016/j.ijpharm.2008.03.032. A study of the structural disruption of skin subjected to high temperature. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Lee WR, Shen SC, Lai HH, Hu CH, Fang JY. Transdermal drug delivery enhanced and controlled by erbium:YAG laser: a comparative study of lipophilic and hydrophilic drugs. J Control Release. 2001;75(1–2):155–166. doi: 10.1016/s0168-3659(01)00391-1. [DOI] [PubMed] [Google Scholar]
  • 35.Nelson JS, McCullough JL, Glenn TC, Wright WH, Liaw LH, Jacques SL. Mid-infrared laser ablation of stratum corneum enhances in vitro percutaneous transport of drugs. J Invest Dermatol. 1991;97(5):874–879. doi: 10.1111/1523-1747.ep12491600. [DOI] [PubMed] [Google Scholar]
  • 36.Gomez C, Costela A, Garcia-Moreno I, Llanes F, Teijon JM, Blanco D. Laser treatments on skin enhancing and controlling transdermal delivery of 5-fluorouracil. Laser Surg Med. 2008;40(1):6–12. doi: 10.1002/lsm.20590. [DOI] [PubMed] [Google Scholar]
  • 37.Arora A, Prausnitz MR, Mitragotri S. Micro-scale devices for transdermal drug delivery. Int J Pharm. 2008;364(2):227–236. doi: 10.1016/j.ijpharm.2008.08.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Henry S, McAllister DV, Allen MG, Prausnitz MR. Microfabricated microneedles: a novel approach to transdermal drug delivery. J Pharm Sci. 1998;87(8):922–925. doi: 10.1021/js980042+. [DOI] [PubMed] [Google Scholar]
  • 39.Sullivan SP, Murthy N, Prausnitz MR. Minimally invasive protein delivery with rapidly dissolving polymer microneedles. Adv Mater. 2008;20(5):933–938. doi: 10.1002/adma.200701205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Banks SL, Pinninti RR, Gill HS, Crooks PA, Prausnitz MR, Stinchcomb AL. Flux across microneedle-treated skin is increased by increasing charge of naltrexone and naltrexol in vitro. Pharm Res. 2008;25(7):1677–1685. doi: 10.1007/s11095-008-9578-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Gill HS, Prausnitz MR. Coated microneedles for transdermal delivery. J Control Release. 2007;117(2):227–237. doi: 10.1016/j.jconrel.2006.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lanke SSS, Kolli CS, Strom JG, Banga AK. Enhanced transdermal delivery of low molecular weight heparin by barrier perturbation. Int J Pharm. 2009;365(1–2):26–33. doi: 10.1016/j.ijpharm.2008.08.028. [DOI] [PubMed] [Google Scholar]
  • 43.Wermeling DP, Banks SL, Hudson DA, et al. Microneedles permit transdermal delivery of a skin-impermeant medication to humans. Proc Natl Acad Sci USA. 2008;105(6):2058–2063. doi: 10.1073/pnas.0710355105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Riviere JE, Heit MC. Electrically-assisted transdermal drug delivery. Pharm Res. 1997;14(6):687–697. doi: 10.1023/a:1012129801406. [DOI] [PubMed] [Google Scholar]
  • 45.Warwick WJ, Huang NN, Waring WW, et al. Evaluation of a cystic fibrosis screening system incorporating a miniature sweat stimulator and disposable chloride sensor. Clin Chem. 1986;32(5):850–853. [PubMed] [Google Scholar]
  • 46.Squire SJ, Kirchhoff KT, Hissong K. Comparing two methods of topical anesthesia used before intravenous cannulation in pediatric patients. J Pediatr Healthcare. 2000;14(2):68–72. doi: 10.1067/mph.2000.103647. [DOI] [PubMed] [Google Scholar]
  • 47.Patel SR, Zhong H, Sharma A, Kalia YN. Controlled non-invasive transdermal iontophoretic delivery of zolmitriptan hydrochloride in vitro and in vivo. Eur J Pharm Biopharm. 2009;72(2):304–309. doi: 10.1016/j.ejpb.2008.08.006. [DOI] [PubMed] [Google Scholar]
  • 48.Nakamura K, Katagai K, Mori K, Higo N, Sato S, Yamamoto K. Transdermal administration of salmon calcitonin by pulse depolarization-iontophoresis in rats. Int J Pharm. 2001;218(1–2):93–102. doi: 10.1016/s0378-5173(01)00615-9. [DOI] [PubMed] [Google Scholar]
  • 49.Pillai O, Panchagnula R. Transdermal delivery of insulin from poloxamer gel: ex vivo and in vivo skin permeation studies in rat using iontophoresis and chemical enhancers. J Control Release. 2003;89(1):127–140. doi: 10.1016/s0168-3659(03)00094-4. [DOI] [PubMed] [Google Scholar]
  • 50.Rastogi SK, Singh J. Transepidermal transport enhancement of insulin by lipid extraction and iontophoresis. Pharm Res. 2002;19(4):427–433. doi: 10.1023/a:1015131325930. [DOI] [PubMed] [Google Scholar]
  • 51.Suzuki Y, Nagase Y, Iga K, et al. Prevention of bone loss in ovariectomized rats by pulsatile transdermal iontophoretic administration of 91(2), human PTH1–34. J Pharm Sci. 2002:350–361. doi: 10.1002/jps.10019. [DOI] [PubMed] [Google Scholar]
  • 52.Nair V, Panchagnula R. Physicochemical considerations in the iontophoretic delivery of a small peptide: in vitro studies using arginine vasopressin as a model peptide. Pharmacol Res. 2003;48(2):175–182. doi: 10.1016/s1043-6618(03)00080-x. [DOI] [PubMed] [Google Scholar]
  • 53.Galinkin JL, Rose JB, Harris K, Watcha MF. Lidocaine iontophoresis versus eutectic mixture of local anesthetics (EMLA®) for IV placement in children. Anesth Analg. 2002;94(6):1484–1488. doi: 10.1097/00000539-200206000-00020. [DOI] [PubMed] [Google Scholar]
  • 54.Runeson L, Haker E. Iontophoresis with cortisone in the treatment of lateral epicondylalgia (tennis elbow) – a double-blind study. Scand J Med Sci Sports. 2002;12(3):136–142. doi: 10.1034/j.1600-0838.2002.02142.x. [DOI] [PubMed] [Google Scholar]
  • 55.Gupta SK, Bernstein KJ, Noorduin H, Van Peer A, Sathyan G, Haak R. Fentanyl delivery from an electrotransport system: delivery is a function of total current, not duration of current. J Clin Pharmacol. 1998;38(10):951–958. doi: 10.1002/j.1552-4604.1998.tb04392.x. [DOI] [PubMed] [Google Scholar]
  • 56.Singh J, Gross M, Sage B, Davis HT, Maibach HI. Regional variations in skin barrier function and cutaneous irritation due to iontophoresis in human subjects. Food Chem Toxicol. 2001;39(11):1079–1086. doi: 10.1016/s0278-6915(01)00057-6. [DOI] [PubMed] [Google Scholar]
  • 57.Prausnitz MR. A practical assessment of transdermal drug delivery by skin electroporation. Adv Drug Deliv Rev. 1999;35(1):61–76. doi: 10.1016/s0169-409x(98)00063-5. [DOI] [PubMed] [Google Scholar]
  • 58.Denet AR, Preat V. Transdermal delivery of timolol by electroporation through human skin. J Control Release. 2003;88(2):253–262. doi: 10.1016/s0168-3659(03)00010-5. [DOI] [PubMed] [Google Scholar]
  • 59.Zewert TE, Pliquett UF, Vanbever R, Langer R, Weaver JC. Creation of transdermal pathways for macromolecule transport by skin electroporation and a low toxicity, pathway-enlarging molecule. Bioelectrochem Bioenerg. 1999;49(1):11–20. doi: 10.1016/s0302-4598(99)00056-2. [DOI] [PubMed] [Google Scholar]
  • 60.Mitragotri S. Effect of therapeutic ultrasound on partition and diffusion coefficients in human stratum corneum. J Control Release. 2001;71(1):23–29. doi: 10.1016/s0168-3659(00)00344-8. [DOI] [PubMed] [Google Scholar]
  • 61.Tang H, Wang CCJ, Blankschtein D, Langer R. An investigation of the role of cavitation in low-frequency ultrasound-mediated transdermal drug transport. Pharm Res. 2002;19(8):1160–1169. doi: 10.1023/a:1019898109793. [DOI] [PubMed] [Google Scholar]
  • 62.Mitragotri S, Blankschtein D, Langer R. Transdermal drug delivery using low-frequency sonophoresis. Pharm Res. 1996;13(3):411–420. doi: 10.1023/a:1016096626810. [DOI] [PubMed] [Google Scholar]
  • 63.Mitragotri S, Blankschtein D, Langer R. Ultrasound-mediated transdermal protein delivery. Science. 1995;269(5225):850–853. doi: 10.1126/science.7638603. [DOI] [PubMed] [Google Scholar]
  • 64.Mitragotri S, Edwards DA, Blankschtein D, Langer R. Mechanistic study of ultrasonically-enhanced transdermal drug-delivery. J Pharm Sci. 1995;84(6):697–706. doi: 10.1002/jps.2600840607. [DOI] [PubMed] [Google Scholar]
  • 65.Mitragotri S, Farrell J, Tang H, Terahara T, Kost J, Langer R. Determination of threshold energy dose for ultrasound-induced transdermal drug transport. J Control Release. 2000;63(1–2):41–52. doi: 10.1016/s0168-3659(99)00178-9. [DOI] [PubMed] [Google Scholar]
  • 66▪.Cross SE, Roberts MS. Physical enhancement of transdermal drug application: is delivery technology keeping up with pharmaceutical development? Curr Drug Delivery. 2004;1(1):81–92. doi: 10.2174/1567201043480045. Extensive review of the physical enhancement methods used in percutaneous drug delivery. [DOI] [PubMed] [Google Scholar]
  • 67.Kost J, Pliquett U, Mitragotri S, Yamamoto A, Langer R, Weaver J. Synergistic effect of electric field and ultrasound on transdermal transport. Pharm Res. 1996;13(4):633–638. doi: 10.1023/a:1016070710397. [DOI] [PubMed] [Google Scholar]
  • 68.Riviere JE, Monteiroriviere NA, Rogers RA, Bommannan D, Tamada JA, Potts RO. Pulsatile transdermal delivery of LHRH using electroporation: drug delivery and skin toxicology. J Control Release. 1995;36(3):229–233. [Google Scholar]
  • 69.Chang SL, Hofmann GA, Zhang L, Deftos LJ, Banga AK. The effect of electroporation on iontophoretic transdermal delivery of calcium regulating hormones. J Control Release. 2000;66(2–3):127–133. doi: 10.1016/s0168-3659(99)00262-x. [DOI] [PubMed] [Google Scholar]
  • 70.Prausnitz MR. Microneedles for transdermal drug delivery. Adv Drug Deliv Rev. 2004;56 (5):581–587. doi: 10.1016/j.addr.2003.10.023. [DOI] [PubMed] [Google Scholar]
  • 71.Chen H, Zhu H, Zheng J, et al. Iontophoresis-driven penetration of nanovesicles through microneedle-induced skin microchannels for enhancing transdermal delivery of insulin. J Control Release. 2009;139(1):63–72. doi: 10.1016/j.jconrel.2009.05.031. [DOI] [PubMed] [Google Scholar]
  • 72▪.Kasting GB, Smith RL, Anderson BD. In: Prodrugs: Topical and Ocular Drug Delivery. Sloan KB, editor. Marcel Dekker; NY, USA: 1992. pp. 117–161. Discussion of prodrug design for transdermal delivery. [Google Scholar]
  • 73.Qandil A, Al-Nabulsi S, Al-Taani B, Tashtoush B. Synthesis of piperazinylalkyl ester prodrugs of ketorolac and their in vitro evaluation for transdermal delivery. Drug Dev Ind Pharm. 2008;34(10):1054–1063. doi: 10.1080/03639040801946681. [DOI] [PubMed] [Google Scholar]
  • 74.Kushnir M, Yaar A, Reichman A, Heldman E. Transdermal delivery of a levodopa prodrug; a pilot clinical trial. Mov Disord. 2008;23:592. [Google Scholar]
  • 75.Thomas JD, Majumdar S, Sloan KB. Soft alkyl ether prodrugs of a model phenolic drug: the effect of incorporation of ethyleneoxy groups on transdermal delivery. Molecules. 2009;14(10):4231–4245. doi: 10.3390/molecules14104231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Strasinger CL, Scheff NN, Stinchcomb AL. Prodrugs and codrugs as strategies for improving percutaneous absorption. Expert Rev Dermatol. 2008;3(2):221–233. [Google Scholar]
  • 77.Vaddi HK, Banks SL, Chen J, Hammell DC, Crooks PA, Stinchcomb AL. Human skin permeation of 3-O-alkyl carbamate prodrugs of naltrexone. J Pharm Sci. 2009;98(8):2611–2625. doi: 10.1002/jps.21594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kiptoo PK, Paudel KS, Hammell DC, et al. Transdermal delivery of bupropion and its active metabolite, hydroxybupropion: a prodrug strategy as an alternative approach. J Pharm Sci. 2009;98(2):583–594. doi: 10.1002/jps.21463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79▪.Murphy M, Carmichael AJ. Transdermal drug delivery systems and skin sensitivity reactions: incidence and management. Am J Clin Dermatol. 2000;1(6):361–368. doi: 10.2165/00128071-200001060-00004. Discusses characteristics, sources, management and prevention of skin irritation reactions caused by drug delivery systems. [DOI] [PubMed] [Google Scholar]
  • 80.Berner B, Wilson DR, Guy RH, Mazzenga GC, Clarke FH, Maibach HI. The relationship of pka and acute skin irritation in man. Pharm Res. 1988;5(10):660–663. doi: 10.1023/a:1015931105660. [DOI] [PubMed] [Google Scholar]
  • 81.Berner BWD, Steffens RJ, Mazzenga GC, Hinz R, Guy RH, Maibach HI. The relationship between pka and skin irritation for a series of basic penetrants in man. Fundam Appl Toxicol. 1990;15(4):760–766. doi: 10.1016/0272-0590(90)90192-m. [DOI] [PubMed] [Google Scholar]
  • 82.Andersen PH, Nangia A, Bjerring P, Maibach HI. Chemical and pharmacologic skin irritation in man. Contact Derm. 1991;25(5):283–289. doi: 10.1111/j.1600-0536.1991.tb01875.x. [DOI] [PubMed] [Google Scholar]
  • 83.Mangia A, Andersen PH, Berner B, Maibach HI. High dissociation constants (pKa) of basic permeants are associated with in vivo skin irritation in man. Contact Derm. 1996;34(4):237–242. doi: 10.1111/j.1600-0536.1996.tb02192.x. [DOI] [PubMed] [Google Scholar]
  • 84.Johnson W. Final report on the safety assessment of capsicum annuum extract, capsicum annuum fruit extract, capsicum annuum resin, capsicum annuum fruit powder, capsicum frutescens fruit, capsicum frutescens fruit extract, capsicum frutescens resin, and capsaicin. Int J Toxicol. 2007;26(Suppl 1):3–106. doi: 10.1080/10915810601163939. [DOI] [PubMed] [Google Scholar]
  • 85.Branco N, Lee I, Hongbo Z, Maibach HI. Long-term repetitive sodium lauryl sulfate-induced irritation of the skin: an in vivo study. Contact Derm. 2005;53(5):278–284. doi: 10.1111/j.0105-1873.2005.00703.x. [DOI] [PubMed] [Google Scholar]
  • 86▪.Fluhr JW, Darlenski R, Angelova-Fischer I, Tsankov N, Basketter D. Skin irritation and sensitization: mechanisms and new approaches for risk assessment. Skin Pharmacol Physiol. 2008;21(3):124–135. doi: 10.1159/000131077. Discusses skin irritation and sensitization in detail as well as current methods used to determine their presence and extent. [DOI] [PubMed] [Google Scholar]
  • 87.Fang JY, Tsai MJ, Huang YB, Wu PC, Tsai YH. Percutaneous absorption and skin erythema: quantification of capsaicin and its synthetic derivatives from gels incorporated with benzalkonium chloride by using non-invasive bioengineering methods. Drug Dev Res. 1997;40(1):56–67. [Google Scholar]
  • 88.Schmid-Wendtner MH, Korting HC. The pH of the skin surface and its impact on the barrier function. Skin Pharmacol Physiol. 2006;19(6):296–302. doi: 10.1159/000094670. [DOI] [PubMed] [Google Scholar]
  • 89.Antoine JL, Contreras JL, Van Neste DJ. pH influence of surfactant-induced skin irritation. Derm Beruf Umwelt. 1989;37(3):96–100. [PubMed] [Google Scholar]
  • 90.Ananthapadmanabhan KP, Lips A, Vincent C, et al. pH-induced alterations in stratum corneum properties. Int J Cosmetic Sci. 2003;25(3):103–112. doi: 10.1046/j.1467-2494.2003.00176.x. [DOI] [PubMed] [Google Scholar]
  • 91.Williams A. Pharmaceutical solvents as vehicles for topical dosage forms. In: Augustijns P, Brewster ME, editors. Solvent Systems and Their Selection in Pharmaceutics and Biopharmaceutics. Vol. 6. Springer; NY, USA: 2007. pp. 403–426. [Google Scholar]
  • 92.Matsumura H, Oka K, Umekage K, et al. Effect of occlusion on human skin. Contact Derm. 1995;33(4):231–235. doi: 10.1111/j.1600-0536.1995.tb00472.x. [DOI] [PubMed] [Google Scholar]
  • 93.Homick JLKR, Reschke MF, Degioanni J, Cintron-Trevino NM. Transdermal scopolamine in the prevention of motion sickness: evaluation of the time course of efficacy. Aviat Space Environ Med. 1983;54(11):994–1000. [PubMed] [Google Scholar]
  • 94.Hurkmans JF, Bodde HE, Driel LM, Doorne HV, Junginger HE. Skin irritation caused by transdermal drug delivery systems during long-term (5 days) application. Br J Dermatol. 1985;112(4):461–467. doi: 10.1111/j.1365-2133.1985.tb02321.x. [DOI] [PubMed] [Google Scholar]
  • 95.Van der Valk PG, Maibach HI. Post-application occlusion substantially increases the irritant response of the skin to repeated short-term sodium lauryl sulfate (SLS) exposure. Contact Derm. 1989;21(5):335–338. doi: 10.1111/j.1600-0536.1989.tb04754.x. [DOI] [PubMed] [Google Scholar]
  • 96.Tsen-Fang T, Maibach HI. How irritant is water? An overview. Contact Derm. 1999;41(6):311–314. doi: 10.1111/j.1600-0536.1999.tb06990.x. [DOI] [PubMed] [Google Scholar]
  • 97.Dwyer CM, Forsyth A. Allergic contact dermatitis from methacrylates in a nicotine transdermal patch. Contact Derm. 1994;30(5):309–310. doi: 10.1111/j.1600-0536.1994.tb00612.x. [DOI] [PubMed] [Google Scholar]
  • 98.Ross D, Rees M, Godfree V, et al. Randomised crossover comparison of skin irritation with two transdermal oestradiol patches. BMJ. 1997;315(7103):288. doi: 10.1136/bmj.315.7103.288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Marier JF, Lor M, Potvin D, Dimarco M, Morelli G, Saedder EA. Pharmacokinetics, tolerability, and performance of a novel matrix transdermal delivery system of fentanyl relative to the commercially available reservoir formulation in healthy subjects. J Clin Pharmacol. 2006;46(6):642–653. doi: 10.1177/0091270006286901. [DOI] [PubMed] [Google Scholar]
  • 100.Wester RC, Patel R, Nacht S, Leyden J, Melendres J, Maibach H. Controlled release of benzoyl peroxide from a porous microsphere polymeric system can reduce topical irritancy. J Am Acad Dermatol. 1991;24(5 Pt 1):720–726. doi: 10.1016/0190-9622(91)70109-f. [DOI] [PubMed] [Google Scholar]
  • 101▪.de Leeuw J, de Vijlder HC, Bjerring P, Neumann HAM. Liposomes in dermatology today. J Eur Acad Dermatol. 2009;23(5):505–516. doi: 10.1111/j.1468-3083.2009.03100.x. Discusses the characteristics of liposome formulations, a synopsis of the state of liposome research and how liposomal formulations are impacting the field of dermal drug delivery. [DOI] [PubMed] [Google Scholar]
  • 102.Schäfer-Korting M, Korting HC, Ponce-Pöschl E. Liposomal tretinoin for uncomplicated acne vulgaris. Clin Invest. 1994;72(12):1086–1091. doi: 10.1007/BF00577761. [DOI] [PubMed] [Google Scholar]
  • 103.Zhai H, Willard P, Maibach HI. Evaluating skin-protective materials against contact irritants and allergens. Contact Derm. 1998;38(3):155–158. doi: 10.1111/j.1600-0536.1998.tb05683.x. [DOI] [PubMed] [Google Scholar]
  • 104.Wigger-Alberti W, Hinnen U, Elsner P. Predictive testing of metalworking fluids: a comparison of 2 cumulative human irritation models and correlation with epidemiological data. Contact Derm. 1997;6(1):14–20. doi: 10.1111/j.1600-0536.1997.tb00916.x. [DOI] [PubMed] [Google Scholar]
  • 105.Ben-Shabat S, Baruch N, Sintov AC. Conjugates of unsaturated fatty acids with propylene glycol as potentially less-irritant skin penetration enhancers. Drug Dev Ind Pharm. 2007;33(11):1169–1175. doi: 10.1080/03639040701199258. [DOI] [PubMed] [Google Scholar]
  • 106.Sintov A, Ben-Shabat S. Design of fatty acid conjugates for dermal delivery and topical therapeutics. Anglais. 2006;23(1):67–87. doi: 10.1615/critrevtherdrugcarriersyst.v23.i1.20. [DOI] [PubMed] [Google Scholar]
  • 107.Karande P, Mitragotri S. Enhancement of transdermal drug delivery via synergistic action of chemicals. Biochim Biophys Acta Biomembranes. 2009;1788(11):2362–2373. doi: 10.1016/j.bbamem.2009.08.015. [DOI] [PubMed] [Google Scholar]
  • 108.Wilson DE, Kaidbey K, Boike SC, Jorkasky DK. Use of topical corticosteroid pretreatment to reduce the incidence and severity of skin reactions associated with testosterone transdermal therapy. Clin Ther. 1998;20(2):299–306. doi: 10.1016/s0149-2918(98)80093-3. [DOI] [PubMed] [Google Scholar]
  • 109.Amkraut AA, Jordan WP, Taskovich L. Effect of coadministration of corticosteroids on the development of contact sensitization. J Am Acad Dermatol. 1996;35(1):27–31. doi: 10.1016/S0190-9622(96)90491-0. [DOI] [PubMed] [Google Scholar]
  • 110.Andersen F, Hedegaard K, Petersen TK, Bindslev-Jensen C, Fullerton A, Andersen KE. Anti-irritants I: Dose-response in acute irritation. Contact Derm. 2006;55(3):148–154. doi: 10.1111/j.1600-0536.2006.00752.x. [DOI] [PubMed] [Google Scholar]
  • 111.Andersen F, Hedegaard K, Petersen TK, Bindslev-Jensen C, Fullerton A, Andersen KE. Comparison of the effect of glycerol and triamcinolone acetonide on cumulative skin irritation in a randomized trial. J Am Acad Dermatol. 2007;56(2):228–235. doi: 10.1016/j.jaad.2006.08.063. [DOI] [PubMed] [Google Scholar]
  • 112.Huang YB, Tsai YH, Chang JS, Liu JC, Tsai MJ, Wu PC. Effect of antioxidants and anti-irritants on the stability, skin irritation and penetration capacity of captopril gel. Int J Pharm. 2002;241(2):345–351. doi: 10.1016/s0378-5173(02)00265-x. [DOI] [PubMed] [Google Scholar]

Websites

RESOURCES