243

Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Embed Size (px)

Citation preview

Page 1: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554
Page 2: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Encapsulation andControlled Release

Technologies in Food Systems

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page i

Page 3: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Encapsulation andControlled Release

Technologies in Food Systems

Edited by Jamileh M. Lakkis

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page iii

Page 4: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Jamileh M. Lakkis, Ph.D., has 14 years experience in the food, dietary supplements, and consumerproducts industries. She served as Senior Project Manager at Pfizer/Cadbury-Schweppes, MorrisPlains, NJ, focusing on designing confectionery products as delivery systems for oral care benefits.As a Senior Encapsulation Specialist for General Mills, Inc., Minneapolis, MN, Dr. Lakkis designedseveral microencapsulation processes for stabilizing and masking the taste/aroma of a variety offunctional and nutraceutical actives for their applications in breakfast cereals, dairy, confections, andshelf-stable bakery products. Her professional experience also includes engagements as SeniorResearch Scientist at Land O’Lakes, Inc., Arden Hills, MN. Dr. Lakkis co-organized the first IFTsymposium on microencapsulation and controlled release applications in food systems. She is anactive member of the Controlled Release Society and serves on the society’s newsletter editorialboard representing the Consumer and Diversified Products Division.

©2007 Blackwell PublishingAll rights reserved

Blackwell Publishing Professional2121 State Avenue, Ames, Iowa 50014, USA

Orders: 1-800-862-6657Office: 1-515-292-0140Fax: 1-515-292-3348Web site: www.blackwellprofessional.com

Blackwell Publishing Ltd.9600 Garsington Road, Oxford OX4 2DQ, UKTel.: +44 (0)1865 776868

Blackwell Publishing Asia550 Swanston Street, Carlton, Victoria 3053, AustraliaTel.: +61 (0)3 8359 1011

Authorization to photocopy items for internal or personal use, or the internal or personal use of specificclients, is granted by Blackwell Publishing, provided that the base fee is paid directly to the CopyrightClearance Center, 222 Rosewood Drive, Danvers, MA 01923. For those organizations that have beengranted a photocopy license by CCC, a separate system of payments has been arranged. The fee codesfor users of the Transactional Reporting Service is ISBN-13: 978-0-8138-2855-8/2007.

First edition, 2007

Library of Congress Cataloging-in-Publication Data

Encapsulation and controlled release technologies in food systems / edited by Jamileh M. Lakkis,Ph. D.—1st ed.

p. cm.Includes bibliographical references and index.ISBN 978-0-8138-2855-8 (alk. paper)1. Controlled release technology. 2. Microencapsulation. 3. Food—Analysis. I. Lakkis,

Jamileh M.

TP156.C64E53 2007664'.024—dc22

2007006839

The last digit is the print number: 9 8 7 6 5 4 3 2 1

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page iv

Page 5: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

I dedicate this book to LEBANONWhich had not been my country, I’d have chosen it to be

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page v

Page 6: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Table of Contents

Dedication vContributors ixPreface xiJamileh M. Lakkis

1. Introduction 1Jamileh M. Lakkis

2. Improved Solubilization and Bioavailability of Nutraceuticals in Nanosized Self-Assembled Liquid Vehicles 13Nissim Garti, Eli Pinthus, Abraham Aserin, and Aviram Spernath

3. Emulsions as Delivery Systems in Foods 41Ingrid A.M. Appelqvist, Matt Golding, Rob Vreeker, and Nicolaas Jan Zuidam

4. Applications of Probiotic Encapsulation in Dairy Products 83Ming-Ju Chen and Kun-Nan Chen

5. Encapsulation and Controlled Release in Bakery Applications 113Jamileh M. Lakkis

6. Encapsulation Technologies for Preserving and Controlling the Release of Enzymes and Phytochemicals 135Xiaoyong Wang, Yan Jiang, and Qingrong Huang

7. Microencapsulation of Flavors by Complex Coacervation 149Curt Thies

8. Confectionery Products as Delivery Systems for Flavors,Health, and Oral-Care Actives 171Jamileh M. Lakkis

9. Innovative Applications of Microencapsulation in Food Packaging 201Murat Ozdemir and Tugba Cevik

10. Marketing Perspective of Encapsulation Technologies in Food Applications 213Kathy Brownlie

Index 235

vii

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page vii

Page 7: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Contributors

ix

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Ingrid A.M.AppelqvistUnilever Food and Health Research

InstituteUnilever R&D VlaardingenThe NetherlandsChapter 3

Abraham AserinCasali Institute of Applied ChemistryThe Institute of ChemistryThe Hebrew University of JerusalemJerusalem, IsraelNutralease Ltd., Mishor Adumim, IsraelChapter 2

Kathy BrownlieManager, Global ProgrammeFrost & SullivanOxford, England, UKChapter 10

Tugba CevikDepartment of Chemical

EngineeringSection of Food TechnologyGebze Institute of TechnologyGebze-Kocaeli, TurkeyChapter 9

Kun-Nan ChenDepartment of Mechanical

EngineeringTung Nan Institute of TechnologyTaipei, TaiwanChapter 4

Ming-Ju ChenDepartment of Animal ScienceNational Taiwan UniversityTaipei, TaiwanChapter 4

Nissim GartiCasali Institute of Applied ChemistryThe Institute of ChemistryThe Hebrew University of JerusalemJerusalem, IsraelNutralease Ltd., Mishor Adumim, IsraelChapter 2

Matt GoldingUnilever Food and Health Research

InstituteUnilever R&D VlaardingenThe NetherlandsChapter 3

Qingrong HuangDepartment of Food ScienceRutgers UniversityNew Brunswick, NJChapter 6

Nicolaas Jan ZuidamUnilever Food and Health Research

InstituteUnilever R&D VlaardingenThe NetherlandsChapter 3

Yan JiangDepartment of Food ScienceRutgers UniversityNew Brunswick, NJChapter 6

Jamileh LakkisSenior Project ManagerFormerly with Pfizer/Cadbury-SchweppesMorris Plains, NJChapter 1Chapter 5Chapter 8

Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page ix

Page 8: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Murat OzdemirDepartment of Chemical EngineeringSection of Food TechnologyGebze Institute of TechnologyGebze-Kocaeli, TurkeyChapter 9

Eli PinthusNutralease Ltd., Mishor Adumim, IsraelAdumim Food IngredientsMishor Adumim, IsraelChapter 2

Aviram SpernathCasali Institute of Applied ChemistryThe Institute of ChemistryThe Hebrew University of JerusalemJerusalem, IsraelChapter 2

Curt ThiesThies TechnologyHenderson, NevadaChapter 7

Rob VreekerUnilever Food and Health Research

InstituteUnilever R&D VlaardingenThe NetherlandsChapter 3

Xiaoyong WangDepartment of Food ScienceRutgers UniversityNew Brunswick, NJChapter 6

x Contributors

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page x

Page 9: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Preface

Encapsulation and controlled release technologies have enjoyed their fastest growth in thelast two decades. These advances, pioneered by pharmaceutical companies, were a resultof: (1) the rapid change in drug development strategies to target specific organs or evencells, (2) physicians’ growing concern about patient non-compliance, and (3) pharmaceuti-cal companies desire to extend their market monopoly on new drugs for a certain period oftime as provided by the US and international patent laws.

Despite this progress, encapsulation and controlled release technologies have only beenrecently adopted by the food industry. Food researchers and technologists have often beenconfronted with the dilemma of how to translate all these advances from the drug arena intopractical applications in food systems. By searching the literature, one can find volumes ofbooks and specialized publications on encapsulation and controlled release technologies.Unfortunately, most of these publications have dealt with theoretical aspects of these tech-nologies with little emphasis on real applications in consumer and food products.

This book attempts to illustrate various aspects of encapsulation and controlled releaseapplications in food systems using practical examples. These examples will give the readeran appreciation for the delicate art of designing encapsulated ingredients and the enormouschallenges in incorporating them into food formulations. Most of the practical examples inthis book were borrowed from the patent literature. This approach might be questionedbased on the fact that patents applications are never peer reviewed, but seems justifiableconsidering the frantic effort by both industry and academia to protect their discoveries andto gain limited-time monopoly on their innovations, thus limiting the availability of suchinformation in peer-reviewed articles.

This publication has several potential uses. It is a reference book for scientists in thefood, nutraceuticals and consumer products industries who are looking to introducemicroencapsulated ingredients into new or existing formulations. It is also a post-graduatetext designed to give students some comprehension of various aspects of encapsulation andcontrolled release in food systems.

This book is organized in such a way that each chapter treats one major application ofencapsulation and controlled release technologies in foods.

Chapter 1 introduces the readers to various encapsulation and controlled release tech-nologies, as well as release mechanisms, suitable for applications in foods, nutraceuticalsand consumer products.

Chapter 2 by Professor Nissim Garti and his collaborators discusses a novel approach toencapsulation and controlled release via reverse microemulsion technique referred to asnanosized self-assembled liquids (NSSL). Such systems are shown to provide exceptionalthermodynamic stability in a wide pH range. In addition to enhancing bioavailability offunctional active ingredients, NSSL systems, by virtue of their unique transparent appear-ance, are excellent candidates for beverage applications.

Chapter 3, by Dr. Klaas-Jan Zuidam and co-workers, presents an elaborate approach tounderstanding emulsions and their benefits as delivery systems in food applications. Thischapter discusses various mechanisms of emulsion stabilization and destabilization and

xi

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page xi

Page 10: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

how they can best be designed for targeted delivery of flavors and functional ingredients inthe human gastrointestinal system.

Chapter 4 on encapsulation and controlled release of probiotics by Drs. Chen and Chenreports on approaches for encapsulating probiotic bacteria in dairy products as well as inthe human gastrointestinal tract. This chapter also discusses novel optimization techniquesfor stabilizing these beneficial bacteria and enhancing their survival rates.

Chapter 5, written by the editor of this book, highlights current approaches to encapsula-tion and controlled release technologies for bakery products applications. Current encapsula-tion practices such as hot-melt particle coating and spray chilling are discussed. Examples ofthe performance of encapsulated leavening agents as well as sweeteners and flavors arepresented in shelf-stable bakery applications.

Chapter 6 on nanoencapsulation technology by Dr. Huang and his collaborators dealswith novel approaches to encapsulate enzymes and nutraceuticals. Specific examples arepresented on stabilization of phytochemicals and their enhanced bioavailability via incor-poration into nanoemulsions and bioconjugation systems.

Chapter 7 on flavor encapsulation via complex coacervation is written by Dr. Curt Thies.Discussion is focused on the basic principle of complex coacervation technique as a liquid–liquid polymer phase separation phenomenon. Guidance on polymer selection and subse-quent implications on the physicochemical properties of capsules as well as their releasebehavior is provided.

Chapter 8, written by the editor of this book, details techniques used for delivering ther-apeutic as well as functional actives and flavors via confectionery products. Technologiesand subsequent applications discussed in this chapter have wide applications in the food,nutraceuticals, as well as pharmaceutical arenas. Mechanisms and challenges specific totargeted release in upper gastrointestinal tract, especially the mouth and throat areas will bedescribed in great detail.

Chapter 9 discusses encapsulation and controlled release of actives in packaging appli-cations by Dr. Ozdemir and collaborator. In this contribution, the authors provide exampleson embedding fragrances, pigments as well as antimicrobial and insect repellent agentsinto food packaging films.

Chapter 10, authored by Ms. Kathy Brownlie, provides a marketing perspective of micro-encapsulation technologies and their potential impact on the food industry. Ms. Brownlieoffers an in-depth assessment of market drivers as well as constraints that are still hinderingwider implementation of these technologies in food manufacturing.

This book has definitely surpassed my vision and expectations thanks to the contributorsthat I am grateful to all of them for their expertise, commitment, and dedication. It is myhope that this book will prove itself a useful source on encapsulation and controlled releasein a wide range of food and consumer product applications.

Many thanks to the editorial staff at Blackwell Publishing Co., especially to MarkBarrett and Susan Engelken for their valuable help and advice throughout this project.

Last but not least, I would like to thank my parents who taught me the importance ofworking hard, having clear goals, and standing for what I believe is right. It is a lesson thatguides me in everything I do.

Jamileh M. Lakkis

xii Preface

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_FM_i-xiv r1.qxd 3/29/07 1:26 PM Page xii

Page 11: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

1 Introduction

Jamileh M. Lakkis

The European Directive (3AQ19a) defines controlled release as a “modification of the rateor place at which an active substance is released.” Such a modification can be made usingmaterials with specific barrier properties for manipulating the release of an active and toprovide unique sensory and/or functional benefits.

Addition of small amounts of nutrients to a food system, for example, may not affect itsproperties significantly; however, incorporating high levels of the nutrient either to meetcertain requirements or to treat an ailment will most often result in unstable and oftenunpalatable foods. Examples of such nutrients include fortification with calcium, vitamins,polyunsaturated fatty acids, and so on, and the associated grittiness, medicinal and oxi-dized taste, respectively. Different types of controlled-release systems have been formu-lated to overcome these challenges and to provide a wide range of release requirements.

The two principal modes of controlled release are delayed and sustained release(Figure 1.1).

• Delayed release is a mechanism whereby the release of an active substance is delayedfrom a finite “lag time” up to a point when/where its release is favored and is no longerhindered. Examples of this category include encapsulating probiotic bacteria for theirprotection from gastric acidity and further release in the lower intestine, flavor releaseupon microwave heating of ready-meals or the release of encapsulated sodium bicarbon-ate upon baking of a dough or cake batter.

• Sustained release is a mechanism designed to maintain constant concentration of anactive at its target site. Examples of this release pattern include encapsulating flavors andsweeteners for chewing gum applications so that their rate of release is reduced to main-tain a desired flavor effect throughout the time of chewing.

A wide range of cores (encapsulants), wall-forming materials (encapsulating agents), andtechnologies for controlling the interactions of ingredients in a given food system and formanufacturing microcapsules and microparticles of different size, shape, and morphologi-cal properties are commercially viable.

Wall-Forming Materials

Materials used in film coating or matrix formation include several categories:

1. Waxes and lipids: beeswax, candelilla and carnauba waxes, wax micro- and wax macro-emulsions, glycerol distearate, natural and modified fats.

2. Proteins: gelatins, whey proteins, zein, soy proteins, gluten, and so on. All these proteinsare available both in native and modified forms.

1

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 1

Encapsulation and Controlled Release: Technologies in Food SystemsEdited by Jamileh M. Lakkis

Copyright © 2007 by Blackwell Publishing

Page 12: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

3. Carbohydrates: starches, maltodextrins, chitosan, sucrose, glucose, ethylcellulose, cel-lulose acetate, alginates, carrageenans, chitosan, and so on.

4. Food grade polymers: polypropylene, polyvinylacetate, polystyrene, polybutadiene, andso on.

Core Materials

Core materials include flavors, antimicrobial agents, nutraceutical and therapeutic actives,vitamins, minerals, antioxidants, colors, acids, alkalis, buffers, sweeteners, nutrients,enzymes, cross-linking agents, yeasts, chemical leavening agents, and so on.

Release Triggers

Encapsulation and controlled-release systems can be designed to respond to one or a com-bination of triggers that can activate the release of the entrapped substance and to meet adesired release target or rate. Triggers can be one or a combination of the following:

• temperature: fat/wax matrices• moisture: hydrophilic matrices• pH: enteric coating, emulsion coalescence, and others.• Enzymes: enteric coating as well as a variety of lipid, starch and protein matrices.• Shear: chewing, physical fracture, and grinding• lower critical solution temperature (LCST) of hydrogels.

Payload is a term used to estimate the amount of active (core) entrapped in a given matrixor wall material (shell). Payload is expressed as:

Payload (%) = [(core)/(core + shell)] × 100

2 Chapter 1

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

time

Sustained (long-lasting) release

Delayed release

Figure 1.1. Generic representation of “sustained” and “delayed” release profiles.

Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 2

Page 13: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Entrapment of Actives in Food Matrices

Entrapment in an Amorphous Matrix

Encapsulation of active into an amorphous matrix, generally, involves melting a crystallinepolymer using heat and/or shear to transform the molecular structure into an amorphousphase. The encapsulant is then incorporated into the metastable amorphous phase followedby cooling to solidify the structure and form glass, thus restricting molecular movements.

Carbohydrates are excellent candidates for encapsulation applications due to the severalattributes possessed by them.

1. They form an integral part of many food systems.2. They are cost-effective.3. They occur in a wide range of polymer sizes.4. They have desirable physicochemical properties such as solubility, melting, phase

change and so on.

Sucrose, maltodextrins, native and modified starches, polysaccharides, and gums have beenused in encapsulating flavors, minerals, vitamins, probiotic bacteria as well as pharmaceu-tical actives. The unique helical structure of the amylose molecule, for example, makesstarch a very efficient vehicle for encapsulating molecules like lipids, flavors, and so on(Conde-Petit et al., 2006). Some carbohydrates such as inulin and trehalose can provideadditional benefits for encapsulation applications. Inulin, for example, is a prebiotic ingre-dient that can enhance survival of probiotic bacteria while trehalose serves as a supportnutrient for yeasts.

Two main technologies—spray drying and extrusion—have been used in large-scaleencapsulation applications into amorphous matrices, though using different mechanisms.In spray drying, for example, the active is trapped within porous membranes of hollowspheres, while in extrusion the goal is to entrap the active in a dense, impermeable glass.

Encapsulating actives via spray drying requires emulsifying the substrate into the encap-sulating agent. This is important for flavor applications, in particular, considering the factthat most flavors are made up of components of various chemistries (polarity, hydrophobicto hydrophilic ratios), thus limiting their stability when dispersed or suspended in differentsolvents. Hydrophobicity is one of the most critical attributes that can play a significant rolein determining flavors’ payload as well as their release in food systems.

The basic principle of spray drying has been adequately covered by Masters (1979).Briefly, the process comprises atomizing a micronized (1–10 micron droplet size) emulsionor suspension of an active and an encapsulating substance and further spraying the sameinto a chamber. Drying takes place at relatively high temperatures (210°C inlet and 90°Coutlet), though the emulsion’s exposure to these temperatures lasts only for few seconds.The process results in free flowing, low bulk density powders of 10–100 micron size.Optimal payloads of 20% can be expected for flavors encapsulated in starch matrices.Maltodextrins and sugars with lower molecular weight, due to their low viscosities andinadequate emulsifying activities, result in lower flavor payloads.

Several factors can impact the efficiency of encapsulation via spray drying, mainly thoserelated to the emulsion (solid content, molecular weight, emulsion droplet size, and viscos-ity) and to the process (feed flow rate, inlet/outlet temperature, gas velocity, and so on).

Introduction 3

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 3

Page 14: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Release of flavors from spray-dried matrices takes place upon reconstitution of the driedemulsion in the release medium, water most often. Reasonable prediction of the releasebehavior should take into consideration the complex chemistry of flavors and the prevailingpartition and phase transport mechanisms between aqueous and non-aqueous phases(Larbouss et al., 1991; Shimada et al., 1991).

Encapsulation into an amorphous matrix via extrusion has gained wide popularity in thelast two decades with applications ranging from entrapping flavors for their controlledrelease to masking the grittiness of minerals and vitamins. Hot melt extrusion is a highlyintegrated process with many unique advantages for encapsulation applications, namely:

1. Extruders are multifunctional systems (many unit operations) that can be manipulatedto provide desired processing temperature and shear rate profiles by varying screwdesign, barrel heating, mixing speed, feed rate, moisture content, plasticizers, and so on.

2. Possibility of incorporating actives and other ingredients at different points of the extru-sion process. Heat-labile actives, for example, can be incorporated via temperature-controlled inlets toward the end of the barrel and their residence time in the extruder canbe minimized to avoid degradation of the active and to preserve its integrity.

3. Extruders are also formers—encapsulated products can be recovered in practically anydesired shape or size (pellets, rods, ropes, and so on).

4. Only very limited amount of water is needed to transform carbohydrates from theirnative crystalline structure to amorphous glassy matrices in an extruder, thus limitingthe need for expensive downstream drying.

5. High payload—up to 30% can be expected when encapsulating solid actives in extrudedpellets.

6. Economics—attributes such as high throughput, continuous mode, and limited needfor drying make extrusion a very attractive process for manufacturing encapsulatedingredients.

Figure 1.2 describes a typical melt extrusion encapsulation process. Carbohydrate (encapsu-lating matrix), a mixture of sucrose and maltodextrin, is dry fed and melted by a combina-tion of heat and shear in the extruder barrel so that the crystalline structure is transformedinto an amorphous phase. The encapsulant (flavor or other active) is added through an open-ing in a cooled barrel situated toward the die to avoid flashing off of low boiling components.The amorphous mixture exits the die in the form of a rope that can be cooled quickly by airor liquid nitrogen to form a solid glassy material. The latter can be ground to a desiredparticle size to form compact microparticles of high bulk density.

Using this technology, encapsulated products can be designed to achieve any desired tar-get glass transition temperature by incorporating plasticizers (reduce Tg) or high-molecularweight polymers (increase Tg). It should be cautioned that although glass transition andassociated microcapsule stability are clearly related to the material properties of the matrixand rates of crystallization, there is growing evidence that in the glass transition regionsmall molecules are more mobile than might be expected from the high viscosity of thematrix (Parker and Ring, 1995). Mechanism of degradation of molecules entrapped in aglassy matrix is not fully understood but is speculated to be due to side-chain flexibility(e.g. enzymes) and/or diffusion of small molecules such as water and oxygen through theglassy matrix. Other deteriorative mechanisms may include Maillard reaction between theactive and the carrier matrix.

4 Chapter 1

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 4

Page 15: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Microcapsules manufactured via spray drying and extrusion may show structural imper-fections, thus limiting their shelf life. While spray-dried microcapsules tend to have lowbulk density, extruded granules may show stickiness and clumping. In addition, the pres-ence of exposed active on the microparticle surface may have detrimental consequencessuch as drifts in the release profile and/or loss of active due to oxidation and other deterio-rative processes.

A limited number of applications have employed freeze drying or other evaporativetechniques to form carbohydrate glasses from solution. Here, the removal of water mole-cules takes place either by freezing the solution and subliming the ice as in freeze dryingor by evaporation. Freeze drying forms porous substrates due to transport of water vapor.Unlike starches, sugars lack fixed molecular structure; thus they collapse upon freezedrying.

Co-crystallization with sugars has been practiced in few unique situations but hasnot found any commercial success. Crystalline sucrose is a poor flavor carrier but co-crystallization with flavors forms aggregates of very small crystals that incorporate the fla-vors either by inclusion within the crystals or by entrapment between them.

Release of actives from amorphous carbohydrate matrices takes place by subjecting thematrix to moisture or high temperatures, that is, by bringing the matrix to a state above itsglass transition temperature. Microcapsules entrapped in amorphous structures are pre-ferred for their ease of manufacturing, scalability and economics compared to other encap-sulation technologies. Their usage has been adapted to a variety of food systems such assurface sprinkle on breakfast cereals, hot instant drinks, soups, tea bags, chewing gum,pressed tablets, and so on.

Complexation of Actives into Cyclodextrins

Entrapment of actives into cyclodextrins is a unique approach to microencapsulation that isbased on molecular selectivity. Cyclodextrins are cyclic oligosaccharides formed of vari-ous numbers of α-(1,4) linked pyranose subunits. The 6-, 7-, and 8-numbered cyclic struc-tures are referred to as α-, β-, and γ-cyclodextrins, respectively; these molecules vary intheir solubility, cavity size, and complexation properties (Table 1.1).

Introduction 5

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Sugar blends(Dry feed)

Active (powder,dispersion, emulsion)

Mixing & heating

EXTRUDER

Amorphousrope

Groundmicroparticles

Figure 1.2. Encapsulation into amorphous carbohydrate matrices using hot melt extrusion.

Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 5

Page 16: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Type and degree of complexation in cyclodextrins are determined by two main factors:(1) steric fit of the guest (encapsulant) to the host (cyclodextrin) and (2) their thermody-namic interactions, mainly hydrophobic type.

Generally, one guest molecule is included in one cyclodextrin molecule, although forsome molecules with low molecular weight, more than one guest molecule may fit into thecavity (Figure 1.3). For molecules with large hydrodynamic radii, more than one cyclodex-trin molecule may bind to the guest. In principle, only a portion of the molecule must fitinto the cavity to form a complex. As a result, one-to-one molar ratios are not alwaysachieved, especially with high- or low-molecular-weight guests.

Guest molecules in cyclodextrins are not permanently entrapped but occur in a dynamicequilibrium. However, once a complex is formed and dried, it is very stable and oftenresults in very long shelf life (up to years at ambient temperatures under dry conditions).Release of the complexed guest takes place by immersing the guest-host complex in aque-ous media to dissolve the complex and further promoting the release of the guest when dis-placed by water molecules.

A wide variety of molecules can be entrapped in cyclodextrins such as fats, flavors, col-ors, and so on (Martin Del Valle, 2004; Parrish, 1988). Complexation of cyclodextrins with

6 Chapter 1

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Attribute �-Cyclodextrin �-Cyclodextrin �-Cyclodextrin

Number of glucopyranose units 6 7 8Molecular weight (g/mol) 972 1135 1297Solubility in water at 25°C (% w/v) 14.5 1.85 23.2Cavity diameter (Å) 4.7–5.3 6.0–6.5 7.5–8.3Cavity volume (Å)3 174 262 427

Table 1.1. Selected physicochemical properties of cyclodextrins (adapted from Martin Del Valle2004)

Figure 1.3. Schematic representation of a molecule entrapped in cyclodextrins.

Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 6

Page 17: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

sweetening agents such as aspartame can also stabilize the molecule and improve its tasteas well as eliminate the bitter aftertaste of other sweeteners such as stevioside and gly-cyrrhizin. Cyclodextrins can entrap undesirable substances such as cholesterol from prod-ucts such as milk, butter, and eggs (Szetjli, 1998; Hedges, 1998).

Encapsulation in Microporous Matrices—Physical Adsorption

Physical adsorption can only be feasible when an active is adsorbed onto a large surfacearea, microporous substrate, commonly referred to as molecular sieve. Examples of thiscategory include activated carbon (500–1400 m2/g) and amorphous silica (100–1000 m2/g)(Cheremisinoff and Morresi, 1978). Despite their efficiency in entrapping volatiles, silicaand activated carbon usage in foods has been discouraged due to regulatory constraints andis currently limited to packaging applications. The effectiveness of these materials isdemonstrated by extensive reduction in equilibrium vapor pressure which accompaniesphysical adsorption of volatile flavors.

Micronized sugars have been used but with limited success in adsorption applications.Dipping capillary-sized droplets of sucrose or lactose solution into liquid nitrogen followedby freeze drying can produce amorphous spheres that have the ability to adsorb aromas.Sorption of vapor causes these materials to revert to the more stable crystalline state withaccompanying loss of porosity.

Encapsulation in Fat- or Wax-Based Matrices

Entrapment of functional actives in fat-based matrices can be achieved using two main tech-nologies, hot-melt fluid bed coating and spray congealing. Actives can best be entrapped viamixing them with a fat/wax carrier followed by spray congealing. These technologies havebeen adequately discussed in Chapter 5 which deals with the encapsulation of bakeryleavening agents.

Encapsulation in Emulsions and Micellar Systems

Encapsulation via micelles is a convenient approach to enhance the solubility of insolubleor slightly soluble actives. This technique involves the simple entrapment of a hydrophobicactive in a hydrophilic shell material, thus rendering the particle or droplet soluble in aque-ous media. This is no trivial matter when considering the problems with bioavailability ofhydrophobic drugs and nutritional actives (fat-soluble vitamins, fish oil, and a host ofwater-insoluble drug actives).

A second important function of micelles is their small size which allows them to evadethe body’s screening mechanism, the reticuloendothelial system (RES). Recognition byRES is the main reason for removal of many drug delivery vehicles from the blood beforereaching their target site (Sagalowicz et al., 2006).

Micelles serve as drug “reservoirs” or “microcontainers” that ultimately release drugsvia diffusional processes. An in-depth discussion on encapsulation into emulsion systemscan be found in Chapters 2 and 3 of this book by Professor Garti and Dr. Zuidam and theirrespective coworkers.

Introduction 7

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 7

Page 18: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Encapsulation in Cross-Linked or Coacervated Polymers

Coacervation, as defined by Speiser (1976), is a process of transferring macromoleculeswith film properties from a solvated state via an intermediate phase, the coacervationphase, into a phase in which a film is formed around each particle and then to a final phasein which this film is solidified or hardened. Two types of coacervation processes are com-monly used in encapsulation applications, namely simple and complex:

1. Simple coacervation is based on “salting out” of one polymer by addition of agents(salts, alcohols) that have higher affinity to water than the polymer. It is essentially adehydration process whereby separation of the liquid phase results in the solid particlesor oil droplets becoming coated and eventually hardened into microcapsules.

2. Complex coacervation, on the other hand, is a process whereby a polyelectrolyte com-plex is formed. It requires the mixing of two colloids at a pH at which one is negativelycharged and the other positively charged, leading to phase separation and formation ofenclosed solid particles or liquid droplets (Rabiskova and Valaskova, 1998).

Several parameters can impact the formation and integrity of coacervates such as the poly-mers’ molecular weight, their w/w ratios, temperature, and processing time. Core materialssuitable for coacervation are solids and liquids that are water-insoluble so that the activewould not dissolve in the aqueous phase. One of the approaches to achieving high oil pay-loads is by using hydrophobic surfactants (Rabiskova and Valeskova, 1998).

The release of actives from coacervated systems is primarily a function of the wall typeand its thickness (slower release with increased wall thickness). Chapter 7 of this bookpresents an in-depth discussion on coacervation for flavor encapsulation applications.

Encapsulation into Hydrogel Matrices

Hydrogels are hydrophilic, three-dimensional network gels that can absorb much morewater than their own weight. Hydrogels consist of (a) polymers, (b) molecular linkers orspacers, and (c) an aqueous solution. Basic high-molecular-weight polymers include poly-saccharides, proteins, chitin, chitosans, hydrophilic polymers, and so on (Shahidi et al.,2006). The affinity of hydrogels to aqueous media makes them ideal absorbing matrices forfood and agricultural actives.

The principle of encapsulation by hydrogels is simply to entrap an active substanceand to further release it via gel-phase changes in response to external stimuli. Grahm andMao (1996) categorized the types of materials that cannot be delivered via hydrogels as:(i) extremely water-soluble actives due to the risk of uncontrollable quick release and(ii) very high-molecular-weight substances due to the extremely slow release rate toachieve a desired benefit.

Release of actives from hydrogels takes place via diffusion. The latter can be impactedby various chemical and physical factors such as the prevailing chemical bonds (H-bonds,ionic bonds, electrostatic interactions, and hydrophobic interactions) between the activeand the matrix. Physical factors include molecular size and conformation. Controlling(extending) the release of an active in a hydrogel matrix can be achieved by decreasing thehydrophilicity and/or diffusivity of the hydrogel structure or by covalently linking theactive to the carrier hydrogel matrix.

8 Chapter 1

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 8

Page 19: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Ideal hydrogels display a sharp phase transition upon swelling in an aqueous solventin response to environmental stimuli such as temperature, pH, electric field, and so on.Release from hydrogels can be predicted from their LCT (lower critical solution tempera-tures) values. As temperature increases to the hydrogel’s LCT, the hydrogel shrinks due todehydration. Below LCT, hydrogels can take up water thus increasing their swelling(Ichikawa et al., 1996).

Overview of Release Mechanisms

Despite the far-reaching applications of encapsulation and controlled-release technologiesin many industries, predicting the release of encapsulated actives, especially in biologicalsystems (foods included), remains a challenge. In the human gastrointestinal tract (GIT),for example, the release of microcapsules is a function of the physiological conditions,presence of food as well as the physicochemical properties of the ingested dosage.

One of the essential requirements for predicting release mechanisms of microencapsu-lated dosages is by identifying parameters involved in mass transport and diffusion of theactives from a region of high concentration (dosage) to a region of low concentration in thesurrounding environment.

Encapsulation and controlled-release systems can be classified into two main types:reservoir and matrix systems and, in some cases, combinations of both.

Reservoir-Type Systems

Reservoir-type systems are simply described as delivery devices where an inert membranesurrounds an active agent which upon activation diffuses through the membrane at a finitecontrollable rate (Figure 1.4a). Reservoir-type systems are capable of achieving zero-orderrates provided that constant thermodynamic activity is maintained inside the coatingmaterial. Reservoir-type systems are subject to shifts to a “burst-like” mechanism due tominor flaws in the membrane integrity.

Matrix Systems

Matrix or monolithic delivery systems can best be represented by microparticles preparedby extrusion or fat-congealed capsules where the actives are dispersed in the encapsulating

Introduction 9

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

(a)Reservoir-type

device(b)

Matrix-typedevice

(c)Combination-type

device

Figure 1.4. Schematic representation of encapsulation systems: (a) reservoir-type, (b) matrix-type,and (c) combination-type.

Lakkis_Ch_01_001-012 r1.qxd 3/30/07 6:23 PM Page 9

Page 20: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

medium (carbohydrate, fat, or other matrices). Matrix systems can be swellable (hydrogel)or non-swellable. Compared to reservoir systems, matrix systems require less quality con-trol, hence lower manufacturing cost (Figure 1.4b).

Combination Release Mechanism

Examples of this category can best be illustrated by congealed microcapsules or extrudedmicroparticles with additional film. coating (enrobing). This technique is most useful formanufacturing extremely “delayed release” profiles (Figure 1.4c).

Burst Release Mechanism

Burst release is simply described by a high initial delivery of an entrapped active, beforethe release reaches a stable profile, thus reducing the system’s effective lifetime and com-plicating the release control. Although burst release may be preferred for flavor high-impact applications, in drugs this mechanism may lead to high toxicity levels andineffective administration of the active.

Burst release can most often take place in reservoir and hydrogel systems, though it canstill take place in matrix designs. Reasons for this range from cracks in the protective cap-sule shell to storage effect where the membrane becomes saturated with the active sub-stances or due to very high active loading. When placed in a release medium, the active canquickly diffuse out of the membrane surface causing a burst effect (Huang and Brazel,2001). Low-molecular-weight actives frequently undergo burst release, a result of highosmotic pressure and increased concentration gradient. Other reasons include: processingconditions, surface characteristics of host material, sample geometry, host/drug interac-tions, morphology, and porous structure of dry material.

Application of a coating material over a monolithic microparticle can help eliminateburst release, though might change the release profile. Other treatments include washingmicroparticles to extract surface droplets of actives.

10 Chapter 1

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

First-order

Zero-order

Brustrelease

Figure 1.5. Release rates (zero-order, first-order, and burst) of microencapsulated systems.

Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 10

Page 21: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Kinetically, two main release patterns are identified, zero-order and first-order (Figure1.5). Other rates can still occur:

Zero-order release equation –dA/dt = kFirst-order release equation –dA/dt = k[C]

where –dA/dt is the change in active concentration over time, k is the rate constant, and [C]is the active’s concentration.

In designing microcapsules with controlled-release systems, it is critical to identifydesirable release profile so that adequate materials and technology can be chosen.

ReferencesBaker, R.W. and Lonsdale, H.K. 1974. Controlled release: mechanisms and rates. In: Controlled Release of

Biologically Active Agents (A.C. Tanquary and R.E. Lacey, eds.), Plenum, New York, pp. 15–71.Cheremisinoff, P.N. and Morresi, A.C. 1978. Carbon adsorption applications. In: Carbon Adsorption Handbook

(P.N. Cheremisinoff and F. Ellerbusch, eds.), Ann Arbor Science Publishers, Inc., Ann Arbor, Michigan, p. 3.Conde-Petit, B., Escher, F. and Nuessli, J. 2006. Structural features of starch-flavor complexation in food model

systems. Trends in Food Science & Technology 17(5): 227–235.Grahm, N.B. and Mao, J. 1996. Controlled drug release using hydrogels based on poly(ethylene glycols): macro-

gels and microgels, pp. 52–64. In: Chemical aspects of Drug Delivery, Karsa, D. and Stephenson, R. (Eds).Royal Society of Chemistry.

Hedges, R.A. 1998. Industrial applications of cyclodextrins. Chem. Rev. 98: 2035–2044.Huang, X. and Brazel, C.S. (2001). On the importance and mechanisms of burst release I matrix-controlled drug

delivery systems. J. Controlled Release 73: 121–136.Ichikawa, H., Kaneko, S. and Fukumori, Y. 1996. Coating performance of aqueous composite lattices with

N-ispropylacrylamide shell and thermosensitive permeation properties of their microcapsule membrane. Chem.Pharm. Bull. 44(2): 383–391.

Larbousse, S., Roos, Y. and Karel, M. 1992. Collapse and crystallization in amorphous matrices with encapsulatedcompounds. Sci. Aliments 12: 757–769.

Martin Del Valle, E.M. 2004. Cyclodextrins and their uses: a review. Process Biochem. 39: 1033–1046.Masters, K. 1979. Spray Drying Handbook, 3rd ed., George Godwinn, London.Parrish, M.A. 1988. Cyclodextrins—A Review. England: Sterling Organics. Newcastle-upon-Tyne NE3 3TT.Parker, R. and Ring, S.G. 1995. Diffusion in maltose-water mixtures at temperatures close to the glass transition.

Carbohydr. Res. 273: 147–155.Rabiskova, M. and Valaskova, J. 1998. The influence of HLB on the encapsulation of oils by complex coacerva-

tion. J. Microencapsul. 15(6): 747–751.Sagalowicz, L., Leser, M.E., Watzke, H.J. and Michel, M. 2006. Monoglyceride self-assembly structures as deliv-

ery vehicles. Trends in Food Science & Technology 17(5): 204–214.Shahidi, F., Arachchi, J.K.V. and Jeon, Y.-J. 2006. Food applications of chitin and chitosans. Trends in Food Sci-

ence & Technology 10(2): 37–51.Shimada, Y., Roos, Y. and Karel, M. 1991. Oxidation of methyl linoleate encapsulated in amorphous lactose-based

food model. J. Agric. Food Chem. 39: 637–641.Speiser, P. 1976. Microencapsulation by coacervation, spray encapsulation and nanoencapsulation. In: Microen-

capsulation, Nixon, J.R. (Ed.), pp. 1–11.Szetjli, J. 1998. Introduction and general overview of cyclodextrin chemistry. Chem. Rev. 98: 1743–1753.

Introduction 11

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_01_001-012 r1.qxd 3/29/07 1:28 PM Page 11

Page 22: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

2 Improved Solubilization and Bioavailabilityof Nutraceuticals in Nanosized Self-Assembled Liquid Vehicles

Nissim Garti, Eli Pinthus, Abraham Aserin, andAviram Spernath

Introduction

Microemulsions have been known for decades to the scientific community and to experts inthe industry. Hundreds of studies have been carried out by experimentalists and many theo-ries have been worked out regarding the self-aggregation of surfactants in aqueous phase aswell as in oil phase, to form micellar or reverse micellar (respectively) structures. Themicellar phases can be swollen by another liquid phase to form a reservoir of insoluble liq-uid phase entrapped by a tightly packed surfactant layer known as water-in-oil (w/o) or oil-in-water (o/w) microemulsions.

Microemulsion, by the most common general definition, is a “structured fluid” (orsolution-like mixture) of two immiscible liquid phases in the presence of a surfactant(sometimes with cosurfactant and cosolvent), which spontaneously form a thermodynami-cally stable isotropic solution-like liquid.

In spite of the numerous studies and pronounced potential applications in foods, pharma-ceuticals, and cosmetics, only a few practical preparations, in which the solubilized moleculesare at very low solubilization levels, are presently available in the market place. It is alwaysan open question as to why these structures did not make their way to final products.

The self-assembled nanosized surfactants and oil can solubilize another liquid immisci-ble phase and/or guest molecules (solubilizates). Droplet sizes are in the range of a few upto a hundred nanometers. In theory, in order to form such nanostructures, it is essential toreduce the interfacial tension between the two phases to a value close to zero. In order to doso, surfactants with the proper hydrophilicity must be utilized. In addition, surfactants musthave the proper geometry to self-organize in curved structures with the proper criticalpacking parameters (CPP).

Microemulsions are best studied by constructing binary, ternary, or multicomponentphase diagrams, which represent the equilibrium situation of the component mixture or thethermodynamic organization of the components. A typical classical phase diagram isshown in Figure 2.1.

Understanding the phase behavior and microstructure of microemulsions is an importantfundamental aspect of the utilization of these structured fluids in industrial applications.Today, we have a more profound understanding of the phase behavior and microstructure ofmicroemulsions (Shinoda and Lindman, 1987; Billman and Kaler, 1991; Kahlweit et al.,1996; Regev et al., 1996; Solans et al., 1997; Ezrahi et al., 1999). However, industrial appli-cations of microemulsions are rarely simple ternary systems, but more often complicatedmulticomponent systems. It is not always clear whether, in the complex systems, droplet

13

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:35 PM Page 13

Encapsulation and Controlled Release: Technologies in Food SystemsEdited by Jamileh M. Lakkis

Copyright © 2007 by Blackwell Publishing

Page 23: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

sizes and shapes are similar and remain intact and the role of the different components instabilizing the interface. Systematic investigations should be carried out to understand themicrostructure and the effect of the different components on the system.

In recent years, few attempts have been made to formulate and characterize microemul-sions that can be used for food, cosmetic, and pharmaceutical purposes (Dungan, 1997;Gasco, 1997).

In this effort, oils acceptable in food industry have replaced normal alkanes. The major-ity of easily made preparations were of oil-continuous phase (w/o). The authors focused onstudying the ability of formulating a microemulsion with triglycerides (Alander and Warn-heim, 1989a, b; Malcolmson and Lawrence, 1995; von Corswant et al., 1997; von Corswantand Söderman, 1998; Warisnoicharoen et al., 2000) and perfumes (Hamdan et al., 1995;Tokuoka et al., 1995; Kanei et al., 1999) as the oil component. Some workers (Joubranet al., 1993; Trevino et al., 1998) have studied the phase behavior and microstructure ofwater-in-triglyceride (w/o) microemulsions based on polyoxyethylene sorbitan hexaoleate.They found that the monophasic area of these systems was strongly dependent on tempera-ture and aqueous phase content. In other studies, o/w microemulsions were used. Lawrenceand coworkers (Malcolmson and Lawrence, 1995; Warisnoicharoen et al., 2000) examinedthe solubilization of a range of triglycerides and ethyl esters in an o/w microemulsion system

14 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 2.1. Typical phase diagram made with water, emulsifiers, and oil phase. Four types ofisotropic regions have been identified. Note that the dilution lines traverse via a two-phaseregion and full dilution to the far corner of the water phase is not possible.

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 14

Page 24: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

with nonionic surfactants. They concluded that the solubilization capacity depends not onlyon the nature of the surfactants but also on the nature of the oil.

There are very few surfactants that can be used in food formulations. In this respect,polysorbates (Tweens, ethoxylated derivatives of sorbitan esters) and sugar esters are inter-esting families of surfactants. The substitution of the hydroxyl groups on the sorbitan ringwith bulky polyoxyethylene groups increases the hydrophilicity of the surfactant. Similarly,monoesterification of sucrose forms hydrophilic emulsifiers. The ability of Tweens to formmicroemulsions for food applications has been studied by several authors (Constantinidesand Scalart, 1997; Trotta et al., 1997; Park and Kim, 1999; Prichanont et al., 2000; Radomskaand Dobrucki, 2000). An increased solubility of lipophilic drugs in the microemulsionregion was observed and explained by the penetration of these drugs into the interfacialfilm (Trotta et al., 1997; Park and Kim, 1999; Radomska and Dobrucki, 2000).

Even though some food-grade emulsifiers have been mentioned as possible microemul-sion-forming amphiphiles, it was almost impossible to use these systems mainly becausethe concentrates of oil/surfactant mixtures could not be fully diluted with water or aqueousphases to form o/w microemulsions. Any such dilution line (composition) is always “cross-ing” the two-phase region, resulting in a fast destabilization process and formation of emul-sions or two phases. Such phase separation leads to rapid precipitation of the solubilizedmatter. Some examples of such discontinued dilution lines illustrate the dilution problem ofthe classical phase diagrams. In Figure 2.1, these dilution lines are marked as dashed lines.

In most studies, the emphasis was on attempts to add just one immiscible liquid such aswater (or oil) to the oil (or water)-continuous surfactant phase, that is, to solubilize the oilin the core (inner phase) of the micelles. Practically very few attempts were made to incor-porate additional guest molecules, such as vitamins, aromas, antioxidants, and bioactivemolecules, into the solubilized core. Very little has been done to solubilize nutraceuticalswithin nanosized liquid vehicles in order to provide some pronounced health benefits tohumans or to treat chronic diseases.

Many structural and compositional limitations, in the presently available food formula-tions, did not permit loading significant amounts of nutraceuticals. It is not an easy task toaccomplish, since there is a need for additional technology to be developed. It is essential tointroduce new ingredients, new surfactants, and new concepts in microemulsion prepara-tion. Some of the cardinal points to be solved include the following:

• Progressively and continuously diluting, by aqueous phase or water, without destroyingthe interface and forming two-phase regions, that is, forming the so-called U-type phase dia-grams that undergo progressive inversion from w/o to o/w microemulsions (Figure 2.2).

• Preparing microemulsions that will be based on the use of permitted food-grade emulsi-fiers, oils, cosurfactants, or cosolvents.

• Facilitating the entrapment (cosolubilization capacity) of large loads of insoluble guestmolecules within the core of the microemulsion or at its interface.

• Providing environmental protection of the active addenda (guest molecules) fromautooxidation or hydrolytic degradation during shelf storage.

• Improving the bioavailability of the entrapped addenda.• Controlling the release from the vehicle to the water-continuous phase or onto human

membranes.• Using microemulsions as microreactors to obtain regioselectivity, fast kinetics, and con-

trolled and triggered reactions of active molecules once applied on the skin.

Improved Solubilization and Bioavailability of Nutraceuticals 15

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 15

Page 25: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

A phase diagram with a very large isotropic one-phase region is typical of the novelmicroemulsions that are made from multicomponents. The isotropic regions represent w/o,bicontinuous mesophase, and o/w microemulsion structures. The phase diagrams areknown as U-type. In such compositions, within the isotropic regions of the phase diagram,the oil/surfactant condensed structured mixtures (denoted condensed reverse micelles, L2)can transform to an L1 phase (direct micelles) via a w/o microemulsion, bicontinuousmesophase, and o/w microemulsion regions progressively, without any phase separation.

To the best of our knowledge, no reports were available in the literature, prior to theestablishment of our formulations as part of the extended new U-type phase diagrams, tocomply with these prerequisites of dilutable large isotropic regions (Garti et al., 2001,2003, 2004a, b; Yaghmur et al., 2002a, b, c, 2003a, b, 2004, 2005; Spernath et al., 2002,2003; de Campo et al., 2004). Most of the early studies were conducted on systems withconstant water content (>70%), low oil content (ca. 5–10%), and large surfactant excess(high surfactant/oil ratios). We enlarged the scope of the understanding and use of suchmicroemulsions to food and cosmetic preparations. Our studies examined various aspectsof solubilization of nutraceuticals, release patterns, and other thermal and environmentalconditions. In some of our studies the role of the surfactant was examined. The maximumsolubilization load was determined, and efforts were made to estimate the total amounts ofactive matter that can be entrapped along any dilution line. We were the first to establish thecorrelation between maximum solubilization capacity and water dilution (Garti et al., 2001,2003, 2004; Spernath et al., 2002, 2003; Yaghmur et al., 2002a, b, c , 2003a, b, 2004, 2005;de Campo et al., 2004).

This review summarizes our efforts to develop modified microemulsions as nanosizedself-assembled liquid (NSSL) vehicles for the solubilization of nutraceuticals and toimprove transmembrane transport for additional health benefits. Attempts were made toachieve solubilization of nonsoluble active ingredients such as aromas and antioxidantsinto clear beverages that are based on water-continuous phase.

16 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 2.2. Typical novel U-type phase diagram composed of selected combinations of cosmetic-grade emulsifiers with progressive full dilution.

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 16

Page 26: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

U-Type Microemulsions, Swollen Micelles, and Progressiveand Full Dilution

Initially we (Garti et al., 2001; Yaghmur et al., 2002a, b) dealt with solubilization of waterand oil in the presence of a new set of nonionic ingredients and emulsifiers to form U-typenonionic w/o and o/w food microemulsion systems. It was recognized that certain mole-cules destabilize the liquid crystalline phases and extend the isotropic region to higher sur-factant concentrations. The ability of these additives to provide large monophasic systems(denoted as the AT region in Figure 2.2), in which the total amounts of solubilized oil andwater should be as high as possible, was studied. The pseudoternary phase diagrams forR(+)-limonene-based systems with food-grade systems were compared with those basedon non-food grade emulsifiers such as Brij 96v, (C18:1(EO)10, Figure 2.2) (Garti et al., 2001;Yaghmur et al., 2002b). These systems offer great potential in practical formulations. Wefollowed the structural evolution and transformation of the microemulsion system fromaqueous phase-poor to aqueous phase-rich regions without encountering phase separation.

Figure 2.3a demonstrates the size distribution of various droplets along dilution line 73(D73; 70 wt% surfactant and 30 wt% oil phase) from 10 to about 90 wt% water. It can beseen that the droplets in the w/o region are smaller than those at higher water content uponinversion to o/w microemulsions. Figure 2.3b represents a typical structure as seen in thecryo-TEM (transmission electron microscopy) photomicrographs of an o/w microemulsiontaken from the rich-in-water region of the U-type diagram (obtained after inversion from anL2 phase into o/w droplets upon dilution with aqueous phase to 90 wt% water). The dropletsizes are ca. 8–10 nm and are mostly monodispersed. It should be noted that mostmicroemulsions, regardless of the type of oil, type of surfactant, and cosolvents, consistof droplets of ca. 5–20 nm in size and do not grow above these sizes at any water or oilcontents.

Various U-type phase diagrams with different types of hydrophilic surfactants, variouscosolvents, and cosurfactants were constructed to form small or large isotropic AT regions.The most desirable phase diagram yielded an isotropic region of AT > 75% from the totalarea of the phase diagram. The dilution lines connecting the oil/surfactant axis with thewater corner were termed Wm lines. Full dilution lines are those that can undergo full andprogressive dilution to the far water corner (Wm = 100%). Wm = 50% means that samplescan be diluted only up to 50 wt% water and if more water is added the microemulsion willundergo phase separation. An example of Wm = 100% dilution line is line 64 in Figure 2.2,in which a mixture of 60 wt% surfactant phase and 40 wt% oil phase is diluted progres-sively and completely with aqueous phase to the far corner (Wm = 100%) aqueous phase. Indilution line 55 (50 wt% surfactant phase and 50 wt% oil phase), the Wm is of ca. 60%aqueous phase, and further dilution will lead to phase separation.

Construction of U-type phase diagrams is essential for formulations of water-dilutablemicroemulsions.

Solubilization of Nonsoluble Nutraceuticals

The growing interest in microemulsions as vehicles for food and cosmetic formulationsarises mainly from the advantages of their physicochemical properties. Microemulsionscan cosolubilize large amounts of lipophilic and hydrophilic nutraceutical and cosmetoceu-tical additives, together with the inner reservoir.

Improved Solubilization and Bioavailability of Nutraceuticals 17

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 17

Page 27: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

The cosolubilization effect has attracted the attention of scientists and technologistsfor more than two decades. Oil-in-water microemulsions loaded with active moleculesopened new prospective opportunities for enhancing the solubility of hydrophobic vita-mins, antioxidants, and other skin nutrients. This is of particular interest, as it can provide awell-controlled way for incorporating active ingredients and may protect the solubilizedcomponents from undesired degradation reactions (Garti et al., 2001; Spernath et al., 2002;Yaghmur et al., 2002a, b, c). Figure 2.4 is a schematic illustration of the loading process ofvarious nutraceuticals onto the o/w microemulsion droplets after inversion.

Solubilization of active addenda may, therefore, be defined as spontaneous molecularentrapment of an immiscible substance (or only slightly miscible or soluble) in self-assembled surfactant mixtures to form a thermodynamically stable, isotropic, structuredsolution, consisting of nanosized liquid structures.

18 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

1

0

0 2 4 6 8r [nm]

10 12 14 16 18

Normalized to 1 at the maximum(a)

(b)

p(r)

[a.u

.]

10% AP

30% AP

40% AP

50% AP

60% AP

70% AP

80% AP

90% AP

Figure 2.3. (a) Droplet size distribution of various dilution points along dilution line 73 in phase diagram depicted in Figure 2.2. (b) Photomicrograph of typical o/w droplets derived froma concentrate of w/o after dilution to 90 wt% water content (AP refers to aqueous phase).(Adapted from Garti, with permission from the publisher.)

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 18

Page 28: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

The solubilized active molecules are compounds with nutritional value to human healththat, in most cases, are used in food applications. We will mention a few such examples thatwere studied in our labs, such as lycopene, phytosterols, lutein, tocopherols, CoQ10, andessential oils.

Lycopene

Food supplements have become very prominent compounds in recent years, due toincreased public awareness of healthy nutrition. The possibility of enhancing the solubilityof lipophilic vitamins, essential oils, aromas, flavors, and other nutrients in o/w microemul-sions is of great interest, as it can provide a well-controlled method for the incorporationof active ingredients and may protect the solubilized components from undesired degradationreactions (Dungan, 1997; Holmberg, 1998; Garti et al., 2000a, b). Lycopene (Figure 2.5) is animportant carotenoid that imparts a characteristic red color to tomatoes. This lipophiliccompound is insoluble in water and in most food-grade oils. For example, lycopene solubil-ity in one of the most efficient edible essential oils, R(+)-limonene, is 700 ppm. Recentstudies have indicated the important role of lycopene in reducing risk factors of chronicdiseases such as cancer, coronary heart disease, and premature aging (Dungan, 1997;Holmberg, 1998). This, in turn, has led to the idea of studying the effect of lycopene uptakeon human health.

Improved Solubilization and Bioavailability of Nutraceuticals 19

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Figure 2.4. A schematic illustration of the loading process of various nutraceuticals onto theo/w microemulsion droplets after inversion. (Adapted from Nutralease and Garti, 2003, withpermission from the publisher.)

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 19

Page 29: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Bioavailability of lycopene is affected by several factors:

• Food matrix containing the lycopene and, as a result, intracellular location of thelycopene, and the intactness of the cellular matrix. Tomatoes converted into tomato pastecan enhance the bioavailability of lycopene, as the processing includes mechanicalparticle size reduction and heat treatment.

• Amount and type of dietary fat present in the intestine. The presence of fat affects the for-mation of the micelles that incorporate the free lycopene.

• Interactions between carotenoids that may reduce absorption of either one of thecarotenoids (Bramley, 2000) owing to competitive absorption between the carotenoids.On the other hand, simultaneous ingestion of various carotenoids may induce antioxidantactivity in the intestinal tract, and thus result in increased absorption of the carotenoids(Rao and Agrawal, 1999; Bramley, 2000).

• Molecular configuration (cis/trans) of the lycopene molecules. The bioavailability of thecis isomer is higher than the bioavailability of the trans isomer. This may result from thegreater solubility of cis isomers in mixed micelles and lower tendency of cis isomers toaggregate (Cooke, 1998; Rao and Agrawal, 1999).

• Decrease in particle size (Van het Hof et al., 2000).

Care must be taken in formulating lycopene as an additive in food systems, since the largenumber of conjugated bonds in this carotenoid causes instability when exposed to light oroxygen. We explored the ability of U-type microemulsions to solubilize lycopene and havealso investigated the influence of solubilized lycopene on the microemulsion microstruc-ture. Phase diagrams have been constructed, lycopene has been solubilized, and severalstructural methods have been utilized including self-diffusion nuclear magnetic resonance(SD-NMR) spectroscopy. This advanced analytical technology was further developed todetermine the microemulsion microstructure at any dilution point.

The influence of microemulsion composition on the solubilization of lycopene in a five-component system consisting of R(+)-limonene, cosurfactant, water, cosolvent, and poly-oxyethylene (20) sorbitan mono-fatty esters (Tweens) is presented in Figures 2.6 and 2.7.

Solubilization capacity was defined (Spernath et al., 2002, 2003) as the quantity oflycopene solubilized in the microemulsion. Figure 2.7 shows the solubilization capacity oflycopene along water dilution line T64 (at this line the constant ratio of R(+)-limonene/ethanol/Tween 60 is 1/1/3, respectively). Four different regions can be identified along thisdilution line. At 0–20 wt% aqueous phase (region Ι), the solubilization capacity oflycopene decreases dramatically, from 500 to 190 ppm (reduction of 62%). This dramaticdecrease in the solubilization capacity can be associated with the increase in interactionsbetween the surfactant and water molecules. Water can also strongly bind to the hydroxyl

20 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 2.5. Molecular structure of lycopene.

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 20

Page 30: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

groups of the surfactant at the interface. When water is introduced to the core, the micellesswell, and more surfactant and co-surfactant participate at the interface, replacing thelycopene, thus decreasing its solubilization. In region Ι, the reverse micelles swell graduallyand become more hydrophobic, causing less free available volume for the solubilizedlipophilic lycopene and a reduction in its solubilization capacity. At 20–50 wt% aqueousphase (region II) the solubilization capacity remains almost unchanged (decreases only byan additional 7%). This fairly small decrease in the solubilization capacity could be associ-ated with the fact that the system transforms gradually into a bicontinuous phase and theinterfacial area remains almost unchanged when the aqueous phase concentration increases.Surprisingly, in region ΙΙΙ (50–67 wt% aqueous phase) the solubilization capacity increases

Improved Solubilization and Bioavailability of Nutraceuticals 21

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Figure 2.6. Pseudoternary phase diagram (25ºC) of water/PG/R(�)-limonene/ethanol/Tween 60system with a constant weight ratio of water/PG (1:1) and a constant weight ratio of R(�)-limonene/ethanol (1:1). Solubilization of lycopene was studied along dilution line T64. (Adaptedfrom Yaghmur and Garti, 2001, with permission from the publisher.)

Figure 2.7. Solubilization capacity of lycopene along dilution line T64 as per phase diagram inFigure 2.6. (Adapted from Garti, with permission from the publisher.)

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 21

Page 31: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

from 160 to 450 ppm (an increase of 180%). In region IV the solubilization capacitydecreases to 312 ppm (a decrease of 30%).

In order to explain the changes in solubilization capacity of lycopene, we characterizedthe microstructure of microemulsions along dilution line T64 using the SD-NMR tech-nique. Figure 2.8 shows the relative diffusion coefficients of water and R(+)-limonene inempty (containing no solubilizates) microemulsions (Figure 2.8a) and microemulsionssolubilizing lycopene (Figure 2.8b), as a function of the aqueous phase concentration(w/w). One can clearly see that the general diffusion coefficient behavior of microemulsioningredients (R(+)-limonene and water), with or without lycopene, is not very different. Thetotal amount of lycopene does not cause dramatic changes in the diffusion patterns of theingredients.

It can also be seen that, in the two extremes of aqueous phase concentrations (up to20 wt% and above 70–80 wt% aqueous phase), the diffusion coefficients are easily inter-preted, while the regions in between are somewhat more difficult to explain, since gradual

22 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

1.000

0.100

DW

/D0W

DO

/D0O

0.010

0.001

1.000

0.000

0.010

0.0010 20 40

Aqueous phase (wt%)60 80 100

(a)

1.000

0.100

DW

/D0W

DO

/D0O

0.010

0.001

1.000

0.000

0.010

0.0010 20 40

Aqueous phase (wt%)60 80 100

(b)

Figure 2.8. Relative diffusion coefficient of water (•) and R(�)-limonene (▲) in microemulsionswithout (a) and with (b) lycopene, as calculated from SD-NMR results at 25ºC. D0

w was measuredin a solution containing water/PG (1:1), and determined to be 55.5�10–11 m2 s–1. D0

o the purediffusion coefficient of R(�)-limonene was determined to be 38.3�10–11m2 s–1. (Adapted fromGarti, with permission from the publisher.)

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 22

Page 32: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

changes take place. Regions ΙΙ and ΙΙΙ are difficult to distinguish. However, the structuralchanges in the presence of lycopene (Figure 2.8b) are more pronounced than those in theabsence of lycopene (Figure 2.8a).

Microemulsions containing up to 20 wt% aqueous phase, and solubilizing lycopene,have a discrete w/o microstructure, since the relative diffusion coefficients of water andR(+)-limonene differ by more than one order of magnitude. Microemulsions solubilizinglycopene and containing 20–50 wt% aqueous phase have a bicontinuous microstructure, asthe diffusion coefficients of water and R(+)-limonene are of the same order of magnitude.Increasing the aqueous phase concentration to above 50 wt% induces the formation of dis-crete o/w microstructures, as the relative diffusion coefficients of water and R(+)-limonenediffer by more than one order of magnitude.

From the solubilization capacity and SD-NMR results, it is clear that lycopene solubi-lization is structure dependent.

The four different regions in the solubilization capacity curve are an indication of themicrostructure transition along the dilution line. The first region indicates the formation ofw/o (L2) microstructure. The second region indicates the transition from L2 microstructureto a bicontinuous microemulsion. In the third region, a transition from a bicontinuousmicroemulsion to an o/w (L1) microstructure occurs. In the fourth region a discrete L1microstructure was found.

While the general behavior of the diffusion coefficients is the same for microemulsionswith or without lycopene, the transition point from one microstructure to another is differ-ent. Lycopene influences the transition from L2 to bicontinuous microstructure and furtherto L1 microstructure. In empty microemulsions the formation of bicontinuous microstruc-ture occurs when the microemulsion contains 40–60 wt% aqueous phase, whereas in amicroemulsion containing lycopene, bicontinuous microstructure starts at low aqueousphase content (20 wt%) and continues up to an aqueous phase content of 50 wt%. It seemsthat as more water is solubilized in the swollen reverse micelles less free interfacial volumeis available for the lycopene. Lycopene appears to disturb both the flexibility of the micelleand the spontaneous curvature. As a result, the interface changes into a flatter curvature(bicontinuous) at an early stage of water concentration, more so in the presence of lycopenethan empty micelles.

The hydrophilic–lipophilic balance (HLB) of the surfactant influences the quantity ofsolubilized lycopene in the aqueous surfactant phase. Tween 60, being a hydrophilic surfac-tant with the lowest HLB value (HLB 14.9), solubilizes 10 wt% more lycopene than Tween80 (HLB 15.2). In Tween 40 (polyoxyethylene sorbitan monomyristate)-based microemul-sions, the solubilization capacity drops even further (30%). Replacing Tween 60 withTween 20, the most hydrophilic surfactant (HLB 16.7), reduces the solubilization capacityof lycopene by 88%.

We have also demonstrated that microemulsions stabilized by mixed surfactants enhancethe solubilization capacity of lycopene by 32–48%, in comparison to microemulsions stabi-lized by Tween 60 alone (Spernath et al., 2002; Garti et al., 2003, 2007), indicating a syner-gistic effect. Microemulsions stabilized by a mixture of three surfactants—Tween 60,sucrose ester, and ethoxylated monodiglyceride—have the highest solubilization capacity oflycopene—an increase of 48%, in comparison to microemulsion based on Tween 60 alone(Spernath et al., 2002; Garti et al., 2003, 2004a, b). Synergism in surfactant mixtures wasattributed to Coulombic, ion-dipole, or hydrogen-bonding interaction (Hou and Shah, 1987;Huibers and Shah, 1997). Therefore, nonionic surfactant mixtures are expected to have a

Improved Solubilization and Bioavailability of Nutraceuticals 23

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 23

Page 33: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

minimum intermolecular interaction and weak synergistic effects. Nevertheless, Huibersand Shah (1997) demonstrated a strong synergism in nonionic surfactant mixtures, similarto the findings in our study. This behavior remains to be explained. Solubilization capacity isdefined as the quantity (mg) of solubilizate entrapped in 100 g microemulsion, and solubi-lization efficiency is the quantity of solubilizate per 100 g of the oil phase or that normalizedto oil content solubilization. Solubilization efficacy is the ratio of the quantity of solubilizedcompound to the quantity of the total amounts of oil and surfactant phase. Microemulsionsexhibit very large solubilization capacities and solubilization efficiencies for lycopene.Lycopene was solubilized in a microemulsion up to 10 times its dissolution capacity inR(+)-limonene or in any other edible solvent. The solubilization capacity and efficiency oflycopene are strongly affected by microstructure transitions from w/o to bicontinuous andfrom bicontinuous to o/w. Solubilization capacity drops significantly with dilution, whilethe efficiency and efficacy increase as the water content increases, indicating that the inter-face plays a significant role in the solubilization of lycopene.

Phytosterols

Elevated serum cholesterol level is a well-known risk factor for coronary heart disease(Hicks and Moreau, 2001). Most strategies for lowering serum cholesterol require dietaryrestrictions and/or medications. The prospect of lowering cholesterol levels by consumingfoods fortified with natural phytonutrients is considered much more attractive.

Phytosterols (plant sterols) are steroid alcohols. Their chemical structure resembleshuman cholesterol, as can be seen in Figure 2.9. Both sterols are made up of a tetracycliccyclopenta[α]phenanthrene ring system and a long flexible side chain at the C17 carbonatom. The four rings have trans configurations, forming a flat α-system (IUPAC, 1989;Piironen et al., 2000). Moreover, sterols create planar surfaces, at both the top and the

24 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 2.9. Molecular structure of cholesterol and some abundant phytosterols (R = H–cholesterol; R = CH2CH3-β-sitosterol; R = CH2CH3 and additional double bond at C22-stigamsterol;R = CH3-campasterol; R = CH3 and additional double bond at C22-brassicasterol. (Adapted fromGarti, 2004, with permission from the publisher.)

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 24

Page 34: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

bottom of the molecules, since the R-conformation is preferred in the side chain linked toC20 carbon atom of the sterol molecule. This allows for multiple hydrophobic interactionsbetween the rigid sterol nucleus (the polycyclic component) and the membrane matrix(Nes, 1987; Bloch, 1988; Piironen et al., 2000). Only side chains of the various sterols aredifferent. These minor configuration differences result in major differences in their biologi-cal function.

Peterson et al. (1951) reported that addition of soy sterols to a cholesterol-enriched dietprevented an increase in the plasma cholesterol level. This effect significantly reduced theincidence of atherosclerotic plaque (Peterson et al., 1951). Since then, numerous clinicalinvestigations have indicated that administration of phytosterols to human subjects reducesthe total plasma cholesterol and LDL cholesterol levels (Pelletier et al., 1995; Jones et al.,1997). Because of their poor solubility and limited bioavailability, high doses were requiredto have a noticeable effect.

Up to 25 g/day of phytosterol esters were recommended in some reports and up to1.3 g/day of phytosterol esters are to be used as per the FDA recommendation for adecrease of up to 15% of the cholesterol in the blood stream.

The exact mechanism by which phytosterols inhibit the uptake of dietary and endoge-nous cholesterol is not completely understood. One theory suggests that cholesterol in thepresence of phytosterols precipitates in a nonabsorbable state. A second theory suggeststhat cholesterol is displaced by phytosterols in the bile salt micelles and phospholipid-containing mixed micelles, thus preventing its absorption (Hicks and Moreau, 2001).Enhanced solubilization of phytosterols in o/w microemulsions has been hypothesized topromote their bioavailability and maximize their absorption in human tissues owing to theirsmall droplet size (in the range of several nanometers). Activity of phytosterols in food for-mulations has not yet been fully studied. Our results (Rozner and Garti, 2006) and that ofother investigators (Ostlund, 2002) indicate that phytosterols do not cross human mem-branes, but they significantly retard (or prevent) the penetration of cholesterol and otherlipids. We explored the ability of the unique dilutable microemulsions to solubilize phytos-terols and studied the correlation between the solubilization capacity of the phytosterolsand the microemulsion microstructure transitions (Spernath, 2003; Garti et al., 2005).

Typical solubilization capacity of phytosterols and cholesterol along dilution line T64 areshown in Figure 2.10. The solubilization capacity of phytosterols in concentrated reversemicelle solution–like systems containing surfactant and oil phase (at 6:4 weight ratio,respectively), is 60,000 ppm (6 wt%). As can be seen from Figure 2.10, the solubilization

Improved Solubilization and Bioavailability of Nutraceuticals 25

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Figure 2.10. Solubilization capacity (SC) of cholesterol (x) and phytosterols (ο) along dilutionline T64 at 25ºC.

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 25

Page 35: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

capacity of phytosterols decreases with the increase in aqueous phase concentration. In amicroemulsion containing 90 wt% aqueous phase, the maximum solubilization capacity isonly 2400 ppm, that is, a decrease of 96% in the solubilization capacity of phytosterols.

A possible explanation for the dramatic decrease in the solubilization capacity could berelated to the nature of the solubilized molecules and to the locus of its solubilization atthe interface. In concentrates (without added water), phytosterols are entrapped at themicelle’s interface. As more aqueous phase is added, water-in-oil swollen reverse micelles(w/o microemulsions) are formed, and the hydrophilic OH groups of the phytosterolsare oriented toward the aqueous phase, causing the molecules to pack between the surfac-tant hydrophobic chains. This change in the locus of solubilization causes a decrease insolubilization capacity of the interface. Suratkar and Mahapatra (2000) observed a similarchange in the locus of solubilization of phenolic compounds in sodium dodecyl sulfate(SDS) micelles.

The decrease in solubilization capacity as the aqueous phase concentration increasesmay be attributed to microstructure transformations. The structural transformation fromw/o to o/w microstructure via bicontinuous mesophase forces the phytosterols to solubilizebetween the hydrophobic amphiphilic chains. This less-preferable location causes adecrease in the solubilization capacity. It seems that the phytosterols have a strong effect onthe spontaneous curvature of the micelles. As a result, the interface curvature decreases atlower water concentration. This effect is more pronounced in the presence of phytosterolsthan in empty micelles or in the presence of lycopene. The effect of phytosterol on choles-terol trans-membrane penetration was extensively studied. Various mechanisms have beensuggested for the decrease in the transport of cholesterol in the presence of phytosterols(Trautwein et al., 2003; Hui and Howles, 2005; Rozner and Garti, 2006). Similarly, thecompetitive adsorption of cholesterol and phytosterols in the microemulsion membraneindicates that reverse microemulsions (w/o) preferentially solubilize more cholesterol overphytosterols. Nevertheless, upon dilution, once inversion to o/w microemulsions occurs,the phytosterols are somewhat better accommodated at the interface and they displacesome of the cholesterol molecules from the interface (Figure 2.11).

Lutein and Lutein Ester

Evidence that the macular pigment carotenoids—lutein and zeaxanthin—play an importantrole in the prevention of age-related-macular degeneration, cataract and other blinding dis-orders, is now available (Vandamme, 2002; Bone et al., 2003; Semba and Dagnelic, 2003;Kim et al., 2006). Carotenoids are situated in the macula (macula lutea, yellow spot)between the incoming photons and the photoreceptors and have maximum absorption at445 nm for lutein and 451 nm for zeaxanthin. As a result, lutein and zeaxanthin can func-tion as a blue light filter (400–460 nm). The blue light enters the inner retinal layers,thereby causing the carotenoids to attenuate their intensity. In addition to the protectiveeffect of the macula from blue wavelength damage, these carotenoids can also improvevisual acuity and scavenge harmful reactive oxygen species that are formed in the photore-ceptors (Bone et al., 2003; Kim et al., 2006).

With aging, some of the eye antioxidant supplies are diminished and antioxidantenzymes are inactivated. This action appears to be related to the accumulation, aggregation,and eventual precipitation in lens opacities of damaged proteins, subsequently leading tonumerous eye disorders (Vandamme, 2002; Semba and Dagnelie, 2003).

26 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 26

Page 36: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

To improve the understanding of the potential benefits of carotenoids in general andlutein in particular, it is important to obtain more insight into their bioavailability and thefactors that determine their absorption and bioavailability.

Lutein, a naturally occurring carotenoid (Figure 2.12), is widely distributed in fruits andvegetables and is particularly concentrated in the Tagetes erecta flower. Epidemiologicalstudies suggest that high lutein intake (6 mg/day) increases serum levels that are associatedwith a lower risk of cataract and age-related-macular degeneration. Lutein can be extractedeither as a free form or as esterified (myristate, palmitate, or stearate) lutein. Both forms arepractically insoluble in aqueous systems, resulting in low bioavailability.

To improve its bioavailability, lutein was solubilized in U-type microemulsions based onR(+)-limonene. Some of the main findings are (Amar-Yuli et al., 2003, 2004; Garti et al.,2003; Amar-Yuli and Garti, 2006): (1) reverse micellar and w/o compositions solubilizedboth lutein and lutein ester better than o/w microemulsions, while maximum solubilizationis obtained within the bicontinuous phase; (2) free lutein is solubilized better than the ester-ified one in the w/o microemulsions, whereas the esterified lutein is better accommodatedwithin the o/w microemulsion; (3) vegetable oils decrease the solubilization of free lutein;(4) glycerol and alcohol enhance the solubilization of both luteins; and (5) solubilization issurfactant-dependent in all mesophase structures, but its strongest effect is in the bicontin-uous phase.

Improved Solubilization and Bioavailability of Nutraceuticals 27

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Figure 2.11. Competitive solubilization of (a, b) cholesterol alone and (c, d) combinedphytosterols and cholesterol in bile salt micelles (wt ratio of 1/1) in U-type microemulsions as afunction of water dilution. (Adapted from Garti, with permission from the publisher.)

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 27

Page 37: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

On the basis of self-diffusion coefficients of each of the ingredients, a schematic modelof the solubilization of lutein in the three possible structures along the dilution line 73(70 wt% surfactant phase and 30 wt% oil phase) was constructed. The schematic locationof the lutein at the structures is shown in Figure 2.13.

Vitamin E

Microemulsions can also serve as reservoirs for enhanced solubility of lipophilic vitaminsor other nutraceuticals within water-based formulations. The pharmaceutical literature isreplete with studies of enhanced micellar delivery of vitamins, in particular vitamin E, vita-min K1, and β-carotene (Winn et al., 1989; Traber, 2004).

Vitamin E (Figure 2.14), the major lipophilic antioxidant in human body, has invoked agreat deal of interest regarding its disease-preventive and health-promoting effects, as wellas its unique chemical structure, as a group of amphiphilic homologues exhibiting importantinterfacial roles in surfactant self-assemblies. Much interest has been devoted to microemul-sions as efficient cosmetic and drug delivery systems, enabling the solubilization ofhydrophobic active matter in aqueous media and improving its bioavailability.

Therefore, we found it imperative to study the effect of microemulsion composition onthe solubilization capacity of different forms of vitamin E and to infer the structural trans-formations from the solubilization data.

Our results (Garti et al., 2004a, b) (Figure 2.15) show the following: (1) The solubiliza-tion capacity of α-tocopherols with free-OH head groups in Tween 60-based micro-emulsions drops abruptly at either of the two dilution lines that have been studied at constantsurfactant-to-oil ratio, signifying structural transformations in the microemulsion structure.(2) The number of methyl groups on the vitamin’s polar head has an influence on the point atwhich the solubilization drop occurs, while nonsaturation of the hydrophobic tail of the vita-min enhances its solubilization capacity with no observable impact on the solubilization pat-tern. (3) In contrast to the free-OH vitamin E forms, the acetate form showed continuousdecreases in solubilization capacity along the dilution line. (4) The type of oil used inthe microemulsion has a strong influence on the solubilization pattern of the vitamin.

28 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

HO

OH

(a)

(b)

Figure 2.12. Chemical structures of (a) free lutein and (b) lutein ester.

Lakkis_Ch_02-013-040 r1.qxd 3/30/07 6:24 PM Page 28

Page 38: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Figure 2.13. Schematic model of lutein solubilization.

CH3(a)

(b)

CH3

CH3

CH3

CH3

CH3

CH3 CH3 CH3

CH3

CH3

H3C

H3C

HOCH3 CH3 CH3

CH38�R

8�R

4�R

4�R

2R

2R

O

OO

O

Figure 2.14. Chemical structures of (a) α-tocopherol and (b) α-tocopherol acetate.

29

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 29

Page 39: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Triacetin attained a higher solubilization capacity of vitamin E than R(+)-limonene with acertain retardation in the structural transformations along the dilution line. Medium-chaintriglycerides (MCT), on the other hand, maintained a constant ratio of TocOH tosurfactant with an increasing level of aqueous phase within a certain range, while the solu-bilization capacity of D-α-tocopherol acetate (TocAc) decreased significantly in the samedilution range. (5) Alcohol cosurfactants and propylene glycol (PG) were found to bevitally important for improving the solubilization capacity of TocAc and TocOH. The lattershowed a higher boost of solubilization at high levels of alcohols. (6) TocAc was found toprefer higher concentrations of Tween 60 for better solubilization, while TocOH prefersmoderate levels. Mixing Tween 60 with diglycerol monooleate (DGM) displayed a pro-nounced enhancement in the solubilization of TocAc, while it caused a significant decreasein that of TocOH. Based on these findings, a commercial vitamin E clear beverage wasdeveloped.

We have demonstrated that molecules such as essential oils, aromas, isoflavones,β-carotene, and lipoic acid have been similarly solubilized in the NSSL vehicles.

Oxidative Stability

In many cases, NSSL vehicles are loaded with nutraceuticals that are very sensitive to oxida-tion. Any preparation containing these formulations should be stable for very long periods oftime on the shelf and within the final product. Therefore, protection against environmentaloxidative attack is essential. Micelles are very dynamic systems with a very fast exchange ofthe surfactant molecules between the interface and the continuous phase.

Microemulsions are swollen micelles with similar fast exchange. However, systems thatare rich in surfactant content form very concentrated phases, where the swollen micelles(the droplets) are tightly packed. Very condensed packed systems with strong inter-dropletinteractions are obtained. In these systems the mobility of the surfactants is very restricted.In addition, stability was found to be dependent on the nature of the surfactant; therefore,even more tightly packed, worm–like, and entangled giant micelles can be formed.

The stability against oxidation of lycopene, known for its poor oxidative stability oncedissolved in solvents, was evaluated. Lycopene, if exposed to air and light, will be much

30 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 2.15. Solubilization capacities of free tocopherol (•) and tocopherol acetate (▲) in U-typemicroemulsions at several dilutions along dilution line 64 (60% surfactant phase and 40 wt% oilphase. (Adapted from Garti, 2002, with permission from the publisher.)

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 30

Page 40: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

more stable against autooxidation when solubilized in NSSL vehicles than if loaded ontoemulsion droplets, as shown in Figure 2.16. After a few weeks, the emulsified lycopene wastotally oxidized, while over 65 wt% of the NSSL lycopene remained stable. Similar resultswere obtained with other nutraceuticals (private communications).

Bioavailability

Some nutraceuticals are known to be practically insoluble in water and, therefore, tablets orcapsules that are taken orally tend to precipitate once the active ingredient is diluted withwater (in human digestive tracts). As a result, the bioavailability is very limited, and theadsorption from the intestine to the blood serum is poorly controlled. Moreover, tablets andcapsules exhibit strong fluctuations and as a result their activity is questionable. Two suchexamples that are discussed are CoQ10 and lycopene.

CoQ10 and Improved Bioavailability

Coenzyme Q10 and related ubiquinones were first discovered in 1955 and were extractedand isolated from the mitochondria. The number of side chain isoprenoid units determinesthe nomenclature. Coenzyme Q6 is found in bacteria, whereas CoQ10 is found in mam-malian mitochondria. CoQ10 is one part of a complex series of reactions that occur withinmitochondria—ultimately linked to the generation of energy within a cell. The chemicalstructure of a CoQ10 is depicted in Figure 2.17.

Improved Solubilization and Bioavailability of Nutraceuticals 31

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

100

75

50

25

0

0 28Time (days)

Emulsion

NSSL

52 72

% o

f lyc

open

e fr

om o

rigin

al

Figure 2.16. Oxidative stability to air and light of 23 mg lycopene emulsified in 10 g of o/wemulsion versus in the NSSL (modified microemulsion) vehicles.

Figure 2.17. Chemical structure of CoQ10.

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 31

Page 41: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Virtually every cell in the human body contains coenzyme Q10. The mitochondria, thearea of cells where energy is produced, contains most of the human coenzyme Q10. Theheart and the liver, due to their high content of mitochondria per cell, contain the greatestquantity of coenzyme Q10. Coenzyme Q10 supplementation has helped some people withcongestive heart failure (Salles et al., 2006; Yamamoto, 2006). Ubiquinone, or coenzymeQ10, is an important heart nutrient, used primarily by those who take pills against high cho-lesterol levels. Certain lipid-lowering drugs, such as statins as well as oral agents, whichlower blood sugar, cause a decrease in serum levels of coenzyme Q10 and reduce the effectsof coenzyme Q10 supplementation (Mortensen et al., 1997; Palomaki et al., 1998; Lankin,2003; Passi et al., 2003; Bettowski, 2005; Cenedella et al., 2005; Hargreaves et al., 2005;Mabuchi et al., 2005; Strey et al., 2005). These drugs inhibit the production of coenzymeQ10 by the liver, and will cause serious complications, unless one supplements coenzymeQ10 back into the diet. A prescription for lipid-lowering statin drugs should always beaccompanied with a recommendation to take coenzyme Q10, because if a person is deficientin coenzyme Q10, heart failure is more likely.

The second major use of coenzyme Q10 would be in the case of congestive heart failure,where it is particularly effective. Its importance to the human heart is illustrated by the factthat the heart may cease to function when coenzyme Q10 levels fall by 75%. Schematicactivity within the mitochondria of CoQ10 is demonstrated in Figure 2.18.

Adenosine triphosphate (ATP) is present in every cell of human organs. It serves as asource of energy for many of the body’s biochemical processes and represents the reserve

32 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N Figure 2.18. Schematic functionality of CoQ10 in mitochondria.

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 32

Page 42: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

energy in the muscles. The heart needs a constant supply of ATP, which cannot be producedwithout coenzyme Q10. Coenzyme Q10 is the catalyst for the creation of ATP. This meansthat coenzyme Q10 plays a vital role in the inner workings of the human body. Several otherchronic diseases are associated with lack of CoQ10 such as Parkinson’s disease (Andrey andGille, 2003; Batandier et al., 2004; Genova et al., 2004; Sharma et al., 2004; Arroyo andNavas, 2005; Ebadi et al., 2005; Dhanasekaran and Ren, 2005; Moriera et al., 2005). It isalso a potent antioxidant since it fights the harmful free radicals generated during normalmetabolism.

The highest dietary sources of CoQ10 come from fresh sardines and mackerel, the heart,the liver, and beef, lamb, and pork, as well as from eggs. There are plenty of vegetablesources of CoQ10, the richest being spinach, broccoli, peanuts, wheat germ, and wholegrains, although the amount is significantly smaller than that found in meat.

Coenzyme Q10 is primarily offered in tablet, capsule, or soft gel forms containing ayellow-orange powder. The tablet form, being much less digestible, is not recommended.Adult levels of supplementation are usually 30–90 mg/day, although individuals with spe-cific health conditions may supplement with higher levels, such as 100 mg 3–4 times perday. Most of the research on heart conditions has used 90–150 mg/day. CoQ10 is fat solubleand, like most other fat-soluble compounds, is poorly absorbed from the gastrointestinaltract, especially when taken on an empty stomach. Therefore, it is recommended thatCoQ10 be taken with a meal or in a formulation, such as oil phase, that will improve itsbioavailability and, hence, absorption. Our studies on humans were conducted at the Tech-nical University of Tokyo by Prof. Yamamoto on eight individuals who were fed for 28 dayswith CoQ10 from a commercial product known as “275% more bioavailable”: and with ourNSSL vehicles incorporated into soft gels. The individual intake was of 150 mg CoQ10 perday (Yamamoto, 2005).

The efficacy of the NSSL-based formulations versus the commercial product is demon-strated in Figures 2.19–2.21. It can be clearly concluded that (1) CoQ10 in the NSSLvehicles is more bioavailable than the commercial product in soft gels (claimed to be 275%more bioavailable than other products in tablets); (2) the ratio of total CoQ10 to total choles-terol in the blood stream derived from the NSSL soft gels is higher than from the commer-cial product, indicating that the NSSL vehicles provide extra activity to the CoQ10, whichassists in maintaining total cholesterol at lower levels; (3) it is well documented that severalnutraceuticals and oil-soluble phytochemicals tend to interfere with the absorption of

Improved Solubilization and Bioavailability of Nutraceuticals 33

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Figure 2.19. Bioavailability of CoQ10 in humans given a total of 150 mg of active matter in two daily doses in two types of formulations, in best commercial formulation in the market place (entitled 275% more bioavailable, filled bar) versus the CoQ10 solubilized in NSSL vehicles(white bar).

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 33

Page 43: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

34 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 2.20. Ratio of CoQ10 (TQ) to total cholesterol (TC) in human blood when given 150 mg of CoQ10 in two daily doses in two types of formulations, in best commercial formulation in themarket place (entitled 275% more bioavailable, filled bar) versus the CoQ10 solubilized in NSSLvehicles (white bar).

Figure 2.21. Ratio of vitamin E (VE) to total cholesterol in human blood given a total of 150 mgof CoQ10 in two daily doses in two types of formulations, in best commercial formulation in themarket place (entitled 275% more bioavailable, filled bar) versus the CoQ10 solubilized in NSSLvehicles (white bar).

vitamins. Therefore, it was expected that the vitamin E levels in the blood stream woulddecrease with the intake of CoQ10. However, it was found in the human blood tests that vita-min E levels did not decrease in the presence of CoQ10 when CoQ10 was taken in the NSSLvehicles. In fact, it remained at higher levels when compared to its levels derived from thecommercial product.

On the basis of these and other findings, we have proposed a highly schematic cartoonedmodel (Figure 2.22) of the transport of the nutraceuticals across human membranes. Themodel shows how the vehicle that is dispersed in the aqueous phase approaches the mem-brane and adheres to it. The CoQ10 is transported across the membrane, while the emptyvehicles depart and are excreted from the digestive tract. It should be noted that the surfac-tants do not cross the membrane.

Water Binding

The activity of water plays a significant role in any reaction (chemical or enzymatic) thatexists in food systems and related products. Microemulsions of w/o can serve as microreac-tors for several such processes, mainly for Maillard reactions (Lutz et al., 2005). Water-in-oilnanodroplets can be free or bound to the head groups of the surfactants. Thus, the ability to

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 34

Page 44: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

estimate the activity of the water and the binding capacity of the surfactants is of highimportance whenever a triggered reaction is required. At certain water levels, the water inthe core of the microemulsion will be bound and the activity will be minimal; thus, the reac-tivity of the ingredients (sugars and proteins in Maillard reactions or enzymes in hydrolysisprocesses) will be low. Upon adding more water and reaching a point where the waterbecomes free, the reactions will be triggered (Yaghmur et al., 2003a, b).

We (Spernath, 2003; Yamomoto, 2006) examined, by a sub-zero differential scanningcalorimeter (DSC) technique, the nature of the water in the confined space of a w/omicroemulsion, to better understand the role of the entrapped water, in order to controlenzymatic reactions carried out in the inner phase (Spernath et al., 2003; Yaghmur et al.,2003). We reported (Figure 2.23) that the surfactant/alcohol/PG can strongly bind water inthe inner phase, so that it freezes below –10°C and acts, in part, as bound water and, in part,as non-freezable water (Spernath et al., 2003). Even after complete inversion to o/w micro-emulsions, the water in the continuous phase strongly interacts with the cosolvent/surfac-tant and remains partially bound.

The water in the core of nonionic microemulsions containing, in addition to the surfac-tants, polyols and alcohol, is strongly bound to the surfactant head group and/or to thepolyol groups and freezes at subzero temperatures. The amount of bound water stronglydepends on the amounts of the surfactants present in each microdroplet, on the nature of thehead groups, and on the amounts of cosolvents (alcohol and PG).

On the basis of these findings, Maillard reactions, model reactions of furfural and cys-teine and glucose and isoleucine (Ezrahi et al., 2001; Fanun et al., 2001; Yaghmur et al.,2002a, b, 2003, 2005; Lutz et al., 2005), as well as hydrolysis of phosphatidylcholine byphospholipase L2 (PLA2) to lysolecithin (Garti et al., 1997) were studied. It was found thatthe reactions do not start (lag time) until sufficient water is added to exceed the free waterthreshold. The reactions are, therefore, very well triggered and controlled by the wateractivity within the core of the microdroplets. The reaction rates can be delayed or speededby immobilizing (confining) or freeing the water in the core of the microdroplets.

Improved Solubilization and Bioavailability of Nutraceuticals 35

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Figure 2.22. Schematic representation of the microemulsion droplet approaching the membraneand releasing the nutraceutical molecules. The surfactant does not cross the membrane.

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 35

Page 45: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Conclusions

Microemulsions have been known for several decades, but their utilization in food systemshas been very limited owing to some major structural limitations and the nature of thesurfactants and the oils. Another major drawback is that in most cases they were undilutablewith water. In recent years, after significant efforts by colloid chemists, experimentalists,and others, some of the key characteristics related to the packing of the surfactant, freeenergy gain, geometries, and so on, shed light on the basic requirements needed to designU-type phase diagrams. The latter consist of large isotropic regions and have provedcapable of making concentrates that can be easily diluted with water and oil phases. In thecourse of our studies we also learned that:

• Self-assembled, hydrophilic surfactant in oil phase, in the presence of cosolvents andcosurfactants, can provide high solubilization capacities for entrapment of immisciblephases and active guest molecules. These microstructures can be diluted with excesswater to form crystal-clear (transparent) solution-like, isotropic phases, loaded with theactive matter.

• If the ingredients composing the microemulsions and the cosolvents and cosurfactantsare carefully selected, one can form a variety of beverage microemulsions.

• Microemulsions of U-type with progressive full dilution with aqueous phase can beformulated.

36 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

35

30

25

20

15

10

Inte

rpha

sal o

r fr

ee w

ater

(w

t%)

5

00 5 10 15 20

Water content (wt%)

25 30 35 40

Figure 2.23. The amounts (weight percent of free and bound) of interphasal water inmicroemulsions based on sugar esters along dilution line 64 (60% surfactant and 40% oil phase). (o) Bulk (free) water and (•) interphasal (bound) water. (Adapted from Garti, 1995, with permission from the publisher.)

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 36

Page 46: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

• Microemulsions of w/o and bicontinuous structures, as well as o/w microemulsions cansolubilize guest molecules at their interface at high solubilization capacities, in somecases up to 100-fold of the solubility of the nutraceuticals in the corresponding solvent!

• Molecules such as lycopene, vitamin E, tocopherols and tocopherol acetate, β-carotene,lutein, phytosterols, and CoQ10 can be quantitatively solubilized.

• Microemulsions provide some oxidative protection to the nutraceuticals.• Various other guest molecules such as aromas, flavors, and antioxidants can be solubi-

lized in the microemulsions.• Water entrapped at the core of a w/o microemulsion can be strongly bound to the surfac-

tant head group that will restrict the water activity. Thus, upon adding more water, thereaction by the enzyme or regents can be triggered.

It seems that we are now ready to start using microemulsions in beverages and other foodsystems and to incorporate active ingredients within high-quality food for the benefit ofhuman nutrition and health.

ReferencesAlander, J.; Warnheim, T. 1989a. Model microemulsions containing vegetable-oils. 1. nonionic surfactant sys-

tems. JAOCS 66:1656–1660.Alander, J.; Warnheim, T. 1989b. Model microemulsions containing vegetable-oils. 2. ionic surfactant systems.

JAOCS 66:1661–1665.Amar-Yuli, I.; Garti, N. 2006. Progress in Structural Transformation in Lyotropic Liquid Crystals. Colloids and

Surfaces 282: 435–456.Amar-Yuli, I.; Aserin, A.; Garti, N. 2003. Solubilization patterns of lutein and lutein esters in food grade nonionic

microemulsions. J. Agric. Food Chem. 51(16):4775–4781.Amar-Yuli, I.; Aserin, A.; Garti, N. 2004. Microstructure transitions derived from solubilization of lutein and

lutein esters in food microemulsions. Colloids Surfaces B, 33:143–150.Andrey, V.; Gille, L. 2003. The biomolecule ubiquinone exerts a variety of biological functions. BioFactors

18(1–4):23–31.Arroyo, A.; Navas, P. 2005. Specificity of coenzyme Q10 for a balanced function of respiratory chain and endoge-

nous ubiquinone biosynthesis in human cells. Biochim. Biophys. Acta, Bioenergetics 1706(1–2):174–183.Batandier, C.; Leverve, X.; Fontaine, E. 2004. Opening of the mitochondrial permeability transition pore induces

reactive oxygen species production at the level of the respiratory chain complex I. J. Biol. Chem.279(17):17197–17204.

Bettowski, J. 2005. Statins and modulation of oxidative stress. Toxicol. Mech. Meth. 15(2):61–92.Billman, J.F.; Kaler, E.W. 1991. Structure and phase-behavior in 4 component nonionic microemulsions.

Langmuir 7:1609–1617.Bloch, K. 1988. Sterol structure and function. J. Am. Oil Chem. Soc. 65:1763–1766.Bone, R.A.; Landrum, J.T.; Guerra, L.H.; Ruiz, C.A. 2003. Lutein and zeaxanthin dietary supplements raise mac-

ular pigment density and serum concentrations of these carotenoids in humans. J. Nutrition 133(4):992–998.Bramley, P.M. 2000. Is lycopene beneficial to human health? Phytochemistry 54:233–236.Cenedella, R.J.; Neely, A.R.; Sexton, P. 2005. Concentration and distribution of ubiquinone (coenzyme Q), the

endogenous lipid antioxidant, in the rat lens: effect of treatment with simvastatin. Mol. Vis. 11:594–602.Constantinides, P.P.; Scalart, J.P. 1997. Formation and physical characterization of water-in-oil microemulsions

containing long- versus medium-chain glycerides. Int. J. Pharm. 158:57–68.Cooke, J.P. 1998. Nutraceuticals for cardiovascular health. Am. J. Cardiol. 82:43S–46S.de Campo, L.; Yaghmur, A.; Garti, N.; Leser, M.E.; Glatter, O. 2004. Food-grade microemulsions: structural char-

acterization. J. Colloid Interface Sci. 274(1):251–267.Dhanasekaran, M.; Ren, J. 2005. The emerging role of coenzyme Q-10 in aging, neurodegeneration, cardiovascu-

lar disease, cancer and diabetes mellitus. Curr. Neurovasc. Res. 2(5):447–459.Dungan, S.R. 1997. “Microemulsions in Foods: Properties and Applications.” In: Industrial Applications of

Microemulsions, edited by C. Solans and H. Kunieda, pp. 148–170. Marcel Dekker Inc.

Improved Solubilization and Bioavailability of Nutraceuticals 37

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 37

Page 47: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Ebadi, M.; Brown-Borg, H.; El Refaely, H.; Singh, B.B.; Garret, S.; Shavali, S.; Sharma, S.K. 2005. Metallothionein-mediated neuro protection in genetically engineered mouse models of Parkinson’s disease. Molecular BrainRes. 134(1):67–75.

Ezrahi, S.; Aserin, A.; Garti, N. 1999. “Aggregation Behavior in One-Phase (Winsor IV) Microemulsion Sys-tems.” In Handbook of Microemulsion Science and Technology, edited by P. Kumar and K.L. Mittal, pp.185–246. Marcel Dekker Inc.

Ezrahi, S.; Aserin, A.; Fanun, M.; Garti, N. 2001. “Subzero Temprature Behavior of Water in Microemulsions.” InThermal Behavior of Dispersed Systems, edited by N. Garti, pp. 59–120. Marcel Dekker Inc.

Fanun, M.; Leser, M.; Aserin, A.; Garti, N. 2001. Sucrose ester microemulsions as microemulsions as microreac-tors for model Maillard reaction. Colloids Surfaces A: Physicochem. Eng. Asp. 194(1–3):175–187.

Garti, N.; Aserin, A.; Mytayeva, U. 2007. Submitted for publication.Garti, N.; Lichtenberg, D.; Silberstein, T. 1997. The hydrolysis of phosphatidylcholine A2 in microemulsion as

microreactor. Colloids Surfaces A: Physicochem. Eng. Asp. 128(1–3):17–25.Garti, N.; Clement, V.; Fanun, M.; Leser, M.E. 2000a. Some characteristics of sugar ester nonionic microemul-

sions in view of possible food applications. J. Agric. Food Chem. 48(9):3945–3956.Garti, N.; Aserin, A.; Fanun, M. 2000b. Non-ionic sucrose esters microemulsions for food applications. Part 1.

Water solubilization. Colloids Surfaces A: Physicochem. Eng. Asp. 164(1):27–38.Garti, N; Yaghmur, A; Leser, M.E; Clement,V; Watzke, H.J. 2001. Improved oil solubilization in O/W food-grade

microemulsions in the presence of polyols and ethanol. J. Agric. Food Chem. 49:2552–2562.Garti, N.; Amar, I.; Yaghmur, A.; Spernath, A. 2003. Interfacial modification and structural transitions induced by

guest molecules solubilized in U-type nonionic food-grade microemulsions. J. Disper. Sci. Technol. 24(3 and4):397–410.

Garti, N.; Yaghmur, A.; Aserin, A.; Spernath, A.; Elfakess, R.; Ezrahi, S. 2004a. Solubilization of active moleculesin microemulsions for improved solubilization, and environmental protection. Colloids Surfaces A: Physic-ochem. Eng. Asp. 230(1–3):183–190.

Garti, N.; Zakharia, I.; Spernath, A.; Yaghmur, A.; Aserin, A.; Hoffman, R.E.; Jacobs, L. 2004b. Solubilization ofwater-insoluble nutraceuticals in nonionic microemulsions for water-based use. Progr. Colloid Polym. Sci.126:184–189.

Garti, N.; Spernath, A.; Aserin, A.; Lutz, R. 2005. Nano-sized self-assemblies of nonionic surfactants as solubi-lization reservoirs and microreactors for food systems. Soft Matter 1:206–218.

Garti, N.; Aserin, A.; Mytayeva, U. 2007. In preparation.Gasco, M.R. 1997. “Microemulsions in the Pharmaceutical Field: Perspectives and Applications.” In Industrial

Applications of Microemulsions, edited by C. Solans and H. Kunieda, pp. 97–122. Marcel Dekker Inc.Genova, M.L.; Pich, M.M.; Bernacchia, A.; Binachi, C.; Biondi, A.; Bovina, C.; Falasca, A.I.; Formiggini, G.;

Castelli, G.P.; Lenaz, G. 2004. The mitochondrial production of reactive oxygen species in relation to aging andpathology. Annals New York Academ. Sci. 1011:86–100.

Hamdan, S.; Lizana, R.; Laili, C.R. 1995. Aqueous and nonaqueous microemulsion systems with a palm oil-baseemollient. JAOCS 72:151–155.

Hargreaves, L.P.; Duncan, A.J.; Heales, S.J.R.; Land, J.M. 2005. The effect of HMG-CoA reductase inhibitors oncoenzyme Q10. Possible biochemical/clinical implications. Drug Safety 28(8):659–676.

Hicks, K.B.; Moreau, R.A. 2001. Phytosterols and phytostanols: functional food cholesterol busters. Food Tech-nol. 55:63–67.

Holmberg, K. 1998. “Quarter Century Progress and New Horizons in Microemulsions.” In Micelles, Microemul-sions, and Monolayers, edited by O. Shah, pp. 161–192. Marcel Dekker Inc.

Hou, M.J.; Shah, D.O. 1987. Effects of the molecular structure of the interface and continuous phase on solubi-lization of water in water/oil microemulsions. Langmuir 3:1086–1096.

Hui, D.Y.; Howles, P.N. 2005. Molecular mechanisms of cholesterol absorption and transport in the intestine.Semin. Dev. Biol. 16(2):183–192.

Huibers, P.D.T.; Shah, D.O. 1997. Evidence for synergism in nonionic surfactant mixtures: enhancement of solu-bilization in water-in-oil microemulsions. Langmuir 13:5762–5765.

IUPAC, The Nomenclature of Steroids (Recommendations 1989), International Union of Pure and Applied Chem-istry and International Union of Biochemistry and Molecular Biology. Available: http:// www.chem.qmw.ac.uk/iupac/steroid/ [1989].

Jones, P.J.H.; MacDougall, D.E.; Ntanios, F.; Vanstone, C.A. 1997. Dietary phytosterols as cholesterol-loweringagents in humans. Can. J. Physiol. Pharmac. 75:217–227.

38 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 38

Page 48: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Joubran, R.F.; Cornell, D.G.; Parris, N. 1993. Microemulsions of triglyceride and non-ionic surfactant-effect oftemperature and aqueous phase composition. Colloids Surf. A 80:153–160.

Kahlweit, M.; Busse, G.; Faulhaber, B.; Jen, J. 1996. Shape changes of globules in nonionic microemulsions.J. Phys. Chem. 100:14991–14994.

Kanei, N.; Tamura, Y.; Kunieda, H. 1999. Effect of types of perfume compounds on the hydrophile-lipophile bal-ance temperature. J. Colloid Interface Sci. 218:13–22.

Kim, S.R.; Nakanishi, K.; Itagaki, Y.; Sparrow, J.R. 2006. Photooxidation of A2-PE, a photoreceptor outer seg-ment fluorophore, and protection by lutein and zeaxanthin. Experimental Eye Research 82(5):828–839.

Lankin, V.Z.; Tikhaze, A.K.; Kukharchuk, V.V.; Konovalova, G.G.; Pisarenko, O.I.; Kaminnyi, A.I.; Shumaev,K.B.; Belenkov, Y.N. 2003. Antioxidants decreases the intensification of low density lipoprotein in vivo peroxi-dation during therapy with statins. Molecular Cell. Biochem. 249(1 and 2):129–140.

Lutz, R.; Aserin, A.; Garti, N. 2005. Maillard reaction between leucine and glucose in o/w microemulsion mediain comparison to aqueous solution. J. Disper. Sci. Tech. 26(5):535–547.

Mabuchi, H.; Higashikata, T.; Kawashiri, M.; Katsuda, S.; Mizuno, M.; Nohara, A.; Inazu, A.; Koizumi, J.;Kobayashi, J. 2005. Reduction of serum ubiquinol-10 and ubiquinone-10 levels by atorvastatin in hypercholes-terolemic patients. J. Atherosclerosis Thrombosis 12(2):111–119.

Malcolmson, C.; Lawrence, M.J. 1995. Three-component non-ionic oil-in-water microemulsions using poly-oxyethylene ether surfactants. Colloids Surfaces B: Biointerfaces 4:97–109.

Moriera, P.I.; Santos, M.S.; Sena, C.; Nunes, E.; Seica, R.; Oliveira, C.R. 2005. CoQ10 therapy attenuatesamyloid—peptides toxicity in brain mitochondria isolated from aged diabetic rats. Exp. Neurol.196(1):112–119.

Mortensen, S.A.; Leth, A.; Agner, E.; Rohde, M. 1997. Dose-related decreases of serum coenzyme Q10 duringtreatment with HMG-CoA reductase inhibitors. Molecular Aspects Med. 18(Suppl.):s137–s144.

Nes, W.R. 1987. “Multiple Roles for Plant Sterols.” In The Metabolism, Structure and Function of Plant Lipids,edited by P.K. Stumpf, B.J. Mudd and W.R. Nes, pp. 3–9. Plenum Press.

Palomaki, A.; Malminiemi, K.; Solakivi, T.; Malminiemi, O. 1998. Ubiquinone supplementation during lovastatintreatment: effect on LDL oxidation ex vivo. J. Lipid Res. 39(7):1430–1437.

Park, K.M.; Kim, C.K. 1999. Preparation and evaluation of flurbiprofen-loaded microemulsion for parenteraldelivery. Int. J. Pharm. 181:173–179.

Passi, S.; Stancato, A.; Aleo, E.; Dmitrieva, A.; Littarru, G.P. 2003. Statins lower plasma and lymphocyteubiquinol/ubiquinone withput affecting and PUFA. Biofactors 18(1–4):113–124.

Pelletier, X.; Belbraouet, S.; Mirabel, D.; Mordret, F.; Perrin, J.L.; Pages, X.; Debry, G. 1995. A diet moderatelyenriched in phytosterols lowers plasma cholesterol concentrations in normocholesterolemic humans. Ann. Nutr.Metab. 39:291–295.

Peterson, D.W.; Nichols, C.W.; Schneour E.W. 1951. Some relationships among dietary sterols, plasma and livercholesterol levels and atherosclerosis in chicks. Proc. Soc. Exp. Biol. Med. 78:1143–1147.

Piironen, V.; Lindsay, D.G.; Miettinen, T.A.; Toivo, J.; Lampi, A.M. 2000. Review—plant sterols: biosynthesis,biological function and their importance to human nutrition. J. Sci. Food Agric. 80:939–966.

Prichanont, S.; Leak, D.J.; Stuckey D.C. 2000. The solubilization of mycobacterium in a water-in-oil microemul-sion for biotransformations: system selection and characterization. Colloids Surfaces A: Physicochem. Eng.Asp. 166:177–186.

Radomska, A.; Dobrucki, R. 2000. The use of some ingredients for microemulsion preparation containing retinoland its esters. Int. J. Pharm. 196:131–134.

Rao, A.V.; Agarwal, S. 1999. Role of lycopene as antioxidant carotenoid in the prevention of chronic diseases: areview. Nutr. Res. 19:305–323.

Regev, O.; Ezrahi, S.; Aserin, A.; Garti, N.; Wachtel, E.; Kaler, E.W.; Khan, A.; Talmon, Y. 1996. A study of themicrostructure of a four-component nonionic microemulsion by cryo-TEM, NMR, SAXS, and SANS. Lang-muir 12:668–674.

Rozner, S.; Garti, N. 2006. The activity and absorption relationship of cholesterol and phytosterols. Colloids andSurfaces A: Physicochem. Eng. Aspects 282:435–456.

Ostlund, R.E. Jr. 2002. Phytosterols in human nutrition. Annu. Rev. Nutr. 22:533–549.Salles, J.E.; Moises, V.A.; Almeida, D.R.; Chacra, A.R.; Moises, R.S. 2006. Myocardinal dysfunction in mito-

chondrial diabetes treated with Coenzyme Q10. Diabet. Res. Clin. Pract. 72(1):100–103.Semba, R.D.; Dagnelie, G. 2003. Are lutein and zeaxanthin conditionally essential nutrients for eye health? Med.

Hypotheses 61(4):465–472.

Improved Solubilization and Bioavailability of Nutraceuticals 39

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 39

Page 49: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Sharma, S.; Kheradpezhou, M.; Shavali, S.; El Refaely, H.; Eken, J.; Hagen, C.; Ebadi, M. 2004. Neuroprotectiveactions of coenzyme Q10 in Parkinson’s disease. Meth. Enzymol. 382(Quinones and Quinone Enzymes,Part B):488–509.

Shinoda, K.; Lindman, B. 1987. Organized surfactant systems: microemulsions. Langmuir 3:135–149.Solans, C.; Pons, R.; Kunieda, H. 1997. “Overview of Basic Aspects of Microemulsions.” In Industrial Applica-

tions of Microemulsions, edited by C. Solans and H. Kunieda, pp. 1–17. Marcel Dekker Inc.Spernath, A.; Yaghmur, A.; Aserin, A.; Hoffman, R.E.; Garti, N. 2002. Food grade microemulsions based on non-

ionic emulsifiers: media to enhance lycopene solubilization. J. Agric. Food Chem. 50:6917–6922.Spernath, A.; Yaghmur, A.; Aserin, A.; Hoffman, R.E.; Garti, N. 2003. Phytosterols solubilization capacity and

microstructure transitions in Winsor IV food-grade microemulsions studied by self-diffusion NMR. J. Agric.Food Chem. 51(8):2359–2364.

Strey, C.H.; Young, J.M.; Molyneux, S.L.; George, P.M.; Florkowski, C.M.; Scott, R.S.; Frampton, C.M. 2005.Endothelium-ameliorating effects of statin therapy and coenzyme Q10 reductions in chronic heart failure. Ath-erosclerosis 179:201–206.

Suratkar, V.; Mahapatra, S. 2000. Solubilization site of organic perfume molecules in sodium dodecyl sulfatemicelles: new insights from proton NMR studies. J. Colloid Interface Sci. 225:32–38.

Tokuoka, Y.; Uchiyama, H.; Abe, M.; Christian, S.D. 1995. Solubilization of some synthetic perfumes by anionic-nonionic mixed surfactant systems 1. Langmuir 11:725–729.

Traber, M.G. 2004. The ABCs of vitamin E and β-carotene absorption. Am. J. Clin. Nutr. 80(1):3–4.Trautwein, E.A.; Duchateau, G.S.M.J.E.; Lin, Y.G.; Mel’nikov, S.M.; Molhuizen, H.O.F.; Ntanios, F.Y. 2003. Pro-

posed mechanisms of cholesterol-lowering action of plant sterols. Eur. J. Lipid Sci. Technol. 105 (3–4):171–185.Trevino, S.F.; Joubran, R.; Parris, N.; Berk, N.F. 1998. Structure of a triglyceride microemulsion: a small-angle

neutron scattering study. J. Phys. Chem. B 102:953–960.Trotta, M.; Morel, S.; Gasco, M.R. 1997. Effect of oil phase composition on the skin permeation of felodipine

from o/w microemulsions. Pharmazie 52:50–53.Vandamme, T.F. 2002. Microemulsions as ocular drug delivery systems: recent developments and future chal-

lenges. Progr. Retinal Eye Res. 21(1):15–34.Van het Hof, K.H.; West, C.E.; Weststrate, J.A.; Hautvast, J.G.A.J. 2000. Dietary factors that affect the bioavail-

ability of carotenoids. J. Nutr. 130:503–506.von Corswant, C.; Söderman, O. 1998. Effect of adding isopropyl myristate to microemulsions based on soybean

phosphatidylcholine and triglyceride. Langmuir 14:3506–3511.von Corswant, C.; Engström, S.; Söderman, O. 1997. Microemulsions based on soybean phophatidylcholine and

triglyceride phase behavior and microstructure. Langmuir 13:5061–5070.Warisnoicharoen, W.; Lansley, A.B.; Lawrence, M.J. 2000. Nonionic oil-in-water microemulsions: the effect of oil

type on phase behavior. Int. J. Pharm. 198:7–27.Winn, M.J.; White, P.M.; Scott, A.K.; Pratt, S.K.; Park, B.K. 1989. The bioavailability of a mixed micellar prepa-

ration of vitamin K1, and its procoagulant effect in anticoagulated rabbits. J. Pharm. and Pharmacol.41(4):257–260.

Yaghmur, A.; Aserin, A.; Garti N. 2002a. Furfural-cysteine model reaction in food-grade nonionic o/wmicroemulsions for selective flavor formation. J. Agric. Food Chem. 50:2878–2883.

Yaghmur, A.; Aserin, A.; Garti N. 2002b. Phase behavior of microemulsions based on food-grade nonionic surfac-tants: effect of polyols and short-chain alcohols. Colloids Surfaces A 209:71–81.

Yaghmur, A.; Aserin, A.; Tiunova, I.; Garti, N. 2002c. Structural behavior of nonionic surfactants in the presenceof propylene glycol in nonionic microemulsions studied by DSC. J. Thermal Anal. Cal. 69:163–177.

Yaghmur, A.; Aserin, A.; Antalek, B.; Garti, N. 2003a. Microstructure of five-component food grade oil-in-watermicroemulsions by PGSE-NMR, conductivity, and viscosity. Langmuir 19(4):1063–1068.

Yaghmur, A.; Fanun, M.; Aserin, A.; Garti, N. 2003b. “Food Grade Microemulsions Based on Nonionic Emulsi-fiers as Microreactors for Selective Flavor Formation by Maillard Reaction.” In Self-Assembly, edited by B.H.Robinson, pp. 144–151, IOS Press.

Yaghmur, A.; de Campo, L.; Glatter, O.; Leser, M.E.; Garti, N. 2004. Structural characterization of five-component food grade oil-in-water nonionic microemulsions. PCCP 6(7):1524–1533.

Yaghmur, A.; Aserin, A.; Abbas, A.; Garti, N. 2005. Reactivity of furfural-cysteine model reaction in food gradefive-component nonionic microemulsions. Colloids and Surfaces A 253(1–3):223–234.

Yamamoto, Y. 2005. Private communication report.Yamamoto, Y. 2006. Coenzyme Q10, free radicals, and heart disease. Oxidative Stress and Disease 21(Molecular

Interventions in Lifestyle-Related Diseases):37–46.

40 Chapter 2

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_02-013-040 r1.qxd 3/29/07 1:36 PM Page 40

Page 50: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

3 Emulsions as Delivery Systems in Foods

Ingrid A.M. Appelqvist, Matt Golding, Rob Vreeker, and Nicolaas Jan Zuidam

Introduction

Many industries use emulsion technology as a delivery vehicle for either aqueous- or oil-based actives (or both). Examples include paint, pharmaceutical and bitumen industries. Inall cases, there are two considerations that must be taken into account when formulating anemulsion for controlled delivery. First, the emulsion system must be (storage) stable rightup to the point of application. Secondly, upon its application the emulsion should behave ina consistent manner so that it achieves the desired delivery. In many (but by no means all)cases this equates to the “making and breaking” of emulsions for stability and subsequentdelivery.

Emulsion systems are, of course, an integral part of food manufacturing. Emulsion tech-nology in the context of foods is not in itself novel—examples include milk, dairy cream,and mayonnaise. The latter can be traced back to the 17th century. However, the use ofemulsions as delivery vehicles represents a rapidly developing area for the application ofemulsions within the food industry.

Similar to other industries, same essential formulation and processing considerationsapply. First, the emulsion should be stable up to the point of application—in other words:shelf stable. This is true for all food emulsions, although there may be significant variationin the length of time that the product is required to be stable. Generally, this is limited by themicrobiological stability of the particular product: pasteurized emulsions, such as milk orcream, may have a two-week shelf life. In contrast, sterilized emulsions such as crèmeliqueurs may be stable for over a year. However, in all cases it is important that during thelifetime of the product the emulsion does not show signs of instability or phase separation.Secondly, the emulsion should be designed so that it performs in a defined manner uponapplication.

In food products, there are effectively two main points of application: consumption anddigestion. From a consumer perspective, the first point of application might be construedas being the most important. The whole sensory experience of food is dominated byits behavior in the mouth. Emulsions play an important role here, both in terms of flavordelivery and release and in terms of textural behavior and response in the mouth. Mouthis a remarkably sensitive tool at differentiating between organoleptic sensations—most of us are able to differentiate between skimmed, semi-skimmed and whole milk.Consequently, even small changes to emulsion composition and in-mouth behaviorcan have a significant impact on whether a particular product is perceived in a positive ornegative way.

41

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 41

Encapsulation and Controlled Release: Technologies in Food SystemsEdited by Jamileh M. Lakkis

Copyright © 2007 by Blackwell Publishing

Page 51: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

In recent years, consumer demand for highly nutritional food products has increased andcan be interpreted as:

1. removal of so-called “bad” ingredients, such as sugar, fat (saturated or trans) and salt;2. enhancement of “good” ingredients, such as fiber, protein or fruit and vegetable content

and;3. direct fortification with actives, such as vitamins, minerals, ω-3 oils.

In all cases it is important that not only there is no compromise in quality but also anyclaimed fortification should have good bioavailability during digestion. Consequently, inaddition to in-mouth behavior, emulsion systems are becoming increasingly utilized in foodproducts as a means of achieving controlled delivery in the gastrointestinal (GI) tract as well.

This chapter highlights recent developments in the application of food emulsions asdelivery vehicles from the consideration of both mouth and gut as areas for targeted delivery.We aim to demonstrate the technical challenges and solutions for delivering both oil- andwater-soluble actives, providing examples from flavor delivery in mouth to delivery of activecompounds and sterols under gastric conditions. We also aim to show how nature can pro-vide solutions for the application of emulsions as delivery systems, as well as looking atfuture developments and opportunities in this richly diverse field.

Stabilization and Destabilization of Emulsion Systems

Emulsion Stabilization

Processed foods are often complex multiphase systems. In the cases where both water andoil are present, emulsification is of course necessary to prevent separation of these twoincompatible phases. Emulsion design within the food industry is not a trivial issue. Thediversity of manufactured food and beverages means that the relative balance of water andoil phases can vary widely depending on product type, and both oil-in-water (o/w) andwater-in-oil (w/o) type emulsions have found a wide variety of applications. Some examplesof food emulsions along with concentrations of water and oil are given in Table 3.1.

Food emulsions are created and stabilized through a combination of process and formu-lation design. Homogenization facilitates droplet break-up to create the dispersed phase,whilst food ingredients displaying appropriate amphipathic properties are able to adsorbonto the newly formed droplet interfaces during homogenization to provide electrostatic

42 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Food Emulsion type Fat/oil content (wt%)

Milk o/w 0–4Ice creama o/w 0–10Cream o/w 20–50Light mayonnaise o/w 20–50Mayonnaise o/w 65–75Butter w/o 80Margarine w/o 80

Table 3.1. Examples of typical food emulsions and their relative concentration of fat

a Ice cream can be considered as a four-phase colloid, comprising dispersed phases of ice, air, and fat in aconcentrated continuous phase.

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 42

Page 52: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

(formation of a charged interface) or steric (formation of a viscoelastic interface) stabiliza-tion against immediate coalescence.

It can be noted that industrial manufacturing of food emulsions generally employs onlya limited number of homogenization technologies, depending on product type. Colloid or Ross mills are commonly used in the manufacture of mayonnaise or similar productswith high oil content. High-pressure homogenizers are used in the manufacture of suchproducts as ice cream, (homogenized) milk, and other beverages as well as many other softsolid products of low-to-intermediate fat content. Water-in-oil emulsions, such as mar-garines, are most commonly prepared on votator lines. Other aspects of processing, too,play an important role in the formation of food emulsions, such as pre-homogenization andthermal treatment (pasteurization, sterilization); however, these will not be discussed aspart of this chapter. More information on the processing aspects of food emulsions can befound in the literature (Paquin, 1999; Schultz et al., 2004; Perrier-Cornet et al., 2005; Lambrich and Schubert, 2005). The specific role and choice of food ingredients in the sta-bilization (and controlled destabilization) of emulsions will be discussed in the section“Release Triggers for Emulsions.”

The most important rule of food emulsion production is that the emulsion should ini-tially be stable. Emulsions are kinetically rather than thermodynamically stable two-phasesystems and, ultimately, both oil and water phases will separate. To understand how to opti-mize emulsion stability, it is necessary to understand the mechanisms by which emulsionsare destabilized. There are four main mechanisms whereby emulsion phase separation maybe accelerated. These are summarized accordingly.

Creaming/Sedimentation

For most food emulsions, the oil phase has a lower density than the aqueous phase and canthus separate out due to gravity. For o/w-type emulsions, creaming specifically refers to themotion of emulsion droplets under gravity to form a concentrated creamy layer at the top ofthe emulsion. Whether or not there is a change in droplet size in this highly concentratedregion depends on the stability of the droplets against coalescence. Creaming of poorly sta-bilized emulsions may result in complete breaking of the emulsion layer, resulting in phaseseparation. For well-stabilized emulsion droplets even an extensively creamed layer can befully re-dispersed. For w/o-type emulsions, the movement of droplets under gravity isreferred to as sedimentation.

The rate of creaming for an individual noninteractive spherical droplet, �s, can bedefined for highly dilute emulsions through Stokes’ Law:

where g is the acceleration due to gravity, r is the radius of the droplet, � is the density ofthe dispersed phase, �0 is the density of the continuous phase and �0 is the Newtonian shearviscosity of the continuous phase. From this equation it can be seen that the rate of cream-ing can be reduced by:

• Reducing droplet size—homogenization of milk typically reduces droplet size fromca. 4 μm in diameter to <1 μm in diameter resulting in a considerable improvement increaming stability of the milk.

�S

��2

9

20

0

r gρ ρη

( )

Emulsions as Delivery Systems in Foods 43

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 43

Page 53: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

• Increasing the viscosity of the continuous phase.• Density matching the continuous and dispersed phases.

From a quantitative perspective, the Stokes’ equation applies only to highly dilute, nonin-teracting emulsions and is therefore not applicable to concentrated, polydisperse or aggre-gated emulsion systems. However, the parameters governing creaming rate, as defined bythe Stokes’ equation, can of course be applied nonquantitatively to improve creaming stability.

The simplest way to measure creaming is by direct observation. However, this relies onthe formation of a well-defined layer between cream-rich and cream-depleted regions.Where there is a more diffuse concentration gradient, it may be impossible to directly oraccurately detect when creaming is occurring. There are several commercially availabletechniques that can be used to provide a more accurate analysis of emulsion creaming.These include ultrasound, magnetic resonance imaging and conductivity. The advantage ofthese techniques is that they are noninvasive and can provide a reasonable approximation ofchanges to dispersed phase volume across a sample (Dickinson et al., 1994; Dickinson,1996).

Flocculation

Flocculation is a general term referring to the various mechanisms for aggregation orassociation of droplets whereby the interfacial layer of the droplets remains intact(Dickinson, 1998). Generally, for dilute emulsions, flocculation results in enhanced cream-ing, since the flocs structures rise more quickly under gravity relative to individualdroplets. However, in more concentrated emulsions, it is possible that flocculation can leadto the formation of a percolating network, which can be controlled to manipulate both sta-bility and rheological properties of the emulsion (Chanamai et al., 2000; Dalgleish, 2006).Flocculation of emulsion droplets takes place when the pair-interaction free energybecomes appreciably negative at a particular separation. This can be achieved througha number of different mechanisms of varying interaction potential. These are brieflysummarized.

Depletion flocculationThis is encountered in emulsion systems containing suitable depletants such as non-interacting polysaccharides (e.g. xanthan) or micellar species (e.g. SDS or sodiumcaseinate) and recently reported to take place in bimodal emulsions, where there is anadequate difference between distributions (Dickinson and Golding, 1997a; Moschakiset al., 2005; Djerdjev et al., 2006). Depletion takes place due to the entropic exclusionof the depletant as droplets approach each other. Due to the exclusion of the depletant,an osmotic pressure gradient forms between droplets resulting in a net attraction therebyleading to flocculation. The strength of the interaction potential is proportional to the sizesof both droplets and depletant as well as the relative concentration of the depletant.Depletion flocculation is termed weak or reversible flocculation, since flocs can be brokenup by simple shaking. However, even though the interaction is weak, depletion can resultin extensive aggregation of droplets leading to rapid creaming and separation in the caseof dilute emulsions, or greatly increased viscosity in the case of more concentratedemulsions.

44 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 44

Page 54: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Bridging flocculationBridging flocculation is a general term to describe the association of emulsion dropletsthrough interfacial interaction. The strength of the interaction depends greatly on the spe-cific nature of the bridging mechanism, which are discussed below.

Electrostatic bridging: Droplets stabilized by charged interfacial layers can be bridgedthrough the addition of a species containing counterions. This can be achieved through theaddition of an appropriate salt or of a charged biopolymer with the capability of multiplebinding sites. Salt bridging requires a minimum divalent ion to enable bridges to formbetween droplets. An example is the calcium-ion bridging of a casein-stabilized emulsionat neutral pH. In this case, the divalent calcium is able to bind to negatively charged aminoacids on the peptide chain of the protein adsorbed onto the interface. This can result ininterfacial cross-linking between droplets or cross-linking between droplets and free pro-tein in the continuous phase (Dickinson and Golding, 1998; Ye and Singh, 2001).

Alternatively, a biopolymer with opposite charge to the interfacial layer can also behaveas an electrostatic bridging link between droplets (Bratskaya et al., 2006). Taking anadsorbed casein interface as an example, at neutral pH, where the net charge at the interfaceis negative, and a cationic biopolymer will be required to form electrostatic cross-links.(Note: There are very few cationic biopolymers available within the foods industry.Examples include acid/high pI gelatin and chitosan.) However, at pH below the pI of theprotein, where the net surface charge is cationic, electrostatic cross-links can be formedusing an anionic biopolymer (such as pectin or carrageenan) in the aqueous phase. Electro-static bridging flocculation provides a stronger inter-droplet interaction relative to deple-tion, although high shear can be used to break up aggregated structures.

Incomplete surface coverage bridging: This takes place for emulsions stabilized byhigher molecular weight emulsifiers, such as proteins. For emulsion droplets to possessgood stability, an adequate surface coverage of the droplet surface is required. If no suffi-cient emulsifying material is present to provide good coverage, it is possible that biopoly-mer molecules will become adsorbed to more than one droplet surface, leading to bridgingflocculation (Dickinson and Golding, 1997b). A similar effect can be observed when emul-sions are homogenized at very high pressures. Under these circumstances, protein canbecome adsorbed to more than one droplet surface during homogenization, leading to theformation of an inter-connected protein network.

Covalent bridging: Droplets stabilized by biopolymer interfaces can also be cross-linkedthrough covalent bridging mechanisms (Dickinson, 1997; Romoscanu and Mezzenga,2005). There are several routes by which covalent cross-linking can be induced. Forexample, covalent cross-linking can be thermally induced for protein-stabilized emulsionswhereby the adsorbed layer contains accessible disulphide peptides capable of intermolecu-lar cross-linking. The use of high static pressure treatments can also act in a similar mannerto induce disulphide cross-linking for appropriate globular protein-stabilized emulsions(Galazka et al., 2000).

Enzymatic covalent cross-linking: This is another mechanism by which droplet bridgingmay take place. One example is the use of the microbial enzyme transglutaminase, whichcatalyses covalent cross-linking between lysine and glutamine amino acids. Consequently,adsorbed protein interfacial layers which display good availability and accessibility ofthese two amino acid residues (such as the casein proteins) may become covalentlycross-linked. For covalent cross-linking to take place, droplets must be in close proximity.Consequently, for dilute emulsions cross-linking may not occur (or interaction with

Emulsions as Delivery Systems in Foods 45

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 45

Page 55: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

cross-linking species present in the continuous phase may take place). At higher dispersedphase volumes, where droplet–droplet separation is sufficiently low for cross-linkingto happen, bridging mechanisms tend to result in the formation of a droplet network(Dickinson and Yamamoto, 1996). In this respect, bridging flocculation is used moreas a structuring pathway to create emulsion-based gel systems rather than as a cause ofinstability, per se.

Coalescence

Coalescence of emulsion droplets occurs when there is film rupture at the interfaces ofadjoining droplets. This leads to the irreversible conjoining of the droplets into a singlelarger entity. Catastrophic coalescence, such as may take place in a homogenized emulsionin the absence of emulsifier or for poorly stabilized emulsions, can rapidly lead to breakingof the emulsion and partitioning into separate (free) oil and aqueous phases. Where morelimited coalescence takes place (such as through controlled shear), the emulsion can thenbecome unstable due to gravity forces enhancing the creaming rate of the larger droplets.

For film rupture to take place there must be sufficient film thinning between droplets.Film thinning depends on the relative hydrodynamics within the film, and is dependent on anumber of factors, such as the rheological properties of the continuous phase, the concen-tration of the dispersed phase and the effective stabilization of the droplets and their abilityto maintain appreciable inter-droplet distance.

Interfacial rupture depends on the mechanical properties of the film and the influence ofshear and temperature. Emulsion droplets can be stabilized through adsorption of a vis-coelastic and/or charged interface, which is often the case with adsorbed protein, or alter-native biopolymer layers. Increasing interfacial viscoelasticity can provide effective dropletstability against coalescence, even at high applied shear forces. Crystalline interfaces canalso provide surface rigidity and effective stabilization against coalescence (Dickinsonet al., 1988; Simovic and Prestidge, 2004; Giermanska-Kahn et al., 2005; Tcholakova et al.,2005).

Emulsions stabilized with small molecule surfactants do not possess viscoelastic orcharge-stabilized (in the case of nonionic emulsifiers) interfaces. For such systems, stabilityagainst coalescence is provided through the Marangoni effect, in which surfactantstreaming at the point of film thinning leads to an osmotic pressure differential between thefilm and the surrounding solvent. Consequently, water is drawn into the film gap, therebypreventing further thinning from taking place.

An intermediate state of coalescence, termed partial coalescence, is often utilized in thefood industry to deliberately induce fat structuring in a number of products, such as icecream, whipping cream and spreads. In such cases, the interface of the emulsion isdesigned through formulation, such that it will rupture under appropriate conditions,thereby resulting in coalescence of the emulsion. However, for emulsion systems where thedispersed phase contains a prerequisite level of solid fat, the presence of the solid fat canprevent formation of a single larger entity. Instead, fat droplets form agglomerated struc-tures, sharing a common interface, but in which a degree of the original droplet integrity ismaintained. The formation of such agglomerated structures is used to improve the qualityof aerated products such as whipped cream and ice cream, by improving the stability offoam structures and reducing drainage (Vanapalli and Coupland, 2001; Coupland, 2002;Hotrum et al., 2005).

46 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 46

Page 56: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Ostwald Ripening

Ostwald ripening is essentially the growth of larger emulsion droplets at the expense ofsmaller ones. It may happen in polydisperse emulsions, and takes place due to the fact thatthe solubility of the oil phase increases with decreasing droplet size. For small droplets thisincrease in solubility may allow the oil to dissolve and diffuse through the aqueous phase,condensing into larger droplets where solubility is lower. It is difficult to see how the mech-anism could be exploited for encapsulation and controlled release; however, for the inter-ested reader additional information can be found in the literature (Taylor, 1998; Hoanget al., 2004; Meinders and van Vliet, 2004; Mun and McClements, 2006).

This section on “Emulsion stabilization” is aimed to highlight the various mechanismsby which emulsions can be destabilized. These are represented in Figure 3.1. Before goingon to explore potential routes for formulating food emulsions, it is important to remind our-selves that use of emulsions for controlled delivery and release depends primarily on twothings: first, the necessity to stabilize the emulsion against separation prior to delivery; and,secondly, to control destabilization of the emulsion using one or more of the above instabil-ity mechanisms to achieve the required release and delivery under the appropriate physio-logical conditions.

Formulation Design for Food Emulsions

One of the more obvious constraints on the formulation of food emulsions is that all com-ponents should be edible, food-grade and approved for use by international legislation.

Emulsions as Delivery Systems in Foods 47

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Two approachingcolloidal particles

Interaction between particlesvan der Waals, electrostatic, steric, depletion

Repulsive forces dominateStable

Attractive forces dominateFlocculation

Coagulation

Hard spheres

Partial coalescence

Solid/liquid

Coalescence

Emulsions

Attractive forces highly dominate

Figure 3.1. Schematic representation of mechanisms for droplet stabilisation and instability.

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 47

Page 57: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Whilst this places certain restrictions on the scope of formulation, it has led to considerablecreativity with regard to the application of these ingredients for food structuring. The for-mulation of a food emulsion needs to be considered from three perspectives: the design ofthe aqueous phase, the nature of the dispersed phase and the composition of the interface.The choice of formulation also depends on whether the emulsion is intended to be o/w orw/o (or on occasion w/o/w or o/w/o). For an o/w-type emulsion, options for formulationcan be summarized as below.

Design of the Aqueous Phase

There are two important aspects to consider when formulating the continuous phase of ano/w food emulsion. The first is microbiological stability, which can be tailored according tothe anticipated shelf life of the product. This has less relevance to encapsulation and con-trolled release, although it is important to note that for products designed with a long closedshelf life in mind (>3 months), there should be no change in the emulsion structure, toensure that the release properties remain consistent over the lifetime of the product. Micro-biological stability can be improved through both thermal treatment (pasteurization/sterili-zation) and aseptic packaging. In addition, emulsions prepared at low pH (<4), high sugaror alcohol content, with low water activity, or containing preservatives will all haveimproved microbiological stability.

The second point of consideration is that manipulation of the continuous phase can havea significant impact on the rheological properties of the emulsion. This can have importantconsequences on delivery, both in terms of oral response behavior of the emulsion and forthe subsequent behavior in the GI tract. Continuous phase properties are very much depen-dent on product type. For example, beverage emulsions are generally low in viscosity, andwhile it is necessary that the dispersed phase remains stable, there is little requirement forthe addition of thickeners or stabilizers. In contrast, a reduced fat mayonnaise requires theaddition of aqueous thickeners to the continuous phase in order to compensate for reducedviscosity through the removal of fat.

Whilst the addition of biopolymers, such as starch and guar gum, can be used to controlaqueous phase rheology independently of the dispersed phase, biopolymer addition canalso result in additional rheological manipulation through structuring of the fat phase. Thismay take place through such effects as depletion flocculation, bridging flocculation orphase separation, and are discussed in the section “Emulsion Stabilization.” A summary ofa number of biopolymers available for aqueous phase structuring is given in Table 3.2. Thislist is not comprehensive and ignores any synergistic effects that may exist for combina-tions of biopolymers.

As can be seen from Table 3.2, a number of functional effects can be achieved throughaqueous phase structuring. Depending on the nature of structuring, and the potentialfor aqueous phase biopolymers to interact with the dispersed phase, it is possible to controlthe release properties of the emulsion, both in terms of emulsion structure failure in mouth(textural response and taste/flavor release) and in terms of emulsion separation undergastric conditions. Examples of how continuous phase behavior can influence releaseproperties will be given in more detail in the sections “Delivery of Water-Soluble Food Actives via Emulsions” and “Delivery of Hydrophobic Food Actives via O/WEmulsions.”

48 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 48

Page 58: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Emulsions as Delivery Systems in Foods 49

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Table 3.2. Examples of aqueous phase structuring ingredients

Interaction with Biopolymer type Biopolymer name Effect dispersed phase

Polysaccharide Guar Thickener, stabilizer NonePectin (low/high methoxy) Stabilizer, thickener, gelling Electrostatic

bridginga anddepletion

Xanthan Thickener, yield stress DepletionCarrageenan Thickener, stabilizer, gelling Bridginga

Locust bean gum Thickener, gelling (cryogelation) NoneStarch Starch (modified) Thickening, gelation (heat set, cold set) NoneProtein Casein (SMP) Thickening, gelation (acid) Bridginga

Casein (SMP, caseinate) Gelation (enzyme) Bridginga, depletion

Whey Gelation (thermal, covalent) Bridginga

Soy Gelation (thermal, covalent) Bridginga

Gelatin Gelation (thermal, coil-helix transition) Bridginga

Egg (ovalbumin) Gelation (thermal, covalent) Bridginga

Choice of Lipid Phase

Many fats and oils are available for use in food emulsions. Whilst the main role of fat in afood product is sensoric, from a product design perspective the use of a particular fat/oil orblending of different lipids can have a significant impact on food emulsion properties.A summary outlining examples of fat types and their composition is given in Table 3.3.

Generally a food fat is any triglyceride composition which is solid at room temperature,whilst oil is liquid. For many food emulsions, where the emulsion has been designed to pro-vide structure, solid or hardened fats are often used. Typical examples include ice creamand whipping cream, where the solid fat droplets are able to stabilize air bubbles in theproduct, as well as generating structure through partial coalescence. This form of fat struc-turing improves product quality through improved stabilization of the air phase and aslower rate of melt. Saturated fats are also utilized extensively in the spreads and margarineindustry, where required crystallization is an essential aspect of continuous phase structur-ing and stabilization for w/o-type emulsions. For both these examples it would be impo-ssible to use unsaturated fats to achieve this degree of structuring. In contrast, a productsuch as mayonnaise uses liquid oils, since it would be impossible to achieve the correctsensory properties with a hardened fat.

In addition to structuring, another functional use of the oil phase for an o/w emulsion isas a carrier for lipophilic actives such as flavors, nutrients (e.g. oil-soluble vitamins) andcolors. Design of the emulsion system is important for how these actives are stabilized inproduct and how they are released on consumption. The technical challenges associatedwith the delivery of lipophilic actives will be discussed in more detail, with examples in thesections “Delivery of Hydrophobic Food Actives via O/W Emulsions” and “Delivery ofDietary Fats as O/W Emulsions and Their Protection against Oxidation”. It should also benoted that w/o and water-in-oil-in-water (w/o/w) type emulsions can be utilized in a similarmanner for the controlled delivery and release of water-soluble actives such as vitamins andenzymes.

a Under appropriate interfacial conditions.

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 49

Page 59: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Finally, it should be noted that the choice of fat/oil is also important from a nutritionalperspective. Generally, highly saturated fats can have a negative impact on health. Oils withhigh polyunsaturated triglyceride content are considered preferential in terms of dietaryintake, and that oil-containing ω-3 fatty acids are often quoted within nutritional literatureas imparting specific health benefits when consumed regularly. However, replacing all satu-rated fats with polyunsaturated oils is not a trivial exercise. As has been mentioned, thedegree of saturation can have an impact on the structuring behavior of the emulsion. Inaddition, highly unsaturated oils are prone to oxidation, which can lead to the developmentof adverse odors and flavors. Inhibition of oxidative processes within emulsion systemsremains one of the key challenges in the move towards a healthier diet, and will be dis-cussed in more detail in the section “Delivery of Dietary Fats as O/W Emulsions and TheirProtection against Oxidation.”

Interfacial Formulation and Design

Arguably the most important aspect of emulsion preparation is the composition of the inter-face. Of course, the number of ingredients available for formulation of food emulsions islimited to what can be considered edible and food-grade, and may vary between countriesdepending on legislation. However, food emulsifiers can broadly be characterized into twocategories: low molecular weight species based on fatty acids and high molecular biopoly-mers with amphipathic properties. Examples of food emulsifiers are given in Table 3.4.

For all food products, choice of emulsifier is a key to achieving the desired textural andsensory properties. The design of the interface will also play a controlling role in the

50 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Table 3.3. Examples of common food fats and oils and their melting points (MP) and fatcomposition (in %). Saturated fatty acid chains of a given chain length are given the suffix Cxx:0,whilst unsaturated fatty acid chains are given the suffice Cxx:1/2/3 depending on the degree ofunsaturation

Other Other (satu- (unsatu-

Fat or oil MP (°C) C12:0 C14:0 C16:0 C18:0 C20:0 rated) C16:1 C18:1 C18:2 C18:3 rated)

Butter fat 32.2 2.5 11.1 29 9.2 2.4 4.8 4.6 26.7 3.6 – 8.5Lard oil 30.5 – 1.3 28.3 11.9 – – – 74–76 – – –Castor oil −18 0.6 0.6 0.6 0.6 – – – 7.4 3.1 – 87Cocoa 34.1 – – 24.4 35.4 – – – 38.1 2.1 – –

butterCoconut oil 25.1 45.4 18 10.5 2.3 0.4 14.6 0.4 7.5 Trace – –Corn oil −20 – 1.4 10.2 3 – – 1.5 49.6 34.3 – –Cottonseed −1 – 1.4 23.4 1.1 1.3 – 2 22.9 47.8 – –Olive oil −6 – Trace 6.9 2.3 0.1 – – 84.4 4.6 – –Palm oil 35 – 1.4 40.1 5.5 – – – 42.7 10.3 – –Palm kernel 24.1 46.9 14.1 8.8 1.3 – 9.7 – 18.5 0.7 – –Peanut 3 1.92 2 1.4 1.9 2.4 – – 17.8 – 17.5 –Rapeseed −10 – – 1 – – – – 32 15 1 50Safflower – 1.1 1.1 1.2 1.1 1.2 – – 18.6 70.1 3.4 –Sesame oil −6 – – 9.1 4.3 0.8 – – 45.4 40.4 – –Soybean −16 0.2 0.1 9.8 2.4 0.9 – 0.4 28.9 50.7 – –Sunflower −17 – – 5.6 2.2 0.9 – – 25.1 66.2 – –

seed

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 50

Page 60: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Emulsions as Delivery Systems in Foods 51

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Table 3.4. Summary of some food emulsifiers available for stabilization of oil-in-water andwater-in-oil food emulsions, including application

Name Type Functionality Application

High-molecular- Skimmed milk Dairy protein O/W emulsion stability Ice cream, yoghurtweight powder at neutral pH, emulsion biopolymer structuring (bridging)

at acid pHSodium caseinate Dairy protein O/W emulsion Cream liqueurs,

stability at neutral pH, meat emulsionsemulsion structuring (bridging) at acid pH

Whey powder Dairy protein O/W emulsion stability Neutral pH at neutral pH, emulsion- beverages, infant based gels under formulationsthermal processing

Gelatin Bovine/Porcine/ Encapsulation, emulsion- Nutritional Fish Protein based gels supplements

Gum arabic Polysaccharide O/W emulsion stability Acid beveragesgalactan protein at low pH

OSA starch Modified starch O/W emulsion stabilizer Acid beverages

Low-molecular- Lecithin Phospholipid O/W and w/o stability Spreads, weight depending on mayonnaiseemulsifier modification

Monoglycerides Glycerol-esterified W/O emulsion Spreads, ice cream,fatty acids and stabilization and o/w cream liqueurstriglycerides destabilization

Sodium stearoyl Lactic acid- O/W emulsion Salad dressings, lactylate esterified stabilization creamers

monoglycerideDatem Diacetyl tartaric O/W emulsion Powder mixes,

acid-esterified stabilization gravymonoglyceride

Tween Polysorbate- O/W emulsion Desserts, ice cream, esterified stabilization, dressingstriglyceride destabilization

PGPR Polyglycerol- W/O emulsion Spreads and other esterified stability oil continuous monoglycerides products

stabilization and breakdown of emulsion structure. High molecular weight amphipathicbiopolymers, in particular milk proteins, are very effective at stabilizing o/w-type emulsions.Here, adsorption of casein and whey protein onto the oil–water interface provides effectivesteric and electrostatic stabilization against coalescence and flocculation. Most dairy-basedfood emulsions, such as milk, dairy and nondairy creams, ice cream and yoghurt are based onmilk protein stabilization of the emulsion. The advantages of using proteins are the nutritionaland clean-label aspects associated with proteins. In addition, caseins (essential milk proteinfraction) are able to maintain functionality as a result of thermal processing. The disadvantageof proteins is that the peptide interface can be sensitive to changes in pH and ionic composi-tion. For example, acidification of the emulsion towards the isoelectric point of the protein oraddition of calcium ions can result in flocculation of the emulsion. In some instances thisresults in undesirable separation of the emulsion. However, aggregation of dairy emulsions

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 51

Page 61: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

through pH and calcium is also deliberately used for specific product design, such as in themanufacture of yoghurt or cheese.

Where pH sensitivity is an issue for emulsion stability, alternative emulsifiers are avail-able. In particular, for beverage emulsions, where the dispersed phase is dilute and requireseffective stabilization, gum arabic or modified starch can be used. In both cases, the struc-ture of the biopolymer allows for effective interfacial stabilization, even at low pH.

Low molecular weight emulsifiers do not have the clean-label perception of proteins.However, they are often able to bring specific functional benefits not achieved by proteinsalone. Most low molecular weight emulsifiers are based on triglycerides or fatty acids thathave undergone an esterification process to produce amphiphilic molecules, whereby thefatty acid chain provides the hydrophobic tail group and the esterified species (such as gly-cerol) provides the more hydrophilic headgroup. Depending on the chain length, degree of(un)saturation of the triglyceride, and the type of headgroup, the amphiphilic properties ofemulsifiers can be controlled to a certain degree.

This classification of emulsifiers according to their amphiphilic properties is known asthe hydrophilic–lipophilic balance (HLB). HLB is a numerical scale for emulsifiers. Emulsi-fiers which are more hydrophobic in nature have low HLB values. Increasing hydrophilicityincreases HLB score. For example, polyglycerol polyricinoleate (PGPR), which is ahydrophobic emulsifier used for the stabilization of w/o-type emulsions, has an HLB valueof 2. In contrast, polysorbate emulsifiers, which are commonly used for the stabilization ofo/w emulsions, have HLB values of around 16.

The choice of an emulsifier for a product is very much dependent on the required func-tionality imparted by the emulsifier. Emulsifiers are mainly used for emulsion stabilizationas well as its controlled destabilization in food products. Other uses include functionalapplications such as fat crystal modification, aeration, wetting, and the formation of stablemesophases. The other main application of emulsifiers, often but not always in combinationwith emulsion systems, is as delivery vehicles for active components. The use of interfacialdesign for both biopolymer- and emulsifier-stabilized interfaces will be discussed in moredetail in subsequent sections.

Release Triggers for Emulsions

To be able to design food emulsions for encapsulation and delivery, it is necessary to iden-tify where and how delivery will take place.

For delivery of actives that are intended to impart a sensory benefit of the product, deliverytakes place on consumption. Behavior of the active in-mouth is therefore of considerableimportance. Mouth is a remarkable receptor for sensory perception. Our preference ordislike for certain foods and drinks is driven by three key attributes:

1. Taste and aroma perception.2. Texture perception: design of food microstructure is responsible for the initial textural

perception of a foodstuff.3. Our like or dislike of a food may derive not only from the initial texture of the product

but also from the way in which it breaks down in the mouth. Consequently, whendesigning food structure, it is necessary to take into account the manner in which theproduct microstructure breaks down on eating. This is often an issue for fat replacementin foods. It may be possible to replace the fat in a product with aqueous fillers such as

52 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 52

Page 62: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

starch, such that the initial rheological properties of the two products are the same.However, the breakdown of the two different structures during mastication may result inthe two products being texturally perceived entirely differently. The breakdown ofmicrostructure in the mouth will also influence both taste perception and flavor percep-tion. The breakdown of food structures in the mouth is dependent on the applied shearforces during mastication, dilution of the structure with saliva and the fact that in-mouthtemperature is generally higher than ambient temperatures ex-vivo. In addition, amylasepresent in saliva will rapidly start the digestion of starch in the mouth, which mayimpact on texture. Physical changes in microstructure on eating, such as melting pointtransitions (fat, gelatin), phase inversions (w/o to o/w emulsions), break-up of aggre-gated emulsion structures, can not only influence textural response, but also be used tocontrol the release of actives, such as tastants or flavors, in the mouth.

Alternatively, delivery of an active for nutritional benefit will need to take place after con-sumption. Consequently, the release properties of the emulsion/active under gastric condi-tions will be paramount. In the GI tract, emulsions face a low pH (around 2.0) and pepsin inthe stomach and lipases and bile salts in the beginning of the small intestine. Lipophilicactives may then be absorbed by the human body in the intestine via dietary micelles. Fur-thermore, since nutritional actives often possess negative taste attributes (e.g., bitterness orastringency), there may also be the additional requirement that release of the active in-mouth should be minimized as much as possible and that the active should be encapsulatedin such a way that it is undetectable on consumption.

Clearly, in designing emulsions with particular delivery and release properties, the phys-iological conditions of the delivery site will need to be taken into consideration. For in-mouth and gastric conditions the delivery environment will be quite different.

Delivery of Water-Soluble Food Actives via Emulsions

Water-in-Oil Emulsions for Controlling Water-Soluble Actives

Most spreads containing 40% or more fat are fat-continuous products with the aqueousphase being dispersed as w/o emulsions. One of the key requirements for a good spread isphase inversion during breakdown in the mouth (see also “Release Triggers for Emul-sions”), thereby releasing, for example, salt. Margarines that do not do that are perceived aswaxy and with very little taste.

In principle, stable w/o emulsions that do not invert in the mouth should be able to trap,for example, bitter hydrophilic molecules (such as proteins) in the water phase withoutbeing perceived.

Effect of O/W Emulsions on Taste Release and Perception

In foods containing both water and lipids, the lipid phase may also take part in the sensoryperception by influencing the distribution of food components in the oil and aqueousphases or at the oil—water interface (Yamamoto and Nakabayashi, 1999). Similarly, fatscan modify the perception of other sapid food components by influencing their partitioningbetween the food matrix, saliva, taste receptors (for taste), or headspace (for aroma) withinthe oral cavity (Forss, 1973). Traditionally the majority of flavor studies in emulsion-based

Emulsions as Delivery Systems in Foods 53

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 53

Page 63: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

foods have focused on aroma release and little has been reported on the effect of fat on tasteperception. Shamil and co-workers (1991) found that the reduction of fat in cheese led to anincrease in bitterness and astringency and a reduction in saltiness. Increased bitterness andastringency were assumed to arise from the hydrophobic character of these ingredients andthe resultant increase in their concentration in the aqueous phase when the fat level wasreduced. Conversely, the decrease in salt intensity when the fat level was reduced wasproposed to be due to the reduced concentration of salt in the aqueous phase. Wendin(Wendin, et al., 1999) also reported that a decrease in fat content led to a decrease in sourtaste due to the reduced concentration of the acid in the aqueous phase. Earlier studies haveshown a correlation between oil mouth coatings and taste perception (Lynch et al., 1993).Valentova and Pokorny (1998) also found that the intensity of sweet, bitter, and astringentcompounds was reduced by the prior consumption of oil, but acidic and salty tastes werenot affected. Lynch and co-workers (1993) found that all the taste modalities were affected,albeit by a small amount, and that coconut oil had a more suppressive effect than sunfloweroil and proposed that fat mouth coatings physically interfere with tastant access to the tastereceptors.

Yamamoto and Nakabayashi (1999) concluded from their work on the effects of increas-ing oil-phase volume on salt perception in o/w emulsions that taste intensity will be influ-enced by a combination of an increased concentration of salt in the aqueous phase and asuppressed contact of salt with taste receptors.

Overall, however, the results cited in the literature on the effects of oil on tastants havebeen inconclusive but suggest that oils and emulsions can influence taste perception. Thetaste-modifying effects of fats may be mediated through:

1. Changes in the partitioning of flavor (taste) compounds between the food (e.g., changesin the aqueous phase concentration as the fat level is altered), saliva and the taste recep-tor cell membranes and pores.

2. Physical interference with diffusion processes affecting access or binding of taste mole-cules to receptors through a mouth coating action by the oil.

3. Changes in the rate of regeneration of interfacial surfaces required for the release ofsapid compounds into the surrounding media.

In order to test these ideas, Malone et al. (2003) investigated the relationship between tasteperception and microstructure and specifically the effect of fat content in o/w emulsions onsalt perception. Their studies involved a sensory analysis of salt perception on isoviscouso/w emulsions with fat contents ranging from 0 to 95%. The results showed that as the fatcontent was increased for products kept at constant salt concentration, the perceived salti-ness increased due to the associated increase in the salt concentration in the aqueous phase(Figure 3.2).

However, when the salt concentration on aqueous phase was kept constant the saltinessdecreased with increasing fat content (Figure 3.2), suggesting that perceived saltiness isnonlinearly dependent on a combination of salt concentration and aqueous phase volume.Based on their results a psychophysical model was developed that related taste intensity tosalt concentration and the phase volume of fat:

I k C mpn n� � ��[ ] ( ) [ exp( )]φ φ

aq aq1

54 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 54

Page 64: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

where I is the taste intensity, k is a constant, n is a power-law number, Cp is the concentra-tion of tastant on product, �aq is the aqueous phase volume and m is the amount of tastant inthe aqueous phase.

Indeed, through this insight, water-in-oil-in-water duplex emulsions have been designedto control taste perception by effectively controlling the extent to which the aqueous phaseis able to interact with the oral surfaces (further described in the section “Control of TasteUsing W/O/W Emulsions”).

Double Emulsions for Controlling Water-Soluble Actives

Double emulsions (also known as “duplex emulsions”) have potentially promising applica-tions in the food industry, primarily for sustained release of active components via a con-trolled transport mechanism (Matsumoto and Kang, 1989; Garti, 1996, 1997a, 1997b,1998; Garti and Aserin, 1996a, 1996b; Garti and Bisperink, 1998; Garti and Benichou,2001), for taste masking (Malone et al., 2003) and for encapsulating sensitive ingredientssuch as flavors and active components (Kim and Lee, 1999; Yoshida et al., 1999; Edris andBergnståhl, 2001; Benichou et al., 2004; Onuki et al., 2004; Shima et al., 2005) in both thewater and oil phases (van der Graaf et al., 2005). Two main types of double emulsion can bedistinguished: w/o/w emulsions, in which a w/o emulsion is dispersed as droplets in anaqueous phase, and oil-in-water-in-oil (o/w/o) emulsions, in which an o/w emulsion is dis-persed in an oil phase. This latter emulsion is less common primarily due to having fewhydrophobic emulsifiers that are food-grade to stabilize water droplets in continuous oilphase (Pays et al., 2002).

The advantage of double emulsion technology is in their double compartment structure,in which they act as reservoirs, encapsulating a range of active components. Actives thatare water-soluble but insoluble in the oil phase can be entrapped in w/o/w emulsions, andactives that are oil-soluble but insoluble in the water phase can be entrapped in o/w/o emul-sions. Actives that are both soluble in oil and water cannot be “encapsulated.” The encapsu-lated actives may be released subsequently under variable conditions. This will be the maintopic covered in this section, although the production and the issues of instability will alsobe addressed briefly.

Emulsions as Delivery Systems in Foods 55

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Figure 3.2. Effect of oil phase volume in o/w emulsions on salty taste perception forconstant salt concentration on product or on aqueous phase.

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 55

Page 65: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Production of W/O/W Emulsion

Membrane emulsification is one of the key technologies for producing stable double emul-sions due to the prevailing mild shear stress conditions (van der Graaf et al., 2005). Usuallydouble emulsions are prepared in a two-step emulsification process using two surfactants:a hydrophobic surfactant to stabilize the internal w/o emulsion and a hydrophilic one forthe external interface of the oil drops with the aqueous environment. In conventional emul-sification processes high shear stresses are needed to reduce the internal droplet size anddroplet size distribution of the emulsion, but this also causes internal streaming in thedroplets, which causes more collisions and therefore coalescence of the internal waterphase with that of the outer aqueous phase (Muguet et al., 1999; Klahn et al., 2002).

Membrane emulsification (Nakashima et al., 1991) is a relatively new method for pro-ducing double emulsions but has added benefits in its low energy consumption, better con-trol of the droplet size and distribution and mildness of processing. In general two methodsare used: cross-flow membrane emulsification and pre-mix membrane emulsification(Suzuki et al., 1998). In the latter case a pre-mix is forced through a membrane, which fur-ther breaks up the droplets. In the cross-flow method the to-be-dispersed phase is pressedthrough a microporous membrane while the continuous phase flows along the membranesurface. Once this primary emulsion is produced, the second emulsification step to producethe o/w/o emulsion can also be carried out using membrane emulsification, which helpsprevent rupture of the double emulsion and inversion into a single o/w emulsion. Mem-brane emulsification for the production of single emulsions has been reviewed by a numberof authors (Joscelyne and Tragardh, 2000; Charcosset et al., 2004) and for microstructuredemulsion systems by Lambrich and Vladisavljevic (2004). Gijsbertsen-Abrahamse et al.(2004) reviewed the current status of membrane emulsification and van der Graaf et al.(2005) have recently reviewed membrane emulsification techniques for the production ofw/o/w emulsions and also highlighted new developments in nanoengineered and micro-engineered membranes, such as microsieves to improve the flux of emulsions through themembrane (van Rijn, 2004).

Instability of W/O/W Emulsions

The main problem with double emulsions though is that they tend to be unstable since theycontain more interface and therefore are more thermodynamically unstable than singleemulsions. Much literature has been published on this subject since around the mid-1980s(Garti et al., 1994; Garti and Aserin, 1996a, 1996b). Florence and Whitehill (1981)described four possible mechanisms for instability of w/o/w emulsions:

1. Coalescence (see “Coalescence”) of the internal aqueous droplets2. Coalescence of the oil droplets3. Rupture of the oil film separating the internal and external aqueous phases4. Migration of water (including the water-soluble ingredients) between the internal and

external water phases through the oil layer.

This migration of the water phase discussed above could be via reverse micellar transport; bydiffusion across the oil layer, where it is at its thinnest; and transport via hydrated surfactant(for further details see “Transport and Release Mechanisms of Water-Soluble Components”).

56 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 56

Page 66: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

An enormous amount of formulations for double emulsions is known in the literaturewith various types of oil, different fractions of phases and different sorts of surfactants invarying concentrations (van der Graaf et al., 2005).

Several methods have been documented in the literature for improving stabilization andslowing solute release (Davis et al., 1985). The methods can be categorized into three mainareas:

1. Stabilization of the internal interface of the inner emulsion2. Selection of appropriate oil phase and components that could structure it and3. Stabilization of the outer interface.

A range of low molecular weight emulsifiers, oils, co-solvents and co-emulsifiers havebeen tried (Benichou et al., 2004). Materials that have been investigated also includebiopolymers, synthetic graft and comb co-polymers and polymerized emulsifiers thatimpart steric or mechanical stabilization. Monomeric nonionic emulsifiers (hydrophobicand hydrophilic) have some limitations in terms of emulsion stability and therefore othermolecules such as naturally occurring macromolecular materials (e.g., gums and proteins)are of interest (Garti, 1997a, 1997b). Macromolecules such as proteins offer excellent sta-bilization effects through electrostatic repulsion in combination with steric contributions.Proteins, polysaccharides and their blends are natural surface-active biopolymers. Underappropriate conditions these may complex through electrostatic interaction, and the newlyformed macromolecular amphiphile can anchor onto oil–water interfaces more strongly(Benichou et al., 2002). Complexation between proteins and polysaccharides at the emul-sion droplet surface can improve steric stabilization. Droplet size can be smaller if thepolysaccharide is present during homogenization, and so rate of creaming may be reducedso long as there is no bridging flocculation (Benichou et al., 2002). Combinations ofsurfactants in the outer water phase have also shown a beneficial effect on stability andthese multianchoring flexible amphiphilic surfactants are effective emulsifiers since theycan improve the steric stabilization by forming thick multilayered coating on the emulsiondroplets and also by providing protection against coalescence by making them resistant toshear (Garti, 1998).

Transport and Release Mechanisms of Water-Soluble Components

Two major release mechanisms involved in the release of a water-soluble active such ascommon salt (NaCl), which is entrapped in the aqueous core of w/o/w double emulsions,have been suggested (Pays et al., 2002). The first one describes the rupture of the thin liquidfilm separating the internal droplets and the double emulsion surfaces. The second is whenthe entrapped species diffuses or permeates through the oil membrane. These two mecha-nisms can be controlled by varying the composition and amounts of surfactant in the sys-tem. In w/o/w emulsions stabilized with sodium dodecyl sulfate below its critical micelleconcentration (cmc), the release of salt occurs by diffusion across the oil membrane. Watertransport also needs to be taken into account and a number of mechanisms have beensuggested (Wen and Papadopoulos, 2000, 2001): through the surfactant thin lamellae, byreverse micelles and via hydrated surfactant. Ficheux et al. (1998) identified two types ofthermodynamic instabilities that allow the (uncontrolled) release of entrapped actives. Thefirst mechanism involves the coalescence of the inner and outer aqueous phase, through the

Emulsions as Delivery Systems in Foods 57

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 57

Page 67: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

rupture of the thin nonaqueous film between them, so that inversion to a single emulsionoccurs. This mechanism is best suited for the release of water-soluble components, and therelease kinetics can be controlled by the hydrophilic surfactant concentration. Dependingon the concentration of surfactant, double emulsions may be destabilized with a time scaleranging from a few months to a few minutes. The second mechanism involves the coales-cence between the smaller droplets inside the oil phase leading to an increase in the internaldroplet size and a reduction in number. Interestingly, even if the osmotic pressure is bal-anced between the internal and external phase, electrolytes can still be transported outthrough the reverse micellar mechanism, which is controlled by the viscosity of the oilphase and the nature of the oil membrane (Garti and Bisperink, 1998). The release ratefrom double emulsions in general tends to follow first-order kinetics (Garti et al., 1994;Sela et al., 1995; Jage-Lezer et al., 1997) with the release rate being controlled primarily byan increase in the diffusion of the active through the oil phase by the selection of appropri-ate secondary hydrophilic emulsifiers.

Delivery of hydrophilic actives in the GI tract has also been studied with w/o/w emul-sions (see “General Applications of W/O/W Emulsions”). The oil layer is supposed to pro-tect the active from inactivation by the digestive process in the GI tract (see also “ReleaseTriggers for Emulsions”). However, osmotic pressures in the GI tract are often not con-trolled and this might limit the use of duplex emulsions.

General Applications of W/O/W Emulsions

The potential applications of double emulsion technology are enormous, with primaryfocus being in the food, cosmetics, medical and pharmaceutical industries. Potential appli-cations have been demonstrated in improved biological availability (Elson et al., 1970;Brodin et al., 1978), delivery of drugs (Pandit et al., 1987), and adsorption of toxic com-pounds (Lata et al., 1987). Garti (1997b) reviewed the progress made up until then withrespect to food applications of double emulsions. Since then many potential applicationsfor double emulsions have been well documented and some have been patented (Thill-Francis, 1993; Gaonkar, 1994; Takahashi et al., 1994). In most cases double emulsions areaimed for slow and sustained release or controlled release of active matter from an internalreservoir into the continuous phase. Double emulsions have also been used to improvedissolution and solubilization of insoluble materials. Application of double emulsions inthe protection of sensitive and active molecules from oxidation (Gallarate et al., 1999;Kim and Lee, 1999; Yoshida et al., 1999; Edris and Bergnståhl, 2001) has also been investi-gated, and double emulsions used to mask acid taste was described by Malone et al. (2003)(see below).

Control of Taste Using W/O/W Emulsions

It has already been mentioned in “Effect of O/W Emulsions on Taste Release and Percep-tion” that the results by Malone et al. (2003) on the effect of oil content on taste perceptionindicated that the perceived intensity of a tastant is dependent on the oil-phase volume �oilso for any given system the taste intensity can be manipulated by making w1/o/w2 duplexemulsions to control the apparent �oil. Upon consumption the external w2 phase will beperceived but the internal w1 phase will be shielded from the taste receptors during the timescales of eating.

58 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 58

Page 68: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

In order to test this hypothesis, a series of w1/o/w2 double emulsions were made havingfixed oil contents of 30% w/w and internal w1 phase volumes ranging from 0 to 50% w/w.The concentration of the solutes, in particular the citric acid, was at the same levels in boththe w1 and w2 phases in order to remove the osmotic and concentration gradients that nor-mally destabilize the emulsions during storage and to maintain the acid functionality inboth phases. Their results showed that as more aqueous phase was incorporated into the oilphase the titratable acidity decreased (Malone et al., 2003). The acid in the internal w1phase was not released from the duplex emulsion over the duration of the experiment(which had typical timescales in minutes). Sensory evaluation showed that the perceivedacidity decreased, clearly demonstrating the dependence of taste perception on the volumeof aqueous phase coming into direct contact with the mouth (i.e., w2).

These results demonstrated that it is possible to manipulate the taste intensity by con-trolling the external w2 phase volume that contacts the taste receptors without resorting totraditional encapsulation approaches. This approach is different in that it provides a meansof controlling active release whilst allowing for the thermodynamic stable distribution ofthe active between the constituent phases of the product. In emulsion-based foods, duplexemulsions also provide the benefit of controlled taste whilst remaining within acceptabletextural limits for the product concerned.

Delivery of Hydrophobic Food Actives via O/W Emulsions

Lipophilic Health Ingredients in O/W Emulsions

Oil-in-water emulsions can be utilized to provide consumers with lipophilic health ingredi-ents, such as dietary fat, antioxidants (such as the arytenoids -carotene and lycopene),vitamins (e.g. vitamin E), or sterols. They dissolve in the oil of the o/w emulsion, whichmay stabilize them during storage and increase their bioavailability. The use of dietaryfat and how to protect these against oxidation are discussed in more detail in “Delivery ofDietary Fats as O/W Emulsions and Their Protection against Oxidation.” This latersection may also illustrate routes to stabilize lipophilic health ingredients against oxidation(if necessary).

Lycopene may have various health benefits, such as antioxidation, induction of cell com-munication and growth control, and lower risk of cancer (Ribeiro et al., 2003 and refer-ences therein). Ribeiro et al. (2003) found that the chemical stability of lycopene wasparticularly high in orange juice (pH 3.7), in contrast to skimmed milk (pH 6.6) or emul-sions in water (pH 5.7). The authors stated that this result might be due to the antioxidant inthe orange juice (ascorbic acid, or vitamin C), presence of iron in the milk, or influence ofpH on oxidation. The use of -tocopherol or of nitrogen strongly inhibited the oxidation inall the three different food systems studied. The use of different emulsifiers (Tween 20,Lamegin ZE609, or lecithin) had little influence on the stability of lycopene. The bioavail-ability of lycopene dissolved in oil is high, in contrast to its crystal form (Ribeiro et al.,2003 and references therein). Therefore, o/w lycopene emulsions could be a base todevelop functional foods.

Plant sterols or phytosterols can reduce serum cholesterol by inhibiting intestinal cho-lesterol absorption (Trautwein et al., 2003). Different mechanisms—such as competingwith cholesterol for absorption into the dietary micelles (the vehicles for the transportof lipophilic compounds in the intestine) or for cholesterol transporters, co-crystallization

Emulsions as Delivery Systems in Foods 59

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 59

Page 69: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

with cholesterol to form insoluble crystals and interference with the hydrolysis process bylipases and cholesterol esterases—are believed to play a role in this process. About 3 g/dayof phytosterols are needed to have a significant effect on cholesterol lowering in the serum.The phytosterols are insoluble in water and poorly soluble in oil. Engel and Schubert(2005) produced a high loading of phytosterols in emulsions by using triglyceride and theemulsifiers lecithin or monoglyceride as crystallization inhibitors. Esterifying phytosterolswith fatty acids increases their solubility in oil dramatically and this allows easy incorpora-tion of plant sterol fatty acid esters into food products (both in fat-based products, such asmargarine and spreads, and in o/w emulsion-based products, such as yoghurt or milk(Trautwein et al., 2003; Noakes et al., 2005 and references therein).

Aroma Release from O/W Emulsions

The concentration of flavor (aroma) reaching the olfactory receptors will be influenced bythe structure and composition of the food and by the physiological environment and masti-cation behavior that impacts on the rate of aroma released from the foodstuff (Harrison andHills, 1997a, 1997b; Harrison, 1998; Malone et al., 2000, 2003). Specifically, the flavor-release kinetics may depend on the flavor concentration, the microstructure and tempera-ture of the food, the occurrence of reversible/irreversible binding, structure breakdownduring mastication, mixing with saliva, and most importantly (for lipophilic flavors) theconcentration of the fat (Delahunty and Piggott, 1995; Overbosch et al., 1991; Maloneet al., 2000, 2003; Le Guen and Vreeker, 2003).

Fat is recognised as playing a critical role in influencing many flavor attributes such asflavor quality, flavor release, flavor stability and the masking of off-flavor (McGorrin andLeland, 1994). Previous studies have demonstrated that when the fat content of a product isreduced the release of lipophilic flavors is altered resulting in changes to the intensity andrelease profiles, which alter the overall flavor balance and acceptability of the product(Overbosch et al., 1991; McGorrin and Leland, 1994; Malone et al., 2000; Doyen et al.,2001). These changes are particularly apparent when the fat content is reduced to below5%, where both the intensity and temporal profile are significantly altered (Malone et al.,2000). These differences arise primarily due to the reduction in absorption of the lipophilicflavors in accordance with simple partition theory.

Among all food constituents, lipids (and thus also emulsions) affect aroma release mostnotably, as they not only lower the aroma partial pressure or the air-product partition coeffi-cient of most of the flavor compounds but also change the time scale of release with vary-ing concentrations (de Roos, 1997; Guichard, 2002; Rabe et al., 2003; McClements, 2005).The higher the lipid content and the lipophilicity of the components (i.e., oil–water parti-tion coefficient, Pow), the stronger the decrease in aroma release (with the exception ofhighly polar compounds possessing log Pow values �0, such as vanillin or acetic acid, seeLeland, 1997). However, this effect is stronger under static conditions than under in vivoand artificial throat conditions (Weel et al., 2004b). Generally, increasing fat concentrationsresult in decreasing flavor release. The affinity of aroma to a lipid phase depends on itschemical composition, degree of saturation, chain length and sequence of fatty acids incor-porated in a triacylglycerol. Hydrophobicity of the aroma compound is the determiningfactor for the distribution of aroma in the oil and water phases. Using homologues series ofhydrocarbons, aldehydes, ketones and alcohols, Jo and Ahn (1999) found that aromarelease decreased linearly with the fat content of the emulsion. The effect was less

60 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 60

Page 70: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

pronounced for ketones and greater for hydrocarbons, which can be explained by their di-fferent solubilities in oil. However, hydrogen bonds between aroma and lipids are an addi-tional parameter (as has been demonstrated for, e.g., 1-octen-3-ol and linoleic acid byLe Thanh et al., 1998). Small changes in oil content can have significant effect on the aromapartial pressure or the air-product partition coefficient of lipophilic aroma compounds, incontrast to hydrophilic ones (Guichard, 2002). As a consequence, the overall aroma percep-tion is changed often contributing to an imbalance to the nature of the overall flavor.

Studies of the influence of the nature and surface area of an oil–water interface onthe volatility of aroma in biphasic systems show that there is no general rule for under-standing the effects of interfaces on the aroma release (Druaux and Voilley, 1997, and refer-ences therein). If the volatile compounds (such as dimethylsulfide) accumulate at theinterface, the aroma concentration in the headspace of an emulsified system is significantlyreduced compared to a two-phase, nonemulsified system. In the case of sunflower-oil/wateremulsions that were stabilized with a sugar ester as the emulsifier, diacetyl displayed ahigher volatility in w/o emulsions than in o/w emulsions. The presence of proteins at theoil–water interface of emulsions may induce retention for the compounds with high bind-ing constants (Guichard, 2002). For example, ethyl hexanoate was significantly betterreleased from emulsions containing -lactalbumin (protein with lower affinity for aromacompounds) than from those with -lactoglobulin. The presence of -lactoglobulin at theoil–water interface increases the resistance to the transfer of hydrophobic aroma com-pounds from oil to water and thus induces a decrease in aroma release (and perception). Onthe other hand, emulsifier concentrations above the cmc of Tween-80 did not influence therelease (Rabe et al., 2003). The results with ionic emulsifiers might be different, due toionic interactions.

In general, droplet size of emulsions normally found in foods (5–100 �m) will not affectthe aroma release of a food product (Rabe et al., 2003; Weel et al., 2004b; McClements,2005), although divergent results have been reported in the literature about the effect ondroplet size on aroma release in vivo (Guichard, 2002; Rabe et al., 2003; Weel et al.,2004a). Smaller droplets may lead to faster mass transfer due to increased interfacial areaand shorter diffusion distance through the oil droplets. However, the exchange of aromasbetween the two phases is generally assumed to be extremely rapid and it is the water–airinterface that is the rate-limiting step for soft solids containing emulsion droplets in thenormal size range (1–100 �m). Moreover, recent data showed that only small amounts ofvolatiles are dynamically released from water within 30 s. Therefore, the re-equilibrationprocess between the lipid and the aqueous phase should not be rate-limiting for the initialrelease process, as the concentration gradients to be adjusted are very flat.

Surveys of food preferences among consumers indicate that the most importantattributes of foods are aroma (flavor), appearance, and taste with flavor being the primarybasis upon which food is selected and reselected. One aspect of flavor delivery that has hadlittle attention is the control of the temporal flavor release profile (i.e., shape of the flavordelivery curve). An increasingly important market requirement is that a new flavor for aparticular product should make a specific perceptual impression, for example a powerfulinitial impact or a novel sensation during eating. Since the rate and duration at which a fla-vor is delivered influence the perception, there is a rational that by controlling the temporalrelease profile the perception of that flavor can be manipulated. Factors that are importantfor flavor delivery in the mouth include the composition and microstructure of the product,dilution and mixing with saliva, changes in temperature, flavor concentration and the

Emulsions as Delivery Systems in Foods 61

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 61

Page 71: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

occurrence of reversible/irreversible flavor binding (Overbosch et al., 1991; de Roos andWolswinkel, 1994; Taylor, 1996; see also “Release Triggers for Emulsions”). Furthermore,mass transfer of flavor in the mouth is affected by the gas and saliva flow rates, the degreeof agitation and the temperature—all affected by the food structure and composition(Overbosch et al., 1991; Harrison and Hills, 1997a; Harrison, 1998; van Ruth et al., 2000).

Aroma release in the mouth is a nonequilibrium process. Consequently, the maximumheadspace concentration of a static system will hardly be reached in the mouth. A number ofmathematical and mechanistic physicochemical models have been developed that describeflavor release from solid and semi-solid food matrices during eating (de Roos and Wol-swinkel, 1994; Hills and Harrison, 1995; Harrison and Hills, 1996; Harrison et al., 1998;Wright et al., 2003, Wright and Hills, 2003). These theories have been based on the fact thatduring mastication, the kinetics of flavor release is primarily dependent on the generation ofnew surfaces (Harrison, 2000) and that the rate-limiting step is the mass transfer of flavorvolatiles across the solid–saliva and saliva–gas interfaces for solid (Hills and Harrison, 1995;Harrison and Hills, 1996; Harrison et al., 1998; Wright and Hills, 2003; Wright et al., 2003)and semi-solid foods (Harrison et al., 1997b; Bakker et al., 1998). Much of the impetus forthis work has been to model the link between the perception of flavor intensity and the foodscomposition, microstructure and breakdown during eating (Wright and Hills, 2003; Wrightet al., 2003). Two important factors for controlling flavor delivery have been identified. Theseare (i) the rate-limiting step for soft-solid materials is the mass transfer of volatiles across thesolid–liquid and liquid–gas interfaces (Harrison et al., 1998) and (ii) these rates are propor-tional to the mass transfer coefficient and the interfacial surface area (Hills and Harrison,1995). This implies that by controlling the interfacial surface area by creating new surfaces(through particle breakdown) the temporal flavor profile could be altered.

Harrison and Hills developed a mathematical model for the release of flavor volatilesfrom solid foods based on the two-film stagnant film theory (Hills and Harrison, 1995;Harrison and Hills, 1997a; Harrison et. al., 1998). This took into account the saliva flow,decrease in particle size, increase in new surfaces (surface area) and mixing with salivaduring mastication. The expression they used based on Euler’s approximation was

where M is the total mass of volatile diffusing across the interface, hD is the mass transfercoefficient of a volatile, Asf is the surface area of the saliva–food interface, Ksf is thesaliva–food partition coefficient, and cf and cs denote the concentration of flavor in the foodand saliva, respectively.

In principle, the same physicochemical model could be used to model the flavor releasedfrom particles that break down, provided that the model takes into account the differentrelease mechanisms that are involved, each of which is rate determining at a different time.

Based on considerations of mass transport between the food surface, the surroundingsaliva layer and the gas phase of the breath, these treatments have drawn attention to theimportance of describing the changing food surface area and saliva content during thecourse of mastication.

One of the biggest challenges and as yet not really solvable with current model con-straints is developing models that can predict the flavor-release behavior when the kinetics

M h A cc t

Kt= −

⎣⎢⎢

⎦⎥⎥D sf f

s

sf

( )

62 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 62

Page 72: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

and mechanism of food structure breakdown are changing and sometimes in dramaticways. Currently, most models are empirical and generally based on a number of assump-tions that have not been fully tested. For example, these models have so far needed toassume idealized spherical shapes for the food particles, which can be related to surfacearea measurements. The goal would be to mathematically predict the effect of food struc-ture and composition, material behavior and breakdown during eating on the time-intensityflavor release profile, based on mathematical models requiring fewer approximations.

The increasing demand for low calorie foods has engendered much interest in the devel-opment of low-fat and sugar-free foods with the required consumer satisfaction. The difficultyin reduced-fat food lies in the multiple functions that fat plays, as it influences all aspects offood perception including appearance, texture, mouthfeel, and flavor. Fat is a source of fla-vor and also influences flavor character, flavor release, off-flavor, and taste perception.Lowering of fat levels is known to reduce the binding of lipophilic flavors to the foodmatrix thereby influencing the flavor balance (Overbosch et al., 1991; Shamil et al., 1991;Hatchwell, 1996; Malone et al., 2000; Doyen et al., 2001). Reduction in fat levels not onlyaffects the intensity of the flavor perception but also influences the temporal profile. Inhigh-fat products the initial impact of the flavor is gradual providing a well-balanced flavorprofile whereas, in fat-free foods the flavor tends to be intense and transient manifestingitself as an “unbalanced” flavor with an “uneven” release profile.

Structured Emulsions in Hydrogels for Controlled Release of Aromas

The microstructure and composition of food affects flavor release since aroma compoundsmay be adsorbed and absorbed by food components (Kinsella, 1990; Bakker, 1995) orinfluenced by the material and rheological properties of the food, which affect its break-down during eating (Baines and Morris, 1987; Malkki et al., 1990; Guinard and Marty,1995; Wilson and Brown, 1997). The relative importance of each of these mechanismsvaries with the properties of the aroma compounds and the physicochemical properties ofthe food. Binding phenomena, however, generally involve interactions that are specific tothe flavor and composition of the food and are not a versatile or practical means of control-ling flavor release in low-fat foods. Likewise, the rheological and material properties havebeen demonstrated to influence flavor release but large textural changes are required inorder to have a noticeable effect; in addition, the scale of these changes is beyond the tex-tural tolerances that would be acceptable for many products.

From the consideration of the partitioning of the flavor between the oil and aqueousphases at equilibrium, it can be shown that a considerable proportion of lipophilic flavorsare present in the oil phase, even at relatively low oil phase volumes (0.5–5%). Hence, onestrategy to control the release of lipophilic flavors would be to inhibit the rate of mass trans-fer of flavor molecules between the oil phase and the continuous aqueous phase. To do thisa new approach was taken in our laboratory in which oil was encapsulated within gelparticles (Malone and Appelqvist, 2003).

In standard o/w emulsions, the rate of inter-phase transport of small solutes from the oilto the water phase occurs on a millisecond timescale (Wedzicha and Couet, 1995). There-fore, during eating, it would appear that the aqueous phase of low-fat o/w emulsions is rap-idly stripped of its flavor, creating a strong driving force for rapid mass transfer of aromafrom the oil phase to the aqueous phase. This rapid replenishment of the aqueous phase isthe principal reason for the increase in maximum flavor intensity (FImax) and the rate of

Emulsions as Delivery Systems in Foods 63

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 63

Page 73: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

release into the headspace. One of the challenges in developing low-fat products is thedesign of microstructures that will reduce the rate of release of lipophilic aroma. Maloneand co-workers (Malone et al., 2003) have demonstrated that one approach to reducingaroma release in low-fat systems is via incorporating the oil droplets into biopolymer gelparticles, termed microstructured emulsions. In such microstructures the oil droplets areenveloped in a gel phase, creating a static diffusion layer around the oil droplets. Thisincreases the path-length through which the aroma must diffuse before coming under theinfluence of the advective conditions existing within the bulk of the product during eating.The result of these structures was to inhibit the rate at which the lipophilic aromas replenishthe continuous phase and reduce the rate of aroma release into the headspace.

The principle behind encapsulating oil within a gelled particle was to increase the effec-tive path-length for diffusion into the aqueous continuous environment to reduce the rate oflipophilic flavor release. A model for describing the flavor release from gel particles wasdeveloped by Lian et al. (2004). The model relates release rates to the composition(oil/water phase volume) and particle size and takes into account the resistance to masstransfer in both the particle and the bulk liquid phase. It should be noted that this approachdiffers from conventional encapsulation techniques in that it is the oil and not the aromathat is encapsulated. The aroma is allowed to reach thermodynamic equilibrium in theproduct, and it is the concentration of lipophilic aroma in the oil phase of the gel particlesthat forms the basis on which the controlled release is achieved. This is an important dis-tinction because this approach does not attempt to resist the thermodynamic distribution ofaroma between the oil and water phases.

Release can be triggered by matrix melting in the mouth (e.g. gelatin), enzymatic break-down (amylase hydrolysis of starch) and compression-induced fracture (chewing). Aromarelease can take place upon breaking the initial equilibrium of aroma between the oil andwater phases. As aroma is stripped from the aqueous phase the system attempts to re-establish the o/w equilibrium by diffusion of aroma from the oil but the additional diffu-sional pathway formed by the surrounding gel increases the half-life (t1/2), which, fordiffusion into an infinite sink, can be approximated by

t1/2

where r is the radius of the particle, Kow is the oil–water partition coefficient of the flavorcompounds, �o is the phase volume of oil in the particle, and D is the diffusion coefficientof the aroma compounds in the particle. From this equation, the key parameters that influ-ence the rate of aroma release are the radius r, the oil content of the particle, and Kow of thearoma species.

It has been possible to make microstructured emulsions from a range of biopolymer gelsincluding Ca-alginate, gellan, gelatin, gelatin/gum arabic, agar, and starch. This providesthe option to design particles that demonstrate controlled breakdown under physiologicalconditions during eating (Malone et al., 2003). Hence, by careful selection and design ofthe gel particle it should be possible to manipulate the shape of the aroma-release profile bythe particle breakdown pattern.

Other mechanisms that might contribute to changes in flavor-release profiles includebrittle/elastic fracture involving syneresis of gel particles. For highly structured foods such

r KD

2

2

0 693 1=

+( ). ow oφ�

64 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 64

Page 74: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

as crackers, flavor release can be triggered by plasticization of the matrix by saliva.Changes in pH, ionic strength, and salt type (Kuhn and Foegeding, 1991) within the mouthenvironment could also be used to trigger physical disruption of particles to release flavormolecules in a controlled manner.

There are a large number of factors such as biopolymer type, concentration, salt and soon that could be adjusted to design structured particles that break down and release underphysiological conditions. By doing this, it is possible to manipulate the flavor release pat-terns, so that novel flavor profiles are obtained that may be appealing to the consumer. Theability to predict the effect of varying composition and structure on flavor-release profileswill be of great value to the food industry, particularly when trying to design new sensa-tions. Combining microstructure design and mathematical modeling, it would be possibleto formulate foods for a desired flavor profile, taking into account both the composition offood and individual or group differences in mastication behavior.

Delivery of Dietary Fats as O/W Emulsions and Their Protectionagainst Oxidation

Dietary fats fall in three main groups: saturated, mono-unsaturated and polyunsaturated. Oliveoil is the best-known example of dietary oil that contains predominantly mono-unsaturated fattyacids. Polyunsaturated fatty acids are further divided into two subgroups called ω-6 and ω-3fatty acids. Here, the term ω-6 means that the first double bond in the carbon backbone of thefatty acid occurs in the sixth carbon–carbon bond (counted from the terminal carbon atomopposite the acid group). Similarly, ω-3 fatty acids have their first double bond in the thirdcarbon–carbon bond. Examples of ω-3 fatty acids are -linolenic acid (ALA), eicosapentaenoicacid (EPA), and docosahexaenoic acid (DHA). ALA is an essential fatty acid (i.e., it is not syn-thesized in the human body and must be obtained from food) and can be found in vegetablesources such as the seeds of flax or wall nut; EPA and DHA are abundantly present in fish oilsand other marine oils and are assumed to play an important role in the prevention of cardiovas-cular diseases and several other disorders (Nestel, 2000). Furthermore, DHA has been proposedto play an important role in neural and visual developments of infants (Conner, 2000). Becauseof their beneficial health properties, ω-3 fatty acids have great potential as functional food ingre-dient (Jacobsen, 2004). Unfortunately, the use of ω-3 fatty acids in foods is limited due to theirsusceptibility to oxidation. Oxidation is a major cause of quality deterioration in foods contain-ing significant amounts of ω-3 fatty acids and gives rise to changes in, for example, flavor (ran-cidity) and nutritional value. Considerable effort has been made to elucidate the mechanisms oflipid oxidation in bulk oils and emulsions.

Lipid oxidation can occur via three mechanisms: autoxidation, photo-oxidation, orenzyme action. Emulsified foods usually do not contain enzymes that catalyze oxidation andtherefore the latter mechanism is probably less relevant. Photo-oxidation occurs in the pres-ence of light (visible or ultraviolet) and photosensitizing pigments (such as chlorophyll orriboflavin). Photo-oxidation can be a major cause of quality deterioration and is efficientlycontrolled by storing foods in the dark. Autoxidation is perhaps the most common mecha-nism in foods. It proceeds via a complex series of free-radical reactions with initiation, pro-pagation, and termination steps (Karel, 1992). Transition metal ions (such as iron or copper)are known to be important catalysts for autoxidation. In theory, these metals are capable ofdirectly breaking down unsaturated lipids (RH) into radicals. This reaction, however, is notbelieved to be important in initiating lipid oxidation. More likely, oxidation is initiated by

Emulsions as Delivery Systems in Foods 65

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 65

Page 75: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

metal-catalyzed decomposition of hydroperoxides (ROOH) (lipid hydroperoxides are foundin small quantities in all food oils). For the initiation reaction, it is necessary that metal ionsand hydroperoxides are in close proximity. In emulsion systems, hydroperoxides tend toaccumulate at the surface of the oil droplets because of their relatively polar nature(McClements and Decker, 2000) and therefore are able to interact with metal ions or otherpro-oxidants in the aqueous phase. This interaction leads to decomposition of the hydroper-oxides and formation of highly reactive peroxyl (ROO•) or alkoxyl (RO•) radicals. Oncethese free radicals have been formed at the droplet surface they will interact with polyunsat-urated lipids in their vicinity. This triggers a complex series of oxidation reactions (thereader is referred to Karel, 1992, for more details). Flavor changes typical for oxidized oilproducts (rancidity, fishy off-flavors, etc.) result from the formation of secondary oxidationproducts (such as aldehydes, ketones, alkanes, etc.) (Belitz et al., 2001).

Various approaches can be taken to retard lipid autoxidation in food products. Removal ofoxygen by packing under vacuum or nitrogen can be effective in certain cases. The use ofhigh quality oils with low levels of hydroperoxides and ingredients with low levels of metal-ion contamination is also important. A well-known strategy of controlling lipid oxidation isby the addition of antioxidants (Frankel, 1996; McClements and Decker, 2000). Antioxi-dants are classified depending on their mechanism of action as either primary or secondaryantioxidants. Primary antioxidants are compounds that react with free radicals and which arecapable of interrupting lipid oxidation chain reactions. Tocopherols and plant polyphenolsare important examples of natural primary antioxidants. Secondary antioxidants can retardlipid oxidation through a number of different mechanisms such as metal chelation, oxygenscavenging, or by replenishing hydrogen to primary antioxidants. Examples of metal chela-tors are ethylenediaminetetraacetic acid (EDTA) and citric acid; various food proteins andpolysaccharides are also known for their excellent metal-chelating properties.

Recently, the impact of microstructure and interfacial characteristics on the oxidativestability of emulsions has been highlighted (see McClements and Decker, 2000, for anexcellent review). Various studies have shown that controlling the type and concentration ofemulsifiers at the droplet interface can influence the rate of oxidation. One of the physico-chemical factors that appear to be important is the electrical charge of the interfacial layer.An electrically charged surface will attract oppositely charged ions (counter ions) in thesurrounding aqueous phase. A negatively charged surface (anionic emulsifier) will thusattract positively charged metal ions and bring them into close proximity of the lipid sub-strate. This is expected to reduce the oxidative stability of the emulsion. On the other hand,a positively charged emulsifier will repel metal ions from the surface and may thus helpto stabilize the emulsion against oxidation. The importance of surface charge was demo-nstrated by Mei et al. (1997) for corn oil-in-water emulsions produced using three differentemulsifiers. Oxidation rates (in the initial stage of the process) were found to be largest foran emulsion stabilized by sodium dodecyl sulfate (a negatively charged emulsifier); emul-sions stabilized by Brij 35 (uncharged) or dodecyltrimethylammonium bromide (positivelycharged) appeared to oxidize at a lower rate (at pH 6.5). The authors suggested that the useof positively charged emulsifiers could be an effective means of retarding iron-catalyzedlipid oxidation. As low molecular weight cationic emulsifiers are not commonly used infoods, it was suggested to use protein stabilized emulsions at a pH below the isoelectricpoint (pI) of the protein. Under these conditions proteins can form a positively chargedinterfacial membrane around the oil droplet that will repel metal ions. Most food proteinshave isoelectric points in the pH range 4.5–5.5 and thus positively charged emulsion

66 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 66

Page 76: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

droplets can only be prepared at relatively low pH (i.e., lower than usually desirable in foodemulsions). Gelatin (produced by acid hydrolysis of collagen) is an exception as this pro-tein has a relatively high isoelectric point (pI � 7–8). Acid-treated gelatin can thus be usedto prepare o/w emulsions with positively charged droplets over a wider range of pH valuesthan is possible with other food proteins (Surh et al., 2006).

The ability of positively charged proteins to retard lipid oxidation was studied by Huet al. (2003a). The authors studied the oxidative stability of salmon oil-in-water emulsionsstabilized by different whey proteins (viz. -lactalbumin, -lactoglobulin, sweet whey, andwhey protein isolate). Oxidative stability was greatest at pH values below the isoelectricpoint of the proteins, which was explained from electrostatic repulsion of metal ions awayfrom the positively charged emulsion droplet surface. The authors noted, however, that theability of whey proteins to alter oxidation rates is not solely due to charge effects, becausethe positive charge (� potential) of the emulsion droplets (at pH 3.0) decreased in the order-lactoglobulin > -lactalbumin > whey protein isolate > sweet whey, whereas the oxida-tive stability decreased in the order -lactoglobulin � sweet whey > -lactalbumin � wheyprotein isolate. This suggests that other factors also influence the ability of adsorbed pro-teins to retard lipid oxidation. In a subsequent study the authors compared oxidation ratesof corn oil-in-water emulsions stabilized by casein, whey protein isolate, and soy proteinisolate (Hu et al., 2003b). The oxidative stability (at pH 3.0) decreased in the order casein >whey protein isolates � soy protein isolate. It was concluded that the magnitude of the posi-tive droplet charge again is not the only factor responsible for differences in oxidative stab-ility and that other membrane properties probably also play a role. One of the factors thatmight be involved is the thickness of the interfacial membrane: a thick layer at the emulsiondroplet interface is assumed to hinder interactions (i.e., acts as a physical barrier) betweenwater-soluble pro-oxidants and lipids inside the emulsion droplets (Silvestre et al., 2000).Caseins form a relatively thick layer on the emulsion droplet interface (as compared to,e.g., whey protein isolate), which might contribute to the lower oxidation rate observed incasein-stabilized emulsions.

Another factor of importance is the metal–ion chelation properties of proteins. Villiereet al. (2005) compared the oxidative stability of sunflower oil-in-water emulsions stabilizedby bovine serum albumin and sodium caseinate. At pH 6.5, emulsions stabilized by sodiumcaseinate were found to oxidize faster than emulsions stabilized by bovine serum albumin.The faster oxidation was attributed to the better chelating properties of sodium caseinate(as compared to bovine serum albumin) and to electrostatic interactions that favor position-ing of metal ions at the interface. The authors suggest that proteins with good metal chela-tion properties, such as sodium caseinate, should not be used as emulsifiers in systemscontaining oxidation sensitive lipids, but preferably should be added to the aqueous phaseas a natural antioxidant after the emulsification process. This does not hold for emulsions inwhich metal ions are deactivated and kept away from the interface by the addition ofEDTA; in the presence of EDTA, emulsions stabilized by sodium caseinate appeared to bemore stable than emulsions stabilized by bovine serum albumin, which was attributed tofree-radical-scavenging properties of sodium caseinate.

In protein-stabilized emulsions, usually only a fraction of the proteins adsorbs at the oildroplet interface, whereas the remaining proteins are located in the continuous water phase.If the proteins in the water phase are able to chelate metals ions, they can remove the ionsaway from the oil droplet and inhibit oxidation. The impact of various continuous phaseproteins (viz., soy protein isolate, casein and whey protein isolate) on the oxidative stability

Emulsions as Delivery Systems in Foods 67

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 67

Page 77: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

of menhaden oil-in-water emulsions was studied by Faraji et al. (2004). In their experi-ments, continuous phase proteins were removed in a number of “washing” steps and theoxidative stability of washed emulsions was compared to those of nonwashed emulsions.Unwashed emulsions (at pH 7.0) were more oxidatively stable than washed emulsions indi-cating that continuous phase proteins are indeed antioxidative and could be used as aneffective means of protecting ω-3 fatty acids. Under the conditions used, soy protein isolatewas found to have the greatest antioxidant activity of all proteins tested, that is, larger thancasein, which was found to have the largest chelation capacity. The authors suggested thatin case of soy, antioxidant activity most likely results from a combination of metal-ionchelation and free-radical scavenging. The latter may be due to the presence of specificamino acids with antioxidant activity (such as free sulfhydryl groups) or antioxidants (e.g.,isoflavones) associated with the soy protein.

Klinkesorn et al. (2005) studied the effect of multilayer membranes on the oxidative stab-ility of tuna oil-in-water emulsions. Multilayer membranes were produced by sequentialdeposition of oppositely charged emulsifiers. First, an emulsion was made by dispersing oilin a solution of an anionic emulsifier (lecithin) and then this emulsion was mixed with asolution of a positively charged polysaccharide (chitosan). This “layer-by-layer depositiontechnique” could be used to produce cationic and relatively thick emulsion droplet inter-faces. The oxidative stability of emulsion droplets coated by a lecithin-chitosan multilayerwas found to be higher than that of emulsion droplets coated with lecithin only. Theimproved stability is likely due to the cationic nature of the droplets that causes repulsion ofthe prooxidative metals and possibly also from a thicker interfacial region that reducesinteractions between lipids and water-soluble prooxidants. According to the authors, pro-duction of emulsion droplets with a multilayer lecithin-chitosan coating might be an excel-lent technology for protecting labile oils.

The previous examples have highlighted the importance of prooxidant location. How-ever, the location of chain-breaking antioxidants can also play a critical role in stabilizingemulsions (Frankel, 1996; McClements and Decker, 2000; Chaiyasit et al., 2005). Chain-breaking antioxidants are expected to be most effective at retarding lipid oxidation whenthey are located in the oil–water interfacial region, where oxidation reactions are initiated.Hydrophilic antioxidants, in general, are less effective than lipophilic antioxidants in o/wemulsions. This is because a significant portion of the hydrophilic antioxidant will partitioninto the aqueous phase, where it is considered to be inactive (Schwarz et al., 2000). Theeffectiveness of chain-breaking antioxidants in general increases as their polaritydecreases, because they are then more likely to be localized in the lipid phase or near thelipid surface (Huang et al., 1996a, 1996b, 1997).

The importance of the electrical charge of chain-breaking antioxidants (relative to thecharge of emulsion droplets) was demonstrated by Mei et al. (1997). The authors measuredoxidation rates for salmon oil-in-water emulsions stabilized by anionic surfactants (sodiumdodecyl sulfate) or uncharged surfactants (Brij 35) containing negatively charged,uncharged, or positively charged phenolic antioxidants. In emulsions stabilized by sodiumdodecyl sulfate (at pH 7), the negatively charged antioxidants were found to be less effect-ive than the positively or uncharged antioxidants, which suggests that the negativelycharged antioxidants are electrostatically repelled from the surface of the emulsiondroplets. In emulsions stabilized by Brij 35, the uncharged phenolic antioxidants werefound to be most effective, which was thought to result from the low solubility ofuncharged phenolic antioxidants (as compared to the charged phenolics) and a tendency to

68 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 68

Page 78: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

accumulate at the oil–water interface. Physical properties, such as polarity and partitioningbetween different phases, are thus important criteria in selecting a proper antioxidant sys-tem. However, as mentioned by Huang et al. (1997), other criteria such as relative oxidativestability and hydrogen-donating ability in different phases should also be considered in theselection of antioxidants.

The literature on oxidation in real food products (e.g., fish-oil enriched mayonnaise,margarine, or milk drinks) is still relatively limited (Jacobsen, 2004). Most studies so farhave concentrated on model emulsion systems. The knowledge gained from model studiesis expected to lead to new product opportunities. In particular, the possibility of designinginterfacial properties (“interfacial engineering”) will enable food scientists to engineerfoods with improved oxidative stability.

Future Trends

Current efforts are focusing on naturalness, convenience, and perfection. The use of “natu-ral emulsions” and the production of monodispersed emulsions are discussed here. The useof nanoemulsions will be discussed in Chapter 2 in this book.

Nature-Made Emulsions

Nature-made emulsions can be used when purified or reconstituted. The idea here is toentrap active components in these pre-formed emulsions. Potentially all plant, animal, andmicrobial cells can be used and as with all release devices selection will be dependent onthe ability of the system to deliver the required release characteristics against a particularapplication. Three types of preformed capsule systems will be briefly discussed here, oil orlipid bodies, yeast cells, and plant cells. Their use may enhance the “natural” image of afood product, in addition to other functional advantages.

Oil or Lipid Bodies

Seed oil bodies (Figure 3.3) are lipid storage organelles of 0.5–2 �m in diameter and com-prise a triacylglycerol matrix shielded by a monolayer of phospholipids and proteins. Theseproteins include abundant structural proteins, oleosins (a structural protein), and at least twominor proteins caleosin (a calcium-binding protein) and steroleosin (an NADP-dependentsterol-binding protein) (Chen et al., 2004). Native oil bodies—modified and reconstructed—can be a useful structure for a range of applications especially as a carrier for hydrophobicmolecules.

The layer of oleosin coating imparts stability to the oil body by protecting the phospho-lipid monolayer both from attack by the phosphorlipases present in the cell and by givingthe oil body a negatively charged surface, which prevents the oil bodies from aggregatingand stops coalescence if the structures touch (Tzen and Huang, 1992). In fact oil bodies areremarkably stable both in and out of the cell due to steric hindrance and electronegativerepulsion provided by the oleosins on the surface of the oil bodies (Tzen et al., 1992).Oleosins are insoluble in aqueous media, have a pI of 5.7–6.6 and make up 8–20% of thetotal seed protein (Murphy, 1993; Huang, 1996).

It is thought that the oil body size is determined by the ratio of oil to oleosin during oilbody formation (Murphy, 1999), which means that it could be possible to control the size of

Emulsions as Delivery Systems in Foods 69

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 69

Page 79: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

oil body by controlling the rate that oleosin is produced. The nature of the oil within the oilbody can also be important both for determining the types of actives that can be encapsu-lated and for the specific application in foods and pharmaceuticals.

During normal extraction of oil from plant materials the oil bodies are normallydestroyed due to the high shear processes of crushing and milling followed by degummingand further refining (Gunstone et al., 1994). In the last ten years a number of companies(e.g., Sembiosys) have developed methods to extract oil bodies from seeds or plants with-out destroying them and in good yield. A number of papers and patents have been publishedconcerning the specific use of oil bodies for therapeutic and nutraceutical purposes byattaching active peptides to the termini of the oleosin protein and using the oil body as acarrier of the active component concerned (Boothe et al., 1997; Deckers et al., 1998, 1999).This type of research has also stimulated many workers in the field to look at a number ofways in which oil bodies can be modified to make them more functional. This has includedimproving the payload of lipophilic material by extracting all of the oil from the oil body toleave an empty ghost (Tzen and Huang, 1992; Tzen et al., 1998), which can be later filledwith a combination of different oils and actives. These regenerated oil bodies possess thesame physiochemical properties as the original oil bodies but now possess higher payloadsof active.

Oil bodies have also been modified to target specific sites for the delivery of an active bymodifying the oleosin proteins, which due to their high level of functional groups makethem very susceptible to alterations. Much is already known about the genetics of differentplant species, and genetically modified oil bodies have already been produced in which, forexample, -glucuronidase enzyme has been fused to an oil body and shown to be active

70 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

(a) (b)

Figure 3.3. Confocal scanning light microscopic images of an intact pine tree seed cell (left) inthe presence of Nile Blue. The dotted line represents the cell wall. Purified oil bodies could beisolated from these cells (right). The light grey spheres in both images depict the oil core of theoil bodies. The white colour represents the protein containing cell structures (hardly visible inthe right picture). These pictures have been kindly provided by our colleagues C.M. Beindorffand E. Drost of Unilever R&D Vlaardingen, The Netherlands.

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 70

Page 80: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

(Abenes et al., 1997). Other forms of modification to the oil bodies have been via chemicalmodification (cross-linked with glutaraldehyde or genipin) to enhance their stability (Penget al., 2003) and self-assembling targeting systems, in which oil bodies can be targetedeffectively to their site of action via multivalent antigen-binding proteins (Frenken et al.,1999) since antibodies are easily raised to oleosin (Cummins and Murphy, 1992; Wuet al., 1997).

Since the constituents of native oil bodies and their proportions are well known, it hasbeen possible to produce stable artificial oil bodies technically reconstituted from theirthree main components: triglycerols, phospholipids, and oleosin protein (Tzen and Huang,1992; Tzen et al., 1998; Tai et al., 2002). Artificial oil bodies were successfully reconsti-tuted with various compositions of these components and compared to native oil bodies forsize and stability. Increasing the size of the oil body led to a decrease in the thermostabilityand structural stability of the reconstituted oil bodies.

Native oil bodies, modified and reconstructed, can be a useful structure for a range ofapplications especially as a carrier for hydrophobic molecules such as flavors, vitamins,nutraceutical actives (e.g., antioxidants) and pharmaceutical drugs (e.g., steroids), and cos-metic lipids (e.g., healthy fatty acids) (Peng et al., 2003). Other applications are as a vehiclefor the production of recombinant proteins (van Rooijen and Moloney, 1995), as a biocap-sule for encapsulation of lactic acid bacteria in dairy products (Hou et al., 2003) and the useof artificial oil bodies reconstituted with olive oil and phospholipid in the presence of cale-osin to elevate the bioavailability of hydrophobic drug cyclosporin A via oral administra-tion (Chen et al., 2005).

Yeast Cells

Yeast cells have been explored recently by a number of workers for their potential as con-trolled delivery devices for flavor release (Bishop et al., 1998; Normand et al., 2005) and toimprove the bioavailability of poorly soluble drugs in the GI tract (Nelson et al., 2006).Indeed, yeast cells have been investigated as early as the 1970s when LaboratoiresSérozym, France (Laboratoires Sérozym, 1973) and Swift and Co., USA (Shark, 1977)patented a technique using specially prepared yeast cells containing >40% loading of lipid.They described the encapsulation of dyes, drugs, and flavors in viable and nonviablemicroorganisms including fungi and protozoa. The mechanism of the encapsulationprocess in yeast cells relies on the relative affinity of would-be encapsulated material forthe internal lipid phase of the yeast cell. Flavor components which display ideal solutionwith this lipid phase will be encapsulated to the greatest degree.

It has been suggested that the internal lipid phase is primarily made of phospholipidbilayer membranes unlike a classic micelle structure. Actives which are extensivelynonpolar (such as -carotene) might be expected to exist in the interior of the micelle(Wedzicha, 1988); however, their molecular size would involve geometric changes tothe micelle and therefore very high molecular weight hydrocarbons may be excluded fromthe cell.

Rebalancing flavors, the use of co-encapsulates to alter the properties of the internallipid phase to compensate for disproportionate uptake, and other cell modifications such asextraction of the cell wall using detergents (Chow and Palecek, 2004) to improve perme-ability have helped extend the allocation range of the yeast cells as preformed capsules.

Emulsions as Delivery Systems in Foods 71

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 71

Page 81: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Indeed, yeast cell wall composition and thickness can be modified using different cellstrains for enzyme expression or by mutating genes involved in cell wall biosynthesis ordegradation (Chow and Palecek, 2004).

Under dry conditions (e.g., water activity below 0.7), release rates are considerablylow due to limited mass transfer. Flavour release can be resumed upon rehydration(Normand et al., 2005). Normand et al. (2005) have used limonene as a model marker forhydrophobic flavors and discussed the flavor-release mechanism with regard to the cell wallstructure and its behavior toward water uptake and also desorption during the drying of theyeast cells. The basis of the driving force for flavor release from hydrated yeast cellsappears in good agreement with the theory describing monolithic solution release, a theoryderived by Crank (1956) and applied to spherical controlled-release devices by Baker andLonsdale (Baker and Lonsdale, 1974; Baker, 1987) demonstrating a biphasic releasepattern. Importantly, the resistance to transfer of flavor materials within the hydrated yeastcell is not rate-determining, and the kinetics of release are dictated by the aqueous phasesolubilities.

Plant Cells

A plant cell in nature is surrounded by a cell wall and therefore not prone to allowingmacromolecules from outside to accumulate within the cell (Rosenbluh et al., 2004).Indeed, cells are protected from the surrounding environment by plasma membrane, whichis impenetrable for most hydrophilic and hydrophobic materials. However, it would appearthat a process resembling cell endocytosis, which occurs in animals, can also occur in plantcells (Robinson et al., 1998; Daelemans et al., 2002) although much less is known about thedetailed mechanism. It has been shown that the addition of macromolecules that have beenbiotinylated such as hemoglobin, BSA or IgG to cultured soybean cells resulted in theirintracellular accumulation (Horn et al., 1990, 1992) and that this process was temperaturedependent indicating a requirement for metabolic energy.

There are, however, certain low molecular weight proteins that appear able to cross theplasma membrane at least for mammalian cells without the involvement of the endocyticpathway (Lindgren et al., 2000) and have been termed “cell-penetrating protein/peptides”(CCP). These types of molecules such as purified core histones (Rosenbluh et al., 2004) arealso capable of crossing plasma membranes of plant cells and acting as CCPs in plant cells.These molecules can be used to mediate the internalization of larger molecules such asoligonucleotides, peptides, proteins, and nanoparticles following their conjugation to theCCP (Fawell et al., 1994; Pooga et al., 1998; Astriab-Fisher et al., 2002). In plant cells it hasbeen confirmed using confocal laser-scanning microscopy that histone-BSA conjugateshave penetrated into protoplasts of petunia plants via direct translocation through theplasma membrane (Rosenbluh et al., 2004). This type of technology therefore gives anapproach that could be used to introduce and deliver a whole range of actives and macro-molecules into plant cells. Although in the biotechnology area, the internalization of CPPsand the attached molecules by plant cells may open up a new method for transfection inplant cells (Mae et al., 2005), this method could also be used to load plant cells with activemolecules such as flavors, vitamins, and so on to be used as controlled delivery devices.Due to the plasma membrane and cell wall structures, plant cells make excellent preformedcapsules that can contain a range of macromolecules in a very natural system, which can beused in a range of foods.

72 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 72

Page 82: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Monodispersed Emulsions

Several technologies have been developed to produce highly uniform emulsion droplets(see Link et al., 2004, and references therein). Technologies to reduce polydispersity ofalready formed emulsions include repeated fractionation and shearing immiscible fluidsbetween uniformly separated plates (Mabille et al., 2003). Alternatively, single-drop tech-nologies are available, such as flow through a micromachined comb, hydrodynamic flowfocusing through a small orifice, and drop break off in co-flowing streams (Figure 3.4).Using microchannel technology, more-complex droplet structures have been prepared:w/o/w emulsions (Okushima et al., 2004; Sugiura et al., 2004), gelled beads with a varietyof shapes (Seo et al., 2005; Dendukuri et al., 2006), Janus particles where the two halvespresent different properties (Nisisako et al., 2004), and a variety of encapsulates. Currently,these single-drop technologies are limited in production rate (in the order of �l–ml perhour). Highly parallel production at a small scale by microfluidic technology may reducethis limitation in the future.

Monodispersed emulsions may have a more defined behavior and release pattern ofentrapped actives than polydispersed ones. This can be very important in pharmaceuticsand when the emulsions are used as a template to make new materials for, for example,electronics. Currently, it is not clear whether or not this would constitute a real advantage infood systems. Using these technologies may allow forming a better picture of the rheologi-cal and organoleptic behavior of monodispersed emulsions by experimentally testing theirproperties.

Emulsions as Delivery Systems in Foods 73

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Oil flow

Oil flow

Tip

Microchannel (100 μm width)

Water flow

(a)

(b)

Figure 3.4. Emulsion production via microfluidic technology. Here a so-called psi-junction isused. Other geometries are possible as well. (a) shows the schematic overview and (b) is amicroscopic “real” picture that has been kindly provided by Conchi Pulido de Torres, UnileverR&D Colworth, UK.

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 73

Page 83: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

ReferencesAbenes, M., Holbrook, L., and Moloney, M. 1997. Transient expression and oil body targeting of an Arabidopsis

oleosin-GUS reporter fusion protein in a range of oilseed embryos, Plant Cell Rep. 17(1), 1–7.Astriab-Fisher, A., Sergueev, D., Fisher M., Shaw, B.R., and Juliano, R.L. 2002. Conjugates of antisense oligonu-

cleotides with the Tat and antennapedia cell-penetrating peptides: effects on cellular uptake, binding to targetsequences, and biologic actions, Pharm. Res. 19, 744–754.

Baines, Z.V., and Morris, E.R. 1987. Flavour/taste perception in thickened systems: The effect of guar gum aboveand below C*, Food Hydrocolloids 1, 197–205.

Baker, R.W. 1987. Controlled release of biologically active agents. Wiley-Interscience, New York.Baker, R.W., and Lonsdale, H.K. 1974. Controlled release of biologically active agents. A.C. Tanquary and R.E.

Lacey (Eds) Plenum Press, New York, 15–22.Bakker, J. 1995. “Flavor interactions with the food matrix and their effects on perception”, in Gaonker, A.G. (Ed.)

Ingredient interactions: Effect on food quality, Marcel Dekker, 411–439.Bakker, J., Boudaud, N., and Harrison, M. 1998. Dynamic release of diacetyl from liquid gelatin in the headspace,

J. Agri. Food Chem. 46, 2714–2720.Belitz, H.D., Grosch, W., and Schieberle, P. 2001. Lehrbuch der lebensmittelchemie. Springer, Heidelberg.Benichou, A., Aserin, A., and Garti, N. 2002. Protein–polysaccharide interactions for stabilization of food emul-

sions, J. Dispersion Sci. Technol. 23, 93–123.Benichou, A., Aserin, A., and Garti, N. 2004. Double emulsions stabilized with hybrids of natural polymers for

entrapment and slow release of active matters, Adv. Colloid Interface Sci. 108–109, 29–41.Bishop, J.R.P., Nelson, G., and Lamb, J. 1998. Microencapsulation in yeast cells, J. Microencapsul. 15(6),

761–773.Boothe, J.G., Saponia, J.A., and Parmenter, D.L. 1997. Molecular farming in plants: Oilseeds as vehicles for the

production of pharmaceutical proteins, Drug Dev. Res. 42, 172–181.Bratskaya, S., Avramenko, V., Schwarz, S., and Philippova, I. 2006. Enhanced flocculation of oil-in-water emul-

sions by hydrophobically modified chitosan derivatives, Colloids Surfaces A-Physicochem. Eng. Aspects275(1–3), 168–176.

Brodin, A.F., Kavaliunas, D.R., and Frank, S.G. 1978. Prolonged drug release from multiple emulsions, ActaPharmacol. 45, 135–136.

Chaiyasit, W., McClements, D.J., and Decker, E.A. 2005. The relationship between the physicochemical proper-ties of antioxidants and their ability to inhibit lipid oxidation in bulk oil and oil-in-water emulsions, J. Agric.Food Chem. 53, 4982–4988.

Chanamai, R., Herrmann, N., and McClements, D.J. 2000. Probing floc structure by ultrasonic spectroscopy, vis-cometry, and creaming measurements, Langmuir 16(14), 5884–5891.

Charcosset, C., Limayem, I., and Fessi, H. 2004. The membrane emulsification process—a review, J. Chem. Tech-nol. Biotechnol. 79, 209–218.

Chen, M.C.M., Chyan, C.L., Lee, T.T.T., Hung, S.H., and Tzen, J.T.C. 2004. Constitution of stable artificial oilbodies with triacylglycerol, phospholipid, and caleosin, J. Agric. Food Chem. 52, 3982–3987.

Chen, M.C.M., Wang, J.L., and Tzen, J.T.C. 2005. Elevating bioavailability of cyclosporine A via encapsulation inartificial oil bodies stabilised by caleosin, Biotechnol. Prog. 21, 1297–1301.

Chow, C.K, and Palecek, S.P. 2004. Enzyme encapsulation in permeabilised Sacchraromyces cerevisiae cells.Biotechnol. Prog. 20(2), 449–456.

Conner, W.E. 2000. Importance of n-3 fatty acids in health and disease, Am. J. Clin. Nutr. 71, 171S–175S.Coupland, J.N. 2002. Crystallization in emulsions, Current Opinion Colloid Interface Sci. 7(5–6), 445–450.Crank, J. 1956. The mathematics of diffusion. Oxford University Press, London.Cummins, I., and Murphy, D.J. 1992. cDNA sequence of a sunflower oleosin and transcript tissue specificity, Plant

Mol. Biol. 19(5), 873–876.Daelemans, D., Afonina, E., Nilsson, J., Werner, G., Kjems, J., De Clercq, E., Pavlakis, G.N., and Vandamme,

A.M. 2002. Synthetic HIV-1 Rev inhibitor interfering with the CRM1-mediated nuclear export, Proc. Natl.Acad. Sci. USA. 99, 14440–14445.

Dalgleish, D.G., 2006. Food emulsions—their structures and structure-forming properties, Food Hydrocolloids20(4), 415–422.

Davis, S.S., Hadgraft, J., and Palin, K.J. 1985. “Medical and pharmaceutical applications of emulsions”, inBecher, P. (Ed.) Encyclopedia of emulsion technology, Volume 2, Applications. New York: Marcel Dekker.159–238.

de Roos, K.B. 1997. How lipids influence food flavor, Food Technol. 51(1), 60–62.

74 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 74

Page 84: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

de Roos, K.B., and Wolswinkel, K. 1994. “Non-equilibrium partition model for predicting flavour release in themouth”, in Maarse, H., van der Heij, D.G. (Eds) Trends in flavour science. Elsevier, Amsterdam. 15–32.

Deckers, H., Moloney, M., and Baum, A. 1999. The case of recombinant production of pharmaceutical proteins inplants, Annu. Rep. Med. Chem. 34, 237–245.

Deckers, H.M., van Rooijen, G., Booth, J., Goll, J., Moloney, M., and Mahmoud, S.S. 1998. Uses of oil bodies,WO9853698.

Delahunty, C.M., and Piggott, J.R. 1995. Current methods to evaluate contribution and interactions of componentsto flavour of solid foods using hard cheese as an example, Int. J. Food Sci. Tech. 30, 555–570.

Dendukuri, D., Pregibon, D.C., Collins, J., Hatton, T.A., and Doyle, P.S. 2006. Continuous-flow lithography forhigh-throughput microparticle synthesis, Nature Materials 5(5), 365–369.

Dickinson, E. 1996. Biopolymer interactions in emulsion systems: Influences on creaming, flocculation, and rhe-ology, ACS Symp. Series 650, 197–207.

Dickinson, E. 1997. Enzymatic crosslinking as a tool for food colloid rheology control and interfacial stabiliza-tion, Trends Food Sci. Technol. 8(10), 334–339.

Dickinson, E. 1998. Structure, stability and rheology of flocculated emulsions, Curr. Opinion Colloid InterfaceSci. 3(6), 633–638.

Dickinson, E., and Golding, M. 1997a. Depletion flocculation of emulsions containing unadsorbed sodiumcaseinate, Food Hydrocolloids 11(1), 13–18.

Dickinson, E., and Golding, M. 1997b. Rheology of sodium caseinate stabilized oil-in-water emulsions, J. ColloidInterface Sci. 191(1), 166–176.

Dickinson, E., and Golding, M. 1998. Influence of calcium ions on creaming and rheology of emulsions contain-ing sodium caseinate, Colloids Surfaces A—Physicochem. Eng. Aspects 144 (1–3), 167–177.

Dickinson, E., and Yamamoto, Y., 1996. Rheology of milk protein gels and protein-stabilized emulsion gels cross-linked with transglutaminase, J. Agric. Food Chem. 44(6), 1371–1377.

Dickinson, E., Ma, J.G., and Povey, M.J.W. 1994. Creaming of concentrated oil-in-water emulsions containingxanthan, Food Hydrocolloids 8(5), 481–497.

Dickinson, E., Murray, B.S., and Stainsby, G. 1988. Coalescence stability of emulsion-sized droplets at a planaroil–water interface and the relationship to protein film surface rheology, J. Chem. Soc. Faraday Trans. I 84,871–883.

Djerdjev, A.M., Hunter, R.J., and Beattie, J.K., 2006. Self-depletion flocculation of tetralin oil-in-water emulsions,Langmuir 22(1), 84–87.

Doyen, K., Carey, M., Linforth, R.S.T., Marin, M., and Taylor, A.J. 2001. Volatile release from an emulsion: Head-space and in-mouth studies, J. Agric. Food Chem. 49(2), 804–810.

Druaux, C., and Voilley, A. 1997. Effect of food composition and microstructure on volatile flavour release,Trends Food Sci. Tech. 8, 364–368.

Edris, A., and Bergnståhl, B. 2001. Encapsulation of orange oil in a spray dried double emulsion, Nahrung Food45(2), 133–137.

Elson, L.A. Mitcheley, B.C.V., Collings, A.J., and Schneider, R. 1970. Chemotherapeutic effect of a water-oil-water emulsion of methotrexate on the mouse L1210 leukemia, Revue Europeene détudes Cliniques etBiologiques 15, 87–90.

Engel, R., and Shubert, H. 2005. Formulation of phytosterols in emulsions for increased dose response in func-tional foods, Innovative Food Sci. Emerg. Technol. 6, 233–237.

Faraji, H., McClements, D.J., and Decker, E.A. 2004. Role of continuous phase protein on the oxidative stabilityof fish oil-in-water emulsions, J. Agric. Food Chem. 52, 4558–4564.

Fawell, S., Seery, J., Daikh, Y., Moore, C., Chen, L.L., Pepinsky, B., and Barsoum, J. 1994. Tat-mediated deliveryof heterologous proteins into cells, Proc. Natl. Acad. Sci. USA. 91, 664–668.

Ficheux, M.F., Bonakdar, L., Leal-Calderon, F., and Bibette, J. 1998. Some stability criteria for double emulsions,Langmuir 14, 2702–2706.

Florence, A.T., and Whitehill, D. 1981. Some features of breakdown in w/o/w multiple emulsions, J. Colloid Inter-face Sci. 79(1), 243–256.

Forss, D.A. 1973. Odour and flavour compounds from lipids, Prog. Chem. Fat Lipids 13, 181–253.Frankel, E.N. 1996. Antioxidants in lipid foods and their impact on food quality, Food Chem. 57, 51–55.Frenken, L.G.J., Howell, S., Ledeboer, A.M., and van der Logt, C.P.E. 1999. Multivalent antigen-binding proteins,

WO9923221 (Galazka et al., 2000).Gallarate, M., Carlotti, M.E, Trotta, M., and Bovo, S. 1999. On the stability of ascorbic acid in emulsified systems

for topical and cosmetic use, Int. J. Pharm. 188, 233–241.Gaonkar, A.G. 1994. Method for preparing a multiple emulsion, US 5322704.

Emulsions as Delivery Systems in Foods 75

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 75

Page 85: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Garti, N. 1996. “Delivery of microparticulated liquid systems in food”, in Barenholz, Y., and Lasic, D.D. (Eds)Handbook of nonmedical applications of liposomes III. CRC Press, Boca Raton. 143–198.

Garti, N. 1997a. Double emulsion—scope, limitation and new achievements, Colloids and Surf B Biointerfaces123, 233–246.

Garti, N. 1997b. Progress in stabilization and transport phenomena of double emulsions in food applications, FoodSci. Technol. 30, 222.

Garti, N. 1998. In Seiller, M., and Grossiord, J.L. (Eds) Multiple emulsions: Structure, properties and applica-tions. Editions de santé, Paris.

Garti, N., and Aserin, A. 1996a. “Double emulsions stabilized by macromolecular surfactants” in Chattopadhyay,A.K., and Mittal, K.L. (Eds) Surfactants in solution. Surfactant Science Series 64. Marcel Dekker, New York.297–332.

Garti, N., and Aserin, A. 1996b. Double emulsions stabilized by macromolecular surfactants, Adv. Colloid Inter-face Sci. 65, 37–69.

Garti, N., and Benichou, A. 2001. “Double emulsions for controlled release applications: Progress and trends”, inSjoblom, J. (Ed.) Encyclopedic handbook of emulsion technology. Marcel Dekker, New York. 377–408.

Garti, N., and Bisperink, C. 1998. Double emulsions: Progress and applications, Curr. Opin. Colloid Interface Sci.3, 657–667.

Garti, N., Aserin, A., and Cohen, Y. 1994. Mechanistic considerations on the release of electrolytes from multipleemulsions stabilised by BSA and non-ionic surfactants, J. Control. Release 29, 41–51.

Giermanska-Kahn, J., Laine, V., Arditty, S., Schmitt, V., and Leal-Calderon, F. 2005. Particle-stabilized emulsionscomprised of solid droplets, Langmuir 21(10), 4316–4323.

Gijsbertsen-Abrahamse, A.J., van der Padt, A., and Boom, R.M. 2004. Status of cross-flow membrane emulsifica-tion and outlook for industrial application, J. Membr. Sci. 230, 149–159.

Guichard, E. 2002. Interactions between flavor compounds and food ingredients and their influence on flavor per-ception, Food Rev. Int. 18(1), 49–70.

Guinard, J.X., and Marty, C. 1995. Time intensity measurements of flavour release from a model gel system:Effect of gelling agent type and concentration, J. Food Sci. 60(4), 727–730.

Gunstone, F.D., Harwood, J.L., and Padley, F.B. 1994. The lipid handbook. Chapman and Hall, London.Harrison, M., 1998. Effect of breathing and saliva flow on flavor release from liquid food, J. Agric. Food Chem. 46,

2727–2735.Harrison, M. 2000. Flavour Release, Roberts, D.D., and Taylor, A.J. (Eds) American Chemical Society.Harrison, M., and Hills, B.P. 1996. A mathematical model to describe flavour release from gelatine gels, Int.

J. Food Sci. Technol. 31, 167–176.Harrison, M., and Hills, B.P. 1997a. Effects of air flow-rate on flavour release from liquid emulsions in the mouth,

Int. J. Food Sci. Technol. 32, 1–9.Harrison, M., and Hills, B.P. 1997b. Mathematical model of flavour release from liquids containing aroma-bind-

ing macromolecules, J. Agric. Food Chem. 45, 1883–1890.Harrison, M., Campbell, S., and Hills, B.P. 1998. Computer simulation of flavor release from solid foods in the

mouth, J. Agric. Food Chem. 46, 2736–2743.Harrison, M., Hills, B.P., Bakker, J., and Clothier, T. 1997. Mathematical models of flavor release from liquid

emulsions, J. Food Sci. 62, 653.Hatchwell, C. 1996. “Implications of fats on flavours”, in McGorrin, R.J., and Leland, J.V. (Eds) Flavor–food

interactions. ACS symposium series 633, Washington DC.Hills, B.P., and Harrison, M. 1995. Two-film theory of flavour release from solids, Int. J. Food Sci. Technol. 30,

425–436.Hoang, T.K.N., Deriemaeker, L., La, V.B., and Finsy, R. 2004. Monitoring the simultaneous Ostwald ripening and

solubilization of emulsions, Langmuir 20(21), 8966–8969.Horn, M., Heinstein, P., and Low, P. 1990. Biotin-mediated delivery of exogenous macromolecules into soybean

cells, Plant Physiol. 93, 1492–1496.Horn, M., Heinstein, P., and Low, P. 1992. Characterization of parameters influencing receptor-mediated endocy-

tosis in cultured soybean cells, Plant Physiol. 98, 673–679.Hotrum, N.E., Stuart, M.A.C., van Vliet, T., Avino, S.F., and van Aken, G.A. 2005. Elucidating the relationship

between the spreading coefficient, surface-mediated partial coalescence and the whipping time of artificialcream, Colloids and Surfaces A-Physicochem and Eng. Aspects 260(1–3), 71–78.

Hou, R.C.W., Lin, M.Y., Wang, M.M.C., and Tzen, J.T.C. 2003. Increase of viability of entrapped cells of Lacto-bacillus delbrueckii ssp. bulgaricus in artificial sesame oil emulsions, J. Dairy Sci. 86, 424–428.

76 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 76

Page 86: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Hu, M., McClements, D.J., and Decker, E.A. 2003a. Impact of whey protein emulsifiers on the oxidative stabilityof salmon oil-in-water emulsions, J. Agric. Food Chem. 51, 1435–1439.

Hu, M., McClements, D.J., and Decker, E.A. 2003b. Lipid oxidation in corn oil-in-water emulsions stabilized bycasein, whey protein isolate and soy protein isolate, J. Agric. Food Chem. 51, 1696–1700.

Huang, A.H.C. 1996. Oleosins and oil bodies in seeds and other organs. Plant Physiol. 110, 1055–1061. In “Watermodel systems”, J. Agric. Food Chem. 45, 1991–1994.

Huang, S.W., Hopia, A., Schwarz, K., Frankel, E.N., and German, J.B. 1996a. Antioxidant activity of α-tocopheroland Trolox in different lipid substrates: Bulk oils vs oil-in-water emulsions, J. Agric. Food Chem. 44, 444–452.

Huang, S.W., Frankel, E.N., Schwartz, K., Aeschbach, R., and German, J.B. 1996b. Antioxidant activity ofcarnosic acid and methyl carnosoate in bulk oil and in oil-in-water emulsion, J. Agric. Food Chem. 45,2951–2956.

Jacobsen, C. 2004. “Developing polyunsaturated fatty acids as functional ingredients”, in Arnoldi, A. (Ed.) Func-tional foods, cardiovascular disease and diabetes. Woodhouse Publishing, Utah.

Jage-Lezer, N., Terrisse, I., Bruneau, F., Tokgoz, S, Ferreirra, L., Clausse, D., Seiller, M., and Grossiord, J.L. 1997.Influence of lipophilic surfactant on the release kinetics of water soluble molecules entrapped in a w/o/w mul-tiple emulsions, J. Control Release 45, 1–13.

Jo, C., and Ahn, D.U. 1999. Fat reduces volatiles production in oil emulsion system analyzed by purge and trapdynamic headspace/gas chromatography, J. Food Sci. 64, 641–643.

Joscelyne, S.M., and Tragardh, G. 2000. Membrane emulsification—a literature review, J. Membr. Sci. 169, 107–117.Karel, M. 1992. “Kinetics of lipid oxidation”, in Schwartzberg, H.G., and Hartel, R.W. (Eds) Physical chemistry

of foods. Marcel Dekker Inc., New York.Kim, S.Y., and Lee, Y.M. 1999. Lipid nanospheres containing vitamin A or vitamin E: Evaluation of their stabili-

ties and in vitro skin permeability, J. Ind. Eng. Chem. 5(4), 306–313.Kinsella, J.E. 1990. Flavour perception and binding, Int. News Fat Oils Relat. Matter 1(3), 215–216.Klahn, J.K., Janssen, J.J.M., Vaessen, G.E.J., de Swart, R., and Agterof, W.G.M. 2002. On the escape process dur-

ing phase inversion of an emulsion, Colloid surf A–Physicochem Eng. Aspects 210, 167.Klinkesorn, U., Sophanodora, P., Chinachoti, P., McClements, D.J., and Decker, E.A. 2005. Increasing the oxidat-

ive stability of liquid and dried tuna oil-in-water emulsions with electrostatic layer-by-layer deposition technol-ogy, J. Agric. Food Chem. 53, 4561–4566.

Kuhn, P.R., and Foegeding, E.A. 1991. Mineral salt effects on whey protein gelation, J. Agric. Food Chem. 39(6),1013–1016.

Laboratoires Sérozym. 1973. Procédé pour faire pénétrer, absorber et/ou fixer par des microorganismes des sub-stances diverses, French Patent no. 72.12622.

Lambrich, U., and Vladisavljevic, G.T. 2004. Emulgieren mit microstrukturierten Systemen, Chem. Ing. Tech.76(4), 376.

Lambrich, U., and Schubert, H. 2005. Emulsification using microporous systems, J. Memb. Sci. 257(1–2), 76–84.Lata, M., Nasa, S.L., and Murphy, R.S. 1987. Microencapsulation of salbutamol sulphate using multiple emulsion

technique, Eastern pharmacist 30, 137–138.Le Thanh, M., Goubet, I., Le Quéré, J.L., Voilley, A. 1998. Interactions between volatilities and lipids in complex

systems, J. Am. Oil Chem. Soc. 75, 441–445.Le Guen, S., and Vreeker, R. 2003. “Interactions between flavour compounds and milk proteins under static and

dynamic conditions”, in Le Quéré, J.L., and Etievant, P.X. (Eds) Flavour research at the dawn of the twenty-firstcentury. Lavoisier, Cachan, France.

Leland, J.V. 1997. Flavor interactions: The greater whole, Food Technol. 51(1), 75–80.Lian, G., Malone, M., Homan, J.E., and Norton, I.T. 2004. A mathematical model of volatile release in mouth

from the dispersion of gelled emulsion particles, J. Control. Release 98, 139–155.Lindgren, M., Hallbrink, M., Prochiantz, A., and Langel, U. 2000. Cell-penetrating peptides, Trends Pharmacol.

Sci. 21, 99–103.Link, D.R., Anna, S.L., Weitz, D.A., and Stone, H.A. 2004. Geometrically mediated breakup of drops in microflu-

idic devices, Phys. Rev. Lett. 92(5), 054503.Lynch, J., Liu, J.-H., Mela, D.J., and MacFie, H. 1993. A time-intensity study of the effect of oil mouth coatings on

taste perception, Chem. Senses 18, 121–129.Mabille, C., Leal-Calderon, L., Bibette, J., and Schmitt, V. 2003. Monodisperse fragmentation in emulsions:

Mechanisms and kinetics, Europhysics Letters 61(5), 708–714.Mae, M., Myrberg, H., Jiang, Y., Paves, H., Valkna, A., and Langel, U. 2005. Internalisation of cell-penetrating

peptides into tobacco protoplasts, Biochimica et Biophysica Biomembranes 1669, 101–107.

Emulsions as Delivery Systems in Foods 77

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 77

Page 87: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Malkki, Y., Heinio, R.L, and Autio, K. 1990. Influence of oat gum, guar gum and carboxymethyl cellulose on theperception of sweetness and flavour, Food Hydrocolloids 6(6), 525–532.

Malone, M.E., and Appelqvist, I.A.M. 2003. Gelled emulsion particles for the controlled release of lipophilicvolatiles during eating, J. Control Release 90(2), 227–241.

Malone, M.E., Appelqvist, I.A.M., and Norton, I.T. 2003. Oral behaviour of food hydrocolloids and emulsions—Part 2: Taste and aroma release, Food Hydrocolloids 17, 775–784.

Malone, M.E., Appelqvist, I.A.M., Goff, T.C., Homan, J.E., and Wilkins, J.P.G. 2000. “A novel approach to theselective control of lipophilic flavour release in low fat foods”, in Roberts, D.D., and Taylor, A.J. (Eds) FlavourRelease. ACS Symposium Series 763, American Chemical Society, USA, 212–227.

Matsumoto, S., and Kang, W.W. 1989. Formation and applications of multiple emulsions, J. Disper. Sci. Technol,10, 455–482.

McClements, D.J. 2005 (2nd edn). Food emulsions: Principles, practices, and techniques. CRC Press, BocaRaton, Florida.

McClements, D.J., and Decker, E.A. 2000. Lipid oxidation in oil-in-water emulsions: Impact of molecular envi-ronment on chemical reactions in heterogeneous food systems, J. Food Sci. 65, 1270–1282.

McGorrin, R.J., and Leland, J.V. 1994. Flavour-food interactions. ACS Symposium Series 633, American Chemi-cal Society, USA.

Mei, L.Y., McClements, D.J., Wu, J.N., and Decker, E.A. 1997. Iron-catalyzed lipid oxidation in emulsion asaffected by surfactant, pH and NaCl, Food Chem. 61, 307–312.

Meinders, M.B.J., and van Vliet, T. 2004. The role of interfacial rheological properties on Ostwald ripening inemulsions, Adv. Colloid Interface Sci. 108, 119–126.

Moschakis, T., Murray, B.S., and Dickinson, E. 2005. Microstructural evolution of viscoelastic emulsions sta-bilised by sodium caseinate and xanthan gum, J. Colloid Interface Sci. 284(2), 714–728.

Muguet, V., Seiller, M., Barratt, G., Clausse, D., Marty J.P., and Grossoird, J.L. 1999. W/O/W multiple emulsionssubmitted to a linear shear flow: Correlation between fragmentation and release, J. Colloid Interface Sci. 218,335–337.

Mun, S.H., and McClements, D.J. 2006. Influence of interfacial characteristics on Ostwald ripening in hydrocar-bon oil-in-water emulsions, Langmuir 22(4), 1551–1554.

Murphy, D.J. 1993. Structure, function and biogenesis of storage lipid bodies and oleosins in plants, Prog. LipidRes. 32(3), 247–280.

Murphy, D.J. 1999 (2nd Edn). “Plant lipids. Their metabolism, function and utilization”, in Lea, P.J., and Leegood,R.C. (Eds) Plant biochemistry and molecular biology. John Wiley and Sons, Chichestor.

Murphy, D.J., Hernandez-Pinzon, I., and Patel, K. 2001. Role of lipid bodies and lipid-body proteins in seeds andother tissues, J. Plant Physiol. 158, 471–478.

Nakashima, T., Shimizu, M., and Kukizaki, M. 1991. Membrane emulsification by microporous glass, Key Eng.Mater. 61/61, 513.

Nelson, G., Duckham, S.C., and Crothers, M.E.D. 2006. Microencapsulation in yeast cells and applications indrug delivery. Polymeric drug delivery, Volume 1 Particulate drug carriers. ACS Symposium Series 923,268–281.

Nestel, P.J. 2000. Fish oil and cardiovascular disease: Lipids and arterial function, Am. J. Clin. Nutr. 71, 228–231.Nisisako, T., Torii, T., and Higuchi, T. 2004. Novel microreactors for functional polymer beads, Chem. Eng. J. 101,

23–29.Noakes, M., Clifton, P.M., Doornbos, A.M.E., Trautwein, E.A. 2005. Plant sterol ester–enriched milk and

yoghurt effectively reduce serum cholesterol in modestly hypercholesterolemic subjects, Eur. J. Nutr. 44,214–222.

Normand, V., Dardelle, G., Bouquerand, P.E., Nicolas, L., and Johnston, D.J. 2005. Flavour encapsulation inyeasts: Limonene used as a model system for characterisation of the release mechanism, J. Agric. Food Chem.53(19), 7532–7543.

Okushima, S., Nisisako, T., Torii, T., and Higuchi, T. 2004. Controlled production of monodisperse double emul-sions by two-step droplet breakup in microfluidic devices, Langmuir 20(23), 9905–9908.

Onuki, Y., Morishita, M., and Takayama, K. 2004. Formulation optimization of water-in-oil-water multiple emul-sion for intestinal insulin delivery, J. Control. Release 97, 91–99.

Overbosch, P., Agterof, W.G.M., and Haring, P.G.M. 1991. Flavour release in the mouth, Food Rev. Int. 7,137–184.

Pandit, J.K., Mishra, B., and Chand, B. 1987. Drug release from multiple w/o/w emulsions, Indian J. Pharm. Sci.49, 103–105.

78 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 78

Page 88: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Paquin, P. 1999. Technological properties of high pressure homogenizers: The effect of fat globules, milk proteins,and polysaccharides, Int. Dairy J. 9(3–6), 329–335.

Pays, K., Giermanska-Kahn, J., Pouligny, B., Bibette, J., and Leal-Caldron, F. 2002. Double emulsions: How doesrelease occur, J. Control. Release, 79(1–3), 193–205.

Peng, C.C., Lin, I.P., Lin, C.K., and Tzen, J.T.C. 2003. Size and stability of reconstituted sesame oil bodies,Biotechnol. Prog. 19, 1623–1626.

Perrier-Cornet, J.M., Marie, P., and Gervais, P. 2005. Comparison of emulsification efficiency of protein-stabilized oil-in-water emulsions using jet, high pressure and colloid mill homogenization, J. Food Eng. 66(2),211–217.

Pooga, M., Soomets, U., Hallbrink, M., Valkna, A., Saar, K., Rezaei, K., Kahl, U., Hao, J.X., Xu, X.J., Wiesenfeld-Hallin, Z., Hokfelt, T., Bartfai, T., and Langel, U. 1998. Cell-penetrating PNA constructs regulate galanin recep-tor levels and modify pain transmission in vivo, Nat. Biotechnol. 16, 857–861.

Rabe, S., Krings, U., and Berger, R.G. 2003. Influence of oil-in-water emulsion characteristics on initial dynamicflavour release, J. Sci. Food Agric. 83, 1124–1133.

Ribeiro, H.S., Ax, K., and Schubert, H. 2003. Stability of lycopene emulsions in foods systems, J. Food Sci. 68(9),2730–2734.

Robinson, D.G., Hinz, G., and Holstein, S.E. 1998. The molecular characterization of transport vesicles, PlantMol. Biol. 38, 49–76.

Romoscanu, A.I., and Mezzenga, R. 2005. Cross-linking and rheological characterization of adsorbed protein lay-ers at the oil–water interface, Langmuir 21(21), 9689–9697.

Rosenbluh, J., Singh, S.K., Gafni, Y., Graessmann, A., and Loyter, A. 2004. Non-endocytic penetration of core his-tones into petunia protoplasts and cultured cell: A novel mechanism for the introduction of macromoleculesinto plant cells, Biochima et Biophysica Acta- Membranes, 1664, 230–240.

Schultz, S., Wagner, G., Urban, K., and Ulrich, J. 2004. High-pressure homogenization as a process for emulsionformation, Chem. Eng. Technol. 27(4), 361–368.

Schwarz, K., Huang, S.W., German, J.B., Tiersch, B., Hartmann, J., and Frankel, E.N. 2000. Activities of antioxi-dants are affected by colloidal properties of oil-in-water and water-in-oil emulsions and bulk oils, J. Agric. FoodChem. 48, 4874–4882.

Sela, Y., Magdessi, S., and Garti, N. 1995. Release of markers from the inner phase of w/o/w emulsions stabilisedby silicone-based polymeric surfactants, J. Control. Release 33, 1–12.

Seo, M., Nie, Z.H., Xu, S.Q., Mok, M., Lewis, P.C., Graham, R., and Kumacheva, E. 2005. Continuous microflu-idic reactors for polymer particles, Langmuir 21, 11614–11622.

Shamil, S., Wyeth, L.J., and Kilcast, D. 1991. Flavour release and perception in reduced fat foods. Food Qual.Pref. 3(1), 51–60.

Shark, J.L. 1977. Encapsulation process utilizing microorganisms and products produced thereby, US Patent4001480.

Shima, M., Tanaka, M., Kimura, Y., Adachi, S., and Matsuno, R. 2005. Enhancement in transport of a hydrophilicmarker through intestinal epithelial cell (Caco-2) monolayer by w/o/w multiple emulsion containing C8TG,Food Hydrocolloids 19, 321–328.

Silvestre, M.P.C., Chaiyasit, W., Brannan, R.G., McClements, D.J., and Decker, E.A. 2000. Ability of surfactanthead group size to alter lipid and antioxidant oxidation in oil-in-water emulsions, J. Agric. Food Chem. 48,2057–2061.

Simovic, S., and Prestidge, C.A. 2004. Nanoparticles of varying hydrophobicity at the emulsion droplet–waterinterface: Adsorption and coalescence stability, Langmuir 20(19), 8357–8365.

Sugiura, S., Nakajima, M., Yamamoto, K., Iwamoto, S., Oda, T., Satake, M., and Seki, M. 2004. Preparation char-acteristics of water-in-oil-in-water multiple emulsions using microchannel emulsification, J. Colloid InterfaceSci. 270, 221–228.

Surh, J., Decker, E.A., and McClements, D.J. 2006. Properties and stability of oil-in-water emulsions stabilized byfish gelatin, Food Hydrocolloids 20, 596–606.

Suzuki, K., Fujiki, I., and Hagura, Y. 1998. Preparation of corn oil/water and water/corn oil emulsions using PTFEmembranes, Food Sci. Technol. Int. (Tokyo) 4(2), 164.

Tai, S.S.K., Chen, M.C.M., Peng, C.C., and Tzen, J.T.C. 2002. Gene family of oleosin isoforms in sesame seed oilbodies and their structural stabilisation to reconstituted oil bodies, Biosci. Biotechnol. Biochem. 66,2146–2153.

Takahashi, Y., Yoshida, T., and Takashi, T. 1994. Process for producing a w/o/w-type multiple emulsion for medi-cines, cosmetics, etc., US Patent 4985173.

Emulsions as Delivery Systems in Foods 79

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 79

Page 89: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Taylor, A.J. 1996. Volatile flavour release from foods during eating, Crit. Rev. Food Sci. Nutr. 36(8), 765–784.Taylor, P. 1998. Ostwald ripening in emulsions, Adv. Colloid Interface Sci. 75(2), 107–163.Tcholakova, S., Denkov, N.D., Sidzhakova, D., Ivanov, I.B., and Campbell, B. 2005. Effects of electrolyte concen-

tration and pH on the coalescence stability of beta-lactoglobulin emulsions: Experiment and interpretation,Langmuir 21(11), 4842–4855.

Thill-Francis, L. 1993. Stable double emulsions containing finely divided particles. WO93/00007.Trautwein, E.A., Duchateau, G.S.M.J.E., Lin, Y., Mel’nikov, S.M., Molhuizen, H.O.F., and Ntanios, F.Y.

2003. Proposed mechanisms of cholesterol-lowering action of plant sterols, Eur. J. Lipid. Sci. Technol. 105,171–185.

Tzen, J.T.C., and Huang, A.H.C. 1992. Surface structure and properties of plant seed oil bodies, J. Cell Biol. 117,327–335.

Tzen, J.T.C., Lie, G.C., and Huang, A.H.C. 1992. Characterisation of the charged components and their topologyon the surface of plant seed oil bodies, J. Biol. Chem. 267, 15626–15634.

Tzen, T.J.C., Chuang, R.L.C., Chen, J.C.F., and Wu, L.S.H. 1998. Coexistence of both oleosin isoforms on the sur-face of seed oil bodies and their individual stabilisation to the organelles, J. Biochem. 123, 319–324.

Valentova, H., and Pokorny, J. 1998. Effect of edible oils and oil emulsions on the perception of basic tastes,Nahrung 42(6), 406–408.

Vanapalli, S.A., and Coupland, J.N. 2001. Emulsions under shear: The formation and properties of partially coa-lesced lipid structures, Food Hydrocolloids 15(4–6), 507–512.

van der Graaf, S., Schroen, C.G.P.H., and Boom, R.M. 2005. Preparation of double emulsions by membrane emul-sification: A review, J. Memb. Sci. 251, 7–15.

van Rijn, C.J.M. 2004. Nano and micro engineered membrane technology. Elsevier, Amsterdam.van Rooijen, G.J.H, and Moloney, M.M. 1995. Plant seed oil bodies as carriers for foreign proteins, Bio/Technol-

ogy 13, 72–77.van Ruth, S.M., O’Connor, C.H., and Delahunty, C.H. 2000. Relationships between temporal release of

aroma compounds in a model mouth system and their physicochemical characteristics, Food Chem. 71,393–399.

Villiere, A., Viau, M., Bronnec, I., Moreau, N., and Genot, C. 2005. Oxidative stability of bovine serumalbumin- and sodium caseinate–stabilized emulsions depends on metal availability, J. Agric. Food Chem. 53,1514–1520.

Wedzicha, B.L. 1988. “Distribution of low-molecular-weight food additives”, in Dickinson, E., and Stainsby,G. (Eds) Advances in food emulsions and foams. Elsevier Applied Science Publishers, London. 329–371.

Wedzicha, B.L., and Couet, C. 1995. Kinetics of transport of benzoic acid in emulsions, Food Chem. 55, 1–6.Weel, K.G.C., Boelrijk, A.E.M., Burger, J.J., Jacobs, M.A., Gruppen, H., Voragen, A.G.J., and Smit, G. 2004a

Effect of emulsion properties on release of esters under static headspace, in vivo and artificial throat conditionsin relation to sensory intensity, J. Agric. Food Chem. 52, 6572–6577.

Weel, K.G.C., Boelrijk, A.E.M., Burger, J.J., Verschueren, M., Gruppen, H., Voragen, A.G.J., and Smit, G. 2004b.New Device to simulate swallowing and in vivo aroma release in the throat from liquid and semiliquid food sys-tems, J. Agric. Food Chem. 52, 6564–6571.

Wen, L., and Papadopoulos, K.D. 2000. Visualization of water transport in w1/o/w2 emulsions, Colloids Surf.174, 159–167.

Wen, L., and Papadopoulos, K.D. 2001. Effects of osmotic pressure on water transport in w1/o/w2 emulsions,J. Colloid Interface Sci. 235, 398–404.

Wendin, K, Ellekjaer, M.R., and Solheim, R. 1999. Fat content and homogenization effects of flavour and textureof mayonnaise with added aroma, Food Sci. Technol. (Lebensmittel Wissenschaft and Technologie) 32,377–383.

Wilson, C.E., and Brown, W.E. 1997. Influence of food matrix structure and oral breakdown during mastication ontemporal perception of flavour, J. Sens. Stud. 12(1), 69–86.

Wright, K.M., and Hills, B.P. 2003. Modelling flavour release from a chewed bolus in the mouth: Part II. Therelease kinetics, Int. J. Food Sci. Technol. 38, 361–368.

Wright, K.M., Sprunt, J., Smith, A.C., and Hills, B.P. 2003. Modelling flavour release from a chewed bolus in themouth: Part I. Mastication, Int. J. Food Sci. Technol. 38, 351–360.

Wu, S.S.H., Platt, K.A., Ratnayake, C., Wang, T.W., Ting, J.T.L., and Huang, A.H.C. 1997. Isolation and charac-terization of neutral-lipid-containing organelles and globuli-filled plastids from Brassica napus tapetum, Proc.Natl. Acad. Sci. USA 94, 12711–12716.

80 Chapter 3

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 80

Page 90: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Yamamoto, Y, and Nakabayashi, M. 1999. Enhancing effect of an oil phase on the sensory intensity of salt taste ofNaCl in oil/water emulsions, J. Food Texture Stud. 30, 581–590.

Ye, A.Q., and Singh, H. 2001. Interfacial composition and stability of sodium caseinate emulsions as influencedby calcium ions, Food Hydrocolloids 15(2) 195–207.

Yoshida, K., Sekine, T., Matsuzaki, F., Yanaki, T., and Yamaguchi, M. 1999. Stability of vitamin A in oil-in-water-in-oil-type multiple emulsions, J. Am. Oil. Chem. Soc. 76(2), 195–200.

Emulsions as Delivery Systems in Foods 81

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_03-041-082 r1.qxd� 3/29/07 2:42 PM Page 81

Page 91: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

4 Applications of Probiotic Encapsulationin Dairy Products

Ming-Ju Chen and Kun-Nan Chen

Introduction

Most probiotics in the food supply are used in fermented milks and dairy products; in fact,dairy products are the major carriers of probiotics available today. Probiotics can be definedas living microbial supplements which can improve the balance of intestinal microorgan-isms (Fuller 1992). This definition was broadened by Havenaar and Huis in’t Veld (1992) toa “mono- or mixed-culture of live microorganisms which benefit man or animals byimproving the properties of the indigenous microflora.” The probiotic effect has beenattributed to the production of acid and/or bacteriocins, competition with pathogens andenhancement of the immune system. Claimed benefits include controlling serum choles-terol levels, preventing intestinal infection, improving lactose utilization in persons who arelactose intolerant, and possessing anticarcinogenic activity.

Good probiotic viability and activity are considered essential for optimal functionality(Mattila-Sandholm et al. 2002; Champagne and Gardner 2005). Furthermore, the ability ofmicroorganisms to survive and multiply in the host strongly influences their probiotic ben-efits. The bacteria in a product should remain metabolically stable and active, survivingpassage through the upper digestive tract in large numbers sufficient enough to producebeneficial effects when in the host intestines (Gilliland 1989). Adequate numbers of viablecells, namely the “therapeutic minimum,” need to be regularly consumed in order to trans-fer the probiotic effect to consumers. Survival of these bacteria during the product shelf lifeuntil being consumed is therefore an important consideration. Suggested beneficial mini-mum level for probiotics in yogurt is 106 cfu/mL (Robinson 1987; Kurman and Rasic 1991)or the daily intake should be about 108 cfu/mL. Earlier studies indicated that some strainsof probiotics, especially Bifidobacterium spp., lack the ability to survive gastrointestinalconditions (Berrada et al. 1991; Lankaputhra and Shah 1995).

Other studies have also reported low viability of probiotics in dairy products such asyogurt and frozen dairy desserts (Iwana et al. 1993; Shah and Lankaputhra 1997; Schillinger1999) due to the concentration of lactic acid and acetic acid (Samona and Robinson 1994),low pH (Martin and Chou 1992; Klaver et al. 1993), the presence of hydrogen peroxide(Lankaputhra and Shah 1996), and the oxygen content (Dave and Shah 1997). Methods forprotecting probiotics including selection of acid-resistant strains, control of over-acidificationof dairy products, and the addition of cysteine or an oxygen scavenger such as ascorbic acid(Dave and Shah 1997) have been proposed by various studies (Dave and Shah 1998;Adhikari et al. 2000; Krasaekoopt et al. 2003). Encapsulation has been investigated forimproving the viability of microorganisms in both dairy products and the intestinal tract(Prevost and Divies 1988; Lacroix et al. 1990; Champagne et al. 1992).

Encapsulation is a physicochemical or mechanical process in which particles containingactive ingredients are covered by a layer of another material, providing protection and

83

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 83

Encapsulation and Controlled Release: Technologies in Food SystemsEdited by Jamileh M. Lakkis

Copyright © 2007 by Blackwell Publishing

Page 92: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

controlled release of the primary ingredients as well as making the ingredients more conve-nient to work with (Thies 1996). The selection of different types of coating materials usu-ally depends on the functional properties of the microcapsules and the coating process used(Hegenbart 1993). For dairy and food applications, probiotic encapsulation in food grade,porous matrices has been most widely used (Champagne et al. 1994). Spherical entrapmentbeads are produced using spray-drying, extrusion, or emulsification techniques.

The following sections describe the techniques, effects, and applications of probioticencapsulation in dairy products. Published data on new techniques of probiotic encapsulationwith survival of probiotic capsules in dairy products and in the intestines are also discussed.

Techniques for Probiotic Encapsulation

Encapsulation of probiotics for use in dairy products or biomass production can beachieved in two ways: physicomechanically and chemically. The probiotics are encapsu-lated in the gas phase during physicomechanical procedures including spray-drying tech-nique whereas, probiotic encapsulation is performed in liquid by thermal or ionotropicgelation of the droplets including extrusion and emulsion techniques. All three techniqueshave been proven to increase the survival of probiotics by up to 90% (Kebary et al. 1998).

Spray-Drying Technique

Among the well-known microencapsulation methods, spray-drying is most widely used inthe chemical, pharmaceutical, and food industries due to its inherent attributes such as highproduction rates and relatively low operational cost (Gibbs et al. 1999). The principle ofspray-drying technique involves dissolving a polymer, in the continuous phase, which sur-rounds the core material particles (encapsulant such as probiotics) inside the sprayeddroplets. The drying process causes this solution to shrink into a pure polymer envelopeenclosing the core material. The resulting capsules are obtained as free-flowing dry powder.

Table 4.1 shows probiotic encapsulation using the spray-drying technique in dairy prod-ucts and biomass production. Various carrier matrices including starch (O’Riordan et al.2001; Lian et al. 2003), gelatin (Lian et al. 2002, 2003), gum arabic (Lian et al. 2002,2003), skim milk (Gardiner et al. 2002; Lian et al. 2003; Ananta et al. 2005), celluloseacetate phthalate (CAP; Favaro-Trindade and Grosso 2002), whey protein (Picot andLacroix 2003, 2004), gum acacia (Desmond et al. 2002), and prebiotics (Ananta et al.2005) have been reported and applied to various dairy products including yogurt (Picot andLacroix 2004), dry dairy beverages (O’Riordan et al. 2001), and cheddar cheese (Gardineret al. 2002). However, exposure to high air temperatures required to facilitate water evapo-ration during the passage of the bacteria in the spray-drying chamber exerts a negativeimpact on their viability and hampers their activity in the spray-dried product (Ananta et al.2005). The survival of encapsulated microorganisms produced by spray-drying will be dis-cussed in more detail in a later section.

Extrusion Technique

Extrusion is the simplest and most common technique used to produce probiotic capsuleswith hydrocolloids (King 1995). The principle of this technique simply involves preparinga hydrocolloid solution, adding the probiotic ingredient to the solution and dripping the cell

84 Chapter 4

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 84

Page 93: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

suspension through a syringe needle or nozzle spray machine in the form of droplets whichare allowed to free-fall into a hardening solution or setting bath. This extrusion techniqueproduces large particles with uniform particle size.

Table 4.2 shows probiotic encapsulation using extrusion techniques in dairy products andbiomass production. The common polymer used to produce probiotic encapsulation matrixby extrusion technique is alginate (Krasaekoopt et al. 2003). Other food-grade encapsulationmaterials like gellan gum and xanthan gum (Sun and Griffiths 2000; McMaster et al. 2005)have also been proposed for encapsulating probiotics. Many dairy products including yogurt(Prevost and Divies 1987; Sun and Griffiths 2000; Krasaekoopt et al. 2004; Iyer and Kailas-apathy 2005), cheese (Prevost and Divies 1988), and cream (Prevost and Divies 1992) carryencapsulated probiotics produced by extrusion. One of the major advantages of this methodis that the viscosity of the fluid does not limit capsule generation (Prüße et al. 2000). Fur-thermore, the biological matter can be treated at lower temperatures.

Emulsion Technique

The emulsion technique has successfully been used to encapsulate lactic acid bacteria inboth batch (Lacroix et al. 1990) and continuous fermentation processes (Audet et al. 1992).

Applications of Probiotic Encapsulation in Dairy Products 85

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Table 4.1. Probiotic encapsulation by spray-drying in dairy products and biomass production

Inlet and outlet Probiotics Carrier matrix (%) temperature Application Reference

Bifidobacterium PL1 10% starch Inlet: 60–140°C Dry beverage O’Riordan Outlet: 45°C et al. (2001)

L. paracasei 20% reconstituted Inlet: 175°C Cheddar Gardinerskim milk Outlet: 68°C cheese et al. (2002)

L. acidophilus Cellulose acetate Inlet: 130°C Favaro-Trindadephthalate and Grosso (2002)

B. lactis Outlet: 75°C

L. paracasei Gum acacia Inlet: 170°C Desmond Outlet: 95–105°C et al. (2002)

B. longum 30% gelatin Inlet: 100°C Lian et al. (2002, 2003)

B. infantis 35% soluble starch Outlet: 50–60°C35% gum arabic15% skim milk

B. breve 85% milk fat/5–15% Inlet: 160°C Picot and Lacroixwhey protein (2003)

B. longum 10% whey protein Outlet: 80°C

B. breve 85% milk fat/5–15% Inlet: 160°C Yogurt Picot and Lacroixwhey protein (2004)

B. longum Outlet: 80°C

L. rhamnosus GG 20% skim milk/ Outlet: 70–100°C Ananta et al. oligofructose or (2005)polydextrose

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 85

Page 94: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Pro

biot

ics

Sup

port

ing

mat

eria

l (%

)H

arde

ning

bat

hS

peci

al tr

eatm

ent

Apl

icat

ion

Ref

eren

ce

L. d

elbr

ueck

ii

1.85

% s

odiu

m a

lgin

ate

1.5

M c

alci

um c

hlor

ide

No

Che

ese

Pre

vost

et a

l. (1

987)

ssp.

bulg

aric

usSt

rept

ococ

cus

ther

mop

hilu

sL

. pla

ntar

um1.

5% s

odiu

m a

lgin

ate

0.05

M c

alci

um c

hlor

ide

No

Bio

mas

s pr

oduc

tion

Kea

rney

et a

l. (1

990)

L. l

acti

s2%

sod

ium

alg

inat

e0.

1M

cal

cium

chl

orid

eN

oC

ream

Pre

vost

and

Div

ies

(199

2)L

. cas

ei0.

6% s

odiu

m a

lgin

ate

1.0

M c

alci

um c

hlor

ide

Chi

tosa

nY

oo e

t al.

(199

6)1.

0M

bar

ium

chl

orid

eL

acto

cocc

us la

ctis

2%

sod

ium

alg

inat

e0.

05M

cal

cium

chl

orid

eC

hito

san

Bio

mas

s pr

oduc

tion

Zho

u et

al.

(199

8)ss

p. c

rem

oris

B. b

ifidu

m2%

sod

ium

alg

inat

e0.

2M

cal

cium

chl

orid

ePo

ly-L

-lys

ine

Cui

et a

l. (2

000)

chit

osan

B. l

ongu

m2–

4% s

odiu

m a

lgin

ate

0.1

M c

alci

um c

hlor

ide

No

Lee

and

Heo

(20

00)

B. i

nfan

tis

0.75

% g

ella

n/1%

0.

1M

cal

cium

chl

orid

eN

oY

ogur

tS

un a

nd G

riffi

ths

(200

0)xa

ntha

n gu

mL

. aci

doph

ilus

0.75

–2%

sod

ium

alg

inat

e0.

1–1.

0M

cal

cium

chl

orid

eN

oC

hand

ram

ouli

et a

l. 20

04L

. aci

doph

ilus

2% s

odiu

m a

lgin

ate

0.05

M c

alci

um c

hlor

ide

Chi

tosa

nY

ogur

tK

rasa

ekoo

pt e

t al.

(200

4)L

. cas

eiB

. bifi

dum

L. a

cido

phil

us2%

sod

ium

alg

inat

e0.

05M

cal

cium

chl

orid

eS

odiu

m a

lgin

ate

Kra

saek

oopt

et a

l. (2

004)

poly

-L-l

ysin

e ch

itos

anL

. cas

eiB

. bifi

dum

L. b

ulga

ricu

s2%

sod

ium

alg

inat

e0.

5M

cal

cium

chl

orid

eC

hito

san

Lee

et a

l. (2

004)

B. l

acti

s0.

75%

gel

lan/

1%0.

1M

cal

cium

chl

orid

eN

oA

mas

i M

cMas

ter

et a

l. (2

005)

xant

han

gum

(sou

r m

ilk

prod

ucts

)L

. aci

doph

ilus

Sod

ium

alg

inat

e C

alci

um c

hlor

ide

Hi-

mai

ze s

tarc

hY

ogur

tIy

er a

nd K

aila

sapa

thy

poly

-L-l

ysin

e(2

005)

Chi

tosa

nR

afti

line

®/R

afti

lose

®

Tab

le 4

.2.

Pro

bio

tic

enca

psu

lati

on

by

extr

usi

on

tec

hn

iqu

e in

dai

ry p

rod

uct

s an

d b

iom

ass

pro

du

ctio

n

86

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 86

Page 95: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Applications of Probiotic Encapsulation in Dairy Products 87

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

The principle of these techniques is based on the relationship between the discontinuousand the continuous phases. A small volume of the cell-polymer suspension (i.e., the discon-tinuous phase) is added to a large volume of vegetable oil (i.e., the continuous phase). Themixture is then homogenized to form a water-in-oil emulsion. Once the water-in-oil emul-sion is formed, the water-soluble polymer must be insolubilized to form tiny gel particleswithin the oil phase. The insolubilization method of choice depends on the type of support-ing material used. The beads are harvested later by filtration. For encapsulation in an emul-sion, an emulsifier and a surfactant are needed. Emulsifiers such as Tween 80 can break upwater and oil emulsions as well as prevent spheres from coalescing before breaking up theemulsion. A surfactant such as sodium lauryl sulfate (SLS) is used to lower the surface ten-sion in the coating matrix in order to reduce the size of the spheres.

Table 4.3 shows probiotic encapsulation using emulsion technique for dairy productsand biomass production.

Various supporting materials have been used to encapsulate probiotics by the emulsionmethod including alginate (Sheu and Marshall 1993; Sultana et al. 2000; Truelstrup et al.2002; Song et al. 2003; Shah and Ravla 2004), �-carrageenan (Dinakar and Mistry 1994;Adhikari et al. 2000, 2003), CAP (Modler and Villa-Garcia 1993), chitosan, and gelatin(Peniche et al. 2003). This type of probiotic beads have been successfully applied to yogurt(Adhikari et al. 2000; Sultana et al. 2000; Adhikari et al. 2003), cheddar cheese (Dinakarand Mistry 1994), milk (Truelstrup et al. 2002), and ice cream (Sheu and Marshall 1993;Shah and Ravla 2004). This technique provides both encapsulated and entrapped corematerials and is easy to scale up for large-scale production.

Advantages and Disadvantages of Various Probiotics Encapsulation Techniques

A comparison of different encapsulation techniques is presented in Table 4.4. Both spray-drying and extrusion (Krasaekoopt et al. 2003) are relatively simple techniques. Con-versely, the emulsion technique based on the relationship between the discontinuous andcontinuous phases is more complex. Although both spray-drying and emulsion techniquesare easier to scale up, Picot and Lacroix (2003) used an emulsification/spray technology toproduce microcapsules containing micronized skim milk powder dispersed in milk fatdroplets surrounded by an insoluble whey protein film. This technique is claimed to besimple and can be easily scaled up for microencapsulation of dry probiotic cultures.

Encapsulation of probiotics using natural biopolymers such as calcium alginate, �-carrageenan, and gellan gum is currently applicable only on a laboratory scale (Doleyresand Lacroix 2005). The high viscosity of these coating materials appears to hamper the effi-ciency of encapsulation (Krasaekoopt et al. 2003). Scale-up production of encapsulatedprobiotics via extrusion is more difficult due to the slow formation of beads (Krasaekooptet al. 2003).

The sizes of beads formed from spray-drying and emulsion are smaller than those pro-duced by the extrusion method. With the extrusion method, the size of the capsules is highlydependent on the viscosity of sodium alginate solution, the extruder orifice diameter, and thedistance between the syringe and the calcium chloride collecting solution (Smidsrod andSkjak-Braek 1990). A higher concentration of sodium alginate results in significantly highviscosity which leads to large particle sizes. Spherical beads, prepared by extrusion, areapproximately 2–3 mm in diameter, while those made by emulsification techniques have

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 87

Page 96: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Con

cent

rati

on o

f P

robi

otic

ssu

ppor

ting

mat

eria

l (%

)C

onti

nuou

s ph

ase

Spe

cial

trea

tmen

tA

ppli

cati

onR

efer

ence

L. b

ulga

ricu

s3%

�-c

arra

geen

an/

Soy

oil

No

Bio

mas

s pr

oduc

tion

Aud

et e

t al.(

1989

)lo

cust

bea

n gu

mS.

ther

mop

hilu

sL

c. la

ctis

L. b

ulga

ricu

s3%

alg

inat

eV

eget

able

oil

No

Ice

mil

kS

heu

and

Mar

shal

l (1

993)

B. b

ifidu

m2%

�-c

arra

geen

anV

eget

able

oil

No

Che

ddar

Din

akar

and

Mis

try

(199

4)B

. lon

gum

3% �

-car

rage

enan

/S

oy o

ilN

oM

aitr

ot e

t al.

(199

7)lo

cust

bea

n gu

mB

. lon

gum

2% �

-car

rage

enan

Veg

etab

le o

ilN

oS

et y

ogur

tA

dhik

ari e

t al.

(200

0)L

. aci

doph

ilus

2% a

lgin

ate

Veg

etab

le o

ilH

i-m

aize

sta

rch

Yog

urt

Sul

tana

et a

l. (2

000)

Bifi

doba

cter

ium

spp

.B

. ado

lesc

enti

s3%

alg

inat

eV

eget

able

oil

No

Mil

kT

ruel

stru

p et

al.

(200

2)B

. bre

veB

. lac

tis

B. l

ongu

mL

. cas

ei1%

alg

inat

eC

orn

sala

d oi

lM

icro

poro

us

Son

g et

al.

(200

3)G

lass

Mem

bran

eB

. lon

gum

2% �

-car

rage

ena

Veg

etab

le o

ilN

oS

tirr

ed y

ogur

tA

dhik

ari e

t al.

(200

3)L

. bul

gari

cus

Art

ifici

al o

ilS

esam

e oi

l/ve

geta

ble

oil

No

Hou

et a

l. (2

003)

L. a

cido

phil

us3%

alg

inat

eV

eget

able

oil

No

Froz

en d

esse

rtS

hah

and

Rav

la

(200

4)B

ifido

bact

eriu

m s

pp.

Tab

le 4

.3.

Pro

bio

tic

enca

psu

lati

on

by

emu

lsio

n in

dai

ry p

rod

uct

s an

d b

iom

ass

pro

du

ctio

n

88

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 88

Page 97: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Applications of Probiotic Encapsulation in Dairy Products 89

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Spray-drying Extrusion Emulsion

Scale-up Easy More difficult EasyEncapsulating process Simple Simple More difficultVariety of coating materials Many Few ManyShape and size Uniform and small Uniform and large Non-uniform and smallSurvival of microorganisms Dependent on the High High

carriers used and temperature

Table 4.4. Advantages and disadvantages of encapsulation methods

bead diameters ranging from 25 µm to 2 mm. The actual bead size can be controlled by vary-ing the speed of agitation and it also depends on the type of emulsifier used.

Probiotics encapsulated via spray-drying technique show lower survival rates duringdrying and lower stability during storage (Ananta et al. 2005) than those produced by emul-sion and extrusion, a result of their exposure to high air temperatures required to facilitatewater evaporation.

Effects of Encapsulation on Probiotic Survival

This section summarizes the factors affecting the survival of encapsulated probiotics.

Effect of Carrier Matrix on Probiotic Survival

Alginate

Alginate is a linear heteropolysaccharide of D-mannuronic and L-guluronic acids extractedfrom various species of algae. The functional properties of alginate as a supporting materialare strongly associated with the composition and sequence of L-guluronic and D-mannuronicacids. Divalent cations such as Ca2� preferentially bind to the polymer of L-guluronic acid(Krasaekoopt et al. 2003). Calcium alginate is preferred over all other supporting materialsfor encapsulating probiotics due to its simplicity, non-toxicity, biocompatibility, and lowcost (Sheu and Marshall 1993; Krasaekoopt et al. 2003). Solubilization of alginate gels bysequestering calcium ions and releasing entrapped cells within the human intestines isanother advantage. The concentrations of sodium alginate and calcium chloride used to formthe beads vary and range between 1 and 3% alginate with 0.05~1.5 M CaCl2 (Prevost et al.1988; Kearney et al. 1990; Cui et al. 2000; Chandramouli et al. 2004; Krasaekoopt et al.2004). A very low level of alginate (0.6% alginate with 0.3 M CaCl2) was used to form a gelby Jankowski et al. (1997). Nevertheless, alginate beads formed using low-viscosity alginatesolutions lack mechanical and physical stability (Smidsrod and Skjak-Braek 1990; Peironeet al. 1998).

The use of alginate, however, is limited due to its low physical stability in the presenceof anti-gelling cations such as sodium and magnesium ions (Lee et al. 2004) or chelatingagents such as phosphate (Krasaekoopt et al. 2006). The latter share an affinity for calcium,thus destabilizing the alginate gel (Smidsrod and Skjak-Braek 1990). Furthermore, underlow pH conditions, cross-linked alginate matrices can undergo degradation of the alginatemolecule and subsequent reduction in its molecular weight causing faster release ofentrapped active ingredients (Gombotz and Wee 1998).

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 89

Page 98: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Specially Treated Alginates

Coating alginate beads with polycations and cross-linking with barium ions (Ba2+) insteadof calcium ions (Ca2�) have been suggested for improving the mechanical stability of algi-nate microcapsules (Thu et al. 1996; Gaumann et al. 2001; Koch et al. 2003; Krasaekooptet al. 2006).

Polycation-coated alginates: Coating alginate beads with polycations such as chitosan andpoly-L-lysine has been studied extensively for encapsulating probiotics (Cui et al. 2000; Canhet al. 2004; Krasaekoopt et al. 2004; Lee et al. 2004; Krasaekoopt et al. 2006). Chitosan-coated alginate capsules were produced by dropping an alginate solution into a mixture ofcalcium chloride and chitosan solution (Krasaekoopt et al. 2004). Since chitosan (poly-(2-amino-2-deoxy-β-D-glucopyranose)) is positively charged, it forms polyelectrolyte com-plexes with alginates resulting in the formation of polyanionic polymer membranes whichare stable in the presence of calcium chelators or antigelling agents (Smidsrod and Skjak-Braek 1990). Zhou et al. (1998) reported that suspending alginate capsules in a low molecu-lar weight chitosan solution reduced cell release by 40%. On the contrary, Lee et al. (2004)indicated that high molecular weight chitosan coating resulted in the highest survival forLactobacillus bulgaricus in simulated gastric juice and better stability at 22°C. Krasaekooptet al. (2006) studied the survival of probiotics encapsulated in chitosan-coated alginatebeads in yogurt and found that the survival of the encapsulated probiotic bacteria was higherthan free cells by approximately 1 log cycle. Lee et al. (2004) indicated that microencapsula-tion of freeze-dried L. bulgaricus by chitosan-coated calcium alginate greatly improved theviability of probiotics in simulated gastric and intestinal juices.

Alginate poly-L-lysine microcapsules’ high biocompatibility and strength make themgood candidates for food applications (Champagne et al. 1992; Larisch et al. 1994;Krasaekoopt et al. 2004). Bifidobacteria loaded onto alginate poly-L-lysine microparticlesdisplayed enhanced survival of the probiotic bacteria during storage at 4°C (Cui et al.2000). Krasaekoopt et al. (2004) compared the survival of microencapsulated probioticsusing different coating materials and found that chitosan-coated alginate beads provide bet-ter protection for Lactobacillus acidophilus and Lactobacillus casei than did poly-L-lysine-coated alginated beads in 0.6% bile salts.

Modification of alginates by succinylation (increased matrix anionic charge) or byacetylation (increased matrix hydrophobicity) has also been suggested for stabilizingencapsulated probiotics in acidic conditions (Le-Tien et al. 2004).

Prebiotics-Coated Alginates

Prebiotics are non-digestible food ingredients that beneficially affect the host by selectivelystimulating the growth and/or activity of one or a limited number of bacteria in the colon(Gibson and Roberfroid 1995). Several studies (Bielecka et al. 2002; Chen et al. 2005a) haveconfirmed that incorporation of prebiotics and calcium alginate as coating materialsprovides better protection for probiotics in food and eventually the intestinal tract. Chen etal. (2005a) incorporated prebiotics as coating materials for probiotic microencapsulationand demonstrated that the addition of fructooligosaccharides (FSO), isomaltooligosaccha-rides (IMO), and peptides in the walls of probiotic microcapsules provided improved protec-tion for the active organisms. Probiotic counts remained at 106�107 cfu g-1 formicrocapsules stored for one month and were then subjected to a simulated gastric fluid test

90 Chapter 4

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 90

Page 99: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

and a bile salt test. Iyer and Kailasapathy (2005) reported that addition of Hi-maize starch tocapsules containing Lactobacillus spp. provided maximum protection under acidic condi-tion. Moreover, by further coating the capsules with chitosan, the survival rate was signifi-cantly increased under acidic and bile salt conditions.

Gellan Gum and Xanthan Gum

Gellan gum, a microbial polysaccharide derived from Pseudomonas elodea, is constitutedof a repeating unit of four monosaccharide molecules (glucose, glucuronic acid, glucose,and rhamnose). The combination of gellan and xanthan gums to form bead is not only acidresistant but also is stabilized by calcium ions (Norton and Lacroix 1990), which can pro-tect cells from acid injury. Sun and Griffiths (2000) encapsulated Bifidobacterium spp. withgellan-xanthan gum as the coating material and reported that gellan-xanthan beads werehighly acid-stable. At pH 2.5, the viable count of encapsulated probiotics decreased by only0.67log in 30 min. while the survival of free cells dropped from 1.23 � 109 cfu mL-1 to anundetectable level in the same period.

�-Carrageenan and Locust Bean Gum

�-Carrageenan is a natural polymer extracted from Irish moss and is commonly used in thefood industry. Formation of a gel using this polymer occurs because of temperaturechanges. The cell suspension is mixed with the heat-sterilized polymer solution at 40–50°Cand gelation occurs on cooling to room temperature. The microcapsules are stabilized byadding potassium ions. The encapsulation of Bifidobacterium bifidum in �-carrageenanbeads maintained the cell viability for as long as 24 weeks of cheddar cheese ripening, withno negative effects on the texture, appearance, or flavor (Dinakar and Mistry 1994). How-ever, �-carrageenan produces brittle gels which are not able to withstand stresses of inter-nal bacterial growth and shear during agitation (Audet et al. 1988).

The combination of �-carrageenan with locust bean gum, which produces more flexiblegels due to specific interactions between the two gums, was recently used to encapsulateprobiotics. The probiotics suspension was mixed with a �-carrageenan-locust bean gumsolution, and the cell-polymer dispersion was then rapidly poured into vegetable oil withagitation. The beads were washed and soaked in sterile KCl solution. Several researchers(Maitrot et al. 1997; Audet, et al. 1988) combined �-carrageenan with locust bean gum assupporting material for encapsulation of probiotics and found that this coating materialwas less sensitive to acid than alginate. Guoqiang et al. (1991) reported that a mixed gelmatrix of �-carrageenan and locust bean gum showed significant stability for 3 monthsin continuous lactic acid fermentation. However, the encapsulation of probiotics using�-carrageenan-locust bean gum as support material required potassium ions which candamage cells of the probiotic bacteria (such as Streptococcus thermophilus, L. bulgaricus,and Bifidobacterium longum) during fermentation (Audet et al. 1988). Furthermore, largeamount of potassium ions are not recommended in human diet.

Cellulose Acetate Phthalate

Cellulose acetate phthalate (CAP) is an enteric coating material used for controlling drugrelease in the intestines and thus has a well-established safety record for pharmaceutical

Applications of Probiotic Encapsulation in Dairy Products 91

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 91

Page 100: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

and dietary supplements applications. CAP is not soluble in water at pH values of less thanabout 5.8.

The advantage of CAP is that it is insoluble at acidic pH (less than 5) but is soluble at pHgreater than 6. Nevertheless, encapsulation of bifidobacteria by CAP was found to be inef-fective in preventing acid injury to bacteria in highly acidic yogurt (Modler and Villa-Garcia1993). Fávaro-Trindade and Grosso (2002) encapsulated Bifidobacterium lactis and Lacto-bacillus acidophilus using CAP as the coating material and concluded that CAP providedgood protection for both microorganisms in acid and bile solutions, conditions similar tothose of the intestine.

Chitosan

Chitosan is a cationic linear polysaccharide composed essentially of β(1-4)-linked glu-cosamine units together with some proportion of N-acetylglucosamine units. Droplets of achitosan solution suspended in an oil phase can be hardened by cross-linking with glu-taraldehyde (suspension cross-linking) via solvent evaporation or by the addition of poly-valent anions such as sodium tripolyphosphate (TPP) or citrate (ionotropic gelation). Thestirring rate, temperature, level of the gelling agent, concentration of the surfactant poly-mer, and the viscosities of the phases were reported to affect the size and morphology of theparticles (Peniche et al. 2003). However, inhibitory effects of chitosan on different types oflactic acid bacteria were reported by Groboillot et al. (1993).

Others

Lian et al. (2002) investigated the survival of bifidobacteria after spray-drying with differ-ent carrier matrices and indicated that the survival of microencapsulated bifidobacteriaafter spray-drying varied with strains and was mainly dependent on the carriers used. Inaddition, use of 10% gelation, gum arabic, and soluble starch resulted in the highest sur-vival of bifidobacteria. O’Riordan et al. (2001) used modified waxy maize starch to encap-sulate Bifidobacterium spp. with an average size of 5 µm by spray-drying and demonstratedthat maximum recovery yields were 30%. However, the starch-encapsulated Bifidobac-terium spp. showed no improvement in viability compared with the control-free cells whenexposed to acidic conditions or when added to yogurt. They concluded that the modifiedstarches might not be suitable for use as an encapsulating material for probiotic strains.Ananta et al. (2005) incorporated oligofructose-based or polydextrose-based skim milk ina carrier matrix which resulted in a high level of survival for Lactobacillus rhamnosus(LGG). A probiotic survival rate of 60% was achieved at an outlet temperature of 80°C.Desmond et al. (2002) studied the survival of Lactobacillus paracasei in a mixture ofreconstituted skim milk and gum acacia followed by spray-drying and found ten-foldgreater survival than in the control group. Hou et al. (2003) developed a technique to pro-tect lactic acid bacteria against simulated gastrointestinal conditions by encapsulating bac-terial cells within artificial sesame oil emulsions.

Effect of Spray-Drying on Probiotic Survival

The survivability of the encapsulated probiotics is most significantly influenced by the exe-cution of the spray-drying process as well as other factors. The survival of various lactic

92 Chapter 4

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 92

Page 101: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

cultures affected by spray-drying have been carried out by various investigators (O’Riordanet al. 2001; Lian et al. 2002; Lian et al. 2003; Picot and Lacroix 2003, 2004; Ananta et al.2005). Different polysaccharides were used as the matrix and the nozzle temperature of thespray dryer as well as the water activity of the microcapsules had a considerable impact onthe survival of probiotics.

The heat resistance of probiotic strains should be taken into account during the spray-dryencapsulation of sensitive microorganisms. Picot and Lacroix (2004) dispersed fresh cells ina heat-treated whey protein suspension followed by spray-drying and found a survival rateof 26% for Bifidobacterium breve after spray-drying and 1.4% for the more heat-sensitiveB. longum. Lian et al. (2002) studied the survival of bifidobacteria after spray-drying andfound that Bifidobacterium longum B6 exhibited the least sensitivity to spray-drying andshowed the highest survival of 82.6% after drying with skim milk.

The outlet-air temperature is another major parameter affecting probiotic survival afterspray-drying with lower temperatures resulting in higher survival rates (Favaro-Trindadeand Grosso 2002; Ananta et al. 2005; Chen et al. 2006). Lian et al. (2002) reported that Bifi-dobacterium spp. had the highest survival after drying at 50°C. Chen et al (2006) studiedthe viability of probiotics after spray-drying at outlet air temperatures of 60, 70, and 80°Cand found that the survival of L. acidophilus and B. longum decreased as the outlet-air tem-perature increased. However, the final total probiotic counts still remained above the rec-ommended therapeutic minimum (107 cfu/g) after spray-drying at various outlet airtemperatures. Gardiner et al. (2002) spray-dried L. paracasei NFBE 338 Rifr with 20%reconstituted skim milk at air inlet and outlet temperatures of 175°C and 68°C, respec-tively, and found a probiotic survival rate of 84.5%. Ananta et al. (2005) assessed probioticinjury sites in spray-drying by flow cytometry and found that the damage to cell mem-branes was the key reason for cell death. Higher outlet temperature used for spry-dryingresulted in more serious disintegration of membranes. On the other hand, inactivationcaused by increased outlet-air temperatures varied with the carrier used. Lian et al. (2002)indicated that using soluble starch as the carrier matrix significantly improved the probioticsurvival at a high outlet-air temperature, whereas skim milk showed the least effect.

Probiotic Survival in Dairy Products

An adequate number of viable cells, namely the “therapeutic minimum,” need to be con-sumed regularly in order for consumers to experience the probiotic effects. Encapsulationhas been investigated for improving the viability of the microorganisms in dairy productsincluding fermented milk (Adhikari et al., 2000; Sultana et al. 2000; Sun and Griffiths2000; Adhikari et al. 2003; Krasaekoopt et al. 2004; Picot and Lacroix 2004; Iyer andKailasapathy 2005), cheese (Dinakar and Mistry 1994; Desmond et al. 2002), and frozendesserts (Sheu and Marshall 1993; Shah and Ravla. 2004).

Cheese

Introducing encapsulated probiotics in cheese not only enhances the storage viabilityof probiotics but also improves the flavor of cheese. Research results (Dinakar and Mistry1994; Desmond et al. 2002) have reported that cheese containing encapsulated Bifidobac-terium spp. and L. paracasei did not differ from the control cheese in soluble protein,flavor, appearance, texture, and normal microflora. The viabilities of both encapsulated

Applications of Probiotic Encapsulation in Dairy Products 93

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 93

Page 102: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Bifidobacterium spp. and L. paracasei in cheese were maintained for at least 6 monthsand 3 months, respectively. In addition, acetic acid, a common metabolite of Bifidobac-terium spp. and not preferred in dairy products, was not detected during ripening.

Frozen Dairy Desserts

It is difficult to incorporate probiotic bacteria into frozen desserts due to the acidity of theproducts, high osmotic pressure, freeze injury, and exposure to air, as air is introduced dur-ing freezing of these products (Shah and Ravla 2004). Thus, the application of microencap-sulated probiotic bacteria to frozen dairy desserts may overcome these difficulties andcould produce useful markets and health benefits. Sheu et al. (1993) studied the survival ofculture bacteria in frozen desserts and indicated that the survival rate for encapsulatedL. bulgaricus in continuously frozen ice milk was approximated at 90% without a measura-ble effect on the sensory characteristics.

Yogurt

Incorporation of probiotics has been shown to enhance the therapeutic value of yogurt.However, the survival of probiotics in yogurt is low due to the prevailing low pH rangingfrom 4.2 to 4.6 (Kailaspathy and Rybka 1997). Many studies have documented the positiveeffects of encapsulation of probiotics and their survival in fermented dairy products(Adhikari et al. 2000; Sultana et al. 2000; Sun and Griffiths 2000; Adhikari et al. 2003;Krasaekoopt et al. 2004; Picot and Lacroix 2004; Iyer and Kailasapathy 2005). Of allencapsulation techniques tested, chitosan-coated alginate beads were reported to offer noenhanced protection for probiotics in yogurt stored at 4°C for 4 weeks (Krasekoopt et al.2006).

Probiotic Survival in Gastrointestinal Conditions

Encapsulated probiotics should survive passage through the upper digestive tract in largenumbers in order to ensure desired beneficial effects in the host intestines (Gilliland 1989).Various effects of encapsulation on the survival of bacteria under gastrointestinal condi-tions have been reported (Table 4.5). The survival of encapsulated cells is strongly depen-dent on the type and concentration of coating materials, bead size, initial cell numbers, andbacterial species. Most studies have proven the advantages of encapsulating probiotics overfree cells under in vitro gastric conditions, others did not find any additional protectionunder strongly acidic conditions (Rao et al. 1989; Sultana et al. 2000; O’Riordan et al.2001; Truelstrup et al. 2002).

Several coating materials including sodium alginate (Lee and Heo 2000; Chandramouliet al. 2004), sodium alginate with a polycation (Cui et al. 2000; Krasaekoopt et al. 2004;Lee et al. 2004; Iyer and Kailasapathy 2005), gellan/xanthan gum (Sun and Griffiths 2000;McMaster et al. 2005), artificial oil (Hou et al. 2003), gum arabic (Lian et al. 2003), andwhey protein (Picot and Lacroix 2004) showed good protection for encapsulating probi-otics under gastrointestinal conditions. Lee and Heo (2000) studied the survival ofB. longum immobilized in alginate beads in simulated gastric juices and bile salt solutionsand found that the death rate of the probiotics in the capsules decreased proportionally withan increase in the alginate concentration (1�3%), bead size (1�3 mm), and initial cell

94 Chapter 4

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 94

Page 103: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Sur

viva

l und

er g

astr

oint

esti

nal

Pro

biot

ics

Enc

apsu

lati

on m

etho

dC

oati

ng m

ater

ials

cond

itio

nsR

efer

ence

B. l

ongu

mE

xtru

sion

2–4%

sod

ium

alg

inat

eD

epen

ding

on

algi

nate

L

ee a

nd H

eo (

2000

)co

ncen

trat

ion

and

bead

siz

eB

. bifi

dum

Ext

rusi

on2%

sod

ium

alg

inat

e w

ith

Hig

her

than

106

cfu

mL

–1C

ui e

t al.

(200

0)po

ly-L

-lys

ine

or c

hito

san

L. a

cido

phil

usE

mul

sion

2% a

lgin

ate

wit

h H

i-m

aize

sta

rch

Hig

her

than

106

cfu

mL

–1S

ulta

na e

t al.

(200

0)B

ifido

bact

eriu

m s

pp.

B. i

nfan

tis

Ext

rusi

on0.

75%

gel

lan/

1% x

anth

an g

umH

ighe

r th

an 1

06cf

u m

L–1

Sun

and

Gri

ffith

s (2

000)

B. r

umin

anti

umS

pray

-dry

ing

10%

sta

rch

No

coun

ts d

etec

tabl

eO

’Rio

rdan

et a

l. (2

001)

B. a

dole

scen

tis

Em

ulsi

on3%

alg

inat

e8.

2–1.

0lo

g cf

u m

L–1

Tru

elst

rup

et a

l. (2

002)

B. b

reve

B. l

acti

sB

. lon

gum

L. b

ulga

ricu

sE

mul

sion

Art

ifici

al o

ilH

ighe

r th

an 1

06cf

u m

L–1

Hou

et a

l. (2

003)

B. l

ongu

mS

pray

-dry

ing

30%

gel

atin

87.1

5%L

ian

et a

l. (2

003)

35%

sol

uble

sta

rch

95.4

7%35

% g

um A

rabi

c93

.53%

15%

ski

m m

ilk

81.2

6%B

. inf

anti

sS

pray

-dry

ing

30%

gel

atin

92.7

3%L

ian

et a

l. (2

003)

35%

sol

uble

sta

rch

92.7

0%35

% g

um A

rabi

c89

.17%

15%

ski

m m

ilk

65.1

6%L

. cas

eiE

mul

sion

1% a

lgin

ate

wit

h m

icro

poro

us

Hig

her

than

106

cfu

mL

–1S

ong

et a

l. (2

003)

glas

s m

embr

ane

L. a

cido

phil

usE

xtru

sion

1.8%

sod

ium

alg

inat

e10

5 –10

6cf

u m

L–1

Cha

ndra

mou

li e

t al.

(200

4)L

. aci

doph

ilus

Ext

rusi

on2%

sod

ium

alg

inat

e w

ith

chit

osan

1.5

×10

6cf

u g–1

Kra

saek

oopt

et a

l. (2

004)

Alg

inat

e1.

104

cfu

g–1

PL

L-a

lgin

ate

1.0

×10

4cf

u g–1

L. c

asei

Ext

rusi

on2%

sod

ium

alg

inat

e w

ith

chit

osan

1.6

×10

6cf

u g–1

Kra

saek

oopt

et a

l. (2

004)

Alg

inat

e6.

103

cfu

g–1

PL

L-a

lgin

ate

7.0

×10

3cf

u g–1

L. b

ulga

ricu

sE

xtru

sion

2% s

odiu

m a

lgin

ate

wit

h ch

itos

anH

ighe

r th

an 1

06cf

u m

L–1

Lee

et a

l. (2

004)

B. b

reve

Em

ulsi

on/s

pray

-dry

ing

10%

hea

t-de

natu

red

whe

y pr

otei

n is

olat

e1.

0log

cfu

mL

–1P

icot

and

Lac

roix

(20

04)

B. l

ongu

m3.

8log

cfu

mL

–1

L. a

cido

phil

usE

xtru

sion

Sod

ium

alg

inat

e w

ith

poly

-L-l

ysin

e H

ighe

r th

an 1

06cf

u m

L–1

Iyer

and

Kai

lasa

path

y (2

005)

or c

hito

san

Add

itio

n of

Hi-

mai

ze s

tarc

hor

Raf

tili

ne®

/Raf

tilo

se®

B. l

acti

sE

xtru

sion

0.75

% g

ella

n/1%

xan

than

gum

Hig

her

than

106

cfu

mL

–1M

cMas

ter

et a

l. (2

005)

Tab

le 4

.5.

The

effe

ct o

f en

cap

sula

tio

n o

n t

he

surv

ival

of

bac

teri

a u

nd

er g

astr

oin

test

inal

co

nd

itio

ns

95

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 95

Page 104: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

numbers. Similar results were also observed by Chandramouli et al. (2004). Further-more, Sultana et al. (2000) reported that survival of probiotics in alginate-starch beads withdiameters of 1.0 mm did not improve after exposure to acidic and bile salt solutions.

Applications of Modern Optimization Techniques on the Optimal Manufacturing Conditions for Probiotic Capsules

Factors that can influence the survival rate of the probiotic capsules have been discussed inthe above sections. Different ingredients constituting the probiotic capsules may also haveprofound effects on the survival rate. In order to clarify the effects of these different ingre-dients, experimental design can be carried out and response surface models developed.Furthermore, modern optimization techniques can be applied to attain the optimal compo-sition of the capsules.

The objective of this section is to demonstrate the application of two modern optimiza-tion techniques for searching the optimal combination of coating materials for probioticmicrocapsules. The whole concept (Figure 4.1) includes:

1. Performing screening experiments and experimental design2. Encapsulating the probiotics according to the experimental design3. Building response surface models and formulating the optimization model4. Performing optimization5. Verifying the optimal manufacturing conditions.

A practical example of incorporating an additional prebiotic component to alginate matrixis presented in the following to illustrate the entire scheme.

Performing Screening Experiments and Experimental Design

Theoretically, all factors that affect the physicochemical properties of a final productshould be included in the experimental design. However, if all the variables are included,the search process may become cumbersome. Therefore, the potentially dominant parame-ters must be identified by a screening process to limit the number of experiments needed toa reasonable extent. After the screening experiments, the remaining screened factors areused in the design.

The experimental design, which applies the statistical principles for data collection priorto the experiment, has the main advantage of reducing the number of experimental trialsneeded to evaluate multiple parameters and to determine their interactions (Porretta et al.1995; Lee et al. 2000; Chen et al. 2005b). The response surface design, including theCentral Composite Design (CCD) and Box-Behnkin Design (BBD; Box and Behnkin1960) provides more informative data from the least number of experimental runsthan from the traditional method. The CCD is a popular class of second-order design. Thisdesign involves the use of a two-level factorial and 2k axial points with k being the numberof factors involved. On the other hand, the BBD is an effective three-level design basedon the construction of a balanced incomplete block design, and is an important alternativeto CCD.

In this study, survival of encapsulated probiotics (Lactobacillus spp. and Bifidobac-terium spp.) was found to be dependent on the concentrations of alginates as well as the

96 Chapter 4

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 96

Page 105: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

three prebiotic coating materials (peptides, FOS, and IMO). These four components wereregarded as independent variables and therefore a four-variable BBD with six replicates atthe center point (total 30 trials) was selected to build the response surface models. Thecoded and the nature variables and their respective levels are shown in Table 4.6.

Applications of Probiotic Encapsulation in Dairy Products 97

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Figure 4.1. Research scheme for application of modern optimization techniques forencapsulating probiotics.

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 97

Page 106: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Encapsulating the Probiotics According to the Experimental Design

A schematic representation of the manufacturing process for probiotic microcapsules isshown in Figure 4.2 and the process can be described as follows. Probiotic microcapsuleswere prepared according to the BBD by mixing 4% (v/v) of culture concentrate (1% eachof L. acidophilus, L. casei, B. bifidum, and B. longum) with sodium alginate and the previ-ously autoclaved (121°C, 15 min) prebiotics, FOS (0�3%), and IMO (0�3%), as well as

98 Chapter 4

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N Figure 4.2. Flow diagram for the preparation of probiotic microcapsules.

Level

Independent variable Symbol Coded Nature

–1 1.00Sodium alginate concentration (%) X1 0 2.00

+1 3.00–1 0.00

Peptides concentration (%) X2 0 0.50+1 1.00–1 0.00

FOS concentration (%) X3 0 1.50+1 3.00–1 0.00

IMO concentration (%) X4 0 1.50+1 3.00

Table 4.6. Process variables and their levels in four variables—Box Behnkin Design

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 98

Page 107: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

peptides (0�1%). The mixture with cell suspension was injected through a 0.11 needle intosterile 0.1 M CaCl2. The beads approximately 0.5 mm in diameter were allowed to stand for1 hr for solidification, and then rinsed with, and subsequently kept in, sterile 0.1% peptonesolution at 4°C. Survival of the microencapsulated probiotics before and after simulated gas-tric fluid test (defined as responses) was determined. The four responses were defined as viabil-ity of Lactobacillus spp (L. acidophilus + L. casei.) before simulated gastric fluid test (SGFT),viability of Bifidobacterium spp. (B. longum + B. bifidum) before SGFT, viability of Lacto-bacillus spp. after SGFT, and viability of Bifidobacterium spp after SG

Building Response Surface Models and Formulating theOptimization Model

Experimental data can be utilized to build mathematical models using linear, quadratic, orcubic functions by the least square regression method, after which the fitted functions aretested for adequacy and fitness using analysis of variance (ANOVA). Once an appropriateapproximating model has been derived, it can then be analyzed using various optimizationtechniques to determine the optimum conditions for the process.

Model analysis and the Lack-of-Fit test can be used for the selection of adequate mod-els, as outlined by Lee et al. (2000) and Weng et al. (2001). The model analysis comparesthe validities of the linear, quadratic, and cubic models for the different responses accord-ing to their F-values. A model with P-values (P>F) below 0.05 is regarded as significantand the highest-order polynomial that is significant will be selected. The Lack-of-Fit testdemonstrates if the lack-of-fit between the experimental values and those calculated basedon the model equations can be explained by the experimental error. The model with no sig-nificant lack-of-fit is appropriate for the description of the response surface.

In this example, the model analysis results (Table 4.7 and Table 4.8) show that the foll-owing four equations, which represent three linear survival models (Lactobacillus spp.before SGFT, Bifidobacterium spp. before SGFT and Bifidobacterium spp. after SGFT)and one cubic model (Lactobacillus spp. after SGFT), appear to be the most accurate withno significant lack-of-fit.

Applications of Probiotic Encapsulation in Dairy Products 99

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

La Bb

Model analysisc Lack-of-Fit testd Model analysis Lack-of-fit test Source (P>F) (P>F) (P>F) (P>F)

Linear 0.0002** 0.3972 0.0013** 0.8444Quadratic 0.5377 0.3595 0.4090 0.8743Cubic 0.5023 0.2509 0.6494 0.9092

Table 4.7. Model analysis and lack-of-fit test for the viability of lactic acid bacteria for beforesimulated gastric fluid test

* Significant at 5% level.** Significant at 1% level.a L: L. acidophilus � L. casei.b B: B. longum � B. bifidum.c Model analysis selects the highest order polynomial where the additional terms are significant.d Lack-of-Fit test wants the selected model to have insignificant lack-of-fit.

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 99

Page 108: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

(1)

(2)

f Laft � 1.41 � 3.53X1 � 8.89X2 � 1.35X3 � 0.68X4 � 0.83X 2

1 � 1.19X 22

� 0.23X23 � 0.074X2

4 � 5.89X1X2 � 0.029X1X3 � 0.65X1X4

� 1.46X2X3 � 0.81X2X4 � 0.14X3X4 � 1.34X1X1X2 � 0.076X1X1X3

� 0.17X1X1X4 � 0.20X1X2X2 � 0.093X1X3X3 � 0.085X2X2X3

� 0.74X2 X2 X4 � 0.48X2X3X3 (3)

f Baft � 7.35 � 0.045X1 � 0.30X2 � 0.065X3 � 0.065X4 (4)

where f Lbef , f

Bbef , f

Laft , and f B

aft represent the functions for the survival of Lactobacillus spp.(superscript L) and Bifid obacterium spp. (superscript B) before (subscript bef) and after(subscript aft) SGFT, respectively. The three-level BBD is incapable of forming the purecubic terms, that is, those with X3

i, and equation (3) confirms this fact.In order to search for a solution maximizing multiple responses, a composite fitness func-tion (CFF) is defined as following:

(5)

where fi represents the ith function (response) and m denotes the total number of functions.The term inside the parentheses in equation (5) is the product of all m functions. The com-posite function combines m responses (m = 4 in our study) into one single function whosemaximum can then be sought by optimization techniques with each response contributingequally to the CFF.

The relationship between the factors and the responses can be investigated by examiningthe CFF contour plots created by holding constant two of the four independent variables.

CFF =⎛

⎝⎜

⎠⎟

=∏ fi

i

m m

1

1

f X X XBbef

� � � � � �7 71 0 098 0 46 0 021 3 45 101 2 3

3. . . . .− XX4

f X X XLbef

� � � � � �8 17 0 075 0 13 0 024 1 05 101 2 3

3. . . . . × XX4

100 Chapter 4

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

L B

Model analysis Lack-of-fit test Model analysis Lack-of-fit test Source (P>F) (P>F) (P>F) (P>F)

Linear 0.0004** 0.0812** 0.0292* 0.4182Quadratic 0.0161* 0.0631** 0.2185 0.4976Cubic 0.0006** 0.1421 0.2918 0.6442

Table 4.8. Model analysis and Lack-of-Fit test for the viability of lactic acid bacteria for aftersimulated gastric fluid test

* Significant at 5% level.** Significant at 1% level.

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 100

Page 109: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Applications of Probiotic Encapsulation in Dairy Products 101

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

By fixing the peptides and FOS at three different levels, a three-dimensional plot of CFFvalues as a function of sodium alginate and IMO can be produced. Figure 4.3 depicts thatthe CFF values increase in accordance with the higher levels of FOS and peptides. On theother hand, the higher IMO and alginate concentrations lead to lower CFF values whenFOS and peptides are 3 and 1%, respectively. Figure 4.3(c) shows clearly an optimal CFFvalue of 8.172.

Performing Optimization

The CFF in equation (5) can be used as the objective function to be maximized in anoptimization problem, and the problem can be solved to find the optimal formulation forprobiotic microcapsules using optimization techniques. Optimization theory consists of abody of numerical methods for finding and identifying the best candidate from a collectionof alternatives without having to explicitly evaluate all possible alternatives (Reklaintis et al.1983). Among the optimization techniques, the steepest ascent (or descent) is commonlyused (see, for example, Stat-Ease, Inc., 2000), but the method is relatively inefficient and is alocal optimization technique capable of finding only local optima. Genetic Algorithms(GAs), although even less efficient than the steepest ascent, are considered as global schemes.The Sequential Quadratic Programming (SQP) technique is very powerful and efficient, andwith some modifications it can also perform global optimizations (Chen 2003).

Optimization Using the SQP Technique

A quadratic programming (QP) problem is an optimization problem involving a quadraticobjective function and linear constraints. The SQP method represents the current state-of-the-art in non-linear programming methods (The Math Work Inc., 2000) and can be used tosolve a series of QP problems approximating the original non-linear programming prob-lem. The basic scheme of an SQP technique can be expressed in the following steps(Reklaintis et al. 1983; Chen 2003):

Step 1: Set up and solve a QP subproblem, giving a search direction.Step 2: Test for convergence, stop if it is satisfied.Step 3: Step forward to a new point along the search direction.Step 4: Update the Hessian matrix in QP and go to step 1.

In order to search for the global optimum, the concept of multi-start global optimizationprocedure (Snyman and Fatti 1987) may be combined with the SQP method. If F* denotesthe global maximum and r, the number of sample points falling within the region of conver-gence of the current overall maximum F after n points have been sampled, then, under sta-tistically non-informative prior distribution, the probability that F be equal to F* satisfiesthe following relationship (Chen 2003):

Pr[F�F*] � q(n, r) � 1�[(n�1)!(2n�r)!]/[(2n�1)!(n�r)!] (6)

A global optimization program equipped with a multi-start SQP technique was coded tosolve for the optimal solution in this example. The modified SQP with the multi-start ability,

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 101

Page 110: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

(a)

(b)

(c)

Figure 4.3. Response surface plots of survivability of probiotic microcapsules showing effects ofsodium alginate and IMO at constant levels of (a) 0% peptides, 0% FOS, (b) 0.5% peptides, 1.5%FOS, (c) 1% peptides, 3% FOS.

102

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 102

Page 111: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

which is capable of reaching the global optimum with great certainty, has been proven to be avery efficient method (Chen et al. 2004). The program generates a series of uniformlydistributed random points for initial search, and then the SQP is applied to find the optimumbased on each subsequent initial point. If the probability of locating the global optimumexceeds a preset value (99.99% in this example), the global optimum is considered found.Otherwise, the next random, initial point is generated and the SQP re-executed.

A very high probability (>0.9999) in equation (6) was set to ensure the global optimumwould be attained. Figure 4.4 shows the evolution of the CFF values for a sequence of ran-domly generated initial searching points and the optimal points found. The optimizationresults clearly show that determination of the optima depends on the initial search points

Applications of Probiotic Encapsulation in Dairy Products 103

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

(a)

(b)

Number of function evaluations

Com

posi

te fi

tnes

s fu

nctio

n (C

FF

)

Initial searching point set

Opt

imal

com

posi

te fu

nctio

n va

lue

Figure 4.4. (a) Evolution curve of CFF with 2% alginate, 0.5% peptides, 1.5% FOS and 1.5% IMOas the initial searching point; (b) evolution curve of optimal CFF for randomly generated initialsearching point using SQP to identify optimal production conditions for probiotic microcapsules.

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 103

Page 112: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

and there are three different local optimal CFF values identified from 20 randomly gener-ated initial points. Of these local optima, the global optimal CFF is 8.172 with 99.99%certainty. The global maximum corresponds to: 8.30log cfu for survival of Lactobacillusspp. before SGFT, 8.01log cfu for survival of Bifidobacterium spp. before SGFT, 8.00logcfu for survival of Lactobacillus spp. after SGFT, and 7.72log cfu for survival of Bifidobac-terium spp. after SGFT. The highest optimal CFF value (8.172) was attained for 10 of 20sets and the optimal point consists of independent variables at X1�1, X2�1, X3�3, andX4 = 0. In other words, the optimal combination of the coating materials for the probioticmicrocapsules is 1% sodium alginate blended with 1% peptides, 3% FOS, and 0% IMO.

Optimization Using the Genetic Algorithms

Genetic Algorithms are search procedures that imitate the natural evolution process andcan be used for the computation of the global maximum or minimum of a function(Mitchell 1996). Genetic algorithms differ from other search techniques in that they searchamong a population of points and use probabilistic rather than deterministic transitionrules. As a result, genetic algorithms search more globally (Wang 1997).

GAs provide a very flexible framework and recently have been regarded as not only aglobal optimization method but also a multi-objective optimization method in variousareas. Generally, the algorithms can be described in the following steps (Goldberg 1989;Mitchell 1996):

Step 1: Start with a randomly generated population of chromosomes, each of whichdefines a combination of the coating materials in this example.Step 2: Calculate the fitness f (x) of each chromosome x in the population, with the fit-ness being the CFF value of that combination of the coating materials.Step 3: Repeat the following substeps until n offsprings have been created: (i) select apair of parent chromosomes from the current population, the probability of selectionbeing an increasing function of fitness; (ii) with crossover rate, cross over the pair at arandomly chosen point to form two offsprings; (iii) mutate the two offsprings at a pre-scribed mutation rate and place the resulting chromosomes in the new population;(iv) replace the current population with the new population. Each iteration of thisprocess is called a generation. The above procedure is called the simple GA (SGA).

The Micro Genetic Algorithm (MGA) is a popular modification to SGA to optimize theprocessing conditions (Chen et al. 2003). The essence of MGA is the lack of mutations andthe presence of re-starts. Due to these features, the algorithm converges rapidly to a local orglobal maximum (Nikitas et al. 2001). The lack of mutations also results in a rapid decreaseof the variance of the cost values of the population. When the variance value falls below acertain limit, a restarting process begins in which the chromosome with the highest CFFvalue is retained and the rest N–1 chromosomes (N is the total number of chromosomes inone generation) are replaced by randomly generated new ones.

The efficiency of the algorithms can be examined by the number of function evaluationsas follows:

Number of function evaluations � Number of generations � Population size (7)

104 Chapter 4

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 104

Page 113: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

A smaller number of function evaluations indicate a higher efficiency.In the study of alginate microcapsules incorporated with prebiotics, the CFF was opti-

mized using MGA. The initial population consisting of 10 chromosomes (population size)was generated at random and the crossover rate was set to 0.5. The chromosomes withhigher CFF values were selected and retained for the next generation. The maximum num-ber of generations was set to 500 for the problem. Figure 4.5 shows the evolution curve ofthe first 3000 function evaluations in searching for the global, optimal value. The MGAproduced rapidly increasing CFF during the early stage of the optimization process consist-ing of a total of 5000 function evaluations, which is typical for MGA.

The chromosomes having the maximum CFF provided the optimal ratio of concentra-tions of the coating materials. The optimal value (CFF = 8.172) was obtained after 1490function evaluations during the process.

Verifying the Optimal Manufacturing Conditions

After the optimal processing condition is found by the SQP or MGA, repeated experimentsbased on the condition should be conducted to verify the predicted optimum. The verifica-tion results can then be analyzed using ANOVA from the SAS software package (SASInstitute Inc., 1990), with Duncan’s multiple range test for significance to detect differencesbetween predicted values and observed values. In this example, the optimal productioncondition for the coating composition, derived from the SQP and MGA, was the same. Theoptimal combination of the coating materials for the probiotic microcapsules is 1% sodiumalginate blended with 1% peptides, 3% FOS, and 0% IMO. The four responses (survival ofLactobacillus spp. and Bifidobacterium spp. before and after SGFT) and the CFF valuederived from the verification experiments are all very close to the SQP- or MGA-based pre-diction, with no apparent significant differences (P�0.05) comparing the two sets. BothSQP and MGA techniques may be used to determine the optimal combination of the coat-ing materials for probiotic microcapsules. By comparing both methods, SQP was deemedto be much more efficient than MGA at such a task.

Applications of Probiotic Encapsulation in Dairy Products 105

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47Figure 4.5. Optimum composite function values using MGA.

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 105

Page 114: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Practical Applications of Encapsulated Probiotics in Dairy Products

As discussed above, encapsulated probiotics have been used for accelerating cheese ripen-ing, fortifying dairy products with beneficial bacteria as well as enhancing the shelf life andbioavailability of this class of microorganisms in dietary supplements.

Several companies are currently involved in designing and manufacturing such productsto meet customers’ needs. Following are few examples of incorporating encapsulated pro-biotics into real food systems:

Yogurt

For manufacturing set yogurt, homogenized whole milk and skim milk powder are blendedfor a total solids content of 15–18%(w/w). The mix is pasteurized at 80–85°C for 30 min.and cooled to 42°C before inoculation with a commercial freeze-dried starter culture con-taining S. thermophilus and L. bulgaricus. The encapsulated probiotic cultures are thenadded and the resulting mix is dispensed into containers and incubated at 42°C for 4–5 hruntil the pH reached 4.5. Finally, yogurts are stored at 4°C (Adhikari et al. 2000; Sultanaet al. 2000; Sun and Griffiths 2000; Krasaekoopt et al. 2004; Picot and Lacroix 2004; Iyerand Kailasapathy 2005).

Stirred yogurt is manufactured in the same way except that the mix after inoculation isincubated at 42°C for 4–5 hr until the pH reaches 4.5, added with 10% microencapsulatedprobiotics, and then stirred and dispensed into containers. The probiotic yogurt is stored at4°C (Adhikari et al. 2003). The probiotic counts of yogurts remained above 106 cfu/mLand the final pH was 3.9–4.1 after one month of storage.

Commercialized yogurt products containing microencapsulated probiotics are alsoavailable. Kaung-Chuan Inc. in Taiwan produces a bio-yogurt drink with probiotic micro-capsules, which incorporate Bifidobacterium spp, are made by gelatin and have an averagesize of 1–2 mm. The company claims that this product has intestinal benefits.

Cheese

Introducing encapsulated probiotics to cheese not only enhances the storage viability ofprobiotics but also improves the cheese flavor.

For manufacturing cheddar cheese, raw whole milk is pasteurized and cooled to 31°C. Thefreeze-dried mesophilis lactic starter culture is added at the rate of 5 g/100 g of milk. Curdforms in approximately 30 min. and is cut with 0.65 cm wire knives. After a 15 min. healingperiod, the temperature of the curd and whey mixture is raised to 37–38°C in 30 min. and thenmaintained at that temperature for an additional 30 min. After the whey is drained, the curd ischeddared to pH 5.2, and then milled, salted, followed by addition of the microencapsulatedprobiotics and packing into hoops that are further ripened at 7°C for 6 months. Cheese con-taining encapsulated Bifidobacterium was shown to possess similar flavor, texture, andappearance compared to the control (Dinakar and Mistry 1994; Desmond et al. 2002).

Frozen Desserts

For manufacturing frozen ice milk, probiotics microencapsulated with 3% calcium alginate(bead diameters > 30 µm) are blended with milk (5% fat) and the mix is frozen continuously

106 Chapter 4

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 106

Page 115: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

in a freezer. Addition of microencapsulated probiotics has no measurable effect on the over-run and the sensory characteristics of the products with 90% probiotic survival (Sheu et al.1993). Sheu et al. (1993) manufactured fermented frozen dairy desserts by blending freezedried microencapsulated probiotics with yogurt and base mix, and then the mix was frozenin a continuous freezer. Figure 4.6 details the process of incorporating encapsulated probi-otic culture to a frozen milk-based dessert system.

Applications of Probiotic Encapsulation in Dairy Products 107

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Figure 4.6. Typical manufacturing process of fermented, frozen dairy desserts with microencap-sulated probiotics.

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 107

Page 116: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Summary

Encapsulated probiotics can be used in many dairy products such as yogurt, frozendesserts, and cheese. In the encapsulated form, these sensitive microorganisms are pro-tected from harsh environments including high levels of lactic and acetic acid, gastrointesti-nal conditions, and freezing temperatures. Among encapsulation methods, spray-drying,extrusion, and emulsion are the most common techniques for probiotic encapsulation.However, the high cost of the process and the technical difficulty limit the large-scale appli-cation of encapsulation technologies in the dairy industry.

Carrier matrices, encapsulation methods, and various dairy products to which the probi-otic capsules are applied can influence the survival rate of the probiotics. Different ingredi-ents constituting the probiotic capsules may also have profound effects on the survival rate.In order to clarify the effects of these different ingredients, experimental design can be car-ried out and response surface models developed. Furthermore, modern optimization tech-niques can be applied to attain the optimal composition of the capsules. The two-stageeffort of obtaining a surface model using RSM and optimizing this model using SQP andMGA techniques has been demonstrated to represent an effective approach.

ReferencesAdhikari, K., A. Mustapha and I. U. Grün. 2000. Viability of microencapsulated Bifidobacteria in set yogurt dur-

ing refrigerated storage. Journal of Dairy Science 83: 1946–1951.Adhikari, K., A. Mustapha and I. U. Grün. 2003. Survival and metabolic activity of microencapsulated Bifidobac-

terium in stirred yogurt. Journal of Food Science 68(1): 275–280.Ananta, E., M. Volkert and K. Knorr. 2005. Cellular injuries and storage stability of spray-dried Lactobacillus

rhamnosus GG. International Dairy Journal 15: 399–409.Audet, P., C. Paquin and C. Lacroix. 1988. Immobilized growing lactic acid bacteria with k-carrageenan-locust

bean gum gel. Applied Microbiology and Biotechnology 29: 11–18.Audet, P., C. Paquin and C. Lacroix. 1989. Sugar utilization and acid production by free and entrapped cells of

Streptococcus salivarius ssp. Thermophilus, Lactobacillus delbrueckii ssp. bulgaricus, and Lactococcus lactisssp. lactis in a whey permeate medium. Applied and Environmental Microbiology 55(1): 185–189.

Audet, P., C. Lacroix and C. Paquin. 1992. Effect of agitation rate on cell release rate and metabolism during con-tinuous fermentation with entrapped growing. Biotechnology Techniques 6(3): 265–270.

Berrada, N., J. F. Lemeland, G. Laroche, P. Thouvenot and M. Piaia. 1991. Bifidobacterium from fermented milks:Survival during gastric transit. Journal of Dairy Science 74: 409–413.

Bielecka, M., E. Biedrzycka and A. Majkowska. 2002. Selection of probiotics and prebiotics for symbiotic andconfirmation of their in vivo effectiveness. Food Research International 35: 125–131.

Box, G. E. P. and E. W. Behnkin. 1960. Some new three level designs for the study of quantitative variables. Tech-nometrics 2: 455–475.

Canh, L., M. Millette, M. Mateescu and M. Lacroix. 2004. Modified alginate and chitosan for lactic acid bacteriaimmobilization. Biotechnology and Applied Biochemistry 39: 347–354.

Champagne, C. P., Gaudy, D. Poncelet and R. J. Neufeld. 1992. Lactococcus lactis release from calcium alginatebeads. Applied and Environmental Microbiology 58: 1429–1434.

Champagne, C. P., Lacroix and I. Sodini-Gallot. 1994. Immobilized cell technologies for the dairy industry. Criti-cal Reviews in Biotechnology 14(2): 109–134.

Champagne, C. P. and N. J. Gardner. 2005. Challenges in the addition of probiotic cultures to foods. CriticalReviews in Food Science and Nutrition 45: 61–84.

Chandramouli, V., K. Kailasapathy, P. Peiris and M. Jones. 2004. An improved method of microencapsulation andits evaluation to protect Lactobacillus spp. in simulated gastric conditions. Journal of Microbiological Methods56: 27–35.

Chen, H. C., C. W. Lin and M. J. Chen. 2006. The effects of freeze drying and rehydration on survival of microor-ganisms in Kefir. Asian-Australian Journal Animal Science 19(1): 126–130.

108 Chapter 4

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 108

Page 117: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Applications of Probiotic Encapsulation in Dairy Products 109

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Chen, M. J., K. N. Chen and C. W. Lin. 2003. Optimization of the viability of probiotics in a new fermented milkdrink by genetic algorithms for response surface modeling. Journal of Food Science 68: 632–638.

Chen, M. J., K. N. Chen and C. W. Lin. 2004. Sequential quadratic programming for development of a new probi-otic dairy tofu with glucono-D-lactone. Journal of Food Science 69(7): 344–350.

Chen, M. J., K. N. Chen and C. W. Lin. 2005a. Optimization of incorporated prebiotics as coating materials forprobiotic microencapsulation. Journal of Food Science 70 (5): 260–267.

Chen, M. J., K. N. Chen and C. W. Lin. 2005b. Optimization on response surface models for the optimal manufac-turing conditions of dairy tofu. Journal of Food Engineering 68(4): 471–480.

Chen, S. 2003. Robust design with dynamic characteristics using stochastic sequential quadratic programming.Engineering Optimization 35(1): 79–89.

Cui, J., J. Goh, P. Kim, S. Choi and B. Lee. 2000. Survival and stability of Bifidobacteria loaded in alginate poly-L-lysine microparticles. International Journal of Pharmaceutics 210: 51–59.

Dave, R. I. and N. P. Shah. 1997. Effectiveness of ascorbic acid as an oxygen scavenger in improving viabilityof probiotic bacteria in yoghurts made with commercial starter cultures. International Dairy Journal7: 435–443.

Dave, R. I. and N. P. Shah. 1998. Ingredients supplementation effects on viability of probiotic bacteria in yogurt.Journal of Dairy Science 81: 2804–2816.

Desmond, C., R. P. Ross, E. O’Callaghan, G. Fitzgerald and C. Stanton. 2002. Improved survival of Lactobacillusparacasei NFBC 338 in spray-dried powders containing gum acacia. Journal of Applied Microbiology93: 1003–1011.

Dinakar, P. and V. V. Mistry. 1994. Growth and viability of Bifidobacterium bifidum in cheddar cheese. Journal ofDairy Science 77: 2854–2864.

Doleyres, Y. and C. Lacroix. 2005. Technologies with free and immobilized cells for probiotic Bifidobacteria pro-duction and protection. International Dairy Journal 15: 973–988.

Favaro-Trindade, C. S. and C. R. F. Grosso. 2002. Microencapsulation of L. acidophilus (La-05) and B. lactis(Bb-12) and evaluation of their survival at the pH values of the stomach and in bile. Journal of Microencapsula-tion 19(4): 485–494.

Fuller, R. 1992. Probiotics: The Scientific Basis. London: Chapman & Hall. p. 125.Gardiner, G. E., P. Bouchier, E. O’Sullivan, J. Kelly, J. K. Collins, B.G. Fitzgerald, R. P. Ross and C. Stanton. 2002.

A spray-dried culture for probiotic cheddar cheese manufacture. International Dairy Journal 12: 749–756.Gaumann, A., M. Laudes, B. Jacob, R. Pommersheilm, C. Laue, W. Vogt and J. Schrezenmeir. 2001. Xenotrans-

plantation of parathyroids in rats using barium alginate and polyacrylic acid multilayer microcapsules. Experi-mental and Toxicologic Pathology 53: 35–43.

Gibbs, B. F., S. Kermasha, I. Alli and C. N. Mulligan. 1999. Encapsulation in the food industry: A review. Inter-national Journal of Food Sciences and Nutrition 50: 213–224.

Gibson, G. R. and M. B. Roberfroid. 1995. Dietary modulation of the human colonic microbial: Introducing theconcept of prebiotics. Journal of Nutrition 125: 1401–1412.

Gilliland, S. E. 1989. Acidophilus milk products: A review of potential benefits to consumers. Journal of DairyScience 72: 2483–2494.

Goldberg, D. E. 1989. Genetic Algorithms in Search, Optimization, and Machine Learning. Massachusetts: Addi-son-Wesley Publishing Company.

Gombotz, W. R. and S. F. Wee. 1998. Protein release from alginate matrices. Advanced Drug Delivery Reviews31: 276–283.

Groboillot, A. F., C. P. Champagne, G. F. Darling and D. Poncelet. 1993. Membrane formation by interfacial cross-linking of chitosan for microencapsulation of Lactococcus lactis. Biotechnology and Bioengineering 42(10):1157–1163.

Guoqiang, D., R. Kaul and B. Mattiasson. 1991. Evaluation of. alginate-immobilized Lactobacillus casei for lac-tate production. Applied Microbiology and Biotechnology 36: 309–314.

Havenaar, R. and J. H. H. Huis in’t Veld. 1992. Probiotics: A General View. New York: Chapman and Hall.pp. 209–224.

Hegenbart, S. 1993. Encapsulated ingredients keep problems conversed. Food Product Design 3: 28–34.Hou, R. C. W., M. Y. Lin, M. M. C. Wang and J. T. C. Tzen. 2003. Increase of viability of entrapped cells of

Lactobacillus delbrueckii ssp. bulgaricus in artificial sesame oil emulsions. Journal of Dairy Science 86:424–428.

Iwana, H., H. Masuda, T. Fujisawa, H. Suzuki and T. Mitsuoka. 1993. Isolation and identification of Bifidobac-terium ssp. in commercial yogurt sold. Europe. Mitsuokifidobacteria Microflora 12: 39–45.

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 109

Page 118: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Iyer, C. and K. Kailasapathy. 2005. Effect of co-encapsulation of probiotics with prebiotics on increasing the via-bility of encapsulated bacteria under in vitro acidic and bile salt conditions and in yogurt. Journal of Food Sci-ence 70(1): 18–23.

Jankowski, T., M. Zielinska and A. Wysakowska. 1997. Encapsulation of lactic acid bacteria with alginate/starchcapsules. Biotechnology Techniques 11(1): 31–34.

Kailaspathy, K. and S. Rybka. 1997. Lactobacillus acidophilus and Bifidobacterium spp.—their therapeuticpotential and survival in yogurt. Australia Journal of Dairy Technology 52: 28–35.

Kearney, L., M. Upton and A. Loughlin. 1990. Enhancing the viability of Lactobacillus plantarum inoculum byimmobilizing the cells in calcium-alginate beads. Applied and Environmental Microbiology 56(10): 3112–3116.

Kebary, K. M., S. A. Hussein and R. M. Badawi. 1998. Improving viability of Bifidobacteria and their effect onfrozen ice milk. Egyptian Journal of Dairy Science 26 (2): 319–337.

King, A. H. 1995. Encapsulation of food ingredients: A review of available technology, focusing on hydrocolloids.In Encapsulation and Control Release of Food Ingredients, eds. S. J. Risch and G. A. Reineccius, pp. 213–220.Washington DC: American Chemical Society.

Klaver, F. A. M., F. Kingma and A. H. Weerkamp. 1993. Growth and survival of bifidobacteria in milk. NetherlandsMilk and Dairy Journal 47: 151–164.

Koch, S., C. Schwinger, J. Kressler, C. H. Heinzen and N. G. Rainov. 2003. Alginate encapsulation of geneticallyengineered mammalian cells: comparison of production devices, methods and microcapsule characteristics.Journal of Microencapsulation 20(3): 303–316.

Krasaekoopt, W., B. Bhandari and H. Deeth. 2003. Evaluation of encapsulation techniques for probiotics foryogurt. International Dairy Journal 13: 3–13.

Krasaekoopt, W., B. Bhandari and H. Deeth. 2004. Comparison of texture of yogurt from conventionally treatedmilk and UHT milk fortified with low-heat skim powder. Journal of Food Science 69(6): E276–E280.

Krasaekoopt, W., B. Bhandari and H. C. Deeth. 2006. Survival of probiotics encapsulated in chitosan-coated algi-nate beads in yoghurt from UHT- and conventionally treated milk during storage LWT 39: 177–183.

Kurman, J. A. and J. L. Rasic. 1991. The health potential of products containing Bifidobacteria. In TherapeuticProperties of Fermented Milk, ed. R. K. Robinson, 115–117. London: Elsevier Application Food ScienceSeries.

Lacroix, C., C. Paquin and J. P. Arnaud. 1990. Batch fermentation with entrapped growing cells of Lactobacilluscasei. I. Optimization of the rheological properties of the entrapment. Applied Microbiology and Biotechnology32: 403–408.

Lankaputhra, W. E. V. and N. P. Shah. 1995. Survival of Lactobacillus acidopilus and Bifidobacterium spp. in thepresence of acid and bile salts. Cultured Dairy Products Journal 30: 2–7.

Lankaputhra, W. E. V. and N. P. Shah. 1996. Survival of Bifidobacteria during refrigerated storage in the presenceof acid and hydrogen peroxide. Milchwissenschaft 51: 65–70.

Larisch, B. C., D. Poncelet and C. P. Champagne. 1994. Microencapsulation of Lactococcus lactis subsp. cre-moris. Journal of Microencapsulation 11(2): 189–195.

Lee, J. S., D. S. Cha and H. J. Park. 2004. Survival of freeze-dried Lactobacillus bulgaricus KFRI 673 in chitosan-coated calcium alginate microparticles. Journal of Agricultural Food Chemistry 52: 7300–7305.

Lee, K. and T. Heo. 2000. Survival of Bifidobacterium longum immobilized in calcium alginate beads in simulatedgastric juices and bile salt solution. Applied and Environmental Microbiology 66(2): 868–873.

Lee, J., L. Ye, W. O. Landen and R. R. Eitenmiller. 2000. Optimization of an extraction procedure for the quantifi-cation of vitamin E in tomato and broccoli using response surface methodology. Journal of Food Compositionand Analysis 13: 45–57.

Le-Tien, C., M. Millette, M. Mateescu and M. Lacroix. 2004. Modified alginate and chitosan for lactic acid bacte-ria immobilization. Biotechnology and Applied Biochemistry 39: 349–354.

Lian, W. D., H. C. Hsiao and C. C. Chou. 2002. Survival of Bifidobacteria after spray-drying. International Journalof Food Microbiology 74: 79–86.

Lian, W. D., H. C. Hsiao and C. C. Chou. 2003. Viability of microencapsulated Bifidobacteria in simulated gastricjuice and bile solution. International Journal of Food Microbiology 86: 293–301.

Maitrot, H., C. Paquin, C. Lacroix and C. P. Champagne. 1997. Production of concentrated freeze-dried culturesof Bifidobacterium longum in k-carrageenan-locust bean gum gel. Biotechnology Techniques 11 (7): 527–531.

Martin, J. H. and K. M. Chou. 1992. Selection of Bifidobacterium ssp. for use as dietary adjuncts in cultured dairyfoods I. Tolerance to pH of yogurt. Cultured Dairy Products Journal 27: 21–26.

Mattila-Sandholm, T., P. Myllärinen, R. Crittenden, G. Mogensen, R. Fonden and M. Saarela. 2002. Technologicalchallenges for future probiotic foods. International Dairy Journal 12: 173–182.

110 Chapter 4

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 110

Page 119: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

McMaster, L. D., S. A. Kokott, S. J. Reid and V. R. Abratt. 2005. Use of traditional African fermented beverages asdelivery vehicles for Bifidobacterium lactis DSM 10140. International Journal of Food Microbiology 102:231–237.

Mitchell, M. 1996. An Introduction to Genetic Algorithms. London: The MIT Press.Modler, H. W. and L. Villa-Garcia. 1993. The growth of Bifidobacterium longum in whey-based medium and via-

bility of this organism in frozen yogurt with low and high levels of developed acidity. Culture Dairy ProductsJournal 28: 4–8.

Nikitas, P., A. Pappa-Louisi, A. Papageorgiou and A. Zitrou. 2001. On the use of genetic algorithms for responsesurface modeling in high-performance liquid chromatography and their combination with the Microsoft Solver.J. Chromatography A 942: 93–105.

Norton, S. and C. Lacroix. 1990. Gellan gum gel as entrapment matrix for high temperature fermentationprocesses: A rheological study. Biotechnology Techniques 4: 351–356.

O’Riordan, K. I., D. Andrews, K. Buckle and P. Conway. 2001. Evaluation of microencapsulation of a Bifidobac-terium strain with starch as an approach to prolonging viability during storage. Journal of Applied Microbiology91: 1059–1066.

Peirone, M., C. J. Ross, G. Hortelano, J. L. Brash and P. L. Chang. 1998. Encapsulation of various recombinant mam-malian cell types in different alginate microcapsules. Journal of Biomedical Materials Research 42: 587–596.

Peniche, C., W. Argüelles-Monal, H. Peniche and N. Acosta. 2003. Chitosan: An attractive biocompatible polymerfor microencapsulation. Macromolecular Bioscience 3: 511–520.

Picot, A. and C. Lacroix. 2003. Production of multiphase water-insoluble microcapsules for cell microencapsula-tion using an emulsification/spray-drying technology. Journal of Food Science 68(9): 2693–2700.

Picot, A. and C. Lacroix. 2004. Encapsulation of Bifidobacteria in whey protein-based microcapsules and survivalin simulated gastrointestinal conditions and in yogurt. International Dairy Journal 14: 505–515.

Porretta, A., A. C. Birzi and E. Vicini. 1995. Effects of ultra-high hydrostatic pressure treatments on the quality oftomato juice. Food Chemistry 52: 35–41.

Prevost, H. and C. Divies. 1987. Fresh fermented cheese production with continuous pre-fermented milk by amixed culture of mesophilis lactic streptococci entrapped in Ca-alginate. Biotechnology Letters 9(11):789–794.

Prevost, H. and C. Divies. 1988. Continuous pre-fermentation of milk by entrapped yogurt bacteria. I. Develop-ment of the process. Milchwissenschaft 43: 621–625.

Prevost, H. and C. Divies. 1992. Cream fermentation by a mixed culture of lactococci entrapped in two-layer cal-cium alginate gel beads. Biotechnology Letters 14(7): 583–588.

Prüße, U., J. Dalluhn, J. Breford and K. Vorlop. 2000. Production of spherical beads by Jet Cutting. ChemicalEngineering Technology 23(12): 1105–1110.

Rao, A. V., N. Shiwnarain and I. Maharaj. 1989. Survival of microencapsulated Bifidobacterium pseudolongum insimulated gastric and intestinal juices. Canadian Institute of Food Science and Technology Journal 22(4):345–349.

Reklaintis, G. V., A. Ravindran and K. M. Ragsdell. 1983. Engineering Optimization: Methods and Applications.New York: John Wiley and Sons.

Robinson, R. K. 1987. Survival of Lactobacillus acidophilus in fermented products. Suid Afrikaans Tydskrif VirSuiwelunde 19(1): 25–27.

Samona, A. and R. K. Robinson. 1994. Effect of yogurt cultures on the survival of Bifidobacteria in fermentedmilks. Journal of the Society of Dairy Technology 47: 58–60.

Schillinger, U. 1999. Isolation and identification of lactobacilli from novel-type probiotic and mild yogurts andtheir stability during refrigerated storage. International Journal of Food Microbiology 47: 79–87.

Shah, N. P. and W. E. V. Lankaputhra. 1997. Improving viability of Lactobacillus acidophilus and Bifidobacteriumssp. in yogurt. International Dairy Journal 7: 349–356.

Shah, N. P. and R. R. Ravla. 2004. Selling the cells in desserts. Dairy Industries International 69(1): 31–32.Sheu, T. Y. and R. T. Marshall. 1993. Microentrapment of lactobacilli in calcium alginate gels. Journal of Food

Science 54: 557–561.Sheu, T. Y., R. T. Marshall and H. Heymann. 1993. Improving survival of culture bacteria in frozen desserts by

microentrapment. Journal of Dairy Science 76: 1902–1907.Smidsrod, O. and G. Skjak-Braek. 1990. Alginate as immobilization matrix for cells. Trends in Biotechnology

8(3): 71–78.Snyman, J. A. and L. P. Fatti. 1987. A multi-start global minimization algorithm with dynamic search trajectories.

Journal of Optimization Theory and Applications 54(1): 121–141.

Applications of Probiotic Encapsulation in Dairy Products 111

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 111

Page 120: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Song, S., Y. Cho and J. Park. 2003. Microencapsulation of Lactobacillus casei YIT 9018 using a microporousglass membrane emulsification system. Journal of Food Science 68(1): 195–200.

Sultana, K., G. Godward, N. Reynolds, R. Arumugaswamy and P. Peiris. 2000. Encapsulation of probiotic bacteriawith alginate-starch and evaluation of survival in simulated gastrointestinal conditions and in yogurt. Inter-national Journal of Food Microbiology 62: 47–55.

Sun, W. and M. W. Griffiths. 2000. Survival of bifidobacteria in yogurt and simulated gastric juice followingimmobilization in gellan-xanthan beads. International Journal of Food Microbiology 61: 17–25.

Thies, C. 1996. A survey of microencapsulation. In Microencapsulation: Methods and Industrial Applications, ed.S. Benita, 1–19. New York: Marcel & Dekker.

Thu, B., P. Bruheim, T. Espevik, O. Smidsrod, P. Soon-Shiong and G. Skjak-Braek. 1996. Alginate polycationmicrocapsules. I. Interaction between alginate and polycation. Biomaterials 17: 1031–1041.

Truelstrup-Hansen, L., P. M. Allan-Wojtas, Y. L. Jin and A. T. Paulson. 2002. Survival of Ca-alginate microencap-sulated Bifidobacterium spp. in milk and simulated gastrointestinal conditions. Food Microbiology 19: 35–45.

Wang, Q. J. 1997. Using genetic algorithms to optimize model parameters. Environmental Modelling& Software12: 27–34.

Weng, W., W. Liu and W. Lin. 2001 Studies on the optimum models of the dairy product. Kou Woan Lao usingresponse surface methodology. Asian-Australasian Journal of Animal Sciences 14(10): 1470–1476.

Yoo, I., G. H. Seong, H. N. Chang and J. K. Park. 1996. Encapsulation of Lactobacillus casei cells in liquid-corealginate capsules for lactic acid production. Enzyme and Microbial Technology 19: 428–433.

Zhou, Y., E. Martins, A. Groboillot, C. P. and R. J. Neufeld. 1998. Spectrophotometric quantification of lactic bac-teria in alginate and control of cell release with chitosan coating. Journal of Applied Microbiology 84(3):342–348.

112 Chapter 4

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_04_083-112 r1.qxd 3/29/07 1:45 PM Page 112

Page 121: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

5 Encapsulation and Controlled Release inBakery Applications

Jamileh M. Lakkis

Introduction

In commercial baking operations, high volumes of dough and batter premixes are preparedfor further distribution to stores and on-site baking. Maintaining good functionality andoverall quality of these products requires careful inactivation of the prevailing leaveningsystems during storage and their controlled reactivation upon preparation and baking.

The basic ingredients in doughs and cake batters include flour, fat, eggs, and sweeteners.These components play an important role in determining the functional and eating qualityof bakery products. Minor ingredients such as yeasts and chemical leavening agents, how-ever, can have more dramatic effects on the overall quality and shelf life of these products.

Recent advances in microencapsulation and controlled release technologies have con-tributed significantly to current availability and wide consumers’ acceptability of shelf-stable bakery products. Bakery manufacturers have been keen on adopting these technologiesdue to the tremendous cost savings provided by extending shelf life, eliminating fermenta-tion stage, and shortening dough proofing time along with minimal impact on processabil-ity of bakery products. These benefits can be better appreciated considering the hugemarket of bakery products that was estimated at $300 billion worldwide in 2005 (SoslandPublishing Co., Kansas City, MO).

This chapter discusses methods for encapsulating and controlling the release of chem-ical and biological leaveners as well as other functional components of bakery systemssuch as sweeteners, antimicrobial agents, dough conditioners, and flavors. Microencapsula-tion technologies as well as coating materials available for bakery applications are alsodiscussed.

Encapsulation Technologies for Bakery Applications

A variety of encapsulation technologies have been adapted for bakery applications, mainlyhot melt particle coating and congealing via spray chilling. Embedding via extrusion andliposome/vesicles, used to a much lesser extent in bakery applications, has been coveredelsewhere in this book; therefore, only particle coating and congealing are discussed here.

Hot Melt Particle–Coating Technology

Fluid bed coating is a well-established technology for encapsulating and controlling therelease of solid actives. The process consists, essentially, of spraying a solution or a moltenfluid onto particles of a substrate material undergoing encapsulation. Application of a filmto a solid is a very complex process and requires careful selection of substrates and coatingmaterials as well as process conditions.

113

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 113

Encapsulation and Controlled Release: Technologies in Food SystemsEdited by Jamileh M. Lakkis

Copyright © 2007 by Blackwell Publishing

Page 122: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

The solid substrate is placed in a container that is typically an inverted truncated conewith a fine retention screen and an air distribution plate at its base. As the warm air flowsthrough the distribution plate, the particles become fluidized and are accelerated in anupward flow where they encounter fine spray of the coating fluid. The coating spray nozzlecan be fitted (1) to the top (top-spray system); (2) to the bottom (bottom-spray systemreferred to as Wurster); or (3) tangential to the base container (Figure 5.1a, b, c). The choiceof a suitable coating configuration should take into consideration the type of solid to becoated (powder, pellets, etc.) as well as the desired film thickness and release properties.

Top-spray fluid beds are favored for high-throughput applications as well as for film uni-formity. Bottom-spray (Wurster) systems are preferred for their high coating effectivenessas well as their ability to form perfectly sealed films. This is critical for controlled releaseapplications. Tangential-spray systems (rotor pellet coating), on the other hand, are suitablefor coating pellets and rods (yeasts) but not small particles (sodium bicarbonate and otherchemical leaveners). In the tangential coating system, rotation of the base plate disc sets thepellets into a spiral motion where they encounter the coating spray, thus coating concur-rently to the powder bed. Very thick film layers can be applied using the rotor configuration.In the Wurster system, film thickness varies with particle size within a batch; top- ortangential-spray fluid beds rarely show this variation. This is due, in part, to the slow circu-lation of lighter and/or smaller particles, a pattern inherent to the Wurster process(Ichikawa et al., 1996).

Regardless of the coating unit configuration chosen, film formation around solidparticles cannot be achieved by a single pass through the coating zone, but requires manysuch passes to produce complete particle coverage. The presence of any loose uncoatedactives can also have detrimental effects on the release mechanism and overall stability ofthe finished product. Figure 5.2 shows a schematic of the steps involved in particle coatingand film formation in a fluid bed–coating unit.

Coat integrity and subsequent release of the active require careful combination of sev-eral parameters such as air velocity, air temperature, spray rate, spray droplet size and soon. Jozwiakowski et al. (1990) published an excellent paper detailing the impact ofsubstrate’s physicochemical properties on coating quality and efficiency in a fluid bed sys-tem. Their study highlighted the importance of two types of interactions, namely

114 Chapter 5

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Top spray

(a)

Bottom spray(Wurster coating)

(b)

Tangential spray(rotor pellet coating)

(c)

Figure 5.1. Various configurations of fluid bed–coating systems: (a) top spray, (b) bottom spray(Wurster) and (c) tangential spray. (Courtesy of Glatt Air Techniques, with permission.)

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 114

Page 123: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

particle–particle and particle–machine, and concluded that an ideal substrate should pos-sess essential attributes such as spherical shape, uniform (high) bulk density, narrowparticle size distribution, and chemical stability.

Effect of Substrate’s Physicochemical Properties

It is critical to point out that film coating in a fluid bed system is applied on a weight basis.Therefore, to achieve same film thickness, larger amounts of shell material are needed to coatsmall particle cores (Madan et al. 1974). Coat thickness has been shown to be directly relatedto substrate’s particle diameter but inversely proportional to its surface area (Table 5.1).Particle shape, porosity, and friability can also play an important role in determining filmquality. Irregular-shaped particles such as crystals (salts, sodium bicarbonate) requirelarger amounts of coating (in excess of 80% of microcapsule’s weight) and can most oftenlead to nonuniform film formation.

In coating applications, particle–particle interactions manifest themselves in two differ-ent phenomena, agglomeration and attrition. Fluidization of wet fine particle cores (<100 µmdiameter) under intensive motion in the bed vortex can lead to particle–particle collision andagglomeration. The latter can be dramatically magnified when the fluid bed is operated attemperatures too close to the melting temperature of the coating material or when using veryhigh rates of spray coating. Ideal core particle size for fluid bed encapsulation ranges from

Encapsulation and Controlled Release in Bakery Applications 115

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Spraying Wetting Congealing Coated particle

Particle coating droplets Film formation

Figure 5.2. Film formation principle in a fluid bed–coating system. (Courtesy of Glatt Airtechniques, with permission.)

Calculated surface Number of particlesParticle diameter (µm) area of particles (n � 10–4) Wall thickness, T (µm)

235 624 17.7 0.26 � 0.02505 414 5.17 0.49 � 0.03715 292 1.82 0.64 � 0.02840 249 1.12 1.31 � 0.13

Table 5.1. Effect of particle size on wall thickness. (Adapted from Madan et al., 1974, withpermission from the publisher.)

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 115

Page 124: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

100 to 800 µm (assuming optimal bulk density). Larger size particles or pellets (>1 mm)can be coated readily, their repeated cycling in the bed may lead to particle abrasion andattrition. Such cores should be coated for only short time intervals with minimum bedmovement during the warming period (Lehmann and Dreher, 1981).

Figure 5.3 shows a scanning electron micrograph of typical coated particle surroundedby a lipid/wax wall material with the substrate particles completely engulfed in thelipid/wax shell.

Spray Chilling

Fluid bed coating described above is, in essence, an enrobing mechanism whereby one orfew particles (100–400 µm) are enveloped in a coating film, forming a reservoir-like sys-tem. As the temperature surrounding the capsule reaches the melting point of the wallmaterial, the entrapped particles are released. However, in the presence of slightest imper-fections in the shell material, the actual release tends to shift to “burst-like” behavior. Thelatter can have detrimental consequences upon storage and preparation of dough or battersystems, resulting in premature or uncontrolled release of the encapsulated active.

Spray chilling is an alternative technique that has been used for years in manufacturingstable pharmaceutical capsules with a unique matrix release mechanism. This technique isa solvent-free spray-drying method for encapsulating water-sensitive actives. In thisprocess, fine particles (typically <100 µm) are dispersed in a hot melt fluid (waxes, fattyacids) to form a homogeneous dispersion. The latter is atomized via spraying through apressurized single nozzle into a cooled chamber. The chamber temperature is set below themelting point of the mixture or its individual components using nitrogen or carbon dioxidegas. Ideally, spray chilling results in the formation of uniform spherical micropellets withsmooth surfaces that are water-impermeable but not water-resistive. These qualities areessential for better mixing owing to reduced surface tension between the microcapsule’shydrophobic surface and the batter’s aqueous environment. Due to the absence of solvent

116 Chapter 5

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 5.3. Surface morphology of a coated solid particle. (Courtesy of Balchem Corp., withpermission.)

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 116

Page 125: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

evaporation in spray chilling operations, the particles are generally non-porous andmechanically-strong so that they remain intact upon agitation (Gherbre-Sellassie, 1989).Nanosized particles can also be prepared using this process (Eldem et al., 1991). Additionalcoating is often applied to spray chilled congealed particles to ensure complete coverage ofthe microparticle and to eliminate undesirable interactions of exposed actives with theirsurroundings during storage and dough/batter preparation.

Conventional spray dryers with cooled air inlets can be used for spray chilling. Theapparatus consists of two main parts: (1) a cooling chamber and (2) an atomizer. For effec-tive spray chilling, it is recommended that the dispersion matrix has a very narrow meltingrange so that the particles can be held together during spraying.

Critical conditions for manufacturing uniform congealed capsules are: (1) low viscosityof the active and molten fat dispersion. Das and Gupta (1988) suggested that an ideal vis-cosity should be around 24 cP at 55°C and (2) high atomization speed to increase the per-centage of small micropellets (Scott et al., 1964; Deasey, 1984).

Release of actives from spray chilled microcapsules takes place via erosion and leachingthrough the matrix. Surfactants (depending on type and concentrations) can also dramati-cally affect matrix dissolution rates. John and Becker (1968) demonstrated that addition of4% of the non-ionic surfactant sorbitan monooleate resulted in enhanced release rates froma wax-congealed matrix; however, increasing the monooleate concentration to 10% led to areduction in rate of release.

Spray chilling suffers from one main drawback, that is, the rapid cooling rates can some-times crystallize the triglyceride matrix in the unstable α-polymorphic form, leading to theformation of disordered chains with undesirable orientation and, subsequently, low barrierproperties.

High-Pressure Congealing (Beta Process)

To overcome these drawbacks, a modified method was advanced by Verion Inc. (Redding,1995; Vaghefi et al., 2001). The process involves forming an active-matrix dispersion (e.g.,sodium bicarbonate dispersed in molten fat or wax) and further subjecting the mixture tohigh pressure (40,000–50,000 psi) for few seconds to intimately compress the mixture. Thesodium bicarbonate/fat mixture is then discharged through a spray nozzle into a chillingzone to congeal the molten fat material around the particles. The mixture is allowed tocycle through the system for multiple passes depending on the active load and/or desiredcapsule matrix consistency. The high pressure and high shear applied result in favorablechanges in the polymorphic structure of the treated fat or wax and in shifting its polymor-phic structure into the stable beta (β)-form, thus the name Beta process.

Redding (1995) studied the impact of heat, pressure, and their combinations on changesin the differential scanning calorimetric (DSC) profiles of tristearin (Figure 5.4) using theβ-process system. Commercially obtained native triglyceride displayed a β-melting peak at72°C. Upon melting and further resolidification, the DSC profile showed, in addition to theβ-peak, a new peak at 59.84°C corresponding to the α-form. Heating the tristearin to145°C along with pressure application (4400 psi) led to the complete elimination of theα-peak and the dominance of a stable β-peak at 75.73°C.

Similar to low-pressure congealing, complete coverage of the active particles locatedon the microparticle surface can be ensured by applying an outer coating layer via fluid-bedor other coating techniques. Capsules prepared, using this process, normally follow true

Encapsulation and Controlled Release in Bakery Applications 117

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 117

Page 126: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

zero-order release mechanism, a result of the slow erosion from the microcapsules thatform “tortuous” paths throughout the matrix. The shell material is not swellable and doesnot rely on osmotic pressure to release the core material.

Film-Forming Materials

A variety of fats and waxes are available for hot melt coating of leavening systems(Table 5.2) Lipid-based coating materials are available as pure components or most often as

118 Chapter 5

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

620 40 60 80 100

20 40 60 80 100

8

10

12

14

16

Temperature (°C)

20 40 60 80 100Temperature (°C)

Hea

t flo

w (w

/g)

Hea

t flo

w (w

/g)

59.84°C

77.86°C

1717.5

1818.5

1919.5

2020.5

Temperature (°C)

Hea

t flo

w (w

/g)

75.73°C

72°C

(a)

(b)

(c)

Figure 5.4. Effect of temperature and pressure on polymorphic profile of stearine: (a) native,(b) melted and resolidified, and (c) treated at 145°C and 4400 lb/in.2. (Reproduced from Redding,1995, with permission from the publisher.)

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 118

Page 127: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

functionally optimized composites (Kanig and Goodman, 1962; Kester and Fennema,1989a). Blends of lipids (different hydrophobicity, chain length, hardness, melting point,etc.), lipids and waxes, and lipids and polysaccharides can be adequately formulated toencapsulate actives for bakery and other food applications.

Waxes

Natural and synthetic waxes have been used in particle-coating applications. The mostcommonly used materials include: paraffin, carnauba, candelilla, beeswaxes and/or waxemulsions.Paraffin wax: Paraffin wax is derived from the wax distillate fraction of crude petroleum. Itis composed of hydrocarbon fractions of generic formula CnH2n+2 ranging from 18 to 32carbon units (Hernandez, 1994). Refined paraffin waxes can be used in specific coatingapplications (21 CFR, Code of Federal Regulations, 184.1973).Carnauba wax: Carnauba is a plant-derived exudate from the leaves of the Tree of Life(Copernica Cerifera) found mostly in Brazil. Carnauba wax consists mostly of saturatedwax acid esters with 24–32 hydrocarbons and saturated long-chain monofunctional alco-hols such as myricyl cerotate alcohols C9H59CH2OH.

Carnauba wax is the hardest natural wax available (hardness 4.7 cm � 10�2 for a 50 g/60 sec/25oC) and has the highest melting point and specific gravity of commonlyfound natural waxes (82–86oC). It is added to other waxes to increase melting point, hard-ness, toughness, and luster. However, carnauba wax is very brittle and lacks elasticity. Car-nauba wax is allowed for specific applications in food systems (21 CFR 184.1978).Beeswax: Beeswax is a secretory product of honey bees and is the basic material for combbuilding. Beeswax is harvested after removal of the honey by draining or centrifuging andfurther melting with hot water, steam, or solar heating. The wax is separated from impuri-ties by treating with diatomaceous earth and activated carbon. Beeswax is made up essen-tially of long-chain alcohols (C24–C33), hydrocarbons (C25–C33), and long-chain acids(C24–C34). Beeswax is very plastic at room temperature (melting point 61–65oC) but

Encapsulation and Controlled Release in Bakery Applications 119

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Table 5.2. Source and melting temperatures of selected group of waxes, lipids, and resincompounds used in particle-coating applications. (Adapted from various sources.)

Product Source Melting point (°C)

Beeswax Bees 61–64Carnauba wax Tree of life 82–86Candelilla wax Candelilla plant 65–69Shellac Laccifer lacca insect 115–120Lauric acid Coconut oil 44Capric acid Coconut oil 31.6Myristic acid Coconut oil, butter fat 54.4Stearic acid Most fats/oils 69.6Behenic acid Peanut oil 80Palmitic Most fats/oils 62.9Stearine Partially hydrogenated 61–64Cottonseed oilStearine Partially hydrogenated 66–70Soybean oil

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 119

Page 128: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

becomes brittle at colder temperatures. It is soluble in most other waxes and oils (Tulloch,1970; Bennett, 1975). Beeswax is considered a GRAS substance and is allowed for directuse with some limitations (21 CFR 184.1973).Candelilla wax: Candelilla wax is obtained from the candelilla plant that grows mostly inMexico and southern Texas. The wax is prepared by immersing the plant in boiling watercontaining sulfuric acid and further skimming off the surface, refining, and bleaching. Itsdegree of hardness is intermediate between beeswax and carnauba. It contains smallamounts of esters and free acids. This wax sets very slowly and takes several days to reachmaximum hardness. Candelilla wax is considered GRAS and is allowed for certain fooduses (21 CFR 184.1976).Wax macro- and microemulsions: Carnauba and beeswax owing to their high content ofalcohol and ether groups, can be microemulsified to form effective coating materials.Waxes are dispersed in water to form macro- or microemulsions via a process commonlyknown as inversion (Wineman, 1984).

Resins and Rosins

Shellac: Shellac resin is a secretion by the insect Laccifer lacca and is mostly produced incentral India. This resin consists of a complex mixture of aliphatic alicyclic hydroxyl acidpolymers, that is, aleuritic and shelloic acids. It is soluble in alcohol and alkaline media.Shellac resins can be blended with waxes to form improved moisture-barrier properties andincreased gloss for coated products. Shellac is not GRAS; it is only permitted as an indirectfood additive in food coatings and adhesives (21 CFR 175.300). Shellac is rich in car-boxylic acid residues and is highly water insoluble (Sward, 1972).

Glycol Polymers

Polyethylene glycols such as Carbowax (different grades, that is, viscosities 3350, 4600,8000) possess desirable coating properties, mainly their resistance to abrasion. Levels of15–40% were found to be useful for coating yeasts and extending their viability (Percel,1988).

Fats and Glycerides

A wide variety of commercially available triglycerides are used in coating applications.Naturally occurring food-grade fats are derived from animal or plant origin. Animaltriglycerides differ from plant triglycerides not only in the ratios of saturated to unsaturatedcarboxylic acids or their chain length but also in the location of the unsaturated fatty acid inthe glyceryl molecule. Variations in the carboxylic acid chain length, their melting profile,degree of saturation, degree of esterification, purity grades as well as their crystalline struc-ture can have a significant impact on coating processability as well as the performance ofthe encapsulated product.Natural oils and fats used in coating applications consist of one ormore of the three major fatty acid groups; these are lauric, palmitic, and oleic–linoleicgroups:Lauric acid group: Fatty acids of this group are highly saturated, rich in short-chain fattyacids (8, 10, and 14 carbon chain length), and are very stable. They contain 40–50% lauricacid on average. Oleic and linoleic acids constitute the majority of the unsaturated

120 Chapter 5

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 120

Page 129: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

fractions, while saturated ones are essentially made up of palmitic and stearic acids.Examples of this group include palm seed oil, canola, coconut, babassu, and palm kerneloils. Lauric-acid-based fatty acids have relatively low melting points (~44°C). Hydro-genated canola oil, for example, is composed of a triglyceride with extremely symmetrical,nonrandom structure, resulting in a hard and dry fat with good flowability at room temper-ature. Coconut oil has melting point of 24°C, while its hydrogenated counterpart melts at33°C (Bailey, 1952).Palmitic acid group: This group is represented by palm oil (from palm pulp) and palm ker-nel oil (from kernel). Palm oil contains 32–47% palmitic acid and 40–52% oleic acid. Palmoil has equal concentrations of the saturated and unsaturated fatty acids. Most of thetriglycerides (85%) of palm oil contain unsaturated fatty acids at the 2-position of the glyc-erol backbone (Bailey, 1952).Oleic/linoleic acid group: Commercially important oils in this group include corn, cotton-seed, peanut, olive, sunflower, safflower, and rice bran oil. These oils can be hydrogenatedto form plastic fats with different degrees of hardness. Most oils in this group are short- andmedium-chain unsaturated fatty acids. Only the highly-hydrogenated versions of thesefatty acids can be effective in particle coating applications.

Characteristics of Wax and Fat-Coating Materials

Chain Length

Long-chain fatty acids such as stearic (C 18:0) and palmitic (C 16:0) acids, by virtue oftheir high melting and apolar properties, have been used extensively in food coating appli-cations (Hagenmaier and Baker, 1991; Greener and Fennema, 1993; Kester and Fennema,1989b; McHugh and Krochta, 1994). Due to their strong H-bonding, long-chain fatty acidssuch as stearyl alcohols, stearic acid and beeswax crystallize into platelet-like densemicrostructures, commonly associated with effective moisture barrier properties (Figure 5.5).Longer-chain triglycerides (higher than 18 carbon atoms) such as arachidonic or behenicacids, however, show higher permeability presumably due to the heterogenous structure ofthe polymer network.

Polarity

Stearyl alcohol, a polar molecule, is a better barrier than its fatty acid counterpart, a resultof the lower affinity of the hydroxyl group for water than carbonyl and carboxyl groups.However, in most applications, other factors such as chain structure and its conformationshould be taken into consideration when choosing a lipid barrier material.

Degree of Unsaturation

The degree of unsaturation plays a considerable role in defining the crystal structure oftriglycerides and their mobility. For example, the area occupied by a molecule of oleic acidin a monolayer film is 0.48 nm2, whereas for stearic acid, it is 0.23 nm2 (Kamper andFennema, 1984). Despite this fact, oleic acid displays greater mobility owing to the doublebond that favors the diffusivity of water molecules compared to stearic acid, which is a fullysaturated carboxylic acid (Gontard et al., 1994).

Encapsulation and Controlled Release in Bakery Applications 121

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 121

Page 130: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Solid Fat Index

Solid fat index (SFI) has been directly related to fat hardness and its spreadability (Bailey,1952; Vreeker et al., 1992). High solid fat concentration (0–30%) improves barrier proper-ties of fat-based films but increases their hardness (Narine and Margoni, 2002). High solidfats have better barrier properties than their corresponding liquid or semi-solids due to theformation of dense structure and greater volume that limits the diffusion of water (Perronand Ollivon, 1992). However, at very high solid fat concentration, higher than the criticalvalue, permeability could increase due to structural defects within the film.

Hydrophobicity

Hydrophobicity has been used as a criterion for predicting water barrier properties of fatsand waxes. Kester and Fennema (1989a, b) proposed the following order of decreasingmoisture barrier effectiveness of waxes and fats: beeswax, stearyl alcohol, acetyl glycerols,hexatriacontane, tristearin, and stearic acid, a reflection of their decreased hydrophobicity.Avner and Blatt (1990) indicated that despite the similarities in melting temperaturesbetween hydrogenated castor oil and calcium stearate/stearic acid blend (meltingpoint~86°C), hydrogenated castor oil provided higher thermal stability and barrier proper-ties due to the superior hydrophobic character of castor oil.

Polymorphism (Crystallization Behavior)

Triglyceride molecules are naturally arranged in a “tuning fork” structure where three fattyacids are more or less parallel, one pointing in the opposite direction of the others. How-ever, the complexity and flexibility of triglyceride molecules allow different crystallinepacking of the same ensemble of molecules, leading to the existence of different poly-morphs. During crystal growth, such molecules will pack more easily side to side than end

122 Chapter 5

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 5.5. Cross section of wax-coated particle showing platelet-like microstructure. (Courtesyof Balchem Corp., with permission.)

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 122

Page 131: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

to end with glyceryl alcohols. The resulting individual crystals appear needle shaped underthe microscope (Bailey, 1952; van de Tempel, 1961).

Fat polymorphism, that is, their ability to crystallize under several forms, also affectstheir barrier efficiency. Triglycerides crystallize into α (hexagonal), β (triclinic), and β′(mainly orthorhombic) forms. The latter two crystals are stabilized by London forcesoccurring between the aliphatic chains, which give rise to dense networks. No such forcesoccur in the α-form. The carbon atom can rotate (small angles), leading to a hexagonalnonstable structure (Sato and Kuroda, 1987 and therein; Ubbelohde, 1978). The β-form,represented by lard and tallow, has the highest melting point and highest order (large andcoarse crystals 25–50 µm long). β′-crystals, however, provide the most functional formowing to their small size (less than 1 µm), thin needle-shaped morphology, and interlockingstructure (Roberts et al., 2000). Polymorphic states of a substance have different physicalproperties but on melting yield identical liquids since these states are merely due to differ-ences in packing of the constituent molecules upon crystallization.

Melting Point

Most naturally occurring fats show multiple melting points reflected in several meltingpeaks in their DSC profile. Low melting fats most often result in inferior coating qualitiesdue to their poor flowability and tendency to form clumps. On the other hand, high meltingfats such as palm stearine may cause difficulties in spray coating encapsulation due to theirstiffness and lack of plasticity. The melting point of polymorphic forms of stearine rangefrom 65°C (α) to 70°C (β′) and 72°C (β) (Bailey, 1952).Low melting lipids are useful forcoating heat-labile actives such as yeasts, enzymes, vitamins, probiotic bacteria and so on.High melting lipids, on the other hand, are suitable for encapsulating chemical leaveningagents, acids, and other less heat-sensitive actives.

Despite the positive impact of high melting fats on fluid bed–coating efficiency, coatingperformance of a given fat is a function of the melting profile and not necessarily the tem-perature of the melting peak. It has been a common practice to source pure fats/waxes(fewer components) in order to provide a single narrow-shaped peak; in practice coatingconditions, in particular the rate of fat/wax cooling, are as critical in determining the sharp-ness or broadness of the melting peak.

The mechanical properties of lipid-based films can be improved by modifying either thefilm’s melting profile or its structural properties. Two classes of materials have been used tomodify the performance of fats, namely, waxes and plasticizers:

1. Waxes can be successfully used to raise the melting point of natural fats, thereby reduc-ing tackiness and enhancing their coating performance. For such applications, it is criti-cal to maintain the fat/wax blend under continuous mixing and at temperatures slightlyabove the wax-melting point to avoid wax gravity settling and subsequent plugging ofthe spray nozzle. In the petroleum industry, the temperature at which wax crystals startto appear when temperature falls to a critical level referred to as wax appearance tem-perature, WAT (Azvedo and Teixeira, 2002). Prolonged heating of wax/fat mixtures,however, may lead to fat oxidation; therefore caution should be exercised and wheneverpossible, incorporating antioxidants or blanketing the hot melt container with nitrogencan be very effective in retarding fat oxidation and/or degradation.

Encapsulation and Controlled Release in Bakery Applications 123

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 123

Page 132: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

2. Plasticizers such as acetoglycerides, glycerin, monoglycerides, alcohols, phospholipids(lecithins) and ester derivatives of glycerols can be used to lower the melting point offats and waxes, thus facilitating their atomization from the spray nozzle. Plasticizers canalso help in reducing film brittleness and enhancing its flexibility and mechanical prop-erties without any significant impact on the melting point of the wax. Litwinenko et al.(2004) reported that addition of glycerol increased the mean crystal size of fats up to aconcentration where the crystal size decreased with increased glycerol concentration(Table 5.3).

Films made from molten beeswax are often smooth and uniform, whereas those made withalcohol are rough and irregular with apparent large size globules (Greener and Fennema,1993). Incorporation of polysaccharide molecules such as ethyl cellulose into fat-basedfilms can be used to provide additional film toughness. The latter is presumably a result ofthe proper orientation of the wax crystallites parallel to the polysaccharide support base.

Ideal Properties of Encapsulated Particles for Bakery Applications

Ideal microcapsules manufactured for baking applications should possess the followingessential properties.

Good Barrier Properties

Fats of large and closely packed crystals are favored for their high moisture barrier proper-ties. As discussed above, barrier properties are a combination of fat/wax chain length,polarity, polymorphic form, film flexibility, and so on.

Flexibility

Fat crystals form a particular class of soft materials that demonstrate yield stress and vis-coelastic properties, that is, plastic-like materials (Narine and Marangoni, 1999; 2002).Waxes, on the other hand, form stiff films that tend to become fragile, especially if storedfor long periods. Film stiffness could also lead to microcapsule fracture and rupture duringblending.

124 Chapter 5

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Table 5.3. Effect of glycerol on microstructural parameters determined by image analysis.(Adapted from Litwinenko et al., 2004, with permission from the publisher.)

Crystallization temperature (�C) Glycerol (%) Mean particle size (µm2) Number of particles (N)

Following storage for 24 h at 5°C 0.00 40 48450.03 30 46300.10 86 33000.25 69 3646

Following storage for 15 min at 5°C 0.00 163 13860.03 302 6850.10 326 6790.25 150 1555

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 124

Page 133: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Mechanical Strength

Good mechanical properties are critical for processing intact microcapsules without fracture.Edible fats have inherently poor mechanical strength. The latter can be enhanced by incorpo-rating waxes, by modifying their hydrophobic character (Narine and Marangoni, 1999), or byaddition of polar substances such as hydrocolloids (Chen and Nussinovitch, 2000).

Surface Morphology

Smooth surfaces that are free of cracks or crevices are ideal for formulating bakery prod-ucts with microencapsulated actives and for controlling their release, that is, by retardingmicrocapsule leaking and premature release.

Particle Size Distribution

Uniform particle size distribution is critical for many applications, especially in situationswhere only a very narrow melting window is available. Very large microparticles or micro-capsules can cause localized effects such as failure to distribute evenly and eventually formimmobile melted masses of barrier material. Very small particles, on the other hand, will havea large surface area which speeds up melting of the coating material upon baking the product.

Film Thickness

In encapsulation and film coating, one critical decision to be made is how much coating isnecessary to achieve desired properties in the finished product. Applied films should bethick enough to overcome surface imperfections but not to modify the release behavior ofthe microcapsule. For stable dough and batter systems, coatings in the range of 50–95% arecommon. It should be noted that high film thickness can modify the release of actives in anunpredictable manner and at extreme levels, the release rate tends to become dependent onthe size of the core, regardless of the film thickness.

Melting Properties

Choice of suitable melting properties of the coating material should take into considerationthe bakery product’s desired storage and processing conditions. Figure 5.6 shows a typicaltemperature/time profile of baked batter in a conventional oven. An ideal leavening micro-capsule for this application should have a melting profile that closely mirrors that of thebaked product. Melting of the shell material should commence as the product temperatureapproaches or attains 200ºF (�93ºC) and should be completed around the first 13–14 minof the baking cycle, that is, before the baked product structure is fully set.

Miscellaneous Examples of Encapsulation and Controlled Release in Bakery Applications

Yeasts

Yeasts used in baking applications include mainly Saccharomyces cerevisiae, Saccha-romyces rosei, Saccharomyces exiguous, and Candida milleri. In the absence of oxygen,

Encapsulation and Controlled Release in Bakery Applications 125

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 125

Page 134: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

yeast cells metabolize carbohydrates, via multiple intermediate steps, to produce alcohol,energy, and carbon dioxide gas (Equation 1). The latter is essential for building volume andcell structure in the baked product. Yeast activity takes place during dough preparation andcan sharply increase as temperature rises upon preparation and baking (rate doubles forevery 10°C rise in temperature).

Yeast fermentationC6H12O6 → 2C2H5OH � 2CO2 � 234 kJ (1)

Baker’s yeast is available in many forms: active dry yeast (ADY), inactive dry yeast,compressed yeast, cream yeast, crumbled yeast, and protected dry yeast (Fleischmann’s,2001). Although active dry yeast (ADY) has a relatively low moisture content (7.3–8.3%),it is sensitive to oxygen and is always distributed in hermetically sealed pouches under vac-uum or flushed with liquid nitrogen. Cream yeast and compressed yeast are usually storedat subfreezing temperatures to maintain their activities. However, problems arise if any ofthese yeast forms is incorporated into a dough system before freezing because of uncon-trolled fermentation and dough expansion (Reed and Nagodawithana, 1991).

Many attempts to improve the keeping qualities of Baker’s yeast have been documented(Pelletier and Roger, 1989). Addition of hydrophilic agents (starch, locust bean gum) toyeast preparations to bind up water and potentially retard endogenous metabolic processeshas not proven to be very effective. Other technical approaches involve immobilization inchemically cross-linked chitosan beads or activated carriers (Donova et al., 1993;Markvicheva et al., 1991; Freeman and Dror, 1994; Shimon et al., 1991). Luca et al.(1979)) patented a process for treating fresh yeast with hydrophobic silicon dioxide(0.2–1%) to form a colloidal dispersion that was claimed to help maintain the yeast stableat high moisture contents. Soltis and Sell (1998) developed a method for encapsulatingBaker’s (cream) yeast and forming a shelf-stable product that can be safely stored at ambi-ent conditions, thus eliminating the need for costly refrigerated storage. Their method con-sists essentially of pre-adsorbing the high moisture cream yeast onto food fiber, such asgrain or bean hulls, followed by coating with a thermoplastic hydrophobic material. Thesystem was claimed to provide a means for delaying the release of active yeast cells untillater in the baking process.

Newly developed forms of ADY (high ADY) possess high fermentative power and areinstantized to allow their incorporation into dough systems without rehydration. However,

126 Chapter 5

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Muffin temperature development

0

50

100

150

200

250

1 3 5 7 9 11 13 15 17 19 21 23 25Time in oven (min)

Bat

ter t

empe

ratu

re (°

F)

Figure 5.6. Temperature development of baked muffin over time.

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 126

Page 135: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

this process results in porous granules that may expose the yeast to undesirable conditions,resulting in their inactivation within three days. Percel (1988) devised a method for preserv-ing the viability of instant yeast via coating (15–40% w/w) with polyethylene glycol of dif-ferent molecular weights (Carbowax 3350, 4600 and 8000) under mild temperatures of54–63°C using fluid bed coating. These experiments showed that Carbowax 4600 providedmost immediate release of the yeast in cold water while Carbowax 8000 resulted in yeastmicropartculates with most abrasion resistance and stability (several weeks in dry mixescompared to few days for the unencapsulated yeast). Fuglsang et al. (2002) used liposomesystems to entrap yeast and inhibit their release at ambient temperature. Upon heating from25 to 60°C, the lipid undergoes phase change, thus releasing the encapsulated yeast material.

Chemical Leaveners

Manufacturing consistent quality bakery products, especially in large volume operations,relies on using chemical leaveners such as baking powders for cakes, muffins, and cookiesand to replace yeasts in bread and dough formulations. Chemical leavening agents operatedifferently from yeast; while yeast requires sitting time after thawing and prior to baking toproduce carbon dioxide, a chemical leavening system produces CO2 gas during baking toexpand the dough and create cellular microstructure.

Chemical leavening agents comprise a long list of acids and alkalis that vary in their reac-tivity, stability, solubility, as well as other attributes. (Church and Dwight, 1999). Leaveningsystems comprise two main components, leavening acid such as sodium acid pyrophosphate(SAP) and base pair such as sodium bicarbonate (soda) that when allowed to react in an aque-ous medium (in some cases in the presence of heat) results in the formation of CO2. In suchsystems, the bicarbonate will provide the gas while the acid will control the reaction rate.

Common leavening agents are classified into three categories: (1) slow acting, (2) fastacting, and (3) double acting. Sodium aluminum sulfate (SAS) and monocalcium phos-phate monohydrate (MCP) form a double-acting system. Slow-acting systems include acombination of soda and SAP, where baking temperatures affect CO2 release. In fast-actingsystems (soda and MCP), CO2 evolution and its release occur effectively during mixing andstanding. Deciding which component of the leavening system to encapsulate will dependgreatly, among other aspects, on whether the product is a dry mix, bread dough, or a highwater activity batter. In high water activity formulations, encapsulating the soda componentis more feasible since the acid component can help provide additional antimicrobial protec-tion to the batters.

Chemical leavening agents are sensitive to moisture but much less to storage and prepara-tion temperatures; therefore, encapsulating chemical leaveners in a hydrogel system, forexample, would not be an option owing to the risk of dissolving the active prior to its point ofapplication. Hydrophobic coatings such as fats and waxes constitute the most commerciallyviable encapsulating media. Upon heating, the shell melts, thus allowing the leaveningactive to become available and ultimately to release CO2 needed for building volume andcell structure of the baked product. The rate of capsule melting and CO2 evolution are criti-cal parameter for determining volume, density, and textural qualities of the baked product.This step must occur within narrow limits for some applications such as in the preparation ofcanned doughs. Manipulating the release rate can be achieved by choosing suitable fatsand/or waxes with adequate melting temperatures as well as melting profile, coat thickness,encapsulation technology, and release mechanism (reservoir vs. matrix vs. combination).

Encapsulation and Controlled Release in Bakery Applications 127

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 127

Page 136: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

For most bakery systems, encapsulating chemical leaveners via particle coating requiresthe application of fairly high levels of coating materials onto the crystalline core; idealcoat:core ratios range from 50:50 to 95:5 to ensure complete coverage of the active’s sur-face. Canned self-sealing doughs represent an exceptional application, where the presenceof small amounts of partially-coated or uncoated leavening is desirable. In such situations,the partially-coated or uncoated agents react to form CO2 during or immediately followingpackaging of the can, thus purging out air and providing a seal from inside the package. Thefully-coated portion of the leavening microcapsules is preserved for further reaction duringbaking. Care should be taken not to allow excessive evolution of CO2 and over-expansionof the pressurized dough.

Huang et al (1989) cited an interesting advantage of encapsulating leaveners formicrowave baked products i.e. their ionic interactions with the dough components and thesubsequent positive impact on reducing gluten toughness and starch firmness, importantattributes to forming stable matrix for stabilizing generated CO2.

Assessing the stability of encapsulated sodium bicarbonate is achieved by tracking therelease of carbon dioxide in sealed packages or containers as well as changes in pH,appearance, and other sensory attributes of the stored product. Figure 5.7 shows a compari-son of the risograph gas evolution in refrigerated doughs made using two commerciallyencapsulated soda products and one unencapsulated control, with E-soda 1 displayinghighest stability (lowest CO2 release during storage) while unencapsulated soda showingthe least stability (Domingues et al., 2003).

Pacifico (2003) suggested that leach rate upon baking (rate at which an encapsulatedagent seeps out from the capsule) can be a useful indicator of the functionality of encapsu-lated leaveners. Accordingly, high leach rate of congealed and coated actives may enhancetheir reactivity and does not necessarily imply ineffective encapsulation.

Several other publications and inventions have surfaced in the last decade claimingingredients and methods for manufacturing chemically-leavened shelf stable bakery

128 Chapter 5

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

0

10

20

30

40

50

60

0 200 400 600 800 1000 1200 1400

Time

CO2

(%)

free CO2

E1-Soda

E2-Soda

Figure 5.7. Release of carbon dioxide from refrigerated dough package made with unencapsulated and two encapsulated soda samples (E1-soda and E2-soda) and stored at 45°F for six weeks. (Reproduced from Domingues, 2003.)

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 128

Page 137: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

products (Book et al., 2000; Chung and Lavault, 1995; El-Afandi and Citti, 2006;Kringelum et al., 1999; LaBell, 1999; Dorko and Penfield, 1993; Redding, 1995; Reddingand Bruce, 2002; Tuazon and Foster, 1992; Wu et al., 2000); each of these inventions isdirected to a specific bakery application.

Dough Conditioners

Bread staling and loss of freshness are major economical hurdles for the baking industry. Sev-eral underlying mechanisms have been speculated, with the amylose molecular rearrange-ments theory being the most plausible. During baking, starch is gradually transitioned froman amorphous structure to a partially crystalline state, a result of inter- or intramolecularinteractions via H-bonding of the amylose and amylopectin fractions. Upon recrystallization(retrogradation), starch releases water and the crumb becomes very firm and stale.

Storing baked products at room temperature or under high (safe) relative humidity candelay staling, but only for few hours. Emulsifiers are commonly used to help retard staling,though not very effectively; their mechanism of action is believed to be via softening thebread and reducing its firmness rather than retarding starch retrogradation.

Amylases that modify starch responsible for staling can be used effectively for increasingshelf life of bread via hydrolysis of the glycosidic linkages in polyglucans. Most commonlyused α-amylases are derived mainly from Aspergillus oryzae, Bacillus subtilis, and Bacillusstearothermophilus. These enzymes act on damaged starch particles, thus lowering doughviscosity and producing fermentable sugars necessary for larger bread volume and softerloaves. The Bacillus-derived amylases, however, are fairly heat stable and therefore do notget inactivated during baking, resulting in excessive breakdown of starch and the formationof very moist and sticky crumbs that are difficult to control. Encapsulating these amylases,therefore, can help control their enzymatic activity and reduce their damaging effect onstarch. Encapsulating amylases in lipid films has been suggested for their sustained releaseduring baking and shelf life of the baked product (Cole, 1983; Horn, 2002; Schuster et al.2001; Mori et al., 2002).

To ensure even distribution of dough ingredients, manufacturers may sometimes extendthe time of mixing. In certain formulations, this overmixing can result in doughs beyondtheir peak viscosity, thus adversely affecting their viscoelastic properties. Fuglsang et al.(2002) developed a process for encapsulating xylanase enzymes (dough strengtheners) intomicelles to help reduce dough stickiness and improve its handling and machinability.Release of the enzyme was designed to be initiated by melting the coating material duringleavening or early baking where enzyme activity is desired. Pan breads and flat breads arethe most common target applications.

Antimicrobial Agents

A wide range of antimicrobial agents has been used traditionally for controlling microbialgrowth in shelf-stable bakery products such as sorbates, benzoates, propionates, paraben,nisin, fumaric acid, and in some cases food grade metabolites produced by Propionibac-terium sp. However, effective concentrations of these substances can dramatically affect theflavor, color, odor, and textural attributes of bakery products. Another drawback of usingantimicrobial agents is their negative impact on viability of yeasts and enzymes used inbakery systems. Acidic antimicrobial agents, sorbates in particular, can also disturb the

Encapsulation and Controlled Release in Bakery Applications 129

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 129

Page 138: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

chemical-leavening balance of doughs and batters. Their reducing power can lead to irre-versible changes in the rheological properties of dough and batter systems.

An alternate approach for retarding microbial growth of doughs and batters is via pHadjustment (reducing pH depending on type of bacteria and product storage conditions).Although reducing pH can inhibit growth of bacteria and other microorganisms, it has noeffect on growth of fungus. Generally, a pH of 5 is acceptable for refrigerated doughs,whereas ambient shelf-stable products require a lower pH (<4). The latter can have an unfa-vorable reducing effect on the rheological properties of the dough or batter systems.

Natamycin is an extremely effective natural antifungal polyene macrolide that can beproduced by fermentation of the bacterium Streptomyces natalensis. However, its activitycan be negatively affected by extreme pH conditions and in the presence of metals. In addi-tion, natamycin is very antagonistic to yeasts and moulds. Several encapsulation and con-trolled release formulations have been documented in the patent literature for mitigatingthese undesirable effects (Kringleum, 1999; Thomas et al., 2005) mainly via coating thepreservative with a food-grade high-melting hydrophobic substance and its further disper-sion into bread dough system. Methods for encapsulating natamycin using a variety ofmatrices via extrusion, liposomes, coacervation have been developed (Thomas et al., 2005)and claimed sustained release of the antifungal agent into yeast-leavened dough with noadverse effects on the yeast. Koontz and Marcy (2003) reported successful entrapment ofnatamycin into a γ-cyclodextrin molecule and formation of stable natamycin/γ-cyclodextrininclusion complex, despite the incomplete lodging of the bulky natamycin in the γ-cyclodextrin host.

Flavors

Encapsulation of flavors has been used in baking applications to retard flavor losses duringbaking and/or eliminate their undesirable interactions with dough components. One groupof flavors including cinnamon, cloves, allspice, and nutmeg is known to have negativeeffect on yeast-leavened doughs, resulting in deteriorating the yeast’s rising properties aswell as overall quality of the final baked product. The S-containing major components ofcinnamon oil can also have a negative impact on gluten development due to their interac-tions with disulfide moieties of gluten. Black et al. (1988), developed an encapsulationcomposition that is claimed to allow incorporation of cinnamon flavor to baked productsand to retard the associated undesirable interactions. Wampler (1995) patented a coacervation-based flavor composition to deliver a high payload (70–95% flavor oil). via a cross-linkedgelatin shell. The composition was claimed to be stable during mixing with other doughingredients and subsequent baking.

Sweeteners

Conklin et al. (1987) developed a microencapsulated particulate sweetening system that isthermally stable and is suitable for bakery applications. The composition is essentially awater-swellable structure containing aspartame/acid core. Co-packing the food acid withaspartame can create adequate pH (<5) conditions in the aspartame’s microenvironment,thus stabilizing the dipeptide. The composition was claimed to be made very compact toreduce wetting via capillary and can also be coated with a hydrophobic substance to delaybut not block the diffusion of water.

130 Chapter 5

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 130

Page 139: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Further Remarks

• Generally speaking, encapsulation of leavening agents, especially chemical leaveners,requires applications of higher levels of coating than most traditional applications (up to95% coating may be required).

• Reproducibility of production-scale microcapsules is often an issue, especially formoisture-labile actives such as sodium bicarbonate. Regardless of the technology ormaterials used, leaky capsules may still be generated. Severity of this problem varieswith the end-product application.

• Manufacturers should be aware of potential differences in performance of encapsulatedingredients in conventional baking compared to microwave baking.

• Judging the stability of an encapsulated leavening system should be done by monitoringchanges in CO2 generated as well as pH of the dough/cake batter. The latter can be mostaccurately be determined few hours after preparing the dough or batter.

• Incorporating additional amounts of leavening acid into the formula (slightly higherthan needed during baking) may help make up for any initial neutralizing reaction thatmight have occurred upon mixing the batter.

• For shelf-stable doughs or batters packaged in containers, injection of an inert gas suchas nitrous oxide that is partially soluble in the dough can help produce extra amounts ofgas bubbles to compensate for any build up of viscosity and density of batters. The lattermay result from reactions with the protein and starch components of the dough/battersystem.

• Chemical reactivity of leavening systems does not necessarily stop in dry mixes. Con-densation reactions can be a problem even at very low moisture levels, which can accel-erate other reactions. The role of water in low moisture systems can be morechallenging.

• The choice of a technology and/or material(s) for encapsulating actives for bakery appli-cations depends on a host of factors such as type of finished product, desired packagingand shelf life, release trigger, site, rate of release, cost and so on. For successful micro-capsule design, it is imperative to determine whether the active is sensitive to moisture,water vapor, oxygen, high temperatures, pH, or other environmental parameters.

ReferencesAvner, R. and Blatt, Y. 1990. Microcapsules containing food additives and their use. EP 411,326 A2.Azvedo, L.F. and Teixeira, A.M. 2002. A critical review of the modeling of wax deposition mechanisms. Presented

at the AIChE 2002 Spring National Meeting, New Orleans, LA, 10–14 March, 2002.Bailey, A.E. 1952. Melting and Solidification of Fats. New York, Interscience Publishers.Bennett, H. 1975. Industrial Waxes. Chemical Publ. Co., New York, NY.Black, M., Popplewell, L.M. and Porzio, M. 1988. Controlled release encapsulation composition. US Patent

5,756,136.Book, S., Corliss, G. and Heidolph, B. 2000. Process and formulation for a chemically leavened dough or bakery

product. US Patent 6,149,960.21 CFR, Code of Federal Regulations(§.184.1973; § 184.1978 and §184.1976).Chen, S. and Nussinovitch, A. 2000. Permeability & roughness determinations of wax-hydrocolloid coatings &

their limitations in determining citrus fruit overall quality. Food Hydrocolloids 15: 127–137.Chung, F.H.Y. and Lavault, S., M.-P. 1995. Novel encapsulated leavening acid composition. EP 0,699,392 A2.Church and Dwight Co, Inc.1999. Leavening Sales brochure.Cole, M. 1983. Antistaling baking composition. US Patent 4,416,903.

Encapsulation and Controlled Release in Bakery Applications 131

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 131

Page 140: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Conklin, J.R., Gressgott, H.L. and Wolford, T.D. 1987. Thermally stable particulate artificial sweeteners. EP 0,229,730 A2.

Das, S.K. and Gupta, B.K. 1988. Optimization of controlled drug release through micropellitization. Drug Dev.Ind. Pharm. 14(12): 1673–1697.

Deasey, P.B. 1984. In Microencapsulation and Related Drug Processes (P.B. Deasy, Ed.), Marcel Dekker, NewYork, p. 181.

Domingues, D.J. 2003. Chemical leavened doughs and related methods. WO 03/020,044 A1.Donova, M.V., Kuz’kina, I.F., Arinbasarova, A.Y., Pashkin, I.I., Markvicheva, E.A., Baklashova, T.G.,

Sukhodoiskaya, G.V., Pokina, V.V., Kirsh, Y.E., Koshcheyenko, K.A. and Zubov, V.P. 1993. poly-N-vinylcapro-lactam gel: a novel matrix for entrapment of microorganisms. Biotechnol. Tech. 7(6): 415-422.

Dorko, C.L. and Penfield, M.P. 1993. Melt point of encapsulated sodium bicarbonates: effect on refrigerated batterand muffins baked in conventional and microwave ovens. J. Food Sci. 58(3):574–578.

El-Afandi, A. and Citti, J. 2006. Refrigerated dough and product in low pressure container. US Patent2006/0,177,558 A1.

Eldem, P., Speiser, P. and Hincal, A.A. 1991. Optimization of spray-dried and –congealed lipid micropellets andcharacterization of their surface morphology by scanning electron microscopy. Pharm. Res. 8(1): 47–54.

Fleischmann’s Yeasts. 2001. Product directory.Freeman, A. and Dror, Y. 1994. immobilization of “disguised” yeast in chemically cross-linked chitosan beads.

Biotechnol and Bioeng. 44: 1083-1088.Fuglsang, C., Callisen, T. and Budolfsen, G. 2002. Dough composition comprising a lipid-encapsulated enzyme.

WO 02/19,828 A1.Gherbe-Sellassie, I. 1989. In Pharmaceutical Pelletization Technology (I. Gherbe-Sellassie, Ed.), Marcel Dekker,

New York, p. i.Gontard, N., Duchez, C., Cuq, J.L. and Guilbert, S. 1994. Edible composite films of wheat gluten and lipids: water

vapor permeability and other physical properties. Int. J. Food Sci. Technol. 29: 39–50.Greener, I. and Fennema, O. 1993. Water vapor and oxygen permeability of wax films. JAOCS 70: 867–873.Hagenmaeir, R. and Baker, R. 1991. Reduction in gas exchange of citrus fruit by wax coating. J. Agric. Food

Chem. 41: 283–287.Hernandez, E. 1994. Edible Coatings from Lipids & Resins. Chapter 10: 279- in Edible Coatings & Films to

improve Food Quality (J. Krochta, E.A. Baldwin and M.O. Nisperos-Carriedo, Eds.). Technomic PublishingCo., Lancaster, Basel., pp. 279–303.

Horn, M.C. 2002. Methods and compositions for retarding the staling of baked goods (US 2002/0,058,086 A1).Huang, V.T., Hoseney, R.C., Graf, E., Ghiasi, K., Miller, L.C., Weber, J.L., Gaertner, K.C., Matson, K., Hunstiger,

A.M., Rogers, D.E. and Saguy, I. 1989. Starch-based products for microwave cooking or heating. EP 0,617,896 A2.Ichikawa, H., Kaneko, S. and Fukumori, Y. 1996. Coating performance of aqueous composite lattices with

N-ispropylacrylamide shell and thermosensitive permeation properties of their microcapsule membrane. Chem.Pharm. Bull. 44(2): 383–391.

John, P.M. and Becker, C.H. 1968. Surfactant effects on spray-congealed formulations of sulfaethylthiadiazole-wax. J. Pharm. Sci. 57(4): 584–589.

Jozwiakowski, M.J., Jones, D.M. and Franz, R.M. 1990. Characterization of a hot-melt fluid bed coating processfor fine granules. Pharm. Res. 7(11): 1119–1126.

Kamper, S. and Fennema, O. 1984. Water vapor permeability of an edible fatty acid bilayer film. J. Food Sci. 49:1482–1485.

Kester, J.J. and Fennema, O. 1989a. Resistance of lipid films to water vapor transmission. JAOCS 66: 1139–1146.Kester, J.J. and Fennema, O. 1989b. Tempering influence on oxygen and water vapor transmission through a

stearyl alcohol film. JAOCS 66: 1154–1157.Koontz, J.L. and Marcy, J.E. 2003. Formation of natamycin:cyclodextrin inclusion complexes and their character-

ization. J. Agric. Food Chem. 51: 7106–7110.Kringelum, E. 1999. Compositions containing encapsulated food additives and their use. WO 99/08,553.LaBell, F. 1999. Encapsulated acid improves flour tortilla quality. Prepared Foods October: 91.Lehmann, K. and Dreher, D. 1981. Coating tablets and small particles with acrylic resins by fluid bed technology.

Int. J. Pharm. Tech. and Prod. Mfr. 2(4): 31–43.Litwinenko, J.W., Singh, A.P. and Marangoni, A.G. 2004. Effects of glycerol and Tween 60 on the crystallization

behavior, mechanical properties and microstructure of a plastic fat. Crystal Growth and Design 4(1): 161–168.Luca, S.F., Thommel, J. and Bronn, W.K. (1979). Free-flowing powdered fresh baker’s yeast preparation and

method of producing it. US Patent 4,160,040.

132 Chapter 5

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 132

Page 141: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Madan et al. 1974. Microencapsulation of a waxy solid: wall thickness and surface appearance studies. J. of Phar-maceutical Sci. 63(2): 280–284.

Markvicheva, E.A., Kuz’kina, Pashkin, I.I., plechko, T.N., Kirsh, Y.E. and Zubov, V.P. 1991. A novel technique forentrapment of hybridome cells in synthetic thermally reversible polymers. Biotechnol. Tech. 5(3): 223-226.

McHugh, T.H. and Krochta, J.M. 1994. Milk protein-based edible films and coatings. Food Technol. 48(1):97–103.

Mori, S., Sato, K. and Tanaka, N. 2002. Dough composition and preparation thereof. US Patent 6,355,282 B1.Narine, S.S. and Marangoni, A.G. 1999. Relating structure of fat crystal networks to mechanical properties: a

review. Food Research International 32: 227–248.Narine, S. S. and Marangoni, A.G. 2002. Structure and mechanical properties of fat crystal networks. Advances in

Food and Nutrition Research 44: 33-145.Pacifico, C.J. 2003. Chemical leavening ingredient. US Patent 2003/0,031,773 A1.Pelletier, R. and Roger, F. 1989. Process for using bakery additives and bakery yeast. WO 89/00,009.Percel, P.J. 1988. Encapsulated yeast. US Patent 4,719,114.Perron, R. and Ollivon, M. 1992. Proptriétés physiques des corps gras. Propriétés générales de la châine hydrocar-

bonée. (A. Karleskind, Ed.), Manuel Des Corps Gras, Paris: Tec & Doc Lavoisier, 433–442.Redding, B.K. 1995. Method for entrapment of liquids in transformed waxes. US Patent 5,460,756.Redding, B.K. and Bruce, K. 2002. Ready-to-use food product. WO 02/11,544 A1.Reed, G. and Nagodawithana, T.W. (Eds.). 1991. Bakers yeast production, pp. 261–314. In Yeast Technology, 2nd

edition. AVI Publishers, New York.Roberts, B.A., Scavone, T. A. and Riedell, S.P. 2000. Beta-stable low-saturate, low trans, all purpose shortening.

US Patent 6,033,703.Sato, K. and Kuroda, T. 1987. kinetics of melt crystallization and transformation of tripalmitin polymorphs.

J AOCS 64(1): 124–127.Schuster, E., Sprossler, B. and Hofmeister, J. 2001. Process for making baked articles that retain freshness. US

Patent 6,254, 903 B1.Scott, M.W., Robinson, M.J., Pauls, J.F. and Lentz, R.J. 1964. Spray congealing: particle size relationships using a

centrifugal wheel atomizer. J. Pharm. Sci. 53: 670–675.Shimon, L.M., Kotorman, M. and Sayani, B. 1991. immobilization of yeast alcohol dehdrogenase on a p-benzo-

quinone-actiavted silicate carrier. Prikladnaya Biokhimiya i Mikrobiologiya 27 (1): 86-90.Soltis, J. and Sell, J.L. 1989. Yeast composition. US Patent 5,70,669.Sward, G.G. 1972. Natural resins. Am. Soc. Test Mat: 77-91.Thomas, L.V., Gouin, S., Tse, K.L. and Hansen, C.B. 2005. Natamycin dosage form, method for preparing same

and use thereof. US Patent 2005/0,042,341 A1.Tuazon, M.T. and Foster, L.C. 1992. Souffle mix. EP 0,545,025 B1.Tulloch, A.P. 1970. The composition of beeswax and other waxes secreted by insects. Lipids 5(2): 247-258.Ubbelohde, A.R. 1978. In The molten State of Matter, John Wiley & Sons, Chichester.Vaghefi, F., Lee, J. and Nalamothu, V. 2001. Zero-order release and temperature-controlled microcapsules and

process for the preparation thereof. US Patent 2001/0044026 A1.Van de Tempel, M. 1961. Mechanical properties of plastic disperse systems at very small deformations. J. Colloid

Sci. 16: 284–296.Vreeker, A., Hoekstra, L.L., deb Boer, D.C. and Agterof, W.G.M. 1992. The fractal nature of fat crystal networks.

Colloids and Surfaces 65: 185–189.Wampler, D. 1993. Aqueous liquid flavor oil capsules, method of making and using in foods. EP 0,633,732 B2.Wetzel, C.R. and Bell, L.N. 1998. Chemical stability of encapsulated aspartame in cakes without added sugar.

Food Chem. 63(1): 33-37.Wineman, R.D. 1984. Water emulsion fruit and vegetable coatings based on waxes. Eastman Chem. Prod. Publi-

cation # F-257A.Wu, C., Creek, J.L., Wang, K., Carlson, R.M., Cheung, S. and Tang, P.J. 2002. Measurement of wax deposition in

paraffin solutions. Presented at the AIChE 2002 Spring National Meeting, New Orleans, LA 10–14 March.

Encapsulation and Controlled Release in Bakery Applications 133

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_05_113-134 r1.qxd 3/29/07 2:53 PM Page 133

Page 142: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

6 Encapsulation Technologies for Preserving and Controlling the Release of Enzymes and Phytochemicals

Xiaoyong Wang, Yan Jiang, and Qingrong Huang

Introduction

According to a report from Business Communication, Inc. (http://www.bccresearch.com),the functional food industry in the US was valued at $20.2 billion in 2002 or 4 percentof the total food industry. Driven by both increasing fortification with healthy food in-gredients and consumer demand for novel food products, the functional food market isexpected to increase at an average growth rate of 13.3 percent, bringing the market valueto $37.7 billion by 2007. The development of functional foods with good bioavailabilityand eating qualities, however, requires methods for protecting these sensitive compo-nents from harmful environmental conditions and masking the taste of some of thesecomponents.

Encapsulation is the technique by which one material or a mixture of materials is coatedwith or entrapped within another material or system (Green and Scheicher, 1955). Encap-sulation can also be used to mask undesirable odors and bitter tastes of food ingredi-ents. The coated material is called active or core material, and the coating material is calledshell, wall material, carrier, or encapsulant. Encapsulation technology is well devel-oped and accepted within the pharmaceutical, chemical, cosmetic, and food industries(Augustin et al., 2001; Heinzen, 2002). Many encapsulation techniques have been devel-oped, such as spray drying, spray chilling and cooling, coacervation, fluidized bed coating,liposome entrapment, rotational suspension separation, and extrusion and inclusioncomplexation (Madene et al., 2006).

The widely used wall materials include polysaccharides and proteins, the keycomponents in both natural and processed foods (Tolstoguzov, 1991). Such polymershave critical impact on the structure and stability of food systems through their gelling,thickening, and surface-stabilizing functional properties. Proteins and polysaccha-rides are usually used in composites, especially when the creation of new productsis required. Intrinsic functional properties of individual components and their interactionsdetermine the final structure, texture, and stability of food materials. Understanding suchinteractions, especially between proteins and polysaccharides, is important not onlyfor manufacturing cost-effective functional ingredients, but also for designing novelfoods and for controlling their structural and textural impact on fabricated foods (Sanchezet al., 1997).

135

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 135

Encapsulation and Controlled Release: Technologies in Food SystemsEdited by Jamileh M. Lakkis

Copyright © 2007 by Blackwell Publishing

Page 143: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Complex Coacervate-Based Controlled Release Systems

One way to create controlled release encapsulation systems is through the use of complexcoacervates formed by proteins and polysaccharides. The basic science behind thecoacervation process is well developed and understood. Coacervation is divided into“simple” or “complex” processes. The former involves only one macromolecule and mayresult from the addition of a dehydrating agent that promotes polymer–polymer interac-tions over polymer–solvent interactions. In the latter case, two or more oppositely chargedmacromolecules or colloidal species are present to generate phase separation. In solution,polysaccharide and proteins may undergo two types of phase separation at above or belowthe isoelectric points of proteins: (i) the solid–liquid phase separation called precipitation(Kokufuta et al., 1981); and (ii) the liquid–liquid phase separation called coacervation(Burgess and Carless, 1984). The coacervate is the denser phase that is relatively concen-trated in macromolecules and is in equilibrium with the relatively dilute macromolecularliquid phase (Bungenberg, 1949).

The general picture for protein/polysaccharide coacervation from previous studies is thatprotein molecules initially bind to polysaccharide chains to form primary soluble complexesat first critical pH (pHc), and complex coacervate droplets, which ultimately settle at the bot-tom to generate the dense coacervate phase, are formed at second critical pH (pH

�). Primary

complex formation, initiated at pHc, is viewed as a microscopic transition on the molecularscales, whereas coacervate droplet formation at pH

�is viewed as a global phase transition.

A typical phase diagram of bovine serum albumin (BSA)/�-carrageenan mixtures isshown in Figure 6.1. Three regions are observed in the pH titration curve: (i) at pH > 6.2,there is no change in turbidity; (ii) at pH < 6.2, turbidity starts to increase, which is identifiedas the intercept of the soluble complex (pHc); and (iii) at pH < 4.8, turbidity increases signifi-cantly with the decrease of pH, which corresponds to the phase separation point (pH

�).

Because coacervates formed by polysaccharides and oppositely charged proteins are mainlydriven by the long-range character of the electrostatic interaction, physicochemical parame-ters affecting such interactions, such as pH, ionic strength, polysaccharide linear charge den-sity, protein surface charge density, rigidity of the polysaccharide chain, size of the protein,

136 Chapter 6

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 6.1. Plot of turbidity versus pH for mixture of bovine serum albumin (BSA) and �-carrageenan (10:1 w/w) in 0.1 M NaCl solution.

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 136

Page 144: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

and protein/polysaccharide ratio, strongly influence the formation of the complexes (Burgessand Singh, 1993; Hansen et al., 1971; Hugerth and Sundelof, 2001; Xia and Dubin, 1994).

Although extensive studies have been focused on the phase boundaries of protein/polysaccharide coacervation, understanding the structure of protein/polysaccharide coacer-vates is still quite lacking (Doublier et al., 2000; Turgeon et al., 2003). With the help of confo-cal scanning laser microscopy, Sanchez et al. (2002) found that the internal structure of�-lactoglobulin/gum arabic coacervates was vesicular or sponge-like, exhibiting numerousspherical inclusions of water depending on the initial mixing ratio. Using small-angle X-rayscattering, Weinbreck et al. (2004a) found that whey protein/gum arabic complex coacervateswere dense and structured and could be tuned by pH, protein/polysaccharide ratio, and ionicstrength. They also studied the viscoelastic properties of whey protein/gum acacia coacer-vates and verified that whey protein/gum acacia complex coacervates had the highest viscos-ity at pH = 4.0, which was ascribed to the strongest electrostatic interaction between wheyprotein and gum acacia (Weinbreck et al., 2004b). Recently, our group has investigated thedynamic rheolgical properties of BSA/�-carrageenan complex coacervates. Figure 6.2 showstypical profile of small deformation oscillatory measurements of BSA/�-carrageenan com-plex coacervates at 0.1 M NaCl concentration and 10:1 protein/polysaccharide ratio, withpH = 4.5. The storage modulus (G�) was found to be more than two times greater than the lossmodulus (G�), wherein the two moduli are almost independent of angular frequency (�) at� > 0.5 rad/s. The high value of G� when compared to G� indicates that BSA/�-carrageenancomplex coacervates have a highly interconnected gel-like network structure with mainlyelastic behavior, which agrees with the rheological properties of simple coacervates like gela-tin (Mohanty and Bohidar, 2005). At similar frequencies, sweep measurements for wheyprotein/gum Arabic coacervates, G� was reported to be three to seven times higher than G�, anindication of the highly viscous character of whey protein/gum Arabic coacervates (Weinbrecket al., 2004a). Coacervates of BSA with synthetic polyelectrolyte poly(diallyldimethylammo-nium chloride) also show viscous nature, with G� larger than G� in the high-frequency range

Preserving and Controlling the Release of Enzymes and Phytochemicals 137

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Figure 6.2. Plot of storage modulus G� and loss modulus G� versus angular frequency �for the coacervates of BSA with �-carrageenan (10:1 w/w) in 0.1 M NaCl at pH � 4.5 (Leeet al., 2003).

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 137

Page 145: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

(Bohidar et al., 2005). Therefore, different viscoelastic properties in different systems reflectthe characteristics of protein/polymer pair and thus distinct coacervate structure.

Small-angle neutron scattering (SANS) experiments, which were performed at theintense pulsed neutron source at Argonne National Laboratory, Argonne, IL, have been usedto illustrate the structure of complex coacervates formed by �-lactoglobulin and pectin. TheSANS results for �-lactoglobulin/pectin coacervates (30:1 w/w) at two different salt concen-trations are shown in Figure 6.3(a). All the curves show a peak of shoulder at intermediatescattering vector range, indicating the electrostatic repulsion of proteins bound onto pectinchains. From the maximum of the peaks in the structure factor curves [Figure 6.3(b)], thedistance between bound proteins (d) could be determined: d = 7.6 nm at 0.05 M NaCl isfound to be smaller than d = 8.7 nm at 0.1 M NaCl. The strong correlation between peakmaxima and their position with salt concentration may be an indication of the more het-erogenous and less-structured nature of these coacervates at higher salt concentration.

The concept behind protein/polysaccharide complex coacervate-based controlled releasedelivery systems arises from the pH-triggered phase separation of protein/polysaccharidecomplexes from the initial mixed solutions, and the subsequent deposition of the newlyformed coacervate phase surrounding the active ingredients (Gouin, 2004). If needed, thecoacervate shell can be cross-linked using an appropriate chemical or enzymatic cross-linker. A large number of protein/polysaccharide complex systems, such as gelatin/gum aca-cia (Ijichi et al., 1997; Rabiskova and Valaskova, 1998), gelatin/carboxymethylcellulose(Bakker et al., 1999), �-lactoglobulin/gum acacia (Schmitt et al., 2000), and guar/dextran(Simonet et al., 2002), have shown good properties for microencapsulation application.

Coacervation is typically used in the encapsulation of flavor oils (Soper, 1995), but canalso be adapted for the encapsulation of fish oils (Lamprecht et al., 2001), vitamins(Junyaprasert et al., 2001), enzymes (Dubin et al., 1998), and dietary supplements.

To improve the appeal of frozen baked foods upon heating, flavor oil was entrapped incomplex coacervate microcapsules using gelatin and gum arabic (Yeo et al., 2005). Thedesign criteria of these systems include: (i) the odor should not be released while the food isfrozen or, if thawed, before it is cooked; and (ii) the odor should be released upon heating.The rate of homogenization was found to affect the size of oil cores encapsulated within themicrocapsules, whereas the polyion concentrations affected the number of core aggregatesconsisting of a microcapsule. The comparison of homogenization speed between 3000 and

138 Chapter 6

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

100

10

I (q)

q (A–1)

1

0.01 0.1 1

(a)0.05 M NaCl0.1 M NaCl 100

10

S (

q)

q (A–1)

0.01 0.1 1

(b)0.05 M NaCl0.1 M NaCl

Figure 6.3. Small-angle neutron scattering intensity profiles from �-lactoglobulin/pectincoacervates (30:1 w/w) at different salt concentrations: (a) scattering curves; (b) structure factor curves.

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 138

Page 146: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

9000 rpm shows that the microcapsules prepared with a lower homogenization speed wereless resistant to heating. A possible explanation for this result is that slower homogenizationformed microcapsules having single large cores, which may make them more vulnerable todamage than multivesicular microcapsules produced by higher speed homogenization.Microcapsules prepared with higher concentrations of polyions were less resistant to therelease conditions. This result may be due to the fact that lower solution concentrationsresulted in bigger agglomerates of oil droplets, which were harder to completely break down.

Encapsulation and Controlled Release of Food Enzymes

Enzymes are catalytic proteins that are capable of great specificity and reactivity underphysiological conditions. Like most proteins, they are highly susceptible to physiologicalparameters such as pH and heat and to chemicals like denaturation agents. They are com-monly encapsulated and immobilized in food processing, biomedical examination, andantibody labeling. However, they are normally contaminated by proteases, which may gen-erate unpredictable or inaccurate results. Most often, these enzymes are obtained from bac-teria or other biomaterials, which subsequently complicates their purification and theformation of protease-free enzymes. The existence of protease in enzymes prevents the useof proteins as wall materials for enzyme encapsulation.

Recently, we have developed an inexpensive, fast, and convenient method for encapsulat-ing food enzyme (Jiang and Huang, 2004) through the direct formation of complex coacer-vate with negatively charged polysaccharide such as �-carrageenan. -Amylase was used asa model enzyme to form coacervates with �-carrageenan to microencapsulate -amylase.The -amylase encapsulation efficiency and free -amylase were defined as:

Encapsulation efficiency % �Encapsulated enzyme

Total enzyme(1)

Free enzyme % �Non-encapsulated enzyme

(2)Total enzyme

Our results show that a �-carrageenan to -amylase ratio of 1 to 2 resulted in very highencapsulation efficiency (>99 percent) of -amylase as shown in Figure 6.4. -Amylasereleased from coacervates also maintained the same catalytic activity as the enzyme control,while unencapsulated -amylase lost most of its enzymatic activities after exposure to low pH(i.e., 3) for half an hour. Enzyme kinetics, therefore, can be described by Michaelis–Mentenequation,

(3)

Here Km is the Michaelis–Menten constant and Vmax is the maximum hydrolysis rate. Km andVmax were determined from equation (3). Table 6.1 shows that for coacervate-encapsulated-amylase, even after being treated with acid, -amylase displayed negligible change in enzy-matic activities after being released. However, -amylase without coacervate protectionalmost lost its enzymatic activities, as evidenced by significantly lower values of Km and Vmax.

1 1 1

V

K

V S V� m

max max[ ]

Preserving and Controlling the Release of Enzymes and Phytochemicals 139

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 139

Page 147: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

These results suggest that the enzyme encapsulation through complex coacervation is an effi-cient method to protect the enzyme from denaturation.

Encapsulation and Controlled Release of Phytochemicals

Phytochemicals have received much attention in recent years from the scientificcommunity, consumers, and food manufacturers due to their potential in lowering bloodpressure, reducing cancer risk factors, regulating digestive tract activity, strengtheningimmune systems, regulating growth, controlling blood sugar concentration, loweringcholesterol levels and serving as antioxidants. The scientific evidence supporting thesehealth-promoting claims of phytochemicals is growing steadily (Wildman, 2001).Although the use of phytochemicals in capsules and tablets is abundant, their effect isfrequently diminished or even lost due to their lack of solubility in water, vegetable oils orother food-grade solvents. In addition, insufficient gastric residence time, low permeabilityand solubility within the gut, as well as instability under conditions encountered in productprocessing (temperature, oxygen, light) or in the gastro-intestinal tract (pH, enzymes,presence of other nutrients) limit the activity and potential health benefits of phytochemicalmolecules (Bell, 2001). The delivery of these molecules will therefore require availabilityof protective mechanisms that can maintain the active molecular form until the time ofconsumption and to deliver this form to the physiological target within the organism.

140 Chapter 6

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 6.4. Encapsulation efficiency curve. Encapsulation has the highest efficiency of about99.3 percent at the ratio of �-carrageenan/-amylase 1:2 in 0.01 M NaCl.

Enzyme Km (g) Vmax (g/min)

1 Untreated enzyme (control) 1.08 0.32 Encapsulated, acid treated, released 1.07 0.283 Unencapsulated, acid treated 0.04 0.0036

Table 6.1. Enzymatic kinetics of -amylase with different treatments

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 140

Page 148: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

To overcome instability, poor water solubility and bioavailability of phytochemicals,encapsulation techniques have been employed to bring about effective amounts of the intactactive component to desired target sites in the body. Ideally, actives such as phytochemicalsshould be stable and intact under stomach acidic conditions, but readily bioavailable underprevailing alkaline conditions of the small intestines (Ho et al., 1992; Salah et al., 1995;Havsteen, 1983).

Tea catechins, one of the typical flavonoid components of green tea, have been shownto possess desirable physiological activities such as antioxidants, anti-AIDS virus, anti-mutagenic, anti-carcinogenic, probiotic, anti-microbial and anti-inflammatory (Havsteen,1983; Nakagawa et al., 1999). One of the major challenges with utilizing tea catechinsis their poor oral bioavailabilities. Epigallocatechin gallate (EGCG), the most importantcomponent of catechins contained in green tea, can readily undergo extensive glucoronida-tion, sulfation, methylation and ring fission in humans, mice and rats (Yang et al., 2002;Nakagawa et al., 1997; Suganuma et al., 1998; Cauturla et al., 2003). In addition, it can eas-ily undergo oxidation at neutral to alkaline pH, especially at high temperatures. Figure 6.5demonstrates progressive increase in color intensity (browning) of EGCG solutions withincreased pH after only one-day storage. Oxidation of EGCG solutions at different pHlevels and temperatures can be accurately monitored by tracing their absorption at wave-length of �290 nm using UV spectroscopy. Upon oxidation of EGCG, its absorption wave-length was found to gradually shift to longer wavelength (317 nm).

In our laboratories, we attempted to preserve the stability and bioavailability of teacatechins (EGCG) via complex coacervation in carrageenan/gelatin-A (Jiang and Huang,2004). The encapsulation efficiency of EGCG in these coacervates was determinedby high performance liquid chromatography (HPLC) and found to be as high as 89.4%(Figure 6.6). In vitro release of coacervate encapsulated EGCG was also studied in artificialstomach and intestinal juices at 37ºC for 2 hrs and 4 hrs, respectively. The active (EGCG)did not show any release under acidic stomach conditions (confirmed by UV spectra),but was totally released in the first 15 minutes of incubation in artificial intestinal juice(Figure 6.7).

Preserving and Controlling the Release of Enzymes and Phytochemicals 141

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Figure 6.5. Color changes of epigallocatechin gallate (EGCG) at different pHs after 1-daystorage.

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 141

Page 149: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Encapsulation of Phytochemicals by Nanoemulsions

Nanoemulsions are a class of extremely small emulsion droplets that can be transparent ortranslucent with a bluish coloration (Nakajima, 1997; Solans et al., 2005; Sonneville-Aubrun et al., 2004). They are usually available in the range of 50-200 nm. Similar to tradi-tional macro-emulsions, two types of nanoemulsions can be prepared, namely oil-in-water(O/W) and water-in-oil (W/O) nanoemulsions. Although emulsions are thermodynamicallyunstable systems, nanoemulsions, owing to their characteristic size, may possess highkinetic stability against sedimentation or creaming. Nanoemulsions can be prepared by the

142 Chapter 6

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

12

10

8

6

mg/

ml

4

2

0Encapsulated

+Un-encap Un-encap

Efficiency89.44%

Figure 6.6. Encapsulation efficiency of complex coacervate-encapsulated epigallocatechingallate (EGCG) as determined by high-performance liquid chromatography (HPLC).

Figure 6.7. In vitro release of tea catechins in artificial stomach and intestinal juice.

No catechinswere released

in 2 hrs.

Within 20 min.,all catechins

were released.

Small intestine

Stomach

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 142

Page 150: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

so-called dispersion or high-energy emulsification methods using high shear stirring, high-pressure homogenization and ultrasound generators (Walstra, 1983). Other methods suchas condensation or low-energy emulsification and phase inversion temperature could pro-duce nanoemulsion almost spontaneously (Rang & Miller, 1999).

Nanoemulsions have been investigated for their ability to transport phytochemicals(Solans et al., 2005). The mechanism takes place via large reduction in gravitational forceand Brownian diffusion, thus preventing any creaming or sedimentation, followed by stericstabilization and prevention of droplet flocculation or its coalescence. Nanoemulsions alsooffer other advantages for encapsulating water-soluble (entrapped in the core) and waterinsoluble (incorporated at the interface or the oil phase) substances that can be designed forslow release applications (Garti et al., 2003; Shefer and Shefer, 2003). This approach wasclaimed to enhance bioavailability of oil-soluble or water-soluble phytochemicals.

Curcumin, an FDA-approved food additive, is widely used as a preservative and yellowcoloring agent for foods, drugs, and cosmetics. Curcumin has also been shown to possessunique anti-inflammatory activity (Reddy et al., 2004; Huang et al., 1988, 1994). However,orally administered curcumin is plagued with low systemic bioavailability (Pan et al.,1999). Recently, we developed o/w nanoemulsion for encapsulating curcumin (Wang andothers, unpublished). Figure 6.8 shows photomicrographs of curcumin regular- and nano-sized- emulsions with the latter exhibiting unique homogeneous droplet size distribution.Using particle size analysis, average diameter of curcumin nanoemulsion droplets wasfound to be 65 nm.

The mouse ear inflammation model is commonly used to test the bioavailability of anti-inflammatory agents in vivo. In such studies, topical application of 12-O-tetrade-canoylphorbol-13-acetate (TPA) can rapidly induce edema of mouse ear in a dose- andtime-dependent manner. Earlier studies in our laboratory have shown that oral administra-tion of anti-inflammatory agents such as aspirin and garcinol can inhibit TPA-inducededema in mouse ears. We have also reported that various levels of garcinol were found inserum, ear, liver, lung and colon after oral administration of garcinol by female CD-1 micefor several hours. In addition, oral administration of aspirin or garcinol by gavages resultedin marked inhibition of TPA-induced edema in mouse ears.

In contrast, oral administration of curcumin, a poor bio-available anti-inflammatoryagent, had little or no effect on TPA-induced edema of mouse ears. However, oral adminis-tration of two different preparations of curcumin emulsion (10 mg curcumin in 1 ml)prepared by either high speed homogenization (regular) or high pressure homogenization(nanoemulsion) to mice by gavages at 30 min prior to topical application of TPA hasmarkedly inhibited TPA-induced edema of mouse ears by 43 and 85%, respectively.

Bioconjugation of Phytochemicals

Nanoparticles are defined as submicronic (<1 μm) colloidal systems made of polymersboth biodegradable and non-biodegradable. Nanocapsules, one type of nanoparticles, arevesicular systems in which actives such as flavonoids can be confined to a cavity, generally,an oily or aqueous core surrounded by a unique polymeric membrane. Nanospheres, on theother hand, are matrix systems in which the active is dispersed throughout the particles.Initial research on colloidal carriers was mainly focused on liposomes which are very diffi-cult to produce or stabilize for practical applications. In contrast, nanopraticles owing totheir unique stability can potentially be superior carriers compared to liposome.

Preserving and Controlling the Release of Enzymes and Phytochemicals 143

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 143

Page 151: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Activities of polyphenols such as their anti-oxidative power, circulation time in thehuman body and other activities can be reduced upon exposure to environmental stressessuch as moisture, heat, and oxidation (Hagerman et al., 1998; Kurisawa et al., 2003). Wehave attempted to preserve polyphenol activities, in particular their anti-oxidative ability bymeans of synthesizing poly(catechin)via enzyme-catalyzed oxidative coupling using horse-radish peroxidase as a catalyst (Shin and Huang, unpublished results). The poly-catechinshowed great improvement in antioxidative activity such as radical scavenging activityagainst the superoxide anion and inhibition effects against free radical induced oxidation oflow-density lipoprotein, compared to the catechin monomer. In addition, poly-(catechin)showed very high inhibition effects on xanthine oxidase activity, whereas the catechinmonomer showed very low inhibition effects.

Conclusion

One of the most important stakes in the health promotion industry is the efficient encapsu-lation of highly valuable phytochemicals. Taking advantage of nanoscale particles,nanoemulsions and nanoparticles, provide excellent vehicles for encapsulating phytochem-icals and to preserve their stability and bioavailability. Another unique encapsulation tech-nique for such applications is complex coacervation. Numerous protein/polysaccharidepairs have been demonstrated to provide controlled-release of phytochemicals in vitro aswell as in vivo. Conjugation of phytochemicals can also play a promising role in encapsu-lating large-scale actives and in their effective utilization in food systems. Indeed, thechoice of an appropriate technique, however, depends on the properties of the active com-pounds, the degree of stability required during storage and processing, desired releaseproperties, maximum obtainable phytochemical load as well as production cost.

ReferencesAugustin, M. A., Sanguansri, L., Margetts, C., Young, B. 2001. Microencapsulation of food ingredients. Food

Aust., 53, 220–223.Bakker, M. A. E., Galema, S. A., Visser, A. 1999. Microcapsules of Gelatin and Carboxymethylcellulose. Euro-

pean Patent Application EP 0 937 496 A2, Unilever NV, NL; Unilever PLC, GB.

144 Chapter 6

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 6.8. Microscope images of curcumin normal emulsions (left) and nanoemulsions (right).

(a) (b)

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 144

Page 152: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Bell, L. N. 2001. Stability testing of nutraceuticals and functional foods. In: Wildman, R. E. C. (Ed.), Handbook ofNutraceuticals and Functional Foods (pp. 501–516). New York: CRC Press.

Bohidar, H., Dubin, P. L., Majhi, P. R., Tribet, C., Jaeger, W. 2005. Effects of protein–polyelectrolyte affinity andpolyelectrolyte molecular weight on dynamic properties of bovine serum albumin-poly (diallyldimethylammo-nium chloride) coacervates. Biomacromolecules, 6, 1573–1585.

Bungenberg de Jong, H. G. 1949. In: Kruyt, H. R. (Ed.), Colloid Science, Vol. II. Amsterdam: Elsevier.Burgess, D. J., Carless, J. E. 1984. Microelectrophoretic studies of gelatin and acacia for the prediction of complex

coacervation. J. Colloid Interface Sci., 98, 1–8.Burgess, D. J., Singh, O. N. 1993. Spontaneous formation of small sized albumin/acacia coacervate particles.

J. Pharm. Pharmacol., 45, 586–591.Cauturla, N., Vera-samper, E., Villalain, J., Mateo, C. R., Micol, V. 2003. The relationship between the antioxidant

and the antibacterial properties of galloylated catechins and the structure of phospholipid model membranes.Free Radic. Biol. Med., 34, 648–662.

Cho, Y. H., Huang, Q. R. (submitted for publication) Temperature Triggered Release of Curcumin from ComplexCoacervation with Gelatin A and Alginate.

Doublier, J.-L., Garnier, C., Renard, D., Sanchez, C. 2000. Protein–polysaccharide interactions. Curr. Opin.Colloid Interface Sci., 5, 202–214.

Dubin, P. L., Muhoberac, B. B., Xia, J. 1998. Preparation of Enzyme–Polyelectrolyte Coacervate Complexes andtheir Properties. US 5834271A.

Garti, N., Aserin, A., Spernath, A., Amar, I. 2003. Nano-Sized Self-Assembled Structured Liquids.US20030232095.

Gouin, S. 2004. Microencapsulation: industrial appraisal of existing technologies and trends. Trends Food Sci.Technol., 15, 330–347.

Green, B. K., Scheicher, L. 1955. Pressure Sensitive Record Materials. US Patent No. 2, 217, 507, NCR C.Hagerman, A. E., Riedl, K. M., Jones, G. A., Sovik, K. N., Ritchard, N. T., Hartzfeld, P. W., Riechel, T. L. 1998.

High molecular weight plant polyphenolics (Tannins) as biological antioxidants. J. Agric. Food Chem., 46(5),1887–1892.

Hansen, P. M. T., Hidalgo, J., Gould, I. 1971. Reclamation of whey protein with carboxymethylcellulose. J. DairySci., 54, 830–834.

Havsteen, B. 1983. Flavonoids, a class of natural products of high pharmacological potency. Biochem. Pharmacol.,32, 1141–1148.

Heinzen, C. 2002. Microencapsulation solves time dependent problems for foodmakers. Eur. Food Drink Rev. 3,27–30.

Ho, C. T., Lee, C. Y., Huang, M. T. (Eds.). 1992. Phenolic Compounds in Food and their Effects on Health.I: Analysis, Occurrence, and Chemistry. ACS Symp. Ser. 506, Washington, D.C.: American Chemical Society.

Huang, M. T., Lou, Y. R., Ma, W., Newmark, H. L., Reuhl, K. R., Conney, A. H. 1994. Inhibitory effects of dietarycurcumin on forestomach, duodenal and colon carcinogenesis in mice. Cancer Res., 54, 5841–5847.

Huang, M. T., Smart, R. C., Wong, C. Q., Cooney, A. H. 1988. Inhibitory effect of curcumin, chlorogenic acid, caf-feic, and ferulic acid on tumor promotion in mouse skin by 12-O-tetradecanoylphorbol-13-aceate, Cancer Res.,48, 5941.

Hugerth, A., Sundelof, L.-O. 2001. The effect of polyelectrolyte counterion specificity, charge density, and confor-mation on polyelectrolyte–amphiphile interaction: The carrageenan/furcellaran-amitriptyline system. Biopoly-mers, 58, 186–194.

Ijichi, K., Yoshizawa, H., Uemura, Y., Hatate, Y., Kawano, Y. 1997. Multi-layered gelatin/acacia microcapsules bycomplex coacervation method. J. Chem. Eng Jpn., 30, 793–798.

Jiang, Y., Huang, Q. R. (submitted for publication) Encapsulation and controlled release of polyphenols using pro-tein/polysaccharide coacervates.

Jiang, Y., Huang, Q. R. 2004. Microencapsulation and controlled-release of food enzyme using protein–-polysaccharide coacervates. Polym. Prepr., 45(2), 464.

Junyaprasert, V. B., Mitrevej, A., Sinchaipanid, N., Boonme, P., Wurster, D. E. 2001. Effect of process variables on themicroencapsulation of vitamin A palmitate by gelatin-acacia coacervation. Drug Dev. Ind. Pharm., 27, 561–566.

Kokufuta, E., Shimizu, H., Nakamura, I. 1981. Salt linkage formation of poly(diallyldimethylammonium chlo-ride) with acidic groups in the polyion complex between human carboxyhemoglobin and potassium poly(vinylalcohol) sulfate. Macromolecules, 14, 1178–1180.

Kurisawa, M., Chung, J. E., Kim, Y. J., Uyama, H., Kobayashi, S. 2003. Amplification of antioxidant activity andxanthine oxidase inhibition of catechin by enzymatic polymerization. Biomacromolecules, 4(3), 469–471.

Preserving and Controlling the Release of Enzymes and Phytochemicals 145

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 145

Page 153: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Lamprecht, A., Schafer, U., Lehr, C. M. 2001. Influences of process parameters on preparation of microparticleused as a carrier system for W-3 unsaturated fatty acid ethyl esters used in supplementary nutrition. J. Microen-capsul., 18, 347–357.

Lee, J. Y., Ruengruglikit, C., Huang, Q. R. 2003. Interactions between Carrageenan and BSA: (1) Effects of linearcharge density and ionic strength. Polym Prepr., 44, 289–290.

Madene, A., Jacquot, M., Scher, J., Desobry, S. 2006. Flavour encapsulation and controlled release. Int. J. FoodSci. Technol., 41, 1–21.

Mohanty, B., Bohidar, H. B. 2005. Microscopic structure of gelatin coacervates. Int. J. Biol. Macromol., 36,39–46.

Nakagawa, K., Miyazawa, T. 1997. Absorption and distribution of tea catechin, (-)-epigallocatechin-3-gallate, inthe rat. Nutr. Sci. Vitaminol., 43, 679–684.

Nakagawa, K., Ninomiya, M., Okubo, T., Aoi, N., Juneja, L. R., Kim, M., Yamanaka, K., Miyazawa, T. 1999. Teacatechin supplementation increases antioxidant capacity and prevents phospholipid hydroperoxidation inplasma of human. J. Agric. Food Chem., 47, 3967–3973.

Nakajima, N. 1997. Microemulsions in cosmetics. In: Solans, C., Kunieda, H. (Eds.), Industrial Applications ofMicroemulsions (pp. 175–197). New York: Marcel Dekker.

Pan, M. H., Huang, T. M., Lin, J. K. 1999. Biotransformation of curcumin through reduction and glucuronidationin mice. Drug Metab. Dispos., 27, 486–494.

Rabiskova, M., Valaskova, J. 1998. The influence of HLB on the encapsulation of oils by complex coacervation.J. Microencapsul., 15, 747–751.

Rang, M. J., Miller, C. A. 1999. Spontaneous emulsification of oils containing hydrocarbon, nonionic surfactant,and oleyl alcohol. J. Colloid Interface Sci., 209, 179–192.

Reddy, R. S., Rao, C. V. 2004. Chemoprevention of colon cancer by curcumin. In: Meskin, M. S., Bidlack, W. R.,Davies, A. J., Lewis, D. S., Randolph, R. K. (Eds.), Phytochemicals: Mechanisms of Action (pp. 177–192).Boca Raton, FL: CRC Press.

Salah, N., Miller, N. J., Paganga, G. 1995. Polyphenolic flavanols as scavengers of aqueous phase radicals and aschain breaking antioxidants. Arch. Biochem. Biophys., 322, 339–346.

Sanchez, C., Schmitt, C., Babak, V. G., Hardy, J. 1997. Rheology of whey protein isolate-xanthan mixed solutionsand gels. Effect of pH and xanthan concentration. Nahrung, 41(6), 336–343.

Sanchez, C., Mekhloufi, G., Schmitt, C., Renard, D., Robert, P., Lehr, C.-M., Lamprecht, A., Hardy, J. 2002. Self-assembly of �-lactoglobulin and acacia gum in aqueous solvent: structure and phase-ordering kinetics. Lang-muir, 18, 10323–10333.

Schmitt, C., Sanchez, C., Despond, S., Renard, D., Thomas, F., Hardy, J. 2000. Effect of protein aggregates on thecomplex coacervation between beta-lactoglobulin and acacia gum at pH 4.2. Food Hydrocolloids, 14, 403–413.

Soper, J. C. 1995. Utilization of coacervated flavors. ACS-Symposium Series, No. 590, 104–112.Shefer, A., Shefer, S. 2003. Multicomponent Controlled Release System for Oral Care, Food Products, Nutraceti-

cal, and Beverages. US20030152629.Simonet, F., Garnier, C., Doublier, J. L. 2002. Description of the thermodynamic incompatibility of the guar-

dextran aqueous two-phase system by light scattering. Carbohydr. Polym., 47, 313–321.Solans, C., Izquierdo, P., Nolla, J., Azemar, N., Garcia-Celma, M. J. 2005. Self-assembly of surfactants and phos-

pholipids at interfaces between aqueous phases and thermotropic liquid crystals. Curr. Opin. Colloid InterfaceSci., 10, 102–110.

Sonneville-Aubrun, O., Simonnet, J. T., L’Alloret, F. 2004. Nanoemulsions a new vehicle for skincare products.Adv. Colloid Interface Sci., 108–109, 145–149.

Suganuma, M., Okabe, S., Oniyama, M., Tada, Y., Ito, H., Fujiki, H. 1998. Wide distribution of [3H](-)-epigallocatechin gallate, a cancer preventive tea polyphenol, in mouse tissue, Carcinogenesis, 19, 1771–1776.

Tolstoguzov, V. B.1991. Functional properties of food proteins and role of protein–polysaccharide interaction.Food Hydrocolloids, 4, 429–468.

Turgeon, S. L., Beaulieu, M., Schmitt, C., Sanchez, C. 2003. Protein–polysaccharide interactions: phase-orderingkinetics, thermodynamic and structural aspects. Curr. Opin. Colloid Interface Sci., 8, 401–414.

Weinbreck, F., Tromp, R. H., de Kruif, C. G. 2004a. Composition and structure of whey protein/gum arabic coac-ervates. Biomacromolecules, 5, 1437–1445.

Weinbreck, F., Wientjes, R. H. W., Nieuwenhuijse, H., Robijn, G. W., de Kruif, C. G. 2004b. Rheological proper-ties of whey protein/gum arabic coacervates. J. Rheol., 48, 1215.

Wildman, R. E. C. 2001. In: Wildman, R. E. C. (Ed.), Handbook of Nutraceuticals and Functional Foods. NewYork: CRC Press.

146 Chapter 6

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 146

Page 154: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Xia, J., Dubin, P. L. 1994. Protein–polyelectrolyte complexes. In: Dubin, P. L., Bock, J., Davis, R., Schulz, D. N.,Thies, C. (Eds.), Macromolecular Complexes in Chemistry and Biology (pp. 247–271). Berlin: Springer-Verlag.

Yang, C. S., Maliakal, P., Meng, X. 2002. Inhibition of carcinogenesis by tea. Annu. Rev. Pharmacol. Toxicol., 42,25–54.

Yeo, Y., Bellas, E., Firestone, W., Langer, R., Kohane, D. E. 2005. Complex coacervates for thermally sensitivecontrolled release of flavor compounds. J. Agric. Food Chem., 53, 7518–7525.

Preserving and Controlling the Release of Enzymes and Phytochemicals 147

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_06_135-148 r1.qxd 3/29/07 1:47 PM Page 147

Page 155: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

7 Microencapsulation of Flavors by Complex Coacervation

Curt Thies

Introduction

The encapsulation of flavors was first reported in the 1930s when it was observed that avolatile substance, isopropanol, was retained by a spray-dried particle (Thies, 1999). Thisobservation catalyzed the development of spray dry flavor encapsulation, a technologyresponsible today for the daily production of tons of encapsulated flavor products globally.Reineccius (2004) and others (Brenner, 1983; Re, 1998; Liu et al., 2001) have discussedspray dry encapsulation technology in some detail.

Although spray drying is currently the dominant flavor encapsulation technique, a num-ber of alternate encapsulation technologies exist and offer a potential means of producingunique flavor-loaded microcapsules. Complex coacervation encapsulation procedures fallinto this category. Accordingly, this contribution is a discussion of various aspects of com-plex coacervation encapsulation technology and the encapsulation of flavors for food prod-ucts. Only complex coacervation processes based on food-grade polymers are consideredhere. Although fragrances are not considered in this contribution, much of the discussion isalso applicable to the encapsulation of fragrances as well as other complex core materials.

Flavor Encapsulation

The preparation of flavor-loaded microcapsules is a complex task. It is much more compli-cated than it appears at first glance, because flavor microcapsules must meet a series ofrequirements. One requirement is the production of microcapsules that retain the desiredproperties of the flavor encapsulated. Each flavor is a unique and complex mixture of manycompounds. These compounds have a broad range of structures with vapor pressures, sol-vent solubility, and stability that differ significantly. Any useful encapsulation technologymust be able to accommodate this variability. Ideally, the chemical composition of the fla-vor is unchanged by the encapsulation process, and the encapsulated flavor is identical inall respects to the unencapsulated flavor. In reality, this is generally not the case. Encapsula-tion processes typically change the chemical composition of a flavor in some way. Loss ofmore volatile or more water-soluble components during an encapsulation process is common.Such losses can have a significant effect on the desired olfactory properties of the flavor.

Flavor-loaded microcapsules must contain enough active agents to cause the desiredeffect. The amount of flavor required varies with the nature of the flavor and intended foodproduct. Although flavor impact can be altered by varying the flavor loading of a capsule offixed size as well as by varying capsule size, the degree of variation may be limited by thenature of the food product. For example, capsule size variations may be limited by the need

149

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 149

Encapsulation and Controlled Release: Technologies in Food SystemsEdited by Jamileh M. Lakkis

Copyright © 2007 by Blackwell Publishing

Page 156: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

to retain structural integrity during processing while having the ability to be ruptured bychewing. Variations in amount of flavor carried by a capsule of fixed size may be limitedby the capsule formation process as well as the retention or barrier properties of the cap-sule shell.

Stability of flavor-loaded capsules during processing and storage is an issue that must beaddressed. Such capsules must have acceptable stability from the time of formation to con-sumption of the food product. Stability during processing of capsules as they are incorpo-rated into a food product can be a problem if this involves high shear or a combination ofhigh temperature and shear. Shelf life stability after incorporation and storage in a foodproduct is important as is the ability of capsules to release their contents during food prepa-ration or consumption. This series of stability requirements is imposing and often limitsactual capsule performance. Factors that affect capsule stability include oxygen, moisture,heat, and light. In principle, the shell of a capsule should protect an encapsulated flavorfrom these agents, but deficiencies in either the capsule shell or the material(s) from whichthe shell is prepared may cause a shell to provide inadequate protection. Shell materialstypically used to form food-grade flavor capsules may experience major property changesduring food-processing steps that involve heat and moisture. Of course, flavor-loadedmicrocapsules must meet specifications imposed by governmental regulatory agenciesresponsible for food safety. This requirement puts a restriction on the shell materials thatcan be used. It also limits the nature and amount of processing agents used in a capsule-formation process.

In summary, the complex series of specifications associated with the formation of flavor-loaded microcapsules makes their preparation an interesting field of study. Much can bedone in order to produce capsules that more closely approach the degree of perfectiondesired. Studies by various workers of the diffusion barrier properties of candidate shellmaterials merit review, because they provide much insight into the properties of such mate-rials and help one develop a realistic appreciation for the limitations of specific shell mate-rials and capsule formation processes (Menting and Hoogstad, 1967; Kerkof and Thijssen,1974; Thijssen, 1975; Rulkans and Thijssen, 1978; Goubet et al., 1998). Although most ofthese involve spray drying and freeze drying studies, the results obtained are applicable tocapsules formed by any encapsulation process.

Complex Coacervation

Before discussing complex coacervation encapsulation processes, it is appropriate to con-sider the nature of complex coacervate systems and some of their characteristic features.Complex coacervation is the liquid/liquid phase separation that occurs when solutionsof two or more oppositely charged polyelectrolytes are mixed under suitable conditions.Two liquid phases are formed: the coacervate phase and the supernatant or equilibriumliquid phase. The coacervate phase is a relatively concentrated polymer solution that par-ticipates in a complex coacervation encapsulation system. It is in this phase that capsuleshell forms. The supernatant phase is a dilute polymer solution and serves as the continu-ous phase in which capsule formation occurs. Dilution favors complex coacervation and isa property that distinguishes complex coacervation from other polymer phase-separationphenomena.

Complex coacervation is affected by many variables. Bungenberg de Jong’s experimentalstudies in the 1930s and 1940s provide much useful background data about the phenomenon

150 Chapter 7

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 150

Page 157: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

(Bungenberg, 1949). Since then, a variety of workers have considered various aspects ofcoacervation including theoretical analyses based on polymer solution thermodynamics(Burgess, 1990; Veis, 1970; Schmitt et al., 1998). Although this information provides a guidefor developing complex coacervation microencapsulation procedures, it is important to rec-ognize that conditions that optimize the degree of coacervation in a specific system may notbe conditions under which useful microcapsules can be formed. For example, these condi-tions may produce a complex coacervate that is too viscous to yield acceptable capsules.

Selected Properties of Complex Coacervates

Many complex coacervation systems suitable for the production of microcapsules exist. Invirtually all cases, gelatin is the polycation used. A wide range of polyanions is used. Eachsystem operates under a unique set of conditions and has a unique set of properties onceformed. This reflects differences in nature and frequency of ionic groups distributed alongthe chains of the polymers involved in a specific complex coacervation procedure. Differ-ences in polymer chain structure and molecular weight (MW) are other factors that influ-ence coacervation.

One of the polyelectrolytes used in a typical complex coacervation encapsulation proce-dure is a natural polymer with a complex molecular structure. For example, gelatin polymermolecules are made up of a number of different amino acids with different pendent groups.Because anionic and cationic pendent groups are distributed along the polymer chain, gela-tin is a polyampholyte. The cationic groups are primary amino groups, while the anionicgroups are carboxyl groups. The degree of ionization of these ionic groups varies with pH,so the net charge carried by a gelatin molecule varies with pH.

Gelatins formed by acid hydrolysis of collagen are classified as Type A or acid precursorgelatins. Alkaline hydrolysis yields Type B or alkaline precursor gelatins. The isoelectricpoint (pI) of Type A gelatins is typically 8–9, while the typical pI of Type B gelatins is 4–5.The reduced pI value of Type B gelatins is caused by hydrolysis of pendant amide groupsunder alkaline conditions. The number of primary amino groups distributed along a gelatinchain is essentially independent of the hydrolysis procedure. Although both types ofgelatins produce complex coacervates suitable for microcapsule formation, Type A gelatinshistorically have been used most. Significantly, for gelatin to carry a net cationic charge,it must be at a pH lower than its pI.

Although gelatin is the polycation involved in the formation of complex coacervatesused in microencapsulation processes, many different polyanions are used. They differgreatly in anion group distribution along a polymer chain as well as the nature of thisgroup. This is particularly true of natural polymers that carry an anionic group. Gum arabic(GA) and alginate, two polysaccharides, derive their anionic character from carboxylgroups distributed along their polymer chains. In GA, such groups are located on short-chain branches hanging off the primary polymer chain. Approximately 20% of the sugarunits in GA contain a carboxyl group. In contrast, alginate molecules are linear polymerchains and every sugar in the chain has a carboxyl group. The degree of ionization ofcarboxyl groups is a strong function of pH and steadily decreases as pH decreases. Theanionic character of sodium polyphosphate, an inorganic material, is due to the phosphategroup. Carrageenan, a polysaccharide with a linear chain, has sulfate groups distributedalong its chain. The average number of sulfate groups per sugar unit varies with the type ofcarrageenan.

Microencapsulation of Flavors by Complex Coacervation 151

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 151

Page 158: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Because the polymers used to form complex coacervates differ significantly in composi-tion and structure, properties of complex coacervates formed by different polymers differsignificantly. In order to illustrate this point, Table 7.1 contains degree of coacervation andenrichment data at 50°C for three gelatin-based complex coacervation systems used to pre-pare microcapsules: gelatin/gum arabic (GGA), gelatin/polyphosphate (GP), and gelatin/sodium alginate (GAlg) (Commandur et al., 1989). Degree of coacervation (ρ) is defined asthe fraction of total polymer in the system that is in the coacervate (Veis and Aryani, 1960).Enrichment (ε) is defined as the ratio of polymer concentration in the coacervate phase tothat in the supernatant phase (Veis and Aryani, 1960). GGA and GP coacervates wereformed by interacting 285 bloom Type A gelatin with GA and sodium hexametaphosphate,respectively. GAlg coacervates were formed by interacting 231 bloom Type A gelatin witha hydrolyzed alginate.

Each coacervation system was studied at three initial solids concentrations and three pHvalues in order to illustrate how changes in these variables affect coacervate formation.

152 Chapter 7

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Coacervate system Initial solids w/v (%) pH Degree of coacervation Enrichment

GGA 3.96 4 0.86 223.3 4 0.88 34.82.83 4 0.89 47.7

3.96 4.2 0.78 11.33.3 4.2 0.81 172.83 4.2 0.81 21.2

3.96 4.4 0.81 12.13.3 4.4 0.8 16.62.83 4.4 0.83 24.2

GAlg 2.11 4 0.86 66.71.81 4 0.83 681.59 4 0.82 71.7

2.11 4.2 0.66 25.31.81 4.2 0.73 42.51.59 4.2 0.7 54.7

2.11 4.4 0.78 35.71.81 4.4 0.81 531.59 4.4 0.79 54.7

GP 5 4 0.74 11.54.54 4 0.78 15.54.17 4 0.78 20.9

5 4.2 0.72 9.44.54 4.2 0.71 11.34.17 4.2 0.74 14.5

5 4.4 0.61 4.84.54 4.4 0.57 5.74.17 4.4 0.65 10.2

Table 7.1. Tabulation of degree of coacervation (ρ) and enrichment data (ε) at 50ºC for severalcoacervation systems

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 152

Page 159: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Indeed, at sufficiently high concentration and pH values, complex coacervation does notoccur. Initial solids content is defined as the total polymer solids in the system at the timewhen complex coacervation occurred. The data in Table 7.1 were obtained by using initialsolids of 1.6–5 w/w% and pH values of 4.0–4.4 pH. Although these initial solids and pHvalues are typical for many coacervation systems used to make microcapsules, situationsexist where suitable coacervates will form when values of one or both parameters fall out-side these ranges.

The gelatin/polyanion ratio used for forming gelatin-based complex coacervates varieswith ionic equivalent weight of the polyanion(s) used. The GGA complex coacervate datain Table 7.1 were formed by using a 1:1 w/w ratio of gelatin and GA, because both poly-mers have an ionic equivalent weight of roughly 1000. Alginates have an ionic equivalentweight of approximately 180, so the gelatin/alginate the ratio used was 3.7:1 w/w. The w/wgelatin/polyphosphate ratio was 9:1. It is high, because polyphosphates have a low ionicequivalent weight.

In all cases, the polyanion ratios reported here are those typically used by the author toproduce complex coacervates suitable for microcapsule formation. Complex coacervateswill form when a coacervation system contains excess gelatin or polyanion, but it is com-mon practice to use gelatin/polyanion ratios that approach ionic equivalency. Although thedata in Table 7.1 are for coacervation systems based on one polyanion, mixtures of severalchemically different polyanions can be used to produce gelatin-based coacervates suitablefor microcapsule formation. This enables one to develop a broad range of complex coacer-vate systems suitable for microcapsule formation.

The data in Table 7.1 (Commandur et al., 1989) show that ρ and ε at 50°C are affected bythe nature of the polyanion involved in coacervate formation, system pH, and initial solidscontent of the system. For all coacervation systems examined, values of ε at constant pHincrease as the initial solids content decreases. This reflects the increase in intensity ofcoacervation upon solution dilution, a characteristic feature of complex coacervation. Incontrast, other polymer phase-separation phenomena such as polymer/polymer incompati-bility and salting out (simple) coacervation are favored by increasing the concentration ofthe molecules involved.

Values of ρ for the GGA system fall between 0.8 and 0.9 over the range of initial solidsand pH values examined. Thus, in these GGA systems, 80–90 w/v% of the polymers wereconcentrated in the coacervate phase. The GGA coacervate phases formed are 15–26 vol%of total system volume and have a solids content of 10–14 w/v%. Solids content of thesupernatant phase was 0.3–0.9 w/v%. Since values of ε found for the GGA systems rangefrom 11 to 35, a high degree of polymer partitioning was achieved. At constant initialsolids, values of ε decrease as pH increases.

GP coacervate phases occupy 13–21 vol% of total system volume and have a solids con-tent of 13–21 w/v%. GP ρ values of 0.6–0.8 and ε values of 5–21 are lower than the rangeof ρ and ε values found for GGA and GAlg systems. Thus, the GP coacervate systemachieves a lower degree of polymer partitioning. The solids content of GP supernatantphases range from 1 to 2.3 w/v%, considerably higher than that observed with GGA andGAlg systems. Because gelatin concentrations of approximately 2 w/v% approach the con-centration at which gelatin solutions gel, operating conditions of an encapsulation processbased on GP must be adjusted to keep the supernatant solids concentration below 2 w/v%.

The solids content of a GAlg coacervate phase at 50°C varies from 15 to 22 w/v% whilethe solids content of a GAlg supernatant phase varies from 0.3 to 0.9 w/v%. The GAlg

Microencapsulation of Flavors by Complex Coacervation 153

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 153

Page 160: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

coacervate is 4–7 vol% of total system volume, considerably lower than the valuesobserved with GGA or GP coacervates. Values of ρ for the GAlg systems are 0.7–0.9, arange similar to but broader than that observed with GGA coacervates. The 25–72 range ofε values for GAlg coacervate systems is higher than that observed with GGA or GP sys-tems. Thus, GAlg coacervate systems more effectively concentrate or partition into thecoacervate phase the polymers involved in complex coacervation. At first glance, this issurprising, because the volume of the GAlg coacervate phase is much smaller than that ofthe GGA and GP coacervate phases, while the solids content of the GAlg coacervate phaseis similar to that of the GGA and GP coacervate phases. Closer analysis leads to the recog-nition that the initial solids content of the GAlg system is measurably lower than that of theGGA and GP systems. Thus, the GAlg coacervate phase contains a higher percentage ofpolymers present in the GAlg system, even though it occupies a smaller volume fractionthan the GGA and GP coacervate phases and has a solids content similar to these systems.

The ρ and ε data reported in Table 7.1 provide valuable insight into the nature of threecomplex coacervate systems, but they reveal nothing about coacervate rheology or the tem-perature at which the coacervates gel. Coacervate rheology is a primary variable thataffects capsule shell formation and capsule aggregation. Accordingly, the viscosity of anumber of coacervate phases was measured over a range of temperatures by capillaryviscometry (Commandur et al., 1989). Table 7.2 summarizes results of these measure-ments. The data show that the sodium alginate and GA solutions used to form GGA and GPcoacervates have a viscosity of 3–6 cS at 50°C. This viscosity increases as the temperatureis reduced to 30°C, but the viscosity increase caused by cooling is not pronounced, becauseneither polymer alone gels on cooling. In contrast, viscosity of the gelatin solutions exam-ined steadily increases as the solution temperature is reduced from 50°C to 35°C. Theviscosity of most such solutions becomes unstable at 32°C. That is, the recorded viscositysteadily increases toward infinity as the time at 32ºC increases. This viscosity increase isdue to the onset of gelation.

Not shown in Table 7.2 are viscosity data for GGA, GP, and GAlg supernatant phasesthat exist in equilibrium with the GGA, GP, and GAlg coacervate phases for which viscos-ity data were obtained. Most supernatant phases have a viscosity below 1 cS at tempera-tures ranging from 50°C to 35°C and provide no indication that they will gel on furthercooling. Exceptions are two GP supernatant solutions isolated from pH 4.4 GP coacervatesystems. The viscosity of both solutions remained below 1.5 cS as they were cooled to35°C, but the upward slope of their temperature–viscosity plots suggests that both will ulti-mately gel.

GGA coacervate viscosity at pH 4.4 and 50°C ranged from 23 to 58 cS, i.e., 2 to 5 timesgreater than the 11.3 cS viscosity of a 10% solution of 285 bloom Type A gelatin at 50ºC.Viscosity of the GGA coacervates steadily increases as the system is cooled. They eithergel or become unstable due to onset of gelation as the temperature falls below 35°C.Decreasing the pH of GGA coacervate formation from 4.4 to 4.0 increases coacervate vis-cosity at all temperatures examined.

The viscosity of GP coacervates at 50°C is 47–373 cS, measurably higher than the vis-cosity of GGA coacervates. Reducing the pH of a GP coacervate from 4.4 to 4.0 causes amajor increase in coacervate viscosity and raises the GP coacervate gelation temperatureabove 35°C.

The viscosity of all GAlg coacervates at 50°C is 20–60 times higher than that of mostGGA and GP coacervate phases at 50°C. Although GAlg coacervate phases have a very

154 Chapter 7

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 154

Page 161: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47Ta

ble

7.2

.C

han

ges

in v

isco

sity

of

vari

ou

s co

acer

vate

sys

tem

s at

dif

fere

nt

tem

per

atu

res

(fro

m C

om

man

du

r et

al.,

1989

, wit

h p

erm

issi

on

)

Init

ial s

olid

s S

olid

sV

isco

sity

(cS

)

Sys

tem

(w/v

%)a

pH(w

/v%

)b50

°C

45°C

40°C

37°C

35°C

32.5

°C30

°C

GG

A c

oace

rvat

e3.

964.

410

.423

2629

3546

107

Gel

led

2.83

4.4

12.1

3942

4762

96U

nsta

ble

Gel

led

GG

A c

oace

rvat

e3.

964

13.2

3942

4879

9714

3G

elle

d2.

834

14.3

5863

74U

nsta

ble

Gel

led

GP

Coa

cerv

ate

54.

412

.347

5698

221

Gel

led

4.17

4.4

14.3

6774

120

679

Gel

led

GP

Coa

cerv

ate

4.55

4.2

19.8

298

342

1782

Gel

led

4.17

420

.937

340

226

06G

elle

dG

Alg

Coa

cerv

ate

2.11

4.4

15.6

679

806

1636

3396

Gel

led

1.59

4.2

18.4

1245

1480

2485

7404

Gel

led

GA

lg C

oace

rvat

e2.

114

2013

9116

6528

0662

20G

elle

d1.

594

18.6

1382

1698

3254

7658

Gel

led

Gel

atin

sol

utio

n Ty

pe A

10

11.5

13.2

14.8

1720

Uns

tabl

eG

elle

d(2

85 b

loom

)15

3135

4052

.166

.6U

nsta

ble

Gel

led

Gel

atin

sol

utio

n Ty

pe A

10

5.2

7.3

89.

210

.311

.516

.2U

nsta

ble

(231

blo

om)

155.

415

.917

.820

.624

.628

.9U

nsta

ble

Gel

led

Sod

ium

alg

inat

e so

luti

on2

5.5

33.

43.

74

4.2

4.8

Gum

ara

bic

solu

tion

104.

24.

14.

54.

95.

45.

56.

3

aTo

tal s

yste

m s

olid

s at

the

tim

e of

coa

cerv

atio

n.bTo

tal s

olid

s of

sol

utio

n us

ed f

or c

apil

lary

vis

cosi

ty m

easu

rem

ent.

155

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 155

Page 162: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

high viscosity at temperatures ranging from 50°C to 40°C, they do not appear to gel untilcooled to 35°C.

As the data in Table 7.2 show, complex coacervate viscosity is a strong function of thecoacervation system, pH, and temperature. Characterizing the effect of temperaturechanges on rheology of a coacervate system is an important task, because all gelatin-basedcomplex coacervation encapsulation protocols involve a cooling step that lowers the sys-tem temperature below the gel temperature of the coacervate. In such processes, complexcoacervate formation always occurs above the coacervate melt temperature so that thecoacervate formed is initially a liquid. It must have a viscosity that is sufficiently low toenable it to engulf dispersed core material droplets or particles, thereby coating them with athin film of liquid coacervate. Once a coacervate is formed, the two-phase system is cooledbelow the gel temperature of the coacervate, thereby setting the gel structure of the coacer-vate. This transforms the thin liquid film that surrounds the small droplets of core materialinto a thin gel coating. Viscosity of a coacervate above its melt temperature and changes inits rheology as cooling occurs have a major impact on the success of a complex coacerva-tion encapsulation process.

The viscosity data in Table 7.2 cover a very large range of values. Although capillaryviscometry is appropriate for measuring the viscosity of Newtonian fluids, it has not beendetermined that all complex coacervates exhibit Newtonian flow behavior, especially attemperatures that approach the gel point. Leuenberger (1991) reported that 10% w/w%solutions of several different gelatin samples at 40°C are linear in the shear rate range of10–350 s–1. Deviations from linearity occur at high shear rates. Because of the relative flu-idity at 50°C of GGA and GP coacervates, it is believed that they exhibit Newtonian behav-ior at this temperature. The long capillary flow times of the GA coacervates at 50°C suggestthat such coacervates exhibit non-Newtonian flow behavior. Although additional measure-ments are needed in order to properly characterize the rheological behavior of a range ofcomplex coacervates, the data in Table 7.2 provide a means of comparing the apparent vis-cosity of several coacervates used to form microcapsules. These data illustrate the signifi-cant effect that composition of a complex coacervate has on its rheological properties. Thishas a profound effect on capsule formation.

It is relevant to note that Koh and Tucker (1988a, b) characterized the gelatin–carboxymethylcellulose (CMC) complex coacervate system. They reported characterizationdata similar to that shown in Tables 7.1 and 7.2 for this system. Although their characteriza-tion data as well as that shown in Tables 7.1 and 7.2 shed valuable insight into the specificcomplex coacervation systems studied, it must be recognized that such data represent typi-cal properties of a given complex coacervate system. Specific ρ and ε values reported forany complex coacervate system can be difficult to precisely reproduce consistently withinthe same laboratory by the same person, let alone different laboratories and different per-sons. This problem is caused by the sensitivity of complex coacervation to many factors.Experimental variations can be minimized by establishing standard experimental protocols.For example, all of the data shown in Table 7.1 were obtained by using aqueous solutionspolymers from the same lot. Distilled water was the solvent. Very different results wouldmost likely occur if tap water is used, because tap water can contain a variety of salt ions,and salt ions repress complex coacervation. Lot-to-lot variations in properties of either thegelatin or the polyanion used will also affect results obtained. In order to minimize varia-tion in reported results, the polymer solution preparation protocol must be standardized asmust the length of solution storage before use.

156 Chapter 7

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 156

Page 163: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

In cases where a flavor is being encapsulated by a process based on complex coacerva-tion, water-miscible or partially water-soluble components present in the flavor can affectthe coacervation process and nature of the coacervate formed. Seemingly small variationsin a coacervate system can have a major effect on complex coacervation results.

Complex Coacervation Encapsulation Processes

Bungenburg de Jong’s studies of the complex coacervation of gelatin and GA formed thebasis for the original GGA complex coacervation encapsulation procedure reported byGreen and Schleicher (1957). Figure 7.1 is a flow diagram of their process. The first step isto emulsify the material being encapsulated in a warm (40–60°C) aqueous gelatin solution.This material is commonly called the core material. It is typically a water-immiscible oil,but could be a water-insoluble solid. Oil emulsification typically is carried out in a warm8–11 w/w% gelatin solution because such concentrated solutions increase the ease of emul-sification to a desired drop size.

The second step is to add GA and dilution water to the system followed by adjustment ofthe pH to a value at which sufficient complex coacervate phase to encapsulate the dispersedoil droplets is formed. As noted in Table 7.1, this pH typically falls in the range of 4.0–4.4,although higher or lower pH values may be needed for a specific core material. Dilutionwater is added to the system at this point in order to lower the total polymer solids contentfrom the 8 to 11 w/v% used in the emulsification step to the 2.83–3.96 w/v% at which com-plex coacervation occurs (see Table 7.1).

The third step is to cool the system below the gel point of the coacervate, thereby caus-ing the coacervate to gel. In order to improve capsule shell stability, capsules with shells

Microencapsulation of Flavors by Complex Coacervation 157

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Water-immiscible

oil

Adjust pH(e.g.,4.0–4.6)

Oil-in-wateremulsion

Cool(to gel

coacervate)

Aqueousgelatinsolution

(40–60°C)

Harvestmicrocapsules

Water(40–60°C)

CrosslinkMixerMixer

Aqueousgum arabic

solution(40–60°C)

Figure 7.1. Flow diagram of gelatin gum arabic (GGA) complex coacervation encapsulationprocedure (from Green and Schleicher, 1957, with permission).

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 157

Page 164: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

formed by the complex coacervation of gelatin are typically chemically cross-linked withglutaraldehyde (glut) before they are isolated.

Today, complex coacervation encapsulation protocols still follow the basic Green andSchleicher (1957) procedure shown in Figure 7.1. Specific concentrations and types of thepolymers involved may vary, but the three-step protocol of emulsion formation, coacervateformation, and coacervate gelation is still used. Numerous variations of this process haveappeared since 1957, because many polyanions other than GA have been shown to interactwith gelatin to produce complex coacervates suitable for microcapsule formation. Thishas led to the development of a broad family of complex coacervation encapsulationprocedures. Specific coacervation procedures acceptable for the formation of flavor-loadedmicrocapsules are those in which gelatin interacts with food-grade polyanions such as GA,sodium alginate, carrageenan, pectin, CMC, gellan, and sodium polyphosphate. Combina-tions of these polyanions can also be used.

Significantly, the data in Table 7.1 show that properties of the coacervate phase formed ineach protocol differs in some manner. Even if the difference is small, considerable time maybe required to experimentally define conditions required for suitable capsule formation.

Table 7.3 is a list of a number of complex coacervation encapsulation protocols reportedby various workers in journals or patents. It is not an exhaustive list, but the references citedprovide information about a number of gelatin-based complex coacervation systems thatare candidates for flavor oil encapsulation. All the protocols are based on materials that theauthor regards as suitable for encapsulating food flavors.

It is interesting that no two protocols disclosed in Table 7.3 are similar. They all have anemulsification, coacervation, and cooling step, but the conditions under which these arecarried out are not standardized; that is, a standard protocol is not followed. Some workersuse Type A gelatin, while others use Type B. Gelatin bloom strength varies. Reported coac-ervation pH values fall between 3.0 and 5.5. Some workers adjust pH with acetic acid,while others use HCl.

Gelatin involved carries a positive charge in the 3.0–5.5 pH range and serves as thepolycation in all but one of the complex coacervation encapsulation systems cited in Table7.3. The sole case where this may not be the case is the coacervation of Type B gelatin withchitosan at pH 5.25–5.5 (Remunan-Lopez and Bodmeier, 1996). If this system is a complex

158 Chapter 7

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Table 7.3. Complex coacervation encapsulation protocols based on food-grade shell materials

Gelatin Polyanion Coacervation (pH) References

Type B, 225 bloom CMC 3.0–4.0 (0.1 M HCl) Koh and Tucker (1988a, b)Type B, 225 bloom Acacia 3.9 (1 M HAc) Jegat and Taverdet (2000)Type A Acacia 4.2 (glacial, acetic acid) Mayya et al. (2003)Type A, 300 bloom CMC 4.4 (10% HAc) Kim et al. (2001)Type A, 275 bloom Pectin NF 3.2–4.6 (0.5 M HCl) McMullen et al. (1984)Type B, 175 and Chitosan glutamate 5.25–5.50 (0.5 M HCl) Remunan-Lopez and

225 bloom Bodmeier (1996)Type A, 175 bloom Gellan 3.5–5.50 (0.5 M HCl) Chilvers and Morris (1995)Type A Sodium alginate 3.5–4.5 (0.5 N HCl) Joseph and Venkataran (1995)Type A, 275 bloom Sodium pyrophosphate 4.5 (10% acetic acid) Yan (2005)Fish gelatin CMC/gum arabic NA Soper (1997)Type A Gum arabic 3.9–4.7 (10% acetic acid) Saeki and Hosoi (1984)

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 158

Page 165: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

coacervation system, the gelatin must act as the polyanion, while chitosan is the polycation.At pH 5.25–5.5, such protocols should be limited to Type B gelatins. The pI of Type A gela-tin is typically 8–9, so its complex coacervation with chitosan should be limited to pHvalues above this.

Several of the studies referenced in Table 7.3 explore how various parameters affectmicrocapsule formation by complex coacervation. For example, Jegat and Taverdet (2000)found that the relationship of stirring rate and size of capsules produced by gelatin-GAcoacervation agrees well with predictions of the inertial breakup theory. Mayya et al.(2003) reported that addition of low concentrations of sodium dodecyl sulfate (SDS) to theaqueous phase during emulsification promotes GGA coacervate capsule shell formation ondispersed paraffin oil droplets. The SDS concentration used was well below its CMC. Theseworkers suggest that SDS causes deposition of a two-layer shell. Duquemin and Nixon(1985) examined the effect of SDS and cetrimide on complex coacervate formation andencapsulation by complex coacervation. Three SDS concentrations were used: 0.07, 0.2,and 0.35 w/v%. The 0.07 w/v% SDS solution was below the CMC of SDS. Although theweight of coacervate obtained at pH 4.35 and 40°C decreased linearly with increasing SDSconcentration, 0.07 w/v% SDS caused very little reduction. Significantly, Duquemin andNixon (1986) reported that 0.07 w/v% SDS caused a major reduction in the amount of corematerial (phenobarbitone) encapsulated.

The author has historically avoided the addition of surfactants to a complex coacerva-tion system. I am concerned that surfactants will have a negative effect on capsule quality.In my experience, nonionic surfactants have consistently had a negative effect on capsulequality; this is consistent with the observations of Luzzu and Gerraughty (1964). Neverthe-less, the positive results with an anionic surfactant reported by Mayya et al. (2003) coupledwith the positive result with a cationic surfactant (cetrimide) reported by Duquemin andNixon (1985) indicate that further studies of how ionically charged surfactants affect com-plex capsule formation are warranted.

Although it is not complex coacervation, the report by Vinietsky and Magdassi (1997)that soybean oil droplets are encapsulated by an SDS–Type A Gelatin complex is interest-ing. Encapsulation occurred at pH 4 and an SDS concentration of 1.5–2.0 mM. This SDSconcentration range is below the CMC of SDS. The gelatin concentration after SDS addi-tion was 0.3 mM.

Yan (2005) claims the formation of a microcapsule structure in which an agglomerationof primary microcapsules is encapsulated by an outer shell. Each individual primary micro-capsule in the agglomeration is claimed to have a primary shell and this agglomeration isencapsulated by an outer shell. Yan (2005) described the formation of this capsule structureby a GP complex coacervation procedure in which the aqueous phase contained 0.5%sodium ascorbate. The first step is to prepare an 8.33 w/w% gelatin and 0.5% sodiumascorbate solution in water at 50°C. A fish oil concentrate is emulsified in this solutionunder high shear. The resulting oil-in-water emulsion has oil droplets with an average sizeof 1 µm. After it is formed, the emulsion is diluted by addition of a 0.5% aqueous sodiumascorbate solution at 50°C. A 5% sodium polyphosphate plus 0.5% sodium ascorbate solu-tion is then added. After the pH is adjusted to 4.5, the system is cooled, thereby formingcoacervates that coats the individual oil droplets, which creates primary microcapsules.The temperature at which this occurs and the rate of cooling to this temperature are notmentioned, but it is noted that the primary microcapsules begin to agglomerate as cooling iscarried out to above the gel point of the coacervate. Upon further cooling of the system,

Microencapsulation of Flavors by Complex Coacervation 159

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 159

Page 166: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

additional coacervate forms and coats the agglomerates of primary microcapsules, therebycreating an agglomerate of primary microcapsules with an outer shell and an average sizeof 50 µm. Once the system is cooled to 5°C, glut is added and allowed to react for 12 h withthe suspended capsules in the system under continuous stirring for 12 h. The microcapsulesuspension is subsequently washed with water and spray dried to give a free-flow powder.

Two interesting complex coacervation encapsulation systems that contain an essentiallynonionic polymer have been reported. Although such polymers are not believed to bedirectly involved in complex coacervate formation, they undoubtedly affect it in some man-ner. One system was reported by Jizomoto (1984). He reported that the addition of smallamounts of a nonionic polymer like polyethylene oxide or poly (ethylene glycol) expandedthe pH range over which a GGA complex formed. By using this approach, it was possibleto prepare GGA capsules loaded with paraffin oil at pH 6.5. The pH range over which GGAcoacervates were formed was 2–9. This type of system offers a possible method of encap-sulating active agents sensitive to the acidic conditions characteristic of a typical GGAencapsulation system. The author views it as a process that combines complex coacervationwith polymer–polymer incompatibility.

The second procedure was reported by Xing et al. (1973). They prepared capsaicin-loaded GGA microcapsules in the presence of low concentrations of hydroxylethyl cellu-lose (HEC), poly (vinyl alcohol) (PVA), and poly (vinyl pyrrolidone) (PVP). It was notedthat the presence of HEC yielded GGA microcapsules with a better morphology and geom-etry than capsules prepared in the presence of PVA or PVP. These polymers were classifiedby the authors as surfactants, but it is possible that differences in their polymer–polymerincompatibility behavior could contribute to the observed results even at the low concentra-tion used.

Cross-Linking of Gelatin-Based Coacervate Capsule Shells

When initially formed, gelatin-based complex coacervate capsule shells are highly waterswollen and melt if reheated. They also dissolve in warm aqueous media, thereby releasingtheir core. This latter property is highly desirable in many food applications, but it alsoposes problems because the isolation of discrete gelatin-based coacervate capsules thathave not been cross-linked in some way is difficult. The shell of such capsules is highlywater swollen and melts if subjected to relatively low levels of thermal energy. Extractivedrying with a water-miscible solvent at low temperatures is one option, but extraction ofcore material by the extractive drying solvent must be minimized.

In any case, drying capsules that have not been cross-linked in some manner is an issue.For this reason, it is common practice to treat gelatin coacervate shells in some way in orderto stabilize them, so that they can be dried using conventional spray-drying techniques andfluidized bed units. Historically, this has been done by using aldehydes or tannins to cross-link gelatin-based capsule shells. Treatment with an aldehyde has been the most commonapproach taken. Formaldehyde and glut are two aldehydes cited in many publications.However, glut, a five-carbon-chain dialdehyde, is used by most commercial capsuleproducers.

Glut effectively cross-links gelatin-based complex coacervates and insolubilizes themunder conditions that rapidly and completely dissolve untreated capsules (1973). Glutuptake at 4°C by acid or alkaline precursor gelatin gels ranges from 0.9 to 1.4 mM/g gela-tin. Initial solids content of these gels varied from 1.4 to 5.5 wt%, significantly lower than

160 Chapter 7

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 160

Page 167: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

the 12–17 wt% initial solids content of the GGA gels treated with glut. GGA gels formedfrom both types of gelatins had a glut uptake of 0.54–0.65 mM/g gelatin when the reactionwas carried out at 4°C. Glut uptake by GGA gels formed with acid precursor gelatin essen-tially doubled when the reaction temperature was increased to 28°C. This increase wasattributed to temperature-dependent changes in the gel structure of the coacervate (1973).Since a 10 wt% GA solution did not react with a significant amount of glut, glut consump-tion by GGA coacervate gels is attributed to reaction with gelatin.

The amount of glut reacting with the gelatin in a GGA gel at acid pH generally does notexceed the titratable amino content of the gelatin. Thus, glut produces a lightly cross-linkedgel structure. Such cross-linked gels are largely insoluble in water, but retain an ability toswell in water. They are also able to absorb moisture at a relative humidity (RH) above70%. Thus, the shell of glut-treated GGA and other complex coacervate capsules remainssensitive to moisture. At high RH, the amount of moisture absorbed by such shells is suffi-cient to plasticize them and thereby reduce their barrier properties significantly. For thisreason, glut-treated complex coacervate microcapsules loaded with volatile flavors are typ-ically unstable at high RH. This should also be true for such capsules loaded with oxygen-sensitive flavors stored in air at high RH.

Although glut-treated complex coacervate capsules have been approved for specific fla-vor uses, the safety of capsules cross-linked with aldehydes such as glut has always beenopen to question. For this reason, interest in an alternate ways to stabilize complex coacer-vate capsule shells has existed for some time. The goal is to produce stabilized capsules thatare broadly accepted as food grade. One of the first alternate approaches involved posttreat-ing complex coacervate capsules with aqueous tannic acid solutions. Tannic acid rapidlyand dramatically shrinks coacervate capsule shells, thereby reducing the water content ofthe capsule shells and greatly increasing their ease of capsule isolation and drying. How-ever, the interaction of tannic acid with gelatin is intense and rapid. This makes it difficultto achieve precise control of the treatment process required in order to obtain reproducibleresults. When capsules with a continuous core/shell structure and high oil loading aretreated, the effective degree of cross-linking achieved can be so intense that the capsulescrack and break open upon drying. Lot-to-lot variations in tannic acid properties areanother issue. Nevertheless, various workers continue to explore the use of tannins and nat-ural phenolic compounds as cross-linking agents for coacervate gels.

Xing et al. (2004) reported that they successfully treated GGA capsules with tannins.The GGA capsules were prepared in the presence of HEC, PVA, or PVP and subsequentlyimmersed for 10 h in pH 8–9 aqueous media that contained 2.4 w/v% tannins. The capsulesso treated contained upon drying 19% core material, much lower than the core content ofcontinuous core/shell capsules typically produced by complex coacervation. Further, thecore material was sonicated in a GA/HEC solution for 30 min. before gelatin was added tothe system and coacervation induced. This suggests that the capsules isolated had a multi-core structure and not a continuous shell/core structure. Stresses that tannins induce whenthey rapidly shrink a water-swollen capsule shell may have less effect on capsule stabilitywhen the core material is distributed in small droplet form throughout the final dry particle.

Strauss and Gibson (2004) reported that treating gelatin and pectin-gelatin coacervategels with plant phenolics increased their mechanical strength and thermal stability as wellas reduced swelling. They interpreted their results as being consistent with a picture ofpolyphenols reacting under oxidizing conditions with gelatin side chains to form covalentcross-links. It was noted that coffee, grape juice, and various other plant materials contain

Microencapsulation of Flavors by Complex Coacervation 161

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 161

Page 168: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

enough phenolics to be effective cross-linking materials, so that isolation of the active com-ponents was not necessary. The effect of plant phenolics on capsules with a gelatin-basedcoacervate shell was not reported. The effect of lot-to-lot variability of the natural productsolutions on degree of effective cross-linking achieved was also not discussed.

Another non-aldehyde route to chemical cross-linking gelatin-based complex coacer-vate capsule shells involves using transglutaminase (TG), an enzyme produced by micro-bial fermentation and sold in the United States by Ajinomoto Food Ingredients, Paramus,NJ. Dickenson reviewed the use of covalent cross-linking enzymes to introduce cross-linksas a tool for controlling the rheology and stability of protein-based foods (Dickinson,1997). He noted that in 1997 TG, an extracellular product, was the only commerciallyavailable cross-linking enzyme, although another enzyme, lysyl oxidase, should be ofinterest to food technologists. TG is commercially available, because it is readily isolatedfrom the broth of fermented Streptoverticillium mobaraense. Ajinomoto literature stressesthat its TG functions without calcium. This is important, because many common foodproteins such as casein tend to precipitate at relatively low calcium ion concentrations(Dickinson, 1997).

TG introduces cross-links between protein molecules, because it catalyzes the acyltransfer reaction between the γ-carboxyamide group of a glutamine residue of a protein andthe ε-amino group of a lysine residue of a protein (Folk and Finlayson, 1977). The twoprotein molecules may be different. Various Ajinomoto data sheets specify the range of pHand temperature conditions under which Activa®, the trademarked name for Ajinomoto’scommercial TG, will function. The optimal temperature range is 50–55°C, while the opti-mal pH range is 6–7. The enzyme functions outside these ranges, although at a reducedactivity level. Since uncross-linked gelatin-based coacervate gels typically melt above35–40°C and swell significantly at neutral pH values, TG –induced cross-linking of suchgels will not be carried out under optimal conditions.

Cho et al. (2003) used TG to crosslink capsules loaded with fish oil that were formedby a double emulsion process. The capsule shell was isolated soy protein (ISP). The fishoil was first emulsified in a 10% ISP aqueous solution that contained 0.025% TG. Thisemulsion was then dispersed in corn oil that contained 3% Span 80. The resulting doubleemulsion was warmed to 37°C and held at this temperature for 4 h. During this time, theprotein solution was converted to a gel.

Soper and Thomas (2000, 2001) disclosed a process in which TG was used to cross-linkthe shell of capsules formed by complex coacervation. The aqueous capsule systemdescribed as the example in their patents was formed by complex coacervation of gelatin bya CMC–GA mixture. The aqueous capsule system produced was cooled to 5–10°C atwhich temperature its pH was adjusted to 7.0. TG was slowly added to the system andallowed to react with the capsules for 16 h at 10°C. The TG was then inactivated by lower-ing the system’s pH to 2.75 with concentrated citric acid. No specific properties of the cap-sules cross-linked by TG were presented.

Complex Coacervation Encapsulation Technology Issues

Complex coacervation encapsulation procedures have existed for 50 years and continue tobe used to produce large amounts of microcapsules for various commercial applications.Nevertheless, such procedures have a number of issues. Consistent quality control of com-plex coacervation encapsulation processes can be a challenging task, because final capsule

162 Chapter 7

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 162

Page 169: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

properties are very sensitive to small changes in the many variables associated with complexcoacervation. Since complex coacervation is a polymer phase-separation process, repro-ducibility is very dependent on solution properties of the polymers involved. Lot-to-lotvariations in properties of the polymers used will impact process control. The degree ofphase separation that occurs in a specific complex coacervate encapsulation system is influ-enced by the polymers involved, their MW, ionic charge density, and concentration. Varia-tions in these properties affect polymer phase-separation behavior. The source, type ofgelatin used, acid or alkaline precursor, and bloom strength are variables that must be con-trolled. Since gelatin is hydrolytically unstable, thermal conditions throughout the encapsu-lation protocol must be controlled. The ratio of polymers present in the complex coacervatesystem is another factor. System pH, temperature, and salt ion content also influence thephenomenon and must be controlled. Other factors that may affect coacervation includethe type of acid used to adjust system pH as well as the presence of surfactants in the sys-tem. Soper et al. (2000) also describe a number of factors that affect flavor encapsulation bycomplex coacervation.

The core material being encapsulated is another process variable. This is particularlytrue when the core material is a complex mixture of many components of differing polar-ity. Flavors are a specific example of such types of core materials. In order to illustratethis point, Table 7.4 lists experimentally determined compositions of a sample oforange, lemon, and mint oils (Arneodo et al., 1988/1989). Although these flavor oils arecomposed of a mixture of hydrocarbons, ketones, aldehydes, alcohols, esters, and otherunidentified components, the dominant component is limonene. The data in Table 7.5(Arneodo et al., 1988/1989) establish that some of the minor components present in orangeand lemon oil partition into the aqueous phase. Limonene glycol, a limonene-degradationproduct that impacts citrus oil quality, was present in the aqueous phase equilibrated withboth oils.

Microencapsulation of Flavors by Complex Coacervation 163

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Table 7.4. Composition of orange oil, lemon oil, and mint oil flavors

Component Orange oil Lemon oil Mint oil

Hydrocarbons (limonene, others) 96 73 0.9Ketones 0.7 21.8 33.5Aldehydes 2 1.8 60.4Alcohols 0.47 1.2 60.4Esters 0.05 0.9 2.9

Table 7.5. Composition of orange oil, lemon oil, and mint oil that partition at 30°C and 50°C(component concentration in aqueous phase, ppm)

Orange oil Lemon oil Mint oil

Component 30°C 50°C 30°C 50°C 30°C 50°C

Hydrocarbons 2(limonene, others)

Ketones 185 185Aldehydes 12 9Alcohols 6 8 36 31 212 245Esters 8 7Limonene glycol 10 12 8 5

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 163

Page 170: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Even though limonene is the major component of orange and lemon oils, the interfacialbehavior of both oils is dominated by polar components present in small amounts. Theinterfacial tension of D-limonene against water at 50°C declines from 35 to 23 mJ/m2 over5 h (Arneodo et al., 1988). These interfacial tension values are much greater than thoseobtained with orange and lemon oils against water at 50°C (46). In contrast, the initialinterfacial tension value of 1-octanol against water at 50°C is 8 mJ/m2 and it declines to2 mJ/m2 after 5 h. The interfacial tension of decanal against water at 50°C declines over 5 hfrom an initial value of 10–6 mJ/m2. Although both of these compounds are present inorange and lemon oils in small amounts, they have an affinity for the oil–water interfaceand undoubtedly impact the interfacial activity of both oils.

Interfacial tension values of GA coacervate and supernatant phases measured at 50°Cagainst D-limonene decay from an initial value of 14–10 mJ/m2 after 5 h (Arneodo et al.,1988). Since the coacervate phase is a much more concentrated polymer solution, it is sur-prising that the two interfacial tension decay curves overlap as well as they do. The slope ofthe interfacial tension decay curve decreases significantly at an interface age of 1 h afterwhich linear slow decay continues up to at least 5 h.

Interfacial tension values of orange and lemon oils measured against water at 25°C,30°C, or 50°C decline significantly as the interface ages over a 5–10 h period (Arneodoet al., 1988). Initial interfacial tension values are 4–8 mJ/m2. Values after interfacial agingrange from 6 mJ/m2 to a value too low to be measured by the Wilhelmy plate method.The rate of interfacial tension decline is reduced but not eliminated when the system tem-perature was lowered to 1.2°C. The interfacial tension behavior of both oils against thesupernatant phases isolated from GGA, GP, and GAlg coacervate systems was similar tothat observed with water. This was not surprising, because the supernatant phases are dilutepolymer solutions. However, it is interesting that the interfacial tension of GGA, GP, andGAlg coacervate phases against orange and lemon oils is similar to that observed withwater. The primary difference with the coacervate phases is that the rate of interfacial ten-sion decline was generally faster at the same temperature than that observed with waterand an interfacial tension too low to measure by the Wilhelmy plate method was observedmore often.

Interfacial tension aging at an oil/aqueous phase interface is an indication of interfaceinstability. The precise cause of this instability is difficult to define because it can be causedby many factors. Furthermore, a very small amount of interfacially active material can havea major effect on interfacial tension. For flavor encapsulation procedures, the possibilitythat interfacial aging is due to one or more chemical reaction(s) at the flavor oil/water inter-face is a concern. Candidate reactions in a gelatin-based complex coacervation encapsula-tion system include oxidation, hydrolysis, and aldehyde condensation with any freeprimary amino groups in the coacervate. However, interfacial aging could also be causedby slow physical adsorption of interfacial active agents at the interface and slow rearrange-ments of the molecules in the adsorbed layer. Although polymer solutions frequently showthis type of behavior, the author favors interfacial chemical reactions as the primary causeof interfacial aging. This point of view is based on several experimental observations. First,dispersed precipitate particles or a continuous intact film often formed at citrus oil/waterinterfaces aged in several hours. When such systems were agitated, the aqueous phasebecame cloudy. A second observation is the significant interfacial tension aging that bothoils experience against highly purified water. Gelatin and other polymers that could con-tribute to prolonged interfacial aging and interfacial film formation are not initially present

164 Chapter 7

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 164

Page 171: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

in such systems. Finally, interfacial aging can be markedly reduced by reducing the temper-ature of the system to 2°C.

Gelatin has been the dominant polycation in complex coacervation encapsulation proto-cols since 1957, because it is readily available at reasonable cost and forms a gel structureupon cooling, thereby setting an embryonic capsule shell. Nevertheless, the use of gelatincreates a number of issues. For example, the appearance of mad cow disease raised seriousconcerns about the safety of bovine gelatin for human consumption. This issue promptedglobal gelatin manufacturers to support extensive studies of the possible transmission ofprions via gelatin consumption. These studies have concluded that commercially availablefood grades of gelatin are not a potential source of mad cow disease transmission. Never-theless, residual concern about its safety remains; if not for mad cow disease, other diseasesmay appear in the future. Web sites posted by the Gelatin Manufacturers of Europe (GME)or Gelatin Manufacturers of Asia Pacific (GMAP) provide a means of monitoring the regu-latory status of various types of gelatin.

Another gelatin issue is the reality that food products must increasingly meet globaldietary requirements. This has promoted a search for materials that can replace beef andpork gelatins. Fish skin gelatin is one candidate. Soper (2001) disclosed the formation ofcapsules by coacervation of warm water fish gelatin with a CMC/GA mixture. Fish skingelatin is currently much more expensive than beef or pork gelatin but can be used.

Residual concern about the safety of gelatin has sparked interest in producing microcap-sules by complex coacervation of other proteins. Weinbreck et al. (2003) disclose an encap-sulation procedure that is based on the complex coacervation of whey protein. Although arange of polyelectrolytes is claimed to be suitable for the coacervation process, GA is citedas the preferred one. The preferred coacervation pH range is 2.5–4.5. The capsules pro-duced can be hardened by treatment with an aldehyde or enzyme. A claimed feature of theirprocess is that it can be carried out at or below room temperatures.

Solvent Exchange: A Unique Property of Complex CoacervateMicrocapsules

An interesting feature of gelatin-based complex coacervate microcapsules is their ability toundergo solvent exchange; that is, a water-immiscible liquid originally encapsulated withina complex coacervate shell can be exchanged with a second, chemically different liquidthat has finite water miscibility. The exchange process occurs by diffusion through intactcapsule shells and broadens the range of core materials that can be incorporated in a com-plex coacervate capsule. Because of solvent exchange, capsules formed by complex coac-ervation can be loaded with liquids and flavors that cannot be encapsulated at the time ofcapsule formation.

Figure 7.2 is a simplified flow diagram of the Brynko and Olderman solvent exchangeprocedure (Brynko and Olderman, 1970). The first step is to prepare complex coacervatemicrocapsules loaded with an oil that is water immiscible. For food applications, the oilwill be edible. Capsules to be subjected to solvent exchange must have a water-swollenshell. It may be chemically cross-linked or uncross-linked. Once such capsules are avail-able, they are subjected to the solvent exchange treatment. In one case, Brynko and Older-man (1970) dispersed a water-wet filter cake of oil-loaded capsules in a concentrated(80 w/w%) aqueous sorbitol solution for a defined time (e.g., 30 min). The purpose of thistreatment was to introduce into the capsule shell a finite amount of sorbitol, a compound

Microencapsulation of Flavors by Complex Coacervation 165

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 165

Page 172: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

designed to plasticize the capsule shell. After excess sorbitol solution was removed fromthe capsule slurry, typically by filtration, a finite volume of ethanol was added to the sys-tem. Food-flavoring agents may be dissolved in the ethanol. After a finite diffusion or sol-vent exchange time (e.g., 30 min), excess ethanol solution may be removed from the systemby decantation or filtration and replaced with a fresh ethanol solution. This cycle isrepeated until the desired extent of solvent exchange is achieved. Brynko and Olderman(1970) completed their solvent exchange process by immersing the capsules in excessanhydrous ethanol free of solute. The objective is to remove the last remaining traces ofwater from the capsule shell, thereby sealing them. After the capsules are dried, they form afree-flow powder.

Brynko and Olderman (1970) teach that the shell of a complex coacervate capsule mustbe water-swollen in order to successfully undergo solvent exchange. The water may bepresent either because the capsule was never dried after formation or because water wasadded after the capsule had been dried. Water is essential. It causes complex coacervateshells to swell and become porous to liquids that have finite water miscibility. This enablesthe diffusion of such liquids into the interior of capsules. Since solvent exchange is a diffu-sion process, a finite time is required. Solvent exchange is a diffusion-controlled equilibra-tion or partitioning process, so complete removal of the oil originally carried by a capsule isdifficult to achieve. A small but finite amount of oil is typically left in a capsule after sol-vent exchange is deemed complete. In some cases, a water-swollen coacervate capsuleshell acts as a semi-permeable membrane that allows diffusion of the solvent exchange liq-uid into the capsule, but not diffusion of the oil out. The resulting increase in liquid contentof the capsule may be so significant that the capsules visibly increase in diameter.

Although water must be present in the shell of a complex coacervate when solventexchange is carried out, its presence in the capsule shell after completion of the solventexchange process is detrimental to prolonged storage stability. Brynko and Olderman(1970) correlated retention of a solvent exchange with its dielectric constant. They report

166 Chapter 7

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Oil-loaded capsulesShell: Water-swollen

Gelatin-based

Solvent exchange liquid withfinite H2O and core miscibility

Agitation for several hours(exchange by diffusion)

Anhydrous water-misciblesolvent (e.g., ethanol)

Brief agitation (extraction of water)

Isolation and dryingDry capsules loaded with

exchanged liquid

Figure 7.2. Simplified flow diagram of solvent exchange coacervation procedure (from Brynkoand Olderman, 1970, with permission).

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 166

Page 173: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

that single liquids with finite water miscibility and a dielectric constant below 20 yieldstable capsule powders, whereas liquids with a dielectric constant above 20 do not. Sincethe dielectric constant of ethanol, a desirable exchange solvent, is 24.3, ethanol-loaded cap-sules prepared by solvent exchange are unstable. The lower the dielectric constant of theexchange liquid, the more stable the capsules. Mixtures of dielectric solvent liquids withlow dielectric constants can be used as long as the dielectric constant of the mixture is <20.Thus, stable capsules that contain a finite concentration of ethanol can be prepared as longas the other solvents present in the capsule have a low dielectric constant and the solventmixture has a dielectric constant below 20, preferably below 14.

The instability of capsules loaded with a high dielectric constant solvent is attributed totheir ability to readily absorb water from the atmosphere in which the capsules are stored.The absorbed water plasticizes the capsule shell, thereby facilitating rapid solvent release.Brynko and Olderman (1970) note that capsules isolated after solvent exchange with butylacetate contain 0.01% water. These have excellent storage stability, because the dielectricconstant of butyl acetate is 5 and butyl acetate does not promote water absorption from theatmosphere under typical conditions (e.g., RH <70%). Although Soper et al. (2000a, b) alsodisclose a solvent exchange procedure for preformed capsules. In one example, capsulesloaded with purified vegetable oil were formed by the complex coacervation of gelatin witha mixture of GA and sodium CMC. Both chemically cross-linked and uncross-linked cap-sules were used. As noted by Brynko and Olderman (1970), Soper et al. (2000a, b) requirethe shell of capsules that are candidates for solvent exchange to be water swollen. Thus,capsules that were dried after formation were placed for 5 min in a flavor-water mixture.They were subsequently transferred to a closed plastic container and incubated for 24 hbefore use. The inventors note that the shell of capsules subjected to their solvent exchangeprocedure can be treated in order to prevent removal of flavor from microcapsules andwater removal from the shells was specifically cited. The importance of residual water todry capsule stability was not referenced, but it is reasonable to suggest that high humiditywill affect storage stability of capsules subjected to solvent exchange, as disclosed bySoper et al. (2000) process. Soper et al. (2000, 2001) do not discuss the need for the solventexchange system to remain in one phase throughout the solvent exchange process nor theeffect of the exchange solvent’s dielectric constant on solvent exchange.

Soper et al. (2000) report that capsules with water-swollen shells can absorb flavorsfrom the gas phase. They placed oil-loaded capsules having a water-swollen shell in a con-tainer and purged them with a loaded gaseous phase that contained an aroma. After a finitepurging time, 0.5–5 h depending on the aroma, the aroma absorbed by the capsules couldbe detected. A variety of applications were cited, including the separation and concentra-tion of volatile hydrophilic aroma components from hydrophobic components.

Summary

Complex coacervation encapsulation technology is versatile and adaptable. Capsules pro-duced by this technology have many interesting features, which continue to attract interestin them. Soper (1995) discusses applications of flavors encapsulated by complex coacerva-tion. Such capsules are prime candidates for flavor oil encapsulation. They offer food tech-nologists a high degree of versatility and flexibility. Microcapsules with sizes ranging froma few microns to over a millimeter in diameter can be produced. Such capsules typicallycarry a flavor payload of 60–90 wt%, although lower and higher payloads can be produced.

Microencapsulation of Flavors by Complex Coacervation 167

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 167

Page 174: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

The capsules can be supplied as an aqueous slurry or dry powder. Drying is typically donein a fluidized bed or spray drier. Capsules supplied as aqueous slurries are able to withstandfinite handling or processing stresses, because water-swollen complex coacervate shells arerubbery and resist failure. Such encapsulation protocols are not free of limitations or issuesincluding: component partitioning during capsule manufacture, moisture sensitivity duringstorage, and reaction of aldehyde components of flavors with gelatin-containing shells onstorage. Nevertheless, the technology continues to diversify and expand. Because many ofits characteristic properties remain uncharacterized and unexploited, it is a fruitful area forfurther study.

ReferencesArneodo, C., Baszkin, A., Benoit, J.-P. and Thies, C. 1988/1989. Interfacial studies of essential oil-water systems.

Colloids and Surfaces 34: 159–169.Arneodo, C., Baszkin, A., Benoit, J.-P. and Thies, C. 1988a. Interfacial tension behavior of citrus oil components,

Absts. 6th International Conference on Surface and Colloid Science, Hakone, Japan, p. 106.Arneodo, C., Baszkin, A., Benoit, J.-P. and Thies, C. 1988b. Interfacial tension behavior of citrus oils against

phases formed by complex coacervation of gelatin, In Flavor Encapsulation, S.J. Risch and G.A. Reineccius,eds., American Chemical Society, Washington, DC, Chapter 15.

Brenner, J. 1983. The essence of spray dried flavors: the state of the art. Perfumer and Flavorist 8: 40–44.Brynko, C. and Olderman, G.M. 1970. Replacement of capsule contents by diffusion, US Patent 3,516,943 (June

23, 1970).Bungenberg de Jong, H.G. 1949. Complex colloidal systems, In Colloid Science, Vol. II, H.R. Kruyt, ed., Elsevier,

Chapter 10.Burgess, D.J. 1990. Practical analysis of complex coacervate systems. J. Colloid and Interface Sci. 140: 227–238.Chilvers, G.R. and Morris, V.J. 1987. Coacervation of gelatin-gellan gum mixtures and their use of microencapsu-

lation. Carbohydrate Polymers 7: 111–120.Cho, Y.-H., Shim, H.K. and Park, J. 2003. Encapsulation of fish oil by an enzymatic gelation process using transg-

lutaminase cross-linked proteins. J. Food Sci. 68: 2717–2723.Commandur, B., Arneodo, C., Benoit, J.-P. and Thies, C. 1989. A viscosity study of gelatin-based complex coacer-

vates. Proceed. Intern. Symp. Control. Rel. Bioact. Mater. 16: 279–280.Duquemin, S.-J. and Nixon, J.R. 1985. The effect of sodium lauryl sulphate, cetrimide and polysorbate 20 surfac-

tants on complex coacervate volume and droplet size. J. Pharm. Pharmacol. 37: 698–702.Duquemin, S.-J. and Nixon, J.R. 1986. The effect of surfactants on the microencapsulation and release of pheno-

barbitone from gelatin-acacia complex coacervate microcapsules. J. Microencapsulation 3: 89–93.Dickinson, E. 1997. Enzymic cross-linking as a tool for food colloid rheology control and interfacial stabilization.

Trends in Food Science and Technology 8: 34–339.Folk, J.E. and Finlayson, J.S. 1977. The ε-(γ-glutamyl)lysine crosslink and the catalytic role of transglutaminase.

Adv. Protein. Chem. 31: 1–133.Goubet, I., Le Quere, J.L. and Voilley, A.J. 1998. Retention of aroma compounds by carbohydrates: influ-

ence of their physicochemical characteristics and their physical state. A review. J. Agric. Food Chem. 46:1981–1990.

Green, B.K. and Schleicher, L. 1957. Oil-containing microscopic capsules and method for making them, USPatent 2,800,457, July 23.

Jegat, C., Taverdet, J.L. 2000. Stirring speed influence study on the microencapsulation process and on the drugrelease from microcapsules. Polymer Bulletin 44, 345–351.

Jizomoto, H. 1984. Phase separation induced in gelatin-base coacervation systems by addition of water-solublenonionic polymers I: Microencapsulation. J. Pharm Sci. 73: 879–882.

Kerkof, P.J. and Thijssen, H.A. 1974. Retention of aroma components in extractive drying of aqueous carbohy-drate solutions. J. Food Technol. 9: 415–423.

Kim, J.-C., Song, M.-E., Lee, E.-J., Park, S.-K., Rang, M.-J. and Ahn, H.-J. 2001. Preparation and characterizationof triclosan-containing microcapsules by complex coacervation. J. Dispersion Science and Technology 22:591–596.

168 Chapter 7

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 168

Page 175: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Koh, G.-L. and Tucker, I.G. 1988a. Characterization of sodium carboxymethylcellulose-gelatin complex coacer-vation by viscosity, turbidity and coacervate wet weight and volume measurements. J. Pharm. Pharmacol. 40:233–236.

Koh, G.-L. and Tucker, I.G. 1988b. Characterization of sodium carboxymethylcellulose-gelatin complex coacer-vation by chemical analysis of the coacervate and equilibrium fluid phases. J. Pharm. Pharmacol. 40: 309–312.

Leuenberger, B.H. 1991. Investigation of viscosity and gelatin properties of different mammalian and fishgelatins. Food Hydrocolloids 5: 353–361.

Liu, X.-D., Atarashi, T., Furuta, T., Yoshii, H., Aishima, S., Ohkawara, M. and Linko, P. 2001. Microencapsulationof emulsified hydrophobic flavors by spray drying. Drying Technology 19: 1361–1374.

Luzzu, L.A. and Gerraughty, R.J. 1964. Effect of selected variables on the extractability of oils from coacervatecapsules. J. Pharm Sci. 53: 429–431.

Mayya, K.S., Bhattacharyya, A. and Argillier, J.-F. 2003. Micro-encapsulation by complex coacervation: influenceof surfactant. Polym. Int. 52: 644–647.

McMullen, J.N., Newton, D.W. and Becker, C.H. 1984. Pectin-gelatin complex coacervates II: effect of microen-capsulated sulfamerazine on size, morphology, recovery, and extraction of water-dispersible microglobules. J. Pharm. Sci. 73: 1799–1803.

Menting, L.C. and Hoogstad, B. 1967. Volatiles retention during the drying of aqueous carbohydrate solution. J. Food Sci. 32: 87–90.

Re, M.I. 1998. Microencapsulation by spray drying. Drying Technology 16: 1195–1236.Reineccius, G. 2004. The spray drying of food flavors. Drying Technology 22: 1289–1324.Remunan-Lopez, C. and Bodmeier, R. 1996. Effect of formulation and process variables on the formation of

chitosan-gelatin coacervates. International Journal of Pharmaceutics 135: 63–72.Rulkans, W.H. and Thijssen, H.A. 1978. Retention of organic volatiles in spray-drying aqueous carbohydrate

solution. J. Food Technol. 7: 95.Saeki, K. and Hosoi, N. 1984. Microencapsulation by a complex coacervation process using acid-precursor

gelatin. Appl. Biochem. Biotechnol. 10: 251–254.Schmitt, C., Sanchez, C., Desobry-Banon, S. and Hardy, J. 1998. Structure and technofunctional properties of

protein-polysaccharide complexes: a review. Critical Rev. Food Sci. and Technol. 38: 689–753.Soper, J.C. 1995. Utilization of coacervated flavors, In Encapsulation and Controlled Release of Food Ingredients,

S.J. Risch and G.A. Reineccius, eds., American Chemical Society, Washington, DC.Soper, J.C. 1997. Method of encapsulating food or flavor particles using warm water fish gelatin and capsules pro-

duced therefrom. US Patent 5,603,952 (February 18, 1997).Soper, J.C. and Thomas, M.T. 2000. Enzymatically protein-encapsulating oil particles by complex coacervation.

US Patent 6,039,901 (March 21, 2000).Soper, J.C. and Thomas, M.T. 2001. Enzymatically protein-encapsulating oil particles by complex coacervation.

US Patent 6,325,951 (December 4, 2001).Soper, J.C., Kim, Y.D. and Thomas, M.T. 2000a. Method of encapsulating flavors and fragrances by controlled

water transport into microcapsules. US Patent 6045835 (April 4, 2000).Soper, J.C., Yang, X. and Thomas, M.T. 2000b. Method of encapsulating flavors and fragrances by controlled

water transport into microcapsules. US Patent 6106875 (August 22, 2000).Strauss, G. and Gibson, S.M. 2004. Plant phenolics as cross-linkers of gelatin gels and gelatin-based coacervates

for use as food ingredients. Food Hydrocolloids 18: 81–89.Thies, C. 1973. The reaction of gelatin-gum Arabic coacervate gels with glutaraldehyde. J. Colloid and Interface

Sci. 44: 133–141.Thies, C. 1999. A Short History of Microencapsulation Technology, Microspheres, In Microcapsules & Lipo-

somes, Vol. 1: Preparation & Chemical Applications, R. Arshady, ed., Citus Books, London, pp. 43–54.Thijssen, H.A.C. 1975. In Freeze Drying and Advanced Food Technology, S.A. Goldblith, L. Rey and W.W.

Rothmayer, eds., Academic Press, London.Veis, A. 1970. Phase equilibria in systems of interacting polyelectrolytes, In Biological Polyelectrolytes, A. Veis,

ed., Marcel Dekker, NY.Veis, A. and Aranyi, C. 1960. Phase separation in polyelectrolye systems. I. Complex coacervates of gelatin 64:

1203–1210.Venkataram, J.S. 1995. Indomethacon sustained release from alginate-gelatin or pectin-gelatin coacervates. Int. J.

Pharm. 126: 161–168.Vinietsky, Y. and Magdassi, S. 1997. Microencapsulation by surfactant-gelatin insoluble complex: effect of pH

and surfactant concentration. J. Colloid and Interface Sci. 189: 83–91.

Microencapsulation of Flavors by Complex Coacervation 169

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 169

Page 176: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Weinbreck, F., De Kruif, C. and Schrooyen, P. 2003. Complex coacervates containing whey proteins. WO03/106014 A1 (December 24, 2003).

Xing, F., Cheng, G., Yang, B. and Ma, L. 2004. Microencapsulation of capsaicin by the complex coacervation ofgelatin, acacia and tannins. J. Applied Poly. Sci. 91: 2669–2675.

Yan, N. 2005. Encapsulated agglomeration of microcapsules and method for the preparation thereof. US Patent6974592 (December 13, 2005).

170 Chapter 7

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_07_149-170 r1.qxd 3/29/07 1:49 PM Page 170

Page 177: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

8 Confectionery Products as Delivery Systemsfor Flavors, Health, and Oral-Care Actives

Jamileh M. Lakkis

Introduction

Despite consumers apprehension about sugar-based products, the confectionery market iscurrently experiencing record growth. Euromonitor International has estimated the globalconfectionery market at more than $142 billion in 2005. This surge is due to two main fac-tors: first, the availability of sugar-free versions of traditional sugared products and second,the new trend in formulating confectionery products with functional actives that can deliverunique health benefits.

The last two decades have witnessed an intense effort to shift the market positioning ofconfectionery products from just pleasantly tasting sweet snacks to a platform for deliveringnutraceuticals, breath fresheners, nicotine, antimicrobial and dental health agents as well asdrug actives. The latter category—including analgesics, insulin, antibiotics, and other phar-maceutical ingredients, although outside the scope of this book—will be referred to helpexplain the mechanisms of delivery and absorption.

Functional confections are currently enjoying an unprecedented acceptability from con-sumers trying to increase their intake of functional and health-promoting ingredients suchas vitamins, minerals, herbal extracts, etc. in a familiar food format, which does not signal ill-ness and can be consumed discreetly. Successful examples of this category include Viactive®

from McNeil, a calcium- and vitamin-containing chewy candy formulated for women, Orbit®

chewing gum from Wrigley’s that claims teeth-cleaning benefits, mentholated lozenges suchas Vicks® and Robitussin® that claim throat relief or nasal decongestion, Listerine Pocket-paks® from Pfizer, and Nicorette®, smoke-cessation chewing gum from GlaxoSmithkline.

The first patent on functional confectionery products was granted to W.F. Semple in thenineteenth century for developing a dentifrice in the form of a chewing gum (Semple, 1869).However, the first commercial product claiming delivery of functional ingredients was a sal-icylic acid-containing chewing gum, Aspergum®, which was marketed in the United Statesin 1924 and is still available today. A breakthrough in utilizing chewing gums as deliverysystems was documented in the clinical finding that smokers may be able to give up smokingby self-titrating the amount of nicotine they absorb (Fernö, 1973; Mulry, 1988; Silagy et al.,2002). This finding was the basis for the development and marketing of nicotine-containingchewing gums, where subjects can chew the gum to release and absorb the needed amountsof nicotine (Benowitz et al., 1987; Mulry, 1988).

Despite these advances, the challenge for true commercial success of functional confec-tions lies mainly in the inability of some formats to deliver therapeutic levels of healthactives and the harsh conditions in the stomach that can sometimes degrade the active beforeit had the chance to reach its target site such as lower intestines or the blood stream. Unlikeflavor delivery, where the only requirement is dissolution of the active from the dosage into

171

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 171

Encapsulation and Controlled Release: Technologies in Food SystemsEdited by Jamileh M. Lakkis

Copyright © 2007 by Blackwell Publishing

Page 178: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

the saliva and its extended release for a desired period of time, delivering therapeutic activesrequires more elaborate capsule or delivery system design and an understanding of the phys-iology of absorption across membranes. Actives incorporated into a confectionery productmust be transported from the dosage carrier to specialized tissues and epithelia and eventu-ally to the target site in the blood stream or the cytoplasm of a particular cell group.

Two types of oral delivery routes can be distinguished; these are local (target release siteis the mouth or throat areas) and systemic (blood stream or specific organ or cell). In design-ing delivery systems, it is imperative to take into consideration not only the physiology andorganizational structure of the oral cavity but also the physicochemical properties of thedelivery system including dose concentration, format, residence time in the mouth, etc.

Physiology and Organization of the Oral Area

Organs that constitute the oral area include the mouth, tongue, and esophagus (Figure 8.1).Within these organs, several regions can be differentiated that are critical for permeabilityand absorption (Squier et al., 1976). The mouth extends from the lips to the oropharynx atthe rear; its temperature and humidity vary greatly during normal activities such as drinkingand eating, thus impacting the active’s dissolution and its absorption. The oral cavity can bedivided into two main regions (Figure 8.2), namely:

1. The oral cavity proper consisting of the tongue, hard and soft palates, and floor of themouth.

2. The outer vestibule consisting of cheeks (buccal mucosa), maxillary (upper jaw), andmandibular (lower jaw) areas.

172 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N Figure 8.1. Views of the oral cavity and pharynx.

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 172

Page 179: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

In humans, the tongue is essential for several processes including moving the food bolusaround in the mouth, chewing, speech, sucking, and swallowing. The latter is achieved byvirtue of the negative pressure created within the oral cavity. The tongue consists of a massof interwoven, striated muscles interspersed with glands and fat and covered with mucusmembrane tissues that are responsible for secreting small amounts of mucus. The tonguesurface contains papillae, which are sensitive to food flavors along with several ridges thathelp grip the food article while the tongue agitates it during chewing.

The tongue is a highly sensitive well-coordinated organ that occupies the middle of themouth; therefore any device placed in the oral cavity should take this into consideration.The sublingual area moves extensively during eating, drinking, and speaking, thus impact-ing the residence time of food bolus or any delivery device placed in the oral cavity (Collinsand Dawes, 1987). The inferior portion of the tongue (under surface leading from the tip ofthe tongue to the floor of the mouth) contains mucus membranes and is smooth and purplein color due to the many blood vessels present. The root contains bundles of nerves, arter-ies, and muscles that branch to the other regions. Nerves from the tongue receive chemicalstimulation from food in solution which gives the sensation of taste.

The esophagus is a muscular tube that connects the pharynx to the stomach. It is approxi-mately 25-cm long and about 2-cm in diameter. Similar to the buccal area, the esophagus islined with stratified squamous epithelium lining whereas the very remote portion (toward thestomach) is lined with columnar epithelia, which are highly specialized for absorption. Themain role of the esophagus is to move ingested materials from the mouth area to the stomachand lower gastrointestinal tract (GIT). The esophageal epithelial area is non-keratinized and

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 173

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Upper lip

Undersideof tongue

Gingiva

Floor of mouth

Lower lip

Alveolar mucosa

Hard palate

Soft palate

Cheek

Tongue

Masticatory mucosa

Lining mucosa

Specialized mucosa

Figure 8.2. Anatomical location and extent of masticatory, lining and specialized mucosa in theoral cavity (Squier and Kremer, 2001 with permission).

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 173

Page 180: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

is lined with mucus-secreting glands that help keep the esophagus moist and protect it fromgastric acidity. Typical food transit time in the esophagus is very short (10–14 seconds).

One peripheral system, the trigeminal nerve, is responsible for specialized sensationsand constitutes an important part of the oral cavity. Its function resembles that of the spinalnerves, which are responsible for the sensation in the rest of the body. The trigeminal nerveis a cranial nerve comprised of three major branches: the ophthalmic nerve, the maxillarynerve, and the mandibular nerve. The sensory function of the trigeminal nerve is to provideconscious awareness of the face and mouth. The maxillary nerve carries sensory informa-tion mainly from the cheek, upper lip, upper teeth and gum, palate and roof of the pharynx.Mandibular nerve carries sensory information from the lower lip, lower teeth and gum, andfloor of the mouth. The mandibular nerve carries touch/position and pain/temperature sen-sations from the mouth but not taste sensations. Unlike touch/position input that takes placeimmediately, pain/temperature sensation experiences a perceptible delay due to theunmyelinated slow-conducting nerve fibers. This type of sensation is mostly caused by aspecific group of chemicals commonly referred to as “sensates” and which include sub-stances that induce cooling, warming, tingle, and similar effects. Sensates have been usedin confectionery formulations to provide perception of refreshment (cooling, tingle) orsoothing (warming) and calming sensations.

Permeability and Barrier Functions of the Oral Cavity

Permeability and barrier selectivity of the oral cavity are complex phenomena. A better appre-ciation for these functions can be gained by understanding the structure and critical functionsof tissues, salivary glands and their secretions as well as their interactions (Rojanasakul et al.,1992). Table 8.1 shows variations in thickness of the oral mucosa in various regions of thehuman oral cavity. However, mucosal thickness does not explain variations in permeability invarious regions of oral cavity.

Physiological and Structural Basis of Transport Routes (Plasma and Epithelial Membranes)

Plasma Membranes

Plasma membranes retain the contents of the cell and act as permeability barriers. Theyallow only certain substances to enter or leave the cell, though the rate of entry is strictlycontrolled. Hydrophobic materials enter the cells easily due to the presence of a lipoidallayer at the cell surface, commonly known as the bilayered lipid membrane, with bands

174 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Table 8.1. Thickness of various regionsof the human oral cavity (Robinson,2000 with permission)

Region (microns) Thickness

Skin 100Hard palate 250Attached gingival 200Buccal mucosa 200–600Floor of mouth 100–200

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 174

Page 181: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

approximately 3 nm in width and an overall thickness of between 8 and 12 nm (Curatolo,1987). Plasma membranes are highly organized structures, where proteins in specific con-formations act as structural elements, transport nutrients, and sample the cell environment.Lipid-soluble substances tend to diffuse along the plasma membranes of the cells whilewater can flow through transcellular routes by virtue of the small polar channels throughthese membranes.

Epithelial Membranes

Most internal and external body surfaces are covered with epithelia, which contain a layer ofbasal lamina and structural collagen underlying layers of epithelial cells. There are severalmorphologically distinct epithelial types, namely, simple squamous (line blood vessels),simple columnar (line stomach and small intestines), and stratified squamous epithelium(line mouth and esophagus).

The epithelium has a vertical dimension of 600 microns through the epithelial ridgesand 250 microns through the areas overlying the connective tissue papillae. The buccalepithelium possesses some net charge, hence its permeability and selective ion transport.Assessing the role of epithelial layers can better be understood by differentiating betweentwo criteria, namely permeability and permselectivity. The former refers to permeationmagnitude (as quantified by electrical resistance), while the latter describes its qualitativeability to show preference for cations or anions or within a series of cations and anions(Fromter and Diamond, 1972; MacKnight and Leader, 1983).

Epithelia of the epidermis, hard palate, and gingivae are keratinized and are known tobe not very permeable to water. Earlier studies showed that these keratinized epithelia con-tain neutral lipids such as acylceramides and ceramides, which have been associated with abarrier function (Wertz and Downing, 1983). Epithelia of the soft palate, sublingual, and buc-cal area as well as those located in the floor of the mouth are nonkeratinized and have shownsignificant permeability to water presumably due to the absence of acylceramides (Squier andHall, 1985).

Oral Mucosa

The oral mucosa represents one type of epithelial membranes that secretes mucus(Figure 8.3). Similar to the skin and intestinal mucosa, oral mucosa mainly protects the oralcavity from harmful substances as well as facilitates absorption of chemical entities. The oralmucosa plays a protective role during mastication, which involves compression and shearforces. Areas such as the hard palate and attached gingivae have a textured surface to resistabrasion and are tightly bound to the underlying bone to resist shear forces. The cheekmucosa is elastic to allow for distension. Like the skin, the human oral mucosa consists ofstratified squamous epithelia. However, unlike the skin, it is always maintained moistbecause of the presence of numerous salivary glands and does not show the presence ofkeratin. These dissimilarities make the oral mucosa more permeable than the skin (Chenand Squier, 1984; Gandhi and Robinson, 1988; Squier and Wertz, 1996).

The mucosa of the human mouth is permeable to various vitamins such as thiamine,ascorbic acid and nicotinic acid. Several investigations have shown that the absorptioncharacteristics of the oral mucosa were broadly similar to those of the small intestine of arat (Evered and Mallett, 1983; Evered et al., 1980).

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 175

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 175

Page 182: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Saliva

Saliva is a mucus, viscous, colorless fluid that originates in the buccal and sublingual glands.It is a unique fluid that plays a significant role in controlling absorption and bioavailability ofingested actives both as an enhancer and as a barrier to permeability. Saliva forms a thin film(0.07–0.10 mm) of hypotonic nature (110–220 milliosmoles/lit) that lubricates and moistensthe inside of the mouth. Saliva is believed to play a significant role in repairing injuries andtears in the oral area due to the abundance of hyaluronic acid molecules. pH of human salivaranges from 7.4 to 6.2 depending on its flow rate (low to high flow rates). Certain foods suchas carbohydrates, due to bacterial action, can reduce saliva pH to 3–4.

Saliva is primarily composed of water, mucus, proteins, glycoproteins, mineral salts, andamylases. The composition of the saliva depends on the rate at which different cell types con-tribute to the final secretion: mucus secretion (due to the glycoprotein and mucin) and waterysecretion (containing salivary amylase). The major ions are sodium, potassium, chloride, andbicarbonates (Weatherell et al., 1994). In the ducts of the salivary glands, sodium and chlo-ride are reabsorbed but potassium and bicarbonates are secreted, thus, the electrolyte balanceis altered depending on the rate of salivary flow. Other salivary enzymes include ptyalin,lingual amylases and so on (Chauncey et al., 1957; Lindqvist and Augustinsson, 1975;Tan, 1976).

In order to be absorbed orally, the active must first dissolve in the saliva. Extremelyhydrophobic materials do not dissolve well and are likely to be swallowed intact unless aspecialized delivery system is used to present them to the mucosa. Saliva containing dis-solved actives is constantly being swallowed, thus competing with buccal absorption.

176 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Epithelium

Lamina

Submucosa

Figure 8.3. Structure of the oral mucosa (Harris and Robinson, 1992 with permission).

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 176

Page 183: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Keratinization

Barrier function of the surface layers of the buccal epithelium depends on the intercellularlipid composition. Epithelia that contain polar lipids notably cholesterol sulfate and gluco-syl ceramides are considerably more permeable to water than keratinized epithelia. Intra-cellular lamellae composed of chemically nonreactive lipids have been identified inthe human buccal mucosa and may be relevant to drug permeability. There are intercellularbarriers in the superficial layers of both keratinized and nonkeratinized oral epitheliathat can limit the penetration of substances traversing the tissue by this route, espe-cially polar molecules and electrolytes. Substances with a preferential solubility aremore likely to pass along membranes and these may be limited by the formation of a ker-atin layer.

Membrane Coating Granules

Membrane coating granules (MCGs) are spherical or oval organelles of about 100–300 nmin diameter found in many stratified epithelia and are believed to form major permeabilitybarriers. MCGs appear to play a major filtration barrier role in the kidneys (Kanwar et al.,1980) by delaying or preventing the movement of large molecules such as proteins. Theyhave also been found in both keratinized (gingivae) and nonkeratinized (buccal) epithelia(Hayward, 1979).

These granules contain glycoproteins, formed by covalent linkage between glyco-saminoglycans, mucopolysaccharide, and proteins. The glycosaminoglycans are high-molecular weight linear molecules with complex sequence. MCGs are also negativelycharged molecules (abundant in sulfate and carboxyl groups). The glycosaminoglycanmolecule occupies a much larger volume than other molecules with comparable size. Thesecharacteristics make the glycosaminoglycan molecule an effective diffusional barrier inparticular against electrolytes and water in extracellular fluids.

Polarity

Permeability routes across the oral mucosa can be classified into nonpolar and polar: (i) thenon-polar route involves lipid elements of the mucosa, which partition the activeinto the lipid bilayer of the plasma membrane or into the lipid of the intercellular matrix;and (ii) the polar route involves passage of hydrophilic materials through aqueous poresin the plasma membranes of individual epithelial cells or ionic channels in the intercel-lular spaces of the epithelium. Whether a given nonelectrolyte will pass rapidly acrossthe oral mucosa is determined by its partitioning between lipid and aqueous phases(Schanker, 1964). Substances with high lipid solubility will be transported across the lipid-rich plasma membranes of the epithelial cells while water-soluble substances will passthrough the intercellular spaces.

An alternative classification involves passage through intercellular spaces between cells(i.e. the paracellular route) or transport into and across the cells (i.e. the transcellular route).The latter involves partitioning, cellular channel diffusion, and carrier-mediated transport(Blanchette et al., 2004). The paracellular route represents diffusive convective transportoccurring through the intercellular space (Figure 8.4).

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 177

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 177

Page 184: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

pH

The buccal epithelium has an isoelectric point (pH at which potential is zero) of 2.6.At neutral pH, the buccal epithelial membrane is negatively charged, relatively imperme-able to anions and therefore functions as an ion-exchange surface for cations. At acidic pHvalues (i.e. below the isoelectric point), the membrane carries a net positive charge andbecomes relatively impermeable to cations and functions as an anion exchanger.

Although diffusion potential experiments have shown a higher relative permeability ofpotassium cation (K�) over the chloride anion (Cl�), information on the absolute permeabil-ities of these ions is lacking (Kaber, 1974; Lesch et al., 1989). As ionic strength increases,resistance decreases due to increased electrostatic shielding and therefore lower electrostaticpotential barrier to permeation of ions and a reduction in membrane resistance.

Transport Mechanisms across Membranes

Drugs and active components, except when given intravenously, must be transported acrossseveral biological barriers before reaching general circulation. Four transport mechanismsare known, namely: simple (passive) diffusion, facilitated diffusion, active transport, andpinocytosis. It is generally believed that most substances passing across the oral mucosamove by simple Fickian diffusion (Siegel et al., 1971). Only qualitative evidence of facili-tated diffusion for small substances has been reported (Siegel, 1984). The oral mucosaemploys an active uptake mechanism for a very few number of small molecules such asmonosaccharides (Manning and Evered, 1976). In buccal epithelia, passive diffusion is,likely, the most frequent mechanism.

1. Passive diffusion is the transport across the cell membrane wherein the driving force forthe movement is the concentration gradient of the solute. In orally administered actives,this absorption occurs in the small intestines.

2. Facilitated diffusion can best be described by the movement of molecules from a higherconcentration to a lower one as a result of their random motion. Depending on the physi-cal or chemical properties of the active, diffusion across biological membranes can takeplace through a lipid phase or along aqueous channels. In either situation, provided that

178 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 8.4. The four mechanisms of transport across a cell monolayer (Blanchette et al., 2004with permission).

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 178

Page 185: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

an adequate concentration of the active is applied to one side of a membrane and there issufficiently rapid removal of it from the other side, then a steady state is reached in whichthe rate of diffusion is directly proportional to the concentration of the active (this isknown as Fick’s law).

3. Active transport involves the movement of molecules against a concentration gradient orof ions against an electrochemical gradient and requires the expenditure of metabolicenergy. Some sugars and amino acids are transported across intestinal epithelia but areunlikely to take place across skin or oral mucosa (Kaaber, 1973).

4. Endocytosis is a process by which a large number of different cell types are capable oftaking up solid particles (phagocytosis) or fluids (pinocytosis) from their external envi-ronment by engulfing the material in membranous vesicles. While cells of the oral epithe-lium are capable of taking up material by endocytosis, particularly in the basal andprickle layers, it does not seem a likely transport mechanism across an entire stratifiedepithelium (Berridge and Oschman, 1972).

Effect of Dosage Position in the Mouth

Within the oral cavity, delivery of drugs can be classified into four categories:

1. Sublingual delivery, in which the dosage form is placed on the floor of the mouth underthe tongue.

2. Buccal delivery, in which the formulation is positioned against the mucus membraneslining the cheeks.

3. Local oropharyngeal delivery, where the delivery vehicle is positioned to treat the mouthand throat.

4. Periodontal delivery to treat below the gum margin.

It has been suggested that drug absorption through the sublingual mucosa is more effectivethan through the buccal mucosa, even though both these regions are nonkeratinized. Thesublingual epithelium is, however, thinner and immersed in saliva, both of which aid absorp-tion (Altman et al., 1960).

When fluoride tablets were placed in the lower mandibular sulcus, fluoride concentra-tions were found to increase significantly in the region of the tablet, but there was no appre-ciable increase in salivary levels. In addition, relatively small amounts of fluoride hadmigrated to the opposite side of the mouth suggesting that the lower mandibular sulci arequite isolated from the remainder of the mouth. However, when the tablet was placed in theupper sulcus, the fluoride migrated some distance from the site of administration (Weath-erell et al., 1984). Glucose was also found to behave in a similar fashion (Weatherell et al.,1989). It may be concluded that the site-specific differences are due to saliva movement anddilution of the test substance rather than the nature of the substance. Thickness of the sali-vary film will vary from place to place depending upon the proximity to the ducts of themajor and minor salivary glands, separation of mucosal layers during speaking and mouthbreathing.

Weatherell et al. (1989) reported that glucose retention in the oral cavity was least underthe tongue presumably due to (i) dilution and flushing by saliva, (ii) mechanical actionof the tongue, and (iii) tendency for some glucose to disappear by absorption through thesublingual mucosa or in certain other areas by metabolism in the plaque. Despite the

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 179

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 179

Page 186: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

widespread use of the buccal absorption test, which expresses results as average “buccalpermeability” through the whole oral mucosa, recent efforts have focused on differentiat-ing permeability between structurally different regions of the oral mucosa (Beckett andMoffat, 1969; Tucker, 1988).

Advantages of the Oral Route for Drug Delivery

Most research has focused on the absorption and bioavailability of actives in the GITepithelial area of the stomach and lower intestines. Orally administered actives, however,and their subsequent transport across the oral cavity are less understood. Transport ofactives across the oral route, nevertheless, has many advantages including:

1. Rapid action or onset of actives: The oral cavity is very rich in blood vessels (Table 8.2).Blood supply from the buccal mucosa, unlike the rest of the gastrointestinal tract, doesnot drain into the hepatic portal vein, since these peripheral areas are not specialized fornutrients absorption. Buccal dose forms have often been found to show the samebioavailability as intravenous formulations, without the need for aseptic preparations.

2. Bioavailability: Absorption of drugs via the oral route can avoid first-pass organs such asthe intestine, liver, and lung (Pang, 2003).

3. Actives can be incorporated into consumer-friendly formats (confectionery products),which may help in masking the taste of some objectionable actives.

Disadvantages of Oral Route Delivery

Despite the role of oral mucosa in transporting nutrients and actives, several structural prob-lems hinder the delivery of active substances across the oral mucosa.

1. Compared to the intestinal lining, the oral cavity occupies a very small surface area(2–5 cm2).

2. Buccal cavity, like the entire alimentary canal, is a lipoidal barrier to the passage ofactive substances. Active transport, pinocytosis, and passage through aqueous pores playonly insignificant roles in moving actives across the oral mucosa; hence the majority ofabsorption is passive and only lipophilic molecules are well absorbed. Polar actives, thatis, those ionized at the pH of the mouth (6.2–7.4), are poorly absorbed.

3. Little intercellular absorption is possible across the cuboid squamous epithelium of theoral cavity. However, some amino acids such as glutamic acid and lysine and some

180 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Region of oral mucosa Blood flow (ml/min/100 cc)

Buccal tissue 2.4Sublingual floor of mouth 0.97Sublingual ventral tongue 1.17Gingival tissue 1.47Palatal tissue 0.89

Table 8.2. Blood flow (ml/min/100 cc) in various regions of theoral mucosa of the rhesus monkey (Veillard et al., 1987 withpermission)

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 180

Page 187: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

vitamins such as L-ascorbic acid, nicotinic acid, and thiamine are transported via a carrier-mediated process.

4. Dose form must be kept in place while absorption is occurring since excessive salivaryflow may wash the substances away.

Dosage Formulation: Physicochemical Properties of the Active and Dosage

A further issue affecting the absorption of orally administered actives is their physicochem-ical properties. Most of these actives are presented in the form of tablets or capsules so inorder to be absorbed, the carriers have to be disintegrated or dissolved. A variety of factorsaffect the dissolution rate and therefore the availability of the actives for absorption. Prod-uct characteristics include format (tablet, lozenge, chewing gum, edible strips, capsule),particle-size distribution of the active, dosage porosity, and presence/absence of coatings.

Chewing Gums

Compared to other confectionery formats, chewing gums provide the most hospitable envi-ronment for encapsulated and unencapsulated ingredients due to the mild preparationconditions, mainly the absence of heat stress or excess moisture. In addition, the physico-chemical properties of gum base can be used effectively to delay/sustain the release ofactives. There is no monograph about chewing gum in any pharmacopoeia, but it is describedin guidelines for pharmaceutical dosage forms issued by the Commission of the EuropeanCommunities (1991) as a “solid preparation with a basis consisting of gum which should bechewed and not swallowed, providing a slow steady release of the medicine contained.”

Controlling the release of flavors and active ingredients from chewing gums can best beaccomplished by a thorough understanding of the complex chemistry of gum bases andtheir binding affinity to those ingredients.

Typical Chewing Gum Composition and Manufacturing

Chewing Gum Composition

Chewing gum preparations involve gum base and nonbase components (flavor, sweeteners,color, etc.). Optional ingredients include vitamins, cooling and warming agents, menthol,and other active ingredients. A typical chewing gum formulation is shown in Table 8.3.

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 181

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Table 8.3. Typical composition of chewing gum formulation

Component Sugared Sugar-free

Gum base 18% 22–30%Glucose syrup 45° 19% —Mannitol 0–5%Powdered sugar (to make 100%) —Liquid sorbitol 70% 15–22%Flavor 1.0% 1–1.5%Color 0.1% 0.1%Salvage 5% 5%Glycerin 2% 1–6%

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 181

Page 188: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Gum base formulations are usually held as trade secrets by confectionery manufacturers;due to the competitive nature of the chewing gum business, most chewing gum companieshave established their own gum base manufacturing factories. Several categories of gumbases can be distinguished, depending on the ultimate application. Gum bases are primarilymade up of hydrophilic and hydrophobic polymers (styrene-butadiene elastomers, polyviny-lacetates (PVA), waxes, elastomer plasticizers, waxes, fats, oils, softeners, emulsifiers,fillers, texturizers (talc, calcium carbonate), hydrogenated soybean oil, sugar, glycerin, fla-vors, color, antioxidants, and other minor ingredients. Gum bases are notorious for theiraffinity to most flavors, thus complicating their release from the chewing gum matrix.

A new category of ingredients referred to as sensates has recently been introducedinto chewing gums and other confectionery products to provide unique trigeminal sensa-tion of cooling, warming, tingle, etc. When combined with flavors, cooling agents such asN-ethyl-p-menthane-3-carboxamide, N,2,3-trimethyl-2-isopropyl-butanamide, menthylglutarate, menthyl lactate, isopulegol, menthone glyceryl ketal, and others have been foundto enhance the pleasant perception of flavors and breath freshening (Johnson et al., 2004;Wolf et al., 2005). Similarly, warming/heating agents such as capsicum oleoresin, cinnamicaldehyde, pepper oleoresin, gingerol, shoagol, etc. are often used to provide unique warm-ing sensation in the mouth and the trigeminal area.

Chewing Gum Manufacture

Gum base is softened or melted (50–70°C) and placed in a kettle/mixer fitted with z-shapedblades for 10–30 minutes. Powdered sweeteners, syrups, active ingredients are added fol-lowing accurate time schedule. Late in the mixing procedure, flavors and cooling/warmingagents are then added and the mixture is cooled to 35–45°C, rolled onto plates, scored intostrips, and cut into pieces to produce sticks or tablets. Recently, extruders have been intro-duced for manufacturing chewing gums due to the efficiency, process flexibility, and cost-effectiveness of such units.

Coating is an essential step in finishing pellet gums and where flavors, colors,and actives can be added. Sugar syrup, gum arabic, starches, and other binders are appliedto the surface of gum pellets placed in rotating basket-type mixing/coating units. Tum-bling continues until sufficient amounts of coating material are applied followed by gentlepolishing to provide smooth surface free of imperfections. A variety of ingredientssuch as waxes, shellac, talc, and emulsifiers can be used in chewing gum polishingapplications.

Coated chewing gums are hardened in the 8 weeks following preparation; the coatingsugars and polyols (sorbitol and xylitol) develop crystals that provide hardness andcrunchy texture. Crystallization, however, creates a porous coating structure with multiplemicrochannels. The latter can allow migration of moisture and oxygen, thus exposing labileactives to possible losses or degradation reactions.

Chewing Gums for Delivering Flavors and Nonmedicated Actives

Significant advances in delivering unique flavors and sensations via chewing gums havebeen documented in the patent literature. Most of these patents have been filed in the lastdecade by gum manufacturers and flavor houses who have been keen on adapting their deliv-ery systems technologies to this highly profitable sector of the confectionery business.

182 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 182

Page 189: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Loss of flavor, either due to degradation from the product or due to its tight binding tothe gum base, remains the most challenging concern in chewing gum manufacture. Severalapproaches have surfaced recently for addressing this problem either directly by (i) acceler-ating flavor release (burst effect) or indirectly (ii) by enhancing flavor perception viaextending the release of physiological cooling agents or sweeteners throughout the prod-uct’s normal chewing time. Encapsulated flavors prepared by coacervation, particle coat-ing, entrapping into liposomes or amorphous matrices can be simply applied to the outercoating of chewing gums. For burst-release effect, it is advisable to entrap flavor compo-nents into a water-soluble matrix so that flavor release can take place instantly as the prod-uct is placed in the mouth (Clark and Shen, 2004). The following citations represent agroup of controlled-release applications of chewing gums designed for local delivery offlavors, sweeteners, breath-fresheners, etc.

A two-step process for controlling the release of flavors from a chewing gum systemwas devised by Merritt et al. (1985), where the flavors were incorporated into an emulsionvia a hydrophilic matrix. The latter is further dried and ground to appropriate particle sizefollowed by coating with a water-impermeable substance (matrix-reservoir combinationsystems). Song and Courtwright (1992) patented a method for manufacturing sustainedflavor releasing structures. The inventive process comprised of blending the flavors andbinding material such as amorphous silicon dioxide hydrates and further coating the com-positions with a barrier material such as polyvinylpyrrolidone (PVP). The amount of flavorreleased was claimed to be about 20% during 20 min. of chewing compared to 35% in aconventional chewing gum. Sustained flavor release was claimed for a process whereby theflavor is partitioned into a water-soluble phase of the gum for immediate release, while thedelayed effect was provided by the other flavor portion embedded into the water-insolublefraction such as polyethylene or polypropylene (Rutherford et al., 1992). Song and Copper(1992) disclosed an innovative approach to controlled release via a fiber structure usingmelt-spinning technique, which was followed by stretching via applying a draw or astretching force. The flavor droplets exposed along the sides of the fiber can be released asthe solvent infuses into the fiber creating channel-like structures. The length of these chan-nels gradually increases as the active agent directly in contact with the solvent is dissolved.The fiber structures can be incorporated directly into a chewing gum where the pressuregenerated from chewing will flatten, stretch, and deform the fibers exposing new surfaceareas of active to the solvent.

Wolf et al. (2005) patented a composition for extending the perception of breath-freshening in a chewing gum by encapsulating physiological cooling compounds into thegum matrix. Monitoring breath-freshening using trained sensory panel showed a signifi-cant increase in perceived breath freshening intensity compared to unencapsulated control(Figure 8.5).

McGrew et al. (2006) formulated a chewing gum containing metal salt, which isclaimed to reduce/eliminate oral malodors associated with bad breath. The controlledrelease of Zn lactate and Cu gluconate claimed to provide breath-freshening benefits bybinding to volatile sulfur compounds generated in the GIT that are commonly associatedwith bad breath.

Manufacturing good-tasting sugar-free chewing gums can be a challenging task becauseof several factors, for instance incompatibility of artificial sweeteners with other compo-nents of the chewing gum system. Aspartame (APM) is a methyl ester of L-aspartyl-L-phenylalanine dipeptide molecule, which exhibits about 180 times the sweetening ability of

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 183

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 183

Page 190: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

sugar on an equal weight basis. APM can be destabilized and its sweetening power can besignificantly reduced in the presence of aldehyde-based flavors such as cinnamon resultingin chewing gums with unacceptable taste, color, and texture. Similarly, APM can degradequickly in chewing gums containing sodium pyrophosphate. The latter is added to chewinggum systems to provide teeth remineralization benefits.

Bunczek and Urnezis (1993) patented a method for stabilizing APM in cinnamon-flavored chewing gums by mixing an aqueous solution of APM with hydrochloric acid anda thickener (gelatin) followed by air-drying and grinding and further incorporation into achewing gum formula (Figure 8.6). Stability of acid-treated APM with or without gelatincoating was found to be superior to the native untreated sweetener. Acesulfame-K (Ace-K)is another sweetener that can be encapsulated to extend its release throughout chewing.Broderick and Record (1992) developed water-insoluble porous beads that comprised acopolymer of divinylbenzene and styrene impregnated with Ace-K. The beads were furthercoated with hydroxypropylmethylcellulsoe (HPMC) prior to incorporation into a chewing

184 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

0 5 10 15 20

Time (min.)

Per

ceiv

ed b

reat

h fre

shne

ss(in

tens

ity)

Control

Encapsulatedcooling agents

012345678910

Figure 8.5. Perceived breath freshening intensity of chewing gum containing encapsulatedcooling agents and control (reproduced from Wolf et al., 2005).

Figure 8.6. Effect of acids and gelatin on aspartame (APM) retention (reproduced from Bunzeckand Urnezis, 1993).

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 184

Page 191: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

gum system. Release of Ace-K and spearmint was found to be about 40% after 5 min. ofchewing compared to 80% from control.

Other interesting controlled release systems have been documented in the patent litera-ture such as those proposed by Johnson and Yatka (2000), Ream et al. (2003), Savage et al.(2002), Sharma and Yang (1986), Song et al. (1992) and many others.

Chewing Gum for Delivering Caffeine

Caffeine is a well-known stimulant, which is used to alleviate the effects of sleep depriva-tion and combat headache and fatigue. The caffeine molecule is completely metabolized bythe liver, its rate of inactivation is unaffected by the delivery to the liver and can only bemodified by a change in hepatic enzyme activity. Incorporating caffeine into beverages andother food products has been very challenging due to many constraints—mainly its aque-ous insolubility (2.1%), objectionable bitterness as well as delayed stimulant activity.

Syed et al. (2005) studied the pharmacokinetics of three doses (50, 100 and 200 mg) ofcaffeine delivered via Stay Alert® chewing gum and proposed a dose-proportionate linearincrease in plasma caffeine levels. Their study showed that delivering caffeine via chewinggums is an effective and convenient means of maintaining desirable levels of alertness andperformance in sleep-deprived individuals. The same chewing gum (Stay Alert®) was alsoused earlier by another research group (Kamimori et al., 2002) to compare the bioavailabil-ity of caffeine delivered via a chewing gum and a capsule. Mean plasma Tmax for individu-als who chewed the gum was found to be in the range 44.2–80.4 min. compared to84.0–120.0 min. for the capsule group, indicating an early onset of pharmacological effectand a faster rate of absorption of the caffeine molecule via the buccal mucosa.

Enhancing buccal absorption of caffeine was attempted both in vivo and in vitro.Donbrow and Freidman (1974) investigated the release properties of caffeine using a diffu-sion cell and concluded that diffusion of caffeine via an ethyl cellulose cell was time-dependent, that is, the diffusion follows a zero-order mechanism. Increasing membranehydrophilicity by incorporating 40% polyethylene glycol (PEG) demonstrated the possibil-ity of enhancing permeability of the caffeine molecule (Table 8.4). An in vitro study byNicolazzo et al. (2003) showed that pretreating porcine buccal mucosa with different levelsof sodium dodecyl sulfate (SDS) (0.05, 0.1, and 1%) significantly enhanced caffeine fluxby a factor of 1.57, 1.63, and 1.81% respectively.

Gudas et al. (2000) developed a chewing gum containing slow-releasing caffeine profileby pre-encapsulating the active (50–100 mg caffeine) into a water-soluble matrix. Con-trolled release of small amounts of caffeine over a longer period of time was designed toreduce the impact on taste. Results from the corresponding clinical trials conducted using6 subjects showed enhanced absorption rate constant (Ka) when caffeine was administeredthrough the chewing gum due to high buccal absorption rate and subsequent fast deliveryinto the systemic circulation. A similar change in the onset of dynamic response wasnoted, for example alertness and performance when caffeine was incorporated at 50–500milligrams levels. Plasma caffeine concentration was also found to be significantly greaterfor gum than caffeinated cola or other beverages within the first 10–30 min. after caffeineintake, that is, faster uptake by the body.

Ream et al. (2001) formulated a chewing gum for delivering caffeine by layering theactive onto the outer shell of the pellet coating. Their study demonstrated significant levelsof buccal absorption presumably due to pressure development in the buccal cavity, a result

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 185

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 185

Page 192: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

of continued chewing, which may have forced the permeation of the released caffeinemolecules through the mucosa.

Chewing Gums for Delivering Vitamins

The release of ascorbic acid from chewing gum formulations was investigated by severalgroups. Ascorbic acid mixed with hydrophobic components has been shown to exhibit aslower but complete release of the drug within 15 min., compared to mixing with hydrophilicones (Odumusu and Wilson, 1977; Sadoogh-Abasian and Evered, 1979). A slightly fasterrelease of L-(�) ascorbic acid was observed in vivo compared to its in vitro release in a masti-cation machine; however, a good correlation was observed between the in vivo and in vitrorelease patterns within the first 5 min. of mastication. Andersen (2004) incorporated vitamin Cinto a chewing gum formulation at two different gum base levels (30 and 45%) and showed avery high level of vitamin recovery especially in the 30% gum base formulation.

Sadoogh-Abasian and Evered (1979) and Stevenson (1974) independently studied thetransport of ascorbic acid across the human mucosal membranes and showed that its absorp-tion is Na ion-dependent. Calcium ions were also found to increase ascorbic acid absorptionpresumably due to a secondary effect of Na ion fluxes. Buccal mucosa was found to bepermeable to dehydroascorbic acid and D-isoascorbic acid. The presence of D-glucose and3-O-methyl-D-glucose increased the absorption of ascorbic acid but D-fructose had littleeffect and D-mannitol had no effect.

The impact of ascorbic acid solubility and its ionization behavior on the vitamin buccalpermeability were also studied by changing pH of the medium stepwise from 3.4 to 9.0.Increasing pH resulted in a gradual decrease in buccal absorption of the vitamin. Vitamin C isonly 13.7% ionized at pH 3.4 but almost fully ionized at pH 9.0, thus indicating passive diffu-sion of the molecule (Odumusu and Wilson, 1977). The process was reported to be not stere-ospecific since both the natural form L-ascorbic acid and the unnatural D-isomer weretransferred across buccal mucosa at similar rates. Glucose was also found to enhance transfer

186 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Table 8.4. Transfer rate of caffeine as a function of film thickness, caffeine concentration andPEG concentration in film (Donbrow and Freidman, 1974 with permission)

Rate of transfer reciprocal Rate of transfer/film thicknessFilm thickness (cm � 104) (mol s�1 � 109) (mol cm s�1 � 107)

26.8 400 1.0732.7 3.32 1.0848.0 2.20 1.06Concentration of caffeine Rate of transfer/caffeine

(mol � 108 l�1) concentration (l s�1 � 107)1.03 0.52 0.5052.06 1.06 0.5094.12 2.10 0.509Concentration of PEG Rate of transfer/PEG concen-

in film (% w/w) tration (mol s�1 � 109)0 0.123 —10 1.15 0.11520 2.06 0.10340 4.45 0.11150 5.40 0.108

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 186

Page 193: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

of ascorbic acid across buccal membranes, though the effect may be due to glucose actingas an energy metabolite (Stevenson, 1974).

Nicotinic acid and nicotinamide displayed similar buccal absorption rates. The latterwere also consistent with those determined across intestinal mucosa (Evered et al., 1980).Despite the differences in their ionization behavior at pH 6.0 (nicotinic acid is 6% union-ized while nicotinamide is almost fully ionized), both forms of the vitamin are soluble inwater and poorly soluble in lipids. Isonicotinic acid and nicotinic acid have identicalmolecular weights and very similar pKa values (4.84 and 4.77, respectively), yet the formershowed a lower rate of absorption suggesting a carrier-mediated transport system (i.e. facil-itated diffusion). These studies revealed for the first time that human buccal mucosa ispermeable to the water-soluble vitamin, thiamine, as with ascorbic acid (Sadoogh-Abasianand Evered, 1979), nicotinic acid and nicotinamide (Evered et al., 1980) showing somesimilarities to absorption from mammalian small intestine (Evered et al., 1980).

Chewing Gums for Delivering Antimicrobial Agents

Epigallocatechin gallate (ECGC) has been studied for its potential benefits in reducing therisk of heart disease and cancer as well as weight loss. However, recent information fromthe Tearrow Co. suggests that tea extracts of Camellia sinesis incorporated into chewinggum can permeate the mucosal barriers to help in treating gingivitis and eliminatingmicrobial growth in the oral area (Gelski, 2006).

Miconazole is a well-known oral care antimicrobial active. Attempts to incorporatemiconazole directly into chewing gums were not very successful due to its strong binding tothe gum base. To promote its release, different solid dispersions of miconazole in PEG 6000,PVP 40,000 and xylitol were tested. Dissolution rate data showed that dispersions ofmiconazole:PEG 6000 (1:4) had the highest level of release from a chewing gum (15-timescompared to pure miconazole) due to enhanced aqueous solubility of the active. Addition oflecithin to the miconazole–PEG chewing gum formulation was found to enhance the releaserate as well as the time of release both in vitro and in vivo. Lecithin may have improvedmiconazole solubility by virtue of its ability to form liposomes in aqueous media (Pedersenand Rassig, 1990). In vitro data using a mastication device of Christup and Møller (1986)correlated well with the in vivo data derived from six healthy volunteers. Release of micona-zole was also shown to be significantly facilitated by the addition of Panodan 165 (acidicsurfactant) to a chewing gum formulation. High surface activity of Panodan 165 as well asits low pH may have increased the solubility of miconazole and/or enhanced saliva absorp-tion into the chewing gum during mastication.

Using a panel of five subjects, Witzel et al. (1980) investigated the in vivo release of nys-tatin, a slightly soluble antifungal agent from chewing gum. Coating the antifungal agentwith gum arabic resulted in 24% release of the nystatin compared to only 4% from uncoatednystatin. Despite the enhanced solubility of nystatin using enhancers such as CremophorRH40, Tween 60 (nonionic surfactants) and Panodan AB 90, in vitro release rate was too fastto provide any significant antifungal effect (up to 99% in the first 10 min. of administration).Lombardy et al. (2001) disclosed an oral hygiene plaque-disrupting chewing gum compris-ing a core containing encapsulated sodium bicarbonate, which is surrounded by a coatingthat contains an encapsulated edible acid. Upon chewing and subsequent effervescencedevelopment, the formed foam penetrates between the teeth and gum crevices to loosenplaque build up.

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 187

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 187

Page 194: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Chewing Gums as Delivery Systems for Oral Health

Tooth decay is mainly caused by Streptococcus mutans metabolizing fermentable carbo-hydrates leading to a drop in pH in the tooth and plaque microenvironment and to gradualdissolution of the hydroxyapatite [calcium phosphate hydroxide, Ca10(PO4)6(OH)2], the pri-mary component of tooth enamel. Natural remineralization process involves, in part, theflow of saliva (saturated with calcium and phosphate) that raises the pH so that the calciumand phosphate ions can precipitate to replace the dissolved hydroxyapatite. However, thisprocess is very inefficient in most individuals.

Sugar-free chewing gums have been promoted for their effectiveness in preventing dentalcaries. Based on clinical trials, it was suggested that mastication of xylitol-based chewinggum may reduce dental caries in children and young adults better than any other sugar-freechewing gum. This improvement has been associated with reduced levels of Streptococcusmutans and Lactobacilli in saliva along with a reduction in plaque build up at neutral pH(Assev and Rølla, 1986; Wennerholm and Emilson, 1989). However, S. mutans may developresistance to xylitol after few months of chewing.

The effect of chewing nonmedicated chewing gums on plaque pH, saliva flow rates andthe incidence of dental caries have been the topic of many studies. Different brands of sugar-free chewing gums claim to stimulate saliva flow rate compared to that of unstimulatedsaliva flow rate. Peak salivary flow rates are known to develop within the first minuteof chewing.

Leach et al. (1989) presented evidence for the remineralization of artificial caries-likelesions in human teeth enamel in situ following mastication of a sorbitol-containing chewinggum. Winston and Usen (2002) patented a confectionery composition containing solublephosphate and calcium salts that are claimed to help remineralize teeth surface lesions andexposed dentin tubules. The composition was described to be applicable to hard candies,chewing gums, lozenges as well as other formats.

Patients with xerostomia (dry mouth) most often show elevated risk of caries. Sugar-freechewing gums have been recommended to patients who still have some capacity to secretesaliva (Jenkins and Edgar, 1989). The saliva produced during mastication can alleviate therisk of caries by reducing plaque pH generally seen in response to a sucrose challenge. To fur-ther increase caries prophylactic activity of chewing gum, addition of an acid-neutralizingagent (e.g. carbamide) may be appropriate.

Chewing Gum for Delivering Acetylsalicylic Acid

The high clearance drug salicylamide has been used as a model substance in chewing gumexperiments (Christup et al., 1988b). The bioavailability of acetylsalicylic acid from Asper-gum® has been compared to the bioavailability of acetylsalicylic acid from pre-oral tablets.The rate of absorption was shown to be faster from chewing gum than from the tablets and itwas concluded that chewing gum might provide a faster relief of pain (Woodford and Lesko,1981). These results were speculated to be due to a reduction in drug metabolism in the GITand the liver.

Christup and co-workers (1990, 1988) studied the in vitro and in vivo release of differentsalicylamide chewing gum formulations and showed higher release from the formulationcontaining less gum base. Micronized salicylamide in a chewing gum showed greaterrelease from hydrophilic formulations compared to their hydrophobic counterparts when the

188 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 188

Page 195: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

gum was chewed for 30 min. (Christup et al., 1988). The release of coarse salicylamideparticles from a chewing gum composition was reported to be limited, but was doubledwhen the active was micronized.

These studies suggest that drug release from chewing gum can be modified through for-mulation or manufacturing processes either by the addition of substances to the gum basewhich exhibit different lipophilic or hydrophilic characteristics or by modifying the physicalcharacteristics of the incorporated drug.

Comparison of Delivery Profiles between Chewing Gum and Lozenge

Christup et al. (1990) used gamma-scintigraphy to examine how effectively drugs werereleased in vivo from chewing gum, their distribution profile once released and the length oftime drug remained in the oral cavity following release. Release profile from chewing gumwas compared to those observed following the administration of lozenges and sublingualtablets. Vitamin C was found to be absorbed better from a chewing gum than from a tablet(Christup et al., 1988)

Non-absorbable, water-soluble compound Tc E-HIDA (N-(N�-(2,6-dimethylphenyl)) car-bamoylmethyl iminoacetic acid) used as a model active showed complete release after10 min. of chewing at a rate of 1 chew/sec. Activity (counts) vs. time (min) profiles of the oralcavity and the stomach of six subjects following administration of the 3 different dosageforms (chewing gum, lozenge and sublingual tablet), did not show any difference in thedistribution of Tc-HIDA within the oral cavity, glottis or upper oesophagus. However,Tc E-HIDA released from sublingual tablets remained for the longest time while Tc E-HIDAreleased from lozenges remained for the shortest period in the oral cavity (Rassing, 1994).

Lozenges (Hard Boiled Confections)

Lozenges have long been used as vehicles for delivering medicaments to alleviate coldsymptoms such as decongestion, to soothe sore throats and clear nasal passages. Suchmedicaments include analgesics, antitussives, expectorants, cooling, warming, numbingand tingle agents.

Lozenges are essentially hard-boiled candies that can be formulated in sugared andsugar-free versions. Lozenge manufacture involves heating a glucose/sucrose mixture toevaporate water and transform the matrix from a crystalline to an amorphous glassy phase.Lozenges can be manufactured using batch or continuous processes. Essential additivesinclude acids, flavors and colors while therapeutic additives include actives such as menthol,benzocaine, dextromethorphan, pectin, vitamins or other nutrients. Lozenge manufactureexposes these actives to harsh moisture and heat environments and often results in partial ortotal degradation of heat-labile components.

Lozenges for Delivering Flavors and Sensates

Release of actives from a lozenge is activated by sucking and gradual dissolution of thesugar (or sugarless) matrix. Menthol released from lozenges, for example, can bind tothermo-receptors located within the free nerve endings of the trigeminal and nasal cavities.The resulting cooling sensations are presumed to provide analgesic effects via modulatingthe sensitivity of cutaneous pain fibers.

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 189

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 189

Page 196: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Due to their water-based nature, lozenges can be most effective in providing “burstrelease” of flavors, sensates or other actives. Their hydrophilicity makes them ideal perme-ability enhancers for hydrophobic actives (e.g. cooling compounds) across membranes ofthe oral area. Incorporating encapsulated particulates into lozenge formulations resultsmost often in premature destruction of the capsule and the release of entrapped sub-stances during processing and before consumption. Micelles and emulsion-based carriersystems are, generally speaking, better suited for lozenge formulations.

An effective “burst” release approach was developed by Clark and Shen (2004) wherebyencapsulated particulates or powders were layered onto the lozenge outer surface toprovide quick dissolving matrix to liberate the entrapped flavor components. Rivier (2005)patented a lozenge-based delivery system to provide “burst” release of a solid active nes-tled into the center of an oval-shaped lozenge. Due to the inherently thin walls at the ends ofthe larger diameter of the oval piece, “burst release” is activated by sucking the lozengeand creating channels for quick diffusion of the active. A wide range of actives can beincorporated into this lozenge design including flavors and sensates for refreshment.A unique type of actives that can be dosed into the center of the oval piece is polyols.By virtue of their negative heat of dissolution in particular xylitol and eryhthritol,their release and high solubility in the saliva can provide a refreshing moist coolingsensation. Therapeutic actives such as analgesics can be also employed to providequick relief.

Lozenges for Delivering Throat Relief Actives

Despite these advantages, sugar- or polyol-based lozenges do not adequately providelong-lasting solutions to problems unique to the mouth and the esophageal area due to thequick dissolution, short residence time and mode of lozenge consumption, that is, movingaround in the mouth and saliva stimulation that can be secreted and swallowed. A recenttrend in lozenge formulations involves incorporating high molecular weight polymers withmucoadhesive properties into sugared/sugar-free formulations. This practice is supported bythe USP monograph permitting the use of mucoadhesive materials (referred to as demul-cents) for providing relief from mucus irritation, pain and discomfort associated with laryn-gopharyngitis (sore throat) and other upper respiratory tract infections. Examples of thesedemulcents include gelatin, pectin, celluloses, and alginates. Other types of nonpolymer-based mucoadhesive agents include titanium and silicon dioxide (Dobrozsi, 2003) and lipidvesicles (Bealin-Kelley et al., 2002).

Lozenges formulated with demulcents, however, are bland tasting and are oftenperceived by consumers as nonefficacious. A new generation of lozenges has been formul-ated with sensates (cooling or warming compounds) to provide an additional sensorycue of soothing. Coincidentally, it was discovered that combining demulcents with sen-sates can extend the perceived soothing warmth from the mouth to the throat (Figure 8.7),an attribute that is highly desirable by consumers (Bealin-Kelley et al., 2002; Lakkis,2006).

Lozenges as Delivery Systems for Dry Mouth Relief

Lozenges have also been formulated to provide relief from dry mouth symptoms referred toas xerostomia. Wolfson (2002) patented a composition using Heliopsis longipes root for

190 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 190

Page 197: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

increasing salivation, thus alleviating dry mouth feeling while maintaining oral hygiene.Tutuncu et al. (2003) developed a food acid-containing lozenge, which claimed mouthmoistening benefits. Kayane et al. (2003) described a throat care lozenge, which promotesthe secretion of mucin to provide bactericidal effect by inhibiting the adhesion of pathogenicbacteria, Pseudomonas aeruginosa, Haemophilus influenzae, or Staphylococcus aureus.Efficacy of the lozenge was confirmed via ELISA testing, which showed increased levels ofIgA and lysozyme content in the mucus secretion.

Lozenges as Delivery Systems for Teeth Remineralization Actives

Calcium phosphate, the main component of dentin and teeth enamel, while insoluble atneutral saliva pH, is readily soluble in acidic media generated by fermentation of ingestedcarbohydrates. To alleviate the impact of these events and slow down the develop-ment of caries and lesions, it is desirable to increase the available concentration of calciumand phosphate ions in the oral cavity to speed up the remineralization process. Severalinventions and commercially available confectionery products (lozenges and chewinggums) that claim teeth remineralization are commercially available (Chow and Takagi,2001; Kaufmann, 2003; Mazurek et al., 2000; Savage et al., 2002; Winston and Usen,2002).

One drawback with using calcium- and phosphate-containing salts and buffers inhard-boiled lozenges is the development of bitterness and grittiness due to complexationof the calcium and phosphate ions in the candy matrix. One approach to overcome thesedrawbacks is to skillfully incorporate these actives into separate components of thecandy matrix so that the actives can be released concurrently in their soluble forms to formthe hydroxyapatite in the generated saliva (Lakkis and Wong, 2007).

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 191

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

0

1

2

3

4time, min

1

2

3

45

6

7

8

0.1% pectin

0.2% pectin

0.3% pectin

Figure 8.7. Effect of pectin level (0.1, 0.2 and 0.3%) on perceived warming in the human throat.Sensory panel ratings scale ranged from 0 (very low warming intensity) to 4 (highest warmingintensity). Graph reproduced from Lakkis (2006 with permission).

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 191

Page 198: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Bioerodible and Bioadhesive Devices

This category encompasses a wide range of weak and strong adhering structures that arecommercially available in the form of films/strips, patches, tablets or other devices. Weakadhesion is desirable for breath freshening and flavor delivery films while strong adhesion iscritical for medicated applications (e.g., antimicrobials and teeth whitening strips).

Bioadhesive and bioerodible devices represent an ideal delivery system due to manyattributes such as (i) ease of consumption by individuals having difficulties in swallowing,(ii) localization in specified regions of the oral or other GIT sites to improve and enhancethe bioavailability of actives, (iii) optimum contact with the absorbing surface to permitmodification of tissue permeability, (iv) reduced need for drug overage and (v) avoidanceof first-pass metabolism.

Mucoadhesion takes place by establishing an adhesive bond between the device andthe mucus membranes resulting in a reduced total surface energy of the system becausetwo free surfaces are replaced by one (Anlar et al., 1984; Guo, 1994). Polymers withhydrophilic moieties such as carboxyl and hydroxyl groups can bind to the sialic acid andother oligosaccharide residues in the mucosal membranes. The process takes place in threestages: hydration, interpenetration, and mechanical interlocking between mucus and thepolymer. Mucoadhesive strength is affected by various factors such as molecular weight ofthe polymer, its swelling power, size and configuration of the device, time of contact with themucus, and the physiological nature of the membrane. Generally, oral bioadhesive and bio-erodible devices can be designed to adhere to the cheek (buccal area), the floor of the mouth(sublingual tissue), gums surrounding the teeth, and the roof of the mouth (palate tissue).

Efficacy of mucoadhesive and bioerodible devices is well documented in many pharma-ceutical and consumer health applications. Examples include teeth-whitening, accel-erated healing of inflamed or damaged tissues, prolonged and improved coating andprotection of the mouth and esophagus (Barklow et al., 2002; Choi and Kim, 2000), sus-tained release of insulin in the stomach via mucoadhesive microspheres of glizpide, whichis a second-generation sulfonylurea used to acutely lower blood glucose levels (Kahn andShechter, 1991).

Confectionery-based devices are available in the form of medicated and nonmedicatedfilms commonly referred to as edible strips. The latter are usually of the size of a postalstamp, which is placed on the tongue to deliver flavors, breath-fresheners, decongestants,etc. Popularity of these films has soared recently with the introduction of Listerine® pocket-paks™ by Pfizer, Eclipse® Flash by Wrigley’s and most recently Theraflu® cough reliefstrips by Novartis, Inc.

A wide variety of water-soluble and/or -insoluble food-grade hydrogels has been used informulating edible strips. The choice of a suitable composition depends largely on desiredfunctionality and residence time in the oral cavity, its solubility, type of active, and requiredpayload. Typical edible-strips formulations comprise a combination of film-forming poly-mers, fillers, plasticizers, colors, and actives (menthol, flavor, cooling/warming compounds,vitamins, analgesics, etc.). Polymers such as pullulan are favored for their film formingproperties and excellent solubility and clean aftertaste (no residual gumminess). Manufac-turing films with pure pullulans, however, has been hindered by its weak mucoadhesiveproperties and cost. Mucoadhesion of pullulans may be improved by incorporating severaladditives such as PEO, mono- or oligosaccharides to the strip formulation (Ozaki and

192 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 192

Page 199: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Miyake, 1995). Alternative economical film materials have been proposed such as cellu-loses, glucans, native, and modified starches.

Edible strips manufacture involves forming a hot, stable aqueous solution of the filmpolymer and the active(s), casting the solution over a conditioned belt followed by heatdrying and cutting into desired strip dimensions. Such conditions may degrade heat andmoisture labile actives. Encapsulating actives (e.g., food acids) prior to their incorporationinto film formulation may help retard their degradation and help maintain the film integrity(Virgallito and Zhang, 2006).

In vivo assessment of release mechanisms from mucoadhesive devices has been hinderedby physiological variables such as amounts and physicochemical properties of human salivaand movement of the device in the mouth. Most investigations indicate that the release frommucoadhesive devices takes place via erosion, diffusion, or a combination of both. Criticalfactors that can have profound impact on release from bioadhesive/bioerodible devices canbe summarized as follows:

1. Film physicochemical properties, mainly active payload, film forming polymer chem-istry, its thickness and solubility (addition of maltodextrin to pullulan films can enhancetheir dissolution and release of the active).

2. Although high viscosity polymers can improve the bioadhesion of films, at very high vis-cosity, nonhomogeneous distribution of the active may result in unpredictable drugrelease rates (Wong et al., 1999).

3. Location of the device in the mouth as well as tongue movements can play a crucial role inthe device’s residence time and the active’s release rate/mechanism. De Vries et al. (1991)showed that application of buccal patch to the palate provided longer adhesion thanthat of the cheek mucosa. Bouckaert et al. (1993) compared the adhesion of miconazolemucoadhesive tablet to the gingival, palatal, and cheek mucosa and concluded that thelongest adhesion was in the gingival area while the shortest was at the cheeks. Whenapplied to the cheek, the tablet is lodged very near the parotid duct, thus the adhesion timemight be reduced by the salivary flow. Subsequently, the polymer mixture may swell morerapidly and the tablet will become prone to erosion. Despite the fact that less saliva ispresent in the palatal and gingival mucosa compared to the buccal mucosa, adhesion timefor the palate was comparable to that for the cheek and significantly lower than thegingival.

4. Excessive matrix swelling, such as the case with native starch, can hinder the controlledrelease of actives. Tuovinen et al. (2003) using two small model molecules (sotalol,m. wt. 308 and timolol m. wt. 332) and two large model molecules (FITC dextran,m. wt. 4400 and BSA m. wt. 68,000) showed that the small molecules, solatol, and tim-olol were released more rapidly than the FITC dextran and BSA from a native potatostarch matrix (PBS buffer pH 7.4 with or without α-amylase) due to excessive starchswelling. Release of the small molecules was continuous whereas the release of macro-molecules showed discontinuities.

5. Hydrophobic/hydrophilic nature of the hydrogel and active also can have a significantimpact on the release behavior. Tuovinen et al. (2003) showed that the model molecule,sotalol, was released faster than timolol (more hydrophobic) from a starch acetate film(hydrophobic) demonstrating stronger interaction between the matrix and active.Release of timolol and sotalol was faster than the weight loss of the corresponding film

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 193

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 193

Page 200: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

presumably due to the hydrophobic nature of the starch acetate film, thus providingevidence for erosion-controlled mechanism.

Seamless Capsules

Seamless capsules represent a class of delivery systems that can provide powerful impact,but has not yet realized its full potential. Soft seamless capsules are ideal carriers for liq-uids or a suspension of solids in a liquid. A variety of applications have been documentedin the patent literature, including delivery of flavors, menthol, and eucalyptus oil for breath-freshening (Karles et al., 2006; Tanner and Shelley, 1996; Yang, 2005) the pro-vitaminA lycopene (Paetau et al., 1999) and concentrated alcoholic and nonalcoholic beverageconcentrates for recreational use (Hutchinson and Garnett, 1999; Sexton and Lakkis,2003).

Ideal seamless capsules are 4–8 mm diameter with a thin shell wall of 300–600 micronsand maximum core-to-shell ratio of 9:1 w/w. This ratio represents the highest payload of anyencapsulation technology known today. Seamless capsule formulations comprise a liquidcenter and a solid soft shell. The latter can be made of gelatin, agar, alginates, celluloses, orother gelling (moldable) polymers in combination with suitable plasticizers (Figure 8.8).

Seamless capsules are manufactured using specialized machines with coaxial multiplenozzles such as the Spherex system (available from Freund Industrial Co. Japan) as well asother suppliers. The outermost shell layer is formed by extrusion of a hot gelatin solution(60°C) from the outer nozzle and the liquid core (immiscible in water) is extruded from aninner nozzle to form a concentric jet. The jet is further injected into a cooled vegetable oilbath (ca.12°C) to harden the shell. Seamless capsules in the form of spheres are formed dueto surface tension.

One of the critical attributes of seamless capsules is the shell quality, which is expected tobe soft and to dissolve readily in the mouth with no residues. For maximum efficacy andclean aftertaste, release of the active from seamless capsules should take place via breaking

194 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Figure 8.8. Cross-section of a typical seamless capsule showing solid shell and a liquid center.

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 194

Page 201: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

the sphere without any perceivable swelling. Few challenges, however, can complicate themanufacture of seamless capsules and their functional performance, mainly:

1. Shell–core interactions: Due to the hydrophilicity of the shell material, this technologycan only accommodate hydrophobic cores. Hydrophilic substances can interact with theshell material resulting in film plasticization and in some cases can help promote micro-bial growth, a major concern upon processing and shipping of the capsules.

2. Shell thickness: Readily dissolvable capsules require the formation of a thin shell.Shell thickness can affect capsule diameter, that is, for a constant core to shell-mass ratio,shell thickness increases with increased diameter of the capsule.

3. Shell (polymer viscosity): Desired film properties can be achieved by maintaining a deli-cate balance among many parameters, mainly between polymer viscosity and shelldissolution in the mouth. Very low viscosity polymers can lead to capsule deforma-tion and crushing while too high viscosity can lead to the formation of satellites extendedfrom the capsule surface. Wonschik et al. (2005) patented a unique shell formula-tion comprising a mixture of high bloom and hydrolyzed gelatin (zero bloom). The highbloom component is claimed to provide solid network, critical for shell processingwhile the hydrolyzed gelatin occupies spaces in the formed network to provide rapid dis-solution by the saliva.

4. Core Physical properties: For effective processing, liquids, solutions, or suspen-sions should flow by gravity at room temperature. In general liquids with a wide range ofviscosity from 0.2 to >3000 cp at 25°C can be encapsulated. Also, encapsulated liquidsmust have a pH from 2.5 to 7.5 beyond which the gelatin shell would deteriorate.

A new generation of seamless multilayered capsules has been commercialized recently(Jintan Co., Japan) that claims a multitude of functions. Sunohara et al. (2002) patented amultilayered seamless capsule design where the outer shell can provide flavor or breath-freshening and the inner layers can be swallowed to treat stomach-originated bad breath.

Pressed Tablets

Pressed tablets include mouth dissolving, fast-dissolving, rapid-melt, porous, orodis-persible, and melt-in-mouth products. A wide range of tablets and capsules are commonlyused for delivering breath freshening (breath mints) and pharmaceutical actives, althoughthe majority of such tablets are designed to be absorbed in the GIT.

Pressed tablets are prepared by dry blending the active ingredients with water-disintegratable compressible carbohydrate and a binder and then compressing into a convex-shaped tablet. Strength/compactness of these tablets can be detrimental to the extent ofdisintegration, dissolution, and absorption of the active. One of the attractive features ofpressed tablets is the possibility of incorporating high levels of actives compared to otherdosage formats.

Haines (2004) investigated the buccal absorption and bioavailability of vitamin B12 frompressed tablets and a nanofluidized B12 suspension (NF®) via spray applicator as an alterna-tive means to deliver this essential nutrient to patients suffering from intestinal disorders suchas celiacs, who cannot absorb this vitamin from food sources. Results from that study showedthat nanodroplets provided a more effective vehicle for delivering the active molecule acrossthe mucosal barriers at a faster and more even rate than from the tablet or even the non-processed or “normal” vitamin B12 solutions.

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 195

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 195

Page 202: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Effervescent Tablets

Effervescent tablets can be designed to provide enhanced release in the mouth as well asthe lower GIT; Effervescence is the reaction (in water) of acids and alkalis to producecarbon- dioxide. Typical acids used in effervescent tablets are citric, malic, tartaric, adipic,and fumaric while sodium bicarbonate and potassium carbonate are the most commonlyused alkalis.

Effervescence can help promote calcium absorption in the stomach as well as sustainbiological activity of probiotic bacteria (Lee, 2004). It has been speculated that CO2 pro-duced by effervescent reactions induce some alteration (widening) of paracellular path-ways, a primary route of absorption of hydrophilic actives (Anderson, 1992; Nuernbergand Brune, 1989). Absorption of hydrophobic species can also be enhanced due to thenonpolar CO2 gas molecules partitioning into the cell membrane to create a hydrophobicenvironment which allows hydrophobic actives to be absorbed (Eichman, 1997). CO2 mayalso help absorption by reducing the thickness and viscosity of the mucus layer adjacent tothe mucosa (Pather et al., 2002).

Chewable Tablets

Most chewable confectionery products are gelatin based. Gelatin is a very reactive surfacethat interacts with mucin rendering bioavailability very difficult.

Conclusions

The oral mucosa responds to the senses of pain, touch, and temperature in addition to itsunique sense of taste. Some physiological processes are triggered by sensory input from themouth such as the initiation of chewing, masticating, swallowing, etc. The most importantphysiological variable, however, that can markedly affect the release characteristics of anactive from a confectionery dosage is whether a person sucks or chews the formulation,since systems designed to be chewed will invariably be sucked and vice versa by some indi-viduals. Chewing gums possess an advantage over other confectionery formats for control-ling the release of drugs such as nicotine, caffeine, or other medicinal substances in that ifthe gum is swallowed, release of the active in the stomach and lower GIT is extremely low;therefore, reducing potential or toxicity.

Drugs and actives ingested via the oral route can be designed for either or both of thefollowing:

• Local Delivery: Confectionery products have demonstrated their practical effectivenessin delivering flavors, cooling and warming agents, antimicrobials, caries prevention andxerostomia relief agents; and

• Systemic Delivery: Nicotine, vitamins, caffeine, salicylic acid are the most commonactives that can be embedded in a confectionery matrix and have the potential to beabsorbed through the oral mucosa into the circulation, thus giving rise to a systemic effect.Actives absorbed directly via the membranes lining the oral cavity avoid metabolism inthe GIT and the first-pass effect of the liver since the oral veins drain directly into thevena cava. Alternatively, actives released from a chewing gum or other confectionerydosage form—but not absorbed through the oral cavity—membranes will be swallowedand enter the stomach in a dissolved or a dispersed form in saliva.

196 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 196

Page 203: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

ReferencesAndersen, C. 2004. Method of preparation of chewing gum with customer acceptable taste. US patent 0028772 A1.Anderson, M.P. 1992. Lack of bioequivalence between disulfiram formulations. Acta Psychiatr. Scand. Suppl. 86:

31–35W.Anlar, S., Capan, Y., Guven, O., Gogus, A., Dlarara, T. and Hincel, A.A. 1984. Formulation and in-vitro in-vivo

evaluation of buccoadhesive morphine sulfate tablet. Pharm. Res. 11: 231–236.Assev, S. and Rølla, G. 1986. Sorbitol increases the growth inhibition of xylitol on Strep. mutans OMZ 176. Acta

Path. Microbiol. Immunol. Scand. Sect. B. 94: 231–237.Barklow, D.G., Chapdelaine, A.H. and Dzjia, M.R. 2002. Pullulan-free edible film compositions and methods of

making the same. US Patent 0131990 A1.Bealin-Kelley, F.J., Hanke, B. and Nienaber, P. 2002. Throat soothing compositions. US Patent 6432441 B1.Beckett, A.H. and Moffat, A.C. 1969. The influence of substitution in phenylacetic acid on their performance in

the buccal absorption test. J. Pharm. Pharmacol. 21: 139S–143S.Benowitz, N., Jacob, P. and Savanapridi, C. 1987. Determination of nicotine intake while chewing nicotine

polacrilex gum. Clin. Pharmacol. Ther. 41: 467–473.Berridge, M.J. and Oschman, T.L. 1972. In Transporting Epithelia, Academic Press, New York and London. Blanchette, J., Kavimandan, N. and Peppas, N.A. 2004. Principles of transmucosal delivery of therapeutic agents.

Biomed. Pharmacother. 58: 142–151.Bouckaert, S., Lefebvre, R.A. and Remon, J-P. 1993. In vitro/in vivo correlation of the bioadhesive properties of a

buccal bioadhesive miconazole slow-release tablet. Pharm. Res. 10(6): 853–856.Broderick, K.B. and Record, D.W. 1992. Gum composition containing dispersed porous beads containing active

chewing gum ingredients and method. US patent 5139787.Bunczek, M.T. and Urnezis, P. 1993. Aspartame stability in chewing gum using an acid gelatin system. US Patent

5192561.Channer, K.S. and Roberts, C.J.C. 1985. Effect of delayed esophageal transit on acetominophen absorption. Clin.

Pharmacol. Ther. 37: 72–76.Chauncey, H.H., Lionetti, F., Winer, R.A. and Lisanti, V.F. 1957. Enzymes of human saliva I. The determination,

distribution and origin of the whole saliva enzymes. J. Dent. Res. 33: 321–334. Chow, L.C. and Takagi, S. 2001. Calcium phosphate hydroxyapatite precursor and methods for making and using

the same. US Patent 6325292 B1.Choi, H-G. and Kim, C-K. 2000. Development of omeprazole buccal adhesive tablets with stability enhancement in

human saliva. J. Control. Release 68: 397–404.Christup, L.L. and Møller, N. 1986. Chewing gum as a drug delivery system. I. in vitro simulation of human masticatin

and influence of formulation upon the release rate of a water soluble drug. Arch. Pharm. Chem. Sci. Edu. 14: 30–36.Christup, L.L., Davis, S.S. and Frier, M. 1990. Deposition of a model substance 99mTcE-HIDA, in the oral cavity

after administration of lozenges, chewing gum and sublingual tablets. Int. J. Pharm. 66: 169–174.Christup, L.L., Rasmussen, S.N. and Rassing, M.R. 1988a. Chewing gum as a drug delivery system. Proc. 3rd Int.

Conf. Drug Absorpt. Edinburgh.Christup, L.L., Bonde, J., Eriksen, H., Rasmussen, S.N., Rasig, M.R. and Simonsen, K. 1988b. Chewing gum as a

drug delivery system. III. Bioavailability of salicylamide administered in tablets and chewing gum. Farmaci. Sci.Ed. 16: 6–14.

Clark, J.C. and Shen, C.A. 2004. Fast flavor release coating for confectionery. US Patent 0175489 A1.Collins, L.M.C. and Dawes, C. 1987. The surface area of the adult human mouth and thickness of the salivary film

covering the teeth and oral mucosa. J. Dent. Res. 66: 1300–1302.Commission of the European Communities. 1991. CPMP list of allowed terms for the pharmaceutical dosage form,

route of administration, container, closure and administration devices, III/3593/91.Curatolo, W. 1987. The lipoidal permeability barriers of the skin and alimentary tract. Pharm. Res. 4: 271–277.De Vries, M.E, Boddé, H.E. and Junginer, H.E. 1991. In vitro and in vivo adhesion performance of buccal

mucoadhesive patches. In thesis: De Vries, M. Buccal drug absorption and development of mucoadhesive poly-mer systems, State University of Leiden, The Netherlands.

Dobrozsi, D.J. 2003. Oral liquid mucoadhesive compositions. US Patent 6638521.Donbrow, M. and Freidman, M. 1974. Permeability of films of ethyl cellulose and PEG to caffeine. Commun.

J. Pharm. Pharmac. 26: 148–150.Eichman, J.D. 1997. Mechanistic studies on effervescent permeability enhancement; PhD dissertation, University

of Wisconsin-Madison.

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 197

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 197

Page 204: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Evered, D.F. and Mallett, C. 1983. Thiamine absorption across human buccal mucosa in vivo. Life Sci. 32:1355–1358.

Evered, D.F, Sadooght-Abasian, F. and Patel, P.D. 1980. Absorption of nicotinic acid and nicotinamide acrosshuman buccal mucosa in vivo. Life Sci. 27: 1649–1651.

Fernö, O. 1973. A substitute for tobacco smoking. Psychopharmacologia 31: 201–204.Fromter, E. and Diamond, J. 1972. Route of passive ion permeation in epithelia. Nature New Biol. 235: 9–13.Gandhi, R.E. and Robinson, J.R. 1988. Bioadhesion in drug delivery. Ind. J. Pharm. Sci. 50: 145–152.Gelski, J. 2006. Tea’s weight loss potential cited as additional benefits. Food Business News 28: 38–40.Gudas, V.V., Reed, M.A., Schnell, P.G., Tyrpin, H.T., Russell, M.P. and Witkewitz, D.L. 2000. Method of controlling

release of caffeine in chewing gum. US Patent 6165516.Guo, J-H. 1994. Investigating the surface properties and bioadhesion of buccal patches. J. Pharm. Pharmacol. 46:

647–650.Haines, H.G. 2004. A pilot study evaluating the bioavailability and absorption rates of two vitamin B12 prepara-

tions in normal human subjects. Health Plus International, Inc. (study protocol # HPI-NF-B12–1).Harris, D. and Robinson, J.R. 1992. Drug delivery via the mucous membranes of the oral cavity. J. Pharm. Sci. 81:

1–10.Hayward, A.F. 1979. Membrane-coating granules. Int. Rev. Cytol. 59: 97–127.Hutchinson, K.G. and Garnett, K.R. 1999. Method of making water-based beverages. US 5871789.Jacob, J.S., Mathiowitz, E., Nangia, A., Shaked, Ze’ev and Moslemy, P. 2006. Controlled regional oral delivery. US

Patent 0045865 A1.Jenkins, G.N. and Edgar, W.M. 1989. The effect of daily gum-chewing on salivary flow rates in man. J. Dent. Res. 5:

786–790.Johnson, S. and Yatka, R.J. 2000. Method of making chewing gum products containing perillratine. US patent

6159509.Johnson, S., Sheldon, G. and Greenberg, M.J. 2004. Oral products containing novel flavor composition. US Patent

0141927.Kaaber, S. 1973. Studies on the permeability of human oral mucosa VI. The mucosal transport of water, sodium and

potassium under varying osmotic pressure. Acta Odont. Scand. 31: 307–316.Kaaber, S. 1974. The permeability and barrier functions of the oral mucosa with respect to water and electrolytes.

Acta Odontol. Scand. 32 (Suppl 66): 26.Kahn, C.R. and Shechter, Y. 1991. “Oral hypoglycemic agents and the pharmacology of the endocrine pancreas”.

In Theodor W.R., Alan, S.N., Taylor, P. and Gilman, A.G., (Eds) Goodman and Gilman’s The PharmacologicalBasis of Therapeutics, 8th ed., New York, NY: McGraw Hill: 1484.

Kamimori, G.H., Karyekar, C.S., Ottersetter, R., Cox, D.S., Balkin, T.J., Belenky, G.L. and Eddington, N.D. 2002.The rate of absorption and relative bioavailability of caffeine administered in chewing gum versus capsules tonormal healthy volunteers. Int. J. Pharm. 234(1–2): 159–167.

Kanwar, Y.S., Linker, A. and Farquhar, M.G. 1980. Increased permeability of the glomerular basementmembrane to ferritin after removal of glucosaminoglycans (heparin sulfate) by enzyme digestion. J. Cell Biol.688–693.

Karles, G., Garthaffner, M., Juper, R., Kellog, D. and Skinner, I. 2006. Flavor capsule for enhanced flavor deliveryin cigarettes. US Patent 082529 A2.

Kaufmann, K. 2003. Hard candy with plaque-neutralizing effect comprising alkali metal monophosphate. USPatent 6582731 B1.

Kayane, S., Tsutomu, I., Iwase, T. and Miura, Y. 2003. Throat care agents. US Patent 0170322 A1.Lakkis, J. 2006. Roadmap to development of healthy food products via encapsulation and controlled release tech-

nologies. Paper presented at the Controlled Release Society Meeting, July 25, 2006, Vienna, Austria.Lakkis, J. and Wong, S. 2007. Multilayered confectionery product with controlled release teeth remineralization

actives (patent submitted).Leach, S.A., Lee, G.T.R. and Edgar, W.M. 1989. Remineralization of artificial caries-like lesions in human enamel

in situ by chewing sorbitol gum. J. Dent. Res. 68(6): 1064–1068.Lee, R.E. 2004. Effervescent tablets: Key facts about a unique effective dosage form. Tablets and Capsules. July:

40–45.Lesch, C.A., Squier, C.A., Cruchley, A., Williamson, D.M. and Speight, P. 1989. The permeability of human oral

mucosa and skin to water. J. Dent. Res. 68: 1345–1349.Lindqvist, L. and Augustinsson, K.B. 1975. Esterases in human saliva. Enzyme 20: 277–291.Lombardy, C.M., Lombardy, D.R. and Liebrecht, J.W. 2001. Effervescent chewing gum. US patent 6235318 B1.

198 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 198

Page 205: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Macknight, A.D.L. and Leader, J.P. 1983. Intra- and trans-epithelial analytical techniques. CRC Crit. Rev. Clin. Lab.Sci. 18: 339–395.

Manning, A.S. and Evered, D.F. 1976. The absorption of sugars from the human buccal cavity. Clin. Sci. Molec.Med. 51: 127–132.

Mazurek, P.M., Reed, M. and Broderick, K. 2000. Chewing gum with dental benfits including calcium in a foodgrade acid. WO 00/62762.

McGrew, G.N., Maxwell, J.R., Tyrpin, H., Greenberg, M.J. and Record, D.W. 2006. Chewing gum that providesbreath freshening characteristics. US patent 7087255 B2.

Merritt, C., Wingerd, W.H. and Keller, D.J. 1985. Encapsulated flavorant material, methods for its preparation andfood and other compositions incorporating the same. US Patent 4515769.

Mulry, J.T. 1988. Nicotine gum dependency: A positive addiction. Drug Intell. Clin. Pharm. 22: 313–314.Nicolazzo, J.A., Reed, B.L. and Finnin, B. 2003. Assessment of the effects of sodium dodecyl sulfate on the buccal

permeability of caffeine and estradiol. J. Pharm. Sci. 93(2): 431–440.Nuernberg, B. and Brune, K. 1989. Buffering the stomach content enhances the absorption of diflunisal in man,

Biopharm. Drug Dispos. 10: 377–387.Odumusu, A. and Wilson, C.W. 1977. The buccal absorption of ascorbic acid and its passage through lipoid mem-

brane. Int. J. Vitam. Nutr. Res. 47: 135–144.Ozaki, Y. and Miyake, T. 1995. Pullulan binder and its uses. US Patent. 5411945.Paetau, I., Rao, D., Wiley, E.R., Brown, E.D. and Clevidence, B.A. 1999. Carotenoids in human buccal mucosa cells

after 4 wk of supplementation with tomato juice or lycopene supplements. Am. J. Clin. Nutr. 70: 490–494.Pang, K.S. 2003. Modeling of intestinal drug absorption: Roles of transporters and metabolic enzymes (for the

Gillette review series). Drug Metab. Dispos. 31: 1507–1519.Pather, S.I., Robinson, J.R., Eichman, J.D., Khankai, R.K., Hontz, J. and Gupta, S.V., 2002. Effervescent drug

delivery systems for oral administration. US Patent 6391335.Pedersen, M. and Rassing, M.R. 1990. Miconazole and miconazole nitrate chewing gum as drug delivery

systems—a practical application of solid dispersion technique. Drug Dev. Ind. Pharm. 16(1): 55–74.Rassing, M.R. 1994. Chewing gum as a drug delivery system. Adv. Drug Deliv. Rev. 13: 89–121.Ream, R., Greenberg, M., Wokas, W. and Corriveau, C.L. 2003. Over-coated chewing gum formulation. US Patent

20030313 A1.Ream, R.L., Corriveau, C.L., Graff, G. and Matulewicz, L. 2001. Over-coated chewing gum formulations including

tabeleted center. US Patent 6322806 B1.Rivier, V. 2005. Confectionery product containing functional ingredients. EP 1280414 B1.Rojanasakul, Y., Wang, L-Y., Bhat, M., Glover, D.D., Malange, C.J. and Ma, J.K.H. 1992. The transport barrier of

epithelia: A comparative study of membrane permeability and charge selectivity in the rabbit. Pharm. Res. 9:1029–1034.

Rutherford, H.J., Desai, N., McDermott, K. and Wiener, C. 1992. Chewing gum containing compositions for con-trolled release of flavor-bearing substances and process for producing same. EP 0502750 A1.

Sadoogh-Abasian, F. and Evered, D.F. 1979. Absorption of vitamin C from the human buccal cavity. Br. J. Nutr. 42:15–20.

Sadoogh-Abasian, F. and Evered, D.F. 1980. Absorption of nicotinic acid and nicotinamide across human buccalmucosa. Life Sci. 27: 1649–1651.

Savage, W.D., Schnell, P.G., Aumann, R.A. and Yatka, R.J. 2002. Chewing gum formulations including encapsu-lated aspartame and sodium phosphate. US Patent 0142068 A1.

Schanker, L.S. 1964. Physiological transport of drugs. Adv. Drug Res. 1: 71–106.Semple, W.F. 1869. Improved chewing gum. US Patent 98304.Sexton, D. and Lakkis, J. 2003. Encapsulated alcoholic beverages and method of making same. US Patent 750802.Sharma, S.C. and Yang, K.Y. 1986. Chewing gum compositions containing novel sweetener delivery systems and

method of preparation. US patent 4597970.Siegel, I.A. 1984. “Permeability of the oral mucosa”. In Meyer, J., Squier, C.A. and Gerson S.J. (Eds) The Structure

and Function of Oral Mucosa. Oxford, Pergamon Press. 95–108.Siegel, I.A., Hall, S.H. and Stambaugh, R. 1971. “Permeability of the oral mucosa”. In Squier C.A. and Meyer J.

(Eds) Current concepts of the histology of oral mucosa. Charles C. Thomas, Springfield. 274–286Silagy, C., Lancaster, T., Stead, L., Mant, D. and Fowler, G. 2002. Nicotine replacement therapy for smoking cessa-

tion. Cochrane Database Syst. Rev. 4: CD000146.Song, J.H. and Copper, C.M. 1992. Gradual release structures for chewing gum. US patent 5165944.Song, J.H. and Courtwright, S.B. 1992. Flavor releasing structures for chewing gum. US Patent 5128155.

Confectionery Products as Delivery Systems for Flavors, Health, and Oral-Care Actives 199

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 199

Page 206: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Song, J.H., Sundstrom, E., Record, D., Townsend, D., Broderick, K.B. and Schnell, P. 1992. Gum compositioncontaining dispersed porous beads containing active ingredients and method. US patent 5154927.

Squier, C.A. and Hall, B. 1985. The permeability of skin and oral mucosa to water and horseradish peroxidase asrelated to the thickness of the permeability barrier. J. Invest. Dermatol. 84: 176–179.

Squier, C.A., Johson, N.W. and Hopps, R.M. 1976. Human Oral mucosa: Development, Structure and Function.Blackwell Scientific, Oxford.

Squier, C.A. and Kremer, M.J. 2001. Biology of oral mucosa and esophagus. J. Natl. Cancer Inst. Monograph 29:7–15.

Squier, C.A. and Wertz, P.W. 1996. Structure and function of the oral mucosa and implications for drug delivery,pp. 1–26, in Oral Mucosal Drug Delivery, Rathbone, M.J. (Ed.), Marcell Dekker, Inc. New York, NY.

Stevenson, N.R. 1974. Active transport of L-ascorbic acid in the human ileum. Gastroenterology 67: 952–956.Sunohara, H., Kamaguchi, R., Kagawa, M., Nishikawa, M. and miura, Y. 2002. Multilayered soft capsule for elimi-

nating bad breath and process for producing the same. US Patent 6426089 B1.Syed, S.A., Kamimori, G., Kelley, W. and Eddington, N.D. 2005. Multiple dose pharmacokinetics of caffeine

administered in chewing gum to normal healthy volunteers. Biopharm. Drug Dispos. 26(9): 403–409.Tabak, L.A., Levine, M.J., Mendell, I.D. and Ellison, S.A. 1982. Role of salivary mucins in the protection of the

oral cavity. J. Oral Pathol. 11: 1–7.Tan, S.C. 1976. Human saliva esterases. Genetic studies. Human Hered. 26: 207–216.Tanner, K. and Shelley, R.S. 1996. Fill compositions for soft elastic gel capsules. US patent 5569466.Tucker, I.G. 1988. A method to study the kinetics of oral mucosal drug absorption from solutions. J. Pharm.

Pharmacol. 40: 679–683.Tuovinen, L., Peltonen, S. and Järvinen, K. 2003. Drug release from starch-acetate films. J. Control. Release 91:

345–354.Tutuncu, N.P., Hartunian, S., Bratton, J.A. and Stanton, K.J. 2003. Confectionery product having a salivation region

and an oral comfort region. US Patent 0118628 A1.Veillard, M.M., Longer, M., Martens, T.W. and Robinson, J.R. 1987. Preliminary studies of oral mucosal delivery of

peptide drugs. J. Control. Release 6: 123–131.Virgallito, M.T. and Zhang, J. 2006. Edible film containing food acid. US Patent 0035008 A1.Weatherell, J.A., Robinson, C. and Rathbone, M.J. 1994. Site-specific differences in the salivary concentration of

substances in the oral cavity—implications for the etiology of oral disease and local drug delivery. Adv. DrugDel. Rev. 13: 23–42.

Weatherell, J.A., Strong, M., Robinson, C., Nakagi, H. and Ralph, J.P. 1984. Migration of fluoride in the mouth.Caries Res. 18: 348–353.

Weatherell, J.A., Strong, M., Robinson, C., Nakagi, H. and Ralph, J.P. 1989. Retention of glucose in oral fluid at dif-ferent sites in the mouth. Caries Res. 23: 399–405.

Wennerholm, K. and Emilson, C.-G. 1989. Effect of sorbitol- and xylitol-containing chewing gum on salivarymicroflora, saliva, and oral sugar clearance. Scand. J. Dent. Res. 97: 257–262.

Wertz, P.W. and Downing, D.T. 1983. Ceramides of pig epidermis: Structure determination. J. Lipid Res. 24:759–765.

Winston, A. and Usen, N. 2002. Products and methods for the remineralization and prevention of demineralizationof teeth. US Patent 6451290 B2.

Witzel, F., Wayne, J.P., Warren, C., Donald, A. and Mackay, M. 1980. Chewing gum capable of releasing finelydivided water-insoluble materials therefrom. US Patent 4238475.

Wolf, F.R., Mcgrew, G.N. and Tyrpin, H.T. 2005. Chewing gum containing controlled release acyclic carboxamides.US 0082271 A1.

Wolfson, P. 2002. Herbal composition for improving oral hygiene, for providing local anesthesia, for use as an oralsensate, flavor enhancer and potentiator, and methods of using same. US patent 0122778 A1.

Wong, F.W., Yuen, K.H. and Peh, K.K. 1999. Formulation and evaluation of controlled release Eudragit buccalpatches. Int. J. Pharm. 178(1): 11–22.

Wonschik, J., Machinek, A. and Koschorreck, C. 2005. Coated spherical: Seamless filled capsules. WO2005/077521 A1.

Woodford, W. and Lesko, L.J. 1981. Relative bioavailability of aspirin gum. J. Pharm. Sci. 70: 1341–1343.Yang, J. H. 2005. Gelatin soft capsule having the properties of removal of oral smell and cleaning of oral cavity. US

Patent 6852309 B2.

200 Chapter 8

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_08_171-200 r1.qxd 3/29/07 1:23 PM Page 200

Page 207: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

9 Innovative Applications of Microencapsulation in Food Packaging

Murat Ozdemir and Tugba Cevik

Introduction

The use of proper packaging materials and methods to minimize food losses to provide safeand wholesome food products has always been the focus of food packaging. In addition,consumer demands for better-quality, fresh-like and convenient products have been intensi-fying in the last two decades. A wide variety of packaging materials and technologies havebeen developed to meet these consumer requirements and to limit package-related environ-mental pollution and disposal problems (Ozdemir and Floros, 2004). Despite these advancesand availability of unique materials such as plastics that can be specifically designed to delayadverse effects of the environment on food products and to extend their shelf-life, novelapproaches to the development of packaging materials containing microencapsulated activeparticles have emerged recently.

Encapsulation is a technique by which a material or a mixture of several materials can becoated or entrapped in another material. The development of a successful microencapsu-lated product primarily depends on:

1. selecting an appropriate shell formulation, usually GRAS (generally recognized as safe)materials that are approved by the Food and Drug Administration (FDA) or other inter-national health authorities;

2. selecting an appropriate process to provide the desired functionality, stability, andrelease mechanism;

3. economic feasibility of large-scale production including capital, operating costs, andother miscellaneous expenses.

An appropriate shell formulation must stabilize the core material, must not react with ordeteriorate the active agent, yet releases it under specific conditions based on the productapplication. Polysaccharides, proteins, waxes, fatty acids, gums and their derivatives arecommon shell materials that are approved for food use.

Microencapsulation of food ingredients can be achieved by either physical or chemicalmethods. Physical methods include extrusion, fluidized bed, spinning disc, and spray drying.Chemical methods include coacervation, gelation, phase separation, and molecular inclu-sion. Microcapsules can be produced by depositing a thin polymer coating on small solidparticles or liquid droplets, or by the process of dispersion of solids in liquids. The corematerial or the active agent may be released by friction, pressure, diffusion through the poly-mer wall, dissolution of the polymer wall coating, or by biodegradation.

201

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 201

Encapsulation and Controlled Release: Technologies in Food SystemsEdited by Jamileh M. Lakkis

Copyright © 2007 by Blackwell Publishing

Page 208: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Microencapsulated Actives for Packaging Applications

Microencapsulation can most often extend the shelf-life of foods while improving theirnutritional quality, appearance, and in various instances inhibiting the growth of pathogenicand spoilage microorganisms, thus ensuring food safety. Important examples of microen-capsulation in food packaging include incorporation of antimicrobial agents, insect and/orrodent repellents, scented fragrance-inserts and flavor-releasing systems, pigments, inks,and time–temperature indicators.

Antimicrobial Food Packaging Materials

Traditional food protection techniques include curing, smoking, or pickling which wereprimarily effective in changing the moisture content or water activity of the foods. In recentyears, more sophisticated preservation methods have been developed to extend shelf-life offoods. Figure 9.1 shows an example of novel microcapsules that can deliver preservativesfrom plastic films or edible coatings that are currently available. Changing lifestyles andthe limited time available for food preparation require an increasing variety of high-quality,safe, nutritious, and convenient food products today.

Allyl isothiocyanate is an effective inhibitor against various pathogens, particularlyEscherichia coli O157:H7. Consumption of undercooked ground beef has been identified

202 Chapter 9

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Plastic film Food Active agent releasedfrom the microcapsule

Core containingactive agent

Microcapsule

(a)

Plastic film Edible coating Food Active agent releasedfrom the microcapsule

Core containingactive agent

Microcapsule

(b)

Figure 9.1. Migration of active substance from (a) plastic film, and (b) edible coating.

Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 202

Page 209: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

as one of the main causes of E. coli O157:H7 outbreaks in North America (Waters et al.,1994). Chacon et al. (2006) microencapsulated allyl isothiocyanate in gum acacia and cornoil prior to incorporating the preparation into aseptically treated chopped beef that wasinoculated with a known concentration of E. coli O157:H7. The system was packed undernitrogen and stored under refrigeration (4°C). After 18 days, the chopped beef was found tobe free of E. coli O157:H7. Similarly, Klein et al. (2004) described a method for microen-capsulating actives such as antibacterial and antifungal agents into a variety of polymericfood packaging materials such as polyethylene, polypropylene, polyester, polycarbonate,polyvinyl chloride, and polyvinylidene chloride. These films were described as especiallyuseful for controlling bacteria and fungi on food surfaces. Agar diffusion tests showedthat microencapsulated antibacterial agents were effective against Staphylococcus aureus,E. coli, Pseudomonas aerugenosa, and Streptococcus spp. Thomas et al. (2005a, b) paten-ted a method in which an antimicrobial agent was microencapsulated prior to being inte-grated into the package structure via extrusion. The method was claimed to be effective inproviding thermal stability during package processing and manufacturing, but readilyreleased the active agent upon contact with moisture.

Avery Dennison Corp. (USA) developed an antimicrobial active label that releases traceamounts of chlorine dioxide (ClO2) gas from the label. Chlorine dioxide is a broad spec-trum antimicrobial agent effective against both bacteria and fungi. It can also be used toeliminate odors and retard microbial growth on perishable food products, thus extendingtheir shelf-life. Laboratory tests showed that the inclusion of one small antimicrobial labelon the inside of rigid plastic packaging can significantly extend the shelf-life of freshberries. The time release delivery of the chlorine dioxide is moisture activated. The mainadvantage of this system is that it does not require direct contact with the food.

A promising application of controlled release is in antimicrobial agents incorporatedinto chewing gums for reducing the growth of microorganisms in the mouth and therebyretarding tooth decay. Barabolak et al. (2005) produced a chewing gum with controlled-release properties in which the antimicrobial agent (chlorhexidine digluconate) was encap-sulated via film coating. Particles containing the encapsulated antimicrobial agent wereclaimed to be adaptable to produce fast or delayed release when the gum is chewed.

Microencapsulating properties of whey proteins have been investigated extensively inrecent years (Ozdemir and Floros, 2001; Rosenberg, 1997). Whey protein concentrate andwhey protein isolate have been shown to exhibit excellent microencapsulating propertiesfor both volatile and non-volatile core materials (Young et al., 1993a; Lee and Rosenberg,2000). The high microencapsulation yield of whey proteins is presumed to be a result oftheir efficient emulsifying capacity, especially in the presence of carbohydrates (Younget al., 1993b). Films and coatings from whey not only degrade more readily than polymericmaterials, but also could supplement the nutritional value and improve the sensoryattributes of coated foods. Ozdemir and co-workers (Ozdemir, 1999; Ozdemir and Floros,2001) developed active antimicrobial films made of whey protein isolate, sorbitol,beeswax, and potassium sorbate. The mechanism and release profile of potassium sorbatein these films were found to follow non-Fickian diffusion model. A mathematical modelderived from Fick’s second law of diffusion with a time-dependent diffusion coefficient wasused to analyze potassium sorbate diffusion. Subsequent analysis showed that diffusioncoefficients of potassium sorbate in whey protein films were ten-fold higher than those inedible wheat gluten and low density polyethylene (LDPE) films, and ten-fold lower thanthose in intermediate moisture foods.

Innovative Applications of Microencapsulation in Food Packaging 203

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 203

Page 210: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

In a recent study, Ozdemir and Floros (2003) investigated the effect of different con-stituents (levels of whey protein, plasticizer, wax, lipid, and antimicrobial agent as encapsu-lant) on the diffusivity of potassium sorbate using mixture response surface methodology.Their study showed that increasing whey protein concentration in the film decreasedpotassium sorbate diffusivity. Sorbate diffusion increased with increasing sorbitol concen-tration but decreased with increased concentration of beeswax in the film. A rise in the ini-tial active (potassium sorbate) concentration in the film resulted in higher diffusioncoefficients. Strong interactions were observed between beeswax and potassium sorbate,and whey protein and beeswax.

Insect and/or Rodent Repellent Food Packages

Insect infestation of produce and food products results in spoilage and subsequent eco-nomic losses. Controlling insect infestation has generally been achieved by fumigationwith methyl bromide. Methyl bromide is a toxic substance that can adversely affect thehuman central nervous and respiratory systems if present at high concentrations. Methylbromide is also known to be a major contributor to the depletion of Earth’s ozone layer. Oneway to overcome the disadvantages of methyl bromide is to find less toxic and less harmfulinsect repellents and to incorporate them into packaging materials to form packages withcontrolled-release properties.

Microencapsulation of pesticides, herbicides, and other pest control agents has been anactive area of development. Pest control agents are currently microencapsulated to prolongtheir activity while reducing mammalian toxicity, volatilization losses, phytotoxicity, andenvironmental degradation. Spector (1981) introduced a low-cost, self-stick tab in which anactive agent-saturated pad can be enveloped within a perforated sac which can then beadhered to a desired site to release the active over an extended period of time. The active agentcan be an insect or animal repellent or a fragrance (Figure 9.2), and the system can be stuckdirectly onto food packages or boxes to prevent their infestation by insects and animals.

The protection of agricultural products with biopesticides has been promoted recently asa means of reducing the adverse effects of chemical insecticides (Marrone, 1999). Develop-ment cost, time and ease of registration, and potential growing markets make biopesticidespopular over chemical pesticides (Brar et al., 2006). A number of biopesticides (bacteria,

204 Chapter 9

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Core containingrepellent material

Microcapsule wall

(a) (b)

Figure 9.2. Description of the action of a system composed of microcapsules containinga repellent material: (a) microcapsule core containing a repellent, and (b) diffusion andevaporation of the repellent material through the wall.

Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 204

Page 211: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

fungi, virus, pheromones, plant extracts) have been already in use to control various types ofinsects responsible for the destruction of agricultural crops. Bacillus thuringiensis-basedbiopesticides are especially important and are estimated to constitute almost 97% of theworld biopesticide market (Cannon, 1993).

A biological pesticide is effective only if it has a potential major impact on the targetpest, market size, cost- effective and is capable of overcoming a number of technologicalchallenges such as fermentation, formulation and delivery systems. Biopesticides havebeen encapsulated in a coating made of gelatin, starch, cellulose, or other polymers, andeven microbial cells (Barnes and Cummings, 1987a, b; Barnes and Edwards, 1989). Boket al. (1993) showed that encapsulated microbial pesticides possess excellent adhesiveness;therefore, they can be applied directly to the soil or the plant. When the encapsulatedbiopesticide is embedded into a plastic film, the film can be applied near the roots or cuts ofthe crops to protect them against pathogens upon storage or during transportation.

Another recent advance in encapsulation is the production of hydrocapsules that arewater-based shellcores, consisting of a polymer membrane surrounding a liquid center.These shells can be produced using UV radiation-initiated free-radical copolymerization offunctionalized prepolymers (silicones, urethanes, epoxys, polyesters, etc.) and/or vinylmonomers such as acrylates for better dispersion and UV radiation protection (Lechelt-Kunze et al., 2000; Toreki et al., 2004).

El-Rehim et al. (2005) formulated a polyacrylamide/polyethylene oxide hydrogel toencapsulate and cross-link bioactives such as Atrazine. The active was incorporated intothe hydrogel matrix via electron beam irradiation process. Results showed that copolymerblend composition, its gel content, and irradiation dose greatly affected the Atrazine releaserate. In addition, Atrazine release rate was found to decrease with increasing pH butincreased at high temperatures.

Packaged products are also susceptible to infestation by many insects and mites whichare capable of perforating the packaging material or use existing holes or openings in thefood package for penetration. Rieth et al. (1986) encapsulated 2-heptanone, an insect repel-lent for bees and other insects, in a polyvinyl chloride–polyvinyl acetate plastic. Jones andHill (1982) added naphthalene flakes and citronella oil in solid form to synthetic resinssuch as polyethylene, polypropylene, and polystyrene to form insect- and animal-repellentplastic films. The resultant films were shown to have lower tensile and tear strengths thanfilms made without the actives (insect and animal repellent additives).

Atkinson (1991) described a microencapsulation process for manufacturing animal-repellent plastic films where terpenes were incorporated into linear low-density polyethyl-ene (LLDPE) melt via extrusion. Radwan and Allin (1997) developed a controlled releaseinsect repellent device that was described as useful for foods, tobacco, and other consum-able items with the active being an essential oil. Navarro et al. (2005) produced a pest-impervious packaging material by combining ar-turmerone, sesquiterpene alcohols, and/orturmeric oleoresin solid residue. These materials were incorporated into plastics, adhe-sives, or printing inks via microencapsulation.

Scented Fragrance-Inserts and Flavor-Releasing Systems

Food packaging materials, particularly plastics, may interact with food flavors, resulting inloss of flavors, known as flavor scalping, therefore the need to replace these lost flavorconstituents. Although the use of high-barrier plastics holds food flavors in the package,

Innovative Applications of Microencapsulation in Food Packaging 205

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 205

Page 212: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

additional flavor-releasing systems may be necessary in some instances, particularly whenheat seal layers of a package have high affinity to flavors. In addition, consumers alwayslike to smell pleasing flavors when they first open a food package (Brody, 1992). Flavor-releasing systems are highly popular in the beverage industry, microwavable foods, andcoffee makers. The beverage industry, particularly the soft drink segment, is highly com-petitive. Manufacturers take great care and make substantial efforts to formulate their prod-ucts in such a way to differentiate them from their competitors’ and to make consumptionof the food product more enjoyable for the consumers. In the soft drink industry, despite thehigh contribution of taste to the overall soft drink experience, its aroma especially when thebottle is first opened is equally important. Ashcraft and Wong (1993) invented a packagethat releases a burst of flavor when the package is opened. The novel flavor-burst structurecomprised a multilayer film with a flavor-carrier layer disposed between the barrier layers.Due to their chemical incompatibility, the flavor agent desorbs from the carrier when one ofthe barrier layers is removed from the carrier. Sun et al. (2000) developed a flavored poly-ethylene terephthalate (PET) packaging system where the aroma is released once the bottlecap was opened. Unfortunately, typical microencapsulated materials do not adhere well toPET; thus the surface of PET bottles must be modified before the microencapsulated mate-rial is applied to the bottles. A successful way to resolve this problem is by treating thesurface with a primer that enables the microencapsulated material to adhere to PET or toroughen the surface using laser etching.

The use of overwraps on packages can improve the appearance and maintain the quality ofmaterials within the package. The use of a tear strip having the ability to release fragrance atthe time of opening of the package can add further benefit to the overwrap; for example,release of a controlled fragrance can give the impression to the user that the ingredients of thepackage are fresh. Fraser (1988) described a package that releases a fragrant liquid frommicrocapsules when a tear strip is removed from the package. In this system, the separation ofmultilayer sheet materials ruptures the microcapsules fitted to the intermediate adhesive lay-ers and subsequently releases the entrapped fragrance. The described packages can be madeof paper, cardboards, polymeric materials, coated paper, foil, composite structures, metal-lized paper, and so on. In a similar application, Sprinkel and Newsome (1988) microencapsu-lated an aromatic substance in a cigarette package–overwrap where the aromatic substance isreleased upon pulling the tear strip. This mechanism was described as useful for releasingaroma of freshness or for adding flavorings to the cigarettes in the package.

CSP Technologies (US) developed an aroma emitting and aroma absorbing package inwhich the active agent was encapsulated within three component plastic system. DisperseTechnologies (UK) combined a patented thin film–encapsulating technology with ultravio-let curing technology to produce films and coatings that have controlled-release properties(Wheeler, 2001). These films and coatings were claimed to possess a very long-lastingimpact for up to 6 months. Arcade Marketing (based in the United States) commercializedthe MicroFragrance label for foods, especially to promote the sales of low calorie cerealbars. MicroFragrance label is printed onto a clear film so that it does not wear down orblend with another smell such as paper.

Driscoll Labels (US) customized long-lasting, scratch-and-sniff labels for the fragranceand food industries. This technology allows the consumer to perceive the aroma of the foodwithout opening the package. Scentisphere developed a printable, scented ink known asrub-and-sniff ink that can be printed directly onto packaging materials. The rub-and-sniffink is claimed to have advantages over the traditional scratch-and-sniff labels since the

206 Chapter 9

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 206

Page 213: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

scented inks can be added to the ink using standard printers without any interruption in theprinting process or the inconvenience of having the ink drying quickly.

The US-based company ScentSational Technologies™ is a pioneering company in micro-encapsulated packaging. A schematic of their CompelAroma® flagship technology forencapsulating flavors into plastic-based packaging films is described in Figure 9.3. Usingthis technology, food grade aromas/flavors can be embedded directly into plastic films dur-ing manufacturing so that they form an integral part of the package. This technology isclaimed to be applicable to most existing manufacturing methods, including blow molding,injection molding, thermoforming, and extrusion, and in gaskets and liners. Com-pelAroma® technology can be used in containers, trays, cups, closures, bottles, and flexiblepackages. The first commercial application of CompelAroma® technology was in Aquaes-cents® refillable water bottles marketed by NutriSystem™ (USA).

Microencapsulated flavors and aromas have also been adapted for microwave and frozenfoods packaging containers. Yeo et al. (2005) encapsulated flavor oil in complex coacer-vates using gelatin and gum Arabic. The resultant microcapsules were incorporated intopackaging films used for frozen or baked foods such as breads, pastries, pizzas, and cookiesto improve their appeal and release the flavor oil of interest during heating.

Microencapsulated Pigments

Coloring agents containing natural or synthetic substances are commonly used as additivesin the manufacture of food products. Commercially available coloring agents can containsynthetic substances including dyes or azodyes or natural pigments. It is a well-knownproblem in the food industry that coloring agents tend to migrate within the food product orinto the environment of the product. This problem is particularly troublesome if it occurs infood products that comprise multiple, separated compartments or layers where the coloringagent is not added to all of such compartments. One typical class of such a compartmental-ized or layered food product is cakes and other desserts, which comprises at least one layer

Innovative Applications of Microencapsulation in Food Packaging 207

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Encapsulatedflavors releasedinto the air

Package

Encapsulated flavorsreleased into thepackage contents

Figure 9.3. Release of microencapsulated flavors from flavor-incorporated plastics.

Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 207

Page 214: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

of fruit filling, e.g., in the form of jelly, to which a coloring agent is added, and one or morelayers of other ingredients also having an aqueous phase to which the coloring agent is notadded. It is evident that migration of coloring agent into the non-colored layers results in ahighly unacceptable appearance of these layered products. Another important product inwhich the migration of coloring agent is not desirable is surimi. Surimi is a stabilized,myofibrillar protein system prepared from fish mince that has been washed with water andblended with cryoprotectants (Park and Morrissey, 2000). Surimi color is a very importantattribute to the product’s overall quality since it is commercially graded based on its color.Surimi loses its color even if it is stored at freezing temperature. Floros et al. (1997) dis-cussed the use of coloring agent–containing film in surimi systems as an alternative to tra-ditional direct color addition and to avoid migration of coloring agent from the film to theproduct. Shahidi and Pegg (1995) described a process in which the coloring agent wasencapsulated within a mixture of carbohydrate-based wall material, a binding agent, and areducing or sequestering agent to improve color stability of surimi as well as other meatproducts. The encapsulated pigment was reported to be effective by imparting the typicalcured color to frankfurters even after 18 months of storage. Popplewell and Porzio (2001)encapsulated various coloring agents in partially hydrogenated vegetable oil as a means ofincorporating them into edible coating for snacks, chicken legs, fish, and similar products.

In human nutrition, astaxanthin (reddish-orange pigment) has been gaining widespreadpopularity as a dietary supplement due to its powerful antioxidant properties. As mostcarotenoids, astaxanthin is a highly unsaturated molecule and thus can easily be degradedby thermal or oxidative processes during the manufacture and storage of foods. This degra-dation can cause the loss of their nutritive and biological value as well as production ofundesirable flavor or aroma compounds. Due to their intrinsic high instability, these com-pounds are not usually handled in their crystalline form, but rather as stabilized emulsionsor microcapsules. Higuera-Ciapara et al. (2002) microencapsulated synthetic astaxanthinin a chitosan matrix cross-linked with glutaraldehyde using the method of multiple emul-sion/solvent evaporation. A powdered product was obtained containing microcapsules witha diameter of 5–50 µ. Stability of the pigment in the microcapsules was studied under stor-age at 25°C, 35°C, and 45°C for 8 weeks by measuring isomerization and loss of concen-tration of the pigment. Results showed that the microencapsulated pigment did not undergoisomerization or other chemical degradation under the investigated storage conditions.

Microencapsulated-Inks and Time–Temperature Indicators

Sakojiri and Takahashi (1990) developed a multicolor imaging material that comprised asubstrate and a photosensitive layer. The latter consisted of a heat-meltable microcapsulelayer and a color forming layer comprising a diazonium compound and a basic substance. Theimaging material can be coated onto paper and polymeric films, and the final product can beused as a food packaging material. Tajiri et al. (1992) formulated microcapsules containingink for flexographic applications in which encapsulation of the ink ensures its adhesion andflowability. Resins used for this application consist of methacrylate or acrylate of molecularweight of 3000 up to 50,000 g/mol. The microcapsule containing ink compositions for flexo-graphic printing were described to be particularly useful for perfume ink compositions.

Chul et al. (2005) prepared an encapsulated color electronic ink by in situ polymeriza-tion utilizing urea/melanine and formaldehyde resin as wall materials. The electrophoretic

208 Chapter 9

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 208

Page 215: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

medium was made of two different types of white and colored pigment particles, bearingcharges of opposite polarity, in a colorless dielectric fluid. Optical contrast was achieved bymoving charged particles separately to the opposite electrode. White or colored particlesheld electrostatically to the common electrode depending on the electric field and theparticle charge. The white particles modified with polymethylmethacrylate copolymer andthe colored ones (magenta, yellow, and cyan) modified with wax material were found tohave superior affinity for the suspending medium.

More sophisticated applications of microencapsulated inks and coloring agents can befound in the area of time–temperature indicator applications. Because temperature abuse iscommon during storage, transportation, and handling, these indicators are designed tomonitor temperature abuses in the shelf-life of food products. Temperature abuse does notonly cause quality and nutritional losses, but also may lead to food poisoning and foodlosses (Ozdemir and Floros, 2004). In these systems, polymers that contain irreversiblethermochromic dyes change color in response to exposure to predetermined temperatureover time. These indicators can be formed into labels that can irreversibly change color andwarn the consumer when a product has been exposed to a temperature/time abuse. Success-ful time–temperature indicators must satisfy basic requirements to be effective as monitor-ing devices (Selman, 1995):

1. They must be easily activated and sensitive.2. They must provide high degree of accuracy and precision.3. They should have tamper-proof and should not be removed from the package.4. Response should be irreversible, reproducible, and should correlate with food quality

changes.5. Response should be easily readable and not be confusing.6. Physical and chemical characteristics of time–temperature indicators should be determined.

Recently, Avery Dennison Corp. (US) introduced a new time–temperature indicator inwhich TT Sensor™ is employed. The TT Sensor™ consists of two labels: an indicator labeland a transparent activator label. The activator label is applied to the indicator label and thenimmediately dispensed onto the package. Once the time–temperature indicator is activated,the indicator label immediately and irreversibly changes color. Activated labels function inthe temperature range from –18°C to 60°C. These indicators are claimed to be simple, reli-able, and cost-effective for monitoring time–temperature abuse that fresh foods are normallyexposed to. In addition, TT Sensor™ labels do not need to be refrigerated prior to applica-tion. Another time–temperature indicator that uses microencapsulation technology is calledThermax™. By measuring the change in temperature that is reflected in irreversible colorchanges, the latter indicator shows whether a food product was exposed to extreme tem-peratures, and this indicator is tamperproof. Similarly, Prusik et al. (2000) patented atime–temperature indicator label to measure the length of time to which a food product isexposed under temperature abuse conditions. The label contained a microencapsulated heat-fusible substance, which can melt and flow when a food product is exposed to temperaturesabove a predetermined level. The indicator can be activated by light finger pressure orpreferably by appropriate automated mechanical means to rupture the capsule containing theheat-fusible substance. A distinct color would develop upon contact of the dye precursor anddye activator due to the migration of the heat-fusible substance.

Innovative Applications of Microencapsulation in Food Packaging 209

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 209

Page 216: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Future Perspective

Microencapsulation has a promising future in food packaging. Attempts at microencapsu-lating flavors, antimicrobials, fragrances, coloring materials, and printing inks into foodpackaging materials will continue to increase in the next decade. There are significantopportunities in microencapsulation for food packaging, such as self adjusting sell-by-datethat senses when the consumer opens a packaged food and flashes if the food in the packageis spoiled or poses a health risk for the consumer. Another potential commercial applicationof microencapsulation in food packaging is package indicators that inform consumers ifthe package is disposable or not. This is especially useful for the elderly and children. Thenext step in encapsulation is the development of smart microcapsules that embody multiplemicro-compartments, each one dynamically interacting with the other, depending on thechanging environmental conditions. This is especially important to form packages thathave “self-pasteurizing” or “self-sterilizing” capabilities. In these systems, microencapsu-lated active agents within a polymer matrix will be released in a controlled manner depend-ing on the characteristics of the food such as microbial load, pH, and water as well as thechanging environmental conditions such as temperature, relative humidity, and so on toachieve a homogeneous pasteurization or sterilization within the package.

ReferencesAshcraft, C. R. and Wong, M. M. L. 1993. Flavor burst structure and method of making the same. US Patent No.

5,249,676. USA.Barabolak, R. M., Zibell, S. E., Witkewitz, D. L., and Greenberg, M. J. 2005. Method of controlling release of

antimicrobial agents in chewing gum. US Patent No. 6,955,827. USA.Barnes, A. C. and Cummings, S. G. 1987a. Cellular encapsulation of biological pesticides. US Patent No.

4,695,462. USA.Barnes, A. C. and Cummings, S. G. 1987b. Cellular encapsulation of pesticides produced by expression of het-

erologous genes. US Patent No. 4,695,455. USA.Barnes, A. C. and Edwards, D. L. 1989. Cellular encapsulation of biologicals for animal and human use. US Patent

No. 4,861,595. USA.Bok, S. H., Lee, H. W., Son, K. H., Kim, S. U., Lee, J. W., Kim, D.Y., and Kwon, Y. K. 1993. Process for preparing

coated microbial pesticides and pesticides produced therefrom. US Patent No. 5,273,749, USA.Brar, S. K., Verma, M., Tyagi, R. D., and Valero, J. R. 2006. Recent advances in downstream processing and for-

mulations of Bacillus thuringiensis based biopesticides. Process Biochemistry, 41(2): 323–342.Brody, A. L. 1992. Flavor, flavor everywhere-but in packaging? Cereal Foods World, 37(11): 834–835.Cannon, R. J. C. 1993. Prospects and progress for Bacillus thuringiensis based pesticides. Pesticide Science, 37:

331–335.Chacon, P. A., Buffo, R. A., and Holley, R. A. 2006. Inhibitory effects of microencapsulated allyl isothiocyanate

(AIT) against Escherichia coli O157:H7 in refrigerated, nitrogen packed, finely chopped beef. InternationalJournal of Food Microbiology, 107(3): 231–237.

Chul, A. K., Meyoung, J. J., Seong, D. A., Gi, H. K., Seung-Youl, K., In-Kyu, Y., Jiyoung, O., Hey, J. M., Kyu, H.B., and Kyung, S. S. 2005. Microcapsules as an electronic ink to fabricate color electrophoretic displays. Syn-thetic Metals, 151(3): 181–185.

El-Rehim, H. A. A., Hegazy, E. A., and El-Mohdy, H. L. A. 2005. Properties of polyacrylamide-based hydrogelsprepared by electron beam irradiation for possible use as bioactive controlled delivery matrices. Journal ofApplied Polymer Science, 98(3): 1262–1270.

Floros, J. D., Dock, L. L., and Han, J. H. 1997. Active packaging technologies and applications. Food, Cosmeticsand Drug Packaging, 20: 10–17.

Fraser, A. D. 1988. Package opening system. US Patent No. 4,720,423. USA.Higuera-Ciapara, I., Felix-Valenzuela, L., Goycoolea, F. M., and Argüelles-Monal, W. 2002. Microencapsulation

of astaxanthin in a chitosan matrix. Carbohydrate Polymers, 56(1): 41–45.

210 Chapter 9

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 210

Page 217: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Jones, L. R. and Hill, J. L. 1982. Composition for pest repellent receptacle. US Patent No. 4,320,112. USA.Klein, R. B., Selph, J. L., Partridge, J. J., and Reinhard, J. 2004. Compounds and methods for controlling fungi,

bacteria and insects. US Patent Application No. 20,040,235,898. USA.Lechelt-Kunze, C., Simon, J., Zitzmann, W., Kalbe, J., Muller, H. P., and Koch, R. 2000. Biological material

embedded in hydrogels, a process for the embedding thereof, and its use as artificial seed. US Patent No.6,164,012. USA.

Lee, S. J. and Rosenberg, M. 2000. Whey protein-based microcapsules prepared by double emulsification and heatgelation. Lebensmittel-Wissenschaft und Technologie, 33: 80–88.

Marrone, P. G. 1999. Microbial pesticides and natural products as alternatives. Outlook on Agriculture, 28(3):149–154.

Navarro, S., Finkelman, S., Zehavi, D., Dias, R., Angel, S., Mansur, F., and Rindner, M. 2005. Pest-imperviouspackaging material and pest-control composition. US Patent Application No. 20,050,208,157. USA.

Ozdemir, M. 1999. Antimicrobial Releasing Edible Whey Protein Films and Coatings. Ph.D. Dissertation. PurdueUniversity, West Lafayette, IN.

Ozdemir, M. and Floros, J. D. 2001. Analysis and modeling of potassium sorbate diffusion through edible wheyprotein films. Journal of Food Engineering, 47(2): 149–155.

Ozdemir, M. and Floros, J. D. 2003. Film composition effects on diffusion of potassium sorbate through whey pro-tein films. Journal of Food Science, 68: 511–516.

Ozdemir, M. and Floros, J. D. 2004. Active food packaging technologies. Critical Reviews in Food Science andNutrition, 44(3): 185–193.

Popplewell, L. M. and Porzio, M. A. 2001. Fat-coated encapsulation compositions and method for preparing thesame. US Patent No. 6,245,366. USA.

Prusik, T., Arnold, R. M., and Fields, S. C. 2000. Time-temperature indicator device and method of manufacture.US Patent No. 6,042,264. USA.

Radwan, M. N. and Allin, G. P. 1997. Controlled-release insect repellent device. US Patent No. 5,688,509. USA.Rieth, J. P., Wilson, W. T., and Levin, M. D., 1986. Repelling honeybees from insecticide-treated flowers with

2-heptanone. Journal of Apicultural Research, 25(2): 78–84.Rosenberg, M. 1997. Milk derived whey protein-based microencapsulating agents and a method of use. US Patent

No. 5,601,760. USA.Sakojiri, H. and Takahashi, H. 1990. Multicolor imaging material. US Patent No. 4,916,042. USA.Selman, J. D. 1995. Time-temperature indicators. In: Active Food Packaging. pp. 215–237. M. L. Rooney (Ed.).

Blackie Academic and Professional, London.Shahidi, F. and Pegg, R. B. 1995. Stabilized cooked cured-meat pigment. US Patent No. 5,425,956. USA.Spector, D. 1981. Self-stick aroma-dispensing tab. US Patent No. 4,277,024. USA.Sprinkel, Jr. F. M. and Newsome, R. W. 1988. Package with means for releasing aromatic substance on opening.

US Patent No. 4,717,017. USA.Sun, R., Quintus-Bosz, H., Given, P., Pineiro, R., and Morrison, A. 2000. Aroma release bottle and cap. US Patent

No. 6,102,224. USA.Tajiri, M., Wakata, K., Shinmitsu, K., and Shioi, S. 1992. Microcapsule-containing ink composition for flexo-

graphic printing. US Patent No. 5,120,360. USA.Thomas, T. R., Belias, W. P., Chen, P. N., and Kolovich, N. A. 2005a. Packages with active agents. US Patent

Application No. 20,050,220,375. USA.Thomas, T. R., Long, S. P., Belias, W. P., and Kolovich, N. A. 2005b. Packages with active agents. US Patent

Application No. 20,050,220,374. USA.Toreki, W., Manukian, A., and Strohschein, R. 2004. Hydrocapsules and method of preparation thereof. US Patent

No. 6,780,507. USA.Waters, J. R., Sharp, J. C., and Dev, V. J. 1994. Infection caused by Escherichia coli O157:H7 in Alberta, Canada,

and in Scotland: A five-year review, 1987–1991. Clinical Infectious Diseases, 19: 834–843.Wheeler, D. A. 2001. Surface coatings. US Patent No. 6,312,760. USA.Yeo, Y., Bellas, E., Firestone, W., Langer, R., and Kohane, D. S. 2005. Complex coacervates for thermally

sensitive controlled release of flavor compounds. Journal of Agricultural and Food Chemistry, 53(19):7518–7525.

Young, S. L., Sarada, X., and Rosenberg, M. 1993a. Microencapsulating properties of whey proteins. 1. Microen-capsulation of anhydrous milk fat. Journal of Dairy Science, 76: 2868–2877.

Young, S. L., Sarada, X., and Rosenberg, M. 1993b. Microencapsulating properties of whey proteins. 2. Combina-tion of whey proteins with carbohydrates. Journal of Dairy Science, 76: 2878–2885.

Innovative Applications of Microencapsulation in Food Packaging 211

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_09-201-212 r1.qxd 3/29/07 1:51 PM Page 211

Page 218: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

10 Marketing Perspective of EncapsulationTechnologies in Food Applications

Kathy Brownlie

Introduction

From being the enabling technology behind carbonless paper and “scratch and sniff ”fragrance sampling, microencapsulation technologies have found a broad range of indus-trial applications in markets as diverse as pesticides and cosmetics.

Microencapsulation has also found widespread use in the food and beverage industry.Flavorings that last longer, both in the mouth and on the shelf, fresher tasting flavoringswhich are clearly distinguishable from each other, innovative new flavor combinations, fla-vors which are released at the ideal time to provide maximum impact for the consumer, andeven products which initially taste of one thing, but then develop a totally different flavor asyou chew, all of these things are being achieved in food products through the innovative useof microencapsulation techniques.

Flavor microencapsulation, for example, entraps tiny volumes of the flavoring substancein a protective layer of another material, creating particles which are only in the micron oreven nano size range, but which can have a huge impact on the flavor profile of the finalproduct they are used in. This ability to isolate tiny amounts of a substance within a protec-tive wall or matrix has played a crucial role in the development of novel groundbreakingproducts, such as the temperature-regulating clothing sold by Outlast and Frisby Technolo-gies and the electronic displays being developed by E Ink.

The combination of a constant flow of innovative new techniques with wider applicationareas and an increased desire for product differentiation will continue to drive growth in theuse of microencapsulation technologies in various markets. But, technology providers mustchoose their end application market with care to ensure sustained growth. Growth rates inthe end-use markets range from negative figures to as much as 30 percent.

Several potentially high-volume applications of the technology are still in development,and the companies involved have strong intellectual property positions. In other markets,the end-product manufacturers perform most of the microencapsulation themselves andallow fewer opportunities for companies outside the industry. However, in areas such as thecosmetics and in some food ingredient markets, most of the microencapsulation is per-formed by smaller technology providers.

From a performance standpoint, encapsulation of active pharmaceutical ingredientsenables the formulator to design a release profile most appropriate for the drug. Thisincludes fast-dissolve formulas, extended release, targeted release, and delayed release.Microencapsulation technologies also enable taste masking of bitter compounds in chew-able tablets, as well as oral dosages that are taken without water.

In the pharmaceutical consumer product industries, oral drug delivery continues todominate the microencapsulated drug market. Although some of the microencapsulation

213

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 213

Encapsulation and Controlled Release: Technologies in Food SystemsEdited by Jamileh M. Lakkis

Copyright © 2007 by Blackwell Publishing

Page 219: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

technologies have existed for decades, the current trend is to explore drug delivery forincreasingly potent or insoluble new compounds or to add value to existing compounds.

Key Challenges

Identification of New Applications

Although the number of markets in which microencapsulation technology has been appliedis already large, the general feeling is that the surface of possible applications has only beenscratched. Companies offering this technology have to continue to invest in research andmonitor markets and emerging technologies where microencapsulation could add value toproducts or solve particular problems. For companies possessing in-house microencapsula-tion capabilities, internal communication between research departments is crucial in orderto fully utilize these resources. For companies offering microencapsulation services, thiscan be a much harder task. It is important for such companies to develop a widely recog-nized reputation as experts in this field, so that industry turns to them when projects whichmight benefit from this expertise arise. Developing close relationships with R&D depart-ments at various companies will be vital.

Raising Awareness of Technique Potential

Closely related to the identification of new applications for the technology is the educationof as wide a customer base as possible of the potential of microencapsulation technologies.The primary approach for achieving this within the industry appears to be simply talking topeople about the possibilities and gaining a reputation as an effective and innovativeprovider of such technology. This can be a laborious and slow process, so companies mayneed to be creative in the ways in which they market their capabilities in this field.

Extra Costs Associated with the Use of Microencapsulation

The design of a microencapsulation process for a substance, the equipment needed to applyit on an industrial scale, and subsequent production all add to the cost of products usingmicroencapsulated products. These issues tend to limit the use of microencapsulation tech-nology to few markets: (i) those with higher value products where the cost added bymicroencapsulation would have less impact, (ii) to those markets where microencapsula-tion is absolutely necessary, or (iii) to a few markets where economies of scale can beapplied, thus reducing the importance of the fixed costs associated with microencapsula-tion. In other markets, microencapsulation must be seen to provide a very clear added valueto the customer, so that they will be willing to pay a higher price. Although this might beclear to the company doing the encapsulation, it is often not easy to sell this to clients andmay take a lot of persuasion and increased sales costs in the process.

High Pace of Technical Innovation

Although many microencapsulation techniques have been around for a number of yearsnow, it is still a very active research area, in which there is a constant stream of new,improved techniques and consequently patent applications. In order for companies to offerthe latest, and best, solutions to industry problems using microencapsulation, and to be able

214 Chapter 10

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 214

Page 220: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

to react to new market opportunities, they need to keep track of new technical innovationsand to develop their own techniques as quickly as possible.

Client Communication

Whether the client be another company or simply another department in the same company,effective communication during a project to make use of microencapsulation technologiesis vital to the project’s success. This can often be no easy matter due to the complex scien-tific nature of many such projects. In addition, a high level of trust needs to be built betweenthe parties concerned, with what may be quite sensitive projects due to the high degree ofinnovation and possible high returns.

Realistic project goals need to be set, including highlighting technical limitations.A thorough initial screening of the technical possibilities is also vital. Care needs to bepaid to communication between technical and marketing departments, so that the resultingproducts are deemed marketable as well as technically innovative.

Scale-up of Processes to Manufacture High-Volume Products

The scale-up of any chemical process to an industrial level always presents new challenges.The physical properties of bulk reactions differ markedly from those present in a laboratoryscale reaction, adding a level of complexity, which needs to be taken into account duringthe design of the process. These scale-up challenges can be significant even for the simplestchemical processes; for microencapsulation, it is even more challenging due to the fact thatmost laboratory scale reactions are not completely understood. The commonly used state-ment that microencapsulation is as much an art as a science bears witness to the problemsthat are faced in scale-up, the major problem being the reproducibility of reactions to pro-duce a consistently good-quality encapsulation of the substance.

Technique Differentiation

Although most microencapsulation techniques can be related to a relatively small numberof basic principles, variations on these have resulted in a quite bewildering array of differ-ent techniques being available on the market. Whichever industry a company is targeting,a company with a particular proprietary technique faces the hard challenge of selling theadvantages of its own technique over others on the market. The best way to do this is toidentify the key defining characteristics of the technology, breaking this down to a few eas-ily recognizable advantages, and putting these to potential clients in a strong and clear mes-sage. Of course, different strategies will need to be employed for the different industries towhich the technique might be applicable. Certain factors may be particularly of interest inone market, and these should be identified and highlighted. The further sections of thischapter should provide a clear indication of what these might be in the markets covered.

Identification of High-Volume End Uses

Many microencapsulation applications are conducted on relatively small scales of perhapsa few tonnes or less. Whilst these may be of a high value, the high cost of developmentultimately reduces the profits available from such applications. It has been mentioned time

Marketing Perspective of Encapsulation Technologies 215

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 215

Page 221: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

and again to Frost and Sullivan during the course of the research that many companies arelooking for the next high-volume industrial use of microencapsulation technology, whichmight rival the volumes used in the carbonless paper industry. The most likely candidatesfor this would appear to be microencapsulated phase change materials or possibly elec-tronic ink applications if both of these markets expand to their full potential.

Identification of Technology Potential by Market Participants

Many companies have developed microencapsulation techniques or brought in technologyfor their own in-house applications. Often, this has been as far as those companies havetaken the process. Without continued R&D in the area, they may find that the techniquethey are applying is no longer the most suitable or has become uncompetitive compared tomicroencapsulated products produced by other companies.

As microencapsulation in many ways is still very high tech, quite expensive, and oftenrequires a lot of experience to perform, the ability to achieve is still of high value and maybe more marketable than some companies realize. It may be that they do not want to go tothe trouble or expense of outsourcing the technology. Such companies may also represent abusiness opportunity for specialist microencapsulators who might be able to demonstratethe cost benefits of a more modern technology, which could be licensed out.

Overview—Microencapsulated Food Ingredients

Microencapsulation of food ingredients is not a new concept. Earlier efforts to encapsulateflavors were based on spray drying using acacia gum as the coating materials. However, theever-increasing complexity of food products is continuing to drive research into novel anddifferent encapsulation techniques and processes. In particular, the rapid growth in functionalfood seen toward the end of the 1990s is continuing apace, and it is the unstable or unpalatablenature of many of the active ingredients used in these products, which will continue to openup new opportunities for the use of microencapsulation technologies in the food industry.

Microencapsulation of food ingredients is performed for a wide number of reasons,including improved substance stability, taste masking, ease of handling, and controlledrelease. Today, a wide range of food ingredients are microencapsulated using one techniqueor another. Table 10.1 lists examples of microencapsulated taste-masking solutions cur-rently on the market.

This chapter by no means provides an exhaustive reference to all the areas in the foodindustry to which microencapsulation is being applied, but will attempt to provide a flavorof a few of the most interesting areas, where high-value microencapsulation technologiesare being applied to bring improvements to the food products we eat.

Reasons for Encapsulation

As has been discussed by other authors in this book, the main reasons for the microencap-sulation of food ingredients can be outlined as follows:

1. Protection—This is surely the primary reason for the microencapsulation of food andfeed ingredients. Protection can be achieved from a wide variety of influences thatmight cause an ingredient to lose its functionality; this might be simply physical

216 Chapter 10

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 216

Page 222: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

processes such as heat, light, or moisture, which might degrade the ingredient before ithas time to act. It may also involve retarding interactions between substances in a givenfood formula, thus ensuring product’s acceptable shelf-life.

2. Controlled release—Microencapsulation can be used in a delivery capacity whereby aningredient is released at the required time and place. This could mean the release of aleavening agent at a certain temperature during baking, a flavor upon chewing, or a pro-biotic culture upon digestion in the small intestine.

3. Processability—Microencapsulation techniques can simply be used to allow easier han-dling of an ingredient during production of the product. The advantages of providinga dry powdered form of a flavor to a bakery that otherwise only uses powdered ingredi-ents are obvious.

4. Taste masking—Vitamins and minerals are increasingly being added to food to increaseactual or the consumer’s perceived health benefits of the product by the consumer. Manyof these ingredients are unpalatable, and their taste needs to be masked until they havepassed the taste buds and the mouth area. This reason for encapsulation is particularlyprevalent in the animal feed industry, where larger concentrations of such ingredientsare added in order to produce healthier and hence better yielding animals.

Modes of Release

If you have encapsulated an ingredient, you will want it to be released from that encapsu-lant in order for it to function within the food product at the desired time and place. Themost common modes of release are detailed below:

1. Thermal release—Whereby the encapsulant melts at a certain temperature, usually dur-ing cooking of the product, releasing the ingredient. By altering the type of coating andits thickness, it is possible to ensure release of an ingredient within a few degrees of therequired temperature.

2. Physical release—Requiring the physical breaking of the microcapsules; usually thismode of release is designed for ingredients that need to be released during chewing.Factors which can be altered to prolong or otherwise the release profile include the sizeof the capsules and the strength and flexibility of the coating.

Marketing Perspective of Encapsulation Technologies 217

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Table 10.1. Microencapsulated food ingredient market: Examples of companies offeringmicroencapsulation taste-masking solutions on the market (source: Frost and Sullivan)

Company Product/brand Specific application

Bio Dar Encapsulates Protected by masking bitterness Chewing gum applicationsof herbal flavors

Coating Place Barrier coatings Tailor-made maskingColorcon Opadry® Taste masking, pharmaceuticals

and foodsBASF Kollicoat® SR 30D Mainly pharmaceuticals with

food capabilitiesFuiz Technology Cerform and Shearform Taste masking using spun

sugars for food and pharmaceutical applications

Particle Dynamics Micromask Pharmaceutical and foods

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 217

Page 223: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

3. Dissolution—Most food products contain at least a small amount of water, which can beused to ensure the release of an ingredient enclosed in a water-soluble coating. Thechemistry of the coating can be designed so that this only occurs at a particular pH, tem-perature, or salt concentration.

Market Drivers

Examples of key market drivers for microencapsulated food ingredients include:

Increased Consumption of Processed Foods which Require Ingredient Stabilization

Sales of processed and pre-prepared foods, commonly known as de-cooking phenomenon,continue to increase. This essentially means that people are devoting less and less time tothe preparation of their food, and so are using less fresh ingredients and more that are pre-pared in some way to speed cooking. The increased processing that these products mayundergo, including pre-cooking and freeze–thaw cycles, can harm many ingredients in thefood products, resulting in lower quality for the consumer. Microencapsulation can protectmany of these ingredients during processing and then be designed so as to release themwhen needed, either during the final cooking process or upon consumption.

Rapid Growth in the Functional Food Market

Sales of functional foods have increased dramatically since the mid 1990s. This category isbest represented by probiotic milk drinks or vitamin fortified sports bars and cereals. Obvi-ously, such products need to contain some ingredients which will impart this benefit, andmany such ingredients are unstable or difficult to handle such as vitamins, minerals, aminoacids and herbal extracts.

Drive for Brand Differentiation Increases Call for Microencapsulated Ingredients

Food manufacturers are responding to more complex consumer requirements and increas-ing competition, particularly from the powerful supermarket’s own innovative brand prod-ucts and product lines. The use of ingredients or processes which impart some novelcharacteristic to a food product, such as a new taste or health benefit, is a key tool in sellingmore expensive branded products to consumers. Many such innovations are always copiedin some way by the supermarkets, creating a cycle of innovation, which can only increasethe use of such high-tech processes within the food industry.

Consumer Demand for Natural Food Products

The continuous string of public health scares surrounding the food industry, as well asgreater awareness among consumers of the benefits of a healthy diet, are driving thedemand for food products which are natural in origin. Many of these natural ingredients arehowever unstable to harsh environment of food processing and cooking, so in order to

218 Chapter 10

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 218

Page 224: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

ensure the survival of such ingredients through consumption, microencapsulation is beingused to protect them.

Microencapsulation Reduces Amount of Ingredient Required for Same Effect

Whilst many microencapsulation techniques are often considered too expensive for appli-cation to food ingredients, on some occasions, the use of such technology can actuallyresult in a cost saving. The protection of food ingredients until they are required can resultin the need for much less of that ingredient in the product, possibly producing significantsavings on raw materials.

Complexity of Technical Requirements Creates Opportunities for Specialist Companies

The complexities of encapsulation processes often mean that an entirely new procedureneeds to be developed for a given application or a new ingredient. Different problems mayalso call for the use of totally different technologies. Food ingredient manufacturers cannotpossibly have all the equipment and technical know-how in-house in order to cater for eachpossible new problem or innovation. This creates an increasing demand for contractmicroencapsulation services. Small contract companies have the flexibility to respond tocustomer needs. They will often produce very small volumes, which would not be econom-ically feasible for a larger company to do.

Tightening of Laws on Product Claims may Force Companies to Use Microencapsulation

The increasing focus on quality of food products, driven by consumer demand for moreinformation about the food they consume, will undoubtedly lead to tighter regulations gov-erning the product claims that food manufacturers make. At present, in many countries, it issimply necessary for companies to introduce an ingredient at the outset of the productionprocess, without ensuring that the stated level is actually present in the finished product.Often, sensitive ingredients such as some vitamins are destroyed by the cooking process,making claims for the extra health benefits of such added substances essentially false. Ifproducers have to ensure that the ingredients survive until consumption, the protection ofthese ingredients would become vital, leading to new opportunities for developing andusing novel microencapsulation technologies.

New Product Development Creates Future Markets

The development of an encapsulation process for a new ingredient or new purpose is oftenan expensive and time-consuming procedure, which will not be undertaken unless a decentreturn on that investment is expected. One of the best ways for companies providingmicroencapsulated food ingredients to identify where to focus their research is by under-taking contract work. Each new request, whether it results in an order or not, brings a newidea or indication of a market need, which can hopefully be exploited.

Marketing Perspective of Encapsulation Technologies 219

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 219

Page 225: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Market Restraints

Examples of the key market restraints for microencapsulated food ingredients include:

Cost of Many Microencapsulation Techniques Reduces Scope for Use in Food Products

The cost of most microencapsulation techniques is a serious constraint to the use of thistechnology in the food industry, where traditionally the end-user markets have been matureand price sensitive. An alternative scenario is its use in those situations where encapsula-tion is the most cost-effective solution to a particular problem.

Lack of Industry Awareness of Microencapsulation

It is still felt that many food processors are not aware of the possible advantages whichmicroencapsulated ingredients can bring to their products. Combined with this are miscon-ceptions about such things as the cost of these ingredients and concerns over the increasedlevel of processing which they might represent. The desired widespread knowledge of thesetechniques within the food industry will undoubtedly take a long time to build up and willlimit the wider use of the technology in the process.

Difficulties in Technical Communication with Customers

Related to the above constraint is a common complaint from many encapsulation special-ists that companies approaching them do not understand the nature of microencapsulation.Whilst they generally welcome any approaches for business, this is obviously a source offrustration within the industry, and the wasted time and effort can have financial implica-tions. In addition, if clients go away with a poor impression of microencapsulation due tounrealistic expectations, this will have an impact on the future market potential for suchtechnology. It is the encapsulation companies themselves, as providers of the service, whoneed to communicate effectively with their potential clients, so that realistic project objec-tives are agreed upon, which will result in satisfaction for both sides.

Limitations on Encapsulant Materials

There are a number of limitations on the materials which can be used to encapsulate activeswhich may ultimately lead to technical and supply problems:

BSE and Foot and Mouth

The Bovine spongiform encephalopathy (BSE) epizoolic has created a consumer confi-dence crisis in the safety of materials derived from animal sources. Products using encap-sulation are often functional foods bought by very health conscious consumers, who areaware of such issues. Thus, the presence of gelatin may cause concern among consumers.The recent foot and mouth disease epizoolic in the UK and the resulting ban on the exportof animal products also caused an increase in gelatin prices by as much as 20 percent.

220 Chapter 10

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 220

Page 226: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

GMO

Consumer demand has led to most food manufacturers in Europe, in particular, being verycareful not to use ingredients that might come from genetically modified organisms in theirproducts. This has affected supplies of certain materials such as maize-derived starchesfrom the US.

Approval of Ingredients

The European Union maintains a list of chemicals that have been approved for use in food.This may limit the development of novel processess that may require materials not on theapproved list.

Competitive Factors

One key competitive factor in this market is technology. The ability to consistently deliveradded value to food products through the use of a microencapsulation is vital for companiesusing this technology. It is not an easy process to sell simple products such as these at muchhigher prices than normal, so customers must be convinced that the microencapsulation isachieving the desired properties in their products. The ability to deliver something differentand superior in terms of performance is the key to attracting more customers in this market.

Another important competitive factor, as in the provision of microencapsulated productsin many other industries, is customer service. With most ingredients being designed forspecific applications, often at the request of customers, close attention to delivering on cus-tomer service and maintaining good relationships is important to maintain market share.

Another key competitive factor is technical expertise, as the microencapsulation has toachieve the desired result in the final product in order for customers to continue to payhigher prices for these products. Properties of the encapsulation are often tuned to the cus-tomer’s requirements for a particular application, so close attention to customer service isvery important to ensure the product is successful and repeat business is gained.

Microencapsulation of flavorings undoubtedly adds some cost to the production, whichis generally reflected in higher prices. Respondents were very unwilling to discuss whatactual price the microencapsulation added to a flavoring, mainly due to the fact that this canvary greatly according to the technique applied, the volume of product made, and the doserate of that flavoring. In addition, the added value of the microencapsulation was often feltto result in cost savings for customers due to lower wastage of the volatile flavorings andbetter impact in the product, resulting in the need to use lower volumes.

Microencapsulation techniques can result in price increases of 2 to 10 times the price ofa non-encapsulated flavoring. The prices for microencapsulated flavorings are not expectedto mirror this decline as the added value they bring often means that companies are notcompeting on price against similar products.

Pricing

The food industry tends to be price sensitive, so the extra cost associated with microencap-sulating an ingredient needs to be fully justified in terms of offering a clear improvement in

Marketing Perspective of Encapsulation Technologies 221

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 221

Page 227: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

performance of that ingredient within the food product. The application of microencapsula-tion is more easily justified with higher value ingredients such as flavorings, vitamins andminerals, and probiotics.

Price is an important competitive factor, for both non-encapsulated and microencapsu-lated ingredients. Higher prices of microencapsulated ingredients must be shown to be jus-tified by their better performance. There can also be significant price differences betweenmicroencapsulated products from different companies, and here also, improved perfor-mance must be demonstrated.

Microencapsulation is performed by both the original ingredient manufacturers andcompanies specializing in microencapsulation technologies. For the latter, it is imperativethat their technical expertise allows them to offer something unique in terms of perfor-mance. Their hardest task lies in convincing clients of this performance benefit in relationto the higher price of the ingredient. If their message is strong enough, they will face fewermarket barriers and lower competition. Companies need to work closely with clients toidentify the end-use products that microencapsulated ingredients could add benefit to anddesign suitable products to meet this need.

Industry Structure

Microencapsulation is performed by both the original ingredient manufacturers and com-panies specializing in microencapsulation technologies. Examples of companies amongstthe latter group include Balchem, Particle Dynamics, Bio Dar, and TasteTech. Most of themajor flavor houses are actively involved in research into the use of microencapsulationtechnologies, as are other major food ingredient manufacturers. At a very simplistic level,companies can be divided into two groups according to their capabilities of adding value totheir businesses. In practice, there is a significant amount of cross-over between these twodefinitions:

1. The first type includes companies that are usually small- or medium-sized enterprises forwhich the ability to do microencapsulation lies at the core of their business and providestheir main revenue stream. Such companies market their ability to perform microencap-sulation in a number of ways. Close cooperation with their customers is vital and caninvolve licensing a production process, toll manufacturing, co-development work, or saleof a bespoke product line. Such companies may also sell standard microencapsulatedproducts, or products in which microencapsulation is the core enabling technology.

2. The second type of companies are usually larger enterprises for which microencapsula-tion is simply another manufacturing technique they have at their disposal and that theycan use in one or more product lines. Such companies have usually developed or pur-chased microencapsulation capabilities for a particular in-house application, but havegone on to apply this knowledge in other areas of their business. They do not generallyoffer their microencapsulation capabilities to other companies. An exception to this trendis companies such as BASF and 3M. These enterprises have been offering their microen-capsulation capabilities to other companies in certain markets. Such companies and fewothers can really be considered providers of a wide range of microencapsulation expertise,which can be applied for virtually any company in any industry. Such companies specifi-cally advertise their ability to apply their techniques to long lists of substances. Examplesof these companies are South West Research Institute, Thies Technology, 3M and Ciba.

222 Chapter 10

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 222

Page 228: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Microencapsulators in Specific Industries

Examples include Coletica and Lipotec in the cosmetic industry. In reality such companies,though specialized in certain applications, are often looking for new business opportunitiesfor applying their techniques and would gladly try to adapt them to other industries. Cross-over capabilities are often seen in the cosmetic and food industries.

Companies for which Microencapsulation Performs a Vital Role in their Main Product Line

For carbonless paper manufacturers, microencapsulation is thus an inherent part of theirproduction lines. Although most companies originally licensed the technology from itsdevelopers, through using it for a number of years, they were able to have build up a highdegree of expertise in the area. Similar examples can be found in the textile industry.

Companies which Apply Microencapsulation to Certain Product Lines

Examples of this category are flavor suppliers that offer some of their flavors for certain enduses in microencapsulated form. Such companies usually bring the technology from an out-side source, but through running the processes for a number of years, they develop theirown expertise and even research capabilities. Table 10.2 gives examples of companies thatare involved in the provision of microencapsulation technologies to the food industry.

Balchem Encapsulates

Balchem Encapsulates is a wholly owned subsidiary of Balchem Corporation, which wasfounded in 1967 by the merger of several entities, including Dr Leslie Balassa who ownedseveral technology inventions in the field of encapsulation. It operates in two business seg-ments, Encapsulated Products and Specialty Products.

Marketing Perspective of Encapsulation Technologies 223

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Table 10.2. Examples of companies active in the food ingredientsmicroencapsulation market (source: various)

Company Website

Aveka www.aveka.comBalchem www.balchem.comBrace www.brace.comCoating Place www.encaps.comMicap www.micap.comParticle Coating Technologies www.pctusa.comParticle Dynamics www.particledynamics.comRonald T. Dodge Co. www.rtdodge.comSono-Tek www.sono-tek.comSouthwest Research Institute www.swri.comTasteTech, UK www.tastetech.co.ukThies Technology www.thiestechnology.com3M www.mmm.com

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 223

Page 229: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Balchem’s Encapsulated Products business utilizes proprietary microencapsulationtechnologies in an ever-expanding variety of applications, with food and feed ingredients atits core business. Its major product line ranges from ingredients for bakery products(Bakeshure), confectionary (Confecshure) and wellness products (Vitashure), as well asencapsulated flavors (Flavourshure) and meat-processing ingredients (Meatshure). Its ani-mal feed additives have the trade name Reashure. Balchem also offers partnerships and tollmanufacturing capabilities to solve particular problems faced by companies, which mightbe solved using its technologies.

Balchems’ acquisition of the encapsulation and agglomeration capabilities of LodersCroklaan in July 2005 underlines the ambition of this company to grow its core business.

Brace

The Germany-based company, Brace, specializes in laminar flow break microcapsuleforming. The firm holds a wide variety of patents worldwide. Examples of their productsinclude microcapsules of gelatin, alginate, and agar, filled with flavors for use in breathfreshening, chewing gum, and ready meal applications.

Karmat Coating Industries Ltd

Karmat is an Israeli company formed in 1993 and jointly owned by Kibbutz RamotMenashe and Coating Place Inc. Karmat’s microencapsulation techniques are mainly basedon fluid-bed technology, which employs an open air system allowing the use of many dif-ferent coating materials including modified starches, peptides, cellulose derivatives, fattyacids, and peptides. It applies these to the encapsulation of ingredients for the food, cos-metic, pharmaceutical, chemical, and feed industries.

Karmat’s main business is in the microencapsulation of vitamins and minerals, which itincorporates into bespoke premixes for its clients in the food industry. Most products it pro-duces are customized specifically for clients. Its premixes for the dairy sector are called Lac-tomix, those for baking applications, Bakeamix, and those for use in baby food, Babymix.Other product lines include microencapsulated citric acid, ascorbic acid, and ferrous sulfate.

Bio Dar

Bio Dar was founded in 1984 as an Israeli–American joint venture. In 1998, the companywas acquired by Lycored, a subsidiary of Makhteshim-Agan and part of the large Israeliholding group, Koor Industries. Bio Dar’s main products are microencapsulated vitaminsand minerals. It also manufactures microencapsulated nutraceuticals such as carnitine,amino acids, and herbal extracts. Over 50 percent of its products are customized to specificcustomer requirements.

Particle Dynamics

Particle Dynamics, Inc. (PDI) is an American-based company acquired in 1972 by KV Phar-maceutical company. PDI markets specialty raw materials for the pharmaceutical, nutri-tional, food, and personal care industries. It divides its business into three main technologylines; Destab, a direct compression technology used to make tablets for pharmaceutical and

224 Chapter 10

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 224

Page 230: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

nutritional supplement purposes, Descote®, a line of microencapsulated vitamins, minerals,and herbal extracts, and MicroMask which encompasses a series of technologies for tastemasking mainly in over-the-counter medicines.

PDI’s proprietary microencapsulation technology is based on the trapping of ingredientsin a matrix, in which particles of the ingredient are effectively embedded. The advantage ofthis technology lies mainly in its physical stability, being able to better withstand processessuch as tabletting. The company follows a lower risk business approach of modifying exist-ing compounds and formulations rather than discovering new molecular entities. ParticleDynamics serves the vitamin, food, and herbal supplement industries and is also involvedin pan coating.

Particle Coating Technologies

Particle Coating Technologies, Inc. (PCT) is a research and development company dedi-cated to microencapsulation technologies. Formerly part of the University of Washington,Department of Chemical Engineering in St. Louis, MO, the company became independentin 1994.

PCT pioneered a spinning-disk coating technique and also specializes in the formationof narrow particle-size distribution products, using a proprietary atomizer. PCT claimsmore than 20 of the world’s largest 100 public companies as customers and has some tollmanufacturing capabilities in addition to its core work in feasibility studies.

TasteTech

TasteTech is a privately owned British company founded in 1992 and currently distributesits ingredients worldwide. It utilizes its proprietary microencapsulation technology toencapsulate flavors, spice extracts, and key ingredients used in the food industry. It alsoapplies its technology in the pharmaceutical and cosmetic industries. TasteTech employsthree main microencapsulation technologies as well as spray drying capabilities. Its con-trolled release techniques are labeled CR100, CR200, and CR300 and are used to encapsu-late the ingredients mainly using vegetable fats and oils. The advantage of using theseencapsulant materials is the ability to control the release of the ingredients by adjusting themelting point of the fat coating.

Key End-User Groups

In terms of the microencapsulation technology itself, almost any company in the chemical,food, and related industries is a potential end user. The number of areas the techniques arebeing applied to continues to grow as more research is carried out and more projectsattempting to harness microencapsulation capabilities are undertaken.

Competitive Factors

Table 10.3 gives examples of some of the factors taken into consideration in the selection ofa microencapsulation technology.

For those companies marketing their ability to perform microencapsulation for othercompanies in any market, an important competitive factor is reputation. If a company is

Marketing Perspective of Encapsulation Technologies 225

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 225

Page 231: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

recognized as an expert in the field and has previously participated in a large number ofsuccessful projects, then approaches from clients are far more likely. Given the unpre-dictable, project-based nature of much of this business, such industry recognition is vital ifa company is to generate a steady stream of work in the area. Other activities which canraise a company’s profile in this way might be to hold as many patents as possible in thearea concerned or to regularly publish scientific papers on the subject and give talks atconferences.

For any company using microencapsulation, innovative ways of applying the technologyare likely ultimately to lead to greater profitability. If the technology can be applied to givea product a competitive edge or to introduce an entirely new concept to the market, highrates of growth in that product may well result.

Examples of Microencapsulation in the Food Ingredient Industry

This section reviews the use of microencapsulation technology in the flavors, vitamins,salts and acids, and probiotics markets. In these markets, the use of microencapsulation hasmoved beyond basic coating for handling purposes toward its use for stability purposes andmore importantly, controlled release of the encapsulated substance.

The largest market using microencapsulation technologies is that for flavorings. Accord-ing to Frost and Sullivan (Oxford, UK), microencapsulation of flavors in the European mar-ket was valued at $340 million in 2001.

Other market sizes and growth rates vary widely, depending on the food ingredientsconcerned, but overall growth in the use of microencapsulation is undoubtedly being expe-rienced as more food and beverage manufacturers begin to recognize the benefits that someof these technologies can bring. The two major growth areas will continue to be in the func-tional food sector, where microencapsulation can be used to stabilize ingredients such asvitamins and minerals, and elsewhere in techniques which offer controlled release of ingre-dients. Both these markets are expected to see double-digit growth in the next few years.

The increased processing of foods, coupled with the desire for increased quality andinnovation, is driving growth in the use of techniques which can effectively stabilize sensi-tive ingredients.

226 Chapter 10

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Table 10.3. Microencapsulated food ingredient market: Important factors to consider (source: Frost and Sullivan)

Issue Questions Importance

Function Primarily taste-masking is the Vital functionraison d’etre of microencapsulation

Cost What is my budget? Is the technology Need to weigh effectiveness, cost-effective? new methods are expensive!

Size How small do my encapsulated Important for mouthfeel, taste,ingredients need to be? and effectiveness

Production/ Will the encapsulated ingredient Microencapsulation is ineffectiveconditions survive processing and storage? unless it can protect the ingredient

from processing and storageEffects How will the encapsulated ingredient Unintentional interactions

interact with other ingredients can ruin a productin the finished product?

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 226

Page 232: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Microencapsulated Flavors Market

Flavor is an essential part of our experience when eating food. Four basic flavor types havebeen identified: sweet, sour, bitter, and salty, with a fifth defined by the Japanese as Umami,a savory flavor. Obviously, many natural foods have their own inherent flavors, but today’scomplex food products often require the addition of one or more flavors in order to satisfythe consumer. The major areas of flavoring use are in dairy foods, confectionary, beverages,bakery foods, and other savory foods. Flavors can be derived by physical extraction fromnatural sources or synthesized.

Why Microencapsulate Flavors?

Microencapsulation can help protect flavors from the rigors of harsh production processesand in providing a much high sensory impact in the final product. In addition, a more natu-ral flavor can be provided by protecting sensitive extracts and further releasing them at thetime of consumption.

Finally, flavor manufacturers are simply looking to offer innovative new productsto their customers. The protection provided by microencapsulation can be harnessed toallow the use of certain flavors or flavor combinations which were not previously possiblein certain products. As discussed earlier, microencapsulating flavors can help in variousways:

1. Protection from degradation.2. Controlled release at a desired time and rate.3. Stabilization and shelf-life extension.

End-Use Applications

The segmentation of market revenues for microencapsulated flavors does not necessarilyfollow that of the total flavor market due to certain end-use requirements. The major end-use markets for microencapsulated flavorings are listed below:

1. Confectionery products—sustained release of flavorings is often required to add valueto the products. The most common type of confectionary that use microencapsulatedflavorings is chewing gums. Confectionery is estimated to account for 35 percent ofEuropean microencapsulated flavorings market revenues.

2. Bakery products—dry powdered flavorings are required for easier mixing with otherpowdered ingredients. Controlled release of flavors in baking is also a growing featureof baked goods.

3. Powdered beverages—encapsulated dry powders are necessary for better mixing withother dry ingredients of the beverage components and for enhanced shelf life of the drymix. Release is achieved through dissolution of the coating. Spray dried flavors arewidely used in this market for ease of handling and instant solubility.

4. Processed foods—all the reasons for microencapsulating flavors mentioned above areapplicable to its application in processed foods. More advanced microencapsulationtechniques are being used in this market sector to address a range of shelf-life and con-trolled release issues.

Marketing Perspective of Encapsulation Technologies 227

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 227

Page 233: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Microencapsulation is a very active area of research at most of the major flavor houses.These companies have large R&D budgets needed to invest in the development of microen-capsulation expertise, which can take a lot of practice to perfect. Such a quality-driven mar-ket sector also presents opportunities for smaller technology providers. Although profitmargins from partnering may be lower than selling your own products, access to the largemarketing and sales forces of the industry leaders will maximize the sales potential of theproduct, which is something that smaller companies can struggle to achieve on limitedbudgets.

Examples of Market Drivers and Restraints in the Flavors Industry

Product innovation gives market advantage. Innovative new food products which capturethe imagination of the public give food manufacturers a competitive edge in the competi-tive food market. Such innovation within the food industry is a major driver of the use ofmicroencapsulated flavors, which can add an extra dimension to a food product. Possibili-ties such as sequential release of flavors, long-lasting flavor effect, or simply the ability tointroduce certain flavors into a new food area can all help to increase the appeal of a foodproduct and help it to capture market share.

Increasing use of processed foods. Sales of processed and pre-prepared foods con-tinue to increase as the phenomenon known as de-cooking increases. Such systems oftenrequire flavors to be protected from the processing they undergo in order that they can bedelivered to the consumer when the product is eaten.

Consumers demand better food quality. In conjunction with the greater consumptionof processed foods, consumers are demanding greater quality from these food products.One particular quality demand is that for more authentic or natural tastes. Microencapsula-tion can be used to deliver individual, authentic tasting flavors at exactly the right time,resulting in greater customer satisfaction with the product.

Examples of market constraints in the flavors industry

Drive for lower process costs. Although microencapsulated flavorings cost more thanthose which are not encapsulated, they can offer cost savings in other ways. Microencapsu-lation of flavors into dry powders can help in this regard. Less expensive equipment mightbe needed and production steps removed due to the easy mixing of different powders ratherthan oil inclusion. In addition, less flavor is likely to be released to the atmosphere duringprocessing, resulting in the need to use less of the product to achieve the same effect.

Environmental problems with spray drying lead producers to search for alternativetechnologies. The use of spray drying, where a fine mist of flavor is heated will inevitablylead to the escape of some of these volatile compounds into the atmosphere, which canrequire the use of expensive scrubbers to control. Even then, certain flavors such as onionor cheese can be particularly troublesome. Companies using spray drying for flavors aremonitored by environmental officials at a national level, who have the ability to imposeinjunctions stopping the use of the equipment for particular flavorings. This has led produc-ers to look to other microencapsulation techniques for flavor applications. It has been sug-gested to Frost and Sullivan that if another suitable technique were available to encapsulateflavorings in water dispersible coatings, it would be take a large share of the market fromspray dryers.

228 Chapter 10

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 228

Page 234: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Consumer and legislative restraints on encapsulant materials. The European approvedingredient list contains details of those substances which are allowed for use as food ingredi-ents and broadly in which applications they can be used in. Such regulatory constraints pres-ent major challenges mainly in limiting the number of materials available, their usage inspecific systems, and sometimes at low concentrations that may render them ineffective.Consumer preference can also limit the types of encapsulating material used. A particularexample is the movement against genetically modified materials.

Microencapsulated Vitamins Market

Vitamins are essential parts of the human diet, without which we are prone to a large num-ber of vitamin deficiency-related diseases and general ill health. Traditionally, we havederived these essential nutrients by consuming a varied diet of natural foods abundant inparticular vitamins, such as many fruits and vegetables. The techniques most widely prac-ticed are physical processes such as spray drying, spray cooling, and fluid-bed techniques.Revenues indicated in this document refer to all coated vitamins.

Why Microencapsulate Vitamins?

The major reasons for microencapsulating vitamins for use in food products are detailedbelow. In addition to processability and stability issues discussed above for encapsulatingflavors, most vitamins have objectionable taste that needs to be masked so that the activecan only be released in the gut. The largest volume of vitamins sold in microencapsulatedform include the fat- and water-soluble types such as A, D and E, K1, and the B-group(notorious for medicinal taste).

Vitamins in Animal Feed

Vitamins are also microencapsulated for use in animal feed products for both taste-maskingand bioavailability reasons. The latter is particularly an issue for ruminants, where rumenby-pass products can utilize microencapsulation to ensure that vitamins reach the second gutand intestines in their intact form.

Microencapsulated Salts and Acids Market

A variety of other food ingredients have been microencapsulated using mainly fluid-bedtechniques; the most common are salts (sodium chloride, sodium bicarbonate, and sodiumdiacetate) and acids (citric, malic, sorbic, tartaric, ascorbic, acetic, fumaric, etc.).

These ingredients are encapsulated for a number of reasons such as controlled release,extended shelf-life, prevention of color degradation, and protection against moisture. Theirmain end-use applications include confectionery, bakery, and processed meat products.

Microencapsulated Probiotics Market

Probiotics are bacteria, yeasts, or other microorganisms which provide health benefits bycontributing to intestinal microbial balance. These live cultures of microorganisms areincluded in a number of food products and supplements and are generally intended to

Marketing Perspective of Encapsulation Technologies 229

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 229

Page 235: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

colonize the large intestine, where they impart such benefits as impeding harmful bacterialgrowth, supporting the immune response, increasing the nutrient absorption, and reducingthe risk of cancer.

Probiotics have been included in a number of food products, which can be broadly clas-sified into two groups—dairy products and other food supplements. Dairy products includeyoghurt, sour cream, cottage cheese, ice cream, and fermented milk drinks. Other dietarysupplements can be capsules, powders, or tablets.

Why Use Microencapsulated Probiotics?

One important constraint in the probiotic market is the technical challenges associated withworking with live cultures of delicate microorganisms. The bacteria are often sensitive tothe conditions found in food products or encountered during processing such as moisture,temperature, oxygen, or pressure. Microencapsulation can protect the bacteria against theseconditions as well as control their delivery.

It is not expected that microencapsulated probiotics will cannibalize existing probioticmarkets, but they will instead open up new markets, either through the inclusion of probioticsin different food products and supplement forms or through the introduction of previouslyunavailable bacterial strains. The food supplement market will be the first to benefit, withimproved probiotic survival rates during tabletting a particularly attractive proposition. Twomain challenges can be faced in this application:

1. Higher price, which may be two or three times that for non-encapsulated bacteria. Thisis undoubtedly the main reason why the microencapsulated products will not affect salesof non-encapsulated ones, where they simply could not compete. However, for thosebacteria which cannot be included in food products in any other way, price is notexpected to be a major constraint. In fact, increasing survival rates of the bacteria wouldreduce the need for overdosing, potentially offering savings.

2. Product development time: It is likely that each new bacterial strain to be microencapsu-lated will need different conditions of encapsulation and will thus pose its own technicalchallenges. This will undoubtedly slow up the time to market for a wider range of poten-tial microencapsulated probiotic products. Table 10.4 gives examples of companies thathave developed microencapsulated probiotic products.

230 Chapter 10

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Table 10.4. Microencapsulated food ingredients market: Examples of microencapsulation of probiotic bacteria (source: Frost and Sullivan)

Company/group Product/brand Specific application

Institut Roselle Probiocap Guarantees probiotic stability and survival in cereal bars and yogurt

Advanced Bionutrition MicroMatrix® Allows continuous encapsulation of functional foods

Rhodia FloraFit® Designed for supplements and nutritional additives

University of Maryland Xanthan-chitosan Protects prebiotic coacervation products at 0–60°C

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 230

Page 236: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Examples of Strategic Recommendations for Companies Specializing in Microencapsulation Technologies

Indeed, changing consumer trends and tastes are primarily responsible for driving innova-tion in the microencapsulation market. Since food manufacturers constantly monitor suchtrends, food ingredient companies are always looking for ways to meet these ever-changingdemands, thereby promoting the need for novel microencapsulation technologies.

With consumers showing a growing preference for functional food, which now accountsfor a substantial chunk of the global nutrition market, food companies are looking for dif-ferent ways to incorporate health-promoting ingredients that deliver some kind of healthbenefit to the consumer.

Business Promotion Strategies

For companies wishing to promote their microencapsulation capabilities, the promotionof these probably represents the major challenge to the success of the business. Potentialclients need be made aware of situations where microencapsulation could add value toa product or solve certain commonly recurring problems. The main method currentlyemployed to get this message across is to personally talk to clients, either through a site visitor at trade shows. Once projects have been undertaken, personal relationships with researchdepartments at clients can hopefully be built, which might result in further businessopportunities.

To initially catch the attention of potential clients, examples of successful projects pro-ducing interesting and groundbreaking results or simply commercially successful productsshould be emphasized. Technical expertise should be stressed through highlighting aca-demic publications and patents held. Another method of business promotion is to take partin interesting projects, which receive press coverage and indirectly promote both microen-capsulation technology and the company involved in the project.

One way to spread the cost of this would be setting up an industry association represent-ing all companies involved in the market and who aim at promoting the potential benefits ofthe technology. There is presently an International Microencapsulation Society, but this isaimed more at academic research than industrial issues. The European Commission hasalso set up a thematic network entitled “Microencapsulation for Low Cost, High Volume,Pharmaceutical Applications,” intended to provide a guidance manual for European Indus-try indicating which technique will be most suitable for a particular application. Such aproject is exactly what is needed by the industry and could well prove to be the catalyst forsignificant market growth.

Product Proposal Strategies

Maximize Focus on a Single Market

Most companies offering microencapsulation technologies start out by applying them to aparticular market area, this usually being the one for which the particular technique turnsout to be most suitable. Such a focus is important for these smaller entrepreneurial compa-nies initially. Trying to sell the technology to a range of industries would simply dilute themarketing message and prove very difficult for a small company to achieve effectively.Focusing on a single market allows limited funds to be concentrated on a particular

Marketing Perspective of Encapsulation Technologies 231

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 231

Page 237: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

customer group, allowing a company to build a reputation in that industry as a reliabletechnology provider more quickly.

Application of Techniques Across Different Markets

Whilst the above approach is important initially, the ability to apply microencapsulationtechniques to areas other than those where they might first have been used can be veryimportant to the long-term growth. High growth markets using the technology may turn outto be part of a trend, which fade away as quickly as they arrived, so companies need to haveother areas to fall back on if this occurs. Particularly susceptible to this are consumer mar-kets, such as the cosmetic area, where trends toward certain ingredients or product typesmove rapidly.

Offer of a Diverse Range of Encapsulation Techniques

For companies looking to do business through their ability to do microencapsulation, it isimportant to be able to offer a broad range of possible solutions to a particular encapsula-tion problem. This will give such companies the ability to serve as many clients as possible.Companies such as South West Research Institute and Thies Technology in the US haveexpertise in many different encapsulation techniques, making the chances of a companyfinding a satisfactory solution more likely. Even for companies focusing mainly on onemarket, a greater number of product lines again increase the chances of a successful matchwith a client’s product and should result in more repeat business.

Examples of Strategic Recommendations for Larger Companies Using Microencapsulation Technologies In-House

Cross-Fertilization of Technology within the Organization

The number of industries, products, and substances to which microencapsulation can beapplied is virtually limitless. The broad applicability of encapsulation technologies shouldbe pursued by larger companies, making sure that research scientists in all business seg-ments are aware of the techniques and what they might be able to achieve. Companieswhich have clearly benefited from such an approach are 3M and BASF. 3M has long beenrecognized as the leading large industrial user of microencapsulation technologies.Although originally translated to such applications as scratch and sniff fragrance sampling,these technologies are now an inherent expertise within 3M and have been applied todeliver greater product performance and to develop innovative new product lines in a vari-ety of industries including pheremones, oil extraction chemistry, and adhesives.

BASF first developed microencapsulation expertise for use in the carbonless paper mar-ket, in which it still licenses the technology to a number of major paper manufacturers.It has applied various microencapsulation techniques in most of the industries covered bythis chapter including its pesticide formulations, cosmetic ingredients, and food ingredi-ents. Its integrated structure has allowed this cross-fertilization of the technology, which isnow being used in unexpected applications such as building insulation via phase changeencapsulation technology.

232 Chapter 10

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 232

Page 238: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Offer of Technical Expertise to Other Organizations

As has been said time and again about microencapsulation, it is considered as much of anart as a science, so there is no substitute for experience in using the techniques reliably to beable to consistently provide high-quality products. Such experience, especially in the appli-cation of techniques to industrial scale production, is a valuable asset and one which com-panies may be able to directly derive revenues from. A microencapsulation service businesscould be set up by combining the experience a company has in applying a number of tech-niques in a range of different sectors and offering them to industry at large.

Despite the successful approach of 3M in lending its technologies to other industriessuch as agrochemicals, cosmetics, food and pharmaceuticals, it did however spin-off someof the business resulting partly in the formation of the company Aveka in 1994, which pro-vides microencapsulation and particle processing to industrial clients. Another large com-pany which has formed a microencapsulation business is Ciba. Ciba offers its expertise in arange of different microencapsulation technologies to other companies and is looking toapply its expertise in particular to high-volume industrial uses.

Marketing Perspective of Encapsulation Technologies 233

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Ch_10_213-234 r1.qxd 3/29/07 1:25 PM Page 233

Page 239: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

235

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Index

Active transport, 179Actives in food matrices, entrapment of

amorphous matrix, 3–5complexation into cyclodextrins, 5–7cross-linked or coacervated polymers, 8emulsions and micellar systems, 7fat- or wax-based matrices, 7hydrogel matrices, 8–9microporous matrices, 7

Alginates, 89polycation-coated, 90prebiotics-coated, 90–91

Allyl isothiocyanate, 210–211Antimicrobial agents, 129–130Aspartame, 184

Balchem Encapsulates, 231–232Beeswax, 119Bifidobacteria, 92Bio Dar, 232Bioadhesive devices, 192–194Bioerodible devices, 192–194Brace, 232

Candelilla wax, 120Carbohydrates

encapsulation in amorphous matrices, 3as wall-forming material, 2

Carnauba wax, 119Cellulose acetate phthalate, 91–92Chemical leaveners, 127–129Chewable tablets, 196Chewing gums, 181

as delivery systems for oral health,188–189

composition, 182–183

for delivering acetylsalicylicacid, 189

for delivering antimicrobial agents,187–188

for delivering caffeine, 185–187for delivering flavors and nonmedicated

actives, 183–185for delivering vitamins, 187manufacture, 183vs. lozenge delivery profile, 189–190

Chitosan, 92Coacervate phase, 150–151Coacervation, 8Co-crystallization, 5Coenzyme Q10, 31–34Complex coacervation, 150–151

cross-linking of gelatin-based coacervatecapsule shells, 160–162

encapsulation process, 157–160encapsulation technology issues,

162–165properties, 151–157solvent exchange, 165–167

Confectionery products as deliverysystems, 181

Controlled release systems, complexcoacervate-based, 136–139

Controlled release, 1Core materials, 2Cosolubilization effect, 18Covalent bridging, 45Cross-linked or coacervated polymers,

encapsulation in, 8Cyclodextrins, 5

guest molecules and, 6molecules, 6–7type and degree of complexation, 6

Lakkis_Index_235-242 r1.qxd 3/30/07 4:09 PM Page 235

Encapsulation and Controlled Release: Technologies in Food SystemsEdited by Jamileh M. Lakkis

Copyright © 2007 by Blackwell Publishing

Page 240: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Dairy products (probiotic survival)cheese, 93–94frozen dairy desserts, 94yogurt, 94

Dietary fats delivery as o/w emulsion, 65–69Dilution lines, 17Dissolution, 225Double emulsions, 55

general applications of w/o/w emulsions, 58taste control using w/o/w emulsions, 58–59transport and release mechanism of

water-soluble components, 57–58w/o/w emulsion instability, 56–57w/o/w emulsion production, 56

Dough conditioners, 129Droplet size distribution, 17, 18Drug delivery via oral route

advantages, 180buccal delivery, 178disadvantages, 180–181local oropharyngeal delivery, 178periodontal delivery, 178sublingual delivery, 178

Drugs, transport mechanisms ofactive transport, 179endocytosis, 180facilitated diffusion, 179passive diffusion, 179

Duplex emulsions. See Double emulsions

Effervescent tablets, 196Electrostatic bridging, 45Emulsion stabilization, 42–43

coalescence, 46creaming/sedimentation, 43–44flocculation, 44–46Ostwald ripening, 47

Emulsionsdelivery of hydrophobic food actives,

59–65delivery of water-soluble food actives,

53–59dietary fats delivery, 65–69formulation design for food, 47–52future trends, 69–73release triggers, 52–53stabilization, 42–47

Emulsions and micellar systems, encapsulation in, 7–8

Emulsions systems, 41–42Encapsulants. See Core materialsEncapsulated particles (bakery applications),

properties foradhesion and cohesiveness, 125film thickness, 125flexibility, 124good barrier properties, 124mechanical strength, 125melting properties, 125particle size distribution, 125surface morphology, 125

Encapsulated probiotics in dairy products(practical applications)

cheese, 106frozen desserts, 106–107yogurt, 106

Encapsulating agents. See Wall-formingmaterials

Encapsulation, 83–84, 135Encapsulation, reasons for

controlled release, 225processability, 225protection, 224–225taste masking, 225

Encapsulation and controlled release in bakeryapplications

antimicrobial agents, 129–130chemical leaveners, 127–129dough conditioners, 129flavors, 130sweeteners, 130yeasts, 125–127

Encapsulation processextrusion, 3–5, 84–85spray drying, 3–4, 5, 84

Encapsulation technologies for bakeryapplications

high-pressure congealing, 117–118hot melt particle-coating technology,

113–115spray congealing, 116–117

Encapsulation technologies in foodapplications (marketing perspective)

business promotion strategies, 239

236 Index

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Index_235-242 r1.qxd 3/30/07 4:09 PM Page 236

Page 241: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

client communication, 223costs associated with use of

microencapsulation, 222cross-fertilization of technology within the

organization, 240identification of high-volume end uses,

223–224identification of technology potential, 224manufacturing high-volume products, 223new applications identification, 222product proposal strategies, 239–240technical expertise to other organizations, 241technical innovation, 222–223technique differentiation, 223technique potential awareness, 222

Endocytosis, 180Enzymatic covalent cross-linking, 45–46Enzymes, encapsulation and controlled

release of food, 139–140. See alsoPhytochemicals

Extrusion, 3–5

Facilitated diffusion, 179Fat- or wax-based matrices, encapsulation in, 7Fats and glycerides

lauric acid group, 120–121oleic/linoleic acid group, 121palmitic acid group, 121

Flavor encapsulation, 149–150. See alsoComplex coacervation

Flavor-loaded microcapsules, 149–150Flavors, 130Flocculation, 44

bridging, 45–46depletion, 44

Fluid bed coating, 113–116Food actives – delivery via emulsions

double emulsions for controllingwater-soluble actives, 55–59

effect of o/w emulsions on taste release andperception, 53–55

water-in-oil emulsions for controllingwater-soluble actives, 53

Food emulsions, formulation design for, 47–48aqueous phase design, 48–49choice of lipid phase, 49–50interfacial formulation and design, 50–52

Food packaging, microencapsulation infuture trends, 218microencapsulated actives for packaging

applications, 210–215microencapsulated pigments,

215–217Freeze drying, 5

Gellan gum, 91Genetic Algorithms, 104

High-pressure congealing, 117–118Hot melt coating (film-forming materials),

118–119fats and glycerides, 120–121glycol polymers, 120resins and rosins, 120waxes, 119–120

Hot melt extrusion, 4Hydrogel matrices, encapsulation in, 8–9Hydrophobic food actives – delivery via

o/w emulsionsaroma release, 60–63lipophilic health ingredients, 59–60structured emulsions in hydrogels for aroma

release, 63–65Hydrophobicity, 122

Incomplete surface coverage bridging, 45

Karmat Coating Industries Ltd., 232κ-Carrageenan and locust bean gum, 91

Lozenges, 190as delivery systems for dry mouth relief,

191–192as delivery systems for teeth

remineralization actives, 192for delivering flavors and sensates, 190for delivering throat relief actives, 191vs. chewing gum delivery profile,

189–190Lutein and leutin ester, 27

bioavailability, 27role in age-related-macular degeneration,

26–27solubilization, 29

Index 237

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Index_235-242 r1.qxd 3/30/07 4:09 PM Page 237

Page 242: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Lycopene, 19bioavalaibility, 20formulation as additive in food systems, 20hydrophilic–lipophilic balance of

surfactant, 23–24solubilization capacity, 20–23

Micellar systems, encapsulation in, 7–8Microcapsules, manufacturing of, 4–5Microemulsion, 13, 30

formulation and characterization, 14–15industrial applications, 13–14phase diagram, 13–14

Microencapsulated actives for packagingapplications

antimicrobial food packaging materials,210–212

insect and/or rodent repellent foodpackages, 212–213

scented fragrance-inserts and flavor-releasing systems, 213–215

Microencapsulated flavors market, 235–237Microencapsulated food ingredients, 224

competitive factors, 233–234industry structure, 230–233key end-user groups, 233market drivers, 226–227market restraints, 228–229pricing, 229–230

Microencapsulated pigments, 215–216inks and time–temperature indicators,

216–217Microencapsulated probiotics market,

237–238Microencapsulated salts and acids market, 237Microencapsulated vitamins market, 237Microporous matrices, encapsulation in, 7Molecular sieve, 7Monodispersed emulsions, 73Mucoadhesion, 192

Nanoemulsions, 143–144Nanoparticles, 144Nature-made emulsions

oil or lipid bodies, 69–71plant cells, 72yeast cells, 71–72

Nonsoluble nutraceuticals, solubilization of,17–19

lutein and lutein ester, 26–28lycopene, 19–24phytosterols, 24–26vitamin E, 28–30

Nutraceuticals in nanosized self-assembledliquid (NSSL) vehicles, 13–17

bioavailability, 31–34oxidative stability, 30–31solubilization of, 15solubilization of nonsoluble.

See Nonsoluble nutraceuticals,solubilization of

U-type microemulsions, swollen micelles,and progressive and full dilution, 17

water binding, 34–35

Oil-in-water microemulsions, 18Oral cavity. See also Drug delivery via oral

route; Oral transport routes,physiological and structural basis of

division, 172esophagus, 173–174permeability and barrier functions, 174tongue, 173trigeminal nerve, 174

Oral delivery routeslocal, 172, 181systemic, 172, 181

Oral transport routes, physiological andstructural basis of

epithelial membranes, 175keratinization, 177membrane coating granules, 177oral mucosa, 175pH, 178plasma membranes, 174–175polarity, 177–178saliva, 176–177

Paraffin wax, 119Particle Coating Technologies, 233Particle Dynamics, Inc., 232–233Passive diffusion, 179Payload, 2Phase diagram, 13–14, 16

238 Index

12345678910111213141516171819202122232425262728293031323334353637383940414243444546S47N

Lakkis_Index_235-242 r1.qxd 3/30/07 4:09 PM Page 238

Page 243: Encapsulation and Controlled Release Technologies in Food Systems, 0813828554

Physical release, 225Phytochemicals

bioconjugation, 144encapsulation and controlled release,

140–143encapsulation by nanoemulsion, 143–144

Phytosterolschemical structure, 24–25solubilization capacity, 25–26

Plasticizers, 123Pressed tablets, 196Probiotic capsules, manufacturing

conditions forencapsulating according to experimental

design, 98–99optimal manufacturing conditions

verification, 105optimization using genetic algorithms,

104–105optimization using SDP technique, 101–104performing optimization, 101response surface models and optimization

model formulation, 99–101screening experiments and experimental

design, 96–98Probiotic encapsulation techniques in dairy

productsadvantages and disadvantages, 87–89emulsion, 85–87extrusion, 84–85spray-drying, 84

Probiotic survival (effect of encapsulation) indairy products

effect of carrier matrix, 89–92effect of spray drying, 92–93in dairy products, 93–94in gastrointestinal conditions, 94–96

Probiotics, 83Protein/polysaccharide coacervation, 136–139

Release mechansismsburst, 10–11combination, 10matrix systems, 9–10reservoir-type systems, 9

Release modes, 225

Release rates, 10–11Release triggers, 2

for emulsions, 52–53Release

delayed, 1sustained, 1

SDP method, 101–104Seamless capsules, 194–196Shellac resin, 119Solid fat index, 122Solubilization capacity, 24Solubilization efficacy, 24Spray congealing, 116–117Spray dryers, 117Spray drying, 3–4, 5, 84Steroid alcohols. See PhytosterolsSurfactants, 13

polysorbates and sugar esters, 15Sweeteners, 130

TasteTech, 233Thermal release, 225

U-type microemulsions, 17U-type phase diagram, 16, 17

Vitamin E, 28solubilization capacity, 28–30

Wall-forming materials, 1–2Water binding, 34–35Wax- and fat-coating material,

characteristics ofchain length, 121degree of unsaturation, 121hydrophobicity, 122melting point, 123–124polarity, 121polymorphism, 122–123solid fat index, 122

Wax macro-microemulsions, 120

Xanthum gum, 91

Yeasts, 125–127

Index 239

123456789

101112131415161718192021222324252627282930313233343536373839404142434445

S46N47

Lakkis_Index_235-242 r1.qxd 3/30/07 4:09 PM Page 239