You are on page 1of 340

Percutaneous

Penetration Enhancers
Chemical Methods in
Penetration Enhancement

Drug Manipulation
Strategies and Vehicle
Effects

Nina Dragicevic-Curic
Howard I. Maibach
Editors

123
Percutaneous Penetration Enhancers
Chemical Methods in Penetration
Enhancement
Nina Dragicevic
Howard I. Maibach
Editors

Percutaneous
Penetration Enhancers
Chemical Methods in
Penetration
Enhancement
Drug Manipulation Strategies
and Vehicle Effects
Editors
Nina Dragicevic Howard I. Maibach
Apoteka "Beograd" San Francisco, CA
Belgrade USA
Serbia

ISBN 978-3-662-45012-3 ISBN 978-3-662-45013-0 (eBook)


DOI 10.1007/978-3-662-45013-0

Library of Congress Control Number: 2014958975

Springer Berlin Heidelberg New York Dordrecht London


© Springer-Verlag Berlin Heidelberg 2015
This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software,
or by similar or dissimilar methodology now known or hereafter developed. Exempted from this
legal reservation are brief excerpts in connection with reviews or scholarly analysis or material
supplied specifically for the purpose of being entered and executed on a computer system, for
exclusive use by the purchaser of the work. Duplication of this publication or parts thereof is
permitted only under the provisions of the Copyright Law of the Publisher’s location, in its
current version, and permission for use must always be obtained from Springer. Permissions for
use may be obtained through RightsLink at the Copyright Clearance Center. Violations are liable
to prosecution under the respective Copyright Law.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
While the advice and information in this book are believed to be true and accurate at the date of
publication, neither the authors nor the editors nor the publisher can accept any legal responsibility
for any errors or omissions that may be made. The publisher makes no warranty, express or
implied, with respect to the material contained herein.

Printed on acid-free paper

Springer-Verlag GmbH Berlin Heidelberg is part of Springer Science+Business Media


(www.springer.com)
Preface

The main function of skin is the protection of the body from the external envi-
ronment by preventing loss of water and the ingress of exogenous substances.
This implies that the skin acts as a barrier for the diffusion of substances into
the underlying tissue. Despite this role, the skin has become recognized as an
important drug delivery route which can be reached directly. It is an ideal site
for the application of drugs for achieving local (topical) and systemic (trans-
dermal) drug effects. Local or topical drug delivery assumes treating various
skin diseases, while transdermal delivery aims to achieve systemically active
drug levels in order to treat systemic diseases. Drugs have been applied to the
skin to achieve also regional drug delivery which involves drug application to
the skin to treat or alleviate disease symptoms in deep tissues beneath the skin
(such as in musculature, etc.). Topical and transdermal drug delivery offer a
number of advantages compared to other conventional routes, and hence they
are of great interest to pharmaceutical research, which explains the increasing
interest in skin as a site of drug application.
However, skin represents a formidable barrier for percutaneous drug
absorption, being of crucial importance for achieving topical and transdermal
effects of drugs. Significant efforts have been devoted to developing strategies
to overcome the impermeability of intact human skin. There are many ways
for circumventing the stratum corneum, which provides the main barrier to
drug penetration. These methods can be divided into chemical and physical
penetration enhancement methods, i.e. percutaneous penetration enhancers,
which are described in this book series Percutaneous Penetration Enhancers.
The aim of this book series is to provide to readers working in academia
and industry, including young researchers, an up-to-date comprehensive
work describing all the important topics required to understand the principles
of enhancing transdermal and dermal drug delivery. The book series contains
five books.
The book Chemical Methods in Penetration Enhancement: Drug
Manipulation Strategies and Vehicle Effects begins with a description of the
skin, as understanding of its structure, function and especially its penetration
pathways is fundamental to understanding how topical and transdermal dos-
age forms work and how different methods may be employed to enhance per-
cutaneous drug penetration. The first two parts of the book devoted to skin and
the stratum corneum, representing its uppermost layer being responsible for
its protection, discuss their structure, the importance of the lipid organization
in the stratum corneum, the different penetration pathways through the skin

v
vi Preface

with an emphasis on the increasing importance of the follicular route, as well


as the influence of different excipients on the skin. The focus of the book is
on the chemical methods used to overcome the impermeability of intact skin,
such as different drug manipulation strategies (drug or prodrug selection,
chemical potential control, eutectic systems, complexes with cyclodextrines,
etc.) and formulation/vehicle effects (influences of: emulsions, nanoemul-
sions, pickering emulsions, microemulsions, emulsifiers, emollients, liquid
crystalline structures, gels, etc.) on the penetration enhancement of drugs.
The book Chemical Methods in Penetration Enhancement: Nanocarriers
describes similarly to the first book chemical methods used in penetration
enhancement of drugs. However, this book is devoted to the application of
different kinds of nanocarriers and represents an attempt to familiarize the
readers with the importance of nanocarriers used to enhance the percutane-
ous penetration of drugs as they have numerous advantages in comparison to
conventional drug formulations. More recently, different types of nanocarri-
ers have been designed by researchers which allow controlled and targeted
drug delivery (dermal or transdermal drug delivery), improved therapeutic
effectiveness and reduced side effects of drugs. As carriers they can be classi-
fied into lipid-based vesicles (e.g. liposomes, transfersomes, invasomes, etc.),
surfactant-based vesicles (e.g. niosomes, novasomes and others), lipid-based
particulate carriers (e.g. solid lipid nanoparticles, nanostructured lipid car-
riers and lipid nanocapsules), polymer-based particulate carriers (e.g. poly-
meric nano- and microparticles, polymeric nanocapsules, polymeric micelles,
dendrimers, dendritic core-multishell nanocarriers, etc.), nanocrystals and
others. This book focusing on the different nanocarriers gives a comprehen-
sive review of their use as promising dermal and transdermal drug delivery
systems. It also considers the use of nanocarriers for cutaneous immuniza-
tion offering the important advantage of being painless and having a stronger
immune response compared to the intramuscular injection of vaccines. In
addition, the book provides insights on the safety of the use of nanoparticles.
The book Chemical Methods in Penetration Enhancement: Modification
of the Stratum Corneum similarly to the aforementioned two books describes
the chemical methods used in penetration enhancement of drugs with an
emphasis on the enhancing methods used to modify the stratum corneum. It
starts with the classification of penetration enhancers, their mode of action
and provides insights on the structure–activity relationship of chemical pen-
etration enhancers. The focus of this book is on the most commonly used
classes of skin penetration enhancers being investigated in scientific litera-
ture and used in commercial topical and transdermal formulations, and their
representatives are discussed in more detail, including their mechanism of
action, where known. The following penetration enhancers are considered in
the book: alcohols (e.g. ethanol, etc.), glycols (e.g. propylene glycol, etc.),
amides (e.g. 1-dodecylazacycloheptan-2-one or laurocapram (Azone®), etc.),
fatty acids (e.g. oleic acid, etc.), fatty acid esters (e.g. isopropyl myristate,
etc.), ether alcohols (e.g. diethylene glycol monoethyl ether (Transcutol®)),
pyrrolidones (e.g. N-methyl-2-pyrrolidone, etc.), sulphoxides (e.g. dimethyl
sulphoxide, etc.), surfactants (e.g. polysorbates, etc.), terpenes (e.g. L-menthol,
etc.), peptides and new classes of enhancers, such as iminosulfuranes,
Preface vii

transcarbams, dimethylamino acid esters and dicarboxylic acid esters. In


addition, synergistic effects of different chemical penetration enhancers have
been discussed in the book as an important feature of chemical penetration
enhancers. Furthermore, the safety profile of chemical penetration enhancers
is considered.
The book Physical Methods in Penetration Enhancement considers the
current status and possible future directions in the emerging area of physi-
cal methods being used as potent enhancers for the percutaneous penetration
of drugs. It gives a comprehensive overview of the most used methods for
enhancing dermal and transdermal drug delivery. It covers sonophoresis, ion-
tophoresis, electroporation, magnetophoresis, microneedles, needle-free jet
injectors, ablation methods (electrical, thermal or laser skin ablation) and oth-
ers. The numerous advantages of these methods have opened new frontiers
in the penetration enhancement of drugs for dermal and transdermal drug
delivery. Cutaneous vaccination and gene delivery by physical methods have
been also discussed in this volume. Consideration was given to new methods,
too, such as a novel electrochemical device for penetration enhancement, dif-
ferent waves (e.g. photoacoustic waves, microwaves, etc.), natural submicron
injectors, moxibustion and others. Furthermore, the combined use of different
physical methods or of physical methods and passive enhancement methods
(chemical penetration enhancement methods) are discussed as they provide,
due to their synergistic effects, higher percutaneous drug penetration when
used together.
The book Drug Penetration Into/Through the Skin: Methodology and
General Considerations provides fundamental principles of the drug pen-
etration into/through the skin, from covering basic mathematics involved in
skin permeation of drugs, influences of drug application conditions and other
factors on drug penetration, mechanistic studies of penetration enhancers,
influences of the type of skin used (human native or reconstructed skin) to
different methods utilized to assess the drug penetration into/through the skin
and to determine the amount of permeated drug (such as tape stripping of the
stratum corneum, electron spin resonance, Raman spectroscopy, attenuated
total reflection, confocal laser scanning microscopy, single and multiphoton
microscopy, etc.). Retardation strategies are also discussed as being impor-
tant for some classes of substances, such as sunscreens. The safety of applied
penetration enhancers as well as the research ethics in the investigation of
dermal and transdermal drug delivery are addressed in this book. The book
ends with the current status and future perspectives of passive/chemical and
active/physical penetration enhancement methods as they are gaining exten-
sive interest as promising tools to enable an efficient dermal or transdermal
drug delivery.
We are very thankful to all the authors who contributed chapters to the
book series Percutaneous Penetration Enhancers, as they found time to work
on the chapters despite having busy schedules and commitments. All the
authors are eminent experts in the scientific field which was the subject of
their chapter, and hence their contribution raised the value of the book. We
also sincerely thank our collaborators from Springer: Ellen Blasig, Isabella
Formento, Sverre Klemp, Srinath Raju, Andre Tournois, Grant Weston and
viii Preface

Portia Formento Wong, for their dedicated work which was necessary to
achieve such a high standard of publication. We highly appreciate readers’
comments, suggestions and criticisms to improve the next edition of the book
series.

Belgrade, Serbia Nina Dragicevic


San Francisco, CA, USA Howard I. Maibach
Contents

Part I The Skin

1 Skin Deep: The Basics of Human Skin Structure


and Drug Penetration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Keng Wooi Ng and Wing Man Lau
2 Epidermal Lipids and the Intercellular Pathway . . . . . . . . . . . 13
Philip W. Wertz
3 The Importance of Stratum Corneum Lipid
Organization for Proper Barrier Function . . . . . . . . . . . . . . . . 19
Annett Schroeter, Adina Eichner, Josefin Mueller,
and Reinhard H.H. Neubert
4 Molecular Structure and Function of the Skin Barrier . . . . . . 39
Lars Norlén
5 The Increasing Importance of the Hair Follicle
Route in Dermal and Transdermal Drug Delivery . . . . . . . . . . 43
Alexa Patzelt and Jürgen Lademann
6 The Correlation Between Transepidermal Water
Loss and Percutaneous Absorption . . . . . . . . . . . . . . . . . . . . . . 55
Jacquelyn Levin and Howard I. Maibach
7 Influence of Excipients on Two Elements of the
Stratum Corneum Barrier: Intercellular Lipids
and Epidermal Tight Junctions . . . . . . . . . . . . . . . . . . . . . . . . . 69
Laurène Roussel, Rawad Abdayem, Elodie Gilbert,
Fabrice Pirot, and Marek Haftek

Part II Penetration Enhancement Techniques


in Skin Delivery

8 Targets in Dermal and Transdermal Delivery and


Classification of Penetration Enhancement Methods . . . . . . . . 93
Jelena Predic Atkinson, Howard I. Maibach,
and Nina Dragicevic

ix
x Contents

9 Formulation Effects in Percutaneous Absorption. . . . . . . . . . . 109


Rina Kuswahyuning, Jeffrey E. Grice, Hamid R. Moghimi,
and Michael S. Roberts

Part III Drug Manipulation Strategies


in Penetration Enhancement

10 Selection of a Proper Prodrug for Penetration


Enhancement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 137
Kenneth B. Sloan, Jennifer Synovec, and Scott C. Wasdo
11 Transdermal Drug Delivery Systems
Using Supersaturation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 151
Vishwas Rai and Lakshmi Raghavan
12 Eutectic Systems for Penetration Enhancement . . . . . . . . . . . . 163
Stuart A. Jones, Sarah Fiala, and Marc B. Brown
13 Formation of Ion Pairs and Complex Coacervates. . . . . . . . . . 175
Liang Fang, Honglei Xi, and Dongmei Cun
14 Formulation of Drug-Cyclodextrin Complexes. . . . . . . . . . . . . 189
Thorsteinn Loftsson

Part IV Influence of Vehicle Effects in Penetration


Enhancement

15 Dermal and Transdermal Formulations:


How They Can Affect the Active Compound . . . . . . . . . . . . . . 209
Jessica Stahl

Part V Emulsions as Vehicles for Skin Delivery

16 The Effects of Emulsifiers and Emulsion Formulation


Types on Dermal and Transdermal Drug Delivery . . . . . . . . . 223
Anja Otto and Jeanetta du Plessis
17 Skin Permeation: Enhancing Ability of Liquid Crystal
Formulations. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 243
Wesam R. Kadhum, Hiroaki Todo, and Kenji Sugibayashi
18 Nanoemulsions in Dermal Drug Delivery . . . . . . . . . . . . . . . . . 255
Victoria Klang, Julia C. Schwarz,
and Claudia Valenta
19 Pickering Emulsions for Controlled Drug
Delivery to the Skin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 267
Yves Chevalier, Marie-Alexandrine Bolzinger,
and Stéphanie Briançon
Contents xi

Part VI Gels as Vehicles for Skin Delivery

20 Hydrogel Vehicles for Hydrophilic Compounds . . . . . . . . . . . . 285


Teresa Cerchiara, Federica Bigucci, and Barbara Luppi
21 Lecithin Organogels in Enhancing Skin
Delivery of Drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 299
I.M. Shaikh, K.R. Jadhav, and V.J. Kadam
22 Thermosensitive Hydrogels in Dermatology:
A Multidisciplinary Overview. . . . . . . . . . . . . . . . . . . . . . . . . . . 315
Damien Salmon, Laurène Roussel, Elodie Gilbert,
Plamen Kirilov, and Fabrice Pirot
23 Liposomal Gels in Enhancing Skin Delivery of Drugs . . . . . . . 329
Lorena Tavano
Contributors

Rawad Abdayem Faculty of Medicine and Pharmacy, University Lyon 1,


Fundamental, Clinical and Therapeutic Aspects of the Skin Barrier Function,
Lyon, France
Jelena Predic Atkinson 56a Dollis Park, London, UK
Federica Bigucci Department of Pharmacy and Biotechnology-FaBiT,
Bologna University, Bologna, Italy
Marie-Alexandrine Bolzinger Laboratoire d’Automatique et de Génie des
Procédés (LAGEP), CNRS UMR 5007, University of Lyon, Université
Claude Bernard Lyon 1, Villeurbanne, France
Stéphanie Briançon Laboratoire d’Automatique et de Génie des Procédés
(LAGEP), CNRS UMR 5007, University of Lyon, Université Claude
Bernard Lyon 1, Villeurbanne, France
Marc B. Brown Unit 3/ Chancellor Court, MedPharm Ltd., Guildford, UK
School of Pharmacy, University of Hertfordshire, Hatfield, Herts, UK
Teresa Cerchiara Department of Pharmacy and Biotechnology-FaBiT,
Bologna University, Bologna, Italy
Yves Chevalier Laboratoire d’Automatique et de Génie des Procédés
(LAGEP), CNRS UMR 5007, University of Lyon, Université Claude
Bernard Lyon 1, Villeurbanne, France
Dongmei Cun Department of Pharmaceutical Sciences, Shenyang
Pharmaceutical University, Shenyang, Liaoning, China
Nina Dragicevic Apoteka “Beograd”, Belgrade, Serbia
Jeanetta du Plessis Unit for Drug Research and Development,
North-West University, Potchefstroom Campus, Potchefstroom,
South Africa
Adina Eichner Institute of Pharmacy, Martin Luther University
Halle-Wittenberg, Halle (Saale), Germany
Liang Fang Department of Pharmaceutical Sciences, Shenyang
Pharmaceutical University, Shenyang, Liaoning, China

xiii
xiv Contributors

Sarah Fiala Institute of Pharmaceutical Science, King’s College London,


London, UK
Elodie Gilbert Laboratoire de Pharmacie Galénique Industrielle – Faculté
de Pharmacie, Lyon, France
Faculty of Medicine and Pharmacy, University Lyon 1, Lyon, France
Jeffrey E. Grice Therapeutics Research Centre, School of Medicine,
University of Queensland, Princess Alexandra Hospital, Woolloongabba,
QLD, Australia
Marek Haftek Faculty of Medicine and Pharmacy, University Lyon 1,
Fundamental, Clinical and Therapeutic Aspects of the Skin Barrier Function,
Lyon, France
K.R. Jadhav Department of Pharmaceutics, Bharati Vidyapeeth’s College
of Pharmacy, Navi Mumbai, India
Stuart A. Jones Institute of Pharmaceutical Science,
King’s College London, London, UK
V.J. Kadam Department of Pharmaceutics, Bharati Vidyapeeth’s College
of Pharmacy, Navi Mumbai, India
Wesam R. Kadhum Faculty of Pharmaceutical Sciences, Josai University,
Sakado, Japan
Plamen Kirilov Laboratoire de Pharmacie Galénique Industrielle – Faculté
de Pharmacie, Lyon, France
Victoria Klang Department of Pharmaceutical Technology
and Biopharmaceutics, University of Vienna, Vienna, Austria
Rina Kuswahyuning Therapeutics Research Centre, School of Pharmacy
and Medical Sciences, University of South Australia, Adelaide,
SA, Australia
Department of Pharmaceutics, Faculty of Pharmacy, University of Gadjah
Mada, Yogyakarta, Indonesia
Jürgen Lademann Department of Dermatology, Venereology and
Allergology, Charité – Universitätsmedizin Berlin, Center of Experimental
and Applied Cutaneous Physiology, Berlin, Germany
Wing Man Lau Reading School of Pharmacy, University of Reading,
Whiteknights, UK
Jacquelyn Levin, DO Department of Dermatology, Largo Medical Center,
Largo, FL, USA
Thorsteinn Loftsson Faculty of Pharmaceutical Sciences,
University of Iceland, Reykjavik, Iceland
Barbara Luppi Department of Pharmacy and Biotechnology-FaBiT,
Bologna University, Bologna, Italy
Howard I. Maibach Department of Dermatology, University of California
Medical School, UCSF, San Francisco, CA, USA
Contributors xv

Hamid R. Moghimi Therapeutics Research Centre, School of Pharmacy


and Medical Sciences, University of South Australia, Adelaide,
SA, Australia
School of Pharmacy, Shahid Beheshti University of Medical Sciences,
Tehran, Iran
Josefin Mueller Institute of Pharmacy, Martin Luther University
Halle-Wittenberg, Halle (Saale), Germany
Reinhard H.H. Neubert Institute of Pharmacy, Martin Luther University
Halle-Wittenberg, Halle (Saale), Germany
Keng Wooi Ng School of Pharmacy and Biomolecular Sciences,
University of Brighton, Brighton, UK
Lars Norlén Department of Cell and Molecular Biology (CMB),
Karolinska Institutet, Stockholm, Sweden
Dermatology Clinic, Karolinska University Hospital, Stockholm, Sweden
Anja Otto Unit for Drug Research and Development, North-West
University, Potchefstroom Campus, Potchefstroom, South Africa
Alexa Patzelt Department of Dermatology, Venereology and Allergology,
Charité – Universitätsmedizin Berlin, Center of Experimental and Applied
Cutaneous Physiology, Berlin, Germany
Fabrice Pirot Laboratoire de Pharmacie Galénique Industrielle – Faculté
de Pharmacie, Lyon, France
Service Pharmaceutique – Unité de préparation et de contrôle des médicaments,
Groupement Hospitalier Edouard Herriot, Lyon, France
Faculty of Medicine and Pharmacy, University Lyon 1, Fundamental, Clinical
and Therapeutic Aspects of the Skin Barrier Function, Lyon, France
Lakshmi Raghavan Department of Research and Development,
DermPathe Pharmaceuticals, Washington, NJ, USA
Vishwas Rai Department of Research and Development, DermPathe
Pharmaceuticals, Washington, NJ, USA
Michael S. Roberts Therapeutics Research Centre, School of Pharmacy
and Medical Sciences, University of South Australia, Adelaide, SA,
Australia
School of Medicine, University of Queensland, Princess Alexandra
Hospital, Woolloongabba, QLD, Australia
Laurène Roussel Laboratoire de Pharmacie Galénique Industrielle –
Faculté de Pharmacie, Lyon, France
Faculty of Medicine and Pharmacy, University Lyon 1, Fundamental, Clinical
and Therapeutic Aspects of the Skin Barrier Function, Lyon, France
Damien Salmon Laboratoire de Pharmacie Galénique
Industrielle – Faculté de Pharmacie, Lyon, France
Service Pharmaceutique – Unité de préparation et de contrôle des médica-
ments, Groupement Hospitalier Edouard Herriot, Lyon, France
xvi Contributors

Annett Schroeter Institute of Pharmacy, Martin Luther University


Halle-Wittenberg, Halle (Saale), Germany
Julia C. Schwarz Research Platform ‘Characterisation of Drug Delivery
Systems on Skin and Investigation of Involved Mechanisms’, University of
Vienna, Vienna, Austria
I.M. Shaikh Pharmaceutical R&D, GlaxoSmithKline Pharmaceuticals Ltd,
Ambad, Nashik, India
Kenneth B. Sloan Department of Medicinal Chemistry, College of
Pharmacy, University of Florida, Gainesville, FL, USA
Jessica Stahl Department of Pharmacology, Toxicology and Pharmacy,
University of Veterinary Medicine Hannover, Hannover, Germany
Kenji Sugibayashi Faculty of Pharmaceutical Sciences, Josai University,
Sakado, Saitama, Japan
Jennifer Synovec Department of Medicinal Chemistry, College of
Pharmacy, University of Florida, Gainesville, FL, USA
Lorena Tavano Department of Pharmacy, Health and Nutrition Sciences,
University of Calabria, Arcavacata di Rende, Cosenza, Italy
Hiroaki Todo Faculty of Pharmaceutical Sciences, Josai University,
Sakado, Saitama, Japan
Claudia Valenta Department of Pharmaceutical Technology
and Biopharmaceutics, University of Vienna, Vienna, Austria
Research Platform ‘Characterisation of Drug Delivery Systems on Skin
and Investigation of Involved Mechanisms’, University of Vienna, Vienna,
Austria
Scott C. Wasdo Department of Anesthesiology, College of Medicine,
University of Florida, Gainesville, FL, USA
Philip W. Wertz Dows Institute for Dental Research and Department of
Oral Pathology, Radiology and Medicine, University of Iowa,
Iowa City, IA, USA
Honglei Xi Department of Pharmaceutical Sciences, Shenyang
Pharmaceutical University, Shenyang, Liaoning, China
Part I
The Skin
Skin Deep: The Basics of Human
Skin Structure and Drug 1
Penetration

Keng Wooi Ng and Wing Man Lau

Contents 1.1 Introduction


1.1 Introduction. . . . . . . . . . . . . . . . . . . . . . . . . 3
The skin is the heaviest organ of the human body,
1.2 The Skin. . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
1.2.1 Structure of the Skin. . . . . . . . . . . . . . . . . . . 4
on average accounting for 10 % of the body mass
1.2.2 Functions of the Skin . . . . . . . . . . . . . . . . . . 7 and covering nearly 2 m2 of the body surface area
(McGrath et al. 2004; Williams 2003). It defines
1.3 Drug Permeation Through the Skin. . . . . 8
1.3.1 Permeation Pathways . . . . . . . . . . . . . . . . . . 9 the boundary between the body and its surround-
ings, thus allowing vital bodily functions to occur
Conclusion. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
within a controlled physiological environment.
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10 However, the skin is more than just a physical
partition; rather, it provides an important inter-
face through which we interact with the world.
One such interaction takes the form of substance
exchange between the body and the surrounding
environment. Substance exchange is usually
finely regulated by the skin, which possesses
some exceptional properties to enable it to carry
out this function. As a result, the skin is highly
selective as to what it lets into, or out of, the body
and at what rate. This presents a challenge to
drug delivery across the skin into the body, as the
molecules in question are likely to be poorly
absorbed due to low skin permeation.
The overarching subject of this book is on skin
K.W. Ng (*) permeation enhancement. Two key concepts, i.e.
School of Pharmacy and Biomolecular Sciences, the properties of the skin and molecular transport
University of Brighton, Huxley Building, through the skin, are fundamental to a full under-
Lewes Road, Brighton BN2 4GJ, UK
e-mail: K.Ng@brighton.ac.uk
standing of the subject. In this introductory chap-
ter, we describe the basic properties of the skin
W.M. Lau
Reading School of Pharmacy, University of Reading,
and its functions, as well as the mechanisms of
Whiteknights, Reading RG6 6AP, UK skin penetration and permeation, and relate them
e-mail: w.lau@reading.ac.uk to the challenges that may be encountered in

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 3


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_1, © Springer-Verlag Berlin Heidelberg 2015
4 K.W. Ng and W.M. Lau

attempts to deliver molecules through the skin. palm and sole of the foot. However, the stratum
The coverage is intentionally brief, since the aim lucidum is often not considered a distinct epider-
is to provide a basic understanding of the subject mal layer but the lower part of the stratum cor-
area to prepare the reader for more advanced dis- neum. In addition, there are appendageal features
cussions in later chapters of this book. Also, including hair follicles and sweat ducts that tra-
although this chapter describes human skin, it is verse various skin layers.
worth noting that many structural and functional
parallels can be drawn between human skin and 1.2.1.1 Stratum Corneum: The Primary
that of certain other animals. This caveat under- Barrier
pins the use of certain animal skin as in vitro The stratum corneum is the outermost layer of
models for human skin research, as will be appar- the skin. It is typically 10–20 μm thick and com-
ent in some of the later chapters. posed of 10–15 layers of corneocytes (Agache
2004a; Williams 2003). Corneocytes are non-
living cells derived from terminally differenti-
1.2 The Skin ated keratinocytes that have originated from the
deeper layers of the epidermis. Morphologically,
1.2.1 Structure of the Skin corneocytes are flattened and elongated, mea-
suring about 0.2 μm thick and 40–60 μm wide
Human skin is a stratified epithelium, each tis- (Kashibuchi et al. 2002). Corneocytes have a cor-
sue layer consisting of different cell types that nified envelope in place of a plasma membrane,
perform distinct functions. It can be broadly which is surrounded by a lipid coat. They lack
divided into the overlying epidermis, dermis and nuclei and cytoplasmic organelles but are filled
underlying hypodermis (or subcutis) (Fig. 1.1). with keratin filaments and are interspersed in a
The ­epidermis can further be subdivided, from lipid-enriched extracellular matrix that also con-
the outside to the inside, into the stratum cor- tains protein/peptide components (Elias 2012).
neum (horny layer), stratum granulosum (granu- This organisation of the stratum corneum is com-
lar layer), stratum spinosum (prickle cell layer) monly referred to as the ‘brick and mortar’ model
and stratum basale (basal layer also called the (Elias 1983; Michaels et al. 1975), where the cor-
stratum germinativum). The stratum basale and neocytes are likened to bricks and the extracel-
stratum spinosum are collectively known as the lular matrix analogous to the mortar in a brick
Malpighian layer. An additional layer, the stra- wall (Fig. 1.3). Corneocytes are connected by
tum lucidum (clear layer) can be observed on corneodesmosomes and are continuously shed
­
parts of the body with thickened skin, such as the from the skin surface via desquamation.

Hair shaft

Sweat pore
Stratum
corneum
Blood Viable Epidermis
capillaries epidermis
Sensory nerve
Sebaceous endings
gland Dermis
Fig. 1.1  A diagrammatic Sweat duct
representation of the
Sweat gland
structure of human skin in Hair follicle
cross section. The epidermis Adipose tissue Hypodermis
is composed of the stratum
corneum and the viable
epidermis. Diagram
is not to scale Blood vessels Arrector pili muscle
1  Skin Deep: The Basics of Human Skin Structure and Drug Penetration 5

Since diffusion across the stratum corneum is stratum corneum lipids are initially secreted by
considered as the major pathway of skin perme- lamellar bodies found in keratinocytes and later
ation (see Sect. 1.3), the structural properties of fuse to form continuous lipid lamellae in the
this layer have been extensively studied to eluci- extracellular space. This model is supported by
date its barrier function. Significant effort has evidence of lipid membrane extrusion and fusion
been focused on elucidating the role of stratum in the stratum corneum under electron micros-
corneum lipids, since the lipid-enriched extracel- copy (Madison et al. 1987). An alternative model,
lular matrix is the only continuous domain in the the membrane folding model, postulates that the
stratum corneum and thus likely to be pivotal to extracellular lipid lamellae forms from single,
its barrier function (Bouwstra et al. 2002). This is coherent lipid structures through a non-fusogenic
supported by empirical data that demonstrate skin process (Norlén 2001a).
penetration predominantly through the extracel- Ceramides, cholesterol and free fatty acids
lular matrix (Labouta et al. 2011), the marked are the main constituents of the extracellular
reduction in the skin’s barrier function following matrix, present in approximately equimolar pro-
stratum corneum lipid extraction (Sweeney and portions (McGrath et al. 2004). Of these, cerami-
Downing 1970), and that chemicals that disrupt des account for approximately 50 % of stratum
stratum corneum lipid organisation enhance skin corneum lipids by mass (Law et al. 1995).
permeation (Williams and Barry 2004). Eleven classes of ceramides, encompassing 342
Through freeze-fracture electron microscopy, individual ceramide species, have been identi-
Breathnach et al. (1973) first observed that the fied in the human stratum corneum (Masukawa
extracellular space in the stratum corneum was et al. 2008). Each ceramide molecule consists
composed of a ‘laminated material’. It is now of a sphingoid moiety (sphingosine, phyto-
understood that stratum corneum lipids are sphingosine, 6-hydroxysphingosine or dihydro-
organised into layers (lamellae) that run parallel sphingosine) containing a polar head group and
to the flat plane of the corneocytes. According to a hydrocarbon chain and another hydrocarbon
the widely accepted Landmann model of skin chain derived from a fatty acid or fatty acid
barrier lipid morphogenesis (Landmann 1986), ester moiety (Fig. 1.2). The polar head groups

Polar head group Hydrocarbon chains


Polar head group

O Hydrocarbon chain (fatty acid moiety) Hairpin/tuning fork


HN
CER2 HO
OH Hydrocarbon chain
(sphingoid moiety)
Extended/splayed chain

O
HN O
CER1 HO O
OH

Fig. 1.2  Chemical structures of two stratum corneum illustrate its exceptionally long fatty acid ester-derived
ceramides and their possible conformations. Ceramide 2 hydrocarbon chain compared to the typical ceramide mol-
(CER2) is shown as a typical ceramide consisting of a ecule (CER2). The inset shows two possible molecular
polar head group (dotted circle) and two hydrocarbon conformations of ceramides, the ‘hairpin’ or ‘tuning fork’
chains derived from a fatty acid and a sphingoid moiety, conformation, where hydrocarbon chains point in the
respectively. In CER2, the hydrocarbon chain derived same direction, and the extended or ‘splayed chain’ con-
from the fatty acid is 24 carbon atoms long, exemplifying formation, where the hydrocarbon chains point in oppo-
the typical hydrocarbon chain length for this portion of site directions
ceramide molecules. Ceramide 1 (CER1) is shown to
6 K.W. Ng and W.M. Lau

of ceramides can form lateral hydrogen bonds • The model proposed by Swartzendruber et al.
when organised in lattice structures within lipid (1989) describes a repeat unit (Landmann unit)
bilayers. The hydrocarbon chains are mostly sat- of the extracellular lipid lamellae c­ omprising
urated, with few exceptions, and exhibit chain two lipid bilayers. Ceramides in each appos-
length distribution (Bouwstra and Gooris 2010; ing bilayer assume the ‘splayed-chain’ confor-
Masukawa et al. 2008). Notably, those cerami- mation (Fig. 1.2) and, in so doing, contribute
des containing a fatty acid ester moiety have an hydrocarbon chains to form a lipid monolayer
exceptionally long hydrocarbon chain. To illus- between the bilayers that holds the two bilay-
trate this, the typical ceramide contains a total ers together.
of 38–54 carbon atoms, whereas fatty acid ester- • The domain mosaic model (Forslind 1994)
containing ceramides contain a markedly larger describes an intracellular lipid matrix where
number (66–72) of carbon atoms (Masukawa polar lipids are segregated in crystalline
et al. 2008). Like ceramides, the free fatty domains surrounded by liquid crystalline
acids found in the human stratum corneum are ‘grain borders’. It is postulated that the liquid
mostly saturated. Cholesterol fluidises the stra- crystalline ‘grain borders’ provide a diffusion
tum corneum lipid bilayers at skin temperature pathway for hydrophobic molecules to pene-
(Zbytovská et al. 2008). There also exists cho- trate the skin.
lesterol sulphate, typically at 2–5 % weight ratio • The ‘sandwich’ model (Bouwstra et al. 2000,
of total stratum corneum lipids, which appears 2001, 2002) describes alternating crystalline
to facilitate the formation of the lipid lamellae and liquid phases within the LPP. The repeat-
and stabilises the stratum corneum by inhibiting ing unit in this model comprises three lipid
enzymatic degradation of corneodesmosomes layers, namely, a middle, narrow liquid phase
(Sato et al. 1998). sandwiched between two adjacent, broad crys-
The precise molecular arrangement of stratum talline phases. Ceramides are packed into a
corneum lipids within the extracellular matrix crystalline lattice within the broader phases,
remains a subject of intense investigation. Under but the long linoleate moieties in ceramide 1
electron microscopy with ruthenium tetroxide and ceramide 4 protrude beyond the thickness
fixation, the extracellular lipid matrix displays of the crystalline phases into the space between
characteristic, alternating ‘broad-narrow-broad’ the crystalline phases to form the narrow liq-
lucent bands (Madison et al. 1987; Swartzendruber uid phase with cholesterol. It is postulated that
et al. 1989). This trilamellar motif, with a repeat the liquid phase represents the main perme-
distance (periodicity) of approximately 13 nm, is ation pathway within the LPP. In this model,
known as the long periodicity phase (LPP). This ceramides are typically depicted in the ‘hair-
is in contrast with the short periodicity phase pin’ (or ‘tuning fork’) conformation (Fig. 1.2).
(SPP), which has a periodicity of approximately • The single gel phase model (Norlén 2001b)
6 nm but has not been observed in some animal describes the extracellular lipid matrix as a
species. For this reason, the LPP is considered to single and coherent lamellar gel phase, with
be more important for the skin’s barrier function no phase separation.
and thus has been the focus of most investiga- • The model proposed by Hill and Wertz (2003)
tions into stratum corneum lipid organisation. describes three lipid layers of equal thickness
X-ray diffraction analysis has revealed that the within the LPP. According to this model, the
­extracellular lipids are packed into hexagonal or broad-narrow-broad motif observed under
orthorhombic lattices along the plane parallel electron microscopy with ruthenium tetroxide
with the lamellae, orthorhombic packing being fixation is an artefact. It is suggested that
denser than hexagonal. Based on these and other unsaturated linoleate or sphingosine moieties
supporting observations, a number of molecular are asymmetrically distributed in the central
models have been proposed to elucidate stratum lamellae but not the outer lamellae, resulting
corneum lipid organisation: in the reduction of a greater amount of the
1  Skin Deep: The Basics of Human Skin Structure and Drug Penetration 7

fixative agent and hence an apparently nar- the skin and is responsible for its elasticity and
rower central band. strength. It is composed principally of fibroblasts
• The model presented by McIntosh (2003) has in an extracellular matrix of structural proteins,
lamellae composed of twin lipid bilayers, with mainly collagen and elastin. It also contains a
an asymmetric distribution of cholesterol and range of immune cells including macrophages
ceramide 1 in apposing monolayers of each and dermal dendritic cells. The dermis can be
lipid bilayer. subdivided into the upper papillary dermis and
• The model of Schröter et al. (2009) suggested the lower reticular dermis, which can be distin-
that the SPP is formed from lipid bilayers guished microscopically from each other by the
composed of short-chain ceramides, with the thinner and looser packing of collagen fibres in
long-chain ceramide 1 spanning multiple lipid the papillary dermis. The papillary dermis con-
bilayers. Cholesterol is distributed homoge- tains papillae that interdigitate with the basal
nously within the lipid bilayers. layer of the epidermis at the dermo-epidermal
More recent work has shown that ceramides junction. The dermis contains hair follicles, sweat
are fully extended in the stratum corneum, with glands, sebaceous glands, sensory nerve endings,
the two hydrocarbon tails of each ceramide mol- lymphatic vessels and blood capillaries which
ecule pointing in opposite directions centred on extend to the dermal side of the dermo-­epidermal
the polar head of the sphingoid moiety (Iwai junction. This allows nutrient and oxygen deliv-
et al. 2012). ery to, as well as waste removal from, the avas-
cular epidermis to occur by diffusion across the
1.2.1.2 Viable Epidermis dermo-epidermal junction.
Excluding the stratum corneum, the rest of the
epidermis is composed of nucleated cells and 1.2.1.4 Hypodermis
therefore collectively referred to as the viable The hypodermis is the innermost layer of the
epidermis. The viable epidermis is typically skin. However, its absence is notable in some
50–100  μm thick (Gentilhomme and Neveux lean skin, such as that on the eyelid. The hypo-
2004) and devoid of blood capillaries and sen- dermis is composed mainly of subcutaneous fat.
sory nerve endings. It is composed primarily of Embedded in this skin layer are larger lymphatic
keratinocytes (95 %), with the remainder being and blood vessels.
Langerhans cells, melanocytes and Merkel
cells. Keratinocytes arise from the stratum
basale and undergo progressive differentiation 1.2.2 Functions of the Skin
whilst migrating towards the stratum corneum.
Keratinocyte differentiation is characterised by The primary function of the skin is to sepa-
increasing keratinisation (formation of intracel- rate the internal physiological environment of
lular networks of keratin fibres), the formation the body from the external non-physiological
of the lamellar bodies that secrete stratum cor- ­environment. To put it plainly, it serves to ‘keep
neum lipids and the loss of intracellular organ- the insides in, and the outsides out’ (Williams
elles and nuclei. The process culminates in the 2003). The skin barrier is physical, chemical and
formation of corneocytes in the stratum cor- immunological in nature. The physical barrier is
neum. Keratinocyte differentiation serves to provided primarily by the stratum corneum, that
maintain the stratum corneum by replenishing is to say, traversing the stratum corneum is usu-
stratum corneum lipids and corneocytes lost via ally the rate-limiting step in substance exchange
desquamation. between the body and the environment via the
skin. This physical barrier is responsible for regu-
1.2.1.3 Dermis lating not only the ingress of exogenous materials
The dermis, typically ≥1 mm thick (Agache but also preventing excessive water loss from the
2004b; Williams 2003), comprises the bulk of body. The chemical barrier is known as the ‘acid
8 K.W. Ng and W.M. Lau

­ antle’. The skin owes its chemical barrier func-


m constriction of the arrector pili muscle. Perspiration
tion to the acidic (pH 4–6) nature of the skin sur- secreted through sweat pores on the skin surface
face which protects the body in two ways. Firstly, helps reduce body temperature by dissipating heat
it confers selective antimicrobial properties to from the body through the evaporation of water in
the skin by maintaining the natural skin micro- sweat. Blood vessels in the skin dilate or constrict
flora, which live optimally in an acidic environ- to adjust the blood flow and heat loss across the
ment, whilst arresting the growth of pathogenic large skin surface area. These thermoregulatory
microorganisms which thrive in alkaline environ- mechanisms work in concert to help maintain a
ments. Secondly, it helps maintain the integrity constant core body temperature of about 37 °C.
of the stratum corneum barrier since many skin Apart from heat and cold, sensory nerve end-
enzymes pivotal to stratum corneum lipid homeo- ings in the dermis detect touch, vibrations and
stasis (e.g. β-glucocerebrosidase and sphingomy- pain. These sensations are critical to other func-
elinase) have a pH optima within this pH range tions of the body, such as locomotion and coordi-
(Bowser and Gray 1978; Takagi et al. 1999). nation. The ability to sense pain alerts us of
Sebaceous glands in the skin, which secrete danger and is crucial to survival.
sebum, perform a similar function. Following Furthermore, the skin carries out important
its secretion to the skin surface, sebum forms metabolic functions. Adipocytes in the hypo-
a greasy film on the skin, which waterproofs dermis store excess energy in the form of sub-
the skin to maintain hydration and suppleness. cutaneous fat, which can be mobilised rapidly
Sebum also contains antimicrobial constituents. during energy deprivation. The epidermis is
The skin is also an immune-competent organ. the primary site of vitamin D synthesis in the
A range of immune cells including Langerhans body (Bikle 2011). The process, photolysed by
cells, dermal dendritic cells and macrophages ultraviolet irradiation, produces a precursor for
are found in the skin (Zaba et al. 2008). These vitamin D in the stratum spinosum and stratum
cells conduct immune surveillance and defend basale, which is then converted into vitamin D by
the body against invading microorganisms. They keratinocytes.
are antigen-presenting cells capable of priming The skin additionally serves an excretory
naïve T lymphocytes to elicit a primary immune function, as minerals and other organic wastes
response against newly encountered antigens. are released through the skin dissolved in sweat.
This is an important role of the skin considering The hypodermis also provides mechanical pro-
a compromised skin barrier is a common route tection to inner organs by cushioning the body
of pathogen entry into the body. There is also an against physical shock.
increasing body of evidence that supports a role
for some skin dendritic cell subsets in inducing
immune tolerance (Romani et al. 2012), which 1.3  rug Permeation Through
D
is equally important for maintaining immune the Skin
homeostasis.
Moreover, the skin has an important role in The skin is a selectively permeable barrier. As
thermoregulation, allowing thermal energy to be such, different drugs permeate through the skin at
dissipated or conserved. Thermoreceptors in the different rates. The rate of drug permeation is
skin detect heat and cold; they provide sensory expressed as the flux (J), i.e. the amount of drug
input to the hypothalamus, which then invokes a permeated per unit area, per unit time (usually μg
range of thermoregulatory mechanisms to achieve cm−2 h−1). The flux is determined by (a) the
temperature homeostasis. Adipose tissue in the ­permeability of the skin to the permeant and (b)
hypodermis insulates the body from cold and pre- the concentration gradient (∆C) of the permeant
vents excessive heat loss from the body. Body across the skin (usually μg ml−1), according to
hairs on the skin provide additional insulation by Eq. 1.1:
trapping a thin layer of air on the skin surface. This
J = K p ⋅ DC (1.1)
effect is maximised by the erecting of hairs, via
1  Skin Deep: The Basics of Human Skin Structure and Drug Penetration 9

In Eq. 1.1, skin permeability is defined by the 1.3.1 Permeation Pathways


permeability coefficient, Kp (usually cm h−1).
Assuming passive drug absorption, the permea- A molecule can permeate through the skin via
bility coefficient is a combined measure of the either the transepidermal pathway (diffusing
partition coefficient (P, which depicts how read- across the skin layers) or the appendageal pathway
ily the permeant partitions from the formulation (through hair follicles or sweat ducts) (Fig. 1.3).
into the skin), the diffusion coefficient (D, which The combined flux of these two pathways deter-
measures how readily the permeant diffuses mines the overall observed flux across the skin.
through the skin) and the diffusional path length
(h), according to Eq. 1.2: 1.3.1.1 Transepidermal Pathway
In the transepidermal pathway, the permeant tra-
P⋅D
Kp = (1.2) verses the intracellular and/or extracellular spaces,
h from the epidermis to the dermis and hypodermis.
The processes of partitioning and diffusion (and The molecule may do so either transcellularly or
thus skin permeability, according to Eq. 1.2) are intercellularly. The transcellular route requires that
highly dependent on the physicochemical proper- the permeant traverse the alternating layers of cells
ties of the permeant, such as molecular mass and and extracellular matrix. This involves a sequence
hydrophilicity. As a general rule, molecules that of partitioning and diffusion into alternating
permeate the skin most readily have a molecular hydrophilic and lipophilic domains. The cells and
mass of <500 Da and are moderately hydrophilic, substances that comprise the hydrophilic or lipo-
with an octanol-water partition coefficient (log philic domains vary between skin layers, but gen-
Poctanol–water) of 1–3. The quantitative relationship erally the interiors of cells are more hydrophilic
between skin permeability (defined by Kp), than the extracellular matrix. In the intercellular
molecular mass (MW) and hydrophilicity route, the permeant navigates the tortuous path
(defined by log Poctanol–water) is widely described within the extracellular matrix, without traversing
using Eq. 1.3 (Potts and Guy 1992): the cells. Small hydrophilic molecules generally
favour the transcellular route over the intercellular
log K p = 0.71 ⋅ log Poctanol − water − 0.0061 ⋅ MW − 2.74 route and vice versa for lipophilic molecules.

(1.3)
1.3.1.2 Appendageal Pathway
Other factors that may influence skin permeation The appendageal (or shunt) pathway encom-
include hydrogen bond activity, molecular vol- passes permeation through hair follicles (the
ume, melting point and solubility. Other mathe- transfollicular route) or sweat ducts. The
matical models have been devised to relate the transfollicular route has gained significant
­
role of these parameters to skin permeation research interest in recent years and is covered in
(Magnusson et al. 2004; Moss et al. 2002). a separate chapter (Chap. 5).

Appendage
Drug formulation Corneocyte
(hair follicle,
sweat duct) (‘brick’)

Stratum
corneum

Extracellular matrix
(a) (b) (c) (‘mortar’)

Fig. 1.3  Drug permeation pathways in the skin (stratum The transcellular and intercellular routes constitute the
corneum shown): (a) the appendageal route, (b) the transepidermal pathway
­transcellular route, and (c) the tortuous extracellular route.
10 K.W. Ng and W.M. Lau

1.3.1.3 Relative Contributions relationships that permit accurate prediction of


of Permeation Pathways drug permeation profiles. However, the chal-
It is widely accepted that the transepidermal lenge remains that the majority of drugs do not
pathway is usually the predominant pathway of exhibit ­satisfactory skin permeation and inno-
skin permeation and that under sink conditions, vative strategies are needed to enhance their
diffusion across the stratum corneum constitutes uptake via the skin. Many such innovations are
the rate-limiting step that determines the overall described in detail within the following special-
flux of the permeant. The contribution of the ised chapters of this book.
appendageal pathway to percutaneous transport
is generally considered secondary, since append-
ageal features typically account for only around
0.1 % of skin surface area (though this is higher References
at some body sites such as the forehead), and
Agache P (2004a) Metrology of the stratum corneum. In:
early studies suggested that the spatial density of Agache P, Humbert P (eds) Measuring the skin: non-­
appendages did not correlate with the flux of per- invasive investigations, physiology, normal constants.
meants across the skin (Scheuplein 1967). Springer, Berlin, pp 101–111
Nonetheless, the relative contribution of these Agache P (2004b) The human skin: an overview. In:
Agache P, Humbert P (eds) Measuring the skin: non-­
pathways will vary depending on the physico- invasive investigations, physiology, normal constants.
chemical properties of the permeant and the for- Springer, Berlin, pp 3–5
mulation. Highly lipophilic drugs may be retained Bikle DD (2011) Vitamin D, metabolism and function in
in the lipophilic stratum corneum and resist parti- the skin. Mol Cell Endocrinol 347:80–89
Bouwstra JA, Gooris GS (2010) The lipid organization in
tioning into the more hydrophilic viable epider- human stratum corneum and model systems. Open
mis. Thus, clearance from, rather than diffusion Derm J 4:10–13
across, the stratum corneum may then become Bouwstra JA, Dubbelaar FE, Gooris GS, Ponec M (2000)
the rate-limiting step for highly lipophilic drugs. The lipid organisation in the skin barrier. Acta Derm
Venereol Suppl (Stockh) 208:23–30
Similarly, the appendageal pathway may be more Bouwstra JA, Gooris GS, Dubbelaar FER, Ponec M
important for highly hydrophilic molecules such (2001) Phase behavior of lipid mixtures based on
as caffeine (Trauer et al. 2010) and electrolytes, human ceramides: coexistence of crystalline and liq-
as well as large molecules with low diffusion uid phases. J Lipid Res 42:1759–1770
Bouwstra JA, Gooris GS, Dubbelaar FE, Ponec M
coefficients which are thus effectively precluded (2002) Phase behavior of stratum corneum lipid mix-
from the transpidermal pathway. The relative tures based on human ceramides: the role of natural
importance of each pathway may also change and synthetic ceramide 1. J Invest Dermatol 118:
with time – various studies have shown that the 606–617
Bowser PA, Gray GM (1978) Sphingomyelinase in pig
appendageal pathway rapidly but transiently pre- and human epidermis. J Invest Dermatol 70:331–335
dominates before being overtaken by the tran- Breathnach AS, Goodman T, Stolinski C, Gross M (1973)
sepidermal pathway at steady state (Liu et al. Freeze-fracture replication of cells of stratum corneum
2011; Saar et al. 2011; Scheuplein 1967). of human epidermis. J Anat 114:65
Elias PM (2012) Structure and function of the stratum
corneum extracellular matrix. J Invest Dermatol
Conclusion 132:2131–2133
The skin owes its barrier properties primarily to Elias PM (1983) Epidermal lipids, barrier function, and
the stratum corneum. The unique lipid compo- desquamation. J Invest Dermatol 80(Suppl):44s–49s
Forslind B (1994) A domain mosaic model of the skin
sition and organisation within the stratum cor- barrier. Acta Derm Venereol 74:1–6
neum are key determinants of skin permeability, Gentilhomme E, Neveux Y (2004) Epidermal physiology.
which has important implications for drug per- In: Agache P, Humbert P (eds) Measuring the skin:
meation through the skin. Painstaking research non-invasive investigations, physiology, normal con-
stants. Springer, Berlin, pp 165–172
in the last few decades has elucidated the mech- Hill J (2003) Molecular models of the intercellular lipid
anisms of drug transport through this highly lamellae from epidermal stratum corneum. Biochim
effective barrier and generated ­structure-activity Biophys Acta BBA – Biomembr 1616:121–126
1  Skin Deep: The Basics of Human Skin Structure and Drug Penetration 11

Iwai I, Han H, den Hollander L, Svensson S, Öfverstedt L-G, Norlén L (2001a) Skin barrier formation: the membrane
Anwar J et al (2012) The human skin barrier is orga- folding model. J Invest Dermatol 117:823–829
nized as stacked bilayers of fully extended ceramides Norlén L (2001b) Skin barrier structure and func-
with cholesterol molecules associated with the ceramide tion: the single gel phase model. J Invest Dermatol
sphingoid moiety. J Invest Dermatol 132:2215–2225 117:830–836
Kashibuchi N, Hirai Y, O’Goshi K, Tagami H (2002) Potts RO, Guy RH (1992) Predicting skin permeability.
Three-dimensional analyses of individual corneocytes Pharm Res 9:663–669
with atomic force microscope: morphological changes Romani N, Brunner PM, Stingl G (2012) Changing views
related to age, location and to the pathologic skin con- of the role of Langerhans cells. J Invest Dermatol
ditions. Skin Res Technol 8:203–211 132:872–881
Labouta HI, El-Khordagui LK, Kraus T, Schneider M Saar BG, Contreras-Rojas LR, Xie XS, Guy RH (2011)
(2011) Mechanism and determinants of nanopar- Imaging drug delivery to skin with stimulated raman
ticle penetration through human skin. Nanoscale scattering microscopy. Mol Pharm 8:969–975
3:4989–4999 Sato J, Denda M, Nakanishi J, Nomura J, Koyama J
Landmann L (1986) Epidermal permeability barrier: (1998) Cholesterol sulfate inhibits proteases that are
transformation of lamellar granule-disks into intercel- involved in desquamation of stratum corneum. J Invest
lular sheets by a membrane-fusion process, a freeze-­ Dermatol 111:189–193
fracture study. J Invest Dermatol 87:202–209 Scheuplein RJ (1967) Mechanism of percutaneous
Law S, Wertz PW, Swartzendruber DC, Squier CA (1995) absorption. II. Transient diffusion and the relative
Regional variation in content, composition and organi- importance of various routes of skin penetration.
zation of porcine epithelial barrier lipids revealed by J Invest Dermatol 48:79–88
thin-layer chromatography and transmission electron Schröter A, Kessner D, Kiselev MA, Hauß T, Dante S,
microscopy. Arch Oral Biol 40:1085–1091 Neubert RHH (2009) Basic nanostructure of stratum
Liu X, Grice JE, Lademann J, Otberg N, Trauer S, Patzelt corneum lipid matrices based on ceramides [EOS]
A, Roberts MS (2011) Hair follicles contribute signifi- and [AP]: a neutron diffraction study. Biophys J
cantly to penetration through human skin only at times 97:1104–1114
soon after application as a solvent deposited solid in Swartzendruber DC, Wertz PW, Kitko DJ, Madison KC,
man: hair follicles contribute to early human skin pen- Downing DT (1989) Molecular models of the intercel-
etration. Br J Clin Pharmacol 72:768–774 lular lipid lamellae in mammalian stratum corneum.
Madison KC, Swartzendruber DC, Wertz PW, Downing J Invest Dermatol 92:251–257
DT (1987) Presence of intact intercellular lipid lamel- Sweeney TM, Downing DT (1970) The role of lipids in
lae in the upper layers of the stratum corneum. J Invest the epidermal barrier to water diffusion. J Invest
Dermatol 88:714–718 Dermatol 55:135–140
Magnusson BM, Pugh WJ, Roberts MS (2004) Simple Takagi Y, Kriehuber E, Imokawa G, Elias PM, Holleran
rules defining the potential of compounds for transder- WM (1999) Beta-glucocerebrosidase activity in mam-
mal delivery or toxicity. Pharm Res 21:1047–1054 malian stratum corneum. J Lipid Res 40:861–869
Masukawa Y, Narita H, Shimizu E, Kondo N, Sugai Y, Trauer S, Lademann J, Knorr F, Richter H, Liebsch M,
Oba T et al (2008) Characterization of overall ceramide Rozycki C et al (2010) Development of an in vitro
species in human stratum corneum. J Lipid Res modified skin absorption test for the investigation of
49:1466–1476 the follicular penetration pathway of caffeine. Skin
McGrath JA, Eady RAJ, Pope FM (2004) Anatomy and Pharmacol Physiol 23:320–327
organization of human skin. In: Rook A, Burns T (eds) Williams A (2003) Transdermal and topical drug delivery
Rook’s textbook of dermatology, 7th edn. Blackwell from theory to clinical practice. Pharmaceutical Press,
Science, Malden London
McIntosh TJ (2003) Organization of skin stratum Williams AC, Barry BW (2004) Penetration enhancers.
­corneum extracellular lamellae: diffraction evidence Adv Drug Deliv Rev 56:603–618
for asymmetric distribution of cholesterol. Biophys J Zaba LC, Krueger JG, Lowes MA (2008) Resident and
85:1675–1681 “inflammatory” dendritic cells in human skin. J Invest
Michaels AS, Chandrasekaran SK, Shaw JE (1975) Drug Dermatol 129:302–308
permeation through human skin: theory and in vitro Zbytovská J, Kiselev MA, Funari SS, Garamus VM,
experimental measurement. AIChE J 21:985–996 Wartewig S, Palát K et al (2008) Influence of choles-
Moss GP, Dearden JC, Patel H, Cronin MTD (2002) terol on the structure of stratum corneum lipid model
Quantitative structure–permeability relationships membrane. Colloids Surfaces Physicochem Eng Asp
(QSPRs) for percutaneous absorption. Toxicol In Vitro 328:90–99
16:299–317
Epidermal Lipids and the
Intercellular Pathway 2
Philip W. Wertz

Contents 2.1 Lipid Alteration


2.1 Lipid Alteration with Keratinization . . . . 13 with Keratinization
2.2 Lamellar Granules . . . . . . . . . . . . . . . . . . . . 14
The pioneering work of G. Maurice Gray and
2.3 Intercellular Lamellae . . . . . . . . . . . . . . . . . 15 Harold Yardley and their associates (Gray
2.4 Covalently Bound and Yardley 1975a, b; Gray et al. 1978a, b;
ω-Hydroxyceramide . . . . . . . . . . . . . . . . . . 16 Gray and White 1978; Yardley and Summerly
2.5 Penetration Pathways . . . . . . . . . . . . . . . . . 16 1981) done during the mid- to late 1970s dem-
onstrated that lipids associated with epidermal
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
keratinocytes increased in amount and altered
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17 dramatically in composition as a function of dif-
ferentiation. Thus, the relatively undifferentiated
basal keratinocytes contain about 7 pg of lipid
per cell consisting of small amounts of choles-
terol and phospholipids including sphingomyelin,
phosphatidyl choline, phosphatidyl ethanolamine,
phosphatidyl inositol, and phosphatidyl serine.
The more mature granular cells contain about
324 pg of lipid per cell, and this includes more
cholesterol and phospholipids of the same types,
but in addition, there are now also significant
amounts of glucosylceramides, ceramides, and
fatty acids. Finally, the terminally differentiated
stratum corneum cells contain about 3,576 pg of
lipid consisting mostly of ceramides, cholesterol,
P.W. Wertz and fatty acids. There are also small proportions
Dows Institute for Dental Research and Department of cholesterol esters and cholesterol sulfate in the
of Oral Pathology, Radiology and Medicine,
cornified layer, but the ceramides, cholesterol, and
University of Iowa, N450 DSB,
Iowa City, IA 52242, USA fatty acids are thought to be mainly responsible
e-mail: Philip-wertz@uiowa.edu for the permeability barrier of the skin.

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 13


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_2, © Springer-Verlag Berlin Heidelberg 2015
14 P.W. Wertz

2.2 Lamellar Granules shown that these organelles are rich in phospho-
glycerides, sphingomyelin, glucosylceramides,
Much of the lipid that accumulates with increas- and cholesterol. Fatty acids, cholesterol esters,
ing differentiation in epidermis is packaged into and ceramides were minor lipid components. The
small organelles known as keratinosomes most abundant of the glucosylceramides is a
(Wilgram 1965), cementsomes (Hashimoto structurally unusual linoleate-containing acylglu-
1971), Odland bodies (Oashi et al. 1973), cosylceramide (Wertz et al. 1984; Madison et al.
membrane-coating granules (Hayward 1974), 1998). This consists of 30- through 34-carbon
lamellar bodies (Elias and Friend 1975), or lamel- ω-hydroxyacids amide-linked to sphingosine (and
lar granules (Landmann 1986). Lamellar gran- dihydrosphingosine) with linoleate ester-linked to
ules are round to ovoid in shape, are about 0.2 μm the ω-hydroxyl group (Wertz and Downing 1983;
in diameter, and consist of a unit bounding mem- Abraham et al. 1985). Linoleate has long been
brane surrounding one, or sometimes several, known to be required for proper formation and
stacks of internal lamellae (Elias and Friend maintenance of the permeability barrier of the
1975; Landmann 1986; Wertz 2000). Several skin, and it has been proposed that this unusual
lamellar granules are shown in Fig. 2.1. In the sphingolipid is directly involved in this require-
uppermost cells of the granular layer, the lamellar ment (Wertz and Downing 1982). Based on exten-
granules migrate to the apical end of the cell, sive electron microscopic studies, Landmann has
where the bounding membrane of the lamellar proposed that the internal lamellae of the lamellar
granule fuses into the cell plasma membrane and granules are stacks of flattened lipid vesicles
the lamellar granule contents are extruded into (Landmann 1986). It was proposed that in assem-
the intercellular space. This not only delivers lip- bly of these stacks of flattened vesicles, the long
ids, which are thought to be flattened vesicles, to ω-hydroxyacyl chain of the acylglucosylceramide
the intercellular space, but also a battery of completely spans one region of bilayer, while the
hydrolytic enzymes are delivered by this mecha- linoleate inserts into an adjacent region of bilayer,
nism (Freinkel and Traczyk 1985; Grayson et al. thus riveting the two sections of bilayer together
1985; Madison et al. 1998). at a molecular level. In support of this suggestion,
Since lamellar granules are lipid-rich, they it was shown that acylglucosylceramide does
have a low buoyant density. This unique property cause flattening and aggregation of synthetic lipid
has been exploited to isolate lamellar granules vesicles. Lamellar granules and acylglucosylce-
(Freinkel and Traczyk 1985; Wertz et al. 1984; ramides have been found in keratinizing oral epi-
Grayson et al. 1985; Madison et al. 1998; Sando thelium, but neither the organelle nor the unusual
et al. 2003). Direct chemical analyses of isolated sphingolipid are present in nonkeratinizing oral
lamellar granules (Freinkel and Traczyk 1985; epithelium or other tissues. Likewise, lamellar
Wertz et al. 1984; Grayson et al. 1985) have granules and acylglucosylceramides have been

Fig. 2.1 Transmission


electron micrograph of
several epidermal lamellar
granules. Bar = 200 nm
2 Epidermal Lipids and the Intercellular Pathway 15

found in epidermis of a number of terrestrial fatty acid mixture that is found in the intercellular
mammals, birds, and reptiles, but both are absent spaces of the stratum corneum. It is this intercel-
from epidermis of fish and amphibians, where lular lipid that determines the permeability bar-
there is no stratum corneum. rier of the skin, under normal circumstances
The hydrolytic enzymes delivered to the inter- (Wertz 2000).
cellular space by lamellar granules, in general,
have low pH optima like lysosomal hydrolases
(Freinkel and Traczyk 1985; Wertz et al. 1984; 2.3 Intercellular Lamellae
Grayson et al. 1985; Madison et al. 1998). The
enzymes detected include acid phosphatase, aryl Among the enzymatic actions that occur at the
sulfatase, galactosidase, galactosaminidase, glu- boundary between the granular layer and the stra-
cosidase, glucosaminosidase, phospholipase A, tum corneum are the deglycosylation of gluco-
sphingomyelinase, carboxypeptidase, cathepsin sylceramides and the hydrolysis of sphingomyelin
B, acid lipase, and ceramide glucosyltransferase. to produce ceramides. Representative structures
Of these enzymes, the specific activities in the of the ceramides found in porcine stratum cor-
isolated lamellar granules were lower than found neum are presented in Fig. 2.2, along with the
in crude homogenates for arylsulfatase (both A ceramide nomenclature system proposed by
and B) as well as sterol sulfatase (Grayson et al. Motta and colleagues (Wertz and Downing
1985). The lipid-hydrolytic enzymes are impor- 1983b; Motta et al. 1993). In the Motta nomen-
tant in converting the initially extruded lamellar clature system, one letter is used to indicate the
granule lipids into the ceramide, cholesterol, and type of fatty acid (N for normal fatty acid, A for

O
O O
HN
OH Cer[EOS]
OH

O
HN Cer[NS]
OH
OH

O
HN
OH Cer[NP]
OH
OH
HO
O
HN Cer[AS]
OH
OH

HO
O
HN Cer[AS]
OH
OH
HO
Fig. 2.2 Structures of stratum O
corneum ceramides and a HN Cer[AP]
proposed system for nomen- OH
clature (Wertz and Downing OH
1983; Motta et al. 1993) OH
16 P.W. Wertz

α-hydroxyacid and O for ω-hydroxyacid), one The lateral packing of the lipids in normal
letter is used to indicate the long-chain base (S human stratum corneum is predominantly ortho-
for sphingosine, P for phytosphingosine, and H rhombic (crystalline) with small amounts of hex-
for 6-hydroxysphingosine), and the presence of agonal packing (gel) (Pilgram et al. 1998; de
an ester-linked fatty acid is indicated by a prefix Jager et al. 2004). In some skin diseases in which
E. Thus, the ceramide-containing saturated fatty lipid composition is altered and barrier function
acids amide-linked to sphingosine bases can be is diminished, such as atopic dermatitis and
designated as Cer[NS]. The unusual ceramide at lamellar ichthyosis, the hexagonal phase
the top of Fig. 2.2 is an acylceramide or Cer[EOS]. increases relative to orthorhombic phase (Pilgram
Most of the ceramides are cylindrical in shape, et al. 2001).
which favors formation of highly ordered, and
thereby impermeable, membrane domains. The
unusual Cer[EOS] is thought to play a central 2.4 Covalently Bound
role in formation of 13 nm trilaminar lipid struc- ω-Hydroxyceramide
tures (Madison et al. 1987; Kuempel et al. 1998;
Groen et al. 2010). In these trilaminar structures, It is thought that approximately two-thirds of the
it has been proposed that the ω-hydroxyacyl por- acylglucosylceramide molecules associated with
tion of Cer[EOS] spans the outer layers, while the lamellar granules are present in the bounding
the linoleates insert into the central lamella (Hill membrane with the glucosyl moiety at the inside
and Wertz 2003). With this arrangement, the surface of the granule (Wertz 2000). When the
outer two lamellae are highly saturated, while the bounding membrane of the lamellar granule fuses
central lamella contains all of the double bonds into the cell plasma membrane, this inverts the
from the linoleate chains. This stabilizes the tri- orientation of the acylglucosylceramide. The glu-
laminar structures into 13 nm units that have cose is then removed, and two stereoselective
been seen using transmission electron micros- lipoxygenase attacks on the linoleate chain pre-
copy with ruthenium tetroxide fixation (Madison cede its removal and transfer of the resulting
et al. 1987; Kuempel et al. 1998) and with X-ray ω-hydroxyceramide to the outer surface of the
diffraction (Groen et al. 2010). One consequence cornified envelope (Zheng et al. 2011). The
of this arrangement is that the central lamella will attachment of the ω-hydroxyceramide to the cor-
reduce more ruthenium than the outer lamellae. nified envelope is through ester-linkages that
This results in alternating broad-narrow-broad may be produced through the action of a
lucent bands in the electron micrographs, as can transglutaminase (Nemes et al. 1999). Thus, the
be seen in Fig. 2.3. stratum corneum becomes an array of flat keratin-
filled cells bounded by a cornified envelope with
a monolayer of covalently bound lipids on the
outer surface embedded in a multilamellar array
of free lipids.

2.5 Penetration Pathways

In a classic review, Scheuplein and Blank con-


sidered potential pathways through which small
molecules could pass through the stratum cor-
neum (Scheuplein and Blank 1971). These
included a paracellular route through the inter-
Fig. 2.3 Transmission electron micrograph of intercel- cellular spaces, a transcellular route, follicular
lular lamellae in the stratum corneum. Bar = 80 nm penetration, and entry through the sweat ducts.
2 Epidermal Lipids and the Intercellular Pathway 17

For human skin, they argued that entry through Conclusions


sweat pores would be insignificant due to the Epidermal keratinocytes differentiate to pro-
small fraction of the skin surface occupied by duce the stratum corneum which consists of
such openings. Although a similar argument flat, keratin-filled cells bounded by a complex
pertained to follicular penetration, it was cornified envelope and embedded in a matrix
acknowledged that under some conditions fol- of lamellar lipid. The lipids in the intercellular
licular penetration could be significant. The spaces of the stratum corneum consist mainly
follicular route has some advantages for drug of a structurally heterogenous group of
delivery in that it can accommodate nanoparti- ceramides, cholesterol, and long, saturated
cles and microparticles with potential slow fatty acids. These lipids are organized into
drug release (Knorr et al. 2009; Lademann trilamellar structures that, under passive con-
et al. 2011). A major question was whether ditions, provide the primary permeability bar-
interfollicular stratum corneum was amenable rier of the skin. There are a number of physical
to transcellular penetration, paracellular pene- methods that are able to bypass this barrier,
tration, or some combination of the two. This which should prove useful in development of
remained a point of contention until 1980, transdermal drug delivery.
when Nemanic and Elias (1980) visualized the
intercellular pathway followed by N-butanol
diffusing across stratum corneum by in situ
precipitation combined with transmission elec- References
tron microscopy. The penetration pathway
Abraham W, Wertz PW, Downing DT (1985) Linoleate-
for this molecule was exclusively via the rich acylglucosylceramides from pig epidermis: struc-
paracellular route. Squier and Lesch (1988) ture determination by proton magnetic resonance.
subsequently demonstrated an exclusively para- J Lipid Res 26:761–766
cellular route for both polar and nonpolar small Ahad A, Aqil M, Kohli K, Chaudhary H, Sultana Y,
Mujeeb M, Talegaonkar S (2009) Chemical penetra-
molecules using microaudioradiography. tion enhancers: a patent review. Expert Opin Ther Pat
So for passive diffusion of drugs across the 19:969–988
stratum corneum, the paracellular route is con- Coulman S, Allender C, Birchall J (2006) Microneedles
sidered the predominant pathway. Chemical and other physical methods for overcoming the stra-
tum corneum barrier for cutaneous gene therapy. Crit
permeability enhancers are thought to act pri- Rev Ther Drug Carrier Syst 23:205–258
marily by fluidizing the intercellular lamellae De Jager MW, Gooris GS, Dolbnya IP, Ponec M, Bouwstra
of the stratum corneum, thereby reducing diffu- JA (2004) Modelling the stratum corneum lipid orga-
sional resistance (Thong et al. 2007; Ahad et al. nization with synthetic lipid mixtures: the importance
of synthetic ceramide composition. Biochim Biophys
2009). Some physical means of enhancing drug Acta 1664:132–140
delivery may also alter the intercellular lamellae; Dixit N, Bali V, Baboota S, Ahuja A, Ali J (2007)
however some physical enhancement methods Iontophoresis – an approach for controlled drug deliv-
do alter pathways. For example, microneedle ery: a review. Curr Drug Deliv 4:1–10
Elias PM, Friend DS (1975) The permeability barrier in
arrays create direct channels across the stratum mammalian epidermis. J Cell Biol 65:180–191
corneum (Coulman et al. 2006). Iontophoresis Freinkel RK, Traczyk TN (1985) Lipid composition and
may effectively deliver drugs through sweat acid hydrolase content of lamellar granules of fetal rat
ducts (Dixit et al. 2007). Sonophoresis may epidermis. J Invest Dermatol 85:295–298
Gray GM, White RJ (1978) Glycosphingolipids and
both alter the physical state of the intercellular ceramides in human and pig epidermis. J Invest
lamellae and create transcellular pathways (Rao Dermatol 70:336–341
and Nanda 2009). Electroporation increases the Gray GM, Yardley HJ (1975a) Lipid composition of cells
permeability of the stratum corneum by open- isolated from pig, human and rat epidermis. J Lipid
Res 16:434–440
ing or creating aqueous channels, through which Gray GM, Yardley HJ (1975b) Different populations of
relatively large water soluble molecules can pass pig epidermal cells: isolation and lipid composition.
(Singh et al. 2012). J Lipid Res 16:441–447
18 P.W. Wertz

Gray GM, White RJ, Majer JR (1978a) 1-(3′-O-acyl)- long-chain omega-hydroxyceramides to involucrin by
beta-glucosyl-N-dihydroxypentatriacontadienoylsphi ester bond formation. Proc Natl Acad Sci U S A
ngosine, a major component of the glucosylceramides 96:8402–8407
of pig and human epidermis. Biochim Biophys Acta Oashi M, Sawada Y, Makita R (1973) Odland body and
528:127–137 intercellular substances. Acta Derm Venereol Suppl
Gray GM, King IA, Yardley HJ (1978b) The plasma 73:47–54
membrane of granular cells from pig epidermis: isola- Pilgram GS, Engelsma-van Pelt AM, Oostergetel GT,
tion and lipid and protein composition. J Invest Koerten HK, Bouwstra JA (1998) Study on the lipid
Dermatol 71:131–135 organization of stratum corneum lipid models by
Grayson S, Johnson-Winegar AG, Wintroub BU, Isseroff (cryo-) electron diffraction. J Lipid Res 39:1669–1676
RR, Epstein EH Jr, Elias PM (1985) Lamellar body- Pilgram GS, Vissers DC, van den Meulen H, Pavel S,
enriched fractions from neonatal mice: preparative Lavrijsen SP, Bouwstra JA, Koerten HK (2001)
techniques and partial characterization. J Invest Aberrant lipid organization in stratum corneum of
Dermatol 85:289–294 patients with atopic dermatitis and lamellar ichthyosis.
Groen D, Gooris GS, Bouwstra JA (2010) Model mem- J Invest Dermatol 117:710–717
branes prepared with ceramide EOS, cholesterol and Rao R, Nanda S (2009) Sonophoresis: recent advancements
free fatty acids form a unique lamellar phase. and future trends. J Pharm Pharmacol 61:689–705
Langmuir 26:4168–4175 Sando GN, Zhu H, Weis JM, Richman JT, Wertz PW,
Hashimoto K (1971) Cementsome, a new interpretation of Madison KC (2003) Caveolin expression and location
the membrane-coating granule. Arch Dermatol Forsch in human keratinocytes suggests a role in lamellar
240:349–364 granule biogenesis. J Invest Dermatol 126:531–541
Hayward AF (1974) Proceedings: membrane-coating Scheuplein RJ, Blank IH (1971) Permeability of the skin.
granules are secondary lysosomes. J Anat 118:364 Physiol Rev 51:702–747
Hill JR, Wertz PW (2003) Molecular models of the inter- Singh N, Kalluri H, Herwadkar A, Badkar A, Banga AK
cellular lipid lamellae from epidermal stratum cor- (2012) Transcending the skin barrier to deliver pep-
neum. Biochim Biophys Acta 1616:121–126 tides and proteins using active technologies. Crit Rev
Knorr F, Lademann J, Patzelt A, Sterry W, Blume-Peytavi Ther Drug Carrier Syst 29:265–298
U, Vogt A (2009) Follicular transport route – research Squier CA, Lesch CA (1988) Penetration pathways of dif-
progress and future perspectives. Eur J Pharm ferent compounds through epidermis and oral epithe-
Biopharm 71:173–180 lia. J Oral Pathol 17:512–516
Kuempel D, Swartzendruber DC, Squier CA, Wertz PW Thong HY, Zhai H, Maibach HI (2007) Percutaneous pen-
(1998) In vitro reconstruction of stratum corneum etration enhancers: an overview. Skin Pharmacol
lipid lamellae. Biochim Biophys Acta 1372:135–140 Physiol 20:272–282
Lademann J, Richter H, Schanzer S, Knorr F, Meinke M, Wertz PW (2000) Lipids and barrier function of the skin.
Sterry W, Patzelt A (2011) Penetration and storage of Acta Derm Venereol Suppl 208:7–11
particles in human skin: perspectives and safety Wertz PW, Downing DT (1982) Glycolipids in mamma-
aspects. Eur J Pharm Biopharm 77:465–468 lian epidermis: structure and function in the water bar-
Landmann L (1986) Epidermal permeability barrier: rier. Science 217:1261–1262
transformation of lamellar granule-disks into intercel- Wertz PW, Downing DT (1983a) Ceramides of pig
lular sheets by a membrane-fusion process, a freeze- epidermis: structure determination. J Lipid Res
fracture study. J Invest Dermatol 87:202–209 24:759–765
Madison KC, Swartzendruber DC, Wertz PW, Downing Wertz PW, Downing DT (1983b) Acylglucosylceramides
DT (1987) Presence of intact intercellular lipid lamel- of pig epidermis: structure determination. J Lipid Res
lae in the upper layers of the stratum corneum. J Invest 24:753–758
Dermatol 88:714–718 Wertz PW, Downing DT, Freinkel RK, Traczyk TN (1984)
Madison KC, Sando GN, Howard EJ, True CA, Gilbert D, Sphingolipids of the stratum corneum and lamellar
Swartzendruber DC, Wertz PW (1998) Lamellar gran- granules of fetal rat epidermis. J Invest Dermatol
ule biogenesis: a role for ceramide glucosyltransfer- 83:193–195
ase, lysosomal enzyme transport and the Golgi. Wilgram G (1965) The keratinosome: a factor in the kera-
J Invest Dermatol Symp Proc 3:80–86 tinization process of the skin. Hautzart 16:377–379
Motta S, Monti M, Sesana S, Caputo R, Carelli S, Ghidoni Yardley HJ, Summerly R (1981) Lipid composition and
R (1993) Ceramide composition of the psoriatic scale. metabolism in normal and diseased epidermis.
Biochim Biophys Acta 1182:147–151 Pharmacol Ther 13:357–383
Nemanic MK, Elias PM (1980) In situ precipitation: Zheng Y, Yin H, Boeglin WE, Elias PM, Crumrine D, Beier
a novel cytochemical technique for visualization DR, Brash AR (2011) Lipoxygenases mediate the
of permeability pathways in mammalian stratum effect of essential fatty acid in skin barrier formation:
corneum. J Histochem Cytochem 28:573–578 a proposed role in releasing omega-hydroxyceramide
Nemes Z, Marekov LN, Fesus L, Steinert PM (1999) for construction of the cornified lipid envelope. J Biol
A novel function for transglutaminase 1: attachment of Chem 286:24046–24056
The Importance of Stratum
Corneum Lipid Organization 3
for Proper Barrier Function

Annett Schroeter, Adina Eichner, Josefin Mueller,


and Reinhard H.H. Neubert

Contents 3.4  ffect of Short-Chain Ceramides


E
to the Structural Organization
3.1 Introduction. . . . . . . . . . . . . . . . . . . . . . . . . 19 of Stratum Corneum Lipids. . . . . . . . . . . . 30
3.1.1 Ceramides of the Stratum 3.4.1 Elucidation of the Nanostructure
Corneum Lipid Matrix . . . . . . . . . . . . . . . . . 20 of Stratum Corneum Lipid Models Based
3.2  tratum Corneum Lipid Nanostructure
S on CER[AP] or CER[NP]. . . . . . . . . . . . . . . 31
Investigated With Neutron Diffraction. . . 20 3.4.2 Investigating the Nanostructure
3.2.1 Basic Principles of Neutron of an Stratum Corneum Substitute. . . . . . . . 35
Diffraction. . . . . . . . . . . . . . . . . . . . . . . . . . . 20 3.5 Summary and Final Remarks. . . . . . . . . . 35
3.2.2 Investigation of Stratum Corneum
Lipid Model Membranes References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
with Neutron Diffraction. . . . . . . . . . . . . . . . 22
3.2.3 Advantages and Disadvantages
of Neutron Diffraction . . . . . . . . . . . . . . . . . 23
3.2.4 X-Ray Diffraction for the Investigation 3.1 Introduction
of the Stratum Corneum Lipids. . . . . . . . . . . 25
3.3 ω
 -acyl Chain Ceramides Obstacles such as the complexity and chemical
and Their Influence on the 
Nanostructure of the Stratum
variability of the lipids present in the stratum
Corneum Lipid Matrix. . . . . . . . . . . . . . . . 26 corneum (SC), disturbing other material like pro-
3.3.1 Physicochemical Aspects. . . . . . . . . . . . . . . 26 teins, ethical issues related to the use of excised
3.3.2 Long-Chain ω-acyl Ceramides human provided by biological material like
Studied Using Native Stratum Corneum. . . . 27
3.3.3 Synthetically Constructed
excised skin, hinder elucidating the molecular
Long-Chain ω-acyl Ceramides. . . . . . . . . . . 28 morphology of the SC lipid matrix. These diffi-
culties led to an increasing use of synthetic SC
lipids in SC research. To allow for a systematic
evaluation of the relevance of single ceramide
(CER) species, multilamellar model membranes
containing simplistic mixtures of synthetic SC
A. Schroeter (*) • A. Eichner
lipids represent a suitable approach as shown in
J. Mueller • R.H.H. Neubert numerous previous works (Kessner et al. 2008a,
Institute of Pharmacy, Martin Luther University b; Kiselev 2007: Kiselev et al. 2005; Wegener
Halle-Wittenberg, Halle (Saale), Germany et al. 1997; Zbytovska et al. 2009). Such a sys-
e-mail: annett.schroeter@pharmazie.uni-halle.de;
adina.eichner@pharmazie.uni-halle.de;
tematic determination of the impact of particu-
josefin.mueller@pharmazie.uni-halle.de; lar CER subclasses is important for a detailed
reinhard.neubert@pharmazie.uni-halle.de understanding of mechanisms in skin diseases.

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 19


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_3, © Springer-Verlag Berlin Heidelberg 2015
20 A. Schroeter et al.

This knowledge supports the development of new acid. In line with the way of labeling, these
therapeutic approaches. In addition, enhanced ceramides received the letter E (=esterified). Up
understanding of the function of different SC lip- to now, 12 major CER classes have been identi-
ids in the process of barrier formation helps to fied within the SC lipid matrix (Holleran et al.
develop new carrier systems being able to over- 2006; Masukawa et al. 2008) (see Fig. 3.1), but
come the penetration barrier more efficiently. detailed information about their specific role for
the barrier formation and function of the SC
needs to be elucidated. Especially the long-chain
3.1.1 Ceramides of the Stratum CER[EOS], [EOP] and [EOH] are discussed to
Corneum Lipid Matrix be of particular relevance because of their unique
structure. As mentioned above these CER have in
It is generally known that the main constituents, addition to the amidation either a phytosphingo-
the ceramides (CER), play a key role in the struc- sine (P), sphingosine (S), or hydroxysphingosine
turing and hence the maintenance of the barrier (H) base, while the ω-hydroxylated fatty acid is
function of the skin (Coderch et al. 2003; Holleran esterified with a linoleic acid (EO) in ω-position
et al. 1991). They are a group of structurally het- (Coderch et al. 2003). Due to the esterified fatty
erogeneous sphingolipids and consist of a long-­ acid, those CER have a chain length of 30–32
chain fatty acid bound to the amino group of a carbon atoms (Raith et al. 2004). However, in
long-chain di- or trihydroxy sphingoid base recent years also the short-chain CER such as
(sphingosine, phytosphingosine, and 6- CER[AP] and CER[NP] have been in the focus
hydroxysphingosine). The bound fatty acid of the of various investigations, and it became evident
CER consists predominantly of a very long that these CER seem to play a more important
almost entirely saturated alkyl chain (Wertz et al. role in the structural organization of the upper-
1987) and can be hydroxylated at the α-position most skin barrier than previously assumed. But, it
to the carbonyl oxygen, at the end of the hydro- is most likely the broad distribution of alkyl chain
carbon chain (ω-position), or it contains no fur- lengths and the heterogeneity in the head groups
ther hydroxyl group (Coderch et al. 2003). The which guarantee the integrity and functionality of
first nomenclature used to label the different CER the lipid lamellae of the SC (Norlen 2001).
was based on their mobility in the thin-layer
chromatography (Wertz and Downing 1983). As
the number of the identified CER increased, this 3.2 Stratum Corneum Lipid
method of labeling was insufficient. Consequently, Nanostructure Investigated
the nomenclature of the CER is based on their With Neutron Diffraction
chemical structure and was developed by Motta
and coworkers (Motta et al. 1993). In this system 3.2.1 Basic Principles of Neutron
the ceramides are labeled with letters, whereby Diffraction
the last letter assigns the type of sphingoid base
(S… sphingosine, P… phytosphingosine, H… The SC research comprises many biophysical
6-hydroxysphingosine). The long-chain fatty approaches such as Fourier transform infrared
acid bound to the amino group can be differenti- spectroscopy, differential scanning calorimetry,
ated due to their hydroxylation. This was taken atomic force microscopy or nuclear magnetic
up in the nomenclature as amid-bound fatty acids resonance spectroscopy. Among those the scat-
without a hydroxyl group were labeled with the tering techniques X-ray and especially neutron
letter N (=non-hydroxy), while ceramides with diffraction are very potent methods to investigate
an omega- and alpha-hydroxylated fatty acid the structure of isolated SC (Bouwstra et al. 1991)
receive either the letter O or A, respectively. as well as SC model membranes constructed
Furthermore, within the group of ceramides, the from extracted and synthetically derived SC lip-
exceptionally long-chain ω-acyl ceramides exist, ids (Bouwstra et al. 1991, 1996, 1998; Friberg
which are esterified with a long unsaturated fatty and Osborne 1987; Kuempel et al. 1998; McIntosh
3  The Importance of Stratum Corneum Lipid Organization for Proper Barrier Function 21

O OH
O O CH3 O CH3
HO NH NH
HO
HO CH3
Ceramide [EOS] (CER 1) Ceramide [AS] (CER 5)
HO CH3

O OH
O CH3 O CH3
O
HO NH HO NH
HO CH3
Ceramide [EOdS] HO CH3 Ceramide [AdS]

O OH
O CH3 O CH3
O
HO NH
HO
Ceramide [EOH] (CER 4) HO CH3 Ceramide [AH] (CER 8)
HO CH3
OH OH
O OH
O CH3 O CH3
O
NH HO NH
HO Ceramide [EOP] (CER 9)
HO CH3 HO CH3 Ceramide [AP] (CER 6)
OH OH

O CH3 O CH3
HO NH NH
HO
HO CH3 Ceramide [NS] (CER 2)
HO CH3
OH Ceramide [NH] (CER 7)
O CH3 O CH3
HO NH NH
HO
HO CH3 Ceramide [NdS] HO CH3 Ceramide [NP] (CER 3)
OH

Fig. 3.1  Chemical structures of the ceramides found in O ω-hydroxy fatty acid, E esterified, D dihydro. In addi-
the human stratum corneum. S Sphingosine, P tion the old nomenclature according to the mobility of the
Phytosphingosine, H 6-Hydroxysphingosine, N non- CER in the thin layer chromatography was included for
hydroxy fatty acid, A alpha-hydroxy fatty acid, clarification

2003; McIntosh et al. 1996). Both neutron and This makes neutron diffraction a particular
X-ray diffraction are similar techniques, with the valuable instrument to investigate structural and
exception of the irradiation source. While X-rays dynamic features especially in biological sam-
primarily interact with the electrons of an atom, ples, which are rich in hydrogen. Moreover,
the interaction of neutrons with the atomic neutrons show isotope sensitivity, i.e., even dif-
nucleus is short-ranged. To explain the several ferent isotopes of one element may have differ-
advantages of neutron over X-ray diffraction a ent scattering power for neutrons, for which
brief explanation of these methods is necessary. hydrogen (1H) and deuterium (2H, D) are the
The technique of neutron diffraction is a versa- most prominent examples (see Fig. 3.2) (Dachs
tile method to study the structure and dynamics, 1978). The possibility to distinguish between
which specifically applies to biological samples. components differing in their ability to scatter
Due to their specific properties, neutrons may pro- neutrons in one single sample, the so-called
vide structural insights that are hardly obtained by neutron contrast, is of great advantage for the
other techniques, e.g., X-ray or light scattering. study of biological systems like lipids and pro-
As non-charged particles, neutrons are enabled to teins (Büldt et al. 1978; Gutberlet et al. 2001;
penetrate matter deeply due to the small probabil- Tomita et al. 1999).
ity of interaction (Harroun et al. 2006). In contrast In a typical scattering experiment, a well-­
to X-rays, which are scattered by the electron collimated neutron beam with a defined wave-
cloud, neutrons interact with the atomic nucleus length λ irradiates a sample, whereby the neutrons
and are scattered isotropically (Dachs 1978). are scattered in all directions depending on the
Hence, while the ability of elements to scatter interactions between the sample material and the
X-rays increases with the atomic number through- neutrons. The incoming neutrons interact with
out the periodic table of elements, such a correla- the sample and thereby experience a change in
tion does not exist for neutrons (Cantor and their momentum, which appears as a change of
Schimmel 1980). Particularly hydrogen, a light neutron direction and/or velocity. Consequently,
atom that is almost invisible for X-rays, is a strong monitoring the alterations of the neutron’s
scatterer for neutrons (see Fig. 3.2). momentum provides information regarding the
22 A. Schroeter et al.

X-ray relative scattering length Neutron scattering length for individual isotope

H C O Ti Fe U

H C O Ti Fe U 1 12 16 46 54 233

2 13 17 47 56 234

3 18 48 57 233

Fig. 3.2  Schematic comparison of the X-ray relative scattering lengths and neutron scattering lengths of different ele-
ments and their isotopes

structure and dynamics of the sample matter. To scattering direction; the interpretation of the data
describe the change in momentum, the so-called offers information about the structure of the
momentum transfer vector, or scattering vector ­analyzed sample.

Q , was introduced and is defined as the differ-
ence between incoming ki and scattered k s wave
  
vectors Q = k − k . In addition
 to a change in 3.2.2 I nvestigation of Stratum
i s
direction, the magnitude of k can also change as Corneum Lipid Model
energy is transferred between incident neutrons Membranes with Neutron
and sample. When no energy is conveyed, the Diffraction
scattering process
 is considered to be totally
 elas-
tic; therefore, ki has to be equal to k s . Taking The initial biological materials, analyzed with

this in account,
 the scattering vector Q can be neutron diffraction, were phospholipids due to
evaluated as Q = 2ki ⋅ sin q , including the Bragg the ability to form stable and highly organized
angle, which in case of crystalline and lamellar multilamellar lipid bilayers necessary for neutron
material appears at Q values equivalent to the diffraction experiments. In recent years this
 2p ­technique has also been successfully introduced
reciprocal spacing of the lattice: Q = , where into the elucidation of the structural arrangement
d of the stratum corneum lipids. The application of
by d denotes the characteristic spacing of a set of neutron diffraction offers new possibilities to
crystal planes. The complete Bragg formula investigate the nanostructure of SC lipid systems
λ= 2d/sin θ can be received when the wave vector and especially to gain information about the
ki is appropriately substituted with ki = 2p / l . impact of the different CER subspecies as shown
Diffraction can be considered as a special type by Charalambopoulou and coworkers on fully
of scattering, whereby an organized structure hydrated human SC (Charalambopoulou et al.
such as a crystal or a lamellar arrangement is ana- 2002) and by Kiselev et al. on well-defined SC
lyzed. According to Bragg’s law, the incident lipid model membranes (Kiselev et al. 2005).
beams are diffracted at a defined angle 2θ, and
due to the interference between the waves, scat- 3.2.2.1 Evaluation of the Neutron
tered from the parallel planes, diffraction occurs Diffraction Data
as depicted in Fig. 3.3. As mentioned above, the scattering process is
The neutron scattering experiment now mea- assumed as an elastic event with no energy
sures the scattering intensity I as a function of the transfer taking place. Consequently, the scattering
3  The Importance of Stratum Corneum Lipid Organization for Proper Barrier Function 23

Fig. 3.3  Schematic drawing of the scattering process difference between the waves interfering constructively is
from ordered material. (Left) Neutrons strike an array of equal to 2d sin θ. (Right) A typical experimental setup of

atoms (spheres) from the left side and are scattered to the a neutron diffraction experiment, whereby ki designates
right. The planes of atoms are separated by the interplanar 
the incoming wave vector, while ks represents the scat-
distance d. The angle θ to the plans of atoms of the inci-
tered neutron wave vector
dent and the scattered beam are identical. The path length

 rounding atmosphere, the so-called contrast


vector Q can be correlated to the scattering angle
2θ. Furthermore, the intensity of the scattered neu- ­variation (Wiener and White 1991), assuming that
tron is measured as a function of the scattering water can penetrate between the bilayer sheets
angle 2θ. As the Bragg condition is complied, the (Franks and Lieb 1979; Worcester 1976). It was
integrated intensities can be calculated by using shown for such symmetrical and hydrated bilayers
Gaussian fits to the received Bragg reflections. In that the phase problem of Fhs simplifies to the
order to gain deeper insight into the nanostructural determination of the sign of + or – (Franks and
arrangement of the SC lipids, it is necessary to Lieb 1979) and can be derived from the slope of
compute the absolute value of the structure factors the correlation of Fh against the D2O content in
Fh from the integrated peak intensities: water vapor as shown in Fig. 3.4.
The NSLD profile offers detailed information
FH = Ah (q ) ⋅ h ⋅ I h with Lorentz correction h
and absorption factor Ah (θ) (Franks and Lieb about the nanostructure of the investigated lipid
1979). The structure factor Fh serves as a mathe- membrane and can also contribute to assign the
matical description in which mode the incoming position and orientation of the bilayer constitu-
neutron wave is scattered by the investigated mate- ents. Furthermore, the evaluation of the NSLD
rial (Franks and Lieb 1979; Nagle and Tristram- profile allows for determining specific membrane
Nagle 2000a, b). The SC lipid multilamellar layers regions such as the polar head groups, CH3
are composed of numerous bimolecular lipid groups, hydrocarbon chain region, and the region
membranes, which in other terms can be described of cholesterol location (Kiselev et al. 2005). Next
as two equal monolayers facing each other. Such to the assignment of the position of these groups
stacks of lipid layers are considered centrosym- in the lipid bilayer membrane, the determination
metric for the neutron diffraction experiment, of parameters such as the region of polar head
which allows for the construction of the neutron group or thickness of the intermembrane space
scattering length density (NSLD) profile ρs(x) further improves and intensifies the knowledge
across the bilayer as Fourier transform about such SC lipid organization.

2 hmax  2 ⋅p ⋅ x 
rs ( x ) = ∑ Fh ⋅ 
d h =1  d 

3.2.3 Advantages and
Disadvantages of Neutron
(Nagle and Tristram-Nagle 2000b). In order to cal- Diffraction
culate the NSLD profile, it is essential to define the
sign of the structure factor Fh. This can be easily As up to now a clear and detailed picture of the
done by variation of the D2O/H2O ratio in the sur- organization of the SC lipid matrix on a molecu-
24 A. Schroeter et al.

Fig. 3.4  Illustration of the dependency of the membrane structure factor Fh of the orders h = 1, 2, 3, 4 and 5 on the D2O
content on the water vapor a SC lipid model system

lar scale has not been elucidated, it is of supreme (see Fig. 3.2). This special feature renders the
importance to comprehend the mode of action of possibility of the contrast variation as described
the different SC lipid classes and particular the before. In the same line, there is another distinct
impact of the different ceramide subspecies. For advantage of the neutron diffraction technique,
the investigation of the driving forces and mech- which is the possibility of specific deuteration, as
anisms that govern the self-assembling process the coherent scattering length bcoh (the scattering
of such lipid layers, the native SC lipid mem- ability) of hydrogen (1H) and its isotope deute-
branes are too complex objects to probe, espe- rium (2H) differs significantly (bcoh(1H) = − 3.741
cially with neutron diffraction. Consequently, fm,  bcoh(2H) = 6.671 fm). Accordingly, hydrogen
for such an approach, model membranes will be atoms in a lipid molecule can be specifically
the objects of choice. Moreover, issues due to ­substituted by deuterium, which does not alter
the variability of the native lipids, for example, the properties of the lipid molecule. When a par-
the variability in the head group architecture, can tially deuterated lipid sample is compared to its
be overcome. protonated counterpart, it is possible to identify
Neutrons as irradiation source are non- the exact position of the labeled group within the
charged particles; they have only small interac- lipid membrane (see Fig. 3.5). This is a distinct
tion potential with matter, which enables a deep advantage of n­ eutron diffraction over X-ray dif-
penetration into the studied material. This makes fraction, which does not allow for such localiza-
this method particularly suitable for biological tion. As the SC lipid molecules are rich in
issues such as the investigation of the structural hydrogen atoms, there is a variety of substitution
arrangement of the SC lipids. In addition, as positions, which then permit to make distinction
mentioned before the neutrons are scattered dif- between different conformational states of the
ferently by different isotopes of the same element studied lipid species. This feature is of high inter-
3  The Importance of Stratum Corneum Lipid Organization for Proper Barrier Function 25

Fig. 3.5  Example of the localization of partially deuter- (dashed line) or protonated BA (solid line). Dotted lines:
ated behenic acid (BA) molecules (d22BA) in a SC lipid corresponding errors, Long dash: difference NSLD pro-
model membrane composed of ceramide [AP], choles- file, Fat solid line: fit of the difference NSLD profile by
terol, d22BA and cholesterol sulfate. The neutron scatter- two Gaussian functions (deuterium distribution). All mea-
ing length density (NSLD) profiles display the comparison surements were carried out at 57 % relative humidity, at
of the sample membrane containing either deuterated 8 % D2O in water vapor and T = 20 °C

est, especially for the investigation of SC lipids, 3.2.4 X-Ray Diffraction


as it is known for CER to exhibit different con- for the Investigation
formational states (Dahlen and Pascher 1972, of the Stratum Corneum
1979; Raudenkolb et al. 2003a, b, 2005). Lipids
The disadvantages are the limiting factors for
application of the neutron diffraction for explora- As mentioned before, X-ray diffraction has been
tion of the SC lipid matrix. When compared to widely used for the investigation of the structural
X-rays the neutron fluxes are relatively small, arrangement of the SC lipids. So Hatta and
which necessitates a much longer experimental coworkers could establish the impact of ethanol
timescale and a higher amount of lipid material on the lipid membranes. They discovered by
to achieve a reasonable signal-to-noise ratio. X-ray diffraction, that lipid compounds can be
Furthermore, when native SC is probed with extracted and even recrystallized as well (Hatta
neutron diffraction, this yields to only one or two et al. 2001). The investigations of Kessner et al.
diffraction orders, which are not sufficient for the could establish by X-rays that the phase behavior
analysis of the NSLD profile (Charalambopoulou of the long-chain CERs is effected by their long
et al. 2002). Consequently only SC lipid model acyl rests (Kessner et al. 2010).
systems can be studied. Compared to other scattering techniques, it has
Another drawback of the neutron diffraction a variety of advantages. In contrast to the above-
technique is its availability, as there exist only a described neutron diffraction technique, X-rays
few neutron facilities at which such an experi- are scattered by the electrons surrounding the
ment can be carried out (e.g., Helmholtz Centre atomic nuclei. This results in peaks of high inten-
Berlin for Material and Energy (HZB) and Institut sity and high resolution. Furthermore, many X-ray
Laue-Langevin (ILL), Grenoble, France). sources for such experiments are accessible.
26 A. Schroeter et al.

Nevertheless, as described above the major key role with regard to some skin diseases. For
drawback of the application of X-ray in the field atopic dermatitis there was found a decrease
of SC research is the low capability to depict light especially in the CER[EOS] content as proposed
atoms such as hydrogen, which is one of the main by Yamamoto and coworkers (Yamamoto et al.
components of a biological relevant material. 1991), whereas psoriatic skin among others is
Furthermore, as the number of electrons between thought to be caused by an increase in the
different isotopes of the same element does not CER[EOS] amount (Motta et al. 1994).
change, X-rays cannot distinguish isotopes. Its
application as an irradiation source does not allow
for the evaluation of the sign of the structure fac- 3.3.1 Physicochemical Aspects
tor, which is essential in order to be able to calcu-
late the scattering length density profile to gain In addition to the amidation of the sphingosine
deeper insight into the arrangement of the lipid backbone, the ω-hydroxylated fatty acid is at its
bilayer. When the lamellar thickness of a lipid ω-position mainly esterified with a linoleic acid
membrane is changed by way of varying the (EO) (Coderch et al. 2003). Nevertheless, Hinder
thickness of the water layer, it is possible to eval- and coworkers identified for CER [EOS] differ-
uate the sign of the structure factor and conse- ent amid-bound fatty acids with chain lengths
quently calculate the electron density profile. varying from 17 to 22 carbon atoms (Hinder et al.
However, it is well known that the repeat dis- 2011). The same chain length ranging was found
tances of SC lipid mixtures prepared from cerami- for the sphingosine part. Therefore, it was con-
des (CER), cholesterol (CHOL), and free fatty cluded that not only linoleic acid as fatty acid
acids (FFAs) are very insensitive to hydration and compound is esterified to the sphingosine back-
that especially for the short periodicity phase bone. Subsequently, the influence of these differ-
(SPP), only a limited number of diffraction orders ent chain lengths was studied by de Sousa Neto
are obtained with X-ray diffraction. Therefore, et al. with small-angle X-ray scattering and
again it is difficult to acquire an electron density Fourier transform infrared spectroscopy (de
profile by X-ray diffraction analysis. Sousa Neto et al. 2011). Their results show an
important influence of chain length on the lipid
organization. Whereas linoleate- and oleate-­
3.3 ω-acyl Chain Ceramides and linked fatty acid CER[EOS] showed both the for-
Their Influence on the mation of the LPP and SPP, the stearate-linked
Nanostructure of the Stratum variety did not form the LPP. Hence, unsaturated
Corneum Lipid Matrix amid-bound fatty acids seem to be crucial for the
formation of the LPP.
The lipids of the SC and particularly the cerami- Due to the high mobility of the exceptionally
des (CER) are responsible to uphold the proper long-chain ω-acyl residue, these CER form less
barrier function as pointed out in the ­introduction. ordered structures than it is known for short-­chain
The CER are a very heterogeneous group of CER (Raudenkolb 2002). Additionally, the melting
sphingolipids (see Fig. 3.1), which can roughly points being 86 °C for CER[EOS] and above 100 °C
be divided into two groups: (1) short-chain CER for CER[EOP], respectively, hardly differ from
such as CER[AP] or CER[NP] and (2) the excep- those found for short-chain CER as stated by
tionally long-chain ω-acyl CER such as Kessner and coworkers (Kessner et al. 2010). The
CER[EOS] or CER[EOP]. So far, there has been discrepancy in melting points of both ω-acyl chain
a general consensus that especially the long-­ CER was argued to be due to the more polar head
chain ω-acyl CER seem to be of particular rele- group structure of CER[EOP] (2 OH groups for
vance because of their unique structure. CER[EOS] versus 3 OH groups for CER[EOP]),
Besides the assumed importance of long-chain which enables this CER to create more hydrogen
ω-acyl CER in forming the so-called long-­ bonds. In contrast to their expectations, Kessner and
periodicity phase (LPP), their presence plays a coworkers could additionally ascertain that the ther-
3  The Importance of Stratum Corneum Lipid Organization for Proper Barrier Function 27

Molecular arrangement of long periodicity phase (12–13 nm)


of mixtures prepared with
pig ceramides

EI. density
.7
.5

4.57
.3

0.432
.1
ro

12.2 nm
2.37
1

4.57
3
5
7

CER1 CER CHOL

Fig. 3.6  Schematic presentation of the molecular arrangement of the long-periodicity phase (LPP). Reprinted from
Bouwstra et al. (2001a) with permission from Karger Publisher

motropic phase behavior of these CER is reversible mixtures of CER, cholesterol (CHOL), and free
and the melting process does not induce major mod- fatty acids (FFAs) with regard to a diminish-
ifications of the lipids (Kessner et al. 2010). This is ing content of CER[EOS] by X-ray and elec-
in line with other discoveries, which found the polar tron diffraction studies (Bouwstra et al. 2001a).
head group architecture to be responsible for poly- According to their assumption, the fraction of
morphism (Raudenkolb et al. 2003a, b, 2005). lipids forming the LPP decreases by reducing
the amount of CER[EOS]. Thus, they concluded
that the presence of the long-chain ω-acyl CER
3.3.2 Long-Chain ω-acyl Ceramides is necessary for the formation of the LPP and
Studied Using Native Stratum subsequently for a proper barrier function of the
Corneum SC lipid matrix. Resulting from different electron
diffraction studies, the LPP is described as a trila-
As there is a high demand to comprehend the struc- mellar broad-narrow-broad arrangement of the
tural arrangement of the different ­components of SC lipids with a membrane thickness or repeat
the SC lipid, especially, the long-chain CER were distance of 13 nm (Madison et al. 1987; White
supposed to be of great influence. Consequently, et al. 1988). Based on these insights Bouwstra
the focus of many researchers was first placed on et al. developed the sandwich model, depicted in
these exceptionally long-chain ω-acyl CER with Fig. 3.6. According to this model the liquid sub-
their extraordinary characteristics concerning SC lattice consists of CHOL and the linoleic acid res-
lipid organization. For example, Bouwstra et al. idues of the long ω-acyl CER[EOS], [EOP], and
stated the importance of CER[EOS] not only for [EOH], which are located in the center of this tril-
the formation of the LPP but also for the forma- amellar structure and encompass nearly 3 nm of
tion of a liquid phase which enables molecules the total 13 nm thickness of the LPP. The adjacent
to permeate along the lipid layer (Bouwstra et al. crystalline phases with a broadness of 5 nm on
2002). In this context the same group analyzed either side are composed of long saturated hydro-
28 A. Schroeter et al.

carbon chains of the CER and FFA (Bouwstra fraction. Additionally, they concluded that there
et al. 2001a). Except the research of McIntosh is a close connection with the formation of the
et al. (1996), in which isolated CER from native LPP and the presence of CER[EOS]. They stated
pig epidermis were employed, all studies men- that, while partial replacement of CER[EOS] by
tioned above directly used native SC derived from CER[EOP] does not influence phase behavior,
a pig or mouse tissue for their investigations. complete substitution leads to a phase separation
But as pig CER differ structurally from CER of CER[EOP] and a reduction of the LPP.
originated from human tissue, Bouwstra and Another study performed by Kessner and
coworkers compared their previous results of coworkers also employed synthetically derived
mixtures containing pig CER with those contain- CER and applied both X-ray diffraction and
ing human CER (Bouwstra et al. 2001b). Similar Fourier transform Raman spectroscopy in order
to pig CER/CHOL mixtures, human CER/CHOL to ascertain these findings. Contrary to the previ-
mixtures showed the formation of the LPP with ously described investigation, the ­physicochemical
the difference, that by addition of FFA the LPP behavior of only the synthetic long ω-acyl
disappeared and has been replaced by a short CER[EOS] and [EOP] (Kessner et al. 2010) was
periodicity phase (SPP) (Bouwstra et al. 2001b). studied without FFA or CHOL. These investiga-
Contrary to these findings various work groups tions revealed remarkable insights in this regard.
could not detect an LPP by cryoelectron micros- While CER[EOS] in dry state only arranges in a
copy (Al-Amoudi et al. 2005; Pfeiffer et al. 2000) SPP, CER[EOP] already forms the LPP. It was
and X-ray diffraction (Garson et al. 1991) in deduced that the differences in the head group
human SC. They explained the discovery of the architectures are responsible as this is the only dis-
LPP in former researches as an artifact due to similarity between both CER species. They argued
the fixation with Ruthenium tetroxide. Ruthenium that the additional hydroxyl group in CER[EOP]
tetroxide is necessary for the electron diffraction is responsible for more hydrogen bonds and there-
and might yield to the misinterpretation of the fore enables the formation of a high-ordered pack-
received data. However, the fixation problem age preventing the ω-acyl chains extending into
does not account for the X-ray diffraction results. the adjacent bilayer. Once both CER are hydrated,
they are able to form the LPP with a repeat dis-
tance of 12 nm (Kessner et al. 2010).
3.3.3 Synthetically Constructed To further examine the significance of these
Long-Chain ω-acyl Ceramides findings for the arrangement of the SC lipids,
model membranes containing synthetic long-­
There are several disadvantages when native SC chain ω-acyl CER, CHOL, and FFA were ana-
lipids are used. For instance, the variability in lyzed. Using X-ray diffraction, mixtures of
chain length of either the FFA or the CER-bound different CER, CHOL, and the FFA palmitic acid
fatty acids and differences in the CER head (PA) in an equimolar ratio were studied by de
group architecture (see Fig. 3.1) can circum- Jager et al. (2003). In mixtures containing
vent the assignment of different characteristics CER[EOS]/CHOL/PA no LPP could be detected,
to individual lipids, especially the CER sub- while the same mixture comprising additionally
classes. In recent years synthetically constructed CER[NP] showed a clear LPP with a repeat dis-
CER with defined chemical structures have been tance of 11.6 nm. The authors concluded that not
introduced into the SC lipid research, enabling a only the presence of one single CER subclass can
more reliable transduction of the physicochemi- induce the formation of the LPP but a mixture of
cal behavior to the structural characteristics of particular CER (de Jager et al. 2003).
special CER. Subsequently, Kessner and coworkers ana-
This was taken into consideration by de Jager lyzed lipid mixtures containing CER[EOS],
et al. (2004), who confirmed the existence of the CER[AP], and CHOL. They detected a lamellar
LPP by investigating synthetic CER in mixtures membrane with a thickness of two opposing
with CHOL and FFA using small-angle X-ray dif- CER[AP] molecules of approximately 45 Å indi-
3  The Importance of Stratum Corneum Lipid Organization for Proper Barrier Function 29

Fig. 3.7 Schematic
presentation of the arrange- CER[AP]
ment of CER[EOS] in the
model matrix composed of CHOL
CER[EOS]/CER[AP]/CHOL/
BA (23/10/33/33, w/w)
according to Schroeter et al.
CER[EOS]
(2008)

Behenic
acid

48.3 A°

Fig. 3.8  Chemical structures a O


of the native CER[EOS] O
O
specie (A) and the synthe- HN OH
sized CER[EOS]_branched
variety according to OH

Engelbrecht et al. (2011)


b O
O
O
H 3C HN OH
OH

cating that no LPP was formed (Kessner et al. et al. 2008). In another approach by Engelbrecht
2008a). Even by adding of the FFA behenic acid and coworkers, an artificial derivative of
(Schroeter et al. 2008), which was often reported CER[EOS], the so-called CER[EOS]_branched
to be required for the formation of the LPP (de with a methyl-branched and saturated ω-acyl
Jager et al. 2003), only a slight increase of the chain, was synthesized and investigated in a
repeat distance to 48 Å was perceived. model membrane applying neutron diffraction
Furthermore, from the neutron scattering length (Engelbrecht et al. 2011) (see Fig. 3.8). This
density profile, it was concluded that the long molecule is less sensitive to oxidative stress and
ω-acyl chain of CER[EOS] protrudes into the therefore more stable and easier to handle as the
adjacent layer in order to fit into the membrane native specie.
size created by CER[AP]. Consequently, To assure the comparability of the native
CER[EOS] is positioned inside a phase with a CER[EOS] and the artificial CER[EOS]_
short periodicity by spanning a bilayer and branched derivative both Fourier transform
extending into adjacent layer (see Fig. 3.7). Raman spectroscopy and differential scanning
It was concluded that the polar short-chain calorimetry were carried out, with the outcome
CER[AP] plays a key role in the formation of that both species show a comparable phase
this lipid system. It dictates the arrangement of and chain packing behavior (Engelbrecht et al.
the other lipids within this mixture in a lamellar 2011). In order to elucidate the arrangement of
membrane, which covers the range of two oppos- this CER[EOS] derivative with respect to the
ing CER[AP] molecules. Consequently, the dis- previously described findings, Engelbrecht et al.
tinct head group polarity of CER[AP] exceeds additionally studied a model membrane system
the influence of the long ω-acyl chain of composed of CER[EOS]_branched, CER[AP],
CER[EOS] (Kessner et al. 2008a; Schroeter CHOL, and behenic acid with neutron diffrac-
30 A. Schroeter et al.

tion (Engelbrecht et al. 2011). They found that containing CER[EOS], CHOL, and different
the synthetically derived CER[EOS]_branched FFA in an equimolar ratio, they could detect a
is able to serve as an appropriate substitute for 14.7 nm lamellar phase. This very long repeat
the native CER[EOS] in terms of lipid arrange- distance was discussed to result from two oppos-
ment and architecture. Even this more stable and ing CER[EOS] molecules with interdigitating
saturated acyl chain of CER[EOS]_branched linoleate residues. Accordingly they proposed a
did not induce a formation of the LPP in the model arrangement in which the lamellae are
presence of CER[AP], as stated above for the divided into three different lipid layers referred to
non-branched species in such mixtures. Again, as (A) (containing the linoleate residues of
the protruding influence of the more polar CER[EOS]), (B) (composing CHOL and
CER[AP] induces the formation of the SPP. To CER[EOS]-ω-bound fatty acids), and (C)
further verify their results, molecular dynamic (buildup of sphingosine r­esidues and FFA) as
simulation was performed and confirmed the depicted in Fig. 3.10. Although a long 14.7 nm
lamellar arrangement of this model membrane lamellar phase could be observed, the LPP with a
(see Fig. 3.9). repeat distance of 13 nm was not detected.
Contrary to these findings there are the results Concluding, it has to be stated that the role of
from X-ray diffraction and FT-IR studies con- long-chain ω-acyl CER both in context with the
ducted by Groen et al. (2010). For mixtures formation of the LPP or in terms of skin diseases
is not completely elucidated and still debated
vigorously. Therefore, these CER are still a sub-
ject of great interest in SC lipid research.

3.4  ffect of Short-Chain


E
Ceramides to the Structural
Organization of Stratum
Corneum Lipids
Fig. 3.9  Snapshot from molecular dynamic simulation of
the lipid system consisting of CER[EOS]_branched/
CER[AP]/behenic acid/CHOL (23/10/ 33/33 m/m) and As outlined previously, the heterogeneous class
H2O. Color code: orange: CER[EOS]_branched, blue of CER can be generally separated into two major
CER[AP]. Modified according to Engelbrecht et al. (2011) subclasses: the very long-chain ω-acyl CER like

8 % D2O

2.5
rs(X), a.u.

2.0

Fig. 3.10  Model calculations 1.5


polar head
of the neutron SLD profile of groups
a SC lipid system composed 1.0
of CER[AP], CHOL, behenic
acid (BA), and cholesterol cholesterol 0.5
sulfate modified according to
Ruettinger et al. (2008). The 0.0
fitted curves for each group -21 -18 -15 -12 -9 -6 -3 0 3 6 9 12 15 18 21
are: polar head groups (blue -0.5 x, Å
dash), CH2-groups (green), CH2 chains
CH3-groups (orange), -1.0 CH3 groups
cholesterol (red)
3  The Importance of Stratum Corneum Lipid Organization for Proper Barrier Function 31

CER[EOS] or [EOP] and the short-chain CER, thickness of about 1 Å at full hydration. From the
which can further be divided into the phytosphin- neutron scattering length density (NSLD) profile,
gosine type such as CER[AP] or CER[NP] and they further derived information about the posi-
the sphingosine-type CER such as CER[AS] or tion of the molecular groups of the lipids within
CER[NS]. Furthermore, both subclasses are the lipid bilayer. This was achieved by fitting the
known to exhibit a broad distribution of their NSLD profile with Gaussian functions, which
alkyl chain length, which is necessary for their resemble the position of the polar head groups,
proper functionality of the SC lipid matrix the CH3 group, the hydrocarbon chain region, and
(Norlen 2001). During the last years, especially the region of cholesterol location, respectively
the short-chain CER of the phytosphingosine (see Fig. 3.10). Furthermore, they established that
variety have gained in interest in the SC lipid a decrease in the amount of CHOL in the model
research, as specific, their role for a proper b­ arrier system correlates with an increase in the mem-
function has not been fully elucidated up to date. brane thickness (Kiselev et al. 2005).
In order to evaluate the specific function of each In order to identify the exact position of the
CER subclass nowadays, well-defined model CHOL molecules in this model membrane based
systems are investigated using different on CER[AP], Kessner et al. employed two par-
techniques. tially deuterated CHOL derivatives (Kessner
The various experimental techniques such as et al. 2008b). From the neutron scattering length
X-ray diffraction, vibrational spectroscopy, or density (NSLD) profiles, they concluded that the
differential scanning calorimetry (DSC) to char- CHOL molecules are immersed in the hydrocar-
acterize the thermotropic and/or lyotropic prop- bon chain region of the membrane bilayer, with
erties of the ceramides as bulk substance and in the isopropyl residue positioned in the center of
different mixtures have been extensively reviewed the membrane as depicted in Fig. 3.11.
before (Kessner et al. 2008c; Wartewig and As the interaction of the different lipid species
Neubert 2007). In order to understand the impact of the SC matrix is of high interest, the influence
of different CER species for the formation of the of the FFA chain length to the above-described
SC lipid matrix, the analysis of the CER as bulk model membrane based on CER[AP], CHOL,
material and then consequently in mixtures with and FFA was investigated also applying neutron
other SC lipids is mandatory for the ­interpretation diffraction (Ruettinger et al. 2008). In this study,
of the behavior in the complex multicomponent within SC lipid model membranes, only the FFA
SC lipid membranes. chain length (C18:0 stearic acid, C22:0 behenic
acid, C24:0 lignoceric acid, C26:0 hexacosanoic
acid) was varied and the results were compared
3.4.1 E
 lucidation of the to each other. The membrane thickness for all
Nanostructure of Stratum investigated model systems was found to be in
Corneum Lipid Models Based the range of two opposing CER[AP] molecules.
on CER[AP] or CER[NP] An increase of the FFA chain length did not cause
an alteration of the internal nanostructure but led
As a first step to investigate the influences of dif- to a slight decrease in the membrane thickness,
ferent short-chain CER, Kiselev and coworkers causing a partial interdigitation of the longer
prepared a SC lipid model membrane composed chained FFA. The reason for the unexpected
of CER[AP], cholesterol (CHOL), the free fatty result was placed on the presence of the polar
acid (FFA) palmitic acid and cholesterol sulfate as short-chain CER[AP]. This molecule establishes
oriented multilamellar membrane and investi- a tight hydrogen bond network between the adja-
gated it with neutron diffraction (Kiselev et al. cent bilayers due to its four OH groups. Thus, the
2005). They showed by determining the internal CER forces the long-chain FFA to incorporate
membrane nanostructure and the water distribu- into the unchanged spacing of the bilayer, thereby
tion across the bilayer, that such model membrane obligating the FFA to protrude partly through
exhibits very low hydration, with a water layer opposing leaflet as represented in Fig. 3.12.
32 A. Schroeter et al.

Fig. 3.11  Difference or


deuterium distribution profile 3

ρs(X), a.u.
of the SC lipid model
D
membrane derived from the 8
9
10
1 2
D
difference between NSLD 7 3 D
2 6 5 4
profile containing the HO
D D
deuterated CHOL derivative D
and the profile containing the
protonated specie. Reprinted
1
from Kessner et al. (2008b)
with permission from
Springer
0
-25 -22.5 -17.5 -15 -12.5 -7.5 -5 -2.5 2.5 5 7.5 12.5 15 17.5 22.5 25
-20 -10 0 10 20
x, Å

-1

CER[AP] CER[AP]

PA TA

CHOL/ ChS CHOL/ ChS

45.6 Å 43.3 Å
membrane thickness membrane thickness

Fig. 3.12 Schematic presentation of the structural brane containing palmitic acid (left) is compared to the
assembly of the SC model matrix based on CER[AP] matrix including tetracosanoic acid (right). CER[AP]
according to Ruettinger et al. (2008). To demonstrate the ceramide [AP], CHOL cholesterol, PA palmitic acid, TA
influence of the longer chained FFA a model for the mem- tetracosanoic acid

Consequently, CER[AP] creates a super-­ same group employed the partially deuterated
stable structure, which is not influenced by the FFA behenic-­ 22,22,22-d3-acid and cerotic-
alteration of the FFA chain length. The resulting 12,12,13,13-d4-­acid (Schroeter et al. 2009). The
free space due to the interdigitation of the FFA results from the neutron diffraction study pro-
can only be compensated by pulling the mem- vided the direct experimental evidence concern-
brane together, hence, the slight decrease in the ing the localization of the FFA in this SC lipid
membrane repeat distance. Additionally, the model system. Both the interdigitation and the
experiments revealed that the longer-chained presence of the FFA-rich phase could be proven
FFA tends to separate in an FFA-rich phase. It by this method.
was reasoned that the elongation of the chain As mentioned earlier, such SC lipid mem-
length of the FFA decreases the solubility of the branes show very small head group hydration.
FFA in the SC model membrane based on the Therefore, Ryabova and coworkers investi-
short-chain CER[AP] (Ruettinger et al. 2008). To gated the kinetics of the exchange of water in
verify the interdigitation of the FFA in the CER[AP]-based SC lipid model membranes
CER[AP]-based SC lipid model membrane, the with real-time neutron diffraction (Ryabova
3  The Importance of Stratum Corneum Lipid Organization for Proper Barrier Function 33

Fig. 3.13 Schematic partially hydrated membrane membrane in excess water


presentation of the armature
reinforcement model and
transformation of SC lipid
membrane from partly
hydrated to fully hydrated
state by the excess of water
modified according to
Kiselev (2007)
n n

fully extended (FE) conformation hair pin conformation

cholesterol free fatty acid


CER[AP]

et al. 2009). The study revealed that the kinetic


hydration comprises a fast initial segment, which
is followed by two slow stages. By increasing
hair pin FE conformation V–shape
the temperature to 57 °C, this process was sig- conformation conformation
nificantly faster in its initial phase. Furthermore,
they found that an irreversible phase separation at Fig. 3.14  Conformational states of the short-chain CER
this temperature and low hydration level occurs,
whereby they argued that the FFA separate. In density reduction increases the mobility of the
a further investigation the same group studied lipids, which further promotes the miscibil-
the influence of both a realistic FFA mixture ity of the lipids. When, on the other hand, the
and cholesterol sulfate (ChS) to the structure of amount of ChS was increased at the expense of
the above-described SC lipid model ­membrane CHOL, the swelling of the membrane increased.
based on CER[AP] (Ryabova et al. 2010). Ryabova et al. argued that the higher ability to
Again, as described by Ruettinger et al. (2008), form hydrogen bonds of ChS is responsible for
not even a mixture of different FFA affects the this effect (Ryabova et al. 2010).
nanostructure of this model system. Once more, As described in the preceding chapter, the
the FFA needs to interdigitate in order to fit into presence of CER[AP] in an SC lipid membrane
the bilayer created by CER[AP]. Nevertheless, composed also of CER[EOS], CHOL, and FFA
they found that their mixture containing six prevents the formation of the LPP, as it forces the
FFAs differing in chain length prevented the long ω-acyl chain of CER[EOS] to protrude
above-described FFA phase separation. Only the through the complete bilayer into the adjacent
hydration behavior was altered due to the FFA layer (Engelbrecht et al. 2011; Kessner et al.
mixture as Ryabova et al. discovered that the 2008a; Schroeter et al. 2008). These experimen-
membrane swelling process was accelerated at tal findings contributed to or are in accordance
low hydration levels, which was attributed to the with the so-called armature reinforcement model,
less dense bilayer packing due to the interdigi- a theoretical model describing the molecular
tation of the FFA (Ryabova et al. 2010). In the arrangement of these lipids (Kiselev 2007) (see
same study, no alteration of the rate of hydra- Fig. 3.13).
tion was observed for the complete substitution Here, the polymorphism of the short-chain
of ChS by CHOL. However, the absence of ChS CER (Pascher 1976; Pascher and Sundell 1992;
caused a phase separation, which was attributed Raudenkolb et al. 2003a, b, 2005) is taken into
to the missing sulfate group of ChS. As the sul- account, as the role of the fully extended con-
fate group is negatively charged, it can increase formation is discussed for the arrangement of
the molecular area per lipid, which subsequently the lipids (Fig. 3.14). This model conception
reduces the density of the lipid packing. This assumes that CER[AP] in its fully extended
34 A. Schroeter et al.

conformation adopts a sort of anchor function above-described CER[AP], this CER also
due to the strong intermembrane attractions, in belongs to the phytosphingosine subclass, but
which it is able to restrain the other lipids inside does not have the α-hydroxy group present in
the membrane and, respectively, forces their CER[AP]. Accordingly, the lamellar nanostruc-
arrangement within the membrane constraints. ture of a SC lipid model membrane based on
Upon hydration, CER[AP] is capable to perform CER[NP] was investigated in an interdisciplinary
a chain-flip transition to the one-sided or hairpin approach using both neutron diffraction and 2H-
conformation, which accounts for the alterations NMR spectroscopy and then compared to the
in the structure observed in the hydrated state above-described CER[AP]-based SC lipid model
(Kiselev et al. 2005). systems by Engelbrecht and coworkers
Next to neutron diffraction, there is also the (Engelbrecht et al. 2012). The authors indicated
very powerful technique of neutron small angle that in the presence of this CER subspecies a
scattering (SANS), which allows for the investiga- highly ordered lipid lamellae is formed and phase
tion of the structure of vesicular lipids in excess of separation occurred, which was even at high tem-
water. With SANS nanostructural parameters such perature in a densely packed and stable bilayer.
as size of the vesicle, thickness of a lipid bilayer, Here, intra- and intermolecular head group inter-
thickness of hydrophobic and hydrophilic regions, actions of CER[NP] prevent the hydration of the
and number of water molecules can be directly head group region. From the neutron diffraction
determined (Kiselev et al. 2006). Applying these data, it was proposed that CER[NP] exhibits a
methods Zemlyanaya and coworkers (Zemlyanaya V-shaped conformation in both lamellar phases,
et al. 2008) investigated CER[AP]-based quater- but with the distinction that one phase is phase-
nary unilamellar vesicles. In their investigation separated CER[NP] as portrayed in Fig. 3.15,
they detected a short-range interaction between which was further corroborated by 2HNMR spec-
the vesicles specimen, which leads to the forma- troscopy study.
tion of clustered structures. Furthermore, with this Moreover, the model system based on
study they confirmed the chain-flip transition of CER[NP] a completely different diffraction pat-
the CER[AP] molecules described above in the tern at higher temperature, when compared to the
armature reinforcement model. above-described CER[AP]-based lipid mem-
As CER[NP] is one of the most abundant CER branes. Thus, it was argued that the absence of
of the SC (Masukawa et al. 2009), the focus was just one OH group induces drastic structural
also placed on this molecule. Compared to the alteration in the membrane arrangement.

PI PII

CHOL SA CER[NP]

Fig. 3.15  Sketch of the assumed lamellar lipid assembly PII is constituted by crystalline CER[NP] showing a
present in the phase-separated domains of PI and PII in V-shaped conformation, PI is formed by CER[NP], SA
the ternary SC lipid model membrane containing and CHOL. Reproduced from Engelbrecht et al. (2012).
CER[NP], CHOL and SA at 80 °C and 99 % RH. While with permission from The Royal Society of Chemistry
3  The Importance of Stratum Corneum Lipid Organization for Proper Barrier Function 35

Fig. 3.16  Comparison of the D 3 D 2 D 2 D2 D 2 D2 D2 D 2 D 2 D 2 D2 D 2


O
HO
Neutron scattering length D2 D2 D2 D2 D2 D2 D2 D2 D2 D2 DH-N
HO N-H OH
density profile of the sample D2 D2 D2 D2 D2 D2 D 2 D2 D2 D2 D2
OH
containing the protonated O D3 D2 D2 D2 D2 D2 D2 D2 D2 D2 D2 D2
CER[NS] (solid line) and the 4
deuterated variety (dashed
line). Taken from Groen et al.
(2011) with permission from 3
Cell press

2
SLD (x10-9Å-2)

-1

-2
-3 -2 -1 0 1 2 3
X [nm]

3.4.2 Investigating However, as the acyl chain of CER[NS] is per-


the Nanostructure of an deuterated, only the presence of this chain can be
Stratum Corneum Substitute deduced from the data. It is true that the chains
need to be partially interdigitated in order to be in
The above-portrayed investigations were mainly agreement with the neutron diffraction data.
focused on the impact of one specific CER species Nevertheless, another arrangement is possible
in order to explicitly recognize the interaction whereby the CER molecules exhibit a fully
occurring between the different lipids and identify extended conformation (see Fig. 3.14), the deu-
the structure-function relationship of different terated chains forming one leaflet, while the
CER subspecies. In another approach Groen and sphingosine backbone chain is pointed in the
coworkers studied the nanostructure of SC lipid opposing direction.
model membranes with neutron diffraction, which Consequently, the drawback of the application
was composed of different CER species, CHOL, of perdeuterated lipids in the neutron diffraction
and FFA to closely mimic the SPP (Groen et al. experiment is the fact that it does not yield to an
2011). In order to localize the CER, they applied a unambiguous location of the labeled lipid ­species,
CER species (CER[NS]) with a perdeuterated and, furthermore, it does not give any informa-
acyl chain. In accordance with the finding tion about the conformational state of the studied
described concerning the short-­chain CER[AP], a lipid.
bilayer arrangement with a membrane thickness
in the range of two opposing CER molecules was
detected. From the neutron scattering length den- 3.5 Summary and Final Remarks
sity profile of the membrane containing the deu-
terated CER[NS] variety, they reasoned that the The intercellular lipid membranes of the SC are
CER exhibits a symmetrical organization and an excellent biological example for the relation-
excluded the asymmetric conformation of the ship between the lipid composition, its physi-
CER inside the bilayer arrangement (Engelbrecht cochemical properties and biological function,
et al. 2012) as schematically displayed in Fig. 3.16. as well as organization. To elucidate the special
36 A. Schroeter et al.

assembly and the properties of the native SC the molecular organization of the stratum corneum lip-
ids. J Lipid Res 39:186–196
lipid matrix is a very difficult task as the natural
Bouwstra J, Pilgram G, Gooris G, Koerten H, Ponec M
SC membranes are very complex. Therefore, in (2001a) New aspects of the skin barrier organiza-
recent years the researches were primarily placed tion. Skin Pharmacol Appl Skin Physiol 14(Suppl 1):
on the investigation of model systems in order to 52–62
Bouwstra JA, Gooris GS, Dubbelaar FE, Ponec M (2001b)
gain deeper insights into the driving forces of the
Phase behavior of lipid mixtures based on human
lipid assembling process. Furthermore, not only ceramides: coexistence of crystalline and liquid
model membranes are currently in the focus of phases. J Lipid Res 42:1759–1770
different investigation but also rather simplis- Bouwstra JA, Gooris GS, Dubbelaar FE, Ponec M (2002)
Phase behavior of stratum corneum lipid mixtures
tic, nonetheless realistic SC model membranes.
based on human ceramides: the role of natural and
As described, this offers the distinct benefit to synthetic ceramide 1. J Invest Dermatol 118:606–617
analyze the different lipid species systemati- Büldt G, Gally HU, Seelig A, Seelig J, Zaccai G (1978)
cally. This approach is very important, as espe- Neutron diffraction studies on selectively deuterated
phospholipid bilayers. Nature 271:184
cially the impact of the various CER subclasses
Cantor CR, Schimmel PR (1980) Biophysical chemistry:
for the proper barrier function of the SC needs to part II: techniques for the study of biological structure
be studied independently as shown for the very and function. Freeman, San Francisco
closely related CER[AP] and CER[NP]. Charalambopoulou GC, Steriotis TA, Hauss T,
Stefanopoulos KL, Stubos AK (2002) A neutron-­
So far a diversity of techniques have been
diffraction study of the effect of hydration on stratum
introduced into this field, whereby neutron dif- corneum structure. Appl Phys A 74:s1245–s1247
fraction with its specific advantages seems to be Coderch L, Lopez O, de la Maza A, Parra JL (2003)
the most promising one for the investigation of the Ceramides and skin function. Am J Clin Dermatol
4:107–129
nanostructure of the SC lipid matrix, as it enables
Dachs H (1978) Principles of neutron diffraction. In:
the use of specifically labeled molecules. However, Dachs H (ed) Neutron diffraction. Springer, Berlin,
only a combination of different methods and p 357
approaches can provide a complete picture of the Dahlen B, Pascher I (1972) Molecular arrange-
ments in sphingolipids - crystal-structure of
molecular arrangement of SC lipid matrix, as one
N-tetracosanoylphytosphingosine. Acta Crystallogr B
technique alone can cover only a small field. Struct B 28:2396
Up to date only a few CER subspecies have Dahlen B, Pascher I (1979) Molecular arrangements in
been investigated independently and are in parts sphingolipids – thermotropic phase-behavior of tetra-
cosanoylphytosphingosine. Chem Phys Lipids
debated controversially (see CER[EOS], for
24:119–133
instance). Consequently, there is still a high demand de Jager MW, Gooris GS, Dolbnya IP, Bras W, Ponec M,
to fully understand the impact of these lipids for a Bouwstra JA (2003) The phase behaviour of skin lipid
proper barrier function of the SC lipid matrix. mixtures based on synthetic ceramides. Chem Phys
Lipids 124:123–134
de Jager M, Gooris G, Ponec M, Bouwstra J (2004)
Acylceramide head group architecture affects lipid
organization in synthetic ceramide mixtures. J Invest
References Dermatol 123:911–916
de Sousa Neto D, Gooris G, Bouwstra J (2011) Effect of
Al-Amoudi A, Dubochet J, Norlen L (2005) Nanostructure the [omega]-acylceramides on the lipid organization
of the epidermal extracellular space as observed by of stratum corneum model membranes evaluated by
cryo-electron microscopy of vitreous sections of X-ray diffraction and FTIR studies (Part I). Chem
human skin. J Invest Dermatol 124:764–777 Phys Lipids 164(3):184–95
Bouwstra JA, Gooris GS, van der Spek JA, Bras W (1991) Engelbrecht T, Hauss T, Suss K, Vogel A, Roark M, Feller
Structural investigations of human stratum corneum SE, Neubert RHH, Dobner B (2011) Characterisation
by small-angle x-ray scattering. J Invest Dermatol of a new ceramide EOS species: synthesis and investi-
97:1005–1012 gation of the thermotropic phase behaviour and influ-
Bouwstra JA, Gooris GS, Cheng K, Weerheim A, Bras W, ence on the bilayer architecture of stratum corneum
Ponec M (1996) Phase behavior of isolated skin lipids. lipid model membranes. Soft Matter 7:8998–9011
J Lipid Res 37:999–1011 Engelbrecht TN, Schroeter A, Hauß T, Deme B, Scheidt
Bouwstra JA, Gooris GS, Dubbelaar FE, Weerheim AM, HA, Huster D, Neubert RHH (2012) The impact of
Ijzerman AP, Ponec M (1998) Role of ceramide 1 in ceramides NP and AP on the nanostructure of stratum
3  The Importance of Stratum Corneum Lipid Organization for Proper Barrier Function 37

corneum lipid bilayer. Part I: neutron diffraction and Kessner D, Brezesinski G, Funari SS, Dobner B, Neubert
2H NMR studies on multilamellar models based on RHH (2010) Impact of the long chain [omega]-acylce-
ceramides with symmetric alkyl chain length distribu- ramides on the stratum corneum lipid nanostructure.
tion. Soft Matter 8:2599 Part 1: thermotropic phase behaviour of CER[EOS]
Franks NP, Lieb WR (1979) The structure of lipid bilay- and CER[EOP] studied using x-ray powder diffrac-
ers and the effects of general anaesthetics. An x-ray tion and FT-raman spectroscopy. Chem Phys Lipids
and neutron diffraction study. J Mol Biol 133:469–500 163:42–50
Friberg SE, Osborne DW (1987) Interaction of a model Kiselev MA (2007) Conformation of ceramide 6 mole-
epidermal lipid with a vegetable oil adduct. J Dispers cules and chain-flip transitions in the lipid matrix of
Sci Technol 8:249–258 the outermost layer of mammalian skin, the stratum
Garson JC, Doucet J, Leveque JL, Tsoucaris G (1991) corneum. Crystallogr Rep 52:525–528
Oriented structure in human stratum corneum revealed Kiselev MA, Ryabova NY, Balagurov AM, Dante S,
by x-ray diffraction. J Invest Dermatol 96:43–49 Hauss T, Zbytovska J, Wartewig S, Neubert RH (2005)
Groen D, Gooris GS, Bouwstra JA (2010) Model mem- New insights into the structure and hydration of a stra-
branes prepared with ceramide EOS, cholesterol tum corneum lipid model membrane by neutron dif-
and free fatty acids form a unique lamellar phase. fraction. Eur Biophys J 34:1030–1040
Langmuir 26:4168–4175 Kiselev MA, Zemlyanaya EV, Aswal VK, Neubert RH
Groen D, Gooris GS, Barlow DJ, Lawrence MJ, van (2006) What can we learn about the lipid vesicle struc-
Mechelen JB, Demé B, Bouwstra JA (2011) Disposition ture from the small-angle neutron scattering experi-
of ceramide in model lipid membranes determined by ment? Eur Biophys J 35:477–493
neutron diffraction. Biophys J 100:1481–1489 Kuempel D, Swartzendruber DC, Squier CA, Wertz PW
Gutberlet T, Heinemann U, Steiner M (2001) Protein (1998) In vitro reconstitution of stratum corneum lipid
crystallography with neutrons – status and perspec- lamellae. Biochim Biophys Acta 1372:135–140
tives. Acta Crystallogr D 57:349–354 Madison KC, Swartzendruber DC, Wertz PW, Downing
Harroun TA, Wignall GD, Katsaras J (2006) Neutron scat- DT (1987) Presence of intact intercellular lipid lamel-
tering for biology. In: Fitter J, Gutberlet T, Katsaras lae in the upper layers of the stratum corneum. J Invest
J (eds) Neutron scattering in biology: techniques and Dermatol 88:714–718
applications. Springer, Berlin Masukawa Y, Narita H, Shimizu E, Kondo N, Sugai Y,
Hatta I, Ohta N, Ban S, Tanaka H, Nakata S (2001) X-Ray Oba T, Homma R, Ishikawa J, Takagi Y, Kitahara T,
diffraction study on ordered, disordered and reconsti- Takema Y, Kita K (2008) Characterization of overall
tuted intercellular lipid lamellar structure in stratum ceramide species in human stratum corneum. J Lipid
corneum. Biophys Chem 89:239–242 Res 49:1466–1476
Hinder A, Schmelzer CEH, Rawlings AV, Neubert RHH Masukawa Y, Narita H, Sato H, Naoe A, Kondo N, Sugai
(2011) Investigation of the molecular structure of the Y, Oba T, Homma R, Ishikawa J, Takagi Y, Kitahara
human stratum corneum ceramides [NP] and [EOS] T (2009) Comprehensive quantification of ceramide
by mass spectrometry. Skin Pharmacol Physiol species in human stratum corneum. J Lipid Res
24:127–135 50:1708–1719
Holleran WM, Man MQ, Gao WN, Menon GK, Elias McIntosh TJ (2003) Organization of skin stratum cor-
PM, Feingold KR (1991) Sphingolipids are required neum extracellular lamellae: diffraction evidence for
for mammalian epidermal barrier function. Inhibition asymmetric distribution of cholesterol. Biophys J
of sphingolipid synthesis delays barrier recovery after 85:1675–1681
acute perturbation. J Clin Invest 88:1338–1345 McIntosh TJ, Stewart ME, Downing DT (1996) X-ray
Holleran WM, Takagi Y, Uchida Y (2006) Epidermal diffraction analysis of isolated skin lipids: reconsti-
sphingolipids: metabolism, function, and roles in skin tution of intercellular lipid domains. Biochemistry
disorders. FEBS Lett 580:5466 35:3649–3653
Kessner D, Kiselev M, Dante S, Hauss T, Lersch P, Motta S, Monti M, Sesana S, Caputo R, Carelli S, Ghidoni
Wartewig S, Neubert RHH (2008a) Arrangement of R (1993) Ceramide composition of the psoriatic scale.
ceramide [EOS] in a stratum corneum lipid model Biochim Biophys Acta 1182:147–151
matrix: new aspects revealed by neutron diffraction Motta S, Monti M, Sesana S, Mellesi L, Ghidoni R,
studies. Eur Biophys J Biophys 37:989–999 Caputo R (1994) Abnormality of water barrier func-
Kessner D, Kiselev MA, Hauss T, Dante S, Wartewig tion in psoriasis. Role of ceramide fractions. Arch
S, Neubert RHH (2008b) Localisation of partially Dermatol 130:452–456
deuterated cholesterol in quaternary SC lipid model Nagle JF, Tristram-Nagle S (2000a) Lipid bilayer struc-
membranes: a neutron diffraction study. Eur Biophys J ture. Curr Opin Struct Biol 10:474–480
Biophys 37:1051–1057 Nagle JF, Tristram-Nagle S (2000b) Structure of lipid
Kessner D, Ruettinger A, Kiselev MA, Wartewig S, bilayers. Biochim Biophys Acta 1469:159–195
Neubert RH (2008c) Properties of ceramides and their Norlen L (2001) Skin barrier structure and function: the
impact on the stratum corneum structure: part 2: stra- single gel phase model. J Invest Dermatol 117:830–836
tum corneum lipid model systems. Skin Pharmacol Pascher I (1976) Molecular arrangements in sphingolipids
Physiol 21:58–74 conformation and hydrogen-bonding of ceramide and
38 A. Schroeter et al.

their implication on membrane stability and permea- brane. BENSC experimental reports. Helmholtz
bility. Biochim Biophys Acta 455:433–451 Zentrum Berlin für Materialien und Energie, Berlin
Pascher I, Sundell S (1992) Molecular arrangements in Schroeter A, Kiselev MA, Hauß T, Dante S, Neubert RHH
sphingolipids: crystal structure of the ceramide N-(2d, (2009) Evidence of free fatty acid interdigitation in
3d-dihydroxyoctadecanoyl)-phytosphingosine. Chem stratum corneum model membranes based on ceramide
Phys Lipids 62:79–86 [AP] by deuterium labelling. BBA Biomembr
Pfeiffer S, Vielhaber G, Vietzke JP, Wittern KP, Hintze U, 1788:2203
Wepf R (2000) High-pressure freezing provides new Tomita M, Hasegawa T, Tsukihara T, Miyajima S, Nagao
information on human epidermis: simultaneous pro- M, Sato M (1999) Two concentric protein shell struc-
tein antigen and lamellar lipid structure preservation. ture with spikes of silkworm Bombyx mori cytoplas-
Study on human epidermis by cryoimmobilization. mic polyhedrosis virus revealed by small-angle
J Invest Dermatol 114:1030–1038 neutron scattering using the contrast variation method.
Raith K, Farwanah H, Wartewig S, Neubert RHH (2004) J Biochem (Tokyo) 125:916–922
Progress in the analysis of stratum corneum cerami- Wartewig S, Neubert RHH (2007) Properties of ceramides
des. Eur J Lipid Sci Technol 106:561–571 and their impact on the stratum corneum structure: a
Raudenkolb S (2002) Untersuchungen zur strukturellen review. Part 1: ceramides. Skin Pharmacol Physiol
und physikochemischen charakterisierung von stratum 20:220–229
corneum lipiden und deren mischsystemen Institute of Wegener M, Neubert R, Rettig W, Wartewig S (1997)
Pharmacy, vol PhD. Martin-Luther-Universität Halle- Structure of stratum corneum lipids characterized
Wittenberg, Halle (Saale) by FT-raman spectroscopy and DSC. III. Mixtures
Raudenkolb S, Hubner W, Rettig W, Wartewig S, of ceramides and cholesterol. Chem Phys Lipids
Neubert RH (2003a) Polymorphism of ceramide 3. 88:73–82
Part 1: an investigation focused on the head group of Wertz PW, Downing DT (1983) Ceramides of pig epider-
N-octadecanoylphytosphingosine. Chem Phys Lipids mis: structure determination. J Lipid Res 24:759–765
123:9–17 Wertz PW, Swartzendruber DC, Madison KC, Downing
Raudenkolb S, Wartewig S, Neubert RH (2003b) DT (1987) Composition and morphology of epidermal
Polymorphism of ceramide 3. Part 2: a vibrational cyst lipids. J Invest Dermatol 89:419–425
spectroscopic and X-ray powder diffraction investiga- White SH, Mirejovsky D, King GI (1988) Structure of
tion of N-octadecanoyl phytosphingosine and the lamellar lipid domains and corneocyte envelopes of
analogous specifically deuterated d(35) derivative. murine stratum corneum. An x-ray diffraction study.
Chem Phys Lipids 124:89–101 Biochemistry 27:3725–3732
Raudenkolb S, Wartewig S, Neubert RH (2005) Wiener MC, White SH (1991) Fluid bilayer structure
Polymorphism of ceramide 6: a vibrational spectro- determination by the combined use of x-ray and
scopic and x-ray powder diffraction investigation of neutron-­diffraction. 1. Fluid bilayer models and the
the diastereomers of N-(alpha-hydroxyoctadecanoyl)- limits of resolution. Biophys J 59:162–173
phytosphingosine. Chem Phys Lipids 133:89–102 Worcester DL (1976) Neutron diffraction studies of
Ruettinger A, Kiselev MA, Hauss T, Dante S, Balagurov biological membranes and membrane components.
­
AM, Neubert RH (2008) Fatty acid interdigitation in Brookhaven Symp Biol 27:III37–III57
stratum corneum model membranes: a neutron diffrac- Yamamoto A, Serizawa S, Ito M, Sato Y (1991) Stratum
tion study. Eur Biophys J 37:759–771 corneum lipid abnormalities in atopic dermatitis. Arch
Ryabova N, Kiselev M, Balagurov A (2009) Transition Dermatol Res 283:219–223
processes in stratum corneum model lipid mem- Zbytovska J, Vavrova K, Kiselev MA, Lessieur P,
branes with a mixture of free fatty acids. Biophysics Wartewig S, Neubert RHH (2009) The effects of trans-
54:598–606 dermal permeation enhancers on thermotropic phase
Ryabova NY, Kiselev MA, Dante S, Hauss T, Balagurov behaviour of a stratum corneum lipid model. Colloids
AM (2010) Investigation of stratum corneum lipid Surf A 351:30–37
model membranes with free fatty acid composition by Zemlyanaya EV, Kiselev MA, Neubert R, Kohlbrecher J,
neutron diffraction. Eur Biophys J 39:1167–1176 Aksenov VL (2008) Investigation of the structure and
Schroeter A, Engelbrecht T, Hauß T, Neubert RHH (2008) properties of model membranes of the stratum cor-
Role of ceramide [AP] and ceramide [EOS] in the neum by small-angle neutron scattering. J Surf Invest
structural assembly of stratum corneum model mem- X-Ray 2:884–889
Molecular Structure and Function
of the Skin Barrier 4
Lars Norlén

Contents 4.1 Introduction


4.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . 39
Terrestrial life was only made possible through
4.2 Skin Lipid Composition
and Phase State . . . . . . . . . . . . . . . . . . . . . . 40 the adaptive evolution of a waterproof barrier
in the integument of organisms. In man, this
4.3 Skin Lipid Structure . . . . . . . . . . . . . . . . . . 40
barrier is constituted by a uniquely organized
4.4 Skin Lipid Formation . . . . . . . . . . . . . . . . . 40 lipid material situated between the cells of the
4.5 Skin Lipid Function . . . . . . . . . . . . . . . . . . . 41 horny layer of the skin (Breathnach et al. 1973;
Elias and Friend 1975). Recently, the lipid
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
material’s molecular organization was deter-
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42 mined in situ with the aid of a novel experimental
approach: high-resolution cryoelectron micros-
copy of vitreous tissue section (CEMOVIS)
defocus series combined with molecular mod-
eling and electron microscopy simulation (Iwai
et al. 2012). The lipid material is organized in
an arrangement not previously described in a
biological system – stacked bilayers of fully
extended ceramides with cholesterol molecules
associated with the ceramide sphingoid moiety
(Iwai et al. 2012). This organization not only
rationalizes the low permeability of the skin
barrier but also its robustness. The new knowl-
L. Norlén edge may serve as a molecular platform for in
Department of Cell and Molecular Biology (CMB),
silico approaches to identify molecules for
Karolinska Institutet, von Euler’s v 1,
Stockholm 171 77, Sweden enhancing skin penetration for percutaneous
drug delivery.
Dermatology Clinic, Karolinska University Hospital,
Stockholm, Sweden Below follows a brief account of the structure-
e-mail: lars.norlen@ki.se function relationships of the human skin barrier.

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 39


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_4, © Springer-Verlag Berlin Heidelberg 2015
40 L. Norlén

4.2 Skin Lipid Composition stacked in an alternate fashion as bilayers rather


and Phase State than as stacked monolayers.

The horny layer lipids consist of a heterogeneous


mixture of saturated, long-chain ceramides, free 4.4 Skin Lipid Formation
fatty acids, and cholesterol in a roughly 1:1:1
molar ratio (Wertz and Norlén 2003). More than In order to appreciate the structure-function rela-
300 different species have been identified in the tionships of the skin barrier in vivo, it is of value
ceramide fraction alone (Masukawa et al. 2009). to understand horny layer lipid formation, as the
The most characteristic features of the horny horny layer’s lipid structure may represent a “fro-
layer lipid composition (Wertz and Norlén 2003) zen-in” or “immobilized” open biological system
are (1) extensive compositional heterogeneity rather than a primary minimum energy order
with broad, but invariable, chain length distribu- equilibrium system. Skin lipid formation is also
tions (20–32 C; peaking at 24 C) in the ceramide central from a dermatological standpoint, since
fatty acid and free fatty acid fractions, (2) almost barrier malformation may be an etiological factor
complete dominance of saturated very long in barrier-deficient skin conditions such as
hydrocarbon chains (C20:0–C32:0), and (3) large eczema, psoriasis, and “dry skin.”
relative amounts of cholesterol (about 30 mol%). It has recently been proposed that skin lipid for-
These compositional features are typically mation proceeds via (1) membrane synthesis in the
those stabilizing lipid gel phases. It has therefore trans-Golgi of a membrane system with cubic-like
been proposed that the horny layer lipid structure symmetry, followed by (2) morphologically con-
exists as a single and coherent gel phase (Norlén tinuous (non-fusion-dependent) secretion of the
2001b). The viscous gel-like behavior of the lipid cubic-like membrane system into the extracellular
structure has recently been demonstrated by its space, (3) phase transition from cubic-like to
remarkable malleability in situ (Iwai et al. 2012). lamellar membrane morphology, (4) dehydration,
(5) condensation, and (6) lipid chain rearrange-
ment from a folded (hairpin) to an extended
4.3 Skin Lipid Structure (splayed chain) stacked bilayer conformation
(Norlén 2001a; Iwai et al. 2012). CEMOVIS
CEMOVIS has recently shown that the extracel-
lular lipid matrix of the horny layer is organized
as a bilayer structure of fully extended (splayed
chain) ceramides with the sphingoid moieties
interfacing. Both cholesterol and the free fatty
acids are distributed selectively: cholesterol at the
ceramide sphingoid end and the free fatty acid at
the ceramide fatty acid end (Iwai et al. 2012)
(Figs. 4.1 and 4.2). A unique feature of the horny
layer lipid organization is that the lipid molecules
are arranged in the splayed chain conformation,
with the two hydrocarbon tails pointing in oppo-
site directions, contrary to conventional biologi-
cal membranes where the lipids are arranged in Fig. 4.1 Molecular organization of the skin barrier. The
the hairpin conformation with the two hydrocar- stratum corneum lipid layer is organized as stacked bilay-
bon tails pointing in the same direction. Further, ers of fully extended ceramides with cholesterol mole-
cules associated with the ceramide sphingoid moiety
the skin barrier organization differs from conven-
(Iwai et al. 2012). Green spheres represent hydrogen and
tional fat crystals arranged in the splayed chain carbon atoms in ceramides, cholesterol, and free fatty
conformation, as the lipid layers in the skin are acids. Red spheres represent oxygen atoms
4 Molecular Structure and Function of the Skin Barrier 41

Skin suface

Stratum
FFA
corneum

CER

CHOL

Viable
epidermis
CHOL

CER

FFA

Dermis

Fig. 4.2 Schematic drawing of the skin. Left part sche- repeating unit, composed of two mirrored subunits, each
matic cellular-scale drawing of epidermis. Middle part composed of one fully extended ceramide (CER), one
molecular-scale drawing of the lamellar lipid structure cholesterol (CHOL), and one free fatty acid (FFA) mole-
occupying the space between the cells of the stratum cor- cule (Adapted from Norlén (2012), with permission)
neum. Right part atomic model of the lipid structure’s

supports the proposed continuity of the lipid secre- towards temperature and pressure changes
tion system as well as the proposed structural asso- because of its heterogeneous lipid composition
ciation of non-lamellar and lamellar lipid and high cholesterol content, which stabilize gel-
morphologies (Norlén et al. 2003; Al-Amoudi like chain packing and thereby prevent both lat-
et al. 2005). However, structure determination of eral domain formation and induction of “pores”
the intermediate stages of skin lipid formation may or non-lamellar morphologies. Further, this
require access to native molecular resolution tomo- bilayer arrangement accounts for stratum cor-
graphic 3D data in situ (molecular tissue TOVIS neum cell cohesion without advocating special-
(cf. Norlén et al. 2009)), a developing technology ized intercellular adhesion structures such as
that may not yet have reached its full potential. desmosomes. The arrangement hence allows for
sliding of stratum corneum cells to accommodate
skin bending. Finally, as the interaction between
4.5 Skin Lipid Function the individual layers of the lipid structure involves
only hydrocarbons, the layers may be relatively
Current knowledge suggests that a stacked, fully free to slide with respect to one another, making
extended (splayed chain) ceramide bilayer the lipid structure pliable. The fully extended
arrangement (Figs. 4.1 and 4.2) with a high cho- ceramide bilayer arrangement with high choles-
lesterol content and a heterogeneous, saturated, terol content and heterogeneous saturated long-
long-chain lipid composition represents an opti- chain lipid composition thus meets the barrier
mized barrier organization for skin. This is needs of the skin by being simultaneously imper-
because it renders skin largely impermeable to meable and robust.
water as well as to both hydrophilic and lipo-
philic substances due to its condensed chain Conclusions
packing and its alternating lipophilic (alkyl It was recently shown that the human skin bar-
chain) and hydrophilic (headgroup) regions. rier is organized as stacked bilayers of fully
Likewise, it is resistant to both hydration and extended ceramides with cholesterol mole-
dehydration because of its lack of exchangeable cules associated with the ceramide sphingoid
water between lipid leaflets. It is also resistant moiety.
42 L. Norlén

The physical state of the skin’s lipid struc- Breathnach AS, Goodman T, Stolinski C, Gross M (1973)
Freeze fracture replication of cells of stratum corneum
ture has been proposed to be that of a single
of human epidermis. J Anat 114:65–81
and coherent gel phase. Further, the lipid Elias PM, Friend DS (1975) The permeability barrier in
structure may be formed via a phase transition mammalian epidermis. J Cell Biol 65:180–191
from cubic-like to stacked lamellar morphol- Iwai I, Han H, den Hollander L, Svensson S, Öfverstedt
LG, Anwar J, Brewer J, Bloksgaard Mølgaard M,
ogy followed by a flip of the constituent lipid
Laloeuf A, Nosek D, Masich S, Bagatolli L, Skoglund
components from a folded (hairpin) to an U, Norlén L (2012) The human skin barrier is orga-
extended (splayed chain) ceramide bilayer nized as stacked bilayers of fully-extended ceramides
conformation. with cholesterol molecules associated with the
ceramide sphingoid moiety. J Invest Dermatol
The skin’s lipid structure is responsible for
132:2215–2225. doi:10.1038/jid.2012.43
both the skin’s low permeability towards water Masukawa Y, Narita H, Sato H, Naoe A, Kondo N, Sugai
and hydrophilic and lipophilic substances and Y, Oba T, Homma R, Ishikawa J, Tagaki Y, Kitahara T
the barrier’s robustness towards environmen- (2009) Comprehensive quantification of ceramide spe-
cies in human stratum corneum. J Lipid Res
tal stress, such as hydration and dehydration,
50:1708–1719
temperature and pressure changes, stretching, Norlén L (2001a) Skin barrier formation: the membrane
compression, bending, and shearing. folding model. J Invest Dermatol 117(4):823–829
The new molecular description of the skin Norlén L (2001b) Skin barrier structure and function: the
single gel-phase model. J Invest Dermatol 117(4):
barrier may serve as a molecular platform for
830–836
in silico approaches such as molecular simula- Norlén, L (2012) Skin Lipids. In Gordon C. K. Roberts
tions, as well as for in vitro modeling, to (ed.) Encyclopedia of Biophysics, Springer-Verlag
underpin interactions of the lipid matrix with Berlin Heidelberg, Vol 5, pp. 2368–2373
Norlén L, Al-Amoudi A, Dubochet J (2003) A cryo-
drugs and other chemicals. For example, it is
transmission electron microscopy study of skin barrier
foreseeable that this knowledge will now formation. J Invest Dermatol 120:555–560
enable in silico screening to identify mole- Norlén L, Öktem O, Skoglund U (2009) Molecular cryo-
cules for enhancing skin penetration for per- electron tomography of vitreous tissue sections: cur-
rent challenges. J Microsc 235:293–307
cutaneous drug delivery.
Wertz P, Norlén L (2003) “Confidence intervals” for the
“true” lipid compositions of the human skin barrier?
In: Forslind B, Lindberg M (eds) Skin, hair, and nails.
Structure and function. Marcel Dekker Inc, New York,
pp 85–106, Biochim Biophys Acta 304:265–275
References
Al-Amoudi A, Dubochet J, Norlén L (2005) Nanostructure
of the epidermal extracellular space as observed by
cryo-electron microscopy of vitreous sections of
human skin. J Invest Dermatol 124:764–777
The Increasing Importance
of the Hair Follicle Route in Dermal 5
and Transdermal Drug Delivery

Alexa Patzelt and Jürgen Lademann

Contents 5.1 Introduction


5.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . 43
For more than 200 years, constant attempts have
5.2 Architectural and Physiological
Features of the Hair Follicle with
been made to administer drugs via the skin and
Regard to Follicular Penetration . . . . . . . 44 to enhance percutaneous penetration by vari-
ous methods including mechanical, physical,
5.3 Mechanisms of Follicular Penetration
and Transfollicular Penetration . . . . . . . . 45 and chemical manipulations to reduce the bar-
5.3.1 Mechanisms of Follicular rier function of the skin (Helmstadter 2011). In
Penetration . . . . . . . . . . . . . . . . . . . . . . . . . 45 total, three potential penetration pathways have
5.3.2 Mechanisms of Transfollicular
been identified. In addition to the well-described
Penetration . . . . . . . . . . . . . . . . . . . . . . . . . 49
intercellular penetration pathway being mainly
5.4 Enhancement of Follicular responsible for the percutaneous penetration
Penetration. . . . . . . . . . . . . . . . . . . . . . . . . 49
effect, also the follicular route has been spotted
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51 to be of considerable interest as especially the
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51 upper portion of the hair follicle – the infundibu-
lum – displays an area of additional absorption.
However, in different skin sites, the size and
number of hair follicles can differ tremendously
(Otberg et al. 2004b); thus, also the influence
of the follicular penetration route can vary. The
transcellular penetration pathway, on the con-
trary, seems to be of inferior importance.
The aim of the present chapter is to describe
and define the role of the hair follicle in the pen-
etration process and to identify mechanisms
which allow follicular penetration enhancement.
A. Patzelt (*) • J. Lademann Due to the architectural structure of the hair
Department of Dermatology, Venereology and follicle, follicular penetration is a complex pro-
Allergology, Charité – Universitätsmedizin Berlin, cess and has to be divided at least into two steps
Center of Experimental and Applied Cutaneous
as illustrated in Fig. 5.1. It has to be distinguished
Physiology, Berlin, Germany
e-mail: alexa.patzelt@charite.de; between the penetration into the hair follicle and,
juergen.lademann@charite.de in the second step, the transfollicular penetration

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 43


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_5, © Springer-Verlag Berlin Heidelberg 2015
44 A. Patzelt and J. Lademann

into the living tissue surrounding the hair follicle, licular penetration is applicable (Otberg et al.
which cannot be observed for every applied sub- 2008) as well as long-term intrafollicular storage
stance, yet. Previous investigations could show (Lademann et al. 2006) if transfollicular penetra-
that mainly the physicochemical properties of a tion cannot be realized. These and more aspects
topically administered substance determine its will be discussed in this chapter.
follicular penetration depth (Patzelt et al. 2011)
and whether or not the substance is able to pene-
trate transfollicularly. Thus, modifications of the 5.2 Architectural
physicochemical properties of the substances and Physiological Features
such as size can be utilized as follicular penetra- of the Hair Follicle
tion enhancers or inhibitors. with Regard to Follicular
In comparison to the intercellular penetration Penetration
process, topical drug delivery via the hair folli-
cles provides additional features such as fast Due to its complex and dynamic architectural
delivery into the systemic circulation if transfol- structure, the hair follicle is predestined as a
penetration and storaging organ, although the
original functions of the hair follicle seem to be
rather those of a sensory organ, sebum excretion
and protection (Krause and Foitzik 2006). The
infundibulum is the upper part of the hair folli-
cle and consists of an upper and lower portion as
depicted in Fig. 5.2. The epithelium of the upper
infundibulum is continuous with the keratinized
1 epidermis and covered by an intact stratum cor-
neum, whereas the differentiation pattern of the
lower infundibulum switches from epidermal
to trichilemmal leading to an interrupted skin
barrier with only few differentiated corneocytes

2 Hair
Stratum corneum

Upper
Infundibulum Blood capillaries &
Lower
Dendritic cells

Sebaceous
gland
Bulge region
with stem cells

Fig. 5.2 Schematic overview of the follicular architec-


Fig. 5.1 Schematic illustration of the follicular penetra- ture especially depicting the structures relevant to the fol-
tion pathway which has to be divided into two steps: (1) licular penetration process such as the infundibulum
intrafollicular penetration and (2) transfollicular penetra- surrounded by the blood capillaries and dendritic cells,
tion if allowed due to size reasons the sebaceous gland, and the bulge region with stem cells
5 The Increasing Importance of the Hair Follicle Route in Dermal and Transdermal Drug Delivery 45

remaining (Blume-Peytavi and Vogt 2011) that (Vogt et al. 2007). A schematic overview of the
are more prone to transfollicular penetration follicular architecture is depicted in Fig. 5.2.
in this region. Moreover, the whole follicular
infundibulum is supplied by a dense capillary
network and surrounded by a high number of 5.3 Mechanisms of Follicular
immune cells, on the one hand allowing the Penetration
rapid systemic uptake of substances once pen- and Transfollicular
etrated transfollicularly and, on the other hand, Penetration
rendering the hair follicle to be a promising tar-
get for immune therapy or topical vaccination 5.3.1 Mechanisms of Follicular
(Patzelt et al. 2011; Vogt et al. 2008). Penetration
Additionally, as depicted in Fig. 5.2, also the
sebaceous gland and the bulge region of the hair The follicular penetration of topically applied
follicle which is hosting the stem cells are inter- substances represents a complex process which
esting structures within the hair follicle, at the has not been clarified in detail until now. Whereas
same time being attractive targets for therapeutic the retention of the particles in the follicular duct
interventions (Patzelt et al. 2011). The sebaceous has been well documented, researchers are con-
gland is the organ of sebum excretion and addi- troversially discussing which substances are able
tionally associated with a diversity of pathologies to penetrate transfollicularly into the deeper skin
such as acne (Thiboutot 2004). It was suggested layers, whereby the size next to other physico-
that by increasing the distribution of correspond- chemical properties seems to be the predominant
ing drugs in the sebaceous gland, the therapeu- parameter (Labouta and Schneider 2013). In the
tic effectivity could be significantly improved last years, predominantly particulates such as
(Rolland et al. 1993). Respective efforts have liposomes and micro- and nanoparticles have
already been made (Morgan et al. 1993; Ridolfi attracted attention as a result of their capabil-
et al. 2012; Rolland et al. 1993), and some prom- ity to improve penetration into the hair follicle.
ising antiacne products are already on the market. Here, a clear size dependency could be observed
The multipotent, highly proliferative, and easily demonstrating that 320 nm sized polymer par-
accessible stem cells located in the bulge region ticles covalently labeled with a fluorescent dye
(Ohyama 2007) are also to be integrated in the penetrated significantly deeper into the hair
therapeutic concepts for cutaneous regenerative follicles than the same fluorescent dye in non-
medicine or gene correction of congenital hair particulate form (Lademann et al. 2007). The
disorders or genetic skin diseases. optimum size for particles to penetrate deeply
In total, more than 20 different cell types are into the hair follicle was determined to be in the
involved in the structure of the hair follicle range of 400–700 nm, whereas larger and smaller
which underlies cyclical activity (Rogers 2004). particles reached significantly lower penetration
Three different hair follicle types, namely, depths (Patzelt et al. 2011) or remained even on
lanugo, vellus, and terminal hair follicles, have the skin surface in the case of very large par-
to be distinguished. Whereas the same hair fol- ticles (Schaefer and Lademann 2001; Toll et al.
licle produces lanugo hairs in the fetal period 2004). As this effect was demonstrated for dif-
and vellus hairs in the childhood, it produces ferent solid particle preparations such as PLGA
terminal hairs in the adulthood (Blume-Peytavi particles and silicium oxide particles, it was
and Vogt 2011). In total, each human individual assumed that follicular particle penetration is a
displays an estimated number of five million predominantly mechanical effect independently
hair follicles (Krause and Foitzik 2006) under- from the particle preparation (Patzelt et al. 2011).
lining its potential in the penetration process. In Fig. 5.3, the dependency of the penetration
The morphometry of vellus and terminal hair depth on the particles’ size is schematically rep-
follicles has been well documented already resented. Lademann et al. (2009) hypothesized
46 A. Patzelt and J. Lademann

3 µm 1 µm 400–700 nm 100 nm Size ?

Fig. 5.3 Dependency of the penetration depth of the par- cantly deeper into the hair follicles than larger or smaller
ticles on the particles’ size. Large particles (>3 μm) are particles. A size threshold below which transfollicular
only located on the skin surface and in the follicular ori- penetration occurs has not been determined, yet
fice. Particles between 400 and 700 nm penetrate signifi-

that the surface structure of the hair and the hair


Thickness of
follicle, which is determined by the thickness of cuticula cell
the keratin cells in the cuticula, being 530 nm in
human hairs and 320 nm in porcine hairs, might
act as a pumping system delivering the particles Cuticula
deeply into the hair follicle as demonstrated Medulla
in Fig. 5.4. Whereas the movement of the hair
occurs physiologically in vivo, it could be shown
Cortex
that this effect can be simulated in vitro by mas-
sage appliance (Lademann et al. 2007; Patzelt
et al. 2011).
Once penetrated into the hair follicles, sub- Fig. 5.4 Schematic cross section of a hair illustrating the
stances are stored over several days (Lademann specific structure of the cuticula cells. It has been hypoth-
et al. 2006) within this protected area if transfol- esized that this surface structure which is determined by
the thickness of the cuticula cells might act as a pumping
licular penetration is not feasible. Whereas the system delivering the particles deeply into the hair folli-
reservoir of the stratum corneum is relatively cles when cuticula thickness and particle size are similar
unprotected and thus easily depleted by daily
processes such as textile or water contact in com-
bination with the physiological desquamation retarded release could diminish the application
process removing one layer of corneocytes per frequency and thus increase the compliance of
day, the hair follicle represents a protected reser- patients and the therapeutic outcome.
voir which can only be depleted by such slow This therapeutic effectivity could even be
outward-directed processes as sebum flow and enhanced when the correct skin site is chosen for
hair growth. Previous investigations could show the application. Otberg et al. (2004b) measured
that a particle-containing formulation was still the follicular density, the volume of the follicular
detectable within the hair follicle after 10 days, infundibula per square centimeter of skin, and the
whereas the stratum corneum reservoir had surface of the follicular infundibula per square
already been almost completely depleted after centimeter of skin. The latter corresponds to the
1 day (Lademann et al. 2006) as presented in additional absorption area provided by the hair
Fig. 5.5. This long-term storage effect could be follicles. They could show that the forehead and
effectively utilized for therapeutic purposes as the calf regions had the highest follicular volume
the application of drug-loaded particles with per square centimeter of skin surface which was
5 The Increasing Importance of the Hair Follicle Route in Dermal and Transdermal Drug Delivery 47

30 min after application 1 d after application 10 d after application

Fig. 5.5 The hair follicle is a long-term reservoir for neum reservoir is significantly depleted, whereas the par-
topically applied substances. Directly after application, ticles are still available within the hair follicle. After
the applied particles are distributed on the skin surface 10 days, part of the particles are still detectable within the
and within the hair follicle. After 1 day, the stratum cor- hair follicle

explained by the high follicle density on the


forehead and the large hair follicles on the calf. It
was estimated that the reservoir volumes of the
hair follicles and of the stratum corneum were
comparable in these body regions as demon-
strated in Fig. 5.6, whereas the reservoir of the Reservoir of stratum corneum
hair follicles in the region of the forearm was sig- =
nificantly lower by a factor of 20 in comparison Reservoir of hair follicles
to the stratum corneum reservoir.
The investigation of follicular penetration still
represents a challenge as it requires spatial reso-
lution. Nowadays, several methods are available
to investigate follicular penetration reasonably.
These methods have been recently summarized
by Meidan et al. (2010) and include, inter alia,
the selective artificial closing technique, where
the hair follicles are selectively blocked with a
varnish-wax mixture and thus excluded from the Fig. 5.6 The largest follicular reservoir can be found on
the calf and the forehead. Here, the follicular reservoir is
penetration process (Teichmann et al. 2006), the
comparable to the stratum corneum reservoir
usage of a sandwich model (Barry 2002), where
the top skin layer blocks the shunts in the bot-
tom layer or the differential stripping method experiments performed on the same volunteers
(Teichmann et al. 2005). Further novel optical and each on the contralateral skin site, the
devices are, e.g., autoradiography (Fabin and in vitro follicular reservoir was only 10 % of the
Touitou 1991), confocal laser scanning micros- in vivo follicular reservoir. It was assumed that
copy (Lademann et al. 2010) or combined con- the elastic fibers surrounding the hair follicle
focal laser scanning microscopy with confocal contract during the excision. Whereas the
Raman spectroscopy (Caspers et al. 2003). removed skin sample can be re-stretched to its
Next to methodological challenges, also the original size by expanding the interfollicular
selection of adequate model systems plays a elastic fibers, those surrounding the hair follicle
superior role as could be demonstrated recently. remain contracted and reduce the follicular
Patzelt et al. (2008) could demonstrate that for reservoir significantly. Based on these observa-
48 A. Patzelt and J. Lademann

tions, it can be stated that excised skin is not an In addition to physicochemical properties of
appropriate model to investigate follicular pene- the applied substance and the pumping effect
tration. However, in vivo investigations are not transporting the substances into the hair follicle
always feasible as substances or methods are by hair movement, also the activity status of the
mostly too injurious. As a result, the porcine ear hair follicles seems to play an important role and
model has been determined to be a suitable decides whether or not follicular penetration
ex vivo model as the full skin can remain fixed occurs at all. As mentioned earlier, each hair fol-
on the underlying cartilage during the experi- licle undergoes continuous cycling, which
ments, and the good and well-documented simi- includes the complete remodeling of its non-
larities of porcine and human skin architecture permanent portion and which influences the hair
and structure allow a reasonable evaluation of growth and sebum excretion activity of each hair
the obtained data (Lademann et al. 2010). follicle. A previous study could demonstrate that
In this context, it has to be emphasized that it can be distinguished between active hair folli-
full skin samples including the subcutaneous tis- cles, which provide hair growth and/or sebum
sue are very important for follicular penetration excretion and are open for penetration, and inac-
investigations. For diffusion cell experiments, tive hair follicles which provide neither hair
however, mostly split skin or epidermal skin is growth nor sebum flow and are thus inaccessible
utilized where at least the subcutaneous tissue is for topically applied substances (Otberg et al.
discarded meaning that the lower part of the hair 2004a). For these inactive follicles a cover con-
follicle which reaches deeply into the subcutane- sisting of dried sebum and desquamated corneo-
ous tissue is cut off. This means that the inferior cytes was detected in the follicular orifices that
part of the hair follicle is open and the topically prevents any penetration process. In the forearm
applied substances can diffuse directly into the region, one fourth of the hair follicles were shown
receptor medium as demonstrated in Fig. 5.7. to be unreceptive for penetration.
Similar concerns have already been raised by
Senzui et al. (2010).

Substance

500 µm

Receptor

Fig. 5.7 Removing the subcutaneous tissue leads to a the receptor medium via the open inferior part of the hair
violation of the distal hair follicle. If, as usual, only the follicle. Thus, the utilization of diffusion cell experiments
upper part of the skin is utilized for diffusion cell experi- is not an appropriate method to investigate follicular
ments, the topically applied substance can just diffuse into penetration
5 The Increasing Importance of the Hair Follicle Route in Dermal and Transdermal Drug Delivery 49

5.3.2 Mechanisms of Transfollicular performed with dermatomed or split skin which


Penetration always includes a violation of the distal hair fol-
licle so that topically applied substances can just
Transfollicular penetration of topically applied diffuse into the receptor medium. Whereas the
substances and mechanisms of transfollicular contraction effect of the elastic fibers surrounding
penetration have not been fully clarified, so far. It the hair follicle assumed by Patzelt et al. (2008)
can be assumed, yet, that transfollicular penetra- might be able to inhibit the diffusion of larger par-
tion occurs mainly in the region of the infundibu- ticles, it might explain that the detected diffusion
lum and specifically in the lower portion of the of very small particles into the receptor medium
infundibulum where the barrier is interrupted due is erroneously interpreted as penetration. This
to the switch of a differentiation pattern as men- theory is supported by the fact that for all human
tioned above and depicted in Figs. 5.1 and 5.3. in vivo studies reported by Labouta and Schneider
For smaller non-particulates, i.e., substances such (2013), no particle penetration or permeation
as caffeine or minoxidil, a rapid transfollicular could be detected. Only some studies were per-
penetration and systemic uptake has already been formed in vivo on animal skin, and within this
reported (Blume-Peytavi et al. 2010; Otberg et al. group, only few studies reported a penetration
2008). Interestingly, the systemic uptake was sig- into deeper skin layers of very small particles.
nificantly faster (already after 5 min) when the The authors assumed that the gold nanoparticles
hair follicles were accessible in comparison to utilized in their study might interact hydrophobi-
skin areas where the hair follicles were selectively cally with the skin lipids leading to a disruption of
blocked previously. Here, the substances needed the skin lipid layer structure, subsequent increased
approximately 20 min to be detectable in the cir- skin permeability and penetration into deeper skin
culation. For particles the situation seems to be layers (Huang et al. 2010). This assumption still
different, yet. There are clear indications that the needs further verification.
transfollicular penetration process is predomi- Summarizing the recent findings on transfol-
nantly determined by the size of the applied par- licular penetration, it can be stated that particles
ticles, although a clear size threshold for are well suitable to deliver active substances into
transfollicular penetration could not be defined so the hair follicle, whereas transfollicular penetra-
far. Recently, Labouta and Schneider (2013) tion is rather unlikely. For particles larger than
reviewed the current literature focusing on skin 100 nm, intercellular or transfollicular perme-
penetration of inorganic particles. It could be ation has not been observed in intact skin, yet; for
shown that about half of the studies reported par- smaller particles further research is necessary to
ticle penetration or permeation. However, most define a clear threshold below which transfollicu-
studies’ protocols involved either mechanical or lar and intercellular penetration can occur, which
chemical penetration enhancers (Dixit et al. 2007; is also an important aspect with regard to risk
Krishnan et al. 2010; Labouta et al. 2011; assessment.
Mortensen et al. 2008; Paliwal et al. 2006;
Upadhyay 2006; Zhang and Monteiro-Riviere
2008) or utilized excised either animal or human 5.4 Enhancement of Follicular
skin in in vitro diffusion cell experiments. With Penetration
regard to risk assessment or mechanistic investi-
gations concerning transfollicular penetration, the As summarized above, particulate nanocarriers
utilization of penetration enhancers or of in vitro are excellent delivery systems for active sub-
diffusion cell experiments seems to be not reason- stances into the hair follicle, which moreover,
able as described above as overestimation of represents an interesting target site and permits
particle penetration can occur if penetration is fast access into the deeper viable skin layers by
either artificially enhanced or investigations are bypassing the complex intercellular penetration
50 A. Patzelt and J. Lademann

pathways. The disadvantage – or advantage in biopsies, chemical enhancers, microneedles, elec-


terms of risk assessment – however is that parti- troporation, or ultrasound (Lawson et al. 2007;
cles have not been reliably demonstrated to pen- Vogt et al. 2008).
etrate intercellularly or transfollicularly. As a noninvasive alternative, the triggered
Therefore, new approaches have to be devel- release of substances from particle preparations
oped to utilize the advantages of particulate has recently been introduced. In this approach,
delivery – such as deep follicular penetration, the particles exclusively serve as delivery sys-
long-term follicular storaging, sustained release, tems to the desired penetration depth within the
and shielding from degradation – also for trans- hair follicle which can be controlled by the par-
follicular transport of active substances. ticle size. Having reached the desired depth, the
One option is to utilize particles exclusively particles release their active agent by a specific
for delivery of substances into the hair fol- triggering signal which can then translocate inde-
licle and its specific target sites. Recently, it pendently to the viable skin. The principle is
was demonstrated that an antiseptic associated illustrated in Fig. 5.8. By utilizing these stimuli-
with approximately 300 nm sized carrier par- responsive controlled release systems, site-
ticles originating from a fat emulsion on the selective, controlled release patterns can be
basis of medium- and long-chain triglycerides achieved leading to increased therapeutic effi-
(Lipofundin® MCT/LCT, Braun, Germany) was cacy and decreased side effects (Zhu et al. 2005).
able to achieve longer lasting antiseptic effects At the moment, research especially
than the same substance in a non-particulate focuses on the identification of appropriate
form (Ulmer et al. 2012). The study was based
on the assumption that about 25 % of the resi-
dent bacteria colonizing the skin reside within
the hair follicles (Lange-Asschenfeldt et al. Triggering signal
2011). Conventional non-particulate antiseptics,
however, are not able to eradicate all bacteria
from this follicular reservoir which leads to a
fast recolonization of the skin. In contrast, it was
shown that the particle-based antiseptic was able
to penetrate deeply into the hair follicle. The
recolonization was consequently retarded. Other
examples for improved follicular penetration and
optimized therapeutic effects include the appli-
cation of encapsulated hair-growing substances
or acne or rosacea therapeutics (Rolland et al.
1993; Shim et al. 2004; Tsujimoto et al. 2007).
Some of the products are already commercially
available (Papakostas et al. 2011).
If intrafollicular penetration of a therapeutic is
not sufficient, strategies have to be developed to
enhance the transfollicular permeation of the par-
ticles or of the active substance alone after par-
ticulate delivery into the hair follicle.
An easy, but invasive approach to translocate
substances to the viable skin is to disturb the skin Fig. 5.8 Principle of stimuli-responsive release systems.
The particles deliver the drug molecules to the desired
barrier prior to the application of the substance or
depth within the hair follicle. After external or internal
particles, respectively. Here, several techniques stimulus, the active drug is released from the particle and
are available such as cyanoacrylate skin surface translocates independently to the deeper viable skin layers
5 The Increasing Importance of the Hair Follicle Route in Dermal and Transdermal Drug Delivery 51

stimuli-responsive release systems. Mak et al. is released by a specific triggering signal and
(2011, 2012) recently introduced a release system can translocate independently to the deeper
based on the interaction of the particles with a viable skin layers surrounding the hair follicles,
protease. When the protease was likewise applied subsequently. The controlled drug release rep-
in particulate form, similar penetration depths resents a promising concept to utilize the
were reached for the particles and the protease advantageous delivering attributes of particles
leading to a release of the active substance from also for transfollicular penetration.
the delivering particles also at significant depths
of the hair follicle. Even an uptake of the model Acknowledgments We would like to thank the
drug by the sebaceous gland could be detected. Foundation “Skin Physiology” of the Donor Association
for German Science and Humanities for financial support.
Additional general approaches of controlled Parts of this work were realized within the BMBF project
drug release include the application of high- “Topische Vakzinierung mit funktionalen Nanopartikeln.”
frequency magnetic fields (Hu et al. 2008), ultra- The authors acknowledge that this work was supported by
sound (Huang 2008), radiofrequency (Brazel 2009), a grant funded by the German Ministry of Education and
Research (BMBF, grant no, 13N9197). Furthermore, we
light (Pissuwan et al. 2011), and pH drifts (Zhu thank Ulrich Schäfer and Claus-Michael Lehr (University
et al. 2005). Controlled drug release could also be of Saarbrücken) for providing particle preparations and
observed after utilizing CdS nanoparticles as caps Reinhardt Renneberg (Hong Kong University of Science
for mesoporous channels and disulfide bond-reduc- and Technology) for providing the particles for the trig-
gered release experiments.
ing molecules physically blocking the drugs of cer-
tain sizes from leaching out (Lai et al. 2003). A new
promising concept is also the application of gold
nanoparticles in combination with near-infrared References
light. Gold nanorods have an absorption band in the
near-infrared region and convert absorbed light Barry BW (2002) Drug delivery routes in skin: a novel
energy into heat and can therefore act as a controller approach. Adv Drug Deliv Rev 54(Suppl 1):S31–S40
Blume-Peytavi U, Vogt A (2011) Human hair
of a drug release system when combined with near- follicle: reservoir function and selective
infrared light (Yamashita et al. 2011). targeting. Br J Dermatol 165(Suppl 2):13–17.
Most of these approaches have still to be veri- doi:10.1111/j.1365-2133.2011.10572.x
fied in combination with particles in the follicular Blume-Peytavi U, Massoudy L, Patzelt A, Lademann J,
Dietz E, Rasulev U, Garcia Bartels N (2010) Follicular
situation, which will certainly be a topic of future and percutaneous penetration pathways of topically
investigations. applied minoxidil foam. Eur J Pharm Biopharm
76(3):450–453. doi:10.1016/j.ejpb.2010.06.010
Conclusion Brazel CS (2009) Magnetothermally-responsive nano-
materials: combining magnetic nanostructures and
The optimization of drug delivery to and via the thermally-sensitive polymers for triggered drug
hair follicle is getting more and more important release. Pharm Res 26(3):644–656. doi:10.1007/
as the hair follicle offers target sites of thera- s11095-008-9773-2
peutic interest and represents a rapid access to Caspers PJ, Lucassen GW, Puppels GJ (2003) Combined
in vivo confocal Raman spectroscopy and confocal
the deeper skin layers and the circulation if microscopy of human skin. Biophys J 85(1):572–580.
transfollicular penetration is feasible. Current doi:10.1016/S0006-3495(03)74501-9
aspects of optimized follicular drug delivery Dixit N, Bali V, Baboota S, Ahuja A, Ali J (2007)
involve the adaptation of particulate carrier sys- Iontophoresis – an approach for controlled drug deliv-
ery: a review. Curr Drug Deliv 4(1):1–10
tems which have been demonstrated to deliver Fabin B, Touitou E (1991) Localization of lipophilic
substances preferably deep into the hair follicle molecules penetrating rat skin in vivo by quantitative
without allowing transfollicular penetration, autoradiography. Int J Pharm 74(1):59–65.
yet. New approaches specifically aim at the doi:10.1016/0378-5173(91)90408-G
Helmstadter A (2011) Endermatic, epidermatic,
development of controlled drug release systems enepidermatic-the early history of penetration enhancers.
where the particles only serve as transporters Int J Pharm 416(1):12–15. doi:10.1016/j.ijpharm.
deep into the hair follicle. Here, the active drug 2011.06.005
52 A. Patzelt and J. Lademann

Hu SH, Liu TY, Huang HY, Liu DM, Chen SY (2008) skin. Skin Pharmacol Physiol 24(6):305–311.
Magnetic-sensitive silica nanospheres for controlled doi:10.1159/000328728
drug release. Langmuir 24(1):239–244. doi:10.1021/ Lawson LB, Freytag LC, Clements JD (2007) Use of
la701570z nanocarriers for transdermal vaccine delivery. Clin
Huang SL (2008) Liposomes in ultrasonic drug and gene Pharmacol Ther 82(6):641–643. doi:10.1038/sj.
delivery. Adv Drug Deliv Rev 60(10):1167–1176. clpt.6100425
doi:10.1016/j.addr.2008.03.003 Mak WC, Richter H, Patzelt A, Sterry W, Lai KK,
Huang Y, Yu F, Park YS, Wang J, Shin MC, Chung HS, Renneberg R, Lademann J (2011) Drug delivery into
Yang VC (2010) Co-administration of protein drugs the skin by degradable particles. Eur J Pharm Biopharm
with gold nanoparticles to enable percutaneous deliv- 79(1):23–27. doi:10.1016/j.ejpb.2011.03.021
ery. Biomaterials 31(34):9086–9091. doi:10.1016/j. Mak WC, Patzelt A, Richter H, Renneberg R, Lai KK, Ruhl
biomaterials.2010.08.046 E, Sterry W, Lademann J (2012) Triggering of drug
Krause K, Foitzik K (2006) Biology of the hair follicle: release of particles in hair follicles. J Control Release
the basics. Semin Cutan Med Surg 25(1):2–10. 160(3):509–514. doi:10.1016/j.jconrel.2012.04.007
doi:10.1016/j.sder.2006.01.002 Meidan VM (2010) Methods for quantifying intrafollicu-
Krishnan G, Edwards J, Chen Y, Benson HA (2010) lar drug delivery: a critical appraisal. Expert Opin
Enhanced skin permeation of naltrexone by pulsed Drug Deliv 7(9):1095–1108. doi:10.1517/17425247.2
electromagnetic fields in human skin in vitro. J Pharm 010.503954
Sci 99(6):2724–2731. doi:10.1002/jps.22024 Morgan AJ, Lewis G, Van den Hoven WE, Akkerboom PJ
Labouta HI, Schneider M (2013) Interaction of inor- (1993) The effect of zinc in the form of erythromycin-
ganic nanoparticles with the skin barrier: current zinc complex (Zineryt lotion) and zinc acetate on
status and critical review. Nanomedicine 9(1):39–54. metallothionein expression and distribution in hamster
doi:10.1016/j.nano.2012.04.004 skin. Br J Dermatol 129(5):563–570
Labouta HI, el-Khordagui LK, Kraus T, Schneider M Mortensen LJ, Oberdorster G, Pentland AP, Delouise LA
(2011) Mechanism and determinants of nanoparticle (2008) In vivo skin penetration of quantum dot
penetration through human skin. Nanoscale nanoparticles in the murine model: the effect of
3(12):4989–4999. doi:10.1039/c1nr11109d UVR. Nano Lett 8(9):2779–2787. doi:10.1021/
Lademann J, Richter H, Schaefer UF, Blume-Peytavi U, nl801323y
Teichmann A, Otberg N, Sterry W (2006) Hair follicles – Ohyama M (2007) Hair follicle bulge: a fascinating reser-
a long-term reservoir for drug delivery. Skin Pharmacol voir of epithelial stem cells. J Dermatol Sci 46(2):81–
Physiol 19(4):232–236. doi:10.1159/000093119 89. doi:10.1016/j.jdermsci.2006.12.002
Lademann J, Richter H, Teichmann A, Otberg N, Blume- Otberg N, Richter H, Knuttel A, Schaefer H, Sterry W,
Peytavi U, Luengo J, Weiss B, Schaefer UF, Lehr Lademann J (2004a) Laser spectroscopic methods for
CM, Wepf R, Sterry W (2007) Nanoparticles–an effi- the characterization of open and closed follicles. Laser
cient carrier for drug delivery into the hair follicles. Phys Lett 1(1):46–49. doi:10.1002/lapl.200310011
Eur J Pharm Biopharm 66(2):159–164. doi:10.1016/j. Otberg N, Richter H, Schaefer H, Blume-Peytavi
ejpb.2006.10.019 U, Sterry W, Lademann J (2004b) Variations
Lademann J, Patzelt A, Richter H, Antoniou C, Sterry W, of hair follicle size and distribution in differ-
Knorr F (2009) Determination of the cuticula thick- ent body sites. J Invest Dermatol 122(1):14–19.
ness of human and porcine hairs and their poten- doi:10.1046/j.0022-202X.2003.22110.x
tial influence on the penetration of nanoparticles Otberg N, Patzelt A, Rasulev U, Hagemeister T, Linscheid
into the hair follicles. J Biomed Opt 14(2):021014. M, Sinkgraven R, Sterry W, Lademann J (2008) The
doi:10.1117/1.3078813 role of hair follicles in the percutaneous absorption of
Lademann J, Richter H, Meinke M, Sterry W, Patzelt A caffeine. Br J Clin Pharmacol 65(4):488–492.
(2010) Which skin model is the most appropriate for doi:10.1111/j.1365-2125.2007.03065.x
the investigation of topically applied substances into Paliwal S, Menon GK, Mitragotri S (2006) Low-frequency
the hair follicles? Skin Pharmacol Physiol 23(1):47– sonophoresis: ultrastructural basis for stratum
52. doi:10.1159/000257263 corneum permeability assessed using quantum dots.
Lai CY, Trewyn BG, Jeftinija DM, Jeftinija K, Xu S, J Invest Dermatol 126(5):1095–1101. doi:10.1038/
Jeftinija S, Lin VS (2003) A mesoporous silica sj.jid.5700248
nanosphere-based carrier system with chemically Papakostas D, Rancan F, Sterry W, Blume-Peytavi U, Vogt
removable CdS nanoparticle caps for stimuli- A (2011) Nanoparticles in dermatology. Arch Dermatol
responsive controlled release of neurotransmitters and Res 303(8):533–550. doi:10.1007/s00403-011-1163-7
drug molecules. J Am Chem Soc 125(15):4451–4459. Patzelt A, Richter H, Buettemeyer R, Huber HJ, Blume-
doi:10.1021/ja028650l Peytavi U, Sterry W, Lademann J (2008) Differential
Lange-Asschenfeldt B, Marenbach D, Lang C, Patzelt A, stripping demonstrates a significant reduction of the
Ulrich M, Maltusch A, Terhorst D, Stockfleth E, hair follicle reservoir in vitro compared to in vivo. Eur
Sterry W, Lademann J (2011) Distribution of bacteria J Pharm Biopharm 70(1):234–238. doi:10.1016/j.
in the epidermal layers and hair follicles of the human ejpb.2008.02.024
5 The Increasing Importance of the Hair Follicle Route in Dermal and Transdermal Drug Delivery 53

Patzelt A, Richter H, Knorr F, Schafer U, Lehr CM, Dahne L, Toll R, Jacobi U, Richter H, Lademann J, Schaefer H,
Sterry W, Lademann J (2011) Selective follicular target- Blume-Peytavi U (2004) Penetration profile of
ing by modification of the particle sizes. J Control Release microspheres in follicular targeting of terminal
150(1):45–48. doi:10.1016/j.jconrel.2010.11.015 hair follicles. J Invest Dermatol 123(1):168–176.
Pissuwan D, Niidome T, Cortie MB (2011) The forthcom- doi:10.1111/j.0022-202X.2004.22717.x
ing applications of gold nanoparticles in drug and gene Tsujimoto H, Hara K, Tsukada Y, Huang CC, Kawashima
delivery systems. J Control Release 149(1):65–71. Y, Arakaki M, Okayasu H, Mimura H, Miwa N (2007)
doi:10.1016/j.jconrel.2009.12.006 Evaluation of the permeability of hair growing ingre-
Ridolfi DM, Marcato PD, Justo GZ, Cordi L, Machado dient encapsulated PLGA nanospheres to hair follicles
D, Duran N (2012) Chitosan-solid lipid nanoparticles and their hair growing effects. Bioorg Med Chem Lett
as carriers for topical delivery of tretinoin. Colloids 17(17):4771–4777. doi:10.1016/j.bmcl.2007.06.057
Surf B: Biointerfaces 93:36–40. doi:10.1016/j. Ulmer M, Patzelt A, Vergou T, Richter H, Muller G,
colsurfb.2011.11.051 Kramer A, Sterry W, Czaika V, Lademann J (2012) In
Rogers GE (2004) Hair follicle differentiation and regula- vivo investigation of the efficiency of a nanoparticle-
tion. Int J Dev Biol 48(2–3):163–170. doi:10.1387/ emulsion containing polihexanide on the human skin.
ijdb.021587gr Eur J Pharm Biopharm. doi:10.1016/j.ejpb.2012.11.011
Rolland A, Wagner N, Chatelus A, Shroot B, Schaefer H Upadhyay P (2006) Enhanced transdermal-immuniza-
(1993) Site-specific drug delivery to pilosebaceous tion with diphtheria-toxoid using local hyperther-
structures using polymeric microspheres. Pharm Res mia. Vaccine 24(27–28):5593–5598. doi:10.1016/j.
10(12):1738–1744 vaccine.2006.04.039
Schaefer H, Lademann J (2001) The role of follicular pen- Vogt A, Hadam S, Heiderhoff M, Audring H, Lademann J,
etration. A differential view. Skin Pharmacol Appl Sterry W, Blume-Peytavi U (2007) Morphometry of
Skin Physiol 14(Suppl 1):23–27, doi: 56386 human terminal and vellus hair follicles. Exp Dermatol
Senzui M, Tamura T, Miura K, Ikarashi Y, Watanabe Y, 16(11):946–950. doi:10.1111/j.1600-0625.2007.00602.x
Fujii M (2010) Study on penetration of titanium diox- Vogt A, Mahe B, Costagliola D, Bonduelle O, Hadam S,
ide (TiO(2)) nanoparticles into intact and damaged Schaefer G, Schaefer H, Katlama C, Sterry W, Autran
skin in vitro. J Toxicol Sci 35(1):107–113 B, Blume-Peytavi U, Combadiere B (2008)
Shim J, Seok Kang H, Park WS, Han SH, Kim J, Chang IS Transcutaneous anti-influenza vaccination promotes
(2004) Transdermal delivery of minoxidil with both CD4 and CD8 T cell immune responses in
block copolymer nanoparticles. J Control Release humans. J Immunol 180(3):1482–1489
97(3):477–484. doi:10.1016/j.jconrel.2004.03.028 Yamashita S, Fukushima H, Niidome Y, Mori T, Katayama
Teichmann A, Jacobi U, Ossadnik M, Richter H, Koch S, Y, Niidome T (2011) Controlled-release system medi-
Sterry W, Lademann J (2005) Differential stripping: deter- ated by a retro Diels-Alder reaction induced by the
mination of the amount of topically applied substances photothermal effect of gold nanorods. Langmuir
penetrated into the hair follicles. J Invest Dermatol 27(23):14621–14626. doi:10.1021/la2036746
125(2):264–269. doi:10.1111/j.0022-202X.2005.23779.x Zhang LW, Monteiro-Riviere NA (2008) Assessment of
Teichmann A, Otberg N, Jacobi U, Sterry W, Lademann J quantum dot penetration into intact, tape-stripped,
(2006) Follicular penetration: development of a method abraded and flexed rat skin. Skin Pharmacol Physiol
to block the follicles selectively against the penetration 21(3):166–180. doi:10.1159/000131080
of topically applied substances. Skin Pharmacol Zhu Y, Shi J, Shen W, Dong X, Feng J, Ruan M, Li Y
Physiol 19(4):216–223. doi:10.1159/000093117 (2005) Stimuli-responsive controlled drug release from
Thiboutot D (2004) Regulation of human sebaceous a hollow mesoporous silica sphere/polyelectrolyte
glands. J Invest Dermatol 123(1):1–12. multilayer core-shell structure. Angew Chem Int Ed
doi:10.1111/j.1523-1747.2004.t01-2-.x Engl 44(32):5083–5087. doi:10.1002/anie.200501500
The Correlation Between
Transepidermal Water Loss 6
and Percutaneous Absorption

Jacquelyn Levin and Howard I. Maibach

Contents 6.1 Introduction


6.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . 55
6.1.1 What Is Transepidermal Water Loss? . . . . . 55 6.1.1 What Is Transepidermal
6.1.2 What Is Percutaneous Absorption? . . . . . . . 56 Water Loss?
6.1.3 What Is the Significance of a Correlation
Between TEWL and Percutaneous
Absorption? . . . . . . . . . . . . . . . . . . . . . . . . . 56 Transepidermal water loss (TEWL) is the
outward diffusion of water through the skin
6.2 Pertinent Studies Investigating the
Correlation Between TEWL and (Oestmann et al. 1993). An evaporimeter
Percutaneous Absorption . . . . . . . . . . . . . 56 determines TEWL by measuring the pressure
6.3 Discussion of the Assumptions Made
gradient of the boundary layer resulting from
the Studies Investigating the Correlation the water gradient between the skin surface
Between TEWL and Percutaneous and ambient air. TEWL measurements can
Absorption . . . . . . . . . . . . . . . . . . . . . . . . . 61 reflect the general health of the skin via the
6.3.1 Using In Vitro Methods to Model
In Vivo Experiments . . . . . . . . . . . . . . . . . . 61
assessment of skin barrier function and also
6.3.2 Using Animal Skin to Model assess treatment effectiveness or skin barrier
Human Skin . . . . . . . . . . . . . . . . . . . . . . . . 62 repair by monitoring the change in TEWL
6.3.3 Differences in TEWL Measurement over time (Nilsson. 1997; Pinnagoda et al.
Methods. . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
6.3.4 Influences of Percutaneous Absorption
1990). However, TEWL measurements cannot
Measurement Methods . . . . . . . . . . . . . . . . 64 be simply compared across multiple experi-
6.3.5 Influence of the Lipophilicity or ments. TEWL measurements are subject intra-
Hydrophilicity of the Compound individual variation based on the anatomic site
Studied TEWL Measurement Methods . . . 65
where the TEWL was measured and interin-
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65 dividual variation based on the extent of skin
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66 perspiration and skin surface temperature of the
individual tested. In addition, TEWL measure-
J. Levin, DO (*) ments can be affected by experimental condi-
Department of Dermatology, tions such as the air convection, the ambient
Largo Medical Center, Largo, FL, USA air temperature and air humidity of the room
e-mail: jlevin@hotmail.com
where the TEWL measurement was taken,
H.I. Maibach, MD and the method and type of instrument used to
Department of Dermatology, University of California
School of Medicine, UCSF, San Francisco, CA, USA
measure TEWL. Although TEWL can be influ-
e-mail: himjlm@itsa.ucsf.edu enced by many variables, experiments show

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 55


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_6, © Springer-Verlag Berlin Heidelberg 2015
56 J. Levin and H.I. Maibach

that evaporimeter measurements generally are investigating the correlation between TEWL and
reproducible in vitro and in vivo (Pinnagoda percutaneous absorption were reviewed. Of the
et al. 1989, 1990; Elkeeb et al. 2010; Fluhr nine studies reviewed, a majority demonstrated
et al. 2006). a significant quantitative correlation, and a few
found no quantitative correlation. At that time it
was thought that the correlation between TEWL
6.1.2 What Is Percutaneous and percutaneous absorption may not hold for
Absorption? in vitro experimentation models, extremely
lipophilic compounds, or possibly experiments
Percutaneous absorption refers to the rate of performed on animal skin. Since then, several
absorption of a topically applied chemical through other studies have been published investigating
the skin. A compound’s absorption rate is impor- the relationship between TEWL and percutane-
tant for determining the effectiveness and/or ous absorption using a very lipophilic compound
potential toxicity of topically applied compounds. (Hui et al. 2012), in vitro models (Elkeeb et al.
Since many topical formulations are used on dis- 2010; Hui et al. 2012; Elmahjoubi et al. 2009),
eased skin, where the integrity of the permeabil- and animal skin (Elmahjoubi et al. 2009), and all
ity barrier is in doubt, the dose absorbed into the studies have demonstrated a significant quantita-
body could vary greatly (Bronaugh and Stewart tive correlation.
1986). One rate-limiting step of a compound’s In the next section, we review 12 studies
absorption through the skin is the rate of diffusion investigating the correlation between TEWL and
through the stratum corneum (SC). The rate of percutaneous absorption.
absorption through the SC cannot be described by
a zero- or first-order mathematical rate equation
because the SC is a complex system variable in 6.2 Pertinent Studies
its penetration properties. Many factors contribute Investigating the Correlation
to the percutaneous absorption of a given chemi- Between TEWL
cal, such as methodology (including the effects of and Percutaneous
application time, method of measurement), physi- Absorption
cochemical properties of the topical compound,
interindividual variation (including the effects of Oestmann et al. (1993) investigated a correla-
skin condition, age of individual, and blood flow), tion between TEWL and hexyl nicotinate (HN)
and intra-individual variation (including the dif- penetration parameters in man. Penetration of
ferences between anatomic sites) (Noonan and HN was indirectly measured by means of laser
Gonzalez 1990; Wester 1993). Doppler flowmetry (LDF), which quantifies the
increase in cutaneous blood blow (CBF) caused
by penetration of HN, being a vasoactive sub-
6.1.3 What Is the Significance stance. Lipophilic HN was chosen over hydro-
of a Correlation Between TEWL philic methyl nicotinate because HN is a slower
and Percutaneous penetrant, hence, making it easier to distinguish
Absorption? an intact barrier from an impaired barrier.
LDF parameter initial response time (to) and
The extensive procedure required to measure per- the time to maximum response (tmax) were com-
cutaneous absorption versus TEWL enhances the pared with corresponding TEWL values, and a
desire to find a correlation between the two mea- weak quantitative negative correlation was found
surements in order to more easily assess skin bar- (r = −0.31, r = −0.32). This correlation suggests
rier function and should aid in the understanding that when an individual’s response time, to, was
and development of penetration enhancers. In a fast, the skin barrier was impaired. The weak
review by Levin and Maibach in 2005, nine studies negative correlation found that maybe LDF is not
6 The Correlation Between Transepidermal Water Loss and Percutaneous Absorption 57

as reproducible as other methods of measuring the lag phase (not a steady state measurement),
percutaneous absorption. Further research should and this study suggested that these two meth-
investigate this weak correlation between TEWL ods, because they are measuring different things,
and penetration of HN. should not be considered as exchangeable alter-
Lamaud et al. (1984) investigated whether natives but rather as complementary tests to
TEWL correlated to the percutaneous absorption assess barrier function. On the basis of results of
of the lipophilic compounds (hydrocortisone). this chapter, however, it could be concluded that
Penetration of 1 % hydrocortisone and TEWL TEWL and HN penetration injunction are suit-
rates were recorded for the hairless rats in vivo able methods to monitor skin barrier function in
before and after UV irradiation (660 J/cm2). keratinization disorders.
The results demonstrated a correlation between Rougier et al. (1988) attempted to establish
TEWL and the percutaneous absorption of hydro- the relationship between the barrier properties
cortisone both before and after UV irradiation for of the horny layer using percutaneous absorp-
application periods up to 1 h. In part two of the tion and TEWL measurements and discern the
experiment, drug penetration was evaluated by surface area of the corneocytes according to ana-
urinary excretion 5 days after a single 24 h appli- tomic site, age, and sex in man.
cation of hydrocortisone on normal, stripped, or The penetration of benzoic acid (BA) was
UV-irradiated skin of hairless rats. In this experi- measured in vivo at seven anatomic sites and
ment the quantity of the drug eliminated corre- compared to its TEWL value measured on the
lated with the level of TEWL for up to 2 days contralateral site. The amount of BA penetrated
for all skin conditions suggesting that TEWL can was measured through urinary extraction up to
predict the changes of skin permeability to lipo- 24 h after application. It was discovered that irre-
philic drugs in normal and damaged skin. spective of the anatomic site and gender, a linear
Lavrijsen et al. (1993) characterized the SC bar- relationship (r = 0.92, p < 0.001) existed between
rier function in patients with various keratinization total penetration of BA and TEWL.
disorders using two noninvasive methods: mea- Comparing corneocyte surface area to perme-
suring outward transport of water through skin ability, Rougier et al. (1988) also found a general
by evaporimetry, i.e., TEWL, and the vascular correlation of increasing permeability for both
response to HN penetration into the skin deter- water and BA with decreasing corneocyte size.
mined by LDF. Three of the five types of keratini- The smaller the volume of the corneocyte, the
zation disorders studied, i.e., autosomal dominant greater the intercellular space available to act as
ichthyosis vulgaris, X-linked recessive ichthyosis, a reservoir for topically applied molecules, result-
and autosomal recessive congenital ichthyosis, ing in a higher absorption (Dupuis et al. 1984).
have impaired barrier function and are a type of This thinking is in accord with other studies who
ichthyosis, while for the other two keratinization have shown that the smaller the capacity of the res-
disorders studied, dyskeratosis follicularis and ervoir, the less the molecule is absorbed (Dupuis
erythrokeratoderma variabilis, there were no prior et al. 1984; Rougier et al. 1983, 1985, 1987a, b).
information available on barrier impairment. In In order to determine the influence of age on cor-
this experiment the two methods of barrier func- neocyte size, Rougier et al. (1988) investigated the
tion assessment, TEWL and LDF, were correlated corneocyte size in the upper-outer arm for three
for all skin diseases and normal skin as a control. groups of six to eight male volunteers: (1) 20–30,
TEWL measurements and the LDF param- (2) 45–55, and (3) 65–80 years. No variation in
eter, to, showed a high negative correlation in corneocyte size up to 55 years was observed.
those with skin disease (r = −0.64) and a weaker The mean corneocyte size for the 20–30-year
negative correlation among the control healthy cohort was 980 ± 34 μm2, and for the 45–55-year
skin group (r = −0.39). Because TEWL reflects cohort, a value of 994 ± 56 μm2 was recorded.
the steady state flux of a compound across SC The group aged 65–80 years did, however, show
and parameter, to is a function of the duration of significantly larger corneocytes (1,141 ± 63 μm2)
58 J. Levin and H.I. Maibach

relative to the other groups. Relatively small dermatitis. All the subjects had widespread
numbers of subjects were used by Rougier et al. dermatitis covering at least 60 % of the total skin
(1988) which may explain the discrepancies when area. Plasma cortisol concentrations were mea-
compared with data from other more recent stud- sured by radioimmunoassay before and 2 and
ies (Leveque et al. 1984). Generally it is now 4 h after hydrocortisone application. TEWL was
understood that corneocytes generally increase in measured in six standard skin areas immediately
size with age (Machado et al. 2010) and TEWL before application of the hydrocortisone cream.
and percutaneous absorption also increases with Each individual TEWL value was calculated as a
age (Roskos and Guy 1989); therefore, it seems mean of these six measurements.
that corneocyte size cannot explain the permeabil- The concordance between the post application
ity changes in mature skin. increment in plasma cortisol and the mean TEWL
Rougier et al. (1988) used a detergent scrub tech- was highly significant resulting in a correlation
nique to collect corneocytes at different anatomic coefficient of r = 0.991 (p < 0.001). In conclusion
sites from a group of six to eight male volunteers, this study found a highly significant correlation
aged 20–30 years. The rank order of the corneocyte between TEWL and percutaneous absorption of
surface area was forearm (ventral elbow) = forearm hydrocortisone.
(ventral-mid) = arm (upper-outer) = abdomen > Tsai et al. (2001) investigated the relation-
forearm (ventral-wrist) > postauricular > forehead. ship between the permeability barrier disrup-
However when Rougier et al. (1988) investigated tion and the percutaneous absorption of various
corneocyte size by anatomic site, he found that for compounds with different lipophilicity. Acetone
certain anatomic sites where corneocyte size was treatment was used in vivo on hairless mice to
similar (980–1,000 μm2), there were large differ- disrupt the normal permeability barrier, and
ences in permeability. Therefore, while percutane- in vivo TEWL measurements were used to gauge
ous absorption and TEWL are quantitatively barrier disruption. The hairless mouse skin was
correlated, corneocyte size only partially explains then excised and placed in diffusion cells for the
the difference in permeability between the different in vitro percutaneous absorption measurements
anatomic sites and different age of the skin. of five model compounds: sucrose, caffeine,
Lotte et al. (1987) examined the relationship hydrocortisone, estradiol, and progesterone. The
between the percutaneous penetration of four partition coefficient or lipophilicity of these com-
chemicals (acetylsalicylic acid, benzoic acid, caf- pounds and compounds used in the subsequent
feine, and sodium salt of benzoic acid) and TEWL studies are summarized in Table 6.1.
in man as a function of anatomic site. The amount The permeability barrier disruption by acetone
of chemical penetrated was measured by urinary treatment and TEWL measurements significantly
excretion for up to 24 h after application. For a correlated with the percutaneous absorption of
given anatomic site, the permeability varied widely the hydrophilic and lipophilic drugs sucrose,
in relation to the nature of the molecule adminis- caffeine, and hydrocortisone. However acetone
tered due to the physicochemical interactions treatment did not alter the percutaneous penetra-
which occur between the molecule, vehicle, and tion of the highly lipophilic compounds estradiol
SC. For all anatomic sites investigated, irrespec- and progesterone, hence, suggesting that there is
tive of the physicochemical properties of the mol- no correlation between TEWL and the percutane-
ecules administered, there was a linear relationship ous absorption of highly lipophilic compounds.
between TEWL and percutaneous absorption. The results imply the need to use both TEWL and
Aalto-Korte et al. (1993) attempted to find drug lipophilicity to predict alterations in skin
the precise relationship between TEWL and permeability.
percutaneous absorption of hydrocortisone in Chilcott et al. (2002) investigated the relation-
patients with active dermatitis. Percutaneous ship between TEWL and skin permeability to
absorption of hydrocortisone and TEWL was tritiated water (3H2O) and the lipophilic sulfur
studied in three children and six adults with mustard (35SM) in vitro. No correlation was found
6 The Correlation Between Transepidermal Water Loss and Percutaneous Absorption 59

between basal TEWL rates and the permeability open and closed chamber evaporimeters in the
of human epidermal membrane to 3H2O (p = 0.72) evaluation of skin barrier function.
or sulfur mustard (p = 0.74). Similarly, there was Atrux-Tallau et al. (2007) demonstrated sig-
no correlation between TEWL rates and the 3H2O nificant correlation between TEWL and the per-
permeability on full-thickness pig skin (p = 0.68). cutaneous absorption of caffeine (a hydrophilic
There was also no correlation between TEWL compound) during an ex vivo experiment on heat
rates and 3H2O permeability following up to 15 separated epidermis and dermatomed human
tape strips (p = 0.64) or up to four needle stick skin (p < 0.001, r2 = 0.88). Since caffeine is a
punctures (p = 0.13). Taken together these results hydrophilic compound and has a relatively small
from this experiment indicate that under these molecular weight of 194 Da, it was not surpris-
experimental circumstances (i.e., in vitro human ing to the authors that the permeation behavior
and pig skin) TEWL cannot be used as a measure resembles that of tritiated water (22 Da).
of the skin’s permeability to topically applied Hui et al. (2012) investigated the correlation
lipophilic or hydrophilic compounds. between TEWL and the percutaneous absorp-
Elkeeb et al. (2010) compared TEWL to the tion of clonidine (a lipophilic compound) and
percutaneous absorption/flux rate of 3H2O in 3
H2O (a hydrophilic compound) in in vitro human
in vitro dermatomed clinically healthy human cadaver skin. The partition coefficient of cloni-
cadaver skin using three different evaporimeters dine is reported in Table 6.1. TEWL measure-
to measure TEWL. Measurements were taken ments were made with a closed chamber TEWL
at baseline (i.e., at the start of the experiment) meter (AquaFlux AF200). With the goal of dis-
and then again at several time points over 24 h. cerning the potential differences in the correla-
The evaporimeters included an open chamber tion between TEWL and lipophilic clonidine, the
evaporimeter A (TEWameter® TM 210 (Courage correlation between TEWL and hydrophilic 3H2O
and Khazaka, Cologne, Germany)) and two percutaneous absorption and general differences
closed chamber evaporimeters B (VapoMeter™ in the percutaneous absorption of clonidine and
3
(Delfin Technologies, Kuopio, Finland)) and C H2O, the flux rate, skin distribution, and total
(AquaFlux AF200, Biox Systems, Ltd, London, amount of absorption for clonidine and tritiated
UK). Open chamber evaporimeters are open to water were recorded and compared. Statistical
the ambient air, while closed chamber evaporim- analysis indicated that the baseline TEWL values
eters are closed systems that are not open to the weakly correlated with the flux of [14C]-clonidine
environment. There has been controversy over (p < 0.03, r2 = 0.36) and 3H2O (r2 = 0.34, p = 0.04).
the years as to whether open and closed chamber The correlation between fluxes of 3H2O and
evaporimeters are equivalent in given accurate and [14C]-clonidine was moderate (correlation coef-
precise TEWL measurements TEWL. Baseline ficient = 0.675, p < 0.001). In addition, TEWL and
TEWL measurements with evaporimeters A permeation data of 3H2O expressed as a percent
(p = 0.04, r2 = 0.34) and C (p = 0.00, r2 = 0.50) dose of the amount in the receptor fluid correlated
correlated with the percutaneous absorption or well throughout the experiment. However, the
flux rate of tritiated H2O, while evaporimeter permeation curve of [14C]-clonidine as a percent
B showed no statistically significant correla- dose in the receptor fluid differed from that of
tion (p = 0.07, r2 = 0.31). However, the pattern of 3
H2O and TEWL. The difference in the curves is
changing TEWL values over 24 h was similar to likely secondary to differences in the hydrophilic/
that of the percutaneous absorption or tritiated lipophilic properties of clonidine versus water.
water flux for all three evaporimeters A, B, and Therefore as Hui suggests, it may be necessary
C (p = 0.04, r2 = 0.34,). The reason why evapo- to combine the TEWL values with factors such as
rimeter B showed no significant correlation for molecular weight and/or hydrophilicity/lipophi-
baseline TEWL measurement remains unknown. licity to gauge percutaneous absorption.
Elkeeb et al. (2010) state that the results of this Elmahjoubi et al. (2009) investigated TEWL
experiment imply the validity of using both (using the AquaFlux evaporimeter) and the
60 J. Levin and H.I. Maibach

Table 6.1 A summary of the compounds used in the correlation studies, their octanol-water partition coefficient,
solubility classification, and whether or not their percutaneous absorption correlated with TEWL (Oestmann et al. 1993;
Nilsson 1997; Elkeeb et al. 2010; Hui et al. 2012; Elmahjoubi et al. 2009; Lamaud et al. 1984; Lavrijsen et al. 1993;
Rougier et al. 1988; Lotte et al. 1987; Aalto-Korte et al. 1987; Tsai et al. 2001; Chilcott et al. 2002; Atrux-Tallau et al.
2007)
Partition coefficient
Compound (log Poctanol/water) Classification Correlation
Sucrose −3.7 Hydrophilic Yes
Caffeine −0.02 Hydrophilic Yes
Water 1 Hydrophilic Yes
Acetylsalicylic acid 1.13 Hydrophilic Yes
Sulfur mustard 1.37 Lipophilic No
Hydrocortisone 1.5 Lipophilic Yes
Benzoic acid 1.87 Lipophilic Yes
Sodium benzoate 1.87 Lipophilic Yes
Estradiol 2.7 Highly lipophilic No
Progesterone 3.9 Highly lipophilic No
Hexyl nicotinate 4 Highly lipophilic Yes (weak)
Clonidine 5.4 Highly lipophilic Yes (weak)

percutaneous absorption/flux of 3H2O in full- surfactants of differing alkyl chain lengths were
thickness in vitro porcine skin both at baseline and applied to the full-thickness porcine skin in vitro to
after physical and chemical barrier disruption in determine if measuring TEWL values could discern
multiple different experiments. The aim of these between mild and severe perturbations to the barrier
experiments was to further investigate the rela- function. TEWL was largely unaffected following
tionship between TEWL and 3H2O flux using the cutaneous exposure to short and long alkyl chain
AquaFlux evaporimeter® (Bio Systems Ltd, USA) surfactants and, however, was significantly elevated
and to evaluate the use of porcine skin in vitro as a over control levels following exposure to those with
model to study the human skin barrier. intermediate chain lengths. Exposure to sodium
The first experiment investigated the relation- lauryl sulfate (SLS), with an intermediate 12 carbon
ship between basal TEWL rates and 3H2O flux in alkyl chain, produced the greatest increase in
in vitro healthy full-thickness porcine skin. The TEWL.
results showed that basal TEWL values were lin- In the fourth experiment, the effect of varying
early correlated with basal 3H2O flux values SLS concentration, volume, and contact time on
(r2 = 0.80, n = 63). the TEWL in vitro in porcine skin was measured.
The second experiment examined the effect of The results showed a linear trend between TEWL
physical barrier disruption with skin punctures and SLS concentration in the 0–1 % w/v con-
on TEWL measurements. The results did not centration range. However, following treatment
show a perfect correlation between skin punctures with 5 % w/v SLS, TEWL readings were only
and TEWL measurements. TEWL increased sig- slightly higher than those following treatment
nificantly after the first skin puncture and then with 1 % w/v surfactant. A linear correlation was
remained constant for punctures 2, 3, and 4. also demonstrated between TEWL and surfactant
Another large increase in TEWL was seen with solution volume (r2 = 0.87), which was statisti-
the fifth puncture. However no changes in TEWL cally significant (p < 0.01). TEWL also increased
values were seen with the sixth or seventh punc- as a function of increasing SLS treatment time,
ture suggesting that a threshold may have been when concentration was fixed at 1 % w/v and
reached after the fifth puncture. volume fixed at 200 μl.
The third and fourth experiments examined In conclusion, Elmahjoubi et al. (2009) found
TEWL changes after chemical barrier disruption that baseline TEWL values correlated with the per-
with surfactants. In the third experiment, anionic cutaneous absorption of 3H2O in vitro in healthy
6 The Correlation Between Transepidermal Water Loss and Percutaneous Absorption 61

porcine skin and the TEWL measurements linearly through the skin is a passive diffusion process and
correlated with the exposure of porcine skin in vitro the stratum corneum is nonliving tissue. Many
to increasing concentrations, time, and volumes of studies comparing in vivo and in vitro TEWL and
SLS. TEWL measurements did not demonstrate a percutaneous absorption measurements have been
linear correlation between skin punctures (i.e., skin conducted, and the results from those experiments
damage) and TEWL. The authors feel that TEWL support the contention that reliable measurements
measurements in vitro in porcine skin may serve as can be obtained from in vitro studies (Elkeeb
a model for future studies in this area in contrast to et al. 2010; Noonan and Gonzalez 1990; Hui et al.
the previous findings by Chilcott et al. (2002). 2012; Elmahjoubi et al. 2009; Nangia et al. 1993;
Brounaugh et al. 1982b). While the consensus is
that in vitro experiments are reasonable models
6.3 Discussion of the for in vivo human experiments, some experi-
Assumptions Made in the ments note significant differences between these
Studies Investigating the methods for measuring skin permeation. The
Correlation Between TEWL most significant study by Bronaugh and Stewart
and Percutaneous Absorption (1985) found that the effects of UV irradiation
could not be duplicated using an in vitro experi-
Many of the experiments investigating TEWL mentation model, hence, suggesting that in vitro
and percutaneous absorption make large assump- experiments examining the TEWL and percuta-
tions which could affect the results and hence neous absorption after barrier damage may not be
be the source of controversy. For example, Tsai an acceptable model for correlation with in vivo
et al. (2001) and Chilcott et al. (2002) assume that studies. In vitro damage to the SC barrier may
in vitro measurements of TEWL and percutaneous not be an accurate model to in vivo SC damage
absorption are equivalent to in vivo measurements, because in vivo exposure to skin irritants results
while Lamaud et al. (1984) assume that animal in a cascade of reactions that do not occur in vitro
skin may serve as a permeability model for human in human cadaver skin (Nangia et al. 1993).
skin. Great sources of error and variation can also Chilcott et al. (2002) investigated the correla-
be induced depending on the measurement device tion between TEWL and percutaneous absorption
used to record TEWL rates and the choice of the in vitro after inducing different types of barrier
compound and/or method used to measure per- damage. This was one of the rare studies which
cutaneous absorption rates. Because we do not did not observe a correlation between TEWL and
completely understand the qualitative relationship percutaneous absorption after barrier damage. It
between TEWL and percutaneous absorption, it is is possible that in vitro methodology in the exper-
hard to determine which assumptions made dur- imental design may be responsible for the lack of
ing the experiment could be affecting the correla- correlation of TEWL to skin damage reported in
tion results. This section investigates the probable this study. However, Fluhr et al. (2006) suggest
causes that could influence the results of the cor- that the conditions used in the study of Chilcott
relation experiments. Provided in Table 6.2 is a et al. (2002), i.e., the use of heat-split human epi-
summary of the major assumptions from 12 stud- dermis and non-pigmented pig skin that had been
ies discussed in this chapter. stored for up to 14 days and penetration studies
which extended over 96 h post-heat separation,
likely contributed to their results. Fluhr (2006)
6.3.1 Using In Vitro Methods states that the extracellular lipid matrix and cor-
to Model In Vivo Experiments neocytes of the SC were potentially compro-
mised from the heat separation. However, it is
Skin permeation can be measured in vivo or this author’s opinion that even if the barrier was
in vitro by using excised skin in diffusion cells. In compromised by heat separation, these changes
theory, studies using in vitro or ex vivo are feasible in barrier function should have been reflected
models for in vivo experiments because passage both in the TEWL and percutaneous absorption
62 J. Levin and H.I. Maibach

Table 6.2 A summary of the major assumptions made by the studies discussed in this chapter (Aalto-Korte et al. 1993;
Atrux-Tallau et al. 2007; Chilcott et al. 2002; Elkeeb et al. 2010; Hui et al. 2012; Elmahjoubi et al. 2009; Lamaud et al.
1984; Lavrijsen et al. 1993; Lotte et al. 1987; Nilsson 1997; Oestmann et al. 1993; Rougier et al. 1988; Tsai et al. 2001)
In vivo vs Percutaneous
in vitro absorption
(percutaneous measurement Healthy skin vs Correlation
Reference absorption)b Skin type method Compoundc damaged skin results
Oestmann et al. In vivo Human LDF Lipophilic Healthy Yes
(1993)
Lamaud et al. In vivo Animal Urinary Lipophilic Both Yes
(1984)
Lavrijsen et al. In vivo Human LDF Lipophilic Damaged Yes
(1993)
Rougier et al. In vivo Human Urinary Lipophilic Healthy Yes
(1988)
Lotte et al. In vivo Human Urinary Hydrophilic and Healthy Yes
(1987) lipophilic
Aalto-Korte In vivo Human Plasma cortisol Lipophilic Damaged Yes
et al. (1993) level
Tsai et al. In vitro Animal Diffusion cell Hydrophilic and Damaged Yes
(2001a)a lipophilic
Tsai et al. In vitro Animal Diffusion cell Highly lipophilic Damaged No
(2001b)a
Chilcott et al. In vitro Both Diffusion cell Hydrophilic and Both No
(2002) lipophilic
Elkeeb et al. In vitro Human Diffusion cell Hydrophilic Healthy Yes
(2010)
Hui et al. (2012) In vitro Human Diffusion cell Hydrophilic and Healthy Yes
lipophilic
Atrux-Tallau Ex vivo Human Diffusion cell Hydrophilic Healthy Yes
et al. (2007)
Elmahjoubi In vitro Animal Diffusion cell Hydrophilic Both Yes
et al. (2009)
a
Reference Tsai et al. was divided into two experiments in this table since the study found a correlation between TEWL
and percutaneous absorption with some compounds and no correlation with others
b
TEWL in vivo and in vitro measurements are considered equivalent. We are only concerned with how percutaneous
absorption measurements were performed
c
Compounds were classified by their octanol-water partition coefficient, log Koctanol/water. See Table 6.1. Compounds pos-
sessing log Koctanol/water values less than one are considered hydrophilic, while compounds with log Koctanol/water higher than
three were considered very lipophilic

and hence should have correlated if both mea- 6.3.2 Using Animal Skin to Model
sured variables truly reflect skin barrier function. Human Skin
However since Chilcott et al.’s original publi-
cation in 2002, many studies demonstrating the Comparing the skin morphology and absorption
correlation between TEWL and percutaneous of chemicals through human versus animal skin,
absorption have been conducted in in vitro mod- it is clear that human skin is unique in both
els (Elkeeb et al. 2010; Hui et al. 2012; aspects and should be used for the most meaning-
Elmahjoubi et al. 2009), and it is more likely that ful results (Bronaugh and Franz 1986). Yet an
the results of Chilcott et al. (2002) were an experiment by Bronaugh et al. (1982a) found that
exception rather than the rule. depending on the compound and the vehicle
6 The Correlation Between Transepidermal Water Loss and Percutaneous Absorption 63

used, permeability values obtained using animal use of animal skin as a model for experimentation
skin can be well within an order of magnitude of seems unlikely. However, further research may
the permeability values for human skin. be warranted.
Independently, in vitro methods and ani-
mal skin models prove to be reliable models to
predict percutaneous absorption in human skin 6.3.3 Differences in TEWL
in vivo. Therefore it seems logical to assume Measurement Methods
that the in vitro condition and the use of animal
skin may be used in unison to accurately model TEWL meters or evaporimeters can have an open
in vivo absorption through human skin. However or closed chamber system. Open chamber TEWL
Rougier et al. (1987a, b) documented a distinct meters are open to the environment, and therefore
difference between animal studies performed their measurements are influenced by environ-
in vivo versus animal studies performed in vitro mental factors such as room temperature or
when compared to the absorption of compounds humidity. Closed chamber devices are closed sys-
through human skin in vivo. This experiment tems that are not dependent on environmental
compared the permeability of human skin to the variables. With adequate control of environmental
hairless rat (Walker et al. 1983) and the hairless variables, open chamber TEWL meters can pro-
mouse (Bronaugh and Stewart 1986) skin using vide reliable and reproducible measurements that
molecules of widely different physicochemical are comparable to closed chamber TEWL meters
properties. The results show that on in vivo ani- (Pinnagoda et al. 1990, 1989; Elkeeb et al. 2010;
mal or human skin, for whatever the molecule Fluhr et al. 2006). Yet, only a limited number of
tested, the permeability ratios remained relatively comparisons between different types of TEWL
constant, while in vitro they do not. Therefore, meters have been described in the literature until
when application conditions are strictly identical the last few years, and TEWL meters are known
in humans and in animals, it may be possible to to differ in their measurement range, speed,
predict percutaneous absorption in human skin repeatability, and reproducibility (Hui et al. 2012).
in vivo by measuring in vivo absorption through Elkeeb et al. (2010) and Fluhr et al. (2006) per-
animal skin, but not using in vitro animal absorp- formed studies which exemplify the general com-
tion. The inaccurate results obtained when con- parability of TEWL meters, but also exemplify
ducting experiments in vitro using animal skin their differences. As mentioned in the previous
may have affected the results studies by Tsai section, Elkeeb et al. (2010) compared TEWL to
et al. (2001) and Chilcott et al. (2002) which the percutaneous absorption/flux rate of 3H2O in
were the only two studies using in vitro animal in vitro human cadaver skin using three differ-
skin and showing no correlation between TEWL ent evaporimeters: open chamber evaporimeter
and percutaneous absorption. A (TEWameter® TM 210, Courage and Khazaka,
However, Laumaud et al. (1984) con- Cologne, Germany; Acaderm Inc., Menlo Park,
ducted their study in porcine skin in vivo, and CA, USA) and two closed chamber evaporim-
Elmahjoubi et al. (2009) conducted their study in eters B (VapoMeter™, Delfin Technologies,
porcine skin in vitro, and both found a correlation Kuopio, Finland) and C (AquaFlux AF200, Biox
between TEWL and percutaneous absorption. Systems, Ltd, London, UK). TEWL values corre-
This suggests that other factors than using ani- lated at baseline and over the 24 h experiment for
mal in vitro model may have played a role in the evaporimeters A and C. However TEWL values
lack of correlation found in studies by Tsai et al. of evaporimeter B only correlated with evaporim-
(2001) and Chilcott et al. (2002). However, there eters A and C during the experiment and did not
is no doubt that there are distinct differences correlate at baseline.
between animal skin and human skin when used An experiment by Fluhr et al. (2006) com-
as a model for human absorption, whether these pared many different TEWL meters in vivo
differences are large enough to invalidate that the in human and murine skin and ex vivo in
64 J. Levin and H.I. Maibach

murine skin. TEWL rates obtained with two 6.3.4 Influences of Percutaneous
closed chamber systems (VapoMeter™ (Delfin Absorption Measurement
Technologies, Kuopio, Finland) and H4300 Methods
(NIKKISO YSI CO., Ltd, Tokyo, Japan)) and
one closed-loop system (MEECO; MEECO, The major factor affecting percutaneous absorp-
Warrington, PA, USA) under different experi- tion measurements is the used methodology
mental in vivo conditions were compared with (Bronaugh and Maibach 1989; Wester and
data from four open-loop instruments, i.e., Maibach 1992). Methods used for percutaneous
TEWameter® TM 210, TEWameter® TM 300 absorption measurements are not equal and hence
(Courage and Khazaka, Cologne, Germany), can give different results. Table 6.2 column 3 sum-
DermaLab (Cortex Technology, Hadsund, marizes the percutaneous absorption measurement
Denmark), and EP 1 (ServoMED, Stockholm, methodology used in these correlation studies.
Sweden). Through his experiments, Fluhr The most common method for determining
demonstrated the ability of most of TEWL percutaneous absorption in vivo is measuring the
meters to detect minor, moderate, and severe radioactivity of excreta following topical applica-
changes in barrier dysfunction; however, none tion of a labeled compound. Determination of
of the devices could detect minor improve- percutaneous absorption from urinary radioactiv-
ments in barrier function, and there were dif- ity does not account for metabolism by the skin
ferences in the TEWL meters’ ability to detect but has been proven to be a reliable method for
differences between severe and very severe absorption measurements and is widely accepted
barrier dysfunction. However, analysis of all as the “gold standard” when available.
the data collected demonstrated a weak cor- The most commonly used in vitro technique
relation between a few TEWL meters, but an involves placing excised skin in a diffusion
overall good correlation between all the TEWL chamber, applying radioactive compound to one
meters. side of the skin and then assaying the radioactiv-
An additional study by Farahmond et al. ity in the collection vessel on the other side of
(2009) found similar results to Fluhr et al. (2006) the skin (Bronaugh and Maibach 1991). The
when studying the differences between two advantages of using this in vitro technique are
closed chamber TEWL measurement instru- that the method is easy to use and that the results
ments. These instruments were designed based are obtained quickly. The disadvantage is that
on different measurement principles and demon- the fluid in the collection bath which bathes the
strated slight differences in their ability to detect skin is saline, which may be appropriate for
changes in skin barrier function despite that the studying hydrophilic compounds, but is not suit-
values of all three instruments correlated well able for hydrophobic compounds. If the parent
with each other (p < 0.001). compound is not adequately soluble in water,
These studies by Elkeeb et al. (2010), Fluhr then determining in vitro permeation into a water
et al. (2006), and Farahmond et al. (2009) reveal receptor fluid will be self-limiting.
that there are potential limitations to TEWL When conducting in vitro experiments, animal
meters in experimentation and the TEWL meter skin often substitutes human skin. Because ani-
must be chosen carefully based on the proposed mal skin has different permeability characteris-
study design. In general, TEWL meters pro- tics than human skin, one should be careful which
duced comparable and reliable results; however, type of animal skin is used (see section on animal
in both Elkeeb et al.’s (2010) and Fluhr et al.’s vs human skin). In addition, proper care should
(2006), experiments there were reported TEWL be taken in skin preparation of excised skin to not
measurements that did not significantly corre- damage the skin barrier integrity. Anatomic site
late with other measurements. These variations is also important, since the skin from different
in measurement have the potential to influence sites shows different permeability as well as
experimentation. using many different donor skin samples.
6 The Correlation Between Transepidermal Water Loss and Percutaneous Absorption 65

The only two experiments which did not find a independent of the physicochemical properties of
correlation between TEWL and percutaneous the compound applied. Namely, can TEWL mea-
absorption, Tsai et al. (2001) and Chilcott et al. surements predict the skin barrier’s permeability
(2002), were experiments that measured percuta- changes to both hydrophilic and very lipophilic
neous absorption in vitro. Perhaps using a diffu- compounds?
sion cell to measure percutaneous absorption is Correlation between TEWL and percutane-
the reason for not finding a correlation. ous absorption was found in many studies, such
Oestmann et al. (1993) and Lavrijsen et al. as Oestmann et al. (1993), Lamaud et al. (1984),
(1993) used laser Doppler flowmeter (LDF) to Lavrijsen et al. (1993), Lotte et al. (1987), Aalto-
measure HN penetration. LDF measures the Korte et al. (1993), Tsai et al. (2001a), Elkeeb et al.
increase in cutaneous blood flow (CBF) caused by (2010), Elmahjoubi et al. (2009), Hui et al. (2012),
the penetration of HN, a vasoactive substance. One and Atrux-Tallau et al. (2007), which suggest that
problem with this method is that LDF measure- TEWL can predict the changes in skin permeability
ments are not only dependent on the amount of HN to topically applied hydrophilic and lipophilic drugs.
absorbed but also on the individual’s vasoreactiv- However, Tsai et al. (2001b) found that the percuta-
ity, gender, and age. This may be the reason why neous absorption of the highly lipophilic progester-
Oestmann et al. (1993) and Lavrijsen et al. (1993) one and estradiol did not correlate with TEWL.
obtained only a weak correlation between TEWL The most common lipophilicity scale of mol-
and percutaneous absorption of HN. Another dis- ecules is defined by the octanol-water partition
advantage of this method is that LDF measure- coefficient (Koct/w = log (poct/w)). Presented in
ments have many sources of variation which make Table 6.1 are the compounds used in the afore-
it difficult to compare inter-laboratory results. mentioned studies, their octanol-water partition
coefficient, their solubility classification, and
whether or not their percutaneous absorption cor-
6.3.5 Influence of the Lipophilicity related with TEWL.
or Hydrophilicity Looking closely at Table 6.1, the highly lipo-
of the Compound Studied philic compounds were the compounds that dem-
onstrated a weaker correlation or no evidence of
The percutaneous absorption rate and/or total a correlation between percutaneous absorption
absorption of a compound varies greatly and TEWL, while the moderately lipophilic com-
depending on the compound and its lipophilicity. pounds such as hydrocortisone and benzoic acid
Yet, many of the papers reviewed did not consider and the hydrophilic compounds did show a cor-
how lipophilicity of the test compound would relation. This should be further investigated. As
affect percutaneous absorption and hence affect stated previously, it may be necessary to use both
the correlation between TEWL and percutane- TEWL and drug lipophilicity to predict altera-
ous absorption. Feldmann and Maibach (1970) tions in skin permeability.
measured both the total absorption and maximal
absorption rate for 20 different compounds of dif-
ferent lipophilicities. The range for total absorp- Conclusion
tion for the 20 compounds tested demonstrated a In 2005, Levin and Maibach reviewed nine
difference greater than 250 times in total absorp- studies investigating the correlation between
tion amounts, while the 20 compounds that had TEWL and percutaneous absorption of
a difference in maximum absorption rate were actives. At that time seven of the nine studies
greater than 1,000-fold (Feldmann and Maibach demonstrated a quantitative correlation, yet
1970). Because of the extreme range of absorp- two studies did not. Those studies that did not
tion for topically applied compounds, it seems confirm a quantitative correlation (Tsia et al.
reasonable to assume that the correlation between 2001b; Chilcott et al. 2002) or only observed
TEWL and percutaneous absorption may not be a weak correlation (Oestmann et al. 1993; Hui
66 J. Levin and H.I. Maibach

et al. 2012; Lavrijsen et al. 1993) used differ- heat separated epidermis and dermatomed skin in per-
ent experimental methods, such as an in vitro cutaneous absorption studies. Dermatol Res 299(10):
507–511
model, animal skin, or extremely lipophilic Bronaugh RL, Franz TJ (1986) Vehicle effects on
compounds compared with the studies which percutaneous absorption: in vivo and in vitro compari-
found a quantitative correlation. The conclu- sons with human skin. Br J Dermatol 115:1–11
sion at this time was that those assumptions Bronaugh RL, Maibach HI (1989) Percutaneous absorp-
tion, 2nd edn. Marcel Dekker, New York
and differences in experimental design were Bronaugh RL, Maibach HI (1991) In vitro percutaneous
likely responsible for the lack of correlation. absorption. CRC Press, Boca Raton
Since then, new studies have been published Bronaugh RL, Stewart RF (1986) Methods for in vitro
investigating the use of lipophilic compounds, percutaneous absorption studies VI: preparation of the
barrier layer. J Pharm Sci 75:487–491
in vitro models, and animal skin as models for Bronaugh RL, Stewart RF, Congdon ER, Giles AL Jr
in vivo human skin barrier study (Elkeeb et al. (1982a) Methods for in vitro percutaneous absorption
2010; Hui et al. 2012; Elmahjoubi et al. 2009). studies I. Comparison with the in vivo results. Toxicol
These studies have demonstrated significant Appl Pharm 62:474–480
Bronaugh RL, Stewart RF, Congdon ER (1982b) Methods
correlation between TEWL and percutaneous for in vitro percutaneous absorption studies II. Animal
absorption in vitro in human and animal skin models for human skin. Toxicol Appl Pharm 62:
for both lipophilic and hydrophilic compounds 481–488
(Elkeeb et al. 2010; Hui et al. 2012; Elmahjoubi Chilcott RP, Dalton CH, Emmanuel AJ, Allen CE, Bradley
ST (2002) Transepidermal water loss does not corre-
et al. 2009). In this updated overview, 10 of late with skin barrier function in vitro. J Invest
the 12 studies discussed here found some Dermatol 118(5):871–875
degree of correlation between TEWL and per- Dupuis D, Rougier A, Roguet R, Lotte C, Kalopissis G
cutaneous absorption. It is uncertain why (1984) In vivo relationship between horny layer reser-
voir effect and percutaneous absorption in human and
these two studies found no correlation; how- rat. J Invest Dermatol 82:353–356
ever, it seems likely after looking at the com- Elkeeb R, Hui X, Chan H, Tian L, Maibach HI (2010)
piled data in Table 6.1 that TEWL can serve as Correlation of transepidermal water loss with skin bar-
a prediction for percutaneous absorption in rier properties in vitro: comparison of three evaporim-
eters. Skin Res Technol 16(1):9–15
both in vivo and in vitro models and in human Elmahjoubi E, Frum Y, Eccleston GM, Wilkinson SC,
and animal skin and those studies which did Meidan VM (2009) Transepidermal water loss for prob-
not report a correlation between TEWL and ing full-thickness skin barrier function: correlation with
percutaneous absorption were the exception tritiated water flux, sensitivity to punctures and diverse
surfactant exposures. In Vitro 23(7):1429–1435
rather than the rule. Furthermore, it may be Farahmand S, Tien L, Hui X, Maibach HI (2009)
that evaporimeter choice may play a more Measuring transepidermal water loss: a comparative
important role in experimental design than in vivo study of condenser-chamber, unventilated-
previously assumed. chamber and open-chamber systems. Skin Res
Technol 15(4):392–398
Taken together, the weight of evidence Feldmann RJ, Maibach HI (1970) Absorption of some
confirms a relationship between TEWL and organic compounds through the skin in man. J Invest
percutaneous penetration of actives, yet, much Dermatol 54:399–404
Fluhr JW, Feingold KR, Elias PM (2006) Transepidermal
remains to be understood.
water loss reflects permeability barrier status: valida-
tion in human and rodent in vivo and ex vivo models.
Exp Dermatol 15(7):483–492
Hui X, Elkeeb R, Chan H, Maibach HI (2012) Ability to
estimate relative percutaneous penetration via a sur-
References rogate maker – trans epidermal water loss? Skin Res
Technol 18(1):108–113
Aalto-Korte K, Turpeinen M (1993) Transepidermal Lamaud E, Lambrey B, Schalla W, Schaefer H (1984)
water loss and absorption of hydrocortisone in wide- Correlation between transepidermal water loss and
spread dermatitis. Br J Dermatol 128(6):663–665 penetration of drugs. J Invest Dermatol 82:556
Atrux-Tallau N, Pirot F, Falson F, Roberts MS, Maibach Lavrijsen LP, Oestmann E, Hermans J, Bodde HE,
HI (2007) Qualitative and quantitative comparison of Vermeer BJ, Ponec M (1993) Barrier function param-
6 The Correlation Between Transepidermal Water Loss and Percutaneous Absorption 67

eters in various keratinization disorders: transepider- Rougier A, Dupuis D, Lotte C, Roguet R, Schaefer H
mal water loss and vascular response to hexyl (1983) In vivo correlation between stratum corneum
nicotinate. Br J Dermatol 129:547–554 reservoir function and percutaneous absorption.
Leveque JL, Corcuff P, de Rigal J, Agache P (1984) In J Invest Dermatol 81:275–278
vivo studies of the evolution of physical properties of Rougier A, Dupuis D, Lotte C, Roguet R (1985) The mea-
the human skin with age. Int J Dermatol 23:322–329 surement of the stratum corneum reservoir. A predic-
Levin J, Maibach H (2005) The correlation between tran- tive method for in vivo percutaneous absorption
sepidermal water loss and percutaneous absorption: an studies: influence of application time. J Invest
overview. J Control Release 103(2):291–299 Dermatol 84:66–68
Lotte C, Rougier A, Wilson DR, Maibach HI (1987) In Rougier A, Lotte C, Maibach HI (1987a) In vivo percuta-
vivo relationship between transepidermal water loss neous penetration of some organic compounds related
and percutaneous penetration of some organic com- to anatomic site in man: predictive assessment by the
pounds in man: effect of anatomic site. Arch Dermatol stripping method. J Pharm Sci 76:451–454
Res 279:351–356 Rougier A, Lotte C, Maibach HI (1987b) The hairless rat:
Machado M, Hadgraft J, Lane ME (2010) Assessment of a relevant model to predict in vivo percutaneous
the variation of skin barrier function with anatomic absorption in humans? J Invest Dermatol 88:577–581
site, age, gender and ethnicity. Int J Cosmet Sci Rougier A, Lotte C, Corcuff P, Maibach I (1988)
32:397–409 Relationship between skin permeability and corneo-
Nangia A, Camel E, Berner B, Maibach H (1993) cyte size according to anatomic site, age and sex in
Influence of skin irritants in percutaneous absorption. man. J Soc Cosmet Chem 39:15–26
Pharm Res 10:1756–1759 Stewart RF, Bronaugh RL (1985) Methods for in vitro
Nilsson GE (1997) Measurement of water exchange percutaneous absorption studies V: permeation
through skin. Med Biol Eng Comput 15:209–218 through damaged skin. J Pharm Sci 74:1062–1066
Noonan P, Gonzalez M (1990) Pharmacokinetics and the Tsai JC, Sheu HM, Hung PL, Cheng CL (2001) Effect of
variability of percutaneous absorption. J Toxicol barrier disruption by acetone treatment on the perme-
9(2):511–516 ability of compounds with various lipophilicities:
Oestmann E, Lavrijsen AP, Hermans J, Ponec M (1993) implications for the permeability of compromised
Skin barrier function in healthy volunteers as assessed skin. J Pharm Sci 90:1242–1254
by transepidermal water loss and vascular response to Walker M, Dugard PH, Scoot RC (1983) In vitro percuta-
hexyl nicotinate: intra- and inter- individual variabil- neous absorption studies: a comparison of human and
ity. Br J Dermatol 128:130–136 laboratory species. Hum Toxicol 2:561–565
Pinnagoda J, Tupker RA, Coenraads PJ, Nater JP (1989) Wester RC, Maibach HI (1992) Percutaneous absorption
Comparability and reproducibility of the results of in diseased skin. In: Maibach HI, Surber C (eds)
water loss measurements: a study of 4 evaporimeters. Topical corticosteriods. Karger, Basel, pp 128–141
Contact Dermatitis 20:241–246 Wester R, Maibach H (1993) Chair’s summary: percuta-
Pinnagoda J, Tupker RA, Agner T, Serup J (1990) neous absorption – in vitro and in vivo correlations.
Guidelines for transepidermal water loss (TEWL) In: Dermatology: progress & perspectives. The pro-
measurement. Contact Dermatitis 22:164–178 ceedings of the 18th World Congress of Dermatology.
Roskos KV, Guy RH (1989) Assessment of skin barrier The Parthenon Publishing Group, New York,
function using transepidermal water loss: effect of pp 1149–1151
age. Pharm Res 6(11):949–953
Influence of Excipients on Two
Elements of the Stratum Corneum 7
Barrier: Intercellular Lipids
and Epidermal Tight Junctions

Laurène Roussel, Rawad Abdayem, Elodie Gilbert,


Fabrice Pirot, and Marek Haftek

Contents 7.4.6 Dimethyl Sulfoxide (DMSO). . . . . . . . . . . . 82


7.4.7 Surfactants: Detergents . . . . . . . . . . . . . . . . 82
7.1 Introduction: Epidermal Barrier 7.4.8 Barrier-Restoring Molecules . . . . . . . . . . . . 83
Structure and Function . . . . . . . . . . . . . . . 69
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
7.2 Excipients: Their Nature, Roles,
and Modes of Action . . . . . . . . . . . . . . . . . 73 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
7.2.1 Excipients’ Action on the SC Lipids . . . . . . 73
7.2.2 Excipients’ Action on the
Tight Junctions. . . . . . . . . . . . . . . . . . . . . . . 74
7.1 Introduction: Epidermal
7.3 Methods to Evaluate Influence Barrier Structure
of Excipients on the Epidermal Barrier. . . 74
7.3.1 Methods to Follow the Structural
and Function
Modifications . . . . . . . . . . . . . . . . . . . . . . . . 74
7.3.2 Methods to Follow Biochemical The skin is one of the most important organs in the
Modifications . . . . . . . . . . . . . . . . . . . . . . . . 78 human body due to its crucial role as an interface
7.3.3 Methods to Assess the Permeability
between the internal and external environments. It
Barrier Function . . . . . . . . . . . . . . . . . . . . . . 79
controls the entry of ultraviolet radiation, chemi-
7.4 Known Effects of Excipients cals, pathogenic agents, free radicals, etc., as well
on the Epidermal Barrier Structure
and Function. . . . . . . . . . . . . . . . . . . . . . . . 80 as the water exchanges. The permeability bar-
7.4.1 Propylene Glycol (PG). . . . . . . . . . . . . . . . . 80 rier homeostasis is sustained by a combination
7.4.2 Fatty Acids: Oleic Acid (OA) . . . . . . . . . . . 80 of several factors, biochemical and structural, in
7.4.3 Terpenes . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81 response to the influences of the environment.
7.4.4 Azone® . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81
7.4.5 Ethanol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81 The permeability barrier function is fulfilled
by the epidermis and mainly its external layer –
the stratum corneum (SC). SC is composed of
L. Roussel (*) • R. Abdayem • E. Gilbert dead cells, corneocytes, issued from the terminal
F. Pirot • M. Haftek differentiation of epidermal keratinocytes. The
Faculty of Medicine and Pharmacy, University Lyon 1, losses of corneocytes by superficial desquama-
EA4169 “Fundamental, Clinical and Therapeutic Aspects
tion are compensated by cornification of kerati-
of the Skin Barrier Function”,
8, Avenue Rockefeller, Lyon 69373, France nocytes from the living layers of the epidermis
e-mail: laurene.roussel@univ-lyon1.fr; and by cell divisions in the germinative layer
rawad.abdayem@univ-lyon1.fr; (stratum basale).
elodie.gilbert@univ-lyon1.fr;
The efficiency of the SC barrier is based on
fabrice.pirot@univ-lyon1.fr;
marek.haftek@univ-lyon1.fr the composition and the correct arrangement of

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 69


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_7, © Springer-Verlag Berlin Heidelberg 2015
70 L. Roussel et al.

Fig. 7.1 In normal human skin (NHS), the SC may be corneodesmosomes. Exposure to propylene glycol (PG)
subdivided into SC compactum, where the principal per- induces basket-weave pattern in the entire SC. Interlayer
meability barrier resides, and SC disjunctum, where the corneodesmosomes become disrupted (arrowheads)
desquamation process takes place. Transition between because of a better accessibility of the catalytic enzymes,
the two regions is visualized in the inset. Several cor- but the cells remain attached laterally (big arrows). This
neodesmosomes (small arrows) link SC compactum phenomenon is likely related to the presence of TJ-like
corneocytes both laterally and vertically. SC disjunctum structures sealing off the corneocyte periphery and therein
is characterized by horizontal splitting between the suc- located corneodesmosomes. Bars = 200 nm in NHS and
cessive corneocyte layers due to the loss of the interlayer 500 nm in PG

its principal elements: (1) corneocytes linked by secretion of these latter organelles is also indis-
corneodesmosomes (Fig. 7.1) and (2) intercel- pensable for the formation and function of the SC
lular lipids covalently bound to the cross-linked barrier. It has been proposed that epidermal tight
corneocyte envelopes and organized in a multi- junctions (TJ) may contribute to the polarization
layered extracellular matrix (Wertz et al. 1989; of the granular layer cells and constitute an addi-
Wertz 2000; Haftek et al. 2006). Molecular tional permeability barrier (Brandner et al. 2010;
composition and structural arrangement of the Kirschner et al. 2010a, b).
hydrophobic lipid matrix are critically involved In transmission electron microscopy, TJ
in the limitation of SC permeability (Bouwstra appear as fusion points between external sheets
et al. 1991). Intercellular spaces of the SC con- of the plasma membranes of two neighboring
tain also hydrophilic poaches or lacunae that cells. Such cell–cell membrane fusions become
may swell upon SC hydration and are compatible cross-linked during the keratinization process
with the presence of catabolic enzymes and their and persist in the SC in a form of TJ-like struc-
natural inhibitors, essential for the regulation tures (Haftek et al. 2011). The resulting subdi-
of epidermal desquamation (Menon and Elias vision of the intercellular spaces of the SC into
1997; Haftek et al. 1998). Most of the compo- independent compartments appears to be impor-
nents of the SC intercellular spaces are produced tant for appropriate regulation of the desquama-
in the last living layer of the epidermis, stratum tion process and, thus, the barrier homeostasis.
granulosum (SG), and are delivered through exo- At the molecular level, TJ are composed of
cytosis of the trans-Golgi-derived lamellar bod- transmembrane and cytoplasmic proteins con-
ies (Ishida-Yamamoto et al. 2004). The oriented nected to the actin cytoskeleton (Brandner et al.
7 Influence of Excipients on Two Elements of the Stratum Corneum Barrier 71

Cr
um
bs

Claudins Pals
ZONAB tj
Pa
Rab 13

ZO
Rho/Rac

3-
ZO-1
Sec 6/8
tin
Ac

JAM Par-3 Par-6

Cd
c4
2
Occludin

Fig. 7.2 Schematic representation of a tight junction (TJ) tein (ZO-1/3, ZONAB) attach TJ to the actin cytoskeleton.
structure and of its cytoplasmic interacting proteins: trans- Regulatory proteins, e.g., Sec 6/8 the secretion complex,
membrane proteins, i.e., claudins and occludin, and junc- Pals linking protein, Patj Pals-linking protein to tight junc-
tional adhesion molecule (JAM) which interact in the tions, Par-3/6 partitioning defective 3 and 6, Crumb, Cdc42
intercellular space where linker proteins zonula occludens cell division control protein 42, Rab, and Rho/Rac small
and zonula occludens-associated nucleic acid binding pro- G-proteins are important for TJ formation and signaling

2002; Niessen 2007; Brandner et al. 2010). revival of epidermal TJ studies. In 2002, Furuse
Several transmembrane TJ-specific proteins have et al. have demonstrated that biotin (557 Da)
been identified, e.g., occludin, several claudins, injected in a mouse dermis ascends to the epi-
crumb, and JAM (junction adhesion molecules) dermis via the intercellular spaces and stops in
(Fig. 7.2). Extracellular domains of these proteins the SG at sites stained by antibody to occludin,
interact to establish cell–cell contacts. The inter- thus indicating the presence of functional TJ at
action between claudins engenders the formation this level (Furuse et al. 2002). Moreover, in clau-
of “pores” selective for various types and sizes din-1 knockout mice that do not survive after
of molecules (Tsukita and Furuse 2000; Furuse birth due to the severe percutaneous water loss,
et al. 2002) (Fig. 7.3). The selectivity is depen- the injected biotin crossed the level of SG, pen-
dent on the concentration and the type of claudins etrating into the SC. This observation proved the
participating in the junction protein complex. presence of functional TJ in murine epidermis,
Thus, the extracellular parts of TJ are not fully capable of controlling the diffusion of molecules
“tight.” However, when distributed as a continu- bigger than 557 Da.
ous band around a cell, TJ efficiently subdivides In man, several authors have reported the
the cell surface into the apical and basal regions presence of TJ-associated proteins in the epi-
thereby contributing to creation of cell polarity. dermis and TJ-like structures in the SG of the
Ken Hashimoto has observed TJ structures fetal and, more recently, adult epidermis (Pummi
at the apical membrane of the SG keratinocytes et al. 2001; Brandner et al. 2002; Langbein et al.
as early as 1971 (Hashimoto 1971). However, 2003; Schluter et al. 2004). These studies indi-
this observation has been neglected for 30 years cate that TJ provide a barrier against the per-
because lipids of the intercellular space of SC meability of solutes, also in human epidermis.
were considered the only efficient barrier to the However, doubt remains, concerning the exact
water loss (Elias and Friend 1975). The beginning role of TJ and their contribution to the barrier
of the twenty-first century was marked by the function, since the main permeability barrier at
72 L. Roussel et al.

Fig. 7.3 Schematic drawing of molecule-


selective “pores” engendered by tight
junction (TJ) proteins in the intercellular
spaces between the interacting cells.
Pore-like structures are responsible for the
control of paracellular diffusion of different
molecules through the junctions. Claudins
control the “tightness” and the selectivity of
the pores within the TJ strands and, thus,
TJ function relays largely on their claudin
composition. A 3D representation of the
arrangement of TJ proteins at the cell
surface as well as the “pores” created after
interaction of the extracellular domains
of claudins and occludin are shown to the
left (a). To the right (b) – distribution of the
lined-up transmembrane proteins as it can
be visualized at the cell surface with
freeze-fracture

this anatomic site is apparently provided by the (CER, approx. 50 % by weight), free fatty acids
SC components. (FFA; approx. 10 % by weight), and choles-
One possibility is that TJ play an important terol (approx. 20 % by weight and its derivate
role in case of rupture of the SC barrier and dur- cholesterol sulfate – approx. 5 %) (Wertz 2000;
ing the formation of this layer. In fact, TJ belts Weerheim and Ponec 2001; Feingold 2009).
distributed around the granular layer keratino- Triglycerides are also present in the SC barrier
cytes could contribute to the apical extrusion of but, unlike viable cell membranes, the SC is
lamellar bodies. The oriented delivery of lipids almost completely deprived of phospholipids.
to the interface between the viable epidermis and Cholesterol plays an essential role in the mainte-
the SC is necessary for filling of the intercellular nance of the membranes’ fluidity, as it promotes
spaces with a hydrophobic material and thus for intermixing of different lipid species, whereas
the correct barrier function (Brandner et al. 2010). cholesterol sulfate takes part in the modulation
As cornified envelopes of corneocytes are of desquamation because of its inhibitory action
impermeable to most diffusing substances, upon serine proteases. FFA consists predomi-
the main penetration pathway through the SC nantly of saturated long-chain species. Oleic acid
remains the intercellular one (Elias 1983). Lipids (6 %) and linoleic acid (2 %) are the only unsatu-
of the SC constitute the main component of the rated fatty acids detected unbound in the SC.
hydrophobic human skin barrier regulating water To date, in human SC, 12 ceramide subclasses
homoeostasis. Lipid composition determines the with a wide distribution of the chain length have
lipid organization in the SC and is therefore a been identified (Masukawa et al. 2009; Van
key factor underlying the skin barrier function Smeden et al. 2011). All ceramides bear a polar
(De Jager et al. 2004). The lipid matrix is mainly head group and two long carbon chains: a sphin-
composed of equimolar mixture of ceramides goid base and a fatty acid. They are necessary for
7 Influence of Excipients on Two Elements of the Stratum Corneum Barrier 73

the formation of the covalently bound lipid enve- is considered as the least permeable structure,
lope of corneocytes (Behne et al. 2000; Zheng whereas the liquid crystalline phase is highly per-
et al. 2011) and play a key role in the functioning meable to compounds (Groen et al. 2011). On the
of the SC (Coderch et al. 2003). Ceramide-1 is other hand, liquid phase seems to be necessary for
particularly important for the intercellular lipid formation of the LPP pattern (Bouwstra et al.
organization, because its long acyl chain is able to 1999). Moreover, in dry skin, a pure liquid crystal
span more than one lipid bilayer and, therefore, it system allows a rapid water loss through the
helps to rivet the multilayered matrix (Bouwstra bilayers with a moderate barrier action. The solid
et al. 2002). In general, a reduction in chain length system could cause a water loss due to breaks in
of CERs has a stronger impact on the lamellar the solid crystalline phase (Fluhr et al. 2008).
lipid organization and permeability than a change Maintaining the balance between the phases of
in the ratio between CER subclasses keeping the different physical properties is probably required
chain length approximately equal (De Jager et al. for optimal barrier function in preventing water
2006; Groen et al. 2011; Neto et al. 2011). In loss (Thau 2002).
atopic dermatitis patients, a reduction in overall
ceramide chain length is observed and correlates
with the impaired barrier function (Di Nardo et al. 7.2 Excipients: Their Nature,
1998; Janssens et al. 2012). Roles, and Modes of Action
In normal human SC, lipids are arranged in
two coexisting lamellar phases, a long periodicity According to the European Medicines Agency
phase (LPP) and a short periodicity phase (SPP) (EMA, earlier EMEA), excipients may be defined
with repeat distances of around 13 and 6 nm, as the constituents of a pharmaceutical formulation
respectively (Madison et al. 1987; Bouwstra et al. which are not the active substance. Excipients
1991). Within the lipid lamellae, lipid head groups include, e.g., coloring matters, antioxidants, preser-
display an orthorhombic lateral organization vatives, adjuvants, stabilizers, thickeners, emulsifi-
(solid crystalline phase) at the skin temperature of ers, solubilizers, penetration enhancers, flavoring
around 30–32 °C. However, a subpopulation of and aromatic substances, as well as constituents of
lipids can also be less densely packed, showing a the outer covering of the medicinal products.
hexagonal pattern (gel crystalline phase) or, the Penetration enhancers are excipients which
most fluid, liquid crystalline phase (Bommannan have the ability to modify the penetration of
et al. 1990; Ongpipattanakul et al. 1994; Pilgram active substances through the skin and therefore
et al. 2001a). Although the orthorhombic packing could significantly influence the in vivo perfor-
is predominant in the SC, the uppermost SC lay- mance of a dermal and transdermal formulation.
ers have the highest extent of the liquid crystalline Development and control of action of these sub-
phase, a consequence of the presence of sebum. stances are essential for all dermal and especially
As shown in in vitro experiments, cholesterol and transdermal formulations, where a constant and
ceramides are very important for the formation of persistent release of active molecules over sev-
the lamellar phases. After addition of FFA, the lip- eral hours or even days is mandatory for thera-
ids are organized in an orthorhombic packing peutic efficacy.
with a small proportion of lipids in a liquid crys-
talline phase. Deficiencies in any one of the three
main lipid species result in barrier abnormalities 7.2.1 Excipients’ Action on the
as well as observable alterations in the ultrastruc- SC Lipids
tural features of the SC extracellular domains
(Holleran et al. 2006). Lamellar lipid organization Most of the skin penetration enhancers are
is considered to play an important role in the bar- designed to modify the intercellular SC domains
rier function of the skin, especially the LPP in order to reduce the resistance of barrier lipid
(Bouwstra and Ponec 2006). Orthorhombic phase bilayers. They affect the strength of interactions
74 L. Roussel et al.

between the polar head groups, the conforma- et al. 2001b). Soaps associated with an extensive
tional state, and/or the lateral packing of the hydration, e.g., during bathing, can also promote
lipids (Tfayli et al. 2012). Recent studies have a less organized state of the SC lipids (Rawlings
also emphasized the effects of detergents on et al. 1994). Moreover, some detergents induce
lipid synthesis, on lipid-metabolizing enzymes horizontal splitting in the lipid layers leading to
and, generally, on keratinocyte differentiation the loosening of the intercorneocyte spaces and
(Wei et al. 2006; Torma and Berne 2009). Several thus facilitate the mobility of the extracellular
mechanisms of action are individualized and may enzymes implicated in the SC barrier modulation
intervene separately, depending on the nature of (Haftek et al. 1998) (Fig. 7.4; see also Fig. 7.1).
the excipient, although combined actions are
most frequently encountered.
7.2.2 Excipients’ Action on
7.2.1.1 Extraction of Lipid Components the Tight Junctions
Ethanol at concentrations between 40 and 80 %
extracts lipids from the skin and promotes the TJ modulators interact with the extracellular
permeation of polar and nonpolar drugs through loops or the membrane domain of TJ. Such mod-
a hypothetical “pore” pathway (Manabe et al. ulators or excipients can belong to various fami-
1996; Levang et al. 1999). Such lipid depletion lies of molecules, i.e., lipids, surfactants, and
or “degreasing” of the skin, i.e., the ability of sol- proteins (Wong and Gumbiner 1997; Kondoh and
vents or detergents to solubilize and remove SC Yagi 2007; Kondoh et al. 2008; Johnson et al.
lipids, may also be involved in the damaging effect 2008). Wong et al. have proved that a 44-amino-
observed after prolonged or repetitive exposure acid-long peptide is able to increase the cellular
to such substances. turnover of occludin, one of the TJ complex pro-
teins (Wong and Gumbiner 1997). This reversible
7.2.1.2 Fluidization of the Intercellular effect results in an increase of the paracellular
Lipid Matrix permeability of TJ.
The enhanced permeation observed in the pre- Another example of a peptide capable of dis-
sence of ethanol was proposed to be associated integrating the TJ structure by simple interaction
with different mechanisms including the fluidiza- with one of its proteins is the C-terminal frag-
tion of SC lipids (Panchagnula et al. 2001). ment of Clostridium perfringens enterotoxin.
Fourier transform infrared (FTIR) experiment This peptide interacts with claudin-4 and leads
showed increase in SC lipid fluidity upon appli- to the internalization of TJ. It could be used as
cation of 20–60 % ethanol, which may enhance a poison that provokes the complete loss of the
the permeation of lipophilic drugs, especially intestinal TJ barrier (Morita et al. 1999; Sonoda
through the intercellular lipid pathway (Manabe et al. 1999).
et al. 1996).

7.2.1.3 Disorganization of the Lipid 7.3 Methods to Evaluate


Lamellae Influence of Excipients
In normal SC, it is thought that the ratio of lipids on the Epidermal Barrier
in ordered and disordered (liquid crystalline)
phases modulates the SC barrier function proper- 7.3.1 Methods to Follow
ties (Boncheva et al. 2008). The orthorhombic the Structural Modifications
packing is the most tightly packed lipid barrier. It
depends on the presence of long-chain fatty acids 7.3.1.1 Microscopy Methods
combined with ceramides in association with Microscopy can be used to detect the influence
cholesterol (Bouwstra et al. 1998). Towards the of excipients on the extracellular lipid matrix as
skin surface, sebum lipids can induce a transition well as on the TJ. Light microscopy combined
to a less tightly packed hexagonal phase (Pilgram with immunodetection is usually employed to
7 Influence of Excipients on Two Elements of the Stratum Corneum Barrier 75

c d

f g

Fig. 7.4 Ultrastructure of human stratum corneum The junctional plugs appear frequently fragmented and
compactum post-fixed with ruthenium tetroxide (RuO4): unevenly stained. At places where the corneodesmo-
(a) nonoccluded; (b, c) simple occlusion; (d–g) permea- some edges remain in direct contact with equally electron
bilization with propylene glycol. Electron-dense lenticu- dense dilations, distinction between the two elements is
lar dilatations (arrowheads) are localized between the difficult (f, g). Typical triple-band structures of the cor-
intercellular lipid lamellae. In the occluded SC, these neodesmosome cores are observable mostly with low
hydrophilic compartments are often found in contact levels of RuO4 staining, rendering lipid lamellae less
with corneodesmosomes (open arrows). Propylene glycol visible (Reproduced with permission from Microscopy
promotes further expansion of the dark pools of inter- Research and Techniques, John Wiley and Sons; Haftek
cellular substance and corneodesmosome degradation. et al. (1998))
76 L. Roussel et al.

follow changes in expression and distribution violet radiation which poorly penetrates tissue
of protein components such as TJ molecules. blocks turned black by the fixatives (Haftek et al.
Immunofluorescent studies performed on frozen 1998). Acrylate-embedded samples are widely
tissue sections allow the evaluation of “native” used for on-section immunolabeling with colloi-
tissue, not fixed chemically and not exposed to dal gold, including that of TJ (Haftek et al. 2011).
solvents during the preparative methods. When Efficiency of ultrastructural immunolabeling can
combined with exploration in confocal laser be further improved by the use of non-resin-
scanning microscopy, where virtual optical sec- embedded tissue ultra-cryosections (Igawa et al.
tions are recorded within the immunolabeled 2011). Transmission electron microscopy is also
tissue, this approach is very useful to evaluate used for observation of freeze-fracture samples,
TJ topography in 3D reconstructions (Brandner with or without immunolabeling (Corcuff et al.
et al. 2002; Kubo et al. 2009). Immunostaining 2002). In this approach, the snap-frozen tissue is
of dewaxed skin sections, following the standard broken into pieces and the resulting fracture pro-
histology protocol involving aldehyde fixation file is covered with metal atoms. After removal
and paraffin embedding, should be interpreted of the underlying tissue, the metallic replica is
with care when the SC is the compartment of examined with an electron microscope. Exposed
interest. Indeed, solvents used during tissue fragments of the plasma membranes, where the
dehydration and then during dewaxing efficiently split occurs preferentially, may show the presence
remove intercorneocyte lipid matrix thus rende- of transmembrane molecules, such as TJ proteins
ring impossible observation of excipient-induced (Kurasawa et al. 2009). The latter are arranged
changes in this localization. On the other hand, in a characteristic crisscrossed linear pattern and
lipid removal may be beneficial for studies of their TJ origin may be confirmed using immuno-
TJ antigens persisting in the SC, as it improves gold labeling of the replicas with specific antibo-
accessibility of the latter to adequate antibodies. dies. Also, freeze-fracture studies efficiently help
Electron microscopy approach gives further to investigate the lipid arrangement within the SC
insight into the structural composition of the extracellular spaces (Van Hal et al. 1996).
SC barrier elements. Its “standard” transmission Scanning electron microscopy methods
electron microscopy version, consisting of alde- appear to be less suited for SC barrier studies
hyde fixation followed by osmium tetroxide post- because of the magnification range, which is
fixation and resin embedding, is less suitable much lower than in the transmission variant, and
for studies of the SC barrier, essentially for the due to accessibility limited to the surface of a
same reasons as the “standard” light microscopy. sample. Nevertheless, recent technological
Although osmium tetroxide is able to partially improvements allowing examination in partial
preserve lipid bilayers from extraction during vacuum with signals obtained with secondary
the subsequent dehydration procedures, visua- and retro-diffused electrons herald new applica-
lization of the fine lamellar structures, within the tions using tape-stripped and immunolabeled SC
extracellular spaces of SC, is not possible with [Haftek 2013, unpublished data].
this technique. Ruthenium tetroxide post-fixation
solves this problem; however, incubation with 7.3.1.2 Vibrational Spectroscopies
this highly oxidizing reagent results in significant Vibrational spectroscopies are powerful nonde-
deterioration of the ultrastructure of living cells structive techniques that detect characteristic
(Swartzendruber et al. 1995). Low-temperature vibrational energy levels of a molecule. Infrared
embedding of samples in partially hydrophilic and Raman spectra contain detailed information
acrylate resins also preserves, to some extent, lip- about the structures and interactions of molecu-
ids from extraction by solvents used during the lar classes of interest. Several studies have
dehydration phase. However, it is not compatible reported the potential of FTIR and Raman spec-
with osmium or ruthenium tetroxide post-fixation troscopies for the characterization of the mole-
because polymerization takes place under ultra- cular and supramolecular organization of lipids
7 Influence of Excipients on Two Elements of the Stratum Corneum Barrier 77

and to follow changes in the lipid chain confor- temperatures below 40 °C, there are two distinct
mational order. More particularly, these tech- bands at approximately 720 and 729 cm−1. The
niques have already been used to characterize latter is a reliable marker of an orthorhombic
phase transitions of the SC lipids (Moore et al. phase in the SC lipids (Pensack et al. 2006).
1997; Lawson et al. 1998; Ponec et al. 2000;
Wartewig and Neubert 2007) of related model 7.3.1.4 Differential Scanning
systems (Bouwstra et al. 2002) and to study Calorimetry (DSC)
polar and nonpolar lipid–lipid interactions DSC is a thermoanalytical technique measuring
(Corbe et al. 2007). heat capacity of a sample. It allows thermal ana-
lysis of an isolated SC and is used to evaluate the
7.3.1.3 FTIR: Fourier Transform interaction of excipients with the skin.
Infrared Spectroscopy Alterations of the characteristic temperatures of
FTIR was used to obtain information on the lat- the endothermic peaks can be observed, relative to
eral lipid organization and conformational order- disorganization of the lamellar structure of lipids
ing of the lipids (Janssens et al. 2012). FTIR and to dehydration or denaturation of the proteins.
spectroscopy can be used to investigate the bio- DSC is less sensitive to phase transitions, presu-
physical alterations taking place in the lipid mably because of relatively low enthalpy changes
bilayer after treatment with penetration enhanc- resulting from the transition, whereas FTIR is able
ers by studying the vibrational modes of its com- to detect them with relative ease.
ponents. Different types of vibrations were
monitored, the CH2 symmetric stretching vibra- 7.3.1.5 Raman Spectroscopy
tion and second derivatives of the scissoring Raman spectroscopy reveals the spectral features
bandwidth being the most informative for lateral specific to the modes of action of the penetration
organization (Boncheva et al. 2008; Damien and enhancers. These features can be simultaneously
Boncheva 2010). For instance, the CH2 symmet- used as direct vibrational descriptors of the
ric and asymmetric stretching frequencies (υsym supramolecular organization, the polar interac-
CH2 and υasym CH2) near 2,850 and 2,920 cm−1, tions, and the conformational order of the CERs
respectively, are primarily sensitive to lipid chain and as a descriptor of the penetration enhancer
conformational order. A low (~2,848 cm−1) wave activity in drug delivery. Raman spectroscopy
number of the CH2 symmetric stretching vibra- can be used to monitor not only the evolution of
tions indicates the presence of a highly ordered the lateral packing and the polar interactions due
lipid organization (either hexagonal or ortho- to penetration enhancer activity but also the intra-
rhombic), while a high (2,853 cm−1) wave num- chain conformational order and the chain-end
ber indicates the disordered liquid phase (Moore conformers, such as resulting from the thermo-
et al. 1997). However, a low average bandwidth tropic behavior of ceramides (Tfayli et al. 2010).
of the scissoring vibrations represents a reduction Such information cannot be obtained with FTIR
of lipids in an orthorhombic organization and spectroscopy.
thus a less dense lipid organization (Janssens
et al. 2012). Extraction of the SC lipids with sol- 7.3.1.6 X-ray Diffraction Studies
vents results in reduction of the CH2 stretching In this method, a primary X-ray beam, emitted by
absorbance. Indeed, the decrease in CH2 stretch- a source, is partially scattered by the structures
ing bandwidths accompanied by a decrease in present in a studied sample. When applied to SC,
CH2 stretching band intensity suggests an overall the obtained diffraction patterns give informa-
extraction of SC lipids (Levang et al. 1999). The tion about organization of the intercellular lip-
CH2 rocking frequencies (710–735 cm−1) are ids. Position and intensity of the observed peaks
much more sensitive than the changes in CH2 are measured and interpreted, in order to evalu-
symmetric frequencies to the aforementioned ate the action of excipient on lipid organization
hexagonal–orthorhombic phase transition. At (Bouwstra and Ponec 2006).
78 L. Roussel et al.

• Small-Angle X-ray Diffraction (SAXD) 7.3.2.1 Molecular Biology


Diffraction pattern measured at low angle, Molecular biology approach is required for the
typically between 0° and 5°, provides infor- comprehension of viable cell responses. As such,
mation about the larger structural units in the it concerns exclusively studies of the living epi-
sample, such as the repeat distance of a lamel- dermal layers and not the SC. However, study of
lar phase (Janssens et al. 2012). RNA gives an idea on the transcription rate of
At room temperature, two lamellar phases are a specific gene and this, in turn, indicates pos-
present in human SC. One shows a periodicity sible repercussions on the subsequent SC forma-
of approximately 6.4 nm and the other of tion and function. For example, quantification of
approximately 13.4 nm (Bouwstra et al. 1991). mRNA coding for TJ proteins is possible using
Since the 13 nm lamellar phase has always real-time polymerase chain reaction (RT PCR).
been present in all species studied so far, this Using this technique, one can monitor the pres-
phase is considered to be important for the ence of mRNA of various TJ proteins in kerati-
skin barrier function. nocyte cultures with different knockdown genes
• Wide-Angle X-ray Diffraction (WAXD) while testing permeability of the expressed
At wider angle, scattered X-rays contain TJ (Kirschner et al. 2013). Already in 2008,
information about smaller structural units in Yamamoto et al. have performed a posttranscrip-
the sample, such as the lateral packing of tional gene silencing of different TJ proteins to
molecules in a lamellar phase. In the liquid study the impact of these proteins on the forma-
crystalline packing, the intermolecular dis- tion of the SC barrier (Yamamoto et al. 2008).
tance is not very well defined. This results in Keratinocytes were transfected with small inter-
a WAXD pattern that is characterized by a ferent RNA (si-RNA) targeting either claudin-1
very broad peak at 0.46 nm. The diffraction mRNA or occludin mRNA. It turns out that
pattern of the two-dimensional hexagonal lat- transfected cultures were not able to develop a
tice is characterized by a strong reflection at functional barrier. Using molecular biology tech-
0.41 nm. In the case of the orthorhombic niques, it would be possible to detect variations
organization, two strong reflections can be of the keratinocyte gene activity that excipi-
detected at a spacing of 0.41 and 0.37 nm. ents may engender when applied on the skin.
However, whether a hexagonal sublattice Hybridization in situ, on tissue sections, is an
coexists with an orthorhombic one cannot be approach combining structural information with
deduced from the diffraction pattern, since mRNA detection. It is, thus, particularly well
the 0.41 nm reflection, characteristic for the suited for studies of locally induced changes fol-
hexagonal lateral packing, is obscured by the lowing topical treatments.
0.41 nm reflection attributed to the ortho-
rhombic phase (Bouwstra and Ponec 2006). 7.3.2.2 Biochemistry
Biochemistry assays are commonly used to
analyze proteins as well as lipids extracted
7.3.2 Methods to Follow from a tissue.
Biochemical Modifications Immunoblotting technique can detect the pre-
sence of a given protein. In fact, the presence of
The presence of proteins of the TJ complex or the mRNA in the cell cytoplasm is insufficient to
enzymes implicated in the synthesis/degrada- predict the presence of the corresponding protein
tion of lipids can be studied in tissue extracts. due to the complexity of the translation machinery.
Similarly, quantitation of the corresponding Western blot is a reliable qualitative and quantita-
messenger ribonucleic acid (mRNA) is also pos- tive technique applicable to TJ protein studies
sible. As initial tissue localization of the obtained (Kurasawa et al. 2009; Kirschner et al. 2013).
profiles is essential, these studies require precise Lipid analysis may be performed on SC
sampling based on the structural data. extracts using high performance thin layer
7 Influence of Excipients on Two Elements of the Stratum Corneum Barrier 79

chromatography (HPTLC) (Rissmann et al. is an essential factor to the successful perfor-


2008; Popa et al. 2012). Combined with sequen- mance of permeation experiments, as specified
tial tape-stripping, HPTLC approach allows fol- in the test guideline OECD 428 (Lévêque et al.
lowing lipid composition of intercellular spaces 1993). The guidance document recommends the
between corneocytes in the SC according to the measurement of TEWL, transepithelial electri-
sample’s depth (Popa et al. 2011). cal resistance, or the use of tritiated water as
permeation markers.

7.3.3 Methods to Assess the 7.3.3.3 Transepithelial Electrical


Permeability Barrier Function Resistance (TEER)
TEER measurement can be used to detect the
7.3.3.1 Transepidermal Water Loss TJ functionality status. Usually the functiona-
(TEWL) Measurement lity of TJ is evaluated in vitro on monolayer of
One of the key parameters to monitor the skin cultured cells or on reconstructed human epi-
barrier function is the transepidermal water dermis. Functional TJ significantly increase the
loss, i.e., the rate of water evaporation through tissue impedance towards a very weak electri-
the epidermis, independent of sweating. TEWL cal current generated by ohmmeters, by virtue
provides a noninvasive method for assessing of limiting ion flux through the intercellular
changes in the barrier properties of the SC. It space (Kurasawa et al. 2009; Abdayem et al.
can be considered as a determinant indicative of 2011). In normal human skin, SC by itself is
the functional state of the epidermal barrier in able to prevent the passage of the testing current.
normal, pathological, and experimental condi- Therefore, to detect any variation of resistance
tions (Janssens et al. 2012). Measurements are due to TJ modulation, the SC has to be partially
performed in standardized ambient conditions or completely removed. This is usually done
according to the well-defined consensus guide- by tape stripping. In the case of in vitro recon-
lines (Pinnagoda et al. 1990). structed epidermis, the extracellular lipid matrix
is not perfectly organized (Ponec et al. 2000) and
7.3.3.2 Diffusion Cells: Ex Vivo the SC is fully hydrated. This results in readable
Permeation Studies TEER measurements, as the electrical current is
Diffusion cell is helpful to study the influence able to cross this imperfect SC barrier (Abdayem
of excipients on drug permeation and pen- et al. 2011). For the detection of the effects of dif-
etration. Indirectly, information is gained on ferent excipients on the TJ barrier, reconstructed
the functional state of the treated skin barrier. epidermis is cultured in plastic inserts equipped
Skin permeation is evaluated by measuring the with a permeable polycarbonate bottom. This
steady-state permeation flux of the tested drug. experimental setting allows positioning of the
An enhancement ratio can be determined in the ohmmeter electrodes on both sides of the cul-
presence of the studied excipient. The diffusion tured tissue. Tested excipients can be added at
cell consists of the upper, i.e., “donor,” and the different doses either topically, i.e., directly on
lower, i.e., “receptor,” chambers, separated by the SC surface, or “systemically,” into the feed-
a tested skin. The epidermis faces the donor ing medium penetrating the reconstructed epi-
chamber where the tested products are applied, dermis from the bottom compartment. In case
and the dermis faces the receptor chamber of “systemic” treatment, excipients are in direct
filled with a receptor fluid, where the drug or interaction with the epidermal TJ and the SC
another tracer will be measured after perme- remains intact. Topically applied agents have
ation. Temperature is controlled throughout the first to cross the SC barrier, before acting on TJ
experiment and should be maintained at in vivo situated in the granular layer. Thus, TEER read-
skin conditions (32 °C). Ensuring that the skin ings in the latter case reflect both TJ and SC per-
used for testing maintains its physical integrity meability modulation.
80 L. Roussel et al.

7.3.3.4 Dye Penetration Test sidered nontoxic and shows low relative irritancy
Intact SC and functional epidermal TJ are able in vivo (Lashmar et al. 1989; Barry 1991; Haftek
to block or to limit the passage of various mol- et al. 1996). Haftek et al. (1998) have evaluated
ecules depending on their size and nature, e.g., the ultrastructural spatial organization of the
lipophilicity, pH, and charge. Following the pas- intercorneocyte spaces before and after applica-
sage of a tracer applied at the surface of the skin tion of PG. When applied ex vivo undiluted on the
or injected at the basal layers of the epidermis is skin, PG was capable of inducing a pronounced
another way to study the functional quality of the loosening of the horny layer, observable in light
complementary SC and TJ barriers. For example, and electron microscopy as the “basket-weave
in the case of topical application of Lucifer yel- pattern.” At the ultrastructural level, the SC dis-
low, detection of this hydrophilic fluorescent dye, sociation could be linked to the expansion of
with a confocal microscope, in the living layers of water-containing domains within the intercor-
epidermis, may indicate the presence of poor SC neocyte spaces associated with disorganization
barrier, either constitutive or induced (Kurasawa of the lamellar lipids and corneodesmosome
et al. 2009). Another widely used small molecular degradation (Fig. 7.4). Thus, incorporation of
marker is biotin (433–666 Da) (Ding et al. 2011). PG into lipid layers does functionally change the
After being injected to the dermis, the tracer properties of the intercorneocyte space and may
diffuses through the intercellular spaces until it increase mobility of the extracellular proteo-
reaches the TJ barrier in the stratum granulosum. lytic enzymes (Haftek et al. 1996). This, in turn,
Immunohistochemical detection of the biotin promotes digestion of the intercorneocyte junc-
on vertical skin sections reveals the sites where tions and leads to desquamation. Interestingly,
the diffusion has been stopped. These points are the PG-induced horizontal splitting occurring
recognized as TJ because of their co-localization between the successive layers of corneocytes is
with anti-occludin staining (Furuse et al. 2002). not extended to the lateral cell–cell attachments
Tracer penetration studies can also be performed (Fig. 7.1). Recent investigations on the TJ struc-
at the ultrastructural level using electron-dense tures persisting in a cross-linked form between
lanthanum salts. Ken Hashimoto was the first to the SC corneocytes shed light on this curious
describe TJ structures in human epidermis using, phenomenon (Haftek et al. 2011). In fact, TJ-like
precisely, this approach (Hashimoto 1971). structures located at the top of and within the
lateral intercorneocyte contacts efficiently limit
accessibility of the intercellular enzymes to
7.4 Known Effects of Excipients the cell periphery and the corneodesmosomes
on the Epidermal Barrier located within it.
Structure and Function PG is also frequently employed to enhance
drug release, from the dosage form (Panchagnula
Excipients influencing skin barrier demonstrate et al. 2001).
properties of penetration enhancers and are
employed accordingly. The modes of action of
skin penetration enhancers involve, in general, 7.4.2 Fatty Acids: Oleic Acid (OA)
either the disruption of skin barrier properties or
the increase of drug partitioning into the various Percutaneous drug absorption is increased by a
layers of the skin, starting with the SC. wide variety of long-chain fatty acids. Among
the most frequently used is OA, as it moderately
increases both drug diffusivity and drug parti-
7.4.1 Propylene Glycol (PG) tioning parameters (Koyama et al. 1994). This
long-chain monounsaturated fatty acid in cis
PG is a small organic and hydrophilic molecule configuration modifies the lipid domains of the
widely used as solvent and excipient. It is con- SC. This effect appears to be due to lipid layers
7 Influence of Excipients on Two Elements of the Stratum Corneum Barrier 81

fluidization and, predominantly, to phase sepa- FTIR and DSC for evaluation of the action of
ration (Naik et al. 1995). Indeed, after in vivo alcoholic terpenes, carvacrol, linalool, and alpha-
application, OA has been found to exist in a liq- Terpineol (Vaddi et al. 2002).
uid phase at all levels of the spectroscopically
examined SC. OA can be applied on the skin in
mixture with ethanol and easily combined with 7.4.4 Azone®
other penetration enhancers, like propylene gly-
col or terpenes (Yamane et al. 1995; Larrucea Azone® (1-dodecylazacycloheptan-2-one or lau-
et al. 2001). rocapram) was the first molecule specifically
designed as a skin penetration enhancer. Most
probably, the bilayer arrangement of lipids is dis-
7.4.3 Terpenes rupted by the presence of this enhancer, capable
of extracting cholesterol, as suggested by calo-
Terpenes are organic compounds found in essen- rimetry results (Kang et al. 2006). However,
tial oils. They are used as co-enhancers of per- its integration into the lipids is unlikely to be
cutaneous penetration, and, thus, their influence homogeneous, considering the variety of com-
is measured as the difference when compared to positional and packing domains within SC lipid
the results obtained with control mixtures that bilayers. Using electron microscopy, a “soup
do not contain them. Koyama et al. evaluated the spoon” model for Azone®’s conformation within
percutaneous absorption-enhancing effects of the SC lipids was observed. This model is com-
d-limonene in oleic acid using three drug models patible with a mechanism of action relaying on
with different lipophilicities (Koyama et al. 1994). the creation of Azone® domains (Hoogstraate
Pretreatment of the skin with limonene resulted et al. 1991). Electron diffraction studies using
in a large penetration enhancement for lipophilic lipids isolated from human SC provide good
and amphiphilic molecules but had little effect evidence that Azone® exists (or partially exists)
on the hydrophilic one. D-limonene increased in form of distinct phases within the SC lipids
mainly drug diffusivity in the nonpolar penetra- (Pilgram et al. 2001b).
tion route. Moreover, carvone, 1,8-cineole, and
thymol were shown to enhance the percutaneous
absorption of hydrophilic 5-fluorouracil in 50 % 7.4.5 Ethanol
ethanol by either increasing the SC lipid fluidity,
as revealed with FTIR, or perturbing the barrier Ethanol is commonly used as a “permeability
integrity of the epidermis, as demonstrated with enhancer” for transdermal drug delivery, as it is
TEWL (Gao and Singh 1997). well known for its ability to increase skin pene-
Limonene, eugenol, and menthone were used tration of drugs (Kwak et al. 2012). Ethanol-
in concentration of 5 % to investigate their input induced permeation of the SC depends on the
in percutaneous absorption of a moderately large alcohol content in the binary ethanol/H2O sys-
molecule (tamoxifen) when applied in 50 % pro- tem. It was found that for several different
pylene glycol. As addition of any of these ter- chemicals, the optimal range of ethanol concen-
penes resulted in a significantly increased tration was between 40 and 80 % (v/v), while
transcutaneous penetration, two mechanisms of smaller and larger proportions of ethanol in
action were suggested: lipid extraction deduced water showed more limited penetration enhanc-
from FTIR spectra (Zhao and Singh 2000) and ing effect (Kuriharabergstrom et al. 1990; Chen
conformation changes of the lipid chains visual- et al. 1995; Panchagnula et al. 2001). The
ized with Raman spectroscopy (Tfayli et al. enhanced permeation observed in the presence
2012). SAXD studies have also indicated that of ethanol in diffusion cell studies was proposed
d-limonene and 1,8-cineole disrupt SC lipid to be associated with various mechanisms. The
bilayers. Similar conclusions were reached using presence of alcohol increases the rotational
82 L. Roussel et al.

freedom of lipid acyl chains leading to an on lipid-metabolizing enzymes and on keratino-


increase in fluidity of the intercellular lipid cyte differentiation (Wei et al. 2006; Torma and
matrix (Panchagnula et al. 2001). Such a fluidi- Berne 2009). Moreover, in vivo studies combined
zation effect of lipid bilayers could be observed with HPTLC analysis suggest surfactant-induced
experimentally using FTIR (Chin and Goldstein alterations in the SC lipid synthesis (Gloor et al.
1977). Extraction of epidermal lipid compo- 2004; Heinemann et al. 2005).
nents by this solvent may also lead to SC deli- Sodium lauryl sulfate (SLS) or sodium
pidization (Levang et al. 1999; Manabe et al. dodecyl sulfate (SDS) is one of the most used
1996; Kuriharabergstrom et al. 1990; Krishnaiah surfactants. SLS is likely to remove epidermal
et al. 2004; Obata et al. 2006). lipids through the solubilizing action. Froebe
et al. (1990) examined this mechanism and
demonstrated that ceramides were not substan-
7.4.6 Dimethyl Sulfoxide (DMSO) tially extracted by SLS. The total lipid removed
after exposing SC to a 2 % solution of SLS
DMSO is one of the earliest but actually unused was less than 4 % of the total lipid content in
penetration enhancer. It exhibits a concentration- the SC. This result (and additional studies) has
dependent effect (Williams and Barry 2012). led to the assumption that there may be other
Several theories have been proposed to explain mechanisms besides lipid depletion. Indeed,
its mechanism of action including extraction SLS treatment attenuates the peak of SPP at
of skin lipids (Embery and Dugard 1971) and 6.5 nm in the X-ray diffraction pattern, thus
interaction with the head groups of lipid bilay- indicating disruption of the intercellular lipid
ers resulting in a change in the lateral packing organization (Ribaud et al. 1994). Moreover,
and reduction of the compactness of lipid bilay- Saad et al. showed, by FTIR study, a loss of the
ers (Elfbaum and Laden 1968). In fact, DMSO orthorhombically packed lipids paralleled by an
affects interactions between the polar groups of increase in lipids in the hexagonal phase (Saad
lipid molecules mainly by reducing the strength et al. 2012). Other experiments on isolated SC
of H-bonds (Tfayli et al. 2012). examined with electron microscopy showed the
disorganization of the intercellular lipid lay-
ers induced by SDS (Jiang et al. 2003). Lipid
7.4.7 Surfactants: Detergents lamellae structures were obliterated and amor-
phous and/or flocculent material appeared in the
Surfactants are one of the chief ingredients in dilated intercellular spaces. Interestingly, the
personal care products. However, skin barrier damage to the lipid lamellar structure induced
damage is observed after extensive exposure by exposure to water was similar to that induced
to detergents (Branco et al. 2005). Varying but by surfactants. In fact, hyperhydration of SC
overall small amounts of SC lipid are extracted may lead to disorganization of the lipid lamellae
by surfactants (Lévêque et al. 1993; Gloor et al. as it increases the volume of the non-lipid frac-
2004). Nevertheless, disruption of lipid structural tion in intercellular spaces (Haftek et al. 1998).
organization, observed using various physical Sodium caprate (C10) and surfactants con-
techniques, such as X-ray diffraction, transmis- taining polyethylene glycol (PEG) are excipients
sion electron microscopy, and DSC, has been known as enhancers of the intestinal absorption
demonstrated upon exposure of the SC to sur- by regulating jejunal TJ.
factants and was accompanied by an increase in Kurasawa et al. (2009) have examined locali-
TEWL (Ribaud et al. 1994; Warner et al. 1999; zation and expression of claudin-1 and of occlu-
Jiang et al. 2003). Recent studies, using RT-PCR, din in keratinocyte cultures after application
have also emphasized the effects of detergents of 10 mM sodium caprate (C10). Using immu-
7 Influence of Excipients on Two Elements of the Stratum Corneum Barrier 83

nofluorescence microscopy, they have found case. Treatment with Labrasol®, instead, led
that in intact cultures, both TJ proteins were to the same final results in terms of the TEER
present at the cell–cell contact regions, sug- loss, but the TJ permeation was more progres-
gesting the occurrence of TJ. Sodium caprate sive and less destructive morphologically. Thus,
application has resulted in the internalization of although both C10 and Labrasol® are able
the proteins. This effect was reversible after dis- to directly modulate epidermal TJ, the latter
continuation of the treatment. Examination of excipient appears to be better suited for in vivo
the cultures with freeze-fracture technique has applications aimed at percutaneous penetration
confirmed the disappearance of TJ strands from enhancement.
the cell surface upon exposure to C10 and their
reexpression after cessation of the treatment
(Kurasawa et al. 2009). 7.4.8 Barrier-Restoring Molecules
Abdayem et al. (2011 and in preparation) have
used in vitro reconstructed human epidermis and 7.4.8.1 Glycerin/Glycerol
normal human skin ex vivo as examples of fully Glycerin/glycerol, as a moisturizer, has been
keratinized tissues and compared the effects of suggested to prevent crystallization of the skin
various excipients on the ultrastructure of epider- lipids at low relative humidity (Froebe et al.
mis and the functionality of epidermal TJ. The 1990; Mattai et al. 1993). It has been hypoth-
results indicated that after 24 h of topical appli- esized that glycerol can interact with polar
cation of 10 mM C10 and of PEG-8 caprylic/cap- head groups of the lipid bilayers rather than
ric glycerides (Labrasol®, Gattefossé, France), by penetrating the alkyl chains. Glycerol exists
the SC and the living layers of the reconstructed in the SC as a natural endogenous humectant
epidermis were altered. However, morphologi- and largely influences skin hydration, cutane-
cal modifications were clearly more pronounced ous elasticity, and epidermal barrier repair
after C10 application, showing extensive disorga- (Verdier-Sevrain and Bonté 2007; Fluhr et al.
nization of the horny layer and vacuolization of 2008). Upon topical application at low concen-
the nucleated cells at all epidermal layers. Topical trations (1–10 %) in water, glycerol is able to
application of these excipients on intact human restore water holding capacity of the skin bar-
skin resulted in no remarkable modification of rier experimentally disrupted with the detergent
the tissue histology and only mild ultrastructural (SLS). Although this effect is not immediately
modifications could be detected in the SC. followed by the barrier repair, as measured
When assayed with TEER measurements, with TEWL, it may constitute an important
the reconstructed epidermis exposed to either step favoring physiological process to reestab-
of the excipients demonstrated time- and lish water barrier function of the impaired skin
dose-dependent loss in electrical resistance (Atrux-Tallau et al. 2010).
indicating permeabilization of the barrier. Such Some moisturizing agents, including gly-
an effect was not observed ex vivo, suggesting cerol, also promote desquamation (Rawlings
that naturally formed SC barrier was much more et al. 1995), probably by additionally increasing
resistant to the treatments than that of the epi- the volume of the non-lipid, hydrophilic fraction
dermal equivalent. The effect of both excipients of the SC intercellular compartment and, thus,
on TJ was verified by TEER measurement after improving local conditions for the activity of
exposure to the excipients added to the feeding endogenous catabolic enzymes. SC hyperhydra-
medium of the reconstructed epidermis. Such a tion may be easily achieved through skin occlu-
“systemic” treatment resulted in dose- and time- sion; however, hydrophilic moisturizers increase
dependent response revealed with TEER. The water levels in the SC without the formation
response to 1 mM C10 was very rapid in every of a water-impenetrable layer on the surface of
84 L. Roussel et al.

the SC (Lodén 2003). An interesting observa- 7.4.8.4 Physiologic Lipid Mixtures


tion is that nonocclusive lipophilic moisturizers (Mixtures of Ceramides, Fatty
penetrate into the SC and, by changing its lipid Acids, and Cholesterol)
organization to LPP and orthorhombic, actu- Physiologic lipid mixtures (mixtures of cerami-
ally increase the barrier function (Caussin et al. des, fatty acids, and cholesterol) have been
2007). Generally speaking, moisturizers may observed to penetrate into the skin deeper than
have favorable or deleterious influence on the SC SC and to improve the skin barrier function
barrier function, depending on the treated skin (Mao-Qiang et al. 1995, 1996; Lodén and
condition and the nature of the employed agent. Bárány 2000; Chamlin et al. 2002; Na et al.
One of the obvious clinical situations is that 2010; Popa et al. 2012). The immediate effect
of atopic dermatitis. Moisturizers with barrier- of physiologic lipid mixtures is attributable to
improving properties may delay relapse of the unspecific replenishment of the SC intercellu-
disease but treatment with moisturizing creams lar spaces, but, contrary to petrolatum, physio-
could also enhance transcutaneous penetration logic lipids in adequate proportions may be
of potential allergens and thus increase the risks assimilated by keratinocytes from the living
of dermatitis and asthma (Buraczewska et al. layers and used for the barrier repair in the lon-
2007; Lodén 2012). ger term. Accordingly, mixtures of ceramides,
fatty acids, and cholesterol, in equimolar pro-
7.4.8.2 Urea portions, allowed normal barrier recovery in
Urea, a small polar molecule, is frequently used acetone-treated murine skin, whereas two-
in dermatologic formulations as a “moisturizer.” component mixtures, e.g., fatty acids plus
It does not seem to significantly affect the lipid ceramides, cholesterol plus fatty acids, or cho-
matrix of the SC. In fact, it is able to replace water lesterol plus ceramides, delayed barrier recov-
while keeping the physical properties unchanged. ery (Mao-Qiang et al. 1993, 1995). Cholesterol,
Urea was shown to protect living cells against applied as the dominant lipid in physiologic
osmotic stress by retaining the liquid crystalline mixtures, accelerated barrier recovery in aged
phase of the lipid membrane even at low humi- human skin, presumably because of a reduc-
dity, down to 64 % (Costa-Balogh et al. 2006). tion in endogenous cholesterol synthesis
Consequently, maintaining the necessary degree underlying chronological skin aging (Zettersten
of fluidity of the extracellular lipids may be the et al. 1997). No acceleration of the barrier
mechanism by which urea improves skin condi- recovery in SLS-damaged human skin was
tions in dry climates. detected after treatment with ceramide 3 alone
in different emulsions (De Paepe et al. 2000).
7.4.8.3 Petrolatum Neither did a “moisturizer” consisting of
Petrolatum also can help to restore the SC ceramide 3, cholesterol, and fatty acids (so-
barrier by penetrating into its upper layers. It called skin-identical lipids) in a petrolatum-
remains the gold standard for barrier repair rich emulsion show superiority to pure
ingredients because it is the nonphysiologic petrolatum in human skin damaged by SLS and
substance closest to the natural intercellular tape-stripping (Lodén and Bárány 2000). The
lipids and it can intercalate into the intercel- absorption of ceramides and the superiority of
lular spaces. It does not penetrate to the living some lipid mixtures to other lipids thus remain
layers and, contrary to the physiologic lipids, to be proven in randomized and controlled
i.e., ceramides, free fatty acids, and choles- studies on humans, and a good guess is that
terol, petrolatum is unable to stimulate the these formulations should be tailored with
endogenous production of the SC intercellular respect to the epidermal abnormality to be
matrix (Grubauer et al. 1987; Ghadially et al. treated (De Boer and Hillier 2001; Lodén 2003;
1992; Mao-Qiang et al. 1995). Popa et al. 2012).
7 Influence of Excipients on Two Elements of the Stratum Corneum Barrier 85

Conclusion Bouwstra JA, Gooris GS, Dubbelaar FER, Ponec M


(1999) Cholesterol sulfate and calcium affect stratum
Excipients play an important role in the corneum lipid organization over a wide temperature
formulation of dermatological products. They range. J Lipid Res 40(12):2303–2312
may enhance transdermal penetration of mole- Bouwstra JA, Gooris GS, Dubbelaar FER, Ponec M (2002)
cules of interest through modulation of the com- Phase behavior of stratum corneum lipid mixtures
based on human ceramides: the role of natural and
position, organization, and function of the synthetic ceramide 1. J Invest Dermatol 118(4):606–
principal skin barrier elements, i.e., the SC inter- 617. doi:10.1046/j.1523-1747.2002.01706.x
cellular lipids and epidermal TJ. On the other Branco N, Lee I, Zhai H, Maibach HI (2005) Long-
hand, several excipients, useful in skin protection term repetitive sodium lauryl sulfate-induced irrita-
tion of the skin: an in vivo study. Contact Dermatitis
and restoration of the damaged barrier function, 53(5):278–284
are also available. Their appropriate use is at the Brandner JM, Kief S, Grund C, Rendl M, Houdek P,
heart of the galenic art applied to the skin. Kuhn C et al (2002) Organization and formation of
the tight junction system in human epidermis and cul-
tured keratinocytes. Eur J Cell Biol 81(5):253–263.
doi:10.1078/0171-9335-00244
Brandner JM, Haftek M, Niessen CM (2010) Adherens
References junctions, desmosomes and tight junctions in epider-
mal barrier function. Open Dermatol 4:7
Abdayem R, Callejon S, Jannin V, Portes P, Padois K, Buraczewska I, Berne B, Lindberg M, Torma H, Lodén M
Pirot F et al (2011) Modulation of the epidermal tight (2007) Changes in skin barrier function following
junctions with the self -emulsifying excipient labrasol. long-term treatment with moisturizers, a randomized
J Invest Dermatol 131(10):2146 controlled trial. Br J Dermatol 156(3):492–498
Atrux-Tallau N, Romagny C, Padois K, Denis A, Caussin J, Gooris GS, Groenink HW, Wiechers JW,
Haftek M, Falson F et al (2010) Effects of glycerol Bouwstra JA (2007) Interaction of lipophilic moistur-
on human skin damaged by acute sodium lauryl sul- izers on stratum corneum lipid domains in vitro and
phate treatment. Arch Dermatol Res 302(6):435–441. in vivo. Skin Pharmacol Physiol 20(4):175–186.
doi:10.1007/s00403-009-1021-z doi:10.1159/000101387
Barry BW (1991) Lipid-protein-partitioning theory of Chamlin SL, Kao J, Frieden IJ, Sheu MY, Fowler AJ,
skin penetration enhancement. J Control Release Fluhr JW et al (2002) Ceramide-dominant barrier
15(3):237–248. doi:10.1016/0168-3659(91)90115-T repair lipids alleviate childhood atopic dermatitis:
Behne M, Uchida Y, Seki T, de Montellano PO, Elias changes in barrier function provide a sensitive indica-
PM, Holleran WM (2000) Omega-hydroxyceramides tor of disease activity. J Am Acad Dermatol 47(2):198–
are required for corneocyte lipid envelope (CLE) 208. doi:10.1067/mjd.2002.124617
formation and normal epidermal permeability bar- Chen GS, Kim DD, Chien YW (1995) Dual-controlled trans-
rier function. J Invest Dermatol 114(1):185–192. dermal delivery of levonorgestrel and estradiol – enhanced
doi:10.1046/j.1523-1747.2000.00846.x permeation and modulated delivery. J Control Release
Bommannan D, Potts RO, Guy RH (1990) Examination of 34(2):129–143. doi:10.1016/0168-3659(95)00005-S
stratum-corneum barrier function in-vivo by infrared- Chin JH, Goldstein DB (1977) Effects of low concentra-
spectroscopy. J Invest Dermatol 95(4):403–408. tions of ethanol on fluidity of spin-labeled erythrocyte
doi:10.1111/1523-1747.Ep12555503 and brain membranes. Mol Pharmacol 13(3):435–441
Boncheva M, Damien F, Normand V (2008) Molecular Coderch L, Lopez O, de la Maza A, Parra JL (2003)
organization of the lipid matrix in intact stratum Ceramides and skin function. Am J Clin
corneum using ATR-FTIR spectroscopy. BBA Dermatol 4(2):107–129. doi:10.2165/00128071-
Biomembranes 1778(5):1344–1355. doi:10.1016/j. 200304020-00004
bbamem.2008.01.022 Corbe E, Laugel C, Yagoubi N, Baillet A (2007) Role
Bouwstra JA, Ponec M (2006) The skin barrier in healthy of ceramide structure and its microenvironment on
and diseased state. BBA Biomembranes 1758(12):2080– the conformational order of model stratum corneum
2095. doi:10.1016/j.bbamem.2006.06.021 lipids mixtures: an approach by FTIR spectroscopy.
Bouwstra JA, Gooris GS, Vanderspek JA, Bras W (1991) Chem Phys Lipids 146(2):67–75. doi:10.1016/j.
Structural investigations of human stratum-corneum chemphyslip.2006.12.010
by small-angle x-ray-scattering. J Invest Dermatol Corcuff P, Fiat F, Minondo AM, Lévêque JL, Rougier A
97(6):1005–1012. doi:10.1111/1523-1747.Ep12492217 (2002) A comparative ultrastructural study of hydroxy-
Bouwstra JA, Gooris GS, Dubbelaar FER, Weerheim acids induced desquamation. Eur J Dermatol
AM, Ponec M (1998) pH, cholesterol sulfate, and fatty 12(4):xxxix–xliii
acids affect the stratum corneum lipid organization. Costa-Balogh FO, Wennerstrom H, Wadso L, Sparr E
J Investig Dermatol Symp Proc 3(2):69–74 (2006) How small polar molecules protect membrane
86 L. Roussel et al.

systems against osmotic stress: the urea-water- alternative mechanism for skin moisturization. J Soc
phospholipid system. J Phys Chem B 110(47):23845– Cosmet Chem 41(1):51–65
23852. doi:10.1021/Jp0632440 Furuse M, Hata M, Furuse K, Yoshida Y, Haratake A,
Damien F, Boncheva M (2010) The extent of Sugitani Y et al (2002) Claudin-based tight junctions
orthorhombic lipid phases in the stratum corneum are crucial for the mammalian epidermal barrier: a
determines the barrier efficiency of human skin in lesson from claudin-1-deficient mice. J Cell Biol
vivo. J Invest Dermatol 130(2):611–614. doi:10.1038/ 156(6):1099–1111. doi:10.1083/jcb.200110122jcb.
Jid.2009.272 200110122
De Boer DJ, Hillier A (2001) The ACVD task force on Gao S, Singh J (1997) Mechanism of transdermal trans-
canine atopic dermatitis (XV): fundamental concepts in port of 5-fluorouracil by terpenes: carvone, 1,8-cineole
clinical diagnosis. Vet Immunol Immunopathol 81(3– and thymol. Int J Pharm 154(1):67–77. doi:10.1016/
4):271–276. doi:10.1016/S0165-2427(01)00312-9 S0378-5173(97)00123-3
De Jager MW, Gooris GS, Dolbnya IP, Ponec M, Ghadially R, Halkier-Sorensen L, Elias PM (1992) Effects
Bouwstra JA (2004) Modelling the stratum corneum of petrolatum on stratum corneum structure and func-
lipid organisation with synthetic lipid mixtures: the tion. J Am Acad Dermatol 26:387–396
importance of synthetic ceramide composition. BBA Gloor M, Wasik B, Gehring W, Grieshaber R, Kleesz P,
Biomembranes 1664(2):132–140. doi:10.1016/j. Fluhr JW (2004) Cleansing, dehydrating, barrier-
bbamem.2004.05.001 damaging and irritating hyperaemising effect of four
De Jager MW, Groenink HW, Guivernau RBI, Andersson detergent brands: comparative studies using stan-
E, Angelova N, Ponec M et al (2006) A novel in vitro dardised washing models. Skin Res Technol 10(1):1–
percutaneous penetration model: evaluation of barrier 9. doi:10.1111/j.1600-0846.2004.00045.x
properties with P-aminobenzoic acid and two of its Groen D, Poole DS, Gooris GS, Bouwstra JA (2011) Is an
derivatives. Pharm Res 23(5):951–960. doi:10.1007/ orthorhombic lateral packing and a proper lamellar orga-
s11095-006-9909-1 nization important for the skin barrier function? BBA
De Paepe K, Derde MP, Roseeuw D, Rogiers V (2000) Biomembranes 1808(6):1529–1537. doi:10.1016/j.
Incorporation of ceramide 3B in dermatocosmetic bbamem.2010.10.015
emulsions: effect on the transepidermal water loss of Grubauer G, Feingold KR, Elias PM (1987) Relationship
sodium lauryl sulphate-damaged skin. J Eur Acad of epidermal lipogenesis to cutaneous barrier function.
Dermatol. 14(4):272–279. doi:10.1046/j.1468-3083. J Lipid Res 28(6):746–752
2000.00103.x Haftek M, Teillon MH, Martini MC, Chambin O, Schmitt
Di Nardo A, Wertz PW, Giannetti A, Seidenari S (1998) D (1996) Structural and biochemical evaluation of the
Ceramide and cholesterol composition of the skin of epidermal barrier in ex-vivo permeabilized human
patients with atopic dermatitis. Acta Derm Venereol skin. In: Brain KR, James VJ, Walters KA (eds)
78(1):27–30 Prediction of percutaneous penetration, vol 4b. STS
Ding L, Zhang Y, Tatum R, Chen YH (2011) Publishing Ltd, Cardiff, pp 311–314
Detection of tight junction barrier function Haftek M, Teillon MH, Schmitt D (1998) Stratum cor-
in vivo by biotin. Methods Mol Biol 762:91–100. neum, corneodesmosomes and ex vivo percutaneous
doi:10.1007/978-1-61779-185-7_7 penetration. Microsc Res Tech 43(3):242–249.
Elfbaum SG, Laden K (1968) Effect of dimethyl sulfoxide doi: 10.1002/(SICI)1097029(19981101)43:3<242::
on percutaneous absorption – a mechanistic study I. AID-JEMT6>3.0.CO;2-G
J Soc Cosmet Chem 19(2):119–127 Haftek M, Simon M, Serre G (2006) Corneodesmosomes :
Elias PM (1983) Epidermal lipids, barrier function, and pivotal actors in the stratum corneum cohesion and
desquamation. J Invest Dermatol 80:S44–S49. desquamation. In: Elias PM, Feingold KR (eds)
doi:10.1111/1523-1747.Ep12537108 Skin barrier. Taylor & Francis, New York,
Elias PM, Friend DS (1975) The permeability barrier in pp 171–190
mammalian epidermis. J Cell Biol 65(1):180–191 Haftek M, Callejon S, Sandjeu Y, Padois K, Falson F,
Embery G, Dugard PH (1971) Isolation of dimethyl sulf- Pirot F et al (2011) Compartmentalization of the
oxide soluble components from human epidermal human stratum corneum by persistent tight junction-
preparations – possible mechanism of action of like structures. Exp Dermatol 20(8):617–621.
dimethyl sulfoxide in effecting percutaneous migra- doi:10.1111/j.1600-0625.2011.01315.x
tion phenomena. J Invest Dermatol 57(5):308–311. Hashimoto K (1971) Intercellular spaces of the human
doi:10.1111/1523-1747.Ep12292362 epidermis as demonstrated with lanthanum. J Invest
Feingold KR (2009) The outer frontier: the importance of Dermatol 57(1):17–31
lipid metabolism in the skin. J Lipid Res 50:S417– Heinemann C, Paschold C, Fluhr JW, Wigger-Alberti W,
S422. doi:10.1194/jlr.R800039-JLR200 Schliemann-Willers S, Farwanah H et al (2005)
Fluhr JW, Darlenski R, Surber C (2008) Glycerol and the Induction of a hardening phenomenon by repeated
skin: holistic approach to its origin and functions. Br J application of SLS: analysis of lipid changes in the
Dermatol 159(1):23–34 stratum corneum. Acta Derm Venereol 85(4):290–295.
Froebe CL, Simion FA, Ohlmeyer H, Rhein LD, Mattai J, doi:10.1080/00015550410026362
Cagan RH et al (1990) Prevention of stratum-corneum Holleran WM, Takagi Y, Uchida Y (2006) Epidermal
lipid phase-transitions in vitro by glycerol – an sphingolipids: metabolism, function, and roles in skin
7 Influence of Excipients on Two Elements of the Stratum Corneum Barrier 87

disorders. FEBS Lett 580(23):5456–5466. skin diffusion-model. Pharm Res 11(3):377–383.


doi:10.1016/j.febslet.2006.08.039 doi:10.1023/A:1018904802566
Hoogstraate AJ, Verhoef J, Brussee J, Ijzerman AP, Spies F, Krishnaiah YSR, Bhaskar P, Satyanarayana V (2004)
Bodde HE (1991) Kinetics, ultrastructural aspects and Penetration-enhancing effect of ethanol-water solvent
molecular modeling of transdermal peptide flux enhance- system and ethanolic solution of carvone on transder-
ment by n-alkylazacycloheptanones. Int J Pharm 76(1– mal permeability of nimodipine from HPMC gel
2):37–47. doi:10.1016/0378-5173(91)90341-K across rat abdominal skin. Pharm Dev Technol
Igawa S, Kishibe M, Murakami M, Honma M, 9(1):63–74. doi:10.1081/Pdt-120027419
Takahashi H, Iizuka H et al (2011) Tight junctions Kubo A, Nagao K, Yokouchi M, Sasaki H, Amagai M
in the stratum corneum explain spatial differences (2009) External antigen uptake by Langerhans cells
in corneodesmosome degradation. Exp Dermatol with reorganization of epidermal tight junction barri-
20(1):53–57. doi:10.1111/j.1600-0625.2010.01170.x ers. J Exp Med 206(13):2937–2946
Ishida-Yamamoto A, Simon M, Kishibe M, Miyauchi Y, Kurasawa M, Kuroda S, Kida N, Murata M, Oba A,
Takahashi H, Yoshida S et al (2004) Epidermal lamel- Yamamoto T et al (2009) Regulation of tight junction
lar granules transport different cargoes as distinct permeability by sodium caprate in human keratino-
aggregates. J Invest Dermatol 122(5):1137–1144. cytes and reconstructed epidermis. Biochem Biophys
doi:10.1111/j.0022-202X.2004.22515.x Res Commun 381(2):171–175. doi:10.1016/j.
Janssens M, van Smeden J, Gooris GS, Bras W, Portale G, bbrc.2009.02.005
Caspers PJ et al (2012) Increase in short-chain cerami- Kuriharabergstrom T, Knutson K, Denoble LJ, Goates CY
des correlates with an altered lipid organization and (1990) Percutaneous-absorption enhancement of an
decreased barrier function in atopic eczema patients. J ionic molecule by ethanol water-systems in human
Lipid Res 53(12):2755–2766. doi:10.1194/Jlr.P030338 skin. Pharm Res 7(7):762–766
Jiang SJ, Zhou XJ, Sun GQ, Zhang Y (2003) Kwak S, Brief E, Langlais D, Kitson N, Lafleur M,
Morphological alterations of the stratum corneum lip- Thewalt J (2012) Ethanol perturbs lipid organiza-
ids induced by sodium lauryl sulfate treatment in hair- tion in models of stratum corneum membranes: An
less mice. J Dermatol Sci 32(3):243–246. doi:10.1016/ investigation combining differential scanning calo-
S0923-1811(03)00134-8 rimetry, infrared and H-2 NMR spectroscopy. BBA
Johnson PH, Frank D, Costantino HR (2008) Discovery Biomembranes 1818(5):1410–1419. doi:10.1016/j.
of tight junction modulators: significance for drug bbamem.2012.02.013
development and delivery. Drug Discov Today 13(5– Langbein L, Pape UF, Grund C, Kuhn C, Praetzel S, Moll
6):261–267. doi:10.1016/j.drudis.2007.10.023 I et al (2003) Tight junction-related structures in the
Kang L, Ho PC, Chan SY (2006) Interactions between a absence of a lumen: occludin, claudins and tight junc-
skin penetration enhancer and the main components of tion plaque proteins in densely packed cell formations
human stratum corneum lipids – isothermal titration of stratified epithelia and squamous cell carcinomas.
calorimetry study. J Therm Anal Calorim 83(1):27– Eur J Cell Biol 82(8):385–400. doi:10.1078/0171-
30. doi:10.1007/s10973-005-7050-8 9335-00330
Kirschner N, Haftek M, Niessen CM, Behne MJ, Furuse Larrucea E, Arellano A, Santoyo S, Ygartua P (2001)
M, Moll I, Brandner JM (2010a) CD44 regulates tight Combined effect of oleic acid and propylene glycol on
junction assembly and barrier function. J Invest the percutaneous penetration of tenoxicam and its
Dermatol 131:932–943. doi:10.1038/jid.2010.390 retention in the skin. Eur J Pharm Biopharm
Kirschner N, Houdek P, Fromm M, Moll I, Brandner JM 52(2):113–119. doi:10.1016/S0939-6411(01)00158-8
(2010b) Tight junctions form a barrier in human epi- Lashmar UT, Hadgraft J, Thomas N (1989) Topical appli-
dermis. Eur J Cell Biol 89(11):839–842. doi:10.1016/j. cation of penetration enhancers to the skin of nude
ejcb.2010.07.010 mice: a histopathological study. J Pharm Pharmacol
Kirschner N, Rosenthal R, Furuse M, Moll I, Fromm M, 41:118–121
Brandner JM (2013) Contribution of tight junction Lawson EE, Anigbogu ANC, Williams AC, Barry BW,
proteins to ion, macromolecule, and water barrier in Edwards HGM (1998) Thermally induced molecular
keratinocytes. J Invest Dermatol 133(5):1161–1169. disorder in human stratum corneum lipids compared
doi:10.1038/jid.2012.507jid2012507 with a model phospholipid system: FT-Raman spec-
Kondoh M, Yagi K (2007) Tight junction modulators: troscopy. Spectrochim Acta A 54(3):543–558.
promising candidates for drug delivery. Curr Med doi:10.1016/S1386-1425(97)00268-0
Chem 14(23):2482–2488 Levang AK, Zhao K, Singh J (1999) Effect of ethanol pro-
Kondoh M, Yoshida T, Kakutani H, Yagi K (2008) pylene glycol on the in vitro percutaneous absorption
Targeting tight junction proteins-significance for drug of aspirin, biophysical changes and macroscopic bar-
development. Drug Discov Today 13(3–4):180–186. rier properties of the skin. Int J Pharm 181(2):
doi:10.1016/j.drudis.2007.11.005 255–263
Koyama Y, Bando H, Yamashita F, Takakura Y, Lévêque JL, Derigal J, Saintleger D, Billy D (1993) How
Sezaki H, Hashida M (1994) Comparative- does sodium lauryl sulfate alter the skin barrier function
analysis of percutaneous-absorption enhance- in man – a multiparametric approach. Skin Pharmacol
ment by D-Limonene and Oleic-acid based on a 6(2):111–115. doi:10.1159/000211095
88 L. Roussel et al.

Lodén M (2003) Role of topical emollients and moistur- Naik A, Pechtold LARM, Potts RO, Guy RH (1995)
izers in the treatment of dry skin barrier disorders. Am Mechanism of oleic acid-induced skin penetration
J Clin Dermatol 4(11):771–788 enhancement in vivo in humans. J Control Release
Lodén M (2012) Effect of moisturizers on epidermal bar- 37(3):299–306. doi:10.1016/0168-3659(95)00088-7
rier function. Clin Dermatol 30(3):286–296. Neto DD, Gooris GS, Bouwstra JA (2011) Effect of the
doi:10.1016/j.clindermatol.2011.08.015 omega-acylceramides on the lipid organization of stra-
Lodén M, Bárány E (2000) Skin-identical lipids versus tum corneum model membranes evaluated by X-ray
petrolatum in the treatment of tape-stripped and diffraction and FTIR studies (Part I). Chem Phys
detergent-perturbed human skin. Acta Derm Venereol Lipids 164(3):184–195. doi:10.1016/j.chemphyslip.
80(6):412–415 2010.12.007
Madison KC, Swartzendruber DC, Wertz PW, Downing Niessen CM (2007) Tight junctions/adherens junctions:
DT (1987) Presence of intact intercellular lipid lamel- basic structure and function. J Invest Dermatol
lae in the upper layers of the stratum-corneum. J Invest 127(11):2525–2532. doi:10.1038/sj.jid.5700865
Dermatol 88(6):714–718. doi:10.1111/1523-1747 Obata Y, Maruyama Y, Takayama K (2006) The mode of
Manabe E, Sugibayashi K, Morimoto Y (1996) Analysis promoting activity of O-Ethylmenthol as a transder-
of skin penetration enhancing effect of drugs mal absorption enhancer. Pharm Res 23(2):392–400.
by ethanol-water mixed systems with hydrody- doi:10.1007/s11095-005-9257-6
namic pore theory. Int J Pharm 129(1–2):211–221. Ongpipattanakul B, Francoeur ML, Potts RO (1994)
doi:10.1016/0378-5173(95)04328-4 Polymorphism in stratum-corneum lipids. BBA
Mao-Qiang M, Feingold KR, Elias PM (1993) Exogenous Biomembranes 1190(1):115–122. doi:10.1016/
lipids influence permeability barrier recovery 0005-2736(94)90040-X
in acetone-treated murine Skin. Arch Dermatol Panchagnula R, Salve PS, Thomas NS, Jain AK, Ramarao
129(6):728–738. doi:10.1001/archderm.129.6.728 P (2001) Transdermal delivery of naloxone: effect of
Mao-Qiang M, Brown BE, Wu-Pong S, Feingold KR, water, propylene glycol, ethanol and their binary
Elias PM (1995) Exogenous nonphysiologic vs physi- combinations on permeation through rat skin. Int J
ologic lipids. Divergent mechanisms for correction of Pharm 219(1–2):95–105. doi:10.1016/S0378-5173
permeability barrier dysfunction. Arch Dermatol (01)00634-2
131(7):809–816 Pensack RD, Michniak BB, Moore DJ, Mendelsohn R
Mao-Qiang M, Feingold KR, Thornfeldt CR, Elias PM (2006) Infrared kinetic/structural studies of barrier ref-
(1996) Optimization of physiological lipid mixtures ormation in intact stratum corneum following thermal
for barrier repair. J Invest Dermatol 106(5):1096– perturbation. Appl Spectrosc 60(12):1399–1404.
1101. doi:10.1111/1523-1747.Ep12340135 doi:10.1366/000370206779321445
Masukawa Y, Narita H, Sato H, Naoe A, Kondo N, Sugai Y Pilgram GSK, Vissers DCJ, van der Meulen H, Pavel S,
et al (2009) Comprehensive quantification of ceramide Lavrijsen SPM, Bouwstra JA et al (2001a) Aberrant
species in human stratum corneum. J Lipid Res lipid organization in stratum corneum of patients with
50(8):1708–1719. doi:10.1194/jlr.M800014-JLR200 atopic dermatitis and lamellar ichthyosis. J Invest
Mattai J, Froebe CL, Rhein LD, Simion FA, Ohlmeyer H, Dermatol 117(3):710–717. doi:10.1046/j.0022-202x.
Su DT et al (1993) Prevention of model stratum- 2001.01455.x
corneum lipid phase-transitions in vitro by cosmetic Pilgram GSK, van der Meulen J, Gooris GS, Koerten HK,
additives – differential scanning calorimetry, optical Bouwstra JA (2001b) The influence of two azones and
microscopy, and water evaporation studies. J Soc sebaceous lipids on the lateral organization of lipids
Cosmet Chem 44(2):89–100 isolated from human stratum corneum. BBA
Menon GK, Elias PM (1997) Morphologic basis for a Biomembranes 1511(2):244–254. doi:10.1016/S0005-
pore-pathway in mammalian stratum corneum. Skin 2736(01)00271-1
Pharmacol 10(5–6):235–246 Pinnagoda J, Tupker RA, Agner T, Serup J (1990)
Moore DJ, Rerek ME, Mendelsohn R (1997) FTIR spec- Guidelines for transepidermal water loss (TEWL)
troscopy studies of the conformational order and phase measurement. A report from the Standardization
behavior of ceramides. J Phys Chem B 101(44):8933– Group of the European Society of Contact Dermatitis.
8940. doi:10.1021/Jp9718109 Contact Dermatitis 22(3):164–178
Morita K, Sasaki H, Furuse M, Tsukita S (1999) Ponec M, Boelsma E, Weerheim AM, Mulder A,
Endothelial claudin: Claudin-5/TMVCF constitutes Bouwstra JA, Mommaas M (2000) Lipid and ultra-
tight junction strands in endothelial cells. J Cell Biol structural characterization of reconstructed skin mod-
147(1):185–194. doi:10.1083/jcb.147.1.185 els. Int J Pharm 203(1–2):211–225. doi:10.1016/S0378-
Na JI, Hwang JS, Park HJ, Kim DH, Park WS, Youn 5173(00)00459-2
SW et al (2010) A new moisturizer containing Popa I, Remoue N, Hoang LT, Pin D, Gatto H, Haftek
physiologic lipid granules alleviates atopic derma- M et al (2011) Atopic dermatitis in dogs is associ-
titis. J Dermatol Treat 21(1):23–27. doi:10.3109/ ated with a high heterogeneity in the distribution of
09546630903085336 protein-bound lipids within the stratum corneum.
7 Influence of Excipients on Two Elements of the Stratum Corneum Barrier 89

Arch Dermatol Res 303(6):433–440. doi:10.1007/ Tfayli A, Guillard E, Manfait M, Baillet-Guffroy A (2010)
s00403-011-1120-5 Thermal dependence of Raman descriptors of cerami-
Popa I, Remoue N, Osta B, Pin D, Gatto H, Haftek M des. Part I: effect of double bonds in hydrocarbon
et al (2012) The lipid alterations in the stratum cor- chains. Anal Bioanal Chem 397(3):1281–1296.
neum of dogs with atopic dermatitis are alleviated doi:10.1007/s00216-010-3614-y
by topical application of a sphingolipid-contain- Tfayli A, Guillard E, Manfait M, Baillet-Guffroy A (2012)
ing emulsion. Clin Exp Dermatol 37(6):665–671. Molecular interactions of penetration enhancers
doi:10.1111/j.1365-2230.2011.04313.x within ceramides organization: a Raman spectroscopy
Pummi K, Malminen M, Aho H, Karvonen SL, Peltonen approach. Analyst 137(21):5002–5010. doi:10.1039/
J, Peltonen S (2001) Epidermal tight junctions: C2an35220f
ZO-1 and occludin are expressed in mature, devel- Thau P (2002) Glycerin (glycerol): current insights into
oping, and affected skin and in vitro differentiating the functional properties of a classic cosmetic raw
keratinocytes. J Invest Dermatol 117(5):1050–1058. material. J Cosmet Sci 53(4):229–236
doi:10.1046/j.0022-202x.2001.01493.x Torma H, Berne B (2009) Sodium lauryl sulphate
Rawlings AV, Scott IR, Harding CR, Bowser PA (1994) alters the mRNA expression of lipid-metabolizing
Stratum-corneum moisturization at the molecular- enzymes and PPAR signaling in normal human
level. J Invest Dermatol 103(5):731–740. doi:10.1007/ skin in vivo. Exp Dermatol 18(12):1010–1015.
s004030050067 doi:10.1111/j.1600-0625.2009.00877.x
Rawlings AV, Harding CR, Watkinson A, Banks J, Tsukita S, Furuse M (2000) Pores in the wall: claudins con-
Ackerman C, Sabin R (1995) The effect of glycerol stitute tight junction strands containing aqueous pores.
and humidity on desmosome degradation in stratum J Cell Biol 149(1):13–16. doi:10.1083/Jcb.149.1.13
corneum. Arch Dermatol Res 287(5):457–464. Vaddi HK, Ho PC, Chan SY (2002) Terpenes in propylene
doi:10.1007/Bf00373429 glycol as skin-penetration enhancers: permeation and
Ribaud C, Garson JC, Doucet J, Lévêque JL (1994) partition of haloperidol, Fourier transform infrared
Organization of stratum-corneum lipids in relation to spectroscopy, and differential scanning calorimetry.
permeability – influence of sodium lauryl sulfate and J Pharm Sci 91(7):1639–1651
preheating. Pharm Res 11(10):1414–1418. doi:10.102 Van Hal DA, Jeremiasse E, Junginger HE, Spies F,
3/A:1018987721531 Bouwstra JA (1996) Structure of fully hydrated human
Rissmann R, Oudshoorn MH, Kocks E, Hennink WE, stratum corneum: a freeze-fracture electron micros-
Ponec M, Bouwstra JA (2008) Lanolin-derived lipid copy study. J Invest Dermatol 106(1):89–95.
mixtures mimic closely the lipid composition and doi:10.1111/1523-1747.ep12328031
organization of vernix caseosa lipids. Biochim Van Smeden J, Hoppel L, van der Heijden R, Hankemeier
Biophys Acta 1778(10):2350–2360. doi:10.1016/j. T, Vreeken RJ, Bouwstra JA (2011) LC/MS analysis
bbamem.2008.06.017 of stratum corneum lipids: ceramide profiling and dis-
Saad P, Flach CR, Walters RM, Mendelsohn R (2012) covery. J Lipid Res 52(6):1211–1221. doi:10.1194/Jlr.
Infrared spectroscopic studies of sodium dodecyl sul- M014456
phate permeation and interaction with stratum cor- Verdier-Sevrain S, Bonté F (2007) Skin hydration: a review
neum lipids in skin. Int J Cosmet Sci 34(1):36–43. on its molecular mechanisms. J Cosmet Dermatol
doi:10.1111/j.1468-2494.2011.00678.x 6(2):75–82. doi:10.1111/j.1473-2165.2007.00300.x
Schluter H, Wepf R, Moll I, Franke WW (2004) Sealing Warner RR, Boissy YL, Lilly NA, Spears MJ, McKillop
the live part of the skin: the integrated meshwork of K, Marshall JL et al (1999) Water disrupts stra-
desmosomes, tight junctions and curvilinear ridge tum corneum lipid lamellae: damage is similar to
structures in the cells of the uppermost granular layer surfactants. J Invest Dermatol 113(6):960–966.
of the human epidermis. Eur J Cell Biol 83(11– doi:10.1046/j.1523-1747.1999.00774.x
12):655–665. doi:10.1078/0171-9335-00434 Wartewig S, Neubert RHH (2007) Properties of ceramides
Sonoda N, Furuse M, Sasaki H, Yonemura S, Katahira J, and their impact on the stratum corneum structure: a
Horiguchi Y et al (1999) Clostridium perfringens review. Skin Pharmacol Physiol 20(5):220–229.
enterotoxin fragment removes specific claudins from doi:10.1159/00010442
tight junction strands: Evidence for direct involvement Weerheim AM, Ponec M (2001) Determination of stratum
of claudins in tight junction barrier. J Cell Biol corneum lipid profile by tape stripping in combination
147(1):195–204 with high-performance thin-layer chromatography.
Swartzendruber DC, Burnett IH, Wertz PW, Madison KC, Arch Dermatol Res 293(4):191–199. doi:10.1007/
Squier CA (1995) Osmium tetroxide and ruthenium s004030100212
tetroxide are complementary reagents for the prepara- Wei TL, Geijer S, Lindberg M, Berne B, Torma H
tion of epidermal samples for transmission electron (2006) Detergents with different chemical properties
microscopy. J Invest Dermatol 104(3):417–420. induce variable degree of cytotoxicity and mRNA
doi:10.1111/1523-1747.ep12665909 expression of lipid-metabolizing enzymes and
90 L. Roussel et al.

differentiation markers in cultured keratinocytes. enhancers towards 5-fluorouracil as assessed with


Toxicol in Vitro 20(8):1387–1394. doi:10.1016/j. time – permeation, partitioning and differential
tiv.2006.06.002 scanning calorimetry. Int J Pharm 116(2):237–51.
Wertz PW (2000) Lipids and barrier function of the skin. doi:10.1016/0378-5173(94)00312-S
Acta Derm Venereol Supplementum 208:7–11 Zettersten EM, Ghadially R, Feingold KR, Crumrine D,
Wertz PW, Madison KC, Downing DT (1989) Covalently Elias PM (1997) Optimal ratios of topical stratum cor-
bound lipids of human stratum corneum. J Invest neum lipids improve barrier recovery in chronologi-
Dermatol 92(1):109–111 cally aged skin. J Am Acad Dermatol 37(3):403–8.
Williams AC, Barry BW (2012) Penetration enhancers. doi:10.1016/S0190-9622(97)70140-3
Adv Drug Deliv Rev 64:128–137. doi:10.1016/j. Zhao KD, Singh J (2000) Mechanism(s) of in vitro percu-
addr.2012.09.032 taneous absorption enhancement of tamoxifen by
Wong V, Gumbiner BM (1997) A synthetic peptide cor- enhancers. J Pharm Sci 89(6):771–80. doi:10.1002/
responding to the extracellular domain of occludin (Sici)1520-6017(200006)89:6<771::Aid-
perturbs the tight junction permeability barrier. J Cell Jps9>3.0.Co;2-Y
Biol 136(2):399–409. doi:10.1083/jcb.136.2.399 Zheng YX, Yin HY, Boeglin WE, Elias PM, Crumrine D,
Yamamoto T, Saeki Y, Kurasawa M, Kuroda S, Arase S, Beier DR et al (2011) Lipoxygenases mediate the
Sasaki H (2008) Effect of RNA interference of tight effect of essential fatty acid in skin barrier formation a
junction-related molecules on intercellular barrier func- proposed role in releasing omega-hydroxyceramide
tion in cultured human keratinocytes. Arch Dermatol for construction of the corneocyte lipid envelope.
Res 300(9):517–524. doi:10.1007/s00403-008-0868-8 J Biol Chem 286(27):24046–56. doi:10.1074/jbc.
Yamane MA, Williams AC, Barry BW (1995) Effects M111.251496
of terpenes and oleic-acid as skin penetration
Part II
Penetration Enhancement Techniques
in Skin Delivery
Targets in Dermal
and Transdermal Delivery 8
and Classification of Penetration
Enhancement Methods

Jelena Predic Atkinson, Howard I. Maibach,


and Nina Dragicevic

Contents 8.1 Introduction


8.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . 93
In the past three decades, the skin has gradually
8.2 Therapeutic Target Sites in Topical
and Transdermal Drug Delivery . . . . . . . 94
become recognized as an important drug delivery
route. Being the most accessible organ in the body,
8.3 The Skin . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
the skin can be reached directly and so drug deliv-
8.4 Drug Transport Routes Through ery to this tissue is assumed to be relatively easy.
the Skin . . . . . . . . . . . . . . . . . . . . . . . . . . . 97 There is considerable interest in the skin as a site
8.4.1 Factors Affecting Drug Permeation
Rate Through the Skin . . . . . . . . . . . . . . . . 98 of drug application for both local (topical) and sys-
temic (transdermal) effect, the first used in the
8.5 Penetration Enhancement Technique
Classification . . . . . . . . . . . . . . . . . . . . . . . 99
treatment of different skin diseases and the latter
8.5.1 Chemical Methods for Penetration as an alternative route for systemic drug adminis-
Enhancement . . . . . . . . . . . . . . . . . . . . . . . . 99 tration. Advantages offered by this kind of drug
8.5.2 Physical Methods for Penetration delivery are numerous compared to other conven-
Enhancement . . . . . . . . . . . . . . . . . . . . . . . . 103
tional routes (Parikh et al. 1984; Guy et al. 1987;
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104 Schreier and Bouwstra 1994; Paudel et al. 2010):
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104 1. Transdermal drug delivery systems (TDDS)
avoid hepatic first pass which allows for lower
doses of drugs to be administered and that
means these methods are safer for patients
with liver diseases.
2. TDDS avoid the gastrointestinal tract and so
J.P. Atkinson (*)
56a Dollis Park, London N3 1BS, UK bypass problems like drastic pH changes, the
e-mail: jelena.atkinson@gmail.com deleterious presence of food enzymes, vari-
H.I. Maibach able transit times and rapidly fluctuating drug
Department of Dermatology, University of California plasma concentrations.
Medical School, UCSF, San Francisco, CA, USA 3. TDDS are an acceptable, pain-free, non-
e-mail: maibachh@derm.ucsf.edu
invasive form of self-administration for patients
N. Dragicevic (*) which ensures easy patient compliance and
Apoteka “Beograd”,
quick ending of the therapy if necessary.
Bojanska 16/IV, 11 000 Belgrade, Serbia
e-mail: ninadragicevic@hotmail.com; 4. TDDS act as a “depot” controlling the rate of
nina.dragicevic-curic@apotekabeograd.co.rs drug input over a prolonged period of time

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 93


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_8, © Springer-Verlag Berlin Heidelberg 2015
94 J.P. Atkinson et al.

and ensuring constant plasma levels even for Topically delivered drugs should have their phar-
drugs with short half-times. macological or other effects confined to the sur-
In the case of drugs with a narrow thera- face of the skin or within the skin (Flynn and
peutic margin, when applied onto the skin in Weiner 1991). Formulations designed to target
TDDS, their undesirable side effects are the skin surface include sunscreens, barrier prod-
reduced, particularly the effects associated ucts, cosmetics and insect repellents (Benson and
with pulsed peak plasma levels. Furthermore, Watkinson 2012). In addition to these, topical
the dose interval can be reduced. formulations can target appendages (hair follicles
5. Topical drug delivery systems allow the drug and sweat pores) and include antiacne products,
to be directly applied to the skin and delivered antiperspirants, hair growth promoters and
to the site of disease in the skin. anti-infectives.
6. Topical drug delivery systems like TDDS are Regional delivery involves the application of a
non-invasive, avoid hepatic first pass as well drug to the skin in order to treat diseases or alle-
as the gastrointestinal tract and problems viate disease symptoms in tissues that lie deeper,
associated with this drug application route, beneath the application site. Pharmacological tar-
increase patient compliance and can be gets of this type of drug delivery are within the
self-administered. musculature, vasculature, joints and tissues
7. Specially designed topical delivery systems beneath and around the site of application. When
(like liposomes) may also form drug “depots” targeting regional sites, drug formulations aim to
in the skin with sustained drug release. have a regionally selective effect. Regional drug
The problems that topical/transdermal drug concentrations upon this route of drug adminis-
delivery systems encounter are the low permea- tration are higher than the ones achieved by sys-
bility of the stratum corneum which limits the temic administration (Flynn and Weiner 1991).
number of drugs available as transdermal prod- For both topical and regional drug delivery, sys-
ucts and the potential interaction of drugs with temic absorption is unwanted but unavoidable.
the skin causing irritation and sensitization. In transdermal delivery drugs are applied to
the skin with the aim of reaching the systemic
circulation. The purpose of this type of drug
8.2 Therapeutic Target Sites delivery is to achieve a therapeutically relevant
in Topical and Transdermal drug level in order to treat a systemic disease.
Drug Delivery Hence, the percutaneous absorption of the drug is
essential, while the local deposition of the drug is
During topical and transdermal drug delivery, unwanted, but unavoidable (Flynn and Weiner
drugs are applied to the skin after which they 1991). The use of transdermal delivery is limited
should follow a route to one of the following tar- to only a small pool of drugs (see Table 8.1) due
get sites (Fig. 8.1): (1) the local tissues immedi- to the selective barrier properties of the skin. The
ately beneath the application site, (2) deep regions small number of candidates for this delivery route
in the vicinity of (but still somewhat remote from) is a result of the fact that only a few drug mole-
the application site and (3) the systemic circula- cules have skin permeability coefficients suffi-
tion (Flynn and Weiner 1991). Therefore, it is ciently high to achieve clinically active plasma
important to develop an adequate formulation levels. Currently, the market for transdermal
which delivers the drug to the desired target in the patches comprises patches with a few low molec-
skin or below the skin, i.e. to differ between topi- ular weight drugs: scopolamine for motion sick-
cal, regional and transdermal drug delivery, since ness, clonidine and nitroglycerin for
each application has its specific requirements. cardiovascular disease, fentanyl for chronic pain,
Topical delivery can be defined as the applica- nicotine to aid smoking cessation, oestradiol
tion of a drug-containing formulation to the skin (alone or in combination with levonorgestrel or
to directly treat cutaneous disorders or the cuta- norethisterone) for hormone replacement and tes-
neous manifestations of a general disease. tosterone for hypogonadism (Benson 2005).
8 Targets in Dermal and Transdermal Delivery and Classification of Penetration Enhancement Methods 95

Fig. 8.1 Targets in dermal and transdermal drug delivery

Since percutaneous absorption is pivotal to the 8.3 The Skin


effectiveness of both topical and transdermal sys-
tems, significant efforts have been devoted to The skin is the largest organ in humans cov-
developing strategies to overcome the imperme- ering approximately 2 m2 in an average-sized
ability of the intact human skin. There are many adult. Its main role is to prevent water loss
ways for circumventing the stratum corneum, and protect our body from undesired outside
which provides the main barrier for drug penetra- influences. This implies that the skin acts as
tion (Barry 2001). a barrier for diffusion of substances into the
96 J.P. Atkinson et al.

Table 8.1 List of marketed transdermal products


Generic drug Indication Product Manufacturer
1. Scopolamine Motion sickness Transderm Scop® Novartis
2. Nitroglycerin Angina pectoris Minitran®, Nitrol®, 3 M, Rorer, Novartis, Key
Transderm-Nitro®, Pharms
Nitro-Dur®
3. Clonidine Hypertension Catapres-TTS® Boehringer Ingelheim
4. Estradiol Postmenopausal related Estraderm®, Climara® Novartis, Bayer
symptoms HealthCare
5. Nicotine Smoking cessation Nicoderm CQ®, Sanofi-Aventis, Novartis
Habitrol®
6. Testosteron Hypogonadism Androderm®, Watson Labs, Alza
Testoderm®
7. Fentanyl Analgesia Duragesic® Janssen Pharmaceuticals
8. Estradiol and Postmenopausal related Climara ProTM Bayer Healthcare
levonorgestrel symptoms
9. Estradiol and Postmenopausal related Combipatch® Novartis
norethindrone symptoms
10. Ethinyl estradiol and Contraception Ortho Evra® Janssen Pharmaceuticals
norelgestromin
11. Buprenorphine Analgesia Bu Trans® Purdue Pharma L.P.
12. Rivastigmine Dementia associated with Exelon® Novartis
Alzheimer’s and Parkinson’s
disease
13. Oxybutynin Overactive bladder Oxytrol®, Kentera® Watson Labs, Orion
Pharma
14. Oxybutynin chloride Overactive bladder Gelnique® Watson Labs,
15. Rotigotine Parkinson’s disease Neupro® UCB Inc
16. Granisetron Nausea, vomiting Sancuso® ProStrakan Inc
17. Methylphenidate Attention deficithyperactivity Daytrana Noven Pharms Inc
disorder
18. Selegiline Depression Emsam® Somerset
19. Lidocaine Postherpetic neuralgia pain Lidoderm® Teikoku Phar
relief
20. Lidocaine and tetracaine Local dermal analgesia Synera® Zars Pharma
21. Capsaicin Postherpetic neuralgia pain Qutenza® NeurogesX
relief
22. Diclofenac epolamine Topical pain relief Flector® Inst Biochem
23. Diclofenac sodium Topical pain relief in Voltaren® Novartis
osteoarthritis

underlying tissue (Schaefer 1996; Bouwstra 100 μm), which consists of stratum granulosum,
et al. 2003). The main problem in the der- stratum spinosum and stratum basale. The viable
mal/transdermal administration of drugs is epidermis is responsible for the generation of the
overcoming this natural barrier (Barry 2001; stratum corneum (Schaefer 1996).
Bouwstra et al. 2003). The stratum corneum (horny layer, SC) is the
The skin is composed of two anatomically dis- final product of keratinocyte differentiation (cor-
tinct layers: the epidermis and the dermis. nification). It is made of layers of metabolically
Beneath the dermis is the fatty subcutaneous inactive cells, embedded in an extracellular
layer hypodermis (See Fig. 8.1). The epidermis is matrix of lamellar lipid bilayers. Corneocytes
composed of the stratum corneum (10–20 μm provide the physical and chemical stability of the
thick) and the underlying viable epidermis (50– SC, while the extracellular matrix gives it the
8 Targets in Dermal and Transdermal Delivery and Classification of Penetration Enhancement Methods 97

rigid structure, impermeable barrier for water and forming the bulk of the skin. It is made of a net-
water-soluble compounds. The SC can be consid- work of connective tissue, and elastic tissue
ered as a wall consisting of polyhedric squeeze- embedded in a mucopolysaccharide gel (Wilkes
protein “bricks” and water-depleted stiff lipid et al. 1973). The collagen fibres in the connective
sheets as “mortar” (Ghyczy 2002). The protec- tissue give the dermis support and the elastic tis-
tion of the skin is provided primarily by the SC, sue provides flexibility. The following structures
which due to its specific structure provides the are embedded in the dermis: blood and lym-
primary barrier to percutaneous absorption of phatic vessels, nerve endings, pilosebaceous
compounds as well as to water loss (Lindberg and units (hair follicles and sebaceous glands) and
Forslind 2000; Bouwstra et al. 2003). In addition sweat glands (eccrine and apocrine). Fibroblasts,
to the stratum corneum, recent findings showed endothelilal cells and mast cells are present in
that the viable epidermis is also a rate-limiting the dermis, and during inflammation or wound
barrier to drug penetration (Andrews et al. 2012). healing, macrophages, lymphocytes and leuko-
Corneocytes represent cell remnants of termi- cytes may infiltrate (Schaefer 1996). Blood car-
nally differentiated keratinocytes of the viable ries the molecules away from near the
epidermis. It is the composition of the corneo- dermo-epidermal layer, making dermal concen-
cytes that gives the SC its strong barrier proper- trations of most permeants low. The formed con-
ties. Corneocyte layers are made up of centration gradient provides the driving force for
cross-linked proteins and covalently bound lip- drug permeation. In addition to blood, the lym-
ids. The proteins resist chemical and physical phatic system may also remove permeated mol-
denaturation and the lipids resist solubilization ecules from the dermis, maintaining a driving
(Schaefer 1996; Downing and Stewart 2000; force for permeation.
Williams 2003). In addition to these there is the In terms of transdermal drug delivery, the der-
corneocyte protein envelope which is added dur- mis provides a minimal barrier to the delivery of
ing the cornification process (Downing and most polar drugs, but may significantly limit the
Stewart 2000). The insoluble cornified envelope penetration of highly lipophilic molecules
is stabilized through core proteins (90 % of its (Williams 2003).
dry mass) cross-linked to the envelope and
through covalently bound lipids (10 % of its dry
mass) (Schaefer 1996). Therefore, the two layers
of the envelope are the layer adjacent to the cyto- 8.4 Drug Transport Routes
plasm which is thick and composed of structural Through the Skin
proteins and the layer on the exterior of the pro-
tein layer which is composed of lipids. The lipid A molecule placed on the skin surface could
layer serves as an anchor to the keratinocytes and reach the viable tissue: (1) via the appendages,
links the proteinaceous domains to the intercel- (2) across the transcellular route and (3) across
lular lipid domains. the intercellular route (Fig. 8.1).
Intercellular lipids are arranged in a crystal- The transappendageal transport (shunt route
line sublattice, with only a small portion of lipids transport) involves the transport through the
in a liquid phase. The crystalline lipid sublattice pilosebaceous unit (hair follicles with sebaceous
is far less permeable to water than the liquid lipid glands) and through sweat ducts. Hair follicles
phase. The low permeability of the SC is due not are the most important appendages in terms
only to the unique lipid composition but also to of surface area (Schaefer 1996). It is generally
the unique structural organization of the lipid assumed that this pathway contributes margin-
phase (Downing and Stewart 2000; Lindberg and ally to the steady-state drug flux (Redelmeier and
Forslind 2000; Bouwstra et al. 2003; Feingold Kitson 1999; Agarwal et al. 2000; Barry 2006).
et al. 1990). The reason for this is that the skin area covered
The dermis (or corium) is typically 3–5 mm with the appendages is proportionally smaller
thick and is the major component of human skin than the total skin surface area (Scheuplein 1967).
98 J.P. Atkinson et al.

However, appendages may function as shunts, (Barry 2006). According to the domain mosaic
which may be important at short times prior to model of the skin barrier (Forslind 1994), the
steady-state diffusion (Barry 2006). Appendages structural organization of the lipids of the SC has
can contribute to transdermal drug delivery to two phases: (1) lipids in crystalline/gel state sur-
a varied degree. Some results show that their rounded by (2) lipids that form more fluid (liquid)
(appendageal) contribution is small (Siddiqui crystalline domains. This second, more fluid lipid
et al. 1989), whilst others showed that these domains provide the pathway by which permeants
shunts are indeed important in skin permeation traverse the SC. A method combining ultradeform-
for a range of drugs (Illel et al. 1991). The same able lipid vesicles (Transfersomes®) with confocal
author (1997) also suggested that hair follicles laser scanning microscopy (CLSM) showed the
and sebaceous glands can be privileged pathways existence of two different hydrophilic pathways in
for some molecules or formulations, which enter the SC: an “intercluster” pathway and an intercor-
faster into these shunts than they do through the neocyte pathway (Schätzlein and Cevc 1998). The
SC. Changing certain parameters in a formula- intercluster route runs between clusters of 3–10
tion (such as pH, solvent, penetration enhancers) neighbouring corneocyte “columns”. This path-
can influence follicular drug permeation (Frum way has low resistance to molecule penetration
et al. 2008). In addition to this some methods and it comprises ≤1 % of the total skin surface or
used for transdermal permeation enhancement, ≤20 % of the pathway area in the skin. The inter-
such as liposomes and iontophoresis, increase corneocyte pathway runs between all the corneo-
the flux of drugs through hair follicles (Li and cytes in a cluster and is therefore very tortuous.
Hoffman 1997; Hoffman 1998; Ciotti and Weiner This transdermal permeation route resists penetra-
2002). Lauer (1999) reviewed in detail the fol- tion better and is more abundant (≥3 % of the skin
licular delivery. or ≥80 % of the pathway area). Van Kuijk-
The transcellular route leads directly across Meuwissen et al. (1998) showed by CLSM that the
the SC, involving the drug transport through liposomally entrapped fluorescent label travelled
keratinocytes and intercellular lipid lamellae. across SC mainly via the intercellular route.
The consecutive partitioning of the drug
between hydrophilic (keratine) and hydropho-
bic (lipids) parts of the SC makes this a difficult 8.4.1 Factors Affecting Drug
pathway. The nature of the permeant and the Permeation Rate Through
partitioning coefficient will influence the the Skin
importance of this route. Hydrophilic mole-
cules may prefer the transcellular route at a Factors affecting the drug permeation rate
pseudo-steady state. However, lipid bilayers are through SC can be considered using the equation
the rate-limiting barrier for permeation via this (Eq. 8.1) for steady-state flux (Barry 1983):
route (Williams 2003).
dm DC0 K
The intercellular route is through the lipid = (8.1)
bilayers, which comprise around 1 % of the SC dt h
diffusional area, yet provide the only continuous where dm/dt is the steady-state flux, presenting
phase within the membrane. It is generally the cumulative mass of the diffusant, m, passing
accepted that, except for some specialized cases per unit area through the membrane; C0 is the
(e.g. highly hydrophilic substances), the intercel- constant donor drug concentration; K is the parti-
lular lipid route is the principal pathway by which tion coefficient of a solute between membrane
most small, uncharged molecules traverse the SC and bathing solution; D is the diffusion coeffi-
(Loth 1992; Abraham et al. 1995; Roberts et al. cient; and h is the membrane thickness. From
1996; van Kuijk-Meuwissen et al. 1998) and many Eq. 8.1, the ideal properties for a molecule in
methods for enhancing the drug penetration dis- order to penetrate SC well would be the follow-
rupt or bypass the intercellular lipid bilayers of SC ing (Barry 2001; Benson 2005):
8 Targets in Dermal and Transdermal Delivery and Classification of Penetration Enhancement Methods 99

• Low molecular mass, preferably less than Prausnitz and Langer (2008) proposed catego-
600 Da, when D tends to be high. rizing TDDS into three generations of develop-
• Adequate solubility in oil and water in order ment. Drugs in the first generation of TDDS have
to achieve a high membrane concentration low molecular weight (Mw), are lipophilic,
gradient, which is the driving force for diffu- achieve efficacy at low doses and generally do
sion (C0 is large). not require penetration enhancement. The second
• High, but balanced (optimal) K, since a too generation of TDDS utilize enhancement, such as
high coefficient may inhibit clearance from chemical enhancers, iontophoresis and ultra-
viable tissues. This parameter is very impor- sound but have been limited to the delivery of
tant in establishing a high initial penetrant small Mw molecules. The third generation of
concentration in the first layer of the TDDS delivers macromolecules to the SC with
SC. Molecules showing intermediate partition the help of novel chemical enhancers, electro-
coefficients (log K octanol/water of 1–3) have poration, cavitational ultrasound, microneedles,
adequate solubility within lipid domains of thermal ablation and microdermabrasion.
the SC (to permit diffusion through this
domain) whilst still having a sufficiently
hydrophilic nature to allow partitioning into 8.5.1 Chemical Methods
the viable epidermis. for Penetration Enhancement
• Low melting point, which correlates with
good solubility as predicted by the ideal solu- Chemical penetration enhancers are defined as
bility theory. agents that partition into and interact with the SC
When a drug possesses ideal physicochemical constituents to induce a temporary, reversible
properties (as in the case of nicotine and nitro- increase in skin permeability. These substances
glycerin), transdermal delivery is feasible. If the temporarily reduce skin resistance and thereby
drug does not match these ideal characteristics, enhance drug flux (Barry 2001). Different groups
manipulation of the drug or vehicle to enhance of structurally related chemical compounds are
diffusion is necessary and/or penetration used as penetration enhancers (see Volume 3,
enhancement techniques are used. Part 2): water, surfactants, essential oils, terpenes
and their derivatives, fatty acids, esters, ethers,
Azone and its derivatives, transkarbams, amides,
8.5 Penetration Enhancement pyrrolidones, sulphoxides and their analogues,
Technique Classification etc. (Buyuktimkin et al. 1997; Williams and
Barry 2004; Babu and Pandit 2005; Bugaj et al.
Lots of techniques reported in literature (Barry 2006; Puglia and Bonina 2008; Karande and
2001; Benson 2005; Rizwan et al. 2009) are suc- Mitragotri 2009; Mittal et al. 2009; Brychtova
cessful in enhancing the drug delivery into/ et al. 2010; Ibrahim and Li 2010; Karakatsani
through the skin. These methods can be grouped et al. 2010; Salerno et al. 2010). Chemical pene-
initially into chemical and physical methods tration enhancers represent the most studied pen-
(Table 8.2). The most extensively studied chemi- etration enhancement method as they have been
cal methods include chemical penetration shown to enhance the topical as well as transder-
enhancers (Williams and Barry 2004; Ahad et al. mal delivery of a broad range of drugs both
2009), vesicles (El Maghraby and Williams lipophilic and hydrophilic. As an example pyr-
2009) and prodrugs (Kasting et al. 1992). rolidones enhance permeation of hydrophilic
Iontophoresis (Costello and Jeske 1995), electro- (e.g. mannitol, 5-fluorouracil and sulphaguani-
poration (Wang et al. 1998), ultrasound (Cancel dine) and lipophilic drugs (betamethasone-17-
et al. 2004) and most recently microneedles benzoate, hydrocortisone and progesterone)
(Sivamani et al. 2009) are the most studied physi- (Williams and Barry 2004), as well as terpenes,
cal methods. showing enhanced skin permeation of lipophilic
100 J.P. Atkinson et al.

Table 8.2 Methods used in transdermal penetration enhancement


Mode of action Reference
Chemical enhancement methods
Skin hydration Increased drug solubility and/or Barry (2001)
disruption of the SC
Chemical penetration enhancers Increased drug partitioning and/or Williams and Barry (2004)
diffusion in the SC
Vesicles Drugs are encapsulated into vesicles Honeywell-Nguyen et al. (2004)
which interact with the skin
Prodrugs Chemical modification of the drug Qandil et al. (2008)
Ion pairs Permeation is increased by neutralizing Ren et al. (2008)
the drug charge with an ion of the
opposite charge
Salt formation Drug is changed into a suitable salt Cheong and Choi (2003)
form to increase its solubility
Supersaturated solutions Thermodynamic activity of the drug Dias et al. (2003)
solution is shifted, thus increasing
penetration rate
Eutectic systems The mixture of drug and another Ehrenstrom and Reiz (1982)
substance lowers the melting point and
increases solubility
Physical enhancement methods
Sonophoresis Creation of microscopic holes for the Tezel and Mitragotri (2003)
transport of drugs
Iontophoresis Cavitational ultrasound generates Costello and Jeske (1995)
shock waves that disrupt the SC lipid
structure
Electroporation Electrically driven transport of charged Denet and Preat (2003), Zewert
drug molecules et al. (1999)
Jet injections Pore formation with short electrical Bremseth and Pass (2001)
pulses
Microneedles High pressure acceleration of drug Gill and Prausnitz (2007)
particles across the SC
Dermabrasion Selective removal of the SC by Andrews et al. (2011)
applying high pressure microparticles
Thermal ablation Short intervals of localized skin Park et al. (2008)
heating that creates micropores
Laser Thermal ablation of SC creating pores Gomez et al. (2008)
Waves (radiofrequency, Disruption of the structure of SC Levin et al. (2005), Lee et al.
photomechanical, microwaves, (1999), Moghimi et al. (2010),
photoacoustic) Sa et al. (2013)
Magnetophoresis Magnetic field is driving drug Benson and Watkinson 2012
movement across SC and alters the SC
structure
Combination of techniques
Chemical enhancers and microneedles, Mutalik et al. (2009), Mitragotri
sonophoresis and electroporation et al. (2000)
Iontophoresis and other physical Hikima et al. (2009), Banga
methods (electroporation, sonophoresis et al. (1999)
or microneedles)
Sonophoresis and other physical Mitragotri et al. (2000)
methods
Electroporation and microneedles Yan et al. (2010)
8 Targets in Dermal and Transdermal Delivery and Classification of Penetration Enhancement Methods 101

Table 8.2 (continued)


Mode of action Reference
Iontophoresis and chemical penetration Wang et al. (2005)
enhancers
Other methods
Moxibustion Increase in skin temperature and skin Cao et al. (2011)
permeation
Submicron injectors Submicron injection system isolated Shaoul et al. (2012)
from sea anemone accelerates the drug
across the SC
Mechanical methods (tape stripping, Different modes of action: removal of Rouse et al. (2007), Abdulmajed
skin flexing, skin stretching, massage) SC layer or reversible formation of and Heard (2008), Benson and
micropathways Watkinson (2012)

drugs, such as ketoprofen (Wu et al. 2001), ibu- theory to categorize chemical penetration enhanc-
profen (Brain et al. 2006), estradiol (Monti et al. ers and to describe the mechanism by which they
2002), tamoxifen (El-Kattan et al. 2001), zidovu- affect skin permeability. According to this theory,
dine (Narishetty and Panchagnula 2004), hydro- enhancers act by one or more of the three modes
cortisone (El-Kattan et al. 2000) and hydrophilic of action: (1) disruption of the intercellular
drugs, e.g. propranolol hydrochloride (Zhao and bilayer lipid structure (lipid modification), (2)
Singh 1999), bupranolol (Babu and Pandit 2005), interaction with the intracellular proteins of the
nicardipine hydrochloride (Krishnaiah et al. SC (protein modification) and (3) improvement
2002, 2003) and others. Azone and its analogues of partitioning of a drug, coenhancer or cosolvent
have been used to enhance a wide range of drugs, into the SC (partitioning promotion).
too (Afouna et al. 2003; Jampilek and Brychtova The aforementioned mechanisms of action of
2012). Oleic acid is also widely studied and is enhancers are direct effects of enhancers on the
one of the leading penetration enhancers used for skin. Chemical enhancers can also act indirectly
transdermal applications (Prausnitz et al. 2004). by modifying the formulation. These mecha-
The limitations of using chemical enhancers nisms include modification of thermodynamic
are that they are not suitable for enhancing the activity of the vehicle, “drag effect” where the
skin penetration of high Mw drugs and that they solvent permeating the skin carries the permeant
often irritate the skin when used at concentra- with it and solubilizing the permeant in the donor
tions necessary for achieving useful levels of (Williams and Barry 2004). For more details see
penetration enhancement (i.e. they have low effi- Vol. 3 describing a vast range of different chemi-
cacy at low doses) (Prausnitz et al. 2004). In cal penetration enhancers.
attempts to solve these problems, researchers Vesicles are colloidal particles, made of water
have tried synthesizing novel chemical penetra- and amphiphilic molecules. The latter form one
tion enhancers (Akimoto and Nagase 2003), with or more bimolecular layers enclosing an equal
optimal enhancer features such as laurocapram number of aqueous compartments. Vesicles can
(Azone), which safely achieves therapeutic trans- encapsulate hydrophilic drugs within the aqueous
port enhancement and its analogues (Jampilek regions and lipophilic molecules within the lipid
and Brychtova 2012), or using two or more pen- bilayers (Bangham et al. 1965; Williams 2003).
etration enhancers together, because of their syn- There is a large body of research that use different
ergistic effect in augmenting the penetration of types of vesicles for dermal and transdermal drug
drugs into/through skin (Furuishi et al. 2010). delivery (see Volume 2): liposomes, transfero-
Barry and co-workers (Barry 1991; Goodman somes, invasomes, ethosomes, niosomes, veso-
and Barry 1988; Williams and Barry 1991) somes, etc. (Schreier and Bouwstra 1994; Touitou
devised the lipid-protein-partitioning (LPP) et al. 2000; Cevc et al. 2008; Dragicevic-Curic
102 J.P. Atkinson et al.

et al. 2008, 2009; El Maghraby et al. 2009). The increased by the creation of a transient, metasta-
results obtained from these studies are still not ble or supersaturated state, whereby the drug’s
consistent and further investigations are needed thermodynamic activity is increased above unity
to fully understand the nature of vesicle transport (Leichtnam et al. 2006). This approach has the
into/across the skin. advantage of providing improvement in perme-
Salt formation is a drug manipulation process ation (proportional to the degree of saturation
where the drug compound is changed into a suit- (DS)) without inducing skin irritation and it is an
able salt form (Cheong and Choi 2003) with a inexpensive enhancement method. The limitation
higher solubility and therefore increased perme- of the supersaturation approach is its inherent
ation through the skin. problem of stabilization, and the need to find a
In ion pair strategy charged drug molecules way to maintain the metastable state for a period
penetrate the SC more easily, because the charge sufficiently long so that an impact on drug trans-
on the drug is neutralized by the molecule with port is apparent (Leichtnam et al. 2007). This
the opposite charge, i.e. they form an ion pair (see period is frequently so short that no impact on
for details Vol. 1, Chapter 13). In the epidermis transport is observed; however, there are exam-
the ion pairs dissociate and the drug then diffuses ples of modest to significant effect, like modest
further (Megwa et al. 2000; Ren et al. 2008). increase of transdermal delivery of testosterone
Eutectic systems are a form of penetration (Leichtnam et al. 2006) or significant transdermal
enhancement method that uses eutectic mixtures delivery of ketotifen (Inoue and Sugibayashi
which are drug formulations that combine two 2012). For more details see Vol. 1, Chapter 11).
substances in an adequate ratio, so that the mix- Hydration (see for details Vol. 3, Chapter 1) of
ture of substances has a lower melting point than the SC can enhance the permeation of a large
each substance alone (see for details Vol. 1, number of drugs used in transdermal delivery,
Chapter 12). The lower melting point of a drug is both hydrophilic and hydrophobic (Benson
a parameter that determines the solubility of the 2005). The mechanisms by which SC hydration
drug (see Eq. 8.1) and therefore influences the increases drug penetration could be by expanding
skin penetration. An example of an eutectic mix- the solubility of the drug and/or by disrupting the
ture for penetration enhancement is the EMLA® structure of the SC due to swelling. The evidence
cream (AstraZeneca), being an eutectic mixture for these mechanisms is not conclusive. In addi-
of lignocaine and prilocaine (1:1) used as a topi- tion to this, hydration does not consistently
cal local anaesthetic, which significantly reduced enhance the penetration of drug molecules and
pain associated with venous cannulation in chil- extended occlusion could trigger skin injuries
dren compared to placebo (Ehrenström-Reiz and (Bucks and Maibach 1999).
Reiz 1982). It was also shown that terpenes form Prodrugs (see for details Vol. 1, Chapter 10) are
binary eutectic mixtures with ibuprofen and that chemically modified drugs that can cross the skin
the resultant melting point depression of the barrier more easily than the original drug (Sloan
delivery system is correlated with a significant et al. 2006). Once the prodrug crosses the SC, an
increase in transdermal permeation (Stott et al. enzymatic and/or chemical transformation will
1998). Further, the itraconazole-phenol eutectic release the active parent drug, which can then exert
formulation enabled, despite the high molecular the desired pharmacological effect. The goal when
weight and hydrophobicity of itraconazole, the designing a transdermal prodrug is to alter the
drug to permeate the skin (Park et al. 2012). physicochemical properties of the drug in such
The use of supersaturated solutions for a way as to increase their lipid and aqueous solu-
enhanced skin delivery of drugs is based on the bility and therefore facilitate the transfer of the
fact that the maximum, passive flux of a drug drug across the skin. Challenges of the prodrug
across the skin is achieved when it is present in approach are an increase in size of the modi-
the applied formulation at its saturation concen- fied drug and gaining Food and Drug administra-
tration, while the drug delivery can further be tion (FDA) approval. In addition to this, many
8 Targets in Dermal and Transdermal Delivery and Classification of Penetration Enhancement Methods 103

transdermal prodrug reports still use penetration molecules by electroporation (Blagus et al. 2013;
enhancement techniques (Juluri et al. 2013; Liu Denet and Preat 2003; Zewert et al. 1999),
et al. 2011; Milewski et al. 2010) showing that this but they are at the moment confined to animal
approach is not completely straightforward. models and in vitro studies.
Sonophoresis (Volume 4, Part 1) is the method
which uses ultrasound to aid topical and transder-
8.5.2 Physical Methods mal drug delivery at high (MHz) and low (kHz)
for Penetration Enhancement frequencies (respectively). Its mode of action is
via disturbing the lipid structure of the SC. The
Iontophoresis (see for details Volume 4, Part 2) is mechanisms associated with high- and low-
an electrically assisted delivery to administer frequency sonophoresis are different; especially,
therapeutic amounts of the drug across the skin, the location of cavitation and the extent to which
which enables a significant increase in drug each process can increase skin permeability are
transport across the skin compared to passive quite dissimilar (Polat et al. 2011). The major
drug permeation (even 184-fold; Kalaria et al. effect of low-frequency ultrasound is cavitation,
2013). Iontophoresis helps both charged and the formation and collapse of air/gas bubbles in
uncharged drug molecules to migrate across the the liquid medium at the skin surface (Tezel and
skin (Costello and Jeske 1995). The penetration Mitragotri 2003). These bubbles oscillate and
enhancing properties of this method are in the collapse forming shock waves which induce tran-
electric driving force and not in changing skin sient structural changes in the nearby tissue, lipid
permeability. A drug-filled electrode is placed on bilayers of the SC. Cavitational ultrasound can
the skin and a low-voltage current is applied. The markedly increase drug flux across the skin
mechanisms that move charged and uncharged (Mitragotri et al. 2000) and is non-invasive.
molecules are electrophoresis and electro- Microneedles are micron-sized needles which
osmosis, respectively (Banga 1998). An advan- can create channels in the skin that penetrate the
tage of this method is that the drug delivery can SC, but do not stimulate the nerves in deeper tis-
be controlled and regulated and application is sues. In this way drug delivery is increased, pain
relatively painless. Limitations of iontophoresis is avoided and skin invasion is minimal.
are its relatively high cost, narrow/fixed/restricted Microneedles can be solid or hollow and can be
drug delivery rates determined by the maximum used in different ways: piercing of the skin fol-
current applied (Prausnitz and Langer 2008) and lowed by drug-loaded patch; inserting solid
molecular size restriction (up to 10–15 kDa) microneedles coated with the drug; the drug can
(Kalluri and Banga 2011). Iontophoresis is often be encapsulated in a biodegradable microneedle;
used synergistically with other penetration or infusion of drug formulation via hollow
enhancers: chemical penetration enhancers microneedles into the tissue. Small drugs as well
(Wang et al. 2005), ultrasound (Mitragotri et al. as high Mw drugs, such as peptides, proteins and
2000) and electroporation (Banga et al. 1999; oligonucleotides, can be transported with this
Alexander et al. 2012). See Volume 4, Part 2 for a technology (Gill and Prausnitz 2007; Liu et al.
detailed explanation of iontophoresis. 2013). See Volume 4, Chapters 21, 22, and 23 for
Electroporation (see Volume 4, Part 3) is further reading.
another electrically assisted penetration enhance- Laser ablation applies laser beams to ther-
ment method where short pulses of high voltage mally erode the SC making micropathways in the
current are applied to the skin. Skin becomes epidermis (Gomez et al. 2008; Paudel et al.
temporarily permeabilized (the structure of the 2010).
SC is disrupted) which facilitates the transport of During thermal ablation skin permeability can
drugs mainly by diffusion and electrophoresis be increased when the skin is heated to hundreds of
(Denet et al. 2004). Studies show effective degrees for a very short time, to avoid damage
penetration enhancement of both small and large to the surrounding tissues (Park et al. 2008).
104 J.P. Atkinson et al.

The authors suggest that skin changes its perme- electroporation and iontophoresis (Banga et al.
ability upon heating because of the lipid and ker- 1999) and microneedle and iontophoresis (Chen
atin disruption in the SC, resulting in the removal et al. 2009). For a detailed explanation see
of SC and formation of microchannels through Volume 4, Chapters 23, 24, and 25.
which a wide range of drug molecules can pass
into the deeper layers of the skin. See Volume 4,
Chapter 17 for further reading on ablation Conclusion
methods. In the last few decades, the research focus of
Radiofrequency waves can also cause ablation transdermal drug delivery has been on improv-
of the SC. Again, microchannels are formed as a ing skin permeability through the develop-
result of localized heating (Sintov et al. 2003; ment of many new methods. Chemical
Levin et al. 2005). See Volume 4, Chapter 10 for enhancement methods, physical enhancement
more details. methods and combination of methods have all
Microdermabrasion often used in dermatol- contributed to the transdermal industry to a
ogy and cosmetic treatments can also serve as a varied degree. A further aim of transdermal
method for the enhancement of drug penetration systems is to extend the list of products on the
into the skin. Removal of the SC is achieved by market, both in terms of diversity of products
applying a stream of small crystals. Andrews and range of indications (diseases) treated.
et al. (2011) showed that microdermabrasion can One way forward would be including more
increase the subcutaneous delivery of insulin in macromolecular drug formulations, and the
diabetic rats more if the epidermis is removed in recent method developments are promising to
addition to the removal of SC. See Volume 4, push these boundaries.
Chapter 17.
Jet injections are needle-free injections where
liquid droplets or solid particles containing a References
drug are directed towards the skin in a pressur-
ized manner. This high-velocity penetration of Abdulmajed K, Heard CM (2008) Topical delivery of reti-
particles into or across the skin can be relevant in nyl ascorbate. 3. Influence of follicle sealing and skin
vaccine administration (Mohammed et al. 2010; stretching. Skin Pharmacol Physiol 21(1):46–49
Abraham MH, Chanda HS, Mitchell RC (1995) The fac-
Kim and Prausnitz 2011). Insulin can be deliv- tors that influence skin penetration of solutes. J Pharm
ered clinically by jet injection (Engwerda et al. Pharmacol 47:8–16
2011, 2013). See Volume 4, Chapter 15 for more Afouna MI, Fincher TK, Zaghloul AAA, Reddy IK (2003)
details. Effect of Azone upon the in vivo antiviral efficacy of
cidofovir or acyclovir topical formulations in treat-
Magnetophoresis (Volume 4, Chapter 13) is a ment/prevention of cutaneous HSV-1 infections and
method that uses static magnetic fields to enhance its correlation with skin target site free drug concentra-
drug penetration (Murthy et al. 2010). Pulsatile tion in hairless mice. Int J Pharm 253:159–168
electromagnetic fields were used to enhance the Agarwal R, Katare OP, Vyas SP (2000) The piloseba-
ceous unit: a pivotal route for topical drug delivery.
penetration of naltrexone in human skin in vitro Methods Find Exp Clin Pharmacol 22(2):129–133
in a process termed dermaportation (Krishnan Ahad A, Aqil M, Kohli K, Chaudhary H, Sultana Y,
et al. 2010; Benson and Watkinson 2012). Mujeeb M et al (2009) Chemical penetration enhanc-
Dermaportation offers also a potential new deliv- ers: a patent review. Expert Opin Ther Pat
19(7):969–988
ery method for skin delivery of peptides for a Akimoto T, Nagase Y (2003) Novel transdermal drug
range of dermatological and cosmetic applica- penetration enhancer: synthesis and enhancing effect
tions (Namjoshi et al. 2008). of alkyldisiloxane compounds containing glucopy-
The above-mentioned physical enhancement ranosyl group. J Control Release 88(2):243–252
Alexander A, Dwivedi S, Ajazuddin, Giri TK, Saraf S,
methods can be used in combination; examples Saraf S, Tripathi DK (2012) Approaches for breaking
are ultrasound and iontophoresis (Le et al. 2000), the barriers of drug permeation through transdermal
electroporation and ultrasound (Kost et al. 1996), drug delivery. J Control Release 164(1):26–40
8 Targets in Dermal and Transdermal Delivery and Classification of Penetration Enhancement Methods 105

Andrews S, Lee JW, Choi S-O, Prausnitz MR (2011) mechanism, methodology, drug delivery, 2nd edn.
Transdermal insulin delivery using microdermabra- Marcel Dekker, New York, pp 77–93
sion. Pharm Res 28(9):2110–2118 Bugaj A, Juzeniene A, Juzenas P, Iani V, Ma LW, Moan J
Andrews SN, Jeong E, Prausnitz MR (2012) Transdermal (2006) The effect of skin permeation enhancers on the
delivery of molecules is limited by full epidermis, not formation of porphyrins in mouse skin during topical
just stratum corneum. Pharm Res 30(4):1099–109 application of the methyl ester of 5-aminolevulinic
Babu RJ, Pandit JK (2005) Effect of penetration enhanc- acid. J Photochem Photobiol B 83(2):94–97
ers on the transdermal delivery of bupranolol through Buyuktimkin N, Buyuktimkin S, Rytting JH (1997)
rat skin. Drug Deliv 12(3):165–169 Chemical means of transdermal drug permeation
Banga AK (1998) Electrically assisted transdermal and enhancement. In: Ghosh TK, Pfister WR, Yum S (eds)
topical drug delivery. Taylor & Francis Group, London Transdermal and topical drug delivery systems,
Banga AK, Bose S, Ghosh TK (1999) Iontophoresis and Informa health care, London, pp 357–447
electroporation: comparisons and contrasts. Int J Cancel LM, Tarbell JM, Ben-Jebria A (2004) Fluorescein
Pharm 179(1):1–19 permeability and electrical resistance of human skin
Bangham AD, Standish MM, Watkins JC (1965) Diffusion during low frequency ultrasound application. J Pharm
of univalent ions across the lamellae of swollen phos- Pharmacol 56(9):1109–1118
pholipids. J Mol Biol 13:238–252 Cao D, Tazawa Y, Ishii H, Todo H, Sugibayashi K (2011)
Barry BW (1983) Dermatological formulations: percuta- Pretreatment effects of moxibustion on the skin per-
neous absorption. Marcel Dekker, New York meation and skin and muscle concentrations of salicy-
Barry BW (1991) Lipid–protein-partititioning theory of late in rats. Int J Pharm 407(1–2):105–110
skin penetration enhancement. J Control Release Cevc G, Mazgareanu S, Rother M (2008) Preclinical char-
15:237–248 acterisation of NSAIDs in ultradeformable carriers or
Barry BW (2001) Novel mechanisms and devices to conventional topical gels. Int J Pharm 360:29–39
enable successful transdermal drug delivery. Eur J Chen H, Zhu H, Zheng J, Mou D, Wan J, Zhang J et al
Pharm Sci 14(2):101–114 (2009) Iontophoresis-driven penetration of nanovesi-
Barry BW (2006) Penetration enhancer classification. In: cles through microneedle-induced skin microchannels
Smith EW, Maibach HI (eds) Percutaneous penetra- for enhancing transdermal delivery of insulin.
tion enhancers. CRC Press, Taylor& Francis Group, J Control Release 139(1):63–72
LLC, Boca Raton, pp 3–15 Cheong H-A, Choi H-K (2003) Effect of ethanolamine
Benson HAE (2005) Transdermal drug delivery: penetra- salts and enhancers on the percutaneous absorption of
tion enhancement techniques. Curr Drug Deliv 2(1): piroxicam from a pressure sensitive adhesive matrix.
23–33 Eur J Pharm Sci 18(2):149–153
Benson HAE, Watkinson AC (2012) Transdermal and Ciotti SN, Weiner N (2002) Follicular liposomal delivery
topical drug delivery: principles and practice. Wiley, systems. J Liposome Res 12(1–2):143–148
Hoboken Costello CT, Jeske AH (1995) Iontophoresis: applications
Blagus T, Markelc B, Cemazar M, Kosjek T, Preat V, in transdermal medication delivery. Phys Ther
Miklavcic D, Sersa G (2013) In vivo real time moni- 75(6):554–563
toring system of electroporation mediated control of Denet A-R, Préat V (2003) Transdermal delivery of timo-
transdermal and topical drug delivery. J Control lol by electroporation through human skin. J Control
Release 172(3):862–71, pii: S0168-3659(13)00827-4 Release 88(2):253–262
Bouwstra JA, Honeywell-Nguyen PL, Gooris GS, Ponec Denet A-R, Vanbever R, Préat V (2004) Skin electropora-
M (2003) Structure of the skin barrier and its modula- tion for transdermal and topical delivery. Adv Drug
tion by vesicular formulations. Prog Lipid Res 42: Deliv Rev 56(5):659–674
1–36 Dias MMR, Raghavan SL, Pellett MA, Hadgraft J (2003)
Brain KR, Green DM, Dykes PJ, Marks R, Bola TS The effect of beta-cyclodextrins on the permeation of
(2006) The role of menthol in skin penetration from diclofenac from supersaturated solutions. Int J Pharm
topical formulations of ibuprofen 5% in vivo. Skin 263(1–2):173–181
Pharmacol Physiol 19:17–21 Downing DT, Stewart ME (2000) Epidermal composition.
Bremseth DL, Pass F (2001) Delivery of insulin by jet In: Loden M, Maibach HI (eds) Dry skin and moistur-
injection: recent observations. Diabetes Technol Ther izers, chemistry and function. CRC Press, Boca Raton,
3(2):225–232 pp 13–26
Brychtova K, Jampilek J, Opatrilova R, Raich I, Farsa O, Dragicevic-Curic N, Scheglmann D, Albrecht V, Fahr A
Csollei J (2010) Synthesis, physico-chemical proper- (2008) Temoporfin-loaded invasomes: development,
ties and penetration activity of alkyl-6-(2,5- characterization and in vitro skin penetration studies. J
dioxopyrrolidin- 1-yl)-2-(2-oxopyrrolidin-1-yl) Control Release 127(1):59–69
hexanoates as potential transdermal penetration Dragicevic-Curic N, Scheglmann D, Albrecht V, Fahr
enhancers. Bioorg Med Chem 18(1):73–79 (2009) Development of liposomes containing ethanol
Bucks D, Maibach HI (1999) Occlusion does not uni- for skin delivery of temoporfin: characterization and
formly enhance penetration in vivo. In: Bronaugh RL, in vitro penetration studies. Colloids Surf B
Maibach HI (eds) Percutaneous absorption: Biointerfaces 74(1):114–122
106 J.P. Atkinson et al.

Ehrenström-Reiz GM, Reiz SL (1982) EMLA–a eutectic Hikima T, Ohsumi S, Shirouzu K, Tojo K (2009) Mechanisms
mixture of local anaesthetics for topical anaesthesia. of synergistic skin penetration by sonophoresis and ion-
Acta Anaesthesiol Scand 26(6):596–598 tophoresis. Biol Pharm Bull 32(5):905–909
El Maghraby GM, Williams AC (2009) Vesicular systems Hoffman RM (1998) Topical liposome targeting of dyes,
for delivering conventional small organic molecules melanins, genes, and proteins selectively to hair folli-
and larger macromolecules to and through human cles. J Drug Target 5(2):67–74
skin. Expert Opin Drug Deliv 6(2):149–163 Honeywell-Nguyen PL, Gooris GS, Bouwstra JA (2004)
El-Kattan AF, Asbill CS, Michniak BB (2000) The effect Quantitative assessment of the transport of elastic and
of terpene enhancer lipophilicity on the percutaneous rigid vesicle components and a model drug from these
permeation of hydrocortisone formulated in HPMC vesicle formulations into human skin in vivo. J Invest
gel systems. Int J Pharm 198:179–189 Dermatol 123:902–910
El-Kattan AF, Asbill CS, Kim N, Michniak BB (2001) Ibrahim SA, Li SK (2010) Efficiency of fatty acids as
The effects of terpene enhancers on the percutaneous chemical penetration enhancers: mechanisms and
permeation of drugs with different lipophilicities. Int J structure enhancement relationship. Pharm Res 27(1):
Pharm 215:229–240 115–125
Engwerda EE, Abbink EJ, Tack CJ, de Galan BE (2011) Illel B (1997) Formulation for transfollicular drug
Improved pharmacokinetic and pharmacodynamic administration: some recent advances. Crit Rev Ther
profile of rapid-acting insulin using needle-free jet Drug Carrier Syst 14(3):207–219
injection technology. Diabetes Care 34(8):1804–1808 Illel B, Schaefer H, Wepierre J, Doucet O (1991) Follicles
Engwerda EE, Tack CJ, de Galan BE (2013) Needle-free play an important role in percutaneous absorption.
jet injection of rapid-acting insulin improves early J Pharm Sci 80(5):424–427
postprandial glucose control in patients with diabetes. Inoue K, Sugibayashi K (2012) In vivo enhancement of
Diabetes Care 36:3436–3441 transdermal absorption of ketotifen by supersaturation
Feingold KR, Man MQ, Menon GK, Cho SS, Brown BE, generated by amorphous form of the drug. Eur J Pharm
Elias PM (1990) Cholesterol synthesis is required for Sci 47(1):228–234
cutaneous barrier function in mice. J Clin Invest Jampilek J, Brychtova K (2012) Azone analogues:
86(5):1738–1745 classification, design, and transdermal penetration
Flynn GL, Weiner ND (1991) Topical and transdermal principles. Med Res Rev 32(5):907–947
delivery-provinces of realism. In: Teubner GR, Juluri A, Peddikotla P, Repka MA, Murthy SN (2013)
Teubner A (eds) Dermal and transdermal delivery. Transdermal iontophoretic delivery of propofol: a
Wissenschaftliche Verlagsgesellschaft GmbH, general anaesthetic in the form of its phosphate salt.
Stuttgart, pp 33–64 J Pharm Sci 102(2):500–507
Forslind B (1994) A domain mosaic model of the skin Kalaria DR, Patel P, Merino V, Patravale VB, Kalia YN
barrier. Acta Derm Venereol 74(1):1–6 (2013) Controlled iontophoretic transport of
Frum Y, Eccleston GM, Meidan VM (2008) Factors influ- Huperzine A across skin in vitro and in vivo: effect of
encing hydrocortisone permeation into human hair delivery conditions and comparison of pharmacoki-
follicles: use of the skin sandwich system. Int J Pharm netic models. Mol Pharm 10:4322–4329
358(1–2):144–150 Kalluri H, Banga AK (2011) Transdermal delivery of pro-
Furuishi T, Fukami T, Suzuki T, Takayama K, Tomono K teins. AAPS PharmSciTech 12(1):431–441
(2010) Synergistic effect of isopropyl myristate and Karakatsani M, Dedhiya M, Plakogiannis FM (2010) The
glyceryl monocaprylate on the skin permeation of effect of permeation enhancers on the viscosity and
pentazocine. Biol Pharm Bull 33(2):294–300 the release profile of transdermal hydroxypropyl
Ghyczy M (2002) Chemical composition of liposomes methylcellulose gel formulations containing diltiazem
and its influence on the humidity of normal skin, HCl. Drug Dev Ind Pharm 36(10):1195–1206
chemical aspects of the skin lipid approach. In: Braun- Karande P, Mitragotri S (2009) Enhancement of transder-
Falco O, Korting HC, Maibach HI (eds) Liposome mal drug delivery via synergistic action of chemicals.
dermatics. Springer, Berlin, pp 308–314 Biochim Biophys Acta 1788(11):2362–2373
Gill HS, Prausnitz MR (2007) Coated microneedles for Kasting GB, Smith RL, Anderson BD (1992) Prodrugs for
transdermal delivery. J Control Release 117(2): dermal delivery solubility, molecular size, and func-
227–237 tional group effects. In: Sloan KB (ed) Prodrugs
Gómez C, Costela A, García-Moreno I, Llanes F, Teijón topical and ocular drug delivery. Marcel Dekker,
JM, Blanco D (2008) Laser treatments on skin enhanc- New York, pp 117–161
ing and controlling transdermal delivery of Kim YC, Prausnitz MR (2011) Enabling skin vaccination
5-fluorouracil. Lasers Surg Med 40(1):6–12 using new delivery technologies. Drug Deliv Transl
Goodman M, Barry BW (1988) Action of penetration Res 1(1):7–12
enhancers on human skin as assessed by the perme- Kost J, Pliquett U, Mitragotri S, Yamamoto A, Langer R,
ation of model drugs 5-fluorouracil and estradiol. I. Weaver J (1996) Synergistic effect of electric field and
Infinite dose technique. J Invest Dermatol 91(4): ultrasound on transdermal transport. Pharm Res
323–327 13(4):633–638
Guy RH, Hadgraft J, Bucks DA (1987) Transdermal drug Krishnaiah YS, Satyanarayana V, Karthikeyan RS (2002)
delivery and cutaneous metabolism. Xenobiotica Penetration enhancing effect of menthol on the percu-
17(3):325–343 taneous flux of nicardipine hydrochloride through
8 Targets in Dermal and Transdermal Delivery and Classification of Penetration Enhancement Methods 107

excised rat epidermis from hydroxypropyl cellulose pegylated naltrexone prodrug across microneedle-
gels. Pharm Dev Technol 7:305–315 treated skin. J Control Release 146(1):37–44
Krishnaiah YS, Satyanarayana V, Bhaskar P (2003) Mitragotri S (2000) Synergistic effect of enhancers for
Enhanced percutaneous permeability of nicardipine transdermal drug delivery. Pharm Res 17(11):
hydrochloride by carvone across the rat abdominal 1354–1359
skin. Drug Dev Ind Pharm 29:191–202 Mitragotri S, Farrell J, Tang H, Terahara T, Kost J,
Krishnan G, Edwards J, Chen Y, Benson HAE (2010) Langer R (2000) Determination of threshold energy
Enhanced skin permeation of naltrexone by pulsed dose for ultrasound-induced transdermal drug trans-
electromagnetic fields in human skin in vitro. J Pharm port. J Control Release 63(1–2):41–52
Sci 99(6):2724–2731 Mittal A, Sara UVS, Ali A, Aqil M (2009) Status of fatty
Lauer AC (1999) Percutaneous drug delivery to the hair acids as skin penetration enhancers-a review. Curr
follicle. In: Bronaugh RL, Maibach HI (eds) Drug Deliv 6(3):274–279
Percutaneous absorption drugs-cosmetics- Moghimi HR, Alinaghi A, Erfan M (2010) Investigating
mechanisms-methodology, 3rd edn. Marcel Dekker, the potential of non-thermal microwave as a novel skin
Inc., New York, pp 427–449 penetration enhancement method. Int J Pharm
Le L, Kost J, Mitragotri S (2000) Combined effect of low- 401(1–2):47–50
frequency ultrasound and iontophoresis: applications Mohammed AJ, AlAwaidy S, Bawikar S, Kurup PJ,
for transdermal heparin delivery. Pharm Res Elamir E, Shaban MMA et al (2010) Fractional
17(9):1151–1154 doses of inactivated poliovirus vaccine in Oman. N
Lee S, Kollias N, McAuliffe DJ, Flotte TJ, Doukas AG Engl J Med 362(25):2351–2359
(1999) Topical drug delivery in humans with a sin- Monti D, Chetoni P, Burgalassi S, Najarro M, Saettone
gle photomechanical wave. Pharm Res 16(11): MF, Boldrini E (2002) Effect of different terpene-
1717–1721 containing essential oils on permeation of estradiol
Leichtnam ML, Rolland H, Wüthrich P, Guy RH (2006) through hairless mouse skin. Int J Pharm 237:209–214
Enhancement of transdermal testosterone delivery by Murthy SN, Sammeta SM, Bowers C (2010)
supersaturation. J Pharm Sci 95(11):2373–2379 Magnetophoresis for enhancing transdermal drug
Leichtnam ML, Rolland H, Wüthrich P, Guy RH (2007) delivery: mechanistic studies and patch design.
Impact of antinucleants on transdermal delivery of tes- J Control Release 148(2):197–203
tosterone from a spray. J Pharm Sci 96(1):84–92 Mutalik S, Parekh HS, Davies NM, Udupa N (2009) A
Levin G, Gershonowitz A, Sacks H, Stern M, Sherman A, combined approach of chemical enhancers and sono-
Rudaev S et al (2005) Transdermal delivery of human phoresis for the transdermal delivery of tizanidine
growth hormone through RF-microchannels. Pharm hydrochloride. Drug Deliv 16(2):82–91
Res 22(4):550–555 Namjoshi S, Chen Y, Edwards J, Benson HA (2008)
Li L, Hoffman RM (1997) Topical liposome delivery of Enhanced transdermal delivery of a dipeptide by der-
molecules to hair follicles in mice. J Dermatol Sci maportation. Biopolymers 90(5):655–662
14(2):101–108 Narishetty STK, Panchagnula R (2004) Transdermal
Lindberg M, Forslind B (2000) The skin as a barrier. In: delivery of zidovudine: effect of terpenes and their
Loden M, Maibach HI (eds) Dry skin and moisturiz- mechanism of action. J Control Release 95:
ers, chemistry and function. CRC Press, Boca Raton, 367–379
pp 27–37 Parikh NH, Babar A, Plakogiannis FM (1984) Transdermal
Liu K-S, Sung KC, Al-Suwayeh SA, Ku M-C, Chu C-C, therapeutic systems (Part 1). Pharm Acta Helv 59:
Wang J-J et al (2011) Enhancement of transdermal 290–292
apomorphine delivery with a diester prodrug strategy. Park J-H, Lee J-W, Kim Y-C, Prausnitz MR (2008) The
Eur J Pharm Biopharm 78(3):422–431 effect of heat on skin permeability. Int J Pharm
Liu S, Jin MN, Quan YS, Kamiyama F, Kusamori K, 359(1–2):94–103
Katsumi H, Sakane T, Yamamoto A (2013) Transdermal Park CW, Mansour HM, Oh TO, Kim JY, Ha JM, Lee BJ,
delivery of relatively high molecular weight drugs Chi SC, Rhee YS, Park ES (2012) Phase behavior of
using novel self-dissolving microneedle arrays fabri- itraconazole-phenol mixtures and its pharmaceutical
cated from hyaluronic acid and their characteristics and applications. Int J Pharm 436(1–2):652–658
safety after application to the skin. Eur J Pharm Paudel KS, Milewski M, Swadley CL, Brogden NK,
Biopharm 86(2):267–76, pii: S0939-6411(13)00326-3 Ghosh P, Stinchcomb AL (2010) Challenges and
Loth H (1992) Percutaneous absorption and conventional opportunities in dermal/transdermal delivery. Ther
penetration enhancers. In: Braun-Falco O, Korting Deliv 1(1):109–131
HC, Maibach HI (eds) Liposome dermatics. Springer, Polat BE, Hart D, Langer R, Blankschtein D (2011)
Berlin, pp 3–10 Ultrasound-mediated transdermal drug delivery:
Megwa SA, Cross SE, Whitehouse MW, Benson HA, mechanisms, scope, and emerging trends. J Control
Roberts MS (2000) Effect of ion pairing with alkyl- Release 152(3):330–348
amines on the in-vitro dermal penetration and local Prausnitz MR, Langer R (2008) Transdermal drug deliv-
tissue disposition of salicylates. J Pharm Pharmacol ery. Nat Biotechnol 26(11):1261–1268
52(8):929–940 Prausnitz MR, Mitragotri S, Langer R (2004) Current sta-
Milewski M, Yerramreddy TR, Ghosh P, Crooks PA, tus and future potential of transdermal drug delivery.
Stinchcomb AL (2010) In vitro permeation of a Nat Rev Drug Discov 3(2):115–124
108 J.P. Atkinson et al.

Puglia C, Bonina F (2008) Effect of polyunsaturated fatty microchanneling as a new way for electrically assisted
acids and some conventional penetration enhancers on transdermal delivery of hydrophilic drugs. J Control
transdermal delivery of atenolol. Drug Deliv 15(2): Release 89(2):311–320
107–112 Sivamani RK, Stoeber B, Liepmann D, Maibach HI
Qandil A, Al-Nabulsi S, Al-Taani B, Tashtoush B (2008) (2009) Microneedle penetration and injection past the
Synthesis of piperazinylalkyl ester prodrugs of ketoro- stratum corneum in humans. J Dermatolog Treat
lac and their in vitro evaluation for transdermal deliv- 20(3):156–159
ery. Drug Dev Ind Pharm 34(10):1054–1063 Sloan KB, Wasdo SC, Rautio J (2006) Design for opti-
Ren C, Fang L, Li T, Wang M, Zhao L, He Z (2008) Effect mized topical delivery: prodrugs and a paradigm
of permeation enhancers and organic acids on the skin change. Pharm Res 23(12):2729–2747
permeation of indapamide. Int J Pharm 350(1–2):43–47 Stott PW, Williams AC, Barry BW (1998) Transdermal
Redelmeier T, Kitson N (1999) Dermatological delivery from eutectic systems: enhanced permeation
Applications of Liposomes. In: Janoff AS (Ed.), of a model drug, ibuprofen. J Control Release
Liposomes. Rational Design, Marcell Dekker, New 50(1–3):297–308
York 283–307 Tezel A, Mitragotri S (2003) Interactions of inertial cavi-
Rizwan M, Aqil M, Talegaonkar S, Azeem A, Sultana Y, tation bubbles with stratum corneum lipid bilayers
Ali A (2009) Enhanced transdermal drug delivery during low-frequency sonophoresis. Biophys J 85(6):
techniques: an extensive review of patents. Recent Pat 3502–3512
Drug Deliv Formul 3(2):105–124 Touitou E, Dayan N, Bergelson L, Godin B, Eliaz M
Roberts MS, Pugh WJ, Hadgraft J (1996) Epidermal (2000) Ethosomes-novel vesicular carriers: character-
permeability-penetrant structure relationships. 2. The ization and delivery properties. J Control Release
effect of H-bonding groups in penetrants on their dif- 65:403–418
fusion through the stratum corneum. Int J Pharm Van Kuijk-Meuwissen ME, Mougin L, Junginger HE,
132:23–32 Bouwstra JA (1998) Application of vesicles to rat
Rouse JG, Yang J, Ryman-Rasmussen JP, Barron AR, skin in vivo: a confocal laser scanning microscopy
Monteiro-Riviere NA (2007) Effects of mechanical study. J Control Release 56(1–3):189–196
flexion on the penetration of fullerene amino acid- Wang S, Kara M, Krishnan TR (1998) Transdermal
derivatized peptide nanoparticles through skin. Nano delivery of cyclosporin-a using electroporation.
Lett 7(1):155–160 J Control Release 50(1–3):61–70
Sá GFF, Serpa C, Arnaut LG (2013) Stratum corneum Wang Y, Thakur R, Fan Q, Michniak B (2005) Transdermal
permeabilization with photoacoustic waves generated iontophoresis: combination strategies to improve
by piezophotonic materials. J Control Release transdermal iontophoretic drug delivery. Eur J Pharm
167(3):290–300 Biopharm 60(2):179–191
Salerno C, Carlucci AM, Bregni C (2010) Study of Wilkes GL, Brown IA, Wildnauer RH (1973) The biome-
in vitro drug release and percutaneous absorption of chanical properties of skin. CRC Crit Rev Bioeng
fluconazole from topical dosage forms. AAPS 1(4):453–495
PharmSciTech 11(2):986–993 Williams A (2003) Transdermal and topical drug delivery
Schaefer H (1996) Skin barrier: principles of percutane- from theory to clinical practice. Pharmaceutical Press,
ous absorption. Karger, Basel London
Schätzlein A, Cevc G (1998) Non-uniform cellular packing Williams AC, Barry BW (1991) Terpenes and the lipid-
of the stratum corneum and permeability barrier func- protein-partitioning theory of skin penetration
tion of intact skin: a high-resolution confocal laser scan- enhancement. Pharm Res 8(1):17–24
ning microscopy study using highly deformable vesicles Williams AC, Barry BW (2004) Penetration enhancers.
(Transfersomes). Br J Dermatol 138(4):583–592 Adv Drug Deliv Rev 56(5):603–618
Scheuplein RJ (1967) Mechanism of percutaneous Wu PC, Chang JS, Huang YB, Chai CY, Tsai YH (2001)
absorption. II. Transient diffusion and the relative Evaluation of percutaneous absorption and skin irrita-
importance of various routes of skin penetration. tion of ketoprofen through rat skin: in vitro and in vivo
J Invest Dermatol 48(1):79–88 study. Int J Pharm 222:225–235
Schreier H, Bouwstra J (1994) Liposomes and niosomes Yan K, Todo H, Sugibayashi K (2010) Transdermal drug
as topical drug carriers: dermal and transdermal drug delivery by in-skin electroporation using a micronee-
delivery. J Control Release 30:1–15 dle array. Int J Pharm 397(1–2):77–83
Shaoul E, Ayalon A, Tal Y, Lotan T (2012) Transdermal Zewert TE, Pliquett UF, Vanbever R, Langer R, Weaver
delivery of scopolamine by natural submicron injec- JC (1999) Creation of transdermal pathways for mac-
tors: in-vivo study in pig. PLoS One 7(2):e31922 romolecule transport by skin electroporation and a low
Siddiqui O, Roberts MS, Polack AE (1989) Percutaneous toxicity, pathway-enlarging molecule. Bioelectrochem
absorption of steroids: relative contributions of epider- Bioenerg 49(1):11–20
mal penetration and dermal clearance. J Pharmacokinet Zhao K, Singh J (1999) In vitro percutaneous absorption
Biopharm 17(4):405–424 enhancement of propranolol hydrochloride through
Sintov AC, Krymberk I, Daniel D, Hannan T, Sohn Z, porcine epidermis by terpenes/ethanol. J Control
Levin G (2003) Radiofrequency-driven skin Release 62:359–366
Formulation Effects
in Percutaneous Absorption 9
Rina Kuswahyuning, Jeffrey E. Grice,
Hamid R. Moghimi, and Michael S. Roberts

Contents 9.5 Drug Candidates.......................................... 115


9.1 Introduction.................................................. 110 9.6 Formulation-Based Strategies for 
Optimised Percutaneous Absorption.......... 116
9.2 Skin Structure and Rate-­Determining
9.6.1 Escaping Tendency from Formulation
Pathways....................................................... 110
(Thermodynamic Activity)............................. 117
9.3 Goals of Skin Delivery................................. 112 9.6.2 Chemical Penetration Enhancers................... 118
9.6.3 Synergistic Thermodynamic Activity
9.4 Mathematical Considerations and Chemical Enhancers................................ 120
in Percutaneous Absorption........................ 112 9.6.4 Hydration....................................................... 120
9.4.1 Principles of Percutaneous Absorption.......... 112 9.6.5 Ion Pairs......................................................... 121
9.4.2 Requirements for Optimised Delivery 9.6.6 Complex Coacervation................................... 121
and Targeting: An Alternative Approach....... 113 9.6.7 pH Adjustment............................................... 121
9.4.3 Skin Pharmacokinetics: Drug 9.6.8 Eutectic Mixtures........................................... 122
Concentration in the Target............................ 115
9.7 Importance of Other Formulation
Ingredients on Percutaneous
Absorption.................................................... 122
9.7.1 Solvent and Co-solvent.................................. 123
R. Kuswahyuning 9.7.2 Viscosity Modifiers........................................ 124
Therapeutics Research Centre, School of Pharmacy 9.7.3 Ingredients Used for Stability........................ 125
and Medical Sciences, University of South Australia, 9.7.4 Ingredients Used for 
Adelaide, SA 5000, Australia Customer Acceptance.................................... 125

Department of Pharmaceutics, Faculty of Pharmacy, 9.8 Formulation-Related Issues


University of Gadjah Mada, Yogyakarta, Indonesia for Nanoparticulate Carriers...................... 125
e-mail: rina.kuswahyuning@mymail.unisa.edu.au;
Conclusion............................................................... 129
rinakus@ugm.ac.id
J.E. Grice References................................................................ 129
Therapeutics Research Centre, School of Medicine,
University of Queensland, Princess Alexandra
Hospital, Woolloongabba, QLD 4102, Australia
e-mail: jeff.grice@uq.edu.au
M.S. Roberts (*)
H.R. Moghimi Therapeutics Research Centre, School of Pharmacy
Therapeutics Research Centre, School of Pharmacy and Medical Sciences, University of South Australia,
and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
Adelaide, SA 5000, Australia
School of Medicine, University of Queensland,
School of Pharmacy, Shahid Beheshti University of Princess Alexandra Hospital,
Medical Sciences, Tehran, Iran Woolloongabba, QLD 4102, Australia
e-mail: hrmoghimi@sbmu.ac.ir e-mail: m.roberts@uq.edu.au

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 109


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_9, © Springer-Verlag Berlin Heidelberg 2015
110 R. Kuswahyuning et al.

9.1 Introduction potential mechanisms related to this ­paradigm. In


addition, formulation-based strategies for opti-
Skin is increasingly exploited as a site of applica- mised delivery as well as importance of other
tion for drug delivery. The ultimate goal is to ingredients commonly added in formulation for
deliver the active ingredient to the site of action ensuring physicochemically stable formulations
in therapeutic amounts over the intended dura- and better product acceptance are discussed.
tion. The outermost layer of the skin, the stratum Finally, we give an overview of the current under-
corneum, is generally accepted as the major bar- standing of formulation effects in percutaneous
rier for such delivery (Scheuplein and Blank absorption from a simple formulation followed
1971; Thong et al. 2007). Over the past decades, by more complex drug delivery systems.
various techniques have been investigated and
developed to promote active ingredient perme-
ation across the skin (Benson 2005). This 9.2  kin Structure and Rate-­
S
includes formulation optimisation, as an approach Determining Pathways
to promoting percutaneous absorption (Barry
2001; Benson 2005). An appropriate choice of A diagrammatic representation of the stratum
ingredients and their concentrations are formu- corneum structure, along with the three recog-
lated together to promote penetration, through nised pathways of penetration through the skin, is
solute-ingredient or solute-ingredient-skin inter- shown in Fig. 9.1. Skin acts as a barrier between
actions mechanisms. Formulation optimisation our body and the environment and covers an area
gives us an opportunity to maximise, minimise or of approximately 2 m2 in an adult (Hadgraft
control the skin penetration of a solute using vari- 2001). It consists of three main layers: the epider-
ous ingredients mixtures. This is a complex pro- mis, dermis and hypodermis (subcutaneous tis-
cess, given that ingredients are used not only for sue). These layers are structurally different and
enhancing skin delivery but also for ensuring the contribute to the overall process of skin transport.
physicochemical stability of the product, as well The stratum corneum, the outermost layer of the
as its acceptance. It is very common that “real” epidermis, is a non-living layer, whereas the rest
formulations in the market are complex systems, of the epidermis is viable tissue, called viable
such as microemulsions, liposomes and lipid epidermis, comprised of nucleated cells, the
nanoparticles. The more complex the ingredients keratinocytes. The stratum corneum is generally
used, the more complex the interactions might considered as the major barrier in skin delivery. It
exist. While the rate of skin delivery from a sim- is composed of 15–25 layers of flattened,
ple formulation such as a single or binary mixture ­hexagonal and cornified cells called corneocytes.
can be estimated, its prediction from more com- The stratum corneum layers are united by stra-
plex formulations remains a challenge. tum corneum lipid bilayers assembled into a
In this chapter, we will review formulation “brick and mortar” structure: corneocytes as the
optimisation for skin delivery. Our focus will be “bricks” and lipid bilayers as the “mortar”
on the current understanding of the formulation (Roberts et al. 2002a; Fluhr and Darlenski 2009).
approach to optimise delivery into or through the The viable epidermis is a site for drug binding
skin, known as dermal and transdermal drug and sequestration, drug metabolism and active
delivery, respectively. We begin with a brief over- transport. The dermis is an acellular layer, which
view of skin structure and formulation in skin is rich of collagen bundles. Below the dermis is
delivery. We then discuss formulation design, the subcutaneous tissue connecting the skin to
starting from theoretical considerations in skin deeper body structures such as muscle and bone.
delivery and its application in comparing various It is composed of fat microlobules and collagen.
complex formulations. An alternative approach is The dermis and subcutaneous layers contain
proposed and current techniques to enhance per- blood vessels, lymphatics and nerve fibres, as
cutaneous absorption are discussed based on their well as skin appendages (Jepps et al. 2013).
9  Formulation Effects in Percutaneous Absorption 111

Intercellular
Trancellular penetration
Follicular
penetration
penetration

Corneodesmosome Corneocyte envelope


Intercellular lipid

Fig. 9.1  Stratum corneum structure and main penetration pathways through skin (Adapted from Roberts et al. 2008)

Penetrants may cross the skin using a number p­ harmacokinetic modelling and a skin sandwich
of routes, i.e. intercellular and transcellular stra- technique, respectively, to show that the extent of
tum corneum pathways, and appendageal routes follicular transport at steady state is between
(eccrine sweat glands or hair follicles). Whilst it about 5 and 30 % of the total flux. Grice et al.
was originally proposed that the transcellular (2010) measured the stratum corneum and the
route was the main penetration pathway in skin follicles separately and showed that follicular
(Scheuplein and Blank 1971), it is now generally transport for minoxidil was more important at
thought that most drugs penetrate via the inter- early times. There are a number of conditions
cellular pathway (Elias 1983; Barry 2001; such as folliculitis, acne, where there is a need for
Hadgraft 2001). Recent work by Nitsche et al. direct follicular targeting. Topical follicular tar-
(Nitsche and Kasting 2008) has suggested that geting can avoid systemic toxicity and possibly
the relative transport through the two pathways is reduce the dose and frequency of application.
dependent on the nature of the compound. The value of the follicular pathway for systemic
Compounds have long been shown to be taken up drug delivery is not well established. Several
into the follicles but this uptake and penetration advantages of follicular drug targeting are (1)
is believed to be dominant only at early times bypassing the tortuous pathway of transepider-
(Scheuplein and Blank 1971). This hypothesis mal absorption, (2) decreasing systemic toxicity
has been recently confirmed in human skin and (3) possibly reducing the applied dose and/or
in vivo by Liu et al., who showed that hair folli- frequency of its administration. Recently, there is
cles contributed significantly to percutaneous a considerable interest in using the follicular
absorption of caffeine only at times soon after pathway as a reservoir for localised therapy as
application (Liu et al. 2011). Siddiqui et al. well as a transport pathway for systemic drug
(1989) and Essa et al. (2002) have used delivery (Lademann et al. 2008).
112 R. Kuswahyuning et al.

9.3 Goals of Skin Delivery of solute penetrating across unit area of skin (Q)
to the duration of application (t), solute diffusivity
Drugs are applied to skin for local, regional and (D), effective path length for the diffusion (h), as
systemic delivery. The intention of local delivery well as gradient concentration difference between
is to directly treat cutaneous disorders on the skin immediately below the area of application Csc(o)
surface or within the skin directly beneath the and the inner side of stratum corneum Csc(i):
application site, such as eczema or psoriasis. Any
systemic absorption that may occur is undesirable
Q=
(
D t Csc ( o ) - Csc ( i ) ) (9.1)
(Roberts et al. 2002a). In contrast to local deliv- h
ery, regional delivery requires deposition of the
drug with deeper penetration beneath the site of Steady state flux, Jss, is calculated from Eq. 9.1 as
application. Examples of target sites for regional
delivery are muscles and joints, beneath and
J ss = =
(
dQ D Csc ( o ) - Csc ( i ) ) (9.2)
around the application site. Systemic absorption dt h
from this form of delivery is undesirable. High
concentrations of drug can be localised directly at Steady state in Eq. 9.2 is only achieved when
the target site, thereby reducing systemic drug the lag time of diffusion has been reached and it is
concentrations and possible systemic side effects assumed that (i) the skin barrier, usually the stra-
(Honeywell-Nguyen and Bouwstra 2005). tum corneum, behaves like a pseudo-­homogenous
Systemic delivery requires application of the drug membrane with no appreciable changes in its bar-
to the skin to treat systemic disorders. Here, the rier properties; (ii) depletion of the product applied
drug should diffuse sufficiently across the skin to does not occur and (iii) the system has perfect sink
achieve therapeutic systemic concentrations conditions (i.e., Csc(i) << Csc(o) and Csc(i) can there-
(Roberts et al. 2002a). Ideally, there should not be fore be disregarded). Accordingly, if the solute
any accumulation of the drug within the skin. As stratum corneum-vehicle partition coefficient (K)
an alternative to oral drug delivery, transdermal is the ratio of Csc(o) and the solute concentration in
delivery avoids gastrointestinal absorption– the vehicle Cv, and the permeability coefficient (kp)
related problems and hepatic first-pass metabolism is a product of KD/h, Eq. 9.2 can be written as
and can provide zero-order drug delivery and
DK Cv
therefore avoid variability of plasma levels J ss = = kp C v (9.3)
h
(Honeywell-Nguyen and Bouwstra 2005).
The stratum corneum kp is related to the per-
meability of the available pathway in the stratum
9.4 Mathematical corneum, e.g. intercellular lipid, polar route and
Considerations transcellular route. For most solutes, it seems that
in Percutaneous Absorption penetration is via the intercellular lipid; there-
fore, kp usually refers to stratum corneum lipid
9.4.1 P
 rinciples of Percutaneous permeability (Roberts and Walters 1998).
Absorption Normally, maximum flux, Jmax, is achieved at
the solute solubility in the stratum corneum (Csc sat)
When a chemical is in contact with skin, quantifi- which also corresponds when solute solubility in
cation of the extent of penetrant absorption the vehicle achieves its saturation condition (Cv sat).
through the skin is usually related to Fick’s first Substituting Csc sat into Eq. 9.2 under sink condition
law. As has been mentioned before, the stratum or Cv sat into Eq. 9.3 indicates that Jmax is expressed
corneum is considered as the main barrier to pen- by either kp and Cv sat or by D, h and Csc sat:
etration of most drugs and barrier effects in the
viable epidermis and dermis are considered negli- D (9.4)
J max = kpCv sat = Csc sat
gible. In such cases, Fick’s law relates the amount h
9  Formulation Effects in Percutaneous Absorption 113

Flux and kp values can be determined practi- epidermis and dermis permeability coefficients,
cally through in vitro or in vivo studies. In a­ ddition, respectively. Now, taking into account the overall
a qualitative structure–penetration relationship route available for penetration, then Eq. 9.8 can
approach uses physicochemical properties of the be modified to account for the ­ permeability
solute in terms of octanol-water partition coeffi- coefficient of appendages (kpapp) (Jepps et al.
cient (log P) and molecular weight (MW), to give 2013):
an approximation of kp. Based on published per- -1
meability coefficient data from aqueous vehicles, æ 1 1 1 ö
k skin
= ç sc + ve + d ÷ + kpapp (9.9)
p çk ÷
Potts and Guy (1992) generated a model to predict
è p kp kp ø
kp (cm h-1) :
Log kp = −6.3 + 0.71logP − 0.0061 MW (9.5)

Recognising that the solubility of solutes in 9.4.2 R
 equirements for Optimised
water can also be expressed in terms of solute Delivery and Targeting:
melting point (MP) and Log P, Eq. 9.5 can also An Alternative Approach
be expressed in terms of maximum flux Jmax
(Milewski and Stinchcomb 2012; Pastore et al. We have now described the various transport
2014, in press): processes required to achieve either topical or
systemic delivery. Whatever the intended target
Log J max (µg cm −2 h −1 )
of the delivery, solute penetration through the
=1.6 + log MW - 0.0086 MW stratum corneum is the prerequisite. During the
-0.01( MP - 25) - 0.219 log P (9.6) formulation development process, particularly,

the ability to predict the rate of solute penetra-
In addition to the stratum corneum being a tion through the stratum corneum would be use-
barrier in skin penetration, other skin layers ful to minimise trial and error and shorten the
might also affect the skin penetration process. optimisation process. Additionally, it is also
Equation 9.5 is frequently modified to adjust for highly relevant for assessing potential dermal
the viable epidermal resistance for lipophilic sol- exposure from industrial and environmental
utes (Cleek and Bunge 1993). Transport in the hazards.
dermis occurs by both diffusion and convective Predictive models for percutaneous
transport by the blood and the lymphatics. It has ­penetration based on the relationship of steady
been suggested that convective transport will state flux, solute concentration in the vehicle and
dominate when the compound is highly protein skin permeability (kp) are up to now perhaps the
bound and this explains why many non-steroidal most widely studied. They assume that steady
drugs provide effective local analgesia after topi- state flux is directly proportional to the skin per-
cal application (Dancik et al. 2012; Anissimov meability coefficient and the concentration of the
et al. 2013). When the viable epidermis or subse- penetrant in the vehicle. When the penetrant is at
quent layers act as extra barriers, Eq. 9.3 for maximum solubility in the vehicle, maximum
steady state flux can be modified as follows: flux is achieved, provided the system behaves
ideally, i.e. no solute-vehicle-skin interactions
J ss = kpiCv (9.7)
exist. Under these conditions, flux at a concentra-
tion below saturation in a given vehicle can be
where approximated if the fractional solubility is
known.
1 1 1 1
= + + (9.8) Conditions required for kp-based validity, how-
kpi kpsc kpve kpd
ever, are not always met. A solute may interact with
where kpi is total skin permeability coefficient vehicle to some degree, resulting in an increased or
and kpsc, kve
p , kp represent stratum corneum, viable
d
decreased effective solute concentration in the
114 R. Kuswahyuning et al.

vehicle. When this situation occurs, flux may no Eq. 9.10 measures what actually happens in the
longer be directly related to vehicle solute con- skin rather than what happens in the formula-
centration. Another scenario potentially affecting tion, this equation enables us to compare the
kp-based validity is when either the solute or the abilities of different formulations to enhance
vehicle interacts with the stratum corneum, caus- the skin delivery of solutes. When solutes are
ing alteration in barrier properties. The vehicle saturated in formulations and formulations do
may enter the stratum corneum altering solute not affect the skin, their Jss will be similar. A
solubility in the skin. Alternatively, the vehicle reduced flux will likely be caused by a lower
may influence stratum corneum lipid packing solute solubility in the formulation (i.e. lower
which in turn may alter solute diffusivity in the thermodynamic activity in the vehicle) and a
stratum corneum. higher value will be caused by formulation-skin
Another limitation for kp-based prediction interactions, to increase either Tr or Rm.
may be faced during formulation development Increased Tr can be achieved by increasing dif-
or optimisation processes. Given the rapid fusion in the membrane, whereas Rm can be
development in the variety of the potential for- increased by promoting solute partitioning
mulation types in topical and transdermal deliv- from the formulation into the stratum
ery, a formulator may be eager to compare corneum.
directly which type of formulation best suits the Given that stratum corneum penetration is the
properties of a given penetrant. The use of kp- first requirement to exert a local, regional or a
based prediction models may be difficult in real systemic effect, an increase in Jss is desirable.
formulations where there are a number of sol- However, the relative merit of enhancing either Tr
vents, as kp is usually derived from aqueous or Rm depends on the aim of delivery. Topical
solutions. delivery is best achieved by a localised solute
We have recently proposed an alternative way concentration at the target skin layer, with mini-
to overcome the limitations of a kp-based mal systemic delivery to prevent unwanted sys-
approach in predicting the extent of penetration temic side effects. This is achieved by maximising
for various different formulations (Wiechers solute retention in the target layer (Rmtarget) and
et al. 2012). It is derived from a more fundamen- the rate of solute transfer to the target layer
tal definition where the steady state flux through (Trtarget). For example, when the stratum corneum
the SC is directly related to diffusivity (D), path is the ­target layer, such as in sunscreens, insect
length of diffusion (h) and solute concentration in repellents and cosmetic preparations, it would be
the stratum corneum (Csc). The solute concentra- desirable to have formulations that maximise sol-
tion in the stratum corneum can be measured ute retention in the stratum corneum ( Rmsc ) ,
when the amount of solute retained in stratum ­preferably without an increase in Trsc. Similar
corneum and the stratum corneum volume are requirements can be then adopted for the other
known. A tape stripping technique can be used to skin layers. For example, dermal targeting with
measure the amount of solute retained in the anaesthetic formulations requires high solute
­stratum corneum. However, as it may be difficult transfer that can facilitate solute diffusion to the
to estimate the solute concentration in the strips, dermal layer (i. e. high Trsc, Trve and Trdermis) and
we have proposed the use of Eq. 9.10, which is a high solute retention in the epidermal/dermal lay-
variation of Eq. 9.2: ers to localise solute accumulation Rmdermis.
Systemic delivery is best achieved by formula-
J ss = Tr Rm (9.10)
tions that can facilitate solute diffusion across the
where Tr is the rate constant of solute transfer in skin to achieve systemic therapeutic solute con-
the stratum corneum, directly related to D/h, centrations. In other words, this requires high
and Rm is the amount of solute that is retained in Trskin with optimized Rmskin (Rmskin = Rmsc + Rmve + 
the stratum corneum, analogous to Csc. As Rmdermis).
9  Formulation Effects in Percutaneous Absorption 115

Table 9.1  Effective plasma concentration, clearances, required steady state flux and physicochemical data used to
predict required solute transdermal flux for passive topical delivery systems (Pastore et al. 2014, in press)
Plasma level Cl systemic Estimated
Solute (ng/mL) (L/h/70 kg) Jss required (μg/h) t1/2 (h) MW MP (°C) Log P
Clonidine 0.2–2.0 15 3–30 8–13 230 130 2.7
Estradiol 0.04–0.06 600–800 24–48 ~1 272 173–179 4.2
Fentanyl 1–3 27–75 27–225 3–12 337 83–84 3.9
Nicotine 5–30 77 385–2,310 2 162 −79 1.1
Nitroglycerin 0.02–0.4 216–3,270 4.32–1,308 0.03–0.05 227 13 1
Scopolamine >0.05 65–121 3.25–6.05 1–5 303 55 0.8
Testosterone 3–10.5 41 123–430.5 0.17–1.7 288 155 3.6

9.4.3 S
 kin Pharmacokinetics: Drug based on drug physicochemical properties, as
Concentration in the Target shown in Table 9.1.

In relation to the goal of skin delivery, the


­concentration at the target site could be mathe- 9.5 Drug Candidates
matically modelled based on the related transport
process. In the viable epidermis, other processes Despite the advantages of using skin as a drug
that may affect solute penetration and distribu- application site, to the present only a few drugs are
tion within the skin include epidermal metabo- marketed commercially in transdermal delivery
lism, solute binding and sequestration. These systems. Some applications include motion sick-
variables may also affect solute availability (F) ness, hypertension, angina, anaesthesia, hormone
for solute penetration into the dermis and beyond. replacement therapy, contraception and smoking
The steady state solute concentration at a site Css cessation therapy (Finnin and Morgan 1999; Thong
is approximated by accounting for input rate and et al. 2007). This limited application is due mainly
local clearance (Anissimov et al. 2013; Jepps to the limited permeability of the stratum corneum.
et al. 2013). In general, the concentration at any Improved understanding of percutaneous transport
skin layer target site is determined by the steady and skin structure has led us to conclude that the
state flux input to the target site ( J sstarget ) , the bio- best candidate drugs for passive skin delivery must
availability (F), the area of application (A) and have certain physicochemical properties: low
the local clearance (Cllocal): molecular weight (<500 Da), moderate lipophilic-
ity, melting point <250 °C and good potency (daily
J sstarget F A kpCv F A
Cskin = = (9.11) systemic dose ≤20 mg) (Finnin and Morgan 1999).
Cllocal Cllocal
Studies on Quantitative Structure–Penetration
Relationships (QSPR) (Anderson and Raykar
For systemic delivery, the concentration of 1989; Magnusson et al. 2004) have shown that sol-
solute in plasma after topical application, Cssp, ute size is important for facilitating the diffusion
can be approximated from Eq. 9.11, in which process while adequate lipophilicity is required to
kpskin is defined as in Eq. 9.9: provide sufficient solubility in the stratum cor-
neum. Maximum flux was shown to have an inverse
skin relationship with molecular weight (Magnusson
J ssskin F A kp Cv FA
Cssp = = (9.12) et al. 2004) and in aqueous vehicles reached a max-
Clsystemic Clsystemic
imum when 2.7 < log P <3.1 (Zhang et al. 2009).
However, it does not necessarily follow that all
Equation 9.12 can be used in finding the best lipophilic solutes with log P > 3 are poor candi-
drug candidate for achieving the required Jsstarget dates for skin delivery, because drug potency must
116 R. Kuswahyuning et al.

a b

Fig. 9.2  Efficacy concept for antimycotics, whereby effi- terms of its minimum inhibitory concentration (MIC, b)
cacy (c) is defined as the ratio of its maximum penetration (Mertin and Lippold 1997)
flux (Jmax, a) from a formulation to its potency defined in

also be considered. Lippold (Wenkers and Lippold 9.6 Formulation-Based


2000) tested the correlation between maximum Strategies for Optimised
flux and potency of a range of anti-inflammatory Percutaneous Absorption
drugs with varying lipophilicities and showed
diclofenac, with a log P > 4, to have the highest Formulation encompasses multiple processes
efficacy coefficient ratio (the ratio of maximum that lead eventually to a successful product in the
flux and drug potency dose). Even though the max- market. The demonstration of product efficacy
imum flux of diclofenac was lower than drugs with and good customer acceptance would perhaps be
log P in the range 2.7–3.1, its potency was greater, key points for achieving such a goal. While in a
since the a­nti-­inflammatory effect of a drug also broader sense, dosage form design, composition
depends on lipophilicity. A similar concept was design, packaging design and industrial scaling-
also recognised for anti-mycotics as shown in ­up are also essential parts of the formulation pro-
Fig. 9.2 (Mertin and Lippold 1997). It is observed cess, we will limit our discussion in this chapter
that nystatin is an inappropriate compound because to the choice of potential techniques and ingredi-
it has a low Jmax due to its large size and at the same ents that can be adopted to optimise dermal and
time is not very potent. Griseofulvin also shows transdermal drug delivery.
poor efficacy due to both poor penetration and a Based on the theoretical requirements
higher MIC. The most ideal compound for efficacy described in previous sections, improved skin
appears to be amorolfine, which has a lower Jmax delivery of a solute can be achieved by increasing
than naftifine but is much more potent (lower mini- its concentration in the stratum corneum and its
mum inhibitory concentration or MIC). rate of transport in this barrier. Table 9.2
9  Formulation Effects in Percutaneous Absorption 117

Table 9.2  Formulation-based enhancement strategies and their possible effects on solute diffusivity and retention in
stratum corneum
Approaches Mechanisms Potential effects
Thermodynamic Supersaturation Increase partitioning; may increase
activity Rm
Formulating for Efficacy Increase partitioning and/or Rm;
(potentially) increase D
Chemical penetration Stratum corneum lipid alteration, e.g. fluidised lipid Increase D
enhancer bilayer, lipid extraction, polar head group alteration,
hydrophobic lipid
Stratum corneum desmosomes and protein alterations Increase D
Corneocytes alteration Increase D
Shift stratum corneum solubility closer to that of Increase Rm and/or partitioning
permeant
Increase solute solubility in the stratum corneum Increase partitioning and/or Rm
because of solvent-drag mechanism
Ion pairs and Increase lipophilicity Increase partitioning
complex coacervates
Eutectic mixture Decrease melting point Increase Rm

s­ummarises formulation-based passive current s­ olubilities. As thermodynamic activity is based


approaches for percutaneous absorption enhance- on the extent of solute solubility in the applied
ment and their possible effect on Rm and/or Tr. ­vehicle/formulation, the degree of solubility or
Other aspects considered later include thermody- saturation provides a useful way of comparing
namic activity, chemical enhancers, hydration, solute thermodynamic activity among different
ion pairs, complex coacervation, pH adjustment vehicles/formulations. Percutaneous absorption
and eutectic mixtures. is directly related to the thermodynamic activity
unless the skin is affected by s­ olute– or vehicle–
skin interactions (Twist and Zatz 1986).
9.6.1 Escaping Tendency The thermodynamic activity concept has been
from Formulation used to develop formulations for many years
(Thermodynamic Activity) (Poulsen et al. 1968; Ostrenga et al. 1971;
Woodford and Barry 1982; Ishii et al. 2010). For
The thermodynamic activity of a solute can be example, Poulsen et al. (1968) optimised the
estimated from the ability of the solute to escape composition of propylene glycol-water to modu-
from the formulation (i.e. its vapour pressure). late the release of 0.025 % fluocinolone and its
Thermodynamic activity also has been referred to ester. It was shown that release of both of the
as the effective solute concentration in the applied drugs was maximal at the least amount of propyl-
formulation. Increasing the solute concentration ene glycol needed to completely dissolve the
is a common way to increase thermodynamic drug, i.e. 20 and 75 % of propylene glycol in
activity. When saturation conditions are reached, water for fluocinolone and fluocinolone ester,
the condition in which the solute has maximum respectively. A high proportion of propylene gly-
thermodynamic activity, the skin delivery rate col in water beyond the required least amount to
will be the highest. This fundamental concept completely dissolve steroids increased the solu-
was clearly shown by Twist and Zatz (1986) with bility of both steroids and thus decreased the par-
their flux comparison of saturated paraben from tition coefficient, leading to lower thermodynamic
various solvents through a silicone membrane. activity. A dependency of drug release on the
The fluxes were similar, despite the concentration vehicle and the nature of the drug could be
difference of paraben due to the variation of ­demonstrated. Similar results have been observed
118 R. Kuswahyuning et al.

by Ostrenga et al. (1971), who found that a rela- therapeutic dose. This second solvent or emol-
tionship could be built between in vitro and lient has a large RPI. Wiechers recently published
in vivo data and emphasised the importance of a practical application of the principles of FFE
thermodynamic activity considerations in the based on maximum flux considerations (Wiechers
development of formulations. et al. 2012).
Another example of the role of thermodynamic
activity as a determinant of skin penetration flux 9.6.1.2 Supersaturation
was reported by Barry et al. (1985a, b). In their Supersaturation is a condition in which an active
work, the thermodynamic activity of benzyl alco- chemical has greater thermodynamic activity
hol, a volatile hydrogen-bonding model penetrant, than that occurring under saturation conditions.
was measured using headspace chromatography A supersaturated formulation can be designed by
from various binary solvents. The vapour flux different methods, including co-solvent mixing.
across human abdominal skin was directly related The major drawback for this system is its insta-
to its thermodynamic activity (Barry et al. 1985b). bility. Crystallisation may occur during storage,
However, when liquid flux was tested, the rela- leading to a change in therapeutic performance of
tionship was less consistent. It was found that the formulation. An anti-nucleating agent such as
some liquid vehicles, such as toluene, damaged a polymer is often added to reduce crystallisa-
the skin (Barry et al. 1985a). This result indicates tion. Supersaturated conditions can also some-
that if skin is affected by vehicle-skin interactions, times be formed after application of the
thermodynamic activity and flux relationship may formulation to the skin surface. Volatile compo-
not be linear. Expanding on the thermodynamic nents such as ethanol or water may evaporate,
activity approach, concepts such as Formulating leading to supersaturated conditions in the
for Efficacy and Supersaturation have been devel- remaining formulation (Pellett et al. 1994, 1997).
oped and applied. As seen in in vitro (Oliveira et al. 2012) and
in vivo experiments (Stinchcomb et al. 1999),
9.6.1.1 Formulating for Efficacy (FFE) there was an initial rapid solute uptake when
This concept was first introduced by the late ­volatile solvent evaporated from the formulation,
Johann Wiechers in 2004 (2004). FFE was devel- leading to increasing solute concentration and
oped to combine two contradictory properties eventual supersaturation. However, after com-
required of by a formulation, namely, (i) the for- plete evaporation, penetration was markedly
mulation has maximum solubility for the active decreased as the solute was deposited as a cryst-
chemical to accommodate the required dose and allised film on the skin surface.
(ii) the formulation should have, at the same time,
minimum active chemical solubility so as to pro-
mote the active chemical leaving the formulation 9.6.2 Chemical Penetration
and partitioning into the stratum corneum. Enhancers
A Relative Polarity Index (RPI) is used to mea-
sure the polarity difference between the active Chemical penetration enhancers are purposely
ingredient and the vehicle. To achieve the first incorporated in topical or transdermal formula-
goal, a solvent or emollient is chosen based on a tions to alter solute skin solubility, partitioning
low RPI. Because the first chosen solvent or from the formulation to the skin and/or diffusivity
emollient has similar polarity to that of the active in the stratum corneum for promoting skin deliv-
chemical, they are mutually soluble; however, the ery. Ideally, they should also be non-irritant, cos-
escaping tendency is low. Therefore, a second metically acceptable (e.g. odourless, tasteless and
solvent or emollient is added to the formulation colourless), rapid acting with predictable dura-
to reduce the active chemical solubility to just tion, reversible and chemically stable. Potential
above the required solubility to solubilise the mechanisms by which solvents and vehicles,
9  Formulation Effects in Percutaneous Absorption 119

commonly used as enhancers, may affect stratum stratum corneum lipid extraction. Some sol-
corneum properties have been summarised in ear- vents such as dimethyl sulphoxide (DMSO) and
lier work (Roberts et al. 2002b). Briefly, altered ethanol may act by this mechanism although
skin barrier properties may result from the extrac- more systematic research needs to be conducted
tion of stratum corneum lipids, the penetration of to confirm the mechanism (Barry 2001; Benson
enhancers into or through the stratum corneum 2005). Alternatively, enhancers such as polar
and their interactions with stratum corneum com- solvents may also interact near the polar head
ponents (Lane 2013). Either intercellular lipids or groups of the stratum corneum lipids.
intracellular proteins of the stratum corneum, as Interactions with keratin in the corneocytes
proposed in the Lipid-Protein-Partitioning theory may open the dense protein structure and
of Barry (1991) could be potential sites of action thereby increase D. Surfactants, DMSO and
for enhancers. The effects of these interactions urea interact with keratin. The peptide/protein
may alter stratum corneum-vehicle partitioning, in the lipid bilayer domain may also be affected
skin solubility and/or penetrant diffusion in the by this type of enhancer (Benson 2005).
stratum corneum.
There is no accepted consensus on the 9.6.2.2 Effects on Solute Partitioning
­classification of enhancers. Nevertheless, simple and Solubility in Stratum
classifications such as organic vs. inorganic or Corneum
polar vs. non-polar have been used. In this review, Enhancers that increase solute solubility in the
enhancers are grouped according to the potential stratum corneum can promote partitioning into
mechanisms by which enhancers affect D, K, Rm the skin and hence improve flux. If enhancers per-
or combinations of those parameters. meate into the stratum corneum and shift the skin
polarity closer to that of the permeant, the perme-
9.6.2.1 Effects on Solute Diffusivity ant skin solubility and hence partitioning will
Enhancers have been postulated to affect diffu- increase (Cross et al. 2001). A similar result may
sivity in the stratum corneum in various ways. be expected via a solvent-drag mechanism, in
They can act on stratum corneum lipids as well which vehicle entering into the stratum corneum
as proteins. They may disturb the lipid organ- carries solute with it (Zhang et al. 2009; 2011). A
isation of the stratum corneum, making it flui- number of solvents have been reported to increase
dised. As the result of the disturbed stratum partitioning and stratum corneum solubility, e.g.
corneum lipid packing, D may be increased. ethanol, propylene glycol, Transcutol® and
Enhancers may act on the hydrophobic stratum N-methyl pyrrolidone (Williams and Barry 2004;
corneum lipid tails and create free volume Benson 2005).
available for a penetrating solute to diffuse.
Examples of common enhancers that work to 9.6.2.3 Synergistic Effects
increase diffusivity are laurocapram (Azone®, on Diffusivity and Solute Skin
Whitby Research Inc., USA) and oleic acid. Solubility
They may be dispersed in the stratum corneum Combinations of enhancers which increase D and
intercellular lipid, such as Azone® (Hoogstraate K or stratum corneum solubility have been
et al. 1991), or may be located in separate reported to give a synergistic effect of enhance-
domain pools within lipid domain, such as oleic ment. Examples for this approach are Azone®/
acid (Ongpipattanakul et al. 1991). Terpenes propylene glycol for metronidazole (Wotton et al.
such as limonene and cineole have also been 1985) and Azone®/diethylene glycol monoethyl
shown to increase drug diffusivity (Moghimi ether (Transcutol®, Gattefosse Co., France) for
et al. 1996a, b). In addition to disturbance of prostaglandin (Watkinson et al. 1991) as well as
the stratum corneum lipid packing, solute oleic acid and propylene glycol (Walker and
diffusivity may also be increased as a result of
­
Smith 1996; Benson 2005).
120 R. Kuswahyuning et al.

9.6.2.4 Formulation Dependency activity and the use of enhancers was shown by
of Enhancement Effects Rhee et al. (2007) in the development of a trans-
It is important to note that enhancers may dermal gel formulation for the anti-emetic drug,
affect skin properties through different mecha- clebopride. The first step was to investigate the
nisms, resulting in either enhancement (Megrab best solvent from a range of vehicles. Using satu-
et al. 1995; Watkinson et al. 2009), retardation rated drug solutions, the best enhancement was
(Hadgraft et al. 1996) or sometimes no significant with isopropyl myristate (IPM), the solvent in
effect (Sheth et al. 1986). The effect may be either which the drug had the lowest solubility. Because
drug dependent (Aungst et al. 1986), concentra- of clebopride’s low solubility in IPM, Transcutol®
tion dependent (Megrab et al. 1995; Watkinson was added as a co-solvent to increase drug solu-
et al. 2009), time dependent (Grice et al. 2010) bility. However, adding too much Transcutol®
or vehicle or formulation dependent (Sheth et al. led to a decreased clebopride flux. An optimal
1986; Rhee et al. 2007). Sheth et al. (1986) com- flux was achieved when 40–60 % Transcutol®
pared the penetration enhancement of trifluorothy- was added, resulting in 80 times greater enhance-
midine (TFT), an anti-viral compound, amongst ment compared to IPM alone. Other studies
propylene glycol, polyethylene ­glycol-300 (PEG showed similar results when combining hydro-
300) and water with and without the addition of philic and lipophilic penetration enhancers (Barry
Azone®. Without Azone®, TFT flux was ranked et al. 1995; Mayorga et al. 1996; Thomas and
in the order of propylene glycol > water > PEG Panchagnula 2003; Suwanpidokkul et al. 2004;
300. With the addition of 5 % Azone®, TFT flux Karande et al. 2006).
increased three- to fourfold as the ratio of PG :
PEG 300 in the vehicle went from 0:100 to 100:0.
This suggested that the extent of enhancement was 9.6.4 Hydration
dependent on the vehicles in which the enhanc-
ers were incorporated. In addition, it seemed that Skin hydration, the extent of hydration of the
PEG 300 was not a good solvent for enhancement stratum corneum, is a major determinant in per-
purposes (Hadgraft 1983). cutaneous absorption. At normal humidity,
15–20 % of the dry weight of stratum corneum is
made up of water. The stratum corneum may
9.6.3 Synergistic Thermodynamic expand by up to 300–400 % of its dry weight
Activity and Chemical when exposed to occlusion, soaking or very high
Enhancers humidity (Roberts et al. 2008). It has been pro-
posed that water taken up by the stratum corneum
Considering that percutaneous delivery involves may interpolate and open up tightly packed inter-
two consecutive processes, each of which may be cellular lipid bilayer structure, resulting in an
rate limiting, it would be beneficial if optimisation increased rate of skin delivery (Hikima and
addressed both thermodynamic activity and stra- Maibach 2006). However, fluidisation of the
tum corneum modification (diffusivity, partitioning intercellular lipid bilayer and partitioning modifi-
and solubility). Supersaturation coupled with oleic cation have also been suggested to explain the
acid, a lipid fluidiser, has been shown to increase effect of hydration on percutaneous absorption
flurbiprofen flux (Pellett et al. 1997). Wiechers’ (Benson 2005). Hydration can be influenced by
Formulating for Efficacy concept might also offer several means, including the presence of ingredi-
potential for synergistic effects of thermodynamic ents in the formulation such as humectants (glyc-
activity and enhancers. If a second chosen solvent erol, glycols). Urea is an example of a humectant,
or emollient were also an enhancer, a multiplicative where as a 10 % cream, it increases the water-­
flux enhancement could be expected. holding capacity of stratum corneum by 100 %
An example of how a formulation might be (Williams and Barry 1989). Urea has been
designed by maximising both thermodynamic reported to be responsible for significantly
9  Formulation Effects in Percutaneous Absorption 121

enhanced ketoprofen diffusion and permeability prodrug approach, and no external driving force,
through excised rat skin, using water as vehicle such as an applied current in iontophoresis, is
(Kim et al. 1993). Another method to increase used (Seung Jin et al. 1987).
hydration is the application of occlusive systems
like occlusive dressings, patches, ointments and
water-in-oil emulsions (Barry 2001; Zhai and 9.6.6 Complex Coacervation
Maibach 2002). However, Treffel et al. (1992)
have suggested that occlusion does not necessar- Another method to improve percutaneous flux of
ily increase penetration, as enhancement was charged chemicals is by complex coacervation.
shown only for lipophilic compounds. In con- Complex coacervation and ion pairing are similar
trast, Wurster and Kramer in 1961 (1961) showed in the use of counter-ions to increase chemical
that hydration promoted the penetration of the lipophilicity. However, the distinction lies in
more polar salicylate esters. Roberts et al. (2008) complex coacervates being two-phase systems,
have reviewed a number of other studies explor- i.e. a dilute aqueous phase and an oil phase with
ing the effect of hydration on skin penetration a high concentration of lipophilic complex. The
and found conflicting results, with no apparent two systems are in equilibrium with interchange
relationship between hydration and penetration of charged chemical from the aqueous phase and
in some cases. Following occlusion, reduced complex in the oil phase. A study using anti-­
paraben partitioning from the formulation to stra- depressants as model drugs suggested that the
tum corneum, without significant changes in dif- increased lipophilicity of complex coacervates
fusivity, was responsible for the decreased flux can increase the transdermal flux of charged spe-
from paraben-loaded ointment, whereas increased cies (Stott et al. 1996).
diffusivities, with relatively similar partitioning,
occurred with paraben-loaded acetone or ethanol
(Cross and Roberts 2000). 9.6.7 pH Adjustment

As mentioned above, many drugs are ionisable


9.6.5 Ion Pairs and therefore sensitive to pH changes. Studies on
the effect of pH on the penetration of ionisable
Many drugs are weak acids or bases that are ion- drug such as salicylic acid (Smith and Irwin
ised at normal physiological pH. Under these 2000), diclofenac (Obata et al. 1993) and ibupro-
conditions, they are generally poorly absorbed by fen (Watkinson et al. 2009) have been reported.
the membrane. Several techniques have been Ionised species are considered to penetrate poorly
studied to increase skin absorption of such mol- compared to non-ionised species (Scheuplein and
ecules, including prodrug design, iontophoresis, Blank 1971; Michaels et al. 1975). Kushla et al.
and ion pairing. In principle, ion pairing involves (Kushla and Zatz 1991) investigated the influence
the addition of an oppositely charged counter-ion of pH on lidocaine flux and permeability. It was
to form a neutral ion pair. This neutral species has found that the permeability coefficient of union-
increased lipophilicity and hence, membrane per- ised lidocaine through human skin was 50 times
meability is increased. The ion pair diffuses into greater than that of ionised lidocaine. It was sug-
the stratum corneum and dissociates to form the gested that pH adjustment in the vehicle can be
parent compound when it reaches the viable epi- one approach to alter the relative amount of non-­
dermis or deeper skin layers (Seung Jin et al. ionised to ionised compound and hence alter the
1987; Hatanaka et al. 2000; Megwa et al. 2000a, extent of penetration. Evidence has shown, how-
b). Table 9.3 lists formulation studies using an ever, that in some cases, the contribution of ion-
ion-pair approach. The ion-pairing technique ised species on the skin penetration cannot be
offers benefits over these two techniques because neglected. For example, steady state flux of
no chemical modification is required, as in the diclofenac (pKa of 4.7) increased abruptly as the
122 R. Kuswahyuning et al.

Table 9.3  Ion-pair studies in skin delivery


Chemical Counterion Membrane Effect References
Salicylate Alkyl amines and Human epidermis Enhancement was Megwa et al. (2000a)
quaternary ammonium ion shown using ternary
amines as counterion
A series of amines Human epidermis and No enhancement of flux Megwa et al.
anaesthetised rat through human (2000b)
epidermis skin in vitro;
but drug was localised
up to the top rat muscle
layer
Methotrexate Monooctyl phosphate, Hairless mouse skin Marked increase in flux Trotta et al. (1996)
monodecyl phosphate, using dodecyl sulphate
monodecyl and dioctyl
glycerophosphate, sulphosuccinate
taurodeoxycholate,
dodecyl sulphate and
dioctyl sulphosuccinate
Cephalexin Alkyl sulphonates, Rats Enhanced flux Hatanaka et al.
tetraalkylammonium (2000)
Piroxicam Monoethanolamine, Hairless mouse skin Enhanced skin Cheong and Choi
diethanolamine, permeability (2002)
triethanolamine
Retinoic acid Phenylalanine methyl Polydimethylsiloxane Permeation significantly Trotta et al. (2003)
ester, phenylalanine ethyl increased
ester, histidine methyl
ester, tryptophan methyl
ester, valine methyl ester
Risedronate L-arginine, L-lysine, Hairless mouse skin Flux enhancement Nam et al. (2011)
diethylenetriamine 36-fold higher
Lornoxicam Triethylamine, Rabbit skin in vitro All amine counterions, Xi et al. (2012)
diethylamine, especially
diethanolamine, triethanolamine,
triethanolamine showed obvious
enhancing effect

pH of the vehicle went from 3 to 7. As the perme- eutectic mixtures have been studied for enhanc-
ability coefficients decreased at the corresponding ing the flux of drugs. Examples are ibuprofen
pHs, the increase of flux was considered to arise (Stott et al. 1998) (from an ibuprofen-terpene
mainly from the increased solubility of the ion- eutectic mixture), testosterone (Kaplun-Frischoff
ised diclofenac at higher pH (Obata et al. 1993). and Touitou 1997) (from a testosterone-menthol
eutectic mixture) and the β-blocker propranolol
(Stott et al. 2001) (from a propranolol-fatty acids
9.6.8 Eutectic Mixtures eutectic mixture).

A eutectic mixture is a mixture of two or more


components that do not interact to form a new 9.7 I mportance of Other
chemical compound but which, in certain ratios, Formulation Ingredients
inhibit crystallisation of one another, resulting in on Percutaneous Absorption
a system with a lower melting point than either of
the components. Since the decrease of melting In addition to maximising skin delivery to
point has been shown to be inversely proportional improve bioavailability, formulation must also
to lipophilicity, and hence transdermal flux, ensure physicochemical stability of the product
9  Formulation Effects in Percutaneous Absorption 123

Table 9.4  Ingredients commonly added to achieve phys- solubility as the amount of ethanol was increased
ically stable and cosmetically and aesthethically accept-
from 0 to 100 % (Watkinson et al. 2009). A simi-
able formulations
lar result was found for oestradiol, where its solu-
Types of ingredient needed
bility was increased by 30-fold (Megrab et al.
Physicochemical Dispersing and/or solubilising
1995), and subsequently, the partition coefficient
stability of the agent, e.g. water, propylene
active chemical and glycol, ethanol was decreased.
formulation Emulsifier, e.g. polysorbates, Many solvents and co-solvents are known to
cetrimide, sodium dodecyl influence the barrier properties of the skin.
sulphate Ethanol was used in a number of studies as a co-­
Increase vesicles stability, e.g. solvent (Pershing et al. 1990; Hatanaka et al.
cholesterol addition in
conventional liposomes 1993; Obata et al. 1993). Megrab et al. (1995)
Preservatives, e.g. benzyl alcohol, investigated the mechanism behind the enhance-
phenol, chlorocresol ment of oestradiol flux using saturated oestradiol
Antioxidant in 0–90 % ethanol : water compositions tested
Improve cosmetic Emollient, humectant, using three different membranes, namely, excised
and aesthetic moisturiser, e.g. urea human skin, silastic membrane and snake skin.
acceptance Colouring Maximum flux was achieved at different ethanol
Fragrance, e.g. terpenes
concentrations, namely, 40–60 %, 80 % and 40 %
Viscosity modifiers, e.g.
carbomer, carrageenan, acacia,
(followed by a constant phase up to 90 % etha-
alginates, xanthan nol) for excised human skin, silastic membrane
and snake skin, respectively. They have sug-
gested that enhancement of oestradiol flux at low
as well as cosmetic and aesthetic acceptance. ethanol concentrations was due to the increased
Various ingredients are usually incorporated into drug solubility in the stratum corneum. The
the formulation to achieve these purposes. decreased oestradiol fluxes at higher ethanol con-
Table  9.4 shows examples of common ingredi- centrations were due to the dehydration effects of
ents for such purposes and their functions in the ethanol on the stratum corneum. In agreement
formulation. The effects of such ingredients on with this, Watkinson et al. (2009) found greater
percutaneous absorption are discussed here. enhancement for ibuprofen flux with 75 % etha-
nol for human skin. This evidence emphasised a
concentration dependence of ethanol for flux
9.7.1 Solvent and Co-solvent enhancement.
Propylene glycol (PG), a colourless and vis-
One of the common ingredients needed in the cous liquid, is commonly used in topical formu-
formulation is a solubilising agent. Active lations (Arellano et al. 1999; Nicolazzo et al.
­ingredients may be hydrophilic or hydrophobic 2005). The binary system PG : water has been
in nature. Due to the principle of “like dissolves shown to enhance ibuprofen flux either in excised
like”, while a hydrophilic chemical may be easily human skin or silicone membranes. A linear
solubilised in a polar vehicle such as water, a enhancement with increasing PG content was
hydrophobic chemical may face difficulty. The seen in human skin membranes, but in the case of
co-solvent method, where two miscible solvents silastic membrane, there was no linear relation-
are mixed, is a common technique used to ship and the optimum enhancement was achieved
increase solubility in a formulation (Rhee et al. at 70 % PG. The effect of PG on human skin was
2007; Watkinson et al. 2009). To increase the primarily on the solubility and partitioning of
solubility of ibuprofen, a lipophilic compound, ibuprofen (Watkinson et al. 2009).
ethanol : water co-solvent mixtures have been Another study investigated the effect of a ter-
employed. Ibuprofen solubility was greatly nary mixture of ethanol : PG : water on drug flux.
increased by 5,500-fold relative to its aqueous Recent research by our colleagues Grice et al.
124 R. Kuswahyuning et al.

(2010) investigated the influence of a combina- maximum flux of hydrocortisone was directly
tion ethanol : PG : water, i.e. 60 : 20 : 20; 80 : 20 related to the volume of vehicle absorbed by the
: 0 and 0 : 80 : 20 on the minoxidil uptake into membrane. However, a dependency of the vehi-
appendages, stratum corneum and through cle volume fraction sorbed into silastic mem-
human skin in vitro. A change in the transport brane on δv for a wide range of structurally
mechanism of minoxidil uptake was noticed. At unrelated vehicles was not seen. A linear observed
early times (before 12 h), formulations contain- versus predicted solvent volume fraction rela-
ing high amounts of ethanol with the least amount tionship was shown after adjusting δv for vehicle
of PG gave higher minoxidil uptake. Evaporation molecular weight and hydrogen-bonding proper-
of volatile solvents, e.g. ethanol, led to increased ties. This result indicates that it is not only δv but
minoxidil concentrations due to the resulting also vehicle molecular size and hydrogen bond-
reduction in volume. After 12 h, on the other ing that determine vehicle-membrane interac-
hand, maximum flux was obtained from the for- tions (Cross et al. 2001). In addition, vehicle
mulation with the least amount of ethanol but the molecular size has been shown to be an important
highest amount of PG. The authors suggested determinant for mobility in the membrane (Most
that PG was slowly taken up by the membrane, 1972). Further, the relative importance of solute
leading to its physical modification and enhanced diffusivity or membrane solubility as dominating
minoxidil flux. A similar result was found by factors responsible for the enhanced flux follow-
Tata et al. (1994). ing solvent uptake is likely to be determined
Vehicle uptake into the membrane can by the combination of solute and solvent used
strongly influence skin penetration. Our recent (Cross et al. 2001). For example, while Sloan
study, for example, showed that maximum flux found that partitioning was the dominant factor,
from similar sized compounds was determined in our hydrocortisone penetration study we
by solute solubility in the stratum corneum observed that flux was dominated by neither
(Zhang et al. 2009) which was dependent on the diffusivity nor membrane vehicle partition.
­
amount of vehicle penetrating into the stratum A combination of both diffusivity and membrane
corneum (Zhang et al. 2011). The ability to pre- partitioning effects determined vehicle-mem-
dict vehicle uptake into the membrane and the brane interactions.
rate of solute penetration into the skin would
have significant impact on clinical and toxico-
logical applications (Sloan et al. 1986). Earlier 9.7.2 Viscosity Modifiers
work by Potts and Guy (1992) to predict the
extent of percutaneous absorption was based on To increase product acceptance, it would be pref-
a QSPR approach. Using Flynn’s (1990) data- erable to thicken low-viscosity formulations,
base generated from in vitro data, it was shown such as co-solvent mixtures (Rhee et al. 2007)
that lipophilicity and molecular weight of the and microemulsions (Chen et al. 2006; 2007; Zhu
solute determine skin penetration (Mitragotri et al. 2009). A number of viscosity modifiers
et al. 2011). have been investigated for this purpose including
Attempts have been made to relate vehicle silicone dioxide (Rhee et al. 2007), carbomer
physicochemical properties and flux across the (Carbopol® 940, Lubrizol Co., USA) (Chen
skin. The use of solubility parameters (δ) has et al. 2007) and carrageenan (Valenta and Schultz
been shown to be useful as a flux descriptor 2004).
(Sloan et al. 1986; Jiang et al. 1998; Cross et al. One of the drawbacks of incorporating viscos-
2001; Dias et al. 2007). Sloan suggested that ity modifiers in a formulation is that they may
uptake of vehicle into a membrane has its maxi- hinder drug diffusion from the formulation into
mum effect in increasing solute flux when the the skin. For example, addition of a 7 % gelling
solubility parameter of the vehicle, δv, is close to agent to thicken a Transcutol® : isopropyl
that of membrane, δs. Our work showed that the myristate binary mixture (40:60) resulted in a
9  Formulation Effects in Percutaneous Absorption 125

significantly reduced steady state clebopride flux, p­ orcine skin. Addition of SLS to a water vehicle
four times lower compared to the un-gelled opti- was generally found to reduce partitioning and
mised mixture. It was suggested that this was due permeability of lipophilic compounds. The pres-
to the increased viscosity of the clebopride gel ence of SLS at a concentration above its critical
formulation (Rhee et al. 2007). A significantly micelle concentration caused the formation of
reduced permeability coefficient was also seen micelles around the non-polar compounds and
when 4 % Carbopol® 940 was added to thicken reduced their availability for absorption. In con-
an oestradiol microemulsion, which could be trast, when SLS was incorporated into an ethanol
partly due to the increased viscosity of the micro- vehicle, it did not affect partitioning. The authors
emulsion (Peltola et al. 2003). hypothesised that since ethanol has a short car-
On the other hand, there are examples in the bon chain and a polar group, it is able to attract
literature where the addition of viscosity modifi- polar and non-polar molecules making partition-
ers has no effect on permeation rates. Chen et al. ing from an ethanol vehicle system not sensitive
(2007) used a hydrogel-thickened microemulsion to SLS addition. Increased permeability from an
for the topical administration of tripolide at an ethanol vehicle system was linked to changes in
extremely low concentration. Even though the diffusivity.
microemulsion viscosity was increased signifi-
cantly by the addition of Carbopol® 940, no sig-
nificant difference in the tripolide steady state 9.7.4 Ingredients Used
flux was observed compared to the microemul- for Customer Acceptance
sion without Carbopol® 940. The authors sug-
gested that close contact of microemulsion Product acceptance is another aspect that can be
droplets to the skin promoted by the adhesiveness improved by the ingredients in formulation.
of Carbopol® 940 might play a role in tripolide Fragrance is the most commonly added in the
penetration across mouse skin. A similar mecha- topical formulations to provide an agreeable
nism has been attributed to the facilitation of odour or mask an unacceptable one. They are
sodium fluorescein penetration from carrageenan-­ usually lipophilic compounds and therefore more
thickened microemulsions (Valenta and Schultz likely to be absorbed into the skin. Diethyl male-
2004). ate, an example of a fragrance compound, was
absorbed at 54 % of the applied dose over 24 h in
a human in vivo under occluded conditions
9.7.3 Ingredients Used for Stability (Bronaugh et al. 1990). Other fragrance examples
are benzyl derivatives (Bronaugh et al. 1990) and
Ingredients may be added to a formulation to aid terpenes (Hotchkiss 1998). It is worth noting that
its physical stability. For instance, emulsions are terpenes used as fragrances have also been shown
formed from mixtures of immiscible liquids, in to increase percutaneous absorption. For instance,
which one phase will be dispersed in the other. 1,8 cineole enhanced the diffusion coefficient of
This requires a third component such as an emul- oestradiol, a lipophilic model drug, and
sifier to lower interfacial tension and ensure 5-­fluorouracil, a hydrophilic model drug
physical stability (Riviere et al. 2010). (Moghimi et al. 1996a, b, 1997).
The effects of surfactants on skin absorption
have been extensively studied (Shokri et al. 2001;
van der Merwe and Riviere 2005; Riviere et al. 9.8 Formulation-Related Issues
2010). For example, van der Merwe et al. (van for Nanoparticulate Carriers
der Merwe and Riviere 2005) studied the influ-
ence of sodium lauryl sulphate (SLS) on the per- The range of dosage forms for skin delivery can
meability and stratum corneum partitioning of 10 be broadly divided into conventional and novel
agricultural and industrial chemicals using delivery systems. Conventional dosage forms
126 R. Kuswahyuning et al.

Fig. 9.3  Some representative a


of conventional (Adapted
Oil Aqueous Powder
from Roberts et al. (2002a)) (including cosolvent)
(a) and nanoparticle novel
delivery systems used as
dermatological carriers
(Adapted from Honeywell-
Nguyen and Bouwstra (2005),
Pardeike et al. (2009)) (b)
Powder in aqueous Powder in oil Aqueous in oil oil in Aqueous

Semisolid Powder in Semisolid

Skin

b
Double chain Single chain surfactant External phase
non-ionic surfactant or lipid
Active
compound

Lipid
Solid lipid nanoparticles Nanostructured lipid carriers
Vesicles
Active compound

Skin

encompass liquids (such as lotions, emulsions), (Yarosh 2001; Cevc 2004; Honeywell-Nguyen
semisolids (such as ointments, pastes, gels and and Bouwstra 2005; Elsayed et al. 2007) and
creams) and solid preparations (such as powders nanoparticles (Souto et al. 2007; Souto and
or patches) (Epstein 2009; Narasimha Murthy Müller 2008; Pardeike et al. 2009; Prow et al.
and Shivakumar 2010). Besides these, there are 2011) for dermal and transdermal delivery. We
different novel and more complicated systems will now give a general description of some novel
that are used to optimise percutaneous absorption delivery systems followed by a discussion of
of drugs like patches, microneedles and nanopar- their application to percutaneous.
ticles. Among these systems, nanoparticles that Microemulsions have gained interest as poten-
have the ability to penetrate skin will be dis- tial drug carriers due to several beneficial features,
cussed in more detail below. Some examples of such as ease of preparation, long-term stability,
conventional formulations and nanoparticulate high solubilisation capacity of hydrophilic and
delivery systems used as dermatological carriers hydrophobic drugs and improved drug delivery.
are shown in Fig. 9.3. However, the mechanisms behind the enhance-
A number of review articles have been pub- ment are still unclear. Some published studies
lished summarising the application of novel attributed the enhancement to the composition as
delivery systems such as microemulsions well as its formulation characteristics (Heuschkel
(Lawrence and Rees 2000; Kogan and Garti et al. 2008). Typically, a microemulsion consists
2006; Heuschkel et al. 2008; Santos et al. 2008), of lipophilic and hydrophilic ingredients, surfac-
vesicle (liposomes and flexible liposomes) tant and co-surfactant. Microemulsion formation
9  Formulation Effects in Percutaneous Absorption 127

is highly dependent on the surfactant capacity liposome formulation, into highly deformable
to reduce interfacial tension between the hydro- liposomes, including Transfersomes (Cevc et al.
philic and lipophilic ingredients. Often, surfactant 1998; 2002; Schätzlein and Cevc 1998; Jain et al.
alone is not sufficient to ensure physical stability 2003), ethosomes (Dayan and Touitou 2000;
of the mixture. Co-surfactant addition will further Touitou et al. 2000; El Zaafarany et al. 2010),
reduce interfacial tension to facilitate microemul- niosomes (Schreier and Bouwstra 1994; Fang
sion formation. Non-ionic surfactants, such as et al. 2001) and SECosomes (Geusens et al.
polyglyceryl-6-isostearate (Plurol® Isostearique, 2010) by the addition of other ingredients.
Gattefosse Co., France), highly purified dieth- Transfersomes are liposomes consisting of sur-
ylene glycol monoethyl ether (Transcutol® P, factant and ethanol, whereas ethosomes are lipo-
Gattefosse Co., France) and caprylcaproyl mac- somes with a high alcohol content. Niosomes
rogol glycerides (Labrasol®, Gattefosse Co., result from the addition of a non-ionic surfactant,
France), are usually considered safe for human whereas SECosomes are produced by the addi-
skin application and less irritating than ionic sur- tion of a combination of surfactant and ethanol
factants. Examples of co-surfactants are short- or (Geusens et al. 2010). Unlike classical liposomes,
medium-chain alcohols such as ethanol, isopro- those highly deformable liposomes are reported
panol and butanol. Isopropyl myristate, isopro- to be able to penetrate stratum corneum and even
pyl palmitate, ethyl oleate and oleic acid are deeper skin layers, carrying encapsulated mole-
commonly used as lipophilic phases, whereas cules with them.
water, viscosity-enhancing agents, sodium chlo- Solid lipid nanoparticles (SLN) consist of a
ride, buffer salts and preservatives are included mixture of solid lipids and surfactants. The ingre-
in the hydrophilic phase. For topical applica- dients are usually those commonly used in topical
tions, microemulsions have been used to deliver cosmetic and pharmaceutical products, which are
model chemicals such as retinoic acid (Trotta considered as generally regarded as safe (GRAS)
et al. 2003) and tea tree oil (Biju et al. 2005). substances, providing a broad range of choices
For transdermal applications, steroids such as which may avoid safety issue problems. In addi-
oestradiol (Peltola et al. 2003) and testosterone tion, the concentrations of the ingredients are simi-
(Malcolmson and Lawrence 1993) as well as beta lar to those used in conventional formulations.
blockers (Kemken et al. 1991; 1992) have been It was shown that incorporation of an active
studied. A major concern over the application of compound into SLN could increase its stabil-
microemulsions, requiring further research, is the ity. The nature of the solid lipids and the con-
irritation potential due to the high surfactant/co- centration of surfactant in the formulation were
surfactant content (Santos et al. 2008). important parameters contributing to the stabil-
Liposomes are colloidal structures consisting ity (Müller et al. 2002). The potential for occlu-
of a mixture of phosphatidylcholine and choles- sion due to the small particle size of SLN and
terol, as typical ingredients, and other substances. the ability to control active compound release
First reported in 1980, liposomes showed supe- are other attractive features of SLN for topical
rior percutaneous enhancement compared to con- application (Müller et al. 2002; Neubert 2011).
ventional dosage forms such as lotions and Nanostructured lipid carriers (NLC) are a new
creams. Despite the potential applications for generation of lipid nanoparticles developed to
delivering chemicals topically or systemically, overcome some limitations of SLN. During stor-
the current weight of research suggests that age, SLN transform into highly ordered crys-
liposomes remain accumulated in the stratum
­ tal forms, resulting in drug expulsion. In NLC,
corneum and upper skin layers. The application however, lipid material consists of a blend of
to transdermal delivery is less promising because solid lipids and liquid lipids (oils) which reduces
of generally minimal drug penetration into deeper crystal order and hence deters drug expulsion.
skin layers and the systemic circulation. Lipid nanoparticles have been used as vehicles
Numerous studies have modified the classical for the topical application of anti-acne drugs and
128 R. Kuswahyuning et al.

Table 9.5  Proposed mechanisms in promoting dermal and transdermal delivery from micellar systems and lipid
nanoparticles
Lipid nanoparticles
Microemulsion (Santos et al. Liposomes (Touitou et al. Flexible liposomes (Schafer-Korting et al.
2008) 1994; Elsayed et al. 2007) (Elsayed et al. 2007) 2007)
High solubilisation capacity Liquid and gel state of Liquid and gel state of Component of formulation
for hydrophilic and liposomes liposomes may penetrate into stratum
lipophilic drug corneum
Formulation component such Component of liposomes Intact vesicles penetrate Release modulation from
as surfactant, cosurfactant may penetrate into stratum into stratum corneum formulation
and oil may act as corneum
penetration enhancer
Increase of hydration Component of liposomes Occlusion
may penetrate into stratum
corneum
Very low interfacial tension
ensures excellent contact
with skin

s­unscreen agents (­Schafer-Korting et al. 2007). A similar situation is seen with NLC, in which
Lipid particles have been tested in vitro, ex vivo liquid or oil lipid is incorporated into the formu-
and in vivo for transdermal delivery of flurbipro- lation mixture to minimise drug expulsion during
fen. Both SLN and NLC exhibited sustained drug storage. While solute solubility in the NLC
release over a period of 24 h (Bhaskar et al. 2009). increases, the thermodynamic activity will also
Although these novel formulations have been change, and hence, solute concentration might
shown to promote penetration across the skin, the require adjustment.
exact mechanism is not yet clearly understood. In Intact vesicles containing entrapped drug have
general, it is usually postulated that the effective- also been proposed to penetrate into the stratum
ness of drug delivery systems is related to a vari- corneum. Studies performed with surfactant-­
ety of factors depending on the composition of based deformable vesicles indicate that intact
the formulation which also may strongly influ- vesicles may partition into the stratum corneum,
ence formulation characteristics. Table 9.5 pres- but show very limited partitioning into the viable
ents proposed mechanisms for potential complex epidermis, if any (Honeywell-Nguyen and
formulations in dermal and transdermal delivery. Bouwstra 2005). Interactions of the formulations
The relationships between thermodynamic with skin may also occur due to increased hydra-
activity and percutaneous absorption are likely to tion following film formation (Müller et al.
be less well defined in such formulations. 2002), water penetration from oil in water sys-
Ingredients that are required to achieve formula- tems and occlusive conditions in patch systems.
tion stability could also affect solute thermody- Despite rapid development in formulation
namic activity in the formulation and, hence, skin technologies for topical and transdermal delivery,
delivery. Surfactant and co-surfactant, for exam- a reliable percutaneous absorption prediction
ples, are required to reduce interfacial tension in from such formulations remains a challenge.
microemulsion formation. At the same time, their Unlike simple formulations, where the dominant
inclusion in the formulation may increase solute factors controlling partitioning from vehicle to
solubility in the formulation. Indeed, there are skin, diffusivity and skin solubility could be dis-
reports indicating that an increased amount of tinguished relatively easily, complex formulations
surfactant in the microemulsion did not increase are more challenging. Expanding on approaches
flux (Chen et al. 2004; Yuan et al. 2006). If a sol- used for simple vehicle mixtures, Riviere et al.
ute is incorporated at or near its saturation limit, (Ghafourian et al. 2010a, b) recently employed a
solute thermodynamic activity will be enhanced. QSPR method to relate skin penetration to the
9  Formulation Effects in Percutaneous Absorption 129

chemical properties of the mixture and the molec- References


ular structure of the penetrants. Permeability coef-
ficients were obtained from 12 different penetrants Anderson BD, Raykar PV (1989) Solute structure-­
permeability relationships in human stratum corneum.
each incorporated in 24 different mixtures of vari- J Invest Dermatol 93(2):280–286
ous solvents, a surfactant (sodium lauryl sulphate) Anissimov YG, Jepps OG, Dancik Y, Roberts MS (2013)
and a vasodilator (methyl nicotinate) measured Mathematical and pharmacokinetic modelling of epi-
from finite-dose application through porcine skin. dermal and dermal transport processes. Adv Drug
Deliv Rev 65(2):169–190
Prediction was performed to relate penetrant Arellano A, Santoyo S, Martín C, Ygartua P (1999)
physicochemical properties (i.e. log P and the 9th Influence of propylene glycol and isopropyl myristate
order path molecular connectivity index) and sol- on the in vitro percutaneous penetration of diclof-
vent properties (i.e. the difference between boil- enac sodium from carbopol gels. Eur J Pharm Sci
7(2):129–135
ing point and melting points). The result suggests Aungst BJ, Rogers NJ, Shefter E (1986) Enhancement
that skin penetration from complex mixtures of naloxone penetration through human skin in vitro
could be well predicted using a QSPR approach using fatty acids, fatty alcohols, surfactants, sulfoxides
and that high skin penetration would be predicted and amides. Int J Pharm 33(1–3):225–234
Barry BW (1991) Lipid-protein-partitioning theory of
from vehicle mixtures having small differences in skin penetration enhancement. J Control Release
their boiling and melting points. 15(3):237–248
Barry BW (2001) Novel mechanisms and devices to
enable successful transdermal drug delivery. Eur J
Conclusion Pharm Sci 14(2):101–114
Barry BW, Harisson SM, Dugard PH (1985a) Vapour and
Drug delivery into or through the skin is an
liquid diffusion of model penetrants through human
attractive route for local or transdermal appli- skin; correlation with thermodynamic activity. J
cations. The skin barrier, particularly of the Pharm Pharmacol 37:226–235
stratum corneum, has yet to be predictably and Barry BW, Harrison SM, Dugard PH (1985b) Correlation
of thermodynamic activity and vapour diffusion
­consistently overcome to increase bioavailabil-
through human skin for the model compound, benzyl
ity. Formulation-based optimisation, using alcohol. J Pharm Pharmacol 37:84–90
various techniques including thermodynamic Barry BW, Yamane MA, Williams AC (1995) Effects of
activity, chemical penetration enhancers, terpenes and oleic acid as skin penetration enhancers
towards 5-fluorouracil as assessed with time; perme-
hydration, ion pairs, complex coacervation, pH
ation, partitioning and differential scanning calorim-
adjustment and eutectic mixtures, is employed etry. Int J Pharm 116(2):237–251
to achieve efficacious skin delivery. In addition Benson H (2005) Transdermal drug delivery: penetration
to this, ingredients commonly used for physi- enhancement techniques. Curr Drug Deliv 2(1):23–33
Bhaskar K, Anbu J, Ravichandiran V, Venkateswarlu
cochemical formulation stability, aesthetic or
V, Rao Y (2009) Lipid nanoparticles for transder-
cosmetic purposes might potentially alter drug mal delivery of flurbiprofen: formulation, & in vitro,
penetration. Potential novel dermal and trans- ex vivo; and in vivo studies. Lipids Health Dis
dermal delivery systems, e.g. microemulsions, 8(1):1–15
Biju SS, Ahuja A, Khar RK (2005) Tea tree oil con-
flexible liposomes and lipid nanoparticles,
centration in follicular casts after topical delivery:
have been shown to promote skin penetration determination by high-performance thin layer chro-
through various mechanisms requiring further matography using a perfused bovine udder model. J
systematic investigation. A reliable method to Pharm Sci 94(2):240–245
Bronaugh RL, Wester RC, Bucks D, Maibach HI, Sarason
predict percutaneous absorption across all
R (1990) In vivo percutaneous absorption of fragrance
varieties of solutes, complex formulation types ingredients in rhesus monkeys and humans. Food
and conditions remains a challenge. Chem Toxicol 28(5):369–373
Cevc G (2004) Lipid vesicles and other colloids as
drug carriers on the skin. Adv Drug Deliv Rev
Acknowledgements  The authors acknowledge financial
56(5):675–711
support from the National Health and Medical Research
Cevc G, Gebauer D, Stieber J, Schätzlein A, Blume G
Council of Australia. R.K. gratefully thanks the Indonesian
(1998) Ultraflexible vesicles, transfersomes, have an
Directorate General of Higher Education (DIKTI) for the
extremely low pore penetration resistance and transport
­
scholarship.
130 R. Kuswahyuning et al.

therapeutic amounts of insulin across the intact mam- Fang J-Y, Hong C-T, Chiu W-T, Wang Y-Y (2001) Effect
malian skin. BBA Biomembr 1368(2):201–215 of liposomes and niosomes on skin permeation of
Cevc G, Schätzlein A, Richardsen H (2002) enoxacin. Int J Pharm 219(1–2):61–72
Ultradeformable lipid vesicles can penetrate the skin Finnin BC, Morgan TM (1999) Transdermal penetration
and other semi-permeable barriers unfragmented. enhancers: applications, limitations, and potential.
Evidence from double label CLSM experiments J Pharm Sci 88(10):955–958
and direct size measurements. BBA Biomembr Fluhr JW, Darlenski R (2009) In: Revuz J, Roujeau J-C,
1564(1):21–30 Kerdel FA, Valeyrie-Allanore L (eds) Skin barrier life-­
Chen H, Chang X, Weng T, Zhao X, Gao Z, Yang Y, Xu threatening dermatoses and emergencies in dermatol-
H, Yang X (2004) A study of microemulsion sys- ogy. Springer, Berlin, pp 3–18
tems for transdermal delivery of triptolide. J Control Flynn GL (1990) Physicochemical determinants of skin
Release 98(3):427–436 absorption. In: Gerrity T, Henry C (eds) Principles
Chen H, Chang X, Du D, Li J, Xu H, Yang X (2006) of route-to-route extrapolation for risk assessment,
Microemulsion-based hydrogel formulation of ibupro- Elsevier, New York, pp 93–127
fen for topical delivery. Int J Pharm 315(1–2):52–58 Geusens B, Van Gele M, Braat S, De Smedt SC, Stuart
Chen H, Mou D, Du D, Chang X, Zhu D, Liu J, Xu H, Yang MCA, Prow TW, Sanchez W, Roberts MS, Sanders NN,
X (2007) Hydrogel-thickened microemulsion for topi- Lambert J (2010) Flexible nanosomes (SECosomes)
cal administration of drug molecule at an extremely enable efficient siRNA delivery in cultured primary
low concentration. Int J Pharm 341(1–2):78–84 skin cells and in the viable epidermis of ex vivo human
Cheong H-A, Choi H-K (2002) Enhanced percutaneous skin. Adv Funct Mater 20(23):4077–4090
absorption of piroxicam via salt formation with etha- Ghafourian T, Samaras EG, Brooks JD, Riviere JE (2010a)
nolamines. Pharm Res 19(9):1375–1380 Modelling the effect of mixture components on per-
Cleek RL, Bunge AL (1993) A new method for estimating meation through skin. Int J Pharm 398(1–2):28–32
dermal absorption from chemical exposure. 1. General Ghafourian T, Samaras EG, Brooks JD, Riviere JE (2010b)
approach. Pharm Res 10(4):497–506 Validated models for predicting skin penetration from
Cross SE, Roberts MS (2000) The effect of occlusion on different vehicles. Eur J Pharm Sci 41(5):612–616
epidermal penetration of parabens from a commercial Grice JE, Ciotti S, Weiner N, Lockwood P, Cross SE,
allergy test ointment, acetone and ethanol vehicles. Roberts MS (2010) Relative uptake of minoxidil into
J Invest Dermatol 115(5):914–918 appendages and stratum corneum and permeation
Cross SE, Pugh W, Hadgraft J, Roberts MS (2001) Probing through human skin in vitro. J Pharm Sci 99(2):712–718
the effect of vehicles on topical delivery: understand- Hadgraft J (1983) Percutaneous absorption: possibilities
ing the basic relationship between solvent and solute and problems. Int J Pharm 16(3):255–270
penetration using silicone membranes. Pharm Res Hadgraft J (2001) Skin, the final frontier. Int J Pharm
18(7):999–1005 224(1–2):1–18
Dancik Y, Anissimov YG, Jepps OG, Roberts MS (2012) Hadgraft J, Peck J, Williams DG, Pugh WJ, Allan G
Convective transport of highly plasma protein bound (1996) Mechanisms of action of skin penetration
drugs facilitates direct penetration into deep tis- enhancers/retarders: azone and analogues. Int J Pharm
sues after topical application. Br J Clin Pharmacol 141(1–2):17–25
73(4):564–578 Hatanaka T, Shimoyama M, Sugibayashi K, Morimoto
Dayan N, Touitou E (2000) Carriers for skin delivery Y (1993) Effect of vehicle on the skin permeability
of trihexyphenidyl HCl: ethosomes vs. liposomes. of drugs: polyethylene glycol 400-water and ethanol-­
Biomaterials 21(18):1879–1885 water binary solvents. J Control Release 23(3):247–260
Dias M, Hadgraft J, Lane ME (2007) Influence of Hatanaka T, Kamon T, Morigaki S, Katayama K, Koizumi
membrane-­solvent-solute interactions on solute per- T (2000) Ion pair skin transport of a zwitterionic drug,
meation in skin. Int J Pharm 340(1–2):65–70 cephalexin. J Control Release 66(1):63–71
El Zaafarany GM, Awad GAS, Holayel SM, Mortada ND Heuschkel S, Goebel A, Neubert RHH (2008)
(2010) Role of edge activators and surface charge in Microemulsions—modern colloidal carrier for dermal
developing ultradeformable vesicles with enhanced and transdermal drug delivery. J Pharm Sci 97(2):603–631
skin delivery. Int J Pharm 397(1–2):164–172 Hikima T, Maibach H (2006) Skin penetration flux and lag-
Elias PM (1983) Epidermal lipids, barrier function, and time of steroids across hydrated and dehydrated human
desquamation. J Invest Dermatol 80(1 Suppl):44s–49s skin in vitro. Biol Pharm Bull 29(11):2270–2273
Elsayed MMA, Abdallah OY, Naggar VF, Khalafallah Honeywell-Nguyen PL, Bouwstra JA (2005) Vesicles as a
NM (2007) Lipid vesicles for skin delivery of drugs: tool for transdermal and dermal delivery. Drug Discov
reviewing three decades of research. Int J Pharm Today 2(1):67–74
332(1–2):1–16 Hoogstraate AJ, Verhoef J, Brussee J, Ijzerman AP, Spies
Epstein H (2009) Cosmeceutical vehicles. Clin Dermatol F, Boddé HE (1991) Kinetics, ultrastructural aspects
27(5):453–460 and molecular modelling of transdermal peptide flux
Essa EA, Bonner MC, Barry BW (2002) Human skin enhancement by N-alkylazacycloheptanones. Int J
sandwich for assessing shunt route penetration during Pharm 76(1–2):37–47
passive and iontophoretic drug and liposome delivery. Hotchkiss SAM (1998) Absorption of fragrance ingredi-
J Pharm Pharmacol 54(11):1481–1490 ents using in vitro models with human skin. In: Frosch
9  Formulation Effects in Percutaneous Absorption 131

P, Johansen J, White I (eds) Fragrances. Springer, ute maximum flux across the skin. J Invest Dermatol
Berlin, pp 125–135 122(4):993–999
Ishii H, Todo H, Sugibayashi K (2010) Effect of ther- Malcolmson C, Lawrence MJ (1993) A comparison of
modynamic activity on skin permeation and skin the incorporation of model steroids into non-ionic
concentration of triamcinolone acetonide. Chem
­ micellar and microemulsion systems. J Pharm
Pharm Bull 58(4):556–561 Pharmacol 45(2):141–143
Jain S, Jain P, Umamaheshwari RB, Jain NK (2003) Mayorga P, Puisieux F, Couarraze G (1996) Formulation
Transfersomes—a novel vesicular carrier for enhanced study of a transdermal delivery system of primaquine.
transdermal delivery: development, characterization, Int J Pharm 132(1–2):71–79
and performance evaluation. DDIP 29(9):1013 Megrab NA, Williams AC, Barry BW (1995) Oestradiol
Jepps OG, Dancik Y, Anissimov YG, Roberts MS (2013) permeation across human skin, silastic and snake skin
Modeling the human skin barrier — towards a better membranes: the effects of ethanol/water co-solvent
understanding of dermal absorption. Adv Drug Deliv systems. Int J Pharm 116(1):101–112
Rev 65(2):152–168 Megwa SA, Cross SE, Benson HAE, Roberts MS
Jiang R, Benson HAE, Cross SE, Roberts MS (1998) (2000a) Ion-pair formation as a strategy to enhance
In vitro human epidermal and polyethylene mem- topical delivery of salicylic acid. J Pharm Pharmacol
brane penetration and retention of the sunscreen 52(8):919–928
benzophenone-­3 from a range of solvents. Pharm Res Megwa SA, Cross SE, Whitehouse MW, Benson HAE,
15(12):1863–1868 Roberts MS (2000b) Effect of ion pairing with alkyl-
Kaplun-Frischoff Y, Touitou E (1997) Testosterone skin amines on the in-vitro dermal penetration and local
permeation enhancement by menthol through forma- tissue disposition of salicylates. J Pharm Pharmacol
tion of eutectic with drug and interaction with skin 52(8):929–940
lipids. J Pharm Sci 86(12):1394–1399 Mertin D, Lippold BC (1997) In-vitro permeability of the
Karande P, Jain A, Mitragotri S (2006) Insights into syn- human nail and of a keratin membrane from bovine
ergistic interactions in binary mixtures of chemical hooves: prediction of the penetration rate of antimy-
permeation enhancers for transdermal drug delivery. cotics through the nail plate and their efficacy. J Pharm
J Control Release 115(1):85–93 Pharmacol 49(9):866–872
Kemken J, Ziegler A, MÜLler BW (1991) Investigations Michaels AS, Chandrasekaran SK, Shaw JE (1975) Drug
into the pharmacodynamic effects of dermally permeation through human skin: theory and in vitro
administered microemulsions containing β-blockers. experimental measurement. Am Inst Chem Eng J 21(5):
J Pharm Pharmacol 43(10):679–684 985–996
Kemken J, Ziegler A, Müller B (1992) Influence of Milewski M, Stinchcomb AL (2012) Estimation of maxi-
supersaturation on the pharmacodynamic effect of mum transdermal flux of nonionizedxenobiotics from
Bupranolol after dermal administration using micro- basic physicochemical determinants. Mol Pharm
emulsions as vehicle. Pharm Res 9(4):554–558 9(7):2111–2120
Kim CK, Kim J-J, Chi S-C, Shim C-K (1993) Effect of Mitragotri S, Anissimov YG, Bunge AL, Frasch HF, Guy
fatty acids and urea on the penetration of ketoprofen RH, Hadgraft J, Kasting GB, Lane ME, Roberts MS
through rat skin. Int J Pharm 99(2–3):109–118 (2011) Mathematical models of skin permeability: an
Kogan A, Garti N (2006) Microemulsions as transder- overview. Int J Pharm 418(1):115–129
mal drug delivery vehicles. Adv Colloid Interface Sci Moghimi HR, Williams AC, Barry BW (1996a) A lamel-
123–126:369–385 lar matrix model for stratum corneum intercellular
Kushla GP, Zatz JL (1991) Influence of pH on lidocaine lipids III. Effects of terpene penetration enhancers on
penetration through human and hairless mouse skin the release of 5-fluorouracil and oestradiol from the
in vitro. Int J Pharm 71(3):167–173 matrix. Int J Pharm 145(1–2):37–47
Lademann J, Knorr F, Richter H, Blume-peytavi U, Moghimi HR, Williams AC, Barry BW (1996b) A lamel-
Vogt A, Antoniou C, Sterry W, Patzelt A (2008) Hair lar matrix model for stratum corneum intercellular lip-
­follicles – an efficient storage and penetration path- ids IV. Effects of terpene penetration enhancers on the
way for topically applied substances. Skin Pharmacol permeation of 5-fluorouracil and oestradiol through
Physiol 21(3):150–155 the matrix. Int J Pharm 145(1–2):49–59
Lane ME (2013) Skin penetration enhancers. Int J Pharm Moghimi HR, Williams AC, Barry BW (1997) A lamel-
447(1–2):12–21 lar matrix model for stratum corneum intercellular
Lawrence MJ, Rees GD (2000) Microemulsion-based lipids. V. Effects of terpene penetration enhancers on
media as novel drug delivery systems. Adv Drug Deliv the structure and thermal behaviour of the matrix. Int J
Rev 45(1):89–121 Pharm 146(1):41–54
Liu X, Grice JE, Lademann J, Otberg N, Trauer S, Patzelt Most CF (1972) Co-permeant enhancement of drug trans-
A, Roberts MS (2011) Hair follicles contribute signifi- mission rates through silicone rubber. J Biomed Mater
cantly to penetration through human skin only at times Res 6(2):3–14
soon after application as a solvent deposited solid in Müller RH, Radtke M, Wissing SA (2002) Solid lipid
man. Br J Clin Pharm 72(5):768–774 nanoparticles (SLN) and nanostructured lipid carriers
Magnusson BM, Anissimov YG, Cross SE, Roberts MS (NLC) in cosmetic and dermatological preparations.
(2004) Molecular size as the main determinant of sol- Adv Drug Deliv Rev 54(Suppl 1):S131–S155
132 R. Kuswahyuning et al.

Nam SH, Xu YJ, Nam H, Jin G-w, Jeong Y, An S, Park Rhee Y-S, Huh J-Y, Park C-W, Nam T-Y, Yoon K-R, Chi
J-S (2011) Ion pairs of risedronate for transdermal S-C, Park E-S (2007) Effects of vehicles and enhanc-
delivery and enhanced permeation rate on hairless
­ ers on transdermal delivery of clebopride. Arch Pharm
mouse skin. Int J Pharm 419(1–2):114–120 Res 30(9):1155–1161
Narasimha Murthy S, Shivakumar HN (2010) Chapter 1 – Riviere JE, Brooks JD, Yeatts JL, Koivisto EL (2010)
topical and transdermal drug delivery. In: Vitthal SK Surfactant effects on skin absorption of model organic
(ed) Handbook of non-invasive drug delivery systems. chemicals: implications for dermal risk assessment
William Andrew Publishing, Boston, pp 1–36 studies. J Toxicol Environ Health A 73(11):725–737
Neubert RHH (2011) Potentials of new nanocarriers for Roberts MS, Walters KA (1998) The relationship between
dermal and transdermal drug delivery. Eur J Pharm structure and barrier function of skin. In: Roberts
Biopharm 77(1):1–2 M, Walters KA (eds) Dermal absorption and toxic-
Nicolazzo JA, Morgan TM, Reed BL, Finnin BC (2005) ity assessment, vol 2. Marcel Dekker, New York,
Synergistic enhancement of testosterone transdermal pp 1–42
delivery. J Control Release 103(3):577–585 Roberts MS, Cross S, Pellett MA (2002a) Skin transport.
Nitsche JM, Kasting GB (2008) Biophysical mod- In: Walters K (ed) Dermatological and transdermal
els for skin transport and absorption. In: Roberts formulations, vol 119. Informa Healthcare, New York,
MS, Walters KA (eds) Dermal absorption and tox- pp 89–194
icity assessment. Informa Healthcare, USA, Inc, Roberts MS, Gierden A, Riviere JE, Monteiro-Riviere NA
New York, pp 251–269 (2002b) Solvent and vehicle effects on the skin. In:
Obata Y, Takayama K, Maitani Y, Machida Y, Nagai T Roberts MS, Walters KA (eds) Dermal absorption and
(1993) Effect of ethanol on skin permeation of nonion- toxicity assessment. 2nd edition, Informa Healthcare,
ized and ionized diclofenac. Int J Pharm 89(3):191–198 New York, pp 433–447
Oliveira G, Hadgraft J, Lane ME (2012) The influence Roberts MS, Bouwstra J, Pirot F, Falson F (2008) Skin
of volatile solvents on transport across model mem- hydration – a key determinant in topical absorption. In:
branes and human skin. Int J Pharm 435(1):38–49 Walters KA, Roberts MS (eds) Dermatologic, cosme-
Ongpipattanakul B, Burnette R, Potts R, Francoeur M ceutic, and cosmetic development. Informa Healthcare,
(1991) Evidence that oleic acid exists in a separate phase New York, pp 115–128
within stratum corneum lipids. Pharm Res 8(3):350–354 Santos P, Watkinson AS, Hadgraft J, Lane ME (2008)
Ostrenga J, Steinmetz C, Poulsen B (1971) Significance Application of microemulsion in dermal and trans-
of vehicle composition I: relationship between topical dermal drug delivery. Skin Pharmacol Physiol
vehicle composition, skin penetrability, and clinical 21:246–259
efficacy. J Pharm Sci 60(8):1175–1179 Schafer-Korting M, Mehnert W, Korting HC, Fer-Korting
Pardeike J, Hommoss A, Müller RH (2009) Lipid nanoparti- M, Mehnert W, Korting H-C (2007) Lipid nanoparti-
cles (SLN, NLC) in cosmetic and pharmaceutical dermal cles for improved topical application of drugs for skin
products. Int J Pharm 366(1–2):170–184 diseases. Adv Drug Deliv Rev 59(6):427–443
Pastore MN, Kalia YN, Horstmann M, Roberts MS (2014) SchÄtzlein, Cevc (1998) Non-uniform cellular packing of
Transdermal patches: history, development and phar- the stratum corneum and permeability barrier function
macology. Br J Pharmacol (in press) of intact skin: a high-resolution confocal laser scan-
Pellett MA, Davis AF, Hadgraft J (1994) Effect of super- ning microscopy study using highly deformable ves-
saturation on membrane transport: 2. Piroxicam. Int J icles (Transfersomes). Br J Dermatol 138(4):583–592
Pharm 111(1):1–6 Scheuplein R, Blank I (1971) Permeability of the skin.
Pellett MA, Roberts MS, Hadgraft J (1997) Supersaturated Physiol Rev 51(4):702–747
solutions evaluated with an in vitro stratum corneum Schreier H, Bouwstra J (1994) Liposomes and niosomes
tape stripping technique. Int J Pharm 151(1):91–98 as topical drug carriers: dermal and transdermal drug
Peltola S, Saarinen-Savolainen P, Kiesvaara J, Suhonen delivery. J Control Release 30(1):1–15
TM, Urtti A (2003) Microemulsions for topical deliv- Seung Jin L, Tamie K-B, Sung Wan K (1987) Ion-paired
ery of estradiol. Int J Pharm 254(2):99–107 drug diffusion through polymer membranes. Int J
Pershing LK, Lambert LD, Knutson K (1990) Mechanism Pharm 39(1–2):59–73
of ethanol-enhanced estradiol permeation across Sheth NV, Freeman DJ, Higuchi WI, Spruance SL (1986)
human skin in vivo. Pharm Res 7(2):170–175 The influence of Azone, propylene glycol and polyeth-
Potts R, Guy R (1992) Predicting skin permeability. ylene glycol on in vitro skin penetration of trifluoro-
Pharm Res 9(5):663–669 thymidine. Int J Pharm 28(2–3):201–209
Poulsen BJ, Young E, Coquilla V, Katz M (1968) Effect Shokri J, Nokhodchi A, Dashbolaghi A, Hassan-Zadeh D,
of topical vehicle composition on the in vitro release Ghafourian T, Barzegar Jalali M (2001) The effect of
of fluocinolone acetonide and its acetate ester. J Pharm surfactants on the skin penetration of diazepam. Int J
Sci 57(6):928–933 Pharm 228(1–2):99–107
Prow TW, Grice JE, Lin LL, Faye R, Butler M, Becker W, Siddiqui O, Roberts MS, Polack AE (1989) Percutaneous
Wurm EMT, Yoong C, Robertson TA, Soyer HP, Roberts absorption of steroids: relative contributions of epider-
MS (2011) Nanoparticles and microparticles for skin mal penetration and dermal clearance. J Pharmacokinet
drug delivery. Adv Drug Deliv Rev 63(6):470–491 Biopharm 17(4):405–424
9  Formulation Effects in Percutaneous Absorption 133

Sloan KB, Koch SAM, Siver KG, Flowers FP (1986) Use from water-oil microemulsions. Pharm Acta Helv
of solubility parameters of drug and vehicle to predict 71(2):135–140
flux through skin. J Investig Dermatol 87(2):244–252 Trotta M, Ugazio E, Peira E, Pulitano C (2003) Influence
Smith JC, Irwin WJ (2000) Ionisation and the effect of of ion pairing on topical delivery of retinoic acid from
absorption enhancers on transport of salicylic acid microemulsions. J Control Release 86(2–3):315–321
through silastic rubber and human skin. Int J Pharm Twist J, Zatz J (1986) Influence of solvents on paraben
210(1–2):69–82 permeation through idealised skin model membranes.
Souto EB, Müller RH (2008) Cosmetic features and appli- J Soc Cosmet Chem 37(6):429–444
cations of lipid nanoparticles (SLN®, NLC®). Int J Valenta C, Schultz K (2004) Influence of carrageenan on
Cosmet Sci 30(3):157–165 the rheology and skin permeation of microemulsion
Souto EB, Almeida AJ, Müller RH (2007) Lipid nanopar- formulations. J Control Release 95(2):257–265
ticles (SLN, NLC) for cutaneous drug delivery: struc- van der Merwe D, Riviere JE (2005) Effect of vehicles
ture, protection and skin effects. J Biomed 3:317–331 and sodium lauryl sulphate on xenobiotic permeabil-
Stinchcomb A, Pirot F, Touraille G, Bunge A, Guy R ity and stratum corneum partitioning in porcine skin.
(1999) Chemical uptake into human stratum corneum Toxicology 206(3):325–335
in vivo from volatile and non-volatile solvents. Pharm Walker RB, Smith EW (1996) The role of percutaneous
Res 16(8):1288–1293 penetration enhancers. Adv Drug Deliv Rev 18(3):
Stott PW, Williams AC, Barry BW (1996) Characterization 295–301
of complex coacervates of some tricyclic antidepres- Watkinson AC, Hadgraft J, Bye A (1991) Aspects of the
sants and evaluation of their potential for enhancing transdermal delivery of prostaglandins. Int J Pharm
transdermal flux. J Control Release 41(3):215–227 74(2–3):229–236
Stott PW, Williams AC, Barry BW (1998) Transdermal Watkinson RM, Herkenne C, Guy RH, Hadgraft J,
delivery from eutectic systems: enhanced perme- Oliveira G, Lane ME (2009) Influence of ethanol on
ation of a model drug, ibuprofen. J Control Release the solubility, ionization and permeation character-
50(1–3):297–308 istics of ibuprofen in silicone and human skin. Skin
Stott PW, Williams AC, Barry BW (2001) Mechanistic Pharmacol Physiol 22(1):15–21
study into the enhanced transdermal permeation Wenkers BP, Lippold BC (2000) Prediction of the efficacy
of a model β-blocker, propranolol, by fatty acids: of cutaneously applied nonsteroidal anti-inflammatory
a melting point depression effect. Int J Pharm drugs from a lipophilic vehicle. Arzneimittelforschung
219(1–2):161–176 50(3):275–280
Suwanpidokkul N, Thongnopnua P, Umprayn K (2004) Wiechers JW, Kelly CL, Blease TG, Dederen JC (2004)
Transdermal delivery of zidovudine (AZT): the Formulating for efficacy. Int J Cosmet Sci 26(4):173–182
effects of vehicles, enhancers, and polymer mem- Wiechers JW, Watkinson AC, Cross SE, Roberts MS
branes on permeation across cadaver pig skin. AAPS (2012) Predicting skin penetration of actives from
PharmSciTech 5(3):82–89 complex cosmetic formulations: an evaluation of inter
Tata S, Weiner N, Flynn G (1994) Relative influence of eth- formulation and inter active effects during formulation
anol and propylene glycol cosolvents on deposition of optimization for transdermal delivery. Int J Cosmet Sci
minoxidil into the skin. J Pharm Sci 83(10):1508–1510 34(6):525–535
Thomas NS, Panchagnula R (2003) Transdermal delivery Williams AC, Barry BW (1989) Urea analogues in pro-
of zidovudine: effect of vehicles on permeation across pylene glycol as penetration enhancers in human skin.
rat skin and their mechanism of action. Eur J Pharm Sci Int J Pharm 56(1):43–50
18(1):71–79 Williams A, Barry B (2004) Penetration enhancers. Adv
Thong HY, Zhai H, Maibach HI (2007) Percutaneous Drug Deliv Rev 56(5):603–618
penetration enhancers: an overview. Skin Pharmacol Woodford R, Barry BW (1982) Optimization of bioavail-
Physiol 20(6):272–282 ability of topical steroids: thermodynamic control.
Touitou E, Junginger HE, Weiner ND, Nagai T, Mezei M J Investig Dermatol 79(6):388–391
(1994) Liposomes as carriers for topical and transder- Wotton PK, Møllgaard B, Hadgraft J, Hoelgaard A (1985)
mal delivery. J Pharm Sci 83(9):1189–1203 Vehicle effect on topical drug delivery. III. Effect of
Touitou E, Dayan N, Bergelson L, Godin B, Eliaz M Azone on the cutaneous permeation of metronidazole
(2000) Ethosomes – novel vesicular carriers for and propylene glycol. Int J Pharm 24(1):19–26
enhanced delivery: characterization and skin penetra- Wurster DE, Kramer SF (1961) Investigation of some fac-
tion properties. J Control Release 65(3):403–418 tors influencing percutaneous absorption. J Pharm Sci
Treffel P, Muret P, Muret-D’Aniello P, Coumes-Marquet 50(4):288–293
S, Agache P (1992) Effect of occlusion on in vitro per- Xi H, Wang Z, Chen Y, Li W, Sun L, Fang L (2012) The
cutaneous absorption of two compounds with different relationship between hydrogen-bonded ion-pair sta-
physicochemical properties. Skin Pharmacol Physiol bility and transdermal penetration of lornoxicam with
5(2):108–113 organic amines. Eur J Pharm Sci 47(2):325–330
Trotta M, Pattarino F, Gasco MR (1996) Influence of Yarosh DB (2001) Liposomes in investigative derma-
counter ions on the skin permeation of methotrexate tology. Photodermatol Photoimmunol Photomed
17(5):203–212
134 R. Kuswahyuning et al.

Yuan Y, Li S-m, Mo F-K, Zhong D-F (2006) Investigation Zhang Q, Li P, Roberts MS (2011) Maximum transepi-
of microemulsion system for transdermal delivery of dermal flux for similar size phenolic compounds is
meloxicam. Int J Pharm 321(1–2):117–123 enhanced by solvent uptake into the skin. J Control
Zhai H, Maibach HI (2002) Occlusion vs. skin barrier Release 154(1):50–57
function. Skin ResTech 8(1):1 Zhu W, Guo C, Yu A, Gao Y, Cao F, Zhai G (2009)
Zhang Q, Grice JE, Li P, Jepps OG, Wang G-J, Roberts Microemulsion-based hydrogel formulation of pen-
MS (2009) Skin solubility determines maximum tran- ciclovir for topical delivery. Int J Pharm 378(1–2):
sepidermal flux for similar size molecules. Pharm Res 152–158
26(8):1974–1985
Part III
Drug Manipulation Strategies
in Penetration Enhancement
Selection of a Proper Prodrug
for Penetration Enhancement 10
Kenneth B. Sloan, Jennifer Synovec,
and Scott C. Wasdo

Contents 10.1 Introduction


10.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . 137
10.1.1 Push Versus Pull Mechanisms
10.1.1 Push Versus Pull Mechanisms
for Penetration Enhancers . . . . . . . . . . . . . 137
for Penetration Enhancers
10.2 Basis for Prodrugs as Penetration
From a mechanistic point of view, there are two
Enhancers . . . . . . . . . . . . . . . . . . . . . . . . 138
general ways to accomplish the task of improving
10.3 Acyl Versus Soft Alkyl Promoieties . . . . 139 topical delivery using a chemical-based approach.
10.4 Mechanisms for Penetration The first approach is to increase the “push” of the
Enhancement . . . . . . . . . . . . . . . . . . . . . . 140 vehicle components on the drug to drive it into
10.4.1 Decrease Crystal Lattice Energy the skin (Kadir et al. 1987). One way to increase the
by Masking Hydrogen Bond “push” of the vehicle is to use vehicle components
Donor Functional Groups . . . . . . . . . . . . . 140 in which the drug is more soluble but which
10.4.2 Incorporation of Water Solubility are more volatile than the other components.
Enhancing Functional Groups Evaporation of the volatile components after appli-
into Promoiety . . . . . . . . . . . . . . . . . . . . . . 144
cation of the drug-vehicle combination leaves a
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149 supersaturated solution of the drug in a state of
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 149 heightened thermodynamic activity in the vehicle
(αVEH) (Coldman et al. 1969), that is, αVEH greater
than one. The second approach is to increase the
“pull” on the drug into the skin by components of
the vehicle that have permeated the skin and have
decreased the resistance of the skin to permeation
by the drug (Kadir et al. 1987) or increased the sol-
ubility of the drug in the skin, SM1: these compo-
K.B. Sloan (*) • J. Synovec nents interact with the skin. Such components of
Department of Medicinal Chemistry, the vehicle do not have to permeate the skin faster
College of Pharmacy, University of Florida,
Gainesville, FL, USA
than the drug. However, another way to increase the
e-mail: sloan@cop.ufl.edu “pull” on the drug by components of the vehicle is
S.C. Wasdo
to use components that do permeate the skin faster
Department of Anesthesiology, College of Medicine, than the drug and pull the drug along with them – a
University of Florida, Gainesville, FL, USA “drag” effect (Friend and Smedley 1993).

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 137


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_10, © Springer-Verlag Berlin Heidelberg 2015
138 K.B. Sloan et al.

The basis for the two chemical-based formulation), increasing the “pull” can be more
approaches to enhancing topical delivery (decreas- easily accomplished using a prodrug approach
ing the solubility of the drug in the vehicle and that changes the solubility properties of the drug.
increasing its solubility in the skin; “push” and A prodrug is a chemically or enzymatically
“pull,” respectively) lies in the form of the equa- reversible derivative of a parent drug that
tion that describes flux. The flux, J, of the drug improves the physicochemical or biological
through skin is directly related to the concentra- properties of the parent drug molecule to over-
tion of the drug in the first layer of the skin, CM1, come some intrinsic problem associated with its
from Fick’s Law: J = (CM1 − CMn) D/L where CMn therapeutic use: in this case, poor solubility in the
is the concentration of the drug in the last layer of skin and hence low topical delivery (Sloan 1992).
skin (and is assumed to approach zero at steady- The particular combination of functional groups
state), D is the diffusion coefficient of the drug in that is added to the parent drug is called the pro-
the skin, and L is the thickness of the membrane. moiety, and the reversible connection between
The concentration of the drug in the skin, CM1, is the promoiety and the parent drug is called the
generated from its equilibrium with the concen- enabling functional group. A prodrug approach,
tration of the drug in the vehicle, CVEH, through then, can be envisaged as a 1:1 molecular combi-
the product of its partition coefficient between the nation of the drug and a promoiety that contains
two phases KM1:VEH and CVEH. The concentration functional groups that will increase its solubility
of the drug in the skin approaches its saturated in the skin (Sloan and Wasdo 2003). This prodrug
solubility in the skin, SM1, and a thermodynamic approach stands in sharp contrast to most formu-
activity (αM1) of one when CVEH approaches the lation approaches where large molar excesses of
saturated solubility of the drug in a noninteractive penetration enhancers as vehicle components are
vehicle, SVEH; that is, αVEH also is one. The flux J is routinely needed to increase SM1 for the drug.
now the maximum possible flux from noninterac- What are the properties of the functional groups
tive vehicles, JM, and Fick’s law can be written as in the promoiety which, when added to the parent
Eq.  10.1. Regardless of the value for SVEH, the drug, could be reasonably expected to cause an
highest concentration of drug in the skin that is increase in SM1 of the resulting prodrug compared
possible from a drug applied in a noninteractive to the parent drug and hence to cause an increase in
vehicle is SM1. As SVEH increases KM1: VEH tends to its maximum flux, JM? Since it is difficult to mea-
decrease and as SVEH decreases KM1: VEH tends to sure SM1 of the prodrug, it is more convenient to
increase. SM1 can only be increased by using an measure its JM in diffusion cell experiments and
interactive component in the vehicle that changes assume, based on Fick’s law Eq. 10.1, that there is
the solubilizing capacity of the skin, the “pull,” or a direct relationship between increased JM and
by increasing the thermodynamic activity of the increased SM1. Using increases in JM as the crite-
drug in the vehicle, αVEH, so that it is greater than rion for increased SM1, it has been observed for
one, the “push,” and hence the activity of the drug quite some time that for homologous series of
in the skin, αM1, is also greater than one. more lipophilic prodrugs that the more water solu-
ble members of the series gave the greatest increase
J M = ( D / L ) ( S MI − CMn ) (10.1)
in JM and not the more lipid soluble members
(Sloan 1989, 1992; Sloan et al. 1984). In order to
account for these qualitative observations, SM1 in
Fick’s law Eq. 10.1 was expanded mathematically
10.2 B
 asis for Prodrugs to include dependence on solubility in a lipid,
as Penetration Enhancers SLIPID, and in water, SAQ. This form of Fick’s law is
the Roberts-Sloan (RS) Eq. 10.2 (Roberts and
Although increasing the “push” can be easily Sloan 1999): a transformation of the popular, but
accomplished by manipulating the components very specific, Potts-Guy (PG) Eq. 10.3 (Potts and
of the vehicle in which the drug is applied (its Guy 1992) into more general, useful terms.
10  Selection of a Proper Prodrug for Penetration Enhancement 139

Log J M = x + y log S LIPID + (1 − y ) log S AQ − zMW and SAQ where x = −1.837, y = 0.742, z = 0.00435,
and r2 = 0.86 for n = 38 (Synovec et al. 2013).
(10.2)
Thus, good fits to Eq. 10.2 are obtained regard-
less of whether the membrane is mouse, human,
Log P = x + y log K OCT:AQ − zMW (10.3)
or silicone and regardless of whether the vehicle
When a database of those homologous series is a lipid or aqueous. Since the solubilities of the
of more lipid soluble prodrugs (n = 42) comprised permeant in a lipid and in water are both neces-
of their molecular weights, MW, their solubilities sary to define maximum flux, functional groups
in isopropyl myristate (IPM), SIPM (SIPM = SLIPID in should be incorporated into the promoieties of
Eq. 10.2), and in water, SAQ, and their maximum prodrugs that can ideally increase both lipid and
fluxes from IPM through hairless mouse skin, aqueous solubilities to increase maximum flux
JMMIPM, were collected and fitted to Eq. 10.2, the and by inference SM1.
values for the coefficients were x = −0.211, The reason that increasing both lipid and
y = 0.534, z = 0.00364, and r  = 0.937 (Roberts and aqueous solubilities of the drug is important to
2

Sloan 1999). The size of the JMMIPM database has increasing its solubility in skin, and hence its
since been increased to n = 94, and the values for topical delivery, can be found in the structure of
the coefficients are now x = −0.377, y = 0.527, the barrier to topical delivery – the intercellular
z = 0.00346, and r2  = 0.900 (Majumdar et al. compartment of the stratum corneum (SC). The
2012). The maximum fluxes of prodrugs and intercellular compartment consists of lamellar
non-prodrug through human skin in vitro and double bilayers comprised of lipid components
in vivo, respectively, from mineral oil (MO), such as ceramides, cholesterol, and fatty acids
JMHMO, their solubilities in mineral oil, SMO which have polar groups attached to them. These
(SMO = SLIPID in Eq. 10.2), and in water, SAQ, and polar head groups have water associated with
their MW also gave good fit to Eq. 10.2: x = −1.83, them so that for a permeant to cross these bilayers
y = 0.462, z = 0.00153, and r2 = 0.80 for n = 30 pro- perpendicular to the axis of the bilayers, it must
drugs (Sloan et al. 2011); x = −1.459, y = 0.72, alternately cross lipid and aqueous phases (Sloan
z = 0.00013, and r2 = 0.934 for n = 10 nonsteriodal and Wasdo 2003; Sloan et al. 1984, 2011a, b).
anti-inflammatory drugs (Wenkers and Lippold Thus, a balance of solubility in both lipid and
1999; Roberts and Sloan 2001). Thus, good fits to aqueous phases by the drug (or increased lipid
Eq. 10.2 are obtained if the vehicle is a lipid (IPM and aqueous solubility by its prodrug) is neces-
or MO) and the lipid solubility of the permeant, sary for its most efficient permeation of the inter-
SLIPID, and SAQ are independent valuables. cellular compartment of the SC. The agreement
A similar strong dependence of maximum between the experimentally measurable physico-
flux through hairless mouse from water, JMMAQ, chemical parameters in the theoretically derived
on SIPM (SIPM = SLIPID in Eq. 10.2) and SAQ for some Roberts-Sloan equation and in the biochemically
of the members of the n = 94 JMMIPM database was based biphasic solubility model (Sloan et al.
observed where x = −2.30, y = 0.575, z = 0.0016, 2011a, b) for the barrier to permeation is
and r2 = 0.903 for n = 32 (Sloan et al. 2003; Wasdo encouraging.
et al. 2009). Also a strong dependence of maxi-
mum flux through human skin in vitro from
water, JMHAQ, on the solubilities of the permeants 10.3 Acyl Versus Soft Alkyl
in octanol, SOCT (SOCT = SLIPID in Eq. 10.2) and SAQ, Promoieties
was observed where x = −2.506, y = 0.538,
z = 0.00402, and r2 = 0.839 for n = 185 (Juntunen The promoieties that have been used to increase
et al. 2008). Even maximum flux through silicone lipid and aqueous solubilities can be divided into
membranes from water, JMPAQ, for some of the two types based on whether they are attached
members of the n = 94 JMMIPM database was found directly to the functional group in the parent drug
to be dependent on SIPM (SIPM = SLIPID in Eq. 10.2) that is to be modified or indirectly through a
140 K.B. Sloan et al.

­ ethylene or vinylogous methylene (aryl methy-


m of the difference in electronegativities between X and
lene) spacer (Sloan 1989, 1992; Sloan and Wasdo H. This polarized drug–X–H bond is capable of
2003). In each type, the enabling functional group forming intermolecular hydrogen bonds within the
is usually a carbonyl-type functional group crystal lattice which leads to low solubilities espe-
because of its sensitivity to cleavage by chemical cially in lipids but also frequently in water. The
or enzymatic hydrolysis. Generally these types polarization is further attenuated if an electron with-
have been referred as acyl and soft alkyl-type pro- drawing carbonyl-­type functional group is attached
moieties, respectively. Cleavage of the acyl-­type to X–H to give drug–(O = C)–X–H. Examples of
promoiety regenerates the parent drug directly this type of drug molecule, which can be measur-
while cleavage of the acyl group in the soft alkyl ably but not highly ionized at physiological pH,
promoiety generates an intermediate drug–X– include heterocycles such as 5-flurouracil (5-FU)
CHR–X′H from drug–X–CHR–X′–(C = X″)– (drug–(O = C)–NH) and 6-mercaptopurine (6-MP)
X′′′R′: X, X′, X″, and X′′′ can be O, N, or S and (drug–(S = C)–NH) which are very high melting
R and R′ can be alkyl or aryl. The intermediate is and exhibit low solubilities in both water and lipids.
designed to be intrinsically unstable and undergo In other examples such as parent drugs containing
rapid and complete chemical hydrolysis to the par- a carboxylic acid functional group (drug–(O = C)
ent drug–X–H. The advantage of the soft alkyl –OH), the functional group is so highly polarized
prodrug approach is that the stability of the pro- that it becomes highly ionized at physiological pH
drug (as well as its attendant physicochemical which does not allow it to readily cross the lipid
properties) is not limited by the functional group phase of the alternating lipid-­ aqueous phases of
in the parent drug to which it is attached. Generally, the biological barrier. An important class of drugs
changing X will change the biochemical and/or that belong to this category is the nonsteroidal anti-
pharmacological activity of the drug, but changing inflammatory drugs. Another example of this class
X′ to obtain a more or less stable or more or less are the nucleotide-­based drugs where the highly ion-
soluble prodrug will not. Of course X″ and X′′′ ized functional group is a phosphate group. Simply
can be changed in the same ways that they could masking the hydrogen bond donating abilities of the
have been if an acyl prodrug approach had been functional group by replacing the H in the drug–X–
used. H with either an acyl or soft alkyl group decreases
the melting point (mp) and increases the lipid solu-
bility (SLIPID) as well as frequently increasing the
10.4 M
 echanisms for Penetration aqueous solubility (SAQ) of the prodrug compared to
Enhancement the parent drug, especially for the shorter alkyl chain
members of a homologous series (Sloan 1989).
10.4.1 Decrease Crystal Lattice Examples of the results that can be obtained
Energy by Masking Hydrogen by masking the polar functional groups in drugs
Bond Donor Functional to increase SLIPID (SIPM) and SAQ and to increase
Groups topical delivery of the parent drug are several
prodrugs of 5-FU.
Regardless of whether the prodrug is derived from The mp, SAQ, SIPM, log partition coefficients
an acyl or soft alkyl-type promoiety, there are between IPM and pH 4.0 buffer (log KIPM:AQ), and
two general mechanisms by which both types of rates of delivery of total 5-FU containing species
promoieties can increase both lipid and aqueous through hairless mouse skin from an IPM vehicle
solubilities. The first mechanism has its basis in in vitro (JMMIPM) for four different series of pro-
decreasing the crystal lattice energy of the parent drug of 5-FU are given in Table 10.1: three acyl
drug by modifying polar groups capable of form- types and a one soft alkyl type. The first acyl type
ing intermolecular hydrogen bonds. In many if not of prodrug of 5-FU that was evaluated for its abil-
most drug molecules, the X in drug–X–H is a het- ity to increase the delivery of 5-FU was the
eroatom which causes X–H to be polarized because alkylaminocarbonyl-­5-FU (1-AAC-5-FU) ­prodrugs
10  Selection of a Proper Prodrug for Penetration Enhancement 141

Table 10.1  Prodrugs of 5-fluorouracil


Prodrugs, R = a mpb SIPMc SAQc, d Log KIPM: AQe JMMIPMf
1-AAC-5-FU
1, C1NHC = O 212 0.30 3.69 −1.09 0.208
2, C2NHC = O 180 2.79 7.76 −0.44 0.600
3, C3NHC = O 139 12.4 8.98 0.14 0.746
4, C4NHC = O 133 24.6 5.11 0.68 0.515
5, C6NHC = O 113 44.9 0.36 2.09 –
6, C8NHC = O 91 46.9 0.030 3.21 0.060
1-AOC-5-FU
7, C1OC = O 160 2.13 112 −1.72 2.62
8, C2OC = O 128 13.1 175 −1.12 5.92
9, C3OC = O 126 15.2 42.2 −0.44 2.31
10, C4OC = O 98 33.8 24.1 0.15 2.23
11, C6OC = O 67 153 4.94 1.49 1.54
12, C8OC = O 98 36.2 0.13 2.45 0.29
1-AC-5-FU
13, C1C = O 130 22.1 120 −0.73 9.3
14, C2C = O 131 36.4 47.6 −0.12 4.3
15, C3C = O 146 17.4 6.50 0.43 1.3
16, C4C = O 121 39.2 3.48 1.05 1.0
17, C5C = O 102 112.7 2.94 1.58 1.1
18, C7C = O 84 110.7 0.15 2.88 0.60
1-ACOM-5-FU
19, C1(C = O)OCH2 124 3.29 183 −1.74 2.88
20, C2(C = O)OCH2 102 9.83 167 −1.23 3.82
21, C3(C = O)OCH2 91 14.4 42.4 −0.47 2.57
22, C4(C = O)OCH2 88 14.8 12.3 0.08 1.29
23, C5(C = O)OCH2 91 14.7 2.23 0.82 0.56
24, C7(C = O)OCH2 108 9.99 0.17 1.77 0.12
5-FU, H 284 0.049 85.4g −3.24h 0.240
a
C1, C2, etc., refer to the number of carbons in alkyl chain
b
Units of °C
c
Solubilities in units of mM
d
Estimated from SIPM/KIPM:AQ
e
Partition coefficient between IPM and pH4.0 buffer at 23 ± 1 °C
f
Values for the delivery of total species containing 5-FU through hairless mouse skin from IPM in vitro in units of μmol
cm−2 h−1
g
Solubility in pH 4.0 buffer
h
Log solubility ratio between pH 4.0 buffer and IPM

(Table 10.1 and Fig. 10.1). Initially only the longer 1,000 times for 6. However, the most lipid solu-
alkyl chain members of the series were evaluated ble member evaluated, 6, gave only 0.25 times
(4–6) (Sasaki et al. 1990), but subsequently the the flux of 5-FU. None of the 1-AAC-5-FU pro-
shorter alkyl chain members (1–3) were evalu- drugs was even as soluble in water as 5-FU, and
ated, and one of them, 3, was found to give the the C3 member (3), not the shortest alkyl chain
greatest increase in the delivery of the total 5-FU member of the series (1), gave the highest SAQ
containing species, JMMIPM (Sloan et al. 1993). value: only 0.11 times SAQ for 5-FU. The C3
All of the 1-AAC-5-FU prodrugs exhibited lower member also gave the greatest increase in JMMIPM
mp than 5-FU and all of them were more soluble values for the series, albeit only three times.
in IPM than 5-FU: from 6 times for 1 to almost Thus, as predicted (Sloan 1992, 1989; Sloan and
142 K.B. Sloan et al.

HC

O C O
O

F
HN R

O N

R O

5-FU, R = H

1-24 26-29

CH3
N O
CH3

O(CH2)n N O(CH2)n N

(CH2)m (CH2)m
O
O O
H3CO H3CO

31-34 36-39

O
O OR

H3C
N
N
O OR
O
N
O N
O
H3C N
H CH3

40-46 47-54

Fig. 10.1  Chemical structures of prodrugs for compounds 1-54

Wasdo 2003; Sloan et al. 1984), for a more lipid series, and the low SAQ values can be attributed to
soluble homologous series of prodrugs, the more the fact that one of the hydrogen bond donor
water soluble member gave the highest JMMIPM functional groups, (O = C)–NH, in 5-FU was
value. The low increase in JMMIPM can be attrib- merely replaced with another hydrogen
uted to the low SAQ values exhibited by the bond donor group, N–(O = C) –NH, in the pro-
1-ACC-5-FU prodrugs compared to subsequent moiety. The potential for forming intermolecular
10  Selection of a Proper Prodrug for Penetration Enhancement 143

hydrogen bonds was not decreased significantly of 5-FU systemically was the target of topical
and the added alkyl group in the promoiety fur- delivery, then the members of the 1-AOC-5-FU
ther depressed SAQ. series performed well. On the other hand, if
The second acyl type of prodrug of 5-FU that delivery into the skin was the target, then a more
was evaluated was the alkyloxycarbonyl-5-FU rapidly hydrolyzing type of prodrug of 5-FU
(1-AOC-5-FU) prodrugs (Table 10.1, Fig. 10.1) would be required.
(Beall et al. 1994). In this series the hydrogen The third acyl type of prodrug 5-FU that was
bond donating group in the parent drug has not evaluated was the alkylcarbonyl-5-FU
been replaced with another hydrogen bond (1-AC-5-FU) prodrugs (Table 10.1, Fig. 10.1)
donating group in the promoiety so the mp are (Beall et al. 1996). The members of this series
somewhat lower than the corresponding mem- were known to hydrolyze quite rapidly
bers in the 1-AAC-5-FU series except for the C8 (t1/2 = 3–5 min), so it was expected that only 5-FU
member of the series. Consequently, the mem- would be delivered through the skin. This expec-
bers of the 1-AOC-5-FU series were also some- tation was realized, and only 5-FU and no intact
what more soluble in IPM than the members of prodrug was observed in the receptor phase after
the 1-AAC-5-FU series except for the C8 mem- application of 1-AC-5-FU prodrugs in IPM in
ber, 12; and the worst member of the series in diffusion cell experiments. All of the members of
terms of increased SIPM was 43 times instead of 6 the 1-AC series were much more soluble in IPM
times more soluble in IPM than 5-FU. However, than 5-FU (355–2,300 times), and one member,
the big difference between the two series was in C1 (13), was more soluble in water than 5-FU
the SAQ values. Not only were two members of (1.4 times). However, direct comparisons
the series more water soluble than 5-FU, 7 and 8 between the 1-AC series and either the 1-AOC or
(1.3 and 2 times, respectively), but they were all the 1-AAC series based only on the alkyl chain
more water soluble than the corresponding length in the promoiety would be misleading
members of the 1-AAC5-FU series (from 30 to without taking into account the added heteroatom
4.3 times). Thus, since the 1-AOC-5-FU series in the latter two series. For example, we will
was more soluble in lipids and in water, as pre- compare the OC1 member (7) of the 1-AOC
dicted (Sloan 1989, 1992; Sloan et al. 1984), series with the C2 member of the 1-AC series
they delivered more total 5-FU species through (14), the OC2 with the C3, the OC3 with the C4,
hairless mouse skin than the 1-AAC-5-FU series the OC4 with the C5, and the OC6 with the C7.
(from 3 to 12.5 times). Also, as predicted (Sloan Using these interseries comparisons, the mem-
1989, 1992; Sloan et al. 1984) the C2 member, 8, bers of the 1-AC series were more soluble in IPM
which was the most water soluble member of the (1.3–17 times) than those of the 1-AOC series,
series gave the greatest increase in JMMIPM com- except for 18 compared to 11. On the other hand,
pared to 5-FU (24.7 times), and not the most the members of the 1-AOC series were more
lipid soluble member of the series, 11. The next ­soluble in water (2.4–33 times) than those of the
most water soluble member, 7, gave the next 1-AC series, and as predicted (Sloan 1989, 1992;
greatest increase in JMMIPM compared to 5-FU Sloan et al. 1984) they all gave higher JMMIPM val-
(11 times). ues than the corresponding members of the 1-AC
Based on previous literature, the 1-AOC series, except for OC1, 7, versus C2, 14. Prodrug
series was expected to be more stable than the 7 was only 2.4 times more soluble in water than
1-AAC series of prodrugs of 5-FU. Whereas the 14, while 14 was 17 times more soluble in IPM
amount of intact prodrug delivered by the 1-AAC than 7. Prodrug 14 exhibited a somewhat better
series was in the 6–10 % range, the amount balance of SAQ and SIPM than 7 and gave a higher
delivered by the 1-AOC series was in the JMMIPM value (1.6 times). However, within the
40–70 % range and was up to 90 % for the best 1-AC series the C1 member, 13, which was the
performing member of the series, 8. If delivery more water soluble member of the series and not
through the skin and subsequent slower release one of the more lipid soluble members, gave the
144 K.B. Sloan et al.

greatest enhancement in JMMIPM (39 times that of 1-AC series, and their JMMIPM values were greater
5-FU). except for the comparison between 23 and 18
In the 1-AC series the effect of the mp on solu- where the JMMIPM values were equivalent. On the
bilities and ultimately on flux can be readily illus- other hand, although the members of the
trated. The C3 member of the series, 15, exhibited 1-ACOM series were less soluble in IPM than
a higher mp than either the shorter, 14, or longer the members of the 1-AOC series, in this com-
alkyl chain member, 16, and hence exhibited a parison only two members of the 1-ACOM
lower SIPM value than those members. The SAQ series, 20 and 21, were substantially more solu-
value for 15 also dropped off more rapidly than ble in water (4.0 and 1.8 times, respectively) and
expected as did its JMMIPM value. On the other hence gave greater JMMIPM values than the corre-
hand, the log K values appeared normally spaced sponding members of the 1-AOC series. In the
and the methylene π values derived from the log comparison of 19 and 8, the SAQ values were
K values only varied by 10 %: π = 0.59 ± 0.05. very close and 8 was four times more lipid solu-
Thus, log K values are no substitute for experi- ble, so 8 gave a two times greater increase in
mental solubilities for purposes of predicting JMMIPM. Similarly, 11 was 2.2 times more water
trends in JM. soluble and ten times more IPM soluble than 23,
The example of the use of a soft alkyl prodrug so 11 gave a three times greater increase in
in the designs of prodrugs to increase SIPM and JMMIPM.
SAQ and to increase the topical delivery of the Thus, the general mechanism for increasing
parent drug is also a 5-FU prodrug: the
­ lipid and aqueous solubilities of a drug by
1-­alkylcarbonyloxymethyl-5-FU (1-ACOM-­ decreasing its ability to form intermolecular
5-FU) prodrugs (Table 10.1, Fig. 10.1) (Taylor hydrogen bonds in the crystal lattice can be very
and Sloan 1998). As expected each of the effective (11–40 times enhancement of flux). But
1-ACOM-5-FU prodrugs exhibited a lower mp it is essential to evaluate the shorter alkyl chain
than 5-FU since a hydrogen bond donor group members of any series to be considered because
had been masked in the prodrug. Also as those are the members that are most likely to be
expected each was much more soluble in IPM more water soluble as well as more lipid soluble.
than 5-FU (67–302 times), and there were mem- In the examples based on 5-FU, the increases in
bers, 19 and 20, that were more soluble in water flux realized with these acyl and soft alkyl pro-
than 5-FU (2.1 and 1.9 times, respectively). As drug approaches are more than sufficient to
predicted (Sloan 1989, 1992; Sloan et al. 1984) enlarge the indicated use of topical 5-FU from
19 and 20 were the members that gave the great- treating only actinic keratoses of the scalp
est enhancement in JMMIPM (12 and 16 times, (Dillaha et al. 1965) to treating recalcitrant pso-
respectively) and not the more lipid soluble, lon- riasis on less permeable areas of the body (Tsuji
ger alkyl chain members of the series. However, and Sugai 1972).
to compare members of the 1-ACOM series with
members of any one of the 1-acyl series, the
added heteroatom and methylene spacer in the 10.4.2 Incorporation of Water
1-ACOM series needs to be taken into account. Solubility Enhancing
Thus, comparison should be made between the Functional Groups into
C1 member of the 1-ACOM series, 19, and the Promoiety
C3 member of the 1-AC, 15; or the C2 member
of the 1-AOC series, 8, the C2 member of the The second general mechanism by which acyl
1-ACOM series, 20, and the C4 member of the and soft alkyl promoieties can be used to increase
1-AC series, 16; or the C3 member of the 1-AOC the lipid and aqueous solubilities of prodrugs
series, 9, etc. Using these interseries compari- compared to their parent drugs is to incorporate
sons, the members of the 1-ACOM series were polar, water solubilizing groups into their pro-
less soluble in IPM but much more soluble in moieties. In the examples illustrating the previ-
water (15.0–48.0 times) than the members of the ous mechanism, the primary effect of the prodrug
10  Selection of a Proper Prodrug for Penetration Enhancement 145

modification was to increase lipid solubility Table 10.2  Prodrugs of levonorgestrel


because the promoiety contained only an enabling Log
functional group and a simple alkyl group. Prodrugs, R = mpa SVEHb KOCT: AQc JMHVEHd
Although large increases in SIPM were realized for 25, levonorgestrel 240 19.2 3.70 0.00019
(100)
all members of homologous series, increases in
26, C5H11 86 604
SAQ were usually modest (less than two times) (95)
and only for the shorter alkyl chain members. In 12.9 0.00058
the examples illustrating the second general (62)
mechanism, the promoiety contains an additional 27, C4H9 170 28.3 0.00026
amine, amide, ether, or diol functional group (95)
which in retrospect could have been designed 28, OCH2CH(OH) 148 30.2 3.22 0.0063
CH2OH (40)
specifically to increase SAQ. However, in most
29, O(CH2)4CH(OH) 53 396 3.75 0.0030
examples SAQ values were not available from the CH2OH (40)
original references. a
Units of °C
The first example is the use of a diol func- b
Solubilities in mixtures of ethanol:water in units of mM
tional group in the promoiety to increase the SAQ where the value in parenthesis is percentage of ethanol in
of the prodrug and hence JM for the delivery of the mixture
c
Partition coefficient between octanol and water at 24 °C
the parent drug. d
Values for delivery of total species containing levonorg-
Although the stated rationale was that more estrel from suspensions in mixtures of ethanol:water
hydrophilic prodrugs could overcome the per- (given in the SVEH column) through rat skin in vitro in units
ceived rate limiting contribution of the aqueous of μmol cm−2 h−1
viable epidermis part of the barrier to permeation
of the skin by highly lipophlic drugs (Friend
et al. 1988), the success of such prodrugs would 26 and 27 were less hydrophilic than 25. Finally,
also support a model for permeation where alter- since the flux of 25 from various ethanol and
nating lipid-aqueous barriers must be crossed in water (40–100 %) mixtures did not vary signifi-
the intercellular compartment of the SC (Sloan cantly (applications of ethanol and water mix-
et al. 1984, Sloan et al. 2011a, b. In Table 10.2 the tures did not change SM1), it can be assumed that
mp (°C), solubilities in mixtures of ethanol and delivery of total species containing 25 by the pro-
water (SVEH), log K between octanol, and pH 7.4 drugs from widely different ethanol and water
buffer (log KOCT:AQ) and fluxes of total species mixtures can be compared to the average flux
delivered from suspensions in ethanol and water generated by the application of 25 (0.00020 μmol
(VEH) through rat skin in vitro (JMHVEH) are given cm−2 h−1) in ethanol and water mixtures. Thus, 26
for the evaluation of four acyl prodrugs of and 27, which were more soluble in lipids but
levonorgestrel. estimated to be less soluble in water, gave 3 and
Two of the prodrugs in Table 10.2 (Fig. 10.1) 1.3 times greater JMHVEH values, respectively, than
were simple alkylcarbonyl prodrugs: 26 and 27. 25. Only 25 was observed in the receptor phases.
Neither was representative of the shorter alkyl By comparison, since the two prodrugs con-
chain members of the series which would have taining a diol functional group in the promoiety, 28
had the greatest potential for increased aqueous and 29, were both more soluble in an ethanol and
as well as lipid solubility. Since 26 and 27 were water mixture that was primarily aqueous in com-
both more soluble in 95 % ethanol than levonorg- position (40 % ethanol) than 25 was in 100 % etha-
estrel, 25, was soluble in 100 % ethanol, it is rea- nol, it can be reasonably assumed that 28 and 29
sonable to assume they would also be more were more soluble in water than 25. In addition,
soluble in octanol and hence be defined as more since 28 and 29 exhibit log KOCT:AQ that were com-
lipophilic than 25. Since partition coefficients for parable to that of 25 and were more soluble in
26 and 27 could not be obtained because no 26 or water than 25, it can be reasonably assumed that
27 could be measured in the aqueous phase 28 and 29 were more soluble in octanol than 25,
(while 25 could), it is reasonable to assume that that is, more lipophilic. Thus, since 28 and 29 were
146 K.B. Sloan et al.

more soluble in a lipid and in water than their par- (4 times). The more lipid soluble but less water
ent drug, as predicted (Sloan 1989, 1992; Sloan soluble members gave lower enhancement of
et al. 1984), they gave much larger increases in JMHAQ. For the naproxen series, the first member
JMHVEH than the simple alkylcarbonyl prodrugs that of the series, 36, was more soluble in water (8
were only more soluble in a lipid (31 and 15 times, times) than naproxen and was more soluble in
respectively). However, because of their greater water than the other members of the series.
stabilities as carbonate esters, they delivered Prodrug 36 was also more soluble in IPM than
mostly intact prodrug through the skin (80 and the other members of the series but none were as
96 %, respectively). soluble as naproxen. Thus, 36, which was more
The second example is the use of an amide func- soluble in lipids and water than the other mem-
tional group in the promoiety to increase SAQ of the bers of the series, gave the greatest enhancement
prodrug and hence JM for the delivery of the parent in JMHAQ (2.7 times) as would be predicted (Sloan
drug. The first report of the synthesis of a promoi- 1989, 1992; Sloan et al. 1984).
ety containing an amide functional group as part of There are two additional observations that can
an effort to increase topical delivery was for the- be made about these two series of prodrugs which
ophylline: 7-(N, N-diethysuccinamoyloxymethyl) have an amide functional incorporated into the
theophylline (Sloan and Bodor 1982). However, promoiety. First, although the SIPM values for
the prodrug was never completely evaluated. More the two series are comparable, the SAQ values for
recently 1-alkylazacycloalkan-2-one esters of indo- the naproxen series (36–39) are almost uniformly
methacin, 30 (Bonina et al. 1991), and naproxen, ten times greater than those for the indomethacin
35 (Bonina et al. 1993), have been synthesized and series (31–34), and consequently the JMHAQ val-
evaluated. ues for the naproxen series are almost uniformly
In Table 10.3 (Fig. 10.1) the values of SIPM, ten times greater. Second, although more labile
SAQ, and rates of delivery of total species soft alkyl-type prodrugs (n = 1) had been synthe-
­containing 30 or 35 from water through human sized, they were never evaluated because they
skin in vitro (JMt) are given. For the indomethacin were considered to be too labile. On the other
series, the second member of the series, 32, was hand, the n = 2 prodrugs were too stable, and only
the only member of the series that exhibited a 10–12 % of either parent drug was observed in
greater SAQ than indomethacin, and although it the receptor phases of the diffusion cell experi-
was barely as soluble in IPM as indomethacin, it ments in which they were evaluated. It would
caused the greatest enhancement of JMHEV have been interesting to have evaluated the n = 1
series of prodrugs using an IPM vehicle, in which
they would have been stable, to determine how
Table 10.3  Prodrugs of indomethacin and naproxen
effective they might have been at delivering the
Prodrugs SIPMa SAQa JMHAQb parent drug.
30, indomethacin 7.82 0.011 0.23 The third example is the use of an amine func-
31, n = 2, m = 3 6.00 0.0096 0.80 tional group in the promoiety to increase the SAQ of
32, n = 2, m = 4 7.34 0.016 0.96 the prodrug and hence JM. Again the first report of
33, n = 2, m = 5 19.0 0.012 0.77 the synthesis of a promoiety containing an amine
34, n = 2, m = 6 27.5 0.0074 0.19
functional group as part of an effort to increase the
35, naproxen 23.5 0.045 5.1
topical delivery of a parent drug was for theophyl-
36, n = 2, m = 3 21.1 0.355 13.8
line: 7-(N, N-dimethylaminoacetyloxymethyl)
37, n = 2, m = 4 18.8 0.249 8.9
38, n = 2, m = 5 16.7 0.032 4.0
theophylline (Sloan and Bodor 1982). However,
39, n = 2, m = 6 7.64 0.011 2.8 again the prodrug was never completely evaluated.
More recently the 17-(4′-dimethylaminobutyrate)
a
Solubilities in units of mM
b
Values for delivery of total species containing parent ester prodrug of testosterone was evaluated using a
drug from water through human skin in vitro in nmol 10 % solution of the prodrug in pH 7.4 buffer
cm−2 h−1 (Milosovich et al. 1993). Compared to the
10  Selection of a Proper Prodrug for Penetration Enhancement 147

d­ elivery from a suspension of testosterone in pH for SLIPID (SOCT, SMO, SIPM), SAQ, and KLIPID:AQ in the
7.4 buffer, the prodrug was 60 times more effec- literature (Bonina et al. 2001). This lack of exper-
tive at d­ elivering testosterone. Although no solu- imental solubility and K data makes it impossible
bility data were reported, a 10 % solution of the to predict changes in the solubility of the pro-
prodrug was evaluated which suggests that it is drug, attributable to the properties of the promoi-
substantially more soluble in water than testos- ety, compared to its parent in the membrane, SM1,
terone which was s­oluble only to the extent of and hence JM in Eq. 10.2.
0.004 %. The 2-diethylaminoethyl ester prodrug However, there are several examples where
of indomethacin was also evaluated by the same those experimental SLIPID and SAQ values for pro-
group (Jona et al. 1995). It was reported that the drugs containing oxyethylene groups in their pro-
prodrug drug was 3.7 times more soluble in pH moieties have been reported together with their
7.4 buffer and its partition coefficient between corresponding maximum flux values, JM. In the
octanol and pH 7.4 buffer was 6.2 times greater first example, the effect of incorporating one oxy-
than that of indomethacin so the prodrug was also ethylene group into carbonate derivatives of acet-
much more soluble in octanol (23 times). Thus, it aminophen, APAP (Fig. 10.1), on their SLIPID and
was entirely predictable (Sloan 1989, 1992; SAQ was compared with the effect of incorporat-
Sloan et al. 1984) that the prodrug gave a 4.3 ing an alkyl group into carbonate derivatives of
times enhancement in the delivery of total indo- APAP on their SLIPID and SAQ (Table 10.4) (Wasdo
methacin containing species through human skin and Sloan 2004).
in vitro. The resulting effect on experimental JMMIPM
The fourth example is the use of an ether func- was predictable based on the fit of the data to
tional group in the promoiety to increase the SAQ Eq.  10.2 (Roberts and Sloan 1999). The best
of the prodrug and hence JM. There are numerous alkyl carbonate in terms of enhancing JMMIPM was
reports in the literature where polyoxyethylene the C1 derivative, and the best oxyethylene car-
(POE) esters have been used as prodrugs to bonate was CH2CH2OCH3. Although the
enhance oral delivery (Greenwald 2001) but only CH2CH2OCH3 carbonate was equally soluble in
a few where POE esters have been used to IPM and somewhat more soluble in water than
enhance topical delivery. One of the limiting fac- the C1 carbonate, the C1 carbonate produced the
tors associated with using data from previous greater JM. The slightly better SAQ of the
reports on the use of prodrugs containing oxyeth- CH2CH2OCH3 carbonate was offset by its higher
ylene groups in their promoieties to enhance the molecular weight which was predicted by
topical delivery of their parent drugs to design Eq. 10.2 (Roberts and Sloan 1999) to reduce the
new prodrugs is the lack of experimental values value of JM. Note that the solubility ratio (SR) for

Table 10.4  Acetaminophen, APAP, prodrugs


4-AOC-APAP MW mpa Log SIPMb Log SAQb Log JMMIPMc
40, C1d 209 115 1.076 1.314 0.00
41, C2d 223 122 0.968 0.577 −0.76
42, C3d 237 106 1.375 0.427 −0.45
43, C4d 251 120 1.143 −0.377 −1.01
44, C6d 279 110 1.220 −1.328 −1.49
45, CH2CH2OCH3 253 81 1.013 1.537 −0.11
46, CH(CH3)CH2OCH3 267 123 0.529 0.516 −1.06
APAP 151 170 0.279 2.000 −0.29
a
°C
b
Units of mM
c
Units of μmole cm−2 h−1
d
C1, C2 indicates the numbers of carbons in alkyl group
148 K.B. Sloan et al.

the CH(CH3)CH2OCH3 carbonate was greater carbamate derivative was only about 0.25 times
than that for the CH2CH2OCH3 carbonate deriva- as soluble in IPM as the C3 alkyl carbamate
tive (log SR = 0.013 and −0.52, respectively), but derivative, it was 11 times more soluble in water.
it was less soluble in both IPM and water than Therefore, the JMMIPM for the (CH2CH2O)2CH3
the CH2CH2OCH3 carbonate derivative so it only carbamate derivative was about three times that
produced about one tenth the maximum flux. of the C3 alkyl derivative regardless of the nega-
This illustrates how misleading SR or K can be in tive effect of its increased molecular weight.
predicting flux and indesigning optimized topi- Among the oxyethylene carbamate derivatives,
cal products. the (CH2CH2O)2CH3 carbamate derivative was
Similarly, in the second example the effect of three times more soluble in water and 30 % more
incorporating one or two oxyethylene groups into soluble in IPM than the CH2CH2OCH3 carbamate
carbamate derivatives of theophylline, Th-H derivative so, as predicted by Eq. 10.2 (Roberts
(Fig.  10.1), on their experimental SIPM and SAQ and Sloan 1999), its JM value was about two times
values was compared with the effect of incorpo- that of the CH2CH2OCH3 carbamate derivative.
rating alkyl groups into carbamate derivatives of Although the CH(CH3)CH2OCH3 carbamate
Th-H on their SIPM and SAQ values (Table 10.5) derivative was almost two times more soluble in
(Majumdar et al. 2012). IPM, it was only 0.40 times as soluble in water as
Again the resulting effect on experimental the CH2CH2OCH3 carbamate derivative so,
JMMIPM was predicted based on the fit of the data together with its increased molecular weight, the
to Eq. 10.2 (Roberts and Sloan 1999). The best effect of its solubilities on JMMIPM led to its lower
alkyl carbamate in terms of increasing JMMIPM JMMIPM value. Again, the log SR value for the
was the C3 derivative and the best oxyethylene CH(CH3)CH2OCH3 carbamate derivative was
carbamate derivative was the (CH2CH2O)2CH3 much more positive than that of the other oxyeth-
derivative. The C3 alkyl carbamate was essen- ylene carbamate derivatives, but its JMMIPM value
tially equal in solubility in water to the C2 alkyl was lower, illustrating the misleading effect of SR
carbamate, but it was about 20 times more solu- and K in predicting flux.
ble in IPM. Therefore, the JMMIPM for the C3 alkyl In both examples, the incorporation of oxyeth-
carbamate was about four times that of the C1 ylene groups into the promoieties of prodrugs led
regardless of the negative effect of its increased to enhanced solubility properties of the prodrugs
molecular weight predicted by Eq. 10.2 (Roberts compared to their parent compounds that led to
and Sloan 1999). Although the (CH2CH2O)2CH3 higher JM values.

Table 10.5  Theophylline, Th-H, prodrugs


7-AOC-Th MW mpa Log SIPMb Log SAQb Log JMMIPMc
47, C1d 238 175 0.28 1.45 −0.54
48, C2d 252 141 0.65 1.18 −0.68
49, C3d 266 87 1.59 1.43 0.03
50, C4d 280 82 1.70 0.93 −0.19
51, C6d 294 79 1.69 −0.27 −0.82
52, CH2CH2OCH3 282 96 0.87 1.99 0.21
53, (CH2CH2O)2CH3 326 64 0.97 2.49 0.56
54, CH(CH3)CH2OCH3 296 104 1.15 1.61 −0.31
Th-H 180 170 −0.47 1.66 −0.32
a
°C
b
Units of mM
c
Units of μmole cm−2 h−1
d
C1, C2 indicates the numbers of carbons in alkyl group
10  Selection of a Proper Prodrug for Penetration Enhancement 149

Conclusion p­rodrug structure on skin permeability in vitro.


J Control Release 7:251–261
Recognizing that one of the mechanisms for Greenwald RB (2001) PEG drugs: an overview. J Control
topical penetration enhancement involves Release 74:159–171
increasing the solubility of the drug in the skin Jona JA, Dittert LW, Crooks PA, Milosovich SM,
and that prodrugs increase the delivery of Hussain AA (1995) Design of novel prodrugs for the
transdermal penetration of indomethacin. Int J Pharm
drugs into and through the skin by achieving 123:127–136
the same, then it is quite clear that prodrugs Juntunen J, Majumdar S, Sloan KB (2008) The effect of
constitute one type of penetration enhancer water solubility of solutes on their flux through human
separate from formulation approaches. An skin in vitro: a prodrug database integrated into the
extended Flynn database. Int J Pharm 351:92–103
even more powerful approach to enhancing Kadir R, Stempler D, Liron Z, Cohen S (1987) Delivery of
topical delivery would be to use combinations theophylline into excised human skin from alkanoic
of prodrugs with formulation approaches to acid solutions: a “push-pull” mechanism. J Pharm Sci
enhancing topical delivery. So far there have 76:774–779
Majumdar S, Mueller-Spaeth M, Sloan KB (2012)
been no reports of the use of such combina- Prodrugs of theophylline incorporating ethyleneoxy-
tions except for simple one-component vehi- groups in the promoiety: synthesis, characterization
cles which have obviously not been optimized and transdermal delivery. AAPS PharmSciTech
(Waranis and Sloan 1987). However, the pos- 13:853–862
Milosovich S, Hussain A, Dittert L, Aungst B, Hussain M
sibilities with the use of such a combination (1993) Testosteronyl-4-dimethylaminobutyrate HCl: a
approach would seem to be limitless. prodrug with improved skin permeation rate. J Pharm
Sci 82:227–228
Potts RO, Guy RH (1992) Predicting skin permeability.
Pharm Res 9:663–669
References Roberts WJ, Sloan KB (1999) Correlation of aqueous and
lipid solubilities with flux of prodrugs of 5-­fluorouracil,
Beall H, Prankerd R, Sloan KB (1994) Transdermal deliv- theophylline and 6-mercaptopurine: a Potts-Guy
ery of 5-fluorouracil (5-FU) through hairless mouse approach. J Pharm Sci 88:515–522
skin by 1-alkyloxycarbonyl-5-FU prodrugs: physico- Roberts WJ, Sloan KB (2001) Application of the trans-
chemical characterization of prodrugs and correlation formed Potts-Guy equation to in vivo human skin data.
with transdermal delivery. Int J Pharm 111:223–233 J Pharm Sci 90:1318–1323
Beall H, Prankerd R, Sloan KB (1996) Transdermal deliv- Sasaki H, Takahashi T, Mori Y, Nakamura J, Shibasaki J
ery of 5-fluorouracil (5-FU) by 1-alkylcarbonyl-5-FU (1990) Transdermal delivery of 5-fluorouracil and
prodrugs. Int J Pharm 129:203–210 alkylcarbamoyl derivatives. Int J Pharm 60:1–9
Bonina FP, Montenegro L, DeCapraris P, Bousquet E, Sloan KB (1989) Prodrugs for dermal delivery. Adv Drug
Tirendi S (1991) 1-Alkylazacycloalkan-2-one esters as Deliv Rev 3:67–101
prodrugs of indomethacin for improved delivery Sloan KB (1992) Functional group considerations in the
through human skin. Int J Pharm 77:21–29 development of prodrug approaches to solving topical
Bonina FP, Montenegro L, Guerrera F (1993) Naproxen delivery problems. In: Sloan KB (ed) Prodrugs: topi-
1-alkylazacycloalkan-2-one esters as dermal prodrugs: cal and ocular drug delivery. Marcel Dekker,
in vitro evaluation. Int J Pharm 100:99–105 New York, pp 17–116
Bonina FP, Puglia C, Barbuzzi T, DeCapraris P, Palagiano Sloan KB, Bodor N (1982) Hydroxymethyl and acyloxy-
F, Rimoli MG et al (2001) In vitro and in vivo evalua- methyl prodrugs of theophylline: enhanced delivery of
tion of polyoxyethylene esters as dermal prodrugs of polar drugs through skin. Int J Pharm 12:299–213
ketoprofen, naproxen and diclofenac. Eur J Pharm Sci Sloan KB, Wasdo S (2003) Designing for topical delivery:
14:123–134 prodrugs can make the difference. Med Res Rev
Coldman MF, Poulson BJ, Higuchi T (1969) Enhancement 23:763–793
of percutaneous absorption by use of volatile: nonvol- Sloan KB, Koch SAM, Siver KG (1984) Mannich base
atile systems as vehicles. J Pharm Sci 58:1098–1102 derivatives of theophylline and 5-fluorouracil: synthe-
Dillaha CJ, Jansen GT, Honeycutt WM, Holt GA (1965) ses, properties and topical delivery characteristics. Int
Further studies with topical 5-fluorouracil. Arch J Pharm 21:251–264
Dermatol 92:410–417 Sloan KB, Getz JJ, Beal HD, Prankerd R (1993)
Friend DR, Smedley SI (1993) Solvent drag in Transdermal delivery of 5-fluorouracil (5-FU) through
ethanol/ethyl acetate enhanced skin permeation of
­ hairless mouse skin by 1-alkylaminocarbonyl-5-FU
­d-­norgestrel. Int J Pharm 97:39–46 prodrugs: physicochemical characterization of pro-
Friend D, Catz P, Heller J, Reid J, Baker R (1988) drugs and correlation with transdermal delivery. Int J
Transdermal delivery of levonorgestrel II: effect of Pharm 93:27–36
150 K.B. Sloan et al.

Sloan KB, Wasdo S, Ezike-Mkparu U, Murray TJ, chemical properties and topical delivery of
Nichels D, Singh S et al (2003) Topical delivery of 5-FU. J Pharm Sci 87:15–20
5-­fluorouracil (5-FU) and 6-mercaptopurine 6-MP) by Tsuji T, Sugai T (1972) Topical administered fluorouracil
their alkylcarbonyloxymethyl (ACOM) prodrugs from in psoriasis. Arch Dermatol 105:208–212
water: vehicle effects on design of prodrugs. Pharm Waranis RP, Sloan KB (1987) The effects of vehicles
Res 20:639–645 and prodrug properties and their interactions on
Sloan KB, Devarajan-Ketha H, Wasdo SC (2011a) Dermal the delivery of 6-mercaptopurine through skin:
and transdermal delivery: prodrugs. Ther Deliv bisacyloxymethyl-­6-mercaptopurine prodrugs.
2:83–105 J Pharm Sci 76:587–595
Sloan KB, Wasdo SC, Majundar S (2011b) Topical and Wasdo SC, Sloan KB (2004) Topical delivery of a model
transdermal delivery using prodrugs. In: Rautio J (ed) phenolic drug: alkyloxycarbonyl prodrugs of acet-
Prodrugs and targeted delivery. Wiley-VCH Verlag, aminophen. Pharm Res 21:940–946
Weinheim, pp 153–179 Wasdo SC, Juntunen J, Devarajan H, Sloan KB (2009) A
Synovec J, Wasdo SC, Sloan KB (2013) The effect of lipid comparison of the fit of flux through hairless mouse
and aqueous solubilities on flux of nicotinic acid esters skin from water data to three model equations. Int
from water through silicone membrane. Drug Dev Ind J Pharm 366:65–73
Pharm 39(9):1494–1497. doi:10.3109/03639045. 2012. Wenkers BP, Lippold BC (1999) Skin penetration of non-
694590 steroidal antiinflammatory drugs out of lipophilic
Taylor HE, Sloan KB (1998) 1-Alkycarbonyloxymethyl vehicle: influence of the viable epidermis. J Pharm Sci
prodrugs of 5-fluorouracil (5-FU): syntheses, physico- 88:1326–1331
Transdermal Drug Delivery
Systems Using Supersaturation 11
Vishwas Rai and Lakshmi Raghavan

Contents 11.1 Introduction


11.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . 151
The stratum corneum (SC) is the outermost layer
11.2 Method of Preparation for
Supersaturated Solutions . . . . . . . . . . . . . 152 of the skin and acts as a primary barrier to the
permeation of chemicals across the skin. In order
11.3 Role of Anti-nucleant Polymers . . . . . . . . 155
to achieve transdermal penetration enhancement
11.4 Determination of Supersaturation across SC, physical and chemical methods are
and Crystal Growth . . . . . . . . . . . . . . . . . 156
applied for effective drug delivery. The physical
11.5 Synergistic Use of Supersaturation enhancement techniques include iontophoresis,
Along with Other Enhancement sonophoresis, electroporation, microneedle-
Technologies/Methods . . . . . . . . . . . . . . . . 157
based devices, liquid jet injectors, powder injec-
11.6 Use of Supersaturation in Other tors, ultrasound, laser radiation, radiofrequency,
Dosage Forms . . . . . . . . . . . . . . . . . . . . . . 157
magnetophoresis, and temperature. The chemical
11.7 Commercialization of Supersaturated penetration enhancement methods involve either
Transdermal Drug Delivery Systems . . . 158 disrupting the stratum corneum by fluidizing or
11.7.1 Manufacture . . . . . . . . . . . . . . . . . . . . . . . . 158
11.7.2 Storage . . . . . . . . . . . . . . . . . . . . . . . . . . . . 158 disrupting the intercellular lipids and are achieved
11.7.3 Supply Chain Management . . . . . . . . . . . . . 158 through the use of different chemical classes
11.7.4 Patient Compliance . . . . . . . . . . . . . . . . . . . 159 including sulfoxides, azone, pyrrolidones, fatty
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 159 acids, alcohols, glycols, and terpenes (Rai 2010).
The physical and chemical enhancement tech-
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 159
niques mentioned above can be expensive and
can potentially give rise to toxicity and irritation
(due to disruption in the SC integrity).
Supersaturation provides a potentially safe, inex-
pensive mechanism for penetration enhancement
of drugs. Also the use of supersaturated systems
does not interfere with the SC integrity.
V. Rai • L. Raghavan (*) A supersaturated system, by definition, is a
Department of Research and Development, system where the concentration of a chemical
DermPathe Pharmaceuticals,
compound in solution exceeds that of a saturated
108 Route 31 South, Washington, NJ 07882, USA
e-mail: raivishwas@gmail.com; solution. This system is thermodynamically unsta-
lraghavan@dermpathe.com ble and, in many cases, results in the formation

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 151


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_11, © Springer-Verlag Berlin Heidelberg 2015
152 V. Rai and L. Raghavan

of crystals spontaneously (labile state). If the Rodríguez-Hornedo and Wu 1991). It has been
compound remains in solution under supersatu- observed that the choice of solvent is critical for
rated conditions, the state is called metastable the type of crystals (e.g., hydrate, alcoholate) and
state. Because of the nature of metastable state, polymorphs formed in the solution (Corrigan and
addition of foreign particles or external forces Timoney 1974; Khoshkoo 1991). It has also been
such as ultrasound, nucleation results in crystal- observed that different polymorphs of a com-
lization. The boundary between the metastable pound can have different saturated solubilities in
and labile states is called the critical degree of a solvent and, therefore, one can be supersatu-
supersaturation (Fig. 11.1). The degree of super- rated with respect to the other. Solvate forms of
saturation (DS) is then dependent on the concen- drugs, sometimes called pseudopolymorphs, can
tration of the drug on the solution and the also have different solubilities and dissolution
temperature of the system. rates to their corresponding non-solvate forms.
The advantages of supersaturated systems for Similar to polymorphs, an anhydrous form of a
percutaneous penetration were first recognized drug can be supersaturated with respect to its
by Higuchi (Higuchi 1960) (Fig. 11.2). The role hydrate form (de Smidt et al. 1986; Davis 1993).
of supersaturation in transdermal delivery has
been tested and found effective consistently in
the past (Coldman et al. 1969; Guy 2007; Morrow 11.2 Method of Preparation
2007); however, in order to achieve supersatura- for Supersaturated Solutions
tion, the choice of optimal solvents or co-solvents
is critical (Poulsen et al. 1968). The concept and There are four primary methods of preparing the
the understanding of supersaturated systems supersaturated solutions, viz., biphasic or binary
were borne out of the crystallization theory, mixing (addition of a substance to a solution
where a solution must be first of all supersatu- which reduces the solubility of the solute); evap-
rated in order for crystals to form. The precise oration; heating and then cooling; and via chemical
mechanism of nucleation and subsequent crystal reactions (of two or more solutes to produce a new
growth is not fully understood. There are multiple compound which is less soluble in solution than
theories proposed in the literature for predicting the original starting solutes), e.g., melt extru-
crystal growth (Frank 1949; Mehta et al. 1970; sion technology, granulation. The supersaturated

n
tio
u ra
s at
p er
f Su
e eo ilit
y
gr b
e olu
lD dS
i ca ate
LABILE rit r
C atu
Concentration

SUPERSATURATED STATE

B LE
TA
T AS
Fig. 11.1 Diagram showing ME STABLE SUBSATURATED STATE
the critical degree of
supersaturation (DS) and the
different stability states of
supersaturated systems Temperature
11 Transdermal Drug Delivery Systems Using Supersaturation 153

Fig. 11.2 Higuchi’s model


of percutaneous absorption
(Higuchi 1960)

Concentration
in the outer
layer of
stratum
corneum

Concentration
in the vehicle

Concentration
in the
epidermis

Vehicle Stratum Epidermis


Corneum

solutions prepared using all the above techniques solutions and that the flux was proportional to the
have shown increased percutaneous permeation degree of saturation. It was also found that a
of drugs in the past (Adrian et al. 2002). 0.02 % supersaturated gel of hydrocortisone ace-
The preparation of biphasic mixtures can be tate was bioequivalent to a 1 % cream in reducing
explained by constructing a curve for the satu- a surfactant-induced erythema (Davis and
rated solubility of the drug in a binary co-solvent Hadgraft 1991; Davis 1993). Similar observation
system where the drug is more soluble in one of with increase in flux from increasing DS has also
the solvents than the other (Fig. 11.3). By prepar- been observed in fentanyl delivery when super-
ing a saturated solution of the drug in solvent B saturated solutions with different DS were pre-
and diluting with solvent A, a point, E, is obtained pared using PG/water and PG/ethanol systems
along the line CD, where C represents 100 % of (Santos et al. 2011). An 18-fold increase in the
solvent A and D represents 100 % solvent B. This concentration of estradiol in the stratum corneum
is a supersaturated solution and its degree of satu- was observed within ten minutes of exposure to
ration is calculated by dividing the amount of a supersaturated solution when compared with
drug in solution by its saturated solubility in the a saturated solution in the same co-solvent
same co-solvent mixture. In this example, the mixture (Megrab et al. 1995). A testosterone-
solution E has two degrees of saturation. supersaturated solution with 2.5° of supersatura-
Crystallization in a biphasic system is inhibited tion in 1/1 ethanol/PG was found to show flux
(or retarded) by addition of an anti-nucleant (across hairless rat skin) similar to that of
polymer to solvent A (Adrian 2002). metered dose transdermal spray system under
The diffusion studies of the biphasic mixture commercial development (MDTS®, Acrux Ltd.,
of water to the solutions of hydrocortisone acetate Australia, www.acrux.com.au) (Leichtnam
in propylene glycol (PG) and the anti-nucleant 2006). The delivery of piroxicam from a fourfold
polymers polyacrylate, PVP, and HPMC indi- supersaturated solution across silicone and human
cated that the supersaturated solutions gave an skin in vitro was increased four times compared
increase in flux when compared with the saturated to the drug delivery of a saturated solution in the
154 V. Rai and L. Raghavan

Fig. 11.3 Graph showing the


solubility of a drug in a
binary co-solvent system and
the method used to obtain a
supersaturated solution
D

2X E
Saturated
Solubility

100% Solvent A % Solvent 100% Solvent B

same vehicle. The solutions up to 4° of saturation evaporation is the lack of control on concentra-
were found to be stable for 16 h and then crystal- tion of the supersaturated solution thereby having
lization was observed (Pellett et al. 1997). It was less control on flux via that solution. The evapo-
also demonstrated via tape stripping technique ration theory can be tested by occluding/un-
that the intercellular regions of the stratum cor- occluding the formulation when volatile solvents
neum were capable of maintaining supersaturated are present in the vehicle (Tanaka et al. 1985).
systems as the drug is transported across the stra- Just like binary systems, control of crystallization
tum corneum (Pellett et al. 1997). In another is important to maintain the supersaturated state
study with delivery of fluocinonide across sili- of drug in solution. In a study where nifedipine
cone membranes using in vitro diffusion cells, was dissolved in combination of vehicles like
supersaturated solutions up to 3.8° of saturation isopropyl myristate, PG, and acetone and studies
were prepared using PVP and a vehicle composi- for permeation across an ethylene-vinyl acetate
tion of ethanol, water, glycerol, and propylene copolymer membrane, the initial flux values in
glycol, and it was found that fluocinonide perme- the system were found to be greater than the final
ation across silicone membranes was linearly steady-state fluxes attributing to the crystalliza-
related to degree of saturation (Schwarb et al. tion of the drug component. The incorporation of
1999). Permeation studies of ketotifen from anti-nucleant polymers into the IPM:PG:acetone
silicon-based pressure-sensitive adhesive (PSA) vehicle led to a three to five times enhancement
with n-hexane and another solvent dichlorometh- in flux (Kondo and Sugimoto 1987). These find-
ane, tetrahydrofuran, acetone, ethyl acetate, or ings of increased permeation across the lipoidal
toluene demonstrated that the formulation was in membrane were confirmed in an in vivo/in vitro
a supersaturated state (Inoue et al. 2005). correlation study where similar observations
Supersaturation can also be achieved by evap- were reported across excised male Wistar rat skin
oration of the volatile components in the formu- in vivo using the nifedipine solution in 75:25
lation resulting in decreased solubility in the ethanol and diethyl sebacate combinations
residual component of the skin surface leading to (Kondo et al. 1987). In another study, hydrocorti-
increased thermodynamic activity. However, the sone was dissolved in a mixture of acetone and
only drawback of supersaturation achieved via water, and up to 3.9° of saturation was achieved
11 Transdermal Drug Delivery Systems Using Supersaturation 155

giving a proportional increase in flux, which was have been shown to inhibit the crystal growth of
monitored across an ethylene-vinyl acetate copo- compounds like paracetamol, nifedipine, spi-
lymer membrane after the evaporation of volatile ronolactone, sulfamethiazole, hydroflumethia-
component (Theeuwes et al. 1976). The increase zide, n-paraffin, estradiol, and griseofulvin from
in delivery of sodium nonivamide acetate has supersaturated solutions in a particular solvent
also been tested across rat skin from supersatu- and delivery systems (Simonelli et al. 1970;
rated solutions prepared by evaporation of the Corrigan and Timoney 1975; Sekikawa et al.
volatile components of an aqueous ethanolic 1978; Holden 1979; Hasegawa et al. 1985; Femi-
vehicle, and two components, methyl cellulose Oyewo and Spring 1994; Kotiyan and Vavia
and hydroxypropyl cellulose, were found to have 2001; Valenta and Auner 2004). Use of copoly-
a stabilizing effect as an anti-nucleant preventing mers of methacrylic acid (Eudragit® E, EuE, and
crystallization within the skin membrane (Fang Eudragit® RL, EuRL) as ibuprofen crystal inhibi-
et al. 1999). tors in the matrices was found to prevent the drug
When a solution is prepared from cooling it to crystallization for more than 12 months (Cilurzo
a temperature below that of the saturated solubil- et al. 2005). In another study, supersaturated
ity, the solution may crystallize or a stable super- solutions of ibuprofen were prepared using 25 %
saturated system may result. A tenfold increase PG, 5 % vitamin ETPGS (d-alpha tocopheryl
in the permeation was observed when an indo- polyethylene glycol 1,000 succinate), and 5 %
methacin gel was prepared by heating a mixture ethylene oxide/propylene oxide block copolymer
of the drug with hydrogenated soybean phospho- (pluronic F127) solvents and their combinations
lipid and liquid paraffin to 95 °C, cooling to to achieve different DS (0.5, 1, 2.5, 5.0, 10.0,
room temperature, placing it in an air incubator 25.0, and 50), and it was found in that study that
at 40 °C for 3 days, and then storing at room vitamin ETPGS improved the flux better com-
temperature, compared to that prepared at room pared to PG and pluronic F127. The optimization
temperature in the same vehicle components. of the vitamin ETPGS/ibuprofen formulation
Supersaturated solutions of flurbiprofen and with polymeric stabilizers like HPMC and PVP
ketoprofen also showed an increase in the per- K-30 resulted in inhibiting crystal growth (HPMC
meation rates from the heated and cooled formu- showed better crystal growth inhibition com-
lations, but no difference in the rates for pared to that of PVP K-30). However, the use of
ibuprofen formulations was observed (Henmi PVP K-30 increased the permeation rate of drug
et al. 1994). through the skin relative to the HPMC (Ghosh
and Michniak-Kohn 2012).
How to select an appropriate polymer for a
11.3 Role of Anti-nucleant particular drug-solvent system is of a concern.
Polymers The concept of nucleation and crystallization is
not completely understood but efforts have been
Supersaturated systems, by definitions, are unsta- made to study the process using techniques like
ble in their native state. In order to stabilize a differential scanning calorimetry (DSC). In a
supersaturated system and to achieve effective study involving isothermal crystallization of lido-
transdermal enhancement, it is absolutely crucial caine (LC) in supersaturated polyacrylate
to keep the process of crystal formation to its pressure-sensitive adhesive (LC/Duro-Tak®
minimum. For this purpose, anti-nucleant poly- 87-2287 (DT2287)) system, some of the common
mers are used in the supersaturated system. Some reasons for crystallization are given (Cui and
of the common polymers used as effective anti- Frank 2005). Multiple theories have been pro-
nucleants are hydroxypropyl methyl cellulose posed for the inhibition of crystal formation by
(HPMC), polyvinyl pyrrolidone (PVP), polyvi- the use of polymer additives and therefore stabi-
nyl alcohol (PVA), polyethylene glycol (PEG), lization of supersaturated solution of a compound
and Eudragits (acrylic polymers). These polymers in a solvent. In a study involving the preparation
156 V. Rai and L. Raghavan

of supersaturated sulfamethiazole solution in or the hairless mouse skin, whereas Loftson


presence of PVP, it was suggested that PVP et al. have reported an increase in permeation of
formed a netlike structure over a growing crystal corticosterone in presence of HPβCD (Loftsson
face with fingerlike crystal growths occurring and Sigurðardóttir 1994; Shaker et al. 2003;
between the pores of the PVP network, and due Torres-Labandeira et al. 1991). Similarly,
to the curvature of these protrusions, a higher use of 2-hydroxypropyl-ß-cyclodextrin has
degree of supersaturation was required to con- also been found not to alter crystal growth in
tinue crystal growth (Simonelli et al. 1970). In ibuprofen solution (Iervolino et al. 2001).
another study involving the investigation of crys- A very similar observation was observed in
tallization and morphology of hydrocortisone a study of testosterone, added in excess to 4:1:1
acetate crystal formation, it was proposed and (v/v) ethanol/PG/water along with multiple anti-
observed via IR spectroscopy that crystal growth nucleant polymers (PVP Kollidon®30, vinyl-
was inhibited by absorption of polymer, PVP into pyrrolidone-vinyl acetate copolymer Kollidon®
the growing crystal surface through hydrogen VA64, a randomly methylated cyclodextrin
bonding (Raghavan et al. 2001). Similar observa- RAMEB, methacrylic acid and ethyl acrylate
tion of presence of hydrogen bond was also copolymer Eudragit®L100-55, nonionic poly
observed in an ibuprofen – HPMC system (ethylene oxide) polymer Polyox®WSR N-10,
(Iervolino et al. 2001). In yet another study, the hydroxypropyl methyl cellulose Methocel®E5,
preparation and stabilization of colloidal disper- hydroxypropyl methyl cellulose phthalate
sions of triclosan, an antimicrobial agent used up HPMCP®50, and hydroxypropyl cellulose
to 2 % in cosmetic and detergent formulations for Klucel®EF) to prevent crystallization. It was
disinfection of the skin, were performed from found that only Kollidon®VA64 and a cyclo-
supersaturated solutions of triclosan either in the dextrin derivative, RAMEB, at 5 % conc.,
absence or presence of the polymer, HPMC, and showed promise in the preparation of supersat-
it was observed that the particle sizes of triclosan urated solution with a degree of supersaturation
were stabilized in the range of 90–250 nm after between 1.4 and 2.6, but the supersaturated
addition of HPMC in a water-propylene glycol state existed only for 6 h (Leichtnam et al.
co-solvent system, whereas large-shaped crystal 2006).
morphology was observed in the absence of poly-
mer (Raghavan et al. 2003). Stabilization of fen-
tanyl in the supersaturated PG/water formulation
with 3DS was achieved by addition of 1 % 11.4 Determination
hydroxypropyl cellulose (HPC) leading to of Supersaturation
improved permeation flux across the skin. and Crystal Growth
Fentanyl crystal growth was retarded minimally
using HPMC or PVP K90 at 1 % (w/v) for the 5 The easiest way to determine crystal formation is
DS formulation compared with HPC (Santos via spreading the solution into a thin film on a
et al. 2011). slide and visually observing the crystals under
However, presence of a polymer in the drug the microscope. Other methods to determine the
solution does not necessarily affect drug perme- solubility of the drug and crystal formation are
ation in all cases. In a study of the effect of via methods like isothermal heat conduction,
hydroxypropyl-β-cyclodextrin (HPβCD) on microcalorimetry, X-ray diffraction, and DSC
in vitro transdermal permeation of corticoste- (Theeuwes et al. 1974; Latsch et al. 2004a, b).
rone through hairless mouse skin, contradictory Along from experimental techniques, computa-
evidences have been observed between the tional techniques like kinetic Monte Carlo
groups. Shaker et al. have been observed that method has been used for predicting of Oswald
use of HPβCD, PVP, or the HPβCD/PVP com- ripening and evolution in crystal formation/
bination does not affect the permeation of corti- growth of a supersaturated solution (Zeng et al.
costerone across synthetic cellulose membrane 2004, 2006).
11 Transdermal Drug Delivery Systems Using Supersaturation 157

11.5 Synergistic Use estradiol into the membrane whereas supersatu-


of Supersaturation Along ration was believed to increase the concentration
with Other Enhancement of compound in the vehicle beyond that of the
Technologies/Methods normally limited saturated solubility leading to
enhanced drug permeation (Megrab et al. 1995).
Synergistic effect of supersaturation along with Similarly, maintenance of supersaturated state of
the use of the chemical enhancer, oleic acid, was estradiol in a drug-adhesive patch system is
observed on the flurbiprofen permeation across important to be controlled and monitored for
human skin. The stratum corneum of human skin proper drug release from patch system. This can
was pretreated for 1 h with a 2.8 % ethanolic be achieved by use of an appropriate polymer,
(EtOH) solution of oleic acid (OA), and a super- casting solvent, drug loading, and thickness (μm)
saturated solution of flurbiprofen with six degrees of the adhesive layer (Imani et al. 2010; Jain and
of saturation was applied on the skin. Compared Banga 2010: Pattnaik et al. 2011).
to ER=1.0 with saturated solution of flurbiprofen It has been observed that it is difficult to main-
in EtOH, the 2.8 % OA in EtOH showed an ER of tain the stability of the testosterone-supersaturated
2.1, whereas the sixfold supersaturated solution solution in a mechanical aerosol delivery system.
showed an ER of 4.5, whereas in presence of both In a study involving preparation/delivery of stable
supersaturation and oleic acid, the enhancement testosterone solution in 3:1 ethanol/PG mixture
ratio (ER) of 9.9 was observed (Pellett 2012). containing a high percentage of propellant (~50 %),
Transdermal films of caffeine and sumatriptan crystallization was observed on the skin surface
have been shown to result in generation of super- from the aerosol and spray system, whereas the
saturated state of drug on the skin surface leading solution did not exhibit this problem after applica-
to increased permeation (Nicoli 2005; Femenía- tion (Leichtnam et al. 2006). In another metered
Font et al. 2006). dose aerosol (MDA) system, consisting of hydro-
Non-entrapped hydrocortisone (HC) liposomal fluoroalkane 134a, ethanol, poly(vinyl pyrrolidone)
supersaturated system has been shown to increase K90, and beclomethasone dipropionate (BDP),
the transdermal flux due to increase in thermody- with different concentration (0, 5 or 10 % w/w) of
namic activity of the drug caused by accumulation polyethylene glycol (PEG) 400, different degree of
of HC in the skin creating a supersaturated state saturation of 6 for 0 and 5 % PG and DS = 10 for
(Barichello et al. 2006). Another similar approach is 10 % PG formulation was achieved. It was found
applied in delivery of paclitaxel using paclitaxel- that the rate of drug release was controlled by the
loaded methoxy poly(ethylene glycol)-block- DS and the increased mobility of the PEG films
poly(d, l lactic acid (PEDELLA) diblock copolymer and slower supersaturation kinetics allowed a more
nanoparticles (PNPs), synthesized from polycon- sustained drug release (Reid et al. 2008). It was
densation of d,l-lactic acid and mPEG. In this observed that the formulations with higher DS
study, PNPs showed better permeation compared to show rapid recrystallization on the skin. However,
that of the supersaturated solution of paclitaxel in in a highly volatile spray system, BDP supersatura-
water. However, such approach can be combined tion for extended periods of time has been consid-
along with the supersaturation to obtain an effective ered less important than generating instantaneous,
transdermal penetration (Li et al. 2008). high levels of supersaturation to enhance drug
Similar observation was observed with estra- release (Reid et al. 2009).
diol permeation across human skin and silastic
membrane when the effect of propylene glycol-
induced enhancement was observed along with 11.6 Use of Supersaturation
the effect of supersaturated solution. Both the in Other Dosage Forms
methods increased the percutaneous permeation
of estradiol separately and showed synergism in Some of the other polymers used for prevention
enhancement when used together. Propylene gly- of crystallization in non-transdermal systems
col was believed to increase the partitioning of are 2-hydroxypropyl-ß-cyclodextrin (to prevent
158 V. Rai and L. Raghavan

crystallization of amorphous nifedipine in patient compliance, chances of irritation, etc.


spray-dried powders) (Uekama et al. 1992), and Review of the literature suggests that supersatu-
ß-cyclodextrins (for the inhibition of crystal ration as an enhancement technique has been
growth of isosorbide 5-mononitrate in tablets or evaluated intensely in the last 20+ years. Most of
powders) (Uekama et al. 1985). Hydroxypropyl the work on supersaturated systems to date has
cyclodextrins have also been used to supersatu- come from academic groups and minimal efforts
rate pancratistatin, an anticancer drug, for par- have gone into developing supersaturated sys-
enteral use (Torres-Labandeira et al. 1991). In a tems into a commercially viable product. The
study involving permeation of supersaturated main bottleneck for a product that contains the
diazepam (DZP) across polydimethylsiloxane drug in a supersaturated state is the stability of
(PDMS) membranes, chosen as an in vitro the product during (1) manufacture, (2) storage,
model for nasal mucosa, it was observed that (3) supply chain management, and (4) end use by
there was a proportional increase in DZP flux the patients.
across PDMS membrane up to 3× supersatura-
tion, and beyond this point only minor incre-
ment in flux was observed. Such supersaturated 11.7.1 Manufacture
solutions of DZP in [glycofurol (GF)/water] co-
solvent systems can result in rapid response to The process for manufacturing the product might
epileptic seizure emergencies (Hou and Siegel involve heating and cooling at different tempera-
2006). The role of excipient selection in the tures, which is conducive to nucleation. Extreme
solid dosage form formulations is also very care must be taken to control the process in order
important. Some of the important properties to avoid any nucleation. Further, mixing usually
taken into consideration for excipient selection involves high shear mixing that can cause crystal-
are molecular weight, topological polar surface lization to occur. Other processes that can induce
area (TPSA), log P, pKa, and molecular volume nucleation are packaging and analytical testing.
for screening the excipient materials. Some of
the known excipients used in the solid dosage
systems to achieve supersaturation – PEG400 in 11.7.2 Storage
solution, HPMC E5 in coated bead, hydrophilic
cyclodextrin, HPβBD in tablets, polyoxyl 40 The stability of a product under the storage con-
hydrogenated castor oil CremophorRH40, and ditions specified in the label claim of a product
tocopheryl polyethylene glycol 1,000 succinate determines its shelf life. It is known that super-
TPGS in capsule – have been tested and found saturated systems can become unstable upon
efficient in increasing the oral bioavailability of storage for long periods of time. In addition,
the formulation (Vandecruys et al. 2007). there are some formulations that require storage
at refrigerated temperatures, which can cause
thermodynamic instability. Based on the stability
11.7 Commercialization studies, a shorter shelf life might be required.
of Supersaturated
Transdermal Drug
Delivery Systems 11.7.3 Supply Chain Management

Increasing thermodynamic activity beyond satu- Following manufacture, the products have to be
ration is an attractive methodology to enhance transported from the manufacturer to the sales
penetration of actives across skin since other and distribution warehouses and to retail stores
technologies that utilize chemical and physical upon sales. Extreme care would be required to
enhancement technique might not be feasible due monitor and conduct thorough stability studies
to bulkiness of devices, higher cost, lack of under different conditions of transport and storage
11 Transdermal Drug Delivery Systems Using Supersaturation 159

in order to make sure the product is thermody- the concepts behind selection of an appropriate
namically stable under these conditions. polymer for a particular supersaturated system
in order to achieve maximum drug delivery are
still not well understood. In spite of the huge
11.7.4 Patient Compliance challenges in developing a stable supersatu-
rated product that can be commercialized,
Upon receiving a prescription, the patient can extensive research and development efforts as
potentially carry the product during travel or can well as innovative designs can exploit the
store under different conditions of temperature potential advantages of the technology.
and humidity. Extensive stability studies would
be required to provide sufficient guidance and
warnings to patients on how to store and use the
References
product. In addition, some drug molecules, when
used excessively, can cause skin irritation and Adrian D et al (2002) The application of supersaturated sys-
hence might not be practical for the use of super- tems to percutaneous drug delivery. In: Transdermal drug
saturated concentrations. delivery systems. Edited by Richard H.Guy and Jonathan
Most of these challenges can be overcome Hadgraft, CRC Press, Informa Healthcare, New York
Barichello JM et al (2006) Inducing effect of liposomal-
through proper design of product including pack- ization on the transdermal delivery of hydrocortisone:
aging. For example, the drug solution can be stored creation of a drug supersaturated state. J Control
in the stable form in a container separated from the Release 115(1):94–102
co-solvent and can be mixed at the time of use. Cilurzo F et al (2005) Polymethacrylates as crystallization
inhibitors in monolayer transdermal patches contain-
Such a methodology has been used in products ing ibuprofen. Eur J Pharm Biopharm 60(1):61–66
such as benzoyl peroxide – clindamycin gel Coldman MF et al (1969) Enhancement of percutaneous
(Benzaclin) and benzoyl peroxide-erythromycin absorption by the use of volatile: nonvolatile systems
gel (Benzamycin), where the benzyl peroxide gel as vehicles. J Pharm Sci 58(9):1098–1102
Corrigan OI, Timoney RF (1974) Anomalous behaviour
is stored in a separate jar and clindamycin or eryth- of some hydroflumethiazide crystal samples. J Pharm
romycin is compounded with the gel by the phar- Pharmacol 26(10):838–840
macist. Extensive research needs to be undertaken Corrigan OI, Timoney RF (1975) The influence of polyvi-
by the industry in order to overcome these chal- nylpyrrolidone on the dissolution properties of hydro-
flumethiazide. J Pharm Pharmacol 27(10):759–764
lenges in order to successfully bring this technol- Cui Y, Frank SG (2005) Isothermal crystallization kinet-
ogy to the product. There is a huge potential for ics of lidocaine in supersaturated lidocaine/polyacry-
this technology to be exploited with proper choice late pressure sensitive adhesive systems. J Pharm Sci
of drug, optimization of the development and 94(9):2039–2048
Davis AF, H.J. (1993) Supersaturated solutions as topical
manufacturing processes, and innovative design. drug delivery systems. Pharm Skin Penetration
Enhancement 59:243–267, J. H. KA Walters. New York,
Conclusions Marcel Dekker
Presently, it is well known that supersaturated Davis AF, Hadgraft J (1991) Effect of supersaturation on
membrane transport: 1. Hydrocortisone acetate. Int J
solutions of different chemical compounds and Pharm 76(1–2):1–8
drugs molecules have been shown to improve de Smidt JH et al (1986) Dissolution of theophylline
transdermal flux (Davis and Hadgraft 1991; monohydrate and anhydrous theophylline in buffer
Moser et al. 2001). The real challenge in main- solutions. J Pharm Sci 75(5):497–501
Fang J-Y et al (1999) Transdermal delivery of sodium
taining a supersaturated state of a chemical nonivamide acetate from volatile vehicles: effects of
compound in solution is to tailoring the solvent polymers. Int J Pharm 176(2):157–167
system and use of anti-nucleant polymers to Femenía-Font A et al (2006) Bioadhesive monolayer film
achieve the maximum thermodynamic activity for the in vitro transdermal delivery of sumatriptan. J
Pharm Sci 95(7):1561–1569
while still maintaining the compound’s stabil- Femi-Oyewo MN, Spring MS (1994) Studies on
ity in solution. Use of anti-nucleant polymers paracetamol crystals produced by growth in aqueous
has been tested and found effective; however, solutions. Int J Pharm 112(1):17–28
160 V. Rai and L. Raghavan

Frank FC (1949) The influence of dislocations on crystal Latsch S et al (2004b) Determination of the physical state
growth. Discuss Faraday Soc 5:48–54 of norethindrone acetate containing transdermal drug
Ghosh I, Michniak-Kohn B (2012) A comparative study delivery systems by isothermal microcalorimetry,
of Vitamin E TPGS/HPMC supersaturated system and X-ray diffraction, and optical microscopy. Eur J Pharm
other solubilizer/polymer combinations to enhance the Biopharm 57(2):383–395
permeability of a poorly soluble drug through the skin. Leichtnam M-L et al (2006a) Enhancement of transder-
Drug Dev Ind Pharm 38(11):1408–1416 mal testosterone delivery by supersaturation. J Pharm
Guy RH (2007) Transdermal science and technology – an Sci 95(11):2373–2379
update. Dryg Deliv Syst 22(4):442–449 Leichtnam M-L et al (2006b) Formulation and evaluation
Hasegawa A et al (1985) Physical properties of solid dis- of a testosterone transdermal spray. J Pharm Sci
persions of poorly water-soluble drugs with enteric 95(8):1693–1702
coating agents. Chem Pharm Bull (Tokyo) 33(8): Li J et al (2008) Methoxy poly(ethylene glycol)-block-
3429–3435 poly(D, L-lactic acid) copolymer nanoparticles as car-
Henmi T et al (1994) Application of an oily gel formed by riers for transdermal drug delivery. Polym Int 57(2):
hydrogenated soybean phospholipids as a percutane- 268–274
ous absorption-type ointment base. Chem Pharm Bull Loftsson T, Sigurðardóttir AM (1994) The effect of poly-
(Tokyo) 42(3):651–655 vinylpyrrolidone and hydroxypropyl methylcellulose
Higuchi T (1960) Physical chemical analysis of percuta- on HPβCD complexation of hydrocortisone and its
neous absorption process from creams and ointments. permeability through hairless mouse skin. Eur J Pharm
J Soc Cosmet Chem 11:85–87 Sci 2(4):297–301
Holden GAT, J. (1979) Inhibition of crystallization by Megrab NA et al (1995) Oestradiol permeation through
polymers. Polym Prepr Am Chem Soc Div Poly Chem human skin and silastic membrane: effects of propyl-
20:766–769 ene glycol and supersaturation. J Control Release
Hou H, Siegel RA (2006) Enhanced permeation of diaze- 36(3):277–294
pam through artificial membranes from supersaturated Mehta SC et al (1970) Rate of crystal growth of sulfathia-
solutions. J Pharm Sci 95(4):896–905 zole and methylprednisolone. J Pharm Sci 59(5):
Iervolino M et al (2001) Penetration enhancement of ibu- 638–644
profen from supersaturated solutions through human Morrow DIJ, McCarron PA, Woolfson AD, Donnelly RF
skin. Int J Pharm 212(1):131–141 (2007) Innovative strategies for enhancing topical
Imani M et al (2010) Effect of adhesive layer thickness and transdermal drug delivery. Open Drug Deliv
and drug loading on estradiol crystallization in a trans- J 1:36–59
dermal drug delivery system. AAPS PharmSciTech Moser K et al (2001) Enhanced skin permeation of a lipo-
11(3):1268–1275 philic drug using supersaturated formulations. J
Inoue K et al (2005) Enhancement of skin permeation of Control Release 73(2–3):245–253
ketotifen by supersaturation generated by amorphous Nicoli S, Colombo P, Santi P (2005) Release and perme-
form of the drug. J Control Release 108(2–3): ation kinetics of caffeine from bioadhesive transder-
306–318 mal films. AAPS J 7(1):E218–E223
Jain P, Banga AK (2010) Inhibition of crystallization in Pattnaik S et al (2011) Effect of casting solvent on crystal-
drug-in-adhesive-type transdermal patches. Int J linity of ondansetron in transdermal films. Int J Pharm
Pharm 394(1–2):68–74 406(1–2):106–110
Khoshkoo SA, J (1991) Crystallization of polymorphs: Pellett MAW, AC, Brain KR, Hadgraft J (2012) Synergism
effects of supersaturation and solvent. J Pharm between supersaturation and chemical enhancement in
Pharmacol 43:36 the permeation of Flurbiprofen through human skin.
Kondo S, Sugimoto I (1987) Enhancement of transdermal Perspectives in Percutaneous Penetration Thirteenth
delivery by superfluous thermodynamic potential. I. International Conference. La Grande Motte, France: 1–4
Thermodynamic analysis of nifedipine transport Pellett MA et al (1997a) The penetration of supersaturated
across the lipoidal barrier. J Pharmacobiodyn 10(10): solutions of piroxicam across silicone membranes and
587–594 human skin in vitro. J Control Release 46(3):205–214
Kondo S et al (1987) Enhancement of transdermal deliv- Pellett MA et al (1997b) Supersaturated solutions evalu-
ery by superfluous thermodynamic potential. II. In ated with an in vitro stratum corneum tape stripping
vitro-in vivo correlation of percutaneous nifedipine technique. Int J Pharm 151(1):91–98
transport. J Pharmacobiodyn 10(11):662–668 Poulsen BJ et al (1968) Effect of topical vehicle composi-
Kotiyan PN, Vavia PR (2001) Eudragits: role as crystalliza- tion on the in vitro release of fluocinolone acetonide
tion inhibitors in drug-in-adhesive transdermal systems and its acetate ester. J Pharm Sci 57(6):928–933
of estradiol. Eur J Pharm Biopharm 52(2):173–180 Raghavan SL et al (2001) Crystallization of hydrocorti-
Latsch S et al (2004a) Use of isothermal heat conduction sone acetate: influence of polymers. Int J Pharm
microcalorimetry, X-ray diffraction, and optical 212(2):213–221
microscopy for characterisation of crystals grown in Raghavan SL et al (2003) Formation and stabilisation of
steroid combination-containing transdermal drug deliv- triclosan colloidal suspensions using supersaturated
ery systems. Eur J Pharm Biopharm 57(2):397–410 systems. Int J Pharm 261(1–2):153–158
11 Transdermal Drug Delivery Systems Using Supersaturation 161

Rai V, Ghosh I, Bose S, Silva SMC, Chandra P, Michniak- Tanaka S et al (1985) Studies on drug release from oint-
Kohn B (2010) A transdermal review on permeation of ments. V. Release of hydrocortisone butyrate propio-
drug formulations, modifier compounds and delivery nate from topical dosage forms to silicone rubber. Int J
methods. J Drug Deliv Sci Tech 20(2):75–87 Pharm 27(1):29–38
Reid M et al (2008) Manipulation of corticosteroid release Theeuwes F et al (1974) Quantitative analytical method
from a transiently supersaturated topical metered dose for determination of drugs dispersed in polymers
aerosol using a residual miscible Co-solvent. Pharm using differential scanning calorimetry. J Pharm Sci
Res 25(11):2573–2580 63(3):427–429
Reid ML et al (2009) Transient drug supersaturation Theeuwes F et al (1976) Transference: a comprehensive
kinetics of beclomethasone dipropionate in rapidly parameter governing permeation of solutes through
drying films. Int J Pharm 371(1–2):114–119 membranes. J Membr Sci 1:3–16
Rodríguez-Hornedo N, Wu H-J (1991) Crystal growth Torres-Labandeira JJ et al (1991) Oversaturated solutions
kinetics of theophylline monohydrate. Pharm Res of drug in hydroxypropylcyclodextrins: parenteral prep-
8(5):643–648 aration of pancratistatin. J Pharm Sci 80(4):384–386
Santos P et al (2011a) Enhanced permeation of fentanyl Uekama K et al (1985) Stabilization of isosorbide
from supersaturated solutions in a model membrane. 5-mononitrate in solid state by β-cyclodextrin com-
Int J Pharm 407(1–2):72–77 plexation. Int J Pharm 25(3):339–346
Santos P et al (2011b) Formulation issues associated with Uekama K et al (1992) Inhibitory effect of
transdermal fentanyl delivery. Int J Pharm 416(1): 2-hydroxypropyl-beta-cyclodextrin on crystal-growth
155–159 of nifedipine during storage: superior dissolution and
Schwarb FP et al (1999) Effect of concentration and oral bioavailability compared with polyvinylpyrrol-
degree of saturation of topical fluocinonide formula- idone K-30. J Pharm Pharmacol 44(2):73–78
tions on in vitro membrane transport and in vivo avail- Valenta C, Auner BG (2004) The use of polymers for der-
ability on human skin. Pharm Res 16(6):909–915 mal and transdermal delivery. Eur J Pharm Biopharm
Sekikawa H et al (1978) Inhibitory effect of polyvinylpyr- 58(2):279–289
rolidone on the crystallization of drugs. Chem Pharm Vandecruys R et al (2007) Use of a screening method to
Bull (Tokyo) 26(1):118–126 determine excipients which optimize the extent and
Shaker DS et al (2003) Mechanistic studies of the effect of stability of supersaturated drug solutions and applica-
hydroxypropyl-β-cyclodextrin on in vitro transdermal tion of this system to solid formulation design. Int J
permeation of corticosterone through hairless mouse Pharm 342(1–2):168–175
skin. Int J Pharm 253(1–2):1–11 Zeng J et al (2004) Numerical simulations of crystal
Shefter E, Higuchi T (1963) Dissolution behavior of crys- growth in a transdermal drug delivery system. J Cryst
talline solvated and nonsolvated forms of some phar- Growth 262(1–4):602–611
maceuticals. J Pharm Sci 52(8):781–791 Zeng J et al (2006) Numerical study of a drug release pro-
Simonelli AP et al (1970) Inhibition of sulfathiazole crys- file in the transdermal drug delivery system. Langmuir
tal growth by polyvinylpyrrolidone. J Pharm Sci 22(3):1333–1340
59(5):633–638
Eutectic Systems for Penetration
Enhancement 12
Stuart A. Jones, Sarah Fiala, and Marc B. Brown

Contents 12.1 Introduction


12.1 Introduction. . . . . . . . . . . . . . . . . . . . . . . . 163
The skin is an extremely important organ of the
12.2 Dual Drug Diffusion. . . . . . . . . . . . . . . . . 164
body, it forms a highly restrictive barrier between
12.3 Dual Drug Partitioning. . . . . . . . . . . . . . . 165 soft human tissues and the external environment,
12.4 Eutectic Combinations. . . . . . . . . . . . . . . 166 and it maintains body fluid homeostasis. In addi-
12.4.1 Chemical Potential. . . . . . . . . . . . . . . . . . . 169 tion, it protects other internal organs from nox-
12.4.2 Association Complexes. . . . . . . . . . . . . . . . 170
ious insults. As a consequence, when breaches in
Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 171 the skin’s barrier occur, they are repaired quickly
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172 using an effective system of enzymes and immune
cells which are within close proximity to the skin
surface. If a medical condition requires treatment
using the skin as a means of drug administration,
this biological context must be understood, the
properties of the applied chemical must be char-
acterized, and the site of action of the applied
agent must be known in order to facilitate effec-
tive clinical therapy.
When a discrepancy exists in the native physi-
S.A. Jones (*)
cochemical properties displayed by a drug applied
Institute of Pharmaceutical Science, King’s College
London, 150 Stamford Street, London SE1 9NH, UK to the skin and the desired properties that theoreti-
e-mail: stuart.jones@kcl.ac.uk cally allow the agent to passively reach its
S. Fiala intended site of action, enhancement strategies
Institute of Pharmaceutical Science, King’s College may be sought to try and optimize the delivery
London, 150 Stamford Street, London SE1 9NH, UK process (Suhonen et al. 1999). A wide range of
e-mail: sarah.fiala@yahoo.co.uk
percutaneous penetration enhancement strategies
M.B. Brown have been developed over the last six decades,
School of Pharmacy, University of Hertfordshire,
and presenting a molecule to the skin as a eutectic
College Lane, Hatfield, Herts AL10 9AB, UK
mixture is one that falls within the category known
Unit 3/ Chancellor Court, MedPharm Ltd.,
as chemical enhancement techniques. A eutectic
50 Occam Road, Surrey Research Park, Guildford
GU2 7AB, UK system is most typically a binary system that
e-mail: marc.brown@medpharm.co.uk when mixed exhibits a melting point that is lower

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 163


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_12, © Springer-Verlag Berlin Heidelberg 2015
164 S.A. Jones et al.

than either of the component agents (Benson they also encounter a series of different diffu-
2005). Studies using several drugs including ibu- sional barriers which have the ability to indepen-
profen (Stott et al. 1998), propranolol (Stott et al. dently influence molecular transport. In the
2001), testosterone (Kaplun-­ Frischoff and discourse that follows, an attempt has been made
Touitou 1997), and lidocaine (Kang et al. 2000), to review the experimental studies that have
combined with a second component (which may investigated the manner in which agents are
be a drug or excipient) to form a eutectic system, transported through hydrophobic barriers when
have been shown to enhance drug penetration into applied as eutectic systems. In addition, using the
the skin compared to control systems containing lidocaine and prilocaine eutectic system as a cen-
only the single therapeutic agent. The enhanced tral theme, what appears to be the most important
permeation into the skin has been attributed to the concepts in this field have been highlighted in an
lower melting point of the combined agents, attempt to advance the current understanding of
which has been said to increase the solubility of how eutectic systems function to enhance drug
penetrants in the lipids of the skin’s outermost delivery into the skin.
layer, the stratum ­corneum (SC). EMLA cream®
(AstraZeneca, UK), a product that displays supe-
rior clinical efficacy compared to either prilocaine 12.2 Dual Drug Diffusion
or lidocaine applied alone (Juhlin et al. 1980), is
the oft-cited example used to highlight the utility The effects of applying multiple drug molecules
of the eutectic system approach, but the deliberate on the process of diffusion through membranes
formation of these systems remains an underused can be studied using porous regenerated cellulose
means to enhance drug delivery into the skin. This membranes (RCM). Regenerated cellulose is an
could be due to the challenges of formulating a inert material that when formed into a membrane
product that contains two important functional has been shown to provide a diffusion barrier
molecules. In addition to the regulatory and man- with minimal membrane-drug interactions with
ufacturing challenges posed by the use of two non-ionized agents (Reid et al. 2008; Fiala 2008).
functional components in a topical formulation, In addition, it has been found that when the polar-
the presence of two important diffusing species, ity of the application vehicle is well matched to
which can enter the body simultaneously, at dif- the membrane, the residence time of the applied
ferent rates, renders the production of new eutec- drug molecules in the membrane is short, drug
tic systems problematic. partitioning is limited, and hence the transport
According to the Higuchi equation (Higuchi rate can be directly related to the rate of molecu-
1962), the flux of a compound from a saturated lar diffusion in the membrane (Reid et al. 2008;
solution is constant, regardless of the saturated Fiala 2008). Using this basis, previous experi-
concentration in a given vehicle because all satu- mental data using RCM has been interpreted to
rated solutions have a thermodynamic activity of suggest that the simultaneous membrane diffu-
one (Twist and Zatz 1986). This theory was sion of lidocaine and prilocaine can be consid-
developed using the context of highly restrictive ered as a competitive process (Fig. 12.1, Fiala
barriers such as the skin, and it has shown to be a et al. 2008).
reliable model for a number of therapeutic deliv- Relating the correlation observed for lidocaine
ery systems (Davis and Hadgraft 1991; Dias et al. and prilocaine between the normalized drug ratio
2007; Iervolino et al. 2001). However, this math- in the applied bulk solution and the species trans-
ematical model was designed to describe the port back to the principles of the Higuchi equa-
mass transfer of a single agent. Tracking the tion allows the reduced diffusion of one agent in
movement of two agents across the human skin the presence of the second to be predicted by the
generates a three-dimensional problem as not initial composition of the applied saturated solu-
only do the molecules have the capacity to change tions using Eq. 12.1, where the rate of transport
their behavior in a ratio-dependent manner, but for an individual molecule (dq/dt) was related to
12  Eutectic Systems for Penetration Enhancement 165

the membrane surface area (A), the membrane created by the water/SC interface and into the
diffusion coefficient (D), the thermodynamic outer layers of the skin. However, when a polar
activity (α), the activity coefficient in the mem- vehicle is used to administer a compound to the
brane (γbar), the diffusion path length (L), and skin’s surface, the hydrophobic SC is sandwiched
the normalized ratio of the applied agent (N): between two hydrophilic phases. The epidermal
tissue underlying the SC contains more water and
dq ND a
=A (12.1) fewer lipids than the skin’s outermost layer, and
dt g bar L as a consequence, it is distinctly more hydro-

philic than the SC (Scheuplein and Blank 1971).
In the case of lidocaine and prilocaine combi- Therefore, even if an agent applied topically to
nations, the normalized solubility was calculated the surface of the skin passes into the SC, its sub-
using Eqs. 12.2 and 12.3, respectively: sequent transport through the tissue into deeper
layers may be retarded by a second energy barrier
Clido
N lido = (12.2) (Williams and Barry 2004). As a consequence, a
Clido + ( Cprilo × Slido / S prilo ) more detailed understanding of the transport pro-

cess can be obtained if the effects of partitioning
Cprilo into a confluent barrier from a specific applica-
N prilo = (12.3)
Cprilo + ( Clido × S prilo / Slido ) tion vehicle are recorded and compared to pre-
dicted values using transport models that employ
log P rather than relying on the latter alone.
where Sprilo, Slido was defined as the solubility of Human skin can be used in such studies, but it
prilocaine and lidocaine in individually saturated suffers from the problem of structural breakdown
solutions and Cprilo, Clido the concentrations of when long equilibration times are required to
prilocaine and lidocaine in the application solu- gain the measurements. Silicone membranes can
tions. The self-diffusion coefficient measure- be a useful alternative to human skin in partition-
ments (which do not change upon the mixing ing experiments. Many types of silicone mem-
of lidocaine and prilocaine at equimolar branes do not have the same structural
ratios ~ 7.5 × 10 −6  cm2/s) were cited in the paper stratifications as skin, but they form a continuous
by Fiala et al. (2008) to suggest the trends in the hydrophobic barrier that is relatively inert and
data were due to a reduction in the capacity of immiscible with polar solvents. They can be
the membrane to allow unhindered diffusion of sandwiched between two water phases using a
one compound in the presence of the second and Franz cell setup in vitro; thus, they can generate
not drug-drug interactions (Nyqvist-Mayer et al. transport and partitioning data using equivalent
1986). experimental parameters.
Transport studies using binary mixtures of
lidocaine and prilocaine dissolved at different
12.3 Dual Drug Partitioning ratios in water (at a pH that retains both agents in
the unionized state) have shown that the linear
Building the effects of drug partitioning into the relationship between drug transport and normal-
dual drug transport process is very important in ized applied concentration observed in the RCM
percutaneous penetration. The partition coeffi- studies (Fig. 12.1) was not conserved when the
cient (log P) of a molecule is typically measured barrier was switched to silicone (Fig. 12.2). The
as a ratio between the affinity for a standard oil reason for this was assigned to the changes that
phase (often octanol) and water. It can be used to occurred during the act of partitioning, which for
predict the ability of a molecule applied to a the confluent silicone membrane, unlike the
hydrophobic barrier to pass into it. In terms of the porous RCM, was predicted to have a significant
human skin, if the log P is high for a particular effect on the transport process (Fiala et al. 2008).
drug, it will move across the first energy barrier The data reported by Fiala et al. (2008) indicated
166 S.A. Jones et al.

Lidocaine normalised ratio in the donor solution


1 0.8 0.6 0.4 0.2 0
800 800

y = 709.68x + 16.542
Steady-state flux of prilocaine (µg cm−2 h−1)

700 R 2 = 0.9963 700

Steady-state flux of lidocaine (µg cm-2 h-1)


600 600

y = 412.61x + 7.3998
500 500
R 2 = 0.9849

400 400

300 300

200 200

100 100

0 0
0 0.2 0.4 0.6 0.8 1
Prilocaine normalised ratio in the donor solution

Fig. 12.1  Relationship between the steady-state flux of A linear trend was observed between the steady-state flux
lidocaine and prilocaine through regenerated cellulose of prilocaine and lidocaine and their solubility-normal-
membrane and their solubility-normalized ratio in phos- ized ratios in the donor solution and was plotted (repre-
phate buffer solution: prilocaine (♦), lidocaine (□). Each sented by black line) (Reproduced with permission from
point represents mean ± 1 standard deviation (n = 5). Fiala et al. (2008))

that prilocaine partitioning remained unchanged that influence of the dual drug application on par-
irrespective of the concentration of lidocaine titioning was relatively minor in comparison to
present in the binary drug mixtures, while lido- drug diffusion when an aqueous application vehi-
caine partitioning was enhanced when increasing cle was used. These findings are not in agreement
amounts of prilocaine were present in the applied with other published work that has suggested
solutions. The solubility parameters of silicone, eutectic system enhancement of drug delivery to
lidocaine, and prilocaine are 7.3 (cal cm−3)1/2, the skin was mediated through changes in the
10.68 (cal cm−3)1/2 and 11.05 (cal cm−3)1/2, partitioning process (Benson 2005; Stott et al.
­respectively (Fedors 1974). According to these 2001). This raises a question as to the particular
values, the presence of either prilocaine or lido- context in which the previous hypothesis regard-
caine in the membrane could theoretically alter ing eutectic penetration enhancement was gener-
the solubility parameter of silicone in a manner ated and the generality of the conclusions from
which may facilitate the passage of the second any of the aforementioned studies.
agent into the barrier when a binary mixture was
applied onto its surface. Experimentally, a more
efficient partitioning was only observed for 12.4 Eutectic Combinations
­lidocaine during the dual drug application pro-
cess (Fiala et al. 2008). This is difficult to logi- Previous literature has suggested that the skin
cally explain using the standard transport theories penetration enhancement effects of eutectic sys-
outlined in the literature. Furthermore, the transport tems can be attributed to superior solubility of the
data generated by Fiala et al. (2008) suggested active molecules presented by theses systems in
12  Eutectic Systems for Penetration Enhancement 167

Lidocaine normalised ratio in the donor solution


1 0.8 0.6 0.4 0.2 0
900 900

Prilocaine
800 800
y = 722.3x + 28.9
Steady-state flux of prilocaine (µg cm−2 h−1)

Lidocaine enhancement ratios R 2 = 0.991

Steady-state flux of lidocaine (µg cm−2 h−1)


700 700

1.29
600 1.08 600

500 1.21 500

400 400

300 300

200 1.14 200

100 100

0 0
0 0.2 0.4 0.6 0.8 1
Prilocaine normalised ratio in the donor solution

Fig. 12.2  Relationship between the steady-state flux of Each point represents mean ± 1 standard deviation (n = 5).
lidocaine and prilocaine through silicone membrane and Enhancement ratios of lidocaine were calculated as the
their solubility-normalized ratio in phosphate buffer solu- ratio of the actual to the theoretical steady-state flux and
tion: prilocaine (♦), lidocaine (□). Theoretical lidocaine are indicated by the numbers on the graphs (Reproduced
flux (■) was calculated assuming that the diffusion vol- with permission from Fiala et al. (2008))
ume was changing as a function of the normalized ratio.

SC lipids (Alexander et al. 2012; Kaplun-­ has a greater consequential effect on the process
Frischoff and Touitou 1997). This has been linked of drug partitioning compared to drug diffusion,
to the “melting point theory” which suggests that but a similar conclusion is difficult to substantiate
agents with a lower melting point (a key for eutectic systems. Both Pugh et al. (1996) and
­characteristic of all eutectic systems) will often Fiala (2008) suggest that eutectic systems may
have a greater propensity to associate with sol- have a negative influence on drug diffusion due to
vent molecules (Benson 2005). It has also been species competition and interaction in the solu-
suggested in the literature that once the agents tion state. This raises a question as to how, in the
delivered by eutectic systems are within the skin, context of a topical formulation applied to the
the co-­localization of the diffusing species aids surface of the skin, the issues of competitive
barrier penetration by interacting and modifying transport, molecular interactions, and facilitative
the skin structure (Watanabe et al. 2009; Woolfson partitioning function to generate the final trans-
et al. 2000). The facilitation of transport by eutec- port rate generated from different eutectic sys-
tic systems through drug-skin interaction seems tems. In an attempt to provide a critique on this
logical from both the perspective of increasing matter, there is a need to highlight the specific
drug partitioning into the barrier and facilitating details of how the various hypotheses surround-
the movement of drugs through the barrier as ing the eutectic systems have been derived.
such effects seem to be similar in nature to when One model that relates melting point to SC solu-
the skin barrier is heated (Stott et al. 1998). bility was proposed in the 1980s by Kasting et al.
According to Wood et al. (2012), heating the skin who suggested a relationship between transdermal
168 S.A. Jones et al.

flux and melting point of the drug based on the con- Stott et al. (2001) generated data to suggest
cept of ideal solution state chemistry (Kasting et al. that the Touitou model (Eq. 12.7) was in good
1987). Using this model, the ideal solubility of a agreement with the skin permeation behavior of a
drug (Sideal assuming thermodynamic ideality in beta-blocker eutectic system and from this con-
terms of intermolecular interactions) in the skin lip- cluded that the compound’s melting point was an
ids can be obtained using Eq. 12.4: important factor in eutectic system enhancement
effects. However, there are two critical details
r
Sideal = noted by Stott et al. (2001) that do not appear in
ì é DS ùü M
1 - í1 - exp ê f (Tm - T ) ú ý 1 some of the subsequent work that cites this study.
î ë RT ûþ M w First, Stott et al. (2001) showed that the correla-
(12.4) tion between melting point reduction and drug
flux enhancement using Eq. 12.7 required
where ρ is the density of the skin lipids, M1 is ­normalizing to take account of the physicochemi-
their average molecular weight, Mw is the molec- cal properties of the compounds. This process of
ular weight, Tm is the melting point in degrees normalization involved generating an enhance-
Kelvin, and ΔSf is the entropy of fusion of the ment ratio against a standard chemical in the data
drug. The ideal solubility was then used to pre- set and effectively negated any effects of the
dict a maximum flux, Jm (Eq. 12.5): eutectic combination on drug diffusion. Second,
Stott et al. (2001) attempted to use the eutectic

log ( J m / Sideal ) = 1.80 - ( 0.0216 / 2.303) M w systems in their pure form, that is, without the
addition of solvent molecules. Hence, the rela-
(12.5)
tionship between melting point and drug flux in
where Mw is the molecular weight. ΔSf shows Stott’s work was particular in this context, i.e.,
limited change as a function of melting point, but the data was derived in the absence of a tradi-
Sideal increases exponentially with decreasing tional semisolid delivery vehicle and when diffu-
melting point for any given molecular weight, sion effects were negated through a process of
and this relationship is thought to drive flux mathematical “normalization.”
across the skin, which would also theoretically In the case of the lidocaine and prilocaine
increase exponentially. Touitou et al. (1994) used eutectic system, in vitro transport studies using
melting temperatures as indices to predict the silicone membrane have shown that prilocaine-­
relative transdermal fluxes of a series of enantio- rich mixtures demonstrate superior total transport
meric eutectic mixtures. Solubility, expressed as when compared to the 1:1 composition (the latter
solute mole fraction (X) in a given solvent, was has the lowest melting point according to Brodin
related to the melting temperature (Tm) and the et al. 1984). This data was generated when lido-
enthalpy of fusion (ΔH) using Eq. 12.6: caine and prilocaine were presented to the mem-
brane in a predominantly unionized state using a
DH æ Tm - T ö pH-modified aqueous solvent (Figs. 12.1 and
ln X = - ç ÷ (12.6)
R è T × Tm ø 12.2). In addition, the TEMPE® spray (Topical

Eutectic Mixture for Premature Ejaculation), a
From this calculation, the maximum fluxes of product in Phase III development by Plethora
one pure enantiomer (Jmax, s) compared to the Solutions Plc (London, UK), uses a 3:1 lidocaine-
racemic mix (Jmax, rs) were predicted (Eq. 12.7): to-­prilocaine ratio when presenting the com-
pounds as an oil (Henry 1999; Henry and Morales
J max, s X max, s
ln = ln 2003; Henry et al. 2008). These two studies sug-
J max, rs X max, rs gest that the melting point theory does not under-
DH rs (Tm, rs - T ) DH s (Tm, s - T ) pin the delivery of molecules from all eutectic
= - systems and that the administration vehicle can
R × Tm, rs × T R × Tm, s × T
have an important influence on the permeation of
(12.7) compounds from eutectic systems. The effects of
12  Eutectic Systems for Penetration Enhancement 169

the delivery vehicle can be further exemplified liquid phase composition at room temperature
through comparison of the data generated in the has confirmed that lidocaine and prilocaine do
two papers by Fiala et al. (2008, 2011) in Fig. 12.3. not attain physical equilibrium after 24 h when
If the melting point theory cannot be used to mixed at room temperature (Fiala et al. 2011).
adequately describe the transport rate changes Previous work has also shown when either prilo-
observed from the lidocaine and prilocaine sys- caine- or lidocaine-rich ratios of the pure molten
tem, it would suggest that in the context of a oils are prepared at room temperature; a second
pharmaceutical preparation the means by which phase, composed of microparticulate matter,
eutectic systems enhance transdermal penetra- which is presumably non-melted drug product
tion of molecules is a little more complex than suspended within the eutectic system, can be
first suggested. There have been two additional observed at several ratios of the two drugs (Fiala
theoretical concepts that have been proposed to et al. 2011). It is possible that the suspension
play a role in eutectic system enhancement, these formed immediately upon mixing a eutectic sys-
are centered on how the system’s chemical poten- tem composed of only the two drugs lidocaine
tial and association of the drug molecules change and prilocaine may exhibit properties that are
when presented as topical formulation. similar to a supersaturated solution, in that a state
of heightened chemical potential may be tempo-
rarily induced. If this was the case, the thermo-
12.4.1 Chemical Potential dynamic activity of the molecule in the eutectic
mixture that was present in excess may increase
When presented as the pure molten oil, thermal transiently and thus generate an unexpectedly
analysis of the lidocaine and prilocaine eutectic high rate of membrane transport. This hypothesis
system has shown that it can take approximately is supported by the observation that eutectic com-
7 days, if stored under refrigeration, to reach a binations of ibuprofen with terpenes enhanced
state of physical equilibrium, i.e., a constant membrane transport of ibuprofen when a second
solid-liquid ratio is attained for the mixtures solid phase existed in the applied molten oil (Stott
(Brodin et al. 1984). Chemical analysis of the et al. 1998). If drug flux through human skin was

Molten Oil Aqueous Solution

JRCM, prilo = 0.52 ± 0.04 JRCM, prilo = 0.18± 0.02


JRCM, lido = 0.36 ± 0.03 JRCM, lido = 0.35± 0.04
1 1
Donor phase Donor phase
SSil, prilo = 1.29 ± 0.02 SSil, prilo = 0.69 ± 0.01
2 5 2 5
SSil, lido = 1.86 ± 0.07 SSil, lido = 1.60 ± 0.02
JSil, prilo = 0.70 ± 0.05 JSil, prilo = 0.20±0.01
3 DSil, prilo = 2.03 ± 0.36 3 DSil, prilo = 1.34 ± 0.03
JSil, lido = 1.23 ± 0.08 JSil, lido = 0.52±0.04
DSil, lido = 1.50 ± 0.21 DSil, lido = 1.44 ± 0.06
Membrane Membrane

4 4

Receptor phase Receptor phase

1 Drug release (measured using RCM, mg cm−2 h−1) 4 Partitioning into the receptor phase (governed mainly by the
concentration-gradient)
2 Partitioning (measured as solubility in silicone, % w/w)

3 Drug diffusivity through the membrane (x 10−3 cm2 h−1) 5 Overall permeation through silicone membrane (mg cm−2 h−1)

Fig. 12.3  Comparative summary of transport results from a series of studies that used a eutectic prilocaine/lidocaine
mixture at a ratio of ca. 2:3 in the form of a molten oil and an aqueous solution
170 S.A. Jones et al.

significantly influenced by the physical equili- spectroscopy evidence to show if this association
bration state of the pure oils, this may provide would be strong enough to form a pure 1:1 com-
one explanation as to why the TEMPE® and plex at equimolar levels of the two drugs or if
EMLA® systems are formulated using different there would be significant levels of free unassoci-
drug ratios. The former forms a eutectic system ated drug present in the mixtures. At ratios other
only upon its application to the skin, and thus it than the eutectic, some levels of unassociated
will most likely not reach a state of solid-liquid molecules are likely to exist, and this provides a
equilibrium during clinical use, while the latter is complex solution state chemistry landscape
produced from a pre-­ equilibrated oil mixture where several species are penetrating the mem-
free from solid particles. More experimental brane and contributing to the total steady-state
studies using human skin and a deeper theoreti- flux. It is interesting to note that silicone mem-
cal consideration as to the chemical potential of brane solubility measurements using pure molten
pure molten oils are needed to understand the lidocaine and prilocaine oils at a ratio of 1:1
influence of physical equilibrium on eutectic showed a higher lidocaine solubility compared to
enhancement, but it is possible that this could be prilocaine, which suggests that either a pure 1:1
an important factor in the development of eutec- mixture does not exist in the donor oil or the
tic systems. silicone-­lidocaine interaction is stronger than the
1:1 complex interaction.
For the lidocaine and prilocaine eutectic sys-
12.4.2 Association Complexes tem, assuming a strong 1:1 complex is formed, an
average log P for the 1:1 complex can be used
The principal components used to form a eutectic with an additive molecule mass in the Potts and
system must interact in their molten state in order Guy (1992) equation to predict the penetration
to generate their unique melting point-lowering rate of the drugs across the skin when applied as a
properties. H-bonding interactions are known to molten oil (Fig. 12.3). As only two diffusing spe-
be important in the formation of eutectic systems cies are assumed to be present (i.e., free and com-
between ibuprofen and terpenes (Stott et al. 1998) plexed), the overall permeability can be calculated
as well as between urethane and various poly- as the sum of transport of the two species relative
mers (Isama et al. 1993); however, the precise to their ratio in the mixture (Eqs. 12.8 and 12.9).
interactions between prilocaine and lidocaine
have not been identified. As noted previously, If Rprilo > Rlido:
Nyqvist-Mayer et al. (1986) suggested that in
aqueous solvents the lidocaine and prilocaine K p = ( K p , comp ) ⋅ ( 2 Rlido ) 
molecules had an equivalent self-diffusion coef- + ( K p , prilo ) ⋅ (1 − 2 Rlido ) 
ficient, and therefore it was deemed that a com- (12.8)
plex was not formed between the two molecules
in an aqueous solvent. In polar vehicles, the If Rlido > Rprilo:
molecular interactions could be influenced by
the ionization of the molecules, and hence in the K p = éë( K p, comp ) × ( 2 Rprilo ) ùû
work by Fiala et al. (2008), the drug ionization
was suppressed in order to reduce the impact of + éë( K p ,lido ) × (1 - 2 Rprilo ) ùû
(12.9)
ion-pair formation and/or molecular association.
The Nyqvist-Mayer study does not provide the where Rprilo and Rlido are prilocaine and lidocaine
details of drug ionization, and thus it is not clear w/w ratios, respectively; Kp, prilo and Kp, lido are the
what microspecies were presented in the test individual permeability coefficients of prilocaine
formulations, and this complicates the review of and lidocaine, respectively; Kp, comp is the perme-
the data in this area. Even if the association of the ability coefficient of the complex; and Kp is the
compounds was considered to occur, there is no overall permeability coefficient of the eutectic
12  Eutectic Systems for Penetration Enhancement 171

Fig. 12.4 Theoretical Kp 1.4


values for prilocaine/lidocaine
mixtures as calculated using 1.2
the Potts and Guy equation

Kp (x 106 cm sec−1)
1.0

0.8

0.6

0.4

0.2

0.0
0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1
Prilocaine:lidocaine ratio (w /w )

mixture. The overall trend of the calculated Kp Conclusions


values (Fig. 12.4) fits well with the competitive Combining two therapeutic agents in a medi-
transport hypothesis applied to the two anesthet- cine is becoming more attractive to both clini-
ics (Figs. 12.1 and 12.2). However, the Potts and cians and patients as the comorbidity rates
Guy Kp values indicate that when lidocaine was continue to increase through the twenty-first
in excess, the free lidocaine molecules drive the century. It would therefore seem sensible to
total drug permeation to be higher at high understand how the inclusion of a second
­lidocaine ratios compared to the high prilocaine agent functions to influence the properties of
ratios. This was not observed practically with the the first agent in relation to the functional
silicone membrane (Fiala et al. 2010), but the activities of the dosage form. In products that
Potts and Guy model does support the lidocaine-­ are applied to the skin, if the agents form a
rich mixture that was employed in the TEMPE® eutectic system, current literature suggests
spray. At the time of writing this chapter, there that this provides additional advantages as
are no publically available data using human eutectic systems can “enhance the penetra-
skin to suggest the ratio for the TEMPE® system tion” of topically applied medicines into the
was selected based upon experimental rather skin. The dominant discourse in the literature
than theoretical observations. In addition, the to this point seems to suggest that a eutectic
modeling performed in this discourse is system reduces the melting point of the drug
extremely simplistic, and again, as stated previ- combination and increases the transport into
ously, more transport data using human skin the skin by facilitating the act of drug parti-
needs to be generated to probe further the issues tioning. However, a number of studies, includ-
of complexation. However, what this discussion ing those which have tested the oft-cited
highlights is that drug-drug complexation must eutectic example of lidocaine and prilocaine,
be considered to be a potential factor in the man- seem to suggest that a direct link between
ner in which eutectic systems enhance skin melting point and transport cannot be applied
delivery. Furthermore, the strength of the com- to a number of contexts important to pharma-
plexes must be measured in the different applica- ceutical dosage forms.
tion vehicles in order for the nature of the The data reviewed in this chapter seems to
permeating species to be identified and allow the suggest that the ability to present actives to the
systems to be optimized. skin as a molten oil, without additional formu-
172 S.A. Jones et al.

lation excipients, is the special property that Fiala S, Brown MB, Jones SA (2011) Dynamic in-situ
underpins eutectic system’s induced skin pen- eutectic formulations. J Pharm Pharmacol
63:1428–1436
etration enhancement. When the molten mix- Henry R. (1999). Prilocaine and hydrofluorocarbon aero-
ture of the lidocaine and prilocaine eutectic sol preparations. US patent no. 5858331, 12 Jan 1999
system was specifically considered, then a Henry R, Morales A (2003) Topical lidocaine-prilocaine
two-phase mixture that was rich in the mole- spray for the treatment of premature ejaculation: a
proof of concept study. Int J Impot Res 15:277–281
cule which penetrates the barrier most effec- Henry R, Morales A, Wyllie MG (2008) TEMPE: topical
tively was shown to be the most efficient eutectic-like mixture for premature ejaculation. Expert
topical formulation and not the system with Opin Drug Deliv 5:251–261
the lowest melting point. The TEMPE® spray Higuchi WI (1962) Analysis of data on the medicament
release from ointments. J Pharm Sci 51:802–804
which uses a 3:1 lidocaine-to-­prilocaine ratio Iervolino M, Cappello B, Raghavan SL, Hadgraft J (2001)
seems to be a more logical presentation format Penetration enhancement of ibuprofen from supersatu-
for these systems compared to a traditional rated solutions through human skin. Int J Pharm
cream, e.g., EMLA®. However, further work 212:131–141
Isama K, Kojima S, Nakamura A (1993) Phase studies of
needs to be undertaken to understand how the a urethane model compound and polyether macrogly-
molecules in a eutectic system interact in the cols by infrared spectroscopy and the relationship
molten and solution states and how presenta- between eutectic composition of soft segment and
tion to the skin affects this in order to advance blood compatibility. J Biomed Mater Res 27:539–545
Juhlin L, Evers H, Broberg F (1980) A lidocaine-­
this very interesting field of research. prilocaine cream for superficial skin surgery and pain-
ful lesions. Acta Derm Venereol 60:544–546
Kang L, Jun HW, McCall JW (2000) Physicochemical
studies of lidocaine-menthol binary systems for
enhanced membrane transport. Int J Pharm
References 206:35–42
Kaplun-Frischoff Y, Touitou E (1997) Testosterone skin
Alexander A, Dwivedi SA, Giri TK, Saraf S, Tripathi DK permeation enhancement by menthol through forma-
(2012) Approaches for breaking the barriers of drug tion of eutectic with drug and interaction with skin
permeation through transdermal drug delivery. J lipids. J Pharm Sci 86:1394–1399
Control Release 164:26–40 Kasting GB, Smith RL, Cooper ER (1987) Effects of lipid
Benson HA (2005) Transdermal drug delivery: penetra- solubility and molecular size on percutaneous absorp-
tion enhancement techniques. Curr Drug Deliv tion. Pharmacol Skin 1:138–153
2:23–33 Nyqvist-Mayer AA, Brodin AF, Frank SG (1986) Drug
Brodin A, Nyqvist-Mayer A, Wadsten T, Forslund B, release studies on an oil-water emulsion based on a
Broberg F (1984) Phase diagram and aqueous solubil- eutectic mixture of lidocaine and prilocaine as the dis-
ity of the lidocaine-prilocaine binary system. J Pharm persed phase. J Pharm Sci 75:365–373
Sci 73:481–484 Potts RO, Guy RH (1992) Predicting skin permeability.
Davis AF, Hadgraft J (1991) Effect of supersaturation on Pharm Res 9:663–669
membrane transport: 1. Hydrocortisone acetate. Int J Pugh WJ, Roberts MS, Hadgraft J (1996) Epidermal per-
Pharm 76:1–8 meability – penetrant structure relationships: 3. The
Dias M, Hadgraft J, Lane ME (2007) Influence of effect of hydrogen bonding interactions and molecular
membrane-­solvent-solute interactions on solute per- size on diffusion across the stratum corneum. Int J
meation in model membranes. Int J Pharm Pharm 138:149–165
336:108–114 Reid ML, Brown MB, Moss GP, Jones SA (2008) An
Fedors RF (1974) Method for estimating both the solubil- investigation into solvent-membrane interactions
ity parameters and molar volumes of liquids. Polym when assessing drug release from organic vehicles
Eng Sci 14:147–154 using regenerated cellulose membranes. J Pharm
Fiala S, Brown MB, Jones SA (2008) An investigation Pharmacol 60:1139–1147
into the influence of binary drug solutions upon diffu- Scheuplein RJ, Blank IH (1971) Permeability of the skin.
sion and partition processes in model membranes. J Physiol Rev 51:702–747
Pharm Pharmacol 60:1615–1623 Stott PW, Williams AC, Barry BW (1998) Transdermal
Fiala S, Brown MB, Jones SA (2010) A fundamental delivery from eutectic systems: enhanced permeation
investigation into the effects of eutectic formation on of a model drug, ibuprofen. J Control Release
transmembrane transport. Int J Pharm 393:68–73 50:297–308
12  Eutectic Systems for Penetration Enhancement 173

Stott PW, Williams AC, Barry BW (2001) Mechanistic Watanabe H, Obata Y, Ishida K et al (2009) Effect of
study into the enhanced transdermal permeation of a l-menthol on the thermotropic behavior of ceramide 2/
model beta-blocker, propranolol, by fatty acids: a cholesterol mixtures as a model for the intercellular lip-
melting point depression effect. Int J Pharm 219: ids in stratum corneum. Colloids Surf B 73:116–121
161–176 Williams AC, Barry BW (2004) Penetration enhancers.
Suhonen TM, Bouwstra JA, Urtti A (1999) Chemical Adv Drug Deliv Rev 56:603–618
enhancement of percutaneous absorption in relation to Wood DG, Brown MB, Jones SA (2012) Understanding
stratum corneum structural alterations. J Control heat facilitated drug transport across human epider-
Release 59:149–161 mis. Eur J Pharm Biopharm 81:642–649
Touitou E, Chow DD, Lawter JR (1994) Chiral β-blockers Woolfson AD, Malcolm RK, Campbell K, Jones DS,
for transdermal delivery. Int J Pharm 104:19–28 Russell JA (2000) Rheological, mechanical and mem-
Twist JN, Zatz JL (1986) Influence of solvents on paraben brane penetration properties of novel dual drug sys-
permeation through idealized skin model membranes. tems for percutaneous delivery. J Contr Release
J Soc Cosmet Chem 37:291 67:395–408
Formation of Ion Pairs
and Complex Coacervates 13
Liang Fang, Honglei Xi, and Dongmei Cun

Contents 13.1 Ion-Pair Formation


13.1 I on-Pair Formation for  for Penetration
Penetration Enhancement . . . . . . . . . . . 175 Enhancement
13.1.1 The Concept of Ion Pairs . . . . . . . . . . . . . 176
13.1.2 Molecules Suitable for Ion-­Pair A diffusing molecule has to cross the skin barrier
Formation . . . . . . . . . . . . . . . . . . . . . . . . . 176
13.1.3 The Confirmation of Ion-Pair composed of the highly lipophilic stratum cor-
Formation . . . . . . . . . . . . . . . . . . . . . . . . . 177 neum (SC) and the hydrophilic viable epidermis
13.1.4 The Effect of Ion-Pair Formation (ED) in order to reach the deep skin layer as well
on Skin Permeation . . . . . . . . . . . . . . . . . . 182 as the layers under the skin. Therefore, only
13.1.5 Ion-Pair Formation vs. Penetration
Enhancers . . . . . . . . . . . . . . . . . . . . . . . . . 184 active compounds with ideal physicochemical
properties, e.g., low molecular weight, suitable
13.2  omplex Coacervates for 
C
Penetration Enhancement . . . . . . . . . . . 185
solubility in oil and water, moderate partition
coefficient, and low melting point, can permeate
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186
through both the lipid and polar microenviron-
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186 ments in the skin (Barry 2001). Obviously,
hydrophilic ionized drugs do not readily distrib-
ute into or penetrate through the lipophilic SC
membranes. On the contrary, the problem for
those very lipophilic drugs, which can easily dis-
tribute into SC membranes, is that they do not
readily translocate from the SC to the relative
hydrophilic ED due to their high solubility in the
intercellular lipids, thus limiting their skin per-
meation. Ion pairing provides a possible approach
to adjust the physicochemical properties of drugs
L. Fang (*) • H. Xi • D. Cun without any changes on the structure and phar-
Department of Pharmaceutical Sciences, macologic actions of the drug compound and
Shenyang Pharmaceutical University, consequently facilitate the penetration of drugs
No.103 Wenhua Road, Shenyang,
across the skin barrier (Neubert 1989). Some
Liaoning 110016, China
e-mail: fangliang2003@yahoo.com; products using ion-pair technique, such as
xihonglei518@gmail.com; cundongmei@163.com Flector® Patch (diclofenac epolamine topical

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 175


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_13, © Springer-Verlag Berlin Heidelberg 2015
176 L. Fang et al.

patch) from IBSA Institut Biochimique SA it has not been paid extensive attention yet com-
(Switzerland) and Voltarol® emulgel (diclofenac pared to the classical ion pairs mentioned above.
diethylamine emulgel) from Novartis Consumer Hydrogen-bonded ion pairs are a special type
Health UK Ltd. (UK) are commercially of ion pairs. The concept of hydrogen-bonded ion
available. pairs was brought out for better understanding
the nature of the protonic acid–base interaction
occurring in non-dissociating solvents (Barrow
13.1.1 The Concept of Ion Pairs 1956). The interaction between sufficiently
strong protonic acids and bases in a solvent envi-
An ion pair is defined as neutral species which ronment, especially in solvents with low dielec-
consists of a pair of oppositely charged ions held tric constant, tends to promote the formation of
together by coulomb attraction without formation ion pairs accompanied by proton transfer of a
of a covalent bond (McNaught and Wilkinson hydrogen bond. According to the Brønsted-­
1997). Practically, an ion pair behaves as one unit Lowry concept of acids and bases in aprotic sol-
in determining conductivity, kinetic behavior, vents, two forms of simple prototropic
osmotic properties, etc. equilibrium exist as follows (Hudson et al. 1972):
In 1926, Bjerrum developed a theory that took
the interaction of ions into account. He intro-
duced, for the first time, the concept of ion pairs
and showed how the mass action constant of the
equilibrium between ions and ion pairs is depen-
dent on the dielectric constant of the solvent as
well as on temperature and the size of the ions
(Kraus 1956). According to the Bjerrum theory, The classical electrostatic attraction was pre-
oppositely charged ions with their centers closer dominant in weak hydrogen bonds, so-called
together than a distance (q) (pm) are considered classical hydrogen bonds (Equilibrium I).
to constitute an ion pair: However, on going to stronger hydrogen bonds,
the contribution of the proton transfer led to the
q = 8.36 × 106 z + z − / ( e r T ) (13.1) formation of ion pairs, so-called hydrogen-­

bonded ion pairs (Equilibrium II) (Arunan et al.
Where z+ and z− are the charge numbers of the 2011; Ratajczak 1972). Such a hydrogen bond is
ions, εr is the relative dielectric constant of extremely polarizable. The formation of a
the medium and T is the absolute temperature. hydrogen-­bonded ion pair between a carboxylic
The resulting ion pairs exhibit stable, thermody- acid and a pyridine base in benzene solution is a
namically distinct species. Depending on the typical example of acid and base interaction
strength of the solvent–ion interactions, ion pairs (Barrow 1956).
can be classified into two types: contact (tight,
intimate) and solvent-separated (loose) ion pairs
(Nagy and Takacs-Novak 2000). 13.1.2 Molecules Suitable for Ion-­
In polar solvents with high dielectric constant Pair Formation
such as water, ion-pair formation is also possibly
obtained. Diamond (1963) demonstrated that the Both drugs and counter ions have to meet cer-
existing of so-called hydrophobic ion pairing tain demands in order to form ion pairs success-
(HIP). HIP is composed of two large hydropho- fully. For hydrophilic ionized drugs, Neubert
bic ions self-assembled together by coulomb (1989) pointed out that the ideal counter ions
attraction, hydrophobic forces, and hydrogen needed to possess high lipophilicity, sufficient
bonding in polar solvents. Even though the HIP solubility in physiological compatibility, and
complexes display enhanced lipophilicity and metabolic stability, which was suitable for ion-
thus are more suited for their potential a­ pplication, pair formation and crossing lipid membranes in
13  Formation of Ion Pairs and Complex Coacervates 177

the form of ion pairs. The ion pairs formed by spectroscopy (ESR), and X-ray crystallography
quaternary ammonium drugs and organic anions could provide insights into ion-pair formation.
are typical examples of ion-pair formation IR and NMR spectroscopy often offer exper-
(Takacs-Novak and Szasz 1999). Similarly, imental proofs to directly indicate the forma-
Miller et al. (2009) performed a study in which tion of ion pairs, and they are especially
three lipophilic acidic counter ions were pronounced for hydrogen-bonded ion pairs
employed to give an understanding of the mech- (Barthel and Deser 1994; Biliškov et al. 2011;
anism of ion-pair mediated membrane transport Habeeb 1997; Pregosin 2009). Recently, by
of low permeable drugs. using IR and chemical exchange two-dimensional
Besides ionized species, for stronger hydro- infrared (2DIR) spectroscopy, Lee et al.
gen bonds like OH∙∙∙N, the contribution of the (2011) investigated the contact ion pairs (CIP)
proton transfer, OH  · · ·  N ⇌ O−  · · ·  HN+, could assembled by Li+ and SCN−ions in N,
lead to a hydrogen-bonded ion-pair formation. N-dimethylformamide. In IR spectrum, the CIP
Sobczyk and Paweła (1974) have demonstrated formation led to a blue shift (~16 cm−1) of the
the existence of proton-transfer equilibrium CN stretch frequency of Li–SCN CIP with
under appropriate conditions by measuring the respect to that of free SCN− ion. Moreover, the
dipole moment of carboxylic acid–pyridine temperature-dependent IR absorption spectra
base complexes. The results indicated that the revealed that the CIP formation was an endo-
dipole moments of these complexes were thermic process. The CIP association and dis-
dependent on the pKa difference (∆pKa) between sociation time constants (165 and 190 ps,
carboxylic acid and pyridine base, and large respectively) were determined by chemical
dipole moment was induced by strong interac- exchange 2DIR spectroscopy. The experimen-
tion of ion pairs. This type of interaction tal results indicated that the ion-pair formation
depends on the following factors: (a) ∆pKa of was a dynamic process in electrolyte solutions
protonic acid and base, (b) specific complex and in biological systems under physiological
solvation by solvent molecules, and (c) the conditions. In the case of hydrogen-bonded ion
influence of solvent expressed by its macro- pairs, a broad continuum, called as the Zundel
scopic dielectric permittivity. Based on the ion- continuum, is often observed in IR spectrum
pair model established by Huyskens and with extensive intermolecular hydrogen bond-
Zeegers-Huyskens (1964), it was predicted that ing, for which proton transfer is valuable. The
a ∆pKa of 3.6–6 between protonic acid and base broad band is caused by the strong hydrogen
could lead to an almost complete shift to the bonds in which a proton is distributed between
proton-­transfer equilibrium. A recent study the two hydrogen-­bonded species by tunneling
(Gilli et al. 2009) also showed that ion-pair for- (Biliškov et al. 2011). According to the classi-
mation could be reliably predicted from ∆pKa cal theory of hydrogen bond, a shift toward
between the donor and acceptor groups. lower fields in the NMR spectrum is suggested
as a criterion to confirm the formation of a
hydrogen bond due to strong deshielding of the
13.1.3 The Confirmation of Ion-Pair protons. Xi et al. (2012a, b) confirmed the for-
Formation mation of hydrogen-bonded ion pairs between
weak acidic drugs and organic amines at 1:1
In theory, ion pairs are defined as binary species molar ratio by IR and 1H-NMR. In this study,
which exist in solution and in solid in the salt teriflunomide (TEF) and lornoxicam (LOX),
form. Such intermolecular interaction can be two weak acidic drugs with OH groups, were
qualitatively inferred from the spectral character- used as the model drugs, and various
istics. A variety of spectroscopic techniques organic amines including triethylamine (TEtA),
including infrared spectroscopy (IR), nuclear diethylamine (DEtA), N-(2′-hydroxyethanol)-
magnetic resonance (NMR), ultraviolet–visible piperidine (NP), diethanolamine (DEA), and
spectroscopy (UV-Vis), electron spin resonance triethanolamine (TEA) were employed as the
178 L. Fang et al.

N O OH
H
N
S
N
H CI
OH O F
N
S
F
F O O

Teriflunomide (TFF) Lornoxicam (LOX)

N N N
H
OH

Triethylamine (TEtA) Diethylamine (DEtA) N-(2’ -hydroxy-ethanol)-


piperdine (NP)

OH

H
N N
HO OH HO OH

Triethanolamine (TEA) Diethanolamine (DEA)

Fig. 13.1 The chemical structure of teriflunomide (TEF), lornoxicam (LOX), triethylamine (TEtA), diethylamine
(DEtA), N-(2′-hydroxyethanol)-piperidine (NP), diethanolamine (DEA), and triethanolamine (TEA)

counter ions, whose structure was shown in of hydrogen bond in NMR. However, actually
Fig.  13.1. CHCl3 and CDCl3 solutions of TEF this phenomenon may be caused by strong
or LOX with or without the adding of equimo- shielding of the proton, which was a direct
lar organic amines were detected, respectively, consequence of the intermolecular hydrogen
by spectroscopic methods. In IR spectra bond interaction between drugs and organic
(Fig.  13.2), the absorption at ~3,400 cm−1 was amines instead of intramolecular hydrogen
assigned to stretching vibration of OH group of bonds in drugs. Notably, the chemical shift of
the two drugs. A continuum gave rise to a very OH group kept almost constant when the stoi-
broad absorption in the 3,300–2,000 cm−1 range chiometric ratios of drug to organic amine
in the presence of most of organic amines. In were varied from 1:1 to 1:100. These results
1
H-NMR study, compared to the signal of the suggested that TEF and LOX have been inte-
proton from OH group of TEF or LOX itself grated sufficiently into ion pairs at the equimo-
(15.35 and 13.02 ppm, respectively), the proton lar ratio.
magnetic resonance of OH in the complexes In addition, UV-Vis, ESR spectra in solution,
has moved toward higher field, as illustrated in and X-ray crystallography also have been
Fig.  13.3. It seemed that the results were con- employed for measuring the electronic changes
tradictory to the abovementioned classical theory in ion-pair formation process (Lü et al. 2005;
13  Formation of Ion Pairs and Complex Coacervates 179

a 3,600 3,200 2,800 2,400 2,000 b 3,600 3,200 2,800 2,400 2,000
100 105
95 100

%T
%T

90 95
90 TEF-DEA
85 DEA 100
100 95

%T
95 90
%T

85 TEF-TEA
90
85 TEA 96
84

%T
100
72
80 60 TEF-NP
%T

60 98
84
40 NP

%T
70
110
56 TEF-DEtA
100 96
%T

90 %T 84
DEtA 72
80
60 TEF-TEtA
100 105
%T

100
90 95
%T

90
80 TEtA 85 TEF
3,600 3,200 2,800 2,400 2,000 3,600 3,200 2,800 2,400 2,000

Wavenumber (cm-1) Wavenumber (cm-1)

c 3,600 3,200 2,800 2,400 2,000


100
96
%T

92
88 LOX-DEA
100
90
%T

80
70 LOX-TEA
96
84
%T

72
LOX-NP
60
99
88
%T

77
66 LOX-DEta
98
84
%T

70
LOX-TEta
56
96
84
%T

72
60 LOX
3,600 3,200 2,800 2,400 2,000
Wavenumber (cm-1)

Fig. 13.2  Infrared spectra of organic amines, TEF and organic amines; (c) LOX with or without equimolar
LOX in CHCl3: (a) organic amines which was equimo- organic amines
lar with test drug; (b) TEF with or without equimolar
180 L. Fang et al.

Fig. 13.3  1H-NMR spectra a


of TEF and LOX with or TEF-DEA
without organic amines in
CDCl3 at different molar TEF-TEA
ratios: (a) TEF, amines = 1:1;
(b) TEF, amines = 1:100; (c) TEF-NP
LOX, amines = 1:1; (d) LOX,
amines = 1:100 TEF-DEtA

TEF-TEtA

TEF
ppm 15 10 5 0
b
TEF-DEA

TEF-TEA

TEF-NP

TEF-DEtA

TEF-TEtA

TEF

ppm 15 10 5 0
c
LOX-DEA

LOX-TEA

LOX-NP

LOX-DEtA

LOX-TEtA

LOX
ppm 15 10 5 0
d
LOX-DEA

LOX-TEA

LOX-NP

LOX-DEtA

LOX-TEtA

LOX
ppm 15 10 5 0

Pal et al. 2010). Hudson et al. (1972) found that absorption spectra were presented in going from
when weak acidic 3,4-dinitrophenol encountered free acid to a hydrogen-bonded complex, to an
organic amines at different molar ratio, a series ion pair, to a solvated ion pair or a solvated anion.
of characteristic bathochromic shifts in UV Lü et al. (2005) determined the crystal structures
13  Formation of Ion Pairs and Complex Coacervates 181

a H1 b H22
O2
O3
C5 H24
C7
C16 H23
O1 H1
C4 N2
C6
O2
O1 C18
C7 C17
C3
C1
H15
N1 C2 C6 H15
C5 C1 C9
N1
C8
C14 C8
C10
C4 C2
C13 C9
C3 C11
C12 C13 C12
C10 C14

C15 C15
C11

cC15 d H29
O4
C11 C19
O2
C12 C5
C10
C4 C18
C13 H25 C7 C6
C9 N2
O4 C19 C16 H1
C14 O1
C21
C8 C18 C3
H15 C1 C17 C20 H30
N1 H26 C2 O5
H15 N1
C1 O1 N2 C14 O3
C2 H1
C16 C8 H28
C6 C13
C3 C7 O3
C9
C17
H24 C12 C10
C5 C15
C4 O2 C11

Fig. 13.4 Stereoscopic view of (a) MH, (b) MH- hydrogen bonds. Dashed lines indicate the intermolecular
­propanolamine, (c) MH-DEA, and (d) MH-TEA, showing hydrogen bonding
atom numbering, 50 % thermal probability ellipsoids, and

of pure ion-pair salts K(LC)+//DNB− and spectral changes. Compared with other methods,
K(LE)+DNB− by X-ray crystallography. In the X-ray crystallography can provide definitive
near-IR spectral analysis, they found that there structural information via analyzing the diffrac-
were the same patterns of vibronic progressions tion pattern of single crystal ion-pair salts. In
for distinguishing the “separated” from the “con- another study, Fang et al. (2004) prepared the
tact” ion pair in both of the crystalline solid state crystals of ion-pair complexes with an equimolar
and THF solution state, which ensured that the ratio of mefenamic acid (MH) and alkanolamines
same X-ray structures persist in solution. Most by removing the solvent in vacuo and subse-
importantly, in this study, the labilities of these quently confirmed that these complexes were
dynamic ion pairs in solution were thoroughly associated with hydrogen bonds using X-ray
elucidated by the temperature-dependent ESR crystallography (Fig. 13.4).
182 L. Fang et al.

13.1.4 The Effect of Ion-Pair enhancing effect on RIS penetration from the
Formation on Skin aqueous solution into hairless mouse skin, and
Permeation RIS-diethylenetriamine ion pair brought out the
largest enhancement ratio (ER), up to 36-fold
13.1.4.1  The Mechanism of Skin compared to only RIS.
Penetration Enhancement by As to lipophilic drugs possessing some polar
Ion-Pair Formation functional groups, e.g., –COOH, −OH, and –NH2,
The effect of ion-pair formation on skin permeation their skin permeation can also be enhanced by
is complex, and its mechanism has not been thor- hydrogen-bonded ion-pair formation (Cheong
oughly clarified. Generally, the skin penetration and Choi 2002; Green et al. 1989; Kamal et al.
enhancement by ion-pair approach with suitable 2007; Nogueira et al. 2011). However, for those
counter ions is mainly dependent on the physico- lipophilic drugs, their lipophilicity can be
chemical properties of the counter ions (e.g., lipo- decreased by ion-pair formation with small
philicity, pKa, and structure) and the solubility of molecular weight relative hydrophilic counter
ion pairs in donor medium. ions (Fang et al. 2003). The effect of the organic
The extent of penetration enhancement by amines including monoethanolamine (MEA),
ion-pair formation is strongly related to the lipo- DEA, TEA, and propanolamine (PPA) on the
philicity of the ion pairs and the properties that penetration of mefenamic acid (MH) across hair-
depend on the lipophilicity of the selected coun- less rat skin from the lipophilic mixed solvent of
ter ions. A series of studies performed by Neubert isopropyl myristate (IPM) and ethanol (9:1). The
et al. (Neubert et al. 1984; Neubert and Dittrich n-octanol/water partition coefficients (log Ko/w) at
1989; Neubert and Fischer 1991) have made 32 °C of MH and its corresponding ion pairs,
great contribution to the understanding of how MH-MEA, MH-DEA, MH-TEA, and MH-PPA,
hydrophilic ionized drugs penetrate across lipid were 3.31, 0.79, 0.74, 1.99, and 0.66, respec-
membranes together with lipophilic counter ions. tively, which indicated that these complexes were
These studies showed that the partition coeffi- relatively hydrophilic compared with MH. Hence,
cient of the hydrophilic drugs, buformine, qui- the transdermal delivery of MH was significantly
nine, pholedrine, and bretylium, was markedly enhanced by the formation of hydrogen-bonded
increased by more than twofold after the forma- ion pairs, and the ER values of these ion pairs
tion of ion pairs with lipophilic ions, and thereby were 279, 48, 84, and 357, respectively. Obviously,
the transport of ionized drugs across an artificial the reduced lipophilicity of the complexes has
lipid membrane (dodecanol collodion mem- facilitated the partition from the SC to the ED and
brane) could be enhanced. Moreover, it was consequently enhanced drug delivery through the
found that the counter ions could be accumulated skin. These results suggested that a major part of
in the lipid membrane due to their high lipid solu- ion pairs remained the integrity of ion pair during
bility and that they acted as carriers for the ion- the process of crossing the lipophilic SC and the
ized drugs. Besides the increased transport of hydrophilic ED until they reach the receptor com-
ionized drugs, the counter transport of protons partment. The subsequent studies done by Fang’s
and lithium ions, respectively, was also observed. group further confirmed this point of view
Nam et al. (2011) also provided a similar result in (Table 13.1).
the skin permeation of hydrophilic and highly In donor medium, the pKa of the counter ions is
ionized risedronate (RIS) with three lipophilic another main factor that influences the skin perme-
basic counter ions, l-arginine, l-lysine, and ation of ion pairs, especially the h­ ydrogen-­bonded
diethylenetriamine, at different molar ratios. To ion pairs. A positive correlation was found between
varying degree, all the counter ions could enhance the skin permeation of ion pairs and the pKa of the
the solubility of RIS in xylene, a lipophilic sol- counter ions was found (Ma et al. 2010; Xi et al.
vent. Although RIS ion pairs are slightly unstable 2012a, b). In other words, the ∆pKa between the
in the aqueous solution, they had a remarkable drug and the counter ion (they are acids and bases,
13  Formation of Ion Pairs and Complex Coacervates 183

Table 13.1  The enhancement ratio (ER) of ion pairs of stability of hydrogen bonds (Guy and Hadgraft
drugs with different amines
1984; Michaels et al. 1975). According to Xi
Drugs Counter ions ERa et al. (2012a, b), the stability parameter of ion
Teriflunomide in None 1.00 pairs (Tlife) can be obtained, based on the follow-
isopropyl palmitateb Diethylamine 2.47 ing Eq. 13.2 and data of 1H-NMR shown in
Triethylamine 12.69 Fig. 13.3.
Triethanolamine 1.44
Diethanolamine 1.15
1
N-(2′-hydroxyethanol)- 4.54 Tlife > (13.2)
piperidine 2p ( Dd ) v0

Lornoxicam in None 1.00
isopropyl palmitatec Diethylamine 13.63 where ν0 was the spectrometer frequency and Δδ
Triethylamine 19.52 was the chemical shift difference (Tubbs and
Triethanolamine 4.92 Hoffmann 2004). The results in Fig. 13.5 showed
Diethanolamine 13.77 that, in general, the ER values were increased
N-(2′-hydroxyethanol)- 12.08 with Tlife. Therefore, the better stability of
piperidine hydrogen-­ bond ion pairs notably facilitated
Flurbiprofen in 10 % None 1.00 transdermal drug delivery.
EtOH/isopropyl Diethylamine 2.02
myristated Megwa et al. (2000a, b) carried out some in vitro
Triethylamine 2.27
studies to evaluate the possibility of improving the
Triethanolamine 2.93
skin permeation of salicylate through human epi-
Diethanolamine 3.76
dermis from an aqueous solution by forming ion
Ethanolamine 2.60
pairs with basic counter ions (alkylamines and qua-
N-(2′-hydroxyethanol)- 1.86
piperidine ternary ammonium ions). The skin permeation of
Glipizide in 20 % Na salt 1.00 the salicylate ion pairs with primary amines and
EtOH/isopropyl Diethylamine 6.79 quaternary ammonium ions was lower than that of
myristatee Triethylamine 19.61 salicylate itself (ER < 1), while all of secondary and
Triethanolamine 2.34 tertiary amines except DEtA (ER = 0.83) signifi-
Diethanolamine 5.77 cantly promoted the skin permeation of salicylate
Ethanolamine 11.92 (ER: 1.34–4.80). The enhancement effect of these
N-(2′-hydroxyethanol)- 3.90 amines on the penetration of salicylate was in the
piperidine (NP)
following order: quaternary < primary < secondary
a
ER, the enhancement ratio of the cumulative amounts of < tertiary. This phenomenon was attributed to the
drug permeated between with and without organic amines
b,c
Xi et al. (2012a, b) fact that the complex of salicylate with tertiary
d
Ma et al. (2010) amines had higher stability than that with primary
e
Tan et al. (2009) or secondary amines.
The skin permeation rate is also dependent on
the concentration of the soluble permeant in the
correspondingly) can directly influence the applied vehicle. For lipophilic drugs with sparingly
strengths (stability) of hydrogen-­bonded ion pairs solubility in both oil and water like piroxicam,
in a nonpolar medium. meloxicam, and LOX, the skin permeation is very
Most intermolecular hydrogen bonds in the poor (Cheong and Choi 2002; Xi et al. 2012b;
liquid state are formed and broken on an extremely Zhang et al. 2009). Improving the ­solubility in
short timescale (e.g., ~10−5 s), even a picosecond donor solution and the partition in the SC and the
scale (Becker 2007; Simon and Peters 1982). It ED by ion-pair formation is the main mechanism
has been widely accepted that there are abundant of penetration enhancement for those drugs. A
hydrogen bond acceptors and donors existing in recent study (Song et al. 2012) showed that biso-
the SC part of the skin and they may disturb the prolol-maleate salt possessed a higher solubility in
binding of ion pairs and further influence the DURO-TAK® 87-4098 acrylate-­ vinyl acetate
184 L. Fang et al.

Fig. 13.5  The relationship a


between the skin enhance- 14
ment of ion pairs of TEF and TEF
LOX and the stability r = 0.8457
parameter Tlife 12

10

8
ER

0
84 86 88 90 92 94 96
Tlife(µS)
b
20
LOX
r = 0.8380

15
ER

10

0
300 350 400 450 500 550 600
Tlife(µS)

adhesive (National Starch & Chemical Co., USA) 13.1.5 Ion-Pair Formation vs.
than bisoprolol-fumarate salt, which was respon- Penetration Enhancers
sible for the higher penetration enhancing effect of
the maleate salt. In addition, ion pairs existing in Currently, the application of chemical enhancers
salt form can obviously decrease the melting point is a frequently used approach to enhance the per-
of the parent drug, which is frequently used as a meation of drugs through biomembranes.
predictor of solubility, and thereby promotes the However, chemical enhancers are not omnipotent
skin permeation of drugs from transdermal formu- in the drug delivery across the skin. Tan et al.
lations, such as patches and emulgels (Cheong and (2009) found that glipizide (GP) ion pairs with
Choi 2003; Wang and Fang 2008). organic amines as the counter ions provided an
13  Formation of Ion Pairs and Complex Coacervates 185

Table 13.2  The enhancement ratio (ER) of ion pairs of drugs with different counter ions and penetration enhancers
Drugs Counter ions ERa Penetration enhancers ER
Indapamide in 30 % (w/w) enthol/IPM b None 1.00 None 1.00
Acetic acid 2.74 5 % Azone® 4.83
Maleic acid 4.34 5 % l-menthol 3.01
Oxalic acid 2.85 5 % oleic acid 2.52
Adipic acid 3.10 5 % N-methyl pyrrolidone 3.88
Lactic acid 8.46
Citric acid 3.47
Succinic acid 5.13
Fumaric acid 6.93
Scutellarin in 20 % enthol/IPM c None 1.00 None 1.00
Diethylamine 4.76 5 % Azone® 5.64
Triethylamine 3.83 5 % l-menthol 3.28
Ethanolamine 7.11 5 % oleic acid 2.60
Diethanolamine 3.68 5 % N-methyl pyrrolidone 1.38
Triethanolamine 1.66
Meloxicam in 10 % propylene glycol /IPM d None 1.00 None 1.00
Diethylamine 3.71 5 % Azone® 2.89
Triethylamine 1.99 5 % l-menthol 1.28
Ethanolamine 1.399 5 % oleic acid 2.29
Diethanolamine 4.20 5 % N-methyl pyrrolidone 5.77
Triethanolamine 3.94
N-(2′- 5.78
hydroxyethanol)-
piperidine
a
ER, the enhancement ratio of the cumulative amounts of drug permeated between with and without counter ions
b
Ren et al. (2007)
c
Wang et al. (2008)
d
Zhang et al. (2009)

obvious enhancement of the skin permeation of 13.2 Complex Coacervates


GP, while five common enhancers, IPM, propyl- for Penetration
ene glycol, N-methyl-2-pyrrolidone (NMP), Enhancement
Azone® (AZ, Tianmen Kejie Pharmaceuticals
Co., Ltd., China), and oleic acid (OA), had no Complex coacervates are a specialized form of
enhancing effect. Moreover, Cheong and Choi ion pairs, which represents the separation of an
(2002) investigated the effect of various enhanc- aqueous phase containing a mixture of oppo-
ers on the permeation of piroxicam (PX) and its sitely, charged ions into a dense coacervate oil
ethanolamine salts (PX-EAs). The results showed phase, rich in ionic complex, and a dilute equilib-
that, in general, ion-pair salts still exerted a great rium phase. The difference between complex
enhancement on the penetration of PX from vari- coacervates and ion pairs is that a complex coac-
ous saturated solution in which the enhancers ervate exists as a binary phase system. However,
also work as the donor mediums. As illustrated in in transdermal delivery systems, complex coac-
Table 13.2, some studies support the finding that ervates behave like ion pairs. Stott et al. (1996)
ion-pair formation shows a better or comparable prepared complex coacervates composed of cat-
penetration enhancing effect compared to classi- ionic amitriptyline (AMI) and counter ions
cal chemical penetration enhancers (Ren et al. including sodium deoxycholate (NaD) or sodium
2008; Wang et al. 2008; Zhang et al. 2009). lauryl sulfate (SLS). The produced complex
186 L. Fang et al.

coacervates AMI-NaD and AMI-SLS were Barry BW (2001) Is transdermal drug delivery research
still important today? Drug Discov Today 6:967–971
employed to investigate their potentials to
Barthel J, Deser R (1994) FTIR study of ion solvation and
enhance transdermal flux of AMI. Although ion-pair formation in alkaline and alkaline earth metal salt
AMI-NaD was separated into two distinct phases solutions in acetonitrile. J Solut Chem 32:1133–1146
(octanol and vehicle), while AMI-SLS was in sol Becker ED (2007) Hydrogen bonding. In: Harris RK,
Wasylishen RE (eds) eMagRes. Wiley, Chichester
state, both of the systems could obviously
Biliškov N, Kojić-Prodić B, Mali G, Molčanov K, Stare J
increase octanol/vehicle partition coefficients of (2011) A partial proton transfer in hydrogen bond O−
AMI. However, in the skin permeation study, H∙∙∙O in crystals of anhydrous potassium and rubidium
only the AMI-NaD coacervate provided a 2.2- complex chloranilates. J Phys Chem A 115:3154–3166
Cheong HA, Choi HK (2002) Enhanced percutaneous
fold increment in drug flux. On the contrary, the
absorption of piroxicam via salt formation with etha-
AMI-SLS coacervate showed a marked reduc- nolamines. Pharm Res 19:1375–1380
tion in drug flux. The results indicated that the Cheong HA, Choi HK (2003) Effect of ethanolamine salts
increased lipophilicity of the coacervate’s oil and enhancers on the percutaneous absorption of
piroxicam from a pressure sensitive adhesive matrix.
phase could contribute to an increase in the trans-
Eur J Pharm Sci 18:149–153
dermal flux of charged species. Diamond RM (1963) The aqueous solution behavior of
large univalent ions. A new type of ion-pairing. J Phys
Chem 67:2513–2517
Fang L, Numajiri S, Kobayashi D, Morimoto Y (2003)
13.3 Summary The use of complexation with alkanolamines to facili-
tate skin permeation of mefenamic acid. Int J Pharm
In conclusion, ion-pair formation is a simple and 262:13–22
useful method for enhancing percutaneous pene- Fang L, Numajiri S, Kobayashi D, Ueda H, Nakayama K,
Miyamae H et al (2004) Physicochemical and crystal-
tration of drugs by modifying the physicochemi-
lographic characterization of mefenamic acid com-
cal properties of parent molecules and by plexes with alkanolamines. J Pharm Sci 93:144–154
regulating the partition of drugs between the Gilli P, Pretto L, Bertolasi V, Gilli G (2009) Predicting
­dosage form, the SC, and the ED. Furthermore, hydrogen-bond strengths from acid-base molecular
properties. The pKa slide rule: toward the solution of a
hydrogen-bonded ion pairs make some unioniz-
long-lasting problem. Acc Chem Res 42:33–44
able molecules become suitable for ion-pair for- Green PG, Hadgraft J, Ridout G (1989) Enhanced in vitro
mation. In order to obtain a maximum skin permeation of cationic drugs. Pharm Res 6:628–632
enhancement in permeation of drugs, the combi- Guy RH, Hadgraft J (1984) Prediction of drug disposition
kinetics in skin and plasma following topical adminis-
nation of ion pairs and penetration enhancers is a
tration. J Pharm Sci 73:883–887
feasible approach. The enhancement mechanism Habeeb MM (1997) Spectroscopic studies of proton trans-
of ion pairs in transdermal drug delivery is worth fer equilibria in hydrogen bonded complexes. Appl
for further study. As to complex coacervates, Spectrosc Rev 32:103–140
Hudson RA, Scott RM, Vinogradov SN (1972) Hydrogen-
although the published data about their applica-
bonded complex-ion-pair equilibriums in 3,
tion in the field of transdermal drug delivery is 4-dinitrophenol-amine-aprotic solvent systems. J Phys
limited, this technology is still a bright prospect Chem 76:1989–1993
for transdermal pharmaceutical formulations. Huyskens PL, Zeegers-Huyskens (1964) Molecular asso-
ciations and acid-base equilibriums. J Chem Phys
Phys-Chem Biol 61:81–86
Kamal MA, Iimura N, Nabekura T, Kitagawa S (2007)
Enhanced skin permeation of diclofenac by ion-pair
References formation and further enhancement by microemul-
sion. Chem Pharm Bull 55:368–371
Arunan E, Desiraju GR, Klein RA, Sadlej J, Scheiner S, Kraus CA (1956) The ion-pair concept: its evolution and
Alkorta I et al (2011) Defining the hydrogen bond: an some application. J Phys Chem 60:129–141
account (IUPAC technical report). Pure Appl Chem Lee KK, Park KH, Kwon D, Choi JH, Son H, Park S et al
83:1619–1636 (2011) Ion-pairing dynamics of Li+ and SCN− in
Barrow GM (1956) The nature of hydrogen bonded ion-­ dimethylformamide solution: chemical exchange two-­
pairs: the reaction of pyridine and carboxylic acids in dimensional infrared spectroscopy. J Chem Phys
chloroform. J Am Chem Soc 78:5802–5806 134:064506
13  Formation of Ion Pairs and Complex Coacervates 187

Lü JM, Rosokha SV, Lindeman SV, Neretin IS, Kochi JK Pregosin PS (2009) NMR spectroscopy and ion pairing:
(2005) “Separated” versus “contact” ion-pair struc- measuring and understanding how ions interact. Pure
tures in solution from their crystalline states: dynamic Appl Chem 81:615–633
effects on dinitrobenzenide as a mixed-valence anion. Ratajczak H (1972) Charge-transfer properties of the
J Am Chem Soc 127:1797–1809 hydrogen bond. I. Theory of the enhancement of
Ma X, Fang L, Guo J, Zhao N, He Z (2010) Effect of coun- dipole moment of hydrogen-bonded systems. J Phys
ter-ions and penetration enhancers on the skin perme- Chem 76:3000–3004
ation of flurbiprofen. J Pharm Sci 99:1826–1837 Ren C, Fang L, Li T, Wang M, Zhao L, He Z (2008) Effect
McNaught AD, Wilkinson A (1997) Ion pair. In: of permeation enhancers and organic acids on the skin
Compendium of chemical terminology, 2nd edn. (the permeation of indapamide. Int J Pharm 350:43–47
“Gold Book”). IUPAC. http://goldbook.iupac.org/I03 Simon JD, Peters KS (1982) Picosecond dynamics of ion
231.html. Accessed 19 Oct 2002. pairs: the effect of hydrogen bonding on ion-pair
Megwa SA, Cross SE, Benson HA, Roberts MS (2000a) intermediates. J Am Chem Soc 104:6542–6547
Ion-pair formation as a strategy to enhance topical deliv- Sobczyk L, Paweła Z (1974) Infra-red spectra and dipole
ery of salicylic acid. J Pharm Pharmacol 52:919–928 moments of hydrogen-bonded complexes. Part 5.—
Megwa SA, Cross SE, Whitehouse MW, Benson HA, proton transfer in carboxylic acid–pyridine complexes.
Roberts MS (2000b) Effect of ion pairing with alkyl- J Chem Soc Faraday Trans 1 70:832–838
amines on the in-vitro dermal penetration and local Song W, Cun D, Xi H, Fang L (2012) The control of skin-
tissue disposition of salicylates. J Pharm Pharmacol permeating rate of bisoprolol by ion-pair strategy for
52:929–940 long-acting transdermal patches. AAPS PharmSciTech
Michaels AS, Chandrasekaran K, Shaw JE (1975) Drug 13:811–815
permeation through human skin: theory and in vitro Stott PW, Williams AC, Barry BW (1996)
experimental measurement. Am Inst Chem Eng J Characterization of complex coacervates of some tri-
21:985–996 cyclic antidepressants and evaluation of their poten-
Miller JM, Dahan A, Gupta D, Varghese S, Amidon GL tial for enhancing transdermal flux. J Control Release
(2009) Quasi-equilibrium analysis of the ion-pair 41:215–227
mediated membrane transport of low-permeability Takacs-Novak K, Szasz G (1999) Ion-pair partition of
drugs. J Control Release 137:31–37 quaternary ammonium drugs: the influence of counter
Nagy PI, Takacs-Novak K (2000) Theoretical and experi- ions of different lipophilicity, size, and flexibility.
mental study on ion-pair formation and partitioning of Pharm Res 16:1633–1638
organic salts in octanol/water and dichloromethane/ Tan Z, Zhang J, Wu J, Fang L, He Z (2009) The enhancing
water systems. J Am Chem Soc 122:6583–6593 effect of ion-pairing on the skin permeation of glipi-
Nam SH, Xu YJ, Nam H, Jin GW, Jeong Y, An S et al zide. AAPS PharmSciTech 10:967–976
(2011) Ion pairs of risedronate for transdermal deliv- Tubbs JD, Hoffmann MM (2004) Ion-pair formation of the
ery and enhanced permeation rate on hairless mouse ionic liquid 1-ethyl-3-methylimidazolium bis(trifyl)
skin. Int J Pharm 419:114–120 imide in low dielectric media. J Solut Chem 33:381–394
Neubert R (1989) Ion pair transport across membranes. Wang ML, Fang L (2008) Percutaneous absorption of
Pharm Res 6:743–747 diclofenac acid and its salts from emulgel. Asian
Neubert R, Dittrich T (1989) Ampicillin ionenpaartrans- J Pharm Sci 3:131–141
port im vergleich mit dem transport weiterer penicil- Wang M, Fang L, Ren C, Li T (2008) Effect of ion-pairing
line. Pharmazie 44:67–68 and enhancers on scutellarin skin permeability.
Neubert R, Fischer S (1991) Influence of lipophilic coun- J Pharm Pharmacol 60:429–435
ter ions on the transport of ionizable hydrophilic Xi H, Cun D, Wang Z, Shang L, Song W, Mu L et al
drugs. J Pharm Pharmacol 43:204–206 (2012a) Effect of the stability of hydrogen-bonded ion
Neubert R, Fürst W, Böhm W, Dabow S (1984) Drug perme- pairs with organic amines on transdermal penetration
ation through artificial lipid membranes. 17. The mecha- of teriflunomide. Int J Pharm 436:857–861
nism of ion pair transport. Pharmazie 39:401–403 Xi H, Wang Z, Chen Y, Li W, Sun L, Fang L (2012b)
Nogueira IR, Carneiro G, Yoshida MI, de Oliveira RB, The relationship between hydrogen-bonded ion-
Ferreira LA (2011) Preparation, characterization, and pair stability and transdermal penetration of lor-
topical delivery of paromomycin ion pairing. Drug noxicam with organic amines. Eur J Pharm Sci 47:
Dev Ind Pharm 37:1083–1089 325–330
Pal D, Goswami D, Nayak SK, Chattopadhyay S, Zhang JY, Fang L, Tan Z, Wu J, He ZG (2009) Influence
Bhattacharya S (2010) Spectroscopic and theoretical of ion-pairing and chemical enhancers on the transder-
insights into the origin of Fullerene − Calix[4]pyrrole mal delivery of meloxicam. Drug Dev Ind Pharm
interaction. J Phys Chem A 114:6776–6786 35:663–670
Formulation of Drug-Cyclodextrin
Complexes 14
Thorsteinn Loftsson

Contents 14.1 Introduction


14.1 Introduction. . . . . . . . . . . . . . . . . . . . . . . . 189
14.1.1 Cyclodextrins and Their Properties. . . . . . . 190 Drugs permeate intact skin as single molecules.
14.1.2 Cyclodextrin Complexes. . . . . . . . . . . . . . . 190
When drug products are applied to the skin sur-
14.2  yclodextrins as Permeability
C face, dissolved drug molecules diffuse through
Enhancers. . . . . . . . . . . . . . . . . . . . . . . . . . 193 the vehicle to the skin where the molecules parti-
14.2.1 Theoretical Background . . . . . . . . . . . . . . . 193
14.2.2 Cyclodextrins and Biological Membranes. 197 tion from the vehicle into the skin and then per-
meate the skin barrier, stratum corneum, into the
14.3 Formulation Optimization . . . . . . . . . . . . 198
14.3.1 When Can Cyclodextrin Help?. . . . . . . . . . 198
more permeable inner skin layers. Most penetra-
14.3.2 What Is the Desired Effect?. . . . . . . . . . . . . 199 tion enhancers, chemical as well as physical,
14.3.3 How to Optimize the Formulation?. . . . . . . 199 enhance drug delivery by making the skin barrier
Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202 more permeable. Cyclodextrins are different.
They enhance drug delivery into and through the
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202
skin by increasing the availability of dissolved
drug molecules right at the skin surface. However,
cyclodextrins can also hamper dermal and trans-
dermal drug delivery by preventing drug mole-
cules from partitioning from the surface into the
skin. Thus, successful employment of cyclodex-
trins in topical drug formulations requires good
understanding of their physicochemical proper-
ties and the way they enhance topical drug
bioavailability.
Numerous books and reviews have been writ-
ten on cyclodextrins, their industrial applications,
and usage in drug formulations (Loftsson
and Brewster 1996; Dodziuk 2006; Douhal 2006;
Brewster and Loftsson 2007; Loftsson and
T. Loftsson
Faculty of Pharmaceutical Sciences, University of
Brewster 2010; Loftsson and Duchêne 2007;
Iceland, Hofsvallagata 53, Reykjavik IS-107, Iceland Stella and He 2008; Uekama et al. 2006; Hedges
e-mail: thorstlo@hi.is 1998; Kurkov and Loftsson 2013; Bilensoy 2011).

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 189


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_14, © Springer-Verlag Berlin Heidelberg 2015
190 T. Loftsson

In this chapter, the effects of cyclodextrins on cyclodextrins, both towards nonenzymatic and
drug delivery through biological membranes are enzymatic degradation. There are no reports of
discussed with emphasis on dermal and transder- transporter-mediated permeation of cyclodex-
mal drug delivery. trins across biological membranes, and in gen-
eral, the oral bioavailability of cyclodextrins is
well below 4 % (Kurkov and Loftsson 2013).
14.1.1 Cyclodextrins and Their After parenteral administration, cyclodextrins
Properties are, like low-molecular-weight dextrins, mainly
excreted unchanged with urine. In humans, their
Cyclodextrins are cyclic oligosaccharides con- biological half-life is about 1.9 h and volume of
taining 6 (αCD), 7 (βCD), 8 (γCD), or more glu- distribution about 0.2 L/kg (Kurkov and Loftsson
copyranose monomers linked via α-1,4-glycoside 2013). The safety and toxicology of cyclodex-
bonds (Table 14.1). These parent cyclodextrins trins have recently been reviewed (Stella and He
are natural products formed by microbial degra- 2008; Arima et al. 2011).
dation of starch. The outer surface of the The regulatory status of cyclodextrins is
doughnut-­ shaped cyclodextrin molecules is slowly evolving as more and more cyclodextrin-­
hydrophilic, bearing numerous hydroxyl groups, containing products are being approved (Hincal
but their central cavity is somewhat lipophilic. et al. 2011). All three parent cyclodextrins and
Although the parent cyclodextrins and their com- many of their derivatives can be found in US
plexes are hydrophilic, their aqueous solubility is Pharmacopeia/National Formulary (USP/NF),
somewhat limited. This is thought to be due to the European Pharmacopoeia (Ph.EUR.), and the
relative strong intermolecular binding in their Japanese Pharmaceutical Codex (JPC). The par-
crystal state. Partial random substitution of the ent cyclodextrins have been included in the “gen-
hydroxy groups will result in significant erally recognized as safe” (GRAS) list of the
improvements in their solubility (Table 14.1). FDA, and they are commonly found in both food
Cyclodextrins possess many of the same physico- and toiletry products throughout the world.
chemical and biological properties as their cor- Worldwide cyclodextrins can be found in about
responding linear dextrins. In their solid state, 40 marketed pharmaceutical products (Loftsson
cyclodextrins are as stable as starch and can be and Brewster 2010; Hincal et al. 2011).
stored for a number of years at room temperature
without any detectable degradation (Szejtli
1988). In aqueous solutions, their degradation 14.1.2 Cyclodextrin Complexes
follows specific acid-catalyzed hydrolysis of the
glycoside bonds to form glucose, maltose, and Cyclodextrins are able to form drug-cyclodextrin
linear dextrins. In pure aqueous solution, the inclusion complexes by taking up somewhat lipo-
half-life for ring opening of βCD was determined philic drug moieties (or even small lipophilic
to be approximately 15 h at pH 1.1 and 70 °C molecules) into the central cavity (Fig. 14.1). No
(Hirayama et al. 1992). αCD is somewhat more covalent bonds are formed or broken during the
stable and γCD somewhat less than βCD complex formation, and drug molecules bound in
(Schönberger et al. 1988). Cyclodextrins are sta- the complex are in very dynamic equilibrium
ble towards β-amylases, but γCD is degraded by with free drug molecules in solution. Thus, cyclo-
salivary α-amylase (Szejtli 1987; Munro et al. dextrin complexes dissociate readily upon simple
2004). αCD, βCD, and γCD, as well as their dilution, for example, upon injection into liquid
derivatives that are currently used in pharma- chromatographic system or after parenteral
ceutical products, undergo bacterial digestion administration.
in the gastrointestinal tract (Irie and Uekama The main purpose for adding cyclodextrins to
1997; Kurkov and Loftsson 2013). Formation percutaneous drug formulations is to enhance
of inclusion complexes increases the stability of aqueous solubility of poorly soluble drugs and,
14  Formulation of Drug-Cyclodextrin Complexes 191

Table 14.1  Characteristics of the most common natural cyclodextrins and some of their derivatives that are of phar-
maceutical interest
Edge of secondary Edge of primary
a hydroxy groups hydroxy groups

Lipophilic
central cavity

Solubilityb
Cyclodextrin n R = H or Abbreviation Synonyms MS a
MW (Da) (mg/ml) LogKo/wc
α-Cyclodextrin 0 αCD Alfadex – 972.8 130 −13
β-Cyclodextrin 1 βCD Betadex – 1,135 18.4 −14
2-Hydroxypropyl- 1 −CH2CHOHCH3 HPβCD Hydroxypropyl 0.65 1,400 >600 −11
β-­cyclodextrin betadex
Sulfobutylether 1 −(CH2)4SO3− Na+ SBEβCD Betadex 0.9 2,163 >500 <−10
β-cyclodextrin sulfobutyl ether
sodium sodium
Randomly 1 −CH3 RMβCD 1.8 1,312 >500 −6
methylated
β-cyclodextrin
γ-Cyclodextrin 2 γCD Gammadex – 1,297 249 −17
2-Hydroxypropyl- 2 −CH2CHOHCH3 HPγCD Hydroxypropyl 0.6 1,576 >500 −13
γ-­cyclodextrin gammadex
a
Molar substitution (MS) is defined as the average number of substituents per glucopyranose repeat unit
b
From references (Loftsson and Brewster 2011; Sabadini et al. 2006)
c
Calculated LogKo/w (octanol-water partition coefficient) at 25 °C (interactive LogKow Calculator, Syracuse Research
Corporation: http://www.srcinc.com/what-we-­do/free-demos.aspx). These are approximate values. The exact values for
the cyclodextrin derivatives depend on their MS as well as the location of the substituents

Fig. 14.1  Formation of


one-to-one (i.e., 1:1)
drug-cyclodextrin inclusion
complex
192 T. Loftsson

thus, increase their topical bioavailability. where, in saturated drug solutions, [D] is the
Higuchi and Connors’ phase-solubility method is intrinsic solubility of the drug (S0), i.e., the solu-
used to study the effect of cyclodextrin concen- bility when no cyclodextrin is present in the
trations on drug solubility (Fig. 14.2) (Higuchi aqueous complexation media. The total drug sol-
and Connors 1965; Loftsson et al. 2005; Loftsson ubility ([D]T) in a given media is then:
and Hreinsdóttir 2006). The complex formation
is a reversible process:
[ D ]T = S0 + [ D / CD ]

(14.4)


m ⋅ D + n ⋅ CD ← 
m:n
→ Dm CD n (14.1)

K
assuming 1:1 D/CD complex formation according

to Eq. 14.3. A plot of [D]T versus [CD]T for the
where m drug molecules (D) associate with n cyclo- formation of a 1:1 D/CD complex should give a
dextrin (CD) molecules to form a complex of m:n straight line (i.e., AL-type phase-solubility dia-
stoichiometry. Km:n is the observed stability constant gram, Fig. 14.2) with the y-intercept representing
of the complex, also known as the binding constant, S0 and K1:1 defined as (Higuchi and Connors 1965):
formation constant, or association constant. The
Slope
stability constant can be written as follows: K11: = (14.5)
S0 × (1 - Slope )

K m: n =
[ Dm CDn ] (14.2)
[ D ] ⋅ [CD]
m n

where Slope is the slope of the linear AL diagram.


The slope is always less than unity when 1:1 com-
where the brackets denote the molar concentra- plex is being formed. Complexes of other stoichi-
tions. Most commonly, one drug molecule forms ometry are less common (Brewster and Loftsson
a complex with one cyclodextrin molecule: 2007; Loftsson and Brewster 2010). AP-­type pro-
file can indicate formation of a complex that is
K11: =
[ D / CD] (14.3) second or higher order with respect to cyclodex-

[ D ] × [CD] trin or that cyclodextrin complex aggregates
(nanoparticles) are being formed. The complex-
ation efficiency (CE) is calculated from the slope
of the phase-solubility diagram. It is independent
of the intercept (or S0) and frequently used when
the influence of various pharmaceutical excipi-
ents on the solubilization is investigated (Loftsson
and Brewster 2010, 2012). For 1:1 D/CD com-
plexes, the CE is calculated as follows:

CE =
[ D / CD] = S0 × K11: =
Slope


[CD] (1 - Slope )
(14.6)

The drug:CD molar ratio in a particular complex-


ation media saturated with the drug can thus be
calculated from the CE:
Fig. 14.2  Phase-solubility diagrams. A-type diagrams
are due to formation of water-soluble complexes and are
usually associated with the water-soluble cyclodextrin ( CE +1)
derivatives. B-type diagrams indicate formation of poorly D : CD molar ratio = 1: (14.7)
CE
soluble complexes that are usually associated with the
poorly soluble parent cyclodextrins. S0 is the intrinsic
drug solubility, i.e., the solubility of the drug in the com- For a more detailed mathematical description of
plexation media when no cyclodextrin is present the complex formation, the reader is referred to
14  Formulation of Drug-Cyclodextrin Complexes 193

recent reviews (Brewster and Loftsson 2007; cyclodextrins either increase or decrease drug
Loftsson and Brewster 2010) and the original pub- permeation through the skin. Still more studies
lication by Higuchi and Connors (1965). can be found on the effects of cyclodextrins on
Additionally, the effects of various pharmaceuti- drug absorption from the gastrointestinal tract
cal excipients on K1:1 and CE and how they can and the buccal cavity through the nasal mucosa as
enhance the solubilizing effects of cyclodextrins well as through other mucosal membranes, all of
have been reviewed (Loftsson and Brewster 2012). which can give us some insight into how cyclo-
dextrins act as penetration enhancers (Loftsson
et al. 2007b, 2008; Loftsson and Brewster 2011;
14.2 Cyclodextrins Loftsson 2012).
as Permeability Enhancers

In general, chemical penetration enhancers, such 14.2.1 Theoretical Background


as sulfoxides, fatty acids, fatty acid esters, alco-
hols, amides, and surfactants, enhance drug per- Drugs permeate the skin via passive diffusion.
meation into and through the skin by permeating The driving force for passive diffusion through
into the skin barrier where they temporarily an aqueous vehicle into the skin and then through
decrease its barrier properties. These penetration the skin is the gradient of chemical potential (μ)
enhancers enhance membrane permeation of (Higuchi 1960; Idson 1971). Likewise, the parti-
both hydrophilic and lipophilic drugs and, in tioning of drug molecules from the skin exterior
most cases, from both nonaqueous and aqueous into the outermost skin layer is controlled by the
vehicles. Studies have shown that the permeation-­ chemical potential. High chemical potential of
enhancing properties of cyclodextrins are quite the drug in topical vehicle is a prerequisite for its
different from these chemical permeation enhanc- good dermal bioavailability:
ers (Masson et al. 1999; Loftsson and Masson
m2 = m2q + RT ln a2 = m2q + RT ln ( g 2 m2 )
2001; Loftsson et al. 2004; Dahan et al. 2010;
Dahan and Miller 2012; Hymas et al. 2012). For (14.8)
example, only negligible amounts of cyclodex- and
trins are able to permeate intact skin and, thus,
a2 = g 2 m2 (14.9)
they do not directly affect the skin barrier. In one
study only 0.02 % of topically applied HPβCD where μ2 is the chemical drug potential in the
was absorbed into intact hairless mouse skin over vehicle, μ2θ is the chemical potential in a given
24 h period, whereas 24 % was absorbed into standard state, a2 is the thermodynamic drug
stripped skin where stratum corneum had been activity, R is the gas constant, T is the tempera-
removed (Tanaka et al. 1995). Another study ture in Kelvin, γ2 is the activity coefficient, and
showed that only 0.3 % of the more lipophilic m2 is the molality of the drug. The thermody-
dimethyl-β-cyclodextrin was absorbed into intact namic definition of the partition coefficient (Ko/w)
rat skin after topical application (Gerlóczy et al. of a drug between organic (o) and aqueous (w)
1988). In addition, cyclodextrins are only able to phases is:
enhance drug permeation from aqueous vehicles
m wq - moq a g ×C g
and in most cases they are only able to enhance = ln o » ln o o = ln o + ln K o/ w
permeation of lipophilic poorly water-soluble RT aw g w × Cw gw

drugs (Loftsson et al. 2007b, 2008; Loftsson and (14.10)
Brewster 2011; Loftsson 2012).
There are numerous reports on the effects of Equation  14.10 states that equilibrium between
cyclodextrins on dermal and transdermal drug the two phases is attained when the chemical
delivery (Table 14.2). Depending on the experi- potential of the drug in one phase (e.g., in water or
mental conditions and vehicle composition, the aqueous membrane exterior (μw)) is equal to
194 T. Loftsson

Table 14.2  Examples of cyclodextrin-containing dermal formulations and transdermal drug delivery studies
Drug Cyclodextrin Reference
Acitretin RMβCD Loftsson et al. (1995)
Alkannin HPβCD Chen et al. (1996)
Avobenzone HPβCD Yang et al. (2008)
Beclomethasone dipropionate γCD Uekama et al. (1985)
4-Biphenylylacetic acid βCD, DMβCD, HPβCD Arima et al. (1990, 1996)
Bupranolol HPβCD, PMβCD Babu and Pandit (2004)
Capsaicin HPβCD Zi et al. (2008)
Celecoxib DMβCD Ventura et al. (2006)
Curcumin HPβCD, HPγCD Hegge et al. (2008)
Dexamethasone acetate βCD, HPβCD Lopez et al. (2000)
17β-Estradiol HPβCD Loftsson et al. (1991)
Fludrocortisone acetate γCD Klang et al. (2012)
Human growth hormone αCD, βCD, HPβCD Shakory et al. (2010)
Hydrocortisone βCD, CMβCD, HPβCD, Loftsson et al. (1991, 1994a, b),
MLβCD, RMβCD Loftsson and Sigurðardottir (1994),
Sigurdardottir and Loftsson (1995),
Preiss et al. (1995), Chang and Banga
(1998), Masson et al. (1999), Kear
et al. (2008)
Ibuprofen HPβCD Iervolino et al. (2000)
Indomethacin βCD, DEβCD, DMβCD Okamoto et al. (1986), Kawahara et al.
(1992)
Ketoprofen HPβCD Batzdorf and Mullergoymann (1993)
Liarozole HPβCD Vollmer et al. (1993)
Lidocaine DMβCD, HPβCD, SBEβCD Dollo et al. (1998)
Loteprednol etabonate DMβCD Loftsson and Bodor (1994)
Melatonin HPβCD Lee et al. (1998)
Metopimazine MβCD Bounoure et al. (2007)
Methyl paraben HPβCD Tanaka et al. (1995)
Miconazole αCD, HPβCD Tenjarla et al. (1998)
Naproxen βCD Celebi et al. (1993)
Piribedil RMβCD Legendre et al. (1995)
Piroxicam HPβCD Doliwa et al. (2000), (2001)
Prednisolone βCD, γCD Uekama et al. (1987)
Progesterone αCD, βCD, γCD Klang et al. (2010)
Prostaglandin E1 αCD, βCD, CMEβCD, Adachi et al. (1992, 1993), Uekama
et al. (1992), Yuzuriha et al. (1999)
Shikonin HPβCD Chen et al. (1996)
Sulfanilic acid βCD, DMβCD Okamoto et al. (1986)
Testosterone HPβCD Loftsson et al. (1991)
Tolnaftate βCD, βCD-polymer Szeman et al. (1987)
Tretinoin βCD, HPβCD, DMβCD Amdidouche et al. (1994), Montassier
et al. (1998), Ascenso et al. (2012)
Triamcinolone HPβCD Kear et al. (2008)
αCD α-cyclodextrin, βCD β-cyclodextrin, CMβCD carboxymethyl-β-cyclodextrin, HPβCD 2-hydroxypropyl-β-­
cyclodextrin, RMβCD randomly methylated β-cyclodextrin, CMEβCD carboxymethyl-ethyl-β-cyclodextrin, DEβCD
diethyl-β-cyclodextrin, DMβCD dimethyl-β-cyclodextrin, MLβCD maltosyl-β-cyclodextrin, PMβCD partially methyl-
ated β-cyclodextrin, SBEβCD sulfobutylether β-cyclodextrin, βCD-polymer β-cyclodextrin polymer, γCD γ-cyclodextrin,
HPγCD 2-hydroxypropyl-γ-cyclodextrin
14  Formulation of Drug-Cyclodextrin Complexes 195

the chemical potential in the other phase (e.g., the analogous to electric circuits (Higuchi 1960).
oil phase or the membrane itself (μo)). Later drug permeation through biological mem-
Thermodynamic activity is equal to unity in satu- branes was described mathematically as drug
rated solutions, and, thus, many ointments and permeation through a lipophilic membrane sand-
creams consist of finely divided drug suspensions. wiched between unstirred water layers (UWLs)
Under such conditions, the vehicle is saturated emphasizing that the UWL must be treated as a
with drug, and dissolved drug molecules are at part of the total membrane barrier (Zwolinski
their highest potential to leave the vehicle and par- et al. 1949; Flynn et al. 1972; Flynn and
tition into the skin. Addition of solubilizers, such Yalkowsky 1972; Loftsson et al. 2007b). Here a
as cyclodextrins, to an aqueous drug solution will simple two-barrier model will be used to explain
lower the drug activity (i.e., lowers γw in Eq. 14.10), how cyclodextrins affect drug permeation from
and, thus, under normal conditions, cyclodextrins an aqueous vehicle into and through the skin or
lower the potential of the drug to exit the formula- other biological membranes (Fig. 14.3) (Loftsson
tion (Másson et al. 2005). However, addition of and Brewster 2011). In this model, the drug mol-
cyclodextrin to aqueous drug suspension, increas- ecules encounter two barriers on their way from
ing the amount of dissolved drug while keeping the vehicle through a lipophilic membrane. The
the solution saturated with drug, will not lower first one is the aqueous boundary layer at the
the drug activity as long as solid drug is present membrane surface, the UWL. The second one is
in the aqueous suspension. Under such condition, the lipophilic membrane itself, frequently identi-
the thermodynamic activity (aw in Eq. 14.10) will fied as the outermost layer of the skin, stratum
remain equal to unity, and, thus, dissolved drug corneum. The total skin barrier towards drug per-
molecules are at their highest “exiting” potential, meation consists of the UWL and the lipophilic
while total amount of dissolved drug is increased. membrane. Assuming independent and additive
Adding too much cyclodextrin to an aqueous der- resistances of the two layers, the total drug per-
mal formulation will, on the other hand, decrease meation resistance (RT) of this simple membrane
the activity (aw) below unity and, consequently, can be defined as:
result in less than optimum topical bioavailability.
RT = RD + RM (14.12)
Although passive diffusion is driven by the gradi-
ent of chemical potential, it is common to replace where RD and RM are the drug permeation resis-
it by the concentration gradient. For example, tances in the UWL at the exterior and within the
according to Fick’s first law, the driving force for lipophilic membrane, respectively. Since the per-
steady-state drug diffusion between two points meability constants (P) are the reciprocals of the
(i.e., from point 1 to point 2) in a solution is the resistances, the following equation is obtained
concentration gradient: assuming sink conditions (i.e., CV – CD ≈ CV and
C1 – C2 ≈ C1 in Fig. 14.3):
D × ( C1 - C2 ) -1
J= (14.11) æ 1 1 ö
J = PT × CV = ( RD + RM )
-1
h × CV = ç + ÷ × CV
P
è D PM ø

where J is the drug flux, D is the drug diffusion
(14.13)
constant, C1 and C2 are the drug concentrations at
point 1 and point 2, respectively, and h is the dis- where J is the drug flux from the aqueous vehicle
tance between the two points. through the membrane, PT is the overall permea-
Most biological membranes are multilayer bility coefficient, CV is the concentration of the
membrane barriers, and most contain various dif- compound in the aqueous vehicle, and PD and PM
fusion pathways and transport systems. Higuchi are the permeability coefficients in the UWL and
described passive drug transport through multi- within the membrane, respectively. Rearranging
layer barriers as series of additive resistances Eq. 14.13 gives:
196 T. Loftsson

Fig. 14.3  Drug permeation through a simple two-layer UWL (D) and the membrane (M), respectively. CV is the
barrier where an unstirred water layer (UWL) forms an drug concentration in the vehicle, CD is the drug concen-
aqueous diffusion barrier at the vehicle – skin surface and tration in the UWL immediate to the membrane surface,
a skin barrier (stratum corneum) that is a lipophilic mem- C1 and C2 are the drug concentrations within the mem-
brane barrier. The vehicle contains the dissolved drug; RD, brane, and KM/D is the drug partition coefficient between
hD, RM, and hM are the resistance and the thickness of the the membrane and the UWL

æ P ×P ö
J = ç D M ÷ × CV (14.14) controlled. The relationship between the perme-
è PD + PM ø ation coefficient (P) and the diffusion coefficient
(D) is given by Eq. 14.17:
If permeation is much slower through the mem-
D⋅K
brane itself than the UWL (i.e., PD > PM), then: P = (14.17)
h
æ P ×P ö where h is the thickness of the UWL (hD) or the
J »ç D M ÷ × CV = PM × CV (14.15)
è PD ø lipophilic membrane (hM) and K is either the par-
tition coefficient between the membrane and the
In that case, stratum corneum is the main barrier, UWL (KM/D) or equal to unity (i.e., K = 1.00) as in
and the UWL has negligible effect on the drug the case of the UWL. Finally, D can be estimated
permeation through the membrane and can be from the Stokes-Einstein equation:
ignored (i.e., RM > RD). If, on the other hand, per-
R ⋅T
meation through the lipophilic membrane, i.e., D≈ (14.18)
the skin itself, is much faster than permeation 6p ⋅h ⋅ r ⋅ N
through the UWL (i.e., PM > PD), then: where R is the molar gas constant, T is the abso-
lute temperature, η is the apparent viscosity
æ P ×P ö within the UWL or the lipophilic membrane, r is
J » ç D M ÷ × CV = PD × CV (14.16)
è PM ø the radius of the permeating drug molecule, and
N is Avogadro’s number. Thus, the diffusion con-
In this case, the UWL is the main barrier (i.e., stant within the UWL (DD) will decrease with
RD > RM), and drug permeation through the increasing viscosity of the layer as well as with
membrane becomes aqueous diffusion layer increasing molecular weight of the drug.
14  Formulation of Drug-Cyclodextrin Complexes 197

14.2.2 Cyclodextrins and Biological triglycerides in the gastrointestinal tract and pre-


Membranes vent their absorption (Comerford et al. 2011;
Artiss et al. 2006). Hydrophilic cyclodextrins
The effects of cyclodextrins on drug permeation have been added to sunscreen formulations to
through the skin, mucus membranes, and various reduce absorption of lipophilic sunscreen agents
artificial and biological membranes have been thor- into the skin (Felton et al. 2002, 2004; Sarveiya
oughly reviewed (Matsuda and Arima 1999; Loftsson et al. 2004; Yang et al. 2008). Cyclodextrins, like
and Masson 2001; Loftsson et al. 2007b; Cal and HPβCD and γCD (calculated LogKo/w ≈ −17),
Centkowska 2008; Loftsson and Brewster 2011). have been used to reduce absorption of the mos-
Based on these studies, some general remarks can be quito repellent N,N-diethyl-3-methylbenzamide
made on how and when cyclodextrins enhance drug (DEET) through the skin (Proniuk et al. 2002).
delivery into and through biological membranes. Cyclodextrins can likewise be used to decrease
dermal and transdermal uptake of sunscreen
14.2.2.1  The Drug Molecules Have agents (Cal and Centkowska 2008; Berbicz et al.
to Be Released 2011). The key factor here is to use excess
from the Complex amounts of cyclodextrins in the aqueous vehicle,
Hydrophilic cyclodextrins and their complexes do i.e., more than what is needed to solubilize the
not, in general, permeate lipophilic biomembranes poorly soluble lipophilic agent. This is done to
(i.e., their KM/D ≈ 0; Fig.  14.3 and Eq. 14.17). The reduce the amount of free agent (i.e., drug, mos-
LogKo/w of cyclodextrins that are currently used in quito repellent, and sunscreen agent) present in
pharmaceutical formulations is very low (≤ −6; the formulation, thus reducing its partition into
Table 14.1 and Eq. 14.10), and, thus, these cyclo- the skin. In other words, addition of excess
dextrins and their complexes have virtually no ten- cyclodextrin to the vehicle will lower the poten-
dency to partition from the aqueous exterior into tial of the drug to exit the formulation (Eq. 14.10).
lipophilic membrane. There are no reports of trans-
porter-mediated permeation of cyclodextrins 14.2.2.3  Cyclodextrins Only Enhance
across biological membranes, and in general, the Drug Permeation
oral bioavailability of cyclodextrins is well below from Aqueous Vehicles
4 % (Kurkov and Loftsson 2013). Only about In general, cyclodextrins are unable to enhance
0.02 % of topically applied HPβCD (calculated drug delivery from nonaqueous vehicles through
LogKo/w ≈ −11) is absorbed into intact hairless biomembranes but enhance delivery of lipophilic
mouse skin (Tanaka et al. 1995). Consequently, the drugs when an aqueous phase is in contact with
drug molecules have to be released from the com- the lipophilic membrane surface. Hydrophilic
plexes before they can permeate biological mem- cyclodextrins can enhance drug release from
branes (Loftsson and Brewster 2011). Some hydrophilic creams, i.e., oil-in-water emulsions,
lipophilic cyclodextrin derivatives are, however, but frequently decrease drug release and perme-
able to penetrate into lipophilic membranes (e.g., ation from lipophilic creams, i.e., water-in-oil
the nasal mucosa) and act as conventional chemi- emulsions (Preiss et al. 1994, 1995; Loftsson and
cal penetration enhancers, increasing drug perme- Brewster 2011). Thus, cyclodextrins can be good
ation by reducing the lipophilic membrane barrier. permeation enhancers for dermal drug delivery
from hydrophilic creams, hydrophilic ointments,
14.2.2.2  Cyclodextrins Can Prevent hydrophilic gels, aqueous lotions, foams, sham-
Drug Permeation poos, and solutions, but they will most likely have
Cyclodextrins can prevent drug permeation no effect when included in lipophilic creams,
through biological membranes. For example, hydrophobic ointments, and lipophilic gels. For
tablets containing large amounts of αCD (calcu- definition of these pharmaceutical vehicles, see
lated LogKo/w ≈  −13) are used to complex the European Pharmacopoeia, 8th Edition, 2014.
198 T. Loftsson

There are few examples where cyclodextrins to enhanced drug stability through complexation,
can enhance drug delivery to the skin from non- especially in the case of proteins and peptides (Irie
aqueous vehicles. Such effects are usually related and Uekama 1997; Loftsson and Brewster 2011).
to increased chemical (e.g., prevention of drug
degradation) or physical (e.g., inhabitation of 14.2.2.6  In Combination
crystal growth) drug stability within the vehicles with Conventional
(Frömming and Szejtli 1994). Penetration Enhancers
Cyclodextrins Can Have
Additive Effect
14.2.2.4  Cyclodextrins Do Not Enhance Cyclodextrins and conventional penetration
Delivery of Hydrophilic Drugs enhancers, like fatty acids, or mechanical enhanc-
In general, hydrophilic water-soluble drugs have ers, like iontophoresis, can have additive or syner-
little tendency to form hydrophilic cyclodextrin gistic effect on drug delivery through biological
complexes, and, in general, cyclodextrins do not membranes (Adachi et al. 1992, 1993; Uekama
enhance transmembrane delivery of water-­ et al. 1992; Loftsson et al. 1998; Sinha et al. 2003;
soluble drugs. However, cyclodextrins can form Karandea and Mitragotri 2009). Most often the
complexes with lipophilic moieties of water-­ cyclodextrins increase drug availability at the skin
soluble drugs, and, thus, in some cases cyclodex- surface, while the other enhancers decrease the
trin can reduce topical availability of water-soluble membrane barrier itself. In some cases, cyclodex-
drugs. For example, cyclodextrins form com- trins increase delivery of a lipophilic p­ enetration
plexes with water-soluble β-blockers (Gagyi enhancer to the skin surface (Adachi et al. 1993).
et al. 2008), and HPβCD has been shown to In other cases, cyclodextrin complexation of a
reduce ocular bioavailability of the water-soluble penetration enhancer decreases its skin-­irritating
β-blocker timolol maleate in aqueous eye drop effect without decreasing its penetration-­
formulation (Loftsson and Stefánsson 1997). The enhancing property (Martini et al. 1996).
HPβCD complexation of timolol does increase
the hydrophilicity of timolol (i.e., lowers KM/D in
Fig. 14.3) and increases the hydrodynamic radius 14.3 Formulation Optimization
(i.e., r in Eq. 14.18) of the permeating species,
both of which will result in lower membrane and In general, stratum corneum is the main barrier
transmembrane diffusion of timolol. Few studies towards drug permeation into and across the skin,
have indicated that the somewhat lipophilic meth- and the UWL at the skin surface is very thin.
ylated cyclodextrins (like RMβCD in Table 14.1) Thus, drug permeation from topically applied
are, under certain conditions, able to act as con- drug formulations through intact skin most often
ventional chemical penetration enhancers, that is, follows Eq. 14.15. However, under certain condi-
by penetrating into the skin, and decrease its tions, cyclodextrins are able to enhance dermal
membrane barrier towards drug penetration and transdermal drug delivery. Furthermore,
(Babu and Pandit 2004; Babu et al. 2008). since cyclodextrin complexes tend to self-­
assemble in aqueous solutions to form nanopar-
14.2.2.5  Cyclodextrins Can Enhance ticles, they are known to target drug delivery to
Transmembrane Delivery the sweat ducts, hair follicles, and sebaceous
of Drugs by Increasing Their glands (i.e., drug delivery via shunt route pene-
Chemical Stability tration) (Konrádsdóttir et al. 2009).
Cyclodextrins are able to increase chemical
stability of drugs in aqueous solutions and prevent
enzymatic degradation of drugs at aqueous mem- 14.3.1 When Can Cyclodextrin Help?
brane exterior (Loftsson 1995; Loftsson and Brewster
1996, 2010). The enzymatic activity at some muco- Cyclodextrins only enhance drug delivery from
sal membranes can be quite high, and, thus, the aqueous vehicles and only when a UWL presents
observed permeation enhancement is sometimes due a barrier towards uptake of drug molecules into
14  Formulation of Drug-Cyclodextrin Complexes 199

the skin. Frequently, dermal formulations contain aqueous vehicle, more than what is needed to
little or no water (e.g., hydrophobic ointments solubilize the drug (i.e., excess cyclodextrin low-
and lipophilic gels), and sometimes the water ers the value of KM/D in Eq. 14.17; Fig. 14.3).
domains are not in contact with the skin surface
(e.g., in lipophilic creams that consist of water-
in-­oil emulsions). Under such conditions, the 14.3.3 How to Optimize
UWL is very thin (hD in Fig. 14.3) and, thus, does the Formulation?
not present a barrier (i.e., the skin permeation fol-
lows Eq. 14.15). However, many aqueous dermal It is important to optimize cyclodextrin-­containing
formulations, such as hydrophilic creams (i.e., vehicles with regard to the vehicle composition
oil-in-water emulsions) and hydrophilic gels, and the desired effect. Too little or too much
increase the thickness of the UWL in which case cyclodextrin will result in less than optimum effect
the resistance of the UWL (RD in Eq. 14.12 and (Loftsson and Brewster 2011). Here we describe
Fig.  14.3) can become comparable or greater step by step the formulation of hydrophilic hydro-
than the resistance of stratum corneum (RM in cortisone gel. The hydrophilic gel (hydrogel)
Eq. 14.12 and Fig. 14.3). Under such conditions, vehicle consists of water within a starch matrix
cyclodextrins can enhance drug permeation from (0.5–2 %) containing HPβCD as a solubilizer/pen-
the surface into the skin. Sweat can also increase etration enhancer. Similar methods are used to
the thickness of the UWL between a water-free optimize other aqueous skin preparations.
drug donor, such as dermal patch, and the skin
surface (i.e., increasing RD). 14.3.3.1  Phase-Solubility Study
Skin damage due to disease or injury can One hydrocortisone molecule (MW 362.5 Da)
reduce its barrier function (i.e., RM in Eq. 14.12) forms an inclusion complex with one HPβCD
and increase drug permeation through the skin molecule (MW 1400 Da). In aqueous solutions,
(i.e., increase PM in Eqs. 14.13 and 14.14). Under the inclusion complexes are constantly being
such conditions, permeation through UWL might formed and dissociated at rates close to the
be the main barrier towards dermal and transdermal diffusion-­controlled limit, and, thus, the com-
drug delivery (i.e., PM > PD) in which case the plexes are in dynamic equilibrium with free
drug flux into and through the skin follows hydrocortisone and HPβCD molecules (Fig. 14.1)
Eq.  14.16, creating conditions where cyclodex- (Stella et al. 1999). The first step is to determine
trins are known to act as penetration enhancers of how much HPβCD is needed to dissolve given
lipophilic and poorly water-soluble drugs. amount of hydrocortisone. This is done by deter-
mining the phase solubility of the drug in aque-
ous solution (Higuchi and Connors 1965;
14.3.2 What Is the Desired Effect? Loftsson et al. 2007a; Loftsson and Brewster
2010; Loftsson and Hreinsdóttir 2006). The
Most often the aim is to deliver drug molecules aqueous solubility of hydrocortisone is deter-
from the vehicle into and through the skin. In that mined as the function of HPβCD concentration.
case, it is important to include in the vehicle suf- From the linear phase-solubility (i.e., AL-type)
ficient amount of cyclodextrin to enhance drug diagram in Fig. 14.4, we see that we will need
delivery to the skin surface and into the skin but about 7 % (w/v) HPβCD to dissolve 10 mg/ml
to avoid excess amounts. For shunt delivery, the (i.e., 1 % w/v) of hydrocortisone, about 11 % to
total cyclodextrin concentration, or rather the dissolve 15 mg/ml, and about 14 % to dissolve
total concentration of drug-cyclodextrin com- 20 mg/ml. To calculate the stability constant
plexes, has to be sufficient for formation of (K1:1) and the complexation efficiency (CE), we
nanoparticles (Messner et al. 2011; Kurkov and need to determine the phase-solubility diagram
Loftsson 2013). Still in other cases the target is using molar concentrations (Fig. 14.5). From the
the skin surface itself, and then cyclodextrins can slope (0.5432), we can determine the CE accord-
be used to prevent drug partition into the skin ing to Eq. 14.6 (CE = 1.19) and the hydrocorti-
adding excess amounts of cyclodextrins to the sone/HPβCD molar ratio in the aqueous HPβCD
200 T. Loftsson

solution saturated with hydrocortisone from 14.3.3.2  The Amount of Cyclodextrin


Eq.  14.7 (about 1:2). Thus, in aqueous HPβCD and Drug Availability
solution at room temperature, at least two HPβCD The starch (e.g., hydroxypropyl cellulose) used
molecules are needed to dissolve one molecule of to form the matrix might decrease or increase the
hydrocortisone. Then according to Eq. 14.5, we amount of HPβCD needed to solubilize hydro-
can calculate K1:1 from the slope and the hydro- cortisone (Loftsson and Brewster 2012).
cortisone solubility in the aqueous complexation However, it can be difficult to determine hydro-
media when no HPβCD is present (1.15·10−3 M), cortisone solubility in a viscous gel. Alternatively,
the observed K1:1 = 1,030  M−1. one can determine the effect of HPβCD
concentration on hydrocortisone release. The
phase-­solubility study shows that about 11 %
(w/v) HPβCD will be needed to dissolve 15 mg/ml
(1.5 % w/v) hydrocortisone in the hydrophilic
gel. To determine the exact amount of HPβCD
needed, a series of gels are prepared, all of which
contain the same amount of starch and hydrocor-
tisone (1.5 % w/v) but different amounts
(5–15 % w/v) of HPβCD, and the hydrocortisone
permeation from the gels through an artificial
membrane was determined (Fig. 14.6). The
membrane consisted of semipermeable cello-
phane membrane with an octanol/nitrocellulose

Fig. 14.4  The phase-solubility diagram of hydrocorti-


sone in pure water-containing HPβCD at room tempera-
ture (22–23 °C)

Fig. 14.6  Effect of HPβCD concentration on the hydro-


cortisone flux from a hydrophilic gel vehicle through an
artificial biomembrane at room temperature (22–23 °C).
The membrane consisted of semipermeable cellophane
membrane (MWCO 12,000–14,000) with an octanol/
nitrocellulose membrane fused to the receptor side. The
gel contained fixed amount of hydrocortisone, 1.5 %
(w/v). Both free hydrocortisone and the hydrocortisone/
Fig. 14.5  The phase-solubility diagram of hydrocorti- HPβCD complex were able to permeate the cellulose
sone in pure water-containing HPβCD at room tempera- membrane, but only the drug was able to permeate the
ture (22–23 °C) octanol/nitrocellulose membrane
14  Formulation of Drug-Cyclodextrin Complexes 201

Fig. 14.7  Permeation of hydrocortisone from a hydro- is unable to permeate into the octanol membrane, while
philic gel containing hydrocortisone/HPβCD complex hydrocortisone is much more lipophilic (KM/D = KO/W ≈ 40)
through a cellophane-octanol membrane. HPβCD is very (Másson et al. 2005) and is able to permeate the octanol
hydrophilic (KM/D = KO/W ≈ 10−11; see Table 14.1) and, thus, membrane. The observed K1:1 = 1,030  M−1

membrane fused to the receptor side. Only the reduces the flux from 205 to 203 mg h−1 cm−2.
free drug is able to permeate the octanol layer The final composition of the hydrophilic gel will
(Fig. 14.7). Permeation of hydrocortisone mole- then be 1.5 % (w/v) hydrocortisone and 12 %
cules from the gel is at its maximum when just (w/v) HPβCD in a hydrophilic gel.
enough HPβCD is present to dissolve all hydro-
cortisone. At lower HPβCD concentration, the 14.3.3.3  What Happens on the Skin?
permeation is lower, and the gel is turbid due to The gel contains about 85 % water. The water
undissolved hydrocortisone. At higher HPβCD content of the gel will decrease relatively rapidly
concentrations, the gel is clear, but excess after its application to the skin surface, due to
amounts of HPβCD will decrease the concentra- both evaporation and water absorption into the
tion of free hydrocortisone at the surface of the skin. However, since hydrocortisone displays AL-­
lipophilic membrane (Fig. 14.7) resulting in type phase-solubility diagram in aqueous HPβCD
decreased hydrocortisone flux through the mem- solutions, decreased amount of water will not
brane. However, to avoid drug precipitation dur- result in hydrocortisone precipitation. The gel
ing storage, the gel should contain a small excess will become stiffer, and the increased viscosity
of HPβCD. Maximum flux (Fig. 14.6) is obtained (η) might decrease hydrocortisone permeation
at 10 % (w/v) HPβCD. Increasing the concentra- from the gel to the skin surface (see Eq. 14.18),
tion to 12 % (w/v) (20 % excess HPβCD) only but increased hydrocortisone concentration could,
202 T. Loftsson

on the other hand, result in increased hydrocorti- Arima H, Motoyama K, Irie T (2011) Recent findings on
safety profiles of cyclodextrins, cyclodextrin conju-
sone permeation. Hence, decreased water content
gates, and polypseudorotaxanes. In: Bilensoy E (ed)
might have less effect than expected. Cyclodextrins in pharmaceutics, cosmetics, and bio-
medicine: current and future industrial applications.
Wiley, Hoboken, pp 91–122
Artiss JD, Brogan K, Brucal M, Moghaddam M, Jen
Conclusions
K-LC (2006) The effects of a new soluble dietary fiber
Cyclodextrins can under certain conditions act on weight gain and selected blood parameters in rats.
as percutaneous penetration enhancers. In Metab Clin Exp 55:195–202
general, cyclodextrins can only enhance drug Ascenso A, Vultos F, Ferrinho D, Salgado A, Filho SG,
Ferrari V et al (2012) Effect of tretinoin inclusion in
delivery through the skin from aqueous vehi-
dimethyl-beta-cyclodextrins on release rate from a hydro-
cles and only when an aqueous diffusion bar- gel formulation. J Incl Phenom Macroc Chem 73:459–465
rier at the skin exterior contributes to the Babu RJ, Pandit JK (2004) Effect of cyclodextrins on the
overall skin permeation barrier. Cyclodextrins complexation and transdermal delivery of bupranolol
through rat skin. Int J Pharm 271:155–165
do not enhance drug penetration from lipo-
Babu RJ, Dhanasekaran M, Vaithiyalingam SR, Singh
philic vehicles or when the skin barrier, i.e., PN, Pandit JK (2008) Cardiovascular effects of trans-
stratum corneum, is the main permeation bar- dermally delivered bupranolol in rabbits: effect of
rier. Cyclodextrins are able to p­ revent drug chemical penetration enhancers. Life Sci 82:273–278
Batzdorf T, Mullergoymann CC (1993) Release of keto-
partition from an aqueous exterior into the
profen from aqueous systems in the presence of hydro-
skin. It is of uttermost importance to optimize philic β−cyclodextrin derivatives. Pharm Ind
composition of cyclodextrin-containing drug 55:857–860
vehicles with regard to drug release and Berbicz F, Nogueira AC, Neto AM, Natali MRM, Baesso
ML, Matioli G (2011) Use of photoacoustic spectros-
permeation.
copy in the characterization of inclusion complexes of
benzophenone-3-hydroxypropyl-β-cyclodextrin and
ex vivo evaluation of the percutaneous penetration of
sunscreen. Eur J Pharm Biopharm 79:499–57
References Bilensoy E (2011) Cyclodextrins in pharmaceutics, cos-
metics, and biomedicine. Current and future industrial
Adachi H, Irie T, Uekama K, Manako T, Yano T, Saita M applications. Wiley, Hoboken
(1992) Inhibitory effect of prostaglandin E1 on Bounoure F, Lahiani-Skiba M, Barbot C, Sughir A, Mallet
laurate-­induced peripheral vascular occlusive sequelae E, Jezequel S et al (2007) Effect of partially methylated
in rabbits; optimized topical formulation with β cyclodextrin on percutaneous absorption of metopi-
β-cyclodextrin derivative and penetration enhancer mazine. J Incl Phenom Macroc Chem 57:191–195
HPE-101. J Pharm Pharmacol 44:1033–1035 Brewster ME, Loftsson T (2007) Cyclodextrins as phar-
Adachi H, Irie T, Uekama K, Manako T, Yano T, Saita M maceutical solubilizers. Adv Drug Deliv Rev
(1993) Combination effects of O-carboxymethyl-O-­ 59:645–666
ethyl-β-cyclodextrin and penetration enhancer HPE-­ Cal K, Centkowska K (2008) Use of cyclodextrins in topi-
101 on transdermal delivery of prostaglandin-E(1) in cal formulations: practical aspects. Eur J Pharm
hairless mice. Eur J Pharm Sci 1:117–123 Biopharm 68:467–478
Amdidouche D, Montassier P, Poelman MC, Duchene D Celebi N, Kislal O, Tarimci N (1993) The effect of
(1994) Evaluation by laser-doppler velocimetry of the β-cyclodextrin and penetration additives on the release of
attenuation of tretinoin induced skin irritation by naproxen from ointment bases. Pharmazie 48:914–917
β-cyclodextrin complexation. Int J Pharm Chang SL, Banga AK (1998) Transdermal iontophoretic
111:111–116 delivery of hydrocortisone from cyclodextrin solu-
Arima H, Adachi H, Kie T, Uekama K, Pitha J (1990) tions. J Pharm Pharmacol 50:635–640
Enhancement of antiinflammatory effect of ethyl Chen C-Y, Chen F-A, Wu A-B, Hsu H-C, Kang J-J, Cheng
4-biphenylyl acetate in ointment by β-cyclodextrin H-W (1996) Effect of hydroxypropyl-β-cyclodextrin
derivatives: increased absorption and localized activa- on the solubility, photostability and in-vitro
tion of the prodrug in rats. Pharm Res 7:1152–1156 ­permeability of alkannin/shikonin enantiomers. Int J
Arima H, Miyaji T, Irie T, Hirayama F, Uekama K (1996) Pharm 141:171–178
Possible enhancing mechanism of the cutaneous per- Comerford KB, Artiss JD, Jen KLC, Karakas SE (2011)
meation of 4-biphenylylacetic acid by β-cyclodextrin The beneficial effects α-cyclodextrin on blood lipids
derivatives in hydrophilic ointment. Chem Pharm Bull and weight loss in healthy humans Obesity
44:582–586 19:1200–1204
14  Formulation of Drug-Cyclodextrin Complexes 203

Dahan A, Miller JM (2012) The solubility-permeability Hegge AB, Schüller RB, Kristensen S, Tønnesen HH
interplay and its implications in formulation design (2008) In vitro release of curcumin from vehicles
and development for poorly soluble drugs. AAPS J ­containing alginate and cyclodextrin. Studies of cur-
14:244–251 cumin and curcuminoides. XXXIII. Pharmazie 63:
Dahan A, Miller JM, Hoffman A, Amidon GE, Amidon 585–592
GL (2010) The solubility–permeability interplay in Higuchi T (1960) Physical chemical analysis of percuta-
using cyclodextrins as pharmaceutical solubilizers: neous absorption process from creams and ointments.
mechanistic modeling and application to progester- J Soc Cosmet Chem 11(2):85–97
one. J Pharm Sci 99:2739–2749 Higuchi T, Connors KA (1965) Phase-solubility tech-
Dodziuk H (ed) (2006) Cyclodextrins and their com- niques. Adv Anal Chem Instrum 4:117–212
plexes. Wiley-VCH Verlag, Weinheim Hincal AA, Eroğlu H, Bilensoy E (2011) Regulatory sta-
Doliwa A, Delgado-Charro B, Santovo S, Ygartua P, Guy tus of cyclodextrins in pharmaceutical products. In:
RH, (eds) (2000) In vitro iontophoretic delivery of Bilensoy E (ed) Cyclodextrins in pharmaceutics, cos-
piroxicam from hydroxypropyl-β-cyclodextrin – metic, and biomedicine: current and future industrial
piroxicam complexes. Proceed. 27th Int'l. Symp. applications. Wiley, Hoboken, pp 123–130
Control. Rel. Bioact. Mater. 7–13 July; Paris: CRS Hirayama F, Yamamoto M, Uekama K (1992) Acid-­
Doliwa A, Santoyo S, Ygartua P (2001) Influence of catalyzed hydrolysis of maltosyl-β-cyclodextrin. J
piroxicam:hydroxypropyl-beta-cyclodextrin complex- Pharm Sci 81:913–916
ation on the in vitro permeation and skin retention of Hymas RV, Ho NFH, Higuchi WI (2012) Transport of a
piroxicam. Skin Pharmacol Appl Skin Physiol lipophilic ionizable permeant (capric acid) across a
14:97–107 lipophilic membrane (silicone polymer membrane)
Dollo G, Corre PL, Chevanne F, Verge RL (1998) from aqueous buffered solutions in the presence of
Complexation between local anaesthetics and hydroxypropyl-β-cyclodextrin. J Pharm Sci
β-cyclodextrin derivatives – relationship between sta- 101:2340–2352
bility constants and in vitro membrane permeability of Idson B (1971) Biophysical factors in skin penetration. J
bupivacaine and lidocaine from their complexes. STP Soc Cosmet Chem 22:615–634
Pharma Sci 8:189–195 Iervolino M, Raghavan SL, Hadgraft J (2000) Membrane
Douhal A (ed) (2006) Cyclodextrin materials photochem- penetration enhancement of ibuprofen using supersat-
istry, photophysics and photobiology. Elsevier, uration. Int J Pharm 198:229–238
Amsterdam Irie T, Uekama K (1997) Pharmaceutical applications of
Felton LA, Wiley CJ, Godwin DA (2002) Influence of cyclodextrins. III. Toxicological issues and safety
hydroxypropyl-β-cyclodextrin on transdermal perme- evaluation. J Pharm Sci 86(2):147–162
ation and skin accumulation of oxybenzone. Drug Karandea P, Mitragotri S (2009) Enhancement of trans-
Devel Ind Pharm 28:1117–1124 dermal drug delivery via synergistic action of chemi-
Felton LA, Wiley CJ, Godwin DA (2004) Influence of cals. Biochim Biophys Acta 1788:2362–2373
cyclodextrin complexation on the in vivo Kawahara K, Ueda H, Tomono K, Nagai T (1992) Effect
­photoprotective effects of oxybenzone. Drug Devel of diethyl β-cyclodextrin on the release and absorption
Ind Pharm 30:95–102 behaviour of indomethacin from ointment bases. STP
Flynn GL, Yalkowsky SH (1972) Correlation and predic- Pharma Sci 2:506–513
tion of mass transport across membranes I: influence Kear CL, Yang J, Godwin DA, Felton LA (2008)
of alkyl chain length on flux-determining properties of Investigation into the mechanism by which cyclodex-
barrier and diffusant. J Pharm Sci 61:838–852 trins influence transdermal drug delivery. Drug Devel
Flynn GL, Carpender OS, Yalkowsky SH (1972) Total Ind Pharm 34:692–697
mathematical resolution of diffusion layer control of Klang V, Matsko N, Zimmermann A-M, Vojnikovic E,
barrier flux. J Pharm Sci 61:312–314 Valenta C (2010) Enhancement of stability and skin
Frömming KH, Szejtli J (1994) Cyclodextrins in phar- permeation by sucrose stearate and cyclodextrins in
macy. Kluwer Academic Publishers, Dordrecht progesterone nanoemulsions. Int J Pharm 393:153–161
Gagyi L, Gyéresi Á, Kilár F (2008) Role of chemical Klang V, Haberfeld S, Hartl A, Valenta C (2012) Effect of
structure in stereoselective recognition of β-blockers γ-cyclodextrin on the in vitro skin permeation of a ste-
by cyclodextrins in capillary zone electrophoresis. J roidal drug from nanoemulsions: impact of experi-
Biochem Biophys Methods 70:1268–1275 mental setup. Int J Pharm 423:535–542
Gerlóczy A, Antal S, Szejtli J (1988) Percutaneous Konrádsdóttir F, Ogmundsdóttir H, Sigurdsson V,
absorption of heptakis-(2,6-di-O-14C-methyl)-β-­ Loftsson T (2009) Drug targeting to the hair follicles:
cyclodextrin in rats. In: Huber O, Szejtli J (eds) a cyclodextrin based drug delivery. AAPS
Proceedings of the fourth international symposium on PharmSciTech 10:266–269
cyclodextrins. Kluwer Academic Publishers, Kurkov SV, Loftsson T (2013) Cyclodextrins. Int J Pharm
Dordrecht, pp 415–420 453:167–180
Hedges AR (1998) Industrial applications of cyclodex- Lee BJ, Cui JH, Parrott KA, Ayres JW, Sack RL (1998)
trins. Chem Rev 98:2035–2044 Percutaneous absorption and model membrane varia-
204 T. Loftsson

tions of melatonin in aqueous-based propylene glycol Loftsson T, Fridriksdottir H, Ingvarsdóttir G, Jónsdóttir B,


and 2-hydroxypropyl-β-cyclodextrin vehicles. Arch Sigurðardottir AM (1994b) The influence of
Pharm Res 21:503–507 2-hydroxypropyl-β-cyclodextrin on diffusion rates
Legendre JY, Rault I, Petit A, Luijten W, Demuynck I, and transdermal delivery of hydrocortisone. Drug Dev
Horvath S et al (1995) Effects of β-cyclodextrins on Ind Pharm 20:1699–1708
skin: implications for the transdermal delivery of piri- Loftsson T, Sigurðardóttir AM, Ólafsson JH (1995)
bedil and a novel cognition enhancing-drug, S-9977. Improved acitretin delivery through hairless mouse
Eur J Pharm Sci 3:311–322 skin by cyclodextrin complexation. Int J Pharm
Loftsson T (1995) Effects of cyclodextrins on chemical 115:255–258
stability of drugs in aqueous solutions. Drug Stab Loftsson T, Masson M, Sigurdsson HH, Magnusson P, Goffic
1:22–33 FL (1998) Cyclodextrins as co-enhancers in dermal and
Loftsson T (2012) Drug permeation through biomem- transdermal drug delivery. Pharmazie 53:137–139
branes: cyclodextrins and the unstirred water layer. Loftsson T, Brewster ME, Másson M (2004) Role of
Pharmazie 67:363–370 cyclodextrins in improving oral drug delivery. Am J
Loftsson T, Bodor N (1994) The pharmacokinetics and Drug Deliv 2:261–275
transdermal delivery of loteprednol etabonate and Loftsson T, Hreinsdóttir D, Másson M (2005) Evaluation
related soft steroids. Adv Drug Deliv Rev 14:293–299 of cyclodextrin solubilization of drugs. Int J Pharm
Loftsson T, Brewster ME (1996) Pharmaceutical applica- 302:18–28
tions of cyclodextrins. 1. Drug solubilization and sta- Loftsson T, Hreinsdóttir D, Másson M (2007a) The com-
bilization. J Pharm Sci 85(10):1017–1025 plexation efficiency. J Incl Phenom Macroc Chem
Loftsson T, Brewster ME (2010) Pharmaceutical applica- 57:545–552
tions of cyclodextrins: basic science and product Loftsson T, Vogensen SB, Brewster ME, Konráðsdóttir F
development. J Pharm Pharmacol 62:1607–1621 (2007b) Effects of cyclodextrins on drug delivery through
Loftsson T, Brewster ME (2011) Pharmaceutical applica- biological membranes. J Pharm Sci 96:2532–2546
tions of cyclodextrins: effects on drug permeation Loftsson T, Sigurðsson HH, Konráðsdóttir F, Gísladóttir
through biological membranes. J Pharm Pharmacol S, Jansook P, Stefánsson E (2008) Topical drug deliv-
63:1119–1135 ery to the posterior segment of the eye: anatomical and
Loftsson T, Brewster ME (2012) Cyclodextrins as func- physiological considerations. Pharmazie 63:171–179
tional excipients: methods to enhance complexation Lopez RFL, Collett JH, Bentley MVLB (2000) Influence
efficiency. J Pharm Sci 101:3019–3032 of cyclodextrin complexation on the in vitro perme-
Loftsson T, Duchêne D (2007) Cyclodextrins and their ation and skin metabolism of dexamethasone. Int J
pharmaceutical applications. Int J Pharm 329:1–11 Pharm 200:127–132
Loftsson T, Hreinsdóttir D (2006) Determination of aque- Martini A, Artico R, Civaroli P, Muggetti L, De Ponti R
ous solubility by heating and equilibration: a technical (1996) Critical micellar concentration shifting as a
note. AAPS PharmSciTech. 7(1): www.aapspharmsci- simple tool for evaluating cyclodextrin/enhancer inter-
tech.org actions. Int J Pharm 127:239–344
Loftsson T, Masson M (2001) Cyclodextrins in topical Masson M, Loftsson T, Masson G, Stefansson E (1999)
drug formulations: theory and practice. Int J Pharm Cyclodextrins as permeation enhancers: some theo-
225:15–30 retical evaluations and in vitro testing. J Control Rel
Loftsson T, Sigurdardottir AM (1994) The effect of poly- 59:107–118
vinylpyrrolidone and hydroxypropyl methylcellulose Másson M, Sigurdardóttir BV, Matthíasson K, Loftsson T
on HPβCD complexation of hydrocortisone and its (2005) Investigation of drug-cyclodextrin complexes
permeability through hairless mouse skin. Eur J Pharm by a phase-distribution method: some theoretical and
Sci 2:297–301 practical considerations. Chem Pharm Bull
Loftsson T, Sigurðardottir AM (1994) The effect of poly- 53:958–964
vinylpyrrolidone and hydroxypropyl methylcellulose Matsuda H, Arima H (1999) Cyclodextrins in transdermal
on HPβCD complexation of hydrocortisone and its and rectal delivery. Adv Drug Deliv Rev 36:81–99
permeability through hairless mouse skin. Eur J Pharm Messner M, Kurkov SV, Brewster ME, Jansook P,
Sci 2:297–301 Loftsson T (2011) Self-assembly of cyclodextrin com-
Loftsson T, Stefánsson E (1997) Effect of cyclodextrins plexes: aggregation of hydrocortisone/cyclodextrin
on topical drug delivery to the eye. Drug Devel Ind complexes. Int J Pharm 407:174–183
Pharm 23:473–481 Montassier P, Duchene D, Poelman MC (1998) In vitro
Loftsson T, Ólafsdóttir BJ, Bodor N (1991) The effects of release study of tretinoin from tretinoin/cyclodextrin
cyclodextrins on transdermal delivery of drugs. Eur J derivative complexes. J Inclus Phenom Mol
Pharm Biopharm 37:30–33 31:213–218
Loftsson T, Fridriksdottir H, Ingvarsdottir G, Jonsdottir B, Munro IC, Newberne PM, Young RR, Bär A (2004)
Sigurdardottir AM (1994a) The influence of Safety assessment of γ-cyclodextrin. Regul Toxicol
2-hydroxypropyl-beta-cyclodextrin on diffusion rates Pharmacol 39(Suppl 1):S3–S13
and transdermal delivery of hydrocortisone. Drug Dev Okamoto H, Komatsu H, Hashida M, Sezaki H (1986)
Ind Pharm 20(9):1699–1708 Effects of β-cyclodextrin and di-O-methyl-β-
14  Formulation of Drug-Cyclodextrin Complexes 205

cyclodextrin on the percutaneous absorption of butyl- Tanaka M, Iwata Y, Kouzuki Y, Taniguchi K, Matsuda H,
paraben, indomethacin and sulfanilic acid. Int J Pharm Arima H et al (1995) Effect of
30:35–45 ­2-hydroxypropyl-β-­ cyclodextrin on percutaneous
Preiss A, Mehnert W, Frömming KH (1994) In-vitro absorption of methyl paraben. J Pharm Pharmacol
hydrocortisone release from ointments in presence of 47:897–900
cyclodextrins. Pharmazie 49:902–906 Tenjarla S, Puranajoti P, Kasina R, Mandal T (1998)
Preiss A, Mehnert W, Frömming K-H (1995) Penetration Preparation, characterization, and evaluation of
of hydrocortisone into excised human skin under the miconazole-cyclodextrin complexes for improved oral
influence of cyclodextrins. Pharmazie 50:121–126 and topical delivery. J Pharm Sci 87:425–429
Proniuk S, Liederer BM, Dixon SE, Rein JA, Kallen MA, Uekama K, Otagiri M, Sakai A, Irie T, Matsuo N,
Blanchard J (2002) Topical formulation studies with Matsuoka Y (1985) Improvement in the percutaneous
DEET (N, N-diethyl-3-methylbenzamide) and cyclo- absorption of beclomethasone dipropionate by
dextrins. J Pharm Sci 91:101–110 γ-cyclodextrin complexation. J Pharm Pharmacol
Sabadini E, Cosgrove T, Egídio FC (2006) Solubility of 37:532–535
cyclomaltooligosaccharides (cyclodextrins) in H2O Uekama K, Arimori K, Sakai A, Masaki K, Irie T, Otagiri
and D2O: a comparative study. Carbohydr Res M (1987) Improvement of percutaneous absorption of
341:270–274 prednisolone by β- and γ-cyclodextrin complexations.
Sarveiya V, Templeton JF, Benson HAE (2004) Inclusion Chem Pharm Bull 35:2910–2913
complexation of the sunscreen 2-hydroxy-4-methoxy Uekama K, Adachi H, Irie T, Yano T, Saita M, Noda K
benzophenone (oxybenzone) with hydroxypropyl-β-­ (1992) Improved transdermal delivery of prostaglan-
cyclodextrin: effect on membrane diffusion. J Incl din E1 through hairless mouse skin: combined use of
Phenom Macroc Chem 49:275–281 carboxymethyl-ethyl-β-cyclodextrin and penetration
Schönberger BP, Jansen ACA, Janssen LHM (eds) (1988) enhancers. J Pharm Pharmacol 44:119–121
The acid hydrolysis of cyclodextrins and linear oligo- Uekama K, Hirayama F, Arima H (2006) Pharmaceutical
saccharides, a comparative study. 4th Int. Symp. on applications of cyclodextrins and their derivatives. In:
Cyclodextrins. Kluwe, Munich Dodziuk H (ed) Cyclodextrins and their complexes.
Shakory R, Khodabandeh M, Toliyat T, Azimzadeh S, Chemistry, analytical methods, applications. Wiley-­
Sadigh Z-A, Badiefar L (2010) Enhancing effect of VCH Verlag, Weinheim, pp 381–422
cyclodextrins on in vitro skin permeation of hGH. Curr Ventura CA, Tommasini S, Falcone A, Giannone I,
Trends Biotechnol Pharm 4:784–790 Paolino D, Sdrafkakis V et al (2006) Influence of mod-
Sigurdardottir AM, Loftsson T (1995) The effect of poly- ified cyclodextrins on solubility and percutaneous
vinylpyrrolidone on cyclodextrin complexation of absorption of celecoxib through human skin. Int J
hydrocortisone and its diffusion through hairless Pharm 314:37–45
mouse skin. Int J Pharm 126:73–78 Vollmer V, Müller BW, Mesens J, Wilffert B, Peters T
Sinha VR, Bindra S, Kumria R, Nanda A (2003) (1993) In vivo skin pharmacokinetics of liarozole: per-
Cyclodextrin as skin-penetration enhancers. Pharm cutaneous absorption studies with different formula-
Technol 27(3):120–138 tions of cyclodextrin derivatives in rats. Int J Pharm
Stella VJ, He Q (2008) Cyclodextrins. Tox Pathol 99:51–58
36:30–42 Yang J, Wiley CJ, Godwin DA, Felton LA (2008)
Stella VJ, Rao VM, Zannou EA, Zia V (1999) Mechanism Influence of hydroxypropyl-β-cyclodextrin on trans-
of drug release from cyclodextrin complexes. Adv dermal penetration and photostability of avobenzone.
Drug Deliv Rev 36:3–16 Eur J Pharm Biopharm 69:605–612
Szejtli J (1987) The metabolism, toxicity and biological Yuzuriha S, Matsuo K, Noguchi M (1999) Topical appli-
effects of cyclodextrins. In: Duchêne D (ed) cation of prostaglandin E1 ointment to cutaneous
Cyclodextrins and their uses. Editions de Santé, Paris, wounds in ischemic rabbit ears. Eur J Plastic Surg
pp 173–212 22:225–229
Szejtli J (1988) Cyclodextrin technology. Kluwer Zi P, Yang X, Kuang H, Yang Y, Yu L (2008) Effect of
Academic Publisher, Dordrecht HPβCD on solubility and transdermal delivery of cap-
Szeman J, Ueda H, Szejtli J, Fenyvesi E, Watanabe Y, saicin through rat skin. Int J Pharm 358:151–158
Machida Y et al (1987) Enhanced percutaneous Zwolinski BJ, Eyring H, Reese CE (1949) Diffusion and
absorption of homogenized tolnaftate/β-cyclodextrin membrane permeability. I. J Phys Coll Chem 53:
polymer ground mixture. Drug Design Deliv 1426–1453
1:325–332
Part IV
Influence of Vehicle Effects in Penetration
Enhancement
Dermal and Transdermal
Formulations: How They Can 15
Affect the Active Compound

Jessica Stahl

Contents 15.1 Introduction


15.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . 209
The development and distribution of dermal and
15.2 Vehicle Composition . . . . . . . . . . . . . . . . 211
15.2.1 Bases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 211
transdermal therapeutics increased in the last
15.2.2 Gelling Agents . . . . . . . . . . . . . . . . . . . . . 212 decades. Their high popularity is based on the easy
15.2.3 Preservatives (Antimicrobial Agents) . . . . 212 way of application, painless treatment, and reduced
15.2.4 Antioxidants . . . . . . . . . . . . . . . . . . . . . . . 212 side effects. Topically applied formulations shall
15.2.5 Emulsifier . . . . . . . . . . . . . . . . . . . . . . . . . 212
exhibit a high compatibility with low side effects,
15.3 Formulation Types . . . . . . . . . . . . . . . . . 212 high patient acceptance, and adequate shelf life.
15.3.1 Semisolid Formulations . . . . . . . . . . . . . . 213
Furthermore, they have to stay on the skin for a suf-
15.3.2 Liquid Formulations . . . . . . . . . . . . . . . . . 213
15.3.3 Special Formulation Types . . . . . . . . . . . . 214 ficient time in order to provide enough time for the
active compound to diffuse through the formulation
15.4  he Influence of Formulations
T
on Drug Diffusion into and through to the skin (Wiechers et al. 2004). Afterward, the
the Skin . . . . . . . . . . . . . . . . . . . . . . . . . . 214 active compound partitions into the skin (drug lib-
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218
eration) (Fig. 15.1). In dependence on the medical
indication, the active compound has to be delivered
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 218 to the site of action inside the epidermis (drug pen-
etration) or dermis (drug permeation) and has to
remain there for a sufficient time, in order to ensure
a local treatment (Wiechers et al. 2004). On the
other hand, a systemic effect can be aimed, when
the active compound has to be delivered through the
skin into systemic circulation (drug resorption) in
enough high concentrations to provide sufficient
plasma levels (Daniels and Knie 2007).
The ability of a drug to be liberated from the
vehicle can be characterized by the ratio of the
solubility of the active compound in the stratum
J. Stahl corneum (CSC) to the solubility of the active com-
Department of Pharmacology, Toxicology and pound in the vehicle (CV). This dependency can
Pharmacy, University of Veterinary Medicine
Hannover, Foundation, Hannover, Germany be described by the partition coefficient KSC/V
e-mail: jessica.stahl@tiho-hannover.de according to Eq. 15.1 (Wiechers et al. 2004).

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 209


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_15, © Springer-Verlag Berlin Heidelberg 2015
210 J. Stahl

Liberation Penetration Permeation Absorption

Stratum corneum

Epidermis

Dermis

Fig. 15.1  Pathways of a compound from the topically applied product to the intended localization (Modified after
Daniels and Knie (2007))

KSC/ V = CSC / CV (15.1) where J is solute flux, D is diffusion coefficient,



Kow is octanol/water partition coefficient, Δc is
Thus, the drug amount which is liberated from drug concentration difference between the for-
the formulation into the stratum corneum can be mulation and the deepest skin layer, and x is the
enhanced either by increasing the solubility of length of the diffusion pathway of the active
the compound in the stratum corneum or by compound.
decreasing the solubility of the compound in the Furthermore, the permeability coefficient Kp
formulation (Wiechers et al. 2004). is often used in order to simplify the permeation
Topically applied compounds have to over- rate of an active compound through the skin
come the main skin barrier, the stratum corneum, (Eq.  15.3) (Hadgraft and Pugh 1998), since the
which represents a complex mixture of various actual diffusion pathway through the skin (x) is
lipid and protein domains (Cross et al. 2001) in unknown. Kp is furthermore the parameter used
order to reach the underlying regions (Hadgraft for statistical analysis of results of skin diffusion
and Pugh 1998). Thus, two processes, partition studies.
and diffusion, are important determinants for
drug delivery into and through the skin (Wiechers K p = ( D ⋅ K ow ) / x cm ⋅ s −1  (15.3)

et al. 2004), which in turn can be found in the flux
of a compound according to Fick’s law of diffu- The number of transdermal applied compounds is
sion (Eq. 15.2) (Scheuplein 1976; Potts and Guy still limited due to the lack of achieving therapeutic
1992): concentrations in the target tissue within the skin or
in other body tissues after systemic circulation via
J = ( D ⋅ K ow ⋅ ∆c ) / x  mol ⋅ s -1 ⋅ cm -2  (15.2) blood or the lymphatic system (Bach and Lippold
1998), usually due to limitations in transdermal
15  Dermal and Transdermal Formulations: How They Can Affect the Active Compound 211

permeation caused by physicochemical drug char- et al. 2004). Potts and Guy (1992) demonstrated
acteristics (Magnusson et al. 2004). Especially the that the permeation of a topically applied drug in an
lipophilicity, the molecular weight (MW), and the aqueous solution can be estimated by two physico-
melting point have to be taken into account in order chemical characteristics, which are combined in
to deliver a compound through the skin (Hadgraft Eq.  15.4 with MW as molecular weight of the
and Pugh 1998; Magnusson et al. 2004; Nielsen active compound:

log kp = −6.3 + 0.71 log K ow − 0.0061 MW cm ⋅ s −1  (15.4)


From Eq. 15.4 it can be concluded that small t­ hermodynamic activity of the active compound
molecules (low MW) with adequate affinity to have to be considered when transdermal and
the horny layer (log Kow from −3 to +6) can be dermal drug delivery is predicted (Bach and
expected to exhibit a sufficient drug penetration Lippold 1998).
(Bach and Lippold 1998; Potts and Guy 1992; According to that, not only the properties of
Wiechers et al. 2004). Compounds with good the active compound but also the vehicle influ-
affinity to both water and oil will show a good ence the diffusion of the active compound
penetration behavior. Such compounds tend to through the skin. Vehicle ingredients can interact
exhibit a low melting point (Hadgraft 2004). with the skin or with the active compound, so that
According to Potts and Guy (1992), Eq. 15.4 can its diffusion is enhanced or decreased.
be used for substances with MW from 18 to >750
and log Kow from −3 to +6.
Furthermore, the rate of diffusion depends on 15.2 Vehicle Composition
the thermodynamic activity of the drug in the
absence of penetration enhancing substances Although there is diverse nature of ingredients
(Bach and Lippold 1998). It is characterized by contained in vehicles used to deliver drugs into/
the ratio of the drug concentration in the vehicle through the skin (Wiechers et al. 2004), the fol-
(CV) to the maximum solubility in the vehicle lowing classification of vehicle components has
(CS) according to Eq. 15.5, with α as thermody- been published by Daniels and Knie (2007) with
namic activity, CV as concentration of the active five major classes.
compound in the vehicle, and CS as saturation
concentration of the active compound in the vehi-
cle (Bach and Lippold 1998): 15.2.1 Bases

a = ( CV / CS ) (15.5) Bases for topical products, representing the



main ingredient of the product, can be either of
In this process the thermodynamic activity rises hydro­philic nature like water or ethanol or they
with the concentration of the compound up to the show hydrophobic characteristics (e.g., petrola-
maximum concentration (saturation) in the vehi- tum, triglycerides) (Daniels and Knie 2007).
cle (Hadgraft 2004). Thus, solutions show a The choice of the base depends on various fac-
decline of activity with the duration of incuba- tors, all of which have to be considered in the
tion, while suspensions (having drug concentra- development of the final product. The base
tions above their solubility) exhibit a constant should exhibit no irritating or sensitizing poten-
activity (Bach and Lippold 1998). tial to the skin, the active compound has to be
Commercially available topical formulations delivered in adequate amounts to the site of
present a complex mixture of several classes of action, and the shelf life of the product has to be
substances (Wiechers et al. 2004). The effect of long enough (International Pharmacopoeia
all added substances in the formulation and the 2014). Furthermore, a desired consistency has
212 J. Stahl

to be created (e.g., hydrophobic lipid bases can Table 15.1  Classification of emulsifiers used in trans-
dermal therapeutic systems according to Daniels and Knie
exhibit a special spreadable behavior over the
(2007)
skin without a greasy feeling) (Daniels and
Compound
Knie 2007).
classification Examples
Anionic Sodium stearate
Sodium dodecyl sulfate
15.2.2 Gelling Agents Cationic Benzalkonium bromide
Cetylpyridinium chloride
In order to enhance the viscosity of the product, Amphoteric Phosphatidylcholine
which in turn improves storage and sensory attri- Betaine monohydrate
butes, macromolecules are added to form three-­ Nonionic Glycerol monostearate
dimensional scaffolds. Popular thickeners are Polysorbate 20 (Tween® 20)
carbomer, hydroxypropyl cellulose, and carmel-
lose sodium (Daniels and Knie 2007).
15.2.5 Emulsifier

15.2.3 Preservatives (Antimicrobial Emulsifiers added to the vehicle enhance the


Agents) thermodynamic stability between hydrophilic
and hydrophobic compounds. They are classified
Preservatives against bacterial growth are neces- as anionic, cationic, amphoteric, and nonionic
sary in order to enhance the shelf life of a product surfactants and accumulate between a lipid-rich
and for consumer protection against a contami- phase and a hydrophilic phase (Daniels and Knie
nated product. Commonly used substances with 2007). Table 15.1 lists some emulsifiers accord-
properties of preservatives are ethanol or isopro- ing to Daniels and Knie (2007).
panol, which have to be used in sufficient con-
centrations that means in concentrations about
20 %, which in turn are high enough to influ- 15.3 Formulation Types
ence the penetration profile of a drug, as well.
Furthermore, phenoxyethanol, benzoic acid, According to the US Food and Drug
and sorbic acid are used in lower concentra- Administration (FDA) (2013), topical dosage
tions (0.1–0.2 % sorbic acid, 0.15–0.5 % ben- forms (Table 15.2) can be divided into semi-
zoic acid, 0.5–1 % phenoxyethanol) (Daniels solid and liquid formulations, which are char-
and Knie 2007). According to the International acterized by their physicochemical nature.
Pharmacopoeia (2014), antimicrobial agents are Especially the localization of the topical treat-
indicated unless the formulation itself has ade- ment and the severity of the disorder determine
quate preservative properties. the kind of formulation, which is characterized
by the content of oil and water (Daniels and
Knie 2007). Ointments, e.g., are traditionally
15.2.4 Antioxidants preferred by clinicians for topical use on hair-
less skin or areas with short hair, while creams
These compounds are used in order to prevent are preferentially applied onto genial or flex-
oxidation reactions of lipids in exposition of ural areas (Huang et al. 2005). Moreover, skin
light, air, and heat. Typical antioxidants are conditions have to be considered not only in
alpha-tocopherol in concentrations of 0.05– healthy skin, but also in diseased skin. Acute
0.075 % and ascorbic acid esters in concentra- or subacute diseases are often treated with for-
tions of 0.01–0.015 % (Daniels and Knie mulations based on liquid bases (shake lotions
2007). or emulsions), while subchronic to chronic
15  Dermal and Transdermal Formulations: How They Can Affect the Active Compound 213

Table 15.2  Dosage forms according to the FDA (2013) hydrophilic, hydrophobic, or water-emulsifying
with modifications
nature are used (International Pharmacopoeia
Semisolid and 2014).
solid dosage Liquid dosage
forms forms Specialties
Creams and Emulsion Microemulsion
15.3.1.2  Gel
ointments A gel is a clear preparation comprising a liquid
Gel Lotion Nanoemulsion phase (solution or colloidal dispersion) within a
Paste Shampoo Patch three-dimensional polymeric matrix caused by
Powder Solution the addition of a gelling agent. It may contain
Spray suspended particles (International Pharmacopoeia
Suspension 2014; FDA 2013). Gels can be divided into
hydrophobic and hydrophilic gels.

diseases are medicated with pastes, rich oint- 15.3.1.3  Paste


ments, or gels. In brief, a more acute disease Pastes are generally intended for topical appli-
requires a high water and a low oil content cation. They are preparations with high con-
(Daniels and Knie 2007). Thus, creams are centrations of insoluble solids (20–50 %) in a
preferentially used for wet conditions and oint- fatty vehicle (Pharmacopoeia Europaea 2011;
ments for dry conditions. International Pharmacopoeia 2014; FDA 2013).

15.3.1.4  Powder
15.3.1 Semisolid Formulations Powders represent a mixture of drugs and/or
chemicals, which are of dry and finely divided
A semisolid preparation exhibits no pourable nature (FDA 2013).
behavior and does not conform to its storage con-
tainer at room temperature. It does not flow, but
exhibits plastic flow behavior. Low shear stress 15.3.2 Liquid Formulations
does not induce a flow of semisolid preparations.
Semisolid dosage forms for topical treatment can A liquid preparation is pourable and conforms to
be classified into creams and ointments, gels, its storage container at room temperature. It flows
pastes, and powders (International Pharmacopoeia and shows pseudoplastic flow and Newtonian
2014; FDA 2013). behavior. The FDA (2013) distinguishes between
emulsions, lotions, shampoos, solutions, sprays,
15.3.1.1  Cream and Ointment and suspensions for external skin treatment.
Creams and ointments are generally used for top-
ical application and are semisolid preparations, in 15.3.2.1  Emulsion
which solid or liquid substances are dispersed Emulsions are characterized by at least two liq-
(Pharmacopoeia Europaea 2011). The vehicle uids that are immiscible, so that one component
contains >20 % water and volatiles and/or <50 % is dispersed in droplets within the other compo-
hydrocarbons, polyols, or waxes (FDA 2013). nent. Thus, emulsions are special two-phase dos-
The consistency and rheological characteristics age forms. Stability is given by the addition of at
of creams depend on the emulsion type (oil in least one emulsifier (FDA 2013).
water or water in oil) and on the physicochemical
properties of the solids used (International 15.3.2.2  Lotion
Pharmacopoeia 2014). Ointments are prepara- This dosage form is generally intended for topi-
tions which are “immiscible, miscible, or emulsi- cal application. A lotion represents an emulsion
fiable with skin secretions.” Therefore, bases of in a liquid dosage form (FDA 2013).
214 J. Stahl

15.3.2.3  Shampoo trolling membrane and a backing membrane, and


Shampoos represent liquid soaps or detergents, (3) drug reservoir within the center of a periph-
both of which are used to clean the hair and scalp eral adhesive ring around the edges (Sachdeva
or to apply special drugs to the skin (Pharmacopoeia et al. 2013).
Europaea 2011; FDA 2013). Furthermore, special liquid preparations have
been generated in the last decades, such as nano-
15.3.2.4  Solution emulsions (Uner et al. 2005; Alves et al. 2007;
This dosage form is clear and homogenous. It Baboota et al. 2007), or microemulsions (Kemken
may contain more than one chemical dissolved in et al. 1991; Boltri et al. 1994; Pattarino et al.
one solvent or a mixture of miscible solvents 1994), etc.
(FDA 2013).

15.3.2.5  Spray 15.4 The Influence


A spray is characterized by a liquid, which is of Formulations on Drug
divided into small components by a jet of air or Diffusion into and through
steam (FDA 2013). the Skin

15.3.2.6  Suspension The impact of different dosage forms on trans-


This dosage form contains a liquid vehicle as the dermal drug delivery has been examined in sev-
basis, in which solid particles are dispersed in eral investigations. An extensively studied
(FDA 2013). topically applied compound is ibuprofen.
Hadgraft et al. (2003) demonstrated that various
5 % ibuprofen-containing products resulted in
15.3.3 Special Formulation Types marked differences in the permeation rates of
ibuprofen through excised human skin with the
Beside the conventional dosage forms, several most efficient permeation observed for a spray, a
new formulation types have been developed. gel, and a mousse, while two other gels, based on
Such a new preparation is a patch, which repre- other ingredients like ethanol or benzyl alcohol,
sents a sophisticated drug delivery system with and a cream led to less efficient permeation rates.
the intent to release the active compound in such Comparable results were found by Herkenne
a manner that the dosing frequency can be et al. (2007) with different permeation rates
reduced in comparison to conventional dosage observed in vitro and in vivo after topical admin-
forms (FDA 2013). Patches are commercially istration of four 5–10 % ibuprofen-containing
available for a constant delivery of various sub- gels with different ingredients. In this study, the
stances like ethinyl estradiol (Sachdeva et al. highest permeation through excised pig ear skin
2013), fentanyl (Lehmann and Zech 1992), lig- was for a 5 % ibuprofen gel containing propylene
nocaine (Kwon et al. 2012), nicotine (Benowitz glycol and isopropanol, followed by the 5 % gel
et al. 1991), nitroglycerol (Minghetti et al. 2001), containing propylene glycol. Lower permeation
rivastigmine (Gauthier et al. 2013), rotigotine rates were observed for the 10 % and the 5 % gel,
(Stiasny-Kolster et al. 2013), scopolamine (Gil both of which were enhancer-free. Interestingly,
et al. 2012), testosterone (James 1995), and oth- in vivo studies in man revealed similar results,
ers. The main advantages of patches are sus- with the highest permeation rate for the 5 % gel
tained and constant plasma levels and the bypass with both enhancers and the lowest permeation
of the first-pass effect in the liver (Kapil et al. rate for ibuprofen from the 5 % gel without
2012). Due to drug storage within the patch, enhancers, while the addition of propylene glycol
transdermal patches can be classified into three did not enhance permeation in vivo against the
types: (1) drug directly dispersed in adhesive 10 % gel without enhancer as observed with pig
polymer, (2) drug reservoir between a rate con- ear skin (Herkenne et al. 2007).
15  Dermal and Transdermal Formulations: How They Can Affect the Active Compound 215

Moreover, the fate of ibuprofen after topical Cab-O-­Sil®, Cabot Carbon Ltd., UK, and
administration depends on the formulation used. hydroxypropyl cellulose (HPC)) on ketoprofen
Comparative studies between different ibuprofen-­ diffusion through synthetic membranes and
containing formulations demonstrated the impact excised pig skin were performed (Gallagher
of the formulation on the amount of ibuprofen et al. 2003; Gallagher and Heard 2005). The con-
within the skin (skin absorption) (Stahl et al. tent of the thickening agents was chosen in order
2011, 2012). The comparison of ibuprofen per- to gain comparable viscosities of the formula-
meation through excised skin from a gel, a cream, tions. Permeation studies with pig ear skin
and a solution, all of which were commercially revealed that an increase in the content of the
available and contained 5 % ibuprofen, revealed thickener was correlated with reduced diffusion
different amounts of skin absorption depending rates of ketoprofen, while the diffusion rate from
on the formulation. The highest absorption (drug the HPC gel was almost doubled compared to
storage in the skin) was observed for the aqueous, that of silica gel.
enhancer-free solution and the gel containing the The skin diffusion of the glucocorticoid beta-
penetration enhancer 2-propanol, followed by the methasone was studied ex vivo in the isolated
cream, which contained the permeation enhancer perfused bovine udder, after topical administra-
propylene glycol. Interestingly, skin diffusion tion of five formulations (cream, gel, ointment
showed a different ranking: cream > solution > with and without propylene glycol, and solu-
gel (Stahl et al. 2011). Other commercially avail- tion). The ointments showed the highest poten-
able 5 % ibuprofen-containing formulations tials to promote betamethasone diffusion,
(cream, gel, “microgel”) were examined in a sec- followed by the cream, gel, and solution.
ond study in vitro with excised bovine udder skin Interestingly, there was no significant difference
and ex vivo with the isolated perfused bovine between the creams, although one formulation
udder (Stahl et al. 2012). Penetration enhancers contained propylene glycol, which is often used
in the formulations were propylene glycol in the as a penetration enhancer (Kietzmann and
cream and 2-propanol in the gel and the “micro- Blume 1997).
gel”. In this study, skin absorption profiles with a The effect of different formulations on the
general rank order, cream < “microgel” < gel, in vitro skin diffusion behavior of the muscle
were obtained, as well as permeation rates of a relaxant thiocolchicoside was studied by Bonina
differing general rank order, “microgel” < gel < et al. (2002) in excised human skin. They exam-
cream. ined the effect of a novel, noncommercially avail-
Various other substances, like ketopro- able foam based on the enhancers propylene
fen, betamethasone, or thiocolchicoside, show glycol dipelargonate and propylene glycol in
formulation-­ dependent skin diffusion rates, as comparison to an ointment and a gel, both
well (Bonina et al. 2002; Guerol et al. 1996; enhancer-free, and found a two- to threefold
Gallagher and Heard 2005; Gallagher et al. 2003; higher flux for the foam in comparison to the
Huang et al. 2005; Kietzmann and Blume 1997; ointment and the gel.
Mitriaikina and Mueller-Goymann 2009; Singh The main problems of such comparative stud-
et al. 2009). ies are that information about the ingredients pro-
The comparison of the influence of four dif- vided to the consumers is only based on the kind
ferent ointment bases on ketoprofen release from of ingredients, but not on their content (except for
the formulation in vitro revealed the following the active compound), and that commercially
general rank order: white petrolatum < cold available topical formulations are based on a high
cream < hydrophilic ointment < carbomer gel, diversity of additives. Consequently, the main
which correlated with the hydrophilicity of the influencing additive of the vehicle can rarely be
formulations (Guerol et al. 1996). Moreover, found out in such studies without systematic
comparison studies with gel formulations com- examinations about the effect of changes of sin-
prising different thickening agents (silica as gle additives.
216 J. Stahl

Table 15.3  Possibilities to enhance transdermal drug the optimized formulation as “the polarity of the
delivery considering Eqs. 15.1, 15.2, 15.3, 15.4 and 15.5
formulation needs to be as far as possible from
Possibilities to change the the polarity of the active ingredient in order to
Parameter Change parameter
increase the driving force of the active ingredient
Kow ↑ Enhance lipophilicity of the
into the skin, but at the same time as close as
compound
D ↑ Decrease viscosity of the
­possible to that of the active ingredient to ensure
vehicle that high concentrations can be reached to ensure
Use of penetration enhancers that enough material penetrates the skin to reach
Δc ↑ Enhance solubility of the effective tissue concentrations at the site of
compound in the vehicle action in the skin.” The effectiveness of their
x ↓ Use of penetration enhancers guideline “Formulating for Efficacy” has been
MW ↓ Change chemical structure of demonstrated for the skin delivery of octadecene
the compound
dioic acid (Wiechers et al. 2004) and should be
KSC/V ↑/↓ Change the polarity or pH of the
vehicle noted in order to choose an appropriate formula-
α ↑ Enhance concentration of the tion for topical skin delivery of drugs. The ion-
compound in the vehicle (up to ization state of a drug has to be considered, since
saturation) unionized molecules exhibit a higher permeation
Modified after Wiechers et al. (2004) rate than the ionized fraction (Hadgraft and
Valenta 2000) due to the fact that unionized com-
pounds exhibit a higher lipophilicity propor-
The dependency of the drug permeation on the tional to Kow (Watkinson et al. 2009).
drug formulation arises the question of how vehi- Consequently, the pH of the formulation has to
cles interact with the active compounds in order be optimized in correlation to the pKa value of
to exhibit an optimized permeation profile. Since the compound with respect to the physiological
formulations are a mixture of various compounds, skin pH (4–7.4) in order to prevent skin damages
many ingredients can influence transdermal drug (Hadgraft and Valenta 2000). Since the pKa
delivery and have to be taken into account value of a compound depends, furthermore, on
(Wiechers et al. 2004). Considering the Eqs. 15.1 the supplementation of cosolvents like ethanol
15.2, 15.3, 15.4 and 15.5, several parameters of a (Watkinson et al. 2009), an addition of cosol-
topical formulation can be changed in order to vents can be used to optimize the solubility of
influence transdermal drug delivery. The results the compound, which in turn affects both the
are listed in Table 15.3 according to Wiechers thermodynamic activity α according to Eq. 15.5
et al. (2004). and the concentration according to Eq. 15.2,
However, the major problem is that the param- as well.
eters listed in Table 15.3 are linked (Hadgraft and The diffusion coefficient D can be altered by
Pugh 1998): Since the alteration of a single supplementation of suitable permeation enhanc-
parameter may affect several other parameters, it ers (such as propylene glycol, ethanol, etc.) or by
is necessary to have a closer look on the effects of variations of the viscosity of the vehicle
variations of single parameters on transdermal (Wiechers et al. 2004). Permeation enhancers can
drug diffusion. interact with the stratum corneum or with the
An alteration of the octanol-water partition active compound and are of such diversity that
coefficient Kow, e.g., can be performed by chang- they are just briefly mentioned in this chapter.
ing the chemical structure of the compound. Viscosity changes are another elegant method to
Since that is not acceptable for various sub- influence D, which is inversely correlated to the
stances, alternatively a formulation with opti- viscosity according to the Stokes-Einstein equa-
mized polarity has to be chosen in order to tion (Eq. 15.6). Thus, a high viscosity results in a
enhance transdermal drug diffusion, in accor- diminished diffusion coefficient of the active
dance with Wiechers et al. (2004). They described compound:
15  Dermal and Transdermal Formulations: How They Can Affect the Active Compound 217

D = k ⋅ T / 6p ⋅h ⋅ R  m ⋅ s −1  (15.6) 1999). Thus, formulations with the same macro-


viscosity may provide different permeation rates
where k is the Boltzmann constant, T represents for the active compound. This could be demon-
the absolute temperature, η is the viscosity, and R strated for ketoprofen permeation after applica-
is the hydrodynamic radius of the diffusing parti- tion of two formulations sharing the same
cle (Raghavan et al. 2001). The difference in vis- macroviscosity due to the addition of 1 % Cab-O-­
cosity of a formulation has been subject of several Sil® or 0.5 % HPC (Gallagher and Heard 2005).
studies. The supplementation of 3 and 10 % After 24 h, the amount of ketoprofen, which per-
hydroxymethyl cellulose to aqueous s­ olutions of meated from the 1 % Cab-O-Sil® gel through pig
flufenamic acid resulted in a viscosity-­dependent ear skin, was nearly twofold higher than that
permeation of flufenamic acid with higher vis- from the 0.5 % HPC gel that leads to the conclu-
cosities correlating with lowered permeation rates sion that complexation (binding interactions
(ENNEN 2009). Diminished permeation rates between compound and thickening agent) of
depending on the degree of viscosity have also some compounds has a greater influence on
been described for other substances. transdermal permeation rate than changes in the
The diffusion coefficient of theophylline, e.g., viscosity (Gallagher and Heard 2005). Thus,
declined exponentially with the HPC concentra- macroviscosity can influence the solubility of the
tion (0–2 %) of five HPC varieties (molecular active compound in such a way that absorption of
weight between 5 × 105 and 1.2 × 106). In case of the active compound to the thickening agent
the lowest-molecular-weight HPC, a constant enhances solubility of the active compound,
diffusion coefficient was observed up to 0.8 %, which in turn changes the thermodynamic activ-
which was explained by a high entanglement ity and the saturation of the compound. In the
concentration of the HPC (Alvarez-Lorenzo et al. end, the diffusion rate of the compound is
1999). The influence of different contents of modified.
polymers on ketoprofen release was studied The concentration gradient Δc can easily be
in vitro, as well as with the result that an enhance- increased by enhancing the total amount of the
ment of the polymer concentration was corre- drug in the formulation. Furthermore, the solubil-
lated with a higher release of ketoprofen from the ity of the compound in the vehicle can be elevated
gels (Chi and Jun 1991). by adding cosolvents (Watkinson et al. 2009), by
Moreover, the release and permeation rate of using microemulsions (Sabale and Vora 2012), by
several model anesthetics decreased with increas- optimizing the polarity of the vehicle, or by dry-
ing macroviscosity of the formulation through ing effects on the skin surface. Immediately after
guinea pig skin and a silicone membrane (Welin-­ application of a topical formulation on the skin,
Berger et al. 2001). drying effects may occur, which have a signifi-
However, viscosity-independent permeation cant impact on the efficacy of the applied com-
behavior was reported for some active com- pound (Bowen and Heard 2006). This could be
pounds, like caffeine or naproxen (Suh and Jun demonstrated for simple gel formulations con-
1996; Ennen 2009). Therefore, the viscosity had taining ketoprofen with different concentrations
to be characterized in a better way: the apparent of propylene glycol as solvent and HPC as gelling
viscosity is represented by the macroviscosity agent (Bowen and Heard 2006). In this study, a
(macroscopic flow properties of a system), which lower content of the solvent propylene glycol cor-
does necessarily not reflect the matrix through related with a higher permeation rate of ketopro-
which the drug diffuses (Alvarez-Lorenzo et al. fen, which was connected with the increase of the
1999; Raghavan et al. 2001). On the other hand, content of ketoprofen and HPC in the formula-
microviscosity (Alvarez-Lorenzo et al. 1999; tion, in order to simulate the concentration effects
Raghavan et al. 2001) is characterized by molec- of ingredients during the drying process of a sol-
ular drug movements of the active compound “at vent. A rapid crystallization process of formula-
the microscopic scale” (Alvarez-Lorenzo et al. tions may also diminish the percutaneous
218 J. Stahl

permeation of a substance, since crystallized drug Baboota S, Shakeel F, Ahuja A, Ali J, Shafio S (2007)
Design, development and evaluation of novel nano-
particles may be unable to pass the stratum cor-
emulsion formulations for transdermal potential of
neum due to steric hindrance (Kietzmann and celecoxib. Acta Pharm 57:315–332
Blume 1997). Thus, formulations drying on the Bach M, Lippold BC (1998) Percutaneous penetration
skin surface after application show altered per- enhancement and its quantification. Eur J Pharm
Biopharm 46:1–13
meation behaviors of the active compound.
Benowitz NL, Chan K, Denaro CP, Jacob P (1991) Stable
Consequently, drying effects will behave in dif- isotope method for studying transdermal drug absorp-
ferent ways depending on the ­composition of the tion: the nicotine patch. Clin Pharmacol Ther
formulation, since fast evaporating solvents may 50:286–293
Boltri L, Morel S, Trotta M, Gasco MR (1994) In vitro
result in rapid crystallization processes (Bowen
transdermal permeation of nifedipine from thickened
and Heard 2006), while a moderate evaporation microemulsions. J Pharm Belg 49:315–320
process enhances the concentration of the active Bonina F, Puglia C, Trombetta D, Dragani MC, Gentile
compound, which in turn increases skin diffusion MM, Clavenna G (2002) Vehicle effects on in vitro
skin permeation of thiocolchicoside. Pharmazie
according to Eq. 15.2.
57:750–752
The diffusion path length x of a compound is Bowen JL, Heard CM (2006) Film drying and complex-
quite longer than the thickness of the stratum ation effects in the simultaneous skin permeation of
corneum, since a topical applied compound pri- ketoprofen and propylene glycol from simple gel for-
mulations. Int J Pharm 307:251–257
marily diffuses through the intercellular lipid-­
Chi SC, Jun HW (1991) Release rates of ketoprofen from
rich spaces (Hadgraft 2004). Possibilities of poloxamer gels in a membraneless diffusion cell. J
shortening the diffusion pathway are independent Pharm Sci 80:280–283
of the formulation and comprise various method- Cross SE, Pugh WJ, Hadgraft J, Roberts MS (2001)
Probing the effect of vehicles on topical delivery:
ologies like abrasion of the skin via tape-­stripping
understanding the basic relationship between solvent
(Tokumura et al. 2006), laser or microdermabra- and solute penetration using silicone membranes.
sion (Lee et al. 2003), or the creation of shunt Pharm Res 18:999–1005
ways via microneedles (Henry et al. 1998). Daniels R, Knie U (2007) Galenics of dermal products-­
vehicles, properties and drug release. J Dtsch Dermatol
Ges 5:367–383
Ennen JG (2009) Influence of pH, viscosity and co-­
Conclusion diffusion on drug permeation through bovine udder
Topical products are of great diversity con- skin and silicone membrane. Thesis. University of
Veterinary Medicine Hannover, Foundation
cerning their ingredients. The complex con-
FDA (U.S. Food and Drug Administration) (2013)
nection between all diffusion parameters Dosage forms.http://www.fda.gov/Drugs/Development­
highlights the importance of their knowledge ApprovalProcess/FormsSubmissionRequirements/
in order to develop an optimized formulation ElectronicSubmissions/DataStandardsManual­
monographs/ucm071666.htm. Accessed 11 Mar 2013
for transdermal drug delivery. Each additive
Gallagher SJ, Heard CM (2005) Solvent content and mac-
even for sensory purposes or a long shelf life roviscosity effects on the in vitro transcutaneous
can interact with either the active compound ­delivery and skin distribution of ketoprofen from sim-
or the skin and has to be chosen with caution ple gel formulations. Skin Pharmacol Physiol 18:
186–194
to produce a suitable formulation.
Gallagher SJ, Trottet L, Heard CM (2003) Ketoprofen:
release from, permeation across and rheology of sim-
ple gel formulations that simulate increasing dryness.
References Int J Pharm 268:37–45
Gauthier S, Robillard A, Cohen S, Black S, Sampalis J,
Alvarez-Lorenzo C, Gomez-Amoza JL, Martinez-­Pacheco Colizza D, de Takacsy F, Schecter R (2013). Real-life
R, Souto C, Concheiro A (1999) Microviscosity of effectiveness and tolerability of rivastigmine transder-
hydroxypropylcellulose gels as a basis for prediction of mal patch in patients with mild-to-moderate Alzheimer's
drug diffusion rates. Int J Pharm 180:91–103 disease: The EMBRACE study, on behalf of the
Alves MP, Scarrone AL, Santos M, Pohlmann AR, EMBRACE investigators. Curr Med Res Opin
Guterres SS (2007) Human skin penetration and distri- 29:989–1000
bution of nimesulide from hydrophilic gels containing Gil A, Nachum Z, Tal D, Shupak A (2012) A comparison
nanocarriers. Int J Pharm 341:215–220 of cinnarizine and transdermal scopolamine for the
15  Dermal and Transdermal Formulations: How They Can Affect the Active Compound 219

prevention of seasickness in naval crew: a double-­ ute maximum flux across the skin. J Invest Dermatol
blind,randomized, crossover study. Clin Neurophar­ 122:993–999
macol 35(1):37–39 Minghetti P, Casiraghi A, Cilurzo F, Montanari L (2001)
Guerol Z, Hekimoglu S, Demirdamar R, Sumnu M (1996) Evaluation of adhesive properties of transdermal ther-
Percutaneous absorption of ketoprofen. I. In vitro apeutic systems containing nitroglycerin. Boll Chim
release and percutaneous absorption of ketoprofen Farm 140:63–67
from different ointment bases. Pharm Acta Helv Mitriaikina S, Mueller-Goymann CC (2009) Comparative
71:205–212 permeation studies of nondiluted and diluted
Hadgraft J (2004) Skin deep. Eur J Pharm Biopharm betamethasone-­ 17-valerate semisolid formulations
58:291–299 through isolated human stratum corneum and artifi-
Hadgraft J, Pugh WJ (1998) The selection and design of cial skin construct. Skin Pharmacol Physiol 22:
topical and transdermal agents: a review. J Investig 142–150
Dermatol Symp Proc 3:131–135 Nielsen JB, Nielsen F, Sorensen JA (2004) In vitro percu-
Hadgraft J, Valenta C (2000) pH, pK(a) and dermal deliv- taneous penetration of five pesticides–effects of
ery. Int J Pharm 200:243–247 molecular weight and solubility characteristics. Ann
Hadgraft J, Whitefield M, Rosher PH (2003) Skin pene- Occup Hyg 48:697–705
tration of topical formulations of ibuprofen 5 %: an Pattarino F, Carlotti ME, Gasco MR (1994) Topical deliv-
in vitro comparative study. Skin Pharmacol Appl Skin ery systems for azelaic acid: effect of the suspended
Physiol 16:142 drug in microemulsion. Pharmazie 49:72–73
Henry S, McAllister DV, Allen MG, Prausnitz MR (1998) Pharmacopoeia Europaea (Europaeisches Arzneibuch)
Microfabricated microneedles: a novel approach to (2011) Deutscher Apotheker Verlag, Govi-Verlag,
transdermal drug delivery. J Pharm Sci 87:922–925 Pharmazeutischer Verlag GmbH, Stuttgart, Eschborn
Herkenne C, Naik A, Kalia YN, Hadgraft J, Guy RH Potts RO, Guy RH (1992) Predicting skin permeability.
(2007) Ibuprofen transport into and through skin from Pharm Res 9:663–669
topical formulations: in vitro-in vivo comparison. J Raghavan SL, Kiepfer B, Davis AF, Kazarian SG,
Invest Dermatol 127:135–142 Hadgraft J (2001) Membrane transport of
Huang X, Tanojo H, Lenn J, Deng CH, Krochmal L ­hydrocortisone acetate from supersaturated solutions;
(2005) A novel foam vehicle for delivery of topical the role of polymers. Int J Pharm 221:95–105
corticosteroids. J Am Acad Dermatol 53:26–38 Sabale V, Vora S (2012) Formulation and evaluation of
James JS (1995) San Francisco area: testosterone replace- microemulsion-based hydrogel for topical delivery. Int
ment study, injection vs. patch. AIDS Treat News J Pharm Invest 2:140–149
233:7–8 Sachdeva V, Bai Y, Kydonieus A, Banga AK (2013)
Kapil RP, Cipriano A, Friedman K, Michels G, Shet MS, Formulation and optimization of desogestrel transder-
Colucci SV, Apseloff G, Kitzmiller J, Harris SC mal contraceptive patch using crystallization studies.
(2012) Once-weekly transdermal buprenorphine Int J Pharm 441(1–2):9–18
application results in sustained and consistent Scheuplein RJ (1976) Permeability of the skin: a review
­steady-­state plasma levels. J Pain Symptom Manage. of major concepts and some new developments.
doi:10.1016/j.jpainsymman.2012.06.014 J Invest Dermatol 67:672–676
Kemken J, Ziegler A, Mueller BW (1991) Pharmacody­ Singh S, Gajra B, Rawat M, Muthu MS (2009) Enhanced
namic effects of transdermal bupranolol and timolol transdermal delivery of ketoprofen from bioadhesive
in vivo: comparison of microemulsions and matrix gels. Pak J Pharm Sci 22:193–198
patches as vehicle. Methods Find Exp Clin Pharmacol Stahl J, Wohlert M, Kietzmann M (2011) The effect of
13:361–365 formulation vehicles on the in vitro percutaneous per-
Kietzmann M, Blume B (1997) Percutaneous absorption meation of ibuprofen. BMC Pharmacol 11(12):1–5
of betamethasone from different formulations using Stahl J, Blume B, Bienas S, Kietzmann M (2012) The
the isolated perfused bovine udder. In Vitro Toxicol comparability of in vitro and ex vivo studies on the
10:11–15 percutaneous permeation of topical formulations con-
Kwon YS, Kim JB, Jung HJ, Koo YJ, Lee IH, Im KT, taining Ibuprofen. Altern Lab Anim 40:91–98
Woo JS, Im KS (2012) Treatment for postoperative Stiasny-Kolster K, Berg D, Hofmann WE, Berkels R,
wound pain in gynecologic laparoscopic surgery: topi- Grieger F, Lauterbach T, Schollmayer E, Bachmann
cal lidocaine patches. J Laparoendosc Adv Surg Tech CG (2013) Effectiveness and tolerability of rotigotine
A 22(7):668–673 transdermal patch for the treatment of restless legs syn-
Lee WR, Shen SC, Kuo-Hsien W, Hu CH, Fang JY (2003) drome in a routine clinical practice setting in Germany.
Lasers and microdermabrasion enhance and control Sleep Med. doi:10.1016/j.sleep.2013.02.013, May 10;
topical delivery of vitamin C. J Invest Dermatol doi: pii: S1389-9457(13)00106-8
121:1118–1125 Suh H, Jun HW (1996) Physicochemical and release stud-
Lehmann KA, Zech D (1992) Transdermal fentanyl: clini- ies of naproxen in polymer gels. Int J Pharm
cal pharmacology. J Pain Symptom Manage 7:8–16 129:13–20
Magnusson BM, Anissimov YG, Cross SE, Roberts MS The International Pharmacopoeia (2014) 4th Supplement
(2004) Molecular size as the main determinant of sol- to the fourth edition. Monographs: dosage forms:
220 J. Stahl

general monographs: topical semi-solid dosage forms. Watkinson RM, Herkenne C, Guy RH, Hadgraft J,
http://apps.who.int/phint/en/p/docf/. Accessed 16 Oct Oliveira G, Lane ME (2009) Influence of ethanol
2014 on the solubility, ionization and permeation
Tokumura F, Yoshiura Y, Homma T, Nukatsuka H (2006) characteristics of ibuprofen in silicone and human
­
Regional differences in adhesive tape stripping of skin. Skin Pharmacol Physiol 22:15–21
human skin. Skin Res Technol 12:178–182 Welin-Berger K, Neelissen JA, Bergenstahl B (2001) The
Uner M, Wissing SA, Yener G, Mueller RH (2005) Skin effect of rheological behaviour of a topical anaesthetic
moisturizing effect and skin penetration of ascorbyl formulation on the release and permeation rates of the
palmitate entrapped in solid lipid nanoparticles (SLN) active compound. Eur J Pharm Sci 13:309–318
and nanostructured lipid carriers (NLC) incorporated Wiechers JW, Kelly CL, Blease TG, Dederen JC (2004)
into hydrogel. Pharmazie 60:751–755 Formulating for efficacy. Int J Cosmet Sci 26:173–182
Part V
Emulsions as Vehicles for Skin Delivery
The Effects of Emulsifiers
and Emulsion Formulation Types 16
on Dermal and Transdermal Drug
Delivery

Anja Otto and Jeanetta du Plessis

Contents 16.6.4 Effects of Emulsion Droplet Charge . . . . 235


16.6.5 Effects of Surfactant Association
16.1 Introduction . . . . . . . . . . . . . . . . . . . . . . 223 Structures . . . . . . . . . . . . . . . . . . . . . . . . . 236
16.2 Emulsions as Topical Delivery 16.6.6 Effects of Solid Particles as
Systems . . . . . . . . . . . . . . . . . . . . . . . . . . 224 Emulsifiers . . . . . . . . . . . . . . . . . . . . . . . . 238

16.3 Emulsifiers . . . . . . . . . . . . . . . . . . . . . . . 224 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 239

16.4 Different Types of Emulsions . . . . . . . . 225 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 239

16.5 The Effects of Emulsion Types


on Dermal and Transdermal
Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . 226 16.1 Introduction
16.5.1 Overview . . . . . . . . . . . . . . . . . . . . . . . . . 226
16.5.2 Hydrophilic Active Ingredients . . . . . . . . 227
16.5.3 Lipophilic Active Ingredients . . . . . . . . . 231 Emulsions are heterogeneous systems consisting
16.5.4 Stabilisation Effects of the of at least two immiscible liquid phases, in which
Formulation . . . . . . . . . . . . . . . . . . . . . . . 231 the one liquid is dispersed as globules (dispersed
16.6 The Effects of Various Emulsifiers phase) in the other (continuous phase). The two
on Dermal and Transdermal immiscible liquids are, in general, an oil phase and
Delivery . . . . . . . . . . . . . . . . . . . . . . . . . . 232 an aqueous phase. However, an emulsion can also
16.6.1 Overview . . . . . . . . . . . . . . . . . . . . . . . . . 232
16.6.2 Effects of Hydrophilic Chain Length consist of other immiscible phases, e.g. polar and
of Nonionic Surfactants . . . . . . . . . . . . . . 232 nonpolar oil phases (Tadros 2009). The droplet size
16.6.3 Effects of Emulsifier Ratios and of macroemulsions (or conventional emulsions) is
HLB Value . . . . . . . . . . . . . . . . . . . . . . . . 233 in the micrometre range and usually comprises a
radius between 0.15 and 100 μm (Friberg 1990).
Nanoemulsions, on the other hand, are emulsions
that contain very small droplets (r <100 nm) and
are also known as submicron emulsions, ultrafine
emulsions or mini-emulsions (McClements 2012).
However, macro- and nanoemulsions cannot be
A. Otto (*) • J. du Plessis distinguished solely on the basis of the droplet size,
Unit for Drug Research and Development, as the physicochemical or thermodynamic proper-
North-West University, Potchefstroom Campus,
ties do not clearly change when the droplet size is
Potchefstroom 2520, South Africa
e-mail: 12878332@nwu.ac.za; reduced from the micrometre to the nanometre
Jeanetta.DuPlessis@nwu.ac.za range, and therefore no definite cut-off value for

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 223


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_16, © Springer-Verlag Berlin Heidelberg 2015
224 A. Otto and J. du Plessis

the droplet size exists. But there are other emulsion 2006). Additionally, they undergo considerable
properties that can be used to differentiate between structural changes after the application onto the
macro- and nanoemulsions. For example, nano- skin, as water and other volatile substances evapo-
emulsions can become translucent or transparent rate within a short period of time. The modified
and exhibit a much higher stability against cream- formulation structure left on the skin after evapora-
ing or sedimentation (McClements 2012). tion of the volatile compounds is important for the
In contrast to microemulsions, which are in a topical performance of emulsions (Friberg and Ma
thermodynamic equilibrium and can hence form 2006). Therefore, not only an understanding of the
spontaneously, macro- and nanoemulsions are microstructure of the initial emulsion but also of
thermodynamically unstable and necessitate the film that is left on the surface of the skin is
external energy for formation. Macro- and nano- important for optimisation of topical emulsions.
emulsions are fundamentally different from Despite of the complexity of emulsions, Bernardo
microemulsions. However, there are similarities and Saraiva (2008) presented a theoretical model
in composition, dimensions, structures and fabri- for transdermal drug delivery from emulsions. The
cation methods between micro- and nanoemul- model incorporated the formulation heterogeneity
sions, e.g. both can contain droplets in a size and was conceived for the prediction of transdermal
below 100 nm and be translucent (McClements delivery of drugs as a function of emulsion compo-
2012). These similarities have led to confusion sition. It allows the investigation of the effect of
about the correct use of the terms in literature, excipients on the drug activity in the two emulsion
and recent articles have reviewed similarities and phases as well as on the drug diffusivity in the con-
differences between micro- and nanoemulsions tinuous phase and the influence of surfactants form-
in order to clarify the terminology (McClements ing interfacial layers with different resistance on
2012; Anton and Vandamme 2011). drug transfer. A simulated case study, for example,
This chapter focuses on macroemulsions, here indicated that the estimated interfacial resistance by
referred to as emulsions, and their effect on der- the surfactant layer may not be negligible with
mal and transdermal delivery. Nano- and micro- regard to its effect on drug delivery rates.
emulsions as topical drug delivery systems are Furthermore, the model could also be used to pre-
discussed in different chapters of this book. dict the effect of the dosing condition (e.g. applied
emulsion volume) on drug absorption. Different
modes of emulsion application could significantly
16.2 Emulsions as Topical affect drug delivery time profiles. At the same time,
Delivery Systems Grégoire et al. (2009) developed a model to predict
the transport of actives into and through the skin
Emulsions are widely used in the cosmetic and from a cosmetic or dermatological formulation,
pharmaceutical fields/industries for the topical addressing in particular simple oil-in-water emul-
administration of both hydrophilic and lipophilic sions. The model assumed, inter alia, that only the
active ingredients, owing to their pleasant skin sen- fraction of the active ingredient in the continuous
sations and their good solubilising effects on these phase of the emulsion was available for partitioning
substances (Förster and Von Rybinski 1998). The into the skin and good correlation was obtained
consistency of topical emulsions ranges from liq- with experimental data.
uid lotions to semisolid ointments and creams
(Eccleston 1997a). Since topical emulsions should
be designed to feature adequate physical and chem- 16.3 Emulsifiers
ical stability, aesthetic acceptability as well as opti-
mal delivery characteristics, they generally are not Since emulsions are thermodynamically unstable,
simply two-phase formulations, but rather complex emulsifiers are required for the formation and sta-
formulations (Eccleston 1997b). For example, bilisation of emulsions. Generally, an emulsifier is
emulsions can also contain solid particles, vesicles, defined as a substance that stabilises emulsions.
liquid crystals or a third liquid (Friberg and Ma However, no absolute classification exists, as
16 The Effects of Emulsifiers and Emulsion Formulation Types on Dermal and Transdermal Drug Delivery 225

Fig. 16.1 Schematic


illustration of breakdown
processes of emulsions

constituents may perform different functions, e.g. 2009). Creaming or sedimentation can be pre-
a fatty alcohol can be incorporated as an emulsi- vented or retarded by increasing the viscosity of
fier, thickener or emollient (Eccleston 1997b). the continuous phase, e.g. by the development of
Different types of emulsifying agents exist, a three-dimensional network of particles or poly-
e.g. ionic and nonionic surfactants, polymers and mers (Eccleston 1997a; Aveyard et al. 2003).
solid particles. In the absence of any emulsifiers, Cosmetic and pharmaceutical emulsions com-
the emulsions tend to break down, e.g. by coales- monly comprise of blends of emulsifiers, instead
cence (fusion of droplets into larger droplets by of a single emulsifying agent. Most of these
thinning and disruption of the emulsifier layer), mixed emulsifiers consist of ionic or nonionic
flocculation (aggregation of droplets without surfactants, in combination with fatty amphiphi-
disrupting the emulsifier layer), creaming/sedi- les, which may be added separately during the
mentation (gravity-induced separation) and/or emulsification process, or as a pre-manufactured
Ostwald ripening (Fig. 16.1) (Tadros 2009). blend (emulsifying wax). In addition to promot-
Emulsifiers are required for the formation of ing the stability of emulsions, mixed emulsifiers
emulsions as they form a film around the newly and emulsifying waxes have the further advan-
formed drops and consequently prevent coales- tages of improving emulsification by stabilising
cence during emulsification and storage. In addi- the oil droplets during formation and by control-
tion, surfactants reduce the interfacial tension ling the rheological properties of the emulsion
and are important for the deformation and break- (Eccleston 1997b). Another example is the
up of droplets during the emulsification process simultaneous inclusion of surfactants and solid
(Tadros 2009). Furthermore, the properties of particles that could yield synergistic stabilisation
the interface are significant for the rate and of the emulsion against coalescence and cream-
extent of coalescence (Friberg and Ma 2006). To ing (Binks and Whitby 2005; Lan et al. 2007).
prevent or retard flocculation and consequently
also coalescence, it is important to keep a mini-
mum distance between the droplets to overcome 16.4 Different Types of Emulsions
the van der Waals attraction. This could be
achieved, for example, by electrostatic repulsion Several types of emulsions can be distinguished,
in the presence of a surface charge (ionic surfac- for instance:
tants or charged particles) or by steric hindrance • Simple emulsions: oil-in-water (o/w) and
(nonionic surfactants or polymers) (Tadros water-in-oil (w/o)
226 A. Otto and J. du Plessis

• Multiple emulsions: oil-in-water-in-oil contact angle θow >90° prefer to stabilise w/o
(o/w/o) and water-in-oil-in-water (w/o/w) emulsions (Fig. 16.2) (Aveyard et al. 2003).
The type of emulsion that is formed mainly
depends on the property of the emulsifier, e.g. the
hydrophilic-lipophilic balance (HLB) value of 16.5 The Effects of Emulsion
the surfactants. The HLB is an arbitrary scale Types on Dermal
(e.g. from 1 to 20 for nonionic surfactants), and and Transdermal Delivery
the higher the HLB, the more hydrophilic the sur-
factant. According to the Bancroft rule, the phase 16.5.1 Overview
in which the emulsifier is more soluble consti-
tutes the continuous phase (Bancroft 1913). For Various studies have been performed to compare
example, lipophilic surfactants with a low HLB different types of emulsions (Dal Pozzo and
(HLB <7) tend to act as w/o emulsifiers, whilst Pastori 1996; Förster et al. 1997; Wiechers 2005).
hydrophilic surfactants with a high HLB (HLB However, not only the type of emulsion was
>7) tend to form o/w emulsions. However, different, the formulation ingredients also varied,
Harusawa et al. (1980) suggested a change to the and their interactions with the active ingredients
Bancroft rule by proposing that the phase in (e.g. solubilisation in micelles, supramolecular
which the surfactant forms micelles constitutes complex formation) may therefore have impacted
the external phase, independent of the solubility on the dermal and transdermal delivery of the
of the surfactant monomers in the oil and aque- active ingredients (Dal Pozzo and Pastori 1996).
ous phases. For particle-stabilised emulsions Consequently, due to the complexity of topical
(also known as Pickering emulsions), it was dem- emulsions, the investigation of the emulsion type
onstrated that the wettability of the solid parti- effects on dermal and transdermal delivery
cles, which is determined by the contact angle, requires a systematic approach.
defines which type of Pickering emulsion will be A study by Lalor et al. (1995), for example,
formed. For example, particles with a contact showed the effects of the incorporation of surfac-
angle at the oil-water interface θow <90° tend to tants in o/w and w/o emulsions (polysorbate 60
form o/w emulsions, whereas particles with a (Tween® 60) in o/w emulsion and sorbitan

Fig. 16.2 (Upper) Position of a small spherical particle at curved interface. For θ <90°, solid-stabilised o/w emul-
a planar oil-water interface for a contact angle (measured sions may form (left). For θ >90°, solid-stabilised w/o
through the aqueous phase) less than 90° (left), equal to emulsions may form (right) (Reprinted from Aveyard
90° (centre) and greater than 90° (right). (Lower) et al. (2003), with permission from Elsevier)
Corresponding probable positioning of particles at a
16 The Effects of Emulsifiers and Emulsion Formulation Types on Dermal and Transdermal Drug Delivery 227

sesquioleate (Arlacel® 83) in w/o emulsion) on the (e.g. o/w, w/o and w/o/w), hence allowing for the
partitioning and permeation of three model systematic investigation of the effect of the type
compounds (methyl-, ethyl- and butyl-p- of emulsion on dermal and transdermal delivery
aminobenzoate), due to the solubilisation capabili- only. These study outcomes are summarised in
ties of the emulsifiers. It was demonstrated that the Table 16.1.
emulsifier (surfactant) and its distribution between The results in Table 16.1 demonstrate that the
the oil and water phase played an important role in type of emulsion significantly influenced both
the solubility and therefore thermodynamic activ- skin penetration and skin permeation of the active
ity of the permeants in the vehicle, i.e. the thermo- ingredients. The effect of the type of emulsion on
dynamic activity of the compounds in the external dermal and transdermal delivery was furthermore
phase of the emulsions was found to be the driving dependent on the dosing condition (finite non-
force for permeation through the polydimethylsi- occluded vs. infinite occluded). With finite dos-
loxane membranes. Polysorbate 60 (Tween® 60), ing under non-occluded conditions, the
the surfactant used in the o/w emulsion, was physicochemical and thermodynamic properties
mainly available in the external aqueous phase of of the formulation modified rapidly after applica-
the emulsion, where it formed micelles and solu- tion onto the skin, whereas at infinite dosing
bilised the three test permeants, methyl-, ethyl- under occluded conditions, the thermodynamic
and butyl-p-aminobenzoate, thereby reducing properties did not alter significantly (Laugel et al.
their thermodynamic activity and the permeability 1998b).
coefficient. The permeability coefficient between
the o/w emulsion and its corresponding, externally
isolated aqueous phase was equal, signifying the 16.5.2 Hydrophilic Active
importance of the thermodynamic activity of the Ingredients
permeants in the external phase for promoting
the permeation process. However, the solubility of In summary, it can be concluded that the percuta-
the three compounds in the oil phase of the same neous absorption, as well as the skin penetration
o/w emulsion was similar to the solubility in the of hydrophilic drugs (e.g. glucose, metronidazole
oil without surfactant, indicating no solubilising and lactic acid), is generally superior for o/w
effect of polysorbate 60 (Tween® 60) in the oil emulsions, compared to w/o/w and w/o emul-
phase of the o/w emulsion, hence resulting in simi- sions (Fig. 16.3). Various suggestions were made
lar permeability coefficients between the internal for the differences in performance between the
oil phase and the pure oil. Analogous results were diverse emulsion types. For example, the higher
obtained with the w/o emulsion in which the emul- skin uptake from o/w emulsions could have been
sifier, sorbitan sesquioleate (Arlacel® 83), was due to a higher concentration of free hydrophilic
nearly entirely distributed in the oil phase of the actives in the external aqueous phase of the emul-
emulsion, whereas the aqueous phase was in effect sions being directly in contact with the skin,
free of sorbitan sesquioleate (Arlacel® 83). This whilst the actives were encapsulated in the inter-
yielded no solubility increase in the internal aque- nal phase of the w/o/w and w/o emulsions and as
ous phase and thus no reduction in permeability, a result not readily available to the stratum cor-
when compared with water. However, the solubil- neum (Ferreira et al. 1995b; Youenang Piemi
ity of each compound increased in the oil phase, et al. 1998; Sah et al. 1998). It was hence sug-
because of the formation of inverse micelles. gested that w/o/w and w/o emulsions could be
Consequently, the permeability of the three test utilised for the controlled release of water-soluble
permeants was reduced from both the w/o emul- actives (Sah et al. 1998). As shown by Ferreira
sion and from the corresponding isolated external et al. (1994, 1995b), the release of glucose and
oil phase, when compared with the pure oil. metronidazole through cellulose membranes and
Several studies have been reported, involving silicone membranes was in the following order:
different emulsion types with identical composition o/w > w/o/w > w/o. The release from the w/o/w
228

Table 16.1 Influence of emulsion type on release, dermal and transdermal delivery – list of examples for different model drugs
Effect on transdermal
Model drug Emulsifier system Dosing F vs. IFa Effect on release Effect on dermal delivery delivery Reference
Glucose Hypermer™ A60, IF o/w > w/o/w > w/o o/w > w/o/w > w/o o/w > w/o/w > w/o Ferreira et al. (1995b)
Synperonic™ PE/ F – o/w = w/o/w = w/o (dermis: o/w > w/o/w = w/o Ferreira et al. (1995a)
F127b o/w > w/o/w)
Lecithin (Emulmetik F – Epidermis: o/w = w/o/w = o/w > w/o/w = w/o Youenang Piemi et al. (1998)
™ 100/300) w/o
Dermis: o/w > w/o/w = w/o
IF – Epidermis: o/w = w/o/w = o/w > w/o/w = w/o
w/o
Dermis: o/w = w/o/w > w/o
Metronidazole Hypermer™ A60, IF o/w > w/o/w > w/o – o/w = w/o/w > w/o Ferreira et al. (1994)
Synperonic™ PE/ F – – o/w = w/o/w = w/o Ferreira et al. (1995a)
F127b (o/w < w/o)
Lactic acid Hypermer™ A60, F – o/w > w/o/w > w/o o/w < w/o/w = w/o Sah et al. (1998)
Synperonic™ PE/
F127b
Hydrocortisone Nonionic surfactantsc F – w/o < o/w/o w/o > o/w/o Laugel et al. (1998b)
IF w/o > o/w/o – w/o = o/w/o
Terpenes (asiatic acid, Mixture of nonionic F – w/o < o/w/o w/o > o/w/o Laugel et al. (1998a)
madecassic acid and surfactants
asiaticoside)
a
Finite (F), infinite (IF)
b
Hypermer™ A60 (a modified polyester)/Synperonic™ PE/F127 (poloxamer 407)
c
Glycerol sorbitan fatty acid ester, hydroxyoctacosanyl hydroxystearate and copolymer of ethylene and propylene oxides
A. Otto and J. du Plessis
16 The Effects of Emulsifiers and Emulsion Formulation Types on Dermal and Transdermal Drug Delivery 229

Fig. 16.3 (a) Percutaneous absorption profiles of glucose (b) Percutaneous absorption profiles of metronidazole from
from w/o/w, o/w and w/o emulsions through hairless rat w/o/w (□), o/w (▲) and w/o (○) emulsions. Values are the
skin; typical plots of the cumulative amount of glucose as a means (n = 5) ± SD. The SD values for the o/w emulsions are
function of time. Values are means (n = 6) (Reproduced from not represented for purposes of clarity (Reproduced from
Ferreira et al. (1995b) with permission from Elsevier). Ferreira et al. (1994), with permission from Elsevier)
230 A. Otto and J. du Plessis

a b

Fig. 16.4 Evaporation of water from emulsions contain- the applied amount (Ferreira et al. (1995a), with permis-
ing (a) metronidazole and (b) glucose as a function of sion from Elsevier)
time. The data are expressed as percentage of water loss of

emulsion was higher than from the w/o emulsion, the external aqueous phase may have contributed
due to the leakage of glucose and metronidazole to the hydration of the stratum corneum, which in
into the external aqueous phase, resulting in a turn may have enhanced the permeability of
higher effective concentration of the hydrophilic hydrophilic compounds (Ferreira et al. 1995b).
actives in the external aqueous phase of w/o/w This effect could be more pronounced for an infi-
emulsions than in the external oil phase of w/o nite, occluded dosing condition than for a finite,
emulsions (Ferreira et al. 1995a, b). It should also non-occluded dosing (Sah et al. 1998).
be noted that the differences in polarity of the It is also important to consider the fate of the
active ingredients further impacted on the perfor- emulsion after application onto the skin and its
mances of the various emulsions. So the differ- effect on the delivery of the actives. Following
ence between the various emulsions was more the application onto the skin, volatile compo-
pronounced for glucose (high polarity) than for nents (e.g. water) can evaporate, and therefore
metronidazole (intermediate polarity), due to a phase transitions, inversion, flocculation and
smaller oil-water partitioning coefficient of coalescence may occur (Friberg and Langlois
glucose, indicating a higher concentration of 1992). In addition, the drug concentration in the
glucose in the aqueous phase, when compared residual film could increase, due to water evapo-
with metronidazole (Ferreira et al. 1995b). ration from a finite dose (Sah et al. 1998).
Furthermore, the partitioning between the oil and Consequently, consideration of the evaporation
water phases in the emulsion was better for met- of volatile components, as well as the vehicle
ronidazole, which in turn yielded a decrease in structure of the remaining film after the evapora-
the internal release barrier and therefore a less tion of volatile components, is of importance
pronounced difference in performance between when investigating skin penetration of actives.
the various metronidazole emulsions (Ferreira Ferreira et al. (1995a) investigated water evapo-
et al. 1995a). ration from three different emulsions (o/w,
Differences in dermal and transdermal delivery w/o/w and w/o) and found that the rate thereof
could also have occurred as a result of different was higher from emulsions with an aqueous con-
partitioning coefficients between the stratum cor- tinuous phase (o/w and w/o/w) than from emul-
neum and the various emulsions, e.g. a higher par- sions with an oily continuous phase (w/o)
titioning between the stratum corneum and (Fig. 16.4), which may partially explain the dif-
external aqueous phase (for o/w and w/o/w ferences in dermal and transdermal delivery.
emulsions) than between the stratum corneum and They also investigated the structure of the resid-
external oil phase (for w/o emulsions). In addition, ual film after evaporation was completed and its
16 The Effects of Emulsifiers and Emulsion Formulation Types on Dermal and Transdermal Drug Delivery 231

effect on the lipid organisation of the stratum w/o emulsion (Laugel et al. 1998a, b). Similarly,
corneum. No differences were detected among a release study showed that the release of hydro-
the three emulsions (o/w, w/o/w and w/o) though. cortisone was slower from a multiple o/w/o emul-
Youenang Piemi et al. (1998) reasoned in their sion than from a simple w/o emulsion, as the
article that the similar performances of w/o/w active needed to diffuse from the internal phase,
and w/o emulsions in the dermal and transder- across the aqueous phase and into the external
mal delivery of glucose could be due to a similar phase (Laugel et al. 1998b). Both studies hence
vehicle structure of the w/o/w and w/o emul- confirmed that multiple o/w/o emulsions could
sions after application onto the skin, because of be used as prolonged, topical delivery systems
the evaporation of the external water phase of the for lipophilic drugs, when incorporated in the
w/o/w emulsion. internal oily phase and applied as a finite dose.
It should be noted that most of the studies on Furthermore, these multiple emulsions exhibited
hydrophilic compounds, as listed in Table 16.1, the advantages of reducing the transdermal deliv-
were performed with the same synthetic emulsi- ery and therefore the systemic effects of the lipo-
fier system Hypermer™ A60 (a modified polyes- philic drugs, whereas the dermal delivery was
ter)/poloxamer 407 (Synperonic™ PE/F127). increased, thus showing a controlled release of
However, the one study by Youenang Piemi et al. the drugs to the site of action (Laugel et al.
(1998) incorporated natural soybean phospho- 1998b). However, it should be noted that no sig-
lipids (lecithin, Emulmetik™ 100/300) as emul- nificant differences in transdermal delivery were
sifier in order to obtain different types of observed between w/o and o/w/o emulsions,
emulsions. Overall, the same trend was observed when an infinite dose of hydroquinone contain-
with the soybean phospholipids as with the syn- ing emulsions was applied onto the skin (Laugel
thetic emulsifiers. This was indicative of the et al. 1998b).
importance of the nature of the continuous phase
of an emulsion on the dermal and transdermal
delivery of hydrophilic drugs (Youenang Piemi 16.5.4 Stabilisation Effects
et al. 1998). Furthermore, the results of the vari- of the Formulation
ous studies with hydrophilic active ingredients
indicated that the type of emulsion (o/w, w/o/w, When investigating the dermal and transdermal
w/o) may not significantly affect the distribution delivery of active ingredients from emulsions,
of the actives between dermal and transdermal consideration of the stabilisation effects of a
delivery, as the order of emulsions was similar formulation on delivering degradation-sensitive
for dermal and transdermal delivery (see active ingredients intact into and/or across the
Table 16.1). skin can prove beneficial. Schmidts et al. (2011)
investigated the stabilisation effects of various
emulsion systems against enzymatic degrada-
16.5.3 Lipophilic Active Ingredients tion of topically applied oligonucleotides. They
found that the enzymatic degradation of water-
As with the hydrophilic drugs, the encapsulation soluble DNAzymes, encapsulated in the inner
of the lipophilic actives (hydrocortisone and aqueous phase of w/o/w and w/o emulsions, was
three different triterpenic derivatives) in the inter- significantly reduced, compared to DNAzymes,
nal oily phase of multiple o/w/o emulsions incorporated in the outer aqueous phase of a
reduced the percutaneous absorption of these microemulsion and a submicron emulsion. The
lipophilic actives, compared to that of simple w/o outcomes of their study suggested that w/o
emulsions, when applied as a finite dose. and w/o/w emulsions are promising formula-
However, the uptake of the lipophilic actives into tions for effective encapsulation of DNAzymes
the epidermis and dermis was higher from the with concurrent protection against enzymatic
multiple o/w/o emulsion than from the simple degradation.
232 A. Otto and J. du Plessis

16.6 The Effects of Various methoxycinnamate (Uvinul MC 80®) across


Emulsifiers on Dermal human skin, whereas the permeation of butyl
and Transdermal Delivery methoxydibenzoylmethane (Uvinul BMBM®)
was insignificantly affected. It was assumed that
16.6.1 Overview changes in the thermodynamic activity in the
emulsion and modification of the interaction
As was mentioned above, various substances between permeant and emulsion components
exist that can be utilised as emulsifiers (e.g. sur- could account for the different effects of the
factants, polymers, solid particles), and often the emulsifiers on skin permeation. Though the
stabilisation effect of emulsifiers can be attrib- inclusion of different silicone emulsifiers altered
uted to more than one method. For instance, some the viscosity of the emulsions and the release of
surfactants may form a monolayer at the oil- the active ingredients, these factors could not be
water interface, whilst an excess thereof may also related to the modification in permeation.
arrange in liquid crystalline structures in the Wiechers et al. (2004) suggested that the
aqueous phase, which could improve emulsion emulsifier system may influence the distribution
stability by preventing coalescence (Friberg and of the active ingredient between dermal and
Solans 1986). Hydrophobically modified water- transdermal delivery, whilst emollients may sig-
soluble polymers can stabilise o/w emulsions by nificantly affect the total skin absorption (dermal
adsorbing at the oil-water interface (hydrophobic + transdermal delivery) thereof. For example, the
part of the polymer), as well as by gelation of the emulsion with the emulsifier system, sorbitan
aqueous continuous phase (hydrophilic part of stearate/sucrose cocoate (Arlatone® 2121), exhib-
the polymer) (Eccleston 1997a). In case of ited a higher transdermal, but lower dermal,
Pickering emulsions, the particles can, in addi- delivery of octadecenedioic acid (Arlatone™
tion to the adsorption at the oil-water interface, DIOIC DCA), in comparison with the emulsion
also form a three-dimensional network in the containing steareth-2/steareth-21 (Brij® 72/721).
continuous phase surrounding the droplets. The No explanations could be given as more investi-
resulting increased viscosity of the emulsions can gations were required to understand the influence
reduce the rate and extent of creaming (Aveyard of the emulsifier system on skin delivery.
et al. 2003). Furthermore, the complexity of These studies, using the same oil and aqueous
emulsions makes it more difficult to study the phases for the emulsions, demonstrated that
exclusive effect of emulsifiers on dermal and emulsifiers significantly affected dermal and
transdermal delivery, as other emulsion ingredi- transdermal delivery. However, the emulsifiers
ents also contribute to interactions with the active being compared varied in structure and physico-
ingredient in the vehicle, as well as with the stra- chemical properties, making it difficult to explain
tum corneum. Therefore, this chapter aimed at their particular effects on skin penetration and
focusing mainly on studies in which a more sys- permeation. The following examples focus more
tematic approach to investigating the effects of on studies that compare emulsifiers with particu-
emulsifiers on dermal and transdermal delivery lar differences (e.g. variation in hydrophilic chain
was followed, such as using emulsions with the length of nonionic surfactants, HLB values,
same oil and aqueous phases and hence reducing emulsion droplet charge).
the influences of varying formulation ingredients
on the active’s transport into and across the skin.
Montenegro et al. (2004) investigated the 16.6.2 Effects of Hydrophilic
effects of various silicone emulsifiers using the Chain Length of Nonionic
same oil and aqueous phase ingredients for Surfactants
the preparation of the emulsions. The study illus-
trated that the type of silicone emulsifier could Oborska et al. (2004) investigated the effect of
significantly affect the permeation of octyl three different polyoxyethylene cetostearyl ethers
16 The Effects of Emulsifiers and Emulsion Formulation Types on Dermal and Transdermal Drug Delivery 233

of various oxyethylene chain lengths (ceteareth-12 retinol between the skin and formulation. These
(Eumulgin® B1), ceteareth-20 (Eumulgin® B2) results were congruent with those from studies by
and ceteareth-30 (Eumulgin® B3)) in o/w emul- Oborska et al. (2004) and Dalvi and Zatz (1981),
sions on the permeation of two flavonoids, quer- demonstrating that drug permeation was inversely
cetin and rutin, through a liposome model related to the hydrophilic chain length of the sur-
membrane. The study revealed that the permea- factants, due to a change in solubilisation capacity
bility coefficient of both permeants decreased of the surfactant micelles, without an apparent
with increasing length of the oxyethylene chain interaction between the surfactants and the stratum
and their effects were more pronounced for rutin, corneum lipids (Dalvi and Zatz 1981).
the more water-soluble flavonoid. No explanation
was given. However, a similar trend was observed
in a study by Dalvi and Zatz (1981), when inves- 16.6.3 Effects of Emulsifier Ratios
tigating the effects of the polyoxyethylene chain and HLB Value
length of nonionic surfactants on benzocaine flux
from aqueous solutions. The reduction in benzo- As mentioned, often emulsifier combinations are
caine flux from non-saturated solutions with used to manufacture and stabilise topical emul-
increasing polyoxyethylene chain length was sions. The studies discussed next illustrate the
explained by a higher micellar entrapment of influence that an emulsifier mixture may exhibit
benzocaine and therefore a lower concentration on dermal and transdermal delivery. These exam-
of free benzocaine giving rise to a lower driving ples also include nanoemulsions, although a sep-
force for permeation. Although this study was arate book chapter deals with nanoemulsions as
performed on aqueous solutions, the solubilisa- skin delivery systems in more detail.
tion effects of the nonionic surfactants with Nam et al. (2012) showed that the incorpora-
increasing length of the oxyethylene chain could tion of a second emulsifier (Phospholipon® 90 G,
have also occurred with quercetin and rutin in the mostly soybean phosphatidylcholine with max.
o/w emulsions used by Oborska et al. (2004). 4.0 % lysophosphatidylcholine) with skin perme-
A study by Förster et al. (2011) tested o/w ation-enhancing capabilities could increase skin
emulsions with different surfactants of the poly- absorption of the active ingredient, tocopheryl
ethylene glycol ester type, which varied in the acetate. Mixtures of Phospholipon® 90 G and a
length of the alkyl- and polyethylene glycol chains polymer (poly(ethylene oxide)-block-poly(ε-
(PEG6C18:1, PEG20C12 and PEG20C18:1), for caprolactone) (PEO-b-PCL)) in various ratios
skin penetration of the lipophilic active, retinol. were used to stabilise o/w nanoemulsions being
The results confirmed that surfactants with a short tested for their skin absorption effects. The per-
polar head group had an enhancement effect on meation-enhancing effects of the unsaturated
penetration into the skin, as the penetration into lipid yielded improved skin absorption of tocoph-
the epidermis and dermis from emulsions with eryl acetate from nanoemulsions, with increasing
PEG6C18:1 was higher than from emulsions with lipid-to-polymer ratios (Fig. 16.5). In compari-
PEG20C12 and PEG20C18:1. Confocal Raman son, the nanoemulsion, solely being stabilised
microspectroscopy revealed that all three tested with the polymer, PEO-b-PCL, exhibited a much
emulsions did not differ in the lateral interaction lower skin penetration. It was furthermore indi-
(ratio of I2880/I2850), an indicator for the lateral cated that not only the permeation-enhancing
packing of lipids. Furthermore, the ratio I2880/I2850 effect of the unsaturated lipid but also the smaller
for all three emulsions did not significantly differ size of the emulsion droplets could have yielded
from the ratio for untreated skin. As there was no an improved skin delivery of tocopheryl acetate.
indication of disruption of the lipid structure of the Another study by Cho et al. (2012) utilised a
stratum corneum by any of the emulsions, the triblock copolymer (poly(ethylene oxide)-block-
increased dermal delivery with PEG6C18:1 was poly ( ε -caprolactone)- block -poly(ethylene
linked to a change in partitioning behaviour of oxide) (PEO-PCL-PEO)) to co-stabilise a retinol
234 A. Otto and J. du Plessis

Fig. 16.5 Cumulative


amount of tocopheryl acetate
absorbed by the hairless
guinea pig skin form
nanoemulsions stabilised
using a mixture of a lipid
(P90G) and a polymer
(PEO45-b-PCL42) as a function
of the lipid-polymer ratio for
12 h (●) and 24 h (■)
(Reproduced from Nam et al.
(2012), with permission from
Elsevier)

emulsion that was primarily stabilised using


polysorbate 20 (Tween® 20). Their work included
an investigation of the effect of the PCL block
length of the triblock copolymer on the topical
delivery of retinol, and it was ascertained that the
accumulation of retinol in the artificial skin was
enhanced by employing the triblock copolymer,
as well as by increasing the PCL block length. It
was, furthermore, found that with increasing
length of the PCL block, the hydrophilic-lipo-
philic balance (HLB) value, as well as the size of
the emulsion droplets decreased. The outcomes
from this study suggested that the PCL block
length and HLB value are important consider-
ations for the topical delivery of actives from
emulsions being co-stabilised with a triblock
copolymer. Fig. 16.6 Correlation between permeation rate of inulin
across hairy mouse skin (expressed as per cent of applied
Wu et al. (2001) found that the rate and extent
formulation per h ± SE) and HLB of surfactant mixture in
of inulin permeation from water-in-oil nano- nanoemulsion or micellar formulation. HLB of surfactant
emulsions were highly dependent on the HLB mixture = ((volume per cent Span® 80 in formulation ×
values of the incorporated surfactant mixtures 4.3) + (volume per cent Tween® 80 in formulation ×
15.0))/100 (Reprinted from Wu et al. (2001), with permis-
(different ratios of sorbitan monooleate (Span®
sion from Elsevier)
80) and polysorbate 80 (Tween® 80)). Surfactant
mixtures with a low HLB resulted in significantly
higher permeation of inulin than surfactant mix- its transport was facilitated by nanoemulsions in
tures with a high HLB (Fig. 16.6). It was sug- which the HLB of the oil phase was compatible
gested that inulin, encapsulated into the aqueous with the sebum environment of the hair follicle.
phase of the w/o nanoemulsions, was mainly These examples illustrate that the type of
transported via the transfollicular route and that emulsifier/emulsifier system, together with the
16 The Effects of Emulsifiers and Emulsion Formulation Types on Dermal and Transdermal Drug Delivery 235

Fig. 16.7 Permeation profiles


of econazole and miconazole
nitrate incorporated in
negatively (−) and positively
(+) charged submicron
emulsion across hairless rat
skin determined by
HPLC. Approximately 37 μg
of drug were applied per cell
with an effective diffusional
area of 0.635 cm2; n = 6; mean
± S.D (Reprinted from
Youenang Piemi et al. (1999),
with permission from
Elsevier)

ratio of the emulsifier mixture, is not only essen- transdermal delivery. On the contrary, Nam et al.
tial to the emulsification and stabilisation effects (2012) showed that the size of the emulsion drop-
but potentially also for modifying skin penetra- lets could affect skin absorption. They prepared
tion significantly. Moreover, the HLB value may nanoemulsions with the same chemical composi-
considerably influence topical performance. tion but different droplet sizes by applying differ-
Some studies reported that the various emulsi- ent pressure in the microfluid process and found
fiers and/or ratios of emulsifier mixtures also that smaller droplet sizes enhanced the skin
altered the droplet size and viscosity of the emul- absorption of tocopheryl acetate. However, trans-
sions, which may have affected dermal and trans- dermal delivery was not determined in this study.
dermal delivery. The study by Klang et al. (2011),
however, revealed that neither skin permeation
nor penetration was influenced by the droplet size 16.6.4 Effects of Emulsion Droplet
and viscosity. Klang et al. (2011) used sucrose Charge
stearate S-970 (Ryoto® Sugar Ester) as emulsifier
to formulate stable o/w emulsions by forming a Positively and negatively charged submicron
hydrophilic network around the oil droplets. emulsions were compared by Youenang Piemi
Through a slight modification of the manufactur- et al. (1999) for their effects on the dermal and
ing process, they were able to produce a highly transdermal delivery of econazole and micon-
viscous macroemulsion and a less viscous, fluid azole nitrate. The composition of the emulsions
nanoemulsion with exactly the same composi- was the same and only differed in the emulsifier
tion, thus enabling the investigation of the influ- component giving the charge to the emulsion
ence of droplet size and viscosity on dermal and droplets, i.e. stearylamine in the case of positively
transdermal delivery. The macro- and nanoemul- charged emulsions and deoxycholic acid in case
sions showed neither significant difference in of negatively charged emulsions. In overall, the
flux, penetration depth nor in accumulation of the positively charged o/w emulsion containing stea-
three model drugs used (curcumin, flufenamic rylamine was more effective in dermal and trans-
acid, diclofenac acid) in the stratum corneum. dermal delivery of econazole and miconazole
The results regarding droplet size are congruent nitrate than the negatively charged o/w emulsion
with the outcome of the study by Izquierdo et al. containing deoxycholic acid (Fig. 16.7). It was
(2007), where also no correlation could be found suggested that positively charged submicron
between the droplet size and the dermal and emulsion droplets could facilitate the transport of
236 A. Otto and J. du Plessis

the permeants into and through the skin, most emulsions without liquid crystals (polysorbate 60
probably owing to a superior binding of the posi- (Tween® 60), poloxamer 407 (Synperonic™ PE/
tively charged droplets to the skin, which is nega- F127) and acrylates/C10-30 alkyl acrylate
tively charged at neutral pH. crosspolymer (Pemulen® TR1)). The differences
Two studies by Ghouchi Eskandar et al. (2009a, in permeation could be due to modified interac-
2010), investigating oil-in-water submicron emul- tions between surfactants and permeant that may
sions, also showed the possible dependency of have influenced the interactions between the sur-
dermal delivery on the emulsifier charge. The skin factants and stratum corneum. Furthermore, the
retention of the tested lipophilic compounds, all- partitioning between the formulation and the skin
trans-retinol and acridine orange 10-nonyl bro- could have been altered, e.g. partitioning between
mide, was significantly higher for those emulsions the skin and the aqueous phase (emulsions with-
being stabilised by the positively charged oleyl- out liquid crystals) and partitioning between the
amine, compared to emulsions, stabilised by the skin and the liquid crystal phase (emulsions with
negatively charged lecithin. Two explanations liquid crystals).
were given. Firstly, the higher skin accumulation The effect of liquid crystalline structures in
of the lipophilic compounds being released from o/w emulsions on dermal and transdermal deliv-
the oleylamine-stabilised emulsions could be ery was also investigated by Otto et al. (2010).
attributed to the electrostatic attraction between Five o/w emulsions were tested:
the positively charged oleylamine droplets and • ‘Hydrosome’ emulsion, stabilised by sorbitan
negatively charged skin surface. Secondly, it could stearate and sucrose cocoate (Arlatone® 2121),
also have been due to reduced skin barrier proper- with lamellar gel structuring of the water
ties, as a result of the disrupting effect of oleyl- phase (Tadros et al. 2006)
amine on the stratum corneum lipid organisation. • ‘Phosphosome EFA’ (essential fatty acid)
emulsion, stabilised by linoleamidopropyl
PG-dimonium chloride phosphate (Arlasilk™
16.6.5 Effects of Surfactant Phospholipid EFA), with structuring of the
Association Structures water phase and lamellar liquid crystalline
phases around the oil droplets
When viewing the effects of surfactants, one • ‘Phosphosome PTC’ (phosphatidylcholine)
should not solely consider the surfactant but also emulsion, stabilised by cocamidopropyl
the association structures that could form in PG-dimonium chloride phosphate (Arlasilk™
emulsions. In addition to the formation of a sur- Phospholipid PTC), with structuring of the
factant monolayer at the oil-water interface, some water phase and lamellar liquid crystalline
surfactants, when in excess, may arrange in liquid phases around the oil droplets
crystalline structures in the aqueous phase, which • ‘Oleosome’ emulsion, stabilised by steareth-
may aid the stabilisation of emulsions (Friberg 2/steareth-21 (Brij® 72/721) incl. cetearyl
and Solans 1986) and could affect skin perme- alcohol (Laurex CS™) and PPG-15 stearyl
ation. Only two studies are mentioned below to ether (Arlamol™ E), with surfactant bilayers
illustrate the effect of the emulsifier association around the oil droplets (Tadros et al. 2006)
structures on skin permeation. For more informa- • ‘Conventional’ emulsion, stabilised by stea-
tion on liquid crystalline structures, the reader is reth-2/steareth-21 (Brij® 72/721), without liq-
referred to a separate chapter in the book. uid crystalline phases
A study by Brinon et al. (1998) illustrated that The study revealed that the hydrosome and
the flux of benzophenone-4 (Uvinul® MS40) was phosphosome emulsions, with lamellar gel struc-
increased by emulsions containing lamellar liq- turing of the water phase, enhanced the dermal and
uid crystals (triethanolamine stearate, sorbitan transdermal delivery of hydroquinone and
stearate/sucrose cocoate (Arlatone® 2121) and octadecenedioic acid (Arlatone™ DIOIC DCA),
steareth-2/-21 (Brij® 72/721) in comparison to when compared with the conventional o/w
16 The Effects of Emulsifiers and Emulsion Formulation Types on Dermal and Transdermal Drug Delivery 237

a
conv o/w Oleosome Hydrosome conv o/w Oleosome Hydrosome
Phosph EFA Phosph PTC Phosph EFA Phosph PTC
200
HQ absorbed (µg/cm2) ± 95% C.I.

140
180 *

HQ permeated (µg/cm2)
120
160 *
100
140
120 * 80
100 60
80
40
60
20
40
20 * * 0
0 2 4 6 8
0
tape strips rest skin receptor time (h)

b conv o/w Oleosome Hydrosome conv o/w


Phosph EFA
Oleosome
Phosph PTC
Hydrosome
Phosph EFA Phosph PTC
DIOIC absorbed (µg/cm2) ± 95% C.I.

70 60
*

DIOIC permeated (µg/cm2)


60 50
50 40
40 30
30 20
20 10
*
10 0
0 5 10 15 20 25
0
rest skin receptor time (h)

c conv o/w Oleosome Phosph EFA Phosph PTC conv o/w Oleosome Phosph EFA Phosph PTC
90 90
SA absorbed (µg/cm2) ± 95% C.I.

80
SA permeated (µg/cm2)

80
70 70
60
60
50
50
40
40
30
30 20
20 10
10 0
* 0 2 4 6 8 10 12
0
tape strips rest skin receptor time (h)

Fig. 16.8 (a) Skin permeation data on hydroquinone obtained after two-way ANOVA. Conv. conventional,
(HQ) expressed as total amount delivered over 8 h of skin Phosph. phosphosome. *Statistically significant compared
penetration as least square means (formulation effect) ± with the conventional o/w emulsion (Reprinted from Otto
95 % confidence interval (CI) obtained after two-way et al. (2010), with permission from Karger). (c) Skin per-
ANOVA. Conv. conventional, Phosph. phosphosome. meation data on salicylic acid (SA) expressed as total
*Statistically significant compared with the conventional amount delivered over 12 h of skin penetration as least
o/w emulsion (Reprinted from Otto et al. (2010), with per- square means (formulation effect) ± 95 % confidence
mission from Karger). (b) Skin permeation data on octa- interval (CI) obtained after two-way ANOVA. Conv. con-
decenedioic acid (DIOIC) expressed as total amount ventional, Phosph. phosphosome. *Statistically significant
delivered over 24 h of skin penetration as least square compared with the conventional o/w emulsion (Reprinted
means (formulation effect) ± 95 % confidence interval (CI) from Otto et al. (2010), with permission from Karger)

emulsion without liquid crystalline phases and the for salicylic acid, where no differences in skin
oleosome emulsion with lamellar liquid crystal- penetration occurred between the o/w emulsions
line phases around the oil droplets (Fig. 16.8a, b). with and without liquid crystalline phases
The increase in skin penetration was ascribed to an (Fig. 16.8c). It was assumed that the interactions
improved partitioning of both permeants into the between the different emulsifiers and active ingre-
skin. However, a different pattern was observed dients varied and therefore the solubilisation
238 A. Otto and J. du Plessis

capacities of the various emulsifiers and their asso- stratum corneum, whereas the conventional
ciation structures, which could have resulted in emulsion increased the transport through the stra-
changes in the thermodynamic activity of the per- tum corneum into the viable epidermis and der-
meants in the emulsions. mis. It was hypothesised that the enhanced
accumulation of all-trans-retinol in the stratum
corneum could be due to a lack of flexibility of
16.6.6 Effects of Solid Particles the Pickering emulsion droplets.
as Emulsifiers The former studies compared Pickering emul-
sions being stabilised solely by solid particles
More recently, new topical emulsion systems with conventional emulsions being stabilised
have been introduced that are surfactant free and solely by surfactants. However, emulsions with
being stabilised by solid particles. These particle- mixed interfacial layers, for instance, including
stabilised emulsions are also known as Pickering both surfactants and solid particles, were also
emulsions. Another chapter in this book dis- tested for their topical performances, as discussed
cusses Pickering emulsions as topical delivery next.
systems; however, a few examples are mentioned Silica nanoparticle coatings of lecithin- and
below to illustrate the differences in topical per- oleylamine-stabilised oil-in-water submicron
formance due to different emulsifiers. emulsions exhibited an enhanced dermal delivery
Frelichowska et al. (2009b) investigated the of all-trans-retinol and acridine orange 10-nonyl
skin absorption of the hydrophilic active caffeine bromide, compared to non-silica-coated emul-
from silica particle-stabilised w/o emulsions sions (Ghouchi Eskandar et al. 2009a, 2010). As
(Pickering emulsions) and compared it to the the silica nanoparticles significantly increased
absorption from surfactant-stabilised w/o emul- the emulsification efficiency, as well as the physi-
sions (conventional emulsions). These emulsions cal stability of the emulsions (Ghouchi Eskandar
had the same physicochemical properties (drop- et al. 2007), they could be considered as emulsi-
let size and viscosity), as well as chemical com- fiers. In both studies it was observed that the
position and only varied in the type of emulsifier. extent and the depth of penetration of lipophilic
The study revealed that the caffeine flux was all-trans-retinol and acridine orange 10-nonyl
threefold higher from the Pickering emulsion bromide into full-thickness porcine skin were
than from the conventional emulsion, although increased by nanoparticle-coated emulsions and
the release was slower from the Pickering emul- they both were affected by the initial loading
sion. The increased skin absorption could be phase of the silica nanoparticles during the prep-
explained by the higher adhesion of the silica aration of the emulsions (e.g. incorporation into
particle-stabilised water droplets onto the skin the water phase vs. oil phase). However, the silica
surface. Furthermore, it was found that silica par- coating did not influence the transdermal delivery
ticles penetrated into the upper layers of the stra- of the two compounds, which was considered
tum corneum and it was hypothesised that negligible. It was further shown that silica
caffeine was transported into the skin by means nanoparticles could also penetrate the skin up to
of adsorption onto the silica particles. the viable epidermis and dermis, with negligible
Another study by Frelichowska et al. (2009a), permeation across the skin. There was a correla-
investigating oil-in-water emulsions, revealed tion found between the skin accumulation and
that the total skin uptake of the lipophilic drug, distribution of acridine orange 10-nonyl bromide
all-trans-retinol, was similar for the conventional and silica nanoparticles, and it was hypothesised
emulsion and the Pickering emulsion and no that the transport of acridine orange 10-nonyl
transdermal delivery was observed. However, the bromide could have been facilitated by electro-
distribution of all-trans-retinol within the skin static complexation with silica nanoparticles
varied. The Pickering emulsion showed an (Ghouchi Eskandar et al. 2010). Furthermore, the
enhanced accumulation of all-trans-retinol in the improved physical stability of silica-coated
16 The Effects of Emulsifiers and Emulsion Formulation Types on Dermal and Transdermal Drug Delivery 239

emulsions and enhanced chemical stability of all- of the resulting emulsion but it is also impor-
trans-retinol (Ghouchi Eskandar et al. 2009b) are tant for the dermal and transdermal perfor-
other possible mechanisms for the increased skin mance that additionally is influenced by the
retention. Additionally, the formation of a thick type of emulsion.
film of silica nanoparticles and emulsion oil
droplets on the skin surface could have had an
occlusive effect that may have increased the
hydration of the stratum corneum, which in turn References
could have enhanced skin penetration (Ghouchi
Anton N, Vandamme TF (2011) Nano-emulsions and
Eskandar et al. 2009a). micro-emulsions: clarifications of the critical differ-
ences. Pharm Res 28:978–985
Aveyard R, Binks BP, Clint JH (2003) Emulsions stabi-
Conclusion lised solely by colloidal particles. Adv Colloid Interf
Sci 100–102:503–546
This review has emphasised that different Bancroft WD (1913) The theory of emulsification. J Phys
parameters/variables require consideration when Chem 17:501–520
deciding on an appropriate emulsion type and Bernardo FP, Saraiva PM (2008) A theoretical model for
emulsifier system. For example, the skin uptake transdermal drug delivery from emulsions and its
dependence upon formulation. J Pharm Sci
into the skin as well as the permeation through 97:3781–3809
the skin of a hydrophilic active could be Binks BP, Whitby CP (2005) Nanoparticle silica-
enhanced, when the active is incorporated stabilized oil-in-water emulsions: improving emulsion
into the continuous phase of the emulsion. stability. Colloids Surf A 253:105–115
Brinon L, Geiger S, Alard V, Tranchant J-F, Pouget T,
Furthermore, multiple o/w/o emulsions, in Couarraze G (1998) Influence of lamellar liquid
comparison to simple w/o emulsions, reduced crystal structure on percutaneous diffusion of a
the transdermal delivery of lipophilic active hydrophilic tracer from emulsions. J Cosmet Sci
ingredients, whereas the dermal delivery was 49:1–11
Cho HK, Cho JH, Choi S-W, Cheong IW (2012) Topical
increased. Therefore, multiple emulsions could delivery of retinol emulsions co-stabilised by PEO-
be considered if prolonged topical delivery or PCL-PEO triblock copolymers: effect of PCL block
the protection of the active ingredient against length. J Microencapsul. doi:10.3109/02652048.2012.
external influences (e.g. enzymatic degrada- 686528
Dal Pozzo A, Pastori N (1996) Percutaneous absorption of
tion, oxidation) is desired. It was demonstrated parabens from cosmetic formulations. Int J Cosmet
that the effects of surfactants, as incorporated in Sci 18:57–66
emulsions, on dermal and transdermal delivery Dalvi UG, Zatz JL (1981) Effect of nonionic surfactants
could vary, depending on their structure and on penetration of dissolved benzocaine through hair-
less mouse skin. J Soc Cosmet Chem 32:87–94
physicochemical properties, e.g. HLB value, Eccleston GM (1997a) Formulating cosmetic emul-
hydrophilic chain length of nonionic surfac- sions: – advances in understanding emulsion technol-
tants, emulsifier charge or their association ogy. Cosmet Toilet 112(12):65–71
structures in the emulsion. However, the emul- Eccleston GM (1997b) Functions of mixed emulsifiers
and emulsifying waxes in dermatological lotions and
sifier could also influence the droplet size and creams. Colloids Surf A 123–124:169–182
viscosity of emulsions, which in turn may addi- Ferreira LAM, Seiller M, Grossiord JL, Marty JP,
tionally affect topical performance. Pickering Wepierre J (1994) Vehicle influence on in vitro release
emulsions, a rather new topical delivery system of metronidazole: role of w/o/w multiple emulsion. Int
J Pharm 109:251–259
type, are not only interesting in terms of Ferreira LAM, Doucet J, Seiller M, Grossiord JL, Marty
improving the physical stability of emulsions JP, Wepierre J (1995a) In vitro percutaneous absorp-
but also show the potential for dermal skin tar- tion of metronidazole and glucose: comparison of o/w,
geting and controlled release. w/o/w, w/o systems. Int J Pharm 121:169–179
Ferreira LAM, Seiller M, Grossiord JL, Marty JP,
In summary, it can be concluded that the Wepierre J (1995b) Vehicle influence on in vitro
choice of the emulsifier is not only critical for release of glucose: w/o, w/o/w and o/w systems com-
the emulsification process and for the stability pared. J Control Rel 33:349–356
240 A. Otto and J. du Plessis

Förster T, Von Rybinski W (1998) Applications of emul- Klang V, Schwarz JC, Matsko N, Rezvani E, El-Hagin N,
sions. In: Binks BP (ed) Modern aspects of emulsion Wirth M, Valenta C (2011) Semi-solid sucrose
science. Royal Society of Chemistry, Cambridge, stearate-based emulsions as dermal drug delivery sys-
pp 395–426 tems. Pharmaceutics 3:275–306
Förster T, Jackwerth B, Pittermann W, Von Rybinski W, Lalor CB, Flynn GL, Weiner N (1995) Formulation fac-
Schmitt M (1997) Properties of emulsions – structure tors affecting release of drug from topical vehicles
and skin penetration. Cosmet Toilet 112(12):73–82 II. Effect of solubility on in vitro delivery of a series of
Förster M, Bolzinger MA, Ach D, Montagnac G, Briançon n-alkyl p-aminobenzoates. J Pharm Sci 84:673–676
S (2011) Ingredients tracking of cosmetic formula- Lan Q, Yang F, Zhang S, Liu S, Xu J, Sun D (2007)
tions in the skin: a confocal Raman microscopy inves- Synergistic effect of silica nanoparticle and cetyltri-
tigation. Pharm Res 28:858–872 methyl ammonium bromide on the stabilization of o/w
Frelichowska J, Bolzinger M-A, Pelletier J, Valour J-P, emulsions. Colloids Surf A 302:126–135
Chevalier Y (2009a) Topical delivery of lipophilic drugs Laugel C, Baillet A, Ferrier D, Grossiord JL, Marty JP
from o/w Pickering emulsions. Int J Pharm 371:56–63 (1998a) Incorporation of triterpenic derivatives within
Frelichowska J, Bolzinger M-A, Valour J-P, Mouaziz H, an o/w/o multiple emulsion: structure and release
Pelletier J, Chevalier Y (2009b) Pickering w/o emul- studies. Int J Cosmet Sci 20:183–191
sions: drug release and topical delivery. Int J Pharm Laugel C, Baillet A, Youenang Piemi MP, Marty JP,
368:7–15 Ferrier D (1998b) Oil-water-oil multiple emulsions for
Friberg SE (1990) Micelles, microemulsions, liquid crys- prolonged delivery of hydrocortisone after topical
tals, and the structure of stratum corneum lipids. J Soc application: comparison with simple emulsions. Int J
Cosmet Chem 41:155–171 Pharm 160:109–117
Friberg SE, Langlois B (1992) Evaporation from emul- McClements DJ (2012) Nanoemulsions versus micro-
sions. J Disp Sci Technol 13:223–243 emulsions: terminology, differences, and similarities.
Friberg SE, Ma Z (2006) Emulsions: factors and issues for Soft Matter 8:1719–1729
skin care. In: Wille JJ (ed) Skin delivery systems: Montenegro L, Paolino D, Puglisi G (2004) Effects of sili-
transdermals, dermatologicals, and cosmetic actives. cone emulsifiers on in vitro skin permeation of sun-
Wiley-Blackwell, Ames, pp 187–209 screens from cosmetic emulsions. J Cosmet Sci
Friberg SE, Solans C (1986) Surfactant association struc- 55:509–518
tures and the stability of emulsions and foams. Nam YS, Kim J-W, Park JY, Shim J, Lee JS, Han SH
Langmuir 2:121–126 (2012) Tocopheryl acetate nanoemulsions stabilized
Ghouchi Eskandar N, Simovic S, Prestidge CA (2007) with lipid-polymer hybrid emulsifiers for effective
Synergistic effect of silica nanoparticles and charged skin delivery. Colloids Surf B 94:51–57
surfactants in the formation and stability of submicron Oborska A, Arct J, Mojski M, Jaremko E (2004) Influence
oil-in-water emulsions. Phys Chem Chem Phys of polyalcohols and surfactants on skin penetration of
9:6426–6434 flavonoids from the emulsion. J Appl Cosmetol
Ghouchi Eskandar N, Simovic S, Prestidge CA (2009a) 22:35–42
Nanoparticle coated submicron emulsions: sustained Otto A, Wiechers JW, Kelly CL, Dederen JC, Hadgraft J,
in-vitro release and improved dermal delivery of all- du Plessis J (2010) Effect of emulsifiers and their liq-
trans-retinol. Pharm Res 26:1764–1775 uid crystalline structures in emulsions on dermal and
Ghouchi Eskandar N, Simovic S, Prestidge CA (2009b) transdermal delivery of hydroquinone, salicylic acid
Chemical stability and phase distribution of all-trans- and octadecenedioic acid. Skin Pharmacol Physiol
retinol in nanoparticle-coated emulsions. Int J Pharm 23:273–282
376:186–194 Sah A, Mukherjee S, Wickett RR (1998) An in vitro study
Ghouchi Eskandar N, Simovic S, Prestidge CA (2010) of the effects of formulation variables and product
Mechanistic insight into the dermal delivery from structure on percutaneous absorption of lactic acid. J
nanoparticle-coated submicron o/w emulsions. J Cosmet Sci 49:257–273
Pharm Sci 99:890–904 Schmidts T, Dobler D, von den Hoff S, Schlupp P, Garn H,
Grégoire S, Ribaud C, Benech F, Meunier JR, Garrigues- Runkel F (2011) Protective effect of drug delivery sys-
Mazert A, Guy RH (2009) Prediction of chemical tems against the enzymatic degradation of dermally
absorption into and through the skin from cosmetic and applied DNAzyme. Int J Pharm 410:75–82
dermatological formulations. Br J Dermatol 160:80–91 Tadros TF (2009) Emulsion science and technology: a
Harusawa F, Saito T, Nakajima H, Fukushima S (1980) general introduction. In: Tadros TF (ed) Emulsion sci-
Partition isotherms of nonionic surfactants in the ence and technology. Wiley VCH, Weinheim, pp 1–56
water-cyclohexane system and the type of emulsion Tadros T, Leonard S, Taelman M-C, Verboom C, Wortel V
produced. J Colloid Interface Sci 74:435–440 (2006) Correlating the structure and rheology of liquid
Izquierdo P, Wiechers JW, Escribano E, García-Celma crystalline phases in emulsions. Cosmet Toilet
MJ, Tadros TF, Esquena J, Dederen JC, Solans C 121(5):89–94
(2007) A study on the influence of emulsion droplet Wiechers JW (2005) Optimizing skin delivery of active
size on the skin penetration of tetracaine. Skin ingredients from emulsions from theory to practice.
Pharmacol Physiol 20:263–270 In: Rosen MR (ed) Delivery system handbook for
16 The Effects of Emulsifiers and Emulsion Formulation Types on Dermal and Transdermal Drug Delivery 241

personal care and cosmetic products. William Andrew, Youenang Piemi MP, De Luca M, Grossiord J-L, Seiller
Norwich, pp 409–436 M, Marty J-P (1998) Transdermal delivery of glucose
Wiechers JW, Kelly CL, Blease TG, Dederen JC through hairless rat skin in vitro: effect of multiple and
(2004) Formulating for efficacy. Int J Cosmet Sci simple emulsions. Int J Pharm 171:207–215
26:173–182 Youenang Piemi MP, Korner D, Benita S, Marty J-P
Wu H, Ramachandran C, Weiner ND, Roessler BJ (2001) (1999) Positively and negatively charged submicron
Topical transport of hydrophilic compounds using emulsions for enhanced topical delivery of antifungal
water-in-oil nanoemulsions. Int J Pharm 220:63–75 drugs. J Control Rel 58:177–187
Skin Permeation: Enhancing
Ability of Liquid Crystal 17
Formulations

Wesam R. Kadhum, Hiroaki Todo,


and Kenji Sugibayashi

Contents 17.1 Introduction


17.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . 243
The primary dosage forms used in drug therapy
17.2 Liquid Crystals (LC) . . . . . . . . . . . . . . . . 244
are oral formulations and injections; however,
17.3 Current Problems in LC for TDDS . . . . 245 oral formulations have disadvantages, such as the
17.4 Structure of Liquid Crystal side effect on the gastrointestinal tract and the
Dispersion . . . . . . . . . . . . . . . . . . . . . . . . . 245 first-pass effect, while injections cause pain by
17.5 Preparation of a Mixture of needle puncture and may cause infection; trans-
Mono-, Di-, and Triesters (1) and dermal drug delivery systems (TDDS) can avoid
Monoesters (2) Composed of these disadvantages. Interestingly, more TDDS
Erythritol and Phytanylacetic Acid . . . . 245
were developed at the beginning of this century
17.6 Preparation of Liquid Crystal than oral DDS. The outermost layer of the skin,
Dispersion . . . . . . . . . . . . . . . . . . . . . . . . . 246 the stratum corneum (SC), has a role as the pri-
17.7 Structure of Liquid Crystal mary barrier against water evaporation from the
Dispersions Observed by body and skin permeation of most drugs into the
Cryo-TEM Microscope . . . . . . . . . . . . . . 247
body. Thus, overcoming the SC barrier is impor-
17.8 Skin Penetration-Enhancing tant to advance the development of TDDS.
Effect of Liquid Crystals . . . . . . . . . . . . . 248
Chemical approaches, such as penetration
17.9 Drug Distribution in Skin After enhancers (Purdon et al. 2004), and physical
Topically Applied Non-lamella LCs . . . . 251 approaches, such as iontophoresis (Banga and
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251 Kasha 2008), phonophoresis (Ogura et al. 2008),
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 252 and electroporation (Tokudome and Sugibayashi
2004; Tokumoto et al. 2005), have been evaluated
to increase the skin permeation of several drugs.
Formulation approaches have also been investi-
gated to increase the skin permeation of drugs.
Liposomes and niosomes are examples for new
topical formulations having high penetration-
W.R. Kadhum • H. Todo • K. Sugibayashi (*) enhancing activity of the entrapped drugs
Faculty of Pharmaceutical Sciences, Josai University,
1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
(Abraham and Downing 1989; Fang et al. 2001;
e-mail: rkwesam@josai.ac.jp; ht-todo@josai.ac.jp; Kirjavainen et al. 1999); in general, however,
sugib@josai.ac.jp such nano-sized materials have poor stability.

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 243


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_17, © Springer-Verlag Berlin Heidelberg 2015
244 W.R. Kadhum et al.

Recently, solid lipid nanoparticles (Dingler and curvature energy are important parameters for the
Gohla 2002; Müller et al. 2000) and liquid crystal structure and nature of LCs (Silver 1985).
(LC) dispersions have been developed for topical An important feature of lyotropics is the self-
formulations, and the latter is highly bioadhesive assembly of the amphiphilic molecules as super-
(Geraghty et al. 1997) and physically stable. The molecular structures, which are the basic units of
LC structure is very similar to the intracellular these mesophases.
lipid structure in the SC (Norlén 2001). Very sta- The major LC structures are lamella, hexago-
ble LC dispersions made of monoolein/oleic acid nal, reverse hexagonal, bicontinuous cubic, and
(Garg et al. 2007; Gustafsson et al. 1996) and micellar cubic. Among them, hexagonal liquid
phytol (Barauskas and Landh 2003) have been crystals, reverse-hexagonal liquid crystals, and
applied to the skin and mucosa. LC nano- cubic liquid crystals are known as non-lamella
dispersions containing monoolein/oleic acid liquid crystals (non-lamella LC).
were used to modify the skin permeation of indo- Hexagonal liquid crystal and reverse-hexagonal
methacin (Esposito et al. 2005), cyclosporine liquid crystals are formed by some amphiphilic
(Lopes et al. 2006a, b), vitamin K (Lopes et al. molecules when they are mixed with water or
2007), and propranolol (Namdeo and Jain 2002). another polar solvent. In this phase, the amphiphilic
These LCs have also been investigated for skin molecules are aggregated into cylindrical structures
care products (Brinon et al. 1999; Esposito et al. of indefinite length and these cylindrical aggregates
2007) and cubic lipid structures have already are disposed on a hexagonal lattice, giving the phase
been used as lotions and creams in cosmetics long-range orientational order (Scheme 17.1).
(Yamaguchi et al. 2006).

17.2 Liquid Crystals (LC)

LCs are self-assembled mesophases, which are


intermediates between isotropic liquids and crys-
talline solids (Gin et al. 2008b). In LC phases,
long-range periodicity exists, although the mole-
cules exhibit a dynamical disorder at atomic dis-
Scheme 17.1 A schematic representation of hexagonal
tances, as is the case of liquids. Therefore, these phase
materials can also be considered ordered fluids
(Larsson 1989). On the other hand, lyotropic LCs Whereas cubic liquid crystals are formed
are materials that are composed from at least two when the concentration of micelles dispersed in a
molecules: an amphiphilic molecule and its sol- solvent is sufficiently high that they are forced to
vent. A hydrophilic solvent, such as water, pack into a structure having long-ranged posi-
hydrates the polar moieties of the amphiphiles tional order (Scheme 17.2).
via hydrogen bonding, while the flexible aliphatic
tails of the amphiphiles aggregate into fused
hydrophobic regions, based on van der Waals
interactions between aliphatic lipids and hydro-
phobic regions. In addition to morphologic
dependence on the chemical composition, lyo-
tropic LCs are also sensitive to external parame-
ters, such as temperature and pressure (Gin et al.
2008b; Larsson 1989; Yuli-Amar 2008).
Therefore, the shape of the amphiphilic mole-
cule, critical packing parameters, and interfacial Scheme 17.2 A schematic representation of cubic phase
17 Skin Permeation: Enhancing Ability of Liquid Crystal Formulations 245

On the other hand, liposomes as lamella LC Lamellar, hexagonal, bicontinuous cubic, and
have an entrapped, discontinuous aqueous phase discontinuous cubic phases are well known and
separated by bilayered lamellae from the contin- there are many research studies in that field. The
uous aqueous phase (Scheme 17.3). structure of self-assembled mesophases is
affected by amphiphile’s concentration. On
increasing the concentration of amphiphiles in
water, liposomes (lamellar), bicontinuous cubic
lamella phases (Q2), and hexagonal phases
(H2) are formed. There have been numerous
descriptions of the liquid crystalline phase
behavior (Hyde 1990). The dimensionless shape
parameter known as critical packing parameter
(CPP) has provided useful information regard-
ing the choice of type of rational design of
Scheme 17.3 A schematic representation of a liposome amphiphile-water phase behavior (e.g., micelle
structure is when CPP <1, lamellar phase is
Thus, the evaluation of lamella LC as a drug when CPP=1, and inverse cubic structure is
carrier might be difficult because entrapped drug when CPP> 1) (Israelachvili 1976). In this
could easily transfer from the internal phase to chapter, we focus on cubic lamella phase and
the dispersion medium. hexagonal phase that show skin permeation
enhancement effects.

17.3 Current Problems in LC


for TDDS 17.5 Preparation of a Mixture
of Mono-, Di-, and Triesters
Phytantriol and glyceryl monooleate (GMO) are (1) and Monoesters (2)
well-known compounds as non-lamella LC- Composed of Erythritol
forming lipoids (Phan et al. 2011). However, not and Phytanylacetic Acid
all of drugs can be entrapped in non-lamella LC
because hydrophilic/lipophilic balance (HLB) and Erythritol (2.50 kg) was dissolved in dimethyl
molecular size of drugs (Charlotte and Drummond sulfoxide (10.8 kg) at 100 °C under nitro-
2013) might affect the self-assemble of non- gen purging, before the addition of anhydrous
lamella LC-forming lipids. Furthermore, conven- calcium carbonate (37.8 g) at 80 °C. Methyl
tional non-lamella LC-forming lipids have so high phytanylacetate (5,9,13,17-tetramethyloctadec-
viscosity that it would be tough to handle them in anoate) (4.83 kg) was added dropwise to the
drug and cosmetic formulations and these lipids solution under reduced pressure over 2.5 h. The
can form non-lamella LC in a narrow temperature reaction mixture was refluxed under reduced
range. These are problems that need to be over- pressure overnight, while the methanol pro-
come for the development of transdermal formula- duced was gradually distilled. After cooling,
tions using non-lamella LCs. the mixture was neutralized by the addition of
formic acid (29 g) and concentrated in vacuo.
The residue (6.1 kg) was diluted with t-butyl
17.4 Structure of Liquid Crystal methyl ether (18.3 kg) and filtered to remove
Dispersion the remaining erythritol. The filtrate was diluted
again with t-butyl methyl ether (24 kg), washed
Figure 17.1 illustrates a scheme of the forma- twice with aqueous sodium bicarbonate, and
tion of lamella LC and non-lamella LC from concentrated in vacuo at 100 °C. The prod-
amphiphiles in water (Gin et al. 2008a). uct obtained (4.7 kg, mixture 1) consisted of
246 W.R. Kadhum et al.

Lamella liquid crystal Non-lamella liquid crystal (NLLC)

Bicontinuous Reverse hexagonal liquid


Discontinuous Reverse
Lamella liquid cubic liquid crystal crystal (H2), hexagonal crystal
cubic liquid micelle (Om)
Liposome crystal (La) (Q2), cubical crystal )
(cylindrical aggregate)
crystal

Hydrophilic group Lipophilic


Hydrophilic
Self-organizable
amphipathic lipids

Hydrophobic group

Fig. 17.1 Change of self-assembled structure of LC and groups (CPP value) and existence of salt or pH values in
non-lamella LC-forming lipids. The states of self- water phases. The arrow shows transitions of liquid crys-
assembled structure depend on balance of polar/nonpolar tals regarding lipophilicity of solvent

Table 17.1 Composition of Ingredients 30 mM calcein- Blank (%)


liquid crystal dispersions entrapped LC-A (%)
1-o-(5,9,13,17-tetramethyloctadecanoyl) 10.0 10.0
erythritol (crude, 1)
Sodium calcein 2.0 –
Pluronic® F127 (10 %) 10.0 10.0
Methyl p-hydroxybenzoate 0.1 0.1
Purified water 77.9 79.9
Total 100 100.0
Ingredients 30 mM calcein- Blank (%)
entrapped LC-B (%)
1-o-(5,9,13,17-tetramethyloctadecanoyl) 10.0 10.0
erythritol (pure, 2)
Calcein (sodium) 0.2 –
Pluronic® F127 (20 %) 10.0 10.0
Methyl p-hydroxybenzoate 0.1 0.1
Purified water 79.7 79.9
Total 100 100.0

monoesters (36 %), diesters (12 %), and tries- 17.6 Preparation of Liquid Crystal
ters (52 %) of erythritol and phytanylacetic acid Dispersion
(the ratio was determined by gas chromatogra-
phy analysis). Mixture 1 was purified by col- Table 17.1 shows the composition of liquid crys-
umn chromatography using silica gel (Wakogel tal A (LC-A) and liquid crystal B (LC-B) nano-
C-300, Wako Pure Chemicals Industries, Ltd., dispersions. Ten grams of crude ester 1 or pure
Osaka, Japan) to afford monoesters 2 of 1-O- ester 2 was used to prepare LC-A and LC-B,
and 2-O-phytanylacetyl-erythritol. respectively. In this step, LC-A and LC-B were
17 Skin Permeation: Enhancing Ability of Liquid Crystal Formulations 247

semisolid and were nano-dispersed using a high- chapter. The crude and pure esters were used to
pressure emulsifier (NM2-L200AR, Yoshida prepare non-lamella LC-A and non-lamella
Kikai Co., Ltd, Nagoya, Japan) or an ultrasonic LC-B, respectively. Non-lamella LC-A- and
homogenizer (USP-50, Shimadzu Corp., Kyoto, non-lamella LC-B-forming lipids were dis-
Japan), respectively, in aqueous solution (90.0 g) persed in aqueous solution containing sodium
containing sodium calcein, Pluronic® F127 and calcein to prepare non-lamella LCs. Sodium
methyl p-hydroxybenzoate. Calcein concentra- calcein (M.W.; 623) is a good indicator used as
tions in LC-A and LC-B were different (see hydrophilic fluorescent marker for skin perme-
Table 17.1). ation experiment. Thus, penetration-enhancing
ability of non-lamella LC was investigated by
evaluating the permeation of calcein through
17.7 Structure of Liquid Crystal skin.
Dispersions Observed by Figure 17.3 shows cryo-transmission electron
Cryo-TEM Microscope microscope (TEM) micrographs of non-lamella
LC-A and non-lamella LC-B. Similar to the cryo-
Skin permeation enhancement effects of non- TEM micrographs of cubic non-lamella LC pre-
lamella LCs, which consist of crude or pure pared by monoolein and oleic acid (Garg et al.
non-lamella LC-forming lipids, such as 2007; Gustafsson et al. 1996), non-lamella LC
1-O-(5,9,13,17-tetramethyloctadecanoyl) structures were observed in non-lamella LC-A
erythritol (Fig. 17.2), are discussed in this and non-lamella LC-B (in white circles in

OH

O
HO

OH O

Fig. 17.2 Chemical structure of 1-o-(5, 9, 13, 17-tetramethyloctadecanoyl) erythritol

a b

Fig. 17.3 Cryo-TEM microscope images of liquid crystal were found in both (a, b) (see circles in images). Each
dispersions. Images (a, b) are non-lamella LC-A and non- white bar indicates 25 nm length
lamella LC-B, respectively. Non-lamella LC structures
248 W.R. Kadhum et al.

a-1 a-2

b-1 b-2

Fig. 17.4 Electron diffraction pattern of liquid crystal LC, having 4.6 nm periodic structure. (b-2) illustrates
dispersions. (a-1) and (a-2) and (b-1) and (b-2) show the cubic LC, having two periodic structures of 6.0 and
same images of LC-A and LC-B, respectively, without 9.0 nm cycles. White bars indicate 5 or 10 nm length
and with auxiliary lines and marks. (a-2) shows hexagonal

Fig. 17.3). Figure 17.4 shows electronic 17.8 Skin Penetration-Enhancing


diffraction patterns of non-lamella LC deter- Effect of Liquid Crystals
mined by cryo-TEM photographs. It was found
from these diffraction patterns that non-lamella The penetration-enhancing effectiveness of non-
LC-A was a hexagonal non-lamella LC, having lamella LC in topical formulations was evaluated
4.6 nm periodic structure (A-2 dashed lines in by measuring permeation of a mal-absorbable
Fig. 17.4), and non-lamella LC-B was a cubic compound, calcein, through excised hairless rat
non-lamella LC, having two periodic structures skin and through the three-dimensional cultured
of 6.0 and 9.0 nm (B-2 dashed lines in Fig. 17.4). human skin model (LSE-high).
17 Skin Permeation: Enhancing Ability of Liquid Crystal Formulations 249

Intact skin Stripped skin


7 7,000

6 6,000
Calcein permeated (nmol/cm2)

5 5,000

4 4,000

* *
3 3,000

2 2,000

1 1,000

0 0 0
0 1 2 3 4 5 6 7 8 0 1 2 3 4 5 6
Time (h) Time (h)

Fig. 17.5 Effect of non-lamella LC-A formulation on the Asterisks mean significant difference between calcein
time course of the cumulative amount of calcein that per- entrapped in non-lamella LC-A and free calcein or free
meated intact and stripped hairless rat skin. Symbols are calcein plus non-lamella LC-A (p <0.05). Each point rep-
as follows: ◆ free calcein, ○ free calcein plus non- resents the mean ± S.E. of at least three to seven
lamella LC-A, ● calcein entrapped in non-lamella LC-A. experiments

Figure 17.5 shows the time course of calcein could improve the calcein distribution into the
permeation through excised hairless rat skin from skin by providing a high affinity to intercellular
the reverse-hexagonal liquid crystal (non-lamella lipid structure in the skin (stratum corneum).
LC-A) formulations. In both intact and stripped Next, the time course of the skin permeation of
skin, the skin permeations of calcein from calcein calcein from the cubic liquid crystal dispersion
entrapped in non-lamella LC-A formulation pro- (non-lamella LC-B) was evaluated; the results
vided a 3 and 10 times higher, respectively, than are shown in Fig. 17.6. Non-lamella LC-B as
the permeation of calcein from free calcein solu- well as non-lamella LC-A enhanced the perme-
tion. In addition, the mixture of blank non-lamella ation of calcein through intact hairless rat skin;
LC-A dispersion and free calcein showed similar Table 17.3 summarizes the permeation parame-
skin permeation to that of free calcein solution. ters for non-lamella LC-B. Increased partition of
Thus, the blank non-lamella LC formulation calcein was observed by non-lamella LC-B, as by
itself did not show any penetration-enhancing non-lamella LC-A. In contrast, no increase in
effect. Permeation parameters were determined drug partition was observed in stripped skin from
from the time course of the cumulative amount of non-lamella LC-B or from non-lamella
the permeated drug. The calculated parameters LC-A. Non-lamella LC-A has a high affinity for
are shown in Table 17.2. Partition coefficient, K, the stratum corneum as well as the viable epider-
of calcein was markedly increased by application mis, whereas non-lamella LC-B shows high
of the calcein entrapped in non-lamella LC-A affinity for the stratum corneum, but not for the
formulation, suggesting that non-lamella LC-A viable epidermis.
250 W.R. Kadhum et al.

Table 17.2 Partition Calcein entrapped in Free calcein and


coefficient (K), diffusion 30 mM calcein non-lamella LC-A non-lamella LC-A
coefficient (D), and
Intact skin
permeability coefficient
(P) of calcein through K 0.10 ± 0.02 0.22 ± 0.04 0.09 ± 0.03
hairless rat skin after D (×10−11 cm2/s) 6.05 ± 0.52 7.54 ± 0.18 6.75 ± 0.68
application of different P (×10−9 cm/s) 2.91 ± 0.55 8.32 ± 1.33 2.80 ± 0.91
formulations of non- Stripped skin
lamella LC-A K 0.52 ± 0.13 2.51 ± 0.87 1.19 ± 0.20
D (×10−7 cm2/s) 1.04 ± 0.07 3.22 ± 1.14 0.89 ± 0.07
P (×10−7 cm/s) 9.41 ± 2.86 110 ± 12.5 18.9 ± 4.51

Intact skin Stripped skin


2.5 300

250
2
Calcein permeated (nmol/cm2)

200
1.5
* *
150

1
100

0.5
50

0 0
0 1 2 3 4 5 6 7 8 0 1 2 3 4 5 6
Time (h) Time (h)

Fig. 17.6 Effect of LC-B formulation on the time course LC-B. Asterisks mean significant difference between cal-
of the cumulative amount of calcein that permeated cein entrapped in LC-A and free calcein or free calcein
through intact and stripped hairless rat skin. Symbols are plus non-lamella LC-B (p <0.05). Each point represents
as follows: ■ free calcein, □ free calcein plus non- the mean ± S.E. of at least 3–12 experiments
lamella LC-B, ▲ calcein entrapped in non-lamella

Table 17.3 Partition coefficient (K), diffusion coefficient (D), and permeability coefficient (P) of calcein through hair-
less rat skin after application of different formulations of non-lamella LC-B
3 mM calcein in 2 % Free calcein and
3 mM calcein Pluronic sol. non-lamella LC-B
Intact skin
K 0.53 ± 0.12 0.59 ± 0.11 0.95 ± 0.10
D (×10−11 cm2/s) 5.95 ± 0.78 6.47 ± 0.74 6.94 ± 0. 39
P (×10−8 cm/s) 1.56 ± 0.38 1.84 ± 0.13 3.23 ± 0.32
Stripped skin
K 2.18 ± 0.08 2.44 ± 0.39 2.53 ± 0.26
D (×10−8 cm2/s) 9.04 ± 1.33 6.92 ± 2.83 10.7 ± 1.45
P (×10−6 cm/s) 3.46 ± 0.65 2.95 ± 0.55 4.48 ± 0.32
17 Skin Permeation: Enhancing Ability of Liquid Crystal Formulations 251

a c e

b d f

Fig. 17.7 Histological observation of LSE-high, 8 h after and (e, f) for physical mixture of free calcein and non-
application of non-lamella LC-A dispersion with lamella LC-A dispersion. Images (a, c, e) show light
entrapped calcein. Images (a, b) are for free calcein, (c, d) micrograph images and (b, d, f) show corresponding fluo-
for calcein entrapped in non-lamella LC-A dispersion, rescent images. Bars indicate 100 mm (vertical slice)

17.9 Drug Distribution in Skin and free calcein plus non-lamella LC-A dispersion
After Topically Applied (Fig. 17.7f). In contrast, a much higher fluorescent
Non-lamella LCs level was found for the calcein entrapped in non-
lamella LC-A (Fig. 17.7d). These results were also
Similar permeation experiments were carried out obtained by the skin permeation study using non-
using a three-dimensional cultured human skin lamella LC-A dispersion with entrapped or free
model (LSE-high). Since no appendages, such as calcein and the calcein solution (Fig. 17.5).
hair follicles and sweat ducts, are present in LSE-
high, the penetration-enhancing effect by non- Conclusion
lamella LC formulations must be due to the It was found from cryo-TEM observation and
enhanced drug penetration through the stratum electron diffraction pattern analysis that these
corneum barrier, i.e., through the intercellular LC-A and LC-B nano-dispersions have
route. Figures 17.7a–f show the cross section of reverse-hexagonal LC and cubic LC disper-
LSE-high, 8 h after application of free calcein and sions, respectively. These LC dispersions were
calcein entrapped in non-lamella LC-A and simul- very stable, since no aggregation (diameter
taneous application of free calcein and non-lamella growth) was observed. The entrapping ratio of
LC-A, respectively. Figures 17.7a, c, and e show a hydrophilic model compound, calcein, in
hematoxylin and eosin (HE)-stained micrographs LC-B was higher than in LC-A, suggesting
and Fig. 17.7b, d, and f show corresponding fluo- that cubic LC has great entrapping potency for
rescent micrographs. Similar fluorescent level was hydrophilic materials. Other drugs with differ-
observed for groups of free calcein (Fig. 17.7b) ent lipophilicity and molecular weights must
252 W.R. Kadhum et al.

be determined in the future. The present skin Garg G, Saraf S, Saraf S (2007) Cubosomes, an overview.
permeation experiments showed that LC-A Biol Pharm Bull 30:350–353
Gustafsson J, Ljusberg-Wahren H, Almgren M, Larsson K
and LC-B nano-dispersions increased the skin (1996) Cubic lipid-water phase dispersed into submi-
permeation of the entrapped drug, probably cron particles. Langumuir 12:4611–4613
due to increased partition of the LC in the skin Hyde ST (1990) Curvature and the global structure of
barrier. Thus, the presently prepared LC for- interfaces in surfactant-water systems. J Phys Colloq
51(C7):209–228
mulations can be utilized as new topical for- Israelachvili JN, Mitchel DJ, Ninham BW (1976) Theory
mulations for therapeutic drugs and cosmetic of self-assembly of hydrocarbons amphiphiles into
ingredients, especially for hydrophilic com- micelles and bilayers. J Chem Soc Faraday Trans
pounds. The skin content of the entrapped 2(72):1525–1568
Kasha PC, Banga AK (2008) A review of patent literature
drugs and detail mechanistic analysis must be for iontophoretic delivery and devices. Recent Pat
determined in the near future. Drug Deliv Formul 2:41–50
Kirjavainen M, Urtti A, Valjakka-Koskelab R, Kiesvaara J,
Mönkkönena J (1999) Liposome–skin interactions and
their effects on the skin permeation of drugs. Eur
References J Pharm Sci 7:279–286
Larsson K (1989) Cubic lipid-water phases: structure and
Abraham W, Downing DT (1989) Preparation of model biomembrane aspects. J Phys Chem 93:7304–7314
membranes for skin permeability studies using stra- Lopes LB, Ferreira DA, de Paula D, Garcia MTJ,
tum corneum lipids. J Invest Dermatol 93:809–813 Thomazini JA, Fantini MCA, Bentley MVLB (2006a)
Barauskas J, Landh T (2003) Phase behavior of phytant- Reverse hexagonal phase nanodispersion of monoolein
riol/water system. Langmuir 19:9562–9565 and oleic acid for topical delivery of peptides: in vitro
Brinon L, Geiger S, Alard V, Doucet J, Tranchant JF, and in vivo skin penetration of cyclosporin A. Pharm
Couarraze G (1999) Percutaneous absorption of sun- Res 23:1332–1342
screens from liquid crystalline phases. J Control Lopes LB, Lopes JLC, Oliveira DCR, Thomazini JA,
Release 60:67–76 Garcia MTJ, Fantini MCA, Collett JH, Bentley MVLB
Conn CE, Drummond CJ (2013) Nanostructured bicon- (2006b) Liquid crystalline phases of monoolein and
tinuous cubic lipid self-assembly materials as matrices water for topical delivery of cyclosporin A, character-
for protein encapsulation. Soft Matter. doi:10.1039/ ization and study of in vitro and in vivo delivery. Eur J
C3SM27743G Pharm Biopharm 63:146–155
Dingler A, Gohla S (2002) Production of solid lipid Lopes LB, Speretta FFF, Bentley MVLB (2007)
nanoparticles (SLN): scaling up feasibilities. J Enhancement of skin penetration of vitamin K using
Microencapsul 19:11–18 monoolein-based liquid crystalline systems. Eur J
Esposito E, Cortesi R, Drechsler M, Paccamiccio L, Pharm Sci 32:209–215
Mariani P, Contado C, Stellin E, Menegatti E, Bonina Müller RH, Mäder K, Gohla S (2000) Solid lipid
F, Puglia C (2005) Cubosome dispersions as delivery nanoparticles (SLN) for controlled delivery – a review
systems for percutaneous administration of indometh- of the state of the art. Eur J Pharm Biopharm
acin. Pharm Res 22:2163–2173 50:161–177
Esposito E, Drechsler M, Mariani P, Sivieri E, Bozzini R, Namdeo A, Jain NK (2002) Liquid crystalline pharmaco-
Montesi L, Menegatti E, Cortesi R (2007) Nanosystem gel based enhanced transdermal delivery of proprano-
for skin hydration: a comparative study. Int J Cosmet lol hydrochloride. J Control Release 82:223–236
Sci 29:39–47 Norlén L (2001) Skin barrier formation, the membrane
Fang J, Hong C, Chiu W, Wang Y (2001) Effect of lipo- folding model. J Invest Dermatol 117:823–829
somes and niosomes on skin permeation of enoxacin. Ogura M, Paliwal S, Mitragotri S (2008) Low-frequency
Int J Pharm 219:61–72 sonophoresis: current status and future prospects. Adv
Geraghty PB, Attwood D, Collett JH, Sharma H, Dandiker Drug Deliv Rev 60:1218–1223
Y (1997) An investigation of the parameters Phan S, Fong WK, Kirby N, Hanley T, Boyd BJ (2011)
influencing the bioadhesive properties of Myverol Evaluating the link between self-assembled meso-
18–99/water gels. Biomaterials 18:63–67 phase structure and drug release. Int J Pharm
Gin DL, Pecinovsky CS, Bara JE, Kerr RL (2008a) 421:176–182
Functional lyotropic liquid crystal materials. Liquid Purdon CH, Azzi CG, Zhang J, Smith EW, Maibach HI
crystalline functional assemblies and their supramo- (2004) Penetration enhancement of transdermal deliv-
lecular structures and bonding. 128:181–222 ery–current permutations and limitations. Crit Rev
Gin DL, Pecinovsky CS, Bara JE, Kerr L (2008b) Ther Drug Carrier Syst 21:97–132
Functional lyotropic liquid crystal materials. Struct Silver B (1985) The physical chemistry of membranes.
Bond 128:181 Solomon Press, Winchester
17 Skin Permeation: Enhancing Ability of Liquid Crystal Formulations 253

Tokudome Y, Sugibayashi K (2004) Mechanism of the Yamaguchi Y, Nakamura N, Nagasawa T, Kitagawa A,


synergic effects of calcium chloride and electropora- Matsumoto K, Soma Y, Matsuda T, Mizoguchi M,
tion on the in vitro enhanced skin permeation of drugs. Igarashi R (2006) Enhanced skin regeneration by
J Control Release 95:267–274 nanoegg formulation of all-trans retinoic acid.
Tokumoto S, Mori K, Higo N, Sugibayashi K (2005) Effect Pharmazie 61:117–121
of electroporation on the electroosmosis across hairless Yuli-Amar I (2008) Ph.D. Dissertation, The Hebrew
mouse skin in vitro. J Control Release 105:296–304 University of Jerusalem
Nanoemulsions in Dermal
Drug Delivery 18
Victoria Klang, Julia C. Schwarz,
and Claudia Valenta

Contents 18.1 Introduction


18.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . 255
18.1.1 Definition and Characterisation 18.1.1 Definition
of Nanoemulsions . . . . . . . . . . . . . . . . . . . 255 and Characterisation
18.1.2 Dermal Application: Advantages of Nanoemulsions
and Limitations . . . . . . . . . . . . . . . . . . . . . 258
18.2 Penetration Enhancement Strategies . . . 259 Classical emulsions are dispersions of a liquid
18.2.1 Role of Droplet Size . . . . . . . . . . . . . . . . . 259
18.2.2 Role of Droplet Surface Charge. . . . . . . . . 262
phase in another immiscible liquid phase. They
18.2.3 Role of Compounds . . . . . . . . . . . . . . . . . . 262 have been employed for the dermal application of
active substances for a long time since they can
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 264
easily be produced through mild energy input and
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 264 can be kinetically stabilised with the help of suit-
able surfactants.
During the last decades, technological and
methodological progress has led to the develop-
ment of oil-in-water emulsions with increasingly
small droplet sizes. The first systems known as
V. Klang (*) • C. Valenta (*) submicron emulsions emerged during the 1950s
Department of Pharmaceutical Technology and were originally employed for parenteral nutri-
and Biopharmaceutics, University of Vienna, tion (Wabel 1998; Calder et al. 2010). They were
Althanstraße 14, Vienna 1090, Austria
composed of natural oils such as soybean oil dis-
Research Platform ‘Characterisation of Drug persed within an aqueous phase with the help of
Delivery Systems on Skin and Investigation
natural lecithin mixtures. The average droplet
of Involved Mechanisms’, University of Vienna,
Althanstraße 14, Vienna 1090, Austria sizes of these systems produced through high-
e-mail: victoria.klang.vie@gmail.com; energy emulsification usually ranged between 100
claudia.valenta@univie.ac.at and 500 nm. During the next decades, different
J.C. Schwarz high-energy production methods were estab-
Research Platform ‘Characterisation of Drug lished, including homogenisation with high-pres-
Delivery Systems on Skin and Investigation
sure homogenisers or microfluidisers as well as
of Involved Mechanisms’, University of Vienna,
Althanstraße 14, Vienna 1090, Austria ultrasonication. Depending on the production
e-mail: julia.schwarz@univie.ac.at technique and the employed compounds, droplet

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 255


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_18, © Springer-Verlag Berlin Heidelberg 2015
256 V. Klang et al.

sizes around 100 nm could be reached, and the the term nanoemulsion is widely – albeit inaccu-
prevailing terminology changed from submicron rately – employed as a synonym for the term
emulsions to nanoemulsions. submicron emulsion today. We would like to
Nanoemulsions of the oil-in-water type have point out this slight discrepancy from nanoscale
been adapted for various routes of drug delivery, conventions: nanoemulsions do not necessarily
including sensitive routes such as intravenous, exhibit droplet sizes below 100 nm.
ocular or dermal application (Benita and Levy To further complicate the terminology issue,
1993; Yang and Benita 2000; Klang and Valenta increasing numbers of publications about ‘nano-
2011) while water-in-oil nanoemulsions have emulsions’ are in fact dealing with microemul-
been investigated for technical applications as sions. As well known, microemulsions are
well as for theoretical surfactant studies (Chiesa thermodynamically stable isotropic systems that
et al. 2008; Porras et al. 2004). In addition to clas- contain large amounts of surfactants and solvents
sical lecithin-based nanoemulsions produced by and form spontaneously. Microemulsions may
high-energy emulsification, new low-energy pro- exhibit complex internal structures including
duction methods have been developed, such as the bicontinuous sponge phases or droplet-shaped
phase inversion temperature method or the solvent phases. Nevertheless, they bear no similarities to
evaporation method (Fernandez et al. 2004; Tadros real emulsions such as nanoemulsions; the well-
et al. 2004; Sole et al. 2010). These methods established term microemulsion is of historical
require the use of specific surfactants or additional nature. Several groups have taken up the task of
solvents to produce nano-sized oil droplets from a clarifying these terminology issues that render
microemulsion matrix due to changes in the spon- literature research increasingly difficult (Klang
taneous surfactant curvature. Conveniently, these and Valenta 2011; Mason et al. 2006; Anton and
methods can be conducted rapidly without the Vandamme 2011; McClements 2012).
need for specific equipment but may involve To avoid confusion, we adhere to the nowa-
strong heating of the pre-emulsion system. days established term nanoemulsion for emul-
Discussions about the exact nature of the pro- sions with droplets in the lower submicron range.
duced systems are still ongoing. Important param- The majority of publications are dealing with oil-
eters such as mean droplet size and droplet size in-water nanoemulsions since they are more rel-
distribution again depend strongly on the evant for most applications than water-in-oil
employed compounds and exact processing condi- systems. Recent developments include adapta-
tions. Progress in the field of nanoemulsion pro- tions such as multiple nanoemulsions for the
duction is constantly being reported for both delivery of drugs of different logP values (Anton
high- and low-energy emulsification methods et al. 2010; Schwarz et al. 2012).
(Cortés-Muñoz et al. 2009; Anton et al. 2007). With decreasing droplet size, the general prop-
In summary, the term nanoemulsion is erties of nanoemulsions start to differ signifi-
employed to describe conventional emulsions cantly from those of conventional emulsions.
with droplet sizes in the lower submicron range. Although nanoemulsions exhibit basic emulsion
It should be noted that droplet sizes of classical properties such as an inherent metastability, they
nanoemulsions, i.e. nanoemulsions stabilised by differ from macroscale emulsions in regard to
lecithin-type surfactants that are produced optical appearance, physicochemical properties
through high-energy emulsification, rarely reach and prevailing destabilisation processes (Klang
mean droplet sizes below 100 nm. The general and Valenta 2011; Mason et al. 2006).
recommendation of accepted guidelines today is Nanoemulsions may exhibit a transparent to
that the prefix nano is to be employed for systems translucent appearance if the droplet size is small
with size ranges between 1 and 100 nm (Mason enough while emulsions are generally creamy
et al. 2006). Accordingly, the term nanoemulsion white due to multiple scattering of light. In addi-
would only be appropriate for submicron emul- tion, nanoemulsions are of a fluid nature and
sions with droplet sizes below 100 nm. However, exhibit near-Newtonian flow behaviour, while
18 Nanoemulsions in Dermal Drug Delivery 257

classical emulsions are usually of higher viscos- with spacious surface-active agents are success-
ity and shear-thinning flow behaviour. Most ful strategies to further optimise nanoemulsion
importantly, nanoemulsions may exhibit signifi- stability (van Nieuwenhuyzen and Szuhaj 1998).
cantly improved physical stability during storage Characterisation of the above-mentioned prop-
when compared to conventional emulsions. erties of nanoemulsions can usually be performed
Classical emulsions are sooner or later destabi- by common analytical techniques for aqueous col-
lised by gravity-induced alterations, coalescence loidal systems. Dynamic light scattering as a popu-
of oil droplets and eventual phase separation. lation-based technique of droplet size
Nanoemulsions are hardly affected by coales- characterisation is frequently employed, ideally in
cence due to their small droplet size. If the dis- combination with static laser diffraction to exclude
persed droplets have a relatively high solubility the presence of individual large droplets. In addi-
within the continuous phase, Ostwald ripening tion, microscopic techniques such as cryo-
may occur: individual molecules of the dispersed transmission electron microscopy are recommended
phase diffuse from smaller to larger droplets due to obtain information about the exact morphology
to differences in Laplace pressure, thus leading to of the system including droplet shape and the pres-
a continuous increase in droplet size. By consid- ence of vesicular structures (Klang et al. 2012a).
erate choice of excipients, however, this Examples of different techniques of analysis
destabilisation mechanism occurring in nano- including electron microscopy and atomic force
emulsions may be largely eliminated (Wabel microscopy are presented in Fig. 18.1.
1998; Mason et al. 2006). Enhanced electrochem- Apart from analysing droplet size distributions,
ical stabilisation through modification of the the droplet surface charge as an indicator for elec-
droplet surface charge and sterical stabilisation trochemical stabilisation can be determined by

a b

c d

Fig. 18.1 (a) Optical appearance and cryo-TEM image of a (2001), p. 400, with permission of Elsevier). (c) Freeze-
translucent nanoemulsion (Reprinted from Sonneville- fracture TEM micrograph of an amphotericin B nanoemul-
Aubrun et al. (2004), p. 146, with permission of Elsevier). sion (Reprinted from Benita and Levy (1993), p. 1078, with
(b) Cryo-TEM image of nano-sized oil droplets (homoge- permission of Wiley). (d) Nanoemulsion morphology visu-
neously filled circles) and vesicles (unfilled circles); the alised by atomic force microscopy (Reprinted from Marxer
scale bar represents 200 nm (Reprinted from Norden et al. et al. (2011), p. 433, with permission of Elsevier)
258 V. Klang et al.

laser Doppler electrophoresis (Mueller 1996). Classical nanoemulsions prepared by high-


Stability investigations are usually performed by pressure homogenisation are usually stabilised
monitoring parameters such as the mean droplet by skin-friendly surfactants such as lecithins and
size, the polydispersity index, droplet surface do not require synthetic surfactants. Thus, skin
charge and pH value of the nanoemulsions in ques- lipids are not washed out during cleaning.
tion. In our experience, the physical stability of Lipophilic drugs can be incorporated into the oil
classical nanoemulsions produced by high-pres- phase according to their solubility. The release of
sure homogenisation can be expected to be around the drugs from the system is generally acknowl-
2 years or longer at slightly refrigerated storage. edged to be quite rapid; however, attempts
However, the chemical stability of individual com- towards retarded or controlled release as well as
pounds has to be taken into account as well. Oil, site-specific targeting have been reported as well
surfactants and incorporated actives may be sub- (Yang and Benita 2000; Eskandar et al. 2009).
jected to chemical degradation by oxidation or Nanoemulsions support the penetration of incor-
hydrolysis, potentially leading to an unpleasant porated actives into the skin and may thus pro-
optical and olfactory appearance despite unchanged mote their accumulation in the skin. In addition,
droplet size (Wabel 1998; Baker and Naguib 2005). the cosmetic effect of the basic vehicles is of fur-
Optical monitoring of the formulations, assess- ther interest. In recent approaches, the possibility
ment of drug stability and monitoring of both zeta to incorporate hydrophilic drugs into nanoemul-
potential and pH are useful tools in detecting such sion systems is being investigated with the aim of
changes (Klang et al. 2011a). promoting the incorporation efficiency, stability
and penetration of water-soluble actives (Anton
et al. 2010; Schwarz et al. 2012).
18.1.2 Dermal Application: Retarded drug delivery from nanoemulsions is
Advantages and Limitations difficult to achieve because of the low viscosity of
the systems. When compared to aqueous solutions
Most marketed nanoemulsion products can be or dispersions, retarded drug release from nano-
found in the realms of intravenous nutrition and emulsions may of course be observed. In general,
drug delivery as well as in dermal drug delivery and a retarded release may be achieved for very lipo-
cosmetics. In terms of dermal drug delivery, nano- philic drugs with high affinity to the oil phase.
emulsions offer distinct advantages: high physical From another viewpoint, the low viscosity of
stability compared to conventional emulsions, high nanoemulsions is an advantageous feature: it ren-
skin friendliness due to the low amount and the mild ders them attractive systems for development of
nature of the employed surfactants and last but not aerosol sprays such as sunscreens that show no
least the ease of preparation and scale-up. They phase separation during storage. If a higher vis-
avoid the limitations of other colloidal drug delivery cosity is required, modification of the nanoemul-
systems, such as the limited drug loading and stabil- sion is possible by incorporation of gelling
ity issues of liposomes and the potentially irritating agents. Successful reports on the development of
compounds required for the production of nanopar- thickened nanoemulsions can be found in the lit-
ticles or microemulsions. Depending on composi- erature (Alves et al. 2005; Mou et al. 2008).
tion and the nature of the employed drug, Regarding transdermal penetration, there is a
nanoemulsions may achieve higher rates of skin certain amount of recent literature proposing
penetration and drug accumulation within the skin nanoemulsions for this task. However, a closer
than lipid nanoparticles (Calderilla-Fajardo et al. look at these articles reveals that the systems in
2006). In a recent study investigating the dermal question are exclusively dealing with thermody-
delivery of lutein, a more rapid release in case of namically stable microemulsion phases.
nanoemulsions was found, which achieved higher Nanoemulsions in the sense of skin-friendly
skin permeation rates than nano-structured lipid car- submicron-sized emulsion systems are not spe-
riers and solid lipid nanoparticles (Mitri et al. 2011). cifically designed for the purpose of delivering
18 Nanoemulsions in Dermal Drug Delivery 259

drugs transdermally. To allow for the passage of Regarding the mechanism of nanoemulsion
intact nanovectors across pores of the skin, much penetration, it can be summarised that no evi-
smaller and deformable carriers are required since dence exists that conventional colloidal systems
the skin is a strong and complex barrier (Cevc and such as nanoemulsions may penetrate into the
Vierl 2010). Small drug amounts may of course skin as intact structures. It may thus be assumed
be found in deeper skin layers after application of that their skin penetration is related to penetra-
a nanoemulsion system due to mechanisms such tion of the system into shunt pathways such as
as penetration across hair follicles (Lademann hair follicles, accumulation of the system
et al. 2001) or a general penetration enhancement between corneocyte clusters or in furrows to
caused by the involved excipients, in particular interact with skin lipids or merging of the system
the involved lecithin phospholipids (Yu et al. into extended lipidic structures on the skin sur-
2009). In general, however, nanoemulsions are face. In this context, penetration enhancement of
not employed for transdermal drug delivery, but drugs may be caused by the occlusion of the skin
rather for targeting the outermost skin layers, for surface (Eskandar et al. 2009; Zhou et al. 2010)
instance, in case of fungal infections or inflamma- as well as the interaction of nanoemulsion excipi-
tion. The versatile nature of nanoemulsions is ents such as phospholipids with skin compounds.
highlighted by another recent study by Spagnul The systems may alter the thermodynamic activ-
et al., who designed a calixarene nanoemulsion ity of incorporated drugs on the skin surface as
aimed at chelating uranium molecules at the skin well (Cevc and Vierl 2010). According to their
surface (Spagnul et al. 2011). The patented for- composition, nanoemulsions may thus be
mulation is a promising approach towards treat- employed to enhance the penetration of incorpo-
ing cutaneous uranium contamination if the rated actives into the skin. Different aspects may
formulation is applied quickly. This study shows influence the success of this strategy, such as the
that nanoemulsions exhibit advantageous proper- exact formulation morphology, the droplet sur-
ties for different applications on skin and can be face charge and the nature of the involved com-
tailor-made according to the envisioned task. pounds. The role of these factors will be discussed
In another recent study, a lecithin-based nano- in the following sections.
emulsion containing 5-aminolevulinic acid
(5-ALA) was developed for dermal application in
photodynamic therapy (Maisch et al. 2010). 18.2 Penetration Enhancement
Significantly deeper skin penetration was found Strategies
for the nanoemulsion when compared to a con-
ventional creamy emulsion (Fig. 18.2). As 18.2.1 Role of Droplet Size
intended in localised therapy, no penetration
beyond the basal cell membrane was observed. The large surface area and low surface tension of
The increased drug transport into the epidermis small nanoemulsion oil droplets has been
may be ascribed to a stabilising effect of the described to improve skin interaction and thus
nanoemulsion on 5-ALA, thus preventing dimer- dermal drug delivery (Benita 1999; Klang et al.
isation. Furthermore, the nanoemulsion formula- 1998). An enhanced local therapeutic effect of
tion seemed to support the cellular uptake of incorporated drugs due to prolonged residence
5-ALA. Recently, the pivotal phase III studies time in the uppermost skin layers may thus be
were completed for the developed system. The expected. One of the earliest studies dealing with
results support the claims of improved stability the effect of nanoscale droplet size in topical
and skin penetration when tested against a con- emulsions was conducted by Friedman et al.
ventional cream and placebo (Dirschka et al. (1995). Both steroidal and nonsteroidal antiin-
2012). The corresponding product has success- flammatory drugs including betamethasone val-
fully been marketed (Ameluz®, Biofrontera erate and dipropionate, naproxen, diclofenac,
Pharma GmbH). indomethacin and piroxicam were incorporated
260 V. Klang et al.

Fig. 18.2 Distribution of protoporphyrin IX after appli- skin with the respective formulation. The blue line repre-
cation of a nanoemulsion-based formulation containing sents the basal membrane, i.e. the border between epider-
10 % 5-ALA hydrochloride (left column) or a commercial mis and dermis. Samples were removed after 3, 8 and
cream containing 16 % w/w aminolevulinate methyl ester 12 h, and the induction of protoporphyrin IX was deter-
hydrochloride (right column). Scale bars represent mined (coloured areas) (Image reprinted from Maisch
100 μm. The images were taken after incubation of the et al. (2010), p. e304, with permission of Wiley)
18 Nanoemulsions in Dermal Drug Delivery 261

into nanoemulsions and regular creamy emul- microviscosity and thus the drug transport were
sions of identical composition. The antiinflam- found to be comparable to that of corresponding
matory efficacy of the systems was compared fluid nano-sized systems (Ullrich et al. 2008). For
against each other and to established marketed the developed sucrose stearate emulsions, droplet
formulations by using the carrageenan-induced sizes around 120–150 nm did not significantly
paw edema rat model. Significantly improved affect the skin penetration of incorporated drugs
antiinflammatory efficacy was found for the when compared to corresponding emulsions with
nanoemulsion systems, especially in case of droplet sizes in the micrometre range. Further
indomethacin, diclofenac and betamethasone comparative studies in this direction using nano-
esters. Noticeable systemic activity of antiinflam- emulsions with droplet sizes below 100 nm
matory drugs formulated in nanoemulsions was would be of interest to gain further information
observed as well. Preliminary in vivo studies on on this matter.
human volunteers showed good acceptability and In another recent study, lecithin nanoemul-
comparable properties to marketed products. The sions for topical delivery were developed using
authors hypothesised that lipid disruption within snake oil, soybean lecithin, glycerol and purified
the stratum corneum caused by the nanoemulsion water (Zhou et al. 2010). When applied in an
phospholipids, the formation of gaps and the O/W cream, the nano-sized droplets were shown
increased swelling of the skin may favour the to enhance skin hydration and skin penetration of
penetration of lipid droplets with diameters the incorporated model dye nile red into the der-
below 100 nm. Under these circumstances, shunt mis when compared to a control O/W cream. The
pathways such as hair follicles or sebaceous authors observed increased skin adhesion of the
channels may lead to enhanced penetration as nanoemulsion formulation to the skin surface and
well. It should be kept in mind, however, that subsequent formation of an occlusive film, thus
these conditions may be particularly encountered increasing skin hydration and consequently skin
in the employed edema model. In a more recent penetration of the model dye. The increased skin
study, Kotyla and co-workers compared the bio- penetration can therefore be ascribed to physical
availability of tocopherol from a nanoemulsion effects caused by the presence of small oil drop-
and a micrometre-sized emulsion of the same lets. In addition, the authors assumed that an
composition. After dermal application of the increased partitioning of lecithin molecules into
nanoemulsion in vivo on golden hamster skin, a the stratum corneum might be responsible for
2.5-fold increase in plasma tocopherol levels was subsequent changes in the barrier properties, i.e.
determined (Kotyla et al. 2008). that the observed effects could be ascribed to the
These studies held aside, literature about the employed nanoemulsion compounds.
role of droplet size on the skin penetration of oth- Another study reported increased in vitro skin
erwise identical emulsions is scarce. However, by permeation rates of camphor, menthol and methyl
experimenting with certain sucrose ester surfac- salicylate through rat skin when the substances
tants with peculiar gelling behaviour, we were were applied in form of a hydrogel-thickened
able to create both fluid nanoemulsions and semi- nanoemulsion instead of a control gel (Mou et al.
solid emulsions of identical composition. When 2008). The authors speculated that different fac-
evaluating the skin penetration potential of these tors, e.g. high concentration gradients, formation
systems in vitro (Klang et al. 2011b) and in vivo of drug reservoirs or embedding of the small oil
(Klang et al. 2012b), we found a highly similar droplets within the stratum corneum lipids, might
penetration behaviour irrespective of the respec- account for the observed enhancement effect.
tive droplet size distribution. This may however In another recent study, penetration enhance-
be related to the specific nature of the developed ment of glycyrrhetic acid incorporated into a
emulsions, in particular the hydrophilic gel net- nanoemulsion was observed in vitro when com-
work of the semisolid emulsions. For aqueous pared to a conventional oil-in-water emulsion of
dispersions based on the same network, the different composition (Puglia et al. 2010). In
262 V. Klang et al.

addition, an increased antiinflammatory activity from negatively charged formulations containing


in vivo was observed for the nanoemulsion. deoxycholic acid (Piemi et al. 1999). The binding
However, the authors stated that the nature of the of surfactants to the skin can be attributed to non-
employed excipients for the different nanoemul- specific hydrophobic interactions with keratin as
sion and emulsion systems may have influenced well as to specific electrostatic interactions in
the obtained results. This again confirms that the case of charged surfactants; the latter phenome-
beneficial effects obtained with nanoemulsion non is strongly dependent on the pH of formula-
depend not only on the droplet size but on the tion and skin. The obtained results suggest that
nature of the developed systems as well as the the surface charge of nanoemulsion droplets may
employed compounds. affect skin penetration of drugs due to increased
interaction with the target site. Similar results
were reported in subsequent studies for stearyl-
18.2.2 Role of Droplet Surface amine (Fang et al. 2004) and phytosphingosine
Charge (Hoeller et al. 2009; Klang et al. 2010), although
the observed enhancement effects were not as
The surface charge of nanoemulsion oil droplets pronounced in case of the latter. Baspinar and co-
in dispersion, often described by the zeta poten- workers likewise employed phytosphingosine to
tial, is an important aspect that may strongly affect produce a positively charged nanoemulsion for
the interaction between formulation and skin and dermal application of prednicarbate. Higher drug
drug penetration. A positive droplet surface charge release was observed for negatively charged
has been observed to enhance penetration of drugs nanoemulsions in vitro when using diffusion
into the nasal and ocular mucosae as well as the cells and a synthetic model membrane. However,
stratum corneum of the skin (Yang and Benita significantly higher amounts of the active ingre-
2000; Mou et al. 2008). A high absolute surface dient were found to penetrate into the skin from
charge of the nanoemulsion droplets is also a cru- the positively charged nanoemulsion when using
cial prerequisite to ensure physical stability upon excised human skin (Baspinar and Borchert
storage. From this viewpoint, a high positive sur- 2012). These results suggest that a positive sur-
face charge may fulfil both the tasks of enhancing face charge in nanoemulsions may indeed be of
stability and promoting skin penetration. value for in vivo skin penetration.
A positive zeta potential can be induced by
using cationic excipients such as lipids, polymers
and surfactants, which can result in an improved 18.2.3 Role of Compounds
interaction with negatively charged membranes
such as the skin or corneal membrane. Negatively 18.2.3.1 Role of Surfactant Type
charged protein or fatty acid residues of the skin A wide range of permeation enhancers are known
as well as the presence of selective active ion to promote drug penetration into the skin or
pumps render the skin surface selective to posi- mucous membranes. Classical nanoemulsion sur-
tively charged substances (Yang and Benita 2000; factants such as phospholipids and lysophospho-
Piemi et al. 1999). It has been established by lipids act by interfering with the structure of skin
Benita and co-workers that positively charged lipids, thereby enhancing the transport of co-
nanoemulsions exhibited better wettability on the applied substances into the skin (Kirjavainen et al.
cornea compared to saline or negatively charged 1999). Current research focuses on identifying
systems (Yang and Benita 2000). In dermal drug further types of skin-friendly, biodegradable sur-
delivery, the same strategy was followed in sev- factants that may be employed for nanoemulsion
eral studies. Piemi et al. found that the in vitro production and exhibit similarly beneficial prop-
penetration of econazole and miconazole nitrate erties for dermal drug delivery. Among others,
into rat skin was higher from positively charged sucrose ester mixtures have been found interesting
nanoemulsions containing stearylamine than compounds for this task (Klang et al. 2011a;
18 Nanoemulsions in Dermal Drug Delivery 263

Calderilla-Fajardo et al. 2006; Cazares-Delgadillo accumulation of all-trans-retinol could be


et al. 2005). achieved by addition of oleylamine and silica
In first studies, synthetic surfactants such as nanoparticles to a conventional nanoemulsion,
polysorbate 80 were found to be more effective thus producing more complex interfacial films
permeation enhancers in comparative in vitro (Eskandar et al. 2009).
experiments than a hydrophilic sucrose laurate
ester of comparable HLB value (Hoeller et al. 18.2.3.2 Role of Additional
2009). This was ascribed to the enhancement Compounds
mechanism of the different surfactants: while Natural, semi-synthetic or synthetic lipids, fatty
sucrose laurate interferes with the lipid chains acids and oils can be employed for the production
and increases their fluidity, polysorbates may of oil-in-water nanoemulsions. For dermal appli-
increase skin penetration both by interacting with cation, skin-friendly cosmetic oils such as soy-
intercellular lipid domains and keratin filaments, bean oil, jojoba oil, castor oil, PCL liquid or
thus disturbing both the lipid matrix and the pro- squalene are popular choices. The presence of
tein domain of the corneocyte layers (Nokhodchi free fatty acids in the oil phase is beneficial for
et al. 2003). Later studies showed that sucrose nanoemulsion stability, as reported for castor oil
stearate mixtures of similar HLB value as lecithin (Jumaa and Mueller 1998). In addition, the polar-
mixtures were equally suitable in terms of drug ity of the oil phase affects the system’s stability
delivery from nanoemulsions and even superior against Ostwald ripening as well as drug release
in terms of physical stability (Klang et al. 2011a). properties. Squalene not only acts as a ripening
As already discussed, surfactants can be used inhibitor due to its hydrophobic nature but also
to modify the surface charge of the droplets. allows for the production of smaller droplet sizes
Cationic surfactants such as stearylamine, oleyl- in lecithin nanoemulsions than most other oils,
amine, chitosan or cetyltrimethylammonium bro- which in turn may affect skin interaction and
mide can be employed for this task. Apart from drug release (Fox 2009; Chung et al. 2001).
the amphiphilic phospholipids, nonionic surfac- Variation of the amount of incorporated oil
tants such as poloxamers or polysorbates are fre- may affect physical system properties such as
quently employed (Yang and Benita 2000). droplet size as well as drug release rates. Higher
Polysorbate-type surfactants possess voluminous amounts of oil may lead to decreased drug release
groups that provide for increased steric stabilisa- rates due to higher retention capacity (Hung et al.
tion. Likewise, surface-active polymers such as 2007). The microviscosity of the system may
the cellulose ether hydroxypropyl methylcellu- likewise be influenced by variations in the oil
lose (HPMC) can be employed to stabilise nano- content. As a result, the wetting properties of
emulsions as additional or even main surfactants nanoemulsions may be superior to those of con-
(Schulz and Daniels 2000). ventional marketed products, thus exhibiting an
Importantly, the nature of the interfacial film increased therapeutic effect particularly in muco-
surrounding the oil droplets may affect drug sal drug delivery (Ibrahim et al. 2009). In addi-
release. Mixtures of surfactants may lead to the tion, viscosity-enhancing additives may increase
formation of more compact interfacial films, the contact time between formulation and the
which consequently form a more efficient barrier skin. For this reason, nanoemulsions with or
against drug release (Ibrahim et al. 2009). without addition of gelling agents may show
Interfacial films composed of lecithin alone do improved penetration of incorporated drugs when
not represent a strong interfacial transport bar- compared to aqueous drug solutions or disper-
rier, thus usually showing burst release proper- sions (Mou et al. 2008).
ties. However, by creating mixed interfacial films If required, penetration enhancers such as
by including further surfactants or nanoparticles, propylene glycol or oleic acid can be incorpo-
controlled release properties can be achieved for rated into nanoemulsions to further enhance skin
topical application. Recently, enhanced dermal penetration (Fang et al. 2004). Addition of
264 V. Klang et al.

cosurfactants such as polysorbates, sucrose the emulsion phase inversion. Int J Pharm 344(1–2):
44–52
esters or alkyl polyglucosides can serve the same
Anton N, Mojzisova H, Porcher E, Benoit JP, Saulnier P
purpose (Schwarz et al. 2012; Hoeller et al. (2010) Reverse micelle-loaded lipid nano-emulsions:
2009; Klang et al. 2010). Recent studies have new technology for nano-encapsulation of hydrophilic
shown that cyclodextrins, i.e. cyclic polysaccha- materials. Int J Pharm 398(1–2):204–209
Baker MT, Naguib M (2005) Propofol: the challenges of
ride molecules, may likewise act as penetration
formulation. Anesthesiology 103(4):860–876
enhancers for specific drugs when incorporated Baspinar Y, Borchert HH (2012) Penetration and release
into nanoemulsions. This effect is most likely studies of positively and negatively charged nano-
caused by improved solubilisation and increased emulsions–is there a benefit of the positive charge? Int
J Pharm 430(1–2):247–252
availability of the drug at the skin surface and/or
Benita S (1999) Prevention of topical and ocular oxidative
involvement of the cyclodextrins in oil droplet stress by positively charged submicron emulsion.
formation (Klang et al. 2010, 2011a). Biomed Pharmacother 53(4):193–206
Benita S, Levy MY (1993) Submicron emulsions as colloi-
dal drug carriers for intravenous administration: com-
Conclusion
prehensive physicochemical characterization. J Pharm
In summary, the benefits of nanoemulsions for Sci 82(11):1069–1079
dermal application lie with their high skin Calder PC, Jensen GL, Koletzko BV, Singer P, Wanten GJ
friendliness and their excellent physical sta- (2010) Lipid emulsions in parenteral nutrition of
intensive care patients: current thinking and future
bility. The employed surfactants may serve to
directions. Intensive Care Med 36(5):735–749
promote skin penetration of incorporated Calderilla-Fajardo SB, Cazares-Delgadillo J, Villalobos-
drugs. Enhanced in vivo efficacy of different Garcia R, Quintanar-Guerrero D, Ganem-Quintanar
drugs has been observed for nanoemulsion A, Robles R (2006) Influence of sucrose esters on the
in vivo percutaneous penetration of octyl methoxycin-
formulations due to their specific morphology
namate formulated in nanocapsules, nanoemulsion,
and composition. Additional factors such as and emulsion. Drug Dev Ind Pharm 32(1):107–113
the droplet surface charge and viscosity of Cazares-Delgadillo J, Naik A, Kalia YN, Quintanar-
nanoemulsions can be modified with the help Guerrero D, Ganem-Quintanar A (2005) Skin perme-
ation enhancement by sucrose esters: a pH-dependent
of different additives to further improve skin
phenomenon. Int J Pharm 297(1–2):204–212
interaction and drug delivery. The possibility Cevc G, Vierl U (2010) Nanotechnology and the
to incorporate various lipophilic as well as Transdermal route: a state of the art review and critical
hydrophilic drugs by means of novel strate- appraisal. J Control Release 141(3):277–299
Chiesa M, Garg J, Kang YT, Chen G (2008) Thermal con-
gies renders nanoemulsions interesting vehi-
ductivity and viscosity of water-in-oil nanoemulsions.
cles for dermal and cosmetic applications. Colloids Surf A Physicochem Eng Asp 326:67–72
Chung H, Kim TW, Kwon M, Kwon IC, Jeong SY (2001)
Acknowledgement The authors would like to thank Dr. Oil components modulate physical characteristics and
Tim Maisch and co-workers for providing Fig. 18.2. function of the natural oil emulsions as drug or gene
delivery system. J Control Release 71(3):339–350
Cortés-Muñoz M, Chevalier-Lucia D, Dumay E (2009)
Characteristics of submicron emulsions prepared by
References ultra-high pressure homogenisation: Effect of chilled
or frozen storage. Food Hydrocoll 23(3):640–654
Alves PM, Pohlmann AR, Guterres SS (2005) Semisolid Dirschka T, Radny P, Dominicus R, Mensing H, Bruning
topical formulations containing nimesulide-loaded H, Jenne L et al (2012) Photodynamic therapy with
nanocapsules, nanospheres or nanoemulsion: develop- BF-200 ALA for the treatment of actinic keratosis:
ment and rheological characterization. Pharmazie results of a multicentre, randomized, observer-blind
60(12):900–904 phase III study in comparison with a registered methyl-
Anton N, Vandamme TF (2011) Nano-emulsions and 5-aminolaevulinate cream and placebo. Br J Dermatol
micro-emulsions: clarifications of the critical differ- 166(1):137–146
ences. Pharm Res 28:978–985 Eskandar NG, Simovic S, Prestidge CA (2009)
Anton N, Gayet P, Benoit JP, Saulnier P (2007) Nano- Nanoparticle coated submicron emulsions: sustained
emulsions and nanocapsules by the PIT method: an in-vitro release and improved dermal delivery of all-
investigation on the role of the temperature cycling on trans-retinol. Pharm Res 26(7):1764–1775
18 Nanoemulsions in Dermal Drug Delivery 265

Fang JY, Leu YL, Chang CC, Lin CH, Tsai YH (2004) characterisation and stability assessment. Micron
Lipid nano/submicron emulsions as vehicles for topi- 43:85–103
cal flurbiprofen delivery. Drug Deliv 11(2):97–105 Klang V, Schwarz JC, Lenobel B, Nadj M, Auböck J,
Fernandez P, André V, Rieger J, Kuehnle A (2004) Nano- Wolzt M et al (2012b) In vitro vs in vivo tape strip-
emulsion formation by emulsion phase inversion. ping: validation of the porcine ear model and penetra-
Colloids Surf A Physicochem Eng Asp 251:53–58 tion assessment of novel sucrose stearate emulsions.
Fox CB (2009) Squalene emulsions for parenteral vaccine Eur J Pharm Biopharm 80:604–614
and drug delivery. Molecules 14(9):3286–3312 Kotyla T, Kuo F, Moolchandani V, Wilson T, Nicolosi R
Friedman DI, Schwarz JS, Weisspapir M (1995) (2008) Increased bioavailability of a transdermal
Submicron emulsion vehicle for enhanced transdermal application of a nano-sized emulsion preparation. Int J
delivery of steroidal and nonsteroidal antiinflamma- Pharm 347(1–2):144–148
tory drugs. J Pharm Sci 84(3):324–329 Lademann J, Otberg N, Richter H, Weigmann HJ,
Hoeller S, Sperger A, Valenta C (2009) Lecithin based Lindemann U, Schaefer H et al (2001) Investigation of
nanoemulsions: a comparative study of the influence follicular penetration of topically applied substances.
of non-ionic surfactants and the cationic phytosphin- Skin Pharmacol Appl Skin Physiol 14(1):17–22
gosine on physicochemical behaviour and skin perme- Maisch T, Santarelli F, Schreml S, Babilas P, Szeimies
ation. Int J Pharm 370(1–2):181–186 RM (2010) Fluorescence induction of protoporphyrin
Hung CF, Fang CL, Liao MH, Fang JY (2007) The effect IX by a new 5-aminolevulinic acid nanoemulsion used
of oil components on the physicochemical properties for photodynamic therapy in a full-thickness ex vivo
and drug delivery of emulsions: tocol emulsion versus skin model. Exp Dermatol 19(8):e302–e305
lipid emulsion. Int J Pharm 335(1–2):193–202 Marxer EE, Brussler J, Becker A, Schummelfeder J,
Ibrahim SS, Awad GA, Geneidi A, Mortada ND (2009) Schubert R, Nimsky C et al (2011) Development and
Comparative effects of different cosurfactants on ster- characterization of new nanoscaled ultrasound active
ile prednisolone acetate ocular submicron emulsions lipid dispersions as contrast agents. Eur J Pharm
stability and release. Colloids Surf B: Biointerfaces Biopharm 77(3):430–437
69(2):225–231 Mason TG, Wilking JN, Meleson K, Chang CB, Graves
Jumaa M, Mueller BW (1998) The effect of oil compo- SM (2006) Nanoemulsions: formation, structure, and
nents and homogenization conditions on the physico- physical properties. J Phys Condens Matter 18:
chemical properties and stability of parenteral fat R635–R666
emulsions. Int J Pharm 163:81–89 McClements DJ (2012) Nanoemulsions versus micro-
Kirjavainen M, Monkkonen J, Saukkosaari M, Valjakka- emulsions: terminology, differences, and similarities.
Koskela R, Kiesvaara J, Urtti A (1999) Phospholipids Soft Matter 8:1719–1729
affect stratum corneum lipid bilayer fluidity and drug Mitri K, Shegokar R, Gohla S, Anselmi C, Mueller RH
partitioning into the bilayers. J Control Release (2011) Lipid nanocarriers for dermal delivery of
58(2):207–214 lutein: preparation, characterization, stability and per-
Klang V, Valenta C (2011) Lecithin-based nanoemul- formance. Int J Pharm 414(1–2):267–275
sions. J Drug Del Sci Tech 21(1):55–76 Mou D, Chen H, Du D, Mao C, Wan J, Xu H et al (2008)
Klang SH, Parnas M, Benita S (1998) Emulsions as drug Hydrogel-thickened nanoemulsion system for topical
carriers – possibilities, limitations and future perspec- delivery of lipophilic drugs. Int J Pharm 353(1-2):
tives. In: Mueller RH, Benita S, Böhm BHL (eds) 270–6
Emulsions and nanosuspensions for the formulation of Mueller RH (1996) Zetapotential und Partikelladung in
poorly soluble drugs. Medpharm Scientific Publishers, der Laborpraxis. Band 37, Paperback APV ed.
Stuttgart, pp 31–56 Stuttgart: Wissenschaftliche Verlagsgesellschaft mbH
Klang V, Matsko N, Zimmermann AM, Vojnikovic E, Nokhodchi A, Shokri J, Dashbolaghi A, Hassan-Zadeh D,
Valenta C (2010) Enhancement of stability and skin per- Ghafourian T, Barzegar-Jalali M (2003) The enhance-
meation by sucrose stearate and cyclodextrins in pro- ment effect of surfactants on the penetration of loraz-
gesterone nanoemulsions. Int J Pharm 393(1–2):1 epam through rat skin. Int J Pharm 250(2):359–369
52–160 Norden TP, Siekmann B, Lundquist S, Malmsten M
Klang V, Matsko N, Raupach K, El-Hagin N, Valenta C (2001) Physicochemical characterisation of a drug-
(2011a) Development of sucrose stearate-based nano- containing phospholipid-stabilised o/w emulsion for
emulsions and optimisation through gamma- intravenous administration. Eur J Pharm Sci 13(4):
cyclodextrin. Eur J Pharm Biopharm 79:58–67 393–401
Klang V, Schwarz JC, Matsko N, Rezvani E, El-Hagin N, Piemi MP, Korner D, Benita S, Marty J-P (1999) Positively
Wirth M et al (2011b) Semi-solid sucrose stearate- and negatively charged submicron emulsions for
based emulsions as dermal drug delivery systems. enhanced topical delivery of antifungal drugs. J Control
Pharmaceutics 3:275–306 Release 58(2):177–187
Klang V, Matsko NB, Valenta C, Hofer F (2012a) Electron Porras M, Solans C, González C, Martinez A, Guinart A,
microscopy of nanoemulsions: an essential tool for Gutiérrez JM (2004) Studies of formation of W/O
266 V. Klang et al.

nano-emulsions. Colloids Surf A Physicochem Eng a calixarene nanoemulsion. Eur J Pharm Biopharm
Asp 249(1–3):115–118 79(2):258–267
Puglia C, Rizza L, Drechsler M, Bonina F (2010) Tadros T, Izquierdo P, Esquena J, Solans C (2004)
Nanoemulsions as vehicles for topical administration Formation and stability of nano-emulsions. Adv
of glycyrrhetic acid: characterization and in vitro and Colloid Interface Sci 108–109:303–318
in vivo evaluation. Drug Deliv 17(3):123–129 Ullrich S, Metz H, Maeder K (2008) Sucrose ester nano-
Schulz MB, Daniels R (2000) Hydroxypropyl methylcel- dispersions: microviscosity and viscoelastic proper-
lulose (HPMC) as emulsifier for submicron emul- ties. Eur J Pharm Biopharm 70(2):550–555
sions: influence of molecular weight and substitution van Nieuwenhuyzen W, Szuhaj BF (1998) Effects of leci-
type on the droplet size after high-pressure homogeni- thins and proteins on the stability of emulsions. Fett-
zation. Eur J Pharm Biopharm 49(3):231–236 Lipid 100(7):282–291
Schwarz JC, Klang V, Karall S, Mahrhauser D, Resch GP, Wabel C (1998) Influence of lecithin on structure and sta-
Valenta C (2012) Optimisation of multiple W/O/W bility of parenteral fat emulsions [Dissertation].
nanoemulsions for dermal delivery of aciclovir. Int J Friedrich-Alexander-Universität, Erlangen-Nürnberg
Pharm 435(1):69–75 Yang SC, Benita S (2000) Enhanced absorption and drug
Sole I, Pey CM, Maestro A, Gonzalez C, Porras M, Solans targeting by positively charged submicron emulsions.
C et al (2010) Nano-emulsions prepared by the phase Drug Dev Res 50(3–4):476–486
inversion composition method: preparation variables Yu C, Meng J, Chen J, Tang X (2009) Preparation of ergol-
and scale up. J Colloid Interface Sci 344(2):417–423 oid mesylate submicron emulsions for enhancing nasal
Sonneville-Aubrun O, Simonnet JT, L’Alloret F (2004) absorption and reducing nasal ciliotoxicity. Int J Pharm
Nanoemulsions: a new vehicle for skincare products. 375(1–2):16–21
Adv Colloid Interface Sci 108–109:145–149 Zhou H, Yue Y, Liu G, Li Y, Zhang J, Gong Q et al (2010)
Spagnul A, Bouvier-Capely C, Phan G, Landon G, Tessier Preparation and characterization of a lecithin
C, Suhard D et al (2011) Ex vivo decrease in uranium nanoemulsion as a topical delivery system. Nanoscale
diffusion through intact and excoriated pig ear skin by Res Lett 5:224–230
Pickering Emulsions for Controlled
Drug Delivery to the Skin 19
Yves Chevalier, Marie-Alexandrine Bolzinger,
and Stéphanie Briançon

Contents 19.1 Introduction


19.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . 267
Pickering emulsions are emulsions of any type,
19.2 Physical Chemistry of Pickering
Emulsions . . . . . . . . . . . . . . . . . . . . . . . . . 268
either oil-in-water (o/w), water-in-oil (w/o), or even
19.2.1 Adsorption of Solid Particles at Interfaces multiple, stabilized by solid particles in place of
and Stabilization of Emulsions . . . . . . . . . 269 surfactants (Aveyard et al. 2003; Binks 2002; Binks
19.2.2 Emulsion Type, Emulsion Inversion, and Horozov 2006). Although such emulsions did
and Double Emulsions . . . . . . . . . . . . . . . 269
19.2.3 Control of Emulsion Properties
not receive much development towards their appli-
by Formulation and Process Parameters . . 270 cation, their properties are quite attractive and
deserve special attention. The interest lays in the
19.3 Skin Delivery of Drugs . . . . . . . . . . . . . . 272
19.3.1 O/W Pickering Emulsions . . . . . . . . . . . . 274 fact that Pickering emulsions essentially remain
19.3.2 W/O Pickering Emulsions . . . . . . . . . . . . 275 emulsions, i.e., they share most properties of emul-
19.3.3 Mechanisms of Enhanced sions with their conventional surfactant-based
Skin Absorption . . . . . . . . . . . . . . . . . . . . 276
homologues. Additionally, they have also few spe-
19.3.4 Fate of the Stabilizing Solid Particles . . . . 278
cific properties that are advantageous in the field of
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 279 drug delivery to the skin. The solid stabilizing par-
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 279 ticles behave in quite a similar way as surfactant
molecules: one part is adsorbed at the oil-water
interface and the residual part is remaining in the
continuous phase according to the adsorption equi-
librium. Solid particles form a dense coating that
can be seen by optical microscopy when the parti-
cles are large enough (Fig. 19.1).
The name “Pickering emulsion” was given
after their early disclosure by S.U. Pickering
(1907). Actually, the adsorption of solid particles
Y. Chevalier (*) • M.-A. Bolzinger • S. Briançon at the air-water interface has been reported earlier
Laboratoire d’Automatique et de Génie des Procédés (Ramsden 1903). However, the merit has been
(LAGEP), CNRS UMR 5007, University of Lyon,
given to S.U. Pickering, because his paper spe-
Université Claude Bernard Lyon 1,
43 bd 11 Novembre, Villeurbanne 69622, France cifically dealt with emulsions stabilized by solid
e-mail: chevalier@lagep.univ-lyon1.fr particles; he reported improved stability for these

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 267


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_19, © Springer-Verlag Berlin Heidelberg 2015
268 Y. Chevalier et al.

where surfactants often cause either irritancy or


hemolysis.
Applications of Pickering emulsions in phar-
maceutical and cosmetic fields rely on either
their specific properties manifested in vivo (low
irritancy related the surfactant-free character,
specific interactions with biological interfaces)
or the development of new dosage forms having
improved ex vivo properties (emulsion stability,
thickening with no polymeric thickener) over
surfactant-based emulsions. The present chapter
is dealing with percutaneous penetration-­
Fig. 19.1  Optical microscopy picture of an o/w Pickering
enhancing properties of Pickering emulsions
emulsion of silicone oil stabilized by zinc oxide (ZnO) par- and covers the former aspect where drug-loaded
ticles. Addition of a large concentration of sodium chloride Pickering emulsions modify the transport of
(NaCl, 1 mol·L−1) caused the partial flocculation of ZnO drug molecules into/through skin. This includes
particles into large aggregates that could be observed at the
surface of oil droplets by optical microscopy. The oil drop-
the classical “penetration enhancer” action simi-
lets are covered by a dense coating of ZnO particles over the lar to that of “penetration enhancer” chemicals
brown areas where large aggregates are also visible. The and also any modification of the drug transport
white areas are bare. The picture also shows free ZnO par- induced by the formulation in a Pickering emul-
ticles dispersed in the aqueous phase which are seen as
aggregates in between the oil droplets
sion. The behavior of Pickering emulsions is
discussed with respect to a reference surfactant-
based emulsion.
emulsions with respect to surfactant-based emul- The physicochemical properties of Pickering
sions and provided a definite proof for the adsorp- emulsions are shortly reviewed in the first part of
tion of solid particles, being the origin for such the chapter. The second part reports on the skin
stabilization. absorption behavior of drugs followed by a dis-
Pickering emulsions are essentially emul- cussion of the mechanism of the effects induced
sions, so that their main properties are common by Pickering emulsions compared to surfactant-­
with classical emulsions stabilized by surfactants based emulsions.
(emulsifiers). Pickering emulsions are prepared
using the same manufacturing processes as for
classical emulsions. There might be a specific 19.2 Physical Chemistry
process for Pickering emulsification; however, of Pickering Emulsions
the most often used preparation methods are the
same as for classical emulsions. As a conse- The origin of emulsion stabilization is the adsorp-
quence, the application domains of Pickering tion of solid particles at the surface of emulsion
emulsions are the same as for classical emul- droplets. The mechanism of adsorption is partial
sions, and an application based on classical wetting of the particles by oil and water, which is
emulsions can easily be switched to Pickering very different of surfactants; the solid particles are
emulsion. Pickering emulsions show some not amphiphilic. Stabilization takes place by pre-
improved properties compared to classical emul- venting destabilization events: coagulation, coales-
sions. Adsorbed solid particles act as a more cence, and Ostwald ripening. There are strong
effective barrier against coalescence than surfac- similarities with classical surfactant-based emul-
tants. Such important benefit opens the possible sions; and there are few differences that provide
stabilization of coarse emulsions and multiple specificities to Pickering emulsions. The manufac-
emulsions. The “surfactant-free” character makes turing processes of Pickering emulsions used so
them attractive to applications to life sciences far are the same as those of classical emulsions.
19  Pickering Emulsions for Controlled Drug Delivery to the Skin 269

19.2.1 Adsorption of Solid Particles not show surface activity, which is a definite dif-
at Interfaces and Stabilization ference compared to surfactants (Vignati et al.
of Emulsions 2003; Dong and Johnson 2003).
Stabilization of Pickering emulsions occurs
Particles adsorbed at the oil-water interface are because the layer of adsorbed solid particles
wet both by oil and water. Wetting conditions forms a rigid coating that acts as a mechanical
depend on interfacial tensions of the solid-water, barrier against coalescence. Such a rigid protec-
solid-oil, and oil-water interfaces, γs-w, γs-o, and tive coating has been compared to an eggshell.
γo-­w. Under partial wetting conditions, the contact The origin of its mechanical strength is the two-­
angle in water, θw, is given by the Young’s law: dimensional aggregation of solid particles at the
droplet surface by means of capillary forces.
g s-o − g s-w
cos (q w ) = (19.1) A supplementary three-dimensional aggregation
g w -o takes place in some instances, building a thick

solid layer of solid particles. Therefore, the sta-
Partial wetting is quite common. Complete wet- bility of Pickering emulsions is very high com-
ting by water occurs for very hydrophilic sur- pared to classical emulsions. It allows the
faces; such particles remain dispersed in the preparation of either concentrated emulsions
aqueous phase. This is the case of silica that can- (high internal phase ratio) or coarse emulsions
not be used as stabilizing particle for most com- (emulsion with droplet size in the millimeter
mon oils (Frelichowska et al. 2009a). Silica is range) that conventional surfactants would not be
very often used however because there are com- able to stabilize with sufficient efficiency. Solid
mercially available grades of “hydrophobized” particles may also prevent coagulation if they
silica having organosilanes chemically grafted to cause thickening of the continuous phase. This
their surface (Barthel 1995). Wetting of such sil- effect is similar to the stabilizing action of poly-
ica surfaces is controlled by the grafting degree meric thickeners that are often included in the
of the organosilane. Adsorption of organic mole- formulation of conventional emulsions.
cules such as surfactants, polymers, or even small Numerous particles are able to stabilize
surfactant-like organic molecules, to the solid Pickering emulsions. They are either inorganic or
particles, allows adjusting the surface properties organic. Hydrophobized silica and clay (mont-
of the stabilizing particles so as to achieve the morillonite, laponite, kaolin) are the most com-
desired emulsion type and optimum stability mon inorganic particles. Latex and carbohydrate
(Gelot et al. 1984; Hassander et al. 1989; nanocrystals (cellulose, chitin) are examples of
Midmore 1998a, b, 1999; Ghouchi Eskandar suitable organic particles.
et al. 2007, 2011; Akartuna et al. 2009; Drelich
et al. 2010). However, surface-active molecules
that adsorb at the surface of solid particles may 19.2.2 Emulsion Type, Emulsion
also adsorb at the oil-water interface and contrib- Inversion, and Double
ute to the emulsion stability in the same way as Emulsions
classical surfactants do. Adsorption of solid par-
ticles is very strong when partial wetting condi- The emulsion type is controlled by the wettability of
tions are met. The strongest adsorption and the the solid particles (Aveyard et al. 2003; Binks and
maximum stability of emulsions are reached Lumsdon 2000): hydrophilic particles stabilize o/w
when the contact angle is 90° (Binks and emulsions and hydrophobic particles stabilize w/o
Lumsdon 2000a). Obviously, large particles hav- emulsions. Examples of hydrophilic particles are
ing a larger area contacting oil and water show silica, clay, titanium dioxide, and zinc oxide.
larger adsorption strength. Even small solid Examples of hydrophobic particles are “hydropho-
nanoparticles also adsorb to the oil-water inter- bic” silica (grafted with organic silanes), “hydro-
face quite strongly. Stabilizing solid particles do phobic bentonite” (montmorillonite coated with
270 Y. Chevalier et al.

fatty quaternary ammonium salts) polystyrene, and acids. The stabilization of multiple emulsions
polytetrafluoroethylene (PTFE, Teflon®). Too much using emulsifiers is a difficult task because the
hydrophilic (or hydrophobic) particles are totally stability is lost when the emulsifiers adsorbed at
wet by water (or oil) however; partial wetting is an the surfaces of the w/o internal and w/o/w drop-
absolute requirement for solid particles adsorption lets mix together. The strong adsorption of solid
at the oil-water interface. This is very similar to the particles to the various oil-water interfaces is an
Bancroft rule for emulsifiers stating that a hydro- obvious benefit because particles keep retained at
philic emulsifier (high HLB) gives rise to an o/w the right interfaces and do not mix.
emulsion type, while a hydrophobic emulsifier (low A classical two-step process was used to pre-
HLB) gives rise to a w/o emulsion type. Accordingly, pare w/o/w multiple emulsions of medium-chain
a HLB-like rule has been introduced for solid parti- triglycerides: a primary w/o emulsion was stabi-
cles by Kruglyakov (2000). The relevant quantity is lized by hydrophobic silica particles (coated with
the contact angle in water, θw, defined by the Young’s grafted dimethylsilyl groups at 49 % coverage);
law (Eq. 19.1): o/w emulsions form when θw <90°, the obtained primary w/o emulsion was then dis-
and w/o emulsions form when θw >90°. In contrast persed in water using hydrophilic silica particles
with the behavior of emulsifiers, the contact angle (coated with dimethylsilyl groups at 21 % cover-
for optimum stability is close to θw = 90°. Therefore, age) as stabilizing particles, yielding a stable
“balanced particles” are efficient stabilizers, w/o/w emulsion (Barthel et al. 2003).
whereas “balanced surfactants” behave as poor
emulsifiers. As an example, the ­well-­known lecithin
is a “balanced surfactant” that cannot stabilize 19.2.3 Control of Emulsion
emulsions; it needs being mixed with either a hydro- Properties by Formulation
philic surfactant in order to stabilize o/w emulsions and Process Parameters
or with a hydrophobic surfactant for o/w emulsions.
In other words, optimum emulsifiers are shifted Once the emulsion type and the nature of the
apart from the HLB of “balanced surfactants”; con- ingredients have been chosen, the droplet size and
versely, “balanced particles” are at optimum. As a the rheological behavior are the main physico-
corollary, the same type of solid particles meets the chemical properties of emulsions that matter in
stabilization criteria of either o/w or w/o emulsion regard to their skin delivery. Such properties are
type. The Bancroft rule for solid particles is less a controlled by the formulation (the type and con-
clear-cut rule than for emulsifiers. Phase inversion centration of the ingredients) and the emulsifica-
does not take place when the contact angle is close tion process.
to 90°. Catastrophic phase inversion upon progres-
sive addition of the continuous phase does not hap- 19.2.3.1  Droplet Size of Pickering
pen as readily as for surfactants. Apart from the Emulsions
wetting behavior, supplementary parameters such The concentration of solid particles controls the
as the relative water and oil contents, the medium droplet size. This meets expectations as it is sim-
where the solid particles have been initially dis- ilar to the effect of the concentration of surfac-
persed, have a definite influence on the emulsion tants. Indeed, a larger amount of stabilizing
type (Binks and Lumsdon 2000a, b, c; Binks and species allows a larger interfacial area to be
Rodrigues 2003). Another consequence of both o/w formed, thus smaller droplets. Under conditions
and w/o emulsion types being at optimum for θw = such that solid particles adsorb as a dense mono-
90° is a strong hysteresis in phase inversion experi- layer, the total oil-water interfacial area is in
ments (Binks and Rodrigues 2003; Kruglyakov and proportion to the amount of solid particles, and
Nushtayeva 2004). the mean droplet diameter is given by the fol-
Multiple emulsions of the w/o/w type are lowing relationship (for an o/w emulsion) based
attractive dosage forms for encapsulation of on simple geometrical considerations (Wiley 1954;
hydrophilic drugs such as proteins and nucleic Arditty et al. 2003; Frelichowska et al. 2010):
19  Pickering Emulsions for Controlled Drug Delivery to the Skin 271

6 M ( oil ) Pickering emulsions is controlled by both formula-


Diam = (19.2) tion and emulsification process parameters.
ρoila solid M ( solid )
On the basis of the geometrical relationship
where M(oil)/M(solid) is the mass ratio of oil between droplet diameter and oil/solid mass ratio
(dispersed phase) to solid particles, ρoil is the den- (Eq. 19.2), very fine emulsions might be prepared
sity of oil (kg m−3), and asolid is the interfacial area at a high content of solid particles. Such expecta-
covered per mass of adsorbed solid particles tion is based on the hypothesis of droplet-size
(m2 kg−1). Such a linear relationship has often control by the amount of stabilizing particles.
been experimentally verified (Arditty et al. 2004, A high-power emulsification process is required
2005); but cases of departure from Eq. 19.2 have for the fragmentation of droplets. In the case of
also been reported (Wang and Hobbie 2003; lack of power, the smallest droplets that the pro-
Binks and Whitby 2004; Frelichowska et al. cess can create do form, and only a part of the
2010). Incomplete coverage by solid particles stabilizing particle is used for reaching full cov-
may also lead to stable Pickering emulsions; erage of the droplets. The remaining part of the
however, there are several microscopic observa- stabilizing particles is not used for the stabiliza-
tions of such emulsions (Fig. 19.1) (Binks and tion; such excess particles are left free in the con-
Kirkland 2002; Horozov and Binks 2006; tinuous phase.
Destribats et al. 2010). As a summary, three distinct regimes are
The mechanisms being responsible for the drop- encountered depending on the mass ratio of solid
let size in surfactant-based and Pickering emulsions particles to the dispersed phase (Fig. 19.2). In the
are different. The reduction of droplet size by first regime at low solid content, either emulsifi-
increasing surfactant concentration results from cation fails or coarse emulsions are prepared pro-
two phenomena: on one hand, the simple geometri- vided a suitable process has been designed. In the
cal argument given above holds; on the other hand, second regime, the droplet size is controlled by
fast surfactant adsorption during the emulsification the ratio M(particles)/M(oil) according to
process causes a decrease of the interfacial tension Eq.  19.2. The third regime is reached when the
that makes the droplets fragmentation easier under emulsification process is not able to create the
shear. On the contrary, adsorption of solid particles interfacial area that the amount of solid particle
does not change the interfacial tension (Vignati might stabilize. The droplet size gets controlled
et al. 2003; Dong and Johnson 2003) (unless the by the emulsification process parameters such as
particles contain surface-­active impurities or they homogenizer type, design of the instruments
are associated with a surfactant). (propeller, rotor-stator, etc.), and stirring speed.
In the case of a low amount of solid particles, a The crossover from the second to the third regime
“limited coalescence” ripening phenomenon may depends on the power of the emulsification
occur: the poorly stabilized emulsion droplets process.
undergo coalescence till the total interfacial area
matches the area that the solid particles can stabi- 19.2.3.2  Rheology of Pickering
lize (Wiley 1954; Arditty et al. 2004). Successful Emulsions
limited coalescence requires that the emulsion The viscosity of emulsions is an important prop-
remains stable during the ripening process; other- erty regarding their use as formulations for topi-
wise emulsification fails as a part of the oil is cal administration. Indeed it controls the materials
released (Frelichowska et al. 2010; Avendaño transfer from the emulsion droplets to the skin.
Juárez and Whitby 2012). Under conditions of low When all solid stabilizing particles are adsorbed
particle content, large droplets can only be stabi- to the emulsion droplets, the rheological behavior
lized. A suitable emulsification process yields sta- of Pickering emulsions is the same as that of
ble coarse emulsions having droplets within surfactant-­based emulsions. Emulsions are fluid
millimeter size range (Zhai and Efrima 1996; as long as the concentration of droplets remains
Arditty 2003). Formation of poorly stabilized low; strong thickening appears in concentrated
272 Y. Chevalier et al.

Fig. 19.2  Scheme of the different behaviors taking place (amounts of solid particles and oil). The droplet size is con-
according to the mass ratio M(particles)/M(oil). In the first trolled by the emulsification process in the third regime at
regime at low particle/oil ratio, either emulsification pro- high particle/oil ratio; excess solid particles is dispersed in
cess fails or a stable coarse emulsion is formed. Full adsorp- the aqueous phase and possibly particles aggregate, form-
tion of solid particles takes place in the second regime ing a percolating continuous network in case of strong
where the droplet size is controlled by the formulation aggregation or large amount of excess particles

emulsions when the volume fraction of droplets stuck to the network of solid particles, so that the
reaches 50–60 %. High internal phase ratio emul- oil droplets were part of the gel (Neuhäusler et al.
sions (HIPEs, gel emulsions) are viscoelastic 1999; Thieme et al. 1999).
materials found for concentrations above The flocculation of excess solid particles can
60–70 %. A specific feature of Pickering emul- be induced in a controlled manner by physico-
sions is the thickening action of excess of free chemical parameters such as salinity of the aque-
particles (non-adsorbed) present in the aqueous ous phase. Thus, low salinity leave well-dispersed
phase (of o/w emulsions). In general, the solid or weakly flocculated particles and the emulsion
particles that are able to adsorb onto oil droplets is fluid (either Newtonian or rheo-thinning fluid);
also tend to self-aggregate in water because they high salinity induces the formation of a continu-
are not hydrophilic enough to remain well dis- ous network of solid particles in the continuous
persed. As an example, the hydrophobic fumed phase that causes gelation of the emulsion. The
silica particles used for the stabilization of rheological properties of practical relevance, in
Pickering emulsions have been primarily made particular the yield stress, can be adjusted by
commercially available as thickening agents means of simple addition of electrolytes (Horozov
(Barthel 1995). et al. 2007; Whitby et al. 2011).
A nice example of gelation induced by solid
particles in Pickering emulsions is given by
Abend and Lagaly (1998, 2001). A percolating 19.3 Skin Delivery of Drugs
network of flocculated solid particles has been
built by heterocoagulation of two types of parti- Topical application of drugs loaded in Pickering
cles. The adsorbed particles prevented coales- emulsions is of particular relevance for pharmaceu-
cence; the remaining part caused thickening and tical and cosmetic applications. It shows advantage
prevented coagulation. Interestingly, X-ray due to the surface properties of droplets covered by
microscopy observations (Fig. 19.3) have shown solid particles and the possible controlled drug
that the oil droplets coated by solid particles were release through the barrier of solid particles present
19  Pickering Emulsions for Controlled Drug Delivery to the Skin 273

Fig. 19.3  X-ray microscopy images of o/w Pickering emul- ous phase, as well as entrapped oil droplets appearing as large
sion stabilized by mixed montmorillonite and layered double black circles. Montmorillonite particles appear as platelets
hydroxide particles that underwent heterocoagulation. A con- and layered double hydroxide appear as particles of nearly
tinuous network of aggregated particles is seen in the continu- spherical shape (Thieme et al. (1999), with permission)

at the surface of droplets. An important benefit for drugs by Pickering emulsions has been approached
cosmetic applications is the surfactant-free charac- in comparative investigations of Pickering and
ter of these emulsions which enables the avoidance surfactant-­based emulsions by several groups as
of irritancy caused by surfactants. Skin delivery of reported in the following.
274 Y. Chevalier et al.

Pickering emulsions behave as penetration ascribed to the penetration enhancer action of the
enhancers in some instances where the skin perme- emulsifiers (Brinon et al. 1998; Montenegro et al.
ation of a drug loaded inside the droplets of a 2008; Otto et al. 2009). Such an effect cannot hold
Pickering emulsion was faster than for a conven- for Pickering emulsions. That a surfactant-free
tional emulsion or a homogeneous solution. emulsion causes high penetration rate casts doubts
Although the mechanism of accelerated transport on the role of emulsifiers as the origin of improved
is not clearly established yet, it is quite obvious that
skin penetration from conventional emulsions.
Pickering emulsions do not behave as c­lassical The total amounts of retinol found in the skin
penetration enhancers that increase the permeabil- were identical for both emulsions, but the distri-
ity of the stratum corneum by fluidization of the bution of retinol along the various skin layers
intercellular lipids. Pickering emulsions cause a showed large differences. Thus, retinol absorbed
faster permeation in some instances. They may also from the Pickering emulsion was strongly retained
cause accumulation of drugs inside the stratum cor- in the stratum corneum, and it consequently
neum. In conclusion, there are definite differences reached the viable epidermis and dermis to a
with respect to surfactant-­based emulsions. Such lesser extent in comparison to the surfactant-
effects are discussed in the following as being a based emulsion. Tape-­stripping experiments have
generalized penetration enhancement behavior. shown that storage of retinol in stratum corneum
took place especially in its outermost layers when
Pickering emulsion was used (Fig. 19.4).
19.3.1 O/W Pickering Emulsions A very similar investigation of skin delivery of
retinol has been carried out independently at the
Skin penetration of a hydrophobic drug, all-trans same time from a medium chain triglycerides
retinol, from o/w Pickering emulsions has been emulsion stabilized by mixed particle/surfactant
compared to the penetration from a classical (fumed silica nanoparticles + either anionic soy-
surfactant-­based emulsion (Frelichowska 2009b). bean lecithin or cationic oleylamine) (Ghouchi
For the comparison to make sense, the fundamental Eskandar et al. 2009). Interestingly, similar conclu-
properties of both emulsions were set identical: sions were reached although the stabilizing system
the two emulsions had the same chemical compo- of the Pickering emulsion was different. Retinol
sition, but the stabilizing ingredients were different was accumulated in the outermost layers of the
(either solid particles or surfactant molecules), skin. The in-depth distribution of retinol in the skin
and they had the same droplet size and the same was measured by cutting the skin into several hori-
viscosity. The purpose was to investigate the influ- zontal 100 μm thick slices that were analyzed for
ence of the droplet coating composed of solid par- their retinol content. A full comparison with the
ticles better than comparing a Pickering emulsion study by Frelichowska et al. (2009b) is difficult
to an emulsion taken from a commercial product. because of the low in-depth spatial resolution of the
Owing to its high lipophilic character (logP​ method based on parallel slicing compared to that
octanol = 5.68), the main part of retinol remains of tape stripping (Touitou et al. 1998). Thus, the
stored in the lipidic medium of stratum corneum. first slice containing the largest amount of retinol
As consequence, permeation is negligible for both included the full stratum corneum and a large part
Pickering and classical emulsions. Accumulated of the viable epidermis. The “Pickering” emulsions
amounts of retinol have been measured in the skin were basically surfactant-­based emulsions to which
layers of excised pig skin after 24 h exposure to silica was added, so that it is difficult to conceive
o/w Pickering and classical emulsions of medium the relative contribution of solid particles and sur-
chain triglycerides loaded with 0.1 % retinol and factant molecules. The skin absorption was larger
a solution in oil as reference. The skin penetration than in the study by Frelichowska et al. (2009b)
from emulsions was much larger than from the pertaining to true Pickering emulsions (free of sur-
solution. A significant contribution to faster factant); in particular, the second and third slices
absorption from classical emulsion has often been corresponding to 100–300 μm depth contained
19  Pickering Emulsions for Controlled Drug Delivery to the Skin 275

Fig. 19.4  Distribution of retinol in skin layers after 24 h viable epidermis, and dermis (applied dose =
exposure to Pickering emulsion (black), classical emul- 400,000 ng·cm−2). Right: Distribution inside the stratum
sion (gray), and solution in medium chain triglycerides corneum obtained by the tape-stripping technique
(dashed). Left: Cumulated amount in stratum corneum,

large amounts of retinol. Such differences with loaded in the lecithin-stabilized emulsion was
respect to surfactant-free Pickering emulsions were mainly retained in the stratum corneum, whereas
probably caused by the penetration-­ enhancing its penetration was deeper in the epidermis when
action of the surfactants (especially the oleyl- loaded in the emulsion stabilized by mixed silica/
chained oleylamine) present at a fairly high con- lecithin. However, the effects of solid particles
centration (1 % for 10 % oil content). Indeed the were hardly perceptible in the same experiments
infrared spectrum of stratum corneum lipids was performed with oleylamine as emulsifier instead
altered by the presence of the oleylamine surfactant of lecithin. Two effects explain this result: loading
in the emulsion. The presence of silica particles sig- of the cationic AONB inside oil droplets was low
nificantly increased the absorption of retinol with because of the electrostatic repulsions with oleyl-
respect to the silica-­free emulsion (only surfactant- amine emulsifier, and the strong penetration-
based). Silica could be considered as a penetration enhancing effect of oleylamine screened the
enhancer in such systems or silica acted in synergy possible effect of the presence of solid particles.
with the surfactants, which showed definite differ- As summary, accumulation inside the stratum
ence with the surfactant-free Pickering emulsions. corneum and less penetration to the deeper skin lay-
Accelerated skin absorption of drugs has ers has been observed for the highly hydrophobic
recently been shown for o/w Pickering emulsions retinol. This is not a general phenomenon however.
loaded with molecules of medium polarity. The Less hydrophobic drugs loaded in o/w Pickering
steady-state permeation flux of methyl salicylate emulsion showed also accelerated penetration to the
(logP = 2.5) loaded in a Pickering emulsion was dermis and permeation into receiver compartment.
twice higher than the aqueous solution (Marku
et al. 2012). The permeation and accumulation
inside skin of the fluorescent probe acridine orange 19.3.2 W/O Pickering Emulsions
10-nonyl bromide (AONB) was enhanced by the
presence of silica particles adsorbed at the surface Skin delivery of a hydrophilic drug, caffeine,
of an o/w emulsion stabilized by the lecithin emul- from a w/o Pickering emulsion has been studied
sifier (Ghouchi Eskandar et al. 2010). AONB in the same way as the retinol absorption from
276 Y. Chevalier et al.

o/w emulsions. Permeation of caffeine was mea- in order to evaluate their relative contributions
sured from a Pickering emulsion compared to a and finally disclose the main origin of the faster
surfactant-based emulsion having the same permeation through the skin. The different path-
chemical composition (but the stabilizing layer ways of the drug molecule from the emulsion
being composed of emulsifier molecules instead droplets to the deep skin layers are considered.
of silica nanoparticles), the same droplet size, The first step is drug release from inside the
and the same viscosity (Frelichowska et al. emulsion droplets to the surrounding medium;
2009c). Results of the in vitro drug diffusion the release behavior is often referred to as the
through excised pig skin revealed a higher trans- bioavailability. Since the dense coating of solid
dermal flux of caffeine through excised skin particles around the emulsion droplets efficiently
(Fig. 19.5). Such higher permeation in an in vitro stabilizes the emulsion with respect to emulsifier-­
experiment suggests that a larger delivery to the based emulsions, it is presumed that such layer
deep dermis and hypodermis would occur with acts as a barrier to diffusion of molecules. The
regard to application to full skin in vivo. A higher Pickering emulsion droplets may behave as an
concentration in the dermis correlates with the encapsulation system that delays the release of
larger permeation. drugs to the continuous phase. Experimental evi-
dence of sustained release has been given for par-
ticular o/w emulsions made of cross-linked
19.3.3 Mechanisms of Enhanced Skin polydimethylsiloxane coated with adsorbed silica
Absorption particles (Simovic and Prestidge 2007). Sustained
release of caffeine from w/o emulsion droplets to
The origin of the faster penetration has mainly a bulk aqueous phase has been indeed measured
been discussed by Frelichowska (2009c) for the by Frelichowska (2009c). Transfer of the drug
skin absorption of caffeine from w/o Pickering from Pickering emulsion droplets to the aqueous
emulsions. Several transport phenomena have phase was slowed down by a factor 1.5 with
been considered; experiments have been performed respect to the emulsifier-based emulsion.

Fig. 19.5  Skin absorption of caffeine from w/o emul- different emulsions. Right: Distribution of caffeine inside
sions: Pickering emulsion (black) and classical emulsion skin layers after 24 h exposure to different emulsions
(gray). Left: Permeation profile over 24 h exposure to (applied dose = 3,000  μg.cm−2)
19  Pickering Emulsions for Controlled Drug Delivery to the Skin 277

Permeation of the drug through the skin should a higher storage of retinol in the stratum cor-
have been slower accordingly; on the contrary, it neum. Indeed retinol probably adsorbs at the sur-
has been measured to be faster. face of silica particles in the lipidic medium of
The direct transfer of drug molecules from stratum corneum in the same way as fatty alco-
emulsion droplets to the skin surface appeared the hols do in nonpolar organic solvents. The larger
most relevant contribution to the accelerated per- storage of retinol in the stratum corneum after
meation of caffeine. Such direct transfer occurs exposure of the skin to the o/w Pickering emul-
when droplets come in contact with the skin sur- sion may result from such adsorption of the drug
face. Stronger adhesion of the w/o Pickering to silica particles that do not penetrate deeper. In
emulsion droplets to the skin has been disclosed case where silica particles penetrate the skin
from measurements of adhesion energy of water deeper, the transport of drug adsorbed to silica
drops to the skin surface immersed in an oil particles may occur. Such situation was claimed
medium. Thus, the adhesion energy was 3.3 J·m−2 by Ghouchi Eskandar et al. (2010) on the basis of
for drops covered with silica particles (“Pickering a correlation between concentration profiles of
drops”) emulsions against 0.27 J·m−2 for drops fluorescent penetrant molecule and silica
from emulsifier-coated drops (and 3.3 J·m−2 for nanoparticles measured from confocal fluores-
bare drops). Higher adhesion energy meant a lon- cence microscopy and SEM on histological
ger contact time of the water droplets to the skin transversal sections, respectively.
surface, and consequently a faster transfer of the Easier direct transfer from emulsion droplets
caffeine molecules contained in water droplets to to the skin may also be operative as in the case of
the skin. According to such mechanism of transfer w/o Pickering emulsions. Indeed, it should com-
during the contact of droplets with the skin sur- pensate the penetration-enhancing activity of the
face, emulsion droplets are not considered as car- surfactant in the classical emulsion taken as ref-
riers, and no penetration of emulsions droplets erence, so that the total amounts of absorbed reti-
into the skin is involved. It is believed that emul- nol measured for Pickering and classical
sion droplets cannot penetrate the skin as intact emulsions are the same. Although the mechanism
particles. Accelerated transfer of the drug from of improved transfer coming from a higher adhe-
the emulsion droplets to the skin overcompen- sion of emulsion droplets has not been studied for
sates the encapsulation-like sustained release of o/w emulsions, this phenomenon is likely operat-
caffeine into the continuous phase, as well as the ing in the same way as for w/o emulsions. This
possible penetration-­ enhancing effect of the holds for o/w Pickering emulsions loaded with
emulsifier contained in the classical emulsion, so drugs of medium polarity that are able to perme-
that the overall effect is an acceleration of the ate through the skin (Ghouchi Eskandar et al.
drug delivery by Pickering emulsions. 2010; Marku et al. 2012).
Penetration of silica particles into the skin The measurement of the adhesion energy of
gives a possible supplementary transport mecha- emulsion droplets to the skin surface can be per-
nism for the drug. Indeed, the polar caffeine mol- formed quite easily taking macroscopic drops as
ecule may adsorb at the surface of silica particles mimicking the emulsion droplets (Frelichowska
that penetrate the nonpolar medium of the stra- 2009c). The size of macroscopic drops is ~1,000
tum corneum. Penetration of silica particles is times larger than the size of emulsion droplets.
limited however as discussed in the next section. The adhesion energy of a droplet deposited on a
The depth of penetration of silica particles upon (skin) surface is
24 h exposure to a Pickering emulsion was 5 μm,
EAdhesion = g w − o (1 + cos q w ) (19.3)
that is, half of the thickness of the stratum cor-
neum. Therefore, the contribution of this phe-
nomenon is limited. where γw-o is the interfacial tension between oil
For the o/w emulsions, storage of silica parti- and water in the presence of either solid particles
cles in the stratum corneum may be the origin of or surfactant molecules and θw is the contact
278 Y. Chevalier et al.

angle of the drop of dispersed phase deposited on dioxide, and zinc oxide (Mavon et al. 2007;
the (flat and smooth) skin surface immersed in Nohynek at al. 2008; Bolzinger et al. 2011).
the continuous phase. Both γw-o and θw can be eas- There is a wealth of experimental data showing
ily measured with the standard laboratory equip- that such inorganic particles do not penetrate the
ment (drop shape analysis tensiometer). skin deeply and that their penetration is restricted
to the stratum corneum upon reasonable expo-
sure durations (hours to days). The same trend
19.3.4 Fate of the Stabilizing has been observed for silica particles used for the
Solid Particles stabilization of Pickering emulsions. Figure 19.6
shows scanning electron microscopy (SEM) pic-
The deep skin penetration of solid particles may tures of corneocytes collected by means of tape
be considered as an issue regarding possible stripping after 24 h exposure of the skin to a w/o
health concerns of nanoparticles. Such topic has Pickering emulsion. Silica particles are readily
been largely addressed concerning inorganic par- visible at the surface of corneocytes; their abun-
ticles used in sunscreen formulations, titanium dance decreases as a function of the skin depth

Fig. 19.6 SEM pictures of corneocytes peeled off the stratum corneum; no more particles are detected beyond
stratum corneum by the tape-stripping technique after the 10th strip corresponding to half the thickness of stra-
24 h exposure to Pickering emulsion. A less amount of tum corneum. From top to bottom: skin surface, 1st, 7th,
silica particles is observed upon going deeper into the 10th (2 pictures), 15th, and 19th strip
19  Pickering Emulsions for Controlled Drug Delivery to the Skin 279

and no more silica particles have been detected the control of the emulsion properties and skin
beyond the 10th strip corresponding to 5 μm skin absorption pertain to both the formulation
depth (half of the stratum corneum thickness). (choice of the ingredients and their concentra-
Ghouchi Eskandar et al. (2010) reached the tion) and the emulsification process. The
same conclusion from observations of histologi- physicochemical properties of the skin surface
cal sections of the skin. Silica particles appeared are such that Pickering emulsion droplets
as white areas in SEM pictures under back- show strong adhesion to the skin, thus allow-
scattered electron detection. The images revealed ing faster transfer of drugs to the skin. The
a uniformly white layer corresponding to the penetration-enhancing activity of Pickering
stratum corneum that contained a high amount of emulsions mainly relies on this adhesion phe-
silica, and several white spots dispersed at ran- nomenon. Other phenomena specific to
dom in the viable epidermis and dermis. The Pickering emulsions modulate skin absorp-
amount in the stratum corneum was obviously tion, even if they are not sensu stricto penetra-
much higher than the cumulated amounts in the tion enhancer actions: lower bioavailability
deeper layers of skin. Unfortunately, the quanti- due to sustained release and penetration of
tative analysis of silicon by EDX has been aver- solid particles inside the stratum corneum
aged over the whole histological section, so that a causing immobilization of adsorbed drug
quantitative assessment of the fractions of silica molecules.
in the stratum corneum and viable layers of skin Manufacture of Pickering emulsions can be
was not available. Their measurements are par- easily implemented since the replacement of
ticularly interesting because in their studies drug classical emulsifiers by solid particles is quite
penetration was favored by other ingredients of a direct substitution, and fabrication process is
the formulation that acted as penetration enhanc- the same as for conventional emulsions.
ers. The stabilizing layer was made of mixed sur-
factants and solid particles, so that the solid
particles might have penetrated easier because of
the penetration-enhancing properties of the sur- References
factant, in particular oleylamine, which acts as a
penetration enhancer. Abend S, Lagaly G (2001) Bentonites and double hydrox-
ides as emulsifying agents. Clay Miner 36:557–570
Alternatives to inorganic nanoparticles are Abend S, Bonnke N, Gutschner U, Lagaly G (1998)
biodegradable organic particles. Biodegradable Stabilization of emulsions by heterocoagulation of
Pickering emulsions are attractive for skin deliv- clay minerals and layered double hydroxides. Colloid
ery applications, as well as for other fields such Polym Sci 276:730–737
Akartuna I, Tervoort E, Studart AR, Gauckler LJ (2009)
as food applications, development of emulsifier-­ General route for the assembly of functional inorganic
free environmentally sustainable formulations, capsules. Langmuir 25:12419–12424
etc. Several types of such particles can stabilize Arditty S, Whitby CP, Binks BP, Schmitt V, Leal-Calderon
Pickering emulsions of edible or biodegradable F (2003) Some general features of limited coalescence
in solid-stabilized emulsions. Eur Phys J E Soft Matter
oils: fat crystals (Rousseau 2000), cellulose 11:273–281
(Kalashnikova et al. 2011, 2012), chitin Arditty S, Schmitt V, Giermanska-Kahn J, Leal-Calderon
(Tzoumakia et al. 2011), or block copolymer F (2004) Materials based on solid-stabilized emulsion.
micelles (Laredj-Bourezg et al. 2012). J Colloid Interface Sci 275:659–664
Arditty S, Schmitt V, Lequeux F, Leal-Calderon F (2005)
Interfacial properties in solid-stabilized emulsions.
Conclusions Eur Phys J E 44:381–393
Solid particles can be advantageously used as Avendaño Juárez J, Whitby CP (2012) Oil-in-water
emulsifiers for the stabilization of Pickering Pickering emulsion destabilisation at low particle con-
centrations. J Colloid Interface Sci 368:319–325
emulsions. They also influence the skin Aveyard R, Binks BP, Clint JH (2003) Emulsions stabi-
absorption of drugs loaded inside the droplets lized solely by solid colloidal particles. Adv Colloid
of dispersed phase. The parameters allowing Interf Sci 100–102:503–546
280 Y. Chevalier et al.

Barthel H (1995) Surface interactions of dimethylsiloxy Gelot A, Friesen W, Hamza HA (1984) Emulsification of
group-modified fumed silica. Colloids Surf A 101: oil and water in the presence of finely divided solids
217–226 and surface-active agents. Colloids Surf 12:271–303
Barthel H, Binks BP, Dyab A, Fletcher P (2003) Multiple Ghouchi Eskandar N, Simovic S, Prestidge CA (2007)
emulsions. Patent EP1350556 B1, US2003/0175317 A1 Synergistic effect of silica nanoparticles and charged
Binks BP (2002) Particles as surfactants – similarities and surfactants in the formation and stability of submicron
differences. Curr Opin Colloid Interface Sci 7:21–41 oil-in-water emulsions. Phys Chem Chem Phys 9:
Binks BP, Horozov TS (eds) (2006) Colloidal particles at liq- 6426–6434
uid interfaces. Cambridge University Press, Cambridge Ghouchi Eskandar N, Simovic S, Prestidge CA (2009)
Binks BP, Kirkland M (2002) Interfacial structure of solid- Nanoparticle coated submicron emulsions: sustained
stabilised emulsions studied by scanning electron in-vitro release and improved dermal delivery of all-
microscopy. Phys Chem Chem Phys 4:3727–3733 trans-retinol. Pharm Res 26:1764–1775
Binks BP, Lumsdon SO (2000a) Influence of particle wet- Ghouchi Eskandar N, Simovic S, Prestidge CA (2010)
tability on the type and stability of surfactant-free Mechanistic insight into the dermal delivery from
emulsions. Langmuir 16:8622–8631 nanoparticle-coated submicron o/w emulsions. J Pharm
Binks BP, Lumsdon SO (2000b) Catastrophic phase inver- Sci 99:890–904
sion of water-in-oil emulsions stabilized by hydropho- Ghouchi Eskandar N, Simovic S, Prestidge CA (2011)
bic silica. Langmuir 16:2539–2547 Interactions of hydrophilic silica nanoparticles and
Binks BP, Lumsdon SO (2000c) Effects of oil type and classical surfactants at non-polar oil-water interface.
aqueous phase composition on oil-water mixtures con- J Colloid Interface Sci 358:217–225
taining particles of intermediate hydrophobicity. Phys Hassander H, Johansson B, Törnell B (1989) The mecha-
Chem Chem Phys 2:2959–2967 nism of emulsion stabilization by small silica (Ludox)
Binks BP, Rodrigues JA (2003) Types of phase inversion particles. Colloids Surf 40:93–105
of silica particle stabilized emulsions containing tri- Horozov TS, Binks BP (2006) Particle-stabilized emul-
glyceride oil. Langmuir 19:4905–4912 sions: a bilayer or a bridging monolayer? Angew
Binks BP, Whitby CP (2004) Silica particle-stabilized Chem Int Ed 45:773–776
emulsions of silicone oil and water: aspects of emulsi- Horozov TS, Binks BP, Gottschalk-Gaudig T (2007)
fication. Langmuir 20:1130–1137 Effect of electrolyte in silicone oil-in-water emulsions
Bolzinger M-A, Briançon S, Chevalier Y (2011) stabilised by fumed silica particles. Phys Chem Chem
Nanoparticles through the skin: managing conflicting Phys 9:6398–6404
results of inorganic and organic particles in cosmetics Kalashnikova I, Bizot H, Cathala B, Capron I (2011) New
and pharmaceutics. WIREs Nanomed Nanobiotechnol Pickering emulsions stabilized by bacterial cellulose
3:463–478 nanocrystals. Langmuir 27:7471–7479
Brinon L, Geiger S, Alard V, Tranchant J-F, Pouget T, Kalashnikova I, Bizot H, Cathala B, Capron I (2012)
Couarraze G (1998) Influence of lamellar liquid crys- Modulation of cellulose nanocrystals amphiphilic
tal structure on percutaneous diffusion of a hydro- properties to stabilize oil/water interface.
philic tracer from emulsions. J Cosmet Sci 49:1–11 Biomacromolecules 13:267–275
Destribats M, Ravaine S, Heroguez V, Leal-Calderon F, Kruglyakov PM (2000) Hydrophile-Lipophile Balance of
Schmitt V (2010) Outstanding stability of poorly-­ surfactants and solid particles. Physicochemical
protected Pickering emulsions. Progr Colloid Polym aspects and applications. Elsevier, Amsterdam
Sci 137:13–18 Kruglyakov PM, Nushtayeva AV (2004) Phase inversion
Dong L, Johnson D (2003) Surface tension of charge-­ in emulsions stabilised by solid particles. Adv Colloid
stabilized colloidal suspensions at the water-air inter- Interf Sci 108–109:151–158
face. Langmuir 19:10205–10209 Laredj-Bourezg F, Chevalier Y, Boyron O, Bolzinger M-A
Drelich A, Gomez F, Clausse D, Pezron I (2010) Evolution (2012) Emulsions stabilized with solid organic parti-
of water-in-oil emulsions stabilized with solid parti- cles. Colloids Surf A 413:252–259
cles. Influence of added emulsifier. Colloids Surf A Marku D, Wahlgren M, Rayner M, Sjöö M, Timgren A
365:171–177 (2012) Characterization of starch Pickering emulsions for
Frelichowska J, Bolzinger M-A, Chevalier Y (2009a) potential applications in topical formulations. Int J Pharm
Pickering emulsions with bare silica. Colloids Surf A 428:1–7
343:70–74 Mavon A, Miquel C, Lejeune O, Payre B, Moretto P
Frelichowska J, Bolzinger M-A, Pelletier J, Valour J-P, (2007) In vitro percutaneous absorption and in vivo
Chevalier Y (2009b) Topical delivery of lipophilic drugs stratum corneum distribution of an organic and a
from o/w Pickering emulsions. Int J Pharm 371:56–63 mineral sunscreen. Skin Pharmacol Physiol 20:​
Frelichowska J, Bolzinger M-A, Valour J-P, Mouaziz H, 10–20
Pelletier J, Chevalier Y (2009c) Pickering w/o emul- Midmore BR (1998a) Synergy between silica and poly-
sions: drug release and topical delivery. Int J Pharm oxyethylene surfactants in the formation of O/W
368:7–15 emulsions. Colloids Surf A 145:133–143
Frelichowska J, Bolzinger M-A, Chevalier Y (2010) Effects Midmore BR (1998b) Preparation of a novel silica-
of solid particle content on properties of o/w Pickering stabilized oil/water emulsion. Colloids Surf A 132:
emulsions. J Colloid Interface Sci 351:348–356 257–265
19  Pickering Emulsions for Controlled Drug Delivery to the Skin 281

Midmore BR (1999) Effect of aqueous phase composition Thieme J, Abend S, Lagaly G (1999) Aggregation in
on the properties of a silica-stabilized W/O emulsion. Pickering emulsions. Colloid Polym Sci 277:
J Colloid Interface Sci 213:352–359 257–260
Montenegro L, Carbone C, Paolino D, Drago R, Touitou E, Meidan VM, Horwitz E (1998) Methods for
Stancampiano AH, Puglisi G (2008) In vitro skin per- quantitative determination of drug localized in the
meation of sunscreen agents from O/W emulsions. Int skin. J Control Release 56:7–21
J Cosmet Sci 30:57–65 Tzoumakia MV, Moschakis T, Kiosseoglou V, Biliaderis
Neuhäusler U, Abend S, Jacobsen C, Lagaly G (1999) CG (2011) Oil-in-water emulsions stabilized by chi-
Soft X-ray spectromicroscopy on solid-stabilized tin nanocrystal particles. Food Hydrocoll 25:
emulsions. Colloid Polym Sci 277:719–726 1521–1529
Nohynek GJ, Dufour EK, Roberts MS (2008) Vignati E, Piazza R, Lockhart TP (2003) Pickering emul-
Nanotechnology, cosmetics and the skin: is there a sions: interfacial tension, colloidal layer morphology, and
health risk? Skin Pharmacol Physiol 21:136–149 trapped-particle motion. Langmuir 19:6650–6656
Otto A, du Plessis J, Wiechers JW (2009) Formulation Wang H, Hobbie EK (2003) Amphiphobic carbon nano-
effects of topical emulsions on transdermal and dermal tubes as macroemulsion surfactants. Langmuir 19:​
delivery. Int J Cosmet Sci 31:1–19 3091–3093
Pickering SU (1907) Emulsions. J Chem Soc 91:2001–2021 Whitby CP, Fischer FE, Fornasiero D, Ralston J (2011)
Ramsden W (1903) Separation of solids in the surface-­ Shear-induced coalescence of oil-in-water Pickering
layers of solutions and ‘suspensions’. Proc Roy Soc emulsions. J Colloid Interface Sci 361:170–177
Lond 72:156–164 Wiley RM (1954) Limited coalescence of oil droplets in
Rousseau D (2000) Fat crystals and emulsion stability – a coarse oil-in-water emulsions. J Colloid Sci 9:​
review. Food Res Int 33:3–14 427–437
Simovic S, Prestidge CA (2007) Nanoparticle layers con- Zhai X, Efrima S (1996) Chemical and physical aspects of
trolling drug release from emulsions. Eur J Pharm macroemulsions stabilized by interfacial colloids. J Phys
Biopharm 67:39–47 Chem 100:11019–11028
Part VI
Gels as Vehicles for Skin Delivery
Hydrogel Vehicles for Hydrophilic
Compounds 20
Teresa Cerchiara, Federica Bigucci,
and Barbara Luppi

Contents 20.1 Introduction


20.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . 285
Hydrogels date back to 1960 when Wichterle and
20.1.1 Physical and Chemical Properties
of Hydrogels . . . . . . . . . . . . . . . . . . . . . . . 286
Lim first proposed the use of hydrophilic net-
works of poly(2-hydroxyethylmethacrylate)
20.2 Applications of Hydrogels
(PHEMA) in contact lenses (Wichterle and Lim
in Transdermal Drug Delivery . . . . . . . . 289
1960). Since then, the use of hydrogels has
20.2.1 Semisolid Vehicles. . . . . . . . . . . . . . . . . . . 291 extended to various biomedical (Hoffman 2002;
20.2.2 Film-Based Vehicles . . . . . . . . . . . . . . . . . 292 Peppas et al. 2006; Kopeceka 2007) and pharma-
Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295 ceutical (Peppas et al. 2000) applications. In par-
ticular, due to their physical properties, similar to
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295
those of human tissues (water content, soft and
pliable consistence), hydrogels have been used
for different administration routes such as oral,
rectal, ocular, epidermal, and subcutaneous
(Peppas et al. 2000; Guy 1996; Jatav et al. 2011).
Hydrogels are composed of hydrophilic mac-
romolecules forming three-dimensional insolu-
ble networks able to imbibe large amounts of
water or biological fluids (Peppas and Mikos
1986). Commonly, the polymers utilized to make
hydrogels are insoluble due to the presence of
permanent or reversible cross-links (Berger et al.
2004). Permanent cross-linked hydrogels
(Wichterle and Lim 1960; Xiao and Zhou 2003;
Brasch and Burchard 1996) are characterized by
T. Cerchiara (*) • F. Bigucci • B. Luppi covalent bonds forming tie points or junctions,
Department of Pharmacy and Biotechnology-FaBiT, whereas reversible cross-linked hydrogels
Bologna University, Via San Donato 19/2,
(Watanabe et al. 1996; Wang et al. 1999; Qu et al.
Bologna 40127, Italy
e-mail: teresa.cerchiara2@unibo.it; 1999) present ionic, hydrophobic, or coiled-coil
federica.bigucci@unibo.it; barbara.luppi@unibo.it physical interactions. These kinds of cross-links

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 285


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_20, © Springer-Verlag Berlin Heidelberg 2015
286 T. Cerchiara et al.

in the polymer structure yield insoluble materials important. In fact, investigators have focused on
able to swell in aqueous environments retaining a controlling degradation behavior of hydrogels to
significant fraction of water in their structure, up design polymers able to be cleared from the body
to thousands of times their dry weight in water. once they complete their roles (Anderson and
Hydrogels can be divided into homopolymer or Shive 1997; Timmer et al. 2002): for this reason
copolymers based on the preparative method, but labile bonds are frequently introduced in the gels.
they can also be natural polymers, synthetic poly- These bonds can be present either in the polymer
mers, or derivatives. In nature hydrogels can be backbone or in the cross-links used to prepare the
found in plants (pectin, pullulan), various species gel. The labile bonds can be broken under physi-
of brown seaweed (alginic acid, agar, carrageenan), ological conditions either enzymatically or
crustaceans (chitin), and animal tissue (hyaluronic chemically, in most cases by hydrolysis (Damink
acid, collagen, fibrin). Typical simple synthetic et al. 1996; Eliaz and Kost 2000; Lee et al. 2004).
materials applied for general-purpose hydrogels
are poly(ethylene oxide), poly(vinyl alcohol),
poly(vinyl pyrrolidone), poly(hydroxyethyl meth- 20.1.1 Physical and Chemical
acrylate), and poly(N-isopropyl acrylamide). Properties of Hydrogels
Moreover, the synthetic pathway offers more pos-
sibilities to create hydrogels with modified func- An important property of hydrogels is their swell-
tional properties. In fact, several physiologically ing behavior: it depends upon the polymer, extent
responsive hydrogels are obtained from chemical of cross-linking, temperature, polymer-solvent
or physical modifications of natural and synthetic interactions, and extent of ionization (Khare et al.
polymers and tested for use in the so-called intel- 1992). In particular the extent of cross-linking
ligent biomaterials (Hoffman 1991; Miyata et al. can be changed to achieve a relatively strong and
2002; Murdan 2003; Chen et al. 2004) because yet elastic hydrogel. Long-chain cross-linkers
they are capable of reacting to various environmen- and low cross-linking ratios (the ratio of moles of
tal stimuli (temperature, pH, ionic strength, solute cross-linking agent to the moles of polymer
concentration, electric radiation, light, sound, etc.). repeating units) produce extremely weak hydro-
Hydrogels can be homogeneous, when the gels, while short-chain cross-linkers and high
pores between polymer chains are the only spaces cross-linking ratios produce extremely tight
available for mass transfer and the pore size is hydrogels. Tightly cross-linked hydrogels will
within the range of molecular dimensions (a few swell less than the same hydrogels with low
nanometers or less) or porous when the effective cross-linking ratios or long cross-linkers chains.
pore size is over 10 nm. In homogeneous hydro- The presence of hydrophilic or hydrophobic
gels the transfer of water or other solutes is groups in the chemical structure of the polymer
achieved by a pure diffusional mechanism, which affects the swelling behavior of hydrogels. When a
restricts the rate of absorption and to some extent dry hydrogel begins to absorb water, the first water
the size of species that are absorbed. molecules entering the matrix will hydrate polar
Porous hydrogels can be made by different hydrophilic groups. As the polar groups are
polymerization methods in the presence of dis- hydrated, the network swells exposing hydropho-
persed porosigens (ice crystals, oil, sucrose crystals) bic groups which also interact with water mole-
which can be removed later to leave an intercon- cules, leading to hydrophobically bound water.
nected meshwork, where the pore size depends on Finally an equilibrium swelling level is reached
the size of the porosigens (Hickey and Peppas when the network imbibes additional water (“free
1995). The introduction of a porosigen reduces water”) which fills the space between the polymer
mechanical strength significantly making porous chains. As hydrophobic groups minimize their
hydrogels weaker than homogeneous hydrogels. exposure to the water molecule, hydrogels con-
In medical, engineering, and pharmaceutical taining hydrophobic groups will swell much less
technology, hydrogel degradation is considerable than hydrogels containing hydrophilic groups.
20 Hydrogel Vehicles for Hydrophilic Compounds 287

As stated earlier, the dissolution of polymer (ethylene glycol dimethacrylate) in the presence
chains and consequently hydrogel swelling abil- of AIBN (2,2′-azo-bis-isobutyronitrile), the radi-
ity is prevented by the presence of cross-linking cal initiator (Fig. 20.1). Chemical cross-linking
in the three-dimensional network. Different agents, such as acyl dichlorides (Fig. 20.2),
chemical and physical cross-linking meth- can establish covalent linkages with functional
ods have been employed to prepare hydrogels groups of polymers, such as activated hydrox-
(Hennink and van Nostrum 2002). In chemi- ylic groups of poly(vinyl alcohol) (Orienti et al.
cally and physically cross-linked gels, dissolu- 2000). Polyaldehydes are utilized to cross-link
tion is prevented by covalent bonds and physical proteins such as albumin (Sahin et al. 2002) and
interactions between different polymer chains, gelatin (Draye et al. 1998) or natural polysaccha-
respectively. Chemically cross-linked gels can rides such as hyaluronic acid (Luo et al. 2000).
be obtained by radical polymerization of low However, a significant disadvantage of chemical
molecular weight monomers in the presence of cross-linking agents is their toxicity. Among vari-
cross-linking agents, chemical reaction of com- ous methods applied for the production of hydro-
plementary groups, and high-energy irradiation. gels, the radiation technique (Safrany 1997) is a
Physically cross-linked gels can be obtained simple, efficient, clean, and environment-friendly
by ionic interactions, hydrogen bonds, crystal- process (Fig. 20.3). Hydrogels can be obtained by
lization, and aggregation of the hydrophobic radiation technique in a few ways, including irra-
segments of multiblock copolymers or graft diation of solid polymer (Nedkov and Tsvetkova
copolymers. An example of cross-linking by 1994), monomer (in bulk or in solution) (Rosiak
radical polymerization is the synthesis of hydro- 1991), or aqueous solution of polymer (Kabanov
gels of Wichterle and Lim (1960), a copolymer- 1998). For irradiation technologies, the main
ization of HEMA with the cross-linker EGDMA irradiating sources include gamma rays from

CH3 CH3 CH3 CH3


AIBN
CH + CH CH2 CH2 n
90°C
CO CO CH CO

O O O O

CH2 CH2 CH2 CH2

CH2 CH2 CH2 CH2

OH O OH O

HEMA CO CO CH3

CH CH2 CH2
n
CH3 CH3 CO

EGDMA O
P(HEMA-co-EGDMA) CH2

CH2

OH

Fig. 20.1 Schematic representation of radical polymerization. Hydrogels are formed by the copolymerization of
HEMA with EGDMA using AIBN as the radical initiator
288 T. Cerchiara et al.

CH2 CH + Cl – CO CH2 CO-Cl CH2 CH


n
x x

OH O

CO
n = 2 SUCCINYL
n = 4 ADIPOYL
n = 8 SEBACOYL
CH2
n

CO

CH CH2
y

Fig. 20.2 Preparation of PVA hydrogels cross-linked by acyl dichlorides

radioactive isotopes such as cobalt 60, electron due to the formation of hydrogen bonds between
beams from electron accelerators, and X-rays the oxygen of poly(ethylene glycol) and the car-
converted from electron beams. boxylic groups of the acrylic polymers (Eagland
Physical cross-linking of hydrogels also avoids et al. 1994). Another physical method for produc-
the use of chemical cross-linking agents. Such ing physically cross-linked hydrogels is the for-
agents can potentially inactivate the active prin- mation of crystalline regions in the polymer
ciple and covalently link it to the hydrogel net- network, obtained by casting dilute, aqueous
work. Examples of ionically cross-linked alginate solutions of poly(vinyl alcohol), then cooling to
hydrogels have been reported (Grant et al. 1973). −20 °C, and thawing back to room temperature
Alginate is a family of linear polysaccharides several times (Stauffer and Peppas 1992). These
composed of mannuronic acid (MA) and gulu- freeze/thawed gels have demonstrated enhanced
ronic acid (GA). The chemical composition and physical properties, such as high mechanical
sequence of MA and GA residues depend on the strength and high elasticity, that make them suit-
source from which the alginate has been extracted. able for biomedical applications.
The gelation of alginate is mainly achieved by the Finally, physically cross-linked hydrogels can
exchange of sodium ions with divalent cations be obtained by hydrophobic modification of
such as Ca2+, Cu2+, Zn2+, or Mn2+, which can form polymers and in particular of polysaccharides
cation bridges between adjacent molecules. The such as chitosan, dextran, pullulan, and carboxy-
“egg-box” model of Grant et al. (1973) is gener- methyl curdlan (Noble et al. 1999; Sludden et al.
ally taken into consideration to explain the forma- 2000; Cerchiara et al. 2002). Glycol chitosan
tion of a rodlike cross-linked complex due to the substituted with palmitoyl chains is an example
bounding of the divalent cations in the interchain of a hydrophobized polysaccharide. The attach-
cavities. Some polymeric complexes can be held ment of hydrophobic groups to glycol chitosan
together by hydrogen bonds: poly(acrylic acid) yields an amphiphilic polymer capable of self-
and poly(methacrylic acid) provide physically assembly into vesicles (Uchegbu et al. 1998).
cross-linked hydrogels with poly(ethylene glycol) Non-covalent cross-linking is achieved by the
20 Hydrogel Vehicles for Hydrophilic Compounds 289

CH2 CH2 CH2 O CH2 CH2 O CH2 CH2 O CH2

High radiation energy

CH2 CH2 CH2 O CH2 CH O CH2 CH2 O CH2

Crosslinking

Fig. 20.3 Schematic representation of the radiation method to design hydrogels

hydrophobic interactions of the palmitoyl groups due to the interaction of the different acyl chains
and a gel matrix is formed. Finally, our research with the stratum corneum.
group (Cerchiara et al. 2002) reported physically
cross-linked chitosan hydrogels with lauric,
myristic, palmitic, or stearic acid prepared by 20.2 Applications of Hydrogels
freeze drying and studied for transdermal use in Transdermal Drug
(Fig. 20.4). These polymers produce hydrogels Delivery
with different functional properties related to the
different acyl chains introduced in the polymer Transdermal delivery is an attractive and promising
structure. In particular, the permeation of hydro- alternative compared to conventional administra-
philic substances through the skin can be modu- tion routes (e.g., oral and injectable) for transport
lated by increased or decreased drug solubility through the skin into the blood circulation of drugs
290 T. Cerchiara et al.

OH OH
O
O
HO H H
H
NH2 OH
OH O
H
O O
HO H
H
NH
H CO
O

CH2OH CH2OH
O O
OH O OH O n=10 lauroyl chain
n=12 miristoyl chain
n=14 palmitoyl chain
NH2 n=16 stearoyl chain
NH
CO
CH2 n

CH3

O
CO H
HN
H
H OH
O O
O H
HO OH H2N
H H
H OH
O
O
HO OH

Fig. 20.4 Structural representation of physically cross-linked chitosan hydrogels


20 Hydrogel Vehicles for Hydrophilic Compounds 291

such as hydrophobic small molecules, hydrophilic in stratum corneum leading to a less organized
molecules, and macromolecules. In fact, this route structure of this protein and enhancing transder-
offers several advantages over conventional routes mal permeation of drugs.
(Peppas et al. 2000; Brown et al. 2006; Prausnitz As regards the transdermal drug delivery sys-
and Langer 2008): tems, the traditional formulations as plasters,
• Is noninvasive and pain-free, with no trauma cream, and ointments have been replaced in order
or risk of infection (Denet et al. 2004), thus to minimize skin irritation, promote adhesion
resulting in an enhanced patient compliance properties, guarantee dosage flexibility, enhance
• Maintains constant drug levels in the blood, patient acceptability, and improve ease of use. In
eliminating plasma peaks and valleys associ- this context, particular attention has been paid to
ated to oral and injectable administration the formulations based on hydrogels such as
• Avoids gastrointestinal tract and circumvents semisolid systems and film-based systems
hepatic first-pass metabolism, thus resulting (matrix-type systems, membrane-coated sys-
in a lower drug amount administered and in a tems, film-forming solution).
reduction of systemic side effects
• Is inexpensive
In spite of the advantages, major disadvantage 20.2.1 Semisolid Vehicles
of transdermal drug delivery is that the drug itself
or the materials used to fabricate the vehicles Hydrogels are three-dimensional networks based
may sometimes induce an irritation or sensitiza- on linear hydrophilic polymers that are able to
tion reaction of the skin (Kurihara-Bergstrom absorb large amounts of water, remaining insolu-
et al. 1991; Murphy and Carmichael 2000; Ale ble due to the presence of chemical or physical
et al. 2009; Wohlrab et al. 2011). Moreover, few cross-links (Peppas et al. 2000). The relatively
molecules have been successfully delivered high water content makes them a good alternative
transdermally, mainly due to the stratum cor- to other dosage forms such as creams, ointments,
neum that forms a barrier to the permeation of and patches, enhancing skin moisturization and
hydrophilic drugs, especially macromolecules elasticity and providing a better feel when applied
such as proteins, peptides, and vaccines. to the skin. The most important and well-known
Consequently, research trends are focusing on polymers used for preparing these hydrogels are
approaches to overcome the barrier presented by biopolymers such as polysaccharides (starch, cel-
the skin, including physical penetration enhancers lulose, chitin, alginate, hyaluronate) or proteins
such as microneedles (Henry et al. 1998), ionto- (collagens, gelatins, caseins, albumins) and syn-
phoresis and electroosmosis (Pikal 2001), electro- thetic polymers such as polyvinyl alcohol, poly-
poration (Hu et al. 2000), radiofrequency energy vinylpyrrolidone, polyethylene glycol, and
(Sintov et al. 2003), and chemical penetration polyacrylates.
enhancers such as sulfoxides, alkanones, alco- Chitosan, a polysaccharide comprising copo-
hols, polyols, amides, fatty acids, fatty acid esters, lymer of glucosamine and N-acetylglucosamine,
surfactants, terpenes, organic acids, and cyclodex- derived by the partial deacetylation of chitin, is a
trins (Thong et al. 2007). Recent studies have nontoxic and bioabsorbable polymer (Muzzarelli
introduced a new category of transdermal penetra- et al. 1988; Luppi et al. 2010a) extensively stud-
tion enhancers such as positively charged poly- ied for the release of many drugs. Our research
mers. In fact, Taveira et al. (2009) described that group (Cerchiara et al. 2002) described physically
chitosan interacts with negative charges in the cross-linked chitosan hydrogels with lauric,
skin improving drug penetration in the deeper lay- myristic, palmitic, or stearic acid able to enhance
ers, while He et al. (2008, 2009) demonstrated the skin permeation of propranolol hydrochloride
that chitosan and its derivatives such as N-trimethyl selected as a hydrophilic model drug. The aim of
chitosan and mono-N-carboxymethyl chitosan are the work was to improve the permeation of drugs
able to change the secondary structure of keratin through biological membranes, using hydrogels
292 T. Cerchiara et al.

made of amphiphilic polymer (Noble et al. 1999). accelerating drug permeation through the skin. In
The concomitant presence of hydrophobic and fact, flux values (mg/h cm2) of pyridoxine hydro-
hydrophilic groups in the polymer influenced the chloride from hydrogels through porcine skin
swelling properties. So, at pH 7.4 all hydrogels were 1.29 ± 0.12 for GZ, 5.83 ± 0.22 for GZ-ET,
swelled slowly and their behavior influenced the 4.91 ± 0.14 for GZ-BUT, 4.32 ± 0.10 GZ-EX,
drug release. Among the different chitosan gels, 3.82 ± 0.09 GZ-OCT, 3.25 ± 0.11 GZ-DEC, and
chitosan laurate and chitosan myristate enhanced 2.42 ± 0.12 GZ-DOD.
drug permeation through the skin with respect to
chitosan palmitate and chitosan stearate hydro-
gels (flux values (mg/h cm2) of propranolol 20.2.2 Film-Based Vehicles
hydrochloride from physically cross-linked chito-
sans oversaturated hydrogels through porcine Film-based vehicles are the widest utilized and
skin were 1.00 ± 0.02 for chitosan laurate, studied transdermal delivery systems based on
0.87 ± 0.05 for chitosan myristate, 0.47 ± 0.03 for hydrogels. They are generally distinguished in
chitosan palmitate, and 0.37 ± 0.01 for chitosan matrix-type systems, membrane-coated systems,
stearate). This could be explained by the interac- and film-forming solution.
tion of the hydrogels with the stratum corneum, The matrix-type systems are essentially a
increasing the solubility of the drug in the skin. polymeric layer containing the drug, eventually
Another example of hydrophilic and biocom- added with an adhesive layer to enhance the
patible polymer used to design hydrogels able to bioadhesion and an impermeable layer to force
release hydrophilic drugs through the skin is the release to the skin. In absence of the adhe-
polyvinyl alcohol. Polyvinyl alcohol cross-linked sive layer and in order to assure a controlled
with succinyl, adipoyl, or sebacoyl chloride was delivery of the drug, it is necessary to choose
employed as a supporting material to release pro- polymers able to guarantee an intimate and pro-
pranolol hydrochloride. In particular, these longed contact with the skin after the applica-
hydrogels increased the transdermal permeation tion and flexibility and elasticity sufficient to
of drug, and as described in the previous work follow the movements of the skin. An adhesive
(Cerchiara et al. 2002), this effect seems to be hydrogel patch based on a hydrophilic matrix
linked to an increased drug solubility in the skin, of poly(N-vinylpyrrolidone) and oligomeric
probably produced by the interaction of the poly- short-chain poly(ethylene glycol) was reported
mer with the stratum corneum. Moreover, the by Feldstein and coworkers (1996). They
maximum enhancement of the drug permeation observed that the delivery rates of drugs with
has been observed in the presence of the higher various chemical structures (propranolol, glyc-
degree of cross-linking and the shorter length of eryl trinitrate, isosorbide dinitrate) from the
the cross-linker acyl chain (Orienti et al. 2000). hydrophilic transdermal systems were higher
Luppi and coworkers (2003) used also the than from the hydrophobic ones (stirene-
cross-linked poly(methyl vinyl ether-co-maleic butadiene rubber/mineral oil, polydimethylsi-
anhydride) (GZ) as a topical vehicles for pyri- loxane/ silicone oil, polyisobutylene/mineral
doxine hydrochloride, selected as a hydro- oil), and the drug delivery from the hydrophilic
philic model drug. In particular, poly(methyl matrix across human cadaver skin epidermis or
vinyl ether-co-maleic anhydride) was cross- skin-imitating Carbosil membrane in vitro was
linked with ethylene glycol (GZ-ET), butane- characterized by zero-order drug delivery
diol (GZ-BUT), 1,6-exandiol (GZ-EX), kinetics up to the point of 75–85 % drug release
1,8-octanediol (GZ-OCT), 1,10-decanediol from initial contents in matrix. Drug delivery
(GZ-DEC), or 1,12-dodecanediol (GZ-DOD). rates from the hydrophilic matrix were con-
In vitro permeation studies were influenced by trolled by the skin or skin-imitating membrane
the nature of the cross-linker: the decrease in permeability and may be described by Fick’s
cross-linker acyl chain length provides vehicles law (Iordanskii et al. 2000).
20 Hydrogel Vehicles for Hydrophilic Compounds 293

Ethylene-vinyl acetate (EVA) matrix was tested context, films based on polyelectrolyte complexes
as a system for transdermal delivery of atenolol in were developed for topical and transdermal admin-
the presence of plasticizers able to increase the istration of drugs by Silva et al. (2008). The com-
rate of drug release. The effects of drug concentra- plexes were prepared with chitosan and different
tion, temperature, and plasticizers on drug release polyacrylic acid polymers, cross-linked with allyl
were investigated. The release rate from EVA pentaerythritol (Carbopol 71G NF®) or divinylg-
matrix was enhanced increasing temperature and lycol (Noveon AA-1®) at different cross-linking
drug concentration. In particular, the release rate densities. The interaction between the polymers
of drug increased about 1.72-fold when the tem- was maximized controlling the preparative condi-
perature of release system was raised from 32 to tions of complexes, and the film properties were
42 °C and about 1.24-fold when the drug loading improved by means of different plasticizers (glyc-
dose was increased from 0.5 to 1.5 %. Moreover, erol or polyethylene glycol 200), a moisturizing
drug release from the polymeric matrix followed a agent (Hydrovance®), and an hydrophilic pressure
diffusion-controlled model, where the quantity sensitive adhesive (polyvinylpyrrolidone/polyeth-
released per unit area was proportional to the ylene glycol 400). Between the different vehicles
square root of time. Among the plasticizers used obtained, the film prepared by cross-linking with
such as alkyl citrates and phthalates, tributyl citrate Noveon, plasticized with glycerol and covered
showed the best enhancing effects: flux values (μg/ with adhesive, has shown very good flexibility,
cm2/h1/2) were 8.056 and 5.327 for EVA matrix resistance, and bioadhesion, making it a good can-
containing tributyl citrate and EVA matrix without didate for further incorporation of drugs for topical
plasticizer, respectively. The results obtained con- and transdermal administration. Recently, chito-
firmed that ethylene-vinyl acetate matrix could be san-polyvinyl alcohol blend reticulated with glu-
used for transdermal delivery of hydrophilic drug taraldehyde has been utilized to prepare transdermal
(Kim and Shin 2004). film suitable for insulin release in diabetes chemo-
Padula and coworkers (2003) proposed a poly- therapy (Zu et al. 2012). The hydrogel obtained
vinyl alcohol film not adhesive in the dry state, but had a honeycomb-like structure and showed good
bioadhesive when applied on wet skin. The film mechanical and thermal properties. Moreover, the
was applied to the skin in the presence of a certain in vitro release studies showed that insulin release
amount of water. Water swelled the film on the is comply with Fick’s first law of diffusion show-
surface in contact with the skin, transforming a ing a high permeation rate (4.421 μg/(cm2h)).
dry polymeric matrix into a jellified polymer Transdermal delivery represents also an
layer. This particular film is flexible, is mechani- important opportunity for vaccine administration
cally resistant, and can avoid skin occlusion (Prausnitz and Langer 2008). In fact, although
because of its permeability to water vapor. vaccines are generally large entities such as mac-
Compared to a typical patch, the bioadhesive film romolecules or viral particles, their small dose
has a monolayer structure which includes back- facilitates transdermal administration. Ishii et al.
ing, adhesive, and drug reservoir functions. This (2008) describes simple, easy-to-use, noninva-
simple delivery system composed of a smaller sive transcutaneous vaccination system, formed
number of layers simplifies the preparation by an adhesive matrix. This patch is composed of
procedure of transdermal patches and represents a cross-linked acrylic medical adhesive, octyldo-
great innovation in the field of transdermal patch. decyl lactate, glycerin, and sodium hyaluronate
Another polymer that represents a good candi- and delivers antigenic proteins to Langerhans
date as a starting material for film-based vehicles cells resident in the epidermal layer without
because of its good film-forming properties is chi- destroying or removing the stratum corneum and
tosan (Mengatto et al. 2012). Frequently, chitosan induces Th2 (Type 2 helper T cells)-dominant
is modified by cross-linking reactions to achieve immune response (production of neutralizing
films with improved mechanical properties and immunoglobulin G1 antibodies), effectively pre-
obtain an efficient control of drug delivery. In this venting viral and bacterial infection.
294 T. Cerchiara et al.

Finally, Luppi et al. (2010b) formulated trans- having high cross-link density released 7.5 mg
dermal hydroxypropyl methylcellulose-based within 24 h. Moreover, devices having a diameter
films containing chlorpromazine hydrochloride of 2.5 cm and characterized by uncross-linked
for the treatment of psychotic disorders. Film chitosan membrane released about 15 mg after
composition was modified by incorporating a 24 h, while the same device having a diameter of
chemical permeation enhancer or binary enhancer 1.5 cm released about 4 mg after 24 h. Finally, all
combinations (oleic acid or polysorbate 80, or the devices delivered propranolol hydrochloride
both) and a plasticizer (propylene glycol). Both in a near zero order fashion suggesting that these
oleic acid and polysorbate 80 had significant chitosan membranes might be used successfully
effect on drug permeation with respect to the for the fabrication of membrane-controlled trans-
control formulation and films containing a dermal delivery systems.
mixture of oleic acid and polysorbate 80 pro- A more innovative approach in transdermal
vided the best enhancement activity for chlor- drug delivery is represented by film-forming
promazine. In fact, the amount of chlorpromazine solutions. These systems are essentially polymer
hydrochloride permeated through pig ear skin solutions, containing dispersed or dissolved drug.
from hydroxypropyl methylcellulose films after After application onto the skin, solvent evapora-
100 h were 15.2 ± 0.7 mg/cm2 for film containing tion guarantees the formation of a bioadhesive
oleic acid (2.15, % w/w on dry basis) and poly- and thin film. Compared with matrix-type vehi-
sorbate 80 (2.15, % w/w on dry basis) and cles and membrane-coated vehicles, these sys-
6.5 ± 0.3 mg/cm2 for control formulation (film tems are very easy to prepare and they possess
with the same composition, but without perme- higher dosage flexibility, less irritation of the
ation enhancers). Moreover, also the hydroxypro- skin, better cosmetic appearance associated with
pyl methylcellulose type and the different high ease of use. Schroeder et al. (2007) tested a
concentration of drug and plasticizer contributed broad range of polymers (acrylates, polyurethane-
to modulate drug permeation. A decrease of acrylates, cellulose derivatives, polyvinylpyrrol-
hydroxypropyl methylcellulose viscosity, as a idones, silicones) as film-forming materials.
function of its molecular weight, and an increase Polymers at different concentration were solubi-
in propylene glycol and chlorpromazine content lized in a volatile solvent (ethanol or volatile sili-
provided higher cumulative amounts of drug per- cone), added with different amounts of plasticizer
meated. These results confirm that chlorproma- (triethyl citrate, triacetin, dibutyl phthalate) and
zine permeation can be easily modulated by possibly with a cross-linking agent (succinic
varying the composition of hydroxypropyl meth- acid). Formulations with adequate properties for
ylcellulose-based films. the application on the skin (low viscosity, short
Another type of film-based vehicles is drying time, low outward stickiness, high cos-
membrane-coated systems that consist of a drug metical attractiveness, and integrity on the skin
depot and an hydrogel-based membrane able to for a prolonged time) were evaluated in terms of
control drug release. Tacharodi and Panduranga mechanical properties (tensile strength and elon-
Rao (1995) proposed a patch consisting of a chi- gation at break), water vapor permeability, and
tosan membrane cross-linked with different con- transepidermal water loss. The positively evalu-
centrations of glutaraldehyde and a chitosan gel ated preparations resulting from these experi-
as propranolol hydrochloride reservoir. Drug ments provide the basis for the development of
release can be easily tailored by changing cross- film-forming polymeric solutions as a transder-
link density within the membrane and strongly mal dosage form.
depends on the area of the device. In fact, devices A novel organic-inorganic hybrid film-
characterized by a diameter of 2.5 cm and chito- forming agent for transdermal drug delivery was
san membrane having low cross-link density prepared by a modified poly(vinyl alcohol) (PVA)
released about 12 mg of drug within 24 h, while gel using γ-(glycidyloxypropyl)trimethoxysi-
devices with diameter of 2.5 cm and membrane lane) (GPTMS) as a cross-linking agent, poly(N-
20 Hydrogel Vehicles for Hydrophilic Compounds 295

vinyl pyrrolidone) as a tackifier, and glycerol as a Cerchiara T, Luppi B, Bigucci F, Orienti I, Zecchi V (2002)
Physically cross-linked chitosan hydrogels as topical
plasticizer. The obtained gels can be applied to
vehicles for hydrophilic drugs. J Pharm Pharmacol
the skin by a coating method and in situ form 54:1453–1459
very thin and transparent films with good flexibil- Chen L, Tian Z, Du Y (2004) Synthesis and pH sensitivity
ity and adhesive properties. Furthermore, the skin of carboxymethyl chitosan-based polyampholyte
hydrogels for protein carrier matrices. Biomaterials
irritation tests showed that the formulations pro-
25(17):3725–3732
duced no skin irritation after topical application, Damink LHHO, Dijkstra PJ, vanLuyn MJA, vanWachem
while the in vitro release studies revealed that PB, Nieuwenhuis P, Feijen J (1996) In vitro degrada-
these films are able to release both hydrophilic tion of dermal sheep collagen cross-linked using a
water-soluble carbodiimide. Biomaterials 17:679–684
drugs such as 5-fluorouracil both lipophilic mol-
Denet AR, Vanbever R, Préat V (2004) Skin electropora-
ecules such as ibuprofen. The cumulative release tion for transdermal and topical delivery. Adv Drug
of hydrophilic 5-fluorouracil was much higher Deliv Rev 56:659–674
than hydrophobic ibuprofen and introduction of Draye JP, Delaey B, van de Voorde A, van den Bulcke A,
Bogdanov B, Schacht E (1998) In vitro release charac-
adequate GPTMS amount (GPTMS/(PVA +
teristics of bioactive molecules from dextran dialde-
GPTMS) ratio = 20–30 %) into PVA matrix can hyde cross-linked gelatin hydrogel films. Biomaterials
decrease the crystalline regions of PVA and 19:99–107
enhance drug diffusion (Guo et al. 2011). Eagland D, Crowther NJ, Butler CJ (1994) Complexation
between polyoxyethylene and polymethacrylic acid –
the importance of the molar mass of polyethylene. Eur
Conclusions Polym J 30:767–773
Transdermal delivery is a major administra- Eliaz RE, Kost J (2000) Characterization of a polymeric
tion route for drugs that are destroyed by the PLGA-injectable implant delivery system for the con-
trolled release of proteins. J Biomed Mater Res
liver when taken orally (Langer 2004).
50:388–396
Recently, much attention has been paid to the Feldstein MM, Tohmakhchi VN, Malkhazov LB, Vasiliev
hydrogels as vehicles for transdermal drug AE, Plate NA (1996) Hydrophilic polymeric matrices
delivery, and their success can be mainly for enhanced transdermal drug delivery. Int J Pharm
131:229–242
attributed to the possibility to modulate drug
Grant G, Morris ER, Rees DA, Smith PJC, Thom D
release kinetics (Kim et al. 1992). Currently, (1973) Biological interaction between polysaccha-
few drugs have been successfully delivered rides and divalent cations: the egg-box model. FEBS
through the skin utilizing hydrogels, and new Lett 32(1):195–198
Guo R, Du X, Zhang R, Deng L, Dong A, Zhang J (2011)
opportunities may be envisaged.
Bioadhesive film formed from a novel organic-
inorganic hybrid gel for transdermal drug delivery sys-
tem. Eur J Pharm Biopharm 79(3):574–583
Guy RH (1996) Current status and future prospects of
References transdermal drug delivery. Pharm Res
13:1765–1769
He W, Guo X, Zhang M (2008) Transdermal permeation
Ale I, Lachapelle JM, Maibach HI (2009) Skin tolerability
enhancement of N-trimethyl chitosan for testosterone.
associated with transdermal drug delivery systems: an
Int J Pharm 356(1–2):82–87
overview. Adv Ther 26(10):920–935
He W, Guo X, Xiao L, Feng M (2009) Study on the mech-
Anderson JM, Shive MS (1997) Biodegradation and bio-
anisms of chitosan and its derivatives used as transder-
compatibility of PLA and PLGA microspheres. Adv
mal penetration enhancers. Int J Pharm 382(1–2):
Drug Deliv Rev 28:5–24
234–243
Berger J, Reist M, Mayer MJ, Felt O, Peppas NA, Gurny
Hennink WE, van Nostrum CF (2002) Novel crosslinking
R (2004) Structure and interactions in covalently
methods to design hydrogels. Adv Drug Deliv Rev
and ionically crosslinked chitosan hydrogels for bio-
54(1):13–36
medical applications. Eur J Pharm Biopharm 57:
Henry S, McAllister DV, Allen MG, Prausnitz MR (1998)
19–34
Microfabricated microneedles: a novel approach to
Brasch U, Burchard W (1996) Preparation and solution
transdermal drug. J Pharm Sci 87:922–925
properties of microhydrogels from poly(vinyl alco-
Hickey AS, Peppas NA (1995) Mesh size and diffusive
hol). Macromol Chem Phys 197:223–235
characteristics of semicrystalline poly(vinyl alcohol)
Brown MB, Martin GP, Jones SA, Akomeah FK (2006)
membranes prepared by freezing/thawing techniques.
Dermal and transdermal drug delivery systems: cur-
J Membr Sci 107:229–237
rent and future prospects. Drug Deliv 13(3):175–187
296 T. Cerchiara et al.

Hoffman AS (1991) Environmentally sensitive polymers Miyata T, Uragami T, Nakamae K (2002) Biomolecule-
and hydrogels – “smart” biomaterials. MRS Bull sensitive hydrogels. Adv Drug Deliv Rev
XVI:42–46 54(1):79–98
Hoffman AS (2002) Hydrogels for biomedical applica- Murdan S (2003) Electro-responsive drug delivery from
tions. Adv Drug Deliv Rev 54:3–12 hydrogels. J Control Release 92(1–2):1–17
Hu Q, Liang W, Bao J, Ping Q (2000) Enhanced transdermal Murphy M, Carmichael AJ (2000) Transdermal drug
delivery of tetracaine by electroporation. Int J Pharm delivery systems and skin sensitivity reactions: inci-
202:121–124 dence and management. Am J Clin Dermatol
Iordanskii AL, Feldstein MM, Markin VS, Hadgraft J, 1(6):361–368
Plate NA (2000) Modeling of the drug delivery from a Muzzarelli R, Baldassarre V, Conti F, Ferrara P, Biagini G,
hydrophilic transdermal therapeutic system across Gazzanelli G et al (1988) Biological activity of chito-
polymer membrane. Eur J Pharm Biopharm san: ultrastructural study. Biomaterials 9:247–252
49:287–293 Nedkov E, Tsvetkova S (1994) Effect of γ-irradiation on
Ishii Y, Nakae T, Sakamoto F, Matsuo K, Quan YS, the crystalline structure of ultra high molecular
Kamiyama F et al (2008) A transcutaneous vaccina- weight poly(ethylene oxide). Radiat Phys Chem
tion system using a hydrogel patch for viral and bacte- 43:397–401
rial infection. J Control Release 131(2):113–120 Noble L, Gray AL, Sadiq L, Uchegbu IF (1999) A non-
Jatav VS, Singh H, Singh SK (2011) Recent trends on covalently cross-linked chitosan based hydrogel. Int J
hydrogels in human body. IJRPBS 2:442–447 Pharm 192(2):173–182
Kabanov VY (1998) Preparation of polymeric biomateri- Orienti I, Di Pietra A, Luppi B, Zecchi V (2000)
als with the aid of radiation-chemical methods. Russ Crosslinked polyvinylalcohol hydrogels as vehicles
Chem Rev 67:783–816 for hydrophilic drugs. Arch Pharm Pharm Med Chem
Khare AR, Peppas NA, Massimo G, Colombo P (1992) 333:421–424
Measurement of the swelling force in ionic polymeric Padula C, Colombo G, Nicoli S, Catellani PL, Massimo G,
networks. I. Effect of pH and ionic content. J Control Santi P (2003) Bioadhesive film for the transdermal deliv-
Release 22:239–244 ery of lidocaine: in vitro and in vivo behaviour. J Control
Kim J, Shin SC (2004) Controlled release of atenolol from Release 88(2):277–285
the ethylene-vinyl acetate matrix. Int J Pharm 273: Peppas NA, Mikos AG (1986) Preparation methods and
23–27 structure of hydrogels. In: Peppas NA (ed) Hydrogels
Kim SW, Bae YH, Okano T (1992) Hydrogels: swelling, in medicine and pharmacy. CRC press, Boca Raton,
drug loading and release. Pharm Res 9:283–290 pp 1–25
Kopeceka J (2007) Hydrogel biomaterials: a smart future? Peppas NA, Bures P, Leobandung W, Ichikawa H (2000)
Biomaterials 28:5185–5192 Hydrogels in pharmaceutical formulations. Eur J Pharm
Kurihara-Bergstrom T, Good WR, Feisulin S, Signur C Biopharm 50:27–46
(1991) Skin compatibility of transdermal drug deliv- Peppas NA, Hilt JZ, Khademhosseini A, Langer R (2006)
ery systems. J Control Release 15:271–278 Hydrogels in biology and medicine: from molecular prin-
Langer R (2004) Transdermal drug delivery: past prog- ciples to bionanotechnology. Adv Mater 18:1345–1360
ress, current status and future prospects. Adv Drug Pikal MJ (2001) The role of electroosmotic flow in trans-
Deliv Rev 56:557–558 dermal iontophoresis. Adv Drug Deliv Rev
Lee KY, Bouhadir KH, Mooney DJ (2004) Controlled 46:281–305
degradation of hydrogels using multi-functional cross- Prausnitz MR, Langer R (2008) Transdermal drug deliv-
linking molecules. Biomaterials 25(13):2461–2466 ery. Nat Biotechnol 26(11):1261–1268
Luo Y, Kirker RK, Prestwich GD (2000) Crosslinked Qu X, Wirsen A, Albertson AC (1999) Synthesis and char-
hyaluronic acid hydrogels films: new biomaterials for acterization of pH-sensitive hydrogels based on chito-
drug delivery. J Control Release 69:169–184 san and D. L-lactic acid. J Appl Polym Sci 74:
Luppi B, Cerchiara T, Bigucci F, Di Pietra AM, Orienti I, 3186–3192
Zecchi V (2003) Crosslinked poly(methyl vinyl ether- Rosiak JM (1991) Hydrogel dressings. In: Clough RL,
co-maleic anhydride) as topical vehicles for hydro- Shalaby SW (eds) Radiation effects on polymers. ACS
philic and lipophilic drugs. Drug Deliv 10:239–244 symposium series 475. American Chemical Society,
Luppi B, Bigucci F, Cerchiara T, Zecchi V (2010a) Washington, DC. pp 271–299
Chitosan-based hydrogels for nasal drug delivery: Safrany A (1997) Radiation processing: synthesis and
from inserts to nanoparticles. Expert Opin Drug Deliv modification of biomaterials for medical use. Nucl Inst
7:811–828 Methods Phys Res B 131(1–4):376–381
Luppi B, Bigucci F, Baldini M, Abruzzo A, Cerchiara T, Sahin S, Selek H, Ponchel G, Ercan MT, Sargon M,
Corace G et al (2010b) Hydroxypropylmethylcellulose Hincal AA et al (2002) Preparation, characterization
films for prolonged delivery of the antipsychotic drug and in vivo distribution of terbutaline sulfate loaded
chlorpromazine. J Pharm Pharmacol 62:305–309 albumin microspheres. J Control Release 82(2–3):
Mengatto LN, Helbling IM, Luna JA (2012) Recent 345–358
advances in chitosan films for controlled release of Schroeder IZ, Franke P, Schaefer UF, Lehr C (2007)
drugs. Recent Pat Drug Deliv Formul 6(2):156–170 Development and characterization of film forming
20 Hydrogel Vehicles for Hydrophilic Compounds 297

polymeric solutions for skin drug delivery. Eur J Timmer MD, Jo S, Wang C, Ambrose CG, Mikos AG
Pharm Biopharm 65(1):111–121 (2002) Characterization of the cross-linked structure
Silva CL, Pereira JC, Ramalho A, Pais AACC, Sousa of fumarate-based degradable polymer networks.
JJS (2008) Films based on chitosan polyelectrolyte Macromolecules 35:4373–4379
complexes for skin drug delivery: development and Uchegbu IF, Schatzlein AG, Tetley L, Gray AI, Sludden J,
characterization. J Membr Sci 320:268–279 Siddique S, Mosha E (1998) Polymeric chitosan-
Sintov AC, Krimberk I, Daniel D, Hannan T, Sohn Z, based vesicles for drug deliver. J Pharm Pharmacol
Levin G (2003) Radiofrequency-driven skin micro- 50:453–458
channeling as a new way for electrically assisted trans- Wang C, Steward RJ, Kopecek J (1999) Hybrid hydrogels
dermal delivery of hydrophilic drugs. J Control assembled from synthetic polymers and coiled-coil
Release 89:311–320 protein domains. Nature 397:417–420
Sludden J, Uchegbu IF, Schatzlein AG (2000) The encap- Watanabe T, Ohtsuka A, Murase N, Barth P, Gersonde K
sulation of bleomycin within chitosan based polymeric (1996) NMR studies on water and polymer diffusion
vesicles does not alter its biodistribution. J Pharm in dextran gels. Influence of potassium ions on micro-
Pharmacol 52:377–382 structure formation and gelation mechanism. Magn
Stauffer SR, Peppas NA (1992) Poly(vinyl alcohol) Reson Med 35:697
hydrogels prepared by freezing-thawing cyclic pro- Wichterle O, Lim D (1960) Hydrophilic gels for biologi-
cessing. Polymers 33(18):3932–3936 cal use. Nature 185:117–118
Taveira SF, Nomizo A, Lopez RFV (2009) Effect of the Wohlrab J, Kreft B, Tamke B (2011) Skin tolerability of
iontophoresis of a chitosan gel on doxorubicin skin pen- transdermal patches. Expert Opin Drug Deliv
etration and cytotoxicity. J Control Release 134:35–40 8(7):939–948
Thacharodi D, Rao KP (1995) Development and in vitro Xiao C, Zhou G (2003) Synthesis and properties of
evaluation of chitosan-based transdermal drug deliv- degradable poly(vinyl alcohol) hydrogel. Polym
ery systems for the controlled delivery of propranolol Degrad Stab 81(2):297–301
hydrochloride. Biomaterials 16:145–148 Zu Y, Zhang Y, Zhao X, Shan C, Zu S, Wang K et al (2012)
Thong HY, Zhai H, Maibach HI (2007) Percutaneous pen- Preparation and characterization of chitosan–polyvinyl
etration enhancers: an overview. Skin Pharmacol alcohol blend hydrogels for the controlled release of
Physiol 20(6):272–282 nano-insulin. Int J Biol Macromol 50:82–87
Lecithin Organogels in Enhancing
Skin Delivery of Drugs 21
I.M. Shaikh, K.R. Jadhav, and V.J. Kadam

Contents 21.8 Characterization of Lecithin


Organogels (LOs) ..................................... 306
21.1 Introduction .............................................. 299 21.8.1 Structural Features ..................................... 306
21.2 Lecithin Organogels (LOs) ...................... 300 21.8.2 Rheological Behavior. . . . . . . . . . . . . . . . . 307
21.8.3 Phase Transition Temperatures . . . . . . . . . 307
21.3 Physicochemical Properties 21.8.4 Water Content . . . . . . . . . . . . . . . . . . . . . . 307
of Lecithin Organogels ............................ 300
21.3.1 Viscoelasticity ............................................ 300 21.9 Mechanism of Penetration Enhancement
21.3.2 Non-birefringence and Optical of Lecithin Organogels (LOs) ................. 307
Transparency .............................................. 301 21.10 Improved Skin Penetration Enhancement
21.3.3 Thermoreversibility/Thermostability ......... 301 and Enhanced Efficacy of Drugs
21.3.4 Biological, Physical, and Chemical with Lecithin Organogels (LOs) as Drug
Safety ......................................................... 301 Carriers .................................................. 309
21.4 Salient Features of Lecithin 21.11 Safety Profile of Lecithin Organogels
Organogels (LOs) ..................................... 302 (LOs) as Drug Carrier ............................. 310
21.5 Preparation of Lecithin Organogels ....... 302 Conclusion .............................................................. 311
21.5.1 Organogellator: Lecithin ............................ 302
21.5.2 Organic Solvents ........................................ 303 References ............................................................... 311
21.5.3 Polar Solvent .............................................. 303
21.6 Identification of Appropriate Quantities
of Lecithin/Organic Solvent/Water
Using Phase Diagram............................... 304
21.7 Mechanism of Formation Lecithin 21.1 Introduction
Organogels (LOs) ..................................... 305
In the past three decades, LOs have emerged as
one of the most effective drug delivery systems
I.M. Shaikh (*)
Pharmaceutical R&D, GlaxoSmithKline
for the transdermal delivery of hydrophobic,
Pharmaceuticals Ltd, A-10, MIDC, hydrophilic, as well as amphoteric drugs, includ-
Ambad, Nashik 422010, India ing enzymes. LOs are gels formed by the addition
e-mail: shaikhpeer17@gmail.com of trace amounts of water to organic/nonaque-
K.R. Jadhav • V.J. Kadam ous lecithin solution. LOs are thermodynami-
Department of Pharmaceutics, Bharati Vidyapeeth’s cally stable, clear, viscoelastic, biocompatible,
College of Pharmacy, Sector-8, CBD Belapur,
Navi Mumbai 400614, India
isotropic gels composed of lecithin, an organic
e-mail: krj24@rediffmail.com; solvent and a polar liquid. It was Scartazzini and
thinkgood786@yahoo.co.in Luisi who first provided information about LOs

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 299


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_21, © Springer-Verlag Berlin Heidelberg 2015
300 I.M. Shaikh et al.

in an article published in 1988. They observed 1990). LOs are formed by a three-dimensional
that adding a small quantity of water into a non- network of entangled reverse cylindrical micelles
aqueous solution of naturally occurring lecithin (Motulsky et al. 2005). First, the lecithin spheri-
resulted in drastic rise in viscosity (Scartazzini cal reverse micelles are formed when lecithin is
and Luisi 1988). Two years later, Schurtenberger solubilized in nonpolar organic liquids such as
et al. (1990) published their findings on struc- isopropyl myristate, isopropyl palmitate, and oth-
tural and dynamic properties of LOs. Following ers (Walde et al. 1990). Later, hydration of these
these reports, LOs have gained attention from spherical reverse micellar leads to the formation of
the researchers across the globe. Willimann and elongated tubular micelles. These then entangle to
Luisi (1991) were the first to unleash the poten- form a three-dimensional network in the solution
tial of LOs as a matrix for transdermal delivery (Shchipunov 2001). The latter serves to immobi-
of drugs. They investigated the ability of LOs as lize the external organic phase, thus producing a
a means to deliver scopolamine across the skin. gel form of the initial nonviscous solution, keep-
A tenfold increase in the percutaneous absorp- ing the transparency and optical isotropy of the
tion of scopolamine was observed when delivered organogel same as the original (Capitani et al.
using LOs as compared to the aqueous solution of 1996). The supramolecularly associated micel-
scopolamine. Since then, many studies have been lar aggregates in the entangled state are similar
conducted by various research groups with LOs to uncrossed polymers in semi-dilute or concen-
as transdermal drug delivery systems. Although trated solutions, due to which these systems are
LOs have demonstrated their efficacy and safety often called polymer-like micelles. They are also
as delivery systems at the lab scale, their util- termed as living or equilibrium polymers, worm-
ity as a drug delivery system needs to be further like or threadlike micelles (Shchipunov 1995;
demonstrated in clinical settings. For this pur- Shchipunov and Shumilina 1995).
pose, the full biocompatibility profile (efficacy
in humans and long-term toxicity) of LOs needs
to be explored. Once clinically proven, LOs will 21.3 Physicochemical Properties
emerge as a promising pharmaceutical carrier sys- of Lecithin Organogels
tem for the transdermal delivery of drugs and will
replace most of the conventional delivery systems 21.3.1 Viscoelasticity
used for the transdermal delivery of drugs.
The viscous and elastic nature of LOs follows the
Maxwell model of viscoelasticity (Toshiyuki et al.
21.2 Lecithin Organogels (LOs) 2003; Shikata et al. 2003). LOs are the three-dimen-
sional structures formed as a result of physical
LOs are, as aforementioned, thermodynami- interactions of lecithin molecules with each other.
cally stable gels which are composed of lecithin LOs show an elastic property at low shear rate. As
(Fig. 21.1), an organic solvent and a polar liquid. the shear rate increases, the physical interactions
LOs exhibit viscoelasticity, biocompatibility, and among lecithin molecules start getting weak, and
isotropicity. They are generally clear in appearance thereby the three-dimensional fibrous structure of
(Willimann and Luisi 1991; Schurtenberger et al. LOs is distorted. Finally, the shear stress reaches
its threshold and destroys the LOs’ structure.
O This behavior is termed as plastic flow behavior
(Abdallah et al. 2000). The viscoelastic behavior
R
O
O−
of LOs depends on the concentration of lecithin
R O O O
P N+(CH)3 and the type of organic solvent (precisely fatty
acid ester) used. The higher the lecithin concentra-
O O
tion, the higher is the viscosity (Schurtenberger et
Fig. 21.1 Structure of lecithin al. 1989). The long-chain fatty acid esters, such
21 Lecithin Organogels in Enhancing Skin Delivery of Drugs 301

as isopropyl palmitate or cetearyl octanoate, pro-


duce LOs with high viscosity, whereas short-chain
esters, such as ethyl and propyl acetate, produce
LOs of relatively lesser viscosity. It is worthwhile
to note that LOs can also be prepared by utiliz-
ing mixtures of solvents rather than using a single
solvent. Using a blend of organic solvents, we can
tailor the viscoelastic properties of LOs based on
our needs. Nastruzzi et al. (1994) has developed
LOs using a combination of organic solvents and
investigated its effect on the viscoelastic properties
of LOs. The viscosity of LOs has an impact on the
release profile of the drug encapsulated into LOs.
In general, the higher the viscosity, the slower is
the release of the drug (Shchipunov and Shumilina
1995; Kumar and Katare 2005). Also, medicated
LOs are reported to have slightly lesser viscosity
than placebo LOs of similar composition (Shaikh Fig. 21.2 Thermal behavior of the organogels. (a) Sol
state of the LO at 60 °C and (b) semisolid state of the LO
et al. 2009; Nastruzzi and Gambari 1994).
at room temperature

21.3.2 Non-birefringence cooling of hot LOs causes regain of physical


and Optical Transparency interaction between lecithin molecules which
results in sol-gel transition (Avramiotis et al.
The LOs when viewed under polarized light 2007). The gelation temperature can be deter-
appear as a dark matrix. This is because the mined visibly. It is essential to determine the
isotropic nature of organogels does not cause gelation temperature of the LOs containing drugs
passage of polarized light through the matrix. as it serves as a guide for recommending storage
This property of organogels is termed as non- conditions for drug-loaded medicated LOs (Díaz
birefringence (Kantaria et al. 1999; Nasseri et al. et al. 2008; Dasgupta et al. 2009; Guenet 2006).
2003). Optically, LOs are transparent and pro-
vide the benefit of visual inspection so that the
presence of any particulate matter can be easily 21.3.4 Biological, Physical,
recognized (Kumar and Katare 2005). and Chemical Safety

Lecithin is an important component of all living


21.3.3 Thermoreversibility/ cells, and it is recognized by the Food and Drug
Thermostability Administration (FDA) as Generally Regarded as
Safe (GRAS) (21 CFR 184, 1400). Compatibility
Heating above critical temperature causes LOs studies with human skin revealed that topical use
to convert from gel state to sol state (Fig. 21.2). of LOs is safe (Dreher et al. 1997; Shchipunov
The temperature at which sol-gel transition takes et al. 2001; Schurtenberger et al. 1990). Vehicles
place is called as gelation temperature. The gela- (organic solvents, such as fatty acid esters, e.g.,
tion temperature varies depending on the solvent isopropyl myristate, ethyl oleate) used for pre-
system used when preparing LOs. The gel-sol paring LOs are also GRAS excipients. LOs are
transition in LOs occurs when increased thermal moisture insensitive, and due to their organic
energy within LOs disrupts physical interac- nature, they resist microbial contamination.
tions between lecithin molecules, and subsequent Thermodynamic stability, ease of preparation and
302 I.M. Shaikh et al.

scale-up, easier quality monitoring, and enhanced state has been demonstrated only for nonaqueous
topical performance along with biocompatibility solutions of naturally occurring unsaturated leci-
and safety upon applications for a prolonged thin (Walde et al. 1990; Shchipunov 2001).
period make the organogels a vehicle of choice
for dermal and transdermal drug delivery.
21.5.1 Organogellator: Lecithin

21.4 Salient Features of Lecithin Lecithin is a trivial name for 1, 2-diacyl-sn-3-


Organogels (LOs) phosphocholine. It belongs to a biologically
essential class of substances termed phospho-
• LOs can encapsulate various substances with glycerides or phospholipids. Lecithin is a com-
diverse physicochemical characters, i.e., having plex mixture of acetone-insoluble phosphatides,
a different solubility, molecular weight, and size. which mainly consist of phosphatidylcholine,
• Self-assembled supramolecular arrangement phosphatidylethanolamine, phosphatidylserine,
of surfactant molecules imparts spontaneity to and phosphatidylinositol combined with differ-
organogel formation and hence the process ent amounts of other substances such as triglyc-
becomes simpler. erides and fatty acids (Reynolds 1996). The main
• LOs remain structurally integrated for a lon- sources of lecithin are soya beans and egg yolk.
ger period of time due to their thermodynamic Lecithin varies greatly in its physical form, from
stability. viscous semiliquid to powder depending on the
• LOs are not moisture sensitive; they also resist content of free fatty acids. It may also vary in
microbial contamination due to their organic color from brown to light yellow depending on
nature. whether it is bleached or unbleached (Wade et al.
• Being well balanced in hydrophilic and lipo- 1994). Lecithin is commercially available on the
philic character, they efficiently partition into market under trade name of Epikuron (Lucas
the skin and enhance the skin penetration and Meyer, Hamburg, Germany), Lipoid S100 (Lipoid
transport of molecules. LOs also provide the GmbH, Ludwigshafen, Germany), and Capcithin
desired hydration of the skin in a lipid- (Lucas Meyer, Hamburg, Germany), which are
enriched environment. derived and purified from either soya bean or eggs.
• The use of biocompatible, biodegradable, and The desired gelation in organic solvent occurs
non-immunogenic materials for their formula- only when the lecithin contains more than 95 %
tion makes them safe for long-term phosphatidylcholine and is free from fat as well
applications (Kumar and Katare 2005).1 as moisture. Lecithin is a multifunctional surface-
active agent. The lecithin molecule consists of two
portions: the nonpolar tail (fatty acid portion) and
21.5 Preparation of Lecithin the polar head (phosphoric acid portion). Because
Organogels of these two aspects, lecithin molecules arrange
themselves at the boundary between immiscible
The LOs are readily obtained by adding a mini- liquids such as oil and water. This arrangement
mal amount of polar solvent, such as water, to a reduces the interfacial tension between oil and
solution of lecithin in organic solvents. They are water and makes relatively stable emulsions
formed by three different components, including (Scartazzini et al. 1988). Its unique lipid molecu-
organogellator (lecithin), a nonpolar organic sol- lar structure performs versatile functions. It has a
vent as external or continuous phase, and a polar wide variety of roles in pharmaceuticals, cosmet-
agent, usually water. The transfer into jelly-like ics, and food industries as an emulsifier, viscosity
modifier, stabilizer, and solubilizer and penetra-
1
The above content is adapted and modified from (Kumar tion enhancer (Szuhaj 1989). They form the lipid
and Katare 2005), with permission. matrix of biological membrane and play a key
21 Lecithin Organogels in Enhancing Skin Delivery of Drugs 303

role in the cellular metabolism (Hanahan 1997). n-pentane, n-hexane, n-hexadecane, and
Due to its biocompatibility, it is widely used in tripropylamine. Among these, the fatty acid esters
human and animal food, medicine, cosmetics, have been widely used for LO formation (due to
and other various industrial applications (Wendel better skin feel), but structural investigations have
1995). No systematic research has been done till only been performed on hydrocarbons (such as iso-
date in order to investigate the effect of unsatu- octane, cyclohexane). Natural oils including soya
ration in phospholipids on organogelling ability. bean oil, sunflower oil, rapeseed oil, and mustard
However, it has been reported that unsaturation in oil are proposed as potentially useful organic sol-
phospholipid molecules affect the nature of self- vents for preparing LOs (Shchipunov 2001).
assembly in which the phospholipid molecules
associate and form the microstructures. The prop-
erty of unsaturation can be interpreted in terms 21.5.3 Polar Solvent
of the degree of hydration of phospholipid mol-
ecules that it provides. Unlike saturated hydroge- The third component, a polar agent, acts as a struc-
nated phospholipids, unsaturation in phospholipid ture forming and stabilizing agent. A series of polar
molecules would result in better hydration of the solvents have been studied in order to check their
polar head group, thereby increasing the area per suitability for producing the thickening effect on
lipid polar head group. Hence, a larger area-to- hydrocarbon and fatty acid ester solutions of leci-
volume ratio would favorably alter the spontane- thin. Water has been used extensively as a polar sol-
ous curvature of lipid monomers for the formation vent for organogel formation. However, it has been
of micelles and subsequently their self-assembly established that glycerol, formamide, and ethylene
to form the micellar network (Shchipunov 2001). glycol have the ability to induce gelation. The gel-
forming ability of the polar solvent is governed by
its physicochemical properties (Shchipunov and
21.5.2 Organic Solvents Shumilina 1995, 1996; Shchipunov and Hoffmann
1998). The ability to promote thickening of lecithin
Organic solvent plays a vital role in organogels by solutions has been correlated with the polarity of the
providing the desired solvent action for the drug solvent used for preparing LOs. This correlation is
(hydrophobic) as well as for lecithin (Kumar and particularly pronounced in the series of structurally
Katare 2005; Moore 1982; Sato et al. 1988). More related solvents such as glycerol and ethylene gly-
than 50 organic solvents have been reported to col. In a proposed model of organogels, the solvent
form organogels with water as an aqueous phase. molecules bridge phosphate groups of neighboring
Among them, there are linear, branched, and cyclic lipid molecules, allowing their association into
alkanes; ethers and esters; fatty acids; and amines. tubular aggregates through an extensive ribbonlike
Specific examples include ethyl laureate, ethyl hydrogen bonding network (Shchipunov 2001).
myristate, isopropyl palmitate, cyclopentane, Figure 21.3 depicts schematic diagram of the prepa-
cyclooctane, trans-decalin, trans- pinane, ration of LOs.

Lecithin
Polar Organogel
Fig. 21.3 Schematic Apolar solvent Soy lecithin
additive
diagram of the preparation of dispersion in organic
LOs solvent
304 I.M. Shaikh et al.

It is important to understand that LOs cannot for ethyl myristate-based LOs with a lecithin
be formed by random mixing of lecithin, an concentration of 200 mM, nw value is reported as 5
organic solvent and polar solvent. It depends on (Nastruzzi and Gambari 1994). LOs exist in a nar-
the mixing of a fixed quantity of these three com- row characteristic range of water-to-lecithin molar
ponents. Selection of suitable amounts of these ratio, which is reported as ncr. It is the critical molar
three components can be done by making use of ratio of polar solvent to lecithin molecules at which
the pseudo ternary phase diagram. A detailed complete gel formation takes place and entrapment
description on how to estimate the determination of maximum amount of external organic phase is
of the required amount of lecithin/water/organic observed. When nw is equal to ncr, LOs with maxi-
solvent is described in the next section. mum viscosity are produced, and when nw exceeds
ncr, phase separation of the LOs occurs (Shchipunov
2001; Shchipunov and Schmiedel 1996a, b). The
21.6 Identification of Appropriate rheological measurements or visual and optical
Quantities of Lecithin/ observations of a ternary system between cross
Organic Solvent/Water Using polarizers are widely used to determine ncr values.
Phase Diagram For a better understanding on the construction of a
phase diagram, we wish to describe an example
The phase diagram provides information for the from our reported paper on LO-containing aceclof-
boundaries of the different phases as a function of enac (Shaikh et al. 2009).
composition variables. The phase behavior of a ter- A pseudo ternary phase diagram was con-
nary system of lecithin/organic solvent/polar sol- structed to determine the concentration range of
vent is mainly governed by the concentration of lecithin, organic solvent (ethyl oleate), and water
polar solvent and lecithin (Shchipunov 2001; required for aceclofenac containing LO. This
Shchipunov and Schmiedel 1996). It is defined in study was carried out for the lecithin concentra-
terms of a parameter, molar ratio of polar solvent to tion ranging from 10 to 60 % w/v because leci-
lecithin (nw = [polar solvent]/[lecithin]). When the thin in proportion above 60 % w/v could not be
polar solvent is water, nw is also termed wo (wo = solubilized. Initially, water in oil microemulsion
[water]/[lecithin]). Nw is the critical molar ratio of was stabilized by lecithin micelles formed with
polar solvent to lecithin molecules where gel forma- a low concentration of water, which is charac-
tion and entrapment of external organic phase occur. terized by optical transparency and low viscos-
For LOs prepared using different organic sol- ity (Fig. 21.4a). As the amount of water was
vents but fixed lecithin concentration, the nw value increased, the microemulsion turned to a vis-
varies from solvent to solvent. For isopropyl cous gel (Fig. 21.4b). Further increase in water
myristate-based LOs with a lecithin concentration resulted in turbidity appearance (Fig. 21.4c) and
of 200 mM, nw value is reported to be 3, whereas finally phase separation (Fig. 21.4d). Organogel

Fig. 21.4 Changes in lecithin-/ethyl oleate-based system emulsion turned into viscous transparent gel at wo = 4, (c)
with addition of water. (a) Water in oil microemulsion turbidity appearance wo > 4, and (d) phase separation at
with low viscosity and optical transparency, (b) micro- wo > 5
21 Lecithin Organogels in Enhancing Skin Delivery of Drugs 305

Fig. 21.5 Ternary phase ORGANOGEL


diagram for ethyl oleate/
lecithin/water system 10% 10%
TURBIDITY
20% 20% APPEARANCE
PHASE
30% 30% SEPARATION

40% 40%

50% 50%

60% 60%

70% 70%

80% 80%

90% 90%

10% 20% 30% 40% 50% 60% 70% 80% 90%

existence area was obtained from the above at a concentration of ~0.01 mM (Shchipunov
pseudo ternary phase diagram (Fig. 21.5). et al. 1998). The enormous uniaxial growth of
Based on the outcome of the phase diagram these spherical reverse micelles and subsequent
experiment, it was inferred that the occurrence of transformation into tubular or cylindrical micellar
the gel phase was lecithin concentration depen- aggregates (sphere-to-cylinder transformation)
dent. The water-holding capacity of lecithin is triggered by the addition of small and critical
organogel increases with the increase in lecithin amounts of polar additive as shown in Fig. 21.7.
concentration. Lecithin concentrations ranging The molecules of polar solvent, on addition,
from 10 % to 60 % w/v were found to be opti- bind in stoichiometric ratios to the hydrophilic head
mum for preparing LOs. portion of the lecithin molecules in such a way
that two adjacent lecithin molecules are bridged
together by one polar molecule (Walde et al. 1990;
21.7 Mechanism of Formation Shchipunov and Shumilina 1995). This leads to
Lecithin Organogels (LOs) the formation of linear networks resulting due to
hydrogen bonds formed by the polar molecules
The first prerequisite for gel formation is the bal- and phosphate groups of lecithin molecules and, in
ance of intermolecular interaction among the turn, to the 1-dimensional uniaxial growth of leci-
gelator molecules (e.g., H bonding, van der thin reverse micelles. If polar additive concentra-
Waals interactions, etc.) and between gelator and tion is increased further, flexible and long tubular
solvent molecules. A comparative increase in the micelles from 2.0 to 2.5 nm in radius and nanome-
intermolecular attraction among the gelator mol- ter length are formed (Shchipunov and Schmiedel
ecules and a comparative decrease in the interac- 1996a, b). After reaching a critical length, these
tion between the gelator molecules and solvent extended micelles begin to overlap, entangle them-
lead to the formation of a molecular dispersion, selves, and build up a transient 3-dimensional net-
which further results in the formation of a three- work (Shchipunov and Hoffmann 2000; Voit and
dimensional network in which the solvent mole- Shchipunov 2000; Shchipunov et al. 2001).
cules are trapped (Fig. 21.6). This marks a crossover to a system characterized
Lecithin forms reverse spherical micelles spon- by increased viscosity and viscoelastic properties.
taneously when dissolved in apolar solvent alone The organogel formed contains a considerable
306 I.M. Shaikh et al.

Addition of Addition of
Soy Locithin Polar Additive

Entrapped extenal
Phase (organic solvent)

Three-dimensional
network of entangled
cylindrical micelles
Apolar solvent Soy Lecithin dispersion in Organogel
apolar solvent
Stage 1. Solubilization of Stage 2. Addition of polar additive Stage 2. Spontaneous formation of organogel
lecithin in organic media with solubilized guest molecules

Fig. 21.6 Entanglements of cylindrical reverse micelles to form gel

Addition of water interactions. Hence, they become highly sen-


sitive and impart difficulties in investigative
studies. It was found that a number of physico-
chemical properties of LOs such as rheological
behavior, physical and mechanical stability, and
drug release behavior depend upon the arrange-
ment of molecules within LOs to provide specific
structural network (Willimann and Luisi 1991;
Schurtenberger et al. 1990; Capitani et al. 1996).

Fig. 21.7 Transformation of spherical micelles into


cylindrical micelles 21.8.1 Structural Features

amount of external organic phase entrapped in the Structural elucidation is important for any organo-
spaces between the entangled reverse micelles. The gel system. The isotropic nature and the optical
hydrogen bonding network built up by molecules clarity of LOs make their study feasible by vari-
of polar additive and phosphate groups is also ous spectroscopic techniques, namely, nuclear
accompanied by stiffness of the phospholipid mol- magnetic resonance (NMR) spectroscopy (i.e.,
2
ecule in the region of phosphate group and glycerol H NMR, 31P NMR), and Fourier transformed
residue, which stabilizes the micellar aggregates. infrared (FTIR) spectroscopy. Various techniques
have been employed to find the nature of binding
forces responsible for the association of mono-
21.8 Characterization of Lecithin mers to form self-assembled structures, and FTIR
Organogels (LOs) (Kumar spectroscopy has been found to be successful in
and Katare 2005)2 establishing the hydrogen bonding as one of the
major driving forces for the self-assembly of
Interior structural design due to self-associated organogelator molecules in the organic solvents
supramolecules makes the characterization of (Aboofazeli et al. 2002; Bonina et al. 1995).
LOs complicated. These microstructures are The molecular packing of organogelator mole-
formed due to the number of polar-nonpolar cules inside the organogel network was studied by
scanning and transmission electron microscopy
2
The content of Sect. 21.8 is partly adapted from the ref- (SEM and TEM, respectively) (Willimann and
erence and suitably modified with due permission. Luisi 1991), dynamic and static light scattering
21 Lecithin Organogels in Enhancing Skin Delivery of Drugs 307

(elastic or quasielastic light scattering (QLS) type of organic solvent, concentration of gelator or
techniques) (Schurtenberger et al. 1990, 1993; cosurfactant, or the type or amount of polar agent),
Capitani et al. 1996; Sato et al. 1988; Aboofazeli which significantly influence the structural stabil-
et al. 2000), small-angle neutron scattering ity and rheological behavior of organogels (Voit
(SANS), small-angle X-ray scattering (SAXS), and Shchipunov 2000).
and atomic force microscopy (AFM) (Zemb et al.
1990; Terech and Weiss 1998; Simmons et al.
2001; Gronwald et al. 2002). These techniques 21.8.3 Phase Transition
allow many features of organogels to be studied Temperatures
at 1–1,000 nm scale. Recently, SAXS and AFM
are used to study the molecular arrangement of The phase transition temperature (PTT) gives an
LOs (Gronwald et al. 2002; Abdallah et al. 2000). idea about the nature of microstructures which are
The scattering information using SAXS and found in the network of organogels. The phase
SANS measurements on organogels combined with behavior of organogels varies on changing tem-
mathematical analysis provides details about the perature conditions. PTTs not only help in optimiz-
static correlation length ξ, mesh size of the network ing the organogels’ composition (Jibry et al. 2004)
(or the number density of entanglements “ν”), and but also reveal the microstructural homogeneity of
diffusion coefficients. They also give information the prepared organogel system. For example, a nar-
on the flexibility of the fibrous network, along with row PTT range (i.e., 3–5 °C) indicates homogenous
the structural features of the cross sections of LOs microstructures within the gel (Terech 1985).
(Terech and Weiss 1998; Simmons et al. 2001; Accurate and sensitive methods like hot stage
VanEsch and Feringa 2000; McAllister et al. 2002). microscopy (HSM) and high sensitivity differential
The direct visualization of the gel in its naïve state is scanning calorimetry (HSDSC) are used to mea-
possible using AFM, which allows observing the sure PTTs. However, the inverse flow method, a
microstructures of the fibrous network throughout simple technique based on visual observations, has
the gel mass. It also provides structural details also been employed (Couffin-Hoarau et al. 2004).
where micellar fibers or chains aggregate into large-
sized bundles. Thus, multiple instrumental tech-
niques based on microscopy along with 21.8.4 Water Content
spectroscopic and scattering analysis can help
reveal the structural details of the LO systems. The water content of an organogel system is
important to be maintained, as evaporation of
water can cause a consequent decrease in viscos-
21.8.2 Rheological Behavior ity, thus affecting the gel stability. Near-infrared
(NIR) spectroscopy has been proposed as a sim-
LOs have been studied extensively for their rheo- ple, rapid, and nondestructive technique for
logical attributes and have been determined to be determining the water content in organogels
viscoelastic in nature (Shchipunov 2001; (Nastruzzi and Gambari 1994). This technique
Shchipunov and Hoffmann 2000). The critical can also identify syneresis in organogels.
parameters such as spreadability, adhesiveness,
cohesiveness, and gel consistency need to be mod-
ified in a favorable manner. It has been reported 21.9 Mechanism of Penetration
that the Maxwell rheology model holds good for Enhancement of Lecithin
systems with supramolecular organization, con- Organogels (LOs)
sisting of a temporal three-dimensional network of
entangled micelles. Also, the desired viscoelastic Drugs administered by conventional means
property can be managed by modifying the vari- often have undesirable side effects and are many
ous formulation components (i.e., selecting the times ineffective. Transdermal delivery of drugs
308 I.M. Shaikh et al.

provides advantages like avoidance of first-pass formulation (Epikuron plus tetraglycol) was
metabolism, increased drug efficacy, etc. But as 5.06 μg/cm2/h. Similarly, the average flux for
the skin is an exceptionally effective barrier to procaterol in the control preparation (propylene
most chemicals, very few drugs can penetrate it glycol alone) was 0.14 μg/cm2/h and that for egg
in a manner sufficient to deliver therapeutic dose. yolk lecithin containing formulation (egg yolk
Therefore, drug delivery systems that have the lecithin plus propylene glycol) was 3.47 μg/cm2/h.
ability to traverse the skin by transient opening As exemplified, their research suggested that both
of pores and thereby delivering the drugs to their commercial soya bean lecithins and egg yolk
site of action are of great interest. Traditionally; lecithins are effective skin penetration enhanc-
to formulate such drug delivery systems; chemi- ers and that the control preparations resulted in
cal penetration enhancers are incorporated in the significantly decreased permeation of the drug
formulations. However; long term use of chemi- molecules. They reported that the enhancement
cal penetration enhancers are associated with effect could be mainly due to the reduction of skin
skin irritation and sensitization. Thus; long term resistance to drug permeation induced by surfac-
use of these chemical penetration enhancers are tant action of phospholipids present in lecithin.
avoided. Lecithin is a biocompatible and bio- Bentley et al. (1997) investigated the permeation
degradable surfactant as well as a cellular com- of hydrocortisone acetate from poloxamer gels
ponent, which acts as a penetration enhancer containing penetration enhancers, such as lecithin
(Schneider 1997). A wide variety of molecules, (8 % w/v) or urea (12 % w/v). Lecithin was found to
such as vitamins A and C, hormones, peptides, significantly increase the permeation of hydrocor-
amino acids, local anesthetics, and antifungal tisone acetate as compared to urea. They investi-
agents were reported to have increased perme- gated the mechanism of penetration enhancement
ation when delivered using LOs as a matrix for by differential scanning calorimetry (DSC) and
transdermal/topical drug delivery. The perme- FTIR spectroscopy using the SC from hairless
ation enhancement induced by LOs is attributed mouse treated with solutions of lecithin and urea.
to (a) lecithin and (b) organic solvents used for The DSC and FTIR results suggested that lecithin
LO preparation. has a more pronounced effect with the intercel-
Lecithin is reported to disorganize the struc- lular lipids than urea and thereby aids in increased
ture of the skin; it opens up the pores of the skin skin penetration of hydrocortisone acetate.
transiently and thus increases the penetration of As to organic solvents, there are a wide
various drugs (Natsuki and Takabatake 1987). range of organic solvents, such as ethyl laure-
Although the exact mechanism of how lecithin ate, ethyl myristate, isopropyl palmitate, isopro-
causes skin alteration is not yet clearly under- pyl myristate, cyclopentane, cyclooctane, and
stood, it is anticipated that it could be due to the n-hexadecane, that can be used for LO prepara-
interaction between the skin lipids and the leci- tion. Organic solvents containing fatty acid esters
thin’s phospholipids. Mahjour et al. (1990) stud- give better feel to the skin as compared to the
ied the effects of commercial lecithin, Epikuron organic solvent containing hydrocarbon oils and,
135f (Lucas Meyer, Hamburg, Germany), in tetra- thus, are preferred for preparing LOs. Moreover,
glycol and egg yolk lecithin in propylene glycol fatty acid esters have shown to increase the
on the in vitro permeation of procaterol, dextro- permeation of drugs, such as aceclofenac, sco-
methorphan, oxymorphone, and diphenhydramine polamine, and broxaterol (Shaikh et al. 2009;
using hairless mouse skin as a model membrane. Willimann and Luisi 1991). The exact mecha-
Vehicles (tetraglycol and propylene glycol) with- nism of skin penetration enhancement effect of
out lecithin were considered as control. The aver- fatty acid esters is not known, but it is proposed
age flux for procaterol in the control preparation that these solvents enhance skin permeation by
(tetraglycol alone) was 0.05 μg/cm2/h and that solubilizing the SC (Fujii et al. 1996; Dreher
for commercial soya bean lecithin-containing et al. 1996, 1997).
21 Lecithin Organogels in Enhancing Skin Delivery of Drugs 309

21.10 Improved Skin Penetration • Bonina et al. (1995) have reported an enhanced
Enhancement and Enhanced in vitro and in vivo skin penetration of methyl
Efficacy of Drugs nicotinate for LOs and lecithin-based lipo-
with Lecithin Organogels somes as compared to phospholipid free vehi-
(LOs) as Drug Carriers cles (carbomer- and carboxymethyl-based
hydrophilic gels). LOs were prepared by using
LOs have shown great promise as a drug matrix lecithin/IPP/water. Their results emphasized
for enhanced skin penetration of drugs. There is the significance of having lecithin in transder-
another drug delivery system related to LO, i.e., mal preparations so as to get better effects.
Pluronic Lecithin Organogel, or PLO, that has The results showed rapid induction and intense
shown the ability to enhance the dermal/transder- persistence of methyl nicotinate-induced ery-
mal delivery of various drugs. However, in this thema when LOs were tested in human sub-
section, we will be dealing only with the applica- jects. The author proposed the supramolecular
tion of LOs as skin penetration-enhancing drug aggregation structure of lecithin (which leads
matrices. The readers are encouraged to refer to to a strong interaction of lecithin with the SC)
reviews on PLO from Kumar and Katare (2005), as a probable cause of enhanced methyl nico-
Murdan (2005), and Almeida (2012) for details tinate skin penetration from LOs.
on preparation and applications of PLO. As to • Dreher et al. (1996) have reported their study
LOs and their application as complex penetration on diclofenac incorporated in LOs. LOs con-
enhancers, examples are given below: sisted of lecithin/IPP/water. Improved skin
• Willimann et al. (1992) have reported LOs con- penetration of diclofenac by 3.5-fold was
taining scopolamine. LOs were prepared with observed for LOs as compared to the vehicle
lecithin/IPP/water. They investigated the com- (IPP alone) alone. In addition, Grace et al.
parative ability of LOs with aqueous solution of (1999) have assessed the therapeutic efficacy
scopolamine as a means to deliver scopolamine of a 2 % diclofenac LO by conducting a
across the skin. They observed a tenfold randomized clinical trial in patients suffering
increase in the percutaneous absorption of sco- from osteoarthritis. A significant reduction of
polamine when delivered using LOs as com- knee pain in osteoarthritic patients was
pared to the aqueous solution of scopolamine. reported with the 2 % diclofenac LOs.
• Nastruzzi and Gambari (1994) have reported • Fujii et al. (1996) proved greater permeation
the transdermal delivery of the aromatic poly- of indomethacin from LOs through rat skin.
amidine TAPP-Br (compounds with antitumor The gels were prepared by lecithin/isoce-
activity) formulated in LOs. LOs were devel- tyl isostearate/water. Their study suggested
oped using lecithin/IPP/water. They demon- that the skin permeation of indomethacin
strated the reduction of the tumor mass was enhanced by fourfold when LOs was
developed in nude mice injected with a used as drug matrix as compared to isocetyl
Ha-ras-1 transformed cell line when treated isostearate-based suspension. They attributed
with LOs containing TAPP-Br. the higher skin penetration of indomethacin in
• Bhatnagar and Vyas (1994) have reported LOs due to higher solubility and better skin
LOs containing propranolol. LOs were devel- permeation of the drug in isocetyl isostearate
oped with lecithin/isooctane/water. They con- and lecithin.
ducted a comparative estimation of propranolol • Aboofazeli et al. (2002) have reported a sig-
permeation with LOs and petroleum jelly. nificant improvement in the transdermal deliv-
They inferred that the permeation rate of pro- ery of nicardipine with LOs as compared to
pranolol was increased by tenfold with LOs pure solvent-based formulations. LOs were
when compared with petroleum jelly. prepared by lecithin/isopropyl myristate/
310 I.M. Shaikh et al.

propylene glycol/oleic acid/dimethyl iso- pure ethyl oleate. Moreover, the paw edema
sorbide/water. The cumulative amount of test conducted in mice demonstrated LOs as a
nicardipine released from gel matrix was better formulation than hydrogels in treating
approximately threefold higher than that from the inflammation.
a tertiary solvent formulation (propylene gly-
col/oleic acid/dimethyl isosorbide) containing
nicardipine. This suggested that the gel matrix 21.11 Safety Profile of Lecithin
possesses a higher skin penetration enhance- Organogels (LOs) as Drug
ment effect compared to the pure solvent Carrier
formulation. The author attributed the skin
enhancement effect of LOs to lecithin as well Lecithin organogels (LOs) contain a fairly high
as the solvent mixture (propylene glycol/oleic level of surfactants and organic solvents.
acid/dimethyl isosorbide) Therefore, it is important to consider the safety
• Nasseri et al. (2003) developed LOs containing and irritancy of the formulation on prolonged
varied concentrations of lecithin/isopropyl use. Skin (human and mice) compatibility studies
myristate/water for topical application of using LOs have been evaluated and some exam-
ketorolac tromethamine (6.5 % w/w). They ples are reported below:
tested the in vitro release profile of various LO 1. Willimann et al. (1992) have conducted a com-
preparations containing ketorolac on guinea parative safety study of LOs containing isopro-
pig skin. Their study suggested that LOs pre- pyl myristate and control samples (physiological
pared with a lecithin/IPM weight ratio of 40:60 NaCl solution) on human skin using micro-
and 0.6 % w/w of water provided maximum scopic (light) investigation. During the study,
skin penetration of ketorolac tromethamine. human skin was treated with LOs and control
• Agrawal et al. (2004) have developed leci- sample and observed for 3 days. The outcome
thin-/IPM-/water-based LOs containing of the study indicated that there were no signifi-
piroxicam. They reported increased solubility cant alterations in the skin before and after treat-
of piroxicam in LOs and its improved in vitro ment of LOs and the LOs applied skin sample
skin permeation. In addition, they investi- was comparable with that of control samples.
gated the anti-inflammatory action of LOs 2. Dreher et al. (1996) has assessed the skin irri-
by conducting the carrageenan-induced rat tation potential of LOs (containing isopropyl
paw edema test and compared it with that of palmitate and lecithin) by conducting cumula-
marketed piroxicam gel. The outcome of the tive irritation tests. A cumulative irritation
rat paw edema test indicated that after 3 h of study was conducted by applying LOs in 20
study, the LOs are twofold more effective than volunteers. The volunteers were observed for
the marketed gel in reducing the edema. They skin irritancy for a period of 21 days. The
attributed this enhanced performance of LOs parameter “IT50” (irritation time of 50 % of
to the lecithin present in the formulation. test population) was used as an index for the
• Shaikh et al. (2009) have reported enhanced safety level. Results indicated a very low
skin penetration of aceclofenac from LOs as cumulative skin irritation potential of LOs
compared to aceclofenac from hydrogels. LOs (IT50 of 13 days) and suggested the suitability
were prepared using lecithin/ethyl oleate/ of LOs as a safe dermal and transdermal drug
water. They reported higher drug flux from delivery system for long-term applications.
LO as compared to hydrogels. It was proposed 3. The safety of ethyl oleate-based LOs in
that skin penetration enhancement of aceclof- hairless mouse skin was investigated by
enac could be due to lecithin and ethyl oleate conducting histopathological studies. The
in LOs. LOs not only provided increased pen- histopathological investigation of mouse skin
etration but also improved the solubility of after application of the LOs demonstrated that
aceclofenac by 13-fold as compared to the the formulation has no toxic effects on the
21 Lecithin Organogels in Enhancing Skin Delivery of Drugs 311

skin and all the surface epithelium lining and Aboofazeli R, Zia H, Needham TE (2002) Transdermal
delivery of nicardipine: an approach to in vitro perme-
the granular structure remained intact. This
ation enhancement. Drug Deliv 9(4):239–247, Epub
ensures the safety of LOs as a formulation for 2003/01/04
the delivery of drugs via the skin (Shaikh et al. Agrawal GP, Juneja M, Agrawal S, Jain SK, Pancholi SS
2009; Jadhav et al. 2009). (2004) Preparation and characterization of reverse
micelle based organogels of piroxicam. Pharmazie
59(3):191–193, Epub 2004/04/13
Almeida H, Amaral MH, Lobão P, Lobo JMS (2012)
Conclusion Pluronic® F-127 and pluronic lecithin organogel
Ease of preparation, biocompatible compo- (PLO): main features and their applications in topical
and transdermal administration of drugs. J Pharm
nents, the ability to accommodate hydrophobic
Pharm Sci 15(4):592–605
or hydrophilic micro/macromolecules, ther- Avramiotis S, Papadimitriou V, Hatzara E, Bekiari V,
moreversible nature, skin permeation-enhanc- Lianos P, Xenakis A (2007) Lecithin organogels used
ing ability, and long shelf life have resulted in as bioactive compounds carriers. A microdomain
properties investigation. Langmuir 23(8):4438–4447,
wide acceptance of LOs as dermal and trans-
Epub 2007/03/07
dermal drug delivery systems. The skin Bentley M, Kedor ER, Vianna RF, Collett JH (1997) The
permeation-enhancing ability of LOs is attrib- influence of lecithin and urea on the in vitro perme-
uted to both lecithin and the organic solvent ation of hydrocortisone acetate through skin from
hairless mouse. Int J Pharm 146(2):255–262
used for preparing LOs. The effect of addition
Bhatnagar S, Vyas S (1994) Organogel-based system for
of small amounts of conventional penetration transdermal delivery of propranolol. J Microencapsul
enhancers into LOs on skin penetration needs 11(4):431–438
to be investigated. Further, the effect of using Bonina F, Montenegro L, Scrofani N, Esposito E, Cortesi R,
Menegatti E et al (1995) Effects of phospholipid based
two organic solvents for preparing of LOs
formulations on in vitro and in vivo percutaneous absorp-
instead of one solvent on formulation attributes tion of methyl nicotinate. J Control Release 34(1):53–63
can also be explored, and its effect on skin per- Capitani D, Segre AL, Dreher F, Walde P, Luisi PL (1996)
meation can be studied. In addition, LOs have Multinuclear NMR investigation of phosphatidylcho-
line organogels. J Phys Chem 100(37):15211–15217
demonstrated their efficacy and safety as a drug
Couffin-Hoarau AC, Motulsky A, Delmas P, Leroux JC
delivery system at the lab scale. However, their (2004) In situ-forming pharmaceutical organogels
utility as a drug delivery system needs to be fur- based on the self-assembly of L-alanine derivatives.
ther demonstrated in clinical studies. For this Pharm Res 21(3):454–457, Epub 2004/04/09
Dasgupta D, Srinivasan S, Rochas C, Ajayaghosh A,
purpose, the full biocompatibility profile (effi-
Guenet JM (2009) Hybrid thermoreversible gels from
cacy in humans and long-term toxicity) of LOs covalent polymers and organogels. Langmuir
needs to be explored. Once clinically proven, 25(15):8593–8598, Epub 2009/03/19
LOs will emerge as a promising pharmaceuti- Díaz DD, Marrero Tellado JJ, Velázquez DG, Ravelo ÁG
(2008) Polymer thermoreversible gels from organo-
cal carrier system for enhancing the skin deliv-
gelators enabled by ‘click’ chemistry. Tetrahedron
ery of a wide range of drug candidates. Lett 49(8):1340–1343
Dreher F, Walde P, Luisi PL, Elsner P (1996) Human skin
irritation studies of a lecithin microemulsion gel and
of lecithin liposomes. Skin Pharmacol 9(2):124–129,
Epub 1996/01/01
Dreher F, Walde P, Walther P, Wehrli E (1997) Interaction
References of a lecithin microemulsion gel with human stratum
corneum and its effect on transdermal transport. J
Abdallah DJ, Sirchio SA, Weiss RG (2000) Control Release 45(2):131–140
Hexatriacontane organogels. The first determination Fujii M, Shiozawa K, Henmi T, Yamanouchi S, Suzuki H,
of the conformation and molecular packing of a low- Yamashita N et al (1996) Skin permeation of indo-
molecular-mass organogelator in its gelled state. methacin from gel formed by fatty-acid ester and
Langmuir 16(20):7558–7561 phospholipid. Int J Pharm 137(1):117–124
Aboofazeli R, Barlow DJ, Lawrence MJ (2000) Particle Grace D, Rogers J, Skeith K, Anderson K (1999) Topical
size analysis of concentrated phospholipid micro- diclofenac versus placebo: a double blind, randomized,
emulsions I. Total intensity light scattering. AAPS clinical trial in patients with osteoarthritis of the knee.
PharmSci 2(2):E13, Epub 2001/12/14 J Rheumatol 26:2659–2663
312 I.M. Shaikh et al.

Gronwald O, Snip E, Shinkai (2002) Gelators for organic Sato K, Sugibayashi K, Morimoto Y (1988) Effect and
liquids based on self-assembly: a new facet of supra- mode of action of aliphatic esters on the in vitro skin
molecular and combinatorial chemistry. Curr Opinion permeation of nicorandil. Int J Pharm 43(1):31–40
Colloid Interface Sci 7:148–156 Scartazzini R, Luisi PL (1988) Organogels from lecithins.
Guenet J-M (2006) Microfibrillar networks: polymer ther- J Phys Chem 92(3):829–833
moreversible gels vs organogels. Macromol Symp Schneider M (1997) Industrial production of phospholipids-
241(1):45–50 lecithin processing. Lipid Technol 9:109–116
Hanahan DJ (1997) A guide to phospholipid chemistry. Schurtenberger P, Scartazzini R, Luisi P (1989)
Oxford University Press, New York, USA Viscoelastic properties of polymerlike reverse
Jadhav KR, Kadam VJ, Pisal SS (2009) Formulation and micelles. Rheol Acta 28(5):372–381
evaluation of lecithin organogel for topical delivery of Schurtenberger P, Scartazzini R, Magid LJ, Leser ME,
fluconazole. Curr Drug Deliv 6(2):174–183 Luisi PL (1990) Structural and dynamic properties of
Jibry N, Heenan RK, Murdan S (2004) Amphiphilogels polymer-like reverse micelles. J Phys Chem 94(9):
for drug delivery: formulation and characterization. 3695–3701
Pharm Res 21(10):1852–1861, Epub 2004/11/24 Schurtenberger P, Peng Q, Leser M, Luisi PL (1993)
Kantaria S, Rees GD, Lawrence MJ (1999) Gelatin- Structure and phase behavior of lecithin-based micro-
stabilised microemulsion-based organogels: rheology emulsions: a study of the chain length dependence.
and application in iontophoretic transdermal drug J Colloid Interface Sci 156(1):43–51
delivery. J Controll Release 60(2–3):355–365, Epub Shaikh I, Jadhav S, Jadhav K, Kadam V, Pisal S (2009)
1999/07/30 Aceclofenac organogels: in vitro and in vivo charac-
Kumar R, Katare OP (2005) Lecithin organogels as a terization. Curr Drug Deliv 6(1):1
potential phospholipid-structured system for topical Shchipunov YA (1995) Lecithin organogels: rheological
drug delivery: a review. AAPS PharmSciTech properties of polymer-like micelles formed in the
6(2):E298–E310, Epub 2005/12/16 presence of water. Colloid J 57:556–560
Mahjour M, Mauser B, Rashidbaigi Z, Fawzi M (1990) Shchipunov YA (2001) Lecithin organogel: a micellar
Effect of egg yolk lecithins and commercial soybean system with unique properties. Colloids Surf A
lecithins on in vitro skin permeation of drugs. J Physicochem Eng Asp 183–185:541–554
Control Release 14(3):243–252 Shchipunov YA, Hoffmann H (1998) Growth, branching
McAllister K, Sazani P, Adam M, Cho MJ, Rubinstein M, and local ordering of lecithin polymer-like micelles.
Samulski RJ et al (2002) Polymeric nanogels pro- Langmuir 14:6350–6360
duced via inverse microemulsion polymerization as Shchipunov YA, Hoffmann H (2000) Thinning and thick-
potential gene and antisense delivery agents. J Am ening effects induced by shearing in lecithin solutions
Chem Soc 124(51):15198–15207 of polymer-like micelles. Rheol Acta 39(6):542–553
Moore J (1982) Final report on the safety assessment of Shchipunov YA, Mezzasalma SA, Koper GJM, Hoffmann
octyl palmitate, cetyl palmitate and isopropyl palmi- H (2001) Lecithin organogel with new rheological and
tate. J Am Coll Toxicol 1:13–35 scaling behavior. J Phys Chem B 105:10484–10488
Motulsky A, Lafleur M, Couffin-Hoarau AC, Hoarau D, Shchipunov YA, Shumilina EV (1995) Lecithin bridging
Boury F, Benoit JP et al (2005) Characterization and by hydrogen bonds in the organogel. Mater Sci Eng C
biocompatibility of organogels based on L-alanine for 3(1):43–50
parenteral drug delivery implants. Biomaterials Shchipunov YA, Shumilina E (1996) Lecithin organogels:
26(31):6242–6253, Epub 2005/05/27 role of polar solvent and nature of intermolecular
Murdan S (2005) Pluronic lecithin organogels: a review of interactions. Colloid J Russ Acad Sci 58(1):117–125
a unique topical drug delivery system. Hosp Pharm Shchipunov YA, Schmiedel P (1996) Phase behavior of
12(7):267–270 lecithin at the oil/water interface. Langmuir
Nasseri AA, Aboofazeli R, Zia H, Needham TE (2003) 12:6443–6445
Lecithin–stabilized microemulsion–based organogels Shchipunov YA, Schmiedel P (1996) Electrorheological
for topical application of ketorolac tromethamine. phenomena in lecithin-decane-water mixtures. J
II. In vitro release study. Iran J Pharm Res 2:117–123 Colloid Interface Sci 179:201–206
Nastruzzi C, Gambari R (1994) Antitumor activity of Shchipunov YA, Shumilina E, Hoffmann H (1998)
(trans) dermally delivered aromatic tetra-amidines. J Lecithin organogels with n-alkyl-D-glucosides and
Control Release 29(1):53–62 n-alkly-D-lactobionamide. Colloid Polym Sci
Natsuki R, Takabatake E (1987) Effect of lecithin on per- 276(4):368–372
cutaneous absorption of drugs. II. Mechanism of Shikata T, Ogata D, Hanabusa K (2003) Viscoelastic
enhancing effect of lecithin on the percutaneous behavior of organogels. Riron Oyo Rikigaku Koenkai
absorption of indomethacin gel-ointment. Yakugaku Koen Ronbunshu 52:477–478
Zasshi 107(8):622–626 Simmons BA, Taylor CE, Landis FA, John VT, McPherson
Reynolds JEE (1996) Martindale, the extra pharmaco- GL, Schwartz DK et al (2001) Microstructure determina-
poeia, 31st edn. Royal Pharmaceutical Society, London tion of AOT + phenol organogels utilizing small-angle
21 Lecithin Organogels in Enhancing Skin Delivery of Drugs 313

X-ray scattering and atomic force microscopy. J Am Wade A, Weller PJ (1994) Handbook of pharmaceutical
Chem Soc 123(10):2414–2421, Epub 2001/07/18 excipients. 2nd ed. Washington: Ame Pharm
Szuhaj BF (1989) Lecithins: sources, manufacture & uses. Association
Amer Oil Chemists Society, Champaign; Illinois, USA Walde P, Giuliani AM, Boicelli CA, Luisi PL (1990)
Terech P (1985) Kinetics of aggregation in a steroid deriv- Phospholipid-based reverse micelles. Chem Phys
ative/cyclohexane gelifying system. J Colloid Interface Lipids 53(4):265–288, Epub 1990/03/01
Sci 107(1):244–255 Wendel A (1995) Kirk-Othmer encyclopedia of chemical
Terech P, Weiss RG (1998) Low molecular mass gelators technology, vol 15. Wiley, New York
of organic liquids and the properties of their gels. Willimann HL, Luisi PL (1991) Lecithin organogels as
ChemInform. 29(11):3133–3159 matrix for the transdermal transport of drugs. Biochem
Toshiyuki S, Daisuke O, Kenji H (2003) Viscoelastic Biophys Res Commun 177(3):897–900, Epub
behavior of organogels Riron Oyo Rikigaku Koenkai 1991/06/28
Koen Ronbunshu, 52:477–478 Willimann H, Walde P, Luisi PL, Gazzaniga A, Stroppolo
van Esch JH, Feringa BL (2000) New functional materials F (1992) Lecithin organogel as matrix for transdermal
based on self-assembling organogels: from serendipity transport of drugs. J Pharm Sci 81(9):871–874, Epub
towards design. Angew Chem Int Ed 39(13):2263–2266 1992/09/01
Voit A, Shchipunov YA (2000) Dynamics of polymer-like Zemb T, Barnes I, Derian P, Ninham B (1990) Scattering
lecithin micelles. Rheological measurements. Colloid as a critical test of microemulsion structural models.
J 62(4):424–430 Trends Colloid Interface Sci IV 81:20–29
Thermosensitive Hydrogels
in Dermatology: 22
A Multidisciplinary Overview

Damien Salmon, Laurène Roussel, Elodie Gilbert,


Plamen Kirilov, and Fabrice Pirot

Contents 22.3 Formulation of Thermosensitive


Polymers . . . . . . . . . . . . . . . . . . . . . . . . . 322
22.1 Introduction . . . . . . . . . . . . . . . . . . . . . . 315
22.3.1 Gel Preparations. . . . . . . . . . . . . . . . . . . . 322
22.2 Thermosensitive Polymers . . . . . . . . . . 317 22.3.2 Carrier Systems . . . . . . . . . . . . . . . . . . . . 323
22.2.1 General Considerations About
22.4 Dermatological Applications. . . . . . . . . 323
Hydrogels. . . . . . . . . . . . . . . . . . . . . . . . . 317
22.4.1 Topical Delivery. . . . . . . . . . . . . . . . . . . . 323
22.2.2 General Considerations About
22.4.2 In Situ Gelling Systems . . . . . . . . . . . . . . 325
Thermosensitive Hydrogels . . . . . . . . . . . 318
22.4.3 Iontophoresis and Electroporation . . . . . . 326
22.2.3 Physicochemical Characteristics
22.4.4 Subcutaneous Surrogates . . . . . . . . . . . . . 326
of Thermosensitive Hydrogels. . . . . . . . . 318
22.2.4 Rheological Properties of Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 326
Thermosensitive Hydrogels . . . . . . . . . . . 318
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 327

D. Salmon • F. Pirot (*) 22.1 Introduction


Laboratoire de Pharmacie Galénique
Industrielle – Faculté de Pharmacie,
EA 4169 “Aspects fondamentaux, cliniques et The hospital triad formed by the dermatologist,
thérapeutique de la fonction barrière cutanée”, the pharmacist, and the nurse is involved in the
8, avenue Rockefeller, Lyon 69373, France prescription, the preparation, and the administra-
Service Pharmaceutique – Unité de préparation et de tion of adapted, efficient, and safe medicines.
contrôle des médicaments, Groupement Hospitalier Each professional defines specific requirements
Edouard Herriot, Plateforme FRIPHARM- Hospices
Civils de Lyon, 5, Place d’Arsonval, Lyon 69437,
concerning dermatological treatments which
France might be schematically conceptualized in
e-mail: damien.salmon01@chu-lyon.fr; fabrice. Fig. 22.1. However, many medical and nursing
pirot@chu-lyon.fr; http://www.fripharm.com requirements concerning topical treatments
L. Roussel • E. Gilbert imply, for hospital pharmacist stuck “between the
Laboratoire de Pharmacie Galénique
Industrielle – Faculté de Pharmacie,
EA 4169 “Aspects fondamentaux, cliniques et
P. Kirilov
thérapeutique de la fonction barrière cutanée”,
Laboratoire de Pharmacie Galénique
8, avenue Rockefeller, Lyon 69373, France
Industrielle – Faculté de Pharmacie,
Faculty of Medicine and Pharmacy, EA 4169 “Aspects fondamentaux, cliniques
University Lyon 1, Lyon, France et thérapeutique de la fonction barrière cutanée”,
e-mail: laurene.roussel@univ-lyon1.fr; elodie. 8, avenue Rockefeller, Lyon 69373, France
gilbert@universite-lyon.fr e-mail: plamen.kirilov@univ-lyon1.fr

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 315


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_22, © Springer-Verlag Berlin Heidelberg 2015
316 D. Salmon et al.

Fig. 22.1 The hospital triad Drug solubility


involved in dermatological Drug-excipent interaction
medicine management and Sterility
major outcome that should be Pharmacist
Pharmaceutical process
considered by each Conditionning
protagonist Compatibility with dressing

Patient

Dermatologist Nurse

Easy handling
Drug
Single dose unit
Dosage
Unique daily application
Bioavailability
Easy removal
Tolerance and efficacy
Easy washing
No stain/color

mortar and the pestle”, numerous practical times located in foreign country limiting
considerations. importation and/or exportation of pharmaceutical
Firstly, active pharmaceutical ingredients products, (3) not permanently approved by local
(APIs), currently prescribed by dermatologist, health and safety regulatory authorities, and (4)
exhibit low aqueous solubility (e.g., class II, not scaled for small production, packaging, and
diclofenac; class IV, sulfadiazine) and low per- shipment of excipients and APIs to health-care
meability (e.g., class III, acyclovir, and class V, hospital or clinical establishments.
acetazolamide) which complicate the selection of Fourthly, the safety of excipients is recurrently
excipients for compounding. Therefore, pre-for- questioned by authorities from the analysis of
mulation studies are usually necessary for screen- notable adverse effects imputable to excipients
ing appropriate excipients when compounded shifting their status from inactive to mystery
preparations prescribed are not detailed or ingredients, reducing again the width of the field
indexed in the Pharmacopeia and the Formulary of choice (Noiles and Vender 2010).
(i.e., national compendia for chemical and bio- Fifthly, the conservation, the packaging, and
logical drug substances, dosage forms, and com- storage of topical formulations is a major con-
pounded preparations, excipients, medical cern since the use and reuse of the preparation is
devices, and dietary supplements). Both national an obvious source of human and exogenous con-
compendia display substantial heterogeneity in tamination, a factor of physicochemical degrada-
their contents over the world. tion (e.g., hydrolysis, oxidation) of APIs and
Secondly, although medical and scientific lit- excipients, and an issue for formulation instabil-
erature detailed many original excipients, alone ity (e.g., syneresis, creaming, sedimentation).
or in combination, enabling the formulation of Again, the degree of purity of excipients, from
APIs for topical treatment, few topical prepara- different origins (i.e., from biological or mineral to
tions are reported in the Pharmacopeia and the chemical-based synthesis), is often weakened by
Formulary. concomitant components or processing aids, and
Thirdly, the suppliers of pharmaceutical grade the final use of excipients is not always known by
excipients necessary for dosage forms and com- the supplier. Therefore, the choice of appropriate
pounded preparations are (1) scarce, (2) some- excipients for topical compounding is also a com-
22 Thermosensitive Hydrogels in Dermatology: A Multidisciplinary Overview 317

promise between pharmaceutical state of the art intrinsic advantages of thermosensitive hydrogel
and the regulatory and availability status of excipi- are as follows: (1) high water content, (2) solubi-
ents. Facing (i) the pharmaceutical compounding lizing properties for hydrophobic APIs, (3) con-
challenge, (ii) the inherent restrictions of available, trol of swelling properties and gelling
authorized, and harmless excipients, (iii) the pack- temperature, (4) adaptation for tailor-made for-
age features, surely, the simplest drug-excipient mulations in specific dermatologic diseases, and
combination for ready-to-use and easy-handling (5) versatile skin drug delivery from either sur-
product is highly needed for the formulation devel- face application, intradermal or subcutaneous
opment of topicals. injection.
However, the pharmacist experiences that, at In the followings sections, physicochemical
some points, the development of topical prepara- properties of current and innovative thermosensi-
tion leads to consider top-ten recommendations: tive polymers are presented, and then the actual
1. Avoiding the use of many excipients, to pre- and prospective dermatological applications of
fer straightforward process of preparation thermosensitive polymer-based formulations are
where APIs are quickly dissolved, miscible, emphasized.
or suspended in aqueous solvent supple-
mented by not more than three excipients
2. To choose excipients insuring both physical 22.2 Thermosensitive Polymers
and chemical stability of APIs, excipients,
and formulation 22.2.1 General Considerations About
3. To reduce pH variation of formulation over Hydrogels
time during skin exposure (skin surface – pH
~5.5) The gelation in the aqueous solvent is a complex
4. To check the probability to reuse and to phenomenon where a polymer initially soluble in
avoid contamination of formulation water becomes more hydrophobic by (1) interac-
5. To guarantee easy spreading and removal, tion with mineral ions (e.g., gellan gum, natural
sustainability, and aesthetical acceptability anionic heteropolysaccharide, sodium alginate,
(i.e., feel, color, fragrance, absorbability) of natural polysaccharide), (2) variation of pH (e.g.,
formulation polymers carrying carboxylic acid, phosphoric
6. To permit optimal API penetration into skin acid, and amine groups) leading to a change of con-
structures (dermal delivery) formation and swelling behavior (Schmaljohann
7. To permit optimal API permeation through 2006), or (3) modification of temperature. As a
skin structures (transdermal delivery) result, a transparent or translucent semisolid poly-
8. To favor or to limit the buildup of APIs and meric matrix is obtained where the fluid flow is
excipients into the skin limited by entrapment and immobilization of the
9. To improve the cutaneous tolerance to APIs solvent molecules and possesses remarkable
and excipients mechanical properties (deformation, viscoelastic
10. To improve the efficacy of APIs into the skin properties) which facilitate further cutaneous
or after percutaneous delivery spreading.
Therefore, few excipients might fulfill prereq- The regional ionic strength upon the outer-
uisites detailed above. Among likely candidates, most layer of the skin, the stratum corneum, is
excipients forming thermosensitive (also called likely insufficient to elicit gelation with ionic-
thermoresponsive or thermoreversible) hydrogels responsive polymers (i.e., making necessary pre-
offer many advantages which have been exten- gelation of formulation containing appropriate
sively detailed in reviews published in the last ionic strength) (Aust et al. 2012), while acidic pH
decade (Jeong et al. 2012; Klouda and Mikos (~5.5) at the skin surface do not allow a gelation
2008; Ruel-Gariépy and Leroux 2004). The main of common acidic polymer (e.g., carbomer).
318 D. Salmon et al.

Besides, the regulation of body temperature, one importance of natural and modified natural poly-
of the major skin functions in homeostasis, might mers in the dermatological research and subse-
be exploited for the successful development of quent clinical outcomes. Although well known and
thermosensitive hydrogels. considerably used, cellulose derivatives and chito-
Moreover, interactions between skin and ther- san still motivate some intellectual property issues.
mosensitive polymers have been of growing
interest in the past decades as (1) intimate proper-
ties and mechanics of such polymers were gradu- 22.2.3 Physicochemical
ally documented and (2) skin is regarded as a Characteristics
promising alternative to traditional oral or paren- of Thermosensitive Hydrogels
teral routes for the administration of active phar-
maceutical ingredients. Furthermore, interesting Many thermosensitive polymers are amphiphilic
parallels between skin or subcutaneous tissues block copolymers with A-B or A-B-A type struc-
and hydrogels in terms of chemical and physical tures (A: hydrophilic block; B: hydrophobic block)
characteristics draw exciting perspectives for that will form micelles in aqueous solvents.
future developments in experimental and clinical Polyethylene glycol (PEG) is commonly used as
fields (Lee et al. 2009). hydrophilic block due to biocompatibility and
high water solubility. Hydrophobic block usually
forms the drug-binding core of the system in
22.2.2 General Considerations About hydrogels showing a large variety of structures
Thermosensitive Hydrogels such as polypropylene glycol and polyesters such
as poly(lactide-co-glycolide) and poly(lactide-co-
The ability for a solution of polymer to modify caprolactone) (Rijcken et al. 2007). Usually, poly-
its bulk viscosity in response to temperature vari- mer concentration in solvent will determine
ation is called thermosensitivity. Generally natu- gelation temperature (Lenaerts et al. 1987). Nature
ral polymer solutions form gels at low and ionic content of thermosensitive polymer gel
temperature and liquefy when temperature rises, solvents is of great importance as it can signifi-
but chemically modified polymers or synthetic cantly modify drug release properties (Pandit and
polymers may exhibit opposite behavior defined Wang 1998; Ur-Rehman et al. 2010) and gelation
as reverse thermosensitivity. As the physical ability (Pandit and Kisaka 1996). Also, ionic con-
state (i.e., free flowing or non-flowing during tent has an influence on pH and drug ionization
usage time) can be controlled by thermal modu- state which is an important issue to consider
lation, formulas containing those polymers may regarding skin-formulation interactions.
have innovating pharmaceutical applications due
to control of solute transport abilities and bio-
compatibility. Various polymeric molecules 22.2.4 Rheological Properties
exhibit thermosensitive properties such as natu- of Thermosensitive Hydrogels
ral polymers (e.g., gelatin, agarose, carrageen-
ans), modified natural polymers (e.g., cellulose The mechanism of this viscosity change, called
derivatives, chitosan, dextran, xyloglucan), syn- sol→gel transition when viscosity increases or
thetic polymers (e.g., N-isopropylacrylamide gel→sol transition when it decreases, is dependent
and its copolymers), or poloxamers (i.e., on molecular interactions within the polymer solu-
poly(ethylene oxide)/poly(propylene oxide), tion. Natural polymer solutions mostly form a gel
polyethylene glycol/polyester copolymers) phase when temperature is low and liquefy on heat-
(Table 22.1) (Jeong et al. 2012; Klouda and ing. In these hydrogels, polymer molecules arrange
Mikos 2008; Ruel-Gariépy and Leroux 2004). in a partial helicoidal architecture that destructures
Figure 22.2 shows the literature content and pat- in a random coil conformation when temperature
ent applications over the last decade underlying the rises (Ruel-Gariépy and Leroux 2004).
Table 22.1 Structures and formulas of polymers currently used for thermosensitive hydrogel formulations in commercial products or pharmaceutical compounding
22

Molecular weight CAS


Polymers Formula (g.mol−1) USP/Eur. Ph number
Gelatin② e.g., -Ala-Gly-Pro-Arg-Gly-Glu-4Hyp-Gly-Pro- 15,000–250,000 Referenced/ 9000-70-8
referenced
Agarose② (Agar) 306 • n Referenced/ 9063-31-4
OH OH referenced
O
O
OH
O O
H O
OH
HO
n
Carrageenans② 451 • n Referenced/not 9000-07-1
OH 6´ referenced
OSO3-
6
4 O 1´
5 O 5´
2 O O
3 1 O 2´

OH 4´
OSO3-
n
Cellulose derivatives
OR
H H H
OR CH2
O O
Thermosensitive Hydrogels in Dermatology: A Multidisciplinary Overview

RO
H H
H H
O O RO
CH2 OR
H H H
OR
n
R = -H, -CH3, -CH2CH2OH,-CH2CH(OH)CH3 -CH2OCH2COONa
(continued)
319
Table 22.1 (continued)
320

Molecular weight CAS


Polymers Formula (g.mol−1) USP/Eur. Ph number
Methylcellulose④ 10,000–220,000 Referenced/ 9004-67-5
referenced
Hydroxyethylcellulose② – Referenced/ 9004-62-0
referenced
Hydroxypropyl- 50,000–1,250,000 Referenced/ 9004-64-2
cellulose① referenced
Hydroxyethyl – Not referenced/not 9032-42-2
methylcellulose① referenced
Hydroxypropyl- 10,000–1.500,000 Not referenced/ 9004-65-3
methylcellulose① referenced
Carboxymethyl 90,000–700,000 Referenced/ 9004-32-4
cellulose sodium② referenced
Chitosan② 320 • n Not referenced/ 9012-76-4
referenced
H NH2 H
H HO
HOH2C C
O
O
H H
H H
O
O
C CH2OH
HO H NH2 H
H

n
Poloxamers CH3

HO O H
O O
a b a

124 a: 10–15; b: 18–23 2,090–2,360 Referenced/ 9003-11-6
referenced
188③ a: 75–85; b: 25–30 7,680–9.510 Referenced/ 9003-11-6
referenced
237③ a: 60–68; b: 35–40 6,840–8,830 Referenced/ 9003-11-6
referenced
D. Salmon et al.
22

338③ a: 137–146; b: 42–47 12,700–17,400 Referenced/ 9003-11-6


referenced
407③ a: 95–105; b: 54–60 9,840–14,600 Referenced/ 9003-11-6
referenced
Polyesters copolymersa 2,000–100,000 or Not referenced/not
higher referenced
Poly(lactide- 30846-39-0
co-glycolide) O 26780-50-7

O
O

CH3 O

X y

Poly(lactide- 70524-20-8
co-caprolactone) O
65408-67-5
O
O

CH3 O
x y
Reference of polymers in the United States (29) and European Pharmacopoeia (7.6) as monographs is reported as well as chemical abstract service (CAS) numbers. Dynamic
viscosity behavior as a function of heating is reported as follows: ① sol→gel transition, ② gel→sol transition, ③ sol→gel→sol transitions, and ④ gel→sol→gel transitions
a
Hydrophobic moieties of polyester copolymers are presented as poly(lactide-co-glycolide) and poly(lactide-co-caprolactone). Hydrophilic moieties linked to hydrophobic
Thermosensitive Hydrogels in Dermatology: A Multidisciplinary Overview

polyesters determining thermosensitive properties are, e.g., polyethylene glycol blocs


321
322 D. Salmon et al.

10000 PubMed ScienceDirect Espacenet


3039
1850 1850 1462
905 886 1111
1000 573

212
115 101
100 62
39 43 41
23 28 23 27 24 29
13
10
10 6 6 5

PEG/polyester copolymer
N-isopropylacrylamid

poloxamer (PEG/PPO)
cellulose derivatives
carrageenan

xyloglucan
agarose

chitosan

dextran
Gelatin

natural polymers modified natural polymers synthetic polymers

Fig. 22.2 Review of the literature was built by using bib- the growing interest for synthetic polymers, research stud-
liographic (PubMed and ScienceDirect) and patent ies, medical applications, and intellectual property still
research (Espacenet) databases running “polymer (e.g., focus on natural and modified natural thermosensitive
gelatin) and skin” keywords for the last decade. In spite of polymers

Oppositely, reverse thermosensitive polymer used to determine transition temperature by


solutions are liquid when temperature is low and detection of gelation endothermic peak and also
gelify on heating. Temperature variations modify gives information about enthalpy of gelation.
the affinity (i.e., hydrophilicity, hydrophobicity) Eventually, dynamic mechanical analysis by a
between polymer solution components (i.e., poly- rheometer in a thermally controlled environment
mer molecules and solvent molecules) resulting in enables to obtain precise data about sol→gel and
changes in the intimate architecture of the solu- gel→sol transition (Jeong et al. 2012).
tion. Globally, an increase in polymer’s solvent
solubility results in low viscosity (sol), whereas a
decrease in polymer’s solvent solubility results in 22.3 Formulation
polymer molecule network formation and there- of Thermosensitive Polymers
fore gelation (Klouda and Mikos 2008).
Experimental determination of the boundary 22.3.1 Gel Preparations
between sol and gel states is compulsory to ther-
mosensitive formulation characterization. The Semisolid formulations, commonly called gels,
simplest method is the tube inversion method are convenient for dermatological applications as
where a test tube containing polymer solution is they may incorporate various APIs within their
tilted to evidence formulation flow under differ- internal network structure and provide physical
ent temperature conditions with standardized test stability towards (1) sedimentation, (2) creaming,
parameters (i.e., time, tilting rate, tube diameter, and (3) flocculation due to high viscosity. Nature
and amount of solution). Another simple method is rich in semisolid elements (e.g., connective
is the falling ball method where a small dense tissues, extracellular matrix), and many natural
ball is deposited on the top of the gelified poly- polymers can be used in pharmaceutical
mer solution. The temperature point at which the formulations. Relatively recent concerns about
ball penetrates the solution under standardized the environment have led to the discard of
conditions is the gel→sol transition temperature. petroleum-based synthetic polymers despite their
Differential scanning calorimetry (DSC) may be low-cost and biochemical inertness. Modern
22 Thermosensitive Hydrogels in Dermatology: A Multidisciplinary Overview 323

analytical methods enable to understand the Recently, Choi et al. described a thermosensi-
microstructure/function relation in polymers and tive polymer-based nanocarrier for transcutane-
open the way to optimized biocompatible modi- ous protein delivery. A chitosan-poloxamer
fied natural polymers (Yu et al. 2006). 407-conjugated nanocarrier was able to effi-
Chen et al. described a PEG/poly(lactide-co- ciently encapsulate insulin due to thermosensi-
glycolide) hydrogel matrix meant for prolonged tive swelling and surfactant properties of
subcutaneous delivery of porcine growth hor- poloxamer 407. Enhanced cutaneous permeation
mone (pGH). The preparation was very simple, was allowed by chitosan’s properties (i.e., loos-
consisting in a single homogenization of polymer ening of keratin structure in the stratum corneum
and pGH in aqueous solution. Subcutaneous and transient opening of tight junctions) (Choi
injection in rabbits of low dose (0.12 %) and high et al. 2012).
dose (0.42 %) pGH-loaded hydrogel matrix Niosome systems formed by poloxamers
enabled constant prolonged delivery of pGH for allowed to improve skin permeation of class IV
about 4 weeks, i.e., 5–15-fold that of aqueous APIs (sulfadiazine) (Muzzalupo et al. 2001), while
solution for subcutaneous injection. Cell viability chlorhexidine-loaded poly-ε-caprolactone nano-
study in growth media containing polymer extract capsule suspension in thermosensitive hydrogel
was the same as control (Chen and Singh 2008). (i.e., carboxymethylcellulose or poloxamers) was
Furthermore, poloxamers can be used to proven efficient in sustaining antimicrobial activ-
enhance aqueous solubility of poorly soluble ity after an unique application onto the skin sur-
drugs such as anti-inflammatory drugs. In an ear- face (Nhung et al. 2007; Pirot and Falson 2009).
lier study, poloxamer 407 aqueous formulation Interestingly, poly-ε-caprolactone nanocapsules
containing various cosolvents and surfactants might be freeze-dried and rehydrated in situ in the
allowed a 2,000-fold increase in tolfenamic acid presence of moisture on the skin surface or wounds
solubility. The release profile of API was inde- and reform locally thermosensitive hydrogel
pendent of bulk viscosity suggesting dependence (unpublished data, Fig. 22.3).
on gel microstructure. Solvent composition is
known to influence gel microstructure and there-
fore can be adapted to obtain desired drug release 22.4 Dermatological Applications
profile from poloxamer 407 hydrogel formula-
tions (Cafaggi et al. 2008; Ivanova et al. 2001). Hydrogels are easy to formulate and handle,
enabling interesting drug therapy applications
in dermatology (Fig. 22.4). Consequently,
22.3.2 Carrier Systems thermosensitive polymers are currently used in
(1) cutaneous, (2) intradermal, or (3) subcutane-
Polymer incorporation into structures of vesicu- ous commercial formulation (Fig. 22.5). At the
lar systems enables to produce nano- or opposite, in developing countries, compounding
microscaled delivery systems that respond to dermatologic preparations for treating the most
environmental signal (e.g., light, temperature, common dermatologic disorders revealed a
chemicals, and biomolecules). Thermosensitive minimal use of thermosensitive polymers as
polymers are especially interesting in this per- compared to, e.g., white petrolatum (Lamarre
spective, and their properties can be combined to et al. 2009).
other responsive polymers (e.g., photosensitive,
pH sensitive) to produce complex vesicular sys-
tems responding to multiple external signals. 22.4.1 Topical Delivery
Desired properties of such polymers are (1) high
selectivity in stimuli recognition, (2) amplifica- Heilmann et al. investigated thermosensitive
tion of the signal, and (3) transduction of the sig- hydrogels (i.e., poloxamer and hydroxyethyl-
nal into changes in the system’s properties cellulose) for opioid local delivery in patients
(Motornov et al. 2010). with severe skin wounds. Such formulation
324 D. Salmon et al.

Natural light UV light (365 nm)

1. Freeze-dried rhodamine base loaded poly-ε-caprolactone nanocapsule suspension in a


poloxamer 188 hydrogel colored with methylene blue. Rhodamine base produces yellow
fluorescence under 365 nm UV light.

2. Rhodamine base loaded poly-ε-caprolactone nanocapsule suspension obtained by


reconstitution of freeze-dried product presented hereabove.

3. Skin application of reconstituted suspension presented hereabove.

Fig. 22.3 Photographic pictures under (1) natural light 188 hydrogel after (step 1) freeze-drying, (step 2) resus-
and (2) UV light (365 nm) of rhodamine-base-loaded pension in water, and (step 3) application onto the skin
poly-ε-caprolactone nanocapsule suspension in poloxamer

should (1) be able to deliver pain relieving ficial skin” in the treatment of severe burns.
amounts of morphine for a long period of time, A 25 % poloxamer 407 hydrogel was found to
avoiding multiple dressing changes and should be an efficient topical formulation vehicle ena-
(2) not impair wound healing. Hydrogels are beling 24 hours sustained release of morphine
known to provide a moist environment that in addition to be conveniently applicable and
favors skin reparation, and poloxamer gels no impairing natural skin healing (Heilmann
have previously been described as potent “arti- et al. 2013).
22 Thermosensitive Hydrogels in Dermatology: A Multidisciplinary Overview 325

Fig. 22.4 Schematic view of Iontophoresis Intradermal injection


possible skin-related
administrations of API-loaded + – Topical application
thermosensitive gel (blue Subcutaneous injection
spot) showing dermatological Epidermis
interest (Presented view is
adapted from http://
whgormanmd.com/ Systemic uptake
plastic-surgery-educational- Dermis
images/)

Hypodermis

50
Number of commercial formulations

45
40
35
30
25
20
15
10
5
0
cellulose poloxamers polyester gelatin chitosan carrageenans agarose
derivatives copolymers

Fig. 22.5 Histogram showing the number of approved (1) cutaneous, (2) intradermal, or (3) subcutaneous commercial
formulations containing thermosensitive polymers (Source www.theriaque.org)

22.4.2 In Situ Gelling Systems tration as a consequence of temperature


dependent rise of polymer hydrophobicity
In situ gelling systems are liquid aqueous (Ruel-Gariépy and Leroux 2004). Local or sys-
formulations that gelify under physiological temic controlled long-lasting delivery of APIs
conditions. They have a wide range of pharma- is therefore made possible with minimal
ceutical and medical applications, notably administration invasivity and frequency.
for (1) drug delivery, (2) cell encapsulation, Applications in local anticancer therapy (Desai
and (3) tissue repair. Thermosensitive in situ et al. 2008) or hormone systemic delivery for
gelling systems are polymeric solutions under- contraception or substitution (Yoshida et al.
going rapid change in viscosity upon adminis- 1991) are described in literature.
326 D. Salmon et al.

22.4.3 Iontophoresis 22.4.4 Subcutaneous Surrogates


and Electroporation
Hydrogels exhibit physical similarities with
Iontophoretic delivery is a promising alternative human soft tissues. The interstitial space of all tis-
to parenteral administration of active com- sues, including skin, is composed of collagen
pounds with poor skin permeation potential, fibers supporting fully charged glycosaminogly-
especially of high-molecular-weight molecules can hydrogel at physiological pH (Porter et al.
like proteins. However, technical issues need to 2001). Physicochemical characteristics (e.g.,
be resolved to make this administration method osmolarity, pH, density, rheology, water content)
clinically usable. Poloxamer 407 hydrogel con- of such hydrogel is possible to approach in vitro
taining insulin and permeation enhancers has using previously described polymers, such as aga-
been developed by Pillai et al. answering many rose, and adjusting their characteristics with adju-
of these technical issues. The reverse thermo- vants. Recently, Ostergaard’s group described
sensitive nature of this formulation enabled piroxicam diffusion from an oil solution into a
facilitated preparation and storage between poloxamer 407 and an agarose gel meant as sub-
+2 °C and +8 °C as it would remain liquid at cutaneous matrix surrogates. Piroxicam diffusion
such temperature. Application onto the skin and inside the hydrogels was monitored in real time
compatibility with iontophoresis device as the by UV imaging (Ye et al. 2012a, b). Yet unpub-
cool solution flowed easily onto the skin, form- lished data from our group have reported the
ing a non-occlusive gel when reaching body effect of poloxamer 407 and hyaluronic acid on
temperature with excellent contact with the skin porcine ear stratum corneum and dermis hydra-
and device. Furthermore, the adaptability of tion and water evaporation. Significant dehydra-
electrical conductivity and pH of hydrogels and tion of skin samples was evidenced with 30 %
their ability to absorb sweat gland secretions, poloxamer 407 gel as compared to 0.5 % hyal-
avoiding irritation under long-term occlusion, uronic acid or 0.9 % sodium chloride solution.
enabled ideal iontophoretic conditions (Pillai The implication on wound healing after gastric
and Panchagnula 2003). endoscopic mucosal resection using poloxamer
Electroporation consists in applying an elec- gels (Fernandez-Esparrach et al. 2009) is dis-
tric field to the skin in order to temporarily cre- cussed. Such experimental approaches could lead
ate pores in the stratum corneum enabling skin to better understanding and in vitro characteriza-
permeation of unabsorbable molecules. tion (e.g., molecular transport, tissular effect) of
Following the administration process, resealing pharmaceutical products meant for subcutaneous
of the pores to restore skin barrier function is or submucosal injection.
desired to prevent further unwanted absorption.
Poloxamer 188 solution applied after electro- Conclusion
poration procedure in vitro enabled (1) signifi- Thermosensitive polymeric systems forming
cantly more rapid and efficient epidermal electric hydrogels offer numerous applications in the
resistance recovery and (2) rapid skin permea- field of skin-related therapy. Their versatility
bility impairment of witness hydrophilic com- enables to consider various delivery systems,
pound (i.e., glucose). The amphiphilic nature of from a simple gel to complex nanostructured
poloxamer is believed to be responsible for this carriers. Moreover, they are relatively low-
effect because of its ability to interact with skin cost ingredients and might conveniently be
lipids and therefore facilitates their reassembly selected and adapted to match suitable
from electroporation-induced vesicular struc- properties.
tures to physiological lamellar structures Conventional natural (e.g., gelatin) or modi-
(Burgess et al. 2007). fied natural polymers (e.g., cellulose derivatives)
22 Thermosensitive Hydrogels in Dermatology: A Multidisciplinary Overview 327

are the most widely used polymers in hospital Heilmann S, Kuchler S, Wischke C, Lendlein A, Stein C,
pharmacy daily practice and still provide the Schaefer-Korting M (2013) A thermosensitive
morphine-containing hydrogel for the treatment of
majority of literature content related to hydro- large-scale skin wounds. Int J Pharm 444:96–102
gels. However, the growing interest in biocom- Ivanova R, Alexandridis P, Lindman B (2001) Interaction
patible synthetic polymers such as poloxamers of poloxamer block copolymers with cosolvents and
and the ability to control intimately their micro- surfactants. Colloids Surf A Physicochem Eng Asp
183–185:41–53
structure and microreactivity make them an Jeong B, Kim SW, Bae YH (2012) Thermosensitive sol-
exciting object of study and application. The gel reversible hydrogels. Adv Drug Deliv Rev 64:154–
amphiphilic nature of these block copolymers 162, Supplement
supports subsequent capacity to interact with a Klouda L, Mikos AG (2008) Thermoresponsive hydrogels
in biomedical applications. Eur J Pharm Biopharm
large range of API classes (including class IV 68:34–45
compounds and proteins of therapeutical inter- Lamarre D, Bertrand ME, Giroux D, Nordlund JJ, Ertle J,
est) and many human body tissues and compo- Charles AJ (2009) Compounding dermatologic prepa-
nents. In the future, hydrogels, oleogels, and rations in developing countries. Dermatol Ther 22(6):
560–563
composite systems with responsive capacities to Lee SJ, Pishko GL, Astary GW, Mareci TH, Sarntinoranont
physiological stimuli will multiply pharmaceuti- M (2009) Characterization of an anisotropic hydrogel
cal applications and open a window to long time tissue substrate for infusion testing. J Appl Polym Sci
announced and plebicited nanomedicine. Symp 114:1992–2002
Lenaerts V, Triqueneaux C, Quartern M, Rieg-Falson F,
Couvreur P (1987) Temperature-dependent rheologi-
cal behavior of Pluronic F-127 aqueous solutions. Int J
References Pharm 39:121–127
Motornov M, Roiter Y, Tokarev I, Minko S (2010)
Aust DT, Jones DP, Jovanovic AV, Kulkarni V, Kumar P, Stimuli-responsive nanoparticles, nanogels and cap-
Shi L (2012). Water-in-oil emulsion compositions sules for integrated multifunctional intelligent sys-
containing gellan gum for topical delivery of active tems. Prog Polym Sci 35:174–211
ingredients to the skin or mucosa, US 20120149783 Muzzalupo R, Tavano L, Cassano R, Trombino S,
A1. Ferrarelli T, Picci N (2001) A new approach for the
Burgess SE, Zhao Y, Sen A, Hui SW (2007) Resealing of evaluation of niosomes as effective transdermal drug
electroporation of porcine epidermis using phospho- delivery systems. Eur J Pharm Biopharm 79:28–35
lipids and poloxamers. Int J Pharm 336:269–275 Nhung DT, Freydiere AM, Constant H, Falson F, Pirot F
Cafaggi S, Russo E, Caviglioli G, Parodi B, Stefani R, (2007) Sustained antibacterial effect of a hand rub gel
Sillo G, Leardi R, Bignardi G (2008) Poloxamer 407 incorporating chlorhexidine-loaded nanocapsules
as a solubilising agent for tolfenamic acid and as a (Nanochlorex). Int J Pharm 334:166–172
base for a gel formulation. Eur J Pharm Sci 35:19–29 Noiles K, Vender R (2010) Are excipients really inert
Chen S, Singh J (2008) Controlled release of growth hor- ingredients? A review of adverse reactions to excipi-
mone from thermosensitive triblock copolymer sys- ents in oral dermatologic medications in Canada. J
tems: in vitro and in vivo evaluation. Int J Pharm Cutan Med Surg 14:105–114
352:58–65 Pandit NK, Kisaka J (1996) Loss of gelation ability of
Choi WI, Lee JH, Kim JY, Kim JC, Kim YH, Tae G Pluronic® F127 in the presence of some salts. Int J
(2012) Efficient skin permeation of soluble proteins Pharm 145:129–136
via flexible and functional nano-carrier. J Control Pandit NK, Wang D (1998) Salt effects on the diffusion
Release 157:272–278 and release rate of propranolol from poloxamer 407
Desai KGH, Mallery SR, Schwendeman SP (2008) Effect gels. Int J Pharm 167:183–189
of formulation parameters on 2-methoxyestradiol Pillai O, Panchagnula R (2003) Transdermal delivery of
release from injectable cylindrical poly(DL-lactide- insulin from poloxamer gel: ex vivo and in vivo skin
co-glycolide) implants. Eur J Pharm Biopharm permeation studies in rat using iontophoresis and
70:187–198 chemical enhancers. J Control Release 89:127–140
Fernandez-Esparrach G, Shaikh SN, Cohen A, Ryan MB, Pirot F, Falson F (2009). Novel method for producing
Thompson CC (2009) Efficacy of a reverse-phase nanocapsules in the absence of an organic solvent, and
polymer as a submucosal injection solution for EMR: nanocapsules produced thereby, WO/2009/138606.
a comparative study (with video). Gastrointest Endosc Porter CJH, Edwards GA, Charman SA (2001) Lymphatic
69:1135–1139 transport of proteins after s.c. injection: implications
328 D. Salmon et al.

of animal model selection. Adv Drug Deliv Rev Ye F, Larsen SW, Yaghmur A, Jensen H, Larsen C,
50:157–171 Ostergaard J (2012a) Drug release into hydrogel-based
Rijcken CJF, Soga O, Hennink WE, van Nostrum CF (2007) subcutaneous surrogates studied by UV imaging.
Triggered destabilisation of polymeric micelles and J Pharm Biomed Anal 71:27–34
vesicles by changing polymers polarity: an attractive Ye F, Larsen SW, Yaghmur A, Jensen H, Larsen C,
tool for drug delivery. J Control Release 120: Ostergaard J (2012b) Real-time UV imaging of piroxi-
131–148 cam diffusion and distribution from oil solutions into
Ruel-Gariépy E, Leroux JC (2004) In situ-forming gels mimicking the subcutaneous matrix. Eur J Pharm
hydrogels-review of temperature-sensitive systems. Sci 46:72–78
Eur J Pharm Biopharm 58:409–426 Yoshida M, Asano M, Kumakura M, Kataki R, Mashimo
Schmaljohann D (2006) Thermo- and pH-responsive T, Yuasa H, Yamanaka H (1991) Thermo-responsive
polymers in drug delivery. Adv Drug Deliv Rev hydrogels based on acryloyl-L-proline methyl ester
58:1655–1670 and their use as long-acting testosterone delivery sys-
Ur-Rehman T, Tavelin S, Grabner G (2010) Effect of tems. Drug Des Deliv 7:159–174
DMSO on micellization, gelation and drug release Yu L, Dean K, Li L (2006) Polymer blends and composites
profile of Poloxamer 407. Int J Pharm 394:92–98 from renewable resources. Prog Polym Sci 31:576–602
Liposomal Gels in Enhancing Skin
Delivery of Drugs 23
Lorena Tavano

Contents Abbreviations
23.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . 329
23.1.1 Liposomes . . . . . . . . . . . . . . . . . . . . . . . . . 330 Chol Cholesterol
23.1.2 Gel Systems . . . . . . . . . . . . . . . . . . . . . . . 331 DSPC-Chol Distearoyl-glycero-PC and
23.2 Liposomal Gel Systems . . . . . . . . . . . . . . 331 cholesterol
23.2.1 Introduction. . . . . . . . . . . . . . . . . . . . . . . . 331 EPC Egg phosphatidylcholine
23.2.2 Overview on Liposomal Gel Systems EPC-Na Egg phosphatidylglycerol sodium
Used for Dermal Drug Delivery . . . . . . . . 333
HEC Hydroxyethyl cellulose
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 339 HPC Hydrogenated PC
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 340 HPMC Hydroxypropyl methylcellulose
PC Phosphatidylcholine

23.1 Introduction

Transdermal drug delivery (TDD) is an admin-


istration route used for potent, low-molecular-
weight therapeutic agents which cannot
withstand the hostile environment of the gastro-
intestinal tract and/or are subject to considerable
first-pass metabolism by the liver (Prausnitz and
Langer 2008). TDD provides controlled and con-
stant administration of the drug, allows continu-
ous input of therapeutics with short biological
half-lives and eliminates pulsed entry into sys-
temic circulation, which often causes undesir-
able side effects. Skin has been considered as a
promising route for the administration of drugs
L. Tavano, PhD
Department of Pharmacy, Health and Nutrition
because of its accessibility and large surface
Sciences, University of Calabria, area. It is a very heterogeneous membrane that
Via P. Bucci, Arcavacata di Rende, provides a very effective barrier towards the pen-
Cosenza 87036, Italy etration of drugs both to and through it, thanks to
e-mail: uclorena@tiscali.it

N. Dragicevic, H.I. Maibach (eds.), Percutaneous Penetration Enhancers 329


Chemical Methods in Penetration Enhancement: Drug Manipulation Strategies and Vehicle Effects,
DOI 10.1007/978-3-662-45013-0_23, © Springer-Verlag Berlin Heidelberg 2015
330 L. Tavano

Particularly, liposomes have been shown to be


Hydrophilic drug a promising skin drug delivery system: their use
may produce severalfold higher drug concentra-
tions in the epidermis and dermis and lower sys-
temic concentrations when compared to
conventional dosage forms (El Maghraby 2008).
Hydrophobic drug Clearly, their topical use depends on their char-
liposome acteristics as size, surface, charge and chemical
composition. Mezei and Gulasekharam in 1980
Fig. 23.1 Schematic representation of a liposome were the first to employ liposomes as skin drug
delivery systems: they demonstrated that vesi-
cles of dipalmitoylphosphatidylcholine (DPPC)
its outermost layer, the stratum corneum (SC), a and cholesterol (CH) (1.1:0.5, molar ratio)
multilayered wall-like structure in which corneo- increased the concentration of triamcinolone
cytes are embed in a matrix of lipids (Bronaugh acetonide in the epidermis and dermis by four- to
and Maibach 1985). Since this barrier represents fivefold and reduced percutaneous absorption
a significant obstacle for transdermal permeation compared with a standard ointment (Mezei and
of most drugs, enhancement strategies are neces- Gulasekharam 1980). Several mechanisms have
sary to overcome it, thus improving drug release: been suggested for liposomes acting as skin drug
among these, penetration enhancers, supersatu- delivery systems. According to the free drug
rated solutions, physical methods and specific mechanism, the drug may permeate the skin
delivery systems such as vesicular systems (lipo- independently after exiting from the vesicles,
somes and niosomes) or vesicular hydrogels since the liposomes themselves enhance the
represent powerful approaches receiving consid- transdermal drug delivery by lowering the per-
erable attention (Barry 1983). meability barrier of the skin, changing the ultra-
structures of the intercellular lipids and the
enthalpy of the lipid-related transitions of the
23.1.1 Liposomes stratum corneum (Bernadete et al. 2011). In
some cases, the vesicles may adsorb to the stra-
Liposomes are microscopic vesicles that contain tum corneum surface with subsequent transfer of
amphipathic phospholipids arranged in one or drug directly from vesicles to the skin (El
more concentric bilayers enclosing an equal in Maghraby et al. 2008). Moreover, vesicles have
number of aqueous compartments (Fig. 23.1): In been reported to fuse and mix with the stratum
this form, as a spherical shell, they resemble bio- corneum lipid matrix, increasing drug partition-
logical membranes (Gregoriadis and Florence ing into the skin (El Maghraby et al. 2008). The
1993). The ability of phospholipids to form possibility that intact vesicles penetrate human
bilayers is due to their amphipathic nature: the skin acting as carriers and go deep enough to be
presence of a hydrophilic or polar region in the absorbed by the systemic circulation has been
head (attracts water) and a nonpolar region or also suggested (El Maghraby et al. 2008).
lipophilic tail (repels water). In this light, drug Different novel vesicular systems derived
molecules can be encapsulated in the aqueous from liposomes have been proposed as a valid
space (hydrophilic compounds) or intercalated alternative and among these, niosomes, nonionic
into the lipid bilayer (lipophilic compounds) surfactant-based vesicles, represent one of the
depending upon their physico-chemical charac- most suitable options: niosomes appear to be
teristics. Due to their entrapping ability, biode- similar in terms of their physical properties to
gradable and non-toxic nature, liposomes have liposomes, but they are preferred in topical deliv-
been reported to have several potential ery because of chemical stability and low cost of
applications (Allen and Curtis 2013). production (Schreier and Bouwstra 1994).
23 Liposomal Gels in Enhancing Skin Delivery of Drugs 331

GELS STRUCTURE
polymers, such as carbomers, impart an aestheti-
LIQUID PHASE
cally pleasing, clear, sparkling appearance to the
products and are easily washed off from the skin
with water; vehicles containing large amounts
of oleaginous substances provide an emollient
effect to dry irritated skin; bases made up of
non-volatile oleaginous substances can form an
occlusive barrier on the skin that prevents the
escape of moisture from the site of application,
causing hydration of the stratum corneum and
CROSSLINKS
POLYMERIC CHAINS increase of opening up of intra- and intercellu-
lar channels for easier passage of drug molecules
(Hoare and Kohane 2008).
Fig. 23.2 Schematic representation of a gel system

23.2 Liposomal Gel Systems


23.1.2 Gel Systems
23.2.1 Introduction
The term gel was introduced in the late 1800s to
name semisolid systems in which a liquid phase Taking into account all the previously mentioned
is constrained within a three-dimensional poly- reasons, the incorporation of vesicular systems
meric matrix of natural or synthetic gums with a into a gel dosage form has been designed as a
high degree of physical or chemical cross links, new strategy to improve drug percutaneous per-
as shown in Fig. 23.2 (Narin 1997). Gel forming meation: the resulting multicomponent systems,
polymers include natural polymer (proteins and named liposomal gel (Fig. 23.3), may possess
polysaccharides), semisynthetic polymers (cellu- the advantages of the individual formulations
lose derivatives), synthetic polymers (carbomers (vesicular suspensions and gel systems) and
and poloxamers) and surfactants (cetostearyl some other important benefits (Foldvari 1996).
alcohol and polyoxyethylene glycol alkyl ethers
(Brij™, Croda International PLC)). Gel also pos-
sess a degree of flexibility very similar to natural
tissue, and due to their high water content, they
resemble natural living tissue more than any
other type of synthetic biomaterial. Moreover,
they have significant roles in pharmaceutical and
cosmetical fields because of their biocompatibil-
ity, non-toxicity and good skin adhesion (Peppas
1986). Additionally, their insoluble cross-linked
structure allows medium for dissolution of
hydrophilic drugs, and since only the dissolved
drug presented to the skin is able to enter the
stratum corneum, gels could represent a strategy
to enhance the percutaneous drug absorption and
release across the skin in well-defined specific
manner (Wichterle and Lim 1960).
Clearly, the type of vehicle used to formulate a
Fig. 23.3 Photomicrographs of a vesicles-gel system as
topical dermatological product greatly influences seen by transmission electron microscopy (TEM)
its effectiveness. Gels prepared with organic (Adapted from reference Antunes et al. 2011)
332 L. Tavano

Since topically applied liposomal suspensions lone acetonide approximately five times higher in
may leak from the application site, they could the epidermis and three times higher in the dermis,
be mixed with gels in order to obtain semi- than application of the conventional drug gel. The
solid formulations. In addition, liposomal gels results of this study and those reported earlier by
were found to enhance the skin retention of the same researchers (Mezei and Gulasekharam
drugs and to provide higher and sustained skin 1980) suggested the inherent potential of lipo-
concentrations of therapeutics compared to con- somes (when applied in a gel) as a drug delivery
ventional gels and creams, without enhancing system for cutaneous application and the role of
their systemic absorption (Pavelic et al. 2005). liposomes in the formation of a large drug reser-
In addition, the stability of the liposomes (mem- voir in the skin, which is useful in local treatments.
brane integrity and mechanical stability) has been Since then, a lot of researchers explored the poten-
reported to increase when incorporated into a gel tial of liposomal gel systems in transdermal drug
matrix (Mourtas et al. 2007). Additionally, lipo- delivery (Gabrijelcic and Sentjurc 1995).
somal gels have been demonstrated to have bet- The most used polymers to obtain liposomal
ter rheological characteristics with respect to the gels are carbomers, cellulose derivatives and
liposomal dispersion (easiness of application and poloxamers. Carbomers are polymers of acrylic
removal from the skin), to ensure an appropriate acid cross-linked with polyalkenyl ethers or divi-
release of the active principle (compatibility with nyl glycol. They swell in water up to 1,000 times
most active substances) and increased skin tol- their original volume to form a gel when exposed
erance and compliance for patients. Clearly the to a pH environment above 4.0–6.0. Because the
type and concentration of the polymer forming pKa of these polymers is 6.0–0.5, the carboxylate
the gel matrix has been reported to influence the groups on the polymer backbone ionize, resulting
stability and release rate of the active substance, in repulsion between the negative charges, which
whereby an assessment of the physico-chemical adds to the swelling of the polymer (Florence
properties of the drugs, liposomes and polymeric and Pu 1994). Carbomer polymers are very well
gel must be made to avoid adverse effect and suited for aqueous formulations of the topical
chemical, physical and biological incompatibility dosage forms: many commercial products avail-
(Mourtas et al. 2008). able today have been formulated with these poly-
Moreover, liposomes in a gel can be more mers, as they provide numerous benefits to topical
stable to environmental stimuli compared with formulations. Carbomer polymer possesses low
bare liposomes in a dispersion, and when a drug toxicity and low irritancy potential and they are
is placed inside the liposomal core and the lipo- non-sensitizing even upon repeated usage. In
somes are included in a gel network, the drug will addition, due to their extremely high molecular
experience a combination of transport resistances weight, they cannot penetrate the skin or affect
due to the liposomal bilayer and the network the activity of the drug. Because of their excel-
itself: this results in a release of the drug over a lent thickening, suspending, emulsification and
longer period of time. Additionally, this can also suitable rheological properties, good tissue com-
avoid the problem of “burst release” seen with patibility and convenience in handing and ease of
some polymer gels where a large bolus of drug application, carbomer gels represent a good alter-
is released initially from the gel, which can cause native to oil-based formulations (Jian Hwa 2003).
toxicity (Lee et al. 2012). Cellulose polymers (hydroxyethyl cellulose,
The first study on the incorporation of lipo- hydroxypropyl cellulose, sodium carboxymethyl
somes in a gel dosage form was reported by cellulose) are examples of polymers that have
Mezei and Gulasekharam in 1982 (Mezei and been reported to possess adhesive properties
Gulasekharam 1982). They compared the perme- (Jones et al. 1997). Chemically, these linear poly-
ation of triamcinolone from plain and liposomal mers are cellulose derivatives possessing various
gel and they found that the application of the lipo- degrees of substitution and may be ionic or non-
somal gel resulted in a concentration of triamcino- ionic; following addition to an aqueous phase,
23 Liposomal Gels in Enhancing Skin Delivery of Drugs 333

these cellulose derivatives undergo swelling prior porated in gels of polyacrylate, i.e. carbomer
to dissolution. Pharmaceutically, water-soluble gels (Carbopol 974P NF or Carbopol 980 NF, BF
cellulose polymers have found widespread appli- Goodrich, Belgium). In vitro release of encapsu-
cations, e.g. in the formulation of solid dosage lated calcein from both liposomal gels was tested
forms, aqueous disperse systems as viscosity and compared with that of liposomes dispersed
enhancing agents and in products for topical in buffer (control). A slower release of calcein
application (Peppas et al. 2000). from liposomal gel was achieved: in fact, after
Poloxamers are polymers consisting of a 24 h, more than 80 % of the originally encapsu-
relatively long hydrophobic poly(propylene lated calcein was retained in liposomes embed-
oxide) (PPO) middle block and two hydrophilic ded in gel with respect to the control (60 %,
poly(ethylene oxide) (PEO) end blocks and are respectively). The authors ascribed these results
commercially available as Pluronics® (BASF, to the increased viscosity of the gel system which
Hanover, Germany). In the presence of a either reduced migration of drug molecules, acting as
solvent selective for the hydrophilic PEO blocks, drug reservoir system, while preserving the struc-
such as water, PEO–PPO–PEO block copolymers ture and integrity of liposomes.
self-organize into a variety mesophases with Similar results were obtained by Glavas-
lamellar, hexagonal or cubic structure (Batrakova Dodov and collaborators in 2002 (Glavas-Dodov
and Kabanov 2008). Pluronics® have attracted et al. 2002). The authors compared the in vitro
particular interest in the design of dermal and drug release properties of free and liposomally
transdermal delivery systems, with a view to pro- entrapped lidocaine hydrochloride hydrogels. As
moting, improving or retarding drug permeation expected, hydrogel formulations showed higher
through the skin. Moreover, they possess spe- release rate of lidocaine hydrochloride compared
cific pharmacological actions, in particular the to liposomal gel. Moreover, the release kinetic in
dynamic PEO chains prevent particle opsoniza- the case of liposomal gels can be described as dif-
tion and render them ‘unrecognizable’ to reticu- fusion controlled, while a steady-state release,
loendothelial system (RES) and macrophages achieved after the third hour, suggested that lipo-
(Tavano et al. 2010). Several studies reported the somes act as a reservoir system for continuous
advantageous interactions between Pluronics® delivery of the drug and for these reasons they
and liposomes: in particular, these polymers have could have a potential as dermal delivery systems
been used to sterically stabilize the vesicles and, with prolonged and sustained drug release.
hence, to prolong their half-life after parenteral Another delivery system based on liposomal
administration (Kostarelos et al. 1999). gels containing vitamin E acetate was designed in
2006 by Padamwar and collaborators to improve
topical drug delivery (Padamwar and Pokharkar
23.2.2 Overview on Liposomal Gel 2006). The prepared liposomal dispersion showed
Systems Used for Dermal Drug sevenfold increase in drug deposition in rat skin
Delivery compared to the control (plain drug dispersion),
and the liposomal gel formulation demonstrated
A summary of recent works in the area of novel sixfold and fourfold increase in drug deposition in
liposomal gel formulations for dermal drug deliv- rat skin compared to the control gel and marketed
ery is provided below. cream, respectively. Moreover, the liposomal gel
Pavelic et al. in 2001 developed a liposo- formulation was found to be more stable than the
mal gel system able to provide sustained and corresponding liposomal in terms of drug entrap-
controlled release of calcein, as model drug, ment efficiency and uniformity up to 3 months.
for local vaginal therapy (Pavelic et al. 2001). Mura et al. in 2007 designed and evaluated
Traditional liposomes were prepared from egg the potential of a liposomal gel formulation for
phosphatidylcholine (EPC) and egg phosphati- the topical delivery of benzocaine as a model
dylglycerol sodium (EPG-Na); they were incor- drug (Mura et al. 2007). Drug permeation from
334 L. Tavano

liposomal dispersions (based on mixtures of one hydrophilic (calcein) and one lipophilic
phosphatidylcholine, cholesterol, ethanol and (griseofulvin, GRF), when dissolved directly in
water) as such or formulated in a carbomer gel hydrogels (control gels) or dispersed in hydro-
was evaluated both through artificial lipophilic gels in the form of liposomes (liposomal gels).
membranes and excised abdominal rat skin, Drug-loaded liposomes based on PC or DSPC/
whereas in vivo anaesthetic effect was tested Chol were dispersed in carbomer, i.e. Carbopol®
in rabbits. Liposomes were prepared with drug 974 (Chemix S.A, Athens, Greece), HEC
encapsulated in the hydrophilic core or incor- (Natrosol 250 HX, Hercules Inc, Athens, Greece)
porated in the hydrophobic bilayer. The results or a mixture of these two hydrogels. Results dem-
of the benzocaine release study across artificial onstrated that depending on the intended use of
membranes showed that the presence of the poly- a liposomal gel formulation, the first parameter
meric network in the case of liposomal gels gave that should be considered is whether the drug
rise to a general reduction of the drug permeation is hydrophilic or lipophilic. In fact, the release
rate and allowed obtainment of a more regular of calcein from liposomal gels was slower com-
release profile as a function of time, with respect pared to that obtained from conventional gels and
to simple liposomal dispersions. Interestingly, strongly dependent on the liposome-membrane
drug-loaded gels showed a faster drug release rigidity. If the drug was encapsulated in DSPC/
in respect to the gel-containing liposomes with Chol liposomes and dispersed in gels, it was
the drug in the lipophilic phase, but slower than released significantly slower compared to the cor-
that obtained from liposomal gels with the drug responding formulation based on PC liposomes.
encapsulated in the aqueous phase. In permeation In the case of GRF, the release from liposomal
studies using rat skin, a higher reduction of drug gels was determined by drug loading. At high
permeation rate was noted, due to the more com- drug loading levels, GRF is released steadily from
plex permeation process through rat skin than liposomal gels irrespective of liposome type (PC
across artificial membranes. In percutaneous or DSPC/Chol). Moreover, in the case of the lipo-
permeation studies across rat skin, the difference philic drug, liposomes provided means for sub-
between the use of the liposomal dispersion as stantially increased drug loading in gels acting as
such or formulated in the carbomer gel was less reservoirs which released the drug in a sustained
evident, probably due to the major controlling manner. Finally, the authors demonstrated that in
effect exerted by the skin on the drug permeation the case of liposomal carbomer (Carbopol®) gels
rate. Moreover, an initial lag phase was present in that behave predominantly as elastic solids and
respect to the permeation studies thought artifi- have substantially different flow properties (com-
cial membranes, which was attributed to the lon- pared with HEC-based gels), increased release
ger time necessary to saturate the skin membrane rates for free calcein and GRF, indicating easier
and to reach a pseudo steady-state flux condi- diffusion of the compounds through this system,
tion between donor and receiver compartments. were obtained.
Finally, the lowest drug permeation was observed In the following year, Mourtas et al. investi-
from the conventional gel in comparison to all gated the effect of added liposomes on the rheo-
liposomal gel formulations (containing drug logical properties of a hydrogel containing
concentrations ranged from 0.05 to 0.5 % w/w), 0.40 % w/v of carbomer, i.e. Carbopol® 974 NF
confirming the hypothesized permeation enhanc- (Chemix S.A., Athens, Greece) and 1.5 % w/v of
ing effect of liposomal vesicles on drug delivery, HEC (Mourtas et al. 2008). PC or HPC lipo-
which cannot be efficiently estimated by using somes, plain or mixed with Chol, were used to
the artificial membrane. prepare liposomes. As reported by other authors,
Mourtas et al. in 2007 evaluated the effect the lipid composition of liposomal bilayers was
of liposomes, drugs and gel properties on drug found to strongly influence the rheological prop-
release kinetics (Mourtas et al. 2007). They erties of liposomal gels. Zero-rate shear viscosity
studied the release of two model compounds, and power law index values revealed that addi-
23 Liposomal Gels in Enhancing Skin Delivery of Drugs 335

tion of PC liposomes to the hydrogel had the hydroxy ethyl methacrylate (p-HEMA) hydrogel
smallest effect on its rheological properties, even affected the loading of cyclosporine A in the matrix
when the highest lipid concentration was used and controlled the drug release rate, depending on
(20 mg/ml). Oppositely, incorporation of HPC Brij™ concentration. At concentrations of Brij™
(or HPC/chol) liposomes into gels resulted in a above the critical micellar concentration (CMC),
significant increase of the elastic character of the the surfactant forms micellar than from the liposo-
gel, which increased with increasing lipid con- mal gel, but the lower value obtained by the mul-
centration. Since drug rate permeation was found ticomponent system was suggestive for prolonged
to be dependent on gel viscosity, authors demon- and sustained drug release, because of the presence
strated that liposomal gel based on HPC could be of several lipid bilayers that released the drug more
used to act as drug reservoir and to ensure a slow aggregates, into which the hydrophobic drugs can
drug release over a prolonged time period, while partition preferentially.
liposomal gel based on PC could be used to The objective of the work performed by Patel
obtain faster drug permeation. in 2009 was to formulate a 1 % carbomer gel-
Manosroi et al. in 2008 developed a new containing ketoconazole-loaded liposomes and
formulation in which elastic (containing etha- to study the in vitro drug release, skin retention
nol) and conventional niosomal vesicles con- and in vitro antifungal activity (Patel et al. 2009).
taining diclofenac diethylammonium (DCFD) The in vitro permeation of ketoconazole from
were incorporated into a gel base containing liposomal gel through wistar albino rat skin was
0.2 % w/w carbomer (Carbopol® 980, Noveon, compared with that of the corresponding conven-
India) (Manosroi et al. 2008). Authors reported tional cream and gel. From the results it can be
that the cumulative amounts of drug in stratum concluded that the cumulative drug permeation
corneum, viable epidermis and dermis and the was significantly higher from the conventional
receiving solution, obtained upon the applica- gel slowly. The higher skin retention of the drug
tion of niosomal gel, were higher compared to achieved in the case of the liposomal gel was
the commercial emulgel (containing 1.16 % found to depend on the creation of the reservoir
(w/w) of DCFD) and conventional gel without effect due to the presence of liposomes. In addi-
vesicles. They suggested a synergistic mecha- tion, the gel-containing liposomal ketoconazole
nism between ethanol, vesicles and skin lipids to showed the highest antifungal activity after 30 h
improve drug permeation. Moreover the devel- compared to the conventional ketoconazole gel
oped niosomal gels showed an 18.92 % of inhibi- and cream formulations, by using the cup plate
tion of rat ear oedema (after 1 h of application) (or cylinder plate) method.
higher than the commercial emulgel (2.70 %) The main aim of the study published by Nina
and conventional gel (5.41 %), indicating an Dragicevic-Curic et al. in 2009 was to develop
in vivo anti-inflammatory activity enhancement a temoporfin-loaded liposomal hydrogel which
of DCFD when entrapped in vesicles. This study would be able to deliver the photosensitizer in
demonstrated that the gel containing the novel an efficient dose into the stratum corneum and
vesicular system entrapped with DCFD may be deeper skin layers (Dragicevic-Curic et al. 2009).
a promising formulation to be used in the topical Results showed that the presence of the poly-
non-invasive treatment of inflammation. meric network in the liposomal gels systems
The effect of incorporation of surfactants into led to a slower drug release and lower perme-
hydrogels to increase drug loading concentration ation rate compared to liposomal dispersions
and attenuate the topical release rates was also and that the increase in polymer concentration
investigated by Kapoor et al. in 2008 (Kapoor from 0.5 to 1 % w/w, resulting in an increase of
and Chauhan 2008). They demonstrated that the the viscosity of the systems, led to lower skin
presence of 0.25, 0.6 and 1.5 % w/w polyoxyeth- penetration of temoporfin. Despite this, authors
ylene (20) oleyl ether (Brij™ 98, Sigma–Aldrich demonstrated that liposomal gel formulations
Chemicals, St. Louis, MO) as surfactant into a poly- were able to deliver a sufficiently high amount
336 L. Tavano

Fig. 23.4 Permeation profile 1.6

Amount of drug permeated (mg)


of different fluconazole 1.4 liposomal dispersion
containing systems (Adapted 1.2 liposomal gel
from reference Mitkari et al. plain carbopol gel
1
2010)
0.8 marketed gel
Aquous solution
0.6
0.4
0.2
0
0 100 200 300 400 500 600
Time (min)

of temoporfin into and also into the deeper skin its anti-inflammatory activity and compared
layers, because they ensure long contact time of it to the traditional gel formulation (Jithan and
the formulation with the skin. For these reasons, Swathi 2010). In vitro drug release and ex vivo
these new formulations have been suggested as permeation studies showed that the liposomal gel
advantageous against cutaneous malignant (basal provided a more sustained drug release and pro-
cell carcinoma) or non-malignant diseases (pso- longed anti-inflammatory effect compared to the
riasis, acne, etc.). control gel formulation.
Several groups developed liposomal gels to be In a recent study, Lingan et al. formulated
used for the dermal delivery of fluconazole. In topical gel-containing clobetasol propionate ves-
2007, Zhao et al. designed a new liposomal gel icles to prolong the duration of the drug effect
based on lecithin/cholesterol liposomes and car- and to prevent its side effects (Lingan et al. 2011).
bomer (Carbopol® 941, Beijing Haidian Huiyou The vesicles were prepared by varying the ratios
Fine Chemical Plant, China). They evaluated flu- between nonionic surfactants such as sorbitan
conazole skin permeation from the liposomal gel monopalmitate, sorbitan monostearate, sorbitan
across rat skin and compared it to the correspond- monooleate (Span® 40, 60, 80, S.D. Fine chemi-
ing conventional gel (Zhao et al. 2007). Results cals, Mumbai, India) and cholesterol. Afterwards,
indicated that the cumulative amount of drug per- the vesicles were incorporated into a 2 % w/w
meated through rat skin in the case of the liposo- carbomer gel. The conventional carbomer gel and
mal gel was lower than that found upon the marketed gel (Clobevate, Stiefel Laboratories
application of the conventional gel. Moreover, as Pakistan (PVT) Pakistan) showed cumulative
reported in analogue studies, the addition of the percentage of drug release (in respect to the ini-
drug into multicomponent systems significantly tial applied dose) of about 98 % in 300 min,
increased the deposited amounts of drug in the rat respectively, while the multicomponent formula-
skin and resulted beneficial for topical use. tion showed 51 % of drug release within 24 h. In
Differently, Mitkari et al. in 2010 designed addition, results demonstrated that the percent-
a liposomal gel based on hydrogenated PC age of reduction in paw oedema was gradually
(Phospholipon 90H, Phospholipids GmbH, increased in the case of niosomal gels up to 8 h,
Germany) and carbomer (Carbopol® 934 NF, whereas in marketed gel it gradually increased up
Noveon, India) and demonstrated that fluconazol to 4 h and later it declined at 6 h and 8 h, reveal-
skin permeation increased in the case of the lipo- ing that the multicomponent systems had a sus-
somal gel compared to the drug solution, plain tained as well as prolonged action.
carbomer gel and marketed gel (Flucos® gel, Recently, Megha et al. examined the ability
Cosme Health Care, Goa, India) as represented of a new developed liposomal carbomer
in Fig. 23.4 (Mitkari et al. 2010). (Carbopol® 934, Noveon, India) gel loading
In 2010, Jithan et al. formulated a diclof- selegiline, to deliver the active substance across
enac sodium-loaded liposomal gel to improve the skin, and demonstrated that this novel sys-
23 Liposomal Gels in Enhancing Skin Delivery of Drugs 337

tem provided a prolonged and sustained selegi- higher than that obtained from the niosomal
line release rate. This gel could represent an dispersion (18.32 ± 0.18 %).
attractive device for transdermal applications In 2011, Vyas et al. developed a new topical nio-
(Megha et al. 2012). somal gel loaded with benzoyl peroxide (com-
The objective of the study performed by monly used for the treatment of acne) to avoid drug
Mansoori et al. in 2012 was to develop a keto- side effects like skin redness, irritation, itching and
profen liposomal gel to enhance the drug’s anti- oedema (Vyas et al. 2011). Several vesicular for-
inflammatory activity and reduce its adverse mulations were prepared to achieve the best perfor-
effects (Mansoori et al. 2012). Carbomer mance in terms of size and entrapment efficiency
(Carbopol® 934) gel was used as a vehicle (1 % and the optimized niosomal formulation was incor-
w/w) and liposomes at different lipid concentra- porated into 2 % w/w HPMC K15 (Colorcon Asia,
tion were incorporated. Drug release profiles of Mumbai). Drug leakage studies revealed that vesi-
the different formulations were determined by cles incorporated in the multicomponent system
using Franz diffusion cell up to 24 h and com- were significantly more stable, because of the pre-
pared with that of a marketed gel (control) The vention of niosomes’ fusion. In vitro drug release
results showed that the marketed gel released profiles revealed higher amount of released drug
approximately 92 % of the drug within 24 h, from plain gel and marketed gel in respect to nio-
while liposomal gel formulations showed per- somal gel, thus demonstrating a prolongation of
centage of drug release ranging from 81 to drug release in the case of the multicomponent sys-
87 %, indicating that the multicomponent for- tem. Drug retention in the skin has been shown to
mulations provided a more retarded and con- be higher (41.53 %) with the niosomal gel in
trolled drug release compared to the commercial respect to the plain gel (21.45 %) and marketed gel
gel. Finally, the ketoprofen liposomal gel (24.88 %) after 24 h. Further, prolonged drug per-
showed a significantly enhanced retention of meation was observed from the niosomal gel in
drug molecules in the skin. respect to the plain gel and marketed gel, which
The aim of a recent study performed in 2012 may be due to slower diffusion of the drug across
by Patel et al. was to develop a niosomal gel as a the skin and to the creation of a drug reservoir.
new transdermal device to improve the systemic Recently, El-Nabarawi and collaborators
availability of lopinavir (Patel et al. 2012). Skin developed a liposomal gel formulation to be
permeation profiles of lopinavir was studied from used for transdermal paroxetine (PAX) delivery
a conventional gel, optimized niosomal and etho- (El-Nabarawi et al. 2013). Selected drug-loaded
somal dispersions, niosomal gel and ethosomal liposomes were incorporated into HPMC E-4M
gel, after their non-occlusive application onto rat gel (Tama, Tokyo, Japan) and then fabricated
abdominal skin ex vivo. Interesting results were into transdermal patches. In vitro release pro-
obtained: in terms of overall skin permeation of files were evaluated and the transdermal patches
lopinavir (including percentage deposited within were applied to rabbits for in vivo bioavailability
the skin and percentage permeated into the accep- study. Authors demonstrated that the diffusion of
tor medium), the ethosomal gel appeared to be paroxetine from different liposomal gels in vitro
more efficient than the niosomal gel. However, through the artificial membrane was apparently
the major fraction of lopinavir delivered via the dependent on the concentration of HPMC E-4M,
ethosomal gel remained deposited within the where the increase in polymer concentration was
skin, while the niosomal gel efficiently delivered associated with a decrease of the permeation rate
the drug deeper into the skin and released due to the increase of gel viscosity. The percent-
21.24 ± 0.23 % of the drug in 24 h, representing ages of paroxetine released from liposomal gels
an amount significantly greater than that released through artificial membrane were compared to
via the ethosomal gel (11.15 ± 0.15 %). Moreover that permeated from liposomal dispersions, and
the percentage of drug permeated and deposited results indicated that the incorporation of lipo-
in the skin when using the niosomal gel was somes into the gel resulted in a more delayed
338 L. Tavano

Fig. 23.5 Cumulative 45


percentage of paroxetine F5
40

% Cumulative PAX Permeated


(PAX) permeated from
different formulations: F5 35
liposomal dispersion, FH1 FH1
30
liposomal gel (2 % of FH2
polymer), FH2 liposomal gel 25
(4 % of polymer), FH3 20
liposomal gel (6 % of FH3
polymer) (Adapted from 15
reference El-Nabarawi et al. 10
2013)
5
0
0 5 10 15 20 25 30
Time (hr)

release due to the presence of the polymer net- Nie and co-workers in 2011 designed an in situ
work that act as an additional diffusion barrier to gel system based on liposome-containing pacli-
the drug release (Fig. 23.5). taxel, by using 18 % w/w poloxamer (Pluronic®
In 2003, liposome gels bearing 5-fluorouracil F127, BASF, Hanover, Germany) as thermorev-
(5-FU) as an antineoplastic agent, intended for ersible gel, with the aim to control drug release
transdermal application, have been prepared and and improve its antitumor efficiency (Nie et al.
in vitro drug release properties have been evalu- 2011). In vitro release experiment showed that
ated by Glavas-Dodov et al. (2003). Different liposomal gel exhibited the longest drug release
liposomal formulations, prepared by varying the period compared to liposomes, conventional gel
lipid phase composition, hydration conditions of and corresponding commercial gel formulation
the dry lipid film and drug concentration, were (Taxol, Bristol-Myers Squibb, China). This effect
incorporated into a chitosan gel base (1 %, w/w, was presumably due to the increased viscosity of
Katakura Chikkarin, Japan). Afterwards, 5-FU the liposomal gel, which has the effect of creat-
release profiles from conventional liposomes, ing a drug reservoir. 3-(4,5-Dimethylthiazol-
conventional hydrogel as well as drug aqueous 2-yl)-2,5-diphenyltetrazolium bromide (MTT)
solutions were estimated. Results demonstrated assay and drug uptake studies showed that the
that 5-FU was fully released from aqueous solu- treatment with the paclitaxel-loaded liposomal
tions within a period of 4 h, while the entrap- Pluronic® F127 gel yielded cytotoxicity, intercel-
ment of 5-FU into conventional chitosan gel lular fluorescence intensity and drug concentra-
resulted in a prolonged release rate, due to the tion in keratinocyte cancer cells lines (KB cells)
polymeric network of the hydrogel. Moreover, much higher than that of conventional liposomes.
the release of 5-FU from the liposomal gel was In 2011, the influence of nonionic niosomes
significantly slower (40 % released within 8 h) on the rheological behaviour of poloxamer
than from all other formulations, confirming (Pluronic® F127, BASF, Hanover, Germany) gel
that its encapsulation into liposomes resulted in was investigated by Antunes and co-workers,
a prolonged and sustained drug release. In addi- with the aim to develop new devices for the con-
tion, drug release profiles of 5-FU obeyed the trolled topical delivery of diclofenac sodium salt
Higuchi diffusion model, thus liposomes have (Antunes et al. 2011). Results showed that the
been demonstrated to act as reservoir systems presence of surfactant aggregates in the polymer
for continuous delivery of the antineoplastic network clearly enhances the network strength,
drug. because of polymer-vesicle hydrophobic associ-
23 Liposomal Gels in Enhancing Skin Delivery of Drugs 339

Fig. 23.6 Cumulative drug 1.0×10–7


amount versus time of perme-
ated diclofenac sodium salt at
37 °C from different formula-
tions (*niosomal gels with 15 8.0×10–8
and 18 % w/w Pluronic®
F127) (Adapted from Antunes
et al. (2011))

Drug Release (moles)


6.0×10–8

4.0×10–8

2.0×10–8

Vesicles-Drug Free-Drug
0.0 VT-F127* 15% VT-F127* 18%

0 2 4 6 8 10 12 14 16 18 20 22 24 26
Time (h)

ation and the increase of density of active links Conclusions


present in the polymer network. This resulted in Over the last two decades, several researchers
a thickening effect, being optimal to enhance the have attempted to combine the properties of
suitability of vesicular suspensions as transder- liposomes and polymer gels within the same
mal delivery devices, since traditional niosomal material, embedding the vesicular systems in
formulations are in liquid state. Interestingly, the the polymer matrix.
niosomal size increased upon polymer addition, These novel materials have demonstrated
probably due to the formation of a polymer layer some attractive features, including the fact
along the vesicle surface. As reported in that liposomes in a gel resulted more stable
(Fig. 23.6) higher percutaneous permeation of to environmental stimuli compared with
diclofenac sodium salt from niosomal formula- liposomes in solution. Moreover, some
tion in comparison to drug solution has been workers demonstrated that when a drug was
found. Interestingly, in the study of Antunes, the placed inside the liposomal core and the
drug cumulative amounts permeated from the liposomes have been included in a gel net-
niosomal gel decreased with increasing the poly- work, the drug experienced a combination of
mer concentration. In fact, vesicle gel system at transport resistances due to the liposomal
15 % w/w of Pluronic® F127 provided higher bilayer and the network itself, ensuring a
amount of permeated drug compared to the mix- release over a longer period of time and in a
ture of 18 % w/w Pluronic® F127 gel and drug sustained manner.
solution. The higher values of cumulative diclof- Finally, liposomal gel formulations have
enac sodium salt permeated were obtained by been reported to provide therapeutically better
the niosomal dispersion. Unexpected, the cumu- effects than the conventional formulations, as
lative permeated drug amount from niosomal gel topical dosage forms with prolonged and con-
at 18 % w/w Pluronic® F127 was found to be trolled drug release, which may lead to
lower than that achieved by using drug solution improved therapeutical effectiveness and bet-
due to the higher viscosity of the network gel. ter patient compliance.
340 L. Tavano

References Hoare TR, Kohane DS (2008) Hydrogels in drug delivery:


progress and challenges. Polymer 49:1993–2007
Jian Hwa G (2003) Carbopol® polymers for pharmaceuti-
Abraham Lingan M, Abdul Hasan Sathali M, Vijaya
cal drug delivery applications. Drug Dev Deliv
Kumar MR, Gokila A (2011) Formulation and evalua-
3:6–13
tion of topical drug delivery system containing clo-
Jithan AV, Swathi M (2010) Development of topical
betasol propionate niosomes. Sci Revs Chem Commun
diclofenac sodium liposomal gel for better antiinflam-
1:7–17
matory activity. Int J Pharm Sci Nanotechnol 3:
Allen TM, Cullis PR (2013) Liposomal drug delivery sys-
986–993
tems: from concept to clinical applications. Adv Drug
Jones DS, Woolfson AD, Brown AF (1997) Textural, vis-
Deliv Rev 65:36–48
coelastic and mucoadhesive properties of pharmaceu-
Antunes FE, Gentile L, Oliviero Rossi C, Tavano L,
tical gels composed of cellulose polymers. Int J Pharm
Ranieri GA (2011) Gels of Pluronic F127 and non-
151:223–233
ionic surfactants from rheological characterization to
Kapoor Y, Chauhan A (2008) Drug and surfactant trans-
controlled drug permeation. Colloids Surf B
port in cyclosporine A and brij 98 laden p-HEMA
Biointerfaces 87:42–48
hydrogels. J Colloid Interface Sci 322:624–633
Barry BW (1983) Dermatological formulations: percuta-
Kostarelos K, Tadros TF, Luckham PF (1999) Physical
neous absorption. Marcel Dekker, New York
conjugation of (tri-) block copolymers to liposomes
Batrakova EV, Kabanov AVJ (2008) Pluronic block copo-
toward the construction of sterically stabilized vesicle
lymers: evolution of drug delivery concept from inert
systems. Langmuir 15:369–376
nanocarriers to biological response modifiers. Control
Lee JH, Oh H, Baxa U, Raghavan SR, Blumenthal R
Release 130:98–106
(2012) Biopolymer-connected liposome networks as
Bernadete M, Pierre R, dos Santos I, Costa M (2011)
injectable biomaterials capable of sustained local drug
Liposomal systems as drug delivery vehicles for der-
delivery. Biomacromolecules 13:3388–3394
mal and transdermal applications. Arch Dermatol Res
Manosroi A, Jantrawuta P, Manosroi J (2008)
303:607–621
Anti-inflammatory activity of gel containing novel
Bronaugh RL, Maibach HI (1985) Percutaneous absorp-
elastic niosomes entrapped with diclofenac diethylam-
tion. Marcel Dekker, New York
monium. Int J Pharm 360:56–63
Dragicevic-Curica N, Winter S, Stupar M, Milic J,
Mansoori MA, Jawade S, Agrawal S, Khan MI (2012)
Krajisnik D, Gitter B, Fahr A (2009) Temoporfin-
Formulation development of ketoprofen liposomal
loaded liposomal gels: viscoelastic properties and
gel. J Pharm Cosmet 2:22–29
in vitro skin penetration. Int J Pharm 373:77–84
Mezei M, Gulasekharam V (1980) Liposomes – a selec-
El Maghraby GM, Barry BW, Williams AC (2008)
tive drug delivery system for the topical route of
Liposomes and skin: from drug delivery to model
administration. I. Lotion dosage form. Life Sci 26:
membranes. Eur J Pharm Sci 34:203–222
1473–1477
El-Nabarawi MA, Bendas ER, Tag R, El Rehem A, Abary
Mezei M, Gulasekharam V (1982) Liposomes-a selective
MYS (2013) Transdermal drug delivery of paroxetine
drug delivery system for the topical route of adminis-
through lipid-vesicular formulation to augment its
tration. J Pharm Pharmacol 34:473–474
bioavailability. Int J Pharm 443:307–317
Megha R, Harsoliya MS, Shraddha J, Azam K.
Florence AT, Jani PU (1994) Novel oral drug formula-
Development of Liposomal Gel for Transdermal
tions. Their potential in modulating adverse-effects.
Delivery of Selegiline. Int J Pharm Biol Arch.
Drug Saf 410:233–266
2012;3:1–3.
Foldvari M (1996) Effect of vehicle on topical liposomal
Mitkari BV, Korde SA, Mahadik KR, Kokare CR (2010)
drug delivery: petrolatum bases. J Microencapsul
Formulation and evaluation of topical liposomal gel
13:589–600
for fluconazole. Ind J Pharm Educ Res 44:324–333
Gabrijelcic V, Sentjurc M (1995) Influence of hydrogels
Mourtas S, Fotopoulou S, Duraj S, Sfika V, Tsakiroglou
on liposome stability and on the transport of liposome
C, Antimisiaris SG (2007) Effect of liposome, drug
entrapped substances into the skin. Int J Pharm
and gel properties on drug release kinetics. Colloids
118:207–212
Surf B Biointerfaces 55:212–221
Glavas-Dodov M, Goracinova K, Mladenovska K, Fredro-
Mourtas S, Haikou M, Theodoropoulou M, Tsakiroglou
Kumbaradzi E (2002) Release profile of lidocaine HCl
C, Antimisiaris SG (2008) The effect of added lipo-
from topical liposomal gel formulation. Int J Pharm
somes on the rheological properties of a hydrogel: a
242:381–384
systematic study. J Colloid Interface Sci
Glavas-Dodov M, Fredro-Kumbaradz E, Goracinova K,
317:611–619
Calis S, Simonoska M, Hincal AA (2003) 5-Fluorouracil
Mura P, Maestrelli F, Gonzalez-Rodrıguez ML,
in topical liposome gels for anticancer treatment – for-
Michelacci I, Ghelardini C, Rabasco AM (2007)
mulation and evaluation. Acta Pharm 53:241–250
Development, characterization and in vivo evaluation
Gregoriadis G, Florence AT (1993) Liposomes in drug
of benzocaine-loaded liposomes. Eur J Pharm
delivery. Clinical, diagnostic and ophthalmic poten-
Biopharm 67:86–95
tial. Drugs 45:15–28
23 Liposomal Gels in Enhancing Skin Delivery of Drugs 341

Narin GJ (1997) Encyclopedia of pharmaceutical technol- Peppas NA (1986) Hydrogels in medicine and pharmacy.
ogy. Marcel Decker, New York CRC Press, Boca Raton
Nie S, Hsiao WLW, Pan W, Yang Z (2011) Peppas N, Bures P, Leobandung W, Ichikawa H (2000)
Thermoreversible Pluronic® F127-based hydrogel Hydrogels in pharmaceutical formulations. Eur J
containing liposomes for the controlled delivery of Pharm Biopharm 50:27–46
paclitaxel: in vitro drug release, cell cytotoxicity, and Prausnitz MR, Langer R (2008) Transdermal drug
uptake studies. Int J Nanomedicine 6:151–166 delivery. Nat Biotechnol 26:1261–1268
Padamwar MN, Pokharkar VB (2006) Development of Schreier H, Bouwstra J (1994) Liposomes and niosomes
vitamin loaded topical liposomal formulation using as drug carriers: dermal and transdermal drug delivery.
factorial design approach: drug deposition and stabil- J Control Rel 30:1–15
ity. Int J Pharm 320:37–44 Tavano L, Muzzalupo R, Trombino S, Cassano R,
Patel RP, Patel HH, Baria AH (2009) Formulation and Pingitore A, Picci N (2010) Effect of formulations
evaluation of carbopol gel containing liposomes of variables on the in vitro percutaneous permeation of
ketoconazole. (Part-II). Int J Drug Deliv Technol sodium diclofenac from new vesicular systems
1:42–45 obtained from pluronic triblock copolymers. Colloids
Patel KK, Kumar P, Thakkar HP (2012) Formulation of Surf B Biointerfaces 79:227–234
niosomal gel for enhanced transdermal lopinavir Vyas J, Vyas P, Raval D, Paghdar P (2011) Development
delivery and its comparative evaluation with etho- of topical niosomal gel of benzoyl peroxide. Int Sch
somal gel. AAPS PharmSciTech 13:1502–1510 Res Netw Nanotechnol 2011:1–6
Pavelic Z, Skalko-Basnet N, Schubert R (2001) Liposomal Wichterle O, Lim D (1960) Hydrophilic gels for biologi-
gels for vaginal drug delivery. Int J Pharm cal use. Nature 185:117–118
219:139–149 Zhao SS, Du Q, Cao DY (2007) Preparation of liposomal
Pavelic Z, Skalko-Basnet N, Jalsenjak I (2005) fluconazole gel and in vitro transdermal delivery.
Characterisation and in vitro evaluation of bioadhesive J Chin Pharm Sci 16:116–118
liposome gels for local therapy of vaginitis. Int J
Pharm 301:140–148

You might also like