You are on page 1of 22

Review

title

Electroporation as an Efficient Physical


Enhancer for Skin Drug Delivery
José Juan Escobar-Chávez, PhD, Dalia Bonilla-Martínez, MSc,
Martha Angélica Villegas-González, MSc, and Alma Luisa Revilla-Vázquez, PhD

Transdermal drug delivery offers an attractive alternative drugs. The in vivo application of high-voltage pulses is
to the conventional drug delivery methods of oral admin- well tolerated, but muscle contractions are usually
istration and injection. However, the stratum corneum induced. The electrode and patch design is an important
acts as a barrier that limits the penetration of substances issue to reduce the discomfort of the electrical treatment
through the skin. Application of high-voltage pulses to the in humans. This review presents the main findings in the
skin increases its permeability (electroporation) and field of electroporation—namely, transdermal drug deliv-
enables the delivery of various substances into and through ery. Particular attention is paid to proposed enhancement
the skin. The application of electroporation to the skin has mechanisms and trends in the field of topical and trans-
been shown to increase transdermal drug delivery. dermal delivery.
Moreover, electroporation, used alone or in combination
with other enhancement methods, expands the range of
drugs (small to macromolecules, lipophilic or hydrophilic, Keywords: Electroporation; skin; physical enhancers;
charged or neutral molecules) that can be delivered trans- transdermal drug delivery
dermally. The efficacy of transport depends on the electri- Journal of Clinical Pharmacology, 2009;49:1262-1283
cal parameters and the physicochemical properties of © 2009 the American College of Clinical Pharmacology

T ransdermal drug delivery offers several advan-


tages over conventional routes.1,2 It avoids the
first-pass metabolism and the gastrointestinal tract.
of infection. Patient compliance may be improved
by this user-friendly method. Despite the advantages
of transdermal delivery, only a small percentage of
Transdermal delivery has the potential for sustained drugs can be delivered transdermally because of the
and controlled drug release. Moreover, it is a nonin- barrier properties of the skin: only small, potent
vasive mode of drug delivery with no trauma or risk lipophilic drugs can be delivered at therapeutic rates
by passive diffusion.3 Moreover, transport of most
drugs across the skin is very slow, and lag times to
From the División de Estudios de Posgrado (Tecnología Farmacéutica), reach steady-state fluxes are in hours. Achievement
Facultad de Estudios Superiores Cuautitlán-Universidad Nacional of a therapeutically effective drug level is therefore
Autónoma de México, Cuautitlán Izcalli, Estado de México, México difficult without enhancing skin permeation.
(Dr Escobar-Chávez); División de Ciencias Químicas, Sección de Química
In the past 30 years, numerous methods of over-
Analítica, Facultad de Estudios Superiores Cuautitlán-Universidad
Nacional Autónoma de México, Cuautitlán Izcalli, Estado de México,
coming the skin barrier have been described, but they
México (Dr Escobar-Chávez, Dr Bonilla-Martínez, Dr Villegas-González); can be broadly divided into 2 main categories defined
and Laboratorio de Desarrollo de Métodos Analíticos-Facultad de as either passive or active methods. Passive methods
Estudios Superiores Cuautitlán-Universidad Nacional Autónoma de for enhancing transdermal drug delivery include the
México, Cuautitlán Izcalli, Estado de México, México (Dr Revilla- use of vehicles such as ointments, creams, gels, and
Vázquez). Submitted for publication April 11, 2009; revised version “passive” patch technology. More recently, such dos-
accepted July 13, 2009. Address for correspondence: José Juan
age forms have been developed and/or modified to
Escobar-Chávez, PhD, División de Estudios de Posgrado (Tecnología
Farmacéutica), Facultad de Estudios Superiores Cuautitlán-Universidad enhance the driving force of drug diffusion (thermo-
Nacional Autónoma de México, Cuautitlán Izcalli, Estado de México, dynamic activity) and/or increase the permeability of
México 54740; e-mail: josejuanescobarchavez@gmail.com. the skin. Such approaches include the use of penetra-
DOI: 10.1177/0091270009344984 tion enhancers,4 supersaturated systems,5 prodrugs

1262  •  J Clin Pharmacol 2009;49:1262-1283


ELECTROPORATION AS A PHYSICAL ENHANCER FOR SKIN DRUG DELIVERY

or metabolic approach,6,7 liposomes, and other


vesicles.8-11 However, the amount of drug that can be
delivered using these methods is still limited because
the barrier properties of the skin are not fundamen-
tally changed. On the other hand, active methods for
enhancing transdermal drug delivery involve the use
of external energy to act as a driving force and/or act
to reduce the barrier nature of the stratum corneum to
enhance permeation of drug molecules into the skin.12
Recent progress in these technologies has occurred as
a result of advances in precision engineering (bioen-
gineering), computing, chemical engineering, and
material sciences, all of which have helped to achieve
the creation of miniature, powerful devices that can
generate the required clinical response. The use of
active enhancement methods has gained importance
because of the advent of biotechnology in the latter
half of the 20th century, which has led to the genera-
tion of therapeutically active, large molecular weight
(>500 Da) polar and hydrophilic molecules, mostly
peptides and proteins.12 However, gastrointestinal
enzymes often cause degradation of such molecules,
and hence there is a need to demonstrate efficient
Figure 1.  Schematic representation of the skin layers.
delivery of these molecules by alternative administra-
tion routes. Passive methods of skin delivery are
incapable of enhancing permeation of such large sol- that enables the body to interact more intimately
utes, which has led to studies involving the use of with its environment. Essentially, the skin consists
alternative active strategies such as iontophoresis,13,14 of 4 layers: the stratum corneum (SC), which is the
sonophoresis,15 and electroporation.16 outer layer of the skin (nonviable epidermis) and
The scope of this article is, first, to detail the elec- forms the rate-controlling barrier for diffusion for
troporation technique; second, to describe the mecha- almost all compounds. It is composed of dead-
nisms by which electroporation enhances transdermal flattened, keratin-rich cells, the corneocytes. These
transport; and third, to review the increasing elec- dense cells are surrounded by a complex mixture of
troporation enhancement literature to assess the intercellular lipids—namely, ceramides, free fatty
potential for practical transdermal applications in acids, cholesterol, and cholesterol sulfate. Their
drug delivery and noninvasive clinical chemistry. most important feature is that they are structured as
A number of excellent reviews that have been ordered bilayer arrays.23 The predominant diffu-
published contain detailed discussions concerning sional path for a molecule crossing the SC appears to
many aspects of electroporation.17-19 The present be intercellular.24-26 The other layers are the remain-
review shows an updated overview of the use of elec- ing layers of the epidermis (viable epidermis), the
troporation in the pharmaceutical field, specifically dermis, and the subcutaneous tissues (Figure 1).
in the area of topical and transdermal drugs. This There are also several associated appendages: hair
focus is justified because of the magnitude of the follicles, sweat ducts, apocrine glands, and nails.
experimental data available with the use of this tech- In a general context, the skin’s functions may be
nique. The use of electroporation in experimental classified as protective, homeostasis-maintaining
medicine and pharmaceutical sciences has a long functions, or sensing.27,28
history. The importance of the protective and homeostatic
role of the skin is illustrated in one context by its
THE SKIN barrier property. This allows survival in an environ-
ment of variable temperature and water content and
The skin is the largest organ of the body,20-22 account- the presence of environmental dangers, such as
ing for more than 10% of body mass and the one chemicals, bacteria, allergens, fungi, and radiation.

Review 1263
ESCOBAR-CHÁVEZ ET AL

Figure 2.  Processes of percutaneous absorption and transdermal


delivery.

In a second context, the skin is a major organ for


maintaining the body’s homeostasis, especially in
terms of its composition, heat regulation, blood pres-
sure control, and excretory roles.28 Third, the skin is
a major sensory organ in terms of sensing environ- Figure 3.  Diagram representing the in vivo experimental elec-
mental influences, such as heat, pressure, pain, troporation. (A) A sampling chamber glues on the skin surface of
allergens, and microorganism entry. Finally, the skin a rat. Ag/AgCl electrodes are placed in the sampling chamber and
is an organ in a continuous state of regeneration and secured on the skin surface (B, C), and then they are connected to
porator (D).
repair. To perform each of these functions, the
skin must be tough, robust, and flexible, with an
effective communication between each of its intrin-
sic components. the skin to increase molecular permeability.42-46
Many agents are applied to the skin either delib- Figure 3 shows a schematic representation of in vivo
erately or accidentally, with either beneficial or del- animal electroporation. It has the potential to
eterious outcomes. The main interest in dermal enhance macromolecular permeability across skin
absorption assessment is related to (a) local effects in for TDD. As to the method, the voltage applied to
dermatology (eg, corticosteroids for dermatitis), (b) skin and new pathways created in skin have raised
transport through the skin seeking a systemic effect important factors for a drug’s permeability flux.45
(eg, nicotine patches, hormonal drug patches), (c) The drug’s molecule can pass not only through
surface effects (eg, sunscreens, cosmetics, and anti- preexisting aqueous pathways within the lipid
infectives),29 (d) targeting of deeper tissues (eg, non- lamellae-corneocytes region of the SC but also
steroidal anti-inflammatory agents),30-39 and (e) through new aqueous pores created by electropora-
unwanted absorption (eg, solvents in the workplace, tion of multilamellar lipid bilayers separating cor-
pesticides, or allergens).40,41 Figure 2 summarizes the neocytes. The voltage applied to skin plays 2 roles
process of percutaneous absorption. in creating new pathways for drug permeability
The skin became popular as a potential site for through skin and raising the force driving the mole-
systemic drug delivery, on one hand, because of the cule motion to pass skin.
possibility of avoiding the problems of stomach The technique of electroporation is normally used
emptying, pH effects, enzyme deactivation associ- on the unilamellar phospholipid bilayers of cell
ated with gastrointestinal passage, and hepatic first- membranes. However, it has been demonstrated that
pass metabolism and, on the other hand, because of electroporation of skin is feasible, even though the
its capability to enable input control. SC contains multilamellar, intercellular lipid bilay-
ers with few phospholipids and no living cells.47
ELECTROPORATION The electrical behavior of the human epidermal
membrane as a function of the magnitude and dura-
The electroporation method for transdermal drug tion of applied voltage closely parallels the electrical
delivery (TDD) consists of applying high-voltage breakdown: recovery of bilayer membranes seen
pulses to skin, then creating new aqueous pores in during electroporation.48 The approximately 100

1264  •  J Clin Pharmacol 2009;49:1262-1283


ELECTROPORATION AS A PHYSICAL ENHANCER FOR SKIN DRUG DELIVERY

multilamellar bilayers of the SC need about 100-V


pulses for electroporation, or about 1 V per bilayer.49
Although iontophoresis involves the use of relatively
low transdermal voltages (<100 V), electroporation of
skin takes place at high transdermal voltages (~100 V
or more). There is considerable indirect evidence
that high-voltage pulses cause changes in the skin
structure.50,51 The use of electroporation in conjunc-
tion with iontophoresis can expand the scope of
transdermal delivery to larger molecules such as
therapeutic proteins and oligonucleotides. Electropo­
ration acts on the skin with some driving force on the
drug during a pulse.47 It has been shown that applica-
tion of continuous low voltage resulted in a calcein
flux 3 orders of magnitude smaller than pulsing at
high voltage under electrophoretically equivalent
conditions, suggesting that structural changes induced
in the skin by pulsing contribute more significantly
than the direct electrophoretic force acting on the
drug.47 However, iontophoresis will have secondary
effects on the skin, just as electroporation also applies
direct electromotive force on the drug during the brief
pulse period. This may be particularly true for the Figure 4.  Schematic drawing showing skin and subcutaneous
thin cell lining of the sweat ducts, which might tissue being clamped between a pair of plate electrodes. During
be electroporated with low voltages as in iontophore- the pulse application, charges build up across the stratum cor-
sis. Besides the model compounds calcein, sulforho- neum (SC).
damine, and caffeine, other drugs that have been
investigated for transdermal delivery by electropora-
tion include fentanyl,52 metoprolol,53 flurbiprofen,54 voltage drop of an applied electric pulse to the skin
cyclosporin,55 heparin,56 oligonucleotides,57,58 and develops across the SC. This voltage distribution
genes.59 Results with many of these drugs have been favors local electric breakdown (electroporation) of
less dramatic than those seen with model compounds. the SC, which is precisely the barrier targeted to be
Although studies with model compounds have permeabilized to facilitate drug transport.60
given excellent mechanistic insights, the magnitude The application of electroporation to transdermal
of flux enhancement observed may not happen for all delivery is a relatively recent development. If the
drugs; thus, each drug needs to be studied as a sepa- voltage of the applied pulses exceeds a voltage
rate case. threshold at 75 to 100 V (equivalent to the break-
down threshold of 8-10 lipid bilayers in the SC),
MECHANISMS OF ACTION microchannels or “local transport regions” are
created through the breakdown sites of the SC60
Mammalian skin has 2 layers, epidermis and dermis. (Figure 4). Transdermal transport of calcein was
The epidermis is a stratified squamous keratinizing enhanced by electroporation by 2 orders of magni-
epithelium (see Figure 1). The uppermost stratum of tude, compared with that by diffusion and iontopho-
the epidermis is the SC, which consists of about 20 resis.61 Subsequently, many small-molecule drugs
layers of flattened, enucleate, keratin-filled corneo- have been successfully delivered through the skin
cytes surrounded by lamellae of an average of 8 lipid by electroporation. Transport efficiency for small
bilayers.1 The SC forms the major barrier to most (molecular weight ≤1000) charged molecules (such
water-soluble and many hydrophobic drugs and as protoporphyrin IX), using the same polarity
contributes the major portion of the electric resis- pulses, was more than an order of magnitude higher
tance of the skin. The mechanism of electroporation than that for uncharged molecules (such as proto-
depicts the transient electric breakdown site to be porphyrin IX methyl ester) or charged molecules
the most resistive component of the electric circuit. with opposite polarity pulses. The results indicated
In transdermal electroporation, the predominant that, besides passive diffusion through electropores,

Review 1265
ESCOBAR-CHÁVEZ ET AL

electrophoretic force of the pulses also contributes electroporation. Further evidence comes from the similar
to the electroporation-enhanced transport of these anodic and cathodic transport of neutral molecules.67
charged molecules.62 Transport occurred during the
10 to 30 minutes after pulse application was found ADVANTAGES AND DISADVANTAGES OF
to be equal to or higher than that which occurred ELECTROPORATION
during pulses, indicating that passive diffusion
through electropores was also an important trans- The advantage common to all physical methods,
port mechanism.63 Auxiliary current was applied including electroporation (excluding methods using
during the recovery period of the skin to further pro- particle carriers), is that the transport mechanism
mote drug transport by electrophoretic flow.64 Yet does not depend on the uptake functions of the cell;
the transport of uncharged or weakly charged mole- therefore, it can be applied equally well to all cell
cules was also enhanced by a current of opposite types and at all stages of the cell cycle. The process is,
polarity, indicating the action of electroosmotic by itself, biochemically and biologically nontoxic.
force.65 This is especially important for delivering All physical methods, including electroporation,
larger and uncharged molecules. involve transient damage of the cell membrane in
order for the gene and drug to be transported into
Mechanisms of Molecular Transport cells. Therefore, collateral damage resulting in some
cell death is inevitable. This is sometimes referred to
Molecular transport through transiently permeabi- as “toxicity” of the physical treatment. All physical
lized skin by electroporation results from different methods, including electroporation, require instru-
mechanisms at different times. Enhanced diffusion, ments that seem less convenient than simple injec-
during and after pulses, and electrically driven tion of naked DNA or with carrier particles.68-73
transport during pulses (ie, electrophoretic move- In addition to the benefits of avoiding the hepatic
ment and very slight electroosmosis) are the main first-pass effect and higher patient compliance, the
mechanisms of transport. The contribution of elec- additional advantages and disadvantages that the
trophoresis and diffusion depends on the physico- electroporation technique offers can be summarized
chemical properties of the molecule. as follows in Table I.
Electrophoretic Movement
APPLICATIONS OF ELECTROPORATION
During high-voltage pulses, the main driving force
for transport of charged molecules is electropho­ Skin electroporation could be particularly appropri-
resis.47,52,53,66 Evidence for this major contribution of ate for topical delivery for the following reasons. (1)
electrophoresis is the drop in the drug transport with Skin electroporation temporarily permeabilizes the
reverse electrode polarity opposing electrophoresis. barrier to drug permeation and therefore could
broaden topical delivery to drugs not suitable for
Diffusion delivery by passive diffusion (ie, hydrophilic,
Molecular transport through skin highly perm­ charged, and/or large molecular mass drugs).18,51,67,78
eabilized by electroporation is also due to enhanced (2) The application of high-voltage pulses can
passive diffusion. Although much higher skin per- also enhance the permeability of viable cells under-
meability is achieved during the pulse, prolonged lying the SC as demonstrated in vivo by electro-
permeabilization and thereby transport occur after chemotherapy (ie, the electropermeabilization of
pulsing, lasting for hours in the in vitro studies. tumors to bleomycin using high-voltage pulses) or
Evidence for the contribution of enhanced postpulse DNA transfection.79,80
diffusion arises from the increased transport seen The unique and promising release of drugs by
(1) with reverse electrode polarity, (2) with neutral electroporation renders it an attractive candidate as
molecules, and (3) when the drug is added after a physical enhancer to administer drugs throughout
application of the pulses.53,66,67 the skin81-108 and for other uses as gene therapy109-111
and wound healing,112 as well as to develop large,
Electroosmosis permeable regions in the SC113 and to reseal the epi-
In contrast with iontophoresis, the contribution of elec- dermis after electroporation.114,115
troosmosis during high-voltage pulses is low. The short This is emphasized in Table II, which summarizes
time of current application (few seconds) limits the the research on electroporation uses in the transdermal
role of electroosmosis in drug transport by skin administration of drugs.

1266  •  J Clin Pharmacol 2009;49:1262-1283


ELECTROPORATION AS A PHYSICAL ENHANCER FOR SKIN DRUG DELIVERY

Table I  Advantages and Disadvantages of Using Electroporation as a Physical Penetration Enhancer


Advantages Disadvantages

Enhanced drug penetration (of selected drugs) over Cell damage: If the pulses are of the wrong length or
passive transport   intensity, some pores may become too large or fail to
  close after membrane discharge, causing cell damage
  or rupture77
Allows strict control of transdermal penetration rates The transport of material into and out of the cell during
  the time of electropermeability is relatively
  nonspecific77
Versatility: electroporation is effective nearly with all
cells and species types74
Efficiency: a large majority of cells take in the target
DNA or molecule75
Permits rapid termination of drug delivery through
termination of electroporation
The procedure may be performed with intact tissue76
Less anxiety provoking or painful than injection
In many cases, greater patient satisfaction
Not immunologically sensitizing

Analgesic and Anti-Inflammatory Drugs and its prodrugs. The enhancement effect was more
pronounced after applying iontophoresis. The com-
The transdermal transport of cyclodextrin (CD) across bination of 2 electrically assisted methods enhanced
the porcine epidermis by electroporation was studied the delivery of NA; however, no such enhancement
by Murthy et al.81 Electroporation increased the per- was observed for the permeation of NAB and SDN.
meation of h-cyclodextrin (BCD) and hydroxy propyl Their results demonstrated that lipophilicity and
h-cyclodextrin (HPCD) by several orders of magni- molecular size, as well as hydrogel compositions,
tude, relative to passive transport. The presence of had significant effects on skin permeation of NA,
BCD and HPCD enhanced the total transport of the NAB, and SDN via passive diffusion or under the
test permeants piroxicam and carboxyfluorescein electric field.
(CF), respectively, from both permeant solutions and Sung et al83 also assessed the effects of electropora-
suspensions. BCD enhanced the fraction of piroxicam tion on transdermal permeation of NA and its prod-
transported across the epidermis into the receiver rugs. The permeation characteristics were investigated
compartment medium. This was most likely due to under various electrical factors and skin barriers to
the prolonged postpulse permeability state of the epi- elucidate the mechanisms involved in transdermal
dermis. The fraction of CF retained in the epidermis delivery of NA and its prodrugs by skin electropo-
was increased by HPCD. The in vivo delivery of CF by ration. The in vitro permeation studies were per-
electroporation in mice demonstrated the potential of formed using side-by-side diffusion cells. The various
HPCD for sustaining the transdermal absorption rate electrical factors investigated were pulse voltage,
of hydrophilic molecules. pulse duration, and pulse number; the different skin
The aim of the study of Huang et al82 was to assess barriers studied were intact hairless mouse skin,
the effects of iontophoresis and electroporation on SC-stripped skin, delipid skin, and furry Wistar rat
transdermal delivery of nalbuphine (NA) and its 2 skin. Application of electroporation significantly
novel prodrugs, nalbuphine benzoate (NAB) and enhanced transdermal permeation of NA and its
sebacoyl dinalbuphine ester (SDN), from solutions prodrugs. The enhancement ratios were highest for
as well as from hydrogels. Hydroxypropyl cellulose NA, and the 4 prodrugs showed the similar permea-
(HPC) and carboxymethyl cellulose (CMC) were bility after electroporation. The permeation amounts
used in hydrogel formulations to evaluate their of NA and its prodrugs may be increased by applica-
feasibility for delivery of NA and its prodrugs. tion of higher pulse voltage, pulse duration, and
Application of iontophoresis or electroporation sig- pulse number. Various kinetics and mechanisms were
nificantly enhanced the in vitro permeation of NA observed for the permeation of the hydrophilic NA

Review 1267
ESCOBAR-CHÁVEZ ET AL

Table II  Research on Uses of Electroporation to Administer Different Drugs Through the Skin
Analgesic and Anti-Inflammatory Drugs

Research Outcome Author

The transdermal transport of cyclo- Electroporation increased the permeation of h-cyclodextrin Murthy et al81
dextrins (CDs) across porcine epi- (BCD) and hydroxy propyl h-cyclodextrin (HPCD) by sev-
dermis by electroporation eral orders of magnitude, relative to passive transport.
The presence of BCD and HPCD enhanced the total
transport of the test permeants piroxicam and
carboxyfluorescein (CF).
To assess the effects of iontophore- The combination of 2 electrically assisted methods Huang et al82
sis and electroporation on trans- enhanced the delivery of NA; however, no such enhance-
dermal delivery of nalbuphine ment was observed for the permeation of NAB and SDN.
(NA) and its 2 novel prodrugs:
nalbuphine benzoate (NAB) and
sebacoyl dinalbuphine ester
(SDN) from solutions as well as
from hydrogels
Effects of electroporation on trans- The permeation amounts of NA and its prodrugs may be Sung et al83
dermal permeation of NA and its increased by application of higher pulse voltage, pulse
prodrugs duration, and pulse number.
Effect of iontophoresis combined Pulsing of high voltages followed by iontophoresis did not Fang et al84
with treatment of other physical result in increased transport over iontophoresis alone.
enhancement methods such as However, electroporation shortened the onset of transder-
electroporation, low-frequency mal iontophoretic delivery of SNA.
ultrasound, and erbium:YAG
(yttrium-aluminum-garnet) laser
on the transdermal delivery of
sodium nonivamide acetate (SNA)
Antidiuretics
The use of macromolecules as Skin electroporation increased transdermal mannitol deliv- Vanbever et al85
chemical enhancers of mannitol ery by approximately 2 orders of magnitude. The addi-
transdermal transport by skin tion of macromolecules further increased transport up to
electroporation 5-fold.

Antiviral drugs
Transdermal delivery of interferon In vivo delivery of IFNα2b was demonstrated through Badkar et al86
alpha-2b (IFNα2b) in hairless rats micropores created in the outer layer of the skin.
through aqueous microchannels Iontophoresis enhanced delivery through microporated
created in the skin and enhanced skin in hairless rats.
by iontophoresis
Examination of parameters influ- It was found that voltage, pulse length (t), and number of Sharma et al87
encing electroporative transder- pulses were the 3 most important parameters influencing
mal delivery of terazosin transdermal delivery of THCl.
hydrochloride (THCl) to hairless
rat skin
To understand the mechanism of Using shorter pulses and large-area electrodes is a safer Sharma et al88
terazosin delivery as a prelimi- technique than large pulses and small-area electrodes
nary indicator of safety when electroporation is used to enhance skin’s permea-
bility for terazosin hydrochloride.
(continued)

1268  •  J Clin Pharmacol 2009;49:1262-1283


ELECTROPORATION AS A PHYSICAL ENHANCER FOR SKIN DRUG DELIVERY

Table II  (continued)


Analgesic and Anti-Inflammatory Drugs

Research Outcome Author

Beta-blocker agents
The effect of electroporation on ion- The iontophoretic transport of timolol was decreased by Denet et al89
tophoretic transport of 2 beta- electroporation because the high accumulation of the
blockers, timolol and atenolol lipophilic cation timolol in the stratum corneum (SC)
resulted in a decrease of electroosmosis. In contrast,
electroosmosis was not affected by atenolol, and the
iontophoretic transport of atenolol was increased by
electroporation.
Anticancer drugs
The influence of iontophoresis and A combination of electroporation pretreatment and subse- Fang et al90
other physical enhancement quent iontophoresis resulted in a higher permeation of
methods such as electroporation 5-FU than either technique alone.
and erbium:YAG laser on the skin
permeation of 5 fluorouracil
(5-FU)
To present an overview of electro- Electrochemotherapy is a new, clinically acknowledged Sersa et al91
chemotherapy, via cell membrane method for the treatment of cutaneous and subcutaneous
permeabilizing electric pulses tumors.
Insulin
Study of transdermal transport of In vivo noninvasive insulin delivery to therapeutic levels Murthy et al92
insulin and extraction of intersti- and glucose extraction may be achieved by combining
tial glucose under anodal ionto- electroporation with anionic lipids and electroosmosis.
phoresis following electroporation
in the presence of 1,2 dimyris-
toylphophatidylserine (DMPS)
Evaluation of the synergistic effect Percutaneous absorption of insulin is synergistically Tokumoto
of electroporation (EP) and ionto- enhanced by a combined use of EP and IP. et al93
phoresis (IP) on the in vivo percu-
taneous absorption of human
insulin in rats
Hormones
Investigation of electronically facili- The flux of hPTH (1-34) with the electroporation pulses of Medi and
tated transdermal delivery of 100 and 300 V, followed by iontophoresis at 0.2 mA/cm2, Singh94
human parathyroid hormone was 10- and 5-fold higher, respectively.
(1-34), hPTH (1-34)
In vitro electrically assisted delivery A combination of electroporation and iontophoresis Chang et al95
by iontophoresis and/or elec- resulted in higher transdermal permeation than either
troporation of calcium-regulating one technique alone.
hormones, salmon calcitonin
(sCT) and parathyroid hormone
(1-34) (PTH), through human
epidermis
Transdermal penetration of estradiol Combination of electroporation and iontophoresis did not Essa et al96
through human epidermis from markedly improve estradiol penetration for ultradeform-
phosphatidylcholine (PC)–based able vesicles.
liposomes and saturated aqueous
estradiol solution
(continued)

Review 1269
ESCOBAR-CHÁVEZ ET AL

Table II  (continued)


Analgesic and Anti-Inflammatory Drugs
Research Outcome Author

Photosensitizers
Electric pulses to increase transder- A greater than 2-fold enhancement of PpIX production Johnson et al97
mal transport of d-aminolevulinic with electroporative delivery was seen versus that
acid (ALA), a precursor to the obtained with passive delivery.
photosensitizer protoporphyrin IX
(PpIX)
Optimize and enhance the in vitro Application of iontophoresis or electroporation alone also Fang et al98
skin permeation of 5-ALA by 2 increased the ALA permeation by approximately 15-fold
resurfacing techniques: and 2-fold, respectively. The incorporation of iontophore-
erbium:YAG laser and micro- sis or electroporation with the resurfacing techniques
dermabrasion caused a profound synergistic effect on ALA permeation.
Oligonucleotids
Topical delivery in the skin of 39 Electroporation increased the topical delivery of the 39 Regnier et al99
end-modified phosphodiester end-modified phosphodiesters by 2 orders of magnitude
oligonucleotides using compared to passive diffusion.
electroporation
A needle-free method based on The efficacy of peptide delivery was comparable to that of Zhao et al100
transcutaneous electroporation is intradermal injection with Freund’s complete adjuvant.
described for delivering peptide
vaccines
Adaptation of the electroporation The pro-photosensitizer drug delta-amino levulinic acid Hui101
and electrofusion technology in 2 and the anticancer drug methotrexate have been deliv-
fronts of cancer research and ered transcutaneously by electroporation. These studies
treatment hold promise for the treatment of cancers in humans.
Tea polyphenols
To assess the effects of electropora- A synergistic effect was detected for (+)-catechin but not Fang et al102
tion, iontophoresis, and their for (–)-epicatechin after application of electroporation
combination on the transdermal followed by iontophoresis.
delivery of tea catechins across
porcine skin

Stimulants
Investigate the effect of electrotreat- Electrotreatment increased the amount of caffeine and Marrat et al103
ment on skin permeability by NAP in the skin. Enhancement factors (EFs) for NAP of
measuring the cumulative deliv- 7.2 and 14.9 were observed following 20 minutes of
ery of caffeine and sodium ascor- electrotreatment.
byl phosphate (NAP)
Dextrans
To test factors affecting the The use of proper lipid enhancers greatly extends the Sen et al104
potency of anionic lipid transport upper size limit of transportable chemicals.
enhancers
The substantial synergic effects of Little release of calcein was observed from SC lipid with- Tokudome and
CaCl2 and EP on in vitro skin per- out EP, whereas higher releases were observed after EP Sugibayashi105
meation of calcein and fluorescein with or without NaCl or CaCl2.
isothiocyanate (FITC) dextrans
(continued)

1270  •  J Clin Pharmacol 2009;49:1262-1283


ELECTROPORATION AS A PHYSICAL ENHANCER FOR SKIN DRUG DELIVERY

Table II  (continued)


Analgesic and Anti-Inflammatory Drugs
Research Outcome Author

To study the influence of sodium It appears that the presence of SDS during electroporation Murthy et al106
dodecyl sulfate (SDS) on transder- helps in achieving the desired transport with less
mal transport of diffusants by electrical exposure dose.
electroporation
Folic acid antagonists
Application of electroporation to Electroporation of MTX with an anion lipid enhancer Wong et al107
increase the transdermal transport under a mild hyperthermic environment provided a
of methotrexate (MTX) significant transdermal delivery.
A microelectrode array to minimize In vivo transdermal electroporation using a microelectrode Wong et al108
the pain sensation of electropora- array with 180 pulses of 150 V, 0.2 ms at 1 Hz, followed
tion for enhancing transdermal by a 30-minute methotrexate occlusion increased more
drug delivery of methotrexate than 4-fold the systemic methotrexate level in mice.
Other uses
Gene transfer
Optimization of electroporation The highest transgene expression and efficiency of Heller et al109
parameters for transgene expres- individual cell transformation with minimal damage was
sion with minimal tissue damage produced with eight 150-ms pulses at a field strength of
with a novel electrode 100 V/cm.
Local injection of a plasmid before Electrotransfer is a promising alternative for drug and gene Préat110
electroporation delivery.
Optimization of parameters to With the optimized conditions, gene transfer mediated by Chesnoy and
obtain reproducible, high-level intradermal injection of naked DNA was comparable in Huang111
gene transfer to the mouse skin efficiency to electroporation.
Wound healing
Application of plasmid DNA Electroporation-assisted transfection with DNA plasmid Ferguson et al112
expression vectors directly into expression vectors for growth factors will be an effective
the wound with electroporation modality for enhancing cutaneous wound healing.
To develop large permeable regions in the SC
A model of pore formation between The model predicts that the initial radius of the first Pliquett and
adjacent corneocytes aqueous pathway is approximately 5 nm for a transdermal Weaver113
voltage of 60 V at room temperature.
To reseal epidermis after electroporation
The resealing of porcine epidermis Both poloxamer 188 and phosphatidylcholines were Burgess et al114
after electroporation effective in resealing in terms of electric conductance
and transport.
Application of high-voltage pulsing Electroporation caused large molecular transport for all Chen et al115
or iontophoresis to human, hair- 3 types of skin.
less rat, or black rat snake skin

and lipophilic nalbuphine enanthate through differ- The effect of iontophoresis combined with treat-
ent skin barriers by applying electroporation. This ment of other physical enhancement methods such
study demonstrated that electroporation may enhance as electroporation, low-frequency ultrasound, and
and control transdermal permeation of NA and its erbium:YAG (yttrium-aluminum-garnet) laser on the
prodrugs. transdermal delivery of sodium nonivamide acetate

Review 1271
ESCOBAR-CHÁVEZ ET AL

(SNA) was examined by Fang et al.84 Iontophoresis macromolecules can be used as transdermal transport
increased the transdermal flux of SNA in vitro as enhancers uniquely suited to skin electroporation.
compared to the passive diffusion without any
enhancement. Furthermore, iontophoresis was always Antiviral Drugs
the most potent enhancement method for SNA per-
meation among the physical enhancement methods Badkar et al86 demonstrated transdermal delivery of
tested. Pulsing of high voltages (electroporation) interferon alpha-2b (IFNα2b) in hairless rats through
followed by iontophoresis did not result in increased aqueous microchannels (micropores) created in the
transport over iontophoresis alone. However, elec- skin and enhanced by iontophoresis.
troporation shortened the onset of transdermal The Altea Therapeutics PassPort System was con-
iontophoretic delivery of SNA. Pretreatment of low- figured to form an array of micropores (2.0 cm2; 72
frequency ultrasound alone on skin did not increase micropores/cm2) on the rat abdomen. The transder-
the skin permeation of SNA. The combination of ion- mal patch (Iomed TransQ1-GS-hydrogel) was satu-
tophoresis and sonophoresis increased transdermal rated with an IFNα2b solution and applied. Delivery
SNA transport more than each method by itself. The was evaluated with and without cathodic iontophore-
enhancement of drug transport across shunt routes sis (0.1 mA/cm2). Intravenous delivery was performed
and reduction of the threshold voltage in the presence to support pharmacokinetic calculations. IFNα2b was
of an electric field may contribute to this synergistic not delivered through intact skin by itself (passive
effect. Use of an erbium:YAG laser was a good method delivery) or during iontophoresis. However, passive
for enhancing transdermal absorption of SNA because delivery through micropores was achieved in vivo in
it allows precise control of SC removal, and this abla- rats. A dose of 397 ng was delivered over 6 hours,
tion of SC could be reversible to the original normal with steady-state serum concentrations reaching
status. The combination of laser treatment and ionto- a plateau at 1 hour postpatch application. These
phoresis also synergized the skin permeation of SNA, levels dropped rapidly after patch removal and
possibly due to a gradual drop in the electric resis- returned to baseline within 2 hours of patch removal.
tance of the skin. The results in this present study Iontophoresis-enhanced delivery through micropores
point out that the choice of certain conditions with resulted in a 2-fold increase in the dose delivered in
suitable physical enhancement methods can induce a the hairless rat. In vivo delivery of IFNα2b was dem-
synergistic effect on transdermal delivery of SNA onstrated through micropores created in the outer
during iontophoresis. layer of the skin. Iontophoresis enhanced delivery
through microporated skin in hairless rats.
Antidiuretic Drugs The use of electroporation pulses as a physical
means of enhancing the permeability of skin to
Macromolecules were investigated as chemical deliver drugs is in the early stages of development. In
enhancers of transdermal transport by skin electropo- this article, a systematic study examining the para­
ration for Vanbever et al.85 Although unable to enhance meters influencing electroporative transdermal deliv-
passive or iontophoretic transport, macromolecules ery of terazosin hydrochloride to hairless rat skin is
are proposed to enhance electroporation-assisted reported by Sharma et al.87 It was found that voltage,
delivery by stabilizing the increased permeability pulse length (t), and number of pulses were the 3
caused by high-voltage pulses. Skin electroporation most important parameters, in that order. For creating
increased transdermal mannitol delivery by approxi- a significant enhancement in drug delivery to the
mately 2 orders of magnitude. The addition of macro- skin, without causing any apparent change in its
molecules further increased transport up to 5-fold. external appearance, it was necessary to deliver 5 or
Macromolecules present during pulsing enhanced more exponentially decaying electroporation pulses,
mannitol transport after pulsing for hours, apparently at 88 ± 2.5 V (voltage across the skin), with a decay
by a macromolecule-skin interaction. No enhance- time constant of 20 ms. Electrodes with larger area
ment was observed during passive diffusion or low- could attain the same voltages across the skin with a
voltage iontophoresis, suggesting that macromolecules much lower applied voltage and possessed other
interact specifically with transport pathways created advantages with regard to performance of the drug
at high voltage. Although all macromolecules studied delivery system.
enhanced transport, those with greater charge and size In another article, Sharma et al88 described a study
were more effective. This study demonstrates that in which the reversibility of the electroporation

1272  •  J Clin Pharmacol 2009;49:1262-1283


ELECTROPORATION AS A PHYSICAL ENHANCER FOR SKIN DRUG DELIVERY

technique is evaluated with in vitro methods. The 5-fluorouracil (5-FU) was examined by Fang et al.90
skin’s reversal from an enhanced permeation mode as Iontophoresis increased the in vitro transdermal
a result of electroporation to the base level was used as transport of both the anionic and nonionic forms of
an index to understand the mechanism of drug deliv- 5-FU. A combination of electroporation pretreat-
ery and also as a preliminary indicator of safety. ment and subsequent iontophoresis resulted in a
Maximum delivery of the model drug, terazosin hydro- higher permeation of 5-FU than either technique
chloride, occurred during the pulsing. Electroporative alone. It appeared that electroporation treatment
delivery with a wire electrode (small-area electrode, exerted a disruptive influence on the SC. The SC
0.56 cm2) using 20 pulses at 88 V and pulse length of layers in the skin were partly ablated by the laser,
20 ms did not cause any damage to the skin. Increasing resulting in a great enhancement effect on the skin
the pulse length to 60 ms, while keeping the rest of the permeation of 5-FU. Application of iontophoresis
parameters fixed, caused a visible change in the exter- further increased the drug permeation across laser-
nal appearance of the skin. However, with the use of a pretreated skin. The laser was consistently the most
spiral electrode (large-area electrode, 2.74 cm2) at a potent technique to enhance 5-FU delivery among
60-ms pulse length, there was minimal damage to the the physical enhancement methods examined in
skin. This may be attributed to the more uniform flow this study.
of current over the whole skin area. Electrochemotherapy is a local drug delivery
These findings indicate that using shorter pulses approach aimed at treatment with palliative intent
and large-area electrodes is a safer technique than of cutaneous and subcutaneous tumor nodules of
large pulses and small-area electrodes when elec- different histologies. Electrochemotherapy, via cell
troporation is used to enhance skin’s permeability membrane permeabilizing electric pulses, poten-
for drug delivery. tiates the cytotoxicity of nonpermeant or poorly
permeant anticancer drugs with high intrinsic cyto-
Beta-Blocker Agents toxicity, such as bleomycin or cisplatin, at the site of
electric pulse application. An overview of preclini-
Denet et al89 studied the effect of electroporation on cal and clinical studies was presented, and the treat-
iontophoretic transport of 2 beta-blockers, timolol ment procedure was further critically evaluated by
(lipophilic) and atenolol (hydrophilic), to have a bet- Sersa et al.91 In clinical studies, electrochemotherapy
ter understanding of the mechanism of combination. has proved to be a highly efficient and safe approach
The transdermal delivery of these beta-blockers for treating cutaneous and subcutaneous tumor nod-
through human SC was studied in 3-compartment ules. The treatment response for various tumors
diffusion cells. The transport of mannitol was evalu- (predominantly melanoma) was approximately 75%
ated to assess the electroosmotic flow. The ionto­ complete and 10% partial response of the treated
phoretic transport of timolol was decreased by nodules. Electrochemotherapy is a new, clinically
electroporation because the high accumulation of acknowledged method for the treatment of cutane-
the lipophilic cation timolol in the SC resulted in a ous and subcutaneous tumors. Its advantages are
decrease of electroosmosis. In contrast, electroosmo- high effectiveness on tumors with different histolo-
sis was not affected by atenolol, and the iontophoretic gies, simple application, minimal side effects, and
transport of atenolol was increased by electropora- the possibility of effective repetitive treatment.
tion. Using 2 different beta-blockers, the authors
showed that lipophilicity and positive charges affect Insulin
the electrotransport of drugs. Understanding the effect
of the physicochemical properties of the drug, as well Transdermal transport of insulin and extraction of
as the electrical parameters, is thus essential for the interstitial glucose under anodal iontophoresis (elec-
optimization of transdermal drug delivery by a com- troosmosis) following electroporation in the presence
bination of electroporation and iontophoresis. of 1,2-dimyristoylphophatidylserine (DMPS) was
studied by Murthy et al.92 An earlier study showed
Anticancer Drugs that DMPS increased the transport of insulin across
porcine epidermis under electroporation by ~4-fold.
The influence of iontophoresis and other physical It was suggested that DMPS increased the lifetime
enhancement methods such as electroporation of electropores in the epidermis, resulting in an
and erbium:YAG laser on the skin permeation of enhanced transport of permeants. When electro­osmosis

Review 1273
ESCOBAR-CHÁVEZ ET AL

was applied across the epidermis follow­ing elec- Hormones


troporation with DMPS, the enhancement of
insulin transport was ~18-fold over electroporation Electronically facilitated transdermal delivery of
alone. When the same strategy was applied to extract human parathyroid hormone (1-34), hPTH (1-34),
interstitial glucose, the enhancement was ~23-fold was investigated in vitro by Medi and Singh94 using
over electroporation alone. Real-time transdermal dermatomed porcine skin. The effect of iontopho-
insulin transport kinetics was measured using fluo- retic current density, electroporative pulse voltages,
rescein isothiocyanate (FITC)–labeled insulin and a and also electroporation followed by iontophoresis
custom-made vertical diffusion apparatus that had a was investigated on the in vitro percutaneous absorp-
fluorescence cuvette as the receiver compartment. tion of hPTH (1-34). Iontophoresis at 0.5 mA/cm2
Insulin transport by electroporation alone showed a current density significantly enhanced the flux
nonlinear kinetics that is most likely due to the of hPTH (1-34) in comparison to passive flux.
resealing of the electropores with time. The transport Electroporation pulses of 100, 200, and 300 V signi­
kinetics was more linear when electroporation was ficantly increased the flux of hPTH (1-34) in com-
carried out in the presence of DMPS, confirming ear- parison with the passive as well as iontophoretic
lier studies that suggested that DMPS stabilizes trans- flux at 0.5 mA/cm2. The electroporative flux of
port paths formed by electroporation. The data suggest hPTH (1-34) was found to vary linearly with the
that in vivo, noninvasive insulin delivery to thera- pulse amplitude. The principal barrier of the skin,
peutic levels and glucose extraction may be achieved SC, was found perturbed following the pulses, as
by combining electroporation with anionic lipids and evident by light microscopy studies. The application
electroosmosis. of electroporation pulses followed by iontophoresis
Tokumoto et al93 evaluated the synergistic effect of further increased the flux by several fold. The flux of
electroporation (EP) and iontophoresis (IP) on the in hPTH (1-34) with the electroporation pulses of 100
vivo percutaneous absorption of human insulin in and 300 V followed by iontophoresis at 0.2 mA/cm2
rats. Passive diffusion and IP alone (0.4 mA/cm2) was 10- and 5-fold higher, respectively, in compari-
resulted in almost no skin permeation of insulin at son to the flux with corresponding pulses alone.
pH 7, whereas EP treatment (150 or 300 V, 10 ms, and This shows the synergistic effect of iontophoresis in
10 pulses) resulted in a high plasma level of insulin, combination with electroporation on skin permea-
and the combined use of EP and IP led to a further bility of hPTH (1-34). The results indicate the pos-
increase of the plasma level of insulin compared sibility of designing controlled transdermal delivery
with that measured after EP alone. Interestingly, a systems for hPTH (1-34) using electroporation fol-
much higher plasma level was observed when the pH lowed by iontophoresis.
of the insulin solution at 7 was increased to 10. The Electrically assisted delivery by iontophoresis
nonassociation ratio of insulin was significantly and/or electroporation was used in vitro to deliver
higher at pH 10 than at pH 7. Insulin monomers and the calcium-regulating hormones, salmon calcitonin
dimers were observed in addition to the normal form (sCT) and parathyroid hormone (1-34) (PTH), through
of insulin, hexamer, albeit in low percentages, at pH human epidermis by Chang et al.95 Such delivery
10, whereas most of the insulin was in the hexamer could be useful for the chronic treatment of post-
form at pH 7. To confirm the influence of the aggrega- menopausal osteoporosis and other clinical indica-
tion properties of insulin, the commercially available tions as a superior alternative to parenteral delivery.
human insulin analog insulin lispro was then evalu- The sCT (50 mg/mL) or PTH (1-34) (100 mg/mL)
ated. Its skin permeation was found to be extremely formulation was prepared in citrate buffer (pH 4.0
high compared to that of conventional human insu- or 5.0, respectively). Epidermis separated from
lin without increasing the solution pH. Marked human cadaver skin was used. Iontophoresis was
decreases in blood glucose levels reflecting the applied using a constant current power source
increases in the plasma concentration of insulin and electroporation with an exponential pulse gen-
were also observed after EP/IP treatment. The pres- erator. A combination of electroporation and ionto-
ent study suggests that percutaneous absorption of phoresis resulted in higher transdermal permeation
insulin is synergistically enhanced by a combined than either one technique alone. Electroporation
use of EP and IP and that altering the aggregation also shortened the lag time of iontophoretic trans-
properties of insulin is important to enhance the per- dermal delivery of salmon calcitonin. Pulsing at
cutaneous absorption of insulin by IP and/or EP. lower voltages followed by iontophoresis did not

1274  •  J Clin Pharmacol 2009;49:1262-1283


ELECTROPORATION AS A PHYSICAL ENHANCER FOR SKIN DRUG DELIVERY

result in increased transport (over iontophoresis 10-mm cross sections were examined autoradio-
alone), perhaps because the transdermal voltage was graphically. As the electrical dose increased, there
very low. The transdermal transport of salmon calci- was an increase in penetration depth. In vivo deliv-
tonin by pulsing with 15 pulses (1 ppm) of 500 V ery was assayed by measuring the fluorescence of
(200 ms) followed by iontophoresis led to a quick PpIX in skin samples. A greater than 2-fold enhance-
input and high flux. The average transdermal volt- ment of PpIX production with electroporative deliv-
age was only about 50 V for a 500-V study. ery was seen versus that obtained with passive
Essa et al96 investigated transdermal penetration delivery. Superimposition of a DC potential resulted
of a model lipophilic drug (estradiol) through human in a nearly 3-fold enhancement of PpIX production
epidermis from phosphatidylcholine (PC)–based versus passive delivery. Levels were higher than the
liposomes and saturated aqueous estradiol solu- sum of PpIX detected after pulse-alone and DC-alone
tion (control). Representative examples of cholate- delivery. Electroporation and electrophoresis are
containing ultradeformable (Transfersomes), nonrigid likely factors in electrically enhanced delivery.
(pure PC), and membrane-stabilized (cholesterol- 5-Aminolaevulinic acid (ALA) is used as a pro-
containing) vesicles were used. Transdermal pene- toporphyrin IX-precursor for the photodynamic
tration studies involved occluded passive penetration therapy of superficial skin cancer and cutaneous
for 12 hours and cathodic iontophoresis (0.8 mA/ metastases of internal malignancies. However, the
cm2) for 8 hours for all systems. Combined elec- permeability of hydrophilic ALA across the skin is
troporation (5 pulses, 100 V, 100 ms, 1-minute spac- very low. The objective of Fang et al98 was to opti-
ing) and iontophoresis (0.8 mA/cm2, for 2 hours) mize and enhance the in vitro skin permeation of
was also employed for ultradeformable vesicles and ALA by 2 resurfacing techniques: erbium:YAG
control. Estradiol penetration parameters from dif- laser and microdermabrasion. Light microscopic
ferent formulations were compared. All vesicles had changes in pig skin caused by these techniques
essentially the same particle size, with ultradeform- were also compared. The electrically assisted
able liposomes showing the highest negative zeta methods, iontophoresis and electroporation, were
potential (–29 mV). Occluded passive penetration also used to facilitate ALA permeation across laser-
improved estradiol skin penetration from liposomes or microdermabrasion-treated skin. Among the
relative to control. Iontophoretic studies revealed modalities tested in this study, the erbium:YAG laser
the superiority of ultradeformable vesicles regarding showed the greatest enhancement of ALA permeation.
drug skin penetration and deposition compared to The laser fluence was found to play an important
traditional liposomes. Combination of electropora- role in controlling the drug flux, producing enhance-
tion and iontophoresis did not markedly improve ment ratios from 4-fold to 246-fold relative to the
estradiol penetration for ultradeformable vesicles. control. The skin permeation of ALA across micro-
The combination results implied repair of the skin dermabrasion-treated skin was approximately 5- to
barrier due to the penetration-retarding effect of PC 15-fold higher than that across intact skin. Both the
monomers released from liposomes. ablated effect of the SC and ALA flux was proportio­
nal to the treatment duration of microdermabrasion.
Photosensitizers The application of iontophoresis or electroporation
alone also increased the ALA permeation by approx-
Selectivity of photodynamic therapy can be improved imately 15-fold and 2-fold, respectively. The incor-
with localized photosensitizer delivery, but topical poration of iontophoresis or electroporation with the
administration is restricted by poor diffusion across resurfacing techniques caused a profound synergis-
the SC. Johnson et al97 used electric pulses to tic effect on ALA permeation. This basic study has
increase transdermal transport of d-aminolevulinic encouraged the further investigation of ALA perme-
acid (ALA), a precursor to the photosensitizer proto- ation by laser or microdermabrasion.
porphyrin IX (PpIX). ALA-filled electrodes were
attached to the surface of excised porcine skin or the Oligonucleotids
dorsal surface of mice. Pulses were administered
and, in some in vivo cases, a continuous DC poten- The feasibility of topical delivery in the skin of 39
tial (6 V) was concomitantly applied. For in vitro end-modified phosphodiester oligonucleotides using
14C ALA penetration, 10-mm layers parallel to the electroporation was investigated by Regnier et al.99
SC were assayed by liquid scintillation analysis, and Experiments were performed in vitro, using hairless

Review 1275
ESCOBAR-CHÁVEZ ET AL

rat skin. Five pulses of (200 V, 450 ms) were applied. natural killer (NK) cells, peripheral blood stem cells,
The 39 end modifications of the 15-mer oligonucle- and bone marrow–derived dendritic cells. Hybrids
otide were (1) 39-aminohexyl; (2) biotin, with a trieth- of tumor cells and bone marrow–derived dendritic
yleneglycol arm; (3) methylphosphonate links cells have been formed by electrofusion
between nucleotides 13, 14, and 15; and (4) 2-O- for the purpose of tumor vaccines. The second front
methyl nucleotides at 13, 14, and 15 positions. All the was the use of transcutaneous electroporation to
modifications were efficient to protect the oligonucle- deliver anticancer drugs and vaccines across the
otides against degradation in the skin. Electroporation skin. Methods to extend the upper molecular weight
increased the topical delivery of the 39 end-modified limit of transcutaneous electroporation have been
phosphodiesters by 2 orders of magnitude compared developed. The pro-photosensitizer drug delta-amino
to passive diffusion, without significant differences levulinic acid, the anticancer drug methotrexate,
between the derivatives. Oligonucleotide concentra- and peptide vaccines designed for cancer prevention
tions in the range of 1 mm could be achieved in the and immunotherapy have been delivered transcuta-
viable skin. The delivery of a phosphorothioate con- neously by electroporation. These studies hold
gener was lower than phosphodiester delivery due to promise for the treatment of cancers in humans.
the interaction of phosphorothioate with the SC.
Consequently, 39 end-protected phosphodiesters Tea Polyphenols
could be an interesting alternative to phosphorothio-
ate oligonucleotides for topical treatment of cutane- Tea polyphenols, including (+)-catechin, (–)-epicat-
ous diseases. echin, and (–)-epigallocatechin-3-gallate (EGCG),
A needle-free method based on transcutaneous have been shown to possess potent antioxidant and
electroporation is described for delivering peptide chemopreventive activities. The aim of the study by
vaccines by Zhao et al.100 The Kb-binding OVA- Fang et al102 was to assess the effects of electropora-
peptide SIINFEKL was used as an example to induce tion, iontophoresis, and their combination on the
the peptide-specific cytotoxic T lymphocyte (CTL) transdermal delivery of tea catechins across porcine
response in mice. A saturated anionic lipid was skin. The permeation characteristics were investi-
added during electroporation, and postpulse elec- gated using various analogs of catechins, pH values,
troosmosis was applied to enhance the vaccine deliv- and modes of electroporation and iontophoresis.
ery. Electroporation was found to stimulate the exodus The mechanisms by which these catechins were
of Langerhans cells (LC) from the skin. The peptide transported via the skin were elucidated by examin-
transported into and through murine skin was mea- ing the electric conductivity, transepidermal water
sured using a Franz diffusion apparatus. Most peptide loss (TEWL), and fusion of SC lipid liposomes
was retained in the skin rather than passing through (SCLL). The isomers, (+)-catechin and (–)-epicate-
the skin in the process. The peptide was delivered to chin, showed different behaviors of skin permeation
the dorsal skin of mice by in vivo electroporation. An and local skin deposition with the electrically
electroporation-transportable oligonucleotide with a assisted methods. The results suggest evidence of
CpG motif was used as adjuvant. The efficacy of pep- selective skin absorption of (–)-epicatechin over
tide delivery was comparable to intradermal injection (+)-catechin. A synergistic effect was detected for
with Freund’s complete adjuvant. Peptide-specific (+)-catechin but not for (–)-epicatechin after applica-
CTL response to the vaccine delivered by needle-free tion of electroporation followed by iontophoresis.
electroporation/electroosmosis was equivalent to that The presence of a gallic acid ester in the structure of
delivered by intradermal injection, as determined by EGCG significantly increased the skin uptake of cat-
production of the peptide-specific interferon gamma echins. However, a negligible amount of or no EGCG
(IFN-γ) in the ELISPOT assay. molecules permeated across the skin. The mecha-
Electroporation and the associated phenomenon nisms involved in the enhancement of electropora-
of electrofusion have been widely adapted as tools to tion may be the skin reservoir effect and an increase
a broad range of biomedical research and therapy. In in skin permeability. The TEWL profiles suggest that
their study, Hui101 summarized the adaptation of the in addition to the force of electrorepulsion, the skin
electroporation and electrofusion technology in 2 hydration effect and structural alterations may also
fronts of cancer research and treatment. The first have contributed to the enhancement by iontophore-
was genetic manipulation of hematopoietic cells for sis. Electroporation did not influence the skin bar-
the purpose of cancer treatment. High-efficiency rier function, although the skin permeability
transfection methods have been developed to transfect increased according to the SCLL fusion study.

1276  •  J Clin Pharmacol 2009;49:1262-1283


ELECTROPORATION AS A PHYSICAL ENHANCER FOR SKIN DRUG DELIVERY

Stimulants microscopy, that transport of water-soluble mole-


cules was localized in local transport spots or regions
Electrically assisted transport can increase the rate (LTRs) created by the electroporation pulses. Anionic
and extent of delivery; moreover, it also enables the phospholipids, especially DMPS, were located at the
administration of polar and charged molecules into center of the LTRs and spanned the entire thickness
the skin. The objective of Marrat et al103 was to inves- of the SC. They deduced that, after being driven into
tigate the effect of electrotreatment on skin permeabil- the epidermis by negative electric pulses, saturated
ity by measuring the cumulative delivery of caffeine anionic phospholipids mix and are retained better
and sodium ascorbyl phosphate. Furthermore, confo- by the SC lipids. Anionic lipids prefer loose layers
cal microscopy was used to visualize the effect of or vesicular rather than multilamellar forms, thereby
electrotreatment on the penetration of calcein. Porcine prolonging the structural recovery of SC lipids to the
ear skin was used for the in vitro permeation studies, native multilamellar form. In the presence of 1 mg/
which involved application of either the caffeine or mL DMPS in the transport milieu, the flux of FITC-
sodium ascorbyl phosphate (NAP)–containing gels Dextran-4k was enhanced by 80-fold and reached
using the roll-on supplied with the electrotreatment 175 µg/cm2/min. Thus, the use of proper lipid
device. Electrotreatment increased the amount of caf- enhancers greatly extends the upper size limit of
feine and NAP in the skin. Enhancement factors (EFs) transportable chemicals. Understanding the mecha-
for NAP of 7.2 and 14.9 were observed following 20 nism of lipid enhancers enables one to rationally
minutes of electrotreatment and either immediate design better enhancers for transdermal drug and
sampling or a further 60 minutes of passive diffusion vaccine delivery by electroporation.
compared with passive diffusion for either 20 or 80 After reporting the substantial synergic effects of
minutes. The effect on caffeine permeation was less CaCl2 and EP on in vitro skin permeation of calcein
significant (EF = 2.1 for a 20-minute electrotreatment and FITC dextrans, Tokudome and Sugibayashi105
compared with passive diffusion for 20 minutes). The investigated the mechanisms for these effects by
confocal microscopy images showed that electrotreat- considering changes in lamellar structure and bar-
ment significantly increased calcein permeation; rier recovery time of the biggest skin barrier, the SC,
fluorescence was observed deep into the viable by this combined treatment. The change in skin
epidermis-reaching depths of up to 60 to 80 microns. lamellar structure was evaluated by lipid mobility in
They have shown that electrotreatment increases skin the SC using attenuated total reflection Fourier
permeability and the cumulative delivery of cosmeti- transform infrared (ATR-FTIR), calcein release from
cals into the skin. SCLL, in vitro skin permeation of calcein, and
TEWL.
Dextrans The C-H stretching band of skin lipids produced
with EP was blue-shifted when compared to that with-
Sen et al104 found that anionic phospholipids, but out EP. Asymmetric C-H stretching was highest with
not cationic or neutral phospholipids, enhance the EP in CaCl2 solution. Little release of calcein was
transdermal transport of molecules by electropora- observed from SCL without EP, whereas higher
tion. DMPS, phosphatidylserine from bovine brain releases were observed after EP with or without
(brain-PS), dioleoylphosphatidylserine (DOPS), and NaCl or CaCl2. In particular, high calcein release
dioleoylphosphatidylglycerol (DOPG) were used to (>20%) was observed over 60 minutes with EP in
test factors affecting the potency of anionic lipid CaCl2 solution. The in vitro permeation study of cal-
transport enhancers. DMPS with saturated acyl cein was conducted through excised hairless rat skin
chains was found to be a much more potent trans- that was pretreated with EP before skin excision.
port enhancer than those with unsaturated acyl Permeation rate was highest in skin excised immedi-
chains (DOPS and DOPG). Saturated DMPS was also ately after in vivo EP, and this rate decreased with
more effective in delaying resistance recovery after time after EP treatment. TEWL recovered to control
pulsing and had a greater affinity in the epidermis levels within 2 hours after EP in distilled water or
after pulsing. Using fluorescent carboxyl fluorescein NaCl solution, whereas high TEWL was maintained
and FITC-labeled dextrans as test water-soluble mol- after EP in CaCl2 solution. These results suggest
ecules for transport, as well as rhodamine-labeled that at least lamellar destruction of SC must be
phospholipids to track anionic phospholipids, they related to the enhanced skin permeation of drugs by
found, by conventional and confocal fluorescence the combination of CaCl2 and EP. On the other hand,

Review 1277
ESCOBAR-CHÁVEZ ET AL

a prolonged enhancing effect on the skin permeation reactive unit was 1.7 mmol/L. Electropo­ration of MTX
of calcein by this combination may be due to a with an anion lipid enhancer under a mild hyperther-
high lamellar destruction and/or delayed barrier mic environment provided a significant transdermal
repair of SC. delivery within a short application time. The method
The objective of the experiment of Murthy et al106 may be an effective means of drug delivery for treating
was to study the influence of sodium dodecyl sulfate psoriasis or other MTX-sensitive disorders and avoids
(SDS) on transdermal transport of diffusants by elec- potential systemic toxicity.
troporation. The resistance of porcine epidermis in In another study, Wong et al108 designed a micro-
contact with SDS solution (0.2% w/v) dropped by electrode array to minimize the pain sensation of
40% within 24 hours. SDS improved the efficiency electroporation for enhancing transdermal drug
of transdermal delivery of glucose and dextrans of delivery. The influence of the size of the electrode-
molecular weight (MW) 4 kDa (FD4K) and 10 kDa skin contact area and of the distance between elec-
(FD10K) by electroporation. However, the transport trodes on the pain sensation was tested on human
of dextran MW 35 kDa (FD35K) was not influenced volunteers. The pain level decreased with the dimen-
significantly. Pretreatment of epidermis with SDS sion of electrode-skin contact area and with inter-
solution reduced its electroporation threshold from electrode distance. When both reached about 0.5
80 to 60 V. It appears that the presence of SDS during mm, the pain level was not perceptible even at the
electroporation helps to achieve the desired trans- threshold of transdermal electroporation level of 60
port with less electrical exposure dose. SDS enhanced electric pulses at 150 V, 1 ms at 1 to 10 Hz. The elec-
the transdermal delivery of molecules by electropo- tric thresholds for effective drug delivery, using
ration most likely by facilitating the barrier disrup- toluidine blue O as a marker on mouse skin, were
tion during pulse application and also by prolonging found to be the same for microelectrode arrays as for
the lifetime of electropores created by the pulse. larger electrodes and wider interelectrode distances.
In vivo transdermal electroporation using a micro-
Folic Acid Antagonists electrode array with 180 pulses of 150 V, 0.2 ms at 1
Hz, followed by a 30-minute methotrexate occlusion
The topical administration of methotrexate (MTX) increased more than 4-fold the systemic methotrex-
for the treatment of psoriasis and neoplastic diseases ate level in mice. The results demonstrated the
is restricted by the poor diffusion of MTX across the potential of painless delivery of significant amounts
SC. Wong et al107 applied electroporation to increase of chemotherapeutic agents through skin with the
the transdermal transport of MTX. Electrodes were new electrode arrays in a clinical setting.
placed either side-by-side on the surface of excised
full-thickness pig skin or on a piece of skin clamped
Other Uses
between compartments of a vertical diffusion cham-
ber. Sixty rectangular electric pulses at 120 V, 1 ms,
and 1 Hz were applied across the skin. MTX was left Gene Transfer
on the skin surface for an additional 10 minutes to The easy accessibility of skin makes it an excellent
take advantage of diffusion through electropores. target for gene transfer protocols. To take advantage
Cumulative drug transport was measured by radio- of skin as a target for gene transfer, it is important to
active tracing, using [3H]-methotrexate, from punch establish an efficient and reproducible delivery
biopsy samples taken from under the cathode. system. Electroporation is an established technique
Using side-by-side electrodes, treatment with the for enhancing plasmid delivery to many tissues in
pulses alone resulted in a 2.5-fold increase; adding vivo. A critical component of this technique is the
anionic lipid enhancers to the pulses resulted in a 4.4- electrode configuration. Electroporation parameters
fold enhancement compared with passive diffusion. were optimized by Heller et al109 for transgene
Concurrent iontophoresis for the 11-minute time expression with minimal tissue damage with a novel
period made a no significant contribution. To reduce electrode. The highest transgene expression and effi-
tissue resistance, we used 40°C hyperthermia in a ver- ciency of individual cell transformation with mini-
tical diffusion chamber; transport was increased mal damage was produced with eight 150-ms pulses
11-fold to 53 µg/cm2 (flux 290 µg/cm2 h). MTX pene- at a field strength of 100 V/cm. This electrode design
tration profiles indicated that more than half of the offers the potential for easier and more reproducible
MTX was confined to the epidermis and papillary electrically mediated cutaneous plasmid delivery
dermis. The tissue concentration in this superficial than the simple electrodes currently commercially

1278  •  J Clin Pharmacol 2009;49:1262-1283


ELECTROPORATION AS A PHYSICAL ENHANCER FOR SKIN DRUG DELIVERY

available. This electrode can be a valuable tool in applications of large amounts of growth factor have
determining the applicability of electrically medi- been required. Gene therapy has the potential to
ated cutaneous gene transfer. produce growth factors deep within the wound,
The use of a low-intensity current (iontophoresis) where they can be effective as well as able to
and high-voltage pulses (electroporation that per- constantly replenish growth factor that is destroyed
meabilizes lipid bilayers) has a potential for the by peptidases. Ferguson et al112 have shown that
administration of conventional and biotechnology- application of plasmid DNA expression vectors
produced drugs. Iontophoresis and electroporation directly into the wound is an inefficient modality.
enhance transdermal delivery of drugs, including Electroporation, the application of an electrical field
peptides and oligonucleotides. Electrochemotherapy across cells to permeabilize the cell membrane, has led
(ie, combination of a systemic or local delivery of a us to explore the possibility of using the technique
nonpermeant cytostatic drug with electroporation) to enhance transfection efficiency. They have identi-
kills tumor cells locally. Recently, Préat110 has shown fied electroporation parameters that improve the
that the local injection of a plasmid before electropo- efficiency of DNA transfection in cutaneous wounds,
ration significantly increases gene transfection. Hence, and they have shown that electroporation itself
electrotransfer is a promising alternative for drug does not impair wound healing. They are now on
and gene delivery. the threshold of exploring whether electroporation-
Intradermal injection of naked DNA results in assisted transfection with DNA plasmid expression
gene transfer to skin cells, but the efficiency of this vectors for growth factors will be an effective modal-
gene transfer method is relatively low and variable. ity for enhancing cutaneous wound healing.
Chesnoy and Huang111 have systematically opti-
To Develop Large Permeable Regions
mized several parameters to obtain reproducible,
high-level gene transfer to the mouse skin. Older in the Stratum Corneum
mice (~7 weeks) showed a significant decrease in The main barrier to transdermal drug delivery in
gene expression compared with younger mice (4-5 human skin is the SC. Pulsed electric fields (PEFs) of
weeks old). The composition of the solvent vehicle sufficient amplitude can create new aqueous path-
(electrolyte vs nonelectrolyte) strongly affected gene ways across this barrier and enhance drug delivery
expression in the skin. A higher level of gene expres- through the skin. Pliquett and Weaver113 described a
sion was achieved when naked DNA was dissolved model of pore formation between adjacent corneo-
in isotonic phosphate-buffered saline solution com- cytes that predicts the following sequence of events:
pared with isotonic dextrose solution. Finally, trans- (1) the PEF rapidly charges the SC near the electrode
fection efficiency in older mice was greatly improved until the transepidermal potential difference is large
by increasing the ionic strength of the solvent vehi- enough to drive water into a small region of the SC,
cle. The improved transfection efficiency was due to creating new aqueous pathways. (2) PEFs then drive
an enhanced DNA uptake by the skin cells. Gene a high-current density through this newly created
transfer was most evident in the subdermal smooth electropore to generate Joule heating that warms the
muscle cells and epidermal cells. With the opti- pore perimeter. (3) This temperature rise at the
mized conditions, gene transfer mediated by intrad- perimeter increases the probability of further elec-
ermal injection of naked DNA was comparable in troporation there as the local sphingolipids reach
efficiency to electroporation. However, cellular dis- their phase transition temperature. (4) This heat-
tributions of the gene transfer of the 2 methods were generated wave of further electroporation propagates
different. outward until the surface area of the pore becomes
so large that the reduced current density no longer
Wound Healing generates sufficient heat to reach the phase transi-
The major goal of wound-healing biology is to tion temperature of the sphingolipids. (5) Cooling
determine how a wound can be induced to repair and partial recovery occur after the field pulse.
damaged tissue faster and more efficiently. Enhance­ This process yields large, high-permeability
ment of dermal and epidermal regeneration is an regions in the SC at which molecules can more read-
extremely important goal for the treatment of many ily cross this skin barrier. A model is presented for
different types of wounds. Exogenous application of this process that predicts that the initial radius of
growth factors to the wound site has been shown to the first aqueous pathway is approximately 5 nm for
have potential to improve wound healing. Frequent a transdermal voltage of 60 V at room temperature.

Review 1279
ESCOBAR-CHÁVEZ ET AL

To Reseal Epidermis After Electroporation CONCLUSIONS


The resealing of porcine epidermis after electropora-
tion was investigated by Burgess et al.114 Porcine It should be evident from this review that electropo-
epidermis was subjected to electroporation (30 ration holds a lot of promise for the future of trans-
pulses at 100 V, 1 ms, and 1 Hz) in a vertical diffu- dermal drug delivery. Electroporation is an efficient
sion apparatus, in the presence of 2 mg/mL dimyris- method for enhancing transdermal drug delivery in
toylphosphatidylserine, to produce a long-lasting vitro and in vivo and expands the range of com-
permeable state. Resealing treatments include incu- pounds delivered transdermally. It could be a prom-
bation in 0.0625 to 0.25 mM poloxamer 188 (P188) ising alternative as a noninvasive delivery of
or incorporation of phosphatidylcholines (PC) and/ macromolecules (up to at least 40 kDa) and fast and/
or cationic lipids with additional pulses. The recov- or pulsatile transdermal delivery.
ery of electric resistance of the epidermis samples The combined use of electroporation with other
after electroporation with or without resealing treat- physical enhancers such as iontophoresis is likely to
ments was monitored. The transports of carboxyflu- yield useful and interesting data, which will inten-
orescein and glucose were measured during the sify the efforts to more fully explore electropora-
recovery process. Both P188 and PC were effective tion as a means of transdermal drug delivery.
in resealing in terms of electric conductance and The reader should realize that electrically assisted
transport, with P188 reacting more rapidly and com- delivery of drugs involves several chemical, bio-
pletely. P188-mediated lipid exchange between SC chemical, and physiological processes, and there is
lipid particles was measured by fluorescence reso- overlap in the mechanisms involved in transport via
nance energy transfer (FRET). Lipid reorganization electroporation.
facilitated by P188 and PC is suggested to be a major In summary, electroporation is one of the physic-
resealing mechanism of electroporation damage. ochemical methods for gene and drug delivery. It is
High-voltage pulsing of human skin (approxi- superior in some aspects but also has several draw-
mately 100 V across the skin, 1-ms pulses) has been backs. In some cases, as attested by the voluminous
hypothesized to cause electroporation of the SC and literature, it is still the preferred method. New devel-
large fluxes of drugs and other molecules across the opments are expected in the future to make this
skin, through newly created aqueous pathways. In method even more versatile. Pulse protocol and
contrast, iontophoresis (<0.5 mA per cm2, <1 V electrode design need to be optimized to reduce the
across the skin) has long been used in transdermal main adverse effect (ie, muscle contraction).
drug delivery and is believed to involve preexisting
Financial disclosure: José Juan Escobar-Chávez acknowledges
pathways associated with hair follicles and sweat
the PROFIP/UNAM grant and PAPITT 209709.
ducts. Either high-voltage pulsing or iontophoresis
was applied to human, hairless rat, or black rat
snake skin by Chen et al.115 Hairless rat skin contains
REFERENCES
more hair follicles than human skin, and snake skin
does not contain any hair follicles. All 3 types of
1. Elias PM. Epidermal lipids, barrier function and desquamation.
skin had comparable electrical resistances at low J Invest Dermatol. 1983;80:44-49.
voltages; however, the iontophoretic transport of 2. Cleary GW. Transdermal delivery systems: a medical rationale.
charged fluorescent molecules was significant for In: Shah VP, Maibach HI, eds. Topical Drug Bioavailability,
human and hairless rat skin, but no transport occu­ Bioequivalence and Penetration. New York: Plenum; 1993:17.
rred across snake skin, indicating that hair follicles 3. Scheuplein RJ, Blank IH. Permeability of the skin. Physiol Rev.
and sweat ducts play a major role in iontophoresis. 1971;51:702-747.
Electroporation caused large molecular transport for 4. Williams AC, Barry BW. Penetration enhancers. Adv Drug Deliv
all 3 types of skin and involved spontaneously form- Rev. 2004;56:603-618.
ing localized transport regions not associated with 5. Pellet M, Raghavan SL, Hadgraft J, Davis AF. The application of
supersaturated systems to percutaneous drug delivery. In: Guy
appendages. These experiments thus provide further
RH, Hadgraft J, eds. Transdermal Drug Delivery. New York:
support for the hypothesis that high-voltage pulsing Marcel Dekker; 2003:305.
causes electroporation in the SC and that this trans- 6. Tsai JC, Guy RH, Thornfeldt CR, et al. Metabolic approaches to
port mechanism is fundamentally different from enhance transdermal drug delivery: 1. Effect of lipid synthesis
iontophoresis. inhibitors. J Pharm Sci. 1998;85:643-648.

1280  •  J Clin Pharmacol 2009;49:1262-1283


ELECTROPORATION AS A PHYSICAL ENHANCER FOR SKIN DRUG DELIVERY

7. Elias PM, Feingold KR, Tsai J, Thornfeldt C, Menon G. 27. Walters KA, Roberts MS. Dermatological and Transdermal
Metabolic approach to transdermal drug delivery. In: Guy RH, Formulations. New York: Marcel Dekker; 2002.
Hadgraft J, eds. Transdermal Drug Delivery. New York: Marcel 28. Nevill AM. The need to scale for differences in body size and
Dekker; 2003:285. mass: and explanation of Klieber’s 0.75 mass exponent. Am
8. Schreier H, Bouwstra J. Liposomes and niosomes as topical Physiol Soc. 1994;77:2870-2873.
drug carriers: dermal and transdermal drug delivery. J Control 29. Olvera-Martínez BI, Cazares-Delgadillo J, Calderilla-Fajardo
Release. 1994;30:1-15. SB, Villalobos-García R, Ganem-Quintanar A, Quintanar-Guerrero
9. Cevc G. Transfersomes, liposomes and other lipid suspensions D. Preparation of polymeric nanocapsules containing octyl
on the skin: permeation enhancement, vesicle penetration, and methoxycinnamate by the emulsification-diffusion technique:
transdermal drug delivery. Crit Rev Ther Drug Carrier Syst. penetration across the stratum corneum. J Pharm Sci. 2005;94:
1996;13:257-388. 1552-1559.
10. Cevc G. Transferosomes: innovative transdermal drug carriers. 30. Escobar-Chávez JJ, Quintanar-Guerrero D, Ganem-Quintanar
In: Rathbone MJ, Hadgraft J, Roberts MS, eds. Modified Release A. In vivo skin permeation of sodium naproxen formulated in
Drug Delivery Technology. New York: Marcel Dekker; 2003:533. PF-127 gels: Effect of Azone and Transcutol. Drug Develop Ind
11. Godin B, Touitou E. Ethosomes: new prospects in transdermal Pharm. 2005;31:447-454.
delivery. Crit Rev Ther Drug Carrier Syst. 2003;20:63-102. 31. Escobar-Chávez JJ, López-Cervantes M, Naïk A, Kalia YN,
12. Brown MB, Traynor MJ, Martin GP, Akomeah FK. Transdermal Quintanar-Guerrero D, Ganem-Quintanar A. Applications of the
drug delivery systems: skin perturbation devices. In: Jain KK, ed. thermoreversible Pluronic F-127 gels in pharmaceutical formula-
Drug Delivery Systems. Vol. 37. Totowa, NJ: Humana; 2008: tions. J Pharm Pharmaceut Sci. 2006;9:339-358.
119-139. 32. Miyazaki S, Yokouchi Ch, Nakamura T, Hashiguchi N,
Hou W-M, Takada M. Pluronic F-127 gels as a novel vehicle for
13. Escobar-Chávez JJ, Merino-Sanjuán V, López-Cervantes M,
rectal administration of indomethacin. Chem Pharm Bull. 1986;
et al. The use of iontophoresis in the administration of nicotine
34:1801-1808.
and new non-nicotine drugs through the skin for smoking cessation.
Curr Drug Discov Technol. 2009 Sep 1. [Epub ahead of print] 33. Chi S-Ch, Do K, Tan HK, Chun HW. Anti-inflammatory and
analgesic transdermal gel. US patent number 5,527,832, 1996.
14. Merino V, Micó-Albiñana T, Nácher A, Díez-Sales O, Herráez
M, Merino-Sanjuán M. Enhancement of nortriptyline penetration 34. Fang JY, Leu YL, Wang YY, Tsai YH. In vitro topical applica-
through human epidermis: influence of chemical enhancers and tion and in vivo pharmacodynamic evaluation of nonivamide
iontophoresis. J Pharm Pharmacol. 2008;60:415-420. hydrogels using Wistar rat as an animal model. Eur J Pharm Sci.
2002;15:417-423.
15. El-Kamel AH, Al-Fagih IM, Alsarra IA. Effect of sonophoresis
and chemical enhancers on testosterone transdermal delivery 35. Shin SC, Cho CW, Oh IJ. Effects of non ionic surfactants as
from solid lipid microparticles: an in vitro study. Curr Drug Deliv. permeation enhancers towards piroxicam from the poloxamer gel
2008;5:20-26. through rat skins. Int J Pharm. 2001;222:199-203.
16. Zhang L, Widera G, Rabussay D. Enhancement of the effective- 36. Liaw J, Lin Y-Ch. Evaluation of poly(ethylene oxide)-
ness of electroporation-augmented cutaneous DNA vaccination poly(propylene oxide)-poly(ethylene oxide) (PEO-PPO-PEO) gels
by a particulate adjuvant. Bioelectrochemistry. 2004;63:369-373. as a release vehicle for percutaneous fentanyl. J Control Release.
2000;68:273-282.
17. Banga AK, Bose S, Ghosh TK. Iontophoresis and electropora-
tion: comparisons and contrasts. Int J Pharm. 1999;179:1-19. 37. Wang YY, Hong CT, Chiu WT, Fang JY. In vitro and in vivo
evaluations of topically applied capsaicin and nonivamide from
18. Prausnitz MR. A practical assessment of transdermal drug
hydrogels. Int J Pharm. 2001;224:1-2.
delivery by skin electroporation. Adv Drug Deliv Rev. 1999;
38. Kattan El AF, Asbill CS, Kim N, Michniak BB. Effect of formu-
35:61-76.
lation variables on the percutaneous permeation of ketoprofen
19. Vanbever R, Préat V. In vivo efficacy and safety of skin elec- from gel formulations. Drug Deliv. 2000;7(3):147-153.
troporation. Adv Drug Deliv Rev. 1999;35:77-88.
39. Curdy C, Kalia YN, Naïk A, Guy RH. Piroxicam delivery into
20. Forslind BA. Domain mosaic model of skin barrier. Acta Derm human stratum corneum in vivo: iontophoresis versus passive
Venereol. 1994;74:1-6. diffusion. J Control Release. 2001;76:73-79.
21. Potts RO, Francoeur ML. The influence of stratum corneum 40. Mattorano DA, Kupper LL, Nylander-French LA. Estimating
morphology on water permeability. J Invest Dermatol. 1991;96: dermal exposure to jet fuel (naphthalene) using adhesive tape
495-499. strip samples. Ann Occup Hyg. 2004;48:139-146.
22. Potts RO, Guy RH. Predicting skin permeability. Pharm Res. 41. Chao Y-Ch, Nylander-French LA. Determination of keratin
1992;9:663-669. protein in a tape-stripped skin sample from jet fuel exposed skin.
23. Ellias PM. Epidermal barrier function: intercellular lamellar Ann Occup Hyg. 2004;48:65-73.
lipid structures, origin, composition and metabolism. J Control 42. Chizmadzhev YA, Zamitsin VG, Weaver JC, Potts UO.
Release. 1991;15:199-208. Mechanism of electroinduced ionic species transport through a
24. Barry BW. Dermatological formulations: percutaneous absorp- multilamellar lipid system. Biophysical J. 1995;68:749-765.
tion. In: Swarbick J, ed. Drugs and the Pharmaceutical Sciences. 43. Chizmadzhev YA, Indenbom AV, Kuzmin PI, Galichenko SV,
New York: Marcel Dekker; 1983:202. Weaver JC, Potts RO. Electrical properties of skin at moderate
25. Hadgraft J. Skin, the final frontier. Int J Pharm. 2001;224 voltages: contribution of appendageal macropores. Biophys J.
(1-2):1-18. 1998;74:843-856.
26. Guy RH, Hadgraft J. Transdermal Drug Delivery. New York: 44. Jiali B, Hong W, Huiping W, Jiguang G. Quantitative descrip-
Marcel Dekker; 2003. tion and model of molecule transport through skin. Presented at:

Review 1281
ESCOBAR-CHÁVEZ ET AL

26th Annual International Conference of the IEEE EMBS; Septem­ 63. Hu Q, Liang W, Bao J, Ping Q. Enhanced transdermal delivery
ber 1-5, 2004; San Francisco, California. of tetracaine by electroporation. Int J Pharm. 2000;202:121-124.
45. Treco DA, Selden RF. Non-viral gene therapy. Mol Med Today. 64. Bommannan DB, Tamada J, Leung L, Potts RO. Effect of elec-
1995;1:314-321. troporation on transdermal iontophoretic delivery of luteinizing
46. Weaver JC. Electroporation theory: concepts and mechanisms. hormone releasing hormone (LHRH) in vitro. Pharm Res. 1994;
In: Nickoloff JA, ed. Electroporation Protocols for Microorganisms. 11:1809-1814.
Totowa, NJ: Humana; 1995:1. 65. Murthy SN, Zhao Y, Hui SW, Sen A. Synergistic effect of
47. Prausnitz, MR, Bose VG, Langer R, Weaver JC. Electroporation anionic lipid enhancer and electroosmosis for transcutaneous
of mammalian skin: a mechanism to enhance transdermal drug delivery of insulin. Int J Pharm. 2006;326(1/2):1-6.
delivery. Proc Natl Acad Sci USA. 1993;90:10504-10508. 66. Regnier V, Préat V. Mechanisms of a phosphorothioate oligo-
48. Inada H, Ghanem AH, Higuchi WI. Studies on the effects of nucleotide delivery by skin electroporation. Int J Pharm. 1999;
applied voltage and duration on human epidermal membrane 184:147-156.
alteration: recovery and the resultant effects upon iontophoresis. 67. Vanbever R, Leroy MA, Préat V. Transdermal permeation of
Pharm Res. 1994;11:687-697. neutral molecules by electroporation. J Control Release. 1998;54:
49. Weaver JC, Chizmadzhev Y. Electroporation. In: Polte C, 243-250.
Postow E, eds. Biological Effects of Electromagnetic Fields. Boca 68. Li LH, McCarthy P, Hui SW. High-efficiency electrotransfec-
Raton, FL: CRC Press; 1996:247. tion of human primary hematopoietic stem cells. FASEB J.
50. Edwards DA, Prausnitz MR, Langer R, Weaver JC. Analysis of 2001;15:586-588.
enhanced transdermal transport by skin electroporation. J Control 69. Yew NS, Zhao H, Przybylska M, et al. CpG-depleted plasmid
Release. 1995;34:211-221. DNA vectors with enhanced safety and long-term gene expression
51. Prausnitz MR. Do high-voltage pulses cause changes in skin in vivo. Mol Ther. 2002;5:731-738.
structure? J Control Release. 1996;40:321-326. 70. Chen S, Shohet RV, Bekeredjian R, Frenkel P, Grayburn PA.
52. Vanbever R, Lecouturier N, Preat V. Transdermal delivery of Optimization of ultrasound parameters for cardiac gene delivery
metoprolol by electroporation. Pharm Res. 1994;11:1657-1662. of adenoviral or plasmid deoxyribonucleic acid by ultrasound-
53. Vanbever R, LeBoulenge E, Preat V. Transdermal delivery of targeted microbubble destruction. J Am Coll Cardiol. 2003;42:
fentanyl by electroporation: 1. Influence of electrical factors. 301-308.
Pharm Res. 1996;13:559-565. 71. Ding Z, Fach C, Sasse A, Godecke A, Schrader J. A minimally
54. Choi MJ, Maibach HI. Topical vaccination of DNA antigens: invasive approach for efficient gene delivery to rodent hearts.
topical delivery of DNA antigens. Skin Pharmacol Appl Skin Gene Ther. 2004;11:260-265.
Physiol. 2003;16:271-282. 72. Tanaka S, Iwai M, Harada Y, et al. Targeted killing of carcino-
55. Wang S, Kara M, Krishnan TR. Topical delivery of cyclosporin embryonic antigen (CEA)–producing cholangiocarcinoma cells by
A coevaporate using electroporation technique. Drug Dev Ind polyamidoamine dendrimer–mediated transfer of an Epstein-Barr
Pharm. 1997;23:657-663. virus (EBV)–based plasmid vector carrying the CEA promoter.
Cancer Gene Ther. 2000;7:1241-1250.
56. Prausnitz MR, Edelman ER, Gimm JA, Langer R, Weaver JC.
Transdermal delivery of heparin by skin electroporation. 73. Keravala A, Groth AC, Jarrahian S, et al. A diversity of serine
Biotechnology. 1995;13:1205-1209. phage integrases mediate site-specific recombination in mamma-
57. Zewert TE, Pliquett UF, Langer R, Weaver JC. Transdermal lian cells. Mol Genet Genomics. 2006;276:135-146.
transport of DNA antisense oligonucleotides by electroporation. 74. Nickoloff JA. Preface. In: Nickoloff JA, ed. Electroporation
Biochem Biophys Res Commun. 1995;212:286-292. Protocols for Microorganisms. Totowa, NJ: Humana; 1995:5.
58. Brand RM, Wahl A, Iverson PL. Effects of size and sequence 75. Miller EM, Nickoloff JA. Escherichia coli electrotransforma-
on the iontophoretic delivery of oligonucleotides. J Pharm Sci. tion. In: Nickoloff JA, ed. Electroporation Protocols for
1998;87:49-52. Microorganisms. Totowa, NJ: Humana; 1995:105.
59. Zhang L, Hofmann GA. Electric pulse mediated transdermal 76. Tetsuichiro S. In Vivo Electroporation. http://www.frontier.
drug delivery. Proc Int Symp Control Release Bioact Mater. kyoto-u.ac.jp/rc01/in_vivo_electroporation.html. Accessed February
1997;24:27-28. 15, 2003.
60. Pliquett U, Gallo S, Hui SW, Gusbeth C, Neumann E. Local and 77. Weaver JC. Electroporation theory: concepts and mechanisms.
transient structural changes in stratum corneum at high electric In: Nickoloff JA, ed. Electroporation Protocols for Microorganisms.
fields: contribution of Joule heating. Bioelectrochemistry. 2005; Totowa, NJ: Humana; 1995:1.
67:37-46. 78. Pliquett U, Langer R, Weaver JC. Changes in the electrical
61. Fang JY, Huang Y-B, Wang H-Y, Tsai Y-H. Electrically-assisted properties of human stratum corneum due to electroporation.
skin permeation of two synthetic capsaicin derivatives, sodium Biochim Biophys Acta. 1995;1239:111-121.
nonivamide acetate and sodium nonivamide propionate, via rate- 79. Mir L, Orlowski S, Belehradek JJ, Paolelli C. Electrochemo­
controlling polyethylene membranes. Biol Pharm Bull. 2005;28: therapy: potentiation of antitumor effect of bleomycin by electric
1695-1701. pulses. Eur J Cancer. 1991;27:68-72.
62. Murthy SN, Sen A, Zhao YL, Hui SW. pH influences the post- 80. Heller R, Jaroszeski R, Glass LF, et al. Phase I/II trial for the
pulse permeability state of skin after electroporation. J Control treatment of cutaneous and subcutaneous tumor using electro-
Release. 2003;93:49-57. chemotherapy. Cancer. 1996;77:964-971.

1282  •  J Clin Pharmacol 2009;49:1262-1283


ELECTROPORATION AS A PHYSICAL ENHANCER FOR SKIN DRUG DELIVERY

81. Murthy SN, Zhao Y-L, Sen A, Hui SW. Cyclodextrin enhanced comparison with iontophoresis and electroporation. Br J Dermatol.
transdermal delivery of piroxicam and carboxyfluorescein by 2004;151:132-140.
electroporation. J Control Release. 2004;99:393-402. 99. Regnier V, Tahiri A, André N, Lemaitrec M, Préat V.
82. Huang JF, Sung KC, Hu O Y-P, Wang JJ, Lin YH, Fang JY. The Electroporation-mediated delivery of 3′-protected phosphodiester
effects of electrically assisted methods on transdermal delivery of oligodeoxynucleotides to the skin. J Control Release. 2000;67:
nalbuphine benzoate and sebacoyl dinalbuphine ester from solu- 337-346.
tions and hydrogels. Int J Pharm. 2005;297:162-171. 100. Zhao YL, Murthy SN, Manjili MH, Guana LJ, Sena A, Hui SW.
83. Sung KC, Fang J-Y, Wang JJ, Hu O Y-P. Transdermal delivery Induction of cytotoxic T-lymphocytes by electroporation-enhanced
of nalbuphine and its prodrugs by electroporation. Eur J Pharm needle-free skin immunization. Vaccine. 2006;24:1282-1290.
Sci. 2003;18:63-70. 101. Hui SW. The application of electroporation to transfect
84. Fang J-Y, Hwang T-L, Huang Y-B, Tsai Y-H. Transdermal hematopoietic cells and to deliver drugs and vaccines transcuta-
iontophoresis of sodium nonivamide acetate: V. Combined effect neously for cancer treatment. Technol Cancer Res Treat.
of physical enhancement methods. Int J Pharm. 2002;235:95-105. 2002;1:373-384.
85. Vanbever R, Prausnitz MR, Préat V. Macromolecules as novel 102. Fang JY, Hung CF, Hwang TL, Wong WW. Transdermal deliv-
transdermal transport enhancers for skin electroporation. Pharm ery of tea catechins by electrically assisted methods. Skin Phar­
Res. 1997;14:638-644. macol Physiol. 2006;19:28-37.
86. Badkar AV, Smith AM, Eppstein JA, Banga AK. Transdermal 103. Marrat F, Levy JL, Santi P, Kalia YN. In vitro evaluation of
delivery of interferon alpha-2B using microporation and ionto- electrotreatment on skin permeability. J Cosmet Dermatol. 2008;7:
phoresis in hairless rats. Pharm Res. 2007;24:1389-1395. 105-111.
87. Sharma A, Kara M, Smith FR, Krishnan TR. Transdermal drug 104. Sen A, Zhao Y, Zhang L, Hui SW. Enhanced transdermal
delivery using electroporation: II. Factors influencing skin revers- transport by electroporation using anionic lipids. J Control
ibility in electroporative delivery of terazosin hydrochloride in Release. 2002;82:399-405.
hairless rats. J Pharm Sci. 2000;89:536-544. 105. Tokudome Y, Sugibayashi K. Mechanism of the synergic
88. Sharma A, Kara M, Smith FR, Krishnan TR. Transdermal drug effects of calcium chloride and electroporation on the in vitro
delivery using electroporation: I. Factors influencing in vitro enhanced skin permeation of drugs. J Control Release. 2004;95:
delivery of terazosin hydrochloride in hairless rats. J Pharm Sci. 267-274.
2000;89:528-535. 106. Murthy SN, Sen A, Hui SW. Surfactant-enhanced transdermal
89. Denet A-N, Ucakar B, Préat V. Transdermal delivery of timolol delivery by electroporation. J Control Release. 2004;98:307-315.
and atenolol using electroporation and iontophoresis in combina- 107. Wong T-W, Zhao Y-L, Sen A, Hui SW. Pilot study of topical
tion: a mechanistic approach. Pharm Res. 2003;20:1946-1951. delivery of methotrexate by electroporation. Br J Dermatol.
90. Fang J-Y, Hung Ch-F, Fang Y-P, Chan T-F. Transdermal ionto- 2005;152:524-530.
phoresis of 5-fluorouracil combined with electroporation and 108. Wong TW, Chen Ch-H, Huang Ch-Ch, Lin Ch-D, Hui SW.
laser treatment. Int J Pharm. 2004;270:241-249. Painless electroporation with a new needle-free microelectrode
91. Sersa G, Miklavcic D, Cemazar M, Rudolf Z, Pucihar G, array to enhance transdermal drug delivery. J Control Release.
Snoj M. Electrochemotherapy in treatment of tumours. Eur J Surg 2006;110:557-565.
Oncol. 2008;34:232-240. 109. Heller LC, Jaroszeski MJ, Coppola D, McCray AN, Hickey J,
92. Murthy SN, Zhao Y-L, Marlan K, Hui SW, Kazim AL, Sen A. Heller R. Optimization of cutaneous electrically mediated plas-
Lipid and electroosmosis enhanced transdermal delivery of insu- mid DNA delivery using novel electrode. Gene Ther. 2007;14:
lin by electroporation. J Pharm Sci. 2006;95:2041-2050. 275-280.
93. Tokumoto S, Higo N, Sugibayashi K. Effect of electroporation 110. Préat V. Drug and gene delivery using electrotransfer. Ann
and pH on the iontophoretic transdermal delivery of human insu- Pharm Fr. 2001;59:239-244.
lin. Int J Pharm. 2006;326:13-19. 111. Chesnoy S, Huang L. Enhanced cutaneous gene delivery fol-
94. Medi BM, Singh J. Electronically facilitated transdermal lowing intradermal injection of naked DNA in a high ionic
delivery of human parathyroid hormone (1-34). Int J Pharm. strength solution. Mol Ther. 2002;5:57-62.
2003;263:25-33. 112. Ferguson M, Byrnes C, Sun L, et al. Wound healing enhance-
95. Chang S-L, Hofmann GA, Zhang L, Deftos LJ, Banga AK. The ment: electroporation to address a classic problem of military
effect of electroporation on iontophoretic transdermal delivery of medicine. World J Surg. 2005;29:S55-S59.
calcium regulating hormones. J Control Release. 2000;66:127-133. 113. Pliquett U, Weaver JC. Feasibility of an electrode-reservoir
96. Essa EA, Bonner MC, Barry BW. Electrically assisted skin device for transdermal drug delivery by noninvasive skin elec-
delivery of liposomal estradiol: phospholipid as damage retar- troporation. IEEE Trans Biomed Eng. 2007;54:536-538.
dant. J Control Release. 2004;95:535-546. 114. Burgess SE, Zhao YL, Sen A, Hui SW. Resealing of electropo-
97. Johnson PG, Hui SW, Oseroff AR. Electrically enhanced per- ration of porcine epidermis using phospholipids and poloxamers.
cutaneous delivery of d-aminolevulinic acid using electric pulses Int J Pharm. 2007;336:269-275.
and a DC potential. Photochem Photobiol. 2002;75:534-540. 115. Chen T, Langer R, Weaver JC. Skin electroporation causes
98. Fang JY, Lee W-R, Shen S-C, Fang Y-P, Hu C-H. Dermatological molecular transport across the stratum corneum through local-
surgery and lasers enhancement of topical 5-aminolaevulinic ized transport regions. J Investig Dermatol Symp Proc. 1998;3:
acid delivery by erbium:YAG laser and microdermabrasion: a 159-165.

Review 1283

You might also like